Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

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

Review Article

Chitosan Anchored Nanoparticles in Current Drug Development Utilizing Computer-Aided Pharmacokinetic Modeling: Case Studies for Target Specific Cancer Treatment and Future Prospective

Author(s): Harekrishna Roy, Bhabani S. Nayak* and Sisir Nandi *

Volume 26, Issue 15, 2020

Page: [1666 - 1675] Pages: 10

DOI: 10.2174/1381612826666200203121241

Price: $65

Abstract

Background: Recently, in the medical and pharmaceutical fields, biopolymers are extensively used for chemical and mechanical modifications of pharmaceutical dosage forms, which add novel properties, functions, and applications. Structural modification of dosage form by polymers along with redesigning in pharmaceutical and tissue engineering fields, presently being the center of analysis for the modern research world, which utilizes the subtle instruments, precise research strategies and most significantly the excipients.

Method: The polymer, chitosan, which is a natural linear polysaccharide composed of randomly distributed β-(1- 4)-linked D-Glucosamine and N-acetyl-D-Glucosamine units. Chitosan has been used by researchers as a network forming or gelling agent as chitosan is economically available, possesses low immunogenicity, biocompatibility, non-toxicity, biodegradability, protects against secretion from irritation and does not suffer the danger of transmission animal infective agent. Recent studies have proved that the chitosan conjugated in various biopharmaceutical drug formulations, such as nanoparticles, have been used for the treatment of breast, skin, colon, pancreatic, prostate and lung cancer. The nanoparticles have gained significant attention of scientific groups for relevant cancer-targeting drugs and dosage form. In this connection, several articles been published on chitosan anchored nanoparticles by suitable techniques, such as ion gelation, complexation, solvent evaporation, emulsion droplet coalescence and polymerization.

Results: The most remarkable point is that chitosan-drug conjugated nanoparticles (CDNP) can target cancer affected cells with the least attempt to killing the neighbor host cell. It is already proved that the CDNP facilitate the more drugs uptaking or cytotoxicity to a cancerous cell. This overcomes the dosage form designing problems of complexity in the biological mechanism and cell specificity. A computer-aided pharmacokinetic study as well as in-silico design with model fitting can provide the possible finding related to target selectivity and interaction. The computer aided study also reduces time and could make the entire process much cheaper till today, very few research has been reported, such as PyRx with AutoDock, response surface methodology and molecular dynamic simulation in drug delivery for chitosan-drug conjugated nanoparticles.

Conclusion: Therefore, cancer cell target-specific drug delivery using a natural biopolymer conjugate with a computer-aided pharmacokinetic model will be the thirst area of future research. To get successful anticancer drug formulation, in-silico pharmacokinetic modeling would minimize labor, and expenses, during and prior to the experiment has been extensively discussed in the present review.

Keywords: Natural biopolymer, chitosan, conjugated nanoparticle drug development, bone repair, cancer treatment, in-silico pharmacokinetic modeling.

