623
Views
18
CrossRef citations to date
0
Altmetric
Original Research

Evaluation of Antitumor Efficacy of Chitosan-Tamarind Gum Polysaccharide Polyelectrolyte Complex Stabilized Nanoparticles of Simvastatin

, , , , , , , , , , ORCID Icon & show all
Pages 2533-2553 | Published online: 29 Mar 2021

References

  • U.S. Breast Cancer Statistics. Ardmore, PA: Breastcancer.org; 2021. Available from: https://www.breastcancer.org/symptoms/understand_bc/statistics. Accessed December 1, 2020.
  • Aung TN, Qu Z, Kortschak RD, et al. Understanding the effectiveness of natural compound mixtures in cancer through their molecular mode of action. Int J Mol Sci. 2017;18:1–20. doi:10.3390/ijms18030656
  • Aravind SR, Joseph MM, Varghese S, et al. Antitumor and immunopotentiating activity of polysaccharide PST001 isolated from the seed kernel of Tamarindus indica: an in vivo study in mice. Sci World J. 2012;2012:1–14. doi:10.1100/2012/361382
  • Walker JF. Simvastatin: the clinical profile. Am J Med. 1989;87:S44–S46. doi:10.1016/s0002-9343(89)80598-4
  • Wang ST, Ho HJ, Lin JT, et al. Simvastatin-induced cell cycle arrest through inhibition of STAT3/SKP2 axis and activation of AMPK to promote p27 and p21 accumulation in hepatocellular carcinoma cells. Cell Death Dis. 2017;8:e2626. doi:10.1038/cddis.2016.472
  • Hwang KE, Na KS, Park DS, et al. Apoptotic induction by simvastatin in human lung cancer A549 cells via Akt signaling dependent down-regulation of survivin. Invest New Drugs. 2011;29:945–952. doi:10.1007/s10637-010-9450-2
  • Joseph J, Kanchalochana SN, Rajalakshmi G, et al. Tamarind seed polysaccharide: a promising natural excipient for pharmaceuticals. Int J Green Pharm. 2012;6:270–278. doi:10.4103/0973-8258.108205
  • Shao H, Zhang H, Tian Y, et al. Composition and rheological properties of polysaccharide extracted from tamarind (Tamarindus indica L.) seed. Molecules. 2019;24:1–13. doi:10.3390/molecules24071218
  • Chawananorasest K, Saengtongdee P, Kaemchantuek P. Extraction and characterization of tamarind (Tamarind indica L.) seed polysaccharides (TSP) from three difference sources. Molecules. 2016;21:1–9. doi:10.3390/molecules21060775
  • Singh R, Malviya R, Sharma PK. Extraction and characterization of tamarind seed polysaccharide as a pharmaceutical excipient. Pharmacogn J. 2011;3:17–19. doi:10.5530/pj.2011.20.4
  • Katiyar N, Malviya R, Sharma PK. Pharmaceutical applications and formulation based patents of tamarindus indica seed polysaccharide and their modified derivatives. Adv Biol Res. 2014;8(6):274–281.
  • Verma S, Bansal J, Kumar N, et al. Isolation and characterization studies of mucilage obtained from trigonella foenum greacum l. seed and Tamarindus indica polysaccharide as a pharmaceutical excipient. J Drug Deliv Ther. 2014;4(3):106–109.
  • Parhi R. Drug delivery applications of chitin and chitosan: a review. Environ Chem Lett. 2020;18:577–594. doi:10.1007/s10311-020-00963-5
  • de Sousa Victor R, Marcelo da Cunha Santos A, Viana de Sousa B, de Araújo Neves G, Navarro de Lima Santana L, Rodrigues Menezes R. A review on Chitosan’s uses as biomaterial: tissue engineering, drug delivery systems and cancer treatment. Materials. 2020;13:4995. doi:10.3390/ma13214995
  • Adhikari HS, Yadav PN. Anticancer activity of chitosan, chitosan derivatives, and their mechanism of action. Int J Biomater. 2018;2018:1–29. doi:10.1155/2018/2952085
  • Wimardhani YS, Suniarti DF, Freisleben HJ, et al. Chitosan exerts anticancer activity through induction of apoptosis and cell cycle arrest in oral cancer cells. J Oral Sci. 2014;56:119–126. doi:10.2334/josnusd.56.119
  • Savjani KT, Gajjar AK, Savjani JK. Drug solubility: importance and enhancement techniques. ISRN Pharm. 2012;2012:195727. doi:10.5402/2012/195727
  • Tambosi G, Coelho PF, Luciano S, et al. Challenges to improve the biopharmaceutical properties of poorly water-soluble drugs and the application of the solid dispersion technology. Matéria. 2018;23(4):e12224. doi:10.1590/s1517-707620180004.0558
  • Pucci C, Martinelli C, Ciofani G. Innovative approaches for cancer treatment: current perspectives and new challenges. E Cancer Med Sci. 2019;13:1–26.
