DOI QR코드

DOI QR Code

Layer-by-layer assembled polymeric thin films as prospective drug delivery carriers: design and applications

  • Park, Sohyeon (Department of Chemical and Biomolecular Engineering, College of Engineering Yonsei University) ;
  • Han, Uiyoung (Department of Chemical and Biomolecular Engineering, College of Engineering Yonsei University) ;
  • Choi, Daheui (Department of Chemical and Biomolecular Engineering, College of Engineering Yonsei University) ;
  • Hong, Jinkee (Department of Chemical and Biomolecular Engineering, College of Engineering Yonsei University)
  • Received : 2018.06.25
  • Accepted : 2018.09.10
  • Published : 2018.12.31

Abstract

Background: The main purpose of drug delivery systems is to deliver the drugs at the appropriate concentration to the precise target site. Recently, the application of a thin film in the field of drug delivery has gained increasing interest because of its ability to safely load drugs and to release the drug in a controlled manner, which improves drug efficacy. Drug loading by the thin film can be done in various ways, depending on type of the drug, the area of exposure, and the purpose of drug delivery. Main text: This review summarizes the various methods used for preparing thin films with drugs via Layer-by-layer (LbL) assembly. Furthermore, additional functionalities of thin films using surface modification in drug delivery are briefly discussed. There are three types of methods for preparing a drug-carrying multilayered film using LbL assembly. First methods include approaches for direct loading of the drug into the pre-fabricated multilayer film. Second methods are preparing thin films using drugs as building blocks. Thirdly, the drugs are incorporated in the cargo so that the cargo itself can be used as the materials of the film. Conclusion: The appropriate designs of the drug-loaded film were produced in consideration of the release amounts and site of the desired drug. Furthermore, additional surface modification using the LbL technique enabled the preparation of effective drug delivery carriers with improved targeting effect. Therefore, the multilayer thin films fabricated by the LbL technique are a promising candidate for an ideal drug delivery system and the development possibilities of this technology are infinite.

Keywords

Acknowledgement

Supported by : Korea Health Industry Development Institute (KHIDI), National Research Foundation of Korea (NRF)

