Next Article in Journal
Identified Seaweed Compound Diphenylmethane Serves as an Efflux Pump Inhibitor in Drug-Resistant Escherichia coli
Next Article in Special Issue
Antimicrobial Potential of the Genera Geobacillus and Parageobacillus, as Well as Endolysins Biosynthesized by Their Bacteriophages
Previous Article in Journal
Artificial Intelligence and Antibiotic Discovery
Previous Article in Special Issue
Efficacy of Cathelicidin LL-37 in an MRSA Wound Infection Mouse Model
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

New Perspectives on Old and New Therapies of Staphylococcal Skin Infections: The Role of Biofilm Targeting in Wound Healing

1
Department of Clinical and Molecular Sciences Clinic of Dermatology, Polytechnic University of Marche, 60020 Ancona, Italy
2
Department of Biomedical Sciences and Public Health Clinic of Infectious Diseases, Polytechnic University of Marche, 60020 Ancona, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Antibiotics 2021, 10(11), 1377; https://doi.org/10.3390/antibiotics10111377
Submission received: 9 October 2021 / Revised: 6 November 2021 / Accepted: 7 November 2021 / Published: 10 November 2021
(This article belongs to the Special Issue Evaluation of New Molecules in Severe Infectious Diseases)

Abstract

:
Among the most common complications of both chronic wound and surgical sites are staphylococcal skin infections, which slow down the wound healing process due to various virulence factors, including the ability to produce biofilms. Furthermore, staphylococcal skin infections are often caused by methicillin-resistant Staphylococcus aureus (MRSA) and become a therapeutic challenge. The aim of this narrative review is to collect the latest evidence on old and new anti-staphylococcal therapies, assessing their anti-biofilm properties and their effect on skin wound healing. We considered antibiotics, quorum sensing inhibitors, antimicrobial peptides, topical dressings, and antimicrobial photo-dynamic therapy. According to our review of the literature, targeting of biofilm is an important therapeutic choice in acute and chronic infected skin wounds both to overcome antibiotic resistance and to achieve better wound healing.

1. Introduction

Staphylococcal skin infections are one of the most common complications of both surgical sites [1] and chronic wounds, such as arterial, venous, and diabetic ulcers [2]. Staphylococcus aureus is one of the top four bacteria in terms of prevalence among chronic wounds [3,4,5], and its ability to produce biofilm is a major virulence factor contributing to wound chronicity and delayed healing [6]. Furthermore, S. aureus has a high public health impact due to increasing antibiotic resistance [7], and methicillin-resistant Staphylococcus aureus (MRSA) represents a therapeutic challenge, with an important role in slowing down the wound healing process [8,9,10,11].
A meta-analysis [6] of several studies reported that bacterial biofilm is present in 78.2% of chronic wounds and contributes to persistent infection. All staphylococcal species are responsible for 65% of persistent infections in chronic wounds [2] and can adhere to wound surface proteins to form colonies embedded in the biofilm that are resistant to antibiotic therapy [12].
Specifically, biofilm consists of microbial populations attached to a surface and immersed in a polymeric, hydrated extracellular fluid, known as extracellular polymeric substance (EPS) [13], which includes extracellular DNA (eDNA), polysaccharides, and proteins [14]. Furthermore, biofilm confers antiphagocytic capabilities, prevents the action of leukocytes [15], and can capture and make both complement and antibiotics ineffective, triggering persistent tissue damage and chronic inflammation [16]. Staphylococcal biofilm development follows a complex pathway that includes attachment, maturation, and dispersion, with an important role of environmental factors, such as the surface of adhesion considered and nutrients available [14].
The pathogenetic role of staphylococcal biofilm in wound healing was evaluated in a study by Roy et al. [17], showing that, in a porcine skin wound model, biofilm produced by S. aureus promotes degradation of collagen type 1 by repressing wound-edge miR-143 (a miRNA sensitive to staphylococcal biofilm) and consequently upregulating metalloproteinase-2 production. The collagen1/collagen3 ratio decreases, altering wound repair capacity, granulation tissue production, and promoting recurrence. This results in poorer healing outcomes and increased costs of care [18]. In addition, increased levels of metalloproteinases are typical in chronic wounds, which fail to heal effectively because the tensile strength of the type 1 collagen scaffold is lost [19]. Consequently, biofilm loading structurally compromises the wound tissue and promotes local infectious recurrence [19].
In addition to S. aureus, other types of Coagulase-negative staphylococci may also produce biofilms and be responsible for chronic wound maintenance, such as S. epidermidis [20] and S. lugdunensis [21]. However, there are different strains of staphylococci with varying degrees of biofilm production capacity, often in conjunction with other pathogens [22,23].
Biofilm frequently develops on the surface of wounds and is responsible for their chronicity [24]. Its removal can considerably improve the speed and quality of wound healing [25,26], using anti-biofilm agents. These are characterized by their low molecular weight and ability to break down biofilm by promoting penetration of antibacterial molecules and restoring leucocyte activity [27,28,29,30]. Given the high impact of chronic infected wounds [31,32] on public health and the increasing number of bacterial resistances to antibiotics, we reviewed the literature both to update the latest evidence on old antibiotics and molecules, and to highlight new molecules to overcome the staphylococcal defence strategies in order to improve wound healing.
We considered both recent and older molecules (Table 1), as we believe that all of them may be useful to overcome the growing antibiotic resistance and improve wound healing, particularly in association with antibiotics.

2. Results

2.1. Antibiotics

The gold standard for treatment of staphylococcal skin infections, including biofilm-forming strains, are systemic antibiotics. Beta-lactams are the first line therapy for both coagulase-negative staphylococci (CNS) and methicillin-sensitive S. aureus (MSSA) [60]. However, infections sustained by MRSA, or methicillin-resistant CNS, require an antibiogram to select the appropriate antibiotic [61]. Adjuvant therapy to eradicate the infection and improve wound healing is recommended in all conditions where there is potential for biofilm formation [60]. Since most antibiotics have been developed for bacteria in planktonic form (free-swimming), complementary strategies are required to target the biofilm and allow the antibiotic to reach the pathogen [62].
Conversely, incorrect use of antibiotics can promote S. aureus biofilm development. Some studies showed that concentrations below the minimum inhibitory concentration (MIC) of cephalothin [63], oxacillin [64], cephalexin [65], vancomycin [63], and Linezolid [66] can stimulate biofilm formation up to four-fold. Furthermore, Kaplan et al. [67] showed that ampicillin, cloxacillin, methicillin and amoxicillin used sub-MIC in vitro favoured biofilm formation in MSSA and MRSA strains by increasing eDNA release [68]. A similar phenomenon has been shown for S. epidermidis with tigecycline, novobiocin, linezolid, vancomycin and fluoroquinolones [69]. Finally, considering MRSA, Majidpour et al. [70] highlighted that vancomycin and azithromycin may have biofilm-inducing effects, while linezolid, clarithromycin, and cefazolin, followed by minocycline and clindamycin may be effective in inhibiting biofilm formation.

2.1.1. Beta-Lactams

The activity of beta-lactams in wounds infected with biofilm-producing staphylococci may be compromised, requiring combination with other molecules or procedures [33]. In addition, low-dose beta lactam can promote S. aureus biofilm production, which is dependent on the level of eDNA. The latter plays the role of matrix adhesin in the biofilm [71,72,73,74,75] and is produced by bacterial cell lysis determined by the autolysin AtlA [73]. Sub-MIC beta lactams appear to induce AtlA and thus increase biofilm production [67].
In an in vitro study by Kirmusaoglu et al. [33], resistance to beta lactams in biofilm-producing strains of MRSA and MSSA is overcome through synergistic action with 2-aminothiazole, highlighting a possible solution to the treatment of biofilm-forming staphylococcal infections.

2.1.2. Macrolides

Macrolides are bacterial protein synthesis inhibiting molecules with an unclear role against staphylococcal biofilm [76]. We found some studies suggesting their antibiofilm activity only when in combination with other antibiotics, as clarithromycin/daptomycin, [76] clarithromycin/vancomycin [77], and roxithromycin/imipenem [78]. In vivo studies referring specifically to skin wound healing and biofilm are lacking.

2.1.3. Teicoplanin

Teicoplanin is a glycopeptide and represents the antibiotic of choice in empirical therapy of MRSA infections [79]. As it is an effective molecule in chronic wounds; it is often the comparator in animal models and shows excellent results in both preventing biofilm formation and treating wounds with an established biofilm. It also demonstrates to be effective in promoting the wound healing process [34].

2.1.4. Daptomycin

Daptomycin is a lipopeptide with activity against Gram-positive bacteria, including MRSA and vancomycin-resistant S. aureus [80,81,82,83], and is recommended for the treatment of skin, soft tissue, and bloodstream infections. It also has a strong activity against staphylococcal biofilm [84].
In a study [35] in a mouse model with MRSA-infected burns, the efficacy of intraperitoneal daptomycin was evaluated in comparison to intraperitoneal teicoplanin and a no-treatment control. The best antimicrobial activity and histological outcome were obtained in the daptomycin-treated group.

2.1.5. Tigecycline

Tigecycline is a glycylcycline antibiotic that has demonstrated efficacy against staphylococci, particularly MRSA, and their biofilms, although the best results have been obtained in combination with other antibiotics [85,86,87,88,89]. However, a direct effect of tigecycline on wound healing of S. aureus-infected wounds in mice was shown through modulation of matrix metalloproteinase-9 expression, proving superior to teicoplanin in comparison [36].

2.1.6. Dalbavancin

Dalbavancin is a novel lipoglycopeptide with a spectrum of action against Gram-positives and shows great penetration of staphylococcal biofilm [90,91,92].
Its role in wound healing was investigated in a mouse model with MRSA-infected wounds [38]. The comparison was made with daily vancomycin (10 mg/kg) and dalbavancin with two administrations on day 1 and day 8. At 14 days, both antibiotics had reduced the bacterial load, with dalbavancin being more effective, which also resulted in healing with normal, well-organised keratinized epithelium, slightly less than the uninfected group but better than the vancomycin-treated group. In addition, epidermal growth factor receptor (EGFR) and vascular endothelium growth factor (VEGF) values were found to be higher than with vancomycin.

2.2. Quorum Sensing Inhibitors

Quorum sensing (QS) is a cell-to-cell bacterial communication mechanism capable of regulating gene expression according to environmental conditions [93]. Staphylococci, particularly S. aureus, are able to regulate their virulence factors, including biofilm and toxin formation, thanks to QS [94,95,96,97,98,99,100,101,102]. By using molecules that can inhibit QS it is possible to circumvent the adaptation of staphylococcal strains to the wound conditions, preventing the formation of biofilm [103,104,105,106,107].

2.2.1. RIP

RNA III inhibiting peptide (RIP) is a seven-amino acid molecule capable of inhibiting the synthesis of RNAIII, a transcriptional unit of the staphylococcal accessory gene regulator (Agr) system responsible for QS and biofilm formation [108,109].
This peptide has been proven to be effective in treating device-associated infections due to MRSA and MSSA [110,111,112,113], and its role in skin wound healing in animal models has also been evaluated. Schiele et al. [39] showed in a mouse model that S. aureus and S. epidermidis biofilm reduced the rate of wound healing significantly compared with an uninfected wound, with a histologically lower degree of re-epithelisation. Infected mice treated with topical RIP 100 mcg for 7 days had a histologically comparable degree of healing to uninfected mice. This underlines the possible topical role of RIP in preventing biofilm and ensuring better wound healing.
The role of topical RIP (20 mcg) in the healing of MRSA-infected wounds in combination with daily teicoplanin (7 mg/kg) for 7 days compared with teicoplanin alone has also been demonstrated in a mouse model, resulting in a lower bacterial load and histological degree of healing (collagen, re-epithelisation, microvascular density and expression of VEGF comparable to non-infected wounds [40]. It is also hypothesised that RIP may induce VEGF by improving the quality and speed of wound healing [39,40,112,113,114].
Topical daily RIP was also used in two cases [115] of patients with chronic diabetic ulcers after failure of conventional antibiotic therapy, in systemic combination with systemic daptomycin, avoiding amputation of the lower limb.

2.2.2. F19, F12 and F1

F19, F12 and F1 are small-molecule biaryl hydroxyketones that inhibit staphylococcal QS and thus also affect biofilm production [116]. Their efficacy was evaluated in a study by Kuo et al. [41] in an animal model. F19, F12 and F1 were applied by injection (20 mg/kg) into MRSA-infected insect larvae, while F12 and F1 were applied topically to mice with MRSA-infected wounds. F19, F12 and F1 provided a survival advantage for treated infected larvae over untreated larvae. In the mouse model, F12 and F1 improved the speed of wound healing. In addition, it was shown that some antibiotics ineffective in monotherapy against MRSA, such as cephalothin and nafcillin, can be restored in combination with F12 and F1, observing a MIC reduction in vitro by 40 and 60 times respectively.

2.2.3. FS10

FS10 is a tetrapeptide with the same mechanism of action as RIP and with enhanced antistaphylococcal activity against MRSA [37,117,118]. Its role in wound healing was assessed histologically in a mouse model, showing that the combination of systemic tigecycline (7 mg/kg) and topical FS10 (20 µg) resulted in better healing and control of both MRSA and MSSA infection than monotherapy [119]. FS10 shows the best results in combination with a systemic antibiotic, particularly tigecycline. The latter can accelerate wound healing by reducing expression of matrix metalloproteinase 9 but is inferior to combination therapy [42]. FS10 and QS inhibitors in general act by inhibiting staphylococcal virulence factors, including biofilm formation.

