Open access peer-reviewed chapter

New Approaches for Competing Microbial Resistance and Virulence

Written By

Mohammed El-Mowafy, Abdelaziz Elgaml and Mona Shaaban

Submitted: 24 July 2019 Reviewed: 05 November 2019 Published: 06 December 2019

DOI: 10.5772/intechopen.90388

From the Edited Volume

Microorganisms

Edited by Miroslav Blumenberg, Mona Shaaban, Abdelaziz Elgaml

Chapter metrics overview

912 Chapter Downloads

View Full Metrics

Abstract

The spread of multidrug-resistant pathogens together with the development of fatal cases of infectious microorganisms is on the rise. Therefore, there must be new approaches for combating pathogenic microorganisms, either by overcoming antibiotic resistance or via inhibiting their virulence factors. Several virulence factors extremely increase the antimicrobial resistance of various species of pathogens; as a result, the screening of antivirulence agents has gained more and more attention recently. In this aspect, non-traditional strategies that are considered promising in overcoming virulence and pathogenicity of microorganisms will be discussed including; quorum sensing inhibition, antibiofilm, control of the global regulators, bacteriocins and bacteriophages. Applying these methods could provide innovative approaches for competing microbial resistance and virulence.

Keywords

  • bacterial virulence
  • resistance
  • quorum sensing inhibition
  • global regulators
  • phage therapy
  • inhibition of biofilm formation
  • bacteriocins

1. Introduction

The high incidence of microbial resistance and the spread of multidrug-resistant and pan drug-resistant pathogens have been developed to threaten human mankind. Fortunately, there are upcoming alternative therapeutic approach for eliminating bacterial virulence and host-pathogen interaction [1, 2]. Quorum sensing signals [3, 4] and global regulators represent the main players to control virulence circuits and coordinate host-pathogen interaction [5]. Thus, targeting these regulators provide a promising trend to overcome microbial pathogenicity. Bacterial cells have the ability to grow in matrices of polysaccharides, proteins and DNA forming biofilm [6]. The cell communities inside the biofilm matrices are highly resistant to antibiotics [7]. In this chapter, we will focus on the agents that are known to exhibit antibiofilm assembly including bacteriocins.

Moreover, bacteriophages have specific ability to infect and lyse bacteria [8]. Hence, phage therapy has many potential applications in the treatment of infectious diseases, with high therapeutic index and diminished adverse effects [9, 10]. Inhibitors of quorum sensing signaling, control of the global regulators, and the development of antibiofilm agents will be discussed in detail in this chapter. Additionally, the use of bacteriophages either for eradication of bacterial infections or as an efficient delivery system for antimicrobial agents will be described in this part.

Advertisement

2. Control of microbial virulence and resistance

2.1 Quorum sensing inhibition

Quorum sensing (QS) is a cellular signaling system, which is developed in response to population cell density [3, 4]. QS cascade relays on the release of signaling molecules called QS autoinducers/signals. The QS signals are produced at low levels with the start of microbial growth and accumulate upon increase in the cell density. Quorum sensing signals coordinate the microbial virulence behaviors such as secretion of toxins, secretion of exoenzymes, microbial motility, adhesion and biofilm assembly [11]. Furthermore, microbial communication systems have been assigned in fungi [12] and viruses [13]. Studies of QS provide significant insights into different mechanisms that control the interactions in microbial communities and how these interactions affect microbial pathogenesis. Several QS systems are well understood including Gram-negative bacteria that produce acyl-homoserine lactone (AHL) signals, including Pseudomonas aeruginosa, Vibrio sp., Acinetobacter baumannii and Serratia marcescens [5, 14, 15]. Alternatively, Gram-positive species such as Staphylococcus aureus utilize autoinducer peptide (AIP)-based QS systems [16].

Various strategies for quorum sensing inhibition have been explored. The quorum sensing inhibition approaches could be accomplished via interference with the synthesis of QS signals, elimination of the signal accumulation and disruption of signal-receptor interaction [17, 18, 19].

2.1.1 Interference with the synthesis of the autoinducing signals

One of the main quorum sensing inhibiting approaches is the interference with the synthesis of the autoinducing signals [20]. AI-2 compounds are considered as “universal” signal molecules of Gram-negative and Gram-positive bacteria [14, 21]. Moreover, they are encountered in species communications. The biosynthesis of AI-2 requires two main enzymes: methylthioadenosine/S-adenosylhomocysteine nucleosidase (MTA/SAH nucleosidase) and LuxS. AI-2 molecules contribute in various virulence behaviors, biofilm formation and host-pathogen interaction. Therefore, targeting AI-2 elaborates broad spectrum quorum sensing inhibition [22, 23]. In this instance, Gutierrez group have identified the transition analogs, 5′-methylthio- (MT-), 5′-ethylthio-(EtT-) and 5′-butylthio- (BuT) DADMe-immucillin, which specifically bind and inhibit MTA enzymes in Escherichia coli O157:H7. Also, 4,5-dihydroxy-2,3-pentanedioneS-ribosyl-homocysteine analogs have been developed as competitive inhibitor of LuxS [24, 25, 26].

On other instance, inhibiting AHL-synthesis has been extensively studied, for instance, triclosan inhibited both N-3-oxo-dodecanoyl-l-homoserine lactone and N-butyryl-l-homoserine lactone [27, 28], anthranilate derivatives are a Pseudomonas quinolone signal inhibitors [28], and proanthocyanidins have been approved as inhibitor of LasI/RhlI AHL synthases expression [29]. Furthermore, precursors of Pseudomonas quinolone signals (PQS) such anthranilatic acid derivatives reduced the pathogenicity of P. aeruginosa in lung-infected mice [15].

2.1.2 Elimination of the QS signals accumulation

Other common strategy is eliminating the accumulation of the QS signals, which have been attained by degrading the QS signal using enzymes or through sequestering the signal by synthetic polymers [30, 31] or utilizing antibodies that bind with the signals. Synthesized monoclonal antibodies (AP4-24H11) by Park group provoke high binding affinity for sequestering AIP-IV and decrease α-hemolysin production in S. aureus with relief of abscess formation in the infected murine model [32]. Kaufmann and coauthors inhibited the P. aeruginosa QS cascade via development of AHL-specific monoclonal antibodies. Synthetic polymers such as itaconic acid sequester the signaling molecules AHL and attenuate QS in V. fischeri [31, 33].

Moreover, disturbing enzymes responsible for biosynthesis of QS signals is a chief method, which affects both production and accumulation of different signals and perturb quorum sensing circuit [30]. Acylases, lactonases and oxidoreductases are the widely identified enzymes that target AHLs. AHL lactonases are broad AHL degrading enzymes, which produce its effect via hydrolyzing the ester bond of the AHL ring [34]. Lactonases have been isolated from various Bacillus sp., which harbor aiiA (autoinducer inactivation gene) [35, 36]. Ulrich study showed that, the heterologous expression of aiiA in Burkholderia thailandensis and P. aeruginosa lowered the levels of AHL and QS-related virulence factors [37]. Other important AHL lactonases are AttM and AiiB, which have been isolated from Agrobacterium sp. [38], AhlD from Arthrobacterium, AhlK from Klebsiella [39] and AidC from Chryseobacterium [40], QsdA from Rhodococcus erythropolis strain W2 [41], AiiM of Microbacterium testaceum [42], AidH of Ochrobactrum sp. T63 [43] and QsdH of Pseudoalteromonas yunnanensis [44]. Furthermore, paraoxonases 1, 2 and 3 (PON1 to −3) are mammalian lactonases were identified in the airway epithelia and mammalian sera [45].

AHL acylases enzymes (aiiD) and homologs were found in Ralstonia [46], Actinoplanes utahensis and Pseudomonas sp. The purified AiiD protein has the ability to degrade 3OC10HSL into HSL and 3-oxodecanoic acid. In addition, PvdQ , QuiP and HacB are specific AHL acylases of P. aeruginosa, in addition, HacA and HacB acylases of Pseudomonas syringae [47, 48]. Furthermore, the broader substrate specificity of AHL acylase (AhlM) was detected in Streptomyces sp. strain M664 with activity towards medium- and long-chain AHLs [49].

Oxidoreductases from Rhodococcus erythropolis inactivates AHLs (oxidation or reduction) with subsequent elimination of bacterial virulence in vivo.Rhizobium strain NGR234 possess diverse AHL-inactivation loci: dhlR, qsdR1 and qsdR2, with lactonases activity, aldR, and hydR-hitR [50]. Enzymatic degradation of other QS autoinducers have been described: carA and carB from Bacillus, E. coli DH10B, Staphylococcus and Pseudomonas as the genes responsible for inhibition of DSF signaling [51]. Hod (3-hydroxy-2-methyl-4(1H)-quinolone 2,4-dioxygenase) stimulates the cleavage of PQS and attenuates PQS-regulated virulence factors. Roy and coauthors elicit the AI-2 activation activity of endogenous LsrK in E. coli, however, exogenously phosphorylation of AI-2 by LsrK eliminates its intracellular transport and hinders subsequent activation of AI-2 [52].