  • Patra JK, Das G, Fraceto LF, et al. Nano based drug delivery systems: recent developments and future prospects. J Nanobiotechnology. 2018;16(1):71.
  • Lai WF, Shum HC. Hymermallose graft chitosan and its polyelectrolyte complex as novel systems for sustained drug delivery. ACS Appl Mater Interfaces. 2015;7:10501–10510. doi:10.1021/acsami.5b01984
  • Lankalapalli S, Kolapalli VR. Polyelectrolyte complexes: a review of their applicability in drug delivery technology. Indian J Pharm Sci. 2009;71(5):481–487. doi:10.4103/0250-474X.58165
  • Akbari-Alavijeh S, Shaddel R, Jafari SM. Nanostructures of chitosan for encapsulation of food ingredients. In: Biopolymer Nanostructures for Food Encapsulation Purposes. Academic Press; 2019:381–418.
  • Rajabi H, Jafari SM, Rajabzadeh G, et al. Chitosan-gum Arabic complex nanocarriers for encapsulation of saffron bioactive components. Colloids Surf A. 2019;578:123644.
  • Patwekar SL, Potulwar AP, Pedewad SR, et al. Review on polyelectrolyte complex as novel approach for drug delivery system. Int J Pharm Pharm Res. 2016;5:98–109.
  • Potaś J, Szymańska E, Winnicka K. Challenges in developing of chitosan – based polyelectrolyte complexes as a platform for mucosal and skin drug delivery. Eur Polym J. 2020;140:110020. doi:10.1016/j.eurpolymj.2020.110020
  • Dakhara SL, Anajwala C. Polyelectrolyte complex: a pharmaceutical review. Sys Rev Pharm. 2010;1:121–127. doi:10.4103/0975-8453.75046
  • Mark HF, Bikales NM, Overberger CG, et al. Encyclopedia of Polymer Science. John Wiley and Sons, A Willey Interscience Publishers; 1987:739.
  • Jindal AB. The effect of particle shape on cellular interaction and drug delivery applications of micro- and nanoparticles. Int J Pharm. 2017;532:450–465. doi:10.1016/j.ijpharm.2017.09.028
  • Venkataramana K. Formulation characterization and in vitro, in vivo evaluation of stabilized rosuvastatin calcium nanosuspension. Int J Res Pharm Sci. 2020;11:2657–2664. doi:10.26452/ijrps.v11i2.2280
  • Verma A, Nagarwal RC, Sharma SD, et al. Preparation and characterization of floating gellan-chitosan polyelectrolyte complex beads. Lat Am J Pharm. 2012;31:138–146.
  • Manchanda R, Arora SC, Manchanda R. Tamarind seed polysaccharide and its modifications-versatile pharmaceutical excipients-A review. Int J Pharm Technol Res. 2014;6:412–420.
  • Malviya R, Sharma PK, Dubey SK. Efficiency of self-assembled etoricoxib containing polyelectrolyte complex stabilized cubic nanoparticles against human cancer cells. Prec Med Sci. 2020;9:9–22. doi:10.1002/prm2.12004
  • Yousefpour P, Atyabi F, Dinarvand R, et al. Preparation and comparison of chitosan nanoparticles with different degrees of glutathione thiolation. DARU: J Fac Pharm Tehr Unv Med Sci. 2011;19:367–375.
  • Cao D, Gong S, Shu X, et al. Preparation of ZnO nanoparticles with high dispersibility based on oriented attachment (OA) process. Nanoscale Res Lett. 2019;14:1–11. doi:10.1186/s11671-019-3038-3
  • Kumar A, Shinde J, Harinath N. Formulation, development and characterization of Simvastatin nanoparticles by solvent displacement method. Der Pharm Lett. 2014;6:145–155.
