902
Views
71
CrossRef citations to date
0
Altmetric
Review

Novel nanomaterial-based antibacterial photodynamic therapies to combat oral bacterial biofilms and infectious diseases

, , , , , , , , & show all
Pages 6937-6956 | Published online: 28 Aug 2019

References

  • Petersen PE. World Health Organization global policy for improvement of oral health – World Health Assembly 2007. Int Dent J. 2008;58(3):115–121.18630105
  • He J, Li Y, Cao Y, Xue J, Zhou X. The oral microbiome diversity and its relation to human diseases. Folia Microbiol. 2015;60(1):69–80. doi:10.1007/s12223-014-0342-225147055
  • Murray CJ, Vos T, Lozano R, et al. Disability-adjusted life years (DALYs) for 291 diseases and injuries in 21 regions, 1990–2010: a systematic analysis for the Global Burden of Disease Study 2010. Lancet. 2012;380(9859):2197–2223. doi:10.1016/S0140-6736(12)61689-423245608
  • Haapasalo M, Endal U, Zandi H, Coil JM. Eradication of endodontic infection by instrumentation and irrigation solutions. Endod Topics. 2005;10(1):77–102. doi:10.1111/j.1601-1546.2005.00135.x
  • Peters L, Wesselink P, Buijs J, Van Winkelhoff A. Viable bacteria in root dentinal tubules of teeth with apical periodontitis. J Endodont. 2001;27(2):76–81. doi:10.1097/00004770-200102000-00002
  • Peters L, Wesselink P, Moorer W. Penetration of bacteria in bovine root dentine in vitro. Int Endodon J. 2000;33(1):28–36. doi:10.1046/j.1365-2591.2000.00268.x
  • Hengzhuang W, Wu H, Ciofu O, Song Z, Høiby N. Pharmacokinetics/pharmacodynamics of colistin and imipenem on mucoid and nonmucoid Pseudomonas aeruginosa biofilms. Antimicrob Agents Ch. 2011;55(9):4469–4474. doi:10.1128/AAC.00126-11
  • Dai T, Huang YY, Hamblin MR. Photodynamic therapy for localized infections – state of the art. Photodiagn Photodyn. 2009;6(3–4):170–188. doi:10.1016/j.pdpdt.2009.10.008
  • Hopper C. Photodynamic therapy: a clinical reality in the treatment of cancer. Lancet Oncol. 2000;1(4):212–219. doi:10.1016/S1470-2045(00)00166-211905638
  • Hamblin MR, Antimicrobial photodynamic inactivation: a bright new technique to kill resistant microbes. Curr Opin Microbiol. 2016;33:67–73. doi:10.1016/j.mib.2016.06.008
  • Carrera E, Dias H, Corbi S, et al. The application of antimicrobial photodynamic therapy (aPDT) in dentistry: a critical review. Laser Phys. 2016;(26):123001. doi:10.1088/1054-660X/26/12/12300129151775
  • Chrepa V, Kotsakis GA, Pagonis TC, Hargreaves KM. The effect of photodynamic therapy in root canal disinfection: a systematic review. J Endodont. 2014;40(7):891–898. doi:10.1016/j.joen.2014.03.005
  • Trindade AC, De Figueiredo JAP, Steier L, Weber JBB. Photodynamic therapy in endodontics: a literature review. Photomed Laser Surg. 2015;33(3):175–182. doi:10.1089/pho.2014.377625719896
  • Javed F, Romanos GE. Does photodynamic therapy enhance standard antibacterial therapy in dentistry? Photomed Laser Surg. 2013;31(11):512–518. doi:10.1089/pho.2012.332924138192
  • Gursoy H, Ozcakir-Tomruk C, Tanalp J, Yılmaz S. Photodynamic therapy in dentistry: a literature review. Clin Oral Invest. 2013;17(4):1113–1125. doi:10.1007/s00784-012-0845-7
  • Shrestha A, Kishen A. Antibacterial nanoparticles in endodontics: a review. J Endodon. 2016;42(10):1417–1426. doi:10.1016/j.joen.2016.05.021
  • Meimandi M, Ardakani MRT, Nejad AE, Yousefnejad P, Saebi K, Tayeed MH. The effect of photodynamic therapy in the treatment of chronic periodontitis: a review of literature. J Lasers Med Sci. 2017;8(Suppl 1):S7. doi:10.15171/jlms.2017.s229263777
  • Mondal D, Bera S. Porphyrins and phthalocyanines: promising molecules for light-triggered antibacterial nanoparticles. Adv Nat Sci. 2014;5(3):033002. doi:10.1088/2043-6262/5/3/033002