2.1.3 Elimination of the QS signal-receptor interaction

Interference with signal detection through eliminating the QS signal-receptor binding represents a successful approach [53, 54]. Various synthetic and natural AHL analogs have been reported to block the binding of the signal with specific receptors in P. aeruginosa and Vibrio sp. The prototype signal inhibitors, halogenated furanones, which are produced from Delisea pulchra represent a good example [55, 56]. Natural analogs have been also isolated with signal-receptor interference including ajoene [57], eugenol [58], flavonoids [59], iberin [60], furocoumarins [61], ellagic acid, penicillanic acid and patulin [62], phenethyl amide [63] and 1H-pyrrole-2-carboxylic acid [64].

The synthetic furanone derivative C-30 interferes and hinders the interaction of AHLs with the receptors [65]. Other furanone analogs have been developed including S-phenyl-l-cysteine sulfoxide and diphenyl disulfide [66] and tetrazole derivatives [67]. Furthermore, synthetic LasR derivatives have been developed such as indole derivatives, non-AHL-like antagonists [68], the synthesized azines derivatives, 4-(alkyloxy)-6-methyl-2H-pyran-2-one [69] and aspirin [70]. Triphenyl hybridγ-butyrolactones and cyclopentanones derivatives are potent inhibitors of LuxR [71]. Putative LasI inhibitors have been identified using molecular docking methods including the trans-cinnamaldehyde [72], (z)-5-octylidenethiazolidine-2, 4-dione [73] and fatty acyl purified from marine Streptomyces sp. [74]. Additionally, meta-bromo-thiolactone is a potent inhibitor of RhiI and subsequent PQS cascade [11].

In S. aureus, the interference with agr system has been accomplished using solonamide A and B that are cyclodepsipeptides derivatives, which purified from marine Photobacterium and reduced the expression of hla and RNAIII. Solonamide can act through competitive inhibition of agr system such as S. aureus agr system via structure similarity to the AIPs [75]. Other S. aureus quorum-sensing inhibitors have been identified including linear peptidomimetics as competitive inhibitors to AgrC [76], savirin as potent inhibitor of AgrA [77] and the polyhydroxy anthraquinone ω-hydroxyemodin as inhibitor of AgrA [78].

2.2 Control of the global regulators

Beside the QS regulons, other global regulators exhibit crucial functions in dominating the expression of various genes in assortment style as a response to environmental stimuli and changes, most notably the temperature change [5]. These so-called global regulators enable the bacterial communities to survive different environmental stresses including starvations, pH changes and temperature fluctuations, through the quick conformation of bacterial physiology and structure [79].

Among many regulators that coordinate gene expression in bacteria, in Gram-negative bacteria, the global regulator termed histone-like nucleoid-structuring (H-NS) protein is relatively significant and of paramount importance [80]. H-NS has been considered as the main model of studying how global regulators can affect bacterial structure and physiology. The H-NS protein is incorporated in the regulation of many genes responsible for controlling the physiological functions of Gram-negative bacterial cells involving cellular functions, survival under different environmental conditions and production of various virulence factors [81, 82]. Moreover, in Gram-positive bacteria, there are several global regulatory loci [83]. Among them in the S. aureus, SarA, a regulatory DNA binding protein involved in controlling the virulence genes expression, is well documented [84]. During regulation of the expression of various genes, these regulators have been demonstrated to act either as a positive regulators through enhancing the stability of the mRNA of expressed genes, resulting in excessive translation, or as a silencer protein that alter and decrease the gene expression by hindering binding of RNA polymerases to the promoters of target genes [85, 86].

This would open up novel approaches for the treatment and eradication of pathogenic bacteria utilizing inhibitors or modulators of these global loci to vanquish the global concerns of antimicrobial resistance and immune evasion of microbial pathogens. Among these approaches, the interesting inhibitor of SarA (SarABI), 4-[(2,4-diflurobenzyl)amino] cyclohexanol, was confirmed as SarA-based new curative medicament against S. aureus-related infections [87]. This might encourage research groups for screening other compounds that might affect global regulators in bacteria to give a new therapy for multi-drug resistant (MDR) bacterial strains.

2.3 Biofilm inhibition and eradication

Biofilm is a sessile community of microbial cells that is found to be attached to animate or inanimate surface, and usually surrounded by a matrix of polysaccharides, proteins and DNA [6]. The cells in these sessile communities differs phenotypically form those present in planktonic communities [88]. Bacterial cells in planktonic forms are almost one thousand times more sensitive to antibiotics than their biofilm counterparts [7]. Additionally, biofilms act as a defense mechanism against different stress conditions or immune cells attack [89].

In this part, we will focus on the agents that are known to exhibit antibiofilm activity.

2.3.1 Antimicrobial peptides

Antimicrobial peptides (AMPs) that are crucial players of innate immunity are reported to prevent biofilm formation in different pathogens. AMPs with antibiofilm activity are either natural or synthetic. The human cathelicidin peptide, LL-37, has been demonstrated to have antibiofilm activity in case of P. aeruginosa (at a concentration of 0.5 μg/mL), while the minimum inhibitory concentration for planktonic cells was 64 μg/mL [90]. In this study, it was reported that LL-37 was able to interfere with the adherence of microbial cells, enhancing twitching motility and downregulation of genes required for biofilm formation via affecting quorum sensing systems (Las and Rhl) [90]. Furthermore, such peptide was shown to prevent biofilm formation in E. coli and S. aureus [91]. The mouse cathelicidin-derived peptide AS10 was reported to exhibit antibiofilm activity in Candida albicans [92]. The synthetic cathelicidin-derived peptides; peptide 1018, DJK5 and DJK6, were reported to prevent biofilm formation in addition to enhancement of biofilm dispersion via prompting the hydrolysis of nucleotide signaling systems, and therefore, leads to its depletion in bacteria [93].

Another synthetic peptide, S4(1–16) M4Ka, has been found to inhibit biofilm formation and detach bacterial cells in P. aeruginosa [94]. The human β-defensin 3 (hBD-3) was found to inhibit the expression of icaA, icaD and icaR genes of Staphylococcus epidermidis, thus interfering with biofilm formation, where biofilm formation in Staphylococci is dependent on the synthesis of the polysaccharide intercellular adhesin PIA encoded by icaADBC locus [95]. Another example of human AMP with antibiofilm activity in S. epidermidis, is the liver-derived hepcidin 20. This peptide can inhibit extracellular matrix formation of biofilms via targeting PIA [95].

The natural AMP piscidin-3, obtained from fish, exhibits nucleosidase activity and can degrade extracellular DNA of P. aeruginosa [96]. Another example of natural AMP, that possesses antibiofilm activity, is esculentin, which is obtained from frog’s skin. It acts by permeabilization of the cellular membrane of P. aeruginosa PAO1 cells in the biofilm [97]. A synthetic peptide P1, derived from a tick antifreeze protein, significantly inhibited biofilm formation in Streptococcus mutans. Such peptide reduced biofilm biomass by about 75% in microtiter plates and in vitro tooth models [98].

2.3.2 Surfactants

The anionic surfactant, sodium dodecyl sulfate, has been reported to destruct biofilm via enhancing the formation of central cavity within biofilm [99]. Cetyltrimethylammonium bromide (Catanionic surfactant), together with application of high shear stress, increased the detachment of Pseudomonas fluorescens biofilms [100]. The non-ionic surfactants, polyoxy ethylene sorbitan monolaurate (Tween-20) and ethoxylated p-tert-octyl phenol (Triton X-100), were demonstrated to cause biofilm detachment [100]. Certain biosurfactants, which are surface active molecules formed by microorganisms, were reported to have antibiofilm activity. For example, surfactin, obtained from Bacillus subtilis, was found to have antibiofilm activity in case of Salmonella enterica in polyvinyl chloride microtiter wells and urethral catheters [101]. Another example is Rhamnolipids, that are produced principally, by P. aeruginosa, were found to promote the dispersal of bacterial biofilm [99]. Additionally, biosurfactants from P. fluorescens prevent the attachment of Listeria monocytogenes to stainless steel surfaces [102].

