4,440
Views
47
CrossRef citations to date
0
Altmetric
Review Article

Possible role of nanocarriers in drug delivery against cervical cancer

&
Article: 1335567 | Received 17 May 2017, Accepted 20 May 2017, Published online: 07 Jul 2017

References

  • Arbyn M, Castellsagué X, de Sanjosé S, et al. Worldwide burden of cervical cancer in 2008. Ann Oncol. 2011;22:1–25.
  • WHO. Projections of mortality and burden of disease, 2004-2030. 2010. Available from www.who.int/healthinfo/global_burden_disease/projections/en/index.html
  • Nature Biotechnology vol 18 supplement 2000, IT18-20. Available from: http://biotech.nature.com
  • American Cancer Society; Cervical Cancer Fact Sheet. Available from: http://www.cancer.org/acs/groups/content/@nho/documents/document/cervicalcancerpdf.pdf
  • Business  Wire India. Available from: http://www.businesswireindia.com/pressRelease.asp?bmid=28027
  • American Cancer Society. Detailed guide: cervical cancer. ACS. [ cited 2009 Nov 18]. Available from: http://www.cancer.org/doctor/CRI/content/CRI_2_4_1X what is the cervical Cancer_8 asp
  • Eifel PJ, Berek JS, Markman MA. Cancer of the cervix, vagina, and vulva. In: DeVita VT, Hellman S, Rosenberg SA, editors. Cancer: principles and practice of oncology. 8th ed. Philadelphia (PA): Lippincott Williams & Wilkins; 2008. p. 1496–1543.
  • Schiffman M, Castle PE, Jeronimo J, et al. Human papillomavirus and cervical cancer. Lancet. 2007;370:890–907.
  • Zeller JL. Carcinoma of the Cervix. JAMA Patient page. JAMA. 2007;298:23–36.
  • Kaku M, Mathew A, Rajan B. Impact of socio-economic factors in delayed reporting and latestage presentation among patients with cervix cancer in a major cancer hospital in South India. Asian Pac J Cancer Prev. 2008;9:589–594.
  • De Villier EM, Fauquet C, Broker HU, et al. Classification of Papillomavirus. Virol. 2004;324:17–27.
  • Adolfo P, Tanya P, Lilia C, et al. Molecular bases of human papillomavirus pathogenesis in the development of cervical cancer. In: Broeck Dv, editors. Human papillomavirus and related diseases- from bench to bedside – research aspects. Rijeka (Croatia): InTech Publisher; 2012.
  • Gomez DT, Santos JL. Human papillomavirus infection and cervical cancer: pathogenesis and Epidemiology. In: Mendez Vilas A, editors. Communicating current research and educational topics and trends in applied microbiology. Badajoz (Spain): Formatex Pub; 2007.
  • Cervical cancer screening and prevention. 2010 Aug 1. Available from: www.clevelandclinimeded.com/medicalpubs/diseasemanagement/womens-health/cervical-cancer
  • Raybould R, Fiander A, Hibbitts S. Human Papillomavirus integration and its role in cervical malignant progression. Open Clin Cancer J. 2011;5:1–71.
  • Available from: http://www.cancerquest.org/how-hpv-causes-cancer.html
  • Available from: www.cancer.org/cancer/cervical_cancer/detailedguide/cervical_cancer_treating/chemotherapy
  • Li HN, Fei FN, Wei L, et al. Apoptosis induction of oroxylin A in human cervical cancer HeLa cell line in vitro and in vivo. Toxicology. 2009;257:80–85.
  • Hu Y, Yang Y, You QD, et al. Oroxylin A induced apoptosis of human hepatocellular carcinoma cell line Hep G2 was involved in its antitumor activity. Biochem Biophys Res Commun. 2006;351:521–527.
  • Ru-Ming L, Ying-Bo L, Jian-Jiang ZL. Cytotoxic and pro-apoptotic effects of noval ganoderic acid derivative on human cervical cancer cells in vitro. Eur J Pharmacol. 2012;681:23–33.
  • Yao Z, Shulan Z. Inhibition effect of Guizhi-Fuling-decoction on the invasion of human cervical cancer. J Ethnopharmacol. 2008;120:25–35.
  • Fu L, Liu Q, Shen L, et al. Proteomic study on sodium selenite-induced apoptosis of human cervical cancer HeLa cells. J Trace Elem Med Biol. 2011;25:130–137.
  • Shen L, Liu Q, Ni J, et al. A proteomic investigation into the human cervical cancer cell line HeLa treated with dicitratoytterbium (III) complex. Chem Biol Interact. 2009;181:455–462.
  • Abdelwahab SI, Abdul AB, Zain ZNM, et al. Zerumbone inhibits interleukin-6 and induces apoptosis and cell cycle arrest in ovarian and cervical cancer cells. Int Immunopharmacol. 2012;12:594–602.