  • Yin R, Agrawal T, Khan U, et al. Antimicrobial photodynamic inactivation in nanomedicine: small light strides against bad bugs. Nanomedicine (London). 2015;10(15):2379–2404. doi:10.2217/nnm.15.67
  • Huang YY, Sharma SK, Dai T, et al. Can nanotechnology potentiate photodynamic therapy? Nanotechno Rev. 2012;1(2):111–146. doi:10.1515/ntrev-2011-0005
  • Ganoth A, Merimi KC, Peer D. Overcoming multidrug resistance with nanomedicines. Expert Opin Drug Del. 2015;12(2):223–238. doi:10.1517/17425247.2015.960920
  • Pellosi DS, De Jesus PCC, Tedesco AC. Spotlight on the delivery of photosensitizers: different approaches for photodynamic-based therapies. Expert Opin Drug Del. 2017;14(12):1395–1406. doi:10.1080/17425247.2017.1307337
  • Huang YY, Sharma SK, Yin R, Agrawal T, Chiang LY, Hamblin MR. Functionalized fullerenes in photodynamic therapy. J Biomed Nanotechnol. 2014;10(9):1918–1936. doi:10.1166/jbn.2014.196325544837
  • Mroz P, Tegos GP, Gali H, Wharton T, Sarna T, Hamblin MR. Photodynamic therapy with fullerenes. Photoch Photobio Sci. 2007;6(11):1139–1149. doi:10.1039/b711141j
  • Hamblin MR. Fullerenes as photosensitizers in photodynamic therapy: pros and cons. Photochem Photobiol Sci. 2018;17(11):1515–1533. doi:10.1039/C8PP00195B30043032
  • Aoshima H, Kokubo K, Shirakawa S, Ito M, Yamana S, Oshima T. Antimicrobial activity of fullerenes and their hydroxylated derivatives. Biocontrol Sci. 2009;14(2):69–72. doi:10.4265/bio.14.6919579658
  • Yin R, Wang M, Huang YY, et al. Antimicrobial photodynamic inactivation with decacationic functionalized fullerenes: oxygen-independent photokilling in presence of azide and new mechanistic insights. Free Radical Biol Med. 2015;79:14–27. doi:10.1016/j.freeradbiomed.2014.10.51425451642
  • Grinholc M, Nakonieczna J, Fila G, et al. Antimicrobial photodynamic therapy with fulleropyrrolidine: photoinactivation mechanism of Staphylococcus aureus, in vitro and in vivo studies. Appl Microbiol Biot. 2015;99(9):4031–4043. doi:10.1007/s00253-015-6539-8
  • Zhang J, Xu J, Ma H, et al. Designing an amino-fullerene derivative C70-(EDA)8 to fight super bacteria. ACS Appl Mater Inter. 2019;11(16):14597–14607. doi:10.1021/acsami.9b01483
  • Huang L, Bhayana B, Xuan W, et al. Comparison of two functionalized fullerenes for antimicrobial photodynamic inactivation: potentiation by potassium iodide and photochemical mechanisms. Photoch Photobio B. 2018;186:197–206. doi:10.1016/j.jphotobiol.2018.07.027
  • Zhang Y, Dai T, Wang M, Vecchio D, Chiang LY, Hamblin MR. Potentiation of antimicrobial photodynamic inactivation mediated by a cationic fullerene by added iodide: in vitro and in vivo studies. Nanomedicine. 2015;10(4):603–614. doi:10.2217/nnm.14.13125723093
  • Ballatore MB, Durantini J, Gsponer NS, et al. Photodynamic inactivation of bacteria using novel electrogenerated porphyrin-fullerene C60 polymeric films. Environ Sci Technol. 2015;49(12):7456–7463. doi:10.1021/acs.est.5b0140725984839
  • Ouyang K, Dai K, Chen H, Huang Q, Gao C, Cai P. Metal-free inactivation of E. coli O157: H7 by fullerene/C3N4 hybrid under visible light irradiation. Ecotoxicol Environ Saf. 2017;136:40–45. doi:10.1016/j.ecoenv.2016.10.03027810579
  • Wang Y, Jin Y, Chen W, et al. Construction of nanomaterials with targeting phototherapy properties to inhibit resistant bacteria and biofilm infections. Chem Eng J. 2019;358:74–90. doi:10.1016/j.cej.2018.10.002
  • Calver CF, Schanze KS, Cosa G. Biomimetic light-harvesting antenna based on the self-assembly of conjugated polyelectrolytes embedded within lipid membranes. ACS Nano. 2016;10(11):10598–10605. doi:10.1021/acsnano.6b0711127934088
  • Jeon S, Haley J, Flikkema J, et al. Linear and nonlinear optical properties of photoresponsive [60] fullerene hybrid triads and tetrads with dual NIR two-photon absorption characteristics. J Phys Chem C. 2013;117(33):17186–17195. doi:10.1021/jp405424q