[1]
Calinoui LF, Stefanescu BE, Pop ID, Muntean L, Vodnar DC. Chitosan coating applications in probiotic microencapsulation. Coatings 2019; 9: 194.
[http://dx.doi.org/10.3390/coatings9030194]
[2]
Elieh-Ali-Komi D, Hamblin MR. Chitin and chitosan: production and application of versatile biomedical nanomaterials. Int J Adv Res (Indore) 2016; 4(3): 411-27.
[PMID: 27819009]
[3]
Barbosa JN, Amaral IF, Águas AP, Barbosa MA. Evaluation of the effect of the degree of acetylation on the inflammatory response to 3D porous chitosan scaffolds. J Biomed Mater Res A 2010; 93(1): 20-8.
[PMID: 19484769]
[4]
Sinha VR, Singla AK, Wadhawan S, et al. Chitosan microspheres as a potential carrier for drugs. Int J Pharm 2004; 274(1-2): 1-33.
[http://dx.doi.org/10.1016/j.ijpharm.2003.12.026] [PMID: 15072779]
[5]
Kumari S, Rath PK. Extraction and characterization of chitin and chitosan from (Labeo rohit) fish scales. Procedia Mater Sci 2014; 6: 482-9.
[http://dx.doi.org/10.1016/j.mspro.2014.07.062]
[6]
Rafique A, Mahmood Zia K, Zuber M, Tabasum S, Rehman S. Chitosan functionalized poly(vinyl alcohol) for prospects biomedical and industrial applications: A review. Int J Biol Macromol 2016; 87: 141-54.
[http://dx.doi.org/10.1016/j.ijbiomac.2016.02.035] [PMID: 26893051]
[7]
LogithKumar R,KeshavNarayan A, Dhivya S, Chawla A, Saravanan S, Selvamurugan N. A review of chitosan and its derivatives in bone tissue engineering. Carbohydr Polym 2016; 151: 172-88.
[http://dx.doi.org/10.1016/j.carbpol.2016.05.049] [PMID: 27474556]
[8]
Vasconcelos DP, Fonseca AC, Costa M, et al. Macrophage polarization following chitosan implantation. Biomaterials 2013; 34(38): 9952-9.
[http://dx.doi.org/10.1016/j.biomaterials.2013.09.012] [PMID: 24074837]
[9]
Salehi E, Daraei P, Arabi Shamsabadi A. A review on chitosan-based adsorptive membranes. Carbohydr Polym 2016; 152: 419-32.
[http://dx.doi.org/10.1016/j.carbpol.2016.07.033] [PMID: 27516289]
[10]
El-Gamal R, Nikolaivits E, Zervakis GI, Abdel-Maksoud G, Topakas E, Christakopoulos P. The use of chitosan in protecting wooden artifacts from damage by mold fungi. Electron J Biotechnol 2016; 19: 70-8.
[http://dx.doi.org/10.1016/j.ejbt.2016.10.006]
[11]
Croisier F, Jérôme C. Chitosan-based biomaterials for tissue engineering. Eur Polym J 2013; 49: 780-92.
[http://dx.doi.org/10.1016/j.eurpolymj.2012.12.009]
[12]
Yang G, Wang J, Wang Y, Li L, Guo X, Zhou S. An implantable active-targeting micelle-in-nanofiber device for efficient and safe cancer therapy. ACS Nano 2015; 9(2): 1161-74.
[http://dx.doi.org/10.1021/nn504573u] [PMID: 25602381]
[13]
Yang SJ, Shieh MJ, Lin FH, et al. Colorectal cancer cell detection by 5-aminolaevulinic acid-loaded chitosan nano-particles. Cancer Lett 2009; 273(2): 210-20.
[http://dx.doi.org/10.1016/j.canlet.2008.08.014] [PMID: 18809246]
[14]
Yang Y, Wang S, Wang Y, Wang X, Wang Q, Chen M. Advances in self-assembled chitosan nanomaterials for drug delivery. Biotechnol Adv 2014; 32(7): 1301-16.
[http://dx.doi.org/10.1016/j.biotechadv.2014.07.007] [PMID: 25109677]
[15]
Badhe RV, Nanda RK, Chejara DR, et al. Microwave-assisted facile synthesis of a new tri-block chitosan conjugate with improved mucoadhesion. Carbohydr Polym 2015; 130: 213-21.
