314
Views
30
CrossRef citations to date
0
Altmetric
Original Research

Targeted anticancer potential against glioma cells of thymoquinone delivered by mesoporous silica core-shell nanoformulations with pH-dependent release

, , , , , , & show all
Pages 5503-5526 | Published online: 19 Jul 2019

References

  • Ostrom QT, Gittleman H, Farah P, et al. CBTRUS statistical report: primary brain and central nervous system tumors diagnosed in the United States in 2006-2010. Neuro-Oncology. 2013;15(Suppl 2):ii1–ii56. doi:10.1093/neuonc/not15124137015
  • Wen PY, Kesari S. Malignant gliomas in adults. N Engl J Med. 2008;359(5):492–507. doi:10.1056/NEJMra070812618669428
  • Arko L, Katsyv I, Park GE, Luan WP, Park JK. Experimental approaches for the treatment of malignant gliomas. Pharmacol Ther. 2010;128(1):1–36. doi:10.1016/j.pharmthera.2010.04.01520546782
  • Stupp R, Mason WP, van Den Bent MJ, et al. Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N Engl J Med. 2005;352(10):987–996. doi:10.1056/NEJMoa04333015758009
  • Hou LC, Veeravagu A, Hsu AR, Tse VC. Recurrent glioblastoma multiforme: a review of natural history and management options. Neurosurg Focus. 2006;20(4):E5. doi:10.3171/foc.2006.20.4.2
  • Kim SS, Harford JB, Pirollo KF, Chang EH. Effective treatment of glioblastoma requires crossing the blood-brain barrier and targeting tumors including cancer stem cells: the promise of nanomedicine. Biochem Biophys Res Commun. 2015;468(3):485–489. doi:10.1016/j.bbrc.2015.06.13726116770
  • Nicholas MK, Lukas R, Bangalore S. Chemotherapy for malignant glioma. Expert Rev Neurother. 2005;5(6 Suppl):S41–S49. doi:10.1586/14737175.5.6.S4116274270
  • Perry J, Laperriere N, Zuraw L, Chambers A, Spithoff K, Cairncross JG. Adjuvant chemotherapy for adults with malignant glioma: a systematic review. Can J Neurol Sci. 2007;34(4):402–410.18062446
  • Schneider-Stock R, Fakhoury IH, Zaki AM, El-Baba CO, Gali-Muhtasib HU. Thymoquinone: fifty years of success in the battle against cancer models. Drug Discov Today. 2014;19(1):18–30. doi:10.1016/j.drudis.2013.08.02124001594
  • Goyal SN, Prajapati CP, Gore PR, et al. Therapeutic potential and pharmaceutical development of thymoquinone: a multitargeted molecule of natural origin. Front Pharmacol. 2017;8:656. doi:10.3389/fphar.2017.0065628983249
  • Gali-Muhtasib H, Ocker M, Kuester D, et al. Thymoquinone reduces mouse colon tumor cell invasion and inhibits tumor growth in murine colon cancer models. J Cell Mol Med. 2008;12(1):330–342. doi:10.1111/j.1582-4934.2007.00095.x18366456
  • Jafri SH, Glass J, Shi R, Zhang S, Prince M, Kleiner-Hancock H. Thymoquinone and cisplatin as a therapeutic combination in lung cancer: in vitro and in vivo. J Exp Clin Cancer Res. 2010;29:87. doi:10.1186/1756-9966-29-8720594324
  • El-Mahdy MA, Zhu Q, Wang QE, Wani G, Wani AA. Thymoquinone induces apoptosis through activation of caspase-8 and mitochondrial events in p53-null myeloblastic leukemia HL-60 cells. Int J Cancer. 2005;117(3):409–417. doi:10.1002/ijc.2120515906362
  • Arafa El SA, Zhu Q, Shah ZI, et al. Thymoquinone up-regulates PTEN expression and induces apoptosis in doxorubicin-resistant human breast cancer cells. Mutat Res. 2011;706(1–2):28–35. doi:10.1016/j.mrfmmm.2010.10.00721040738
