LIMITATIONS OF PEGYLATED NANOCARRIERS: UNFAVOURABLE PHYSICOCHEMICAL PROPERTIES, BIODISTRIBUTION PATTERNS AND CELLULAR AND SUBCELLULAR FATES

Authors

  • Aya Ahmed Sebak Pharmaceutical Technology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC). Main Entrance of El-Tagamoa Al-Khames, New Cairo City, Egypt

DOI:

https://doi.org/10.22159/ijap.2018v10i5.27568

Keywords:

PEG, PEGylation, Nanoparticles, Nanovesicles, Drawbacks, Limitations, Alternatives

Abstract

Assuming that polyethylene glycol (PEG)-conjugated or PEGylated nanocarriers always offer outstanding physicochemical properties and pharmacokinetics profiles when compared to non-PEGylated ones, is not always accurate. For example, drug-loaded PEGylated nanocarriers for the treatment of cancer will not magically escape the reticuloendothelial system (RES) sequestration and clearance, benefit from the enhanced permeability and retention (EPR) effect of the tumor leaky vasculature and preferentially accumulate in the target tissue or cells. This is too good to be true. In this review, several drawbacks of PEGylation will be discussed; for example, how PEGylation can give rise to unfavourable physicochemical characteristics (e. g. particle size and release patterns) and post in vivo administration limitations of the formulated nanocarriers (e. g. limited evasion of RES uptake, development of hypersensitivity reactions, reduced intracellular accumulation and interference with the subcellular processing of nanocarriers necessary to produce the intended pharmacological effect).

This review aims at providing better understanding of the pros and cons of PEGylation, encouraging the use of PEGylation with caution, avoiding the assumption that PEGylation will provide all advantages needed to deliver nanocarriers to the target tissue and looking for alternatives to optimize nanocarriers' utilization especially in the delivery of chemotherapeutic agents for the treatment of different types of cancer. This review comprises a summary of some of the reported literature between 2013 and 2018 using different search engines; PubMed, Science Direct and Google Scholar, and the keywords listed below.

Downloads

Download data is not yet available.

References

Kudgus RA, Walden CA, Mcgovern RM, Reid JM, Robertson JD, Mukherjee P. Tuning pharmacokinetics and biodistribution of a targeted drug delivery system through the incorporation of a passive targeting component. Sci Rep 2014;4:5669.

Lázaro I, Haddad S, Sacca S, Orellana Tavra C, Fairen Jimenez D, Forgan RS. Selective surface PEGylation of UiO-66 nanoparticles for enhanced stability, cell uptake, and pH-responsive drug delivery. Chem 2017;2:561–78.

Barnard AS. Heterogeneous PEGylation of diamond nanoparticles. Nanoscale 2017;9:70–4.

Suk JS, Xu Q, Kim N, Hanes J, Ensign LM. PEGylation as a strategy for improving nanoparticle-based drug and gene delivery. Adv Drug Delivery Rev 2016;99:28–51.

Shaji J, Menon I. Recent advances in nanocarrier-based therapeutic and diagnostic tools for colorectal cancer. Int J Curr Pharm Res 2015;7:9–16.

Zhang F, Liu M, Wan H. Discussion about several potential drawbacks of PEGylated therapeutic proteins. Biol Pharm Bull 2014;37:335–9.

Sadat SM, Jahan ST, Haddadi A. Effects of size and surface charge of polymeric nanoparticles on in vitro and in vivo applications. J Biomater Nanobiotechnol 2016;7:91–108.

Mohammad AK, Reineke JJ. Quantitative detection of PLGA nanoparticle degradation in tissues following intravenous administration. Mol Biol Cell 2013;10:2183–9.

Rafiei P, Haddadi A. Docetaxel-loaded PLGA and PLGA-PEG nanoparticles for intravenous application: pharmacokinetics and biodistribution profile. Int J Nanomed 2017;12:935–47.

Xu J, Gattacceca F, Amiji M. Biodistribution and pharmacokinetics of EGFR-targeted thiolated gelatin nanoparticles following systemic administration in pancreatic tumour-bearing mice. Mol Pharm 2013;10:2031–44.

Behzadi S, Serpooshan V, Tao W, Hamaly MA, Alkawareek MY, Dreaden EC, et al. Cellular uptake of nanoparticles: a journey inside the cell. Chem Soc Rev 2017;46:4218–44.

