Next Article in Journal
Phosphatidic Acid in Plant Hormonal Signaling: From Target Proteins to Membrane Conformations
Previous Article in Journal
Repositioning the Role of Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL) on the TRAIL to the Development of Diabetes Mellitus: An Update of Experimental and Clinical Evidence
Previous Article in Special Issue
Systematical Screening of Intracellular Protein Targets of Polyphemusin-I Using Escherichia coli Proteome Microarrays
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Use of Photodynamic Therapy Associated with Antimicrobial Peptides for Bacterial Control: A Systematic Review and Meta-Analysis

by
Luana Mendonça Dias
1,
Túlio Morandin Ferrisse
1,
Karine Sousa Medeiros
1,
Eduardo Maffud Cilli
2 and
Ana Claudia Pavarina
1,*
1
Laboratory of Applied Microbiology, Department of Dental Materials and Prosthodontics, School of Dentistry, São Paulo State University (UNESP), Araraquara 14801-903, Brazil
2
Department of Biochemistry and Organic Chemistry, Institute of Chemistry, São Paulo State University (UNESP), Araraquara 14800-900, Brazil
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(6), 3226; https://doi.org/10.3390/ijms23063226
Submission received: 31 January 2022 / Revised: 2 March 2022 / Accepted: 9 March 2022 / Published: 17 March 2022
(This article belongs to the Special Issue Antimicrobial Peptides and Antimicrobial Chemokines)

Abstract

:
Considering the challenges related to antimicrobial resistance, other strategies for controlling infections have been suggested, such as antimicrobial photodynamic therapy (aPDT) and antimicrobial peptides (AMP). This study aims to perform a systematic review and meta-analysis to obtain evidence on the antimicrobial effectiveness of aPDT associated with AMP and establish in vitro knowledge on this topic for further study designs. The PubMed, Scopus, Web of Science, Science Direct, Scielo, and Cochrane Library databases were searched. Two independent and calibrated researchers (Kappa = 0.88) performed all the systematic steps according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA). The odds ratio (OR) was used as the effect measure. The Peto method was used to perform the meta-analysis due to the sparse data. Twenty studies were included in the present review. The result was significant (OR = 0.14/p = 0.0235/I-squared = 0%), showing better outcomes of aPDT associated with peptides than those of aPDT alone for controlling the microbial load. Only 20% of the studies included evaluated this approach in a biofilm culture. Combined treatment with aPDT and AMP highly increased the ability of microbial reduction of Gram-positive and Gram-negative bacteria. However, additional blind studies are required to evaluate the efficacy of this therapy on microbial biofilms.

1. Introduction

Antimicrobial resistance to conventional drugs has resulted in high global rates of recurrent invasive infections, facilitating disease progression and reducing the likelihood of effective treatments [1]. In 2020, the World Health Organization warned about the appearance of strains increasingly resistant and difficult to control. The indiscriminate use of antimicrobial drugs is facilitated by inadequate medical prescriptions and substandard medications [2].
Considering the challenges related to antimicrobial resistance, other strategies for controlling infections have been suggested [3,4,5,6]. Antimicrobial photodynamic therapy (aPDT) has been used to inactivate microorganisms and treat infections [3,4,5,6]. aPDT involves the application of a photosensitizing agent (PS), an LED source corresponding to the absorption band of the PS, and the presence of oxygen. This therapy has several advantages in the treatment of infections from microorganisms, such as the wide spectrum of action and a low mutagenic potential in exposed cells [6].
When comparing aPDT with other therapies, it has the advantage of local PS application, restricting the treatment to the area of interest, thus preventing systemic side effects. There is also an immediate onset of action and elimination of virulence factors secreted by resistant microorganisms [7]. Lastly, the literature did not report the development of bacteria and fungi resistance to aPDT [4,8].
Studies have shown that microbial biofilms reduce the susceptibility to aPDT compared to planktonic cultures [4]. Considering the protection endowed by the extracellular matrix (ECM), it is difficult for the PS to penetrate the deeper layers of the microbial biofilm, impairing aPDT activity [9]. To overcome this limitation, aPDT associated with enzymes or antifungal agents was more effective for microbial inactivation than aPDT alone [5,9]. Additionally, antimicrobial peptides (AMP) have been used alone [10,11], combined with aPDT [12,13], or by conjugating a PS to the AMP molecule [14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31], presenting satisfactory results in pathogenic microorganism inactivation.
AMP are molecules expressed by all living organisms and responsible for the innate defense system against pathogen infection, including viruses, bacteria, fungi, and parasites [32]. AMP are oligopeptides with up to 50 amino acids with a broad spectrum of action against microorganisms [33,34]. This new class of compounds has boosted science for new methodologies for synthesizing, isolating, purifying, analyzing, and quantifying peptides [35]. The presence of cationic residues (Arg and Lys) in AMP promotes a positive liquid charge for this structure, resulting in the interaction with the negative cell membrane of the target organism, such as bacteria [35]. Another important aspect of the construction of the AMP amphipathic structure is the high fraction of hydrophobic amino acids (>50%) [36], which is vital for membrane penetration. The biological activity of AMP is closely related to their structure, and these could be classified as α-helix, β-sheet, extended peptides, and both α-helix and β-sheet peptides [37], with the first two appearing more frequently [38]. Although the molecular target of some peptides is inside the cell, as non-membrane disruptive AMP [39], most peptides interact with the anionic components of the membranes of microorganisms and damage this structure [32].
The literature has described the association of AMP and aPDT to explore the best properties of both treatments, increasing the effectiveness and decreasing the time of application [12,13]. AMP can form pores in cell membranes and present biofilm activity [11], which leads to the penetration of the PS into the membrane, facilitating the inactivation of structures through LED photoexcitation [12]. Other advantages of association treatments are reduced effective dose, minimized toxicity potential, and reduced treatment costs [12,40].
To elucidate the antimicrobial efficacy of aPDT associated with AMP, this study performed a systematic review and a meta-analysis by searching the existing literature. The data synthesis provided in the present study establishes in vitro knowledge on this topic for different study designs.

2. Results

2.1. Search Results

The flowchart in Figure 1 shows the process of article selection. The preliminary electronic search yielded 213 articles. After excluding duplicates, 36 studies remained. The titles and abstracts were read, and no article was excluded. After evaluating the abstracts, 25 studies were considered for a full-text evaluation. Then, 5 articles were excluded because they did not report details about the predetermined microbiological assays.