  • Andrade EB, Martínez A, Free radical scavenger properties of metal-fullerenes: C60 and C82 with Cu, Ag and Au (atoms and tetramers). Comput Theor Chem. 2017;1115:127–135. doi:10.1016/j.comptc.2017.06.015
  • Bai RG, Ninan N, Muthoosamy K, Manickam S. Graphene: a versatile platform for nanotheranostics and tissue engineering. Prog Mater Sci. 2018;(91):24–69. doi:10.1016/j.pmatsci.2017.08.004
  • Ristic BZ, Milenkovic MM, Dakic IR, et al. Photodynamic antibacterial effect of graphene quantum dots. Biomaterials. 2014;35(15):4428–4435. doi:10.1016/j.biomaterials.2014.02.01424612819
  • Xie X, Ma C, Liu X, et al. Synergistic bacteria killing through photodynamic and physical actions of graphene oxide/Ag/collagen coating. ACS Appl Mater Inter. 2017;9(31):26417–26428. doi:10.1021/acsami.7b06702
  • Kuo WS, Chen HH, Chen SY, et al. Graphene quantum dots with nitrogen-doped content dependence for highly efficient dual-modality photodynamic antimicrobial therapy and bioimaging. Biomaterials. 2017;120:185–194. doi:10.1016/j.biomaterials.2016.12.02228063357
  • Santos CI, Gonçalves G, Cicuéndez M, et al. Biocompatible hybrids based on nanographene oxide covalently linked to glycolporphyrins: synthesis, characterization and biological evaluation. Carbon. 2018;135:202–214. doi:10.1016/j.carbon.2018.04.040
  • Xu X, Ho W, Zhang X, Bertrand N, Farokhzad O. Cancer nanomedicine: from targeted delivery to combination therapy. Trends Mol Med. 2015;21(4):223–232. doi:10.1016/j.molmed.2015.01.00125656384
  • Tian B, Wang C, Zhang S, Feng L, Liu Z. Photothermally enhanced photodynamic therapy delivered by nano-graphene oxide. ACS Nano. 2011;5(9):7000–7009. doi:10.1021/nn201560b21815655
  • Sah U, Sharma K, Chaudhri N, Sankar M, Gopinath P. Antimicrobial photodynamic therapy: single-walled carbon nanotube (SWCNT)-Porphyrin conjugate for visible light mediated inactivation of Staphylococcus aureus. Colloids Surf B. 2018;162:108–117. doi:10.1016/j.colsurfb.2017.11.046
  • Banerjee I, Mondal D, Martin J, Kane RS. Photoactivated antimicrobial activity of carbon nanotube−porphyrin conjugates. Langmuir. 2010;26(22):17369–17374. doi:10.1021/la103298e20931992
  • Xiao H, Zhu B, Wang D, et al. Photodynamic effects of chlorin e6 attached to single wall carbon nanotubes through noncovalent interactions. Carbon. 2012;50(4):1681–1689. doi:10.1016/j.carbon.2011.12.013
  • Anju VT, Paramanantham A, Sharan A, et al. Antimicrobial photodynamic activity of rose bengal conjugated multi walled carbon nanotubes against planktonic cells and biofilm of Escherichia coli. Photodiagn Photodyn. 2018;24:300–310. doi:10.1016/j.pdpdt.2018.10.013
  • Sáfar GA, Gontijo RN, Fantini C, et al. Enhanced oxygen singlet production by hybrid system of porphyrin and enriched (6, 5) single-walled carbon nanotubes for photodynamic therapy. J Phys Chem C. 2015;119(8):4344–4350. doi:10.1021/jp5111289
  • Zhu Z, Tang Z, Phillips JA, Yang R, Wang H, Tan W. Regulation of singlet oxygen generation using single-walled carbon nanotubes. J Am Chem Soc. 2008;130(33):10856–10857. doi:10.1021/ja802913f18661988
  • Sarwar S, Chakraborti S, Bera S, Sheikh IA, Hoque KM, Chakrabarti P. The antimicrobial activity of ZnO nanoparticles against Vibrio cholerae: variation in response depends on biotype. Nanomedicine. 2016;12(6):1499–1509. doi:10.1016/j.nano.2016.02.00626970029
  • Colmenares JC, Luque R. Heterogeneous photocatalytic nanomaterials: prospects and challenges in selective transformations of biomass-derived compounds. Chem Soc Rev. 2014;43(3):765–778. doi:10.1039/c3cs60262a24217399
  • Podporska-Carroll J, Myles A, Quilty B, et al. Antibacterial properties of F-doped ZnO visible light photocatalyst. J Hazard Mater. 2017;324:39–47. doi:10.1016/j.jhazmat.2015.12.03826782784
