Next Article in Journal
Effect of Near-Infrared Blood Photobiomodulation on Red Blood Cell Damage from the Extracorporeal Circuit during Hemodialysis In Vitro
Next Article in Special Issue
The Effect of 5-Aminolevulinic Acid Photodynamic Therapy in Promoting Pyroptosis of HPV-Infected Cells
Previous Article in Journal
Spurious Tone Reduction and Signal Stabilization of Optoelectronic Oscillators by Low-Frequency RF Signal Modulation
Previous Article in Special Issue
Review of Virus Inactivation by Visible Light
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Randomized and Controlled Clinical Studies on Antibacterial Photodynamic Therapy: An Overview

by
Fernanda Alves
1,
Mirian D. Stringasci
1,
Michelle B. Requena
1,
Kate C. Blanco
1,2,
Lucas D. Dias
1,
Thaila Q. Corrêa
1 and
Vanderlei S. Bagnato
1,2,*
1
São Carlos Institute of Physics, University of São Paulo, São Carlos 13566-590, SP, Brazil
2
Department of Biomedical Engineering, Texas A&M University, College Station, TX 77843, USA
*
Author to whom correspondence should be addressed.
Photonics 2022, 9(5), 340; https://doi.org/10.3390/photonics9050340
Submission received: 31 March 2022 / Revised: 6 May 2022 / Accepted: 11 May 2022 / Published: 13 May 2022

Abstract

:
The emergence of drug-resistant bacteria is considered a critical public health problem. The need to establish alternative approaches to countering resistant microorganisms is unquestionable in overcoming this problem. Among emerging alternatives, antimicrobial photodynamic therapy (aPDT) has become promising to control infectious diseases. aPDT is based on the activation of a photosensitizer (PS) by a particular wavelength of light followed by generation of the reactive oxygen. These interactions result in the production of reactive oxygen species, which are lethal to bacteria. Several types of research have shown that aPDT has been successfully studied in in vitro, in vivo, and randomized clinical trials (RCT). Considering the lack of reviews of RCTs studies with aPDT applied in bacteria in the literature, we performed a systematic review of aPDT randomized clinical trials for the treatment of bacteria-related diseases. According to the literature published from 2008 to 2022, the RCT study of aPDT was mostly performed for periodontal disease, followed by halitosis, dental infection, peri-implantitis, oral decontamination, and skin ulcers. A variety of PSs, light sources, and protocols were efficiently used, and the treatment did not cause any side effects for the individuals.

1. Introduction

Currently, one of the most important clinical challenges in the world is the increasing resistance of bacteria to antibiotics. According to recent reports about drug-resistant infections, the actual scenario shows a risk is being posed to the ability to treat common infections with any single kind of antibiotic, with antimicrobial resistance being one of the top 10 global public health threats facing humanity [1,2]. Humans have exposed microorganisms, specifically pathogenic microbial populations, to antimicrobial agents, such as antibiotics and antiseptics, to control infectious diseases. The overuse of these substances makes disease-causing microorganisms develop various resistance mechanisms to drugs commonly used to treat them, which is a severe worldwide threat to managing infectious diseases [3]. To overcome the resistance problem, the search for alternative approaches is necessary. Antimicrobial photodynamic therapy (aPDT) has become a promising and potential treatment, since it is nontoxic, noninvasive, and has presented effective results against microorganisms, while not causing them to quickly develop resistance [4].
The mechanisms of aPDT are based on the interaction of the photosensitizer (PS) molecule with light in a compatible wavelength in the presence of molecular oxygen (Figure 1). When the PS molecule in its singlet ground state (S0) absorbs a photon (hν), it transitions to the singlet excited state (Sn). From Sn, the PS can release energy through fluorescence emission (f) or through internal conversion, releasing heat, then returning to the ground state, S0. The PS molecule may, from Sn, still undergo an intersystem crossing (ISC) to a triplet excited state (T1) with a longer lifetime. From T1, the PS may return to S0 after emitting phosphorescence (P) or after participating in reactions that lead to the generation of reactive oxygen species (ROS): type I reactions and type II reactions. In a type I reaction, when the PS is in the T1, it can transfer a proton or an electron to the substrate to form a radical anion or radical cation; these radicals may react with oxygen to produce ROS. In a type 2 reaction, the PS in the T1 can directly transfer energy to molecular oxygen (a triplet in the ground state), producing excited-state singlet oxygen (1O2). Both type 1 and 2 reactions occur simultaneously, but depending on the chemical structure of the PS, one of the reactions will be preferential. The efficiency of the aPDT is often related to the 1O2 quantum yield of the PS [5,6,7,8]. The photoproducts species are responsible for inducing the death of the target cell. Then, aPDT represents a multi-target damaging process, since the reactive oxygen species generated can interact and damage all structures that are close to them. For this reason, the effectiveness of the aPDT is also related to the PS localization and uptake, where the oxidative process will occur [9,10]. Moreover, the ability of the ROS to damage a nonspecific site means that aPDT is unlikely to induce resistance in the microorganisms, and this characteristic is an advantage of aPDT over antibiotics. Antibiotics, regardless of the class, bind to and act on a specific target, to that the bacteria is more likely to develop resistance to them.
Photodynamic action has been applied to non-melanoma skin lesions with well-established protocols and is strongly recommended by the American and European academies of dermatology [11,12,13]. Moreover, many other areas have promisingly benefited from this technique with marked improvement in human-health-concerning infections; however, these do not have defined, validated, effective, and secure protocols.
For example, some aPDT applications, such as the control of disease vectors, are also a great gain that photodynamics can offer. Through the use of appropriate PSs, it is possible to place breeding sites for vectors of the main diseases, such as dengue, etc., in suitable forms that allow the larvae to absorb the substance. With this and with the help of sunlight, the elimination of these larvae can reach 90%, without any aggression to the environment [14,15]. Decontamination of blood banks is also a source of infection for humans [16] and photodynamic with riboflavin and UVA light allows a considerable decrease in the viral and bacterial load of blood bags, thus decreasing the chances of contamination of receptors [17].
Additionally, many infectious diseases are caused by contamination in food [18]. In the case of raw foods (meats, grains, vegetables, and fruits), the number of contaminants that remain in the food when it reaches the consumer’s hands are large. The action of aPDT has been shown to be adequate for the preservation and elimination of infectious factors in food. Finally, water-borne diseases are also a global burden that is estimated to cause several million deaths and innumerable cases of sickness every year. The application of photodynamic processes has been exploited to address the decontamination of waters, where sunlight-mediated aPDT can eliminate pathogens present in municipal and other water supplies [19]. These examples are some cases on the subject, which demonstrate that photodynamic inhibition can go far beyond human health.
The antimicrobial potential of PDT has also been widely and successfully applied for the management of bacterial infectious diseases, such as for the oral decontamination of orthodontic patients [20,21]; for the inactivation of Streptococcus mutans biofilm [22,23] and Staphylococcus aureus biofilm in in vitro and in vivo studies [24]; in the treatment of pharyngotonsillitis [25,26]; against bacteria that cause pulmonary diseases [27,28,29]; in the decontamination of blood [16,30,31]; and in cooperative and competitive aPDT effects [32]. Some studies have focused on the development of new compounds for the enhancement of aPDT [33]. In this context, researchers have synthesized nanoparticle and dye diffusion in bacterial biofilms for aPDT applications [34], such as the use of superhydrophobic sensitizer techniques in the treatment of periodontitis [35].
The natural evolution of these in vitro and in vivo aPDT findings is to translate them to clinical trials aiming to define and validate effective and secure protocols. There are several types of clinical studies, such as randomized controlled trials (RCT), cohort studies, case–control studies, case series, case reports, and opinion reports. Considering the hierarchy of evidence parameters among them, RCT can be considered the gold standard of clinical trials, providing the most reliable evidence of the effectiveness of interventions. RCTs are designed to have the participants randomly assigned to one of two or more clinical interventions which minimize the risk of confounding factors influencing the results [36].
Considering the lack in the literature of reviews reuniting RCTs studies with aPDT applied in bacteria, we performed a systematic review aiming at aPDT randomized clinical trials to treat bacteria-related diseases.

2. Materials and Methods

The present systematic review searched aPDT randomized clinical trials for the treatment of bacteria-related diseases. The studies were collected from The Web of Science database, using the keywords “Photodynamic”, “bacteria”, and “randomized controlled”, from 2008 to 2022.

