Acessibilidade / Reportar erro

Drug delivery systems for oral disease applications

Abstract

There are many restrictions on topical medications for the oral cavity. Various factors affect the topical application of drugs in the oral cavity, an open and complex environment. The complex physical and chemical environment of the oral cavity, such as saliva and food, will influence the effect of free drugs. Therefore, drug delivery systems have served as supporting structures or as carriers loading active ingredients, such as antimicrobial agents and growth factors (GFs), to promote antibacterial properties, tissue regeneration, and engineering for drug diffusion. These drug delivery systems are considered in the prevention and treatment of dental caries, periodontal disease, periapical disease, the delivery of anesthetic drugs, etc. These carrier materials are designed in different ways for clinical application, including nanoparticles, hydrogels, nanofibers, films, and scaffolds. This review aimed to summarize the advantages and disadvantages of different carrier materials. We discuss synthesis methods and their application scope to provide new perspectives for the development and preparation of more favorable and effective local oral drug delivery systems.

Keywords:
Delivery systems; Dental caries; Hydrogels; Nanoparticles; Periodontal disease

Introduction

The oral cavity is a complex environment which communicates with the external environment, the upper respiratory tract, and the digestive system.11 Kamrani P, Sadiq NM. Anatomy, head and neck, oral cavity (mouth) [Internet]. In: StatPearls; 2021 [cited January 24, 2022]. Treasure Island (FL): StatPearls Publishing; 2022. Available from: https://www.ncbi.nlm.nih.gov/books/NBK545271/
https://www.ncbi.nlm.nih.gov/books/NBK54...
,22 Kitamoto S, Nagao-Kitamoto H, Hein R, Schmidt TM, Kamada N. The bacterial connection between the oral cavity and the gut diseases. J Dent Res. 2020;99(9):1021-9. doi: 10.1177/0022034520924633.
https://doi.org/10.1177/0022034520924633...
A wide variety of microorganisms is present in the oral cavity, including bacteria and fungi, such as Streptococcus mutans (S. mutans), Lactobacillus spp. Porphyromonas gingivalis (P. gingivalis), and Candida albicans.33 Rathee M, Sapra A. Dental caries [Internet]. In: StatPearls. In: StatPearls; 2021 [cited January 24, 2022. Treasure Island (FL): StatPearls Publishing; 2022. Available from: https://www.ncbi.nlm.nih.gov/books/NBK551699/
https://www.ncbi.nlm.nih.gov/books/NBK55...
55 Dashper SG, Mitchell HL, KA LC, Carpenter L, Gussy MG, Calache H, et al. Temporal development of the oral microbiome and prediction of early childhood caries. Sci Rep. 2019;9(1):19732. doi: 10.1038/s41598-019-56233-0.
https://doi.org/10.1038/s41598-019-56233...
These microorganisms colonize different parts of the oral cavity, including tooth surfaces and the periodontium, and form biofilms which invade oral cavity tissues. Periodontitis is a consequence of alterations in the ecology of resident microbial communities. The intimate interaction of bacteria with the host leads to an inflammatory reaction.66 Abusleme L, Dupuy AK, Dutzan N, Silva N, Burleson JA, Strausbaugh LD, et al. The subgingival microbiome in health and periodontitis and its relationship with community biomass and inflammation. Isme J. 2013;7(5):1016-25. doi: 10.1038/ismej.2012.174
https://doi.org/10.1038/ismej.2012.174...

Dental caries is a consequence of dietary, sugar-driven biofilm accumulation and localized acidification. Due to frequent sugar consumption, the development of a predominantly acidic environment will favor the growth of aciduric bacteria in the biofilm. Aciduric bacteria include Streptococcus spp., Lactobacillus spp., etc. As a result, the dynamic balance between commensals and opportunistic pathogens is disrupted, causing deleterious microbial community shifts and disrupting tooth-enamel mineral homeostasis.77 Philip N, Suneja B, Walsh LJ. Ecological approaches to dental caries prevention: paradigm shift or shibboleth? Caries Res. 2018;52(1-2):153-65. doi:10.1159/000484985
https://doi.org/10.1159/000484985...
1010 Lamont RJ, Koo H, Hajishengallis G. The oral microbiota: dynamic communities and host interactions. Nat Rev Microbiol. 2018;16(12):745-59. doi:10.1038/s41579-018-0089-x
https://doi.org/10.1038/s41579-018-0089-...

According to the global burden of disease (GBD) study, permanent tooth caries is among the ten diseases with the highest incidence for years lived with disability.1111 Vos T, Abajobir AA, Abbafati C, Abbas KM, Abate KH, Abd-Allah F, et al. Global, regional, and national incidence, prevalence, and years lived with disability for 328 diseases and injuries for 195 countries, 1990-2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet. 2017;390(10100):1211-59. doi: 10.1016/S0140-6736(17)32154-2
https://doi.org/10.1016/S0140-6736(17)32...
Periodontal diseases comprise a wide range of inflammatory conditions affecting tooth-supporting structures (the gingiva, bone, and periodontal ligaments), which starts with the localized inflammation of the gingiva, initiated by a microbial biofilm that forms on the teeth and gingiva.1212 Kinane DF, Stathopoulou PG, Papapanou PN. Periodontal diseases. Nat Rev Dis Primers. 2017;3:17038. doi: 10.1038/nrdp.2017.38
https://doi.org/10.1038/nrdp.2017.38...
1414 Kornman KS, Page RC, Tonetti MS. The host response to the microbial challenge in periodontitis: assembling the players. Periodontol 2000. 1997;14:33-53. doi: 10.1111/j.1600-0757.1997.tb00191.x
https://doi.org/10.1111/j.1600-0757.1997...
Periodontal diseases lead to teeth loss and contribute to systemic inflammation, which is highly prevalent worldwide.1515 Hajishengallis G. Periodontitis: from microbial immune subversion to systemic inflammation. Nat Rev Immunol. 2015;15(1):30-44. doi: 10.1038/nri3785
https://doi.org/10.1038/nri3785...
Thus, periodontal diseases represent a significant public health problem.1616 Petersen PE, Ogawa H. The global burden of periodontal disease: towards integration with chronic disease prevention and control. Periodontol 2000. 2012;60(1):15-39. doi: 10.1111/j.1600-0757.2011.00425.x
https://doi.org/10.1111/j.1600-0757.2011...
Regarding the topical application of drugs to the oral cavity, the influence of the oral environment on the drugs should be considered. Its complex physical and chemical environment and the complexity of bacterial biofilm affect drug application via the oral cavity.1717 Paula AJ, Koo H. Nanosized building blocks for customizing novel antibiofilm approaches. J Dent Res. 2017;96(2):128-36. doi: 10.1177/0022034516679397
https://doi.org/10.1177/0022034516679397...
For example, salivary flow rates in the mouth might affect the efficacy of topical anesthesia to some extent, whereas side effects and drug resistance are inevitable in systemic administration.1818 Makvandi P, Josic U, Delfi M, Pinelli F, Jahed V, Kaya E, et al. Drug delivery (nano)platforms for oral and dental applications: tissue regeneration, infection control, and cancer management. Adv Sci (Weinh). 2021;8(8):2004014. doi: 10.1002/advs.202004014
https://doi.org/10.1002/advs.202004014...
Salivary clearance might dilute and weaken the active ingredients in the oral environment. Therefore, carrier systems had to be designed to consistently release active ingredients so their estimated concentrations can be effective. Recently, emerging advanced biomaterials, including hydrogels, films, nanofibers, and particles hold great potential as cell/drug carriers for local drug delivery and biomimetic scaffolds.1919 Chen X, Wu G, Feng Z, Dong Y, Zhou W, Li B, et al. Advanced biomaterials and their potential applications in the treatment of periodontal disease. Crit Rev Biotechnol. 2016;36(4):760-75. doi: 10.3109/07388551.2015.1035693
https://doi.org/10.3109/07388551.2015.10...
Biofilms decrease the effects of drugs on microorganisms, and antibacterial substances are easily metabolized. Moreover, there is no perfect way of delivering drugs to sites such as the periodontal pocket and the periapical area. Drug delivery systems have many advantages, such as increasing drug solubility, prolonging drug action time, improving drug targeting, and reducing cytotoxicity. Moreover, delivery systems play a role in inhibiting bacteria by releasing active ingredients, such as chlorine, and can serve as engineering scaffolds to promote tissue regeneration. For example, patients receiving ibuprofen-incorporated, chitosan-based microspheres show better anti-inflammatory properties than the orally administered ibuprofen group.2020 Kp K, R B. Evaluation and comparison of anti-inflammatory properties of ibuprofen using two drug delivery systems after third molar surgery: using chitosan microspheres as a carrier for local drug delivery in to the third molar socket and through the oral route. Br J Oral Maxillofac Surg. 2021;59(2):191-6. doi: 10.1016/j.bjoms.2020.08.025
https://doi.org/10.1016/j.bjoms.2020.08....
Moreover, bone morphogenetic protein 2 (BMP-2) loaded into calcium silicate scaffolds showed continuous release and better stimulation for the induction of mesenchymal stem cells.2121 Huang KH, Chen YW, Wang CY, Lin YH, Wu YA, Shie MY, et al. Enhanced capability of bone morphogenetic protein 2-loaded mesoporous calcium silicate scaffolds to induce odontogenic differentiation of human dental pulp cells. J Endod. 2018;44(11):1677-85. doi: 10.1016/j.joen.2018.08.008
https://doi.org/10.1016/j.joen.2018.08.0...
In the field of anesthesia, experiments have also proved that appropriate hybrid nanofilms containing the eutectic mixture of 5% lidocaine-prilocaine (LDC-PLC) show a better anesthetic effect, with higher permeability and no cytotoxicity.2222 Ribeiro LN, Franz-Montan M, Alcântara AC, Breitkreitz MC, Castro SR, Guilherme VA, et al. Hybrid nanofilms as topical anesthetics for pain-free procedures in dentistry. Sci Rep. 2020;10(1):11341. doi: 10.1038/s41598-020-68247-0
https://doi.org/10.1038/s41598-020-68247...

In the reviewing process, “oral/oral cavity/mouth/mouth cavity” and “drug delivery systems/carrier materials/drug targeting” were used as research items in the Pubmed, MEDLINE, and Web of Science databases. Relevant articles with an impact factor greater than five and published after 2015 were included. Moreover, we added some articles about the progress of experiments based on previous classic reviews. All drug delivery systems were classified as nanoparticles, hydrogels, nanofibers, and films according to their material properties. The carrier materials used as oral drug delivery systems in recent years are summarized in Table 1, and their different forms are described in Figure 1. This review discusses drug delivery systems in the oral cavity, which are prepared by various methods to adapt to complex situations. For example, they can be used as support structures to promote regeneration in defective tissues or as drug carriers to release active ingredients which control infection. Moreover, to support the future development of carrier materials, we summarized their composition and application.

Table 1
Carrier materials used as oral drug delivery systems in recent years
Figure 1
Models of different material forms. a: Nanoparticles include inorganic and organic carriers. As shown in the figure, these are mesoporous nanoparticles. b: Hydrogels are usually made from 3D polymers and can be loaded with drugs or nanoparticles. c: Nanofibers are fibers of approximately 100 nm in diameter which can carry drugs. d: Films are often used for oral mucosal drug delivery. e: Scaffolds are loaded with growth factors and usually used in tissue regeneration

Nanoparticles

Nanoparticles have widely served as drug delivery systems in the oral cavity.2323 Benoit DS, Sims KR Jr, Fraser D. Nanoparticles for oral biofilm treatments. ACS Nano. 2019;13(5):4869-75. doi: 10.1021/acsnano.9b02816
https://doi.org/10.1021/acsnano.9b02816...
,2424 Raura N, Garg A, Arora A, Roma M. Nanoparticle technology and its implications in endodontics: a review. Biomater Res. 2020;24(1):21. doi: 10.1186/s40824-020-00198-z
https://doi.org/10.1186/s40824-020-00198...
Nanoparticles include inorganic carriers, such as mesoporous silica and mesoporous calcium silicate and organic carriers, such as poly (d,l-lactide-co-glycolide acid) (PLGA) and chitosan. Nanoparticle size lies mostly between 1-100 nm, which results in good diffusion properties and loading performances.1717 Paula AJ, Koo H. Nanosized building blocks for customizing novel antibiofilm approaches. J Dent Res. 2017;96(2):128-36. doi: 10.1177/0022034516679397
https://doi.org/10.1177/0022034516679397...
,2525 European Commission. Commission Recommendation of 18 October 2011 on the definition of nanomaterial Text with EEA relevance [Internet]. In: Official Journal of the European Union; 2011 [cited 2021 March 21]. Available from: https://eur-lex.europa.eu/eli/reco/2011/696.
https://eur-lex.europa.eu/eli/reco/2011/...
Inorganic nanoparticles are mainly composed of inorganic materials which are crystallized and amorphous solids at ambient temperature. Their reactivity is different in various solution environments, which significantly influence their toxicity. Organic nanoparticles mainly consist of organic substances such as lipids, proteins, etc. Depending on their composition, organic nanoparticles can be degraded and digested in different parts in vivo. Organic nanoparticles are generally less toxic than inorganic nanoparticles, but they might cause toxicity under particular circumstances.2626 McClements DJ, Xiao H. Is nano safe in foods? Establishing the factors impacting the gastrointestinal fate and toxicity of organic and inorganic food-grade nanoparticles. NPJ Sci Food. 2017;1:6. doi: 10.1038/s41538-017-0005-1
https://doi.org/10.1038/s41538-017-0005-...
In recent years, inorganic nanoparticles have received attention due to their unique materials, which are more inert, stable, and easy to functionalize.2727 Lohse SE, Murphy CJ. Applications of colloidal inorganic nanoparticles: from medicine to energy. J Am Chem Soc. 2012;134(38):15607-20. doi: 10.1021/ja307589n
https://doi.org/10.1021/ja307589n...