  • Kim HM, Lim J, Kang JS, et al. Inhibition of human cervical carcinoma growth by cytokine-induced killer cells in nude mouse xenograft model. Int Immunopharmacol. 2009;9:375–380.
  • You BR, Moon HJ, Han YH, et al. Gallic acid inhibits the growth of HeLa cervical cancer cells via apoptosis and/or necrosis. J Food Chem Toxicol. 2010;8:1334–1340.
  • Ting HC, Yen CC, Chen WK, et al. Humic acid enhances the cytotoxic effects of arsenic trioxide on human cervical cancer cells. Environ Toxicol Pharmacol. 2010;29:117–125.
  • Milrot E, Jackman A, Kniazhanski T, et al. Methyl jasmonate reduces the survival of cervical cancer cells and downregulates HPV E6 and E7, and surviving. Cancer Lett. 2012;319:31–38.
  • Kniazhanski T, Jackman A, Heyfets A, et al. Methyl jasmonate induces cell death with mixed characteristics of apoptosis and necrosis in cervical cancer cells. Cancer Lett. 2008;271:34–46.
  • Bertrand N, Leroux JC. The journey of a drug carrier in the body: an anatomo-physiological perspective. J Control Release. 2012;161:152–163.
  • Yohe ST, Herrera VLM, Colson YL, et al. 3D superhydrophobic electrospun meshes as reinforcement materials for sustained local drug delivery against colorectal cancer cells. J Control Release. 2012;162:92–101.
  • Wolinsky JB, Colson YL, Grinstaff MW. Local drug delivery strategies for cancer treatment: gels, nanoparticles, polymeric films, rods, and wafers. J Control Release. 2012;159:14–26.
  • McConville C. The use of localised vaginal drug delivery as part of a neoadjuvant chemotherapy strategy in the treatment of cervical cancer. Gynecol Obstet Res Open J. 2015;2:26–28.
  • Kijanka M, Dorresteijn B, Oliveira S, et al. Nanobody-based cancer therapy of solid tumors. Nanomedicine. 2015;10:161–174.
  • Ye H, Karim AA, Loh XJ. Current treatment options and drug delivery systems as potential therapeutic agents for ovarian cancer: A review. Mater Sci Eng. 2014;45:609–619.
  • Parveen S, Misra R, Sahoo SK. Nanoparticles: a boon to drug delivery, therapeutics, diagnostics and imaging. Nanomedicine. 2012;8:147–166.
  • Ma Y, Huang L, Song C, et al. Nanoparticle formulation of poly(“-caprolactone-colactide)-d–tocopheryl polyethylene glycol 1000 succinate random copolymer for cervical cancer treatment. Polymer. 2010;51:5952–5959.
  • Sun L, Wu Q, Peng F, et al. Strategies of polymeric nanoparticles for enhanced internalization in cancer therapy. Colloids Surf B Biointerfaces. 2015;135:56–72.
  • Siegel RA, Rathbone MJ. Overview of controlled release mechanisms. In: Siepmann J, Siegel AR, Rathbone JM, editors. Fundamentals and applications of controlled release drug delivery. Boston (MA): Springer US; 2012. p. 19–43.
  • Movahedi F, Hu RG, Becker DL, et al. Stimuli-responsive liposomes for the delivery of nucleic acid therapeutics. Nanomedecine. 2015;11:1575–1584.
  • Ta T, Porter TM. Thermosensitive liposomes for localized delivery and triggered release of chemotherapy. J Control Release. 2013;169:112–125.
  • Chen MX, Li BK, Yin DK, et al. Layer-bylayer assembly of chitosan stabilized multilayered liposomes for paclitaxel delivery. Carbohydr Polym. 2014;111:298–304.
  • Casagrande N, De Paoli M, Celegato M, et al. Preclinical evaluation of a new liposomal formulation of cisplatin, lipoplatin, to treat cisplatin-resistant cervical cancer. Gynecol Oncol. 2013;131:744–752.
  • Dou YN, Zheng J, Foltz WD, et al. Heat-activated thermosensitive liposomal cisplatin (HTLC) results in effective growth delay of cervical carcinoma in mice. J Control Release. 2014;178:69–78.
  • Chen JR, Yang YC, Chen TC, et al. Salvage chemotherapy in recurrent cervical cancer with biweekly pegylated liposomal doxorubicin (lipo-dox). Taiwan J Obstet Gynecol. 2008;47:322–326.
  • Corona-Ortega T, Rangel-Corona R, Hernandez-Jimenez M, et al. Characterization of cationic liposomes having IL-2 expressed on their external surface, and their affinity to cervical cancer cells expressing the IL-2 receptor. J Drug Target. 2009;17:496–501.