2.3. Antimicrobial Peptides

Antimicrobial peptides (AMPs) are a large group of molecules that are generally part of the innate immunity of all life forms and are becoming increasingly important in the era of antibiotic resistance [120]. Their mechanism of action differs depending on the type of AMPs and may result in killing bacteria by lysis or by targeting intracellular components [121]. In addition, AMPs have also shown immunomodulatory properties, reducing the inflammatory component, inducing epithelial cell migration and neoangiogenesis [122,123,124,125]. Their role in local biofilm inhibition and wound healing has been shown in the literature [126,127,128], allowing destruction of the polymeric matrix biofilm by binding with anionic bacterial Lipopolysaccharides [129].

2.3.1. Innate Defence Regulator (IDR)-1018

IDR-1018, a synthetic cationic peptide, showed strong antibiofilm action by promoting degradation of the nucleotide guanosine penta- and tetra-phosphate (p)ppGpp, which regulates biofilm formation in staphylococci and participates in antibiotic resistance [130,131]. In a study on a mouse model with S. aureus-infected wounds, IDR-1018 injected subcutaneously was shown to reduce bacterial load and eliminate biofilm [43].

2.3.2. LL-37

LL-37 is a natural cathelicidin-derived AMP that is important in both inhibiting staphylococcal biofilm formation and wound healing. LL-37 is present in non-infected wounds and promotes the reparative process [132], whereas the presence of antibodies to LL-37 inhibits re-epithelisation and its expression is reduced in chronic wounds [133]. Adenovirus-mediated gene transfer for LL-37 was observed to result in improved wound healing in obese mice [44]. In addition, LL-37 appears to promote non-infected wound healing in a dexamethasone-treated mouse model [45].
The anti-biofilm activity is due to the prevention of its formation by impairing bacterial adhesion and staphylococcal QS, while also increasing bacterial motility [46]. A limitation of this AMP is that it can be inactivated by endogenous, bacterial proteases and sub-physiological salt concentrations [46,47,48,49,134].

2.3.3. SHAP1

SHAP1 is a synthetic peptide (APKAMKLLKKLLKLQKKGI) that has shown excellent antimicrobial activity against both bacteria and fungi, maintaining great stability both in the presence of proteases and in a salty environment [50]. One relevant feature is its ability to promote wound healing both in vitro and in the S. aureus-infected mouse model by activating the EGFR pathway. It proved superior in this compared to LL-37, which is inactivated in a salty or protease-rich environment [50,135,136]. SHAP1, applied topically (1µM) to the wounds of S. aureus-infected and uninfected mice, accelerated the healing process in both cases. Complete closure was achieved in 3 days [50].

2.3.4. DRGN-1

DRGN-1 is a synthetic cationic AMP derived from an H1 histone of Komodo dragon and has been shown to be effective as an anti-biofilm, anti-staphylococcal and wound healing agent in a mouse model [51]. Topically applied DRGN-1 showed to promote keratinocyte migration in vitro and act on the EGFR-Signal transducer and activator of transcription (STAT)1/3 pathway, while its antimicrobial activity consists of permeabilising bacterial cell membranes and disrupting biofilm [51].

2.3.5. Dermaseptin Peptide2 (DMS-PS2)

DMS-PS2 is a synthetic cationic AMP belonging to the dermaseptins with potent anti-biofilm, anti-MRSA and wound healing actions [52]. In a mouse model with MRSA-infected wounds, DMS-PS2 was applied topically, resulting in a drastic reduction in the bacterial load after 1 day and a completely increased rate of re-epithelialisation compared with untreated wounds.

2.3.6. Cell-Free Supernatant (CFS) of Lactobacillus plantarum USM8613

In this study, a CFS of Lactobacillus plantarum USM8613 was proven to inhibit S. aureus growth and biofilm formation on a porcine skin wound model by increasing beta-defensin levels. The protein-rich fraction of L. plantarum USM8613 was shown to be effective in promoting wound healing in a mouse model, and it increased the immune response and cytokine and chemokine production [137].

2.3.7. Def-1

Honey defensin-1 is a recombinant molecule derived from honey (honeydew and manuka type) and was tested in a chronic wound model with biofilm formed by various pathogens, including S. aureus. Def-1 was effective in significantly reducing mature biofilm by 24 and 48 h [53]. In vivo studies to confirm its action on wound healing are still not available.

2.4. Other Topical Dressing

Octenidine Dihydrochloride (OCT)

OCT is a cationic surfactant with antiseptic properties. OCT-impregnated gauze dressing was used in a murine model with MRSA-infected skin wounds, resulting in accelerated wound healing after 24 h compared to phosphate-buffered saline controls and the 2% mupirocin group. In addition, histologically fewer inflammatory cells, better re-epithelialisation, and more mature collagen fibres were found in the OCT group [54].

2.5. Antimicrobial Photo Dynamic Therapy (APDT)

APDT may represent an alternative for the treatment of localised skin infections, demonstrating in vitro efficacy for different types of micro-organisms [138]. Its mechanism of action is based on exposing the bacteria-infected surface to light of a defined wavelength, after applying a photosensitiser capable of inducing reactive oxygen species and leading to a cytotoxic effect against the pathogens [138,139,140,141,142,143,144,145,146,147,148].

2.5.1. RLP068/Cl

RLP068/Cl is a photosensitiser derived from tetracationic Zn (II) phthalocyanine [149] that was used in a biofilm-producing MRSA-infected mouse model with 689 nm light. A single session of APDT was performed 2 days after infection, following biofilm production, with a marked reduction in bacterial load on the same day compared to untreated subjects and those treated with systemic teicoplanin (7 mg/kg). At 7 days after therapy, the reduction in bacterial counts was similar between APDT and teicoplanin, suggesting the efficacy of RLP068/Cl in controlling infection. In addition, histological evaluation showed complete re-epithelialisation in APDT-treated mice at 7 days and good healing in the teicoplanin group. This highlighted the role of RLP068/Cl in treating chronic infections with significant biofilm formation [55].

2.5.2. Curcumin Encapsulated in Silica Nanoparticles

Cucurmin is known to be a photosensitiser [150,151,152,153,154], and an encapsulated form of it in silica nanoparticles was used with APDT in an in vitro study on human fibroblast cultures, demonstrating its efficacy in reducing both the bacterial count of S. aureus and the production of biofilm, while also allowing improved human fibroblast activity [56].

2.5.3. Aminolevulinic Acid (ALA)

A study by Lin et al. [57] showed the efficacy of ALA in three patients with chronic leg ulcers resistant to conventional therapy. One to three sessions of APDT were carried out, and after the first session no more staphylococci or other bacteria were isolated. The patients did not present recurrences for 29 months, highlighting the action on wound healing.

2.5.4. Hypericin Nanoparticles

Hypericin is a natural photosensitiser with also an antibacterial and anti-biofilm action [155,156,157,158]. Nanoparticles are an effective vehicle that makes it possible to limit the lipophilicity and therefore the toxicity of this photosensitiser to the microbial target only [157,158,159,160]. In a study by Nafee et al. [58], the efficacy of hypericin nanoparticles on MRSA was evaluated in two models. In the in vitro model, an excellent ability to inhibit biofilm formation was demonstrated, and in the murine model, infected and treated wounds healed faster and histologically better than untreated wounds.

2.5.5. Methylene Blue aPDT (MB-aPDT)

In a study by Pérez et al. [59], the efficacy of MB-aPDT alone or in combination with mupirocin was evaluated in a mouse model of a S. aureus-infected wound. A single session with MB-aPDT alone showed better cosmetic (no residual scarring) and healing (loss of crusts and reduction in extent) results than untreated control. Furthermore, MB-aPDT determined histological evidence of improved connective tissue production and cellularity in the dermis compared to mupirocin alone. The combination of mupirocin and MB-aPDT did not improve the effect on wound healing.

3. Discussion

In our review of the literature, we evaluated the efficacy of targeting staphylococcal biofilms in skin wounds, considering mainly the effect on wound healing in in vivo studies, where histological assessment was also reported. For this reason, studies without skin histological evaluation were not included, while others with in vitro relevance (e.g., human fibroblast) were cited. Therefore, considering exclusively the selection criteria, this review is not exhaustive for every existing therapy that could have an anti-biofilm effect and represents a selection of all available studies.
Regarding antibiotics, beta lactams are indicated in combination with adjuvant therapy for chronic wounds with biofilm-forming staphylococcal infection [33]. To the best of our knowledge, in vivo studies with histological assessment of wound healing in wounds infected with biofilm-producing staphylococci are lacking. However, many adjuvants showed histological benefits in wound healing, and thus, potentially, all the topical therapies described previously may be suitable. (Table 1) Further studies are needed to provide histological evidence of the efficacy of these combinations in wound healing. In addition, teicoplanin, daptomycin and dalbavancin also showed to improve wound healing in vivo in mouse models. For tigecycline better control of infection was achieved in combination with daptomycin or rifampicin, although superiority to teicoplanin in wound healing was demonstrated through modulation of metalloproteinase-9 in a mouse model infected with biofilm-producing S. aureus [36]. This may suggest that tigecycline has a greater effect on wound healing than one of the leading antibiotics for MRSA, teicoplanin, but both antibiotics are effective in either controlling infection or promoting better wound healing. In our opinion, tigecycline or teicoplanin may be used in biofilm-forming MRSA skin infections as first line. In the case of non-responsive and non-healing infections, we suggest evaluating the combination with one of the described adjuvant therapies. However, most of these have never been tested in humans and are not readily available.
For example, quorum sensing inhibitors are also promising treatment options, but there is a lack of studies in humans to assess their efficacy. RIP has been used topically in two cases of chronic diabetic ulcers in combination with daptomycin, with clinical improvement [115]. Although they have been shown to modulate VEGF expression and wound healing, the best results were obtained in combination with systemic antibiotics, such as FS10 and tigecycline. These molecules can help increase MRSA sensitivity towards antibiotics to which they are normally resistant. Specifically, F12 and F1 reduced the MIC for cephalothin and nafcillin by about 50-fold [41]. Increased use of these molecules topically, especially in association with a systemic antibiotic, may therefore both promote wound healing and increase the eradicating capacity of the antibiotic used.
Similarly, better results are possible for AMPs in combination with systemic antibiotics. In our review of the literature, many studies do not consider the histological evaluation of wound healing, preventing an assessment of their effect on the healing process and their direct correlation with the anti-biofilm action. However, many molecules have been referred with excellent potential against the staphylococcal biofilm and represent an important therapeutic possibility worthy of further studies, including applications on humans [43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164].
For instance, naja atra cathelicidin (NA-CATH):ATRA1-ATRA1, a highly cationic synthetic peptide derived from a natural snake cathelicidin, showed exceptional anti-biofilm properties against S. aureus [161,162] and has been found to be superior to LL-37 in inhibiting biofilm under saline environmental conditions [162]. This would overcome the limitations of using LL-37 due to environmental factors, but studies demonstrating an effect on wound healing are still lacking. We believe that this molecule deserves further in vivo studies before clinical application.
Better results on biofilm control can be achieved by combining AMPs with different mechanisms of action, as well as AMPs and antibiotics. In a mouse model study [165], topical RIP plus Temporin A, an AMP effective against biofilm and promoting wound healing [166], was shown to result in better control of glycopeptides-intermediate S. aureus-infected wounds compared with monotherapy and rifampicin. This study suggests that the combination of an anti-QS agent and a specific anti-biofilm agent further increases efficacy against antibiotic-resistant staphylococci. Another interesting molecule is 1.037, a synthetic peptide of 9 amino acids, which can inhibit biofilm formation in staphylococci by increasing bacterial motility and reducing the expression of genes responsible for biofilm formation and QS but has little antimicrobial activity [161]. Even in this study, data on an in vivo model of wound healing are lacking. In our opinion, the combination of AMPs and other molecules with a different mechanism of action could be, in the next future, the solution for difficult multifailure conditions.
We also want to mention bacteriocins, ribosomally synthesized bacterial peptides with important activity against staphylococcal biofilm. For example, Garvicin KS and microcin P1 showed in vitro anti-biofilm activity against both MSSA and MRSA. It was also observed that the combination of these two bacteriocins reduced MRSA resistance to penicillin G, revealing both antibiofilm and anti-MRSA activity [167]. The use of these molecules may therefore help to make antibiotics effective against resistant strains, although there is a lack of studies on wound healing.
With regard to S. epidermidis, one study [168] showed that Nisin A M17Q, a bacteriocin-derivative produced by Lactococcus lactis, can inhibit biofilm formation and the growth of S. epidermidis in an in vitro wound model more than wild type Nisin A. [168] This is one of the few studies considering the action of biofilm-producing S. epidermidis on wounds, although in an in vitro model [168]. Further animal studies and comparisons with other promising molecules are needed to assess its effective action on wound healing.
Other molecules have promising actions against staphylococci in the literature, in particular, defensins, proteins that mediate the innate immunity of organisms against bacteria. One example is the defensin-like peptide (DLP)-P2, a fungal-derived molecule that was shown to be effective in controlling multidrug-resistant S. aureus infections and biofilm formation both in vitro and in a mouse model with peritoneal infection [169]. A study of wound healing has not been carried out, but the prospects for using this molecule are interesting, especially in cases with extreme resistance to antibiotics.
Regarding the most common topical dressings already known, in a study by Brackman et al. [170] their anti-biofilm efficacy was evaluated in an in vitro model of S. aureus and S. epidermidis. Referring to inhibition of biofilm formation, dressings without antimicrobial agents and with only alginate fibres, carboxymethylcellulose (CMC), cotton, or hydrocellular foam proved ineffective or poorly effective. Conversely, dressings with antimicrobial agents such as povidone-iodine (PVP-iodine), hydroactive colloid gel, silver dihydrogen citrate, fusidic acid (20 mg/g) and polyhexanide have been effective in preventing biofilm formation. Finally, dressings containing ionic silver, metallic silver and silver sulphate, silver sulphadiazine, PVP-iodine (10 g/100 mL) and ozonated olive oil were effective in inhibiting biofilm formation.
With regard to biofilm-eradicating activity, fusidic acid, ozonated olive oil and silver dihydrogen citrate were effective for both S. aureus and S. epidermidis, while Betadine (0.1% w/w) with polyhexanide (0.1% w/w) and Hydroactive colloid gel were effective only for S. epidermidis. However, these dressings alone are often not sufficient to eradicate infection, and the possibility of combining an antibiotic or AMPs may improve the outcome of wound healing. Again, in vivo studies are required to evaluate efficacy. We suggest the early use of dressings with silver derivates, PVP-iodine or ozonate olive oil, considering their widespread availability.
An important role in the treatment of wounds infected with biofilm-producing staphylococcal strains is played by APDT, which was proven, in association with RLP068/CI, [152] to be faster than teicoplanin in reducing bacterial load and more effective in wound healing on human fibroblast cultures. In vivo studies are needed to evaluate its efficacy in wound healing. In the literature, we found a case report of APDT with ALA on a patient with a chronic ulcer infected with S. aureus resistant to conventional therapy, with excellent clinical results on wound healing and a long period without relapse (29 months) [158]. This suggests that in the most difficult cases, APDT may be a valid treatment option. There are promising studies in the literature demonstrating in vitro anti-biofilm staphylococcal action of APDT with various photosensitisers and blue light, [171,172,173,174,175,176,177,178,179] but which are lacking in vivo histological evaluation of wound healing.
Other promising treatments have not been investigated because there is no evaluation of wound healing, such as antibodies against staphylococcal antigens [174,175,176,177,178], nanotechnology [179,180,181,182], and new genetic approaches [183,184]. In addition, bacteriophages are viruses with a predatory action against specific bacteria [185]. Their effectiveness in eradicating biofilms has been evaluated in combination with other molecules, such as antibiotics [186,187,188]. It was also shown that the bacteriophage phiIPLA-RODI can be useful in eradicating S. aureus biofilms 24 h per day both in vitro and in an ex vivo pig model, especially in combination with a phage-derived lytic protein CHAPSH3b. The latter resulted in a reduction in the S. aureus population up to 7 h after exposure, followed by bacteriophage activity, which limits bacterial regrowth [185]. All of these methods have been shown to have an anti-biofilm effect, but no studies were conducted to assess their impact on wound healing.
Currently, only RIP and APDT with ALA were used in human patients, albeit only in case reports. In our opinion, these are promising and will help to overcome bacterial resistance, especially in difficult multifailure cases. We hope that other therapies will soon be proved in humans, allowing the treatment of chronic wounds with difficult tissue healing.