References

  1. Prausnitz MR, Mitragotri S, Langer R. Current status and future potential of transdermal drug delivery. Nat Rev Drug Discov. 2004;3:115. https://doi.org/10.1038/nrd1304
  2. Komiyama M, et al. Chemistry can make strict and fuzzy controls for biosystems: DNA nanoarchitectonics and cell-macromolecular nanoarchitectonics. Bull Chem Soc Jpn. 2017;90:967-1004. https://doi.org/10.1246/bcsj.20170156
  3. Tibbitt MW, Dahlman JE, Langer R. Emerging frontiers in drug delivery. J Am Chem Soc. 2016;138:704-17. https://doi.org/10.1021/jacs.5b09974
  4. Yan W, et al. Towards nanoporous polymer thin film-based drug delivery systems. Thin Solid Films. 2009;517:1794-8. https://doi.org/10.1016/j.tsf.2008.09.080
  5. Ariga K, et al. What are the emerging concepts and challenges in NANO? Nanoarchitectonics, hand-operating nanotechnology and mechanobiology. Polym J. 2016;48:371. https://doi.org/10.1038/pj.2016.8
  6. Zelikin AN. Drug releasing polymer thin films: new era of surface-mediated drug delivery. ACS Nano. 2010;4:2494-509. https://doi.org/10.1021/nn100634r
  7. Lavan DA, McGuire T, Langer R. Small-scale systems for in vivo drug delivery. Nat Biotechnol. 2003;21:1184. https://doi.org/10.1038/nbt876
  8. Quinn JF, et al. Next generation, sequentially assembled ultrathin films: beyond electrostatics. Chem Soc Rev. 2007;36:707-18. https://doi.org/10.1039/b610778h
  9. Howarth V, et al. Infrared studies of valinomycin-containing Langmuir-Blodgett films. Langmuir. 1989;5:330-2. https://doi.org/10.1021/la00086a006
  10. Park MH, et al. Controlled and sustained release of drugs from dendrimer-nanoparticle composite films. Adv Mater. 2011;23:2839-42. https://doi.org/10.1002/adma.201004409
  11. Pannier AK, Anderson BC, Shea LD. Substrate-mediated delivery from selfassembled monolayers: effect of surface ionization, hydrophilicity, and patterning. Acta Biomater. 2005;1:511-22. https://doi.org/10.1016/j.actbio.2005.05.004
  12. Mani G, et al. Drug delivery from gold and titanium surfaces using selfassembled monolayers. Biomaterials. 2008;29:4561-73. https://doi.org/10.1016/j.biomaterials.2008.08.014
  13. Vazquez E, et al. Construction of hydrolytically-degradable thin films via layer-by-layer deposition of degradable polyelectrolytes. J Am Chem Soc. 2002;124:13992-3. https://doi.org/10.1021/ja026405w
  14. Tang Z, et al. Biomedical applications of layer-by-layer assembly: from biomimetics to tissue engineering. Adv Mater. 2006;18:3203-24. https://doi.org/10.1002/adma.200600113
  15. Buck ME, Lynn DM. Reactive layer-by-layer assembly of suspended thin films and semipermeable membranes at interfaces created between aqueous and organic phases. Adv Mater. 2010;22:994-8. https://doi.org/10.1002/adma.200903054
  16. Park S, et al. Drug loading and release behavior depending on the induced porosity of chitosan/cellulose multilayer Nanofilms. Mol Pharmaceutics. 2017;14:3322-30. https://doi.org/10.1021/acs.molpharmaceut.7b00371
  17. Choi D, et al. Multifunctional collagen and hyaluronic acid multilayer films on live mesenchymal stem cells. ACS Appl Mater Interfaces. 2017;9:12264-71. https://doi.org/10.1021/acsami.7b00365
  18. Choi M, et al. Inkjet-based multilayered growth factor-releasing nanofilms for enhancing proliferation of mesenchymal stem cells in vitro. J Ind Eng Chem. 2017;50:36-40. https://doi.org/10.1016/j.jiec.2017.02.014
  19. Jeong H, et al. Electronic activation of a DNA nanodevice using a multilayer nanofilm. Small. 2016;12:5572-8. https://doi.org/10.1002/smll.201601273
  20. Heo J, Hong J. CO2 bubble assisted layer-by-layer self-assembly of weak polyelectrolyte multilayer film. J Ind Eng Chem. 2016;42:126-30. https://doi.org/10.1016/j.jiec.2016.07.032
  21. Heo J, Choi D, Hong J. Layer-by-layer self-assembled ferrite multilayer nanofilms for microwave absorption. J Nanomater. 2015;16:350.
  22. Lin X, Choi D, Hong J. Insulin particles as building blocks for controlled insulin release multilayer nano-films. Mater Sci Eng Proc Conf. 2015;54:239-44. https://doi.org/10.1016/j.msec.2015.05.046