  • Sethi D, Sakthivel R. ZnO/TiO2 composites for photocatalytic inactivation of Escherichia coli. Photoch Photobio B. 2017;168:117–123. doi:10.1016/j.jphotobiol.2017.02.005
  • Reddy KR, Karthik K, Prasad SB, Soni SK, Jeong HM, Raghu AV. Enhanced photocatalytic activity of nanostructured titanium dioxide/polyaniline hybrid photocatalysts. Polyhedron. 2016;120:169–174. doi:10.1016/j.poly.2016.08.029
  • Suketa N, Sawase T, Kitaura H, et al. An antibacterial surface on dental implants, based on the photocatalytic bactericidal effect. Clin Implant Dent R. 2005;7(2):105–111. doi:10.1111/j.1708-8208.2005.tb00053.x
  • Lilja M, Forsgren J, Welch K, Åstrand M, Engqvist H, Strømme M. Photocatalytic and antimicrobial properties of surgical implant coatings of titanium dioxide deposited though cathodic arc evaporation. Biotechnol Lett. 2012;34(12):2299–2305. doi:10.1007/s10529-012-1040-222941372
  • Huang YY, Choi H, Kushida Y, Bhayana B, Wang Y, Hamblin MR. Broad-spectrum antimicrobial effects of photocatalysis using titanium dioxide nanoparticles are strongly potentiated by addition of potassium iodide. Antimicrob Agents Ch. 2016;60(9):5445–5453. doi:10.1128/AAC.00980-16
  • Gao L, Giglio KM, Nelson JL, Sondermann H, Travis AJ. Ferromagnetic nanoparticles with peroxidase-like activity enhance the cleavage of biological macromolecules for biofilm elimination. Nanoscale. 2014;6(5):2588–2593. doi:10.1039/c3nr05422e24468900
  • Tassa C, Shaw SY, Weissleder R. Dextran-coated iron oxide nanoparticles: a versatile platform for targeted molecular imaging, molecular diagnostics, and therapy. Accounts Chem Res. 2011;44(10):842–852. doi:10.1021/ar200084x
  • Choi KH, Lee HJ, Park BJ, et al. Photosensitizer and vancomycin-conjugated novel multifunctional magnetic particles as photoinactivation agents for selective killing of pathogenic bacteria. Chem Commun. 2012;48(38):4591–4593. doi:10.1039/c2cc17766h
  • Pan WY, Huang CC, Lin TT, et al. Synergistic antibacterial effects of localized heat and oxidative stress caused by hydroxyl radicals mediated by graphene/iron oxide-based nanocomposites. Nanomedicine. 2016;12(2):431–438. doi:10.1016/j.nano.2015.11.01426711965
  • Cui S, Yin D, Chen Y, et al. In vivo targeted deep-tissue photodynamic therapy based on near-infrared light triggered upconversion nanoconstruct. ACS Nano. 2012;7(1):676–688. doi:10.1021/nn304872n23252747
  • Haase M, Schäfer H. Upconverting nanoparticles. Angew Chem Int Edit. 2011;50(26):5808–5829. doi:10.1002/anie.201005159
  • Zhang Y, Huang P, Wang D, et al. Near-infrared-triggered antibacterial and antifungal photodynamic therapy based on lanthanide-doped upconversion nanoparticles. Nanoscale. 2018;10(3):15485–15495. doi:10.1039/c8nr01967c29881851
  • Xu F, Hu M, Liu C, Choi SK. Yolk-structured multifunctional up-conversion nanoparticles for synergistic photodynamic–sonodynamic antibacterial resistance therapy. Biometer Sci. 2017;5(4):678–685. doi:10.1039/C7BM00030H
  • Grüner MC, Arai MS, Carreira M, Inada N, Camargo ASS. Functionalizing the mesoporous silica shell of upconversion nanoparticles to enhance bacterial targeting and killing via photosensitizer-induced antimicrobial photodynamic therapy. ACS Appl Bio Mater. 2018;1(4):1028–1036. doi:10.1021/acsabm.8b00224
  • Yin M, Li Z, Ju E, et al. Multifunctional upconverting nanoparticles for near-infrared triggered and synergistic antibacterial resistance therapy. Chem Commun. 2014;50(72):10488–10490. doi:10.1039/C4CC04584J
  • Amini SM, Kharrazi S, Hadizadeh M, Fateh M, Saber R. Effect of gold nanoparticles on photodynamic efficiency of 5-aminolevolenic acid photosensitiser in epidermal carcinoma cell line: an in vitro study. IET Nanobiotechnol. 2013;7(4):151–156. doi:10.1049/iet-nbt.2013.002124206772