  • Why are DLS measurements in high concentration solutions difficult?. Holtsville, NY: Brookhaven Instruments Corporation; 2020. Available from: https://www.spectraresearch.com/wp-content/uploads/2019/08/DLS-High-Concentration.pdf. Accessed November 15, 2020.
  • Guide for Dynamic Light Scattering (DLS) sample preparation. Holtsville, NY: Brookhaven Instruments Corporation; 2020. Available from: https://www.brookhaveninstruments.com/guide-for-dls-sample-preparation/. Accessed November 15, 2020.
  • Sondi I, Salopek-Sondi B. Silver nanoparticles as antimicrobial agent: a case study on E. coli as a model for Gram-negative bacteria. J Colloid Interface Sci. 2004;275:177–182. doi:10.1016/j.jcis.2004.02.012
  • Shid RL, Dhole SN, Kulkarni N, et al. Formulation and evaluation of nanosuspension delivery system for simvastatin. Int J Sci Nanotech. 2014;7:2459–2476.
  • Ong SGM, Ming LC, Lee KS, et al. Influence of the encapsulation efficiency and size of liposome on the oral bioavailability of griseofulvin-loaded liposomes. Pharmaceutics. 2016;8:1–17. doi:10.3390/pharmaceutics8030025
  • Modi S, Anderson BD. Determination of drug release kinetics from nanoparticles: overcoming pitfalls of the dynamic dialysis method. Mol Pharm. 2013;10:3076–3089. doi:10.1021/mp400154a
  • Malviya R. Green approach for fabrication of chitosan-neem gum polyelectrolyte stabilized penta and hexagonal nanoparticles and in-vitro cytotoxic potential toward breast cancer (MCF-7) cells. Prec Med Sci. 2020;9:68–82. doi:10.1002/prm2.12025
  • Weng J, Tong HH, Chow SF. In vitro release study of the polymeric drug nanoparticles: development and validation of a novel method. Pharmaceutics. 2020;12:1–18. doi:10.3390/pharmaceutics12080732
  • Costa P, Lobo JMS. Modeling and comparison of dissolution profiles. Eur J Pharm Sci. 2001;13:123–133. doi:10.1016/S0928-0987(01)00095-1
  • Simionato LD, Petrone L, Baldut M, et al. Comparison between the dissolution profiles of nine meloxicam tablet brands commercially available in Buenos Aires, Argentina. Saudi Pharm J. 2018;26:578–584. doi:10.1016/j.jsps.2018.01.015
  • Dash S, Murthy PN, Nath L, et al. Kinetic modeling on drug release from control drug delivery systems. Acta Pol Pharm. 2010;67:217–223.
  • Mircioiu C, Voicu V, Anuta V, et al. Mathematical modeling of release kinetics from supramolecular drug delivery systems. Pharmaceutics. 2019;11:1–45. doi:10.3390/pharmaceutics11030140
  • Gohel MC, Sarvaiya KG, Shah AR, et al. Mathematical approach for the assessment of similarity factor using a new scheme for calculating weight. Indian J Pharm Sci. 2009;71:142–144. doi:10.4103/0250-474X.54281
  • Muller M. Sizing, shaping and pharmaceutical applications of polyelectrolyte complex nanoparticles. In: Polyelectrolyte Complexes in the Dispersed and Solid State II. Berlin, Heidelberg: Springer; 2012:197–260.
  • Makoni PA, WaKasongo K, Walker RB. Short term stability testing of efavirenz-loaded solid lipid nanoparticle (SLN) and nanostructured lipid carrier (NLC) dispersions. Pharmaceutics. 2019;11:1–21. doi:10.3390/pharmaceutics11080397
  • Rubin H. Deprivation of glutamine in cell culture reveals its potential for treating cancer. Proc Natl Acad Sci. 2019;116:6964–6968. doi:10.1073/pnas.1815968116
  • Rana K, Arora A, Bansal S, et al. Synthesis, in vitro anticancer and antimicrobial evaluation of novel substituted dihydropyrimidines. Indian J Pharm Sci. 2014;76:339–347.