2.2. Synthesis of Results

The results of the systematic review show that all articles had an in vitro experimental design and 3 of them were both in vitro and in vivo experimental studies [28,30,31]. Moreover, of the 20 articles analyzed, 18 performed the therapy with a portion of the PS redirected to AMP and only 2 studies performed the therapy combined with AMP [12,13]. The shortest and longest irradiation times were 30 s [14] and 20 h [21,22], respectively. The most commonly used PS were chlorin e6 [12,13,24,28,30,31] and porphyrin [14,16,17,19,21,22,25,26]. Additionally, the most frequently used microorganism in the assay was Staphylococcus aureus [12,14,15,17,18,19,20,21,22,24,26,27,29,30,31], followed by Escherichia coli [12,14,16,17,18,19,20,21,22,26,27,30,31]. Most of the studies analyzed evaluated the microorganisms in suspension (planktonic culture) and only 4 evaluated the therapy in a biofilm culture [12,24,28,31] (Table 1).

2.3. Risk of Bias Assessments for In Vitro Studies

The criteria from the OHAT Rob tool were applied to all articles included in the systematic review. The most frequent biases regarded blinding procedures. Moreover, the problem with internal validity was the lack of methodological details in the statical analyses and the performance of treatments only in microorganism suspensions (Table 2).

2.4. Meta-Analysis

The meta-analysis was performed only in 3 studies [13,15,23]. The reduced number of studies included in the quantitative analysis is due to the lack of data (e.g., sample size) and the absence of a study group evaluating only aPDT application. The experimental group included microorganisms treated with aPDT associated with peptides (aPDT + AMP), while the control group included microorganisms treated only with aPDT (aPDT). The microbial load was the outcome evaluated in the meta-analysis.
The Peto method was used to perform the meta-analysis due to the sparse data. The results were transformed into odds, and, therefore, the odds ratio (OR) was used as the effect measure. The result was significant (OR = 0.14/p = 0.0235/I-squared = 0%), showing better outcomes for aPDT associated with peptides than those for aPDT alone for controlling the microbial load (Figure 2A). Moreover, small-study effects in the meta-analysis and consequently publication and meta-analysis biases were verified with the trim-and-fill method. However, there were no biases (Figure 2B).

3. Discussion

The exposure of bacteria to conventional antimicrobial agents often leads to a selection of strains that are more resistant to many of these drugs [1,41]. To inactivate the microorganism and overcome the progress of the infection, alternative strategies may be suggested, such as using aPDT and AMP [12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31]. This study performed a systematic review and a meta-analysis to elucidate the antimicrobial efficacy of aPDT associated with AMP by searching the existing literature. To achieve better results for antimicrobial inactivation, all variable situations of these two treatments require a precise control [6,7]. For instance, irradiation time and type of photosensitizer are among the main variables associated with the treatment success of aPDT [7]. Moreover, in the clinical application of this therapy, success is determined based on infection remission and consequently the restoration of site function [5,7]. Additionally, in AMP treatments, peptide size, positive charge, conformation, and stability are characteristics related to antimicrobial achievement [35]. The association of AMP with aPDT has been described in the literature to explore the best properties of both treatments, increasing the effectiveness against microorganisms.
To improve the efficacy of aPDT in reducing the microbial load, the N-terminal of AMP has been chosen to conjugate with the photosensitizer molecule [14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31]. This approach may increase water solubility and facilitate the penetration of the component into bacterial membranes through pore formation [26]. Furthermore, AMP-like compounds represent a promising alternative to broad-spectrum antibiofilm agents, with synergistic activities against persistent infections caused especially by biofilm formation [35]. Among the studies evaluated, 80% used the conjugated therapy approach [14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31].
aPDT and AMP may also be associated by combining individual treatments. Two studies included in the present systematic review investigated this approach [12,13]. The success of the combined therapy can be attributed to the possibility of targeting different cell compartments to increase the damage to target cells. It is also possible to extend the spectrum of action of the therapeutic response [42]. Additionally, the meta-analysis results strengthen the findings supporting the combined therapy (aPDT + AMPs) when compared with a single treatment (aPDT). Combining two or more antimicrobial therapies with different action mechanisms can decrease therapeutic failure due to the reduced likelihood of microorganisms presenting antimicrobial resistance and tolerance to both treatments simultaneously [43,44].
The association of aPDT and AMPs resulted in microbial load reduction by 100% for Staphylococcus aureus [12,15,26,30], Pseudomonas aeruginosa [28], and Enterococcus faecium [12]. A high rate of microbial reduction (>90%) was also found for Acinetobacter baumannii [12,20], Escherichia coli [16,18,20,27,30], Staphylococcus epidermidis [18], and Enterococcus faecalis [13]. Lastly, microbial reductions between 50% and 90% were found for Mycobacterium smegmatis [25], Salmonella enteric [25], and Klebsiella pneumoniae [25]. S. aureus, followed by E. coli, were the bacteria most frequently evaluated in the articles included. This preference can be attributed to the pathogenicity and consequently a high degree of infection of these microorganisms. These microorganisms are also recognized as a major threat to human and animal health [30,31]. For instance, E. coli is known to inhabit mainly the lower intestinal tract of humans, causing intestinal and urinary infections. In addition, the increased pathogenicity of E. coli can affect the central nervous system of the host, causing inflammation and meninges [45]. Gram-positive S. aureus bacteria are associated with persistent nosocomial colonization in up to 25% of the healthy adult population, potentially causing bacteremia and subcultural abscesses [46]. Studies have shown that persistent chronic infections caused by S. aureus are related to bacterial growth in a biofilm model and may be fixed in bone and heart valves or implanted materials [47].
A biofilm is defined as a sessile microbial community with cells adhered to a surface and incorporated by a polymeric extracellular matrix (ECM) [48]. The ECM composition varies among strains and may contain host factors, polysaccharides, proteins, and extracellular DNA (eDNA) [48,49,50]. These components provide immune protection and antibiotic resistance and tolerance to microorganisms growing inside the biofilm [51]. Reproducing this culture is relevant because of the potential microbial growth in any humid biotic and abiotic surface [52], which makes it more representative than suspension cultures. Nevertheless, among the articles evaluated in the present study, only 20% performed the research in a biofilm culture. This is because suspension cultures are commonly used in preliminary studies, selecting the better approaches for antimicrobial treatment.
The AMPs presented different action mechanisms against planktonic cells and biofilms. The main action mechanisms in planktonic cultures are cytoplasmic membrane effects, cell envelope targets, and intracellular targets [36]. Most of the studies included in the present systematic review used AMPs with membrane effects [12,13,14,15,16,17,18,19,20,21,23,24,26,27,28,29,30,31]. This preference occurs because PS entry is facilitated after the disruption of the membrane surface of microorganism cells. Additionally, the cationic charge in the PS + AMP association increases the possibility of high PS concentration available to enter the cells [36]. Only two studies evaluated AMPs related to cell envelope targets [22,25]. All articles that evaluated the efficacy of AMPs on biofilms in their study design used AMPs with cytoplasmic membrane effects (maturation phase of a biofilm) [13,24,28,31]. In contrast, the scientific literature describes three additional action mechanisms against biofilms, namely the block attachment of cells (attachment phase of a biofilm), matrix disruption (biofilm development), and cell dispersal (dispersal phase of a biofilm) [36]. It has been suggested that increasing membrane permeability would be the most suitable action mechanism of AMPs against biofilm formation. This would facilitate the entry of antibiotics into cells and access to specific intracellular targets [53].
In some cases, AMPs did not present good anti-biofilm responses because it was difficult to inhibit biofilm formation. This can be explained by the minimization of the process related to microorganism adhesion and differences in the AMP amino acid sequence in peptides [13]. The literature reports that a microbial biofilm is more difficult to eliminate with aPDT than planktonic cultures [4]. The extracellular matrix may have an important part in protecting a biofilm against aPDT [3]. This structure complicates the penetration of PS in the existing multilayer of this model, which consequently interferes with the production of ROS and cellular lysis [54]. To overcome this limitation, the association of aPDT with AMPs should be further investigated in biofilm cultures.
In the present systematic review, the most frequently used PS were chlorin e6 [12,13,28,30,31], followed by porphyrins [14,16,17,19,20,21,22,25]. First-generation PS (porphyrin derivatives) have been replaced with second-generation PS (chlorins and phthalocyanines). Chlorins are reduced hydrophilic porphyrins with a strong absorption band in the red region of the spectrum (540–700 nm), resulting in a high therapeutic response even at lower PS concentrations [55]. Considering the action of chlorin in this absorption band, the light penetrates deeper into the tissue [56]. Additional advantages of using chlorins are the shorter photosensitization period, higher quantum yield in singlet oxygen production, and more favorable light absorption characteristics than first-generation PS [55].
The therapeutic light doses of the studies analyzed ranged from 390 to 750 nm. The optimal condition for the photodynamic therapy is the length of light ranging from 400 to 800 nm, known as the therapeutic window, in which there is maximum tissue light transmittance, representing an advantage for the treatment of infections. Above 800 nm, radiation is absorbed by water, which restricts the wavelength to this upper limit [57]. Wavelength radiation lower than 400 nm undergoes greater scattering. The presence of endogenous chromophores in the tissues, which absorb at shorter wavelengths, mainly hemoglobin, reduces light penetration [58].
All articles included in the systematic review have failed to use blinding. This approach is important because it can eliminate biases related to effect size estimates. Thus, the magnitude of the effect remains accurate, the observational bias may be eliminated, and consequently, the results will be more reliable [59]. For potential threats related to internal validity, details on the statistical approaches were considered, but all articles have also failed in this item. Typically, power analysis and sample size estimations are crucial points for rejecting and accepting the null hypothesis [60]. Data normality and homoscedasticity verification are essential steps for making a correct inference [61]. In the present systematic review, only three articles were included in the meta-analysis. Therefore, the results should be interpreted with caution. The main reason for such a small number of studies meta-analyzed was the absence of sample size data reported in the articles.
In short, further studies should be developed evaluating the association between AMPs and aPDT against microorganisms in a biofilm, in addition to blind studies and using AMPs with different action mechanisms. Moreover, only bacterial species were evaluated in the studies analyzed. Therefore, further research using this therapeutic approach against other microorganisms (e.g., fungal species, parasites, and viruses) would be highly recommended.