  • Darabpour E, Kashef N, Amini SM, Kharrazi S, Djavid GE. Fast and effective photodynamic inactivation of 4-day-old biofilm of methicillin-resistant Staphylococcus aureus using methylene blue-conjugated gold nanoparticles. J Drug Deliv Sci Tec. 2017;37:134–140. doi:10.1016/j.jddst.2016.12.007
  • Maliszewska I, Leśniewska A, Olesiak-Bańska J, Matczyszyn K, Samoć M. Biogenic gold nanoparticles enhance methylene blue-induced phototoxic effect on Staphylococcus epidermidis. J Nanopart Res. 2014;16(6):2457. doi:10.1007/s11051-014-2457-4
  • Turcheniuk K, Turcheniuk V, Hage CH, et al. Highly effective photodynamic inactivation of E. coli using gold nanorods/SiO2 core–shell nanostructures with embedded verteporfin. Chem Commun. 2015;51(91):16365–16368. doi:10.1039/C5CC06738C
  • Planas O, Bresolí-Obach R, Nos J, et al. Synthesis, photophysical characterization, and photoinduced antibacterial activity of methylene blue-loaded amino-and mannose-targeted mesoporous silica nanoparticles. Molecules. 2015;20(4):6284–6298. doi:10.3390/molecules2004628425859784
  • Brevet D, Gary-Bobo M, Raehm L, et al.Mannose-targeted mesoporous silica nanoparticles for photodynamic therapy. Chem Commun. 2009;(12):1475–1477. doi:10.1039/b900427k
  • Wysocka-Król K, Olsztyńska-Janus S, Plesch G, Plecenik A, Podbielska H, Bauer J. Nano-silver modified silica particles in antibacterial photodynamic therapy. Appl Surf Sci. 2018;461:260–268. doi:10.1016/j.apsusc.2018.05.014
  • Lin J, Li J, Gopal A, et al. Synthesis of photo-excited Chlorin e6 conjugated silica nanoparticles for enhanced anti-bacterial efficiency to overcome methicillin-resistant Staphylococcus aureus. Chem Commun. 2019;55(18):2656–2659. doi:10.1039/C9CC00166B
  • Sadasivam M, Avci P, Gupta GK, et al. Self-assembled liposomal nanoparticles in photodynamic therapy. Eur J Nanomed. 2013;5(3):115–129. doi:10.1515/ejnm-2013-0010
  • Mesquita MQ, Dias CJ, Neves M PMS, Almeida A, Faustino MAF. Revisiting current photoactive materials for antimicrobial photodynamic therapy. Molecules. 2018;23(10):2424. doi:10.3390/molecules23102424
  • Yang K, Gitter B, Rüger R, et al. Antimicrobial peptide-modified liposomes for bacteria targeted delivery of temoporfin in photodynamic antimicrobial chemotherapy. Photoch Photobio Sci. 2011;10(10):1593–1601. doi:10.1039/c1pp05100h
  • Jia Y, Joly H, Omri A. Characterization of the interaction between liposomal formulations and Pseudomonas aeruginosa. J Liposome Res. 2010;20(2):134–146. doi:10.3109/0898210090321889219831502
  • Jin CS, Zheng G. Liposomal nanostructures for photosensitizer delivery. Laser Surg Med. 2011;43(7):734–748. doi:10.1002/lsm.21101
  • Nitzan Y, Dror R, Ladan H, Malik Z, Kimel S, Gottfried V. Structure‐activity relationship of porphines for photoinactivation of bacteria. Photochem Photobiol. 1995;62(2):342–347. doi:10.1111/j.1751-1097.1995.tb05279.x7480142
  • Yang YT, Chien HF, Chang PH, et al. Photodynamic inactivation of chlorin e6‐loaded CTAB‐liposomes against Candida albicans. Laser Surg Med. 2013;45(3):175–185. doi:10.1002/lsm.22124
  • Park H, Lee J, Jeong S, et al. Lipase‐sensitive transfersomes based on photosensitizer/polymerizable lipid conjugate for selective antimicrobial photodynamic therapy of acne. Adv Healthc Mater. 2016;5(24):3139–3147. doi:10.1002/adhm.20160081527863184
  • Deng X, Liang Y, Peng X, et al. A facile strategy to generate polymeric nanoparticles for synergistic chemo-photodynamic therapy. Chem Commun. 2015;51(20):4271–4274. doi:10.1039/C4CC10226F
  • Conte C, Maiolino S, Pellosi DS, Miro A, Ungaro F, Quaglia F. Polymeric nanoparticles for cancer photodynamic therapy In: Sortino S, editor. Light-responsive Nanostructured Systems for Applications in Nanomedicine. Springer International Publishing Switzerland; 2016:61–112.