2.3.3 Free fatty acids

Free fatty acids obtained via hydrolysis of lipids by enzymes [103]. Certain members of free fatty acids are reported to exhibit antibiofilm activity [104]. For example, cis-2-decenoic acid from P. aeruginosa enhanced the dispersal of biofilms and inhibited its formation in different pathogens, such as Klebsiella pneumoniae, E. coli, Proteus mirabilis, Streptococcus pyogenes, B. subtilis and S. aureus, in addition to C. albicans [105]. Another example is cis-9-octadecenoic acid (oleic acid) that was reported to repress biofilm formation in S. aureus by interference with the initial attachment of bacterial cells [106]. The diffusible signal factor; cis-11-methyl-2-dodecenoic acid, from Xanthomonas campestris inhibits biofilm formation in case of Bacillus cereus [107]. This study showed also that diffusible signal factor or its structural analogs increased the antibiotic susceptibility of numerous bacterial pathogens, by inhibition of biofilm formation [107].

2.3.4 Metal chelators

Removal of metals from the microbial environment via metal chelators renders bacteria more susceptible to antimicrobial agents, as metals are essential for different cellular processes [108]. Ethylenediaminetetraacetic acid (EDTA), the most-known metal chelator, has been reported to exhibit antibiofilm activity against S. aureus, and to eradicate the in vivo biofilm models on catheters [109]. Combination of EDTA with minocycline has effectively reduced the colonization of S. epidermidis, S. aureus and C. albicans on catheters [110]. Similarly, the combination of EDTA and fluconazole remarkably inhibited biofilm assembly in C. albicans [111].

2.3.5 Enzymes

Based on their target, the antibiofilm enzymes are classified into three types: polysaccharide-degrading enzymes, nucleases and proteases.

2.3.5.1 Polysaccharide-degrading enzymes

Alpha amylase enzyme was found to inhibit biofilm formation by S. aureus through the detachment of biofilm and interfering with aggregation of cells [112]. Dispersion B, a bacterial glycoside hydrolase, degrades poly-N-acetylglucosamine (PNAG), a main matrix exopolysaccharide of S. aureus and E. coli [113]. Such polysaccharide is produced by many bacteria and fungi and plays an important role in surface adhesion, and biofilm formation. Furthermore, PNAG was reported to successfully disrupt the biofilm matrix of S. epidermidis [114]. Moreover, the combination of dispersion B and triclosan was reported to significantly reduce biofilm formation of E. coli, S. aureus and S. epidermidis [115].

2.3.5.2 Nucleases enzymes

Deoxyribonuclease I (DNase I) degrades DNA in biofilm matrix [104]. Moreover, it was shown to have antibiofilm activity and to detach the biofilms produced by different bacterial species [116]. Such nuclease can prevent the initial adherence of microbial cells to surfaces via the degradation of cell surface-associated nucleic acids that act as surface adhesins [117]. Furthermore, DNase I has been found to increase the sensitivity of bacterial cells in biofilm matrix to antibiotics, resulting in reduction of biofilm mass [118].

2.3.5.3 Proteases

Proteases act as antibiofilm agents because they are able to inhibit cell-cell communication, in biofilms, via hydrolysis of extracellular protein fibers and surface adhesins [104]. Subtilisins, a class of serine proteases produced by Bacillus species, were reported to prevent the adherence of microorganisms to surfaces [119]. The coating of silicone surfaces with multiple layers of amylase or acylase has been found to inhibit biofilm formation in case of P. aeruginosa and S. aureus [120]. Another example is lysostaphin, a metalloprotease produced by Staphylococcus simulans, was shown to prevent the adherence of S. aureus to lysostaphin-coated catheters [121].

2.3.6 Amino acids

d-Amino acids have been shown to inhibit biofilm formation in B. subtilis, via activating the release of amyloid fibers [122]. Such inhibitory effect was reversed by their cognate l-amino acids [123]. Furthermore, d-amino acids were shown to have antibiofilm activity in case of P. aeruginosa and S. aureus [122].

2.3.7 Nitric oxide generators

Exogenous generation of nitric oxide (NO) by agents, for example, sodium nitroprusside has been shown to trigger the bacterial growth from the biofilm form to the planktonic form via the reduction of the level of cyclic di-GMP inside the bacterial cells [104]. Further NO-generators, for example, S-nitroso-N-acetyl penicillamine and S-nitroso-l-glutathione were found also to induce the dispersion of P. aeruginosa biofilm [124]. The dispersion of biofilm by NO-generators was also demonstrated in B. subtilis [125]. Recently, it has been reported that catheters charged with NO prevented the adherence and the colonization of P. aeruginosa, E. coli and C. albicans on their surfaces [126].

2.3.8 Natural agents

Alkaloids are a group of natural organic compounds that contain a nitrogen atom and are present in different species of plants. The alkaloid berberine has been reported to inhibit biofilm formation in S. epidermidis biofilm at a concentration of 30 μg/mL, possibly via binding to the amyloid proteins in the biofilm matrix [127]. Reserpine has been shown to effectively prevent biofilm formation in K. pneumoniae at a concentration of 0.0156 mg/mL, which was 64-fold lower than its minimum inhibitory concentration [128]. Tetrandrine inhibited biofilm formation of C. albicans at a concentration of 32 mg/L, which is the MIC50 of that alkaloid against C. albicans SC5314 [129].

Guaijaverin, a flavonoid obtained from the leaves of Psidium guajava, has been shown to prevent the attachment of S. mutans to smooth surfaces by 83.7% at a concentration of 500 μg/mL. Eembelin, which is isolated from Embelia ribes, has been shown to inhibit biofilm formation in S. mutans [130]. Macelignan, isolated from the nutmegs of Myristica fragrans, was shown to reduce more than 50% of S. mutans biofilm at a concentration of 10 μg/mL [131].

Terpenes are a large class of natural hydrocarbons that are synthesized in microorganisms, plants and animals. Bakuchiol, isolated from the seeds of Psoralea corylifolia, has been shown to inhibit the adherence of S. mutans [132]. Other examples for terpenes that inhibit biofilm formation in S. mutans, are Xanthorrhizol (in combination with chlorhexidine gluconate) and casbane diterpene [133, 134].

2.4 Bacteriocins

Bacteriocins are proteins or peptides that are produced by bacteria or archaea, and are usually active against strains of bacteria that are related or unrelated to the producer strain [135]. Several bacteriocins are reported to exhibit antibiofilm activity and/or antimicrobial activity. The results of some these reports are summarized in Table 1.

BacteriocinSourceAntimicrobial activityAntibiofilm activity
Mutacin 1140Streptococcus mutansOral biofilm-associated with Streptococcus sobrinus, Streptococcus oralis [136]
Nisin ALactococcus lactis subsp. lactisEnterococcus faecalis and Streptococcus gordonii [137]Listeria monocytogenes [138]
GalliderminStaphylococcus gallinarumStaphylococcus aureus and Staphylococcus epidermidis [139, 140]
SonorensinBacillus sonorensis MT93Listeria monocytogenes and Staphylococcus aureus [141]Staphylococcus aureus [141]
Epidermicin NI0Staphylococcus epidermidisMRSA, Enterococci [142]Staphylococcus epidermidis [142]
AmylolysinBacillus amyloliquefaciens GA1Listeria monocytogenes, Staphylococcus aureus and Staphylococcus epidermidis [143]
PhilipimycinActinoplanes philippinensis MA7347MRSA [144]

Table 1.

Bacteriocins produced from different sources and exhibit antimicrobial and antibiofilm activity.

2.5 Phage therapy

Phage therapy, which is also termed viral phage therapy, is the utilization of bacteriophages as medicaments for controlling and treating diseases brought by pathogenic bacterial infections [145]. Bacteriophages, like other viruses, are obligate intracellular parasites that utilize the enzymatic machinery of their hosts for establishing their physiological functions and replication [131]. The hosts for bacteriophages are bacteria, and phages have unique ability to specifically infect bacterial hosts resulting in their lysis [8].

There are many conceivable usages for phage therapy in the treatment of crucial diseases in plants, animals as well as human [8, 145]. An outstanding advantage of utilizing bacteriophages over commonly used antibiotics, during treating infectious diseases, is their selectivity and specificity to infect and lyse infectious bacteria only without harming the host [9]. Besides, bacteriophages cause no harm to other organisms that live in a commensalism within hosts, such as the normal flora in human, which decreases significantly the incidence of superinfections or other opportunistic infections [10]. Moreover, due to their mode of action that phages replicate in vivo within their bacterial hosts, they can be used in modicum concentrations, which results in decreasing any side effects may rise during therapy and giving them a high therapeutic index [9, 10]. In addition, the capability of bacteriophages to penetrate bacterial biofilms that act as shields during the conventional antibiotic therapy, gives phages a superiority in controlling and treating diseases brought by pathogenic bacterial infections [146]. As living organisms, the capability of bacteriophages of continuous evolution, gives them the ability to overcome any resistance that can be developed by the evolution of pathogenic bacteria [146, 147]. All these tremendous advantages put the bacteriophage treatment as a superior alternative for treating diseases brought about multidrug resistant MDR bacterial pathogens [132]. On the other hand, the high bacterial host specificity of bacteriophages is encountered as a disadvantage during therapy, where, a phage can kill only its specific bacterial strain. However, this drawback can be solved by utilizing mixtures of bacteriophages, which is termed phage cocktails that have different pathogenic specific bacterial hosts as targets, to enhance the opportunities of unguis complete treatment [148]. Attention must be given, during the preparation of these cocktails, to the fact of continuous evolution of new MDR strains, so the cocktails must be updated periodically to be sufficient enough to treat infections brought by these strains [148, 149].