3. Results and Discussion

Figure 2 summarizes the percentage of each aPDT application in the studies evaluated. Periodontal disease was the most common application explored in the studies (65%), followed by halitosis (14%), dental infection (10%), peri-implantitis (6%), oral decontamination (2%), and skin ulcers (2%), respectively. Table 1, Table 2, Table 3, Table 4, Table 5 and Table 6 show the studies’ details organized by diseases found in the present search.
About 65% of the studies (32/49) we reviewed were related to periodontal disease and they are presented in Table 1. Periodontal disease affects the gingiva, the supporting connective tissue, and the alveolar bone, which anchor the teeth in the jaws. Gingivitis is the mildest form of periodontal disease and is caused by bacterial biofilm (dental plaque) that accumulates on teeth surface adjacent to the gingiva (gums). Periodontitis causes loss of connective tissue and bone support, being a significant cause of tooth loss in adults. In addition to pathogenic microorganisms in the biofilm, genetic and environmental factors, especially tobacco use, contribute to the cause of these diseases [86,87]. The treatment of disease is directed at slowing the progression of the disease process, with mechanical removal of the bacteria. The treatment regime depends on the severity of the disease, the presence or absence of periodontal pockets, and the extent of the loss of alveolar bone; the more advanced the destruction, the more mechanical intervention is necessary [86]. Most of the studies presented in Table 1 compare the association of aPDT with conventional methods: ultrasonic debridement and/or scaling and root planning (SRP).
Among the aPDT protocols used, phenothiazine chlorine, methylene blue, toluidine blue, and indocyanine green (ICG) were adopted as PSs with a maximum incubation period of 5 min. The regions treated with ICG were irradiated at 810 nm, while the irradiations were performed at ranges between 628 and 680 nm to use the other PSs. It is also possible to verify the irradiation protocols, with irradiance varying between 2 and 100 mW/cm2 and fluences between 20 and 320 J/cm2. The trials presented follow-up reviews of 21 days–12 months.
Several studies point out that there is no significant difference between the conventional procedure groups without or with the association of aPDT. In contrast, several other studies observed significant clinical differences as a reduction in bleeding scores, gingival inflammation, and some of the critical periodontal pathogens. All protocols that used ICG as PSs in association with conventional treatment observed improvement in the clinical aspect, concluding that aPDT is a promising adjunct to nonsurgical periodontal therapy [51,65,68]. A single study was performed comparing conventional ultrasonic and aPDT alone and both therapies resulted in the same clinical effect; however, aPDT was less harmful to teeth than ultrasonic therapy [41].
Moreover, aPDT has been applied to treat other infectious diseases, such as halitosis (Table 2), peri-implantitis (Table 3), dental infection (Table 4), oral decontamination (Table 5), and ulcers on the skin (Table 6). Concerning halitosis, it is an oral condition that is characterized by unpleasant odors emanating from the oral cavity caused by deep carious lesions, peri-implant disease, periodontal disease, oral infections, mucosal ulcerations, pericoronitis, and impacted food. [88] In order to develop a safe and effective protocol to treat halitosis, authors from Brazil and Saudi Arabia evaluated (2014–2021) different clinical photodynamic protocols/conditions for treating the tongue, e.g., light parameters (395–660 nm, 36–318 J/cm2), PS type, and concentration and with different follow-up periods (7 days–3 months). These studies reported the effectiveness (reduction in oral pathogens) of aPDT against halitosis. Methylene blue has been applied as a PS to treat halitosis due to its low toxicity and high efficiency. Additionally, the authors concluded that aPDT is a useful and efficient option for treating halitosis without mechanical aggression of the lingual papillae.
Furthermore, aPDT was evaluated in treatment of peri-implantitis (Table 3). This is a pathological health condition in tissues (around dental implants), characterized by an inflammation process in the peri-implant tissue and loss of supporting bone [89]. According to our knowledge, there are three randomized controlled clinical studies concerning the application of aPDT to treat peri-implantitis. In these studies, authors reported the use of aPDT as an adjunctive option or as the primary treatment option to treat initial peri-implantitis, decontamination of the implant surface, and peri-implant diseases. Only two different PSs were applied to treat peri-implantitis, namely phenothiazine chloride and Fotosan. The authors observed a significant reduction in bleeding on probing compared with chlorhexidine (control group) and substantial decontamination of implant surfaces. Moreover, the authors observed an effective reduction in mucosal inflammation process.
Besides periodontal disease, halitosis, and peri-implantitis, there are a set of randomized controlled clinical studies reporting the use of aPDT against dental infections (Table 4) and oral decontamination (Table 5). The authors used different parameters, PSs (toluidine blue, methylene blue, and curcumin), and control groups (e.g., antibiotics, calcium hydroxide therapy, and others) to compare the results obtained. In addition, the authors reported that aPDT can disaggregate oral plaque and reduce the pathogenic microorganisms load present in the oral cavity. In this regard, aPDT was also applied to treat ulcers (chronic diabetic foot) and the authors demonstrated a significant photoinactivation of bacteria (Table 6).

4. Conclusions

Finally, it is possible to conclude that, from 2008 to 2022, aPDT was studied mainly for dentistry applications and was demonstrated to have promising clinical results. A variety of PSs, light sources, and protocols were efficiently used, and the treatment did not cause any side effects for the individuals. However, it is important to emphasize that the lack of standardization in the studies hinders the comparison among them and hinders the translation of preclinical results to clinical studies, which can lead to the failure of the treatment. Future studies should consider performing randomized clinical trials evaluating other infectious diseases, since this treatment has demonstrated antimicrobial effectiveness in in vitro, in vivo, and clinical reports; however, there are few RCT studies that could help the feasibility of this therapy for the management of other infectious diseases. Besides that, the constant emergence of resistant bacteria to the conventional antibiotics and the improbably improbable development of resistance to aPDT, turn photodynamic therapy into a powerful alternative antimicrobial method.

Author Contributions

Conceptualization, F.A., M.D.S., M.B.R., K.C.B., L.D.D., T.Q.C. and V.S.B.; methodology, F.A. and L.D.D.; writing—original draft preparation, F.A., M.D.S., M.B.R., K.C.B., L.D.D. and T.Q.C.; writing—review and editing, F.A., M.D.S., M.B.R., K.C.B., L.D.D., T.Q.C. and V.S.B.; supervision, V.S.B.; project administration, F.A.; funding acquisition, V.S.B. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the São Paulo Research Foundation (FAPESP/CEPOF), Proc. Nº 13/07276-1. Blanco K.C. thanks FAPESP for Post-doc grant 2019/12694-3 and 2021/09952-0. L.D. Dias thanks FAPESP for Post-doc grant 2019/13569-8. F.A. thanks FAPESP for Post-doc grant 2021/01324-0.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors acknowledge the free data base used to create part of the Figure 1 and Figure 2. The websites are: https://www.authoritydental.org/, https://commons.wikimedia.org/wiki/File:Porphyrin-3D-spacefill.png, https://commons.wikimedia.org/wiki/File:Oxygen_molecule.svg, accessed on 2 April 2022.

Conflicts of Interest

There are no conflict of interest.