  • Li X, Ding L, Xu Y, et al. Targeted delivery of doxorubicin using stealth liposomes modified with transferrin. Int J Pharm. 2009;373:116–123.
  • Sriraman SK, Salzano G, Sarisozen C, et al. Anti-cancer activity of doxorubicin-loaded liposomes co-modified with transferrin and folic acid. Eur J Pharm Biopharm. 2016;105:40–49.
  • Rangel-Corona R, Corona-Ortega T, Del Río-Ortiz I, et al. liposomes bearing IL-2 on their external surface induced mice leukocytes to kill human cervical cancer cells in vitro, and significantly reduced tumor burden in immunodepressed mice. J Drug Target. 2011;19:79–85.
  • 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;5:1161–1169.
  • Zeng X, Tao W, Mei L, et al. Cholic acid functionalized nanoparticles of star-shaped PLGA-vitamin E TPGS copolymer for docetaxel delivery to cervical cancer. Biomaterials. 2013;34(25):6058–6067.
  • Dixit N, Kumar V, Pandey RS, et al. Improved cisplatin delivery in cervical cancer cells by utilizing folate-grafted non-aggregated gelatin nanoparticles. Biomed Pharmacother. 2015;69:1–10.
  • Vivero-Escoto JL, Slowing II, Lin VS. Tuning the cellular uptake and cytotoxicity properties of oligonucleotide intercalate or functionalized mesoporous silica nanoparticles with human cervical cancer cells HeLa. Biomaterials. 2010;31:1325–1333.
  • Saini J, Bansal V, Chandra A, et al. Bleomycin sulphate loaded nanostructured lipid particles augment oral bioavailability, cytotoxicity and apoptosis in cervical cancer cells. Colloids Surf B Biointerfaces. 2014;118:101–110.
  • Tran TH, Nguyen CT, Gonzalez-Fajardo L, et al. Long circulating self-assembled nanoparticles from cholesterol-containing brushlike block copolymers for improved drug delivery to tumors. Biomacromolecules. 2014;15:4363–4375.
  • Zhang P, Wu T, Kong JL. In-situ monitoring of intracellular controlled drug release from mesoporous silica nanoparticles coated with pH-responsive charge-reversal polymer. ACS Appl Mater Interfaces. 2014;6:17446–17453.
  • Wang Z, Zeng X, Ma Y, et al. Antitumor efficiency of D-alphatocopheryl polyethylene glycol 1000 succinate-b-poly(epsiloncaprolactone-ran-lactide) nanoparticle-based delivery of docetaxel in mice bearing cervical cancer. J Biomed Nanotechnol. 2014;10:1509–1519.
  • Xiong Q, Zhang M, Zhang Z, et al. Anti-tumor drug delivery system based on cyclodextrin-containing pHresponsive star polymer: in-vitro and in-vivo evaluation. Int J Pharm. 2014;474:232–240.
  • Zhao C, Liu X, Liu J, et al. Transferrin conjugated poly (γ-glutamic acid-maleimide-co-llactide)-1,2-dipalmitoylsn-glycero-3-phosphoethanolamine copolymer nanoparticles for targeting drug delivery. Colloids Surf B Biointerfaces. 2014;14:571–572.
  • Byagari K, Shanavas A, Rengan AK, et al. Biocompatible amphiphilic pentablockcopolymeric nanoparticles for anti-cancer drug delivery. J Biomed Nanotechnol. 2014;10:109–119.
  • Kim TH, Lee GJ, Kang JH, et al. Anticancer drug-incorporated layered double hydroxide nanohybrids and their enhanced anticancer therapeutic efficacy in combination cancer treatment. Biomed Res Int. 2014;2014:193401.
  • Hiorth M, Liereng L, Reinertsen R, et al. Formulation of bioadhesive hexyl amino levulinate pellets intended for photodynamic therapy in the treatment of cervical cancer. Int J Pharm. 2013;441:544–554.
  • Sarisozen C, Abouzeid AH, Torchilin VP. The effect of co-delivery of paclitaxel and curcumin by transferring-targeted PEG-PE-based mixed micelles on resistant ovarian cancer in 3-D spheroids and in vitro tumors. Eur J Pharm Biopharm. 2014;88:539–550.
  • Tan YL, Liu CG. Preparation and characterization of self-assembled nanoparticles based on folic acid modified carboxymethyl chitosan. J Mater Sci Mater Med. 2011;22:1213–1220.
  • Liu CG, Desai KG, Chen XG, et al. Linolenic acid-modified chitosan for formation of self-assembled nanoparticles. J Agric Food Chem. 2005;53:437–441.