  • Asaduzzaman Khan M, Tania M, Fu S, Fu J. Thymoquinone, as an anticancer molecule: from basic research to clinical investigation. Oncotarget. 2017;8(31):51907–51919. doi:10.18632/oncotarget.1720628881699
  • Gurung RL, Lim SN, Khaw AK, et al. Thymoquinone induces telomere shortening, DNA damage and apoptosis in human glioblastoma cells. PLoS One. 2010;5(8):e12124. doi:10.1371/journal.pone.001212420711342
  • Kolli-Bouhafs K, Boukhari A, Abusnina A, et al. Thymoquinone reduces migration and invasion of human glioblastoma cells associated with FAK, MMP-2 and MMP-9 down-regulation. Invest New Drugs. 2012;30(6):2121–2131. doi:10.1007/s10637-011-9777-322170088
  • Racoma IO, Meisen WH, Wang QE, Kaur B, Wani AA. Thymoquinone inhibits autophagy and induces cathepsin-mediated, caspase-independent cell death in glioblastoma cells. PLoS One. 2013;8(9):e72882. doi:10.1371/journal.pone.007288224039814
  • Pazhouhi M, Sariri R, Rabzia A, Khazaei M. Thymoquinone synergistically potentiates temozolomide cytotoxicity through the inhibition of autophagy in U87MG cell line. Iran J Basic Med Sci. 2016;19(8):890–898.27746872
  • Elmaci I, Altinoz MA. Thymoquinone: an edible redox-active quinone for the pharmacotherapy of neurodegenerative conditions and glial brain tumors. A short review. Biomed Pharmacother. 2016;83:635–640. doi:10.1016/j.biopha.2016.07.01827459120
  • Chowdhury FA, Hossain MK, Mostofa AGM, Akbor MM, Bin Sayeed MS. Therapeutic potential of thymoquinone in glioblastoma treatment: targeting major gliomagenesis signaling pathways. Biomed Res Int. 2018;2018:4010629. doi:10.1155/2018/401062929651429
  • Jakaria M, Cho D-Y, Ezazul Haque M, et al. Neuropharmacological potential and delivery prospects of thymoquinone for neurological disorders. Oxid Med Cell Longev. 2018;2018:17. doi:10.1155/2018/1209801
  • Banerjee I, De K, Mukherjee D, et al. Paclitaxel-loaded solid lipid nanoparticles modified with Tyr-3-octreotide for enhanced anti-angiogenic and anti-glioma therapy. Acta Biomater. 2016;38:69–81. doi:10.1016/j.actbio.2016.04.02627109765
  • Orunoğlu M, Kaffashi A, Pehlivan SB, et al. Effects of curcumin-loaded PLGA nanoparticles on the RG2 rat glioma model. Mater Sci Eng C. 2017;78:32–38. doi:10.1016/j.msec.2017.03.292
  • Lee ES, Gao Z, Bae YH. Recent progress in tumor pH targeting nanotechnology. J Controlled Release. 2008;132(3):164–170. doi:10.1016/j.jconrel.2008.05.003
  • Liu J, Huang Y, Kumar A, et al. pH-Sensitive nano-systems for drug delivery in cancer therapy. Biotechnol Adv. 2014;32(4):693–710. doi:10.1016/j.biotechadv.2013.11.00924309541
  • Wirries A, Breyer S, Quint K, Schobert R, Ocker M. Thymoquinone hydrazone derivatives cause cell cycle arrest in p53-competent colorectal cancer cells. Exp Ther Med. 2010;1(2):369–375. doi:10.3892/etm_0000005822993551
  • Odeh F, Ismail SI, Abu-Dahab R, Mahmoud IS, Al Bawab A. Thymoquinone in liposomes: a study of loading efficiency and biological activity towards breast cancer. Drug Deliv. 2012;19(8):371–377. doi:10.3109/10717544.2012.72750023043626