  • Xiao F, Cao B, Wang C, et al. Pathogen-specific polymeric antimicrobials with significant membrane disruption and enhanced photodynamic damage to inhibit highly opportunistic bacteria. ACS Nano. 2019;13(2):1511–1525. doi:10.1021/acsnano.8b0725130632740
  • Darabpour E, Kashef N, Mashayekhan S. Chitosan nanoparticles enhance the efficiency of methylene blue-mediated antimicrobial photodynamic inactivation of bacterial biofilms: an in vitro study. Photodiagn Photodyn. 2016;14:211–217. doi:10.1016/j.pdpdt.2016.04.009
  • Pourhajibagher M, Rostami Rad M, Bahador A. Monitoring of virulence factors and metabolic activity in Aggregatibacter actinomycetemcomitans cells surviving antimicrobial photodynamic therapy via nano-chitosan encapsulated indocyanine green. Front Phys. 2018;6:124. doi:10.3389/fphy.2018.00124
  • Shrestha A, Kishen A. Polycationic chitosan‐conjugated photosensitizer for antibacterial photodynamic therapy. Photochem Photobiol. 2012;88(3):577–583. doi:10.1111/j.1751-1097.2011.01026.x22044238
  • Chen CP, Chen CT, Tsai T. Chitosan nanoparticles for antimicrobial photodynamic inactivation: characterization and in vitro investigation. Photochem Photobiol. 2012;88(3):570–576. doi:10.1111/j.1751-1097.2012.01101.x22283820
  • Choi SS, Lee HK, Chae HS. Synergistic in vitro photodynamic antimicrobial activity of methylene blue and chitosan against Helicobacter pylori 26695. Photodiagn Photodyn. 2014;11(4):526–532. doi:10.1016/j.pdpdt.2014.08.005
  • Carpenter BL, Feese E, Sadeghifar H, Argyropoulos DS, Ghiladi RA. Porphyrin‐cellulose nanocrystals: a photobactericidal material that exhibits broad spectrum antimicrobial activity. Photochem Photobiol. 2012;88(3):527–536. doi:10.1111/j.1751-1097.2012.01117.x22360680
  • Pagonis TC, Chen J, Fontana CR, et al. Nanoparticle-based endodontic antimicrobial photodynamic therapy. J Endodon. 2010;36(2):322–328. doi:10.1016/j.joen.2009.10.011
  • González-Delgado JA, Castro PM, Machado A, et al. Hydrogels containing porphyrin-loaded nanoparticles for topical photodynamic applications. Int J Pharm. 2016;510(1):221–231. doi:10.1016/j.ijpharm.2016.06.03727321129
  • de Freitas L, Calixto G, Chorilli M, et al. Polymeric nanoparticle-based photodynamic therapy for chronic periodontitis in vivo. Int J Mol Sci. 2016;17(5):769. doi:10.3390/ijms17050769
  • Liu S, Qiao S, Li L, et al. Surface charge-conversion polymeric nanoparticles for photodynamic treatment of urinary tract bacterial infections. Nanotechnology. 2015;26(49):495602. doi:10.1088/0957-4484/26/49/49560226572164
  • Felgenträger A, Maisch T, Späth A, et al. Singlet oxygen generation in porphyrin-doped polymeric surface coating enables antimicrobial effects on Staphylococcus aureus. Phys Chem Chem Phys. 2014;16(38):20598–20607. doi:10.1039/c4cp02439g25155698
  • Ma BC, Ghasimi S, Landfester K, et al. Enhanced visible light promoted antibacterial efficiency of conjugated microporous polymer nanoparticles via molecular doping. J Mater Chem B. 2016;4(30):5112–5118. doi:10.1039/C6TB00943C
  • Stanley S, Scholle F, Zhu J, et al. Photosensitizer-embedded polyacrylonitrile nanofibers as antimicrobial non-woven textile. Nanomaterials. 2016;6(4):77. doi:10.3390/nano6040077
  • de Avila ED, Lima BP, Sekiya T, et al. Effect of UV-photofunctionalization on oral bacterial attachment and biofilm formation to titanium implant material. Biomaterials. 2015;67:84–92. doi:10.1016/j.biomaterials.2015.07.03026210175
  • D’Orazio J, Jarrett S, Amaro-Ortiz A, Scott T. UV radiation and the skin. Int J Mol Sci. 2013;14(6):12222–12248. doi:10.3390/ijms14061222223749111
  • Shi CH, Zhu NW, Cao YL, Wu PX. Biosynthesis of gold nanoparticles assisted by the intracellular protein extract of Pycnoporus sanguineus and its catalysis in degradation of 4-nitroaniline. Nanoscale Res Lett. 2015;10(1):147. doi:10.1186/s11671-015-0856-925852436
  • Tim M. Strategies to optimize photosensitizers for photodynamic inactivation of bacteria. J Photoch Photobio B. 2015;150:2–10. doi:10.1016/j.jphotobiol.2015.05.010
  • Kishen A. Advanced Therapeutic Options to Disinfect Root Canals In: Chávez de Paz LE, Sedgley CM, Kishen A, editors. The Root Canal Biofilm. Springer-Verlag Berlin Heidelberg; 2015:334–335.