Historically, the first trials for the utilization of bacteriophages as medicaments for treating bacterial pathogens was reported in the Eastern world before the discovery of marvelous medicaments so-called antibiotics; however, there was any report of their usage in the Western world [150, 151]. The ability of bacteriophages to infect and lyse pathogenic bacteria was discovered by the scientists Frederick Twort and Felix D’Hérelle, who worked on Shigella dysenteriae [152]. They found that the cultures of stool specimens recovered from convalescent patients who were suffering from Shigella dysentery always depicting a high titer of phages [153]. Subsequently, they recorded that phages are the most abundant organisms in the environment and there are many sources where they can be found combined with their bacterial hosts; including gut and feces of convalescent patients as well as sewages [153]. Thereafter, due to their ubiquity especially in sewages, bacteriophages were widely utilized as medicaments for controlling and eradication of diseases brought by pathogenic bacteria [8].

It has been estimated that there are more than 100 different phage species and at least 10 phages for each bacterium. The International Committee for the Taxonomy of Viruses (ICTV) was affirmed at 1971 with the objective to always bring to date the taxonomic guidelines of viruses. The ICTV classified tailed bacteriophages (bacterial infecting phages) under the order of viruses which is termed Caudovirales. In this respect, three main families are involved within this order named Siphoviridae, Myoviridae and Podoviridae. The main difference between bacteriophages belonging to each of these families is the characteristics of the tail. Phages under the Siphoviridae family have long and non-contractile tails, and those belong to Myoviridae family have long and contractile tails, while those belong to the Podoviridae family have short, stubbed tails and a striking lack of features. Each of these three families can also be divided into different genera [8].

Compared with antibiotics and other therapeutic regimens, the steps and cost of production of bacteriophages are much easier and cheaper, respectively [10]. The easiest process for capturing of bacteriophages is done through collecting samples that seem to involve high titers of phages like sewage water samples. The collected samples are inoculated with the host bacterium, which seems to be infected by phages, on suitable growth medium. The successful isolation of certain lytic phage is depicted by the presence of clear inhibition zones in which bacteria cannot grow termed plaques; which indicates the lytic power of the isolated phage. Thereafter, the titer of isolated phage is increased by passing the phage in its specific bacterial strain several times to increase its concentration. Then, the pure supernatants containing phages are gained by centrifugation of bacterial/phage mixture, filtered through bacterial filters to remove any bacterial debris and pure phages are participated using special solutions containing NaCl and polyethylene glycol 8000 (PEG8000) [154].

Caution must be given during isolation of phages as a type called lysogenic bacteriophage may be isolated rather than the required bacterial pathogen killing type, which is called lytic bacteriophage. Lysogenic bacteriophages do not lyse bacterial cells, but they perform as tools for transfer of genetic elements of the nucleic acid between bacteria; including the genes responsible for antibiotic resistance. Fortunately, the most abundant phages are of the lytic type not the lysogenic [8, 145, 150].

Practically, bacteriophages can be dispensed and used through many routes including; less commonly oral or systemic route and most commonly topical route as sprays, liquid solutions or their application on surgical dressings for the treatment of wound infections [154]. The possibility of their clearance during the presence in blood stream by immune system or presence of any trace hazards of chemicals or parts of the bacterial host used during their production, made bacteriophage usage as intravenous injections uncommon and very rare [148, 149]. Lyophilization of bacteriophages and their production as solid dosage forms as pills or tablets do not decrease their potency and increase their shelf life as oral dosage forms [155, 156]. The supplementation of oral forms of phages, either solid or liquid, with antacid increases its stability, as it protect them from the high acidity during their bypassing in the stomach [155, 156].

The application of bacteriophages as therapeutic medicaments has been extensively reported. For example, in the field of human health promotion and food protection, different bacteriophages have been employed to eradicate common bacterial pathogens that may cause food spoilage as Listeria sp. and Campylobacter sp. [157, 158]. In the fields of veterinary medicine and agriculture different bacteriophages were employed to control and eradicate bacterial pathogens like Xanthomonas, Escherichia, Campylobacter and Salmonella [159]. Moreover, in the field of fish production and aquacultures, different bacteriophages were employed to control and eradicate bacterial pathogens like Vibrio sp. [160]. In the field of human medicine, different bacteriophages were employed to control and eradicate bacterial pathogens including P. aeruginosa, Staphylococci, Streptococci, E. coli, Vibrio and Shigella and Mycobacterium sp. [161, 162]. Most recent application of bacteriophages in human medicine is their utilization as drug delivery system, which is very interesting as they can be used for the delivery of common antibiotics [163, 164] or antitumor agents [165].

A more recent policy, termed enzybiotic, for using phages as therapeutic agents is the utilization of their enzymes only, which are produced by recombinant technology, combined with other antibacterial agents or as a separate antibacterial agents [166].

As other therapeutic regimens for controlling bacterial pathogens, the patients may develop extensive fever and shock, when the bacteria are lysed due to the release of what is called pyrogens or endotoxins within the patient [167]. This problem can be coped during phage therapy through the utilization of genetically modified phages that harbor enzymes having the ability to lyse these endotoxins and the other bacterial structures into harmless products [168].

Examples of therapeutic approaches of bacteriophages and their enzymes are illustrated in Table 2.

Infection/diseaseModelCausative agentRoute of administration of phages/enzymesTreatment outcomesReference
Chronic otitisHumanPseudomonas aeruginosaOral administration of phagesSuccessful treatment[169]
TyphoidHumanSalmonella typhiOral administration of phagesSuccessful treatment[170]
Diabetic foot ulcerHumanStaphylococcus aureusTopical application of phagesSuccessful treatment[171]
SepsisMurineVibrio parahaemolyticusIntraperitoneal and oral administration of phagesSuccessful treatment[172]
PneumoniaMurinePseudomonas aeruginosaIntranasal administration of phagesSuccessful treatment[154]
Ulcers and woundsHumanProteus vulgarisTopical application of phagesSuccessful treatment[173]
MeningitisMurineEscherichia coliIntraperitoneal or subcutaneous administration of phages.Successful treatment[174]
SepsisMurineAcinetobacter baumanniiIntraperitoneal administration of phagesSuccessful treatment[175]
BacteremiaMurineEnterococcus faeciumIntraperitoneal administration of phagesSuccessful treatment[176]
IleocecitisHamsterClostridium difficileOral administration of phagesSuccessful treatment[177]
DysenteryHumanShigella dysenteriaeOral administration of phagesSuccessful treatment[178]
CholeraHumanVibrio choleraeOral administration of phagesSuccessful treatment[178]
PneumoniaMurineStreptococcus pneumoniaeIntraperitoneal administration of Cpl-1 lysin enzymeSuccessful treatment[179]
BacteremiaMurineStreptococcus pyogenesIntraperitoneal administration of PlySs2 lysin enzymeSuccessful treatment[179]
In vitroIn vitroBacillus anthracisApplication of PlyG lysin enzymeSignificant reduction in bacterial density[180]
EndophthalmitisMurineStaphylococcus aureusApplication of Ply187 lysin as eye dropsSuccessful treatment[181]
BacteremiaMurineAcinetobacter baumanniiAdministration of PlyF307 lysin enzymeSuccessful treatment[182]
In vitroIn vitroPseudomonas aeruginosa and Salmonella typhimuriumApplication of ABgp46 lysin enzymeSignificant reduction in bacterial density[183]

Table 2.

Therapeutic approaches of bacteriophages and their enzymes.

Advertisement

3. Conclusion

Various approaches have been developed for competing microbial virulence and resistance. Quorum sensing signals and global regulators play an essential role in controlling the gene expression of virulence factors, and the expression of proteins required for adaptation to environmental and stress condition. Therefore, control of these regulators will stop the microbial pathogenicity. In addition, biofilms act as a defense mechanism against host immunity and antimicrobial therapy. Natural and synthetic compounds have approved activities in eradication of biofilm formation. Besides, phage therapy, which is currently successful in destruction of bacterial pathogens that do not respond to conventional antimicrobials. These methods would open up new perspectives for management the up growing problem of microbial resistance. Further, in vivo studies are required for real applications of these trends in eradication of microbial infections.