Chen S, Yang K, Tuguntaev RG, Mozhi A, Zhang J, Wang PC, et al. Targeting tumor microenvironment with PEG-based amphiphilic nanoparticles to overcome chemoresistance. Nanomedicine 2016;12:269–86.

Verhoef JJF, Anchordoquy TJ. Questioning the use of PEGylation for drug delivery. Drug Delivery Transl Res 2013;3:499–503.

Kobayashi H, Watanabe R, Choyke PL. Improving conventional enhanced permeability and retention (EPR) effects; what is the appropriate target? Theranostics 2014;4:81–9.

D’souza AA, Shegokar R. Polyethylene glycol (PEG): a versatile polymer for pharmaceutical applications. Expert Opin Drug Delivery 2016;13:1257–75.

Jenkins SI, Weinberg D, Al-Shakli AF, Fernandes AR, Yiu HHP, Telling ND, et al.Stealth†nanoparticles evade neural-immune cells but also evade major brain cell populations: implications for PEG-based neurotherapeutics. J Controlled Release 2016;224:136–45.

Saraiva C, Praça C, Ferreira R, Santos T, Ferreira L, Bernardino L. Nanoparticle-mediated brain drug delivery: overcoming blood-brain barrier to treat neurodegenerative diseases. J Controlled Release 2016;235:34–47.

Tamba BI, Streinu V, Foltea G, Neagu AN, Dodi G, Zlei M, et al. Tailored surface silica nanoparticles for blood-brain barrier penetration: preparation and in vivo investigation. Arab J Chem 2018. https://doi.org/10.1016/j.arabjc.2018.03.019

Xu Q, Ensign LM, Boylan NJ, Schön A, Gong X, Yang JC, et al. Impact of surface polyethylene glycol (PEG) density on biodegradable nanoparticle transport in mucus ex vivo and distribution in vivo. ACS Nano 2015;9:9217–27.

Rattan R, Bhattacharjee S, Zong H, Swain C, Siddiqui MA, Visovatti SH, et al. Nanoparticle-macrophage interactions: a balance between clearance and cell-specific targeting. Bioorg Med Chem 2017;25:4487–96.

Yang X, Wu S, Wang Y, Li Y, Chang D, Luo Y, et al. Evaluation of self-assembled HCPT-loaded PEG-b-PLA nanoparticles by comparing with HCPT-loaded PLA nanoparticles. Nanoscale Res Lett 2014;9:1–8.

Chen F, Yin G, Liao X, Yang Y, Huang Z, Gu J. Preparation, characterization and in vitro release properties of morphine-loaded PLLA-PEG-PLLA microparticles via solution enhanced dispersion by supercritical fluids. J Mater Sci Mater Med 2013;24:1693–705.

Voon SH, Tiew SX, Kue CS, Lee HB, Kiew LV, Misran M, et al. Chitosan-coated poly(lactic-co-glycolic acid)-diiodinated boron-dipyrromethene nanoparticles improve tumor selectivity and stealth properties in photodynamic cancer therapy. J Biomed Nanotechnol 2016;12:1431–52.

Perche F, Torchilin VP. Recent trends in multifunctional liposomal nanocarriers for enhanced tumor targeting. J Drug Delivery 2013;201:3705265.

Yang Q, Lai SK. Anti-PEG immunity: emergence, characteristics, and unaddressed questions. Wiley Interdiscip Rev Nanomed Nanobiotechnol 2015;7:655–77.

Zhang P, Sun F, Liu S, Jiang S. Anti-PEG antibodies in the clinic: current issues and beyond PEGylation. J Controlled Release 2016;244:184–93.

Wan X, Zhang J, Yu W, Shen L, Ji S, Hu T. Effect of protein immunogenicity and PEG size and branching on the anti-PEG immune response to PEGylated proteins. Process Biochem 2017;52:183–91.

Dhand C, Prabhakaran MP, Beuerman RW, Lakshminarayanan R, Dwivedi N, Ramakrishna S. Role of the size of drug delivery carriers for pulmonary and intravenous administration with an emphasis on cancer therapeutics and lung-targeted drug delivery. RSC Adv 2014;4:32673–89.

Hoshyar N, Gray S, Han H, Bao G. The effect of nanoparticle size on in vivo pharmacokinetics and cellular interaction. Nanomedicine (Lond) 2016;11:673–92.

Hickey JW, Santos JL, Williford JM, Mao HQ. Control of polymeric nanoparticle size to improve therapeutic delivery. J Controlled Release 2015;219:536–47.