  • Ng WK, Saiful Yazan L, Yap LH, Wan Nor Hafiza WAG, How CW, Abdullah R. Thymoquinone-loaded nanostructured lipid carrier exhibited cytotoxicity towards breast cancer cell lines (MDA-MB-231 and MCF-7) and cervical cancer cell lines (HeLa and SiHa). Biomed Res Int. 2015;2015:10. doi:10.1155/2015/263131
  • Dehghani H, Hashemi M, Entezari M, Mohsenifar A. The comparison of anticancer activity of thymoquinone and nanothymoquinone on human breast adenocarcinoma. IJPR. 2015;14(2):539–546.25901162
  • Vallet-Regi M, Rámila A, Del Real RP, Pérez-Pariente J. A new property of MCM-41: drug delivery system. Chem Mater. 2001;13(2):308–311. doi:10.1021/cm0011559
  • Kwon S, Singh RK, Perez RA, Abou Neel EA, Kim H-W, Chrzanowski W. Silica-based mesoporous nanoparticles for controlled drug delivery. J Tissue Eng. 2013;4:2041731413503357. doi:10.1177/204173141350335724020012
  • Wang Y, Zhao Q, Han N, et al. Mesoporous silica nanoparticles in drug delivery and biomedical applications. Nanomedicine. 2015;11(2):313–327. doi:10.1016/j.nano.2014.09.01425461284
  • Watermann A, Brieger J. Mesoporous silica nanoparticles as drug delivery vehicles in cancer. Nanomaterials (Basel, Switzerland). 2017;7(7). doi:10.3390/nano7120458
  • Baghirov H, Karaman D, Viitala T, et al. Feasibility study of the permeability and uptake of mesoporous silica nanoparticles across the blood-brain barrier. PLoS One. 2016;11(8):e0160705. doi:10.1371/journal.pone.016070527547955
  • Tamba BI, Streinu V, Foltea G, et al. Tailored surface silica nanoparticles for blood-brain barrier penetration: preparation and in vivo investigation. Arab J Chem. 2018;11(6):981–990. doi:10.1016/j.arabjc.2018.03.019
  • Shi J, Hou S, Huang J, et al. An MSN-PEG-IP drug delivery system and IL13Rα2 as targeted therapy for glioma. Nanoscale. 2017;9(26):8970–8981. doi:10.1039/c6nr08786h28443896
  • Zhang H, Zhang W, Zhou Y, Jiang Y, Li S. Dual functional mesoporous silicon nanoparticles enhance the radiosensitivity of VPA in glioblastoma. Transl Oncol. 2017;10(2):229–240. doi:10.1016/j.tranon.2016.12.01128193559
  • You Y, Yang L, He L, Chen T. Tailored mesoporous silica nanosystem with enhanced permeability of the blood–brain barrier to antagonize glioblastoma. J Mat Chem B. 2016;4(36):5980–5990. doi:10.1039/C6TB01329E
  • Mo J, He L, Ma B, Chen T. Tailoring particle size of mesoporous silica nanosystem to antagonize glioblastoma and overcome blood–brain barrier. ACS Appl Mater Interfaces. 2016;8(11):6811–6825. doi:10.1021/acsami.5b1173026911360
  • Song Y, Du D, Li L, Xu J, Dutta P, Lin Y. In vitro study of receptor-mediated silica nanoparticles delivery across blood-brain barrier. ACS Appl Mater Interfaces. 2017;9(24):20410–20416. doi:10.1021/acsami.7b0350428541655
  • Salmani JM, Asghar S, Lv H, Zhou J. Aqueous solubility and degradation kinetics of the phytochemical anticancer thymoquinone; probing the effects of solvents, pH and light. Molecules. 2014;19(5):5925–5939. doi:10.3390/molecules1905592524815311
  • El-Toni AM, Khan A, Ibrahim MA, et al. Synthesis of double mesoporous core-shell silica spheres with tunable core porosity and their drug release and cancer cell apoptosis properties. J Colloid Interface Sci. 2012;378(1):83–92. doi:10.1016/j.jcis.2012.04.00622551476