Inorganic nanoparticles

In general, mesoporous materials are used as inorganic nanoparticle carrier systems. Mesoporous materials include mesoporous silica nanoparticles (MSNs) and mesoporous calcium silicate nanoparticles (MCSNs).2828 Vallet-Regí M, Colilla M, Izquierdo-Barba I, Manzano M. Mesoporous silica nanoparticles for drug delivery: current insights. Molecules. 2017;23(1). doi: 10.3390/molecules23010047
https://doi.org/10.3390/molecules2301004...
3030 Leng D, Li Y, Zhu J, Liang R, Zhang C, Zhou Y, et al. The antibiofilm activity and mechanism of nanosilver- and nanozinc-incorporated mesoporous calcium-silicate nanoparticles. Int J Nanomedicine. 2020;15:3921-36. doi: 10.2147/ijn.S244686
https://doi.org/10.2147/ijn.S244686...

MSNs have been modified by amination to make them more suitable for use in the oral environment. In recent studies, the amine-functionalized expanded pore mesoporous silica (aMSN) was synthesized to load polyacrylic acid–stabilized amorphous calcium phosphate (PAA-ACP). PAA-ACP loaded aMSN has the potential to treat white enamel spot lesions and serve as filler in resin adhesives.3131 Hua F, Yan J, Zhao S, Yang H, He H. In vitro remineralization of enamel white spot lesions with a carrier-based amorphous calcium phosphate delivery system. Clin Oral Investig. 2020;24(6):2079-89. doi: 10.1007/s00784-019-03073-x
https://doi.org/10.1007/s00784-019-03073...
,3232 Luo XJ, Yang HY, Niu LN, Mao J, Huang C, Pashley DH, et al. Translation of a solution-based biomineralization concept into a carrier-based delivery system via the use of expanded-pore mesoporous silica. Acta Biomater. 2016;31:378-87. doi: 10.1016/j.actbio.2015.11.062
https://doi.org/10.1016/j.actbio.2015.11...
Regarding their antimicrobial properties, Lu, et al.3333 Lu MM, Ge Y, Qiu J, Shao D, Zhang Y, Bai J, et al. Redox/pH dual-controlled release of chlorhexidine and silver ions from biodegradable mesoporous silica nanoparticles against oral biofilms. Int J Nanomedicine. 2018;13:7697-709. doi: 10.2147/ijn.S181168
https://doi.org/10.2147/ijn.S181168...
(2018). incorporated silver ions into MSNs, synthesizing silver-decorated mesoporous silica nanoparticles (Ag-MSNs) and using them to load chlorhexidine (CHX). They showed redox/pH-responsive release properties due to CHX and silver ions which could inhibit S. mutans biofilm growth. Ag-MSNs@CHX were more effective than an equivalent amount of free CHX in limiting S. mutans biofilm formation since they induced bacterial cell death, particularly in the long term. Though S. mutans was only used as a model to evaluate the effect of Ag-MSNs@CHX, the material may be considered for treating diseases caused by bacterial biofilms, such as periodontal diseases.

A practical antibacterial component, silver ions were also loaded into mesoporous calcium silicate to prepare nano-silver-incorporated mesoporous calcium-silicate nanoparticles (Ag-MCSNs), resulting in lower cytotoxicity. The nanoparticles slowly released silver ions which could inhibit Enterococcus faecalis (E. faecalis) growth and colonization.3434 Fan W, Wu D, Tay FR, Ma T, Wu Y, Fan B. Effects of adsorbed and templated nanosilver in mesoporous calcium-silicate nanoparticles on inhibition of bacteria colonization of dentin. Int J Nanomedicine. 2014;9:5217-30. doi: 10.2147/ijn.S73144
https://doi.org/10.2147/ijn.S73144...
CHX can be successfully loaded onto MCSNs by the mixing-coupling method. These nanoparticles released CHX, showing low cytotoxicity and excellent anti-E. faecalis properties. Moreover, they can also release Ca2+/SiO32− ions, showing an in vitro mineralization property. These nanoparticles could be developed into bone-defect-filling material or intracanal medications in dentistry.3535 Fan W, Li Y, Sun Q, Ma T, Fan B. Calcium-silicate mesoporous nanoparticles loaded with chlorhexidine for both anti- Enterococcus faecalis and mineralization properties. J Nanobiotechnology. 2016;14(1):72. doi: 10.1186/s12951-016-0224-7
https://doi.org/10.1186/s12951-016-0224-...

Huang, et al.3636 Huang CY, Huang TH, Kao CT, Wu YH, Chen WC, Shie MY. Mesoporous calcium silicate nanoparticles with drug delivery and odontogenesis properties. J Endod. 2017;43(1):69-76. doi: 10.1016/j.joen.2016.09.012
https://doi.org/10.1016/j.joen.2016.09.0...
(2017) loaded gentamicin and fibroblast growth factor 2 (FGF-2) with mesoporous calcium silicate nanoparticles, which could sustain the release of FGF-2 and gentamicin. These nanoparticles showed bone/cementum tissue regeneration and inhibited bacterial viability, suggesting their use in endodontics as a valuable, biocompatible dental pulp tissue regenerative material capable of odontogenesis. MCSNs can also be loaded with BMP-2 and prepared into a three-dimensional scaffold by 3D printing, inducing the differentiation of dental pulp cells and odontoblasts. The BMP-2-loaded mesoporous calcium silicate 3D scaffold could serve as an intracanal scaffold for both hemostatic clot retention and odontogenesis in reparative endodontic therapy.2121 Huang KH, Chen YW, Wang CY, Lin YH, Wu YA, Shie MY, et al. Enhanced capability of bone morphogenetic protein 2-loaded mesoporous calcium silicate scaffolds to induce odontogenic differentiation of human dental pulp cells. J Endod. 2018;44(11):1677-85. doi: 10.1016/j.joen.2018.08.008
https://doi.org/10.1016/j.joen.2018.08.0...
Nano-metal particles, such as Ag, Cu, copper oxide (CuO), and titanium oxide (TiO2), can serve as antibacterial agents in the oral cavity.3737 Allaker RP. The use of nanoparticles to control oral biofilm formation. J Dent Res. 2010;89(11):1175-86. doi: 10.1177/0022034510377794
https://doi.org/10.1177/0022034510377794...
,3838 Allaker RP, Memarzadeh K. Nanoparticles and the control of oral infections. Int J Antimicrob Agents. 2014;43(2):95-104. doi: 10.1016/j.ijantimicag.2013.11.002
https://doi.org/10.1016/j.ijantimicag.20...
In recent years, the toxic effects of metals and metal oxides have attracted significant attention.3939 Dizaj SM, Lotfipour F, Barzegar-Jalali M, Zarrintan MH, Adibkia K. Antimicrobial activity of the metals and metal oxide nanoparticles. Mater Sci Eng C Mater Biol Appl. 2014;44:278-84. doi: 10.1016/j.msec.2014.08.031
https://doi.org/10.1016/j.msec.2014.08.0...
,4040 Mathur P, Jha S, Ramteke S, Jain NK. Pharmaceutical aspects of silver nanoparticles. Artif Cells Nanomed Biotechnol. 2018;46(sup1):115-26. doi: 10.1080/21691401.2017.1414825
https://doi.org/10.1080/21691401.2017.14...
Thus, the safety of metal nanoparticles should be further investigated.4141 Tang H, Xu M, Zhou X, Zhang Y, Zhao L, Ye G, et al. Acute toxicity and biodistribution of different sized copper nano-particles in rats after oral administration. Mater Sci Eng C Mater Biol Appl. 2018;93:649-63. doi: 10.1016/j.msec.2018.08.032
https://doi.org/10.1016/j.msec.2018.08.0...

Although many articles have highlighted the role of antimicrobials, there are still many disadvantages in using antibacterial drugs, including drug resistance, dysbiosis, etc. Therefore, active agents are needed to inhibit bacterial biofilms without affecting the ecological balance of flora, such as probiotics and prebiotics, thus reestablishing ecological balance or the biodiversity of oral microbiota.

Organic nanoparticles

In recent years, nanoparticle carriers have commonly been prepared by the self-assembly of organic molecules, such as PLGA, chitosan, and poly (lactic acid).4242 Ding D, Zhu Q. Recent advances of PLGA micro/nanoparticles for the delivery of biomacromolecular therapeutics. Mater Sci Eng C Mater Biol Appl. 2018;92:1041-60. doi: 10.1016/j.msec.2017.12.036
https://doi.org/10.1016/j.msec.2017.12.0...
4545 Rizeq BR, Younes NN, Rasool K, Nasrallah GK. Synthesis, bioapplications, and toxicity evaluation of chitosan-based nanoparticles. Int J Mol Sci. 2019;20(22). doi: 10.3390/ijms20225776
https://doi.org/10.3390/ijms20225776...

PLGA is a conventionally used component in preparing organic nanocarriers. In previous studies,4040 Mathur P, Jha S, Ramteke S, Jain NK. Pharmaceutical aspects of silver nanoparticles. Artif Cells Nanomed Biotechnol. 2018;46(sup1):115-26. doi: 10.1080/21691401.2017.1414825
https://doi.org/10.1080/21691401.2017.14...
,4343 Kumar G, Shafiq N, Malhotra S. Drug-loaded PLGA nanoparticles for oral administration: fundamental issues and challenges ahead. Crit Rev Ther Drug Carrier Syst. 2012;29(2):149-82. doi: 10.1615/critrevtherdrugcarriersyst.v29.i2.20
https://doi.org/10.1615/critrevtherdrugc...
PLGA-nanoparticles were prepared to encapsulate CHX. These nanoparticles could be delivered to the demineralized dentin matrix and the resin-dentin interface by dentinal tubules, in which they might play a role not only in inhibiting bacteria but also matrix metalloproteinase (MMP).4646 Priyadarshini BM, Mitali K, Lu TB, Handral HK, Dubey N, Fawzy AS. PLGA nanoparticles as chlorhexidine-delivery carrier to resin-dentin adhesive interface. Dent Mater. 2017;33(7):830-46. doi: 10.1016/j.dental.2017.04.015
https://doi.org/10.1016/j.dental.2017.04...
PLGA is used to load grapeseed extract (GSE), which could improve the biodegradation resistance of demineralized dentin. Biodegradable polymer nanoparticles can deliver GSE via demineralized dentinal tubules, reducing the biodegradability of the dentin collagen matrix.4747 Fawzy AS, Priyadarshini BM, Selvan ST, Lu TB, Neo J. Proanthocyanidins-loaded nanoparticles enhance dentin degradation resistance. J Dent Res. 2017;96(7):780-9. doi: 10.1177/0022034517691757
https://doi.org/10.1177/0022034517691757...
PLGA is also used to load lovastatin and control its release, which can induce dentin differentiation at appropriate concentrations. Thus, these nanoparticles could serve as an adjunctive treatment in indirect pulp capping procedures.4848 Lin HP, Tu HP, Hsieh YP, Lee BS. Controlled release of lovastatin from poly(lactic-co-glycolic acid) nanoparticles for direct pulp capping in rat teeth. Int J Nanomedicine. 2017;12:5473-85. doi: 10.2147/ijn.S138410
https://doi.org/10.2147/ijn.S138410...
PLGA is used to deliver metronidazole or N-phenacylthiazolium bromide to modulate periodontitis progression.4949 Lin JH, Feng F, Yu MC, Wang CH, Chang PC. Modulation of periodontitis progression using pH-responsive nanosphere encapsulating metronidazole or N-phenacylthialzolium bromide. J Periodontal Res. 2018;53(1):22-8. doi: 10.1111/jre.12481
https://doi.org/10.1111/jre.12481...
PLGA-chitosan nanoparticles are also loaded with simvastatin and doxycycline to promote the repair of infected periodontal sites and non-infected osseous defects.5050 Chang PC, Tai WC, Luo HT, Lai CH, Lin HH, Lin ZJ, et al. Core-Shell poly-(D,l-Lactide-co-Glycolide)-chitosan Nanospheres with simvastatin-doxycycline for periodontal and osseous repair. Int J Biol Macromol. 2020;158:627-35. doi: 10.1016/j.ijbiomac.2020.04.183
https://doi.org/10.1016/j.ijbiomac.2020....
Compared with gel carriers, a PLGA particle carrier prolonged the release of active components, indicating its functional suitability for clinical applications.5151 Spicer PP, Shah SR, Henslee AM, Watson BM, Kinard LA, Kretlow JD, et al. Evaluation of antibiotic releasing porous polymethylmethacrylate space maintainers in an infected composite tissue defect model. Acta Biomater. 2013;9(11):8832-9. doi: 10.1016/j.actbio.2013.07.018
https://doi.org/10.1016/j.actbio.2013.07...