4. Materials and Methods

4.1. Protocol and Registration

The present systematic review was performed according to the Preferred Reporting Items for Systematic Reviews (PRISMA) statement [62]. The present study was registered in the Open Science Framework (OSF) (registration doi:10.17605/OSF.IO/2BWDH).

4.2. Data Extraction and Research Question

The research question was based on the PICO strategy for systematic exploratory reviews, where P = microorganism, I = aPDT combined with antimicrobial peptides (dual therapy) or aPDT conjugated with peptides, C = isolated therapy (aPDT), and O = reduction in microbial load. The present study aimed to answer the following focused questions: “Does the association between aPDT and AMPs increase the effectiveness of the therapy in reducing the microbial load”? Further data on the name of the first author, the date of publication, study design, peptides used, the sample size, the photosensitizer, the wavelength, the irradiation time, and the microorganism evaluated were extracted from the articles included in this systematic review according to the eligibility criteria.

4.3. Eligibility Criteria

The inclusion criteria for this systematic review were the use of aPDT associated with AMPs to reduce the microbial load, including the combined therapy, or PS conjugated with peptides. There were no restrictions on study design (e.g., inclusion of in vitro and in vivo studies, observational human studies, and randomized clinical trials), language, and microorganisms. The exclusion criteria were review articles, case reports, other modalities of treatment using AMPs, and aPDT combined with other modalities of treatment.

4.4. Search Strategy

Two independent examiners (L.M.D and T.M.F) were calibrated to select the articles. Thus, the independent examiners conducted an electronic search in PubMed, Web of Science, Scopus, Scielo, Lilacs, and Cochrane Library databases. The search terms were “antimicrobial peptide” and “antimicrobial photodynamic therapy”. A manual search was also performed in other relevant journals in the field of photodynamic therapy and at ClinicalTrials.gov. Based on the titles and abstracts, the same two independent examiners selected and classified the articles as included in or excluded from the review (Kappa score = 0.88). The Rayyan for Systematic Reviews™ software was used to delete duplicate articles [63]. The data were extracted from the articles selected after concluding the eligibility step (Kappa score = 0.87). The studies were analyzed and discussed. Any disagreement during the process was solved by reaching a consensus before proceeding to the next steps.

4.5. Meta-Analysis and Quantitative Approaches

The meta-analysis was performed with the R software (version 3.6.3) at α = 0.05. The viability of microbial cells (frequency of positive cells) was the outcome used in the meta-analysis. The experimental group included aPDT associated with AMPs (aPDT + AMPs), while the control group included only aPDT application (aPDT). The random-effects model and the Peto method (presence of sparse data) were used to perform the meta-analysis. The odds ratio was the effect measure selected to perform the quantitative analysis of the binary outcome. To detect the publication bias related to the small-study effect in the meta-analysis, the trim-and-fill method was performed. A high level of heterogeneity was considered for I-squared > 50%.

5. Conclusions

Combined treatment with aPDT and AMPs is effective because it increases the ability of microbial reduction for Gram-positive and Gram-negative bacteria. However, additional blind studies are required to evaluate the efficiency of the association between AMPs and aPDT against microorganisms in a biofilm, in addition to blind studies and using AMPs with different action mechanisms. Therefore, further research using this therapeutic approach against other microorganisms would be highly recommended.