  • Shen D, Yang J, Li X, et al. Biphase stratification approach to three-dimensional dendritic biodegradable mesoporous silica nanospheres. Nano Lett. 2014;14(2):923–932. doi:10.1021/nl404316v24467566
  • Hu F-Q, Zhao M-D, Yuan H, You J, Du Y-Z, Zeng S. A novel chitosan oligosaccharide–stearic acid micelles for gene delivery: properties and in vitro transfection studies. Int J Pharm. 2006;315(1):158–166. doi:10.1016/j.ijpharm.2006.02.02616632285
  • Yuan H, Bao X, Du Y-Z, You J, Hu F-Q. Preparation and evaluation of SiO2-deposited stearic acid-g-chitosan nanoparticles for doxorubicin delivery. Int J Nanomedicine. 2012;7:5119–5128. doi:10.2147/IJN.S3557523055724
  • Yang T-S, Liu -T-T, Lin IH. Functionalities of chitosan conjugated with stearic acid and gallic acid and application of the modified chitosan in stabilizing labile aroma compounds in an oil-in-water emulsion. Food Chem. 2017;228:541–549. doi:10.1016/j.foodchem.2017.02.03528317761
  • Klein M, Aserin A, Ben Ishai P, Garti N. Interactions between whey protein isolate and gum Arabic. Colloids Surf B. 2010;79(2):377–383. doi:10.1016/j.colsurfb.2010.04.021
  • Franz TJ. Percutaneous absorption on the relevance of in vitro data. J Invest Dermatol. 1975;64(3):190–195.123263
  • Salama RO, Traini D, Chan HK, Young PM. Preparation and characterisation of controlled release co-spray dried drug-polymer microparticles for inhalation 2: evaluation of in vitro release profiling methodologies for controlled release respiratory aerosols. Eur J Pharm Biopharm. 2008;70(1):145–152. doi:10.1016/j.ejpb.2008.04.00918534832
  • Ammar HO, Ghorab MM, Mahmoud AA, Higazy IM. Lamotrigine loaded poly-varepsilon-(d,l-lactide-co-caprolactone) nanoparticles as brain delivery system. Eur J Pharm Sci. 2018;115:77–87. doi:10.1016/j.ejps.2018.01.02829341900
  • Mosmann T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J Immunol Methods. 1983;65(1–2):55–63.6606682
  • Yergeau E, Arbour M, Brousseau R, et al. Microarray and real-time PCR analyses of the responses of high-arctic soil bacteria to hydrocarbon pollution and bioremediation treatments. Appl Environ Microbiol. 2009;75(19):6258–6267. doi:10.1128/AEM.01029-0919684169
  • Qu F, Zhu G, Huang S, et al. Controlled release of Captopril by regulating the pore size and morphology of ordered mesoporous silica. Microporous Mesoporous Mater. 2006;92(1–3):1–9. doi:10.1016/j.micromeso.2005.12.004
  • AbouAitah K, Farghali A, Swiderska-Sroda A, Lojkowski W, Razin A, Khedr M. pH-controlled release system for curcumin based on functionalized dendritic mesoporous silica nanoparticles. J Nanomed Nanotechnol. 2016;7:1. doi:10.4172/2157-7439.1000351
  • AbouAitah K, Farghali A, Swiderska-Sroda A, Lojkowski W, Razin A. Mesoporous silica materials in drug delivery system: pH/glutathione-responsive release of poorly water-soluble pro-drug quercetin from two and three-dimensional pore-structure nanoparticles. J Nanomed Nanotechnol. 2016;7:1. doi:10.4172/2157-7439.1000360