4. Materials and Methods

A narrative review of the literature was performed on Pubmed using as keywords individually or in combination: antistaphylococcal drugs, antistaphylococcal peptides, antistaphylococcal therapy, staphylococcal skin infection, staphylococcal, treatment, quorum sensing, biofilm, and wound healing. The aim of this review is to bring together the latest evidence on new and old anti-staphylococcal therapies, assessing their anti-biofilm properties and their effect on skin wound healing. We gave more consideration to studies performed in vivo with a histological evaluation of wound healing. Only English-language studies were included.
We did not consider a timeframe or a time limit, since even if a molecule was old, it would still be possible to use it to overcome growing bacterial resistance, maybe in combination with an antibiotic.

5. Conclusions

From our review of the literature, prevention and eradication of biofilm is an important therapeutic target in acute and chronic infected skin wounds both to achieve better wound healing and to overcome antibiotic resistance.
The best therapeutic and tissue-repair effect is obtained by combining systemic antibiotic therapy with a local agent that can act directly on the biofilm by breaking it down or preventing its formation. One of the limitations of the reviewed data is that most of them refer to in vitro studies or animal models, and human studies lack adequate sample size.
In conclusion, targeting biofilm can be an effective strategy not only to overcome antibiotic resistance conditions but also to significantly improve the outcome of wound healing.