Liposomes are often used as drug carriers to encapsulate active ingredients.5252 Li M, Du C, Guo N, Teng Y, Meng X, Sun H, et al. Composition design and medical application of liposomes. Eur J Med Chem. 2019;164:640-53. doi: 10.1016/j.ejmech.2019.01.007
https://doi.org/10.1016/j.ejmech.2019.01...
They were used to develop curcumin-loaded solid lipid nanoparticles (CurSLN) to treat oral mucosal infections, showing increased antibacterial activity.5353 Hazzah HA, Farid RM, Nasra MM, Hazzah WA, El-Massik MA, Abdallah OY. Gelucire-based nanoparticles for curcumin targeting to oral mucosa: preparation, characterization, and antimicrobial activity assessment. J Pharm Sci. 2015;104(11):3913-24. doi: 10.1002/jps.24590
https://doi.org/10.1002/jps.24590...
Furthermore, liposomes have served as carriers to prepare pH-responsive quaternary ammonium chitosan-liposome nanoparticles by first loading doxycycline with liposomes, then coating the latter with N, N, N-Trimethyl chitosan (TMC). When pH is low, the free amino group in the quaternary ammonium chitosan on the surface of nanoparticles will be protonated. The positive charge on the TMC surface could be increased to destabilize the nanoparticles, thereby triggering DOX release. These nanoparticles could be closely linked to biofilms and effectively destroy their membrane structure and disrupt bacterial biofilms in vivo.5454 Hu F, Zhou Z, Xu Q, Fan C, Wang L, Ren H, et al. A novel pH-responsive quaternary ammonium chitosan-liposome nanoparticles for periodontal treatment. Int J Biol Macromol. 2019;129:1113-9. doi: 10.1016/j.ijbiomac.2018.09.057
https://doi.org/10.1016/j.ijbiomac.2018....

Chitosan is usually used as a carrier to load active ingredients. Chitosan nanoparticles loaded with toothpaste actives such as sodium fluoride (NaF) and cetylpyridinium chloride (CPC) are prepared by emulsion dispersion or ionic gelation. These nanoparticles continuously released their active ingredients, which makes them highly promising as dental delivery systems in the protection against caries.5555 Liu H, Chen B, Mao Z, Gao C. Chitosan nanoparticles for loading of toothpaste actives and adhesion on tooth analogs. 2007;106(6):4248-56. doi: https://doi.org/10.1002/app.27078
https://doi.org/10.1002/app.27078...
,5656 Nguyen S, Escudero C, Sediqi N, Smistad G, Hiorth M. Fluoride loaded polymeric nanoparticles for dental delivery. Eur J Pharm Sci. 2017;104:326-34. doi: 10.1016/j.ejps.2017.04.004
https://doi.org/10.1016/j.ejps.2017.04.0...
After functionalization with glutathione, chitosan showed a stronger ability to bind to chlorhexidine, resulting in a better release of chlorhexidine within 48 h.5757 Chronopoulou L, Nocca G, Castagnola M, Paludetti G, Ortaggi G, Sciubba F, et al. Chitosan based nanoparticles functionalized with peptidomimetic derivatives for oral drug delivery. N Biotechnol. 2016;33(1):23-31. doi: 10.1016/j.nbt.2015.07.005
https://doi.org/10.1016/j.nbt.2015.07.00...
Moreover, polycaprolactone has been used to encase chlorhexidine to form Poly (ε-caprolactone) (PCL)-coated CHX nanocapsules. In this process, the nanocapsules could reach the demineralized dentin region via micron-scale dentin tubules, slowly releasing chlorhexidine, thereby playing an antibacterial role.5858 Priyadarshini BM, Selvan ST, Lu TB, Xie H, Neo J, Fawzy AS. Chlorhexidine nanocapsule drug delivery approach to the resin-dentin interface. J Dent Res. 2016;95(9):1065-72. doi: 10.1177/0022034516656135
https://doi.org/10.1177/0022034516656135...

Furthermore, chitosan has been used as a carrier to load dexamethasone (DEX), and alginate solution, containing transforming growth factor-β1(TGF-β1), to wrap the resulting nanoparticles. In turn, the core-shell nanosystem (TD-NS) could release TGF-β1 and DEX so materials might treat periapical inflammation.5959 Shrestha S, Kishen A. Temporal-controlled bioactive molecules releasing core-shell nano-system for tissue engineering strategies in endodontics. Nanomedicine. 2019;18:11-20. doi: 10.1016/j.nano.2019.02.013
https://doi.org/10.1016/j.nano.2019.02.0...
Zhu, et al.6060 Zhu B, Li X, Xu X, Li J, Ding C, Zhao C, et al. One-step phosphorylated poly(amide-amine) dendrimer loaded with apigenin for simultaneous remineralization and antibacterial of dentine. Colloids Surf B Biointerfaces. 2018;172:760-8. doi: 10.1016/j.colsurfb.2018.09.036
https://doi.org/10.1016/j.colsurfb.2018....
(2018) used the phosphorylated poly (amide-amine) dendrimer (PAMMA) as a carrier to load apigenin, a type of antibiotic insoluble in water. The material can both play an antibacterial role and promote remineralization. Polylactic acid (PLA) and PLGA have been used to load cells and BMP-2, respectively, promoting odontogenic differentiation and the formation of dentoid tissue via human stem cells of apical papilla (SCAP). The data showed great promise in promoting dentin tissue regeneration.6161 Wang W, Dang M, Zhang Z, Hu J, Eyster TW, Ni L, et al. Dentin regeneration by stem cells of apical papilla on injectable nanofibrous microspheres and stimulated by controlled BMP-2 release. Acta Biomater. 2016;36:63-72. doi: 10.1016/j.actbio.2016.03.015
https://doi.org/10.1016/j.actbio.2016.03...

Moreover, continuous release systems for CHX were developed based on chitosan (CS) and montmorillonite (MMT). The CHX-hybrid nanosystem containing positively-charged chitosan showed good mucoadhesion properties which enabled the drug delivery system to stay longer at the absorption site and, therefore, contributed to a beneficial effect on drug bioavailability.6262 Onnainty R, Onida B, Páez P, Longhi M, Barresi A, Granero G. Targeted chitosan-based bionanocomposites for controlled oral mucosal delivery of chlorhexidine. Int J Pharm. 2016;509(1-2):408-18. doi: 10.1016/j.ijpharm.2016.06.011
https://doi.org/10.1016/j.ijpharm.2016.0...
A pH-responsive and redox-sensitive polymer-based AmB-delivery carrier system was developed based on O-Carboxymethyl chitosan (OCMCH). This system was functionalized by conjugation with the antifungal peptide histatin 5, which targeted ligands and synergistic antifungal molecules against Candida albicans.6363 Park SC, Kim YM, Lee JK, Kim NH, Kim EJ, Heo H, et al. Targeting and synergistic action of an antifungal peptide in an antibiotic drug-delivery system. J Control Release. 2017;256:46-55. doi: 10.1016/j.jconrel.2017.04.023
https://doi.org/10.1016/j.jconrel.2017.0...
The cellulose acetate phthalate nanoparticle is used to load chlorhexidine, enabling much greater infiltration into the subgingival tissue.6464 Vidal-Romero G, Zambrano-Zaragoza ML, Martínez-Acevedo L, Leyva-Gómez G, Mendoza-Elvira SE, Quintanar-Guerrero D. Design and evaluation of ph-dependent nanosystems based on cellulose acetate phthalate, nanoparticles loaded with chlorhexidine for periodontal treatment. Pharmaceutics. 2019;11(11). doi: 10.3390/pharmaceutics11110604
https://doi.org/10.3390/pharmaceutics111...
Diblock copolymers underwent ultrasound treatment to form nanoparticle carriers by self-assembly. The recent nanoparticle carrier (NPC) DDS is flexible enough to co-load farnesol and myricetin. Therefore, nanoparticles could reduce S. mutansbiofilm organization and acid production.6565 Sims KR Jr, Maceren JP, Liu Y, Rocha GR, Koo H, Benoit DS. Dual antibacterial drug-loaded nanoparticles synergistically improve treatment of Streptococcus mutans biofilms. Acta Biomater. 2020;115:418-31. doi: 10.1016/j.actbio.2020.08.032
https://doi.org/10.1016/j.actbio.2020.08...
Some inorganic nanoparticles are also used in oral mucosal diseases, such as hydroxyapatite nanoparticles and hyperbranched core-multishell nanocarriers.6666 Murata T, Kutsuna T, Kurohara K, Shimizu K, Tomeoku A, Arai N. Evaluation of a new hydroxyapatite nanoparticle as a drug delivery system to oral squamous cell carcinoma cells. Anticancer Res. 2018;38(12):6715-20. doi: 10.21873/anticanres.13040
https://doi.org/10.21873/anticanres.1304...
,6767 Jager J, Obst K, Lohan SB, Viktorov J, Staufenbiel S, Renz H, et al. Characterization of hyperbranched core-multishell nanocarriers as an innovative drug delivery system for the application at the oral mucosa. J Periodontal Res. 2018;53(1):57-65. doi: 10.1111/jre.12487
https://doi.org/10.1111/jre.12487...

Hydrogels

Hydrogels composed a natural or synthetic 3D network by physical or chemical strategies and could absorb a large amount of water (up to 1000-fold compared to the dry weight). Hydrogels consist of long polymer chains filled with water molecules between them.6868 Chyzy A, Plonska-Brzezinska ME. Hydrogel properties and their impact on regenerative medicine and tissue engineering. molecules. 2020;25(24). doi: 10.3390/molecules25245795.
https://doi.org/10.3390/molecules2524579...
,6969 Fennell E, Huyghe JM. Chemically responsive hydrogel deformation mechanics: a review. Molecules. 2019;24(19). doi: 10.3390/molecules24193521
https://doi.org/10.3390/molecules2419352...
,7070 Sharma S, Tiwari S. A review on biomacromolecular hydrogel classification and its applications. Int J Biol Macromol. 2020;162:737-47. doi: 10.1016/j.ijbiomac.2020.06.110
https://doi.org/10.1016/j.ijbiomac.2020....
Natural hydrogels are biocompatible and can degrade into nontoxic by-products which can interact with biological macromolecules. However, their limitations lie in their weak mechanical strength and immunogenicity.7171 Kirchhof S, Goepferich AM, Brandl FP. Hydrogels in ophthalmic applications. Eur J Pharm Biopharm. 2015;95(Pt B):227-38. doi: 10.1016/j.ejpb.2015.05.016
https://doi.org/10.1016/j.ejpb.2015.05.0...
By contrast, synthesized hydrogels can be better regulated and have better stability, but they might degrade into toxic by-products.7272 Cooper RC, Yang H. Hydrogel-based ocular drug delivery systems: emerging fabrication strategies, applications, and bench-to-bedside manufacturing considerations. J Control Release. 2019;306:29-39. doi: 10.1016/j.jconrel.2019.05.034
https://doi.org/10.1016/j.jconrel.2019.0...
Hydrogels can control drug release due to changes (swelling, dissolution or degradation) in the gel structure in response to internal or external stimuli.7373 Narayanaswamy R, Torchilin VP. Hydrogels and their applications in targeted drug delivery. Molecules. 2019;24(3). doi: 10.3390/molecules24030603
https://doi.org/10.3390/molecules2403060...
With appropriate release mechanisms, active drugs can be maintained locally at high concentrations for a longer period. Currently, the clinical applications of hydrogels for drug delivery include ophthalmology, cardiovascular diseases, and cancer, having attracted the attention of oral medicine in recent years.7474 Mou J, Liu Z, Liu J, Lu J, Zhu W, Pei D. Hydrogel containing minocycline and zinc oxide-loaded serum albumin nanopartical for periodontitis application: preparation, characterization and evaluation. Drug Deliv. 2019;26(1):179-87. doi: 10.1080/10717544.2019.1571121
https://doi.org/10.1080/10717544.2019.15...
,7575 Hosny KM, Aldawsari HM, Bahmdan RH, Sindi AM, Kurakula M, Alrobaian MM, et al. Preparation, optimization, and evaluation of hyaluronic acid-based hydrogel loaded with miconazole self-nanoemulsion for the treatment of oral thrush. AAPS PharmSciTech. 2019;20(7):297. doi: 10.1208/s12249-019-1496-7
https://doi.org/10.1208/s12249-019-1496-...
,7676 Franz-Montan M, Ribeiro LN, Volpato MC, Cereda CM, Groppo FC, Tofoli GR, et al. Recent advances and perspectives in topical oral anesthesia. Expert Opin Drug Deliv. 2017;14(5):673-84. doi: 10.1080/17425247.2016.1227784
https://doi.org/10.1080/17425247.2016.12...
,7777 Pagano C, Giovagnoli S, Perioli L, Tiralti MC, Ricci M. Development and characterization of mucoadhesive-thermoresponsive gels for the treatment of oral mucosa diseases. Eur J Pharm Sci. 2020;142:105125. doi: 10.1016/j.ejps.2019.105125
https://doi.org/10.1016/j.ejps.2019.1051...
Compared to nanopore carriers, hydrogel carriers can maintain or trigger drug delivery and enable multi-drug delivery. At the same time, hydrogels avoid intravenous injection and have better local application prospects, making them interesting platforms for drug delivery in the oral cavity.7878 Zhang M, Liang J, Yang Y, Liang H, Jia H, Li D. Current trends of targeted drug delivery for oral cancer therapy. Front Bioeng Biotechnol. 2020;8:618931. doi: 10.3389/fbioe.2020.618931
https://doi.org/10.3389/fbioe.2020.61893...