  23. Hong J, et al. Carbon-based layer-by-layer nanostructures: from films to hollow capsules. Nanoscale. 2011;3:4515-31. https://doi.org/10.1039/c1nr10575b
  24. Ariga K, et al. Layer-by-layer self-assembled shells for drug delivery. Adv Drug Deliv Rev. 2011;63:762-71. https://doi.org/10.1016/j.addr.2011.03.016
  25. Heo J, et al. Highly permeable graphene oxide/polyelectrolytes hybrid thin films for enhanced CO 2/N 2 separation performance. Sci Rep. 2017;7:456. https://doi.org/10.1038/s41598-017-00433-z
  26. Heo J, Hong J. Effects of CO2 bubbles on layer-by-layer assembled hybrid thin film. Chem Eng J. 2016;303:433-8. https://doi.org/10.1016/j.cej.2016.06.030
  27. Wohl BM, Engbersen JF. Responsive layer-by-layer materials for drug delivery. J Control Release. 2012;158:2-14. https://doi.org/10.1016/j.jconrel.2011.08.035
  28. Min J, Braatz RD, Hammond PT. Tunable staged release of therapeutics from layer-by-layer coatings with clay interlayer barrier. Biomaterials. 2014;35:2507-17. https://doi.org/10.1016/j.biomaterials.2013.12.009
  29. Petrak K. Essential properties of drug-targeting delivery systems. Drug Discov Today. 2005;10:1667-73. https://doi.org/10.1016/S1359-6446(05)03698-6
  30. Li BL, et al. Directing assembly and disassembly of 2D MoS2 nanosheets with DNA for drug delivery. ACS Appl Mater Interfaces. 2017;9:15286-96. https://doi.org/10.1021/acsami.7b02529
  31. Goldberg M, Langer R, Jia X. Nanostructured materials for applications in drug delivery and tissue engineering. J Biomater Sci Polym Ed. 2007;18:241-68. https://doi.org/10.1163/156856207779996931
  32. Han U, Seo Y, Hong J. Effect of pH on the structure and drug release profiles of layer-by-layer assembled films containing polyelectrolyte, micelles, and graphene oxide. Sci Rep. 2016;6:24158. https://doi.org/10.1038/srep24158
  33. Kim B-S, Park SW, Hammond PT. Hydrogen-bonding layer-by-layerassembled biodegradable polymeric micelles as drug delivery vehicles from surfaces. ACS Nano. 2008;2:386-92. https://doi.org/10.1021/nn700408z
  34. Smith RC, et al. Layer-by-layer platform technology for small-molecule delivery. Angew Chem. 2009;121:9136-9. https://doi.org/10.1002/ange.200902782
  35. Shu X, Zhu K, Song W. Novel pH-sensitive citrate cross-linked chitosan film for drug controlled release. Int J Pharm. 2001;212:19-28. https://doi.org/10.1016/S0378-5173(00)00582-2
  36. Luo Y, Kirker KR, Prestwich GD. Cross-linked hyaluronic acid hydrogel films: new biomaterials for drug delivery. J Control Release. 2000;69:169-84. https://doi.org/10.1016/S0168-3659(00)00300-X
  37. Jiang B, Li B. Tunable drug loading and release from polypeptide multilayer nanofilms. Int J Nanomedicine. 2009;4:37.
  38. Kreft O, et al. Polymer microcapsules as mobile local pH-sensors. J Mater Chem. 2007;17:4471-6. https://doi.org/10.1039/b705419j
  39. Stuart MAC, et al. Emerging applications of stimuli-responsive polymer materials. Nat Mater. 2010;9:101. https://doi.org/10.1038/nmat2614
  40. Poon Z, et al. Layer-by-layer nanoparticles with a pH-sheddable layer for in vivo targeting of tumor hypoxia. ACS Nano. 2011;5:4284-92. https://doi.org/10.1021/nn200876f
  41. Manju S, Sreenivasan K. Enhanced drug loading on magnetic nanoparticles by layer-by-layer assembly using drug conjugates: blood compatibility evaluation and targeted drug delivery in cancer cells. Langmuir. 2011;27:14489-96. https://doi.org/10.1021/la202470k
  42. Uhrich KE, et al. Polymeric systems for controlled drug release. Chem Rev. 1999;99:3181-98. https://doi.org/10.1021/cr940351u
  43. Zhu Y, et al. Stimuli-responsive controlled drug release from a hollow mesoporous silica sphere/polyelectrolyte multilayer core-shell structure. Angew Chem. 2005;117:5213-7. https://doi.org/10.1002/ange.200501500
  44. Acharya G, Park K. Mechanisms of controlled drug release from drug-eluting stents. Adv Drug Deliv Rev. 2006;58:387-01. https://doi.org/10.1016/j.addr.2006.01.016