References

  1. World Health Organization. Antimicrobial Resistance: Global Report on Surveillance; World Health Organization: Geneva, Switzerland, 2014. [Google Scholar]
  2. O’neill, J. Tackling Drug-Resistant Infections Globally: Final Report and Recommendations: Review on Antimicrobial Resistance; Government of the United Kingdom: London, UK, 2018.
  3. Kashef, N.; Hamblin, M.R. Can microbial cells develop resistance to oxidative stress in antimicrobial photodynamic inactivation? Drug Resist. Updat. 2017, 31, 31–42. [Google Scholar] [CrossRef] [PubMed]
  4. Youf, R.; Müller, M.; Balasini, A.; Thétiot, F.; Müller, M.; Hascoët, A.; Jonas, U.; Schönherr, H.; Lemercier, G.; Montier, T.; et al. Antimicrobial Photodynamic Therapy: Latest Developments with a Focus on Combinatory Strategies. Pharmaceutics 2021, 13, 1995. [Google Scholar] [CrossRef] [PubMed]
  5. Cieplik, F.; Deng, D.; Crielaard, W.; Buchalla, W.; Hellwig, E.; Al-Ahmad, A.; Maisch, T. Antimicrobial photodynamic therapy-what we know and what we don’t. Crit. Rev. Microbiol. 2018, 44, 571–589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Klausen, M.; Ucuncu, M.; Bradley, M. Design of Photosensitizing Agents for Targeted Antimicrobial Photodynamic Therapy. Molecules 2020, 25, 5239. [Google Scholar] [CrossRef] [PubMed]
  7. Chandna, S.; Thakur, N.S.; Kaur, R.; Bhaumik, J. Lignin–Bimetallic Nanoconjugate Doped pH-Responsive Hydrogels for Laser-Assisted Antimicrobial Photodynamic Therapy. Biomacromolecules 2020, 21, 3216–3230. [Google Scholar] [CrossRef]
  8. Dąbrowski, J.M. Reactive Oxygen Species in Photodynamic Therapy: Mechanisms of Their Generation and Potentiation. In Advances in Inorganic Chemistry; Academic Press: Cambridge, MA, USA, 2017; pp. 343–394. [Google Scholar]
  9. Jori, G.; Fabris, C.; Soncin, M.; Ferro, S.; Coppellotti, O.; Dei, D.; Fantetti, L.; Chiti, G.; Roncucci, G. Photodynamic therapy in the treatment of microbial infections: Basic principles and perspective applications. Lasers Surg. Med. 2006, 38, 468–481. [Google Scholar] [CrossRef]
  10. Bacellar, I.O.L.; Oliveira, M.C.; Dantas, L.S.; Costa, E.B.; Junqueira, H.C.; Martins, W.K.; Durantini, A.M.; Cosa, G.; Di Mascio, P.; Wainwright, M.; et al. Photosensitized Membrane Permeabilization Requires Contact-Dependent Reactions between Photosensitizer and Lipids. J. Am. Chem. Soc. 2018, 140, 9606–9615. [Google Scholar] [CrossRef]
  11. Morton, C.A.; Szeimies, R.M.; Basset-Séguin, N.; Calzavara-Pinton, P.G.; Gilaberte, Y.; Hædersdal, M.; Hofbauer, G.F.L.; Hunger, R.E.; Karrer, S.; Piaserico, S.; et al. European Dermatology Forum guidelines on topical photodynamic therapy 2019 Part 2: Emerging indications–field cancerization, photorejuvenation and inflammatory/infective dermatoses. J. Eur. Acad. Dermatol. Venereol. 2020, 34, 17–29. [Google Scholar] [CrossRef]
  12. Morton, C.A.; Szeimies, R.M.; Basset-Seguin, N.; Calzavara-Pinton, P.; Gilaberte, Y.; Hædersdal, M.; Hofbauer, G.F.L.; Hunger, R.E.; Karrer, S.; Piaserico, S.; et al. European Dermatology Forum guidelines on topical photodynamic therapy 2019 Part 1: Treatment delivery and established indications–actinic keratoses, Bowen’s disease and basal cell carcinomas. J. Eur. Acad. Dermatol. Venereol. 2019, 33, 2225–2238. [Google Scholar] [CrossRef]
  13. Braathen, L.R.; Szeimies, R.-M.; Basset-Seguin, N.; Bissonnette, R.; Foley, P.; Pariser, D.; Roelandts, R.; Wennberg, A.-M.; Morton, C.A. Guidelines on the use of photodynamic therapy for nonmelanoma skin cancer: An international consensus. J. Am. Acad. Dermatol. 2007, 56, 125–143. [Google Scholar] [CrossRef]
  14. Mezzacappo, N.F.; Souza, L.M.; Inada, N.M.; Dias, L.D.; Garbuio, M.; Venturini, F.P.; Corrêa, T.Q.; Moura, L.; Blanco, K.C.; de Oliveira, K.T.; et al. Curcumin/d-mannitol as photolarvicide: Induced delay in larval development time, changes in sex ratio and reduced longevity of Aedes aegypti. Pest Manag. Sci. 2021, 77, 2530–2538. [Google Scholar] [CrossRef] [PubMed]
  15. De Souza, L.M.; Inada, N.M.; Venturini, F.P.; Carmona-Vargas, C.C.; Pratavieira, S.; de Oliveira, K.T.; Kurachi, C.; Bagnato, V.S. Photolarvicidal effect of curcuminoids from Curcuma longa Linn. against Aedes aegypti larvae. J. Asia. Pac. Entomol. 2019, 22, 151–158. [Google Scholar] [CrossRef]
  16. Corrêa, T.Q.; Blanco, K.C.; Soares, J.M.; Inada, N.M.; Kurachi, C.; Golim, M.D.A.; Deffune, E.; Bagnato, V.S. Photodynamic inactivation for in vitro decontamination of Staphylococcus aureus in whole blood. Photodiagn. Photodyn. Ther. 2019, 28, 58–64. [Google Scholar] [CrossRef] [PubMed]
  17. Zhu, L.; Li, C.; Wang, D. A novel ultraviolet illumination used in riboflavin photochemical method to inactivate drug-resistant bacteria in blood components. J. Photochem. Photobiol. B Biol. 2020, 204, 111782. [Google Scholar] [CrossRef] [PubMed]
  18. Corrêa, T.Q.; Blanco, K.C.; Garcia, E.B.; Perez, S.M.L.; Chianfrone, D.J.; Morais, V.S.; Bagnato, V.S. Effects of ultraviolet light and curcumin-mediated photodynamic inactivation on microbiological food safety: A study in meat and fruit. Photodiagn. Photodyn. Ther. 2020, 30, 101678. [Google Scholar] [CrossRef] [PubMed]
  19. Jori, G.; Magaraggia, M.; Fabris, C.; Soncin, M.; Camerin, M.; Tallandini, L.; Coppellotti, O.; Guidolin, L. Photodynamic Inactivation of Microbial Pathogens: Disinfection of Water and Prevention of Water-Borne Diseases. J. Environ. Pathol. Toxicol. Oncol. 2011, 30, 261–271. [Google Scholar] [CrossRef] [PubMed]
  20. Panhóca, V.H.; Luis Esteban Florez, F.; Quatrini Corrêa, T.; Paolillo, F.R.; Oliveira de Souza, C.W.; Bagnato, V.S. Oral decontamination of orthodontic patients using photodynamic therapy mediated by blue-light irradiation and curcumin associated with sodium dodecyl sulfate. Photomed. Laser Surg. 2016, 34, 411–417. [Google Scholar] [CrossRef]
  21. Al-Shammery, D.; Michelogiannakis, D.; Ahmed, Z.U.; Ahmed, H.B.; Rossouw, P.E.; Romanos, G.E.; Javed, F. Scope of antimicrobial photodynamic therapy in Orthodontics and related research: A review. Photodiagn. Photodyn. Ther. 2019, 25, 456–459. [Google Scholar] [CrossRef]
  22. Panhóca, V.H.; Carreira Geralde, M.; Corrêa, T.Q.; Carvalho, M.T.; Wesley, C.; Souza, O.; Bagnato, V.S. Enhancement of the Photodynamic Therapy Effect on Streptococcus Mutans Biofilm. J. Phys. Sci. Appl. 2014, 4, 107–114. [Google Scholar]
  23. Nie, M.; Deng, D.M.; Wu, Y.; de Oliveira, K.T.; Bagnato, V.S.; Crielaard, W.; de Souza Rastelli, A.N. Photodynamic inactivation mediated by methylene blue or chlorin e6 against Streptococcus mutans biofilm. Photodiagn. Photodyn. Ther. 2020, 31, 101817. [Google Scholar] [CrossRef]
  24. Romero, M.P.; Alves, F.; Stringasci, M.D.; Buzzá, H.H.; Ciol, H.; Inada, N.M.; Bagnato, V.S. One-Pot Microwave-Assisted Synthesis of Carbon Dots and in vivo and in vitro Antimicrobial Photodynamic Applications. Front. Microbiol. 2021, 12, 662149. [Google Scholar] [CrossRef] [PubMed]
  25. Blanco, K.C.; Inada, N.M.; Carbinatto, F.M.; Giusti, A.L.; Bagnato, V.S. Treatment of recurrent pharyngotonsillitis by photodynamic therapy. Photodiagn. Photodyn. Ther. 2017, 18, 138–139. [Google Scholar] [CrossRef] [PubMed]
  26. Soares, J.M.; Inada, N.M.; Bagnato, V.S.; Blanco, K.C. Evolution of surviving Streptoccocus pyogenes from pharyngotonsillitis patients submit to multiple cycles of antimicrobial photodynamic therapy. J. Photochem. Photobiol. B Biol. 2020, 210, 111985. [Google Scholar] [CrossRef]
  27. Geralde, M.C.; Leite, I.S.; Inada, N.M.; Salina, A.C.G.; Medeiros, A.I.; Kuebler, W.M.; Kurachi, C.; Bagnato, V.S. Pneumonia treatment by photodynamic therapy with extracorporeal illumination—An experimental model. Physiol. Rep. 2017, 5, e13190. [Google Scholar] [CrossRef] [PubMed]
  28. Leite, I.S.; Geralde, M.C.; Salina, A.C.G.; Medeiros, A.I.; Dovigo, L.N.; Bagnato, V.S.; Inada, N.M. Near–infrared photodynamic inactivation of S. pneumoniae and its interaction with RAW 264.7 macrophages. J. Biophotonics 2018, 11, e201600283. [Google Scholar] [CrossRef]
  29. Kassab, G.; Geralde, M.C.; Inada, N.M.; Achiles, A.E.; Guerra, V.G.; Bagnato, V.S. Nebulization as a tool for photosensitizer delivery to the respiratory tract. J. Biophotonics 2019, 12, e201800189. [Google Scholar] [CrossRef]
  30. Sousa, V.; Gomes, A.T.; Freitas, A.; Faustino, M.A.; Neves, M.G.; Almeida, A. Photodynamic inactivation of candida albicans in blood plasma and whole blood. Antibiotics 2019, 8, 221. [Google Scholar] [CrossRef] [Green Version]
  31. Spesia, M.B.; Rovera, M.; Durantini, E.N. Photodynamic inactivation of Escherichia coli and Streptococcus mitis by cationic zinc (II) phthalocyanines in media with blood derivatives. Eur. J. Med. Chem. 2010, 45, 2198–2205. [Google Scholar] [CrossRef]
  32. Dias, L.D.; Correa, T.Q.; Bagnato, V.S. Cooperative and competitive antimicrobial photodynamic effects induced by a combination of methylene blue and curcumin. Laser Phys. Lett. 2021, 18, 075601. [Google Scholar] [CrossRef]
  33. Pratavieira, S.; Uliana, M.P.; dos Santos Lopes, N.S.; Donatoni, M.C.; Linares, D.R.; de Freitas Anibal, F.; de Oliveira, K.T.; Kurachi, C.; de Souza, C.W.O. Photodynamic therapy with a new bacteriochlorin derivative: Characterization and in vitro studies. Photodiagn. Photodyn. Ther. 2021, 34, 102251. [Google Scholar] [CrossRef]
  34. Tonon, C.C.; Ashraf, S.; Alburquerque, J.Q.; Souza Rastelli, A.N.; Hasan, T.; Lyons, A.M.; Greer, A. Antimicrobial Photodynamic Inactivation Using Topical and Superhydrophobic Sensitizer Techniques: A Perspective from Diffusion in Biofilms. Photochem. Photobiol. 2021, 97, 1266–1277. [Google Scholar] [CrossRef] [PubMed]
  35. Pushalkar, S.; Ghosh, G.; Xu, Q.; Liu, Y.; Ghogare, A.A.; Atem, C.; Greer, A.; Saxena, D.; Lyons, A.M. Superhydrophobic Photosensitizers: Airborne 1 O 2 Killing of an in Vitro Oral Biofilm at the Plastron Interface. ACS Appl. Mater. Interfaces 2018, 10, 25819–25829. [Google Scholar] [CrossRef] [PubMed]
  36. Akobeng, A.K. Understanding randomised controlled trials. Arch. Dis. Child. 2005, 90, 840–844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Christodoulides, N.; Nikolidakis, D.; Chondros, P.; Becker, J.; Schwarz, F.; Rössler, R.; Sculean, A. Photodynamic Therapy as an Adjunct to Nonsurgical Periodontal Treatment: A Randomized, Controlled Clinical Trial. J. Periodontol. 2008, 79, 1638–1644. [Google Scholar] [CrossRef]