  • Danhier F, Lecouturier N, Vroman B, et al. Paclitaxel-loaded PEGylated PLGA-based nanoparticles: in vitro and in vivo evaluation. J Control Release. 2009;133:11–17.
  • Yuandong MA, Zheng Y, Liu K, et al. Nanoparticles of Poly(Lactide-Co-Glycolide)-d-a-Tocopheryl Polyethylene Glycol 1000 succinate random copolymer for cancer treatment. Nanoscale Res Lett. 2010;5:1161–1169.
  • Kim SY, Cho SH, Lee YM. Biotin-conjugated block copolymeric nanoparticles as tumor-targeted drug delivery systems. Macromolecular Res. 2007;15:646–655.
  • Gu Q, Xing JZ, Huang M, et al. Nanoformulation of paclitaxel to enhance cancer therapy. J Biomater Appl. 2013;28:298–307.
  • Zhang J, Chen XG, Li YY, et al. Self-assembled nanoparticles based on hydrophobically modified chitosan as carriers for doxorubicin. Nanomedicine: Nanotechnology, Biol Med. 2007;3:258–265.
  • 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;6:666.
  • 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;10:1509–1519.
  • Pimentel RC, Martínez ESM, García AM, et al. Silver nanoparticles nanocarriers, synthesis and toxic effect on cervical cancer cell lines. Bio Nano Science. 2013;3:198–207.
  • 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;23:1830–1837.
  • Ditto AJ, Shah KN, Robishaw NK, et al. The interactions between L-tyrosine based nanoparticles decorated with folic acid and cervical cancer cells under physiological flow. Mol Pharm. 2012;9:3089–3098.
  • Yu B, Li H, Zhang J, et al. Rational design and fabrication of a cancer-targeted chitosan nanocarrier to enhance selective cellular uptake and anticancer efficacy of selenocystine. J Mater Chem B. 2015;3:2497–2504.
  • Ji J, Zuo P, Wang YL. Enhanced antiproliferative effect of carboplatin in cervical cancer cells utilizing folate-grafted polymeric nanoparticles. Nanoscale Res Lett. 2015;10:453.
  • Tang H, Feng X, Zhang T, et al. Stability, pharmacokinetics, biodistribution and safety assessment of folate-conjugated pullulan acetate nanoparticles as cervical cancer targeted drug carriers. J Nanosci Nanotechnol. 2015;15:6405–6412.
  • Iijima S, Ichihashi T. Single-shell carbon nanotubes of 1-nm diameter. Nature. 1993;363:603–605.
  • Morinobu E, Dresselhaus MS. Carbon fibers and carbon nanotubes. Nagano & Cambridge (MA); 2002. Available from https://stuff.mit.edu/
  • Abrahamsona J, Wilesa PG, Rhoadesb BL. Structure of carbon fibres found on carbon arc anodes. Carbon. 1999;37:1873–1874.
  • Zhang X, Meng L, Lu Q, et al. Targeted delivery and controlled release of doxorubicin to cancer cells using modified single wall carbon nanotubes. Biomaterials. 2009;30:6041–6047.
  • Mahmood M, Karmakar A, Fejleh A, et al. Synergistic enhancement of cancer therapy using a combination of carbon nanotubes and anti-tumor drug. Nanomedicine. 2009;4:883–893.
  • Bhatnagar I, Venkatesan J, Kiml SK. Polymer functionalized single walled carbon nanotubes mediated drug delivery of gliotoxin in cancer cells. J Biomed Nanotechnol. 2014;10:120–130.
  • Hu Q, van Gaal EVB, Brundel P, et al. A novel approach for the intravenous delivery of leuprolide using core-cross-linked polymeric micelles. J Control Release. 2015;205:98–108.
  • Sutton D, Nasongkla N, Blanco E, et al. Functionalized micellar systems for cancer targeted drug delivery. Pharm Res. 2007;24:1029–1046.
  • Chen F, Dong D, Fu F, et al. Anti-tumor activity of biodegradable polymer-paclitaxel conjugated micelle against mice U14 cervical cancers. Chem Res Chin Univ. 2012;28:656–661.
  • Guo Q, Guan D, Dong B, et al. Charge-conversional binary drug delivery polymeric micelles for combined chemotherapy of cervical cancer. Int J Polym Mater Polym Biomater. 2015;64:978–987.
  • Melo-Diogo D, Gaspar VM, Costa EC, et al. Combinatorial delivery of crizotinib-palbociclib-sildenafil using TPGS-PLA micelles for improved cancer treatment. Eur J Pharm Biopharm. 2014;88:718–729.
  • Feng C, Dan D, Yan FU, et al. Anti-tumor activity of biodegradable polymer-paclitaxel conjugated micelle against mice U14 cervical cancers. Chem Res Chinese Universities. 2012;28:656–661.