  45. Yoo HS, Kim TG, Park TG. Surface-functionalized electrospun nanofibers for tissue engineering and drug delivery. Adv Drug Deliv Rev. 2009;61:1033-42. https://doi.org/10.1016/j.addr.2009.07.007
  46. Katagiri K, et al. Preparation of pH-responsive hollow capsules via layer-bylayer assembly of exfoliated layered double hydroxide nanosheets and polyelectrolytes. J Nanosci Nanotechnol. 2018;18:110-5. https://doi.org/10.1166/jnn.2018.14590
  47. Vodouhe C, et al. Control of drug accessibility on functional polyelectrolyte multilayer films. Biomaterials. 2006;27:4149-56. https://doi.org/10.1016/j.biomaterials.2006.03.024
  48. Anandhakumar S, Raichur AM. Polyelectrolyte/silver nanocomposite multilayer films as multifunctional thin film platforms for remote activated protein and drug delivery. Acta Biomater. 2013;9:8864-74. https://doi.org/10.1016/j.actbio.2013.06.012
  49. Yang M, et al. Nanoporous multilayer films for controlled antigen protein release. J Ind Eng Chem. 2016;33:221-5. https://doi.org/10.1016/j.jiec.2015.10.009
  50. Serpe MJ, et al. Doxorubicin uptake and release from microgel thin films. Biomacromolecules. 2005;6:408-13. https://doi.org/10.1021/bm049455x
  51. Shukla A, et al. Tunable vancomycin releasing surfaces for biomedical applications. Small. 2010;6:2392-404. https://doi.org/10.1002/smll.201001150
  52. Shukla A, Fuller RC, Hammond PT. Design of multi-drug release coatings targeting infection and inflammation. J Control Release. 2011;155:159-66. https://doi.org/10.1016/j.jconrel.2011.06.011
  53. Min J, et al. Designer dual therapy nanolayered implant coatings eradicate biofilms and accelerate bone tissue repair. ACS Nano. 2016;10:4441-50. https://doi.org/10.1021/acsnano.6b00087
  54. Ma L, et al. Incorporation of basic fibroblast growth factor by a layer-bylayer assembly technique to produce bioactive substrates. J Biomed Mater Res, Part B. 2007;83:285-92.
  55. Su X, et al. Layer-by-layer-assembled multilayer films for transcutaneous drug and vaccine delivery. ACS Nano. 2009;3:3719-29. https://doi.org/10.1021/nn900928u
  56. Hong J, et al. Inherent charge-shifting polyelectrolyte multilayer blends: a facile route for tunable protein release from surfaces. Biomacromolecules. 2011;12:2975-81. https://doi.org/10.1021/bm200566k
  57. Chuang HF, Smith RC, Hammond PT. Polyelectrolyte multilayers for tunable release of antibiotics. Biomacromolecules. 2008;9:1660-8. https://doi.org/10.1021/bm800185h
  58. Macdonald ML, et al. Characterization of tunable FGF-2 releasing polyelectrolyte multilayers. Biomacromolecules. 2010;11:2053-9. https://doi.org/10.1021/bm100413w
  59. Choi D, et al. Controlled surface functionality of magnetic nanoparticles by layer-by-layer assembled nano-films. Nanoscale. 2015;7:6703-11. https://doi.org/10.1039/C4NR07373H
  60. Wiltsey C, et al. Thermogelling bioadhesive scaffolds for intervertebral disk tissue engineering: preliminary in vitro comparison of aldehyde-based versus alginate microparticle-mediated adhesion. Acta Biomater. 2015;16:71-80. https://doi.org/10.1016/j.actbio.2015.01.025
  61. Palivan CG, et al. Bioinspired polymer vesicles and membranes for biological and medical applications. Chem Soc Rev. 2016;45:377-411. https://doi.org/10.1039/C5CS00569H
  62. Elsabahy M, Wooley KL. Design of polymeric nanoparticles for biomedical delivery applications. Chem Soc Rev. 2012;41:2545-61. https://doi.org/10.1039/c2cs15327k
  63. Ma N, et al. Polymer micelles as building blocks for layer-by-layer assembly: an approach for incorporation and controlled release of water-insoluble dyes. Chem Mater. 2005;17:5065-9. https://doi.org/10.1021/cm051221c
  64. Benkirane-Jessel N, et al. Build-up of polypeptide multilayer coatings with anti-inflammatory properties based on the embedding of piroxicam- cyclodextrin complexes. Adv Funct Mater. 2004;14:174-82. https://doi.org/10.1002/adfm.200304413
