361
Views
27
CrossRef citations to date
0
Altmetric
Original Research

In vivo Bio-Distribution and Toxicity Evaluation of Polymeric and Lipid-Based Nanoparticles: A Potential Approach for Chronic Diseases Treatment

ORCID Icon, ORCID Icon, ORCID Icon, ORCID Icon, , ORCID Icon, ORCID Icon, ORCID Icon & show all
Pages 8609-8621 | Published online: 05 Nov 2020

References

  • Semete B, Booysen L, Lemmer Y, et al. In vivo evaluation of the biodistribution and safety of PLGA nanoparticles as drug delivery systems. Nanomedicine. 2010;6(5):662–671. doi:10.1016/j.nano.2010.02.00220230912
  • Oroojalian F, Charbgoo F, Hashemi M, et al. Recent advances in nanotechnology-based drug delivery systems for the kidney. J Control Release. 2020;321:442–462. doi:10.1016/j.jconrel.2020.02.02732067996
  • Sahle FF, Kim S, Niloy KK, et al. Nanotechnology in regenerative ophthalmology. Adv Drug Deliv Rev. 2019;148:290–307. doi:10.1016/j.addr.2019.10.00631707052
  • Ramalho MJ, Andrade S, Loureiro JA, Do Carmo Pereira M. Nanotechnology to improve the Alzheimer’s disease therapy with natural compounds. Drug Deliv Transl Res. 2019.
  • Loureiro JA, Gomes B, Coelho MA, Do Carmo Pereira M, Rocha S. Targeting nanoparticles across the blood-brain barrier with monoclonal antibodies. Nanomedicine (Lond). 2014;9(5):709–722.24827845
  • Nakamura T, Yamada Y, Sato Y, Khalil IA, Harashima H. Innovative nanotechnologies for enhancing nucleic acids/gene therapy: controlling intracellular trafficking to targeted biodistribution. Biomaterials. 2019;218:119329. doi:10.1016/j.biomaterials.2019.11932931306827
  • Marques AC, Costa PJ, Velho S, Amaral MH. Functionalizing nanoparticles with cancer-targeting antibodies: a comparison of strategies. J Control Release. 2020;320:180–200. doi:10.1016/j.jconrel.2020.01.03531978444
  • Vieira DB, Gamarra LF. Getting into the brain: liposome-based strategies for effective drug delivery across the blood-brain barrier. Int J Nanomedicine. 2016;11:5381–5414.27799765
  • Su H, Wang Y, Liu S, et al. Emerging transporter-targeted nanoparticulate drug delivery systems. Acta Pharm Sin B. 2019;9(1):49–58.30766777
  • Karlsson J, Vaughan HJ, Green JJ. Biodegradable polymeric nanoparticles for therapeutic cancer treatments. Annu Rev Chem Biomol Eng. 2018;9:105–127. doi:10.1146/annurev-chembioeng-060817-08405529579402
  • Mokhtarzadeh A, Alibakhshi A, Yaghoobi H, Hashemi M, Hejazi M, Ramezani M. Recent advances on biocompatible and biodegradable nanoparticles as gene carriers. Expert Opin Biol Ther. 2016;16(6):771–785.26998622
  • Gajbhiye KR, Pawar A, Mahadik KR, Gajbhiye V. PEGylated nanocarriers: a promising tool for targeted delivery to the brain. Colloids Surf B Biointerfaces. 2020;110770.31926790
  • Zheng Y, Li Z, Chen H, Gao Y. Nanoparticle-based drug delivery systems for controllable photodynamic cancer therapy. Eur J Pharm Sci. 2020;144:105213.31926941
  • Zhi D, Yang T, Yang J, Fu S, Zhang S. Targeting strategies for superparamagnetic iron oxide nanoparticles in cancer therapy. Acta Biomater. 2020;102:13–34. doi:10.1016/j.actbio.2019.11.02731759124