  • Gatoo MA, Naseem S, Arfat MY, Mahmood Dar A, Qasim K, Zubair S. Physicochemical properties of nanomaterials: implication in associated toxic manifestations. Biomed Res Int. 2014;2014:1–8. doi:10.1155/2014/498420
  • Chi M, Qi M, Lan A, et al. Novel bioactive and therapeutic dental polymeric materials to inhibit periodontal pathogens and biofilms. Int J Mol Sci. 2019;20(2):278. doi:10.3390/ijms20020278
  • Beytollahi L, Pourhajibagher M, Chiniforush N, et al. The efficacy of photodynamic and photothermal therapy on biofilm formation of Streptococcus mutans: an in vitro study. Photodiagn Photodyn. 2017;17:56–60. doi:10.1016/j.pdpdt.2016.10.006
  • Araújo N, Fontana CR, Bagnato V, Gerbi M. Photodynamic antimicrobial therapy of curcumin in biofilms and carious dentine. Laser Med Sci. 2014;29(2):629–635. doi:10.1007/s10103-013-1369-3
  • Misba L, Kulshrestha S, Khan AU. Antibiofilm action of a toluidine blue O-silver nanoparticle conjugate on Streptococcus mutans: a mechanism of type I photodynamic therapy. Biofouling. 2016;32(3):313–328. doi:10.1080/08927014.2016.114189926905507
  • Parker S. The use of diffuse laser photonic energy and indocyanine green photosensitiser as an adjunct to periodontal therapy. Brit Dent J. 2013;215(4):167. doi:10.1038/sj.bdj.2013.79023969654
  • Gholibegloo E, Karbasi A, Pourhajibagher M, et al. Carnosine-graphene oxide conjugates decorated with hydroxyapatite as promising nanocarrier for ICG loading with enhanced antibacterial effects in photodynamic therapy against Streptococcus mutans. J Photoch Photobio B. 2018;181:14–22. doi:10.1016/j.jphotobiol.2018.02.004
  • Siqueira JF Jr. Endodontic infections: concepts, paradigms, and perspectives. Oral Surg Oral Med O. 2002;94(3):281–293. doi:10.1067/moe.2002.126163
  • Stojanović N, Krunić J, Popović B, Stojičić S, Živković S. Prevalence of Enterococcus faecalis and Porphyromonas gingivalis in infected root canals and their susceptibility to endodontic treatment procedures: a molecular study. Srp Ark Celok Lek. 2014;142(9–10):535–541. doi:10.2298/SARH1410535S
  • Komiyama EY, Lepesqueur LSS, Yassuda CG, et al. Enterococcus species in the oral cavity: prevalence, virulence factors and antimicrobial susceptibility. PLoS One. 2016;11(9):e0163001. doi:10.1371/journal.pone.016300127631785
  • Slutzky-Goldberg I, Maree M, Liberman R, Heling I. Effect of sodium hypochlorite on dentin microhardness. J Endodon. 2004;30:880–882. doi:10.1097/01.DON.0000128748.05148.1E
  • Ari H, Yaşar E, Bellí S. Effects of NaOCl on bond strengths of resin cements to root canal dentin. J Endodon. 2003;29(4):248–251. doi:10.1097/00004770-200304000-00004
  • Shrestha A, Kishen A. Antibacterial efficacy of photosensitizer functionalized biopolymeric nanoparticles in the presence of tissue inhibitors in root canal. J Endodon. 2014;40(4):566–570. doi:10.1016/j.joen.2013.09.013
  • Shrestha A, Kishen A. Antibiofilm efficacy of photosensitizer-functionalized bioactive nanoparticles on multispecies biofilm. J Endod. 2014;40(10):1604–1610. doi:10.1016/j.joen.2014.03.00925260731
  • Shrestha A, Hamblin MR, Kishen A. Photoactivated rose bengal functionalized chitosan nanoparticles produce antibacterial/biofilm activity and stabilize dentin-collagen. Nanomedicine. 2014;10(3):491–501. doi:10.1016/j.nano.2013.10.01024200522
  • Shrestha A, Cordova M, Kishen A. Photoactivated polycationic bioactive chitosan nanoparticles inactivate bacterial endotoxins. J Endodon. 2015;41(5):686–691. doi:10.1016/j.joen.2014.12.007
  • Akbari T, Pourhajibagher M, Hosseini F, et al. The effect of indocyanine green loaded on a novel nano-graphene oxide for high performance of photodynamic therapy against Enterococcus faecalis. Photodiagn Photodyn. 2017;20:148–153. doi:10.1016/j.pdpdt.2017.08.017
  • Afkhami F, Akbari S, Chiniforush N. Entrococcus faecalis elimination in root canals using silver nanoparticles, photodynamic therapy, diode laser, or laser-activated nanoparticles: an in vitro study. J Endodon. 2017;43(2):279–282. doi:10.1016/j.joen.2016.08.029