  • Waldmann TA. Immunotherapy: past, present and future. Nat Med. 2003;9:269–277.
  • Janeway CA, Travers P, Walport M. Immunobiology, 6th ed. London: Churchill Livingstone; 2005. [ISBN 0-443-07310-4].
  • 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;101:782–791.
  • Monk BJ, Mas Lopez L, Zarba JJ, et al. Phase II, open-label study of pazopanib or lapatinib monotherapy compared with pazopanib plus lapatinib combination therapy in patients with advanced and recurrent cervical cancer. J Clin Oncol. 2010;28:3562–3569.
  • Shen L, Liu Q, Ni J, et al. A proteomic investigation into the human cervical cancer cell line HeLa treated with dicitratoytterbium (III) complex. Chem Biol Interact. 2009;181:455–462.
  • Abdelwahab SI, Abdul AB, Zain ZN, et al. Zerumbone inhibits interleukin-6 and induces apoptosis and cell cycle arrest in ovarian and cervical cancer cells. Int Immunopharmacol. 2012;12:594–602.
  • Liping F, Liu Q, Shen L, et al. Proteomic study on sodium selenite-induced apoptosis of human cervical cancer HeLa cells. J Trace Elem Med Biol. 2011;25:130–137.
  • Kesharwani P, Banerjee S, Gupta U, et al. Pamam dendrimers as promising nanocarriers for RNAi therapeutics. Mater. Today. 2015;18:565–572.
  • Ordikhani F, Erdem Arslan M, Marcelo R, et al. Drug delivery approaches for the treatment of cervical cancer. Pharmaceutics. 2016;8:23–37.
  • Liu TY, Hussein WM, Jia Z, et al. Self-adjuvanting polymer–peptide conjugates as therapeutic vaccine candidates against cervical cancer. Biomacromol. 2013;14:2798–2806.
  • Mekuria SL, Debele TA, Chou HY, et al. Il-6 antibody and RGD peptide conjugated poly(amidoamine) dendrimer for targeted drug delivery of HeLa cells. J Phys Chem B. 2016;120:123–130.
  • Sarbolouki MN, Sadeghizadeh M, Yaghoobi MM, et al. Dendrosomes: a novel family of vehicles for transfection and therapy. J Chem Tech Biotechnol. 2000;75:919–922.
  • Tathagata D, Aghase HB, Vijayarajkumar P, et al. Dendrosome based gene delivery. J Exp Nanoscience. 2006;1:235–248.
  • Dutta T, Burgess M, McMillan NA, et al. Dendrosome-based delivery of siRNA against E6 and E7 oncogenes in cervical cancer. Nanomedicine: Nanotechnology, Biol Med. 2010;6:463–470.
  • Wolinsky JB, Colson YL, Grinstaff MW. Local drug delivery strategies for cancer treatment: gels, nanoparticles, polymeric films, rods, and wafers. J Control Release. 2012;159:14–26.
  • Williams NL, Werner TL, Jarboe EA, et al. Adenocarcinoma of the cervix: should we treat it differently? Curr Oncol Rep. 2015;17:1–10.
  • Keskar V, Mohanty PS, Gemeinhart EJ, et al. Cervical cancer treatment with a locally insertable controlled release delivery system. J Control Release. 2006;115:280–288.
  • Blakney AK, Ball C, Krogstad EA, et al. Electrospun fibers for vaginal anti-HIV drug delivery. Antivir Res. 2013;100(Suppl.):S9–S16.
  • Caramella CM, Rossi S, Ferrari F, et al. Mucoadhesive and thermogelling systems for vaginal drug delivery. Adv Drug Deliv Rev. 2015;92:39–52.
  • Ensign LM, Cone R, Hanes J. Nanoparticle-based drug delivery to the vagina: a review. J Control Release. 2014;190:500–514.
  • McConville C. The use of localised vaginal drug delivery as part of a neoadjuvant chemotherapy strategy in the treatment of cervical cancer. Gynecol Obstet Res Open J. 2015;2:26–28.
  • Bahram M, Hoseinzadeh F, Farhadi K, et al. Synthesis of gold nanoparticles using pH-sensitivehydrogel and its application for colorimetric determination of acetaminophen, ascorbic acid and folic acid. Colloids Surf A Physicochem Eng Asp. 2014;441:517–524.
  • Caramella CM, Rossi S, Ferrari F, et al. Mucoadhesive and thermogelling systems for vaginal drug delivery. Adv Drug Deliv Rev. 2015;92:39–52.
  • Giri TK, Thakur A, Alexander A, et al. Modified chitosan hydrogels as drug delivery and tissue engineeringsystems: present status and applications. Acta Pharm Sin B. 2012;2:439–449.