  • Polexe RC, Delair T. Elaboration of stable and antibody functionalized positively charged colloids by polyelectrolyte complexation between chitosan and hyaluronic acid. Molecules. 2013;18:8563–8578. doi:10.3390/molecules18078563
  • Kumar R, Siril PF, Javid F. Unusual anti-leukemia activity of nanoformulated naproxen and other non-steroidal anti-inflammatory drugs. Mater Sci Eng C. 2016;69:1335–1344. doi:10.1016/j.msec.2016.08.024
  • Jindal AB, Devarajan PV. Asymmetric lipid-polymer particles (LIPOMER) by modified nanoprecipitation: role of non-solvent composition. Int J Pharm. 2015;489:246–251. doi:10.1016/j.ijpharm.2015.04.073
  • Oh WK, Kim S, Yoon H, et al. Shape-dependent cytotoxicity and proinflammatory response of poly(3,4-ethylenedioxythiophene) nanomaterials. Small. 2010;6:872–879. doi:10.1002/smll.200902074
  • Jansch M, Jindal AB, Sharmila BM, et al. Influence of particle shape on plasma protein adsorption and macrophage uptake. Pharmazie. 2013;68:27–33.
  • Caldorera-Moore M, Guimard N, Shi L, et al. Designer nanoparticles: incorporating size, shape, and triggered release into nanoscale drug carriers. Expert Opin Drug Deliv. 2010;7:479–495. doi:10.1517/17425240903579971
  • Akiyama Y, Mori T, Katayama Y, et al. Conversion of rod-shaped gold nanoparticles to spherical forms and their effect on biodistribution in tumor-bearing mice. Nanoscale Res Lett. 2012;7:565. doi:10.1186/1556-276X-7-565
  • Vacha R, Martinez-Veracoechea FJ, Frenkel D. Receptor-mediated endocytosis of nanoparticles of various shapes. Nano Lett. 2011;11:5391–5395. doi:10.1021/nl2030213
  • Wang THY, Shen Y, Ao H, et al. Preparation of high drug-loading celastrol nanosuspensions and their anti-breast cancer activities in vitro and in vivo. Sci Rep. 2020;10:1–9. doi:10.1038/s41598-019-56847-4
  • Ahuja M, Verma P, Bhatia M. Preparation and evaluation of chitosan–itraconazole co-precipitated nanosuspension for ocular delivery. J Exp Nanosci. 2015;10:209–221. doi:10.1080/17458080.2013.822108
  • Dekate S, Bhairy S, Hirlekar R. Preparation and characterization of oral nanosuspension loaded with curcumin. Int J Pharm Pharm Sci. 2018;10:90–95. doi:10.22159/ijpps.2018v10i6.22027
  • Karfa P, Madhuri R, Sharma PK. Is shape of Ag/AgCl nanoparticle responsible for femtogram detection of alpha-feto protein: comparison between round and cube-shaped nanoparticle modified imprinted polymer. J Mater Chem B. 2016;4(33):5534–5547. doi:10.1039/C6TB01306F
  • Zhao J, Baibuz E, Vernieres J, et al. Formation mechanism of Fe nanocubes by magnetron sputtering inert gas condensation. ACS Nano. 2016;10(4):4684–4694. doi:10.1021/acsnano.6b01024
  • Kolhatkar AG, Nekrashevich I, Litvinov D, Willson RC, Lee TR. Cubic silica-coated and amine-functionalized FeCo nanoparticles with high saturation magnetization. Chem Mater. 2013;25(7):1092–1097. doi:10.1021/cm304111z
  • Tığlı Aydın RS, Pulat M. 5-Fluorouracil encapsulated chitosan nanoparticles for ph-stimulated drug delivery: evaluation of controlled release kinetics. J Nanomater. 2012;2012:1–10. doi:10.1155/2012/313961
  • Shoaib MH, Tazeen J, Merchant HA, et al. Evaluation of drug release kinetics from ibuprofen matrix tablets using HPMC. Pak J Pharm Sci. 2006;19:119–124.
  • Grassi M, Grassi G. Mathematical modelling and controlled drug delivery: matrix systems. Curr Drug Deliv. 2005;2:97–116. doi:10.2174/1567201052772906
  • Chan KKW, Oza AM, Siu LL. The statins as anticancer agent. Clin Cancer Res. 2003;9:10–19.