  • Yan H, Xue Z, Xie J, et al. Toxicity of carbon nanotubes as anti-tumor drug carriers. Int J Nanomedicine. 2019;14:10179–10194.32021160
  • Akbarzadeh A, Rezaei-Sadabady R, Davaran S, et al. Liposome: classification, preparation, and applications. Nanoscale Res Lett. 2013;8(1):102.23432972
  • Sercombe L, Veerati T, Moheimani F, Wu SY, Sood AK, Hua S. Advances and challenges of liposome assisted drug delivery. Front Pharmacol. 2015;6:286.26648870
  • Loureiro JA, Gomes B, Coelho MA, Do Carmo Pereira M, Rocha S. Immunoliposomes doubly targeted to transferrin receptor and to alpha-synuclein. Future Sci OA. 2015;1(4):FSO71. doi:10.4155/fso.15.7128031922
  • Danhier F, Ansorena E, Silva JM, Coco R, Le Breton A, Préat V. PLGA-based nanoparticles: an overview of biomedical applications. J Controlled Release. 2012;161(2):505–522. doi:10.1016/j.jconrel.2012.01.043
  • Song X, Wang J, Xu Y, Shao H, Gu J. Surface-modified PLGA nanoparticles with PEG/LA-chitosan for targeted delivery of arsenic trioxide for liver cancer treatment: inhibition effects enhanced and side effects reduced. Colloids Surf B Biointerfaces. 2019;180:110–117. doi:10.1016/j.colsurfb.2019.04.03631030022
  • Loureiro JA, Gomes B, Fricker G, Coelho MAN, Rocha S, Pereira MC. Cellular uptake of PLGA nanoparticles targeted with anti-amyloid and anti-transferrin receptor antibodies for Alzheimer’s disease treatment. Colloids Surf B Biointerfaces. 2016;145:8–13.27131092
  • Deepika MS, Thangam R, Sheena TS, et al. Dual drug loaded PLGA nanospheres for synergistic efficacy in breast cancer therapy. Mater Sci Eng C Mater Biol Appl. 2019;103:109716. doi:10.1016/j.msec.2019.05.00131349431
  • Geszke-Moritz M, Moritz M. Solid lipid nanoparticles as attractive drug vehicles: composition, properties and therapeutic strategies. Mater Sci Eng C Mater Biol Appl. 2016;68:982–994. doi:10.1016/j.msec.2016.05.11927524099
  • Naseri N, Valizadeh H, Zakeri-Milani P. Solid lipid nanoparticles and nanostructured lipid carriers: structure, preparation and application. Adv Pharm Bull. 2015;5(3):305–313. doi:10.15171/apb.2015.04326504751
  • Loureiro JA, Andrade S, Duarte A, et al. Resveratrol and grape extract-loaded solid lipid nanoparticles for the treatment of Alzheimer’s disease. Molecules. 2017;22:2. doi:10.3390/molecules22020277
  • Zheng G, Zheng M, Yang B, Fu H, Li Y. Improving breast cancer therapy using doxorubicin loaded solid lipid nanoparticles: synthesis of a novel arginine-glycine-aspartic tripeptide conjugated, pH sensitive lipid and evaluation of the nanomedicine in vitro and in vivo. Biomedicine & Pharmacotherapy = Biomedecine & Pharmacotherapie. 2019;116:109006. doi:10.1016/j.biopha.2019.10900631152925
  • Guney Eskiler G, Cecener G, Dikmen G, Egeli U, Tunca B. Solid lipid nanoparticles: reversal of tamoxifen resistance in breast cancer. Eur J Pharm Sci. 2018;120:73–88. doi:10.1016/j.ejps.2018.04.04029719240
  • Mishra B, Patel BB, Tiwari S. Colloidal nanocarriers: a review on formulation technology, types and applications toward targeted drug delivery. Nanomedicine. 2010;6(1):9–24. doi:10.1016/j.nano.2009.04.00819447208