  65. Lin M, et al. Facial layer-by-layer engineering of upconversion nanoparticles for gene delivery: near-infrared-initiated fluorescence resonance energy transfer tracking and overcoming drug resistance in ovarian cancer. ACS. Appl Mater Interfaces. 2017;9:7941-9. https://doi.org/10.1021/acsami.6b15321
  66. Chluba J, et al. Peptide hormone covalently bound to polyelectrolytes and embedded into multilayer architectures conserving full biological activity. Biomacromolecules. 2001;2:800-5. https://doi.org/10.1021/bm015529i
  67. Jessel N, et al. Bioactive coatings based on a polyelectrolyte multilayer architecture functionalized by embedded proteins. Adv Mater. 2003;15:692-5. https://doi.org/10.1002/adma.200304634
  68. Li Q-L, et al. Mesoporous silica nanoparticles coated by layer-by-layer selfassembly using cucurbit [7] uril for in vitro and in vivo anticancer drug release. Chem Mater. 2014;26:6418-31. https://doi.org/10.1021/cm503304p
  69. Feng W, et al. Effect of pH-responsive alginate/chitosan multilayers coating on delivery efficiency, cellular uptake and biodistribution of mesoporous silica nanoparticles based nanocarriers. ACS Appl Mater Interfaces. 2014;6:8447-60. https://doi.org/10.1021/am501337s
  70. Schmaljohann D. Thermo-and pH-responsive polymers in drug delivery. Adv Drug Deliv Rev. 2006;58:1655-70. https://doi.org/10.1016/j.addr.2006.09.020
  71. Caruso F, et al. 2. Assembly of alternating polyelectrolyte and protein multilayer films for immunosensing. Langmuir. 1997;13:3427-33. https://doi.org/10.1021/la9608223
  72. Qi W, et al. Triggered release of insulin from glucose-sensitive enzyme multilayer shells. Biomaterials. 2009;30:2799-806. https://doi.org/10.1016/j.biomaterials.2009.01.027
  73. Hu Y, et al. Fabrication of galactosylated polyethylenimine and plasmid DNA multilayers on poly (D, L-lactic acid) films for in situ targeted gene transfection. Adv Eng Mater. 2009;11:B30-4. https://doi.org/10.1002/adem.200800342
  74. Choi CHJ, et al. Mechanism of active targeting in solid tumors with transferrin-containing gold nanoparticles. Proc Natl Acad Sci. 2010;107:1235-40. https://doi.org/10.1073/pnas.0914140107
  75. Dreaden EC, et al. Bimodal tumor-targeting from microenvironment responsive hyaluronan layer-by-layer (LbL) nanoparticles. ACS Nano. 2014;8:8374-82. https://doi.org/10.1021/nn502861t
  76. Choi KY, et al. Smart nanocarrier based on PEGylated hyaluronic acid for cancer therapy. ACS Nano. 2011;5:8591-9. https://doi.org/10.1021/nn202070n
  77. Zoller M. CD44: can a cancer-initiating cell profit from an abundantly expressed molecule? Nat Rev Cancer. 2011;11:254. https://doi.org/10.1038/nrc3023
  78. Zhou J, et al. Layer by layer chitosan/alginate coatings on poly (lactide-coglycolide) nanoparticles for antifouling protection and folic acid binding to achieve selective cell targeting. J Colloid Interface Sci. 2010;345:241-7. https://doi.org/10.1016/j.jcis.2010.02.004
  79. Sun C. R. Sze, and M. Zhang, folic acid-PEG conjugated superparamagnetic nanoparticles for targeted cellular uptake and detection by MRI. J. Biomed Mater Res Part A. 2006;78:550-7.
  80. Zhang F, et al. Thermal treatment of galactose-branched polyelectrolyte microcapsules to improve drug delivery with reserved targetability. Int J Pharm. 2008;357:22-31. https://doi.org/10.1016/j.ijpharm.2008.01.021
  81. Hashida M, et al. Targeted delivery of drugs and proteins to the liver via receptor-mediated endocytosis. J Control Release. 1997;46:129-37. https://doi.org/10.1016/S0168-3659(96)01577-5
  82. Cook MT, et al. Layer-by-layer coating of alginate matrices with chitosan-alginate for the improved survival and targeted delivery of probiotic bacteria after oral administration. J Mater Chem B. 2013;1:52-60. https://doi.org/10.1039/C2TB00126H
  83. Anal AK, Singh H. Recent advances in microencapsulation of probiotics for industrial applications and targeted delivery. Trends Food Sci Technol. 2007;18:240-51. https://doi.org/10.1016/j.tifs.2007.01.004