Chitosan hydrogels

In recent years, chitosan/β-glycerophosphate (CS/β-GP) thermosensitive hydrogels have served as drug delivery systems to load bone morphogenetic protein-7 (BMP-7) and ornidazole (ORN), continuously releasing them to treat periodontal diseases.7979 Zang S, Mu R, Chen F, Wei X, Zhu L, Han B, et al. Injectable chitosan/β-glycerophosphate hydrogels with sustained release of BMP-7 and ornidazole in periodontal wound healing of class III furcation defects. Mater Sci Eng C Mater Biol Appl. 2019;99:919-28. doi: 10.1016/j.msec.2019.02.024
https://doi.org/10.1016/j.msec.2019.02.0...
Moreover, chitosan/β-glycerophosphate (CS/β-GP) thermosensitive hydrogels were loaded with aspirin to control inflammation and erythropoietin. Hydrogels can promote periodontal regeneration.8080 Xu X, Gu Z, Chen X, Shi C, Liu C, Liu M, et al. An injectable and thermosensitive hydrogel: Promoting periodontal regeneration by controlled-release of aspirin and erythropoietin. Acta Biomater. 2019;86:235-46. doi: 10.1016/j.actbio.2019.01.001
https://doi.org/10.1016/j.actbio.2019.01...
They are also used to load naringin, a naturally derived polymethoxylated flavonoid compound with anti-inflammatory properties that inhibit the inflammatory state of periodontitis.8181 Chang PC, Chao YC, Hsiao MH, Chou HS, Jheng YH, Yu XH, et al. Inhibition of periodontitis induction using a stimuli-responsive hydrogel carrying naringin. J Periodontol. 2017;88(2):190-6. doi: 10.1902/jop.2016.160189
https://doi.org/10.1902/jop.2016.160189...
Regarding other types of thermosensitive hydrogels, a thermosensitive micellar hydrogel was prepared from amphiphilic copolymer poly (ε-caprolactone-co-1,4,8-trioxa [4.6]spiro-9-undecanone)-poly(ethylene glycol)-poly (ε-caprolactone-co-1,4,8-trioxa [4.6]spiro-9-undecanone) (PECT) nanoparticles. Ibuprofen and basic fibroblast growth factor (bFGF) were encapsulated in these thermosensitive micellar hydrogels. Thus, these materials can promote anti-inflammatory activity and soft tissue healing.8282 Chen W, Zhi M, Feng Z, Gao P, Yuan Y, Zhang C, et al. Sustained co-delivery of ibuprofen and basic fibroblast growth factor by thermosensitive nanoparticle hydrogel as early local treatment of peri-implantitis. Int J Nanomedicine. 2019;14:1347-58. doi: 10.2147/ijn.S190781
https://doi.org/10.2147/ijn.S190781...

Other hydrogels

There are other components of hydrogels. To promote tissue regeneration, biocompatible hydrogelator Nap-Phe-Phe-Tyr-OH (NapFFY) hydrogels were loaded with stromal cell-derived factor-1 (SDF-1) and bone morphogenetic proteins (BMPs) to promote and accelerate periodontal bone regeneration.8383 Tan J, Zhang M, Hai Z, Wu C, Lin J, Kuang W, et al. Sustained release of two bioactive factors from supramolecular hydrogel promotes periodontal bone regeneration. ACS Nano. 2019;13(5):5616-22. doi: 10.1021/acsnano.9b00788
https://doi.org/10.1021/acsnano.9b00788...
Pan, et al.8484 Pan J, Deng J, Luo Y, Yu L, Zhang W, Han X, et al. Thermosensitive hydrogel delivery of human periodontal stem cells overexpressing platelet-derived growth factor-bb enhances alveolar bone defect repair. Stem Cells Dev. 2019;28(24):1620-31. doi: 10.1089/scd.2019.0184
https://doi.org/10.1089/scd.2019.0184...
(2019) used PLGA–PEG–PLGA triblock copolymers as a carrier to load human periodontal ligament stem cells, which had been modified to overexpress platelet-derived growth factor-BB. These cells are capable of mediating enhanced alveolar bone regeneration in vivo. High-stiffness transglutaminase crosslinked gelatins (TG-gels) were developed, incorporated with IL-4 and stromal cell-derived factor (SDF)-1α, and used for periodontal tissue regeneration. The presence of IL-4 might promote M2 polarization of macrophages (Mφs) and osteogenesis in bone marrow-derived stromal cells (BMSCs).8585 He XT, Li X, Xia Y, Yin Y, Wu RX, Sun HH, et al. Building capacity for macrophage modulation and stem cell recruitment in high-stiffness hydrogels for complex periodontal regeneration: experimental studies in vitro and in rats. Acta Biomater. 2019;88:162-80. doi: 10.1016/j.actbio.2019.02.004
https://doi.org/10.1016/j.actbio.2019.02...
In the field of dental pulp regeneration, hyaluronic acid (HA) hydrogels were reinforced with cellulose nanocrystals (CNCs) and then, enriched with platelet lysate (PL) and GFs. The PL-loaded hydrogels showed preferential supportive properties for encapsulated human dental pulp cells (hDPCs) in in vitro culture conditions. These hydrogels could serve as scaffold for GF delivery and cell recruitment, with great potential in future developments for regenerative dentistry.8686 Silva CR, Babo PS, Gulino M, Costa L, Oliveira JM, Silva-Correia J, et al. Injectable and tunable hyaluronic acid hydrogels releasing chemotactic and angiogenic growth factors for endodontic regeneration. Acta Biomater. 2018;77:155-71. doi: 10.1016/j.actbio.2018.07.035
https://doi.org/10.1016/j.actbio.2018.07...
Moreover, antibiotics and nitric oxide (NO), releasing biomimetic nanomatrix gel, were synthesized by the self-assembly of peptide amphiphiles, which could promote tooth revascularization with the maturation of root canals.8787 Moon CY, Nam OH, Kim M, Lee HS, Kaushik SN, Cruz Walma DA, et al. Effects of the nitric oxide releasing biomimetic nanomatrix gel on pulp-dentin regeneration: Pilot study. PLoS One. 2018;13(10):e0205534. doi: 10.1371/journal.pone.0205534
https://doi.org/10.1371/journal.pone.020...
Hydrogels can also be used to treat fungal infections and deliver anesthetics. Hydroxypropyl methylcellulose (HPMC) was used to load Histatin-5 (Hst-5), an antimicrobial peptide, which could treat oral candidiasis.8888 Kong EF, Tsui C, Boyce H, Ibrahim A, Hoag SW, Karlsson AJ, et al. Development and in vivo evaluation of a novel histatin-5 bioadhesive hydrogel formulation against oral candidiasis. Antimicrob Agents Chemother. 2016;60(2):881-9. doi: 10.1128/aac.02624-15.
https://doi.org/10.1128/aac.02624-15...
A nanostructured lipid-biopolymer hydrogel was developed to continuously deliver lidocaine-prilocaine for trans-buccal preanesthesia, which showed stability (for 6 months in critical conditions) and suitable mechanical properties for oral administration.8989 Ribeiro LN, Franz-Montan M, Breitkreitz MC, Rodrigues da Silva GH, Castro SR, Guilherme VA, et al. Nanohybrid hydrogels designed for transbuccal anesthesia. Int J Nanomedicine. 2018;13:6453-63. doi: 10.2147/ijn.S180080
https://doi.org/10.2147/ijn.S180080...
Natural products can also be used to make thermosensitive hydrogels for oral applications.9090 H RR, Dhamecha D, Jagwani S, Patil D, Hegde S, Potdar R, et al. Formulation of thermoreversible gel of cranberry juice concentrate: Evaluation, biocompatibility studies and its antimicrobial activity against periodontal pathogens. Mater Sci Eng C Mater Biol Appl. 2017;75:1506-14. doi: 10.1016/j.msec.2017.03.054
https://doi.org/10.1016/j.msec.2017.03.0...

Nanofibers

Nanofibers are commonly produced from ceramic materials, metallic compounds, and synthetic polymers by electrospinning, phase separation, self-assembly or laser spinning.9191 Sousa MG, Maximiano MR, Costa RA, Rezende TM, Franco OL. Nanofibers as drug-delivery systems for infection control in dentistry. Expert Opin Drug Deliv. 2020;17(7):919-30. doi: 10.1080/17425247.2020.1762564
https://doi.org/10.1080/17425247.2020.17...
Nanofibers are defined as fibers approximately 100 nm in diameter, which can carry diverse antimicrobial molecules, thereby showing biocompatibility and a porous structure.9191 Sousa MG, Maximiano MR, Costa RA, Rezende TM, Franco OL. Nanofibers as drug-delivery systems for infection control in dentistry. Expert Opin Drug Deliv. 2020;17(7):919-30. doi: 10.1080/17425247.2020.1762564
https://doi.org/10.1080/17425247.2020.17...
,9292 Monfared M, Taghizadeh S, Zare-Hoseinabadi A, Mousavi SM, Hashemi SA, Ranjbar S, et al. Emerging frontiers in drug release control by core-shell nanofibers: a review. Drug Metab Rev. 2019;51(4):589-611. doi: 10.1080/03602532.2019.1642912
https://doi.org/10.1080/03602532.2019.16...
High surface area and porosity are the main advantages of nanofibers. Drug release can be regulated by changing the porosity of nanofibers. Porous nanofibers can promote cell adhesion and shorten the drug release cycle, which is conducive to wound healing.9393 Chen X, Li H, Lu W, Guo Y. Antibacterial porous coaxial drug-carrying nanofibers for sustained drug-releasing applications. Nanomaterials (Basel). 2021;11(5). doi: 10.3390/nano11051316
https://doi.org/10.3390/nano11051316...
In the oral cavity, nanofibers have structural characteristics similar to dental pulp and periodontal tissues when compared with the most prevalent nanoparticles. Moreover, nanofibers have a larger contact surface which helps distant delivery.9191 Sousa MG, Maximiano MR, Costa RA, Rezende TM, Franco OL. Nanofibers as drug-delivery systems for infection control in dentistry. Expert Opin Drug Deliv. 2020;17(7):919-30. doi: 10.1080/17425247.2020.1762564
https://doi.org/10.1080/17425247.2020.17...
Their advantage of easy modulation of drug release profiles depends on the properties of polymer/polymeric blends/other materials used.9494 Thakkar S, Misra M. Electrospun polymeric nanofibers: new horizons in drug delivery. Eur J Pharm Sci. 2017;107:148-67. doi: 10.1016/j.ejps.2017.07.001
https://doi.org/10.1016/j.ejps.2017.07.0...
To treat Actinomyces naeslundii, P. gingivalis or Enterococcus faecalis infections, polydioxanone nanofibers were used to load antibiotics, such as metronidazole (MET), ciprofloxacin (CIP), and minocycline (MINO). The use of nanofibers suggested a significant potential in the eradication/elimination of bacterial biofilms.9595 Bottino MC, Albuquerque MT, Azabi A, Münchow EA, Spolnik KJ, Nör JE, et al. A novel patient-specific three-dimensional drug delivery construct for regenerative endodontics. J Biomed Mater Res B Appl Biomater. 2019;107(5):1576-86. doi: 10.1002/jbm.b.34250
https://doi.org/10.1002/jbm.b.34250...
,9696 Bottino MC, Kamocki K, Yassen GH, Platt JA, Vail MM, Ehrlich Y, et al. Bioactive nanofibrous scaffolds for regenerative endodontics. J Dent Res. 2013;92(11):963-9. doi: 10.1177/0022034513505770
https://doi.org/10.1177/0022034513505770...
,9797 Albuquerque MT, Ryan SJ, Münchow EA, Kamocka MM, Gregory RL, Valera MC, et al. Antimicrobial effects of novel triple antibiotic paste-mimic scaffolds on actinomyces naeslundii biofilm. J Endod. 2015;41(8):1337-43. doi: 10.1016/j.joen.2015.03.005
https://doi.org/10.1016/j.joen.2015.03.0...
Moreover, a type of polydioxanone nanofiber was developed to load vascular endothelial growth factor (VEGF), which might be a promising scaffold for additional optimization in endodontic regenerative procedures.9898 Yadlapati M, Biguetti C, Cavalla F, Nieves F, Bessey C, Bohluli P, et al. Characterization of a vascular endothelial growth factor-loaded bioresorbable delivery system for pulp regeneration. J Endod. 2017;43(1):77-83. doi: 10.1016/j.joen.2016.09.022
https://doi.org/10.1016/j.joen.2016.09.0...