Shalgunov V, Zaytseva-zotova D, Zintchenko A, Levada T, Shilov Y, Andreyev D, et al. Comprehensive study of the drug delivery properties of poly (L-lactide)-poly (ethylene glycol) nanoparticles in rats and tumour-bearing mice. J Controlled Release 2017;261:31–42.

Ray S, Mishra A, Mandal TK, Sa B, Chakraborty J. Optimization of the process parameters for the fabrication of a polymer coated layered double hydroxide-methotrexate nanohybrid for the possible treatment of osteosarcoma. RSC Adv 2015;5:102574–92.

Voigt J, Christensen J, Shastri VP. Differential uptake of nanoparticles by endothelial cells through polyelectrolytes with affinity for caveolae. Proc Natl Acad Sci 2014;111:2942–7.

Yoo W, Yoo D, Hong E, Jung E, Go Y, Singh SVB, et al. Acid-activatable oxidative stress-inducing polysaccharide nanoparticles for anticancer therapy. J Controlled Release 2018;269:235–44.

Moayedian T, Mosaffa F, Khameneh B, Tafaghodi M. Combined effects of PEGylation and particle size on the uptake of PLGA particles by macrophage cells. Nanomed J 2015;2:299–304.

Kaur S, Vinay C, Narang R, Geeta A. Comparative mucopenetration ability of metronidazole-loaded chitosan and PEGylated chitosan nanoparticles. Asian J Pharm Clin Res 2017;10:125–30.

Srichaivatana K, Ounaroon A, Tiyaboonchai W. Development and characterization of piper retrofractum extract loaded mucoadhesive nanostructured lipid carriers for topical oral drug delivery. Int J Pharm Pharm Sci 2017;9:79–86.

Jahan ST, Sadat SM, Walliser M, Haddadi A. Targeted therapeutic nanoparticles: an immense promise to fight against cancer. J Drug Delivery 2017:9090325. https://doi.org/ 10.1155/2017/9090325

Kettiger H, Schipanski A, Wick P, Huwyler J. Engineered nanomaterial uptake and tissue distribution: from cell to organism. Int J Nanomed 2013;8:3255–69.

Chen BM, Su YC, Chang CJ, Burnouf PA, Chuang KH, Chen CH, et al. Measurement of pre-existing IgG and IgM antibodies against polyethylene glycol in healthy individuals. Anal Chem 2016;88:10661–6.

Hatakeyama H, Akita H, Harashima H. The polyethyleneglycol dilemma: advantage and disadvantage of PEGylation of liposomes for systemic genes and nucleic acids delivery to tumors. Biol Pharm Bull 2013;36:892–9.

Seicean A, Jinga M. Harmonic contrast-enhanced endoscopic ultrasound fine-needle aspiration: fact or fiction? Endosc Ultrasound 2017;6:31–6.

van der Sluis IM, Vrooman LM, Pieters R, Baruchel A, Escherich G, Goulden N, et al. Consensus expert recommendations for identification and management of asparaginase hypersensitivity and silent inactivation. Haematologica 2016;101:279–85.

Browne E, Moore C, Lu Z, Sykes A, Jeha S, Mandrell BN. Clinical characteristics of intravenous PEG-asparaginase hypersensitivity reactions in patients undergoing treatment for acute lymphoblastic leukemia. J Pediatr Oncol Nurs 2016;137:10160–3.

Dubey SK, Samanta MK, Dubey SK, Mishra P. Monoclonal antibody conjugated nanoparticles targeted to prostate tumor cells. Indian J Pharm Biol Res 2014;2:21–9.

Mao Z, Zhou X, Gao C. Influence of structure and properties of colloidal biomaterials on cellular uptake and cell functions. Biomater Sci 2013;1:896.

Oh N, Park JH. Endocytosis and exocytosis of nanoparticles in mammalian cells. Int J Nanomed 2014;9:51–63.

Qi Y, Chilkoti A. Protein-polymer conjugation-moving beyond PEGylation. Curr Opin Chem Biol 2015;28:181–93.

Hadjesfandiari N, Parambath A. Stealth coatings for nanoparticles. In: Parambath A. Engineering of Biomaterialfor Drug Delivery Systems: Beyond Polyethylene Glycol. Elsevier; 2018. p. 345–61.