References

  1. 1. Reuter K, Steinbach A, Helms V. Interfering with bacterial quorum sensing. Perspectives in Medicinal Chemistry. 2016;8:1-15
  2. 2. Finch RG, Pritchard DI, Bycroft BW, Williams P, Stewart GS. Quorum sensing: A novel target for anti-infective therapy. The Journal of Antimicrobial Chemotherapy. 1998;42(5):569-571
  3. 3. Miller MB, Bassler BL. Quorum sensing in bacteria. Annual Review of Microbiology. 2001;55:165-199
  4. 4. Geske GD, O’Neill JC, Miller DM, Mattmann ME, Blackwell HE. Modulation of bacterial quorum sensing with synthetic ligands: Systematic evaluation of N-acylated homoserine lactones in multiple species and new insights into their mechanisms of action. Journal of the American Chemical Society. 2007;129(44):13613-13625
  5. 5. Elgaml A, Miyoshi SI. Regulation systems of protease and hemolysin production in Vibrio vulnificus. Microbiology and Immunology. 2017;61(1):1-11
  6. 6. Bogino PC, Oliva Mde L, Sorroche FG, Giordano W. The role of bacterial biofilms and surface components in plant-bacterial associations. International Journal of Molecular Sciences. 2013;14(8):15838-15859
  7. 7. Hengzhuang W, Wu H, Ciofu O, Song Z, Høiby N. Pharmacokinetics/pharmacodynamics of colistin and imipenem on mucoid and nonmucoid Pseudomonas aeruginosa biofilms. Antimicrobial Agents and Chemotherapy. 2011;55(9):4469-4474
  8. 8. Abedon ST. Phage therapy: Various perspectives on how to improve the art. Methods in Molecular Biology. 1734;2018:113-127
  9. 9. Grasis JA. Host-associated bacteriophage isolation and preparation for viral metagenomics. Methods in Molecular Biology. 1746;2018:1-25
  10. 10. Nobrega FL, Costa AR, Kluskens LD, Azeredo J. Revisiting phage therapy: New applications for old resources. Trends in Microbiology. 2015;23(4):185-191
  11. 11. O’Loughlin CT, Miller LC, Siryaporn A, Drescher K, Semmelhack MF, Bassler BL. A quorum-sensing inhibitor blocks Pseudomonas aeruginosa virulence and biofilm formation. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(44):17981-17986
  12. 12. Hornby JM, Jensen EC, Lisec AD, Tasto JJ, Jahnke B, Shoemaker R, et al. Quorum sensing in the dimorphic fungus Candida albicans is mediated by farnesol. Applied and Environmental Microbiology. 2001;67(7):2982-2992
  13. 13. Erez Z, Steinberger-Levy I, Shamir M, Doron S, Stokar-Avihail A, Peleg Y, et al. Communication between viruses guides lysis-lysogeny decisions. Nature. 2017;541(7638):488-493
  14. 14. Schaefer AL, Val DL, Hanzelka BL, Cronan JE Jr, Greenberg EP. Generation of cell-to-cell signals in quorum sensing: Acyl homoserine lactone synthase activity of a purified Vibrio fischeri LuxI protein. Proceedings of the National Academy of Sciences of the United States of America. 1996;93(18):9505-9509
  15. 15. Calfee MW, Coleman JP, Pesci EC. Interference with pseudomonas quinolone signal synthesis inhibits virulence factor expression by Pseudomonas aeruginosa. Proceedings of the National Academy of Sciences of the United States of America. 2001;98(20):11633-11637
  16. 16. Xiong YQ , Willard J, Yeaman MR, Cheung AL, Bayer AS. Regulation of Staphylococcus aureus alpha-toxin gene (hla) expression by agr, sarA, and sae in vitro and in experimental infective endocarditis. The Journal of Infectious Diseases. 2006;194(9):1267-1275
  17. 17. Kong C, Neoh HM, Nathan S. Targeting Staphylococcus aureus toxins: A potential form of anti-virulence therapy. Toxins. 2016;8(3): pii: E72; 1-21
  18. 18. Remy B, Mion S, Plener L, Elias M, Chabriere E, Daude D. Interference in bacterial quorum sensing: A biopharmaceutical perspective. Frontiers in Pharmacology. 2018;9:203
  19. 19. Shaaban M, Elgaml A, Habib EE. Biotechnological applications of quorum sensing inhibition as novel therapeutic strategies for multidrug resistant pathogens. Microbial Pathogenesis. 2019;127:138-143
  20. 20. Sedlmayer F, Hell D, Muller M, Auslander D, Fussenegger M. Designer cells programming quorum-sensing interference with microbes. Nature Communications. 2018;9(1):1822
  21. 21. Xavier KB, Bassler BL. LuxS quorum sensing: More than just a numbers game. Current Opinion in Microbiology. 2003;6(2):191-197
  22. 22. Guo M, Gamby S, Nakayama S, Smith J, Sintim HO. A pro-drug approach for selective modulation of AI-2-mediated bacterial cell-to-cell communication. Sensors. 2012;12(3):3762-3772
  23. 23. Ren D, Li C, Qin Y, Yin R, Li X, Tian M, et al. Inhibition of Staphylococcus aureus adherence to Caco-2 cells by lactobacilli and cell surface properties that influence attachment. Anaerobe. 2012;18(5):508-515
  24. 24. Gutierrez JA, Crowder T, Rinaldo-Matthis A, Ho MC, Almo SC, Schramm VL. Transition state analogs of 5′-methylthioadenosine nucleosidase disrupt quorum sensing. Nature Chemical Biology. 2009;5(4):251-257
  25. 25. Malladi VL, Sobczak AJ, Meyer TM, Pei D, Wnuk SF. Inhibition of LuxS by S-ribosylhomocysteine analogues containing a [4-aza]ribose ring. Bioorganic & Medicinal Chemistry. 2011;19(18):5507-5519
  26. 26. Wnuk SF, Lalama J, Garmendia CA, Robert J, Zhu J, Pei D. S-Ribosylhomocysteine analogues with the carbon-5 and sulfur atoms replaced by a vinyl or (fluoro)vinyl unit. Bioorganic & Medicinal Chemistry. 2008;16(9):5090-5102
  27. 27. Hoang TT, Schweizer HP. Characterization of Pseudomonas aeruginosa enoyl-acyl carrier protein reductase (FabI): A target for the antimicrobial triclosan and its role in acylated homoserine lactone synthesis. Journal of Bacteriology. 1999;181(17):5489-5497
  28. 28. Coleman JP, Hudson LL, McKnight SL, Farrow JM 3rd, Calfee MW, Lindsey CA, et al. Pseudomonas aeruginosa PqsA is an anthranilate-coenzyme A ligase. Journal of Bacteriology. 2008;190(4):1247-1255
  29. 29. Maisuria VB, Los Santos YL, Tufenkji N, Deziel E. Cranberry-derived proanthocyanidins impair virulence and inhibit quorum sensing of Pseudomonas aeruginosa. Scientific Reports. 2016;6:30169
  30. 30. LaSarre B, Federle MJ. Exploiting quorum sensing to confuse bacterial pathogens. Microbiology and Molecular Biology Reviews. 2013;77(1):73-111
  31. 31. Piletska EV, Stavroulakis G, Karim K, Whitcombe MJ, Chianella I, Sharma A, et al. Attenuation of Vibrio fischeri quorum sensing using rationally designed polymers. Biomacromolecules. 2010;11(4):975-980
  32. 32. Park J, Jagasia R, Kaufmann GF, Mathison JC, Ruiz DI, Moss JA, et al. Infection control by antibody disruption of bacterial quorum sensing signaling. Chemistry & Biology. 2007;14(10):1119-1127
  33. 33. Cavaleiro E, Duarte AS, Esteves AC, Correia A, Whitcombe MJ, Piletska EV, et al. Novel linear polymers able to inhibit bacterial quorum sensing. Macromolecular Bioscience. 2015;15(5):647-656
  34. 34. Dong YH, Xu JL, Li XZ, Zhang LH. AiiA, an enzyme that inactivates the acylhomoserine lactone quorum-sensing signal and attenuates the virulence of Erwinia carotovora. Proceedings of the National Academy of Sciences of the United States of America. 2000;97(7):3526-3531
  35. 35. Dong YH, Gusti AR, Zhang Q , Xu JL, Zhang LH. Identification of quorum-quenching N-acyl homoserine lactonases from Bacillus species. Applied and Environmental Microbiology. 2002;68(4):1754-1759
  36. 36. Liu D, Momb J, Thomas PW, Moulin A, Petsko GA, Fast W, et al. Mechanism of the quorum-quenching lactonase (AiiA) from Bacillus thuringiensis. 1. Product-bound structures. Biochemistry. 2008;47(29):7706-7714