Author Contributions

Conceptualization, O.S. and G.R. (Giulio Rizzetto); methodology, O.S. and G.R. (Giulio Rizzetto); software, G.R. (Giulio Rizzetto); validation, O.C., A.G. and G.R. (Giulia Radi); data curation, G.R. (Giulio Rizzetto) and E.M.; writing—original draft preparation, G.R. (Giulio Rizzetto); writing—review and editing, O.S.; visualization, O.S.; supervision, A.O.; project administration, A.O. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Humphreys, H.; Becker, K.; Dohmen, P.; Petrosillo, N.; Spencer, M.; van Rijen, M.; Wechsler-Fördös, A.; Pujol, M.; Dubouix, A.; Garau, J. Staphylococcus aureus and surgical site infections: Benefits of screening and decolonization before surgery. J. Hosp. Infect. 2016, 94, 295–304. [Google Scholar] [CrossRef]
  2. James, G.A.; Swogger, E.; Wolcott, R.; Pulcini, E.D.; Secor, P.; Sestrich, J.; Costerton, J.W.; Stewart, P.S. Biofilms in chronic wounds. Wound Repair Regen. 2007, 16, 37–44. [Google Scholar] [CrossRef] [PubMed]
  3. E Dowd, S.; Sun, Y.; Secor, P.R.; Rhoads, D.D.; Wolcott, B.M.; A James, G.; Wolcott, R.D. Survey of bacterial diversity in chronic wounds using Pyrosequencing, DGGE, and full ribosome shotgun sequencing. BMC Microbiol. 2008, 8, 43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Diekema, D.J.; Pfaller, M.A.; Schmitz, F.J.; Smayevsky, J.; Bell, J.; Jones, R.N.; Beach, M.; SENTRY Partcipants Group. Survey of infections due to Staphylococcus species: Frequency of occurrence and antimicrobial susceptibility of isolates collected in the United States, Canada, Latin America, Europe, and the Western Pacific region for the SENTRY Antimicrobial Surveillance Program, 1997–1999. Clin. Infect. Dis. 2001, 32 (Suppl. 2), S114–S132. [Google Scholar] [PubMed] [Green Version]
  5. Fridkin, S.K.; Hageman, J.C.; Morrison, M.; Sanza, L.T.; Como-Sabetti, K.; Jernigan, J.A.; Harriman, K.; Harrison, L.H.; Lynfield, R.; Farley, M.M. Methicillin-Resistant Staphylococcus aureus Disease in Three Communities. N. Engl. J. Med. 2005, 352, 1436–1444. [Google Scholar] [CrossRef] [PubMed]
  6. Malone, M.; Bjarnsholt, T.; McBain, A.J.; James, G.A.; Stoodley, P.; Leaper, D.; Tachi, M.; Schultz, G.; Swanson, T.; Wolcott, R.D. The prevalence of biofilms in chronic wounds: A systematic review and meta-analysis of published data. J. Wound Care 2017, 26, 20–25. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Norris, G.R.; Checketts, J.X.; Scott, J.T.; Vassar, M.; Norris, B.L.; Giannoudis, P.V. Prevalence of Deep Surgical Site Infection After Repair of Periarticular Knee Frac-tures: A Systematic Review and Meta-analysis. JAMA Netw. Open 2019, 2, e199951. [Google Scholar] [CrossRef]
  8. Wolcott, R. Disrupting the biofilm matrix improves wound healing outcomes. J. Wound Care 2015, 24, 366–371. [Google Scholar] [CrossRef] [PubMed]
  9. Percival, S.; Mccarty, S.M.; A Lipsky, B. Biofilms and Wounds: An Overview of the Evidence. Adv. Wound Care 2015, 4, 373–381. [Google Scholar] [CrossRef] [Green Version]
  10. Barki, K.G.; DAS, A.; Dixith, S.; Das Ghatak, P.; Mathew-Steiner, S.; Schwab, E.; Khanna, S.; Wozniak, D.J.; Roy, S.; Sen, C.K. Electric Field Based Dressing Disrupts Mixed-Species Bacterial Biofilm Infection and Restores Functional Wound Healing. Ann. Surg. 2019, 269, 756–766. [Google Scholar] [CrossRef] [PubMed]
  11. Roy, S.; Elgharably, H.; Sinha, M.; Ganesh, K.; Chaney, S.; Mann, E.; Miller, C.; Khanna, S.; Bergdall, V.K.; Powell, H.; et al. Mixed-species biofilm compromises wound healing by disrupting epidermal barrier function. J. Pathol. 2014, 233, 331–343. [Google Scholar] [CrossRef]
  12. Kowalewska-Grochowska, K.; Richards, R.; Moysa, G.; Lam, K.; Costerton, J.; King, E. Guidewire Catheter Change in Central Venous Catheter Biofilm Formation in a Burn Population. Chest 1991, 100, 1090–1095. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Wolcott, R.; Costerton, J.W.; Raoult, D.; Cutler, S.J. The polymicrobial nature of biofilm infection. Clin. Microbiol. Infect. 2013, 19, 107–112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Kranjec, C.; Morales Angeles, D.; Torrissen Mårli, M.; Fernández, L.; García, P.; Kjos, M.; Diep, D.B. Staphylococcal Biofilms: Challenges and Novel Therapeutic Perspectives. Antibiotics 2021, 10, 131. [Google Scholar] [CrossRef]
  15. Coates, R.; Moran, J.; Horsburgh, M.J. Staphylococci: Colonizers and pathogens of ma skin. Future Microbiol. 2014, 9, 75–91. [Google Scholar] [CrossRef]
  16. Schultz, G.S.; Sibbald, R.G.; Falanga, V.; Ayello, E.A.; Dowsett, C.; Harding, K.; Romanelli, M.; Ds, M.C.S.; Teot, L.; Vanscheidt, W. Wound bed preparation: A systematic approach to wound management. Wound Repair Regen. 2003, 11, S1–S28. [Google Scholar] [CrossRef] [PubMed]
  17. Roy, S.; Santra, S.; Das, A.; Dixith, S.; Sinha, M.; Ghatak, S.; Ghosh, N.; Banerjee, P.; Khanna, S.; Mathew-Steiner, S.; et al. Staphylococcus aureus Biofilm Infection Compromises Wound Healing by Causing Deficiencies in Granulation Tissue Collagen. Ann. Surg. 2020, 271, 1174–1185. [Google Scholar] [CrossRef]
  18. Sen, C.K.; Gordillo, G.M.; Roy, S.; Kirsner, R.; Lambert, L.; Hunt, T.K.; Gottrup, F.; Gurtner, G.C.; Longaker, M.T. Human skin wounds: A major and snowballing threat to public health and the economy. Wound Repair Regen. 2009, 17, 763–771. [Google Scholar] [CrossRef] [Green Version]
  19. Trengove, N.J.; Stacey, M.C.; Macauley, S.; Bennett, N.; Gibson, J.; Burslem, F.; Murphy, G.; Schultz, G. Analysis of the acute and chronic wound environments: The role of proteases and their inhibitors. Wound Repair Regen. 1999, 7, 442–452. [Google Scholar] [CrossRef]
  20. Rogers, K.L.; Fey, P.D.; Rupp, M.E. Coagulase-Negative Staphylococcal Infections. Infect. Dis. Clin. N. Am. 2009, 23, 73–98. [Google Scholar] [CrossRef]
  21. Argemi, X.; Hansmann, Y.; Riegel, P.; Pre’vost, G. Is Staphylococcus lugdunensis significant in clinical samples? J. Clin. Microbiol. 2017, 55, 3167–3174. [Google Scholar] [CrossRef] [Green Version]
  22. Jenkins, T.C.; Knepper, B.C.; Jason Moore, S.; Saveli, C.C.; Pawlowski, S.W.; Perlman, D.M.; McCollister, B.D.; Burman, W.J. Comparison of the microbiology and anti-biotic treatment among diabetic and nondiabetic patients hospitalized for cellulitis or cutaneous abscess. J. Hosp. Med. 2014, 9, 788–794. [Google Scholar] [CrossRef] [Green Version]
  23. Nguyen, K.T.; Seth, A.K.; Hong, S.J.; Geringer, M.R.; Xie, P.; Leung, K.P.; Mustoe, T.A.; Galiano, R.D. Deficient cytokine expression and neutrophil oxidative burst con-tribute to impaired cutaneous wound healing in diabetic biofilm-containing chronic wounds. Wound Repair. Regen. 2013, 21, 833–841. [Google Scholar] [CrossRef]
  24. Ammons, M.C. Anti-Biofilm Strategies and the Need for Innovations in Wound Care. Recent Patents Anti-Infective Drug Discov. 2010, 5, 10–17. [Google Scholar] [CrossRef]
  25. Percival, S.L.; Hill, K.E.; Williams, D.; Hooper, S.J.; Thomas, D.; Costerton, J.W. A review of the scientific evidence for biofilms in wounds. Wound Repair Regen. 2012, 20, 647–657. [Google Scholar] [CrossRef] [PubMed]
  26. Malik, A.; Mohammad, Z.; Ahmad, J. The diabetic foot infections: Biofilms and antimicrobial resistance. Diabetes Metab. Syndr. Clin. Res. Rev. 2013, 7, 101–107. [Google Scholar] [CrossRef] [PubMed]
  27. Percival, S.L.; Hill, K.E.; Malic, S.; Thomas, D.; Williams, D. Antimicrobial tolerance and the significance of persister cells in recalcitrant chronic wound biofilms. Wound Repair Regen. 2011, 19, 1–9. [Google Scholar] [CrossRef] [PubMed]
  28. Bjarnsholt, T.; Kirketerp-Møller, K.; Jensen, P.; Madsen, K.G.; Phipps, R.K.; Krogfelt, K.A.; Høiby, N.; Givskov, M. Why chronic wounds will not heal: A novel hypothesis. Wound Repair Regen. 2008, 16, 2–10. [Google Scholar] [CrossRef] [PubMed]
  29. Watters, C.; DeLeon, K.; Trivedi, U.; Griswold, J.A.; Lyte, M.; Hampel, K.J.; Wargo, M.; Rumbaugh, K.P. Pseudomonas aeruginosa biofilms perturb wound resolution and antibiotic tolerance in diabetic mice. Med. Microbiol. Immunol. 2012, 202, 131–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Ortiz Balbuena, J.; Garcıa Madero, R.; Segovia Gomez, T.; Cantero Caballero, M.; Sánchez Romero, I.; Ramos Martínez, A. Microbiology of pressure and vascular ulcer infections. Rev. Esp. Geriatr. Gerontol. 2015, 50, 5–8. [Google Scholar] [CrossRef] [PubMed]
  31. Church, D.; Elsayed, S.; Reid, O.; Winston, B.; Lindsay, R. Burn wound infections. Clin. Microbiol. Rev. 2006, 19, 403–434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Davis, S.C.; Ricotti, C.; Cazzaniga, A.; Welsh, E.; Eaglstein, W.H.; Mertz, P.M. Microscopic and physiologic evidence for biofilm-associated wound colonization in vivo. Wound Repair Regen. 2008, 16, 23–29. [Google Scholar] [CrossRef] [PubMed]
  33. Natsis, N.E.; Cohen, P.R. Coagulase-Negative Staphylococcus Skin and Soft Tissue Infections. Am. J. Clin. Dermatol. 2018, 19, 671–677. [Google Scholar] [CrossRef] [PubMed]
  34. Huda, S.; Azmiza, S.J.; Tengku, J.; Rosni, I. A review of Staphylococcal cassette chromosome mec (SCCmec) types in coagulasenegative staphylococci (CoNS) species. Malays. J. Med. Sci. 2017, 24, 7–18. [Google Scholar]
  35. Mohammed, Y.H.E.; Manukumar, H.; Rakesh, K.; Karthik, C.; Mallu, P.; Qin, H.-L. Vision for medicine: Staphylococcus aureus biofilm war and unlocking key’s for anti-biofilm drug development. Microb. Pathog. 2018, 123, 339–347. [Google Scholar] [CrossRef] [PubMed]
  36. Subrt, N.; Mesak, L.R.; Davies, J. Modulation of virulence gene expression by cell wall active antibiotics in Staphylococcus aureus. J. Antimicrob. Chemother. 2011, 66, 979–984. [Google Scholar] [CrossRef] [PubMed]
  37. Mirani, Z.A.; Jamil, N. Effect of sub-lethal doses of vancomycin and oxacillin on biofilm formation by vancomycin intermediate resistant Staphylococcus aureus. J. Basic Microbiol. 2010, 51, 191–195. [Google Scholar] [CrossRef] [PubMed]
  38. Haddadin, R.N.; Saleh, S.; Al-Adham, I.S.; Buultjens, T.E.; Collier, P.J. The effect of subminimal inhibitory concentrations of antibiotics on virulence factors expressed by Staphylococcus aureus biofilms. J. Appl. Microbiol. 2010, 108, 1281–1291. [Google Scholar] [CrossRef]
  39. Frank, K.L.; Reichert, E.J.; Piper, K.E.; Patel, R. In Vitro Effects of Antimicrobial Agents on Planktonic and Biofilm Forms of Staphylococcus lugdunensis Clinical Isolates. Antimicrob. Agents Chemother. 2007, 51, 888–895. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Kaplan, J.B.; Izano, E.A.; Gopal, P.; Karwacki, M.T.; Kim, S.; Bose, J.L.; Bayles, K.W.; Horswill, A.R. Low Levels of β-Lactam Antibiotics Induce Extracellular DNA Release and Biofilm Formation in Staphylococcus aureus. mBio 2012, 3, e00198-12. [Google Scholar] [CrossRef] [Green Version]
  41. Kaplan, J.B.; Jabbouri, S.; Sadovskaya, I. Extracellular DNA-dependent biofilm formation by Staphylococcus epidermidis RP62A in response to subminimal inhibitory concentrations of antibiotics. Res. Microbiol. 2011, 162, 535–541. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Pérez-Giraldo, C.; Rodríguez-Benito, A.; Morán, F.J.; Hurtado, C.; Blanco, M.T.; Gómez-García, A.C. In-vitro slime production by Staphylococcus epidermidis in presence of subinhibitory concentrations of ciprofloxacin, ofloxacin and sparfloxacin. J. Antimicrob. Chemother. 1994, 33, 845–848. [Google Scholar] [CrossRef] [PubMed]
  43. Majidpour, A.; Fathizadeh, S.; Afshar, M.; Rahbar, M.; Boustanshenas, M.; Heidarzadeh, M.; Arbabi, L. Dose-Dependent Effects of Common Antibiotics Used to Treat Staphylococcus aureus on Biofilm Formation. Iran J. Pathol. 2017, 12, 362–370. [Google Scholar] [CrossRef] [PubMed]
  44. Kirmusaoglu, S. Improved β-Lactam Susceptibility Against ica-Dependent Biofilm-Embedded Staphylococcus aureus by 2-Aminothiazole. Clin. Lab. 2020, 66. [Google Scholar] [CrossRef]
  45. Izano, E.A.; Amarante, M.A.; Kher, W.B.; Kaplan, J.B. Differential roles of poly-N acetylglucosamine surface polysaccharide and extracellular DNA in Staphylococcus aureus and Staphylococcus epidermidis biofilms. Appl. Environ. Microbiol. 2008, 74, 470–476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Rice, K.C.; Mann, E.E.; Endres, J.L.; Weiss, E.C.; Cassat, J.E.; Smeltzer, M.; Bayles, K.W. The cidA murein hydrolase regulator contributes to DNA release and biofilm development in Staphylococcus aureus. Proc. Natl. Acad. Sci. USA 2007, 104, 8113–8118. [Google Scholar] [CrossRef] [Green Version]
  47. Houston, P.; Rowe, S.E.; Pozzi, C.; Waters, E.M.; O’Gara, J.P. Essential Role for the Major Autolysin in the Fibronectin-Binding Protein-Mediated Staphylococcus aureus Biofilm Phenotype. Infect. Immun. 2011, 79, 1153–1165. [Google Scholar] [CrossRef] [Green Version]