Cited by

  1. Design of Therapeutic Self-Assembled Monolayers of Thiolated Abiraterone vol.8, pp.12, 2018, https://doi.org/10.3390/nano8121018
  2. Potential of Manuka Honey as a Natural Polyelectrolyte to Develop Biomimetic Nanostructured Meshes With Antimicrobial Properties vol.7, pp.None, 2018, https://doi.org/10.3389/fbioe.2019.00344
  3. Polysaccharide Thin Solid Films for Analgesic Drug Delivery and Growth of Human Skin Cells vol.7, pp.None, 2018, https://doi.org/10.3389/fchem.2019.00217
  4. Improvements on biological and antimicrobial properties of titanium modified by AgNPs-loaded chitosan-heparin polyelectrolyte multilayers vol.30, pp.5, 2018, https://doi.org/10.1007/s10856-019-6250-x
  5. Ampholytic and Polyelectrolytic Starch as Matrices for Controlled Drug Delivery vol.11, pp.6, 2019, https://doi.org/10.3390/pharmaceutics11060253
  6. Layer‐by‐Layer Formation of Polyamine‐Salt Aggregate/Polyelectrolyte Multilayers. Loading and Controlled Release of Probe Molecules from Self‐Assembled Supramolecular Networks vol.220, pp.15, 2019, https://doi.org/10.1002/macp.201900094
  7. Anti-inflammatory Surface Coatings Based on Polyelectrolyte Multilayers of Heparin and Polycationic Nanoparticles of Naproxen-Bearing Polymeric Drugs vol.20, pp.10, 2018, https://doi.org/10.1021/acs.biomac.9b01098
  8. Sustained delivery of growth factors with high loading efficiency in a layer by layer assembly vol.8, pp.1, 2018, https://doi.org/10.1039/c9bm00979e
  9. Polyelectrolytic BSA nanoparticles containing silicon dihydroxide phthalocyanine as a promising candidate for drug delivery systems for anticancer photodynamic therapy vol.31, pp.11, 2018, https://doi.org/10.1080/09205063.2020.1760702
  10. Dynamics of water trapped in transition metal oxide-graphene nano-confinement vol.32, pp.32, 2020, https://doi.org/10.1088/1361-648x/ab814f
  11. Ultrasonic Microplotting of Microgel Bioinks vol.12, pp.42, 2018, https://doi.org/10.1021/acsami.0c15056
  12. Biopolymer Coatings for Biomedical Applications vol.12, pp.12, 2020, https://doi.org/10.3390/polym12123061
  13. Recent Advances in Antiinflammatory Material Design vol.10, pp.1, 2021, https://doi.org/10.1002/adhm.202001373
  14. Layer-by-Layer assembled nano-drug delivery systems for cancer treatment vol.28, pp.1, 2018, https://doi.org/10.1080/10717544.2021.1905748
  15. Development of pH-Responsive Polymer Coating as an Alternative to Enzyme-Based Stem Cell Dissociation for Cell Therapy vol.14, pp.3, 2021, https://doi.org/10.3390/ma14030491
  16. Self-healing multilayer films for simultaneous release of hydrophilic and hydrophobic drugs vol.19, pp.2, 2021, https://doi.org/10.1080/1539445x.2020.1828099
  17. Investigation of the Structural Mechanism and Film Growth on Cytoprotective Type I Collagen-Based Nanocoating of Individual Cellular Surfaces vol.37, pp.15, 2018, https://doi.org/10.1021/acs.langmuir.1c00276
  18. Defect Repair of Polyelectrolyte Bilayers Using SDS: The Action of Micelles Versus Monomers vol.37, pp.17, 2021, https://doi.org/10.1021/acs.langmuir.1c00392
  19. pH-Responsive Chitosan/Alginate Polyelectrolyte Complexes on Electrospun PLGA Nanofibers for Controlled Drug Release vol.11, pp.7, 2021, https://doi.org/10.3390/nano11071850
  20. Chitosan/Cellulose-Based Porous Nanofilm Delivering C-Phycocyanin: A Novel Platform for the Production of Cost-Effective Cultured Meat vol.13, pp.27, 2021, https://doi.org/10.1021/acsami.1c07385
  21. Polyelectrolyte Gels: Fundamentals, Fabrication and Applications vol.7, pp.3, 2021, https://doi.org/10.3390/gels7030148