Moreover, polyvinyl alcohol, polylactic acid, and some natural products are also used to prepare nanofiber carriers. Thiolated chitosan (CS-SH) blended with polyvinyl alcohol (PVA) was used as the carrier to load α-mangostin, an antibacterial substance, and prepared into nanofibers. These nontoxic nanofibers could rapidly adhere to the buccal mucosa in the oral cavity, in which α-mangostin was released to inhibit oral bacterial flora and promisingly prevent the formation of dental caries.9999 Samprasit W, Rojanarata T, Akkaramongkolporn P, Ngawhirunpat T, Kaomongkolgit R, Opanasopit P. Fabrication and in vitro/in vivo performance of mucoadhesive electrospun nanofiber mats containing α-mangostin. AAPS PharmSciTech. 2015;16(5):1140-52. doi: 10.1208/s12249-015-0300-6
https://doi.org/10.1208/s12249-015-0300-...
Resorbable PLA fibers based on electrospinning were developed, containing the antibiotic metronidazole. These fibers could slowly release metronidazole for the topical treatment of periodontitis.100100 Reise M, Wyrwa R, Müller U, Zylinski M, Völpel A, Schnabelrauch M, et al. Release of metronidazole from electrospun poly(L-lactide-co-D/L-lactide) fibers for local periodontitis treatment. Dent Mater. 2012;28(2):179-88. doi: 10.1016/j.dental.2011.12.006
https://doi.org/10.1016/j.dental.2011.12...
Cellulose nanofibers (CNF) were modified with κ-carrageenan oligosaccharides (CO) to deliver drugs, load surfactin, and manufacture surfactin-loaded CO-CNF. The surface-loaded CO-CNF showed antioxidant activity and inhibited the growth of S. mutansand P. gingivalis by preventing biofilm formation and reducing metabolic activity, which promoted oxidative stress in a concentration-dependent manner.101101 Johnson A, He JL, Kong F, Huang YC, Thomas S, Lin HV, et al. Surfactin-loaded ĸ-carrageenan oligosaccharides entangled cellulose nanofibers as a versatile vehicle against periodontal pathogens. Int J Nanomedicine. 2020;15:4021-47. doi: 10.2147/ijn.S238476
https://doi.org/10.2147/ijn.S238476...
However, most polymers currently used in nanofibers are expensive and not suitable for patients who have financial difficulties. At the same time, the stability of nanofibers and hosts’ immune responses should be further evaluated.9191 Sousa MG, Maximiano MR, Costa RA, Rezende TM, Franco OL. Nanofibers as drug-delivery systems for infection control in dentistry. Expert Opin Drug Deliv. 2020;17(7):919-30. doi: 10.1080/17425247.2020.1762564
https://doi.org/10.1080/17425247.2020.17...

Films

In general, oral films serve as carriers of antibacterial or antimycotic agents for gradual and direct release at the target area in the oral cavity, which might be suitable for long-term local effects and improve therapy effectiveness.102102 Yir-Erong B, Bayor MT, Ayensu I, Gbedema SY, Boateng JS. Oral thin films as a remedy for noncompliance in pediatric and geriatric patients. Ther Deliv. 2019;10(7):443-64. doi: 10.4155/tde-2019-0032
https://doi.org/10.4155/tde-2019-0032...
106106 Ibrahim YH, Regdon G Jr, Kristó K, Kelemen A, Adam ME, Hamedelniel EI, et al. Design and characterization of chitosan/citrate films as carrier for oral macromolecule delivery. Eur J Pharm Sci. 2020;146:105270. doi: 10.1016/j.ejps.2020.105270
https://doi.org/10.1016/j.ejps.2020.1052...
Mucoadhesive oral films (MOF) were prepared from carmellose by incorporating a nanotechnologically modified clay mineral intercalated with antiseptic drugs (chlorhexidine diacetate and digluconate) which could inhibitthe growth of Staphylococcus spp. and Candida spp.107107 Gajdziok J, Holešová S, Štembírek J, Pazdziora E, Landová H, Doležel P, et al. Carmellose mucoadhesive oral films containing vermiculite/chlorhexidine nanocomposites as innovative biomaterials for treatment of oral infections. Biomed Res Int. 2015;2015:580146. doi: 10.1155/2015/580146
https://doi.org/10.1155/2015/580146...
Gelatin films, as carriers to load econazole nitrate (ECN), were used as an imidazole antifungal agent to treat skin infections and mucosal candidiasis.108108 Dolci LS, Liguori A, Panzavolta S, Miserocchi A, Passerini N, Gherardi M, et al. Non-equilibrium atmospheric pressure plasma as innovative method to crosslink and enhance mucoadhesion of econazole-loaded gelatin films for buccal drug delivery. Colloids Surf B Biointerfaces. 2018;163:73-82. doi: 10.1016/j.colsurfb.2017.12.030
https://doi.org/10.1016/j.colsurfb.2017....
The plasma treatment completely dissolved the gelatin within 48 hours in a simulated saliva solution, which suggested a potential application for gelatin films as the buccal delivery of econazole to treat oral candidiasis.9797 Albuquerque MT, Ryan SJ, Münchow EA, Kamocka MM, Gregory RL, Valera MC, et al. Antimicrobial effects of novel triple antibiotic paste-mimic scaffolds on actinomyces naeslundii biofilm. J Endod. 2015;41(8):1337-43. doi: 10.1016/j.joen.2015.03.005
https://doi.org/10.1016/j.joen.2015.03.0...
Different hybrid nanofilms, composed of biopolymer matrices (chitosan, pectin, and chitosanpectin), were prepared and blended with nanostructured lipid carriers (NLC) and loaded in a eutectic mixture of 5% lidocaine-prilocaine (LDC–PLC). These nanohybrid films prolonged LDC-PLC release profile for more than 8 h in vitro. LDC-PLC shows higher drug permeation values across porcine oral mucosa and longer-lasting anesthesia without compromising its safety profile, indicating its significant potential to deliver local anesthetics.2222 Ribeiro LN, Franz-Montan M, Alcântara AC, Breitkreitz MC, Castro SR, Guilherme VA, et al. Hybrid nanofilms as topical anesthetics for pain-free procedures in dentistry. Sci Rep. 2020;10(1):11341. doi: 10.1038/s41598-020-68247-0
https://doi.org/10.1038/s41598-020-68247...

Other Drug-delivery Systems

In dental tissue engineering, growth factors and scaffolds are widely used to provide a 3D-scaffold structure with a highly porous, interconnected network that enables the transport of cellular nutrients.109109 Duruel T, Çakmak AS, Akman A, Nohutcu RM, Gümüşderelioğlu M. Sequential IGF-1 and BMP-6 releasing chitosan/alginate/PLGA hybrid scaffolds for periodontal regeneration. Int J Biol Macromol. 2017;104(Pt A):232-41. doi: 10.1016/j.ijbiomac.2017.06.029
https://doi.org/10.1016/j.ijbiomac.2017....
111111 Liu J, Ruan J, Weir MD, Ren K, Schneider A, Wang P, et al. Periodontal bone-ligament-cementum regeneration via scaffolds and stem cells. Cells. 2019;8(6). doi: 10.3390/cells8060537
https://doi.org/10.3390/cells8060537...
Polycaprolactone (PCL)/polylactic-co-glycolic acid was combined with amorphous polycaprolactone (PCL) to synthesize scaffolds which were immobilized with adenoviruses onto the scaffold surface to locally deliver gene vectors encoding platelet-derived growth factor-BB or bone morphogenetic protein-7. Controlled scaffold microtopography, combined with localized growth factor gene delivery, improved the regeneration of periodontal bone–periodontal ligament (PDL) interfaces.112112 Pilipchuk SP, Fretwurst T, Yu N, Larsson L, Kavanagh NM, Asa’ad F, et al. Micropatterned scaffolds with immobilized growth factor genes regenerate bone and periodontal ligament-like tissues. Adv Healthc Mater. 2018;7(22):e1800750. doi: 10.1002/adhm.201800750
https://doi.org/10.1002/adhm.201800750...
Chitosan/alginate/PLGA hybrid scaffolds were prepared for control and the sequential delivery of competence factors, such as insulin-like growth factor (IGF-1), progression factor, and bone morphogenetic factor-6 (BMP-6), which induced proliferation and osteoblastic differentiation of cementoblasts for periodontal tissue regeneration.109109 Duruel T, Çakmak AS, Akman A, Nohutcu RM, Gümüşderelioğlu M. Sequential IGF-1 and BMP-6 releasing chitosan/alginate/PLGA hybrid scaffolds for periodontal regeneration. Int J Biol Macromol. 2017;104(Pt A):232-41. doi: 10.1016/j.ijbiomac.2017.06.029
https://doi.org/10.1016/j.ijbiomac.2017....

Conclusion

Oral drug delivery systems have been designed in different ways to adapt to the physical and chemical environment of oral cavities for more suitable clinical applications. Carrier materials must have good biocompatibility and stable physical and chemical properties. The drugs loaded onto carriers are primarily antibacterial agents and bioactive substances that promote tissue growth. Temporary treatment with antibiotics is helpful in cases with severe symptoms of oral disease. Antibiotics can be used temporarily to suppress the dominant microbiota when severe dysbiosis occurs.

Indeed, we need to be alert to the abuse of antibiotics, which may lead to the dysbiosis of oral microbiota. Therefore, researchers need to pay attention to this issue when preparing carrier materials. Other drugs that preserve ecological balance may be considered for delivery in the future, such as prebiotic substrates.

Drug delivery systems can protect active substances in the complex oral environment and control their release to maintain an effective concentration. In general, hydrogels, scaffolds, and nanofibers can be used in tissue regeneration to treat periodontal and jaw defects because of their advantageous cell growth support. Carrier materials with good mucosal adhesion, such as films and hydrogels, can be used for mucosal drug delivery. Nanoparticles are often used to load active substances due to their strong dispersion, targeting bacteria or inflammatory cells. They have been widely used as drug delivery systems in the oral cavity to treat oral infectious diseases, such as caries and periodontal diseases. Therefore, effective carrier materials should continuously and steadily release effective drugs and target pathogenic bacteria, especially oral biofilms, to reduce cariogenic conditions without affecting the homeostasis of the oral flora and adapting to the oral environment.

Targeted and specific delivery systems may be the future development direction. For example, using the targeted delivery systems to adjust biofilm pH may prevent caries.

Researchers mainly use organic compounds to prepare carrier materials, but their biocompatibility and degraded byproducts need further verification. However, at present, drug delivery still faces challenges to its design and synthesis, and we lack a clear conclusion about the durability of its effect on resistant bacteria, hindering its clinical delivery. At the same time, there are few in vivo studies and animal experiments, and the biocompatibility of different drug delivery systems should be investigated further. The complexity of the oral environment and the lack of detailed studies on release rates and stability of specific drugs in drug delivery systems have become obstacles to their clinical application.