  48. Boles, B.R.; Horswill, A.R. agr-Mediated Dispersal of Staphylococcus aureus Biofilms. PLOS Pathog. 2008, 4, e1000052. [Google Scholar] [CrossRef]
  49. Kiedrowski, M.; Kavanaugh, J.S.; Malone, C.L.; Mootz, J.M.; Voyich, J.M.; Smeltzer, M.; Bayles, K.W.; Horswill, A.R. Nuclease Modulates Biofilm Formation in Community-Associated Methicillin-Resistant Staphylococcus aureus. PLoS ONE 2011, 6, e26714. [Google Scholar] [CrossRef] [Green Version]
  50. Fujimura, S.; Sato, T.; Hayakawa, S.; Kawamura, M.; Furukawa, E.; Watanabe, A. Antimicrobial efficacy of combined clarithromycin plus daptomycin against biofilms-formed methicillin-resistant Staphylococcus aureus on titanium medical devices. J. Infect. Chemother. 2015, 21, 756–759. [Google Scholar] [CrossRef]
  51. Fujimura, S.; Sato, T.; Kikuchi, T.; Zaini, J.; Gomi, K.; Watanabe, A. Efficacy of clarithromycin plus vancomycin in mice with implant-related infection caused by bio-film-forming Staphylococcus aureus. J. Orthop. Sci. 2009, 14, 658–661. [Google Scholar] [CrossRef]
  52. Yamasaki, O.; Akiyama, H.; Toi, Y.; Arata, J. A combination of roxithromycin and imipenem as an antimicrobial strategy against biofilms formed by Staphylococcus aureus. J. Antimicrob. Chemother. 2001, 48, 573–577. [Google Scholar] [CrossRef] [Green Version]
  53. Pace, J.L.; Yang, G. Glycopeptides: Update on an old successful antibiotic class. Biochem. Pharmacol. 2006, 71, 968–980. [Google Scholar] [CrossRef] [PubMed]
  54. Ghiselli, R.; Cirioni, O.; Giacometti, A.; Scalise, A.; Simonetti, O.; Mocchegiani, F.; Orlando, F.; Goteri, G.; Della Vittoria, A.; Filosa, A.; et al. Comparative Efficacy of Topical Versus Systemic Teicoplanin in Experimental Model of Wound Infections. J. Surg. Res. 2008, 144, 74–81. [Google Scholar] [CrossRef] [PubMed]
  55. Hamed, K.; Gonzalez-Ruiz, A.; Seaton, A. Daptomycin: An evidence-based review of its role in the treatment of Gram-positive infections. Infect. Drug Resist. 2016, 9, 47–58. [Google Scholar] [CrossRef] [Green Version]
  56. He, W.-Q.; Zhang, Y.; Chen, H.; Zhao, C.; Wang, H. Efficacy and safety of daptomycin for the treatment of infectious disease: A meta-analysis based on randomized controlled trials. J. Antimicrob. Chemother. 2014, 69, 3181–3189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Pierpaoli, E.; Orlando, F.; Cirioni, O.; Simonetti, O.; Giacometti, A.; Provinciali, M. Supplementation with tocotrienols from Bixa orellana improves the in vivo efficacy of daptomycin against methicillin-resistant Staphylococcus aureus in a mouse model of infected wound. Phytomedicine 2017, 36, 50–53. [Google Scholar] [CrossRef] [PubMed]
  58. Robbel, L.; Marahiel, M.A. Daptomycin, a Bacterial Lipopeptide Synthesized by a Nonribosomal Machinery. J. Biol. Chem. 2010, 285, 27501–27508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Boudjemaa, R.; Briandet, R.; Revest, M.; Jacqueline, C.; Caillon, J.; Fontaine-Aupart, M.-P.; Steenkeste, K. New Insight into Daptomycin Bioavailability and Localization in Staphylococcus aureus Biofilms by Dynamic Fluorescence Imaging. Antimicrob. Agents Chemother. 2016, 60, 4983–4990. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Simonetti, O.; Lucarini, G.; Orlando, F.; Pierpaoli, E.; Ghiselli, R.; Provinciali, M.; Castelli, P.; Guerrieri, M.; Di Primio, R.; Offidani, A.; et al. Role of Daptomycin on Burn Wound Healing in an Animal Methicillin-Resistant Staphylococcus aureus Infection Model. Antimicrob. Agents Chemother. 2017, 61, e00606-17. [Google Scholar] [CrossRef] [Green Version]
  61. Silvestri, C.; Cirioni, O.; Arzeni, D.; Ghiselli, R.; Simonetti, O.; Orlando, F.; Ganzetti, G.; Staffolani, S.; Brescini, L.; Provinciali, M.; et al. In vitro activity and in vivo efficacy of tigecycline alone and in combination with daptomycin and rifampin against Gram-positive cocci isolated from surgical wound infection. Eur. J. Clin. Microbiol. Infect. Dis. 2011, 31, 1759–1764. [Google Scholar] [CrossRef] [PubMed]
  62. Szczuka, E.; Kaznowski, A. Antimicrobial activity of tigecycline alone or in combination with rifampin against Staphylococcus epidermidis in biofilm. Folia Microbiol. 2014, 59, 283–288. [Google Scholar] [CrossRef] [PubMed]
  63. Rose, W.E.; Poppens, P.T. Impact of biofilm on the in vitro activity of vancomycin alone and in combination with tigecycline and rifampicin against Staphylococcus aureus. J. Antimicrob. Chemother. 2008, 63, 485–488. [Google Scholar] [CrossRef] [Green Version]
  64. Aybar, Y.; Ozaras, R.; Besirli, K.; Engin, E.; Karabulut, E.; Salihoglu, T.; Mete, B.; Tabak, F.; Mert, A.; Tahan, G.; et al. Efficacy of tigecycline and vancomycin in experi-mental catheter-related Staphylococcus epidermidis infection: Microbiological and electron microscopic analysis of biofilm. Int. J. Antimicrob. Agents 2012, 39, 338–342. [Google Scholar] [CrossRef] [PubMed]
  65. Simonetti, O.; Morroni, G.; Ghiselli, R.; Orlando, F.; Brenciani, A.; Xhuvelaj, L.; Provinciali, M.; Offidani, A.; Guerrieri, M.; Giacometti, A.; et al. In vitro and in vivo activity of fosfomycin alone and in combination with rifampin and tigecycline against Gram-positive cocci isolated from surgical wound infections. J. Med. Microbiol. 2018, 67, 139–143. [Google Scholar] [CrossRef] [PubMed]
  66. Simonetti, O.; Cirioni, O.; Lucarini, G.; Orlando, F.; Ghiselli, R.; Silvestri, C.; Brescini, L.; Rocchi, M.; Provinciali, M.; Guerrieri, M.; et al. Tigecycline accelerates staphylococcal-infected burn wound healing through matrix metalloproteinase-9 modulation. J. Antimicrob. Chemother. 2011, 67, 191–201. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Simonetti, O.; Rizzetto, G.; Molinelli, E.; Cirioni, O.; Offidani, A. Review: A Safety Profile of Dalbavancin for On- and Off-Label Utilization. Ther. Clin. Risk Manag. 2021, 17, 223–232. [Google Scholar] [CrossRef] [PubMed]
  68. Silva, V.; Antão, H.S.; Guimarães, J.; Prada, J.; Pires, I.; Martins, A.; Maltez, L.; E Pereira, J.; Capelo, J.L.; Igrejas, G.; et al. Efficacy of dalbavancin against MRSA biofilms in a rat model of orthopaedic implant-associated infection. J. Antimicrob. Chemother. 2020, 75, 2182–2187. [Google Scholar] [CrossRef] [PubMed]
  69. Knafl, D.; Tobudic, S.; Cheng, S.C.; Bellamy, D.R.; Thalhammer, F. Dalbavancin reduces biofilms of methicillin-resistant Staphylococcus aureus (MRSA) and methicillin-resistant Staphylococcus epidermidis (MRSE). Eur. J. Clin. Microbiol. Infect. Dis. 2016, 36, 677–680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Simonetti, O.; Lucarini, G.; Morroni, G.; Orlando, F.; Lazzarini, R.; Zizzi, A.; Brescini, L.; Provinciali, M.; Giacometti, A.; Offidani, A.; et al. New Evidence and Insights on Dalbavancin and Wound Healing in a Mouse Model of Skin Infection. Antimicrob. Agents Chemother. 2020, 64, e02062-19. [Google Scholar] [CrossRef] [PubMed]
  71. Warrier, A.; Satyamoorthy, K.; Murali, T.S. Quorum-sensing regulation of virulence factors in bacterial biofilm. Future Microbiol. 2021, 16, 1003–1021. [Google Scholar] [CrossRef] [PubMed]
  72. Recsei, P.; Kreiswirth, B.; O’Reilly, M.; Schlievert, P.; Gruss, A.; Novick, R.P. Regulation of exoprotein gene expression in Staphylococcus aureus by agr. Mol. Gen. Genet. 1986, 202, 58–61. [Google Scholar] [CrossRef] [PubMed]
  73. Rooijakkers, S.H.; van Kessel, K.P.; van Strijp, J.A. Staphylococcal innate immune evasion. Trends Microbiol. 2005, 13, 596–601. [Google Scholar] [CrossRef] [PubMed]
  74. Foster, T.J.; Hook, M. Surface protein adhesins of Staphylococcus aureus. Trends Microbiol. 1996, 6, 484–488. [Google Scholar] [CrossRef]
  75. Otto, M. Staphylococcus aureus toxins. Curr. Opin. Microbiol. 2014, 17, 32–37. [Google Scholar] [CrossRef] [Green Version]
  76. Zhang, L.; Lin, J.; Ji, G. Membrane Anchoring of the AgrD N-terminal Amphipathic Region Is Required for Its Processing to Produce a Quorum-sensing Pheromone in Staphylococcus aureus. J. Biol. Chem. 2004, 279, 19448–19456. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Thoendel, M.; Kavanaugh, J.S.; Flack, C.E.; Horswill, A.R. Peptide Signaling in the Staphylococci. Chem. Rev. 2010, 111, 117–151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Ji, G.; Beavis, R.C.; Novick, R. Cell density control of staphylococcal virulence mediated by an octapeptide pheromone. Proc. Natl. Acad. Sci. USA 1995, 92, 12055–12059. [Google Scholar] [CrossRef] [Green Version]
  79. Otto, M.; Sussmuth, R.; Jung, G.; Gotz, F. Structure of the pheromone peptide of the Staphylococcus epidermidis agr system. FEBS Lett. 1998, 424, 89–94. [Google Scholar] [CrossRef] [Green Version]
  80. Krucke, G.W.; Grimes, D.E.; Grimes, R.M.; Dang, T.D. Antibiotic resistance in Staphylococcus aureus–containing cutaneous abscesses of patients with HIV. Am. J. Emerg. Med. 2009, 27, 344–347. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Balaban, N.; Cirioni, O.; Giacometti, A.; Ghiselli, R.; Braunstein, J.B.; Silvestri, C.; Mocchegiani, F.; Saba, V.; Scalise, G. Treatment of Staphylococcus aureus biofilm in-fection by the quorum-sensing inhibitor RIP. Antimicrob. Agents Chemother. 2007, 51, 2226–2229. [Google Scholar] [CrossRef] [Green Version]
  82. Kiran, M.D.; Adikesavan, N.V.; Cirioni, O.; Giacometti, A.; Silvestri, C.; Scalise, G.; Ghiselli, R.; Saba, V.; Orlando, F.; Shoham, M.; et al. Discovery of a quorum-sensing inhibitor of drug-resistant Staphylococcal infections by structure-based virtual screening. Mol. Pharmacol. 2008, 73, 1578–1586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Giacometti, A.; Cirioni, O.; Ghiselli, R.; Dell’Acqua, G.; Orlando, F.; D’Amato, G.; Mocchegiani, F.; Silvestri, C.; Del Prete, M.S.; Rocchi, M.; et al. RNAIII-inhibiting peptide improves efficacy of clinically used antibiotics in a murine model of Staphylococcal sepsis. Peptides. 2005, 26, 169–175. [Google Scholar] [CrossRef] [PubMed]
  84. Cirioni, O.; Ghiselli, R.; Minardi, D.; Orlando, F.; Mocchegiani, F.; Silvestri, C.; Muzzonigro, G.; Saba, V.; Scalise, G.; Balaban, N.; et al. RNAIII-Inhibiting Peptide Affects Biofilm Formation in a Rat Model of Staphylococcal Ureteral Stent Infection. Antimicrob. Agents Chemother. 2007, 51, 4518–4520. [Google Scholar] [CrossRef] [Green Version]
  85. Simonetti, O.; Cirioni, O.; Mocchegiani, F.; Cacciatore, I.; Silvestri, C.; Baldassarre, L.; Orlando, F.; Castelli, P.; Provinciali, M.; Vivarelli, M.; et al. The Efficacy of the Quorum Sensing Inhibitor FS8 and Tigecycline in Preventing Prosthesis Biofilm in an Animal Model of Staphylococcal Infection. Int. J. Mol. Sci. 2013, 14, 16321–16332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Ciulla, M.; Di Stefano, A.; Marinelli, L.; Cacciatore, I.; Di Biase, G. RNAIII Inhibiting Peptide (RIP) and Derivatives as Potential Tools for the Treatment of S. aureus Biofilm Infections. Curr. Top. Med. Chem. 2019, 18, 2068–2079. [Google Scholar] [CrossRef] [PubMed]
  87. Yarwood, J.M.; Schlievert, P.M. Quorum Sensing in Staphylococcus Infections. J. Clin. Investig. 2003, 112, 1620–1625. [Google Scholar] [CrossRef] [PubMed]
  88. Anguita-Alonso, P.; Giacometti, A.; Cirioni, O.; Ghiselli, R.; Orlando, F.; Saba, V.; Scalise, G.; Sevo, M.; Tuzova, M.; Patel, R.; et al. RNAIII-inhibiting-peptide-loaded polymethylmethacrylate prevents in vivo Staphylococcus aureus biofilm formation. Antimicrob. Agents Chemother. 2007, 51, 2594–2596. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Balaban, N.; Giacometti, A.; Cirioni, O.; Gov, Y.; Ghiselli, R.; Mocchegiani, F.; Viticchi, C.; Del Prete, M.S.; Saba, V.; Scalise, G.; et al. Use of the Quorum-Sensing Inhibitor RNAIII-Inhibiting Peptide to Prevent Biofilm Formation In Vivo by Drug-Resistant Staphylococcus epidermidis. J. Infect. Dis. 2003, 187, 625–630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Balaban, N.; Goldkorn, T.; Nhan, R.T.; Dang, L.B.; Scott, S.; Ridgley, R.M.; Rasooly, A.; Wright, S.C.; Larrick, J.W.; Rasooly, R.; et al. Autoinducer of Virulence As a Target for Vaccine and Therapy Against Staphylococcus aureus. Science 1998, 280, 438–440. [Google Scholar] [CrossRef] [PubMed]
  91. Cirioni, O.; Giacometti, A.; Ghiselli, R.; Dell’Acqua, G.; Orlando, F.; Mocchegiani, F.; Silvestri, C.; Licci, A.; Saba, V.; Scalise, G.; et al. RNAIII-inhibiting peptide signifi-cantly reduces bacterial load and enhances the effect of antibiotics in the treatment of central venous catheter-associated Staphylococcus aureus infections. J. Infect. Dis. 2006, 193, 180–186. [Google Scholar] [CrossRef] [Green Version]
  92. Schierle, C.F.; De La Garza, M.; Mustoe, T.A.; Galiano, R.D. Staphylococcal biofilms impair wound healing by delaying reepithelialization in a murine cutaneous wound model. Wound Repair Regen. 2009, 17, 354–359. [Google Scholar] [CrossRef] [PubMed]