[http://dx.doi.org/10.1016/j.carbpol.2015.05.027] [PMID: 26076619]
[16]
H P S AK, Saurabh CK, A S A, et al. A review on chitosan-cellulose blends and nanocellulose reinforced chitosan biocomposites: Properties and their applications. Carbohydr Polym 2016; 150: 216-26.
[http://dx.doi.org/10.1016/j.carbpol.2016.05.028] [PMID: 27312632]
[17]
Al-Manhel AJ, Al-Hilphy AR, Niamah AK. Extraction of chitosan, characterisation and its use for water purification. J Saudi Soc Agric Sci 2018; 17: 186-90.
[http://dx.doi.org/10.1016/j.jssas.2016.04.001]
[18]
Xu Y, Asghar S, Yang L, et al. Lactoferrin-coated polysaccharide nanoparticles based on chitosan hydrochloride/hyaluronic acid/PEG for treating brain glioma. Carbohydr Polym 2017; 157: 419-28.
[http://dx.doi.org/10.1016/j.carbpol.2016.09.085] [PMID: 27987946]
[19]
Chiappisi L, Gradzielski M. Co-assembly in chitosan-surfactant mixtures: thermodynamics, structures, interfacial properties and applications. Adv Colloid Interface Sci 2015; 220: 92-107.
[http://dx.doi.org/10.1016/j.cis.2015.03.003] [PMID: 25865361]
[20]
Giri TK, Thakur A, Alexander A, Badwaik H, Tripathi DK. Modified chitosan hydrogels as drug delivery and tissue engineering systems: present status and applications. Acta Pharm Sin B 2012; 2(5): 439-49.
[http://dx.doi.org/10.1016/j.apsb.2012.07.004]
[21]
Fu S, Xia J, Wu J. Functional chitosan nanoparticles in cancer treatment. J Biomed Nanotechnol 2016; 12(8): 1585-603.
[http://dx.doi.org/10.1166/jbn.2016.2228] [PMID: 29341581]
[22]
Li J, Cai C, Li J, et al. Chitosan-based nanomaterials for drug delivery. Molecules 2018; 23(10): 26-61.
[http://dx.doi.org/10.3390/molecules23102661] [PMID: 30332830]
[23]
Rajitha P, Gopinath D, Biswas R, Sabitha M, Jayakumar R. Chitosan nanoparticles in drug therapy of infectious and inflammatory diseases. Expert Opin Drug Deliv 2016; 13(8): 1177-94.
[http://dx.doi.org/10.1080/17425247.2016.1178232] [PMID: 27087148]
[24]
Arjunan N, Kumari HL, Singaravelu CM, Kandasamy R, Kandasamy J. Physicochemical investigations of biogenic chitosan-silver nanocomposite as antimicrobial and anticancer agent. Int J Biol Macromol 2016; 92: 77-87.
[http://dx.doi.org/10.1016/j.ijbiomac.2016.07.003] [PMID: 27381584]
[25]
Khan MA, Zafaryab M, Mehdi SH, Ahmad I, Rizvi MMA, Moshahid A. Physicochemical characterization of curcumin loaded chitosan nanoparticles: implications in cervical cancer. Anticancer Agents Med Chem 2018; 18(8): 1131-7.
[http://dx.doi.org/10.2174/1871520618666180412114352] [PMID: 29651963]
[26]
Varshosaz J, Hassanzadeh F, Aliabadi HS, Khoraskani FR, Mirian M, Behdadfar B. Targeted delivery of doxorubicin to breast cancer cells by magnetic LHRH chitosan bioconjugated nanoparticlesInt J Biol Macromol 2016; 93(Pt. A): 1192-205.
[http://dx.doi.org/10.1016/j.ijbiomac.2016.07.025] [PMID: 27693338]
[27]
Nayak D, Minz AP, Ashe S, et al. Synergistic combination of antioxidants, silver nanoparticles and chitosan in a nanoparticle based formulation: Characterization and cytotoxic effect on MCF-7 breast cancer cell lines. J Colloid Interface Sci 2016; 470: 142-52.