  38. Chondros, P.; Nikolidakis, D.; Christodoulides, N.; Rössler, R.; Gutknecht, N.; Sculean, A. Photodynamic therapy as adjunct to nonsurgical periodontal treatment in patients on periodontal maintenance: A randomized controlled clinical trial. Lasers Med. Sci. 2009, 24, 681–688. [Google Scholar] [CrossRef]
  39. Lulic, M.; Leiggener Görög, I.; Salvi, G.E.; Ramseier, C.A.; Mattheos, N.; Lang, N.P. One-year outcomes of repeated adjunctive photodynamic therapy during periodontal maintenance: A proof-of-principle randomized-controlled clinical trial. J. Clin. Periodontol. 2009, 36, 661–666. [Google Scholar] [CrossRef]
  40. Dai, T.; Huang, Y.Y.; Hamblin, M.R. Photodynamic therapy for localized infections-State of the art. Photodiagn. Photodyn. Ther. 2009, 6, 170–188. [Google Scholar] [CrossRef] [Green Version]
  41. Rühling, A.; Fanghänel, J.; Houshmand, M.; Kuhr, A.; Meisel, P.; Schwahn, C.; Kocher, T. Photodynamic therapy of persistent pockets in maintenance patients—A clinical study. Clin. Oral Investig. 2010, 14, 637–644. [Google Scholar] [CrossRef]
  42. Theodoro, L.H.; Silva, S.P.; Pires, J.R.; Soares, G.H.G.; Pontes, A.E.F.; Zuza, E.P.; Spolidório, D.M.P.; de Toledo, B.E.C.; Garcia, V.G. Clinical and microbiological effects of photodynamic therapy associated with nonsurgical periodontal treatment. A 6-month follow-up. Lasers Med. Sci. 2012, 27, 687–693. [Google Scholar] [CrossRef]
  43. Balata, M.L.; Andrade, L.P.D.; Santos, D.B.N.; Cavalcanti, A.N.; Tunes, U.D.R.; Ribeiro, E.D.P.; Bittencourt, S. Photodynamic therapy associated with full-mouth ultrasonic debridement in the treatment of severe chronic periodontitis: A randomized-controlled clinical trial. J. Appl. Oral Sci. 2013, 21, 208–214. [Google Scholar] [CrossRef]
  44. Mongardini, C.; Di Tanna, G.L.; Pilloni, A. Light-activated disinfection using a light-emitting diode lamp in the red spectrum: Clinical and microbiological short-term findings on periodontitis patients in maintenance. A randomized controlled split-mouth clinical trial. Lasers Med. Sci. 2014, 29, 1–8. [Google Scholar] [CrossRef] [PubMed]
  45. Macedo, G.D.O.; Novaes, A.B.; Souza, S.L.S.; Taba, M.; Palioto, D.B.; Grisi, M.F.M. Additional effects of aPDT on nonsurgical periodontal treatment with doxycycline in type II diabetes: A randomized, controlled clinical trial. Lasers Med. Sci. 2014, 29, 881–886. [Google Scholar] [CrossRef] [PubMed]
  46. Jung, G.U.; Kim, J.W.; Kim, S.J.; Pang, E.K. Effects of adjunctive daily phototherapy on chronic periodontitis: A randomized single-blind controlled trial. J. Periodontal Implant Sci. 2014, 44, 280–287. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Betsy, J.; Prasanth, C.S.; Baiju, K.V.; Prasanthila, J.; Subhash, N. Efficacy of antimicrobial photodynamic therapy in the management of chronic periodontitis: A randomized controlled clinical trial. J. Clin. Periodontol. 2014, 41, 573–581. [Google Scholar] [CrossRef] [PubMed]
  48. Carvalho, V.F.; Andrade, P.V.C.; Rodrigues, M.F.; Hirata, M.H.; Hirata, R.D.C.; Pannuti, C.M.; De Micheli, G.; Conde, M.C. Antimicrobial photodynamic effect to treat residual pockets in periodontal patients: A randomized controlled clinical trial. J. Clin. Periodontol. 2015, 42, 440–447. [Google Scholar] [CrossRef]
  49. Moreira, A.L.; Novaes, A.B.; Grisi, M.F.; Taba, M.; Souza, S.L.; Palioto, D.B.; de Oliveira, P.G.; Casati, M.Z.; Casarin, R.C.; Messora, M.R. Antimicrobial Photodynamic Therapy as an Adjunct to Nonsurgical Treatment of Aggressive Periodontitis: A Split-Mouth Randomized Controlled Trial. J. Periodontol. 2015, 86, 376–386. [Google Scholar] [CrossRef]
  50. Birang, R.; Shahaboui, M.; Kiani, S.; Shadmehr, E.; Naghsh, N. Effect of nonsurgical periodontal treatment combined with diode laser or photodynamic therapy on chronic periodontitis: A randomized controlled split-mouth clinical trial. J. Lasers Med. Sci. 2015, 6, 112–119. [Google Scholar] [CrossRef] [Green Version]
  51. Srikanth, K.; Chandra, R.V.; Reddy, A.A.; Reddy, B.H.; Reddy, C.; Naveen, A. Effect of a single session of antimicrobial photodynamic therapy using indocyanine green in the treatment of chronic periodontitis: A randomized controlled pilot trial. Quintessence Int. 2015, 46, 391–400. [Google Scholar] [CrossRef]
  52. Giannelli, M.; Formigli, L.; Lorenzini, L.; Bani, D. Efficacy of Combined Photoablative-Photodynamic Diode Laser Therapy Adjunctive to Scaling and Root Planing in Periodontitis: Randomized Split-Mouth Trial with 4-Year Follow-Up. Photomed. Laser Surg. 2015, 33, 473–480. [Google Scholar] [CrossRef]
  53. Pulikkotil, S.J.; Toh, C.G.; Mohandas, K.; Leong, K.V.G. Effect of photodynamic therapy adjunct to scaling and root planing in periodontitis patients: A randomized clinical trial. Aust. Dent. J. 2016, 61, 440–445. [Google Scholar] [CrossRef] [Green Version]
  54. Castro dos Santos, N.C.; Andere, N.M.R.B.; Araujo, C.F.; de Marco, A.C.; dos Santos, L.M.; Jardini, M.A.N.; Santamaria, M.P. Local adjunct effect of antimicrobial photodynamic therapy for the treatment of chronic periodontitis in type 2 diabetics: Split-mouth double-blind randomized controlled clinical trial. Lasers Med. Sci. 2016, 31, 1633–1640. [Google Scholar] [CrossRef] [PubMed]
  55. Talebi, M.; Taliee, R.; Mojahedi, M.; Meymandi, M.; Torshabi, M. Microbiological efficacy of photodynamic therapy as an adjunct to nonsurgical periodontal treatment: A clinical trial. J. Lasers Med. Sci. 2016, 7, 126–130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Martins, F.; Simões, A.; Oliveira, M.; Luiz, A.C.; Gallottini, M.; Pannuti, C. Efficacy of antimicrobial photodynamic therapy as an adjuvant in periodontal treatment in Down syndrome patients. Lasers Med. Sci. 2016, 31, 1977–1981. [Google Scholar] [CrossRef] [PubMed]
  57. Matarese, G.; Ramaglia, L.; Cicciu, M.; Cordasco, G.; Isola, G. The effects of diode laser therapy as an adjunct to scaling and root planing in the treatment of aggressive periodontitis: A 1-Year Randomized Controlled Clinical Trial. Photomed. Laser Surg. 2017, 35, 702–709. [Google Scholar] [CrossRef]
  58. Isola, G.; Matarese, G.; Williams, R.C.; Siciliano, V.I.; Alibrandi, A.; Cordasco, G.; Ramaglia, L. The effects of a desiccant agent in the treatment of chronic periodontitis: A randomized, controlled clinical trial. Clin. Oral Investig. 2018, 22, 791–800. [Google Scholar] [CrossRef]
  59. Cadore, U.B.; Reis, M.B.L.; Martins, S.H.L.; Invernici, M.D.M.; Novaes, A.B., Jr.; Taba, M., Jr.; Palioto, D.B.; Messora, M.R.; Souza, S.L.S. Multiple sessions of antimicrobial photodynamic therapy associated with surgical periodontal treatment in patients with chronic periodontitis. J. Periodontol. 2019, 90, 339–349. [Google Scholar] [CrossRef]
  60. Borekci, T.; Meseli, S.E.; Noyan, U.; Kuru, B.E.; Kuru, L. Efficacy of adjunctive photodynamic therapy in the treatment of generalized aggressive periodontitis: A randomized controlled clinical trial. Lasers Surg. Med. 2019, 51, 167–175. [Google Scholar] [CrossRef]
  61. Grzech-Leśniak, K.; Gaspirc, B.; Sculean, A. Clinical and microbiological effects of multiple applications of antibacterial photodynamic therapy in periodontal maintenance patients. A randomized controlled clinical study. Photodiagn. Photodyn. Ther. 2019, 27, 44–50. [Google Scholar] [CrossRef]
  62. Niazi, F.H.; Koppolu, P.; Tanvir, S.B.; Samran, A.; Alqerban, A. Clinical efficacy of photodynamic therapy in the treatment of necrotizing ulcerative periodontitis among HIV seropositive patients: A randomized controlled clinical trial. Photodiagn. Photodyn. Ther. 2020, 29, 101608. [Google Scholar] [CrossRef]
  63. Rosa, E.P.; Murakami-Malaquias-Silva, F.; Schalch, T.O.; Teixeira, D.B.; Horliana, R.F.; Tortamano, A.; Tortamano, I.P.; Buscariolo, I.A.; Longo, P.L.; Negreiros, R.M.; et al. Efficacy of photodynamic therapy and periodontal treatment in patients with gingivitis and fixed orthodontic appliances: Protocol of randomized, controlled, double-blind study. Medicine 2020, 99, e19429. [Google Scholar] [CrossRef]
  64. Al Nazeh, A.; Alshahrani, A.; Almoammar, S.; Kamran, M.A.; Togoo, R.A.; Alshahrani, I. Application of photodynamic therapy against periodontal bacteria in established gingivitis lesions in adolescent patients undergoing fixed orthodontic treatment. Photodiagn. Photodyn. Ther. 2020, 31, 101904. [Google Scholar] [CrossRef] [PubMed]
  65. Karmakar, S.; Prakash, S.; Jagadeson, M.; Namachivayam, A.; Das, D.; Sarkar, S. Clinico-microbiological efficacy of indocyanine green as a novel photosensitizer for photodynamic therapy among patients with chronic periodontitis: A split-mouth randomized controlled clinical trial. J. Pharm. Bioallied Sci. 2021, 13, 143. [Google Scholar] [CrossRef] [PubMed]
  66. Patyna, M.; Ehlers, V.; Bahlmann, B.; Kasaj, A. Effects of adjunctive light-activated disinfection and probiotics on clinical and microbiological parameters in periodontal treatment: A randomized, controlled, clinical pilot study. Clin. Oral Investig. 2021, 25, 3967–3975. [Google Scholar] [CrossRef] [PubMed]
  67. Llanos do Vale, K.; Ratto Tempestini Horliana, A.C.; Romero dos Santos, S.; Oppido Schalch, T.; Melo de Ana, A.; Agnelli Mesquita Ferrari, R.; Kalil Bussadori, S.; Porta Santos Fernandes, K. Treatment of halitosis with photodynamic therapy in older adults with complete dentures: A randomized, controlled, clinical trial. Photodiagn. Photodyn. Ther. 2021, 33, 102128. [Google Scholar] [CrossRef]
  68. Al-Momani, M.M. Indocyanine-mediated antimicrobial photodynamic therapy promotes superior clinical effects in stage III and grade C chronic periodontitis among controlled and uncontrolled diabetes mellitus: A randomized controlled clinical trial. Photodiagn. Photodyn. Ther. 2021, 35, 102379. [Google Scholar] [CrossRef]
  69. Lopes, R.G.; de Godoy, C.H.L.; Deana, A.M.; de Santi, M.E.S.O.; Prates, R.A.; França, C.M.; Fernandes, K.P.S.; Mesquita-Ferrari, R.A.; Bussadori, S.K. Photodynamic therapy as a novel treatment for halitosis in adolescents: Study protocol for a randomized controlled trial. Trials 2014, 15, 443. [Google Scholar] [CrossRef] [Green Version]