Lowe S, O’Brien-Simpson NM, Connal LA. Antibiofouling polymer interfaces: poly(ethylene glycol) and other promising candidates. Polym Chem 2015;6:198–212.

Shimizu T, Abu Lila AS, Fujita R, Awata M, Kawanishi M, Hashimoto Y, et al. A hydroxyl PEG version of PEGylated liposomes and its impact on anti-PEG IgM induction and on the accelerated clearance of PEGylated liposomes. Eur J Pharm Biopharm 2018;127:142–9.

Saez Martinez V, Olalde B, Martinez Redondo D, Braceras I, Morin F, Valero J, et al. Degradable poly(ethylene glycol)-based hydrogels: synthesis, physicochemical properties and in vitro characterization. J Bioact Compat Polym 2014;29:270–83.

Ulbricht J, Jordan R, Luxenhofer R. On the biodegradability of polyethylene glycol, polypeptoids and poly(2-oxazoline)s. Biomaterials 2014;35:4848–61.

Murthy AK, Stover RJ, Hardin WG, Schramm R, Nie GD, Gourisankar S, et al. Charged gold nanoparticles with essentially zero serum protein adsorption in undiluted fetal bovine serum. J Am Chem Soc 2013;135:7799–802.

Zhou Y, Peng Z, Seven ES, Leblanc RM. Crossing the blood-brain barrier with nanoparticles. J Controlled Release 2018;270:290–303.

Garg NK, Tandel N, Jadon RS, Tyagi RK, Katare OP. Lipid–polymer hybrid nanocarrier-mediated cancer therapeutics: current status and future directions. Drug Discovery Today 2018. Doi:10.1016/j.drudis.2018.05.033

Dong W, Wang X, Liu C, Zhang X, Zhang X, Chen X, et al. Chitosan-based polymer-lipid hybrid nanoparticles for oral delivery of enoxaparin. Int J Pharm 2018;547:499-505.

Bose RJC, Ravikumar R, Karuppagounder V, Bennet D, Rangasamy S, Thandavarayan RA. Lipid–polymer hybrid nanoparticle-mediated therapeutics delivery: advances and challenges. Drug Discovery Today 2017;22:1258–65.

Date T, Nimbalkar V, Kamat J, Mittal A, Mahato RI, Chitkara D. Lipid-polymer hybrid nanocarriers for delivering cancer therapeutics. J Controlled Release 2018;271:60–73.

D’Souza AA, Shegokar R. Polymer: lipid hybrid nanostructures in cancer drug delivery: successes and limitations. In: Holban AM, Grumezescu A. Nanoarchitectonics for smart delivery and drug targeting. Elsevier; 2016. p. 431–63.

Li Q, Xia D, Tao J, Shen A, He Y, Gan Y, et al. Self-assembled core-shell-type lipid-polymer hybrid nanoparticles: intracellular trafficking and relevance for oral absorption. J Pharm Sci 2017;106:3120–30.

Salatin S, Khosroushahi AY. Overviews on the cellular uptake mechanism of polysaccharide colloidal nanoparticles. J Cell Mol Med 2017;20:1–19.

Levine RM, Kokkoli E. Dual-ligand α5β1 and α6β4 integrin targeting enhances gene delivery and selectivity to cancer cells. J Controlled Release 2017;10:24–36.

Sun C, Ding Y, Zhou L, Shi D, Sun L, Webster TJ, et al. Noninvasive nanoparticle strategies for brain tumor targeting. Nanomedicine 2017;13:2605–21.

Li J, Zhang C, Li J, Fan L, Jiang X, Chen J, et al. Brain delivery of NAP with PEG-PLGA nanoparticles modified with phage display peptides. Pharm Res 2013;30:1813–23.

Agrawal U, Chashoo G, Sharma PR, Kumar A, Saxena AK, Vyas SP. Tailored polymer–lipid hybrid nanoparticles for the delivery of drug conjugate: Dual strategy for brain targeting. Colloids Surf B 2015;12:414–25.

Published

07-09-2018

How to Cite

Sebak, A. A. (2018). LIMITATIONS OF PEGYLATED NANOCARRIERS: UNFAVOURABLE PHYSICOCHEMICAL PROPERTIES, BIODISTRIBUTION PATTERNS AND CELLULAR AND SUBCELLULAR FATES. International Journal of Applied Pharmaceutics, 10(5), 6–12. https://doi.org/10.22159/ijap.2018v10i5.27568

Issue

Section

Review Article(s)