Acknowledgments

This work was supported by West China Hospital of Stomatology, Research and Develop Program, (RD-03-202002); Sichuan University new Century Higher Education Teaching Reform Project (the ninth phase) research project NO. SCU9361

References

  • 1
    Kamrani P, Sadiq NM. Anatomy, head and neck, oral cavity (mouth) [Internet]. In: StatPearls; 2021 [cited January 24, 2022]. Treasure Island (FL): StatPearls Publishing; 2022. Available from: https://www.ncbi.nlm.nih.gov/books/NBK545271/
    » https://www.ncbi.nlm.nih.gov/books/NBK545271/
  • 2
    Kitamoto S, Nagao-Kitamoto H, Hein R, Schmidt TM, Kamada N. The bacterial connection between the oral cavity and the gut diseases. J Dent Res. 2020;99(9):1021-9. doi: 10.1177/0022034520924633.
    » https://doi.org/10.1177/0022034520924633
  • 3
    Rathee M, Sapra A. Dental caries [Internet]. In: StatPearls. In: StatPearls; 2021 [cited January 24, 2022. Treasure Island (FL): StatPearls Publishing; 2022. Available from: https://www.ncbi.nlm.nih.gov/books/NBK551699/
    » https://www.ncbi.nlm.nih.gov/books/NBK551699/
  • 4
    Taylor M, Raja A. Oral candidiasis [Internet]. In: StatPearls; 2021 [cited January, 24, 2022]. Treasure Island (FL): StatPearls Publishing; 2022. Available from:. https://www.ncbi.nlm.nih.gov/books/NBK545282/
    » https://www.ncbi.nlm.nih.gov/books/NBK545282/
  • 5
    Dashper SG, Mitchell HL, KA LC, Carpenter L, Gussy MG, Calache H, et al. Temporal development of the oral microbiome and prediction of early childhood caries. Sci Rep. 2019;9(1):19732. doi: 10.1038/s41598-019-56233-0.
    » https://doi.org/10.1038/s41598-019-56233-0
  • 6
    Abusleme L, Dupuy AK, Dutzan N, Silva N, Burleson JA, Strausbaugh LD, et al. The subgingival microbiome in health and periodontitis and its relationship with community biomass and inflammation. Isme J. 2013;7(5):1016-25. doi: 10.1038/ismej.2012.174
    » https://doi.org/10.1038/ismej.2012.174
  • 7
    Philip N, Suneja B, Walsh LJ. Ecological approaches to dental caries prevention: paradigm shift or shibboleth? Caries Res. 2018;52(1-2):153-65. doi:10.1159/000484985
    » https://doi.org/10.1159/000484985
  • 8
    Rosier BT, Marsh PD, Mira A. Resilience of the oral microbiota in health: mechanisms that prevent dysbiosis. J Dent Res. 2018;97(4):371-80. doi:10.1177/0022034517742139
    » https://doi.org/10.1177/0022034517742139
  • 9
    Manji F, Dahlen G, Fejerskov O. Caries and periodontitis: contesting the conventional wisdom on their aetiology. Caries Res. 2018;52(6):548-64. doi:10.1159/000488948
    » https://doi.org/10.1159/000488948
  • 10
    Lamont RJ, Koo H, Hajishengallis G. The oral microbiota: dynamic communities and host interactions. Nat Rev Microbiol. 2018;16(12):745-59. doi:10.1038/s41579-018-0089-x
    » https://doi.org/10.1038/s41579-018-0089-x
  • 11
    Vos T, Abajobir AA, Abbafati C, Abbas KM, Abate KH, Abd-Allah F, et al. Global, regional, and national incidence, prevalence, and years lived with disability for 328 diseases and injuries for 195 countries, 1990-2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet. 2017;390(10100):1211-59. doi: 10.1016/S0140-6736(17)32154-2
    » https://doi.org/10.1016/S0140-6736(17)32154-2
  • 12
    Kinane DF, Stathopoulou PG, Papapanou PN. Periodontal diseases. Nat Rev Dis Primers. 2017;3:17038. doi: 10.1038/nrdp.2017.38
    » https://doi.org/10.1038/nrdp.2017.38
  • 13
    Armitage GC. Periodontal diagnoses and classification of periodontal diseases. Periodontol 2000. 2004;34:9-21. doi: 10.1046/j.0906-6713.2002.003421.x
    » https://doi.org/10.1046/j.0906-6713.2002.003421.x
  • 14
    Kornman KS, Page RC, Tonetti MS. The host response to the microbial challenge in periodontitis: assembling the players. Periodontol 2000. 1997;14:33-53. doi: 10.1111/j.1600-0757.1997.tb00191.x
    » https://doi.org/10.1111/j.1600-0757.1997.tb00191.x
  • 15
    Hajishengallis G. Periodontitis: from microbial immune subversion to systemic inflammation. Nat Rev Immunol. 2015;15(1):30-44. doi: 10.1038/nri3785
    » https://doi.org/10.1038/nri3785
  • 16
    Petersen PE, Ogawa H. The global burden of periodontal disease: towards integration with chronic disease prevention and control. Periodontol 2000. 2012;60(1):15-39. doi: 10.1111/j.1600-0757.2011.00425.x
    » https://doi.org/10.1111/j.1600-0757.2011.00425.x
  • 17
    Paula AJ, Koo H. Nanosized building blocks for customizing novel antibiofilm approaches. J Dent Res. 2017;96(2):128-36. doi: 10.1177/0022034516679397
    » https://doi.org/10.1177/0022034516679397
  • 18
    Makvandi P, Josic U, Delfi M, Pinelli F, Jahed V, Kaya E, et al. Drug delivery (nano)platforms for oral and dental applications: tissue regeneration, infection control, and cancer management. Adv Sci (Weinh). 2021;8(8):2004014. doi: 10.1002/advs.202004014
    » https://doi.org/10.1002/advs.202004014
  • 19
    Chen X, Wu G, Feng Z, Dong Y, Zhou W, Li B, et al. Advanced biomaterials and their potential applications in the treatment of periodontal disease. Crit Rev Biotechnol. 2016;36(4):760-75. doi: 10.3109/07388551.2015.1035693
    » https://doi.org/10.3109/07388551.2015.1035693
  • 20
    Kp K, R B. Evaluation and comparison of anti-inflammatory properties of ibuprofen using two drug delivery systems after third molar surgery: using chitosan microspheres as a carrier for local drug delivery in to the third molar socket and through the oral route. Br J Oral Maxillofac Surg. 2021;59(2):191-6. doi: 10.1016/j.bjoms.2020.08.025
    » https://doi.org/10.1016/j.bjoms.2020.08.025
  • 21
    Huang KH, Chen YW, Wang CY, Lin YH, Wu YA, Shie MY, et al. Enhanced capability of bone morphogenetic protein 2-loaded mesoporous calcium silicate scaffolds to induce odontogenic differentiation of human dental pulp cells. J Endod. 2018;44(11):1677-85. doi: 10.1016/j.joen.2018.08.008
    » https://doi.org/10.1016/j.joen.2018.08.008
  • 22
    Ribeiro LN, Franz-Montan M, Alcântara AC, Breitkreitz MC, Castro SR, Guilherme VA, et al. Hybrid nanofilms as topical anesthetics for pain-free procedures in dentistry. Sci Rep. 2020;10(1):11341. doi: 10.1038/s41598-020-68247-0
    » https://doi.org/10.1038/s41598-020-68247-0
  • 23
    Benoit DS, Sims KR Jr, Fraser D. Nanoparticles for oral biofilm treatments. ACS Nano. 2019;13(5):4869-75. doi: 10.1021/acsnano.9b02816
    » https://doi.org/10.1021/acsnano.9b02816
  • 24
    Raura N, Garg A, Arora A, Roma M. Nanoparticle technology and its implications in endodontics: a review. Biomater Res. 2020;24(1):21. doi: 10.1186/s40824-020-00198-z
    » https://doi.org/10.1186/s40824-020-00198-z
  • 25
    European Commission. Commission Recommendation of 18 October 2011 on the definition of nanomaterial Text with EEA relevance [Internet]. In: Official Journal of the European Union; 2011 [cited 2021 March 21]. Available from: https://eur-lex.europa.eu/eli/reco/2011/696
    » https://eur-lex.europa.eu/eli/reco/2011/696
  • 26
    McClements DJ, Xiao H. Is nano safe in foods? Establishing the factors impacting the gastrointestinal fate and toxicity of organic and inorganic food-grade nanoparticles. NPJ Sci Food. 2017;1:6. doi: 10.1038/s41538-017-0005-1
    » https://doi.org/10.1038/s41538-017-0005-1
  • 27
    Lohse SE, Murphy CJ. Applications of colloidal inorganic nanoparticles: from medicine to energy. J Am Chem Soc. 2012;134(38):15607-20. doi: 10.1021/ja307589n
    » https://doi.org/10.1021/ja307589n
  • 28
    Vallet-Regí M, Colilla M, Izquierdo-Barba I, Manzano M. Mesoporous silica nanoparticles for drug delivery: current insights. Molecules. 2017;23(1). doi: 10.3390/molecules23010047
    » https://doi.org/10.3390/molecules23010047
  • 29
    Chen L, Zhou X, He C. Mesoporous silica nanoparticles for tissue-engineering applications. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2019;11(6):e1573. doi: 10.1002/wnan.1573
    » https://doi.org/10.1002/wnan.1573
  • 30
    Leng D, Li Y, Zhu J, Liang R, Zhang C, Zhou Y, et al. The antibiofilm activity and mechanism of nanosilver- and nanozinc-incorporated mesoporous calcium-silicate nanoparticles. Int J Nanomedicine. 2020;15:3921-36. doi: 10.2147/ijn.S244686
    » https://doi.org/10.2147/ijn.S244686
  • 31
    Hua F, Yan J, Zhao S, Yang H, He H. In vitro remineralization of enamel white spot lesions with a carrier-based amorphous calcium phosphate delivery system. Clin Oral Investig. 2020;24(6):2079-89. doi: 10.1007/s00784-019-03073-x
    » https://doi.org/10.1007/s00784-019-03073-x
  • 32
    Luo XJ, Yang HY, Niu LN, Mao J, Huang C, Pashley DH, et al. Translation of a solution-based biomineralization concept into a carrier-based delivery system via the use of expanded-pore mesoporous silica. Acta Biomater. 2016;31:378-87. doi: 10.1016/j.actbio.2015.11.062
    » https://doi.org/10.1016/j.actbio.2015.11.062
  • 33
    Lu MM, Ge Y, Qiu J, Shao D, Zhang Y, Bai J, et al. Redox/pH dual-controlled release of chlorhexidine and silver ions from biodegradable mesoporous silica nanoparticles against oral biofilms. Int J Nanomedicine. 2018;13:7697-709. doi: 10.2147/ijn.S181168
    » https://doi.org/10.2147/ijn.S181168
  • 34
    Fan W, Wu D, Tay FR, Ma T, Wu Y, Fan B. Effects of adsorbed and templated nanosilver in mesoporous calcium-silicate nanoparticles on inhibition of bacteria colonization of dentin. Int J Nanomedicine. 2014;9:5217-30. doi: 10.2147/ijn.S73144
    » https://doi.org/10.2147/ijn.S73144
  • 35
    Fan W, Li Y, Sun Q, Ma T, Fan B. Calcium-silicate mesoporous nanoparticles loaded with chlorhexidine for both anti- Enterococcus faecalis and mineralization properties. J Nanobiotechnology. 2016;14(1):72. doi: 10.1186/s12951-016-0224-7
    » https://doi.org/10.1186/s12951-016-0224-7
  • 36
    Huang CY, Huang TH, Kao CT, Wu YH, Chen WC, Shie MY. Mesoporous calcium silicate nanoparticles with drug delivery and odontogenesis properties. J Endod. 2017;43(1):69-76. doi: 10.1016/j.joen.2016.09.012
    » https://doi.org/10.1016/j.joen.2016.09.012
  • 37
    Allaker RP. The use of nanoparticles to control oral biofilm formation. J Dent Res. 2010;89(11):1175-86. doi: 10.1177/0022034510377794
    » https://doi.org/10.1177/0022034510377794
  • 38
    Allaker RP, Memarzadeh K. Nanoparticles and the control of oral infections. Int J Antimicrob Agents. 2014;43(2):95-104. doi: 10.1016/j.ijantimicag.2013.11.002
    » https://doi.org/10.1016/j.ijantimicag.2013.11.002
  • 39
    Dizaj SM, Lotfipour F, Barzegar-Jalali M, Zarrintan MH, Adibkia K. Antimicrobial activity of the metals and metal oxide nanoparticles. Mater Sci Eng C Mater Biol Appl. 2014;44:278-84. doi: 10.1016/j.msec.2014.08.031
    » https://doi.org/10.1016/j.msec.2014.08.031
  • 40