[http://dx.doi.org/10.1016/j.jcis.2016.02.043] [PMID: 26939078]
[28]
Mansouri M, Nazarpak MH, Solouk A, Akbari S, Hasani-Sadrabadi MM. Magnetic responsive of paclitaxel delivery system based on SPION and palmitoyl chitosan. J Magn Magn Mater 2017; 421: 316-25.
[http://dx.doi.org/10.1016/j.jmmm.2016.07.066]
[29]
Costa IdosS, Abranches RP, Garcia MT, Pierre MB. Chitosan-based mucoadhesive films containing 5-aminolevulinic acid for buccal cancer’s treatment. J Photochem Photobiol B 2014; 140: 266-75.
[http://dx.doi.org/10.1016/j.jphotobiol.2014.08.005] [PMID: 25190225]
[30]
Wu J, Jiang W, Shen Y, Jiang W, Tian R. Synthesis and characterization of mesoporous magnetic nanocomposites wrapped with chitosan gatekeepers for pH-sensitive controlled release of doxorubicin. Mater Sci Eng C 2017; 70(Pt 1): 132-40.
[http://dx.doi.org/10.1016/j.msec.2016.08.054] [PMID: 27770872]
[31]
Wu J, Tang C, Yin C. Co-delivery of doxorubicin and interleukin-2 via chitosan based nanoparticles for enhanced antitumor efficacy. Acta Biomater 2017; 47: 81-90.
[http://dx.doi.org/10.1016/j.actbio.2016.10.012] [PMID: 27729232]
[32]
Anandha kumar S, Krishna moorthy G, Ramkumar KM, Raichur AM. Preparation of collagen peptide functionalized chitosan nanoparticles by ionic gelation method: an effective carrier system for encapsulation and release of doxorubicin for cancer drug delivery. Mater Sci Eng C 2017; 70: 378-85.
[http://dx.doi.org/10.1016/j.msec.2016.09.003]
[33]
Wang M, Zhao T, Liu Y, et al. Ursolic acid liposomes with chitosan modification: Promising antitumor drug delivery and efficacy. Mater Sci Eng C 2017; 71: 1231-40.
[http://dx.doi.org/10.1016/j.msec.2016.11.014] [PMID: 27987679]
[34]
Rajan M, Murugan M, Ponnamma D, Sadasivuni KK, Munusamy MA. Poly-carboxylic acids functionalized chitosan nanocarriers for controlled and targeted anti-cancer drug delivery. Biomed Pharmacother 2016; 83: 201-11.
[http://dx.doi.org/10.1016/j.biopha.2016.06.026] [PMID: 27470566]
[35]
Snima KS, Jayakumar R, Unnikrishnan AG, Nair SV, Lakshmanan VK. O-carboxymethyl chitosan nanoparticles for metformin delivery to pancreatic cancer cells. Carbohydr Polym 2012; 89(3): 1003-7.
[http://dx.doi.org/10.1016/j.carbpol.2012.04.050] [PMID: 24750892]
[36]
David KI, Jaidev LR, Sethuraman S, Krishnan UM. Dual drug loaded chitosan nanoparticles-sugar--coated arsenal against pancreatic cancer. Colloids Surf B Biointerfaces 2015; 135: 689-98.
[http://dx.doi.org/10.1016/j.colsurfb.2015.08.038] [PMID: 26340358]
[37]
Qi L, Xu Z, Chen M. In vitro and in vivo suppression of hepatocellular carcinoma growth by chitosan nanoparticles. Eur J Cancer 2007; 43(1): 184-93.
[http://dx.doi.org/10.1016/j.ejca.2006.08.029] [PMID: 17049839]
[38]
Patel NV, Sheth NR, Mohddesi B. Formulation and evaluation of genistein–a novel isoflavone loaded chitosan and eudragit® nanoparticles for cancer therapy. Mater Today 2015; 2: 4477-82.
[39]
Ekinci M, Ilem-Ozdemir D, Gundogdu E, Asikoglu M. Methotrexate loaded chitosan nanoparticles: preparation, radiolabeling and in vitro evaluation for breast cancer diagnosis. J Drug Deliv Sci Technol 2015; 30: 107-13.