  93. Simonetti, O.; Cirioni, O.; Ghiselli, R.; Goteri, G.; Scalise, A.; Orlando, F.; Silvestri, C.; Riva, A.; Saba, V.; Madanahally, K.D.; et al. RNAIII-Inhibiting Peptide Enhances Healing of Wounds Infected with Methicillin-Resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 2008, 52, 2205–2211. [Google Scholar] [CrossRef] [Green Version]
  94. Naldini, A.; Carraro, F. Role of inflammatory mediators in angiogenesis. Curr. DrugTargets Inflamm. Allergy 2005, 4, 3–8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Wolcott, R.D. Clinical Wound Healing Using Signal Inhibitors. In Control of Biofilm Infections by Signal Manipulation; Naomi, B., Ed.; Springer: Berlin/Heidelberg, Germany, 2008; Volume 2, pp. 157–170. [Google Scholar]
  96. Khodaverdian, V.; Pesho, M.; Truitt, B.; Bollinger, L.; Patel, P.; Nithianantham, S.; Yu, G.; Delaney, E.; Jankowsky, E.; Shoham, M. Discovery of antivirulence agents against methicillin-resistant Staphylococcus aureus. Antimicrob. Agents Chemother. 2013, 57, 3645–3652. [Google Scholar] [CrossRef] [Green Version]
  97. Kuo, D.; Yu, G.; Hoch, W.; Gabay, D.; Long, L.; Ghannoum, M.; Nagy, N.; Harding, C.V.; Viswanathan, R.; Shoham, M. Novel Quorum-Quenching Agents Promote Methicillin-Resistant Staphylococcus aureus (MRSA) Wound Healing and Sensitize MRSA to β-Lactam Antibiotics. Antimicrob. Agents Chemother. 2014, 59, 1512–1518. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Baldassarre, L.; Fornasari, E.; Cornacchia, C.; Cirioni, O.; Silvestri, C.; Castelli, P.; Giocometti, A.; Cacciatore, I. Discovery of novel RIP derivatives by alanine scanning for the treatment of S. aureus infections. MedChemComm 2013, 4, 1114–1117. [Google Scholar] [CrossRef]
  99. González, J.E.; Keshavan, N.D. Messing with Bacterial Quorum Sensing. Microbiol. Mol. Biol. Rev. 2006, 70, 859–875. [Google Scholar] [CrossRef] [Green Version]
  100. Dell’Acqua, G.; Giacometti, A.; Cirioni, O.; Ghiselli, R.; Saba, V.; Scalise, G.; Gov, Y.; Balaban, N. Suppression of Drug-Resistant Staphylococcal Infections by the Quorum-Sensing Inhibitor RNAIII-Inhibiting Peptide. J. Infect. Dis. 2004, 190, 318–320. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Simonetti, O.; Cirioni, O.; Cacciatore, I.; Baldassarre, L.; Orlando, F.; Pierpaoli, E.; Lucarini, G.; Orsetti, E.; Provinciali, M.; Fornasari, E.; et al. Efficacy of the Quorum Sensing Inhibitor FS10 Alone and in Combination with Tigecycline in an Animal Model of Staphylococcal Infected Wound. PLoS ONE 2016, 11, e0151956. [Google Scholar] [CrossRef]
  102. Donlan, R.M.; Costerton, J.W. Biofilms: Survival Mechanisms of Clinically Relevant Microorganisms. Clin. Microbiol. Rev. 2002, 15, 167–193. [Google Scholar] [CrossRef] [Green Version]
  103. Kang, H.-K.; Kim, C.; Seo, C.H.; Park, Y. The therapeutic applications of antimicrobial peptides (AMPs): A patent review. J. Microbiol. 2016, 55, 1–12. [Google Scholar] [CrossRef] [PubMed]
  104. Mookherjee, N.; Hancock, R.E.W. Cationic host defence peptides: Innate immune regulatory peptides as a novel approach for treating infections. Experientia 2007, 64, 922–933. [Google Scholar] [CrossRef] [PubMed]
  105. Koczulla, R.; von Degenfeld, G.; Kupatt, C.; Krotz, F.; Zahler, S.; Gloe, T.; Issbrücker, K.; Unterberger, P.; Zaiou, M.; Lebherz, C.; et al. An angiogenic role for the human peptide antibiotic LL-37/hCAP-18. J. Clin. Investig. 2003, 111, 1665–1672. [Google Scholar] [CrossRef]
  106. Mookherjee, N.; Brown, K.L.; Bowdish, D.M.; Doria, S.; Falsafi, R.; Hokamp, K.; Roche, F.M.; Mu, R.; Doho, G.H.; Pistolic, J.; et al. Modulation of the TLRmediated in-flammatory response by the endogenous human host defense peptide LL-37. J. Immunol. 2006, 176, 2455–2464. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Shaykhiev, R.; Beisswenger, C.; Kändler, K.; Senske, J.; Püchner, A.; Damm, T.; Behr, J.; Bals, R. Human endogenous antibiotic LL-37 stimulates airway epithelial cell proliferation and wound closure. Am. J. Physiol. Cell. Mol. Physiol. 2005, 289, L842–L848. [Google Scholar] [CrossRef] [PubMed]
  108. Niyonsaba, F.; Ushio, H.; Nakano, N.; Ng, W.; Sayama, K.; Hashimoto, K.; Nagaoka, I.; Okumura, K.; Ogawa, H. Antimicrobial peptides human beta-1s stimulate epi-dermal keratinocyte migration, proliferation and production of proinflammatory cytokines and chemokines. J. Investig. Dermatol. 2007, 127, 594–604. [Google Scholar] [CrossRef] [Green Version]
  109. Cirioni, O.; Wu, G.; Li, L.; Orlando, F.; Silvestri, C.; Ghiselli, R.; Shen, Z.; Scalise, A.; Gabrielli, E.; Scuppa, D.; et al. S-thanatin enhances the efficacy of tigecycline in an experimental rat model of polymicrobial peritonitis. Peptides 2010, 31, 1231–1236. [Google Scholar] [CrossRef]
  110. Simonetti, O.; Arzeni, D.; Ganzetti, G.; Silvestri, C.; Cirioni, O.; Gabrielli, E.; Castelletti, S.; Kamysz, W.; Kamysz, E.; Scalise, G.; et al. In vitro activity of the lipopeptide derivative (Pal-Lys-Lys-NH), alone and in combination with antifungal agents, against clinical isolates of dermatophytes. Br. J. Dermatol. 2009, 161, 249–252. [Google Scholar] [CrossRef] [PubMed]
  111. Simonetti, O.; Cirioni, O.; Ghiselli, R.; Goteri, G.; Orlando, F.; Monfregola, L.; Luca, S.; Zizzi, A.; Silvestri, C.; Veglia, G.; et al. Antimicrobial properties of distinctin in an experimental model of MRSA-infected wounds. Eur. J. Clin. Microbiol. Infect. Dis. 2012, 31, 3047–3055. [Google Scholar] [CrossRef]
  112. Park, S.-C.; Park, Y.; Hahm, K.-S. The Role of Antimicrobial Peptides in Preventing Multidrug-Resistant Bacterial Infections and Biofilm Formation. Int. J. Mol. Sci. 2011, 12, 5971–5992. [Google Scholar] [CrossRef] [Green Version]
  113. De La Fuente-Núñez, C.; Reffuveille, F.; Haney, E.F.; Straus, S.; Hancock, R. Broad-Spectrum Anti-biofilm Peptide That Targets a Cellular Stress Response. PLoS Pathog. 2014, 10, e1004152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Reffuveille, F.; De La Fuente-Núñez, C.; Mansour, S.; Hancock, R.E.W. A Broad-Spectrum Antibiofilm Peptide Enhances Antibiotic Action against Bacterial Biofilms. Antimicrob. Agents Chemother. 2014, 58, 5363–5371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Etayash, H.; Pletzer, D.; Kumar, P.; Straus, S.K.; Hancock, R. Cyclic Derivative of Host-Defense Peptide IDR-1018 Improves Proteolytic Stability, Suppresses Inflammation, and Enhances In Vivo Activity. J. Med. Chem. 2020, 63. [Google Scholar] [CrossRef] [PubMed]
  116. Niyonsaba, F.; Kiatsurayanon, C.; Chieosilapatham, P.; Ogawa, H. Friends or Foes? Host defense (antimicrobial) peptides and proteins in human skin diseases. Exp. Dermatol. 2017, 26, 989–998. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Heilborn, J.D.; Nilsson, M.F.; Sorensen, O.E.; Ståhle-Bäckdahl, M.; Kratz, G.; Borregaard, N. The Cathelicidin Anti-Microbial Peptide LL-37 is Involved in Re-Epithelialization of Human Skin Wounds and is Lacking in Chronic Ulcer Epithelium. J. Investig. Dermatol. 2003, 120, 379–389. [Google Scholar] [CrossRef] [Green Version]
  118. Carretero, M.; Escámez, M.J.; García, M.; Duarte, B.; Holguín, A.; Retamosa, L.; Jorcano, J.L.; del Río, M.; Larcher, F. In vitro and In vivo Wound Healing-Promoting Activities of Human Cathelicidin LL-37. J. Investig. Dermatol. 2008, 128, 223–236. [Google Scholar] [CrossRef] [Green Version]
  119. Ramos, R.; Silva, J.P.; Rodrigues, A.C.; Costa, R.; Guardão, L.; Schmitt, F.; Soares, R.; Vilanova, M.; Domingues, L.; Gama, M. Wound healing activity of the human an-timicrobial peptide LL37. Peptides 2011, 32, 1469–1476. [Google Scholar] [CrossRef] [Green Version]
  120. Overhage, J.; Campisano, A.; Bains, M.; Torfs, E.C.W.; Rehm, B.; Hancock, R.E.W. Human Host Defense Peptide LL-37 Prevents Bacterial Biofilm Formation. Infect. Immun. 2008, 76, 4176–4182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  121. Simonetti, O.; Cirioni, O.; Goteri, G.; Lucarini, G.; Kamysz, E.; Kamysz, W.; Orlando, F.; Rizzetto, G.; Molinelli, E.; Morroni, G.; et al. Efficacy of the cathelicidin LL-37 in a MRSA wound infection mice model. Antibiotics 2021, in press. [Google Scholar] [CrossRef] [PubMed]
  122. Park, I.Y.; Cho, J.H.; Kim, K.S.; Kim, Y.-B.; Kim, M.S.; Kim, S.C. Helix Stability Confers Salt Resistance upon Helical Antimicrobial Peptides. J. Biol. Chem. 2004, 279, 13896–13901. [Google Scholar] [CrossRef] [Green Version]
  123. Pasupuleti, M.; Schmidtchen, A.; Chalupka, A.; Ringstad, L.; Malmsten, M. End-Tagging of Ultra-Short Antimicrobial Peptides by W/F Stretches to Facilitate Bacterial Killing. PLoS ONE 2009, 4, e5285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Sieprawska-Lupa, M.; Mydel, P.; Krawczyk, K.; Wojcik, K.; Puklo, M.; Lupa, B.; Suder, P.; Silberring, J.; Reed, M.; Pohl, J.; et al. Degradation of human antimicrobial pep-tide LL-37 by Staphylococcus aureus-derived proteinases. Antimicrob. Agents Chemother. 2004, 48, 4673–4679. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Kim, D.J.; Lee, Y.W.; Park, M.K.; Shin, J.R.; Lim, K.J.; Cho, J.H.; Kim, S.C. Efficacy of the designer antimicrobial peptide SHAP1 in wound healing and wound infection. Amino Acids 2014, 46, 2333–2343. [Google Scholar] [CrossRef] [Green Version]
  126. Tokumaru, S.; Sayama, K.; Shirakata, Y.; Komatsuzawa, H.; Ouhara, K.; Hanakawa, Y.; Yahata, Y.; Dai, X.; Tohyama, M.; Nagai, H.; et al. Induction of keratinocyte mi-gration via transactivation of the epidermal growth factor receptor by the antimicrobial peptide LL-37. J. Immunol. 2005, 175, 4662–4668. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Rawlings, N.D.; Barrett, A.J.; Bateman, A. MEROPS: The peptidase database. Nucleic Acids Res. 2010, 38, D227–D233. [Google Scholar] [CrossRef]
  128. Chung, E.M.C.; Dean, S.N.; Propst, C.N.; Bishop, B.M.; Van Hoek, M.L. Komodo dragon-inspired synthetic peptide DRGN-1 promotes wound-healing of a mixed-biofilm infected wound. NPJ Biofilms Microbiomes 2017, 3, 1–13. [Google Scholar] [CrossRef]
  129. Song, X.; Pan, H.; Wang, H.; Liao, X.; Sun, D.; Xu, K.; Chen, T.; Zhang, X.; Wu, M.; Wu, D.; et al. Identification of new dermaseptins with self-assembly tendency: Membrane disruption, biofilm eradication, and infected wound healing efficacy. Acta Biomater. 2020, 109, 208–219. [Google Scholar] [CrossRef]
  130. Ong, J.S.; Taylor, T.D.; Yong, C.C.; Khoo, B.Y.; Sasidharan, S.; Choi, S.B.; Ohno, H.; Liong, M.T. Lactobacillus plantarum USM8613 Aids in Wound Healing and Suppresses Staphylococcus aureus Infection at Wound Sites. Probiotics Antimicrob. Proteins 2020, 12, 125–137. [Google Scholar] [CrossRef]
  131. Sojka, M.; Valachova, I.; Bucekova, M.; Majtan, J. Antibiofilm efficacy of honey and bee-derived defensin-1 on multispecies wound biofilm. J. Med. Microbiol. 2016, 65, 337–344. [Google Scholar] [CrossRef]
  132. Huang, J.; Fan, Q.; Guo, M.; Wu, M.; Wu, S.; Shen, S.; Wang, X.; Wang, H. Octenidine dihydrochloride treatment of a meticillin-resistant Staphylococcus aureus biofilm-infected mouse wound. J. Wound Care 2021, 30, 106–114. [Google Scholar] [CrossRef]
  133. Tavares, A.; Carvalho, C.M.B.; Faustino, M.A.; Neves, M.G.P.M.S.; Tomé, J.P.C.; Tomé, A.C.; Cavaleiro, J.A.S.; Cunha, A.; Gomes, N.C.M.; Alves, E.; et al. Antimicrobial Photodynamic Therapy: Study of Bacterial Recovery Viability and Potential Development of Resistance after Treatment. Mar. Drugs 2010, 8, 91–105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  134. Hamblin, M.R.; Hasan, T. Photodynamic therapy: A new antimicrobial approach to infectious disease? Photochem. Photobiol. Sci. 2004, 3, 436–450. [Google Scholar] [CrossRef] [Green Version]
  135. Silva, E.F.F.; Serpa, C.; Dąbrowski, J.M.; Monteiro, C.J.P.; Formosinho, S.J.; Stochel, G.; Urbanska, K.; Simões, S.; Pereira, M.M.; Arnaut, L.G. Mechanisms of Singlet-Oxygen and Superoxide-Ion Generation by Porphyrins and Bacteriochlorins and their Implications in Photodynamic Therapy. Chem.-Eur. J. 2010, 16, 9273–9286. [Google Scholar] [CrossRef]
  136. Sharman, W.M.; Allen, C.M.; van Lier, J.E. Photodynamic therapeutics: Basic principles and clinical applications. Drug Discov. Today 1999, 4, 507–517. [Google Scholar] [CrossRef]
  137. Wainwright, M. Photodynamic antimicrobial chemotherapy (PACT). J. Antimicrob. Chemother. 1998, 42, 13–28. [Google Scholar] [CrossRef]
  138. Zeina, B.; Greenman, J.; Purcell, W.; Das, B. Killing of cutaneous microbial species by photodynamic therapy. Br. J. Dermatol. 2001, 144, 274–278. [Google Scholar] [CrossRef] [PubMed]
  139. Pérez, C.; Zúñiga, T.; Palavecino, C.E. Photodynamic therapy for treatment of Staphylococcus aureus infections. Photodiagnosis Photodyn. Ther. 2021, 34, 102285. [Google Scholar] [CrossRef]