  70. Lopes, R.G.; da Mota, A.C.C.; Soares, C.; Tarzia, O.; Deana, A.M.; Prates, R.A.; França, C.M.; Fernandes, K.P.S.; Ferrari, R.A.M.; Bussadori, S.K. Immediate results of photodynamic therapy for the treatment of halitosis in adolescents: A randomized, controlled, clinical trial. Lasers Med. Sci. 2016, 31, 41–47. [Google Scholar] [CrossRef]
  71. Da Mota Ciarcia, A.C.C.; Gonçalves, M.L.L.; Horliana, A.C.R.T.; Suguimoto, E.S.A.; Araujo, L.; Laselva, A.; Mayer, M.P.A.; Motta, L.J.; Deana, A.M.; Mesquita-Ferrari, R.A.; et al. Action of antimicrobial photodynamic therapy with red leds in microorganisms related to halitose: Controlled and randomized clinical trial. Medicine 2019, 98, e13939. [Google Scholar] [CrossRef]
  72. Romero, S.D.S.; Schalch, T.O.; Do Vale, K.L.; Ando, E.S.; Mayer, M.P.A.; Feniar, J.P.G.; Fernandes, K.P.S.; Bussadori, S.K.; Motta, L.J.; Negreiros, R.M.; et al. Evaluation of halitosis in adult patients after treatment with photodynamic therapy associated with periodontal treatment: Protocol for a randomized, controlled, single-blinded trial with 3-month follow up. Medicine 2019, 98, e16976. [Google Scholar] [CrossRef]
  73. Gonçalves, M.L.L.; da Mota, A.C.C.; Deana, A.M.; de Souza Cavalcante, L.A.; Horliana, A.C.R.T.; Pavani, C.; Motta, L.J.; Fernandes, K.P.S.; Mesquita-Ferrari, R.A.; da Silva, D.F.T.; et al. Antimicrobial photodynamic therapy with Bixa orellana extract and blue LED in the reduction of halitosis—A randomized, controlled clinical trial. Photodiagn. Photodyn. Ther. 2020, 30, 101751. [Google Scholar] [CrossRef]
  74. Alshahrani, A.A.; Alhaizaey, A.; Kamran, M.A.; Alshahrani, I. Efficacy of antimicrobial photodynamic therapy against halitosis in adolescent patients undergoing orthodontic treatment. Photodiagn. Photodyn. Ther. 2020, 32, 102019. [Google Scholar] [CrossRef] [PubMed]
  75. Romero, S.S.; do Vale, K.L.; Remolina, V.G.; Silva, T.G.; Schalch, T.O.; Ramalho, K.M.; Negreiros, R.M.; Ando, E.S.; Mayer, M.P.A.; Mesquita Ferrari, R.A.; et al. Oral hygiene associated with antimicrobial photodynamic therapy or lingual scraper in the reduction of halitosis after 90 days follow up: A randomized, controlled, single-blinded trial. Photodiagn. Photodyn. Ther. 2021, 33, 102057. [Google Scholar] [CrossRef] [PubMed]
  76. Bassetti, M.; Schär, D.; Wicki, B.; Eick, S.; Ramseier, C.A.; Arweiler, N.B.; Sculean, A.; Salvi, G.E. Anti-infective therapy of peri-implantitis with adjunctive local drug delivery or photodynamic therapy: 12-month outcomes of a randomized controlled clinical trial. Clin. Oral Implants Res. 2014, 25, 279–287. [Google Scholar] [CrossRef] [PubMed]
  77. Rakašević, D.; Lazic, Z.; Rakonjac, B.; Soldatovic, I.; Jankovic, S.; Magic, M.; Aleksic, Z. Efficiency of photodynamic therapy in the treatment of peri-implantitis: A three-month randomized controlled clinical trial. Srp. Arh. Celok. Lek. 2016, 144, 478–484. [Google Scholar] [CrossRef] [Green Version]
  78. Karimi, M.R.; Hasani, A.; Khosroshahian, S. Efficacy of Antimicrobial Photodynamic Therapy as an Adjunctive to Mechanical Debridement in the Treatment of Peri-implant Diseases: A Randomized Controlled Clinical Trial. J. Lasers Med. Sci. 2016, 7, 139–145. [Google Scholar] [CrossRef] [Green Version]
  79. Ichinose-Tsuno, A.; Aoki, A.; Takeuchi, Y.; Kirikae, T.; Shimbo, T.; Lee, M.-C.; Yoshino, F.; Maruoka, Y.; Itoh, T.; Ishikawa, I.; et al. Antimicrobial photodynamic therapy suppresses dental plaque formation in healthy adults: A randomized controlled clinical trial. BMC Oral Health 2014, 14, 152. [Google Scholar] [CrossRef]
  80. Melo, M.A.S.; Rolim, J.P.M.L.; Passos, V.F.; Lima, R.A.; Zanin, I.C.J.; Codes, B.M.; Rocha, S.S.; Rodrigues, L.K.A. Photodynamic antimicrobial chemotherapy and ultraconservative caries removal linked for management of deep caries lesions. Photodiagn. Photodyn. Ther. 2015, 12, 581–586. [Google Scholar] [CrossRef] [Green Version]
  81. Asnaashari, M.; Ashraf, H.; Rahmati, A.; Amini, N. A comparison between effect of photodynamic therapy by LED and calcium hydroxide therapy for root canal disinfection against Enterococcus faecalis: A randomized controlled trial. Photodiagn. Photodyn. Ther. 2017, 17, 226–232. [Google Scholar] [CrossRef]
  82. Costa-Santos, L.; Silva-Júnior, Z.S.; Sfalcin, R.A.; da Mota, A.C.C.; Horliana, A.C.R.T.; Motta, L.J.; Mesquita-Ferrari, R.A.; Fernandes, K.P.S.; Prates, R.A.; Silva, D.F.T.; et al. The effect of antimicrobial photodynamic therapy on infected dentin in primary teeth: A randomized controlled clinical trial protocol. Medicine 2019, 98, e15110. [Google Scholar] [CrossRef]
  83. Okamoto, C.B.; Bussadori, S.K.; Prates, R.A.; da Mota, A.C.C.; Tempestini Horliana, A.C.R.; Fernandes, K.P.S.; Motta, L.J. Photodynamic therapy for endodontic treatment of primary teeth: A randomized controlled clinical trial. Photodiagn. Photodyn. Ther. 2020, 30, 101732. [Google Scholar] [CrossRef]
  84. Leite, D.P.V.; Paolillo, F.R.; Parmesano, T.N.; Fontana, C.R.; Bagnato, V.S. Effects of Photodynamic Therapy with Blue Light and Curcumin as Mouth Rinse for Oral Disinfection: A Randomized Controlled Trial. Photomed. Laser Surg. 2014, 32, 627–632. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Morley, S.; Griffiths, J.; Philips, G.; Moseley, H.; O’Grady, C.; Mellish, K.; Lankester, C.L.; Faris, B.; Young, R.J.; Brown, S.B.; et al. Phase IIa randomized, placebo-controlled study of antimicrobial photodynamic therapy in bacterially colonized, chronic leg ulcers and diabetic foot ulcers: A new approach to antimicrobial therapy. Br. J. Dermatol. 2013, 168, 617–624. [Google Scholar] [CrossRef] [PubMed]
  86. Williams, R.C. Periodontal Disease. N. Engl. J. Med. 1990, 322, 373–382. [Google Scholar] [CrossRef] [PubMed]
  87. Pihlstrom, B.L.; Michalowicz, B.S.; Johnson, N.W. Periodontal diseases. Lancet 2005, 366, 1809–1820. [Google Scholar] [CrossRef] [Green Version]
  88. Cortelli, J.R.; Barbosa, M.D.S.; Westphal, M.A. Halitosis: A review of associated factors and therapeutic approach. Braz. Oral Res. 2008, 22, 44–54. [Google Scholar] [CrossRef] [Green Version]
  89. Schwarz, F.; Derks, J.; Monje, A.; Wang, H. Peri-implantitis. J. Clin. Periodontol. 2018, 45, S246–S266. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Scheme of the antimicrobial photodynamic therapy (aPDT) components: interaction of PS, molecular oxygen, and light, causing bacterial death.
Figure 1. Scheme of the antimicrobial photodynamic therapy (aPDT) components: interaction of PS, molecular oxygen, and light, causing bacterial death.
Photonics 09 00340 g001
Figure 2. The percentage of targets in the papers reviewed showed periodontal disease as the most common application found in the studies, followed by halitosis, dental infection, peri-implantitis, oral decontamination, and skin ulcers, respectively.
Figure 2. The percentage of targets in the papers reviewed showed periodontal disease as the most common application found in the studies, followed by halitosis, dental infection, peri-implantitis, oral decontamination, and skin ulcers, respectively.
Photonics 09 00340 g002
Table 1. Randomized clinical trials that evaluated aPDT for the treatment of periodontal disease. SRP—scaling and root planning.
Table 1. Randomized clinical trials that evaluated aPDT for the treatment of periodontal disease. SRP—scaling and root planning.
Ref.Year of PublicationCountryNumber of PatientsTreatment SiteGoalClinical ConditionProtocolsLight ParametersPS, Concentration, and DLIFollow-UpMain Results
[37]2008Netherlands24Periodontal pocket and gingivaEvaluate the clinical and microbiological adjunctive uses of aPDT for nonsurgical periodontal treatment in chronic periodontitisChronic periodontitisControl Group (n = 12): SRP using hand instruments and sonic instrumentation; aPDT Group (n = 12)670 nm, 75 mW, for 1 minHELBO Blue Photosensitizer, (HELBO Photodynamic Systems, Austria) for 3 min3 and 6 months after treatmentA single aPDT session added to SRP failed to improve probing depth reduction and clinical attachment level gain, but it resulted in a greater reduction in bleeding scores compared with SRP alone
[38]2009Germany24Periodontal pocketEvaluate the clinical and microbiological effects of the adjunctive use of aPDT in nonsurgical periodontal treatmentPatients receiving supportive periodontal therapyRandomly treated with either subgingival SRP followed by a single episode of aPDT (test) or subgingival SRP alone (control)670 nm, power density of 75 mW/cm2Phenothiazine chloride (HELBO Blue Photosensitizer®, HELBO Photodynamic Systems)6 monthsThe additional application of a single episode of PDT to SRP failed to result in an additional improvement in terms of PPD reduction and CAL gain, but it resulted in a significantly higher reduction in bleeding scores than following SRP alone
[39]2009China10Periodontal pocketPossible added benefits of repeated adjunctive aPDT to conventional treatment of residual pockets in patients enrolled in periodontal maintenancePatients with residual pocketsTreatment randomly assigned 5 times in 2 weeks (Days 0, 1, 2, 7, 14) with aPDT (test) or nonactivated laser (control) following debridement670 nm, irradiance of 75 mW/cm2Phenothiazine chloride (HELBO Blue Photosensitizer, HELBO® Photodynamic Systems GmbH) for 3 min12 monthsRepeated (5 times) aPDT adjunctive to debridement yielded improved clinical outcomes in residual pockets in maintenance patients
[40]2009Austria58Periodontal sitesTo evaluate aPDT for its bactericidal potential and clinical effect in the treatment of periodontitisPeriodontitisGroup 1 (control group) was managed with an EMS Piezon Master 600 ultrasonic system (EMS Electro Medical Systems, Nyon, Switzerland);
Group 2 (laser group) was managed by aPDT in addition to ultrasonic treatment
680 nm, 75 mWMethylene blue at 0.005% for 3 min3 months after treatmentThe application of a single cycle of aPDT was not effective as an adjunct to ultrasonic periodontal treatment