[http://dx.doi.org/10.1016/j.jddst.2015.10.004]
[40]
Garg NK, Dwivedi P, Campbell C, Tyagi RK. Site specific/targeted delivery of gemcitabine through anisamide anchored chitosan/poly ethylene glycol nanoparticles: an improved understanding of lung cancer therapeutic intervention. Eur J Pharm Sci 2012; 47(5): 1006-14.
[http://dx.doi.org/10.1016/j.ejps.2012.09.012] [PMID: 23041219]
[41]
Huang Z, Dong L, Chen J, et al. Low-molecular weight chitosan/vascular endothelial growth factor short hairpin RNA for the treatment of hepatocellular carcinoma. Life Sci 2012; 91(23-24): 1207-15.
[http://dx.doi.org/10.1016/j.lfs.2012.09.015] [PMID: 23044224]
[42]
Kuppusamy S, Karuppaiah J. Antioxidant and cytotoxic efficacy of chitosan on bladder cancer. Asian Pac J Trop Dis 2012; 2: S769-73.
[http://dx.doi.org/10.1016/S2222-1808(12)60262-X]
[43]
Ma JX, Qian L, Zhou Y. Stimulation effect of chitosan on the immunity of radiotherapy patients suffered from lung cancer. Int J Biol Macromol 2015; 72: 195-8.
[http://dx.doi.org/10.1016/j.ijbiomac.2014.08.027] [PMID: 25175732]
[44]
Chen G, Gong R. Study on fluorouracil-chitosan nanoparticle preparation and its antitumor effect. Saudi Pharm J 2016; 24(3): 250-3.
[http://dx.doi.org/10.1016/j.jsps.2016.04.008] [PMID: 27275110]
[45]
Zhou W, Wang Y, Jian J, Song S. Self-aggregated nanoparticles based on amphiphilic poly(lactic acid)-grafted-chitosan copolymer for ocular delivery of amphotericin B. Int J Nanomedicine 2013; 8: 3715-28.
[PMID: 24106427]
[46]
Anitha A, Uthaman S, Nair SV, Jayakumar R, Lakshmanan VK. Enhanced delivery system of flutamide loaded chitosan-dextran sulphate nanoparticles for prostate cancer. J Biomed Nanotechnol 2013; 9(3): 335-47.
[http://dx.doi.org/10.1166/jbn.2013.1558] [PMID: 23620988]
[47]
Shering MA, Kannan C, Kumar KS, Kumar VS, Suganeshwari M. Formulation of 5-fluorouracil loaded chitosan nanoparticles by emulsion droplet coalescence method for cancer therapy. Int J Pharm Biol Arch 2011; 2: 926-31.
[48]
Talaei F, Azizi E, Dinarvand R, Atyabi F. Thiolated chitosan nanoparticles as a delivery system for antisense therapy: evaluation against EGFR in T47D breast cancer cells. Int J Nanomedicine 2011; 6: 1963-75.
[PMID: 21976973]
[49]
Le TM, Pham VP, Dang TM, La TH, Le TH, Le QH. Preparation of curcumin-loaded pluronic F127/chitosan nanoparticles for cancer therapy. Adv Nat Sci-Nanosc 2013; 4: 25001-4.
[http://dx.doi.org/10.1088/2043-6262/4/2/025001]
[50]
Boca SC, Potara M, Gabudean AM, Juhem A, Baldeck PL, Astilean S. Chitosan-coated triangular silver nanoparticles as a novel class of biocompatible, highly effective photothermal transducers for in vitro cancer cell therapy. Cancer Lett 2011; 311(2): 131-40.
[http://dx.doi.org/10.1016/j.canlet.2011.06.022] [PMID: 21840122]
[51]
Akhlaghi SP, Saremi S, Ostad SN, Dinarvand R, Atyabi F. Discriminated effects of thiolated chitosan-coated pMMA paclitaxel-loaded nanoparticles on different normal and cancer cell lines. Nanomedicine (Lond) 2010; 6(5): 689-97.
[http://dx.doi.org/10.1016/j.nano.2010.01.011] [PMID: 20172052]
[52]