Author Contributions

Conceptualization, L.M.D., T.M.F. and A.C.P.; methodology, L.M.D. and T.M.F.; software, T.M.F.; validation, L.M.D., T.M.F. and K.S.M.; formal analysis, L.M.D. and T.M.F.; investigation, L.M.D. and T.M.F.; resources, L.M.D.; data curation, L.M.D.; writing the original draft, L.M.D., T.M.F. and K.S.M.; writing, review, and editing, L.M.D., T.M.F., K.S.M. and A.C.P.; visualization, L.M.D.; supervision, A.C.P. and E.M.C.; project administration, A.C.P. and E.M.C.; funding acquisition, A.C.P. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the São Paulo Research Foundation [Fundação de Amparo à Pesquisa do Estado de São Paulo FAPESP # 2013/07276-1 (CePID CePOF)] and FAPESP# 2020/16227-8 to L.M.D.; FAPESP# 2021/06476-3 to K.S.M. and FAPESP# 2021/01191-0 to T.M.F.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Christaki, E.; Marcou, M.; Tofarides, A. Antimicrobial Resistance in Bacteria: Mechanisms, Evolution, and Persistence. J. Mol. Evol. 2020, 88, 26–40. [Google Scholar] [CrossRef] [PubMed]
  2. Watkins, R.R.; Bonomo, R.A. Overview: Global and Local Impact of Antibiotic Resistance. Infect. Dis. Clin. N. Am. 2016, 30, 313–322. [Google Scholar] [CrossRef] [PubMed]
  3. Garcia, B.A.; Panariello, B.H.D.; de Freitas-Pontes, K.M.; Duarte, S. Candida biofilm matrix as a resistance mechanism against photodynamic therapy. Photodiagnosis Photodyn. Ther. 2021, 36, 102525. [Google Scholar] [CrossRef]
  4. Dias, L.M.; Klein, M.I.; Jordão, C.C.; Carmello, J.C.; Bellini, A.; Pavarina, A.C. Successive applications of Antimicrobial Photodynamic Therapy effects the susceptibility of Candida albicans grown in medium with or without fluconazole. Photodiagnosis Photodyn. Ther. 2020, 32, 102018. [Google Scholar] [CrossRef]
  5. Janeth Rimachi Hidalgo, K.; Carmello, J.C.; Carolina Jordão, C.; Aboud Barbugli, P.; de Sousa Costa, C.A.; Mima, E.; Pavarina, A.C. Antimicrobial Photodynamic Therapy in Combination with Nystatin in the Treatment of Experimental Oral Candidiasis In-duced by Candida albicans Resistant to Fluconazole. Pharmaceuticals 2019, 12, 140. [Google Scholar] [CrossRef] [Green Version]
  6. Gilaberte, Y.; Rezusta, A.; Juarranz, A.; Hamblin, M.R. Editorial: Antimicrobial Photodynamic Therapy: A New Paradigm in the Fight against Infections. Front. Med. 2021, 8, 788888. [Google Scholar] [CrossRef] [PubMed]
  7. Dai, T.; Huang, Y.-Y.; Hamblin, M.R. Photodynamic therapy for localized infections—State of the art. Photodiagnosis Photodyn. Ther. 2009, 6, 170–188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  8. Giuliani, F.; Martinelli, M.; Cocchi, A.; Arbia, D.; Fantetti, L.; Roncucci, G. In Vitro Resistance Selection Studies of RLP068/Cl, a New Zn(II) Phthalocyanine Suitable for Antimicrobial Photodynamic Therapy. Antimicrob. Agents Chemother. 2010, 54, 637–642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  9. Panariello, B.; Klein, M.I.; Alves, F.; Pavarina, A.C. DNase increases the efficacy of antimicrobial photodynamic therapy on Candida albicans biofilms. Photodiagnosis Photodyn. Ther. 2019, 27, 124–131. [Google Scholar] [CrossRef]
  10. Aida, K.L.; Kreling, P.F.; Caiaffa, K.S.; Calixto, G.M.F.; Chorilli, M.; Spolidorio, D.M.; Santos-Filho, N.A.; Cilli, E.M.; Duque, C. Antimicrobial peptide-loaded liquid crystalline precursor bioadhesive system for the prevention of dental caries. Int. J. Nanomed. 2018, 13, 3081–3091. [Google Scholar] [CrossRef] [Green Version]
  11. Caiaffa, K.S.; Dos Santos, V.R.; Abuna, G.F.; Santos-Filho, N.A.; Cilli, E.M.; Sakai, V.T.; Cintra, L.; Duque, C. Cytocom-patibility and Synergy of EGCG and Cationic Peptides Against Bacteria Related to Endodontic Infections, in Planktonic and Biofilm Conditions. Probiotics Antimicrob. Proteins 2021, 13, 1808–1819. [Google Scholar] [CrossRef]
  12. De Freitas, L.M.; Lorenzon, E.; Santos-Filho, N.; Zago, L.H.D.P.; Uliana, M.P.; de Oliveira, K.T.; Cilli, E.M.; Fontana, C.R. Antimicrobial Photodynamic therapy enhanced by the peptide aurein 1.2. Sci. Rep. 2018, 8, 4212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. De Freitas, L.M.; Lorenzón, E.N.; Cilli, E.M.; de Oliveira, K.T.; Fontana, C.R.; Mang, T.S. Photodynamic and pep-tide-based strategy to inhibit Gram-positive bacterial biofilm formation. Biofouling 2019, 35, 742–757. [Google Scholar] [CrossRef]
  14. Bourré, L.; Giuntini, F.; Eggleston, I.M.; Mosse, C.A.; MacRobert, A.J.; Wilson, M. Effective photoinactivation of Gram-positive and Gram-negative bacterial strains using an HIV-1 Tat peptide–porphyrin conjugate. Photochem. Photobiol. Sci. 2010, 9, 1613–1620. [Google Scholar] [CrossRef] [PubMed]
  15. Yang, K.; Gitter, B.; Rüger, R.; Wieland, G.D.; Chen, M.; Liu, X.; Albrecht, V.; Fahr, A. Antimicrobial peptide-modified lipo-somes for bacteria targeted delivery of temoporfin in photodynamic antimicrobial chemotherapy. Photochem. Photobiol. Sci. 2011, 10, 1593–1601. [Google Scholar] [CrossRef] [PubMed]