  • Perez E, Fernandez A, Olmo R, et al. pH and glutathione-responsive hydrogel for localized delivery of paclitaxel. Colloids Surf B Biointerfaces. 2014;116:247–256.
  • Jaiswal MK, Pradhan A, Banerjee R, et al. Dual pH and temperature stimuli-responsive magnetic nanohydrogels for thermo-chemotherapy. J Nanosci Nanotechnol. 2014;14:4082–4089.
  • Curry T, Epstein T, Smith R, et al. Photothermal therapy of cancer cells mediated by blue hydrogel nanoparticles. Nanomedicine. 2013;8:1577–1586.
  • Sami H, Kumar A. Tunable hybrid cryogels functionalized with microparticles as supermacroporous multifunctional biomaterial scaffolds. J Biomater Sci Polym Ed. 2013;24:1165–1184.
  • Nazli C, Ergenc TI, Yar Y, et al. RGDS functionalized polyethylene glycol hydrogel-coated magnetic iron oxide nanoparticles enhance specific intracellular uptake by HeLa cells. Int J Nanomed. 2012;7:1903–1920.
  • Bilensoy E, Cirpanli Y, Sen M, et al. Thermosensitive mucoadhesive gel formulation loaded with 5-Fu: cyclodextrin complex for HPV-induced cervical cancer. J Incl Phenom Macrocycl Chem. 2007;57:363–370.
  • Chun CJ, Lee SM, Kim SY, et al. Thermosensitive poly(organophosphazene)–paclitaxel conjugate gels for antitumor applications. Biomaterials. 2009;30:2349–2360.
  • Seo SH, Han HD, Noh KH, et al. Chitosan hydrogel containing GMCSF and a cancer drug exerts synergistic anti-tumor effects via the induction of CD8+ T cell-mediated anti-tumor immunity. Clin Exp Metastasis. 2009;26:179–187.
  • Collaud S, Peng Q, Gurny R, et al. Thermosetting gel for the delivery of 5-aminolevulinic acid esters to the cervix. J Pharm Sci. 2008;97:2680–2690.
  • Hani U, Osmani RA, Bhosale RR, et al. Current perspectives on novel drug delivery systems and approaches for management of cervical cancer: a comprehensive review. Curr Drug Targets. 2016;17:337–352.
  • Saltzman WM, Olbricht WL. Building drug delivery into tissue engineering. Nat Rev Drug Discov. 2002;1:177–186.
  • Langer R. Biomaterials in drug delivery and tissue engineering: one laboratory’s experience. Acc Chem Res. 2000;33:94–101.
  • Atala A. Tissue engineering and regenerative medicine: concepts for clinical application. Rejuvenation Res. 2004;7:15–31.
  • Garg T, Singh O, Arora S, et al. Scaffold: a novel carrier for cell and drug delivery. Critical reviews™. Ther Drug Carrier Syst. 2012;29:1–63.
  • Hu X, Liu S, Zhou G, et al. Electrospinning of polymeric nanofibers for drug delivery applications. J Control Release. 2014;185:12–21.
  • Li SF, Liu DX, Zheng YH, et al. Inhibitory effects of paclitaxel-loaded PLA nanofibers against mice cervical cancers. Acta Polym Sin. 2012;12:1029–1034.
  • Zong S, Wang X, Yang Y, et al. The use of cisplatin-loaded mucoadhesive nanofibers for local chemotherapy of cervical cancers in mice. Eur J Pharm Biopharm. 2015;93:127–135.
  • Keskar V, Mohanty PS, Gemeinhart EJ, et al. Cervical cancer treatment with a locally insertable controlled release delivery system. J Control Release. 2006;115:280–288.
  • Woolfson AD, McCafferty DF, McCarrona PA, et al. A bioadhesive patch cervical drug delivery system for the administration of 5-fluorouracil to cervical tissue. J Control Release. 1995;35:49–58.
  • Iwata M, Machida Y, Nagai T, et al. Double-layered stick-type formulation of bleomycin for treatment of uterine cervical cancer. Drug Des Deliv. 1987;1:253–260.
  • McCarron PA, Woolfson AD, McCafferty DF, et al. Cytotoxicity of 5-fluorouracil released from a bioadhesive patch into uterine cervical tissue. Int J Pharm. 1997;151:69–74.
  • Keskar V, Mohanty PS, Gemeinhart EJ, et al. Cervical cancer treatment with a locally insertable controlled release delivery system. J Control Release. 2006;115:280–288.
  • Choi SH, Lee SH, Park TG. Temperature-sensitive Pluronic/poly(ethylenimine) nanocapsules for thermally triggered disruption of intracellular endosomal compartment. Biomacromolecules. 2006;7: 1864–1870.