  140. 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] [Green Version]
  141. Hu, X.; Huang, Y.; Wang, Y.; Wang, X.; Hamblin, M.R. Antimicrobial Photodynamic Therapy to Control Clinically Relevant Biofilm Infections. Front. Microbiol. 2018, 9, 1299. [Google Scholar] [CrossRef] [Green Version]
  142. 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]
  143. Woźniak, A.; Grinholc, M. Combined Antimicrobial Activity of Photodynamic Inactivation and Antimicrobials–State of the Art. Front. Microbiol. 2018, 9, 930. [Google Scholar] [CrossRef]
  144. Fabris, C.; Soncin, M.; Mazzon, E.; Calzavara-Pinton, P.; Lia, F.; Giacomo, C.; Dei, D.; Tampucci, S.; Roncucci, G.; Jori, G. A novel tetracationic phthalocyanine as a potential skin phototherapeutic agent. Exp. Dermatol. 2005, 14, 675–683. [Google Scholar] [CrossRef] [PubMed]
  145. Simonetti, O.; Cirioni, O.; Orlando, F.; Alongi, C.; Lucarini, G.; Silvestri, C.; Zizzi, A.; Fantetti, L.; Roncucci, G.; Giacometti, A.; et al. Effectiveness of antimicrobial photodynamic therapy with a single treatment of RLP068/Cl in an experimental model of Staphylococcus aureus wound infection. Br. J. Dermatol. 2011, 164, 987–995. [Google Scholar] [CrossRef]
  146. Araújo, N.C.; Fontana, C.R.; Bagnato, V.S.; Gerbi, M.E.M. Photodynamic antimicrobial therapy of curcumin in biofilms and carious dentine. Lasers Med. Sci. 2013, 29, 629–635. [Google Scholar] [CrossRef]
  147. Lee, H.-J.; Kang, S.-M.; Jeong, S.-H.; Chung, K.-H.; Kim, B.-I. Antibacterial photodynamic therapy with curcumin and Curcuma xanthorrhiza extract against Streptococcus mutans. Photodiagnosis Photodyn. Ther. 2017, 20, 116–119. [Google Scholar] [CrossRef]
  148. Spaeth, A.; Graeler, A.; Maisch, T.; Plaetzer, K. CureCuma–cationic curcuminoids with improved properties and enhanced antimicrobial photodynamic activity. Eur. J. Med. Chem. 2018, 159, 423–440. [Google Scholar] [CrossRef]
  149. Araújo, T.S.D.; Rodrigues, P.L.F.; Santos, M.S.; de Oliveira, J.M.; Rosa, L.P.; Bagnato, V.S.; Blanco, K.C.; da Silva, F.C. Reduced methicillin-resistant Staphylococcus aureus biofilm formation in bone cavities by photodynamic therapy. Photodiagnosis Photodyn. Ther. 2018, 21, 219–223. [Google Scholar] [CrossRef] [PubMed]
  150. Méndez, D.A.C.; Gutierrez, E.; Lamarque, G.C.C.; Rizzato, V.L.; Buzalaf, M.A.R.; Machado, M.A.A.M.; Cruvinel, T. The effectiveness of curcumin-mediated antimicrobial photodynamic therapy depends on pre-irradiation and biofilm growth times. Photodiagn. Photodyn. Ther. 2019, 27, 474–480. [Google Scholar] [CrossRef]
  151. Mirzahosseinipour, M.; Khorsandi, K.; Hosseinzadeh, R.; Ghazaeian, M.; Shahidi, F.K. Antimicrobial photodynamic and wound healing activity of curcumin encapsu-lated in silica nanoparticles. Photodiagn. Photodyn. Ther. 2020, 29, 101639. [Google Scholar] [CrossRef] [PubMed]
  152. Lin, M.-H.; Lee, J.Y.-Y.; Pan, S.-C.; Wong, T.-W. Enhancing wound healing in recalcitrant leg ulcers with aminolevulinic acid-mediated antimicrobial photodynamic therapy. Photodiagn. Photodyn. Ther. 2020, 33, 102149. [Google Scholar] [CrossRef]
  153. Saddiqe, Z.; Naeem, I.; Maimoona, A. A review of the antibacterial activity of Hypericum perforatum L. J. Ethnopharmacol. 2010, 131, 511–521. [Google Scholar] [CrossRef]
  154. Jacobson, J.M.; Feinman, L.; Liebes, L.; Ostrow, N.; Koslowski, V.; Tobia, A.; Cabana, B.E.; Lee, D.; Spritzler, J.; Prince, A.M. Pharmacokinetics, safety, and antiviral ef-fects of hypericin, a derivative of St. John’s Wort Plant, in patients with chronic hepatitis C virus infection. Antimicrob. Agents Chemother. 2001, 45, 517–524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Kadam, N.; Chaudhari, H.; Parikh, J.; Modi, V.; Kokil, S.; Balaramnav, V. De novo Combination Therapy in Retroviral Infection. Int. J. Virol. 2010, 6, 219–223. [Google Scholar] [CrossRef]
  156. García, I.; Ballesta, S.; Gilaberte, Y.; Rezusta, A.; Pascual, A. Antimicrobial photodynamic activity of hypericin against methicillin-susceptible and resistant Staphylococcus aureus biofilms. Futur. Microbiol. 2015, 10, 347–356. [Google Scholar] [CrossRef]
  157. Cheng, Y.; Burda, C. 2.01—Nanoparticles for photodynamic therapy. In Comprehensive Nanoscience and Technology; Andrews, D.L., Scholes, G.D., Wiederrecht, G.P., Eds.; Academic Press: Amsterdam, The Netherlands, 2011; pp. 1–28. [Google Scholar]
  158. Paszko, E.; Ehrhardt, C.; Senge, M.O.; Kelleher, D.P.; Reynolds, J.V. Nanodrug applications in photodynamic therapy. Photodiagn. Photodyn. Ther. 2011, 8, 14–29. [Google Scholar] [CrossRef] [PubMed]
  159. Nafee, N.; Youssef, A.; El-Gowelli, H.; Asem, H.; Kandil, S. Antibiotic-free nanotherapeutics: Hypericin nanoparticles thereof for improved in vitro and in vivo antimicrobial photodynamic therapy and wound healing. Int. J. Pharm. 2013, 454, 249–258. [Google Scholar] [CrossRef] [PubMed]
  160. Pérez, M.; Robres, P.; Moreno, B.; Bolea, R.; Verde, M.T.; Pérez-Laguna, V.; Aspiroz, C.; Gilaberte, Y.; Rezusta, A. Comparison of Antibacterial Activity and Wound Healing in a Superficial Abrasion Mouse Model of Staphylococcus aureus Skin Infection Using Photodynamic Therapy Based on Methylene Blue or Mupirocin or Both. Front. Med. 2021, 8. [Google Scholar] [CrossRef]
  161. De La Fuente-Núñez, C.; Korolik, V.; Bains, M.; Nguyen, U.; Breidenstein, E.B.M.; Horsman, S.; Lewenza, S.; Burrows, L.; Hancock, R. Inhibition of Bacterial Biofilm Formation and Swarming Motility by a Small Synthetic Cationic Peptide. Antimicrob. Agents Chemother. 2012, 56, 2696–2704. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Dean, S.N.; Bishop, B.M.; van Hoek, M.L. Natural and synthetic cathelicidin peptides with anti-microbial and anti-biofilm activity against Staphylococcus aureus. BMC Microbiol. 2011, 11, 114. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Ovchinnikov, K.V.; Kranjec, C.; Thorstensen, T.; Carlsen, H.; Diep, D.B. Successful Development of Bacteriocins into Therapeutic Formulation for Treatment of MRSA Skin Infection in a Murine Model. Antimicrob. Agents Chemother. 2020, 64. [Google Scholar] [CrossRef]
  164. Fernandez, J.; Martin-Serrano, A.; Gómez-Casanova, N.; Falanga, A.; Galdiero, S.; de la Mata, F.J.; Heredero-Bermejo, I.; Ortega, P. Effect of the Combination of Levofloxacin with Cationic Carbosilane Dendron and Peptide in the Prevention and Treatment of Staphylococcus aureus Biofilms. Polymers 2021, 13, 2127. [Google Scholar] [CrossRef] [PubMed]
  165. Cirioni, O.; Giacometti, A.; Ghiselli, R.; Dell’Acqua, G.; Gov, Y.; Kamysz, W.; Lukasiak, J.; Mocchegiani, F.; Orlando, F.; D’Amato, G.; et al. Prophylactic efficacy of topical temporin A and RNAIII-inhibiting peptide in a subcutaneous rat Pouch model of graft infection attributable to staphylococci with intermediate resistance to glycopeptides. Circulation 2003, 108, 767–771. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Golda, A.; Kosikowska-Adamus, P.; Kret, A.; Babyak, O.; Wójcik, K.; Dobosz, E.; Potempa, J.; Lesner, A.; Koziel, J. The Bactericidal Activity of Temporin Analogues Against Methicillin Resistant Staphylococcus aureus. Int. J. Mol. Sci. 2019, 20, 4761. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Kranjec, C.; Ovchinnikov, K.V.; Grønseth, T.; Ebineshan, K.; Srikantam, A.; Diep, D.B. A bacteriocin-based antimicrobial formulation to effectively disrupt the cell via-bility of methicillin-resistant Staphylococcus aureus (MRSA) biofilms. NPJ Biofilms Microbiomes 2020, 6, 58. [Google Scholar] [CrossRef] [PubMed]
  168. Twomey, E.; Hill, C.; Field, D.; Begley, M. Bioengineered Nisin Derivative M17Q Has Enhanced Activity against Staphylococcus epidermidis. Antibiotics 2020, 9, 305. [Google Scholar] [CrossRef]
  169. Yang, N.; Teng, D.; Mao, R.; Hao, Y.; Wang, X.; Wang, Z.; Wang, X.; Wang, J. A recombinant fungal defensin-like peptide-P2 combats multidrug-resistant Staphylococcus aureus and biofilms. Appl. Microbiol. Biotechnol. 2019, 103, 5193–5213. [Google Scholar] [CrossRef]
  170. Brackman, G.; De Meyer, L.; Nelis, H.; Coenye, T. Biofilm inhibitory and eradicating activity of wound care products against Staphylococcus aureus and Staphylococcus epidermidis biofilms in an in vitro chronic wound model. J. Appl. Microbiol. 2013, 114, 1833–1842. [Google Scholar] [CrossRef]
  171. Reynoso, E.; Ferreyra, D.D.; Durantini, E.N.; Spesia, M.B. Photodynamic inactivation to prevent and disrupt Staphylococcus aureus biofilm under different media conditions. Photodermatol. Photoimmunol. Photomed. 2019, 35, 322–331. [Google Scholar] [CrossRef]
  172. Halstead, F.D.; Thwaite, J.E.; Burt, R.; Laws, T.R.; Raguse, M.; Moeller, R.; Webber, M.A.; Oppenheim, B.A. Antibacterial Activity of Blue Light against Nosocomial Wound Pathogens Growing Planktonically and as Mature Biofilms. Appl. Environ. Microbiol. 2016, 82, 4006–4016. [Google Scholar] [CrossRef] [Green Version]
  173. Dai, T.; Gupta, A.; Huang, Y.; Sherwood, M.E.; Murray, C.K.; Vrahas, M.S.; Kielian, T.; Hamblin, M.R. Blue Light Eliminates Community-Acquired Methicillin-Resistant Staphylococcus aureusin Infected Mouse Skin Abrasions. Photomed. Laser Surg. 2013, 31, 531–538. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  174. Belyi, Y.; Rybolovlev, I.; Polyakov, N.; Chernikova, A.; Tabakova, I.; Gintsburg, A. Staphylococcus Aureus Surface Protein G is An Immunodominant Protein and a Possible Target in An Anti-Biofilm Drug Development. Open Microbiol. J. 2018, 12, 94–106. [Google Scholar] [CrossRef] [PubMed]
  175. Domanski, P.J.; Patel, P.R.; Bayer, A.S.; Zhang, L.; Hall, A.E.; Syribeys, P.J.; Gorovits, E.L.; Bryant, D.; Vernachio, J.H.; Hutchins, J.T.; et al. Characterization of a Humanized Monoclonal Antibody Recognizing Clumping Factor A Expressed by Staphylococcus aureus. Infect. Immun. 2005, 73, 5229–5232. [Google Scholar] [CrossRef] [Green Version]
  176. Tkaczyk, C.; Kasturirangan, S.; Minola, A.; Jones-Nelson, O.; Gunter, V.; Shi, Y.Y.; Rosenthal, K.; Aleti, V.; Semenova, E.; Warrener, P.; et al. Multimechanistic Mono-clonal Antibodies (MAbs) Targeting Staphylococcus aureus Alpha-Toxin and Clumping Factor A: Activity and Efficacy Comparisons of a MAb Combination and an Engineered Bispecific Antibody Approach. Antimicrob. Agents Chemother. 2017, 61, e00629-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  177. Varshney, A.K.; Kuzmicheva, G.A.; Bowling, R.A.; Sunley, K.M.; Bowling, R.A.; Kwan, T.-Y.; Mays, H.R.; Rambhadran, A.; Zhang, Y.; Martin, R.L.; et al. A natural human monoclonal antibody targeting Staphylococcus Protein A protects against Staphylococcus aureus bacteremia. PLoS ONE 2018, 13, e0190537. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  178. França, A.; Vilanova, M.; Cerca, N.; Pier, G.B. Monoclonal Antibody Raised against PNAG Has Variable Effects on Static S. epidermidis Biofilm Accumulation In Vitro. Int. J. Biol. Sci. 2013, 9, 518–520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  179. Natan, M.; Banin, E. From Nano to Micro: Using nanotechnology to combat microorganisms and their multidrug resistance. FEMS Microbiol. Rev. 2017, 41, 302–322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  180. Banerjee, S.; Ghosh, D.; Vishakha, K.; Das, S.; Mondal, S.; Ganguli, A. Photodynamic antimicrobial chemotherapy (PACT) using riboflavin inhibits the mono and dual species biofilm produced by antibiotic resistant Staphylococcus aureus and Escherichia coli. Photodiagn. Photodyn. Ther. 2020, 32, 102002. [Google Scholar] [CrossRef]
  181. Ramasamy, M.; Lee, J. Recent Nanotechnology Approaches for Prevention and Treatment of Biofilm-Associated Infections on Medical Devices. BioMed Res. Int. 2016, 2016, 1851242. [Google Scholar] [CrossRef] [Green Version]
  182. Malaekeh-Nikouei, B.; Bazzaz, B.S.F.; Mirhadi, E.; Tajani, A.S.; Khameneh, B. The role of nanotechnology in combating biofilm-based antibiotic resistance. J. Drug Deliv. Sci. Technol. 2020, 60, 101880. [Google Scholar] [CrossRef]
  183. Stamsås, G.A.; Myrbråten, I.S.; Straume, D.; Salehian, Z.; Veening, J.-W.; Håvarstein, L.S.; Kjos, M. CozEa and CozEb play overlapping and essential roles in controlling cell division in Staphylococcus aureus. Mol. Microbiol. 2018, 109, 615–632. [Google Scholar] [CrossRef]
  184. DeFrancesco, A.S.; Masloboeva, N.; Syed, A.K.; Deloughery, A.; Bradshaw, N.; Li, G.-W.; Gilmore, M.S.; Walker, S.; Losick, R.M. Genome-wide screen for genes involved in eDNA release during biofilm formation by Staphylococcus aureus. Proc. Natl. Acad. Sci. USA 2017, 114, E5969–E5978. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Duarte, A.C.; Fernández, L.; De Maesschalck, V.; Gutiérrez, D.; Campelo, A.B.; Briers, Y.; Lavigne, R.; Rodríguez, A.; García, P. Synergistic action of phage phiIPLA-RODI and lytic protein CHAPSH3b: A combination strategy to target Staphylococcus aureus biofilms. npj Biofilms Microbiomes 2021, 7, 39. [Google Scholar] [CrossRef] [PubMed]