Jain A, Jain SK, Ganesh N, Barve J, Beg AM. Design and development of ligand-appended polysaccharidic nanoparticles for the delivery of oxaliplatin in colorectal cancer. Nanomedicine (Lond) 2010; 6(1): 179-90.
[http://dx.doi.org/10.1016/j.nano.2009.03.002] [PMID: 19447205]
[53]
Chung CW, Chung KD, Jeong YI, Kang DH. 5-aminolevulinic acid-incorporated nanoparticles of methoxy poly(ethylene glycol)-chitosan copolymer for photodynamic therapy. Int J Nanomedicine 2013; 8: 809-19.
[http://dx.doi.org/10.2147/IJN.S39615] [PMID: 23589688]
[54]
Campuzano S, Pelling A. Scaffolds for 3D cell culture and cellular agriculture applications derived from non-animal sources. Front Sustain Food Syst 2019; 3(38): 1-9.
[55]
Derakhshandeh K, Fathi S. Role of chitosan nanoparticles in the oral absorption of Gemcitabine. Int J Pharm 2012; 437(1-2): 172-7.
[http://dx.doi.org/10.1016/j.ijpharm.2012.08.008] [PMID: 22909993]
[56]
Ruano-Ordás D, Yevseyeva I, Fernandes VB, Méndez JR, Emmerich MT. Improving the drug discovery process by using multiple classifier systems. Expert Syst Appl 2019; 121: 292-303.
[http://dx.doi.org/10.1016/j.eswa.2018.12.032]
[57]
Abel R, Mondal S, Masse C, et al. Accelerating drug discovery through tight integration of expert molecular design and predictive scoring. Curr Opin Struct Biol 2017; 43: 38-44.
[http://dx.doi.org/10.1016/j.sbi.2016.10.007] [PMID: 27816785]
[58]
Ooms F. Molecular modeling and computer aided drug design. Examples of their applications in medicinal chemistry. Curr Med Chem 2000; 7(2): 141-58.
[http://dx.doi.org/10.2174/0929867003375317] [PMID: 10637360]
[59]
Sacan A, Ekins S, Kortagere S. Applications and limitations of in silico models in drug discovery. In: bioinformatics and drug discovery.Totowa, NJ: Humana Press. 2012; 87-124.
[http://dx.doi.org/10.1007/978-1-61779-965-5_6]
[60]
Kumar A, Rathi E, Kini SG. E-pharmacophore modelling, virtual screening, molecular dynamics simulations and in-silico ADME analysis for identification of potential E6 inhibitors against cervical cancer. J Mol Struct 2019; 1189: 299-306.
[http://dx.doi.org/10.1016/j.molstruc.2019.04.023]
[61]
Yu X, Trase I, Ren M, Duval K, Guo X, Chen Z. Design of nanoparticle-based carriers for targeted drug delivery. J Nanomater 2016; 2016: 1-15.
[http://dx.doi.org/10.1155/2016/1087250] [PMID: 27398083]
[62]
Duarte TL, Lunec J. Review: When is an antioxidant not an antioxidant? A review of novel actions and reactions of vitamin C. Free Radic Res 2005; 39(7): 671-86.
[http://dx.doi.org/10.1080/10715760500104025] [PMID: 16036346]
[63]
Stephenson CM, Levin RD, Spector T, Lis CG. Phase I clinical trial to evaluate the safety, tolerability, and pharmacokinetics of high-dose intravenous ascorbic acid in patients with advanced cancer. Cancer Chemother Pharmacol 2013; 72(1): 139-46.
[http://dx.doi.org/10.1007/s00280-013-2179-9] [PMID: 23670640]
[64]
Rossi S, Marciello M, Sandri G, Bonferoni MC, Ferrari F, Caramella C. Chitosan ascorbate: a chitosan salt with improved penetration enhancement properties. Pharm Dev Technol 2008; 13(6): 513-21.
[http://dx.doi.org/10.1080/10837450802288865] [PMID: 18720244]
[65]