  16. Liu, F.; Soh Yan Ni, A.; Lim, Y.; Mohanram, H.; Bhattacharjya, S.; Xing, B. Lipopolysaccharide neutralizing pep-tide-porphyrin conjugates for effective photoinactivation and intracellular imaging of gram-negative bacteria strains. Bioconjugate Chem. 2012, 23, 1639–1647. [Google Scholar] [CrossRef] [PubMed]
  17. Dosselli, R.; Tampieri, C.; Ruiz-González, R.; De Munari, S.; Ragàs, X.; Sánchez-García, D.; Agut, M.; Nonell, S.; Reddi, E.; Gobbo, M. Synthesis, Characterization, and Photoinduced Antibacterial Activity of Porphyrin-Type Photosensitizers Conjugated to the Antimicrobial Peptide Apidaecin 1b. J. Med. Chem. 2013, 56, 1052–1063. [Google Scholar] [CrossRef]
  18. Johnson, G.A.; Muthukrishnan, N.; Pellois, J.-P. Photoinactivation of Gram Positive and Gram Negative Bacteria with the Antimicrobial Peptide (KLAKLAK)2 Conjugated to the Hydrophilic Photosensitizer Eosin Y. Bioconjugate Chem. 2013, 24, 114–123. [Google Scholar] [CrossRef]
  19. Dosselli, R.; Ruiz-González, R.; Moret, F.; Agnolon, V.; Compagnin, C.; Mognato, M.; Sella, V.; Agut, M.; Nonell, S.; Gobbo, M.; et al. Synthesis, Spectroscopic, and Photophysical Characterization and Photosensitizing Activity toward Prokaryotic and Eukaryotic Cells of Porphyrin-Magainin and -Buforin Conjugates. J. Med. Chem. 2014, 57, 1403–1415. [Google Scholar] [CrossRef]
  20. Johnson, G.A.; Ellis, E.A.; Kim, H.; Muthukrishnan, N.; Snavely, T.; Pellois, J.-P. Photoinduced Membrane Damage of E. coli and S. aureus by the Photosensitizer-Antimicrobial Peptide Conjugate Eosin-(KLAKLAK)2. PLoS ONE 2014, 9, e91220. [Google Scholar] [CrossRef] [Green Version]
  21. Le Guern, F.; Sol, V.; Ouk, C.; Arnoux, P.; Frochot, C.; Ouk, T.-S. Enhanced Photobactericidal and Targeting Properties of a Cationic Porphyrin following the Attachment of Polymyxin B. Bioconjugate Chem. 2017, 28, 2493–2506. [Google Scholar] [CrossRef]
  22. Le Guern, F.; Ouk, T.-S.; Ouk, C.; Vanderesse, R.; Champavier, Y.; Pinault, E.; Sol, V. Lysine Analogue of Polymyxin B as a Significant Opportunity for Photodynamic Antimicrobial Chemotherapy. ACS Med. Chem. Lett. 2018, 9, 11–16. [Google Scholar] [CrossRef] [PubMed]
  23. Nakonieczna, J.; Wolnikowska, K.; Ogonowska, P.; Neubauer, D.; Bernat, A.; Kamysz, W. Rose Bengal-Mediated Photoin-activation of Multidrug Resistant Pseudomonas aeruginosa Is Enhanced in the Presence of Antimicrobial Peptides. Front. MiCrobiol. 2018, 9, 1949. [Google Scholar] [CrossRef] [PubMed]
  24. Gao, Y.; Wang, J.; Hu, D.; Deng, Y.; Chen, T.; Jin, Q.; Ji, J. Bacteria-Targeted Supramolecular Photosensitizer Delivery Vehicles for Photodynamic Ablation Against Biofilms. Macromol. Rapid Commun. 2019, 40, e1800763. [Google Scholar] [CrossRef] [PubMed]
  25. Feese, E.; Gracz, H.S.; Boyle, P.D.; Ghiladi, R.A. Towards microbe-targeted photosensitizers: Synthesis, characterization and in vitro photodynamic inactivation of the tuberculosis model pathogen M. smegmatis by porphyrin-peptide conjugates. J. Porphyr. Phthalocyanines 2019, 23, 1414–1439. [Google Scholar] [CrossRef]
  26. Zhang, A.N.; Wu, W.; Zhang, C.; Wang, Q.Y.; Zhuang, Z.N.; Cheng, H.; Zhang, X.Z. A versatile bacterial mem-brane-binding chimeric peptide with enhanced photodynamic antimicrobial activity. J. Mater. Chem. B 2019, 7, 1087–1095. [Google Scholar] [CrossRef]
  27. Chu, J.C.H.; Chin, M.L.; Wong, C.T.T.; Hui, M.; Lo, P.; Ng, D.K.P. One-Pot Synthesis of a Cyclic Antimicrobial Peptide-Conjugated Phthalocyanine for Synergistic Chemo-Photodynamic Killing of Multidrug-Resistant Bacteria. Adv. Ther. 2021, 4, 202000204. [Google Scholar] [CrossRef]
  28. Gao, Q.; Huang, D.; Deng, Y.; Yu, W.; Jin, Q.; Ji, J.; Fu, G. Chlorin e6 (Ce6)-loaded supramolecular polypeptide micelles with enhanced photodynamic therapy effect against Pseudomonas aeruginosa. Chem. Eng. J. 2021, 417, 129334. [Google Scholar] [CrossRef]
  29. Judzewitsch, P.R.; Corrigan, N.; Wong, E.H.H.; Boyer, C.A.J.M. Photo-Enhanced Antimicrobial Activity of Polymers Containing an Embedded Photosensitiser. Angew. Chem. Int. Ed. 2021, 60, 24248–24256. [Google Scholar] [CrossRef]
  30. Qiu, L.; Wang, C.; Lei, X.; Du, X.; Guo, Q.; Zhou, S.; Cui, P.; Hong, T.; Jiang, P.; Wang, J.; et al. Gelatinase-responsive release of an antibacterial photodynamic peptide against Staphylococcus aureus. Biomater. Sci. 2021, 9, 3433–3444. [Google Scholar] [CrossRef]
  31. Qiu, L.; Wang, C.; Lan, M.; Guo, Q.; Du, X.; Zhou, S.; Cui, P.; Hong, T.; Jiang, P.; Wang, J.; et al. Antibacterial Photodynamic Gold Nanoparticles for Skin Infection. ACS Appl. Bio Mater. 2021, 4, 3124–3132. [Google Scholar] [CrossRef] [PubMed]
  32. Mishra, B.; Epand, R.F.; Epand, R.; Wang, G. Structural location determines functional roles of the basic amino acids of KR-12, the smallest antimicrobial peptide from human cathelicidin LL-37. RSC Adv. 2013, 3, 19560–19571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Reinhardt, A.; Neundorf, I. Design and Application of Antimicrobial Peptide Conjugates. Int. J. Mol. Sci. 2016, 17, 701. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Nayab, S.; Aslam, M.A.; Rahman, S.U.; Sindhu, Z.U.D.; Sajid, S.; Zafar, N.; Razaq, M.; Kanwar, R.; Amanullah. A Review of Antimicrobial Peptides: Its Function, Mode of Action and Therapeutic Potential. Int. J. Pept. Res. Ther. 2022, 28, 46. [Google Scholar] [CrossRef]