  • Sahoo B, Devi KSP, Sahu SK, et al. Facile preparation of multifunctional hollow silica nanoparticles and their cancer specific targeting effect. Biomater Sci. 2013;1(6):647–657. doi:10.1039/c3bm00007a
  • Möller K, Kobler J, Bein T. Colloidal suspensions of nanometer-sized mesoporous silica. Adv Funct Mater. 2007;17(4):605–612
  • Poudel BK, Soe ZC, Ruttala HB, et al. In situ fabrication of mesoporous silica-coated silver-gold hollow nanoshell for remotely controllable chemo-photothermal therapy via phase-change molecule as gatekeepers. Int J Pharm. 2018;548(1):92–103. doi:10.1016/j.ijpharm.2018.06.05629959089
  • Sun Y, Sun Y-L, Wang L, Ma J, Yang Y-W, Gao H. Nanoassembles constructed from mesoporous silica nanoparticles and surface-coated multilayer polyelectrolytes for controlled drug delivery. Microporous Mesoporous Mater. 2014;185:245–253. doi:10.1016/j.micromeso.2013.11.020
  • Khan KA. The concept of dissolution efficiency. J Pharm Pharmacol. 1975;27(1):48–49.235616
  • Rinaki E, Dokoumetzidis A, Macheras P. The mean dissolution time depends on the dose/solubility ratio. Pharm Res. 2003;20(3):406–408.12669960
  • Korsmeyer RW, Gurny R, Doelker E, Buri P, Peppas NA. Mechanisms of solute release from porous hydrophilic polymers. Int J Pharm. 1983;15(1):25–35. doi:10.1016/0378-5173(83)90064-9
  • Karasulu HY, Ertan G, Kose T. Modeling of theophylline release from different geometrical erodible tablets. Eur J Pharm Biopharm. 2000;49(2):177–182. doi:10.1016/S0939-6411(99)00082-X10704902
  • Surekha R, Aishwarya V, Sumathi T. Thymoquinone loaded solid lipid nanoparticle: formulation, characterization and in-vitro cell viability assay. Int J Pharm Bio Sci. 2014;6:449–464.
  • Ritger PL, Peppas NA. A simple equation for description of solute release II. Fickian and anomalous release from swellable devices. J Controlled Release. 1987;5(1):37–42. doi:10.1016/0168-3659(87)90035-6
  • Wang C, Fu X, Yang L. Water-soluble chitosan nanoparticles as a novel carrier system for protein delivery. Chin Sci Bull. 2007;52(7):883–889. doi:10.1007/s11434-007-0127-y
  • Barahuie F, Dorniani D, Saifullah B, et al. Sustained release of anticancer agent phytic acid from its chitosan-coated magnetic nanoparticles for drug-delivery system. Int J Nanomedicine. 2017;12:2361–2372. doi:10.2147/IJN.S12624528392693
  • Williams PA, Phillips GO. 11 - Gum arabic In: Phillips GO, Williams PA, editors. Handbook of Hydrocolloids. 2nd ed. Cambridge: Woodhead Publishing; 2009:252–273.
  • Cornelsen PA, Quintanilha RC, Vidotti M, Gorin PA, Simas-Tosin FF, Riegel-Vidotti IC. Native and structurally modified gum arabic: exploring the effect of the gum’s microstructure in obtaining electroactive nanoparticles. Carbohydr Polym. 2015;119:35–43. doi:10.1016/j.carbpol.2014.11.02025563942
  • Gunasekaran S, Ko S, Xiao L. Use of whey proteins for encapsulation and controlled delivery applications. J Food Eng. 2007;83(1):31–40. doi:10.1016/j.jfoodeng.2006.11.001
  • Abu-Dahab R, Odeh F, Ismail SI, Azzam H, Al Bawab A. Preparation, characterization and antiproliferative activity of thymoquinone-β-cyclodextrin self assembling nanoparticles. Die Pharm-Int J Pharm Sci. 2013;68(12):939–944.