  186. Dickey, J.; Perrot, V. Adjunct phage treatment enhances the effectiveness of low antibiotic concentration against Staphylococcus aureus biofilms in vitro. PLoS ONE 2019, 14, e0209390. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Akturk, E.; Oliveira, H.; Santos, S.B.; Costa, S.; Kuyumcu, S.; Melo, L.D.R.; Azeredo, J. Synergistic action of phage and antibiotics: Parameters to enhance the killing efficacy against mono and dual-species biofilms. Antibiotics 2019, 8, 103. [Google Scholar] [CrossRef] [Green Version]
  188. Rahman, M.; Kim, S.; Kim, S.M.; Seol, S.Y.; Kim, J. Characterization of induced Staphylococcus aureus bacteriophage SAP-26 and its anti-biofilm activity with rifampicin. Biofouling 2011, 27, 1087–1093. [Google Scholar] [CrossRef] [PubMed]
Table 1. Summary of reviewed therapies.
Table 1. Summary of reviewed therapies.
TherapyExperimental ModelWound Healing AssessmentHistological FindingsAdvantages/Disadvantages
ANTIBIOTICS
Kirmusaoglu S. et al., 2020 [33]Beta-lactamsIn vitro, MRSA ATCC43300, MRSA, MSSA, beta-lactams combined with 2-aminothiazole as adjuvantNot availableNot available
-
First line treatment for MSSA and CNSA
-
Low efficacy alone versus biofilm-producing strains, needed association with adjuvants
-
Sub- MIC concentration may induce biofilm production [34]
Simonetti et al., 2008 [34]TeicoplaninIn vivo murine model with MRSA infected skin wound; placebo vs. aPDT vs. aPDT + RLP068 vs. teicoplanin intra peritoneal (i.p.) vs. non infectedBetter wound-healing response compared to placebo
-
complete and normal epithelialization,
-
thick granulation tissue
-
regular collagen deposition
-
First line treatment for MRSA
-
Improve wound healing
-
aPDT + RLP068 showed better results for wound healing
Simonetti et al., 2017 [35]DaptomycinIn vivo murine model with S. aureus ATCC43300 infected skin wound (burn); daptomycin i.p. vs. teicoplanin i.p. vs. placebo vs. non infectedBetter overall healing of Daptomycin group
-
better epithelization
-
significantly higher collagen scores
-
higher immunohistochemical expression of wound healing markers (EGFR and FGF-2)
-
better in vivo efficacy than teicoplanin
-
treatment option in more serious cases
Simonetti et al., 2011 [36]TigecyclineIn vivo murine model with S. aureus ATCC43300 infected skin wound (burn); uninfected control group vs. infected no treatment vs. tigecycline i.p. vs. teicoplanin i.p.Tigecycline showed better impact on wound healing
-
Significant decrease in MMp-9 expression
-
Faster re-epithelisation vs. teicoplanin
-
Earlier Collagen better organised in the dermis vs. teicoplanin
-
Poor inflammatory response
-
Modulatory effect on MMP-9 expression and accelerated wound healing compared to teicoplanin
-
Therapeutic option in more severe cases
-
Improved wound healing in combination with topical FS10 [37]
Simonetti et al., 2020 [38]DalbavancinIn vivo murine model with MRSA infected skin wound; vancomycin i.p. vs. dalbavancin i.p. vs. uninfected vs. untreatedfaster healing after dalbavancin treatment
-
robust epidermal coverage, regular and keratinized epidermal lining
-
well-organized granulation tissue with numerous blood vessels
-
Immunohistochemistry with higher levels of EGFR and VEGF, reduction of MMP-9 and MMP-1
-
Faster wound healing than vancomycin
-
Treatment option in case of vancomycin resistance
-
Both effective in controlling infection
QUORUM SENSING INHIBITORS
Schierle et al., 2009 [39]RIPIn vivo murine model with S. aureus and S. epidermidis biofilm producers; uninfected vs. RIP topically (100 mcg for 7 days) vs. untreatedBetter wound healing vs. untreated
-
Re-epithelialization significantly more rapid with RIP
-
Topical RIP restores normal wound healing kinetics
-
Useful only in localized infections
Simonetti et al., 2008 [40] In vivo murine model with MRSA infected skin wound; topical RIP (20 mcg), teicoplanin i.p., allevyn, allevyn + teicoplanin i.p., topical RIP + teicoplanin i.p.Better wound healing with topical RIP + teicoplanin
-
only topical RIP + teicoplanin restored epithelial, granulation, and collagen scores, as well as microvessel density and VEGF expression
-
Possible association with systemic antibiotic (teicoplanin) improves both infection control and wound healing
-
Alternative to overcome antibiotic resistance
-
RIP can induce VEGF, improving quality and speed of wound healing
-
Only 2 case reports on chronic diabetic ulcers in combination with daptomycin
Kuo et al., 2014 [41]F19,F12, and F1In vivo (1) MRSA-infected insect larvae; F19, F12 and F1 injection (20 mg/kg)
(2) in vivo murine model with MRSA-infected wounds; topical F12 and F1 vs. untreated
(1) F19,F12, and F1 improved survival of larvae
(2) F12 and F1 improved the speed of wound healing
-
not available, only wound size considered
-
Useful in improving wound healing
-
Possible contribution in reducing MICs by 50-fold for resistant antibiotics, such as cephalothin and nafcillin
Simonetti et al., 2016 [42]FS10in vivo murine model with MRSA and MSSA-infected wounds; topical FS10 (20mcg) + tigecycline i.p. (7 mg/kg) vs. monotherapy vs. untreated vs. uninfectedFS10 + tigecycline showed better wound healing and infection control
-
robust epidermal coverage, regular epidermal lining, evident keratinization
-
the dermal papillae were still few.
-
Thick granulation tissue with many vessels and fibres
-
Collagen more organized and regular collagen fibres
-
Not evident inflammatory response
-
Improvement even in FS10 monotherapy comparable to tigecycline monotherapy
-
Best result in combination therapy
ANTIMICROBIAL PEPTIDES
Etayash H. et al., 2020 [43]IDR-1018In vivo murine model with MRSA infection abscess; IDR-1018 injected subcutaneouslyNot availableNot available
-
Reduction of bacterial load and elimination of biofilm
-
Further studies are needed to confirm action on wound healing
Carretero M. et al., 2008 [44]LL-37In vivo murine model non infected wound, adenoviral transfer of LL-37Improved wound healing compared to untreated
-
Significant increase in re-epithelialisation
-
Significant increase in granulation tissue
-
Improves wound healing
-
No conclusive histological data on infected wounds anti-biofilm action [45]
-
inactivated by endogenous, bacterial proteases and sub-physiological salt concentrations. [45,46,47,48,49]
Kim DJ et al., 2014 [50]SHAP1In vivo murine model with S. aureus (ATCC 29213) infected wounds; topical shap1 vs. LL-37 vs. PBSPromote and accelerate wound healing
-
activation EGFR pathway
-
migliore riepitelizzazione rispetto a PBS e LL-37
-
SHAP1 more resistant to protease and wound salt environment than LL-37
-
LL-37 showed no difference in wound area compared to PBS, endogenous protease role
Chung EMC et al., 2017 [51]DRGN-1(1) in vivo murine model with S. aureus infected wound;
Topical DRGN-1 vs. VK25 vs. LL-37 vs. PBS
(2) in vivo murine model non-infected, Topical DRGN-1 vs. VK25 vs. PBS
(1–2) Wound healing significantly faster with DRGN-1, wound size considered(1) skin layers were completely rehabilitated
-
(1–2) DRGN-1 accelerates wound healing in both infected and non-infected wounds
-
(2)direct action on re-epithelialisation
-
(1) More effective than monotherapy with LL-37
-
EGFR-STAT1/3 pathway activation
-
Anti-biofilm activity and antibacterial activity through membrane permeabilization
Song X. et al., 2020 [52]DMS-PS2In vivo murine model with MRSA infected wounds;
Topical MDS-PS2 vs. untreated
DMS-PS2 improved wound healing Not available, clinically increased rate of re-epithelialisation
-
Broad-spectrum antimicrobial activities
-
Low toxicity mammalian blood
-
Important anti-biofilm action
-
Strong inhibition of bacterial growth
Cell-free supernatant (CFS) of Lactobacillus plantarum USM8613(1) porcine skin wound model infected with S. aureus; CFS vs. untreated
(2) in vivo murine model infected with S. aureus; CFS vs. untreated control
(2) CFS enhanced wound contraction percentage (54%)(2) accelerated keratinocyte migration over the wound edge towards the centre area over time
(2) achieved better wound closure and complete re-epithelisation
-
(1) lower bacterial count with CFS
-
(1) reduced biofilm thickness
-
(2) CFS increased the immune response (β-defensin), cytokine and chemokine production
Sojka M. et al., 2016 [53]Def-1In vitro Lubbock chronic wound biofilm model, S. aureus among other bacteriaNot availableNot available
-
reduced the viability of S. aureus
-
possible role in controlling biofilm in chronic wounds
TOPICAL
Huang J. et al., 2021 [54]Octenidine dihydrochlorideIn vivo murine model with MRSA infected skin woundAccelerated healing and reduced bacterial counts versus control (PBS)
-
Reduction of inflammatory cells
-
More mature collagen fibres
-
Well-defined epithelisation
-
Useful for difficult to treat chronic wound
-
Possible adjuvant therapy
APDT
Simonetti et al., 2011 [55]RLP068/ClIn vivo murine model with MRSA-infected wound;
RLP068/Cl + aPDT (689 nm) vs. untreated vs. teicoplanin i.p.
Better results in wound healing with RLP068/CI
-
RLP068/CI complete re-epithelialisation
-
RLP068/CI Faster than teicoplanin in controlling infection
-
Better re-epithelisation than teicoplanin
Mirzahosseinipour M. et al., 2020 [56]Curcumin encapsulated in silica nanoparticles (CEN)In vitro human dermal fibroblast culture infected with S. aureus; CEN + APDT (465 nm) vs. curcumin vs. untreatedCEN Improved human fibroblast activitythe denuded region of wounds treated with curcumin and CEN was narrower than that of untreated wounds (in vitro scratch assay)
-
reduction of planktonic bacteria and bacterial biofilm production
-
no significant fibroblast toxicity
-
lack of in vivo studies
Lin et al., 2020 [57]ALA3 patients with chronic leg ulcers resistant to conventional therapy (S. aureus isolated 1 patient); ALA + APDTClinically evident improvement without recurrences for 29 monthsNot available
-
the only study on patients with chronic ulcers and wound healing assessment
-
no bacteria isolated after treatment
-
lasting remission
-
probable direct action on wound healing, (IL-6 dependent migration of keratinocytes in vitro)
Nafee et al., 2013 [58]Hypericin nanoparticles (HN)In vivo murine model with MRSA infected wound; HN vs. Hypericin vs. untreatedHN showed faster wound healing better epithelialization, keratinization, and development of collagen fibres
-
direct effect on wound healing
-
in vitro excellent biofilm inhibition
Pérez et al., 2021 [59]Methylene Blue (MB)-aPDTIn vivo murine model with S. aureus ATCC29213 infected wound;
Topical MB-APDT vs. mupirocin (MU) vs. MB-APDT + MU vs. untreated
MB-aPDT improves quick mild wound contraction at 24 h, better wound healing (reduction of size, crust loss) and cosmetics results (no scar).mild acanthosis and mild undulation of the epidermis, a thicker dermis with moderate dermal fibrosis and more dilated follicles with abundant keratin and granulomatous inflammation.
-
MB-aPDT provided best clinical healing
-
MU enhances antimicrobial activity but not improved relevantly wound healing
-
No synergistic effects
CNS: coagulase-negative staphylococci, PBS: phosphate-buffered saline, EGFR: epidermal growth factor receptor, FGF-2: fibroblast growth factor, 2 DLP: defensin-like peptide, DMS-PS2: dermaseptin peptide2, ALA: aminolevulinic acid.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Simonetti, O.; Rizzetto, G.; Radi, G.; Molinelli, E.; Cirioni, O.; Giacometti, A.; Offidani, A. New Perspectives on Old and New Therapies of Staphylococcal Skin Infections: The Role of Biofilm Targeting in Wound Healing. Antibiotics 2021, 10, 1377. https://doi.org/10.3390/antibiotics10111377

AMA Style

Simonetti O, Rizzetto G, Radi G, Molinelli E, Cirioni O, Giacometti A, Offidani A. New Perspectives on Old and New Therapies of Staphylococcal Skin Infections: The Role of Biofilm Targeting in Wound Healing. Antibiotics. 2021; 10(11):1377. https://doi.org/10.3390/antibiotics10111377

Chicago/Turabian Style

Simonetti, Oriana, Giulio Rizzetto, Giulia Radi, Elisa Molinelli, Oscar Cirioni, Andrea Giacometti, and Annamaria Offidani. 2021. "New Perspectives on Old and New Therapies of Staphylococcal Skin Infections: The Role of Biofilm Targeting in Wound Healing" Antibiotics 10, no. 11: 1377. https://doi.org/10.3390/antibiotics10111377

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