  • Mandal CC, Ghosh-Choudhury N, Yoneda T, et al. Simvastatin prevents skeletal metastasis of breast cancer by an antagonistic interplay between p53 and CD44. J Biol Chem. 2011;286:11314–11327. doi:10.1074/jbc.M110.193714
  • Kotamraju S, Willams CL, Kalyanaraman B. Statin-induced breast cancer cell death: role of inducible nitric oxide and arginase-dependent pathways. Cancer Res. 2007;67:7386–7394. doi:10.1158/0008-5472.CAN-07-0993
  • Greenaway JB, Virtanen C, Osz K, et al. Ovarian tumour growth is characterized by mevalonate pathway gene signature in an orthotopic, syngeneic model of epithelial ovarian cancer. Oncotarget. 2016;7:47343–47365. doi:10.18632/oncotarget.10121
  • Duncan RE, Lau D, El-Sohemy A, et al. Geraniol and β-ionone inhibit proliferation, cell cycle progression, and cyclin-dependent kinase 2 activity in MCF-7 breast cancer cells independent of effects on HMG-CoA reductase activity. Biochem Pharmacol. 2004;68:1739–1747. doi:10.1016/j.bcp.2004.06.022
  • Huang X, Teng X, Chen D, et al. The effect of the shape of Mesoporous silica nanoparticles on cellular uptake and cell function. Biomaterials. 2010;31:438–448. doi:10.1016/j.biomaterials.2009.09.060
  • Godin B, Chiappini C, Srinivasan S, et al. Discoidal porous silicon particles: fabrication and biodistribution in breast cancer bearing mice. Adv Funct Mater. 2012;22:4225–4235. doi:10.1002/adfm.201200869
  • Zhao X, Ng S, Heng BC, et al. Cytotoxicity of hydroxyapatite nanoparticles is shape and cell dependent. Arch Toxicol. 2013;87:1037–1052. doi:10.1007/s00204-012-0827-1
  • Li Y, Kroger M, Liu WK. Shape effect in cellular uptake of PEGylated nanoparticles: comparison between sphere, rod, cube and disk. Nanoscale. 2015;7:16631–16646. doi:10.1039/C5NR02970H
  • Shimoni O, Yan Y, Wang Y, et al. Shape-dependent cellular processing of polyelectrolyte capsules. ACS Nano. 2013;7:522–530. doi:10.1021/nn3046117
  • Attallah OA, Shetta A, Elshishiny F, Mamdouh W. Essential oil loaded pectin/chitosan nanoparticles preparation and optimization via Boxȃ “Behnken design against MCF-7 breast cancer cell lines. RSC Adv. 2020;10(15):8703–8708. doi:10.1039/c9ra10204c
  • Guo H, Li F, Qiu H, et al. Chitosan-based nanogel enhances chemotherapeutic efficacy of 10-hydroxycamptothecin against human breast cancer cells. Int J Polym Sci. 2019;2019:1–6. doi:10.1155/2019/1914976
  • Nascimento AV, Gattacceca F, Singh A, et al. Biodistribution and pharmacokinetics of Mad2 siRNA-loaded EGFR-targeted chitosan nanoparticles in cisplatin sensitive and resistant lung cancer models. Nanomedicine. 2016;11:767–781. doi:10.2217/nnm.16.14
  • Nascimento AV, Singh A, Bousbaa H, Ferreira D, Sarmento B, Amiji MM. Overcoming cisplatin resistance in non-small cell lung cancer with Mad2 silencing siRNA delivered systemically using EGFR-targeted chitosan nanoparticles. Acta Biomater. 2017;47:71–80. doi:10.1016/j.actbio.2016.09.045
  • Jain A, Jain SK. In vitro and cell uptake studies for targeting of ligand anchored nanoparticles for colon tumors. Eur J Pharm Sci. 2008;35(5):404–416. doi:10.1016/j.ejps.2008.08.008
  • Zhang H, Mardyani S, Chan WCW, Kumacheva E. Design of biocompatible chitosan microgels for targeted pH-mediated intracellular release of cancer therapeutics. Biomacromolecules. 2006;7(5):1568–1572. doi:10.1021/bm050912z
  • Yang H, Liu Y, Qiu Y, Ding M, Zhang Y. MiRNA-204-5p and Oxaliplatin-loaded silica nanoparticles for enhanced tumor suppression effect in CD44-overexpressed colon adenocarcinoma. Int J Pharm. 2019;566:585–593. doi:10.1016/j.ijpharm.2019.06.020
  • Cui Y, Zhang M, Zeng F, Jin H, Xu Q, Huang Y. Dual-targeting magnetic PLGA nanoparticles for codelivery of paclitaxel and curcumin for brain tumor therapy. ACS Appl Mater Interfaces. 2016;8(47):32159–32169. doi:10.1021/acsami.6b10175