  • Helgason T, Awad TS, Kristbergsson K, McClements DJ, Weiss J. Effect of surfactant surface coverage on formation of solid lipid nanoparticles (SLN). J Colloid Interface Sci. 2009;334(1):75–81. doi:10.1016/j.jcis.2009.03.01219380149
  • Loureiro JA, Gomes B, Fricker G, et al. Dual ligand immunoliposomes for drug delivery to the brain. Colloids Surf B Biointerfaces. 2015;134:213–219. doi:10.1016/j.colsurfb.2015.06.06726204501
  • Ramalho MJ, Loureiro JA, Coelho MAN, Pereira MC. Factorial design as a tool for the optimization of PLGA nanoparticles for the co-delivery of temozolomide and O6-benzylguanine. Pharmaceutics. 2019;11:8. doi:10.3390/pharmaceutics11080401
  • Stetefeld J, McKenna SA, Patel TR. Dynamic light scattering: a practical guide and applications in biomedical sciences. Biophys Rev. 2016;8(4):409–427.28510011
  • Keenan CM, Baker JF, Bradley AE, et al. International Harmonization of Nomenclature and Diagnostic Criteria (INHAND) progress to date and future plans. J Toxicol Pathol. 2015;28(1):51–53. doi:10.1293/tox.2014-004926023262
  • Wohlfart S, Gelperina S, Kreuter J. Transport of drugs across the blood-brain barrier by nanoparticles. J Control Release. 2012;161(2):264–273. doi:10.1016/j.jconrel.2011.08.01721872624
  • Hoshyar N, Gray S, Han H, Bao G. The effect of nanoparticle size on in vivo pharmacokinetics and cellular interaction. Nanomedicine (Lond). 2016;11(6):673–692. doi:10.2217/nnm.16.527003448
  • Gaumet M, Vargas A, Gurny R, Delie F. Nanoparticles for drug delivery: the need for precision in reporting particle size parameters. Eur J Pharm Biopharm. 2008;69(1):1–9. doi:10.1016/j.ejpb.2007.08.00117826969
  • Blanco E, Shen H, Ferrari M. Principles of nanoparticle design for overcoming biological barriers to drug delivery. Nat Biotechnol. 2015;33(9):941–951. doi:10.1038/nbt.333026348965
  • Danaei M, Dehghankhold M, Ataei S, et al. Impact of particle size and polydispersity index on the clinical applications of lipidic nanocarrier systems. Pharmaceutics. 2018;10:2. doi:10.3390/pharmaceutics10020057
  • Ong SG, Chitneni M, Lee KS, Ming LC, Yuen KH. Evaluation of extrusion technique for nanosizing liposomes. Pharmaceutics. 2016;8:4.
  • Bhattacharjee S. DLS and zeta potential - What they are and what they are not? J Control Release. 2016;235:337–351. doi:10.1016/j.jconrel.2016.06.01727297779
  • Gelperina S, Maksimenko O, Khalansky A, et al. Drug delivery to the brain using surfactant-coated poly(lactide-co-glycolide) nanoparticles: influence of the formulation parameters. Eur J Pharm Biopharm. 2010;74(2):157–163. doi:10.1016/j.ejpb.2009.09.00319755158
  • Scholer N, Olbrich C, Tabatt K, Muller RH, Hahn H, Liesenfeld O. Surfactant, but not the size of solid lipid nanoparticles (SLN) influences viability and cytokine production of macrophages. Int J Pharm. 2001;221(1–2):57–67. doi:10.1016/S0378-5173(01)00660-311397567
  • Morris A. Microbiota drives sex-specific differences. Nat Rev Endocrinol. 2018;15(1):4.