    Mathur P, Jha S, Ramteke S, Jain NK. Pharmaceutical aspects of silver nanoparticles. Artif Cells Nanomed Biotechnol. 2018;46(sup1):115-26. doi: 10.1080/21691401.2017.1414825
    » https://doi.org/10.1080/21691401.2017.1414825
  • 41
    Tang H, Xu M, Zhou X, Zhang Y, Zhao L, Ye G, et al. Acute toxicity and biodistribution of different sized copper nano-particles in rats after oral administration. Mater Sci Eng C Mater Biol Appl. 2018;93:649-63. doi: 10.1016/j.msec.2018.08.032
    » https://doi.org/10.1016/j.msec.2018.08.032
  • 42
    Ding D, Zhu Q. Recent advances of PLGA micro/nanoparticles for the delivery of biomacromolecular therapeutics. Mater Sci Eng C Mater Biol Appl. 2018;92:1041-60. doi: 10.1016/j.msec.2017.12.036
    » https://doi.org/10.1016/j.msec.2017.12.036
  • 43
    Kumar G, Shafiq N, Malhotra S. Drug-loaded PLGA nanoparticles for oral administration: fundamental issues and challenges ahead. Crit Rev Ther Drug Carrier Syst. 2012;29(2):149-82. doi: 10.1615/critrevtherdrugcarriersyst.v29.i2.20
    » https://doi.org/10.1615/critrevtherdrugcarriersyst.v29.i2.20
  • 44
    Mir M, Ahmed N, Rehman AU. Recent applications of PLGA based nanostructures in drug delivery. Colloids Surf B Biointerfaces. 2017;159:217-31. doi: 10.1016/j.colsurfb.2017.07.038
    » https://doi.org/10.1016/j.colsurfb.2017.07.038
  • 45
    Rizeq BR, Younes NN, Rasool K, Nasrallah GK. Synthesis, bioapplications, and toxicity evaluation of chitosan-based nanoparticles. Int J Mol Sci. 2019;20(22). doi: 10.3390/ijms20225776
    » https://doi.org/10.3390/ijms20225776
  • 46
    Priyadarshini BM, Mitali K, Lu TB, Handral HK, Dubey N, Fawzy AS. PLGA nanoparticles as chlorhexidine-delivery carrier to resin-dentin adhesive interface. Dent Mater. 2017;33(7):830-46. doi: 10.1016/j.dental.2017.04.015
    » https://doi.org/10.1016/j.dental.2017.04.015
  • 47
    Fawzy AS, Priyadarshini BM, Selvan ST, Lu TB, Neo J. Proanthocyanidins-loaded nanoparticles enhance dentin degradation resistance. J Dent Res. 2017;96(7):780-9. doi: 10.1177/0022034517691757
    » https://doi.org/10.1177/0022034517691757
  • 48
    Lin HP, Tu HP, Hsieh YP, Lee BS. Controlled release of lovastatin from poly(lactic-co-glycolic acid) nanoparticles for direct pulp capping in rat teeth. Int J Nanomedicine. 2017;12:5473-85. doi: 10.2147/ijn.S138410
    » https://doi.org/10.2147/ijn.S138410
  • 49
    Lin JH, Feng F, Yu MC, Wang CH, Chang PC. Modulation of periodontitis progression using pH-responsive nanosphere encapsulating metronidazole or N-phenacylthialzolium bromide. J Periodontal Res. 2018;53(1):22-8. doi: 10.1111/jre.12481
    » https://doi.org/10.1111/jre.12481
  • 50
    Chang PC, Tai WC, Luo HT, Lai CH, Lin HH, Lin ZJ, et al. Core-Shell poly-(D,l-Lactide-co-Glycolide)-chitosan Nanospheres with simvastatin-doxycycline for periodontal and osseous repair. Int J Biol Macromol. 2020;158:627-35. doi: 10.1016/j.ijbiomac.2020.04.183
    » https://doi.org/10.1016/j.ijbiomac.2020.04.183
  • 51
    Spicer PP, Shah SR, Henslee AM, Watson BM, Kinard LA, Kretlow JD, et al. Evaluation of antibiotic releasing porous polymethylmethacrylate space maintainers in an infected composite tissue defect model. Acta Biomater. 2013;9(11):8832-9. doi: 10.1016/j.actbio.2013.07.018
    » https://doi.org/10.1016/j.actbio.2013.07.018
  • 52
    Li M, Du C, Guo N, Teng Y, Meng X, Sun H, et al. Composition design and medical application of liposomes. Eur J Med Chem. 2019;164:640-53. doi: 10.1016/j.ejmech.2019.01.007
    » https://doi.org/10.1016/j.ejmech.2019.01.007
  • 53
    Hazzah HA, Farid RM, Nasra MM, Hazzah WA, El-Massik MA, Abdallah OY. Gelucire-based nanoparticles for curcumin targeting to oral mucosa: preparation, characterization, and antimicrobial activity assessment. J Pharm Sci. 2015;104(11):3913-24. doi: 10.1002/jps.24590
    » https://doi.org/10.1002/jps.24590
  • 54
    Hu F, Zhou Z, Xu Q, Fan C, Wang L, Ren H, et al. A novel pH-responsive quaternary ammonium chitosan-liposome nanoparticles for periodontal treatment. Int J Biol Macromol. 2019;129:1113-9. doi: 10.1016/j.ijbiomac.2018.09.057
    » https://doi.org/10.1016/j.ijbiomac.2018.09.057
  • 55
    Liu H, Chen B, Mao Z, Gao C. Chitosan nanoparticles for loading of toothpaste actives and adhesion on tooth analogs. 2007;106(6):4248-56. doi: https://doi.org/10.1002/app.27078
    » https://doi.org/10.1002/app.27078
  • 56
    Nguyen S, Escudero C, Sediqi N, Smistad G, Hiorth M. Fluoride loaded polymeric nanoparticles for dental delivery. Eur J Pharm Sci. 2017;104:326-34. doi: 10.1016/j.ejps.2017.04.004
    » https://doi.org/10.1016/j.ejps.2017.04.004
  • 57
    Chronopoulou L, Nocca G, Castagnola M, Paludetti G, Ortaggi G, Sciubba F, et al. Chitosan based nanoparticles functionalized with peptidomimetic derivatives for oral drug delivery. N Biotechnol. 2016;33(1):23-31. doi: 10.1016/j.nbt.2015.07.005
    » https://doi.org/10.1016/j.nbt.2015.07.005
  • 58
    Priyadarshini BM, Selvan ST, Lu TB, Xie H, Neo J, Fawzy AS. Chlorhexidine nanocapsule drug delivery approach to the resin-dentin interface. J Dent Res. 2016;95(9):1065-72. doi: 10.1177/0022034516656135
    » https://doi.org/10.1177/0022034516656135
  • 59
    Shrestha S, Kishen A. Temporal-controlled bioactive molecules releasing core-shell nano-system for tissue engineering strategies in endodontics. Nanomedicine. 2019;18:11-20. doi: 10.1016/j.nano.2019.02.013
    » https://doi.org/10.1016/j.nano.2019.02.013
  • 60
    Zhu B, Li X, Xu X, Li J, Ding C, Zhao C, et al. One-step phosphorylated poly(amide-amine) dendrimer loaded with apigenin for simultaneous remineralization and antibacterial of dentine. Colloids Surf B Biointerfaces. 2018;172:760-8. doi: 10.1016/j.colsurfb.2018.09.036
    » https://doi.org/10.1016/j.colsurfb.2018.09.036
  • 61
    Wang W, Dang M, Zhang Z, Hu J, Eyster TW, Ni L, et al. Dentin regeneration by stem cells of apical papilla on injectable nanofibrous microspheres and stimulated by controlled BMP-2 release. Acta Biomater. 2016;36:63-72. doi: 10.1016/j.actbio.2016.03.015
    » https://doi.org/10.1016/j.actbio.2016.03.015
  • 62
    Onnainty R, Onida B, Páez P, Longhi M, Barresi A, Granero G. Targeted chitosan-based bionanocomposites for controlled oral mucosal delivery of chlorhexidine. Int J Pharm. 2016;509(1-2):408-18. doi: 10.1016/j.ijpharm.2016.06.011
    » https://doi.org/10.1016/j.ijpharm.2016.06.011
  • 63
    Park SC, Kim YM, Lee JK, Kim NH, Kim EJ, Heo H, et al. Targeting and synergistic action of an antifungal peptide in an antibiotic drug-delivery system. J Control Release. 2017;256:46-55. doi: 10.1016/j.jconrel.2017.04.023
    » https://doi.org/10.1016/j.jconrel.2017.04.023
  • 64
    Vidal-Romero G, Zambrano-Zaragoza ML, Martínez-Acevedo L, Leyva-Gómez G, Mendoza-Elvira SE, Quintanar-Guerrero D. Design and evaluation of ph-dependent nanosystems based on cellulose acetate phthalate, nanoparticles loaded with chlorhexidine for periodontal treatment. Pharmaceutics. 2019;11(11). doi: 10.3390/pharmaceutics11110604
    » https://doi.org/10.3390/pharmaceutics11110604
  • 65
    Sims KR Jr, Maceren JP, Liu Y, Rocha GR, Koo H, Benoit DS. Dual antibacterial drug-loaded nanoparticles synergistically improve treatment of Streptococcus mutans biofilms. Acta Biomater. 2020;115:418-31. doi: 10.1016/j.actbio.2020.08.032
    » https://doi.org/10.1016/j.actbio.2020.08.032
  • 66
    Murata T, Kutsuna T, Kurohara K, Shimizu K, Tomeoku A, Arai N. Evaluation of a new hydroxyapatite nanoparticle as a drug delivery system to oral squamous cell carcinoma cells. Anticancer Res. 2018;38(12):6715-20. doi: 10.21873/anticanres.13040
    » https://doi.org/10.21873/anticanres.13040
  • 67
    Jager J, Obst K, Lohan SB, Viktorov J, Staufenbiel S, Renz H, et al. Characterization of hyperbranched core-multishell nanocarriers as an innovative drug delivery system for the application at the oral mucosa. J Periodontal Res. 2018;53(1):57-65. doi: 10.1111/jre.12487
    » https://doi.org/10.1111/jre.12487
  • 68
    Chyzy A, Plonska-Brzezinska ME. Hydrogel properties and their impact on regenerative medicine and tissue engineering. molecules. 2020;25(24). doi: 10.3390/molecules25245795.
    » https://doi.org/10.3390/molecules25245795
  • 69
    Fennell E, Huyghe JM. Chemically responsive hydrogel deformation mechanics: a review. Molecules. 2019;24(19). doi: 10.3390/molecules24193521
    » https://doi.org/10.3390/molecules24193521
  • 70
    Sharma S, Tiwari S. A review on biomacromolecular hydrogel classification and its applications. Int J Biol Macromol. 2020;162:737-47. doi: 10.1016/j.ijbiomac.2020.06.110
    » https://doi.org/10.1016/j.ijbiomac.2020.06.110
  • 71
    Kirchhof S, Goepferich AM, Brandl FP. Hydrogels in ophthalmic applications. Eur J Pharm Biopharm. 2015;95(Pt B):227-38. doi: 10.1016/j.ejpb.2015.05.016
    » https://doi.org/10.1016/j.ejpb.2015.05.016
  • 72
    Cooper RC, Yang H. Hydrogel-based ocular drug delivery systems: emerging fabrication strategies, applications, and bench-to-bedside manufacturing considerations. J Control Release. 2019;306:29-39. doi: 10.1016/j.jconrel.2019.05.034
    » https://doi.org/10.1016/j.jconrel.2019.05.034
  • 73
    Narayanaswamy R, Torchilin VP. Hydrogels and their applications in targeted drug delivery. Molecules. 2019;24(3). doi: 10.3390/molecules24030603
    » https://doi.org/10.3390/molecules24030603
  • 74
    Mou J, Liu Z, Liu J, Lu J, Zhu W, Pei D. Hydrogel containing minocycline and zinc oxide-loaded serum albumin nanopartical for periodontitis application: preparation, characterization and evaluation. Drug Deliv. 2019;26(1):179-87. doi: 10.1080/10717544.2019.1571121
    » https://doi.org/10.1080/10717544.2019.1571121
  • 75
    Hosny KM, Aldawsari HM, Bahmdan RH, Sindi AM, Kurakula M, Alrobaian MM, et al. Preparation, optimization, and evaluation of hyaluronic acid-based hydrogel loaded with miconazole self-nanoemulsion for the treatment of oral thrush. AAPS PharmSciTech. 2019;20(7):297. doi: 10.1208/s12249-019-1496-7
    » https://doi.org/10.1208/s12249-019-1496-7
  • 76
    Franz-Montan M, Ribeiro LN, Volpato MC, Cereda CM, Groppo FC, Tofoli GR, et al. Recent advances and perspectives in topical oral anesthesia. Expert Opin Drug Deliv. 2017;14(5):673-84. doi: 10.1080/17425247.2016.1227784
    » https://doi.org/10.1080/17425247.2016.1227784
  • 77
    Pagano C, Giovagnoli S, Perioli L, Tiralti MC, Ricci M. Development and characterization of mucoadhesive-thermoresponsive gels for the treatment of oral mucosa diseases. Eur J Pharm Sci. 2020;142:105125. doi: 10.1016/j.ejps.2019.105125
    » https://doi.org/10.1016/j.ejps.2019.105125
  • 78
    Zhang M, Liang J, Yang Y, Liang H, Jia H, Li D. Current trends of targeted drug delivery for oral cancer therapy. Front Bioeng Biotechnol. 2020;8:618931. doi: 10.3389/fbioe.2020.618931