  35. Boparai, J.K.; Sharma, P.K. Mini Review on Antimicrobial Peptides, Sources, Mechanism and Recent Applications. Protein Pept. Lett. 2020, 27, 4–16. [Google Scholar] [CrossRef]
  36. Hancock, R.; Alford, M.A.; Haney, E.F. Antibiofilm activity of host defence peptides: Complexity provides opportunities. Nat. Reviews. Microbiol. 2021, 19, 786–797. [Google Scholar] [CrossRef]
  37. Huan, Y.; Kong, Q.; Mou, H.; Yi, H. Antimicrobial Peptides: Classification, Design, Application and Research Progress in Multiple Fields. Front. Microbiol. 2020, 11, 582779. [Google Scholar] [CrossRef]
  38. Haney, E.F.; Mansour, S.C.; Hancock, R.E.W. Antimicrobial Peptides: An Introduction; Methods in Molecular Biology; Humana Press: New York, NY, USA, 2017; Volume 1548. [Google Scholar] [CrossRef]
  39. Santos-Filho, N.A.; Righetto, G.M.; Pereira, M.R.; Piccoli, J.P.; Almeida, L.M.; Leal, T.C.; Cilli, E.M. Effect of C-terminal and N-terminal dimerization and alanine scanning on antibacterial activity of the analogs of the peptide p-BthTX-I. Pept. Sci. 2021, e24243. [Google Scholar] [CrossRef]
  40. Li, J.; Fernández-Millán, P.; Boix, E. Synergism between Host Defence Peptides and Antibiotics against Bacterial Infections. Curr. Top. Med. Chem. 2020, 20, 1238–1263. [Google Scholar] [CrossRef]
  41. Andersson, D.I.; Hughes, D. Selection and Transmission of Antibiotic-Resistant Bacteria. Microbiol. Spectr. 2017, 5, 101128. [Google Scholar] [CrossRef]
  42. Kmeid, J.G.; Youssef, M.M.; Kanafani, Z.A.; Kanj, S.S. Combination therapy for Gram-negative bacteria: What is the evidence? Expert Rev. Anti-Infect. Ther. 2013, 11, 1355–1362. [Google Scholar] [CrossRef] [PubMed]
  43. Kumar, A.; Safdar, N.; Kethireddy, S.; Chateau, D. A survival benefit of combination antibiotic therapy for serious infections associated with sepsis and septic shock is contingent only on the risk of death: A meta-analytic/meta-regression study. Crit. Care Med. 2010, 38, 1651–1664. [Google Scholar] [CrossRef] [PubMed]
  44. Carneiro, V.A.; De Oliveira, S.T.; Silva, R.L.; Duarte, H.D.S.; Silva, M.L.; Matos, M.N.C.; Cavalcante, R.M.B.; Figueira, C.S.; Lorenzón, E.N.; Cilli, E.M.; et al. Antimicrobial and Antibiofilm Activity of Lys-[Trp6]hy-a1 Combined with Ciprofloxacin against Gram-Negative Bacteria. Protein Pept. Lett. 2020, 27, 1124–1131. [Google Scholar] [CrossRef] [PubMed]
  45. Jang, J.; Hur, H.-G.; Sadowsky, M.; Byappanahalli, M.; Yan, T.; Ishii, S. Environmental Escherichia coli: Ecology and public health implications—A review. J. Appl. Microbiol. 2017, 123, 570–581. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Lister, J.L.; Horswill, A.R. Staphylococcus aureus biofilms: Recent developments in biofilm dispersal. Front. Cell. Infect. Microbiol. 2014, 4, 178. [Google Scholar] [CrossRef] [Green Version]
  47. Maiti, P.; Chatterjee, S.; Dey, R.; Kundu, A. Biofilms on indwelling urologic devices: Microbes and antimicrobial management prospect. Ann. Med Health Sci. Res. 2014, 4, 100–104. [Google Scholar] [CrossRef] [Green Version]
  48. Rabin, N.; Zheng, Y.; Opoku-Temeng, C.; Du, Y.; Bonsu, E.; Sintim, H.O. Biofilm formation mechanisms and targets for developing antibiofilm agents. Future Med. Chem. 2015, 7, 493–512. [Google Scholar] [CrossRef]
  49. Parsek, M.R.; Singh, P.K. Bacterial Biofilms: An Emerging Link to Disease Pathogenesis. Annu. Rev. Microbiol. 2003, 57, 677–701. [Google Scholar] [CrossRef]
  50. Kiedrowski, M.; Horswill, A.R. New approaches for treating staphylococcal biofilm infections. Ann. N. Y. Acad. Sci. 2011, 1241, 104–121. [Google Scholar] [CrossRef]
  51. Archer, N.K.; Mazaitis, M.J.; Costerton, J.W.; Leid, J.G.; Powers, M.E.; Shirtliff, M.E. Staphylococcus aureus biofilms: Properties, regulation, and roles in human disease. Virulence 2011, 2, 445–459. [Google Scholar] [CrossRef] [Green Version]
  52. Solano, C.; Echeverz, M.; Lasa, I. Biofilm dispersion and quorum sensing. Curr. Opin. Microbiol. 2014, 18, 96–104. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Crusca, E., Jr.; Rezende, A.A.; Marchetto, R.; Mendes-Giannini, M.J.; Fontes, W.; Castro, M.S.; Cilli, E.M. Influence of N-terminus modifications on the biological activity, membrane interaction, and secondary structure of the antimicrobial peptide hylin-a1. Pept. Sci. 2011, 96, 41–48. [Google Scholar] [CrossRef] [PubMed]
  54. Demidova, T.N.; Hamblin, M.R. Effect of cell-photosensitizer binding and cell density on microbial photoinactivation. Antimicrob. Agents Chemother. 2005, 49, 2329–2335. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Banfi, S.; Caruso, E.; Caprioli, S.; Mazzagatti, L.; Canti, G.; Ravizza, R.; Gariboldi, M.; Monti, E. Photodynamic effects of porphyrin and chlorin photosensitizers in human colon adenocarcinoma cells. Bioorganic Med. Chem. 2004, 12, 4853–4860. [Google Scholar] [CrossRef]
  56. Ferreira, J.; Menezes, P.F.C.; Kurachi, C.; Bagnato, V.S.; Sibata, C.; Allison, R.R. Photostability of different chlorine photo-sensitizers. Laser Phys. Lett. 2008, 5, 156–161. [Google Scholar] [CrossRef]