  • Bhattacharya S, Ahir M, Patra P, et al. PEGylated-thymoquinone-nanoparticle mediated retardation of breast cancer cell migration by deregulation of cytoskeletal actin polymerization through miR-34a. Biomaterials. 2015;51:91–107. doi:10.1016/j.biomaterials.2015.01.00725771001
  • AbouAitah K, Swiderska-Sroda A, Farghali AA, et al. Folic acid-conjugated mesoporous silica particles as nanocarriers of natural prodrugs for cancer targeting and antioxidant action. Oncotarget. 2018;9(41):26466–26490. doi:10.18632/oncotarget.2547029899871
  • Choi JY, Ramasamy T, Kim SY, et al. PEGylated lipid bilayer-supported mesoporous silica nanoparticle composite for synergistic co-delivery of axitinib and celastrol in multi-targeted cancer therapy. Acta Biomater. 2016;39:94–105. doi:10.1016/j.actbio.2016.05.01227163403
  • Park EJ, Chauhan AK, Min KJ, Park DC, Kwon TK. Thymoquinone induces apoptosis through downregulation of c-FLIP and Bcl-2 in renal carcinoma Caki cells. Oncol Rep. 2016;36(4):2261–2267. doi:10.3892/or.2016.501927573448
  • Zhang M, Du H, Huang Z, et al. Thymoquinone induces apoptosis in bladder cancer cell via endoplasmic reticulum stress-dependent mitochondrial pathway. Chem Biol Interact. 2018;292:65–75. doi:10.1016/j.cbi.2018.06.01329981725
  • Gali-Muhtasib H, Diab-Assaf M, Boltze C, et al. Thymoquinone extracted from black seed triggers apoptotic cell death in human colorectal cancer cells via a p53-dependent mechanism. Int J Oncol. 2004;25(4):857–866.15375533
  • Xu D, Ma Y, Zhao B, et al. Thymoquinone induces G2/M arrest, inactivates PI3K/Akt and nuclear factor-kappaB pathways in human cholangiocarcinomas both in vitro and in vivo. Oncol Rep. 2014;31(5):2063–2070. doi:10.3892/or.2014.305924603952
  • Alhosin M, Ibrahim A, Boukhari A, et al. Anti-neoplastic agent thymoquinone induces degradation of alpha and beta tubulin proteins in human cancer cells without affecting their level in normal human fibroblasts. Invest New Drugs. 2012;30(5):1813–1819. doi:10.1007/s10637-011-9734-121881916
  • Paramasivam A, Sambantham S, Shabnam J, et al. Anti-cancer effects of thymoquinone in mouse neuroblastoma (Neuro-2a) cells through caspase-3 activation with down-regulation of XIAP. Toxicol Lett. 2012;213(2):151–159. doi:10.1016/j.toxlet.2012.06.01122732633
  • Rajput S, Kumar BN, Dey KK, Pal I, Parekh A, Mandal M. Molecular targeting of Akt by thymoquinone promotes G(1) arrest through translation inhibition of cyclin D1 and induces apoptosis in breast cancer cells. Life Sci. 2013;93(21):783–790. doi:10.1016/j.lfs.2013.09.00924044882
  • Xie Y-T, Du Y-Z, Yuan H, Hu F-Q. Brain-targeting study of stearic acid-grafted chitosan micelle drug-delivery system. Int J Nanomedicine. 2012;7:3235–3244. doi:10.2147/IJN.S3270122802685
  • Ha H-K, Kim JW, Lee M-R, Jun W, Lee W-J. Cellular uptake and cytotoxicity of β-lactoglobulin nanoparticles: the effects of particle size and surface charge. Asian-Aust J Anim Sci. 2015;28(3):420–427. doi:10.5713/ajas.14.0761
  • Bounous G. Whey protein concentrate (WPC) and glutathione modulation in cancer treatment. Anticancer Res. 2000;20(6c):4785–4792.11205219
  • Traverso N, Ricciarelli R, Nitti M, et al. Role of glutathione in cancer progression and chemoresistance. Oxid Med Cell Longev. 2013;2013:10. doi:10.1155/2013/972913
  • Ramasamy T, Ruttala HB, Gupta B, et al. Smart chemistry-based nanosized drug delivery systems for systemic applications: a comprehensive review. J Controlled Release. 2017;258:226–253. doi:10.1016/j.jconrel.2017.04.043