Sekar V, Rajendran K, Vallinayagam S, Deepak V, Mahadevan S. Synthesis and characterization of chitosan ascorbate nanoparticles for therapeutic inhibition for cervical cancer and their in silico modeling. J Ind Eng Chem 2018; 62: 239-49.
[http://dx.doi.org/10.1016/j.jiec.2018.01.001]
[66]
Trott O, Olson AJ. AutoDock Vina: improving the speed and accuracy of docking with a new scoring function, efficient optimization, and multithreading. J Comput Chem 2010; 31(2): 455-61.
[PMID: 19499576]
[67]
Yadav P, Bandyopadhyay A, Chakraborty A, Sarkar K. Enhancement of anticancer activity and drug delivery of chitosan-curcumin nanoparticle via molecular docking and simulation analysis. Carbohydr Polym 2018; 182: 188-98.
[http://dx.doi.org/10.1016/j.carbpol.2017.10.102] [PMID: 29279114]
[68]
Rejinold NS, Muthunarayanan M, Divyarani VV, et al. Curcumin-loaded biocompatible thermoresponsive polymeric nanoparticles for cancer drug delivery. J Colloid Interface Sci 2011; 360(1): 39-51.
[http://dx.doi.org/10.1016/j.jcis.2011.04.006] [PMID: 21549390]
[69]
Segerman ZJ, Roy B, Hecht SM. Characterization of bleomycin-mediated cleavage of a hairpin DNA library. Biochemistry 2013; 52(31): 5315-27.
[http://dx.doi.org/10.1021/bi400779r] [PMID: 23834496]
[70]
Bayer RA, Gaynor ER, Fisher RI. Bleomycin in non-Hodgkin’s lymphoma. Semin Oncol 1992; 19(2)(Suppl. 5): 46-52.
[PMID: 1384144]
[71]
Chen JK, Yang D, Shen B, Murray V. Bleomycin analogues preferentially cleave at the transcription start sites of actively transcribed genes in human cells. Int J Biochem Cell Biol 2017; 85: 56-65.
[http://dx.doi.org/10.1016/j.biocel.2017.02.001] [PMID: 28167289]
[72]
Blenoxane. Bristol-Myers Squibb Company Princeton, NJ 08543 USA 2010. Available at: http://www.accessdata.fda.gov/drugs atfda_docs/label/2010/050443s036lbl.pdf
[73]
Bleomycin for Injection USP. Faulding pharmaceutical co 2002.Available at:. http://oncozine.com/wpcontent/uploads/2017/07/Bleo mycin-for-Injection-USP.pdf
[74]
Kaur S, Kaur S. Bacteriocins as potential anticancer agents. Front Pharmacol 2015; 6: 272.
[http://dx.doi.org/10.3389/fphar.2015.00272] [PMID: 26617524]
[75]
Paiva AD, de Oliveira MD, de Paula SO, Baracat-Pereira MC, Breukink E, Mantovani HC. Toxicity of bovicin HC5 against mammalian cell lines and the role of cholesterol in bacteriocin activity. Microbiology 2012; 158(Pt 11): 2851-8.
[http://dx.doi.org/10.1099/mic.0.062190-0] [PMID: 22956757]
[76]
Chumchalová J, Smarda J. Human tumor cells are selectively inhibited by colicins. Folia Microbiol (Praha) 2003; 48(1): 111-5.
[http://dx.doi.org/10.1007/BF02931286] [PMID: 12744087]
[77]
Hetz C, Bono MR, Barros LF, Lagos R. Microcin E492, a channel-forming bacteriocin from Klebsiella pneumoniae, induces apoptosis in some human cell lines. Proc Natl Acad Sci USA 2002; 99(5): 2696-701.
[http://dx.doi.org/10.1073/pnas.052709699] [PMID: 11880624]
[78]
Lagos R, Tello M, Mercado G, García V, Monasterio O. Antibacterial and antitumorigenic properties of microcin E492, a pore-forming bacteriocin. Curr Pharm Biotechnol 2009; 10(1): 74-85.
[http://dx.doi.org/10.2174/138920109787048643] [PMID: 19149591]
[79]