  57. Kwiatkowski, S.; Knap, B.; Przystupski, D.; Saczko, J.; Kędzierska, E.; Knap-Czop, K.; Kotlińska, J.; Michel, O.; Kotowski, K.; Kulbacka, J. Photodynamic therapy—Mechanisms, photosensitizers and combinations. Biomed. Pharmacother. 2018, 106, 1098–1107. [Google Scholar] [CrossRef]
  58. Bock, G.R.; Harnett, S. Photosensitizing Compounds: Their Chemistry, Biology and Clinical Use; John Wiley & Sons: Hoboken, NJ, USA, 2008; Volume 146. [Google Scholar]
  59. Saltaji, H.; Armijo-Olivo, S.; Cummings, G.G.; Amin, M.; Da Costa, B.R.; Flores-Mir, C. Influence of blinding on treatment effect size estimate in randomized controlled trials of oral health interventions. BMC Med Res. Methodol. 2018, 18, 42. [Google Scholar] [CrossRef] [Green Version]
  60. Greene, T. Randomized controlled trials 5: Determining the sample size and power for clinical trials and cohort studies. Methods Mol. Biol. 2015, 1281, 225–247. [Google Scholar]
  61. Vetter, T.R. Fundamentals of Research Data and Variables: The Devil Is in the Details. Anesth. Analg. 2017, 125, 1375–1380. [Google Scholar] [CrossRef]
  62. Shamseer, L.; Moher, D.; Clarke, M.; Ghersi, D.; Liberati, A.; Petticrew, M.; Shekelle, P.; Stewart, L.A.; The PRISMA-P Group. Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015: Elaboration and explanation. BMJ 2015, 349, g7647. [Google Scholar] [CrossRef] [Green Version]
  63. Ouzzani, M.; Hammady, H.; Fedorowicz, Z.; Elmagarmid, A. Rayyan—A web and mobile app for systematic reviews. Syst. Rev. 2016, 5, 210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Flowchart based on the PRISMA statement.
Figure 1. Flowchart based on the PRISMA statement.
Ijms 23 03226 g001
Figure 2. Ilustration of the results of the quantitative analysis. The experimental group (positive events) included microorganisms that received the association therapy (aPDT + AMP), while the control group included microorganisms that received only aPDT. (A) results of the meta-analysis illustrated in a forest plot. OR: odds ratio; CI: confidence interval; W: weight, [13,15,23]. (B) trim-and-fill method results illustrated in a forest plot. TE: estimated mean; seTE: estimated standard deviation; OR: odds ratio; CI: confidence interval; W: weight, [13,15,23].
Figure 2. Ilustration of the results of the quantitative analysis. The experimental group (positive events) included microorganisms that received the association therapy (aPDT + AMP), while the control group included microorganisms that received only aPDT. (A) results of the meta-analysis illustrated in a forest plot. OR: odds ratio; CI: confidence interval; W: weight, [13,15,23]. (B) trim-and-fill method results illustrated in a forest plot. TE: estimated mean; seTE: estimated standard deviation; OR: odds ratio; CI: confidence interval; W: weight, [13,15,23].
Ijms 23 03226 g002
Table 1. Summary of the characteristics of the studies included.
Table 1. Summary of the characteristics of the studies included.
Study (Year)Study DesignPeptideIrradiation TimeWavelengthPhotosensitizerMicroorganismCulture TypeSample SizeOutcomes
Bourré et al. 2010 [14]In vitroTat30, 43, 60, and 120 s410 nmTetracks (phenol) and porphyrinEscherichia coli
Staphylococcus aureus
Pseudomonas aeruginosa
Streptococcus pyogenes
SuspensionNDReduction in the concentration of 1 uM from 3 to 6 log10 CFU/mL. The greatest effect was in the first 30 s.
Yang et al. 2011 [15]In vitroWLBU2100 s652 nmTemoporfin + WLBU2S. aureus (methicillin resistant)
P. aeruginosa
Suspension3Reduction by 100% for S. aureus (aPDT only and aPDT + peptide) and reduction by 2 log10 CFU/mL for P. aeruginosa (aPDT + peptide).
Liu et al. 2012 [16]In vitro WI13WF (YVLWKRKRKFCFI-amide)2, 5, and 10 min400 to 900 nmProtoporphyrin IXE. coli
Salmonella enteric
Klebsiella pneumoniae
SuspensionNDPeptide and PS conjugate 99% lethal.
Dosseli et al. 2013 [17]In vitroApidaecinND600–750 nm
390–460 nm
PorphyrinE. coli
S. aureus
SuspensionNDReduction by 100% for E. coli.
Johnson et al. 2013 [18]In vitro(KLAKLAK)230 min525 nm(KLAKLAK)2 + Eosin YAcinetobacter baumannii
P. aeruginosa
E. coli
S. aureus
Staphylococcus epidermidis
SuspensionNDReduction by 99% for all microorganisms.
Dosseli et al. 2014 [19]In vitroMagainin
Buforin
ND390–460 nmPorphyrinE. coli
S. aureus (methicillin resistant)
SuspensionNDReduction by 100% for all microorganisms.
Johnson et al. 2014 [20]In vitro(KLAKLAK)22 min
5 min
30 min
525 nm(KLAKLAK)2 + Eosin YE. coli
S. aureus
Suspension3Reduction by 50% for all microorganisms (2 min of irradiation).
Reduction by 90% (5 min of irradiation).
Reduction by 99.99% (30 min of irradiation).
Le guern et al. 2017 [21] In vitroPolymyxin B20 h420 nmPorphyrinS. aureus
E. coli
P. aeruginosa
SuspensionNDAntibactericidal activity of the PS and peptide association on 3 strains.
De Freitas et al. 2018 [12]In vitroAurein 1.2 (AU)ND660 nm Methylene blue
Chlorin e6
S. aureus
A. baumannii
E. coli
Enterococcus faecium
Suspension9S. aureus reduction
-
MB ~ 1.0 log10 CFU/mL
-
MB + AU ~ 6.0 log10 CFU/mL
-
Ce6 and Ce6 + Au = total reduction