[41]2010Germany60Periodontal pocketEvaluate whether aPDT can reduce probing depth in persistent periodontal pockets, change the microbial composition, and decrease the total load of subgingival bacteria more than conventional mechanical debridementChronic periodontitisControl Group (n = 29) treated with conventional ultrasonic debridement; aPDT Group (n = 25) treated with PS and light635 nm, 100 mW, for 1 minTolonium chloride (Asclepion-Meditec, UK) at 5% concentration for 30 s3 months after treatmentBoth therapies resulted in the same clinical effect; however, aPDT is less harmful to the teeth and microbial counts were reduced by about 30%–40%, but returned to baseline values after 3 months, irrespective of treatment
[42]2012Brazil33Periodontal sitesEvaluate the long-term clinical and microbiological effects of aPDT associated with nonsurgical periodontal
treatment
Chronic periodontitis(1) SRP group; (2) SRP and irrigation with toluidine blue O (TBO group); and (3) SRP, irrigation with TBO, and low level
laser irradiation (aPDT group)
660 nm, power 30 mW, spot size 0.07 cm2, energy 4.5 JToluidine blue O phenothiazine dye (100 μg/mL; Sigma Chemical Co., St Louis, MO), for 1 min180 daysaPDT as an adjunct to periodontal treatment produced statistically significant reductions in some of the key periodontal pathogens but produced no statistically significant benefit in terms of clinical outcome
[43]2013Brazil22Periodontal sitesTo evaluate an aPDT protocol as an adjunct to ultrasonic debridement in patients with severe chronic periodontitisChronic periodontitis(1) Patients were submitted to full-mouth ultrasonic debridement with an ultrasonic scaler; (2) aPDT protocol was carried out (one side of the mouth) with 0.005% methylene blue; (3) after 2 min, the light (660 nm) was applied660 nm, 100 mW, light dose = 320 J/cm20.005% methylene blue3 monthsaPDT and ultrasonic debridement showed good clinical improvements; however, aPDT did not provide any additional benefit when used with ultrasonic debridement
[44]2014Italy30Periodontal sitesTo study the potential adjunctive effect of microbiological/clinical photodynamic protocol using an LED lamp (red spectrum) and to compare it to SRPChronic periodontitis(1) Patients were treated with SRP; (2) treatment sites were allocated by a toss; (3) toluidine blue was applied and the location was illuminated by using an LED lamp (red spectrum—628 nm)628 nm, 2000 mW/cm2, light dose = 20 J/cm2Toluidine blue-Fotosan Agent®-(0.1 mg/mL) in 1% xanthan gel-A single protocol using an LED light system (red spectrum) and toluidine blue enhance short-term clinical and microbiological outcomes of the mechanical procedure
[45]2014Brazil30Periodontal sitesTo evaluate the aPDT combined with nonsurgical periodontal and doxycycline on clinical and metabolic effects in patients that show type 2 diabetes mellitusChronic periodontitisAll subjects of both groups (SRP and SRP + aPDT) were treated with SRP in combination with doxycycline (100 mg/day, for 2 weeks); (1) in the SRP + aPDT group, a diode laser (660 nm) for 1 min and phenothiazine chloride as PS were applied660 nm, 28 mW/cm2, light dose = 16.72 J/cm2Phenothiazine chloride (10 mg/mL)3 monthsaPDT (single use) did not show any clinical improvement as an adjunct to SRP but significantly reduced the glycated hemoglobin levels (HbA1c)
[46]2014Korea41Periodontal sitesTo elucidate clinical and antimicrobial effects of daily phototherapy (PT) as an adjunct to SRP in patients with chronic periodontitisChronic periodontitisAll participants underwent full-mouth SRP with periodontal curettes and an ultrasonic device: (1) SRP + PT group assigned electric toothbrushes with embedded LEDs (single frequency, 635 nm wavelength, 13 mW/cm2); (2) SRP group assigned electric toothbrushes without LEDs; irradiation time was 3 min per sessionSingle frequency, 635 nm, 13 mW/cm2, 3 min-4 weeksThe clinical parameters were improved in both groups; probing pocket depth (PPD) was significantly decreased in the SRP + PT group at the follow-up; furthermore, PPD and clinical attachment levels showed greater changes in the SRP + PT group than in the SRP group; no significant antimicrobial intergroup differences were noted
[47]2014India88Periodontal pocket and gingivaEvaluate whether adjunctive use of aPDT to SRP has any short-term effectiveness in chronic periodontitisChronic periodontitisGroup 1(n = 44) assigned SRP by hand scalers, universal curettes, and ultrasonic scaler; Group 2 (n = 44) assigned SRP according to Group 1 + aPDT655 nm, 60 mW/cm2, for 60 sMethylene blue (Sigma-Aldrich, St. Louis, MO, USA) at 10 mg/mL concentration for 3 min1, 3, and 6 months after treatmentA single application of this aPDT protocol was found to be effective in reducing gingival inflammation and probing pocket depth, evaluated over 6 months
[48]2015Brazil34Periodontal pocketEvaluate the clinical and microbiological effects of aPDT in the treatment of residual pockets of patients with chronic periodontitis subjected to supportive therapyChronic periodontitisControl group (n = 16) assigned saline solution; aPDT group (n = 18) assigned PS + light660 nm, 90 J/cm2, 40 mW, for 90 sMethylene blue (Chimiolux®, Hypofarma, Brazil) at 0.01% concentration for 5 min7 days, 3, 6, and 12 months after treatmentaPDT protocol used failed to demonstrate additional clinical and bacteriological benefits in residual pockets treatment
[49]2015Brazil20Periodontal sitesTo study the efficiency of
multiple sessions of aPDT in combination with SRP
versus SRP in patients that show AgP
Aggressive periodontitis(1) Patients received full-mouth supragingival scaling; (2) SRP was carried out; (3) phenothiazine chloride was added; (4) a diode soft-laser light (670 nm) was applied subgingivally670 nm, 0.25 W/cm2, light dose = 2.49 J/cm2Phenothiazine chloride (10 mg/mL)-The application of aPDT (4 sessions) as an adjunctive protocol to SRP, promotes additional microbiologic, clinical, and immunologic benefits
[50]2015Iran20Periodontal sitesTo evaluate the impact of adjunctive laser therapy (LT) and aPDT on patients with chronic periodontitis.Chronic periodontitisAll patients received SRP; (1) only SRP; (2) SRP with laser therapy—810 nm; (3) SRP + aPDT mediated by Emundo® mixtureStep 1—transgingival irradiation by bleaching handpiece (0.5 W, 10 s); Step 2—irradiation by a 300 µm
bare fiber in a circular pattern (0.5 W, 15 s); (c) Step 3—Granulation tissue removal using a 300 µm bare fiber (0.5 W, 25 s)
Emundo® mixture3 monthsAll groups showed improvements in terms of clinical attachment level (CAL) gain, periodontal pocket depth (PPD) reduction, papilla bleeding index, and microbial count compared with baseline; the results showed more significant improvement in the 6-week evaluation in terms of CAL in groups 2 and 3 than in group 1; group 2 also revealed a greater reduction in PPD than the other treatment modalities
[51]2015India60Periodontal sitesTo evaluate the effects of indocyanine green as an adjunct to nonsurgical periodontal therapy in terms of reduction in the percentage of viable bacteria and host tissue injuryPeriodontitisSRP group (only SRP); laser group—SRP and application of diode laser at 810 nm for 5 s;test group—indocyanine green (SRP) and application of diode laser beam at 810 nm in a continuous wave mode with 0.7 W output for 5 s along with 0.5 mL of 5 mg/mL ICG solution810 nm with 0.7 W output for 5 s0.5 mL indocyanine green, 5 mg/mL injected through a blunt end cannula till the pocket was overfilled6-month period after treatmentLaser-activated ICG dye may enhance the potential benefits of SRP and can be used as an adjunct to nonsurgical periodontal therapy
[52]2015Italy26Periodontal pocketReport the 4-year follow-up results of multiple aPDT cycles (PAPD) associated with SRP compared to sham treatment associated with SRP aloneChronic periodontitisControl group (n = 138 teeth)—sham + SRP; aPDT group (n = 138 teeth)—PAPD + SRPaPDT: 635 nm, 100 mW
Noncontact—gingival pocket external 11.6 W/cm2, 3.8 J/cm2 each passage
Photoablative 810 nm, 1 W
Contact—gingival pocket internal + external: 353.4 W/cm2, 66.7 J/cm2
Contact—gingival pocket internal: 35.3 W/cm2, 6.7 J/cm2 each passage
Methylene blue at 0.3% concentration for 5 min4 years (every 3 months during the 1st year and then every 6 months until the end)PAPD + SRP provided a significant and durable improvement compared with sham + SRP alone
[53]2016Malaysia20Periodontal sitesTo evaluate the efficacy of aPDT in reducing Aggregatibacter actinomycetemcomitans (Aa) in periodontitis patientsPeriodontitisConventional nonsurgical periodontal therapy (NSPT) was performed; in addition, the test side received adjunct aPDTA red LED lamp with a frequency of 628 Hz; gingiva and pocket were irradiated for 10 sMethylene blue, 1 min of DLI7 days, 1 and 3 monthsThere was a clinical improvement in 1 and 3 months compared with baseline, while the bleeding on probing was reduced only in the aPDT group in month 3; however, no difference in the quantification of Aa was detected between the
groups
[54]2016Brazil20Periodontal pocketsInvestigate the local effect of adjunct aPDT to ultrasonic periodontal debridement (UPD) and compare it to UD onlyModerate to severe generalized chronic periodontitis in type 2 diabetic patientsControl group (n = 20)—UPDT; test group (n = 20)—UPD + aPDT660 nm, 60 mW with irradiance of 2.15 W/cm2, total energy of 3.6 J and fluency of 129 J/cm20.005% methylene blue—Chimiolux DMC for 60 s180 daysAfter 180 days, there were statistically in the UPD group and the UPD + aPDT group; however, the intergroup analysis did not reveal statistically significant differences in any of the evaluated clinical parameters
[55]2016Iran18Periodontal pocketCompare the microbiologic effectiveness of the aPDT as an adjunctive treatment modality for nonsurgical treatment in chronic periodontitisModerate–severe chronic periodontitis, presence of at least 2 teeth with a pocket depth of 4–10 mm in each quadrant, gingival bleeding, and presence of at least 5 natural teeth in each quadrantFour quadrants were randomly treated by SRP, diode laser (810 nm wavelength, 1.5 W, and 320 μm fiber, contact, and sweeping technique), SRP + aPDT (with diode laser 808 nm, 0.5 W), and laser + SRP (with diode laser 808 nm, 1 W) in each patient808 nm, 0.2 W power-3 monthsaPDT was more effective as an adjunctive treatment to SRP than SRP alone; however, no distinct differences were found between both treatment modalities regarding the reduction in certain pathogen bacteria