  • Mauvais-Jarvis F, Arnold AP, Reue KA. Guide for the design of pre-clinical studies on sex differences in metabolism. Cell Metab. 2017;25(6):1216–1230. doi:10.1016/j.cmet.2017.04.03328591630
  • Scotland RS, Stables MJ, Madalli S, Watson P, Gilroy DW. Sex differences in resident immune cell phenotype underlie more efficient acute inflammatory responses in female mice. Blood. 2011;118(22):5918–5927. doi:10.1182/blood-2011-03-34028121911834
  • Uner M, Yener G. Importance of solid lipid nanoparticles (SLN) in various administration routes and future perspectives. Int J Nanomedicine. 2007;2(3):289–300.18019829
  • Sonavane G, Tomoda K, Makino K. Biodistribution of colloidal gold nanoparticles after intravenous administration: effect of particle size. Colloids Surf B Biointerfaces. 2008;66(2):274–280. doi:10.1016/j.colsurfb.2008.07.00418722754
  • Kim JH, Kim JH, Kim KW, Kim MH, Yu YS. Intravenously administered gold nanoparticles pass through the blood-retinal barrier depending on the particle size, and induce no retinal toxicity. Nanotechnology. 2009;20(50):505101. doi:10.1088/0957-4484/20/50/50510119923650
  • Chen YS, Hung YC, Liau I, Huang GS. Assessment of the in vivo toxicity of gold nanoparticles. Nanoscale Res Lett. 2009;4(8):858–864. doi:10.1007/s11671-009-9334-620596373
  • Cho WS, Kim S, Han BS, Son WC, Jeong J. Comparison of gene expression profiles in mice liver following intravenous injection of 4 and 100 nm-sized PEG-coated gold nanoparticles. Toxicol Lett. 2009;191(1):96–102.19695318
  • De Jong WH, Hagens WI, Krystek P, Burger MC, Sips AJ, Geertsma RE. Particle size-dependent organ distribution of gold nanoparticles after intravenous administration. Biomaterials. 2008;29(12):1912–1919. doi:10.1016/j.biomaterials.2007.12.03718242692
  • Semmler-Behnke M, Kreyling WG, Lipka J, et al. Biodistribution of 1.4- and 18-nm gold particles in rats. Small. 2008;4(12):2108–2111.19031432
  • Zhang XD, Wu HY, Wu D, et al. Toxicologic effects of gold nanoparticles in vivo by different administration routes. Int J Nanomedicine. 2010;5:771–781. doi:10.2147/IJN.S842821042423
  • Koziara JM, Lockman PR, Allen DD, Mumper RJ. In situ blood-brain barrier transport of nanoparticles. Pharm Res. 2003;20(11):1772–1778. doi:10.1023/B:PHAM.0000003374.58641.6214661921
  • Lockman PR, Mumper RJ, Khan MA, Allen DD. Nanoparticle technology for drug delivery across the blood-brain barrier. Drug Dev Ind Pharm. 2002;28(1):1–13.11858519
  • Vergoni AV, Tosi G, Tacchi R, Vandelli MA, Bertolini A, Costantino L. Nanoparticles as drug delivery agents specific for CNS: in vivo biodistribution. Nanomedicine. 2009;5(4):369–377. doi:10.1016/j.nano.2009.02.00519341816
  • Cho WS, Cho M, Jeong J, et al. Acute toxicity and pharmacokinetics of 13 nm-sized PEG-coated gold nanoparticles. Toxicol Appl Pharmacol. 2009;236(1):16–24.19162059
  • Balasubramanian SK, Jittiwat J, Manikandan J, Ong CN, Yu LE, Ong WY. Biodistribution of gold nanoparticles and gene expression changes in the liver and spleen after intravenous administration in rats. Biomaterials. 2010;31(8):2034–2042. doi:10.1016/j.biomaterials.2009.11.07920044133
  • Balogh L, Nigavekar SS, Nair BM, et al. Significant effect of size on the in vivo biodistribution of gold composite nanodevices in mouse tumor models. Nanomedicine. 2007;3(4):281–296. doi:10.1016/j.nano.2007.09.00117962085
  • Oberdorster G, Maynard A, Donaldson K, et al. Principles for characterizing the potential human health effects from exposure to nanomaterials: elements of a screening strategy. Part Fibre Toxicol. 2005;2:8. doi:10.1186/1743-8977-2-816209704
  • Choi J, Rui Y, Kim J, et al. Nonviral polymeric nanoparticles for gene therapy in pediatric CNS malignancies. Nanomedicine. 2020;23:102115. doi:10.1016/j.nano.2019.10211531655205
  • Pinheiro RGR, Granja A, Loureiro JA, et al. Quercetin lipid nanoparticles functionalized with transferrin for Alzheimer’s disease. Eur J Pharm Sci. 2020;148:105314. doi:10.1016/j.ejps.2020.10531432200044
  • Gupta R, Xie H. Nanoparticles in daily life: applications, toxicity and regulations. J Environ Pathol Toxicol Oncol. 2018;37(3):209–230. doi:10.1615/JEnvironPatholToxicolOncol.201802600930317972
  • Jeevanandam J, Barhoum A, Chan YS, Dufresne A, Danquah MK. Review on nanoparticles and nanostructured materials: history, sources, toxicity and regulations. Beilstein J Nanotechnol. 2018;9:1050–1074.29719757