  37. 37. Ulrich RL. Quorum quenching: Enzymatic disruption of N-acylhomoserine lactone-mediated bacterial communication in Burkholderia thailandensis. Applied and Environmental Microbiology. 2004;70(10):6173-6180
  38. 38. Zhang HB, Wang LH, Zhang LH. Genetic control of quorum-sensing signal turnover in Agrobacterium tumefaciens. Proceedings of the National Academy of Sciences of the United States of America. 2002;99(7):4638-4643
  39. 39. Park SY, Lee SJ, Oh TK, Oh JW, Koo BT, Yum DY, et al. AhlD, an N-acylhomoserine lactonase in Arthrobacter sp., and predicted homologues in other bacteria. Microbiology. 2003;149(Pt 6):1541-1550
  40. 40. Wang WZ, Morohoshi T, Someya N, Ikeda T. AidC, a novel N-acylhomoserine lactonase from the potato root-associated cytophaga-flavobacteria-bacteroides (CFB) group bacterium Chryseobacterium sp. strain StRB126. Applied and Environmental Microbiology. 2012;78(22):7985-7992
  41. 41. Uroz S, Oger PM, Chapelle E, Adeline MT, Faure D, Dessaux Y. A Rhodococcus qsdA-encoded enzyme defines a novel class of large-spectrum quorum-quenching lactonases. Applied and Environmental Microbiology. 2008;74(5):1357-1366
  42. 42. Wang WZ, Morohoshi T, Ikenoya M, Someya N, Ikeda T. AiiM, a novel class of N-acylhomoserine lactonase from the leaf-associated bacterium Microbacterium testaceum. Applied and Environmental Microbiology. 2010;76(8):2524-2530
  43. 43. Mei GY, Yan XX, Turak A, Luo ZQ , Zhang LQ. AidH, an alpha/beta-hydrolase fold family member from an Ochrobactrum sp. strain, is a novel N-acylhomoserine lactonase. Applied and Environmental Microbiology. 2010;76(15):4933-4942
  44. 44. Huang W, Lin Y, Yi S, Liu P, Shen J, Shao Z, et al. QsdH, a novel AHL lactonase in the RND-type inner membrane of marine Pseudoalteromonas byunsanensis strain 1A01261. PLoS One. 2012;7(10):e46587
  45. 45. Yang F, Wang LH, Wang J, Dong YH, Hu JY, Zhang LH. Quorum quenching enzyme activity is widely conserved in the sera of mammalian species. FEBS Letters. 2005;579(17):3713-3717
  46. 46. Lin YH, Xu JL, Hu J, Wang LH, Ong SL, Leadbetter JR, et al. Acyl-homoserine lactone acylase from Ralstonia strain XJ12B represents a novel and potent class of quorum-quenching enzymes. Molecular Microbiology. 2003;47(3):849-860
  47. 47. Bokhove M, Nadal Jimenez P, Quax WJ, Dijkstra BW. The quorum-quenching N-acyl homoserine lactone acylase PvdQ is an Ntn-hydrolase with an unusual substrate-binding pocket. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(2):686-691
  48. 48. Huang JJ, Petersen A, Whiteley M, Leadbetter JR. Identification of QuiP, the product of gene PA1032, as the second acyl-homoserine lactone acylase of Pseudomonas aeruginosa PAO1. Applied and Environmental Microbiology. 2006;72(2):1190-1197
  49. 49. Park SY, Kang HO, Jang HS, Lee JK, Koo BT, Yum DY. Identification of extracellular N-acylhomoserine lactone acylase from a Streptomyces sp. and its application to quorum quenching. Applied and Environmental Microbiology. 2005;71(5):2632-2641
  50. 50. Krysciak D, Schmeisser C, Preuss S, Riethausen J, Quitschau M, Grond S, et al. Involvement of multiple loci in quorum quenching of autoinducer I molecules in the nitrogen-fixing symbiont Rhizobium (Sinorhizobium) sp. strain NGR234. Applied and Environmental Microbiology. 2011;77(15):5089-5099
  51. 51. Llamas I, Suarez A, Quesada E, Bejar V, del Moral A. Identification and characterization of the carAB genes responsible for encoding carbamoylphosphate synthetase in Halomonas eurihalina. Extremophiles. 2003;7(3):205-211
  52. 52. Roy V, Fernandes R, Tsao CY, Bentley WE. Cross species quorum quenching using a native AI-2 processing enzyme. ACS Chemical Biology. 2010;5(2):223-232
  53. 53. Koch B, Liljefors T, Persson T, Nielsen J, Kjelleberg S, Givskov M. The LuxR receptor: The sites of interaction with quorum-sensing signals and inhibitors. Microbiology. 2005;151(Pt 11):3589-3602
  54. 54. Singh RP, Desouky SE, Nakayama J. Quorum quenching strategy targeting Gram-positive pathogenic bacteria. Advances in Experimental Medicine and Biology. 2016;901:109-130
  55. 55. Manefield M, de Nys R, Kumar N, Read R, Givskov M, Steinberg P, et al. Evidence that halogenated furanones from Delisea pulchra inhibit acylated homoserine lactone (AHL)-mediated gene expression by displacing the AHL signal from its receptor protein. Microbiology. 1999;145(Pt 2):283-291
  56. 56. Givskov M, de Nys R, Manefield M, Gram L, Maximilien R, Eberl L, et al. Eukaryotic interference with homoserine lactone-mediated prokaryotic signalling. Journal of Bacteriology. 1996;178(22):6618-6622
  57. 57. Fong J, Yuan M, Jakobsen TH, Mortensen KT, Delos Santos MM, Chua SL, et al. Disulfide bond-containing Ajoene analogues As novel quorum sensing inhibitors of Pseudomonas aeruginosa. Journal of Medicinal Chemistry. 2017;60(1):215-227
  58. 58. Zhou L, Zheng H, Tang Y, Yu W, Gong Q. Eugenol inhibits quorum sensing at sub-inhibitory concentrations. Biotechnology Letters. 2013;35(4):631-637
  59. 59. Paczkowski JE, Mukherjee S, McCready AR, Cong JP, Aquino CJ, Kim H, et al. Flavonoids suppress Pseudomonas aeruginosa virulence through allosteric inhibition of quorum-sensing receptors. The Journal of Biological Chemistry. 2017;292(10):4064-4076
  60. 60. Jakobsen TH, Bragason SK, Phipps RK, Christensen LD, van Gennip M, Alhede M, et al. Food as a source for quorum sensing inhibitors: Iberin from horseradish revealed as a quorum sensing inhibitor of Pseudomonas aeruginosa. Applied and Environmental Microbiology. 2012;78(7):2410-2421
  61. 61. Girennavar B, Cepeda ML, Soni KA, Vikram A, Jesudhasan P, Jayaprakasha GK, et al. Grapefruit juice and its furocoumarins inhibits autoinducer signaling and biofilm formation in bacteria. International Journal of Food Microbiology. 2008;125(2):204-208
  62. 62. Rasmussen TB, Skindersoe ME, Bjarnsholt T, Phipps RK, Christensen KB, Jensen PO, et al. Identity and effects of quorum-sensing inhibitors produced by Penicillium species. Microbiology. 2005;151(Pt 5):1325-1340
  63. 63. Teasdale ME, Liu J, Wallace J, Akhlaghi F, Rowley DC. Secondary metabolites produced by the marine bacterium Halobacillus salinus that inhibit quorum sensing-controlled phenotypes in gram-negative bacteria. Applied and Environmental Microbiology. 2009;75(3):567-572
  64. 64. Hassan R, Shaaban MI, Abdel Bar FM, El-Mahdy AM, Shokralla S. Quorum sensing inhibiting activity of Streptomyces coelicoflavus isolated from soil. Frontiers in Microbiology. 2016;7:659
  65. 65. Wu H, Song Z, Hentzer M, Andersen JB, Molin S, Givskov M, et al. Synthetic furanones inhibit quorum-sensing and enhance bacterial clearance in Pseudomonas aeruginosa lung infection in mice. The Journal of Antimicrobial Chemotherapy. 2004;53(6):1054-1061
  66. 66. Cady NC, McKean KA, Behnke J, Kubec R, Mosier AP, Kasper SH, et al. Inhibition of biofilm formation, quorum sensing and infection in Pseudomonas aeruginosa by natural products-inspired organosulfur compounds. PLoS One. 2012;7(6):e38492
  67. 67. Muh U, Schuster M, Heim R, Singh A, Olson ER, Greenberg EP. Novel Pseudomonas aeruginosa quorum-sensing inhibitors identified in an ultra-high-throughput screen. Antimicrobial Agents and Chemotherapy. 2006;50(11):3674-3679