    » https://doi.org/10.3389/fbioe.2020.618931
  • 79
    Zang S, Mu R, Chen F, Wei X, Zhu L, Han B, et al. Injectable chitosan/β-glycerophosphate hydrogels with sustained release of BMP-7 and ornidazole in periodontal wound healing of class III furcation defects. Mater Sci Eng C Mater Biol Appl. 2019;99:919-28. doi: 10.1016/j.msec.2019.02.024
    » https://doi.org/10.1016/j.msec.2019.02.024
  • 80
    Xu X, Gu Z, Chen X, Shi C, Liu C, Liu M, et al. An injectable and thermosensitive hydrogel: Promoting periodontal regeneration by controlled-release of aspirin and erythropoietin. Acta Biomater. 2019;86:235-46. doi: 10.1016/j.actbio.2019.01.001
    » https://doi.org/10.1016/j.actbio.2019.01.001
  • 81
    Chang PC, Chao YC, Hsiao MH, Chou HS, Jheng YH, Yu XH, et al. Inhibition of periodontitis induction using a stimuli-responsive hydrogel carrying naringin. J Periodontol. 2017;88(2):190-6. doi: 10.1902/jop.2016.160189
    » https://doi.org/10.1902/jop.2016.160189
  • 82
    Chen W, Zhi M, Feng Z, Gao P, Yuan Y, Zhang C, et al. Sustained co-delivery of ibuprofen and basic fibroblast growth factor by thermosensitive nanoparticle hydrogel as early local treatment of peri-implantitis. Int J Nanomedicine. 2019;14:1347-58. doi: 10.2147/ijn.S190781
    » https://doi.org/10.2147/ijn.S190781
  • 83
    Tan J, Zhang M, Hai Z, Wu C, Lin J, Kuang W, et al. Sustained release of two bioactive factors from supramolecular hydrogel promotes periodontal bone regeneration. ACS Nano. 2019;13(5):5616-22. doi: 10.1021/acsnano.9b00788
    » https://doi.org/10.1021/acsnano.9b00788
  • 84
    Pan J, Deng J, Luo Y, Yu L, Zhang W, Han X, et al. Thermosensitive hydrogel delivery of human periodontal stem cells overexpressing platelet-derived growth factor-bb enhances alveolar bone defect repair. Stem Cells Dev. 2019;28(24):1620-31. doi: 10.1089/scd.2019.0184
    » https://doi.org/10.1089/scd.2019.0184
  • 85
    He XT, Li X, Xia Y, Yin Y, Wu RX, Sun HH, et al. Building capacity for macrophage modulation and stem cell recruitment in high-stiffness hydrogels for complex periodontal regeneration: experimental studies in vitro and in rats. Acta Biomater. 2019;88:162-80. doi: 10.1016/j.actbio.2019.02.004
    » https://doi.org/10.1016/j.actbio.2019.02.004
  • 86
    Silva CR, Babo PS, Gulino M, Costa L, Oliveira JM, Silva-Correia J, et al. Injectable and tunable hyaluronic acid hydrogels releasing chemotactic and angiogenic growth factors for endodontic regeneration. Acta Biomater. 2018;77:155-71. doi: 10.1016/j.actbio.2018.07.035
    » https://doi.org/10.1016/j.actbio.2018.07.035
  • 87
    Moon CY, Nam OH, Kim M, Lee HS, Kaushik SN, Cruz Walma DA, et al. Effects of the nitric oxide releasing biomimetic nanomatrix gel on pulp-dentin regeneration: Pilot study. PLoS One. 2018;13(10):e0205534. doi: 10.1371/journal.pone.0205534
    » https://doi.org/10.1371/journal.pone.0205534
  • 88
    Kong EF, Tsui C, Boyce H, Ibrahim A, Hoag SW, Karlsson AJ, et al. Development and in vivo evaluation of a novel histatin-5 bioadhesive hydrogel formulation against oral candidiasis. Antimicrob Agents Chemother. 2016;60(2):881-9. doi: 10.1128/aac.02624-15.
    » https://doi.org/10.1128/aac.02624-15
  • 89
    Ribeiro LN, Franz-Montan M, Breitkreitz MC, Rodrigues da Silva GH, Castro SR, Guilherme VA, et al. Nanohybrid hydrogels designed for transbuccal anesthesia. Int J Nanomedicine. 2018;13:6453-63. doi: 10.2147/ijn.S180080
    » https://doi.org/10.2147/ijn.S180080
  • 90
    H RR, Dhamecha D, Jagwani S, Patil D, Hegde S, Potdar R, et al. Formulation of thermoreversible gel of cranberry juice concentrate: Evaluation, biocompatibility studies and its antimicrobial activity against periodontal pathogens. Mater Sci Eng C Mater Biol Appl. 2017;75:1506-14. doi: 10.1016/j.msec.2017.03.054
    » https://doi.org/10.1016/j.msec.2017.03.054
  • 91
    Sousa MG, Maximiano MR, Costa RA, Rezende TM, Franco OL. Nanofibers as drug-delivery systems for infection control in dentistry. Expert Opin Drug Deliv. 2020;17(7):919-30. doi: 10.1080/17425247.2020.1762564
    » https://doi.org/10.1080/17425247.2020.1762564
  • 92
    Monfared M, Taghizadeh S, Zare-Hoseinabadi A, Mousavi SM, Hashemi SA, Ranjbar S, et al. Emerging frontiers in drug release control by core-shell nanofibers: a review. Drug Metab Rev. 2019;51(4):589-611. doi: 10.1080/03602532.2019.1642912
    » https://doi.org/10.1080/03602532.2019.1642912
  • 93
    Chen X, Li H, Lu W, Guo Y. Antibacterial porous coaxial drug-carrying nanofibers for sustained drug-releasing applications. Nanomaterials (Basel). 2021;11(5). doi: 10.3390/nano11051316
    » https://doi.org/10.3390/nano11051316
  • 94
    Thakkar S, Misra M. Electrospun polymeric nanofibers: new horizons in drug delivery. Eur J Pharm Sci. 2017;107:148-67. doi: 10.1016/j.ejps.2017.07.001
    » https://doi.org/10.1016/j.ejps.2017.07.001
  • 95
    Bottino MC, Albuquerque MT, Azabi A, Münchow EA, Spolnik KJ, Nör JE, et al. A novel patient-specific three-dimensional drug delivery construct for regenerative endodontics. J Biomed Mater Res B Appl Biomater. 2019;107(5):1576-86. doi: 10.1002/jbm.b.34250
    » https://doi.org/10.1002/jbm.b.34250
  • 96
    Bottino MC, Kamocki K, Yassen GH, Platt JA, Vail MM, Ehrlich Y, et al. Bioactive nanofibrous scaffolds for regenerative endodontics. J Dent Res. 2013;92(11):963-9. doi: 10.1177/0022034513505770
    » https://doi.org/10.1177/0022034513505770
  • 97
    Albuquerque MT, Ryan SJ, Münchow EA, Kamocka MM, Gregory RL, Valera MC, et al. Antimicrobial effects of novel triple antibiotic paste-mimic scaffolds on actinomyces naeslundii biofilm. J Endod. 2015;41(8):1337-43. doi: 10.1016/j.joen.2015.03.005
    » https://doi.org/10.1016/j.joen.2015.03.005
  • 98
    Yadlapati M, Biguetti C, Cavalla F, Nieves F, Bessey C, Bohluli P, et al. Characterization of a vascular endothelial growth factor-loaded bioresorbable delivery system for pulp regeneration. J Endod. 2017;43(1):77-83. doi: 10.1016/j.joen.2016.09.022
    » https://doi.org/10.1016/j.joen.2016.09.022
  • 99
    Samprasit W, Rojanarata T, Akkaramongkolporn P, Ngawhirunpat T, Kaomongkolgit R, Opanasopit P. Fabrication and in vitro/in vivo performance of mucoadhesive electrospun nanofiber mats containing α-mangostin. AAPS PharmSciTech. 2015;16(5):1140-52. doi: 10.1208/s12249-015-0300-6
    » https://doi.org/10.1208/s12249-015-0300-6
  • 100
    Reise M, Wyrwa R, Müller U, Zylinski M, Völpel A, Schnabelrauch M, et al. Release of metronidazole from electrospun poly(L-lactide-co-D/L-lactide) fibers for local periodontitis treatment. Dent Mater. 2012;28(2):179-88. doi: 10.1016/j.dental.2011.12.006
    » https://doi.org/10.1016/j.dental.2011.12.006
  • 101
    Johnson A, He JL, Kong F, Huang YC, Thomas S, Lin HV, et al. Surfactin-loaded ĸ-carrageenan oligosaccharides entangled cellulose nanofibers as a versatile vehicle against periodontal pathogens. Int J Nanomedicine. 2020;15:4021-47. doi: 10.2147/ijn.S238476
    » https://doi.org/10.2147/ijn.S238476
  • 102
    Yir-Erong B, Bayor MT, Ayensu I, Gbedema SY, Boateng JS. Oral thin films as a remedy for noncompliance in pediatric and geriatric patients. Ther Deliv. 2019;10(7):443-64. doi: 10.4155/tde-2019-0032
    » https://doi.org/10.4155/tde-2019-0032
  • 103
    Karki S, Kim H, Na S-J, Shin D, Jo K, Lee J. Thin films as an emerging platform for drug delivery. Asian J Pharmac Sci. 2016;11(5):559-74. doi: https://doi.org/10.1016/j.ajps.2016.05.004
    » https://doi.org/10.1016/j.ajps.2016.05.004
  • 104
    Castro PM, Sousa F, Magalhães R, Ruiz-Henestrosa VMP, Pilosof AM, Madureira AR, et al. Incorporation of beads into oral films for buccal and oral delivery of bioactive molecules. Carbohydr Polym. 2018;194:411-21. doi: 10.1016/j.carbpol.2018.04.032
    » https://doi.org/10.1016/j.carbpol.2018.04.032
  • 105
    Arafa MG, Ghalwash D, El-Kersh DM, Elmazar MM. Propolis-based niosomes as oromuco-adhesive films: A randomized clinical trial of a therapeutic drug delivery platform for the treatment of oral recurrent aphthous ulcers. Sci Rep. 2018;8(1):18056. doi: 10.1038/s41598-018-37157-7
    » https://doi.org/10.1038/s41598-018-37157-7
  • 106
    Ibrahim YH, Regdon G Jr, Kristó K, Kelemen A, Adam ME, Hamedelniel EI, et al. Design and characterization of chitosan/citrate films as carrier for oral macromolecule delivery. Eur J Pharm Sci. 2020;146:105270. doi: 10.1016/j.ejps.2020.105270
    » https://doi.org/10.1016/j.ejps.2020.105270
  • 107
    Gajdziok J, Holešová S, Štembírek J, Pazdziora E, Landová H, Doležel P, et al. Carmellose mucoadhesive oral films containing vermiculite/chlorhexidine nanocomposites as innovative biomaterials for treatment of oral infections. Biomed Res Int. 2015;2015:580146. doi: 10.1155/2015/580146
    » https://doi.org/10.1155/2015/580146
  • 108
    Dolci LS, Liguori A, Panzavolta S, Miserocchi A, Passerini N, Gherardi M, et al. Non-equilibrium atmospheric pressure plasma as innovative method to crosslink and enhance mucoadhesion of econazole-loaded gelatin films for buccal drug delivery. Colloids Surf B Biointerfaces. 2018;163:73-82. doi: 10.1016/j.colsurfb.2017.12.030
    » https://doi.org/10.1016/j.colsurfb.2017.12.030
  • 109
    Duruel T, Çakmak AS, Akman A, Nohutcu RM, Gümüşderelioğlu M. Sequential IGF-1 and BMP-6 releasing chitosan/alginate/PLGA hybrid scaffolds for periodontal regeneration. Int J Biol Macromol. 2017;104(Pt A):232-41. doi: 10.1016/j.ijbiomac.2017.06.029
    » https://doi.org/10.1016/j.ijbiomac.2017.06.029
  • 110
    Saranya N, Saravanan S, Moorthi A, Ramyakrishna B, Selvamurugan N. Enhanced osteoblast adhesion on polymeric nano-scaffolds for bone tissue engineering. J Biomed Nanotechnol. 2011;7(2):238-44. doi: 10.1166/jbn.2011.1283
    » https://doi.org/10.1166/jbn.2011.1283
  • 111
    Liu J, Ruan J, Weir MD, Ren K, Schneider A, Wang P, et al. Periodontal bone-ligament-cementum regeneration via scaffolds and stem cells. Cells. 2019;8(6). doi: 10.3390/cells8060537
    » https://doi.org/10.3390/cells8060537
  • 112
    Pilipchuk SP, Fretwurst T, Yu N, Larsson L, Kavanagh NM, Asa’ad F, et al. Micropatterned scaffolds with immobilized growth factor genes regenerate bone and periodontal ligament-like tissues. Adv Healthc Mater. 2018;7(22):e1800750. doi: 10.1002/adhm.201800750
    » https://doi.org/10.1002/adhm.201800750

Publication Dates

  • Publication in this collection
    09 Mar 2022
  • Date of issue
    2022

History

  • Received
    08 June 2021
  • Reviewed
    07 Nov 2021
  • Accepted
    14 Dec 2021
Faculdade De Odontologia De Bauru - USP Serviço de Biblioteca e Documentação FOB-USP, Al. Dr. Octávio Pinheiro Brisolla 9-75, 17012-901 Bauru SP Brasil, Tel.: +55 14 32358373 - Bauru - SP - Brazil
E-mail: jaos@usp.br