A. baumannii reduction
-
MB ~ 1.0 log10 CFU/mL
-
MB + AU ~ 6.0 log10 CFU/mL
-
Ce6 and Ce6 + AU no significant results

E. coli reduction
-
MB ~ 4.0 log10 CFU/mL
-
MB + AU ~ 4.0 log10 CFU/mL
-
Ce6 and Ce6 + AU no significant results

E. faecium reduction
-
MB ~ 1.0 log10 CFU/mL
-
MB + AU ~ 3.0 log10 CFU/mL
-
Ce6 ~ 1.0 log10 CFU/mL
-
Ce6 + AU = total reduction
Le guern et al. 2018 [22]In vitroPolymyxin B modified by lysine20 h420 nmPorphyrinS. aureus
E. coli
P. aeruginosa
SuspensionNDReduced antibacterial activity of polymyxin modified by lysine.
Nakonieczana et al. 2018 [23]In vitroCAMEL
Pexiganan
668 s
1335 s
2668 s
514 nmRose-bengal (RB)P. aeruginosaSuspension3Reduction by 2.06 log10 CFU/mL for RB + CAM.
Reduction by 6.00 log10 CFU/mL for RB + PEX.
Gao et al. 2019 [24]In vitroMagainin I2 min
4 min
8 min
660 nm Magainin I + Chlorin e6P. aeruginosa
S. aureus (methicillin resistant)
BiofilmNDP. aeruginosa
2 min (0.385 log10 CFU/mL reduction)
4 min (1.645 log10 CFU/mL reduction)
8 min (6.724 log10 CFU/mL reduction)
S. aureus
2 min (0.922 log10 CFU/mL reduction)
4 min (3.796 log10 CFU/mL reduction)
8 min (6.586 log10 CFU/mL reduction)
De Freitas et al. 2019 [13]In vitroAU (GLFDIIKKIAESF-NH2)
(AU)2K[(GLFDIIKKIAESF)2-k]
ND664 nmMethylene blue
Chlorin e6
Enterococcus faecalis
S. aureus
E. faecium
Biofilm9Reducing the early biofilm stage
-
95.5%—(Ce6-aPDT + (AU)2K)
-
78%—Ce6-aPDT
-
30%—MB-aPDT + AU
-
20%—MB-aPDT
-
30%—AU
-
70%—(AU)2K)
Feese et al. 2019 [25In vitroAlkyne 1-Zn
TMPYP
5, 15, and 30 min400 to 700 nmPorphyrin Mycobacterium smegmatisSuspension Inactivation of 4 Log10 CFU/mL when associated with porphyrin and 1-Zn.
Zhang et al. 2019 [26]In vitro(KLAKLAK)2(KLA)5 min (in vivo)
10 min (in vitro)
660 nm PpIX
PPK = PpIX + (KLAKLAK)2(KLA)
S. aureus
E. coli
SuspensionNDInhibition rate
S. aureus = 100% for both PS
E. coli = 100% (PPK)/50% (PpIX)
Chu et al. 2021 [27]In vitroBacitracin5 and 30 min 610 nmPhthalocyanineE. coli
S. aureus
Suspension9High phototoxicity of the Peptide with PS.
The group without light 99% reduced.
Gao et al. 2021 [28]In vitro/in vivoPEGylated polypeptide5 min660 nmPEGylated polypeptide + Chlorin e6P. aeruginosaBiofilmNDTotal eradication of P. aeruginosa biofilms.
Judzewitsch et al. 2021 [29]In vitroZnTTP-AC30 minGreen-light irradiationZnTTP-ACS. aureus
P. aeruginosa
Suspension34.5 log10 CFU/mL reduction for S. aureus.
Total reduction for P. aeruginosa.
Qiu et al. 2021 [a] [30]In vitro/in vivoGKRWWKWWR-RPLGVRG5 min660 nmGKRWWKWWR-RPLGVRG + Chlorin e6S. aureus
E. coli
Suspension3Total reduction for S. aureus
90% reduction for E. coli
Qiu et al. 2021 [b] [31]In vitro/in vivoGKRWWKWWRR10 min
20 min
30 min
660 nmGKRWWKWWRR + Chlorin e6 + AuNPsS. aureus
E. coli
Biofilm3S. aureus
10 min (~50% viability)
20 min (~20% viability)
30 min (~2.5% viability)
E. coli
10 min (~60% viability)
20 min (~42.5% viability)
30 min (~10% viability)
ND: not documented; s: seconds; min: minutes: h: hour; PS: photosensitizer; ~: approximately; MB: methylene blue; RB: rose-bengal; Ce6: chlorin e6.
Table 2. Risk of bias assessment in the articles included, according to the OHAT criteria.
Table 2. Risk of bias assessment in the articles included, according to the OHAT criteria.
Studies/QuestionsWas the Dose or Exposure Level Administered Adequately Randomized?Was the Allocation to Study Groups Adequately Concealed?Were the Experimental Conditions Identical Across Study Groups?Were Research Personnel Blind to the Study Group During the Study?Were the Outcome Data Complete without Attrition or Exclusion from the Analysis?Is the Exposure Characterization Reliable?Is the Outcome Assessment (Including Blinding of Assessors) Reliable?Were There No Other Potential Threats to Internal Validity?
Bourré et al. 2010 [14]++++++--++++----
Yang et al. 2011 [15]++++++--++++----
Liu et al. 2012 [16]++++++--++++----
Dosseli et al. 2013 [17]++++++----++----
Johnson et al. 2013 [18]++++++--++++----
Dosseli et al. 2014 [19]++++++--++++----
Johnson et al. 2014 [20]++++++--++++----
Le Guern et al. 2017 [21]++++++--++++----
De Freitas et al. 2018 [12]++++++--++++----
Le Guern et al. 2018 [22]++++++--++++----
Nakonieczana et al. 2018 [23]++++++--++++----
Gao et al. 2019 [24]++++++--++++----
De Freitas et al. 2019 [13]++++++--++++----
Fesse et al. 2019 [25]++++++--++++----
Zhang et al. 2019 [26]++++++--++++----
Chu et al. 2021 [27]++++++--++++----
Gao et al. 2021 [28]++++++--++++----
Judzewitsch et al. 2021 [29]++++++--++++----
Qiu et al. 2021a [30]++++++--++++----
Qiu et al. 2021b [31]++++++--++++----
++: direct evidence of positive finding; --: direct evidence of negative finding.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Dias, L.M.; Ferrisse, T.M.; Medeiros, K.S.; Cilli, E.M.; Pavarina, A.C. Use of Photodynamic Therapy Associated with Antimicrobial Peptides for Bacterial Control: A Systematic Review and Meta-Analysis. Int. J. Mol. Sci. 2022, 23, 3226. https://doi.org/10.3390/ijms23063226

AMA Style

Dias LM, Ferrisse TM, Medeiros KS, Cilli EM, Pavarina AC. Use of Photodynamic Therapy Associated with Antimicrobial Peptides for Bacterial Control: A Systematic Review and Meta-Analysis. International Journal of Molecular Sciences. 2022; 23(6):3226. https://doi.org/10.3390/ijms23063226

Chicago/Turabian Style

Dias, Luana Mendonça, Túlio Morandin Ferrisse, Karine Sousa Medeiros, Eduardo Maffud Cilli, and Ana Claudia Pavarina. 2022. "Use of Photodynamic Therapy Associated with Antimicrobial Peptides for Bacterial Control: A Systematic Review and Meta-Analysis" International Journal of Molecular Sciences 23, no. 6: 3226. https://doi.org/10.3390/ijms23063226

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