  68. 68. Biswas NN, Kutty SK, Barraud N, Iskander GM, Griffith R, Rice SA, et al. Indole-based novel small molecules for the modulation of bacterial signalling pathways. Organic & Biomolecular Chemistry. 2015;13(3):925-937
  69. 69. Park S, Kim HS, Ok K, Kim Y, Park HD, Byun Y. Design, synthesis and biological evaluation of 4-(alkyloxy)-6-methyl-2H-pyran-2-one derivatives as quorum sensing inhibitors. Bioorganic & Medicinal Chemistry Letters. 2015;25(15):2913-2917
  70. 70. El-Mowafy SA, Abd El Galil KH, El-Messery SM, Shaaban MI. Aspirin is an efficient inhibitor of quorum sensing, virulence and toxins in Pseudomonas aeruginosa. Microbial Pathogenesis. 2014;74:25-32
  71. 71. O’Reilly MC, Blackwell HE. Structure-based design and biological evaluation of triphenyl scaffold-based hybrid compounds as hydrolytically stable modulators of a LuxR-type quorum sensing receptor. ACS Infectious Diseases. 2016;2(1):32-38
  72. 72. Chang CY, Krishnan T, Wang H, Chen Y, Yin WF, Chong YM, et al. Non-antibiotic quorum sensing inhibitors acting against N-acyl homoserine lactone synthase as druggable target. Scientific Reports. 2014;4:7245
  73. 73. Lidor O, Al-Quntar A, Pesci EC, Steinberg D. Mechanistic analysis of a synthetic inhibitor of the Pseudomonas aeruginosa LasI quorum-sensing signal synthase. Scientific Reports. 2015;5:16569
  74. 74. Kamarudheen N, Rao KVB. Fatty acyl compounds from marine Streptomyces griseoincarnatus strain HK12 against two major bio-film forming nosocomial pathogens; an in vitro and in silico approach. Microbial Pathogenesis. 2019;127:121-130
  75. 75. Mansson M, Nielsen A, Kjaerulff L, Gotfredsen CH, Wietz M, Ingmer H, et al. Inhibition of virulence gene expression in Staphylococcus aureus by novel depsipeptides from a marine photobacterium. Marine Drugs. 2011;9(12):2537-2552
  76. 76. Karathanasi G, Bojer MS, Baldry M, Johannessen BA, Wolff S, Greco I, et al. Linear peptidomimetics as potent antagonists of Staphylococcus aureus agr quorum sensing. Scientific Reports. 2018;8(1):3562
  77. 77. Sully EK, Malachowa N, Elmore BO, Alexander SM, Femling JK, Gray BM, et al. Selective chemical inhibition of agr quorum sensing in Staphylococcus aureus promotes host defense with minimal impact on resistance. PLoS Pathogens. 2014;10(6):e1004174
  78. 78. Daly SM, Elmore BO, Kavanaugh JS, Triplett KD, Figueroa M, Raja HA, et al. Omega-hydroxyemodin limits Staphylococcus aureus quorum sensing-mediated pathogenesis and inflammation. Antimicrobial Agents and Chemotherapy. 2015;59(4):2223-2235
  79. 79. Lee SE, Kim SY, Kim CM, Kim MK, Kim YR, Jeong K, et al. The pyrH gene of Vibrio vulnificus is an essential in vivo survival factor. Infection and Immunity. 2007;75(6):2795-2801
  80. 80. Elgaml A, Miyoshi S. Role of the histone-like nucleoid structuring protein (H-NS) in the regulation of virulence factor expression and stress response in Vibrio vulnificus. Biocontrol Science. 2015;20(4):263-274
  81. 81. Dorman CJ. H-NS: A universal regulator for a dynamic genome. Nature Reviews. Microbiology. 2004;2(5):391-400
  82. 82. Dorman CJ, Deighan P. Regulation of gene expression by histone-like proteins in bacteria. Current Opinion in Genetics & Development. 2003;13(2):179-184
  83. 83. Zheng W, Liang Y, Zhao H, Zhang J, Li Z. 5,5′-methylenedisalicylic acid (MDSA) modulates SarA/MgrA phosphorylation by targeting Ser/Thr phosphatase Stp1. Chembiochem. 2015;16(7):1035-1040
  84. 84. Cheung AL, Nishina KA, Trotonda MP, Tamber S. The SarA protein family of Staphylococcus aureus. The International Journal of Biochemistry & Cell Biology. 2008;40(3):355-361
  85. 85. Ono S, Goldberg MD, Olsson T, Esposito D, Hinton JC, Ladbury JE. H-NS is a part of a thermally controlled mechanism for bacterial gene regulation. The Biochemical Journal. 2005;391(Pt 2):203-213
  86. 86. Brescia CC, Kaw MK, Sledjeski DD. The DNA binding protein H-NS binds to and alters the stability of RNA in vitro and in vivo. Journal of Molecular Biology. 2004;339(3):505-514
  87. 87. Arya R, Ravikumar R, Santhosh RS, Princy SA. SarA based novel therapeutic candidate against Staphylococcus aureus associated with vascular graft infections. Frontiers in Microbiology. 2015;6:416
  88. 88. Donlan RM, Costerton JW. Biofilms: Survival mechanisms of clinically relevant microorganisms. Clinical Microbiology Reviews. 2002;15(2):167-193
  89. 89. Roy R, Tiwari M, Donelli G, Tiwari V. Strategies for combating bacterial biofilms: A focus on anti-biofilm agents and their mechanisms of action. Virulence. 2018;9(1):522-554
  90. 90. Overhage J, Campisano A, Bains M, Torfs EC, Rehm BH, Hancock RE. Human host defense peptide LL-37 prevents bacterial biofilm formation. Infection and Immunity. 2008;76(9):4176-4182
  91. 91. Aka ST. Killing efficacy and anti-biofilm activity of synthetic human cationic antimicrobial peptide cathelicidin hCAP-18/LL37 against urinary tract pathogens. Journal of Microbiology and Infectious Diseases. 2015;5:15-20
  92. 92. De Brucker K, Delattin N, Robijns S, Steenackers H, Verstraeten N, Landuyt B, et al. Derivatives of the mouse cathelicidin-related antimicrobial peptide (CRAMP) inhibit fungal and bacterial biofilm formation. Antimicrobial Agents and Chemotherapy. 2014;58(9):5395-5404
  93. 93. De la Fuente-Núñez C, Reffuveille F, Haney EF, Straus SK, Hancock RE. Broad-spectrum anti-biofilm peptide that targets a cellular stress response. PLoS Pathogens. 2014;10(5):e1004152
  94. 94. Quiles F, Saadi S, Francius G, Bacharouche J, Humbert F. In situ and real time investigation of the evolution of a Pseudomonas fluorescens nascent biofilm in the presence of an antimicrobial peptide. Biochimica et Biophysica Acta. 2016;1858(1):75-84
  95. 95. Rohde H, Frankenberger S, Zahringer U, Mack D. Structure, function and contribution of polysaccharide intercellular adhesin (PIA) to Staphylococcus epidermidis biofilm formation and pathogenesis of biomaterial-associated infections. European Journal of Cell Biology. 2010;89(1):103-111
  96. 96. Libardo MDJ, Bahar AA, Ma B, Fu R, McCormick LE, Zhao J, et al. Nuclease activity gives an edge to host-defense peptide piscidin 3 over piscidin 1, rendering it more effective against persisters and biofilms. The FEBS Journal. 2017;284(21):3662-3683
  97. 97. Luca V, Stringaro A, Colone M, Pini A, Mangoni ML. Esculentin(1-21), an amphibian skin membrane-active peptide with potent activity on both planktonic and biofilm cells of the bacterial pathogen Pseudomonas aeruginosa. Cellular and Molecular Life Sciences: CMLS. 2013;70(15):2773-2786
  98. 98. Ansari JM, Abraham NM, Massaro J, Murphy K, Smith-Carpenter J, Fikrig E. Anti-biofilm activity of a self-aggregating peptide against Streptococcus mutans. Frontiers in Microbiology. 2017;8:488
  99. 99. Boles BR, Thoendel M, Singh PK. Rhamnolipids mediate detachment of Pseudomonas aeruginosa from biofilms. Molecular Microbiology. 2005;57(5):1210-1223
  100. 100. Simoes M, Pereira MO, Vieira MJ. Action of a cationic surfactant on the activity and removal of bacterial biofilms formed under different flow regimes. Water Research. 2005;39(2-3):478-486
  101. 101. Mireles JR 2nd, Toguchi A, Harshey RM. Salmonella enterica serovar typhimurium swarming mutants with altered biofilm-forming abilities: Surfactin inhibits biofilm formation. Journal of Bacteriology. 2001;183(20):5848-5854