  • Lee Y, Park SY, Kim C, et al. Thermally triggered intracellular explosion of volume transition nanogels for necrotic cell death. J Control Release. 2009;135:89–95.
  • Choi SH, Lee JH, Choi SM, et al. Thermally reversible Pluronic/heparin nanocapsules exhibiting 1000-fold volume transition. Langmuir. 2006;22:1758–1762.
  • Saengkrit N, Saesoo S, Srinuanchai W, et al. Influence of curcumin-loaded cationic liposome on anticancer activity for cervical cancer therapy. Colloids Surf B Biointerfaces. 2014;114:349–356.
  • Berginc K, Suljakovic S, Skalko-Basnet N, et al. Mucoadhesive liposomes as new formulation for vaginal delivery of curcumin. Eur J Pharm Biopharm. 2014;87:40–46.
  • Krishnakumar N, Sulfikkarali N, Prasad NR, et al. Enhanced anticancer activity of naringenin-loaded nanoparticles in human cervical. Biomed Prev Nutr. 2011;1:223–231.
  • Jeyaraj M, Rajesh M, Arun R, et al. An investigation on the cytotoxicity and caspase-mediated apoptotic effect of biologically synthesized silver nanoparticles using Podophyllum hexandrum on human cervical carcinoma cells. Colloids Surf B Biointerfaces. 2013;102:708–717.
  • Jeyaraj M, Arun R, Sathishkumar G, et al. An evidence on G2/M arrest, DNA damage and caspase mediated apoptotic effect of biosynthesized gold nanoparticles on human cervical carcinoma cells (HeLa). Materials Res Bull. 2014;52:15–24.
  • Punfa W, Suzuki S, Pitchakarn P, et al. Curcumin-loaded PLGA nanoparticles conjugated with anti-P-glycoprotein antibody to overcome multidrug resistance. Asian Pac J Cancer Prev. 2014;15:9249–9258.
  • Yang R, Chen D, Li M, et al. 20(s)-ginsenoside Rg3-loaded magnetic human serum albumin nanospheres applied to HeLa cervical cancer cells in-vitro. Biomed Mater Eng. 2014;24(6):1991–1998.
  • Namvar F, Rahman HS, Mohamad R, et al. Cytotoxic effect of magnetic iron oxide nanoparticles synthesized via seaweed aqueous extract. Int J Nanomed. 2014;9:2479–2488.
  • Ignatova M, Manolova N, Toshkova R, et al. Quaternized chitosan coated nanofibrous materials containing gossypol: preparation by electrospinning, characterization and antiproliferative activity towards HeLa cells. Int J Pharm. 2012;436:10–24.
  • Punfa W, Yodkeeree S, Pitchakarn P, et al. Enhancement of cellular uptake and cytotoxicity of curcumin-loaded PLGA nanoparticles by conjugation with anti-P-glycoprotein in drug resistance cancer cells. Acta Pharmacol Sin. 2012;33:823–831.
  • Nair KL, Thulasidasan AK, Deepa G, et al. Purely aqueous PLGA nanoparticulate formulations of curcumin exhibit enhanced anticancer activity with dependence on the combination of the carrier. Int J Pharm. 2012;425:44–52.
  • Altunbas A, Lee SJ, Rahasekaran SA, et al. Encapsulation of curcumin in self-assembling peptide hydrogels as injectable drug delivery vehicles. Biomaterials. 2011;32:5906–5914.
  • Wu W, Shen J, Banerjee P, et al. Water-dispersible multifunctional hybrid nanogels for combined curcumin and photothermal therapy. Biomaterials. 2011;32:598–609.
  • Deepa G, Thulasidasan AK, Anto RJ. Crosslinked acrylic hydrogel for the controlled delivery of hydrophobic drugs in cancer therapy. Int J Nanomed. 2012;7:4077–4088.
  • Gonçalves C, Pereira P, Schellenberg P, et al. Self-assembled dextrin nanogel as curcumin delivery system. J Biomater Nanobiotechnol. 2012;3:178–184.
  • Goncalves C, Martins JA, Gama FM. Self-assembled nanoparticles of dextrin substituted with hexa-decanethiol. Biomacromolecules. 2007;8:392–398.
  • Hani U, Shivakumar HG, Anjum H, et al. Preparation and optimization of curcumin-hydroxypropyl-cyclodextrin bioadhesive vaginal films for human papilloma virus-induced cervical cancer. J Biomater Tissue Eng. 2014;4:796–803.
  • Li GL, Jiang W, Xia Q, et al. HPV E6 down-regulation and apoptosis induction of human cervical cancer cells by a novel lipid-soluble extract (PE) from Pinellia pedatisecta Schott in vitro. J Ethnopharmacol. 2010;132:56–64.