[56]2016Brazil13Periodontal pocketEvaluate the efficacy of aPDT as an adjuvant to conventional periodontal treatment in down syndrome patientsDown syndrome patients who presented at least one tooth in each quadrant of the mouth with probing pocket depth equal to or greater than 5 mm were includedConventional treatment with SRP + a sham procedure and the experimental treatment SRP + aPDT660 nm, 120 J/cm2 divided into 4 points of 30 J/cm2 per tooth (1.2 J per point); the application time was 30 s/point with a spot size of 0.04 cm2Methylene blue 0.01% (Chimiolux®, DMC, São Carlos, SP, Brazil), for 4 min1 monthBoth types of periodontal treatment, with and without aPDT, were similarly effective and were associated with good clinical response
[57]2017Italy31Periodontal sitesTo further evaluate the effects of SRP + diode laser for the treatment of generalized aggressive periodontitis.Generalized aggressive periodontitisSRP + diode laser or SRP alone810 nm laser, set at 1 W in pulsating mode at 50 Hz, toff = 100 msec, ton = 100 msec, and an energy density of 24.84 J/cm2, with a 300 µm fiber optic delivery system-1 yearBoth treatments demonstrated an improvement in periodontal parameters at 1 year; however, SRP + diode laser produced a significant improvement in probing depth and in clinical attachment level; however, microbial and inflammatory mediator changes were not significantly reduced compared to SRP alone
[58]2018Italy36Periodontal sitesTo investigate and compare a desiccant agent as an adjunct to SRP versus SRP alone for the treatment of chronic periodontitisChronic periodontitis----No aPDT or light therapy was performed
[59]2019Brazil16Periodontal sitesTo evaluate the clinical effects and the subgingival microbiota after multiple sessions of aPDT associated with surgical treatment of severe chronic periodontitis (SCP)Chronic periodontitisAll participants underwent 4 sessions of full-mouth SRP: test group (TG)—multiple sessions of aPDT and surgical periodontal treatment (ST); control group (CG)—ST only, in a split-mouth designLaser diode, 660 nm, 60 mW/cm2, 0.6 J/cm2, 60 s per site10 mg/mL of phenothiazine chloride for 5 minBaseline (preintervention), 60 days (30 days after the end of nonsurgical therapy), and at 150 days (90 days after surgery)A reduction in probing depth was observed at 150 days for the TG, when compared with the CG; clinical attachment level gain was higher in the TG at 60 and 150 days; changes in the subgingival microbiota were similar between the groups, but the TG revealed a larger number of bacteria associated with periodontal disease at the end of the experiment
[60]2019Turkey24Periodontal sitesTo evaluate the microbiological and clinical effects of aPDT as an adjunctive tool to the nonsurgical periodontal protocol in patients that show aggressive periodontitis (AgP)Generalized aggressive periodontitis(1) SRP was applied to 12 subjects with ultrasonic; (2) toluidine blue was applied at the bottom of the periodontal pocket; (3) an LED source (625–635 nm) was inserted parallel to the root surface and the illumination was performed625–635 nm, 2000 mW/cm2, light dose = 20 J/cm2Toluidine blue O (0.1 mg/mL)-The use of the aPDT (two sessions) as an adjunct to SRP did not show superior to SRP regarding microbiological and clinical results
[61]2019Finland20Periodontal sitesTo evaluate clinically and microbiologically the outcomes following one single session of subgingival mechanical debridementPeriodontitis40 patients were randomly assigned 2 treatments: 1. SRP using ultrasonic and hand instruments followed by one single session of SRP followed by 1 x immediate application of aPDT and 2 x subsequent applications of aPDT without SRP (test); 2. SRP alone (control)635 nm, 117.64 J/mmToluidine blue 0.1% for 1 min6 months after treatmentEnhanced the clinical and microbiological outcomes compared with SRP alone
[62]2020Finland30Periodontal sitesTo evaluate clinical periodontal and microbiological parameters after the treatment with adjunctive antimicrobial aPDT among HIV-seropositive and -seronegative patients with necrotizing ulcerative periodontitisNecrotizing ulcerative periodontitisGroup I—provision of treatment through aPDT on the dorsum of tongue; group II—provision of treatment with the help of tongue scrappers (TS); group III—provision of treatment with the help of TS and adjunctive aPDT670 nm, 22 J/cm2Methylene blue (Helbo Blue photosensitizer) with 0.005%.3 and 6 months after treatmentaPDT was effective in improving clinical periodontal parameters and bacterial levels
[63]2020Brazil-Periodontal sitesTo evaluate the impact of photodynamic therapy (aPDT) as an adjuvant treatment in patients with gingivitis and fixed orthodontic appliancesGingivitis and fixed orthodontic appliances---21 daysNot performed
[64]2020Saudi Arabia22Periodontal sitesTo evaluate the effectiveness of aPDT as an adjunct to ultrasonic scaling (in the reduction in gingival inflammatory parameters and periodontal pathogensGingivitis lesionsUS group—patients receiving ultrasonic scaling (US) with usual oral hygiene in- instructions; aPDT group—in which patients received adjunctive aPDT with US670 nm, 22 J/cm2Methylene blue (0.0005%) photo- sensitizer (HELBO Blue) for 3 min6 months or 12 monthsaPDT was effective in significantly reducing periodontal pathogens in established gingivitis lesions
[65]2021India20Periodontal sitesTo determine the clinical and microbiological efficacy of aPDT using Indocyanine green (ICG) as a novel PS for the treatment of chronic periodontitisChronic periodontitisAll patients received full-mouth supragingival scaling; (1) SRP + aPDT mediated by ICG; (2) only SRPSoft-tissue diode laser unit (300 mW, 810 nm); each site was irradiated for 30 sICG tablet was suspended in distilled water at a concentration of 1 mg/mL, with a DLI of 2 min3 monthsSites additionally treated with ICG-mediated aPDT presented a statistically significant reduction in PD and CAL when compared with sites treated with only SRP after 3 months of treatment; adjunctive aPDT can be advocated as a treatment option for chronic periodontitis
[66]2021Germany48Periodontal sitesTo evaluate the microbiological and clinical effects of aPDT procedure alone or in combination with probiotics as an adjunct to nonsurgical periodontal treatmentChronic periodontitis(1) Subgingival mechanical debridement was carried out; (2) toluidine blue was applied in the periodontal pockets; (3) an LED device (628 nm) (2000–4000 mW/cm2) was applied subgingivally for 10 s at each side of the tooth628 nm, 2000–4000 mW/cm2, time of application = 10 sToluidine blue O, Fotosan Agent®, (0.1 mg/mL)3–6 monthsThe combined use of subgingival mechanical debridement, aPDT, and probiotics did not lead to significant improvements in the treatment of chronic periodontitis when compared to subgingival mechanical debridement plus aPDT and subgingival mechanical debridement alone
[67]2021Brazil62Periodontal sites and gingivaTo compare the effect of aPDT and tongue scraping (standard treatment) in older people with complete dentures diagnosed with halitosisPeriodontitisGroup I—provision of treatment through aPDT on the dorsum of tongue; group II—provision of treatment with the help of tongue scrappers (TS); group III—provision of treatment with the help of TS and adjunctive aPDT660 nm, 3183 J/cm2Methylene blue 0.005%, Brazil) for 5 min3 and 6 months after treatmentThe oral hygiene behavior associated with aPDT or tongue scraper was not able to reduce halitosis after a 90-day follow-up
[68]2021Saudi Arabia51Periodontal pocket and papillaEvaluate the efficacy of ICG/aPDT in the treatment of chronic periodontitis in terms of clinical, microbiological, and immune-inflammatory parameters in patients with well-controlled and poorly controlled forms of type-2 diabetes mellitus (T2DM)Chronic periodontitisSplit-mouth design—one site for control and the other for treatment (n = 17); control group—only root surface debridement (RSD); treatment group—ICG/aPDT + RSD
  • Controlled T2DM
  • Uncontrolled T2DM
  • Nondiabetic
810 nm, 200 mW, 4 JIndocyanine green (Sigma Aldrich, SA, St. Louis, MO, USA) at 0.5 mg/mL concentration for 30 s in the papilla and 10 s inside the periodontal pocket depth3 and 6 months after treatmentICG/aPDT improved clinical and antimicrobial parameters in well-controlled and poorly controlled T2DM; glycemic status did not interfere with the reduction in periodontal parameters in either type of T2DM
Table 2. Randomized clinical trials that evaluated aPDT for the treatment of halitosis.
Table 2. Randomized clinical trials that evaluated aPDT for the treatment of halitosis.
Ref.Year of PublicationCountryNumber of PatientsTreatment SiteGoalClinical ConditionProtocolsLight ParametersPS, Concentration, and DLIFollow-UpMain Results
[69]2014Brazil-TongueTo evaluate the antimicrobial effect of aPDT on halitosis in adolescentsHalitosis----Not performed
[70]2016Brazil45Tongue surfaceEvaluate the aPDT effect for halitosis in adolescents through the analysis of volatile sulfur compoundsHalitosis stemming from lingual bacteriaGroup 1 (n = 16)—aPDT; group 2 (n = 15)—tongue scraper; group 3 (n = 14)—tongue scraper and aPDT660.52 nm, 3537 mW/cm2 for 90 s/region; 6 regionsMethylene blue at 0.005% (165 µM) for 5 minImmediately after treatmentA novel option (Group 2) for the treatment of halitosis with an immediate effect without involving mechanical aggression of the lingual papillae
[71]2019Brazil39TongueTo evaluate the effectiveness of the application of aPDT in the tongue coating
as a new way to control halitosis
Halitosis(1) aPDT in 4 points of the tongue, E = 36 J, T = 90 s/point; (2) tongue scraper—10 scrapes in the tongue dorsum (Halitus); (3) tongue scraper—10 scrapes in the tongue dorsum and aPDT in 4 points, E = 36 J, T = 90 s/pointRed LED (660 nm) and tip of 2.84 cm2 in diameter; power of 400 mW, E = 36 J, T = 90 s/pointMethylene blue 0.005% (165 µM), 2 min of DLI7, 14, and 30 daysNot performed
[72]2019Brazil40TongueTo treat oral halitosis in healthy adults with aPDT, associated with periodontal treatmentHalitosisThe participants (n = 40) with halitosis will be randomized into 2 groups: G1—treatment with aPDT (n = 20); G2—cleaning of the tongue with a tongue scraper (n = 20)660 nm, 318 J/cm2Methylene blue, 0.005% for 3 min3 months after treatmentThis protocol determined the effectiveness of aPDT in the reduction in halitosis in adults