Zhao H, Sood R, Jutila A, et al. Interaction of the antimicrobial peptide pheromone Plantaricin A with model membranes: implications for a novel mechanism of action. Biochim Biophys Acta 2006; 1758(9): 1461-74.
[http://dx.doi.org/10.1016/j.bbamem.2006.03.037] [PMID: 16806056]
[80]
Abdi-Ali A, Worobec EA, Deezagi A, Malekzadeh F. Cytotoxic effects of pyocin S2 produced by Pseudomonas aeruginosa on the growth of three human cell lines. Can J Microbiol 2004; 50(5): 375-81.
[http://dx.doi.org/10.1139/w04-019] [PMID: 15213746]
[81]
Watanabe T, Saito H. Cytotoxicity of pyocin S2 to tumor and normal cells and its interaction with cell surfaces. Biochim Biophys Acta 1980; 633(1): 77-86.
[http://dx.doi.org/10.1016/0304-4165(80)90039-2] [PMID: 6256005]
[82]
Bernardes N, Chakrabarty AM, Fialho AM. Engineering of bacterial strains and their products for cancer therapy. Appl Microbiol Biotechnol 2013; 97(12): 5189-99.
[http://dx.doi.org/10.1007/s00253-013-4926-6] [PMID: 23644748]
[83]
Mehta RR, Yamada T, Taylor BN, et al. A cell penetrating peptide derived from azurin inhibits angiogenesis and tumor growth by inhibiting phosphorylation of VEGFR-2, FAK and Akt. Angiogenesis 2011; 14(3): 355-69.
[http://dx.doi.org/10.1007/s10456-011-9220-6] [PMID: 21667138]
[84]
Hindupur SK, Colombi M, Fuhs SR, et al. The protein histidine phosphatase LHPP is a tumour suppressor. Nature 2018; 555(7698): 678-82.
[http://dx.doi.org/10.1038/nature26140] [PMID: 29562234]
[85]
Kiaie N, Aghdam RM, Tafti SH, Emami SH. Statistical optimization of chitosan nanoparticles as protein vehicles, using response surface methodology. J Appl Biomater Funct Mater 2016; 14(4): e413-22.
[http://dx.doi.org/10.5301/jabfm.5000278] [PMID: 27647390]
[86]
Thanou M, Verhoef JC, Junginger HE. Chitosan and its derivatives as intestinal absorption enhancers. Adv Drug Deliv Rev 2001; 50(1)(Suppl. 1): S91-S101.
[http://dx.doi.org/10.1016/S0169-409X(01)00180-6] [PMID: 11576697]
[87]
Nam KS, Shon YH. Suppression of metastasis of human breast cancer cells by chitosan oligosaccharides. J Microbiol Biotechnol 2009; 19(6): 629-33.
[PMID: 19597323]
[88]
Xu Y, Wen Z, Xu Z. Chitosan nanoparticles inhibit the growth of human hepatocellular carcinoma xenografts through an antiangiogenic mechanism. Anticancer Res 2009; 29(12): 5103-9.
[PMID: 20044623]
[89]
Svirshchevskaya EV, Gracheva IA, Kuznetsov AG, Myrsikova EV. Antitumor activity of furanoallocolchicinoid-chitosan conjugate. Med Chem 2016; 6(9): 571-7.
[90]
Zhang H, Wu F, Li Y, et al. Chitosan-based nanoparticles for improved anticancer efficacy and bioavailability of mifepristone. Beilstein J Nanotechnol 2016; 7: 1861-70.
[http://dx.doi.org/10.3762/bjnano.7.178] [PMID: 28144535]
[91]
Kuppusamy S, Karuppaiah J. Screening of antiproliferative effect of chitosan on tumor growth and metastasis in T24 urinary bladder cancer cell line. Austrl Asian J Cancer 2013; 12(3): 145-9.
[92]
Park JK, Chung MJ, Choi HN, Park YI. Effects of the molecular weight and the degree of deacetylation of chitosan oligosaccharides on antitumor activity. Int J Mol Sci 2011; 12(1): 266-77.
[http://dx.doi.org/10.3390/ijms12010266] [PMID: 21339986]

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