  • Golmohamadpour A, Bahramian B, Khoobi M, Pourhajibagher M, Barikani HR, Bahador A. Antimicrobial photodynamic therapy assessment of three indocyanine green-loaded metal-organic frameworks against Enterococcus faecalis. Photodiagn Photocyn. 2018;23:331–338. doi:10.1016/j.pdpdt.2018.08.004
  • Klepac‐Ceraj V, Patel N, Song X, et al. Photodynamic effects of methylene blue‐loaded polymeric nanoparticles on dental plaque bacteria. Laser Surg Med. 2011;43(7):600–606. doi:10.1002/lsm.21069
  • Nagahara A, Mitani A, Fukuda M, et al. Antimicrobial photodynamic therapy using a diode laser with a potential new photosensitizer, indocyanine green‐loaded nanospheres, may be effective for the clearance of Porphyromonas gingivalis. J Periodontal Res. 2013;48(5):591–599. doi:10.1111/jre.1204223317284
  • Sasaki Y, Hayashi J, Fujimura T, et al. New irradiation method with indocyanine green-loaded nanospheres for inactivating periodontal pathogens. Int J Mol Sci. 2017;(18):154. doi:10.3390/ijms18010154
  • Sun X, Wang L, Lynch C, et al. Nanoparticles having amphiphilic silane containing Chlorin e6 with strong anti-biofilm activity against periodontitis-related pathogens. J Dent. 2019;81:70–84. doi:10.1016/j.jdent.2018.12.01130593855
  • Socransky SS, Haffajee AD, Cugini MA, Smith C, Kent RL. Microbial complexes in subgingival plaque. J Clin Periodontol. 1998;25(2):134–144. doi:10.1111/j.1600-051X.1998.tb02419.x9495612
  • Mombelli A, Lang NP. The diagnosis and treatment of peri‐implantitis. Periodontol 2000. 1998;17(1):63–76. doi:10.1111/j.1600-0757.1998.tb00124.x10337314
  • Besinis A, Hadi SD, Le H, Tredwin C, Handy R. Antibacterial activity and biofilm inhibition by surface modified titanium alloy medical implants following application of silver, titanium dioxide and hydroxyapatite nanocoatings. Nanotoxicology. 2017;11(3):327–338. doi:10.1080/17435390.2017.129989028281851
  • Coronado-Castellote L, Jiménez-Soriano Y. Clinical and microbiological diagnosis of oral candidiasis. J Clin Exp Dent. 2013;5(5):e279–e286. doi:10.4317/jced.5124224455095
  • Colomba C, Trizzino M, Imburgia C, Madonia S, Siracusa L, Giammanco GM. Candida glabrata meningitis and endocarditis: a late severe complication of candidemia. Int J Infect Dis. 2014;29:174–175. doi:10.1016/j.ijid.2014.04.03225449253
  • Williams D, Lewis M. Pathogenesis and treatment of oral candidosis. J Oral Microbiol. 2011;3(1):5771. doi:10.3402/jom.v3i0.5771
  • Mthethwa T, Nyokong T. Photoinactivation of Candida albicans and Escherichia coli using aluminium phthalocyanine on gold nanoparticles. Photoch Photobio Sci. 2015;14:1346–1356. doi:10.1039/C4PP00315B
  • Sherwani MA, Tufail S, Khan AA, Owais M. Gold nanoparticle-photosensitizer conjugate based photodynamic inactivation of biofilm producing cells: potential for treatment of C. albicans infection in BALB/c mice. PLoS One. 2015;10(7):e0131684. doi:10.1371/journal.pone.013168426148012
  • Teerakapong A, Damrongrungruang T, Sattayut S, Morales NP, Sangpanya A, Tanapoomchai M. Fungicidal effect of combined nano TiO2 with erythrosine for mediated photodynamic therapy on Candida albicans: an in vitro study. Laser Dent Sci. 2017;1(2–4):101–106. doi:10.1007/s41547-017-0014-z
  • Pastar I, Nusbaum AG, Gil J, et al. Interactions of methicillin resistant Staphylococcus aureus USA300 and Pseudomonas aeruginosa in polymicrobial wound infection. PLoS One. 2013;8(2):e56846. doi:10.1371/journal.pone.005684623451098
  • Xu Z, Wang X, Liu X, et al. Tannic acid/Fe3+/Ag nanofilm exhibiting superior photodynamic and physical antibacterial activity. ACS Appl Mater Inter. 2017;9(45):39657–39671. doi:10.1021/acsami.7b10818
  • Li S, Cui S, Yin D, et al. Dual antibacterial activities of a chitosan-modified upconversion photodynamic therapy system against drug-resistant bacteria in deep tissue. Nanoscale. 2017;9(11):3912–3924. doi:10.1039/c6nr07188k28261736
  • Dong K, Ju E, Gao N, Wang Z, Ren J, Qu X. Synergistic eradication of antibiotic-resistant bacteria based biofilms in vivo using a NIR-sensitive nanoplatform. Chem Commun. 2016;52(30):5312–5315. doi:10.1039/C6CC00774K