  • Kajani AA, Zarkesh-Esfahani SH, Bordbar A, et al. Anticancer effects of silver nanoparticles encapsulated by Taxus baccata extracts. J Mol Liq. 2016. DOI:10.1016/j.molliq.2016.08.064
  • Maeda H, Wu J, Sawa T, et al. Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review. J Control Rel. 2000;65(1–2):271–284.
  • Foulsham A, Trussler C, Barker C, et al. Radiotherapy for cervix cancer. 2012 ed. Oxford: Oxford University Hospitals NHS Trust; 2012.
  • Huang Y, Luo Y, Zheng W, et al. Rational design of cancertargeted BSA protein nanoparticles as radiosensitizer to overcome cancer radioresistance. ACS Appl Mater Interfaces. 2014;6:19217–19228.
  • Chechetka SA, Pichon B, Zhang M, et al. Multifunctional carbon nanohorn complexes for cancer treatment. Chem Asian J. 2015;10:1160–1165.
  • Geng F, Xing JZ, Chen J, et al. Pegylated glucose gold nanoparticles for improved in-vivo bio-distribution and enhanced radiotherapy on cervical cancer. Biomed Nanotechnol. 2014;10:1205–1216.
  • Topete A, Alatorre-Meda M, Villar-Alvarez EM, et al. Polymericgold nanohybrids for combined imaging and cancer therapy. Adv Healthc Mater. 2014;3:1309–1325.
  • Bai J, Liu Y, Jiang X. Multifunctional PEG-GO/CuS nanocomposites for near-infrared chemo-photothermal therapy. Biomaterials. 2014;35:5805–5813.
  • Liu Y, Bai J, Jia X, et al. Fabrication of multifunctional SiO2-GN-Serum composites for chemo-photothermal synergistic therapy. ACS Appl Mater Interfaces. 2015;7:112–121.
  • Lu W, Zhang G, Zhang R, et al. Tumor site-specific silencing of NF-kappaB p65 by targeted hollow gold nanospheres mediated photothermal transfection. Cancer Res. 2010;70:3177–3188.
  • Abdulla-Al-Mamun M, Kusumoto Y, Mihata A, et al. Plasmon-induced photothermal cell-killing effect of gold colloidal nanoparticles on epithelial carcinoma cells. Photochem Photobiol Sci. 2009;8:1125–1129.
  • Diniz MO, Cariri FA, Aps LR, et al. Enhanced therapeutic effects conferred by an experimental DNA vaccine targeting human papillomavirus-induced tumors. Hum Gene Ther. 2013;24:861–870.
  • Sadraeian M, Rasoul-Amini S, Mansoorkhani MJ, et al. Induction of antitumor immunity against cervical cancer by protein HPV-16 E7 in fusion with ricin B chain in tumor-bearing mice. Int J Gynecol Cancer. 2013;23:809–814.
  • Martin Caballero J, Garzon A, Gonzalez-Cintado L, et al. Chimeric infectious bursal disease virus-like particles as potent vaccines for eradication of established HPV-16 E7-dependent tumors. Plos One. 2012;7:e52976.
  • Banerjee S, Sahoo AK, Chattopadhyay A, et al. Recombinant IκBα-loaded curcumin nanoparticles for improved cancer therapeutics. Nanotechnology. 2014;25:345102.
  • Liu B, Han SM, Tang XY, et al. Cervical cancer gene therapy by gene loaded PEG-PLA nanomedicine. Asian Pac J Cancer Prev. 2014;15:4915–4918.
  • Zhen S, Hua L, Takahashi Y, et al. In-vitro and in-vivo growth suppression of human papillomavirus 16-positive cervical cancer cells by CRISPR/Cas9. Biochem Biophys Res Commun. 2014;50:1422–1426.
  • Madrigal M, Janicek MF, Sevin BU, et al. In-vitro antigene therapy targeting HPV-16 E6 and E7 in cervical carcinoma. Gynecol Oncol. 1997;64:18–25.
  • Baldwin PJ, van der Burg SH, Boswell CM, et al. Vacciniaexpressed human papillomavirus 16 and 18 e6 and e7 as a therapeutic vaccination for vulval and vaginal intraepithelial neoplasia. Clin Cancer Res. 2003;9:5205–5213.
  • Wang XG, Jandl T, Dadachova E, et al. Effect of naive and radiolabeled rhTRAIL on the cervical cancer xenografts in mice. Ther Deliv. 2014;5:139–147.
  • Zheng Y, Chen H, Zeng X, et al. Surface modification of TPGS-b-(PCL-ran PGA) nanoparticles with polyethyleneimine as a co-delivery system of TRAIL and endostatin for cervical cancer gene therapy. Nanoscale Res Lett. 2013;8:161.