  102. 102. Meylheuc T, van Oss CJ, Bellon-Fontaine MN. Adsorption of biosurfactant on solid surfaces and consequences regarding the bioadhesion of Listeria monocytogenes LO28. Journal of Applied Microbiology. 2001;91(5):822-832
  103. 103. Desbois AP, Smith VJ. Antibacterial free fatty acids: Activities, mechanisms of action and biotechnological potential. Applied Microbiology and Biotechnology. 2010;85(6):1629-1642
  104. 104. Li X-H, Lee J-H. Antibiofilm agents: A new perspective for antimicrobial strategy. Journal of Microbiology. 2017;55(10):753-766
  105. 105. Davies DG, Marques CN. A fatty acid messenger is responsible for inducing dispersion in microbial biofilms. Journal of Bacteriology. 2009;191(5):1393-1403
  106. 106. Stenz L, Francois P, Fischer A, Huyghe A, Tangomo M, Hernandez D, et al. Impact of oleic acid (cis-9-octadecenoic acid) on bacterial viability and biofilm production in Staphylococcus aureus. FEMS Microbiology Letters. 2008;287(2):149-155
  107. 107. Deng Y, Lim A, Lee J, Chen S, An S, Dong YH, et al. Diffusible signal factor (DSF) quorum sensing signal and structurally related molecules enhance the antimicrobial efficacy of antibiotics against some bacterial pathogens. BMC Microbiology. 2014;14:51
  108. 108. Gadd GM. Metals, minerals and microbes: Geomicrobiology and bioremediation. Microbiology (Reading, England). 2010;156(Pt 3):609-643
  109. 109. Kite P, Eastwood K, Sugden S, Percival SL. Use of in vivo-generated biofilms from hemodialysis catheters to test the efficacy of a novel antimicrobial catheter lock for biofilm eradication in vitro. Journal of Clinical Microbiology. 2004;42(7):3073-3076
  110. 110. Raad I, Chatzinikolaou I, Chaiban G, Hanna H, Hachem R, Dvorak T, et al. In vitro and ex vivo activities of minocycline and EDTA against microorganisms embedded in biofilm on catheter surfaces. Antimicrobial Agents and Chemotherapy. 2003;47(11):3580-3585
  111. 111. Casalinuovo IA, Sorge R, Bonelli G, Di Francesco P. Evaluation of the antifungal effect of EDTA, a metal chelator agent, on Candida albicans biofilm. European Review for Medical and Pharmacological Sciences. 2017;21(6):1413-1420
  112. 112. Craigen B, Dashiff A, Kadouri DE. The use of commercially available alpha-amylase compounds to inhibit and remove Staphylococcus aureus biofilms. Open Microbiology Journal. 2011;5:21-31
  113. 113. Ramasubbu N, Thomas LM, Ragunath C, Kaplan JB. Structural analysis of dispersin B, a biofilm-releasing glycoside hydrolase from the periodontopathogen Actinobacillus actinomycetemcomitans. Journal of Molecular Biology. 2005;349(3):475-486
  114. 114. Chaignon P, Sadovskaya I, Ragunah C, Ramasubbu N, Kaplan JB, Jabbouri S. Susceptibility of staphylococcal biofilms to enzymatic treatments depends on their chemical composition. Applied Microbiology and Biotechnology. 2007;75(1):125-132
  115. 115. Darouiche RO, Mansouri MD, Gawande PV, Madhyastha S. Antimicrobial and antibiofilm efficacy of triclosan and DispersinB combination. Journal of Antimicrobial Chemotherapy. 2009;64(1):88-93
  116. 116. Kaplan JB. Therapeutic potential of biofilm-dispersing enzymes. The International Journal of Artificial Organs. 2009;32(9):545-554
  117. 117. Qin Z, Ou Y, Yang L, Zhu Y, Tolker-Nielsen T, Molin S, et al. Role of autolysin-mediated DNA release in biofilm formation of Staphylococcus epidermidis. Microbiology (Reading, England). 2007;153(Pt 7):2083-2092
  118. 118. Tetz GV, Artemenko NK, Tetz VV. Effect of DNase and antibiotics on biofilm characteristics. Antimicrobial Agents and Chemotherapy. 2009;53(3):1204-1209
  119. 119. Leroy C, Delbarre C, Ghillebaert F, Compere C, Combes D. Effects of commercial enzymes on the adhesion of a marine biofilm-forming bacterium. Biofouling. 2008;24(1):11-22
  120. 120. Ivanova K, Fernandes MM, Francesko A, Mendoza E, Guezguez J, Burnet M, et al. Quorum-quenching and matrix-degrading enzymes in multilayer coatings synergistically prevent bacterial biofilm formation on urinary catheters. ACS Applied Materials & Interfaces. 2015;7(49):27066-27077
  121. 121. Shah A, Mond J, Walsh S. Lysostaphin-coated catheters eradicate Staphylococccus aureus challenge and block surface colonization. Antimicrobial Agents and Chemotherapy. 2004;48(7):2704-2707
  122. 122. Kolodkin-Gal I, Romero D, Cao S, Clardy J, Kolter R, Losick R. D-amino acids trigger biofilm disassembly. Science. 2010;328(5978):627-629
  123. 123. Leiman SA, May JM, Lebar MD, Kahne D, Kolter R, Losick R. D-amino acids indirectly inhibit biofilm formation in Bacillus subtilis by interfering with protein synthesis. Journal of Bacteriology. 2013;195(23):5391-5395
  124. 124. Barraud N, Schleheck D, Klebensberger J, Webb JS, Hassett DJ, Rice SA, et al. Nitric oxide signaling in Pseudomonas aeruginosa biofilms mediates phosphodiesterase activity, decreased cyclic di-GMP levels, and enhanced dispersal. Journal of Bacteriology. 2009;191(23):7333-7342
  125. 125. Schreiber F, Beutler M, Enning D, Lamprecht-Grandio M, Zafra O, Gonzalez-Pastor JE, et al. The role of nitric-oxide-synthase-derived nitric oxide in multicellular traits of Bacillus subtilis 3610: Biofilm formation, swarming, and dispersal. BMC Microbiology. 2011;11:111
  126. 126. Margel D, Mizrahi M, Regev-Shoshani G, Ko M, Moshe M, Ozalvo R, et al. Nitric oxide charged catheters as a potential strategy for prevention of hospital acquired infections. PLoS One. 2017;12(4):e0174443
  127. 127. Wang X, Yao X, Zhu Z, Tang T, Dai K, Sadovskaya I, et al. Effect of berberine on Staphylococcus epidermidis biofilm formation. International Journal of Antimicrobial Agents. 2009;34(1):60-66
  128. 128. Magesh H, Kumar A, Alam A, Priyam SU, Sumantran VN, et al. Identification of natural compounds which inhibit biofilm formation in clinical isolates of Klebsiella pneumoniae. Indian Journal of Experimental Biology. 2013;51(9):764-772
  129. 129. Zhao L-X, Li D-D, Hu D-D, Hu G-H, Yan L, Wang Y, et al. Effect of tetrandrine against Candida albicans biofilms. PLoS One. 2013;8(11):e79671
  130. 130. Dwivedi D, Singh V. Effects of the natural compounds embelin and piperine on the biofilm-producing property of Streptococcus mutans. Journal of Traditional and Complementary Medicine. 2015;6(1):57-61
  131. 131. Rukayadi Y, Kim KH, Hwang JK. In vitro anti-biofilm activity of macelignan isolated from Myristica fragrans Houtt. against oral primary colonizer bacteria. Phytotherapy Research. 2008;22(3):308-312
  132. 132. Katsura H, Tsukiyama RI, Suzuki A, Kobayashi M. In vitro antimicrobial activities of bakuchiol against oral microorganisms. Antimicrobial Agents and Chemotherapy. 2001;45(11):3009-3013
  133. 133. Rukayadi Y, Hwang JK. In vitro activity of xanthorrhizol against Streptococcus mutans biofilms. Letters in Applied Microbiology. 2006;42(4):400-404
  134. 134. Sá NC, Cavalcante TTA, Araújo AX, Santos HS, Albuquerque MRJR, Bandeira PN, et al. Antimicrobial and antibiofilm action of Casbane Diterpene from Croton nepetaefolius against oral bacteria. Archives of Oral Biology. 2012;57(5):550-555
  135. 135. Santos V, Nardi R, Dias-Souza M. Bacteriocins as antimicrobial and antibiofilm agents. In: Current Developments in Biotechnology and Bioengineering