[73]2020Brazil44Six points on the back of the tongueEvaluate the reduction in halitosis using aPDT with Bixa orellana extract and blue LED, compare it to the tongue scraping, and verify the association of both treatmentsDiagnosis of sulfide (H2S) ≥ 112 ppb in gas chromatographyGroup 1 (n = 15)—aPDT with annatto and LED; group 2 (n = 14)—tongue scraping; group 3 (n = 15)—tongue scraping and aPDT395−480 nm for 20 s, 9.6 J per pointBixa orellana extract in spray at a concentration of 20% w/v for 2 min7 daysThere was an immediate reduction in halitosis, but the reduction was not maintained after 7 days
[74]2020Saudi Arabia45TongueTo evaluate the efficacy of aPDT on halitosis in adolescent patients undergoing fixed orthodontic treatmentHalitosisGroup I—provision of treatment through aPDT on the dorsum of tongue; group II—provision of treatment with the help of tongue scrappers (TS); group III—provision of treatment with the help of TS and adjunctive aPDT660 nm, 317.43 J/cm2 was kept 0.028 cmMethylene blue at 0.005% for 5 min2 weeks after treatmentaPDT along with tongue scraping showed effective immediate reduction in H2S concentration and reduction in oral pathogens
[75]2021Brazil40TongueVerify whether modification of oral hygiene behavior associated with aPDT or lingual scraper can reduce halitosis after a 90-day follow-upHalitosisSplit-mouth design—one site for control and the other for treatment (n = 17); control group—only root surface debridement (RSD); treatment group—ICG/aPDT + RSD; controlled T2DM; uncontrolled T2DM; nondiabetic660 nm, 318 J/cm2Methylene blue, 0.005%90 days after treatment.aPDT improved clinical and antimicrobial parameters in well-controlled and poorly controlled T2DM
Table 3. Randomized clinical trials that evaluated aPDT for the treatment of peri-implantitis.
Table 3. Randomized clinical trials that evaluated aPDT for the treatment of peri-implantitis.
Ref.Year of PublicationCountryNumber of PatientsTreatment SiteGoalClinical ConditionProtocolsLight ParametersPS, Concentration, and DLIFollow-UpMain Results
[76]2014Switzerland40Dental implantsTo compare the clinical, microbiological, and host-derived effects in the nonsurgical treatment of initial peri-implantitis with either adjunctive local drug delivery or adjunctive aPDT after 12 monthsInitial peri-implantitis(1) aPDT group—the dye phenothiazine chloride was used as PS (3 min), then the pockets were irrigated with 3% hydrogen peroxide and exposed to the laser light for 10 s and aPDT was repeated 1 week later; (2) one unit dosage of minocycline hydrochloride microspheres (1 mg)Hand-held diode laser, 660 nm, power density of 100 mW, for 10 sPhenothiazine chloride (3 min)3, 6, 9, and 12 months from baselineNonsurgical mechanical debridement with adjunctive aPDT was equally as effective in the reduction in mucosal inflammation as with adjunctive delivery of minocycline microspheres up to 12 months
[77]2016Serbia52Peri-implantitis sitesEvaluate early clinical and microbiological outcomes of peri-implantitis after surgical therapy with adjuvant aPDTDecontamination of the implant surfaceControl group used chlorhexidine gel (CHX) followed by saline irrigation;
study group used aPDT for decontamination of the implant surface
660 nm, 100 mW for 30 s/spotPhenothiazine chloride (HELBOR Blue Photosensitizer, bredent medical GmbH&Co. KG) was applied onto implant surface, bone, and peri-implant soft tissue, for 3 min3 monthsaPDT resulted in a significant decrease in bleeding on probing in comparison with CHX (p < 0.001) and showed significant decontamination of implant surfaces with complete elimination of anaerobic bacteria immediately after surgical procedure and three months later
[78]2016Iran1030 dental implantsAssess the clinical effects of aPDT after closed surface scaling in the treatment of peri-implant diseasesPeri-implant diseasesControl group (n = 15)—only closed-surface scaling; aPDT group (n = 15)—aPDT after closed-surface scaling630 nm, 2000 mW/cm2 for 120 sFotösan (CMS Dental, Denmark) for 3 min1.5 and 3 months after treatmentImprovement of clinical parameters, in the treatment of peri-implant diseases
Table 4. Randomized clinical trials that evaluated aPDT for the treatment of dental infections.
Table 4. Randomized clinical trials that evaluated aPDT for the treatment of dental infections.
Ref.Year of PublicationCountryNumber of PatientsTreatment SiteGoalClinical ConditionProtocolsLight ParametersPS, Concentration, and DLIFollow-UpMain Results
[79]2014Japan11Premolar surfacesInvestigate the inhibitory effects of aPDT in the oral cavity of healthy volunteersDental plaque depositionControl group (n = 11)—no treatment; aPDT group (n = 11)—PS + light660 nm, 1100 mW/cm2 for 20 s/surface of each toothToluidine blue ortho (Sigma-Aldrich, USA) at 1 mg/mL concentration for 10 sEvery day until 4 days after treatmentThe plaque formation on the aPDT group was inhibited after day 4 and the percentages of plaque deposition areas to total buccal and lingual tooth surfaces were significantly reduced compared with the control group
[80]2015Brazil4590 deep carious lesionsTest whether photochemistry-based treatment (PACT) reduces bacterial viability in remaining dentinDeep carious lesionsControl group (n = 45)—0.89% NaCl solution + light; experimental group PACT (n = 45)—PS + light630 nm, 150 mW, 94 J/cm2Toluidine blue ortho (Sigma, St. Louis, MO, USA) at 100 g/mL concentration for 5 minImmediately after treatmentPACT led to statistically significant reductions in mutans streptococci, Lactobacillus spp. and total viable bacteria compared with the control
[81]2017Iran20MolarsInvestigate the role of aPDT as a bactericidal agent in infected canals compared with calcium hydroxide therapyMolars requiring endodontic retreatmentGroup 1 (n = 10)—aPDT; group 2 (n = 10)—calcium hydroxide therapy635 nm, 200 mW/cm2 for 60 sToluidine blue (MDD, CMS Dental Denmark, Korea) at 0.1 mg/mL concentration for 5 minImmediately after treatmentaPDT presented better disinfectant performance than calcium hydroxide therapy
[82]2019Brazil32TeethTo evaluate the clinical effect of aPDT on infected dentin in dental caries lesions in primary teethInfection32 primary molars with deep occlusal dental caries will be selected and divided into 2 groups: G1—caries removal with a low-speed drill; G2—application of aPDT with PapacarieMBlue660 nm, 6 J, 60 sMethylene blue for 5 min12 months after treatmentAdding methylene blue dye to the formula of PapacarieMBlue might potentiate the antimicrobial action of aPDT and work more effectively on the infected dentin combined with a conservative, minimally invasive treatment
[83]2020Brazil30TeethTo evaluate the reduction in bacterial load following conventional endodontic treatment with and without antimicrobial aPDT in primary teethEndodontic treatment of primary teethGroup I—patients undergoing conventional root canal therapy (n = 15); group II—patients undergoing conventional root canal therapy combined with antimicrobial aPDT (n = 15)660 nm, J/cm2Methylene blue, 0.005% for 3 min3 months after treatmentThis study proved effective (aPDT) but presented the equal efficacious capability to conventional endodontic treatment alone
Table 5. Randomized clinical trials that evaluated aPDT for oral decontamination.
Table 5. Randomized clinical trials that evaluated aPDT for oral decontamination.
Ref.Year of PublicationCountryNumber of PatientsTreatment SiteGoalClinical ConditionProtocolsLight ParametersPS, Concentration, and DLIFollow-UpMain Results
[84]2014Brazil27Oral cavityEvaluate the effects of the aPDT with blue light and curcumin on oral disinfection for 2 h after treatmentOral cavity decontaminationLight group (n = 9)—only light; Curcumin group (n = 9); aPDT group (n = 9)455 nm, 300 mW/cm2, 5 min, 200 J/cm2Curcumin (aPDT Pharma, Brazil) at 30 mg/L concentration for 5 minImmediately, 1 h, and 2 h after treatmentCurcumin has the potential to disaggregate oral plaque; aPDT protocol may be used for the reduction in salivary microorganisms to overall mouth disinfection before intraoral surgical procedures
Table 6. Randomized clinical trials that evaluated aPDT for the treatment of ulcers on the skin.
Table 6. Randomized clinical trials that evaluated aPDT for the treatment of ulcers on the skin.
Ref.Year of PublicationCountryNumber of PatientsTreatment SiteGoalClinical ConditionProtocolsLight ParametersPS, Concentration, and DLIFollow-UpMain Results
[85]2013Scotland32Legs and footTo determine whether aPDT in bacterially colonized chronic leg ulcers and chronic diabetic foot ulcers can reduce bacterial load, and potentially lead to accelerated wound healingChronic leg and diabetic foot ulcersAll patients (cationic photosensitizer-PPA); G1—placebo; G2—patients with leg ulcer PPA904; G3—patients with diabetic foot ulcer PPA904570–670 nm at a total dose of 50 J/cm2.[3,7-bis (N,N-dibutylamino) pheno- thiazin-5-ium bromide] for 15 min; 500 μmol/L3 months after treatmentThis first controlled study of aPDT in chronic wounds demonstrated a significant reduction in bacterial load
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Alves, F.; Stringasci, M.D.; Requena, M.B.; Blanco, K.C.; Dias, L.D.; Corrêa, T.Q.; Bagnato, V.S. Randomized and Controlled Clinical Studies on Antibacterial Photodynamic Therapy: An Overview. Photonics 2022, 9, 340. https://doi.org/10.3390/photonics9050340

AMA Style

Alves F, Stringasci MD, Requena MB, Blanco KC, Dias LD, Corrêa TQ, Bagnato VS. Randomized and Controlled Clinical Studies on Antibacterial Photodynamic Therapy: An Overview. Photonics. 2022; 9(5):340. https://doi.org/10.3390/photonics9050340

Chicago/Turabian Style

Alves, Fernanda, Mirian D. Stringasci, Michelle B. Requena, Kate C. Blanco, Lucas D. Dias, Thaila Q. Corrêa, and Vanderlei S. Bagnato. 2022. "Randomized and Controlled Clinical Studies on Antibacterial Photodynamic Therapy: An Overview" Photonics 9, no. 5: 340. https://doi.org/10.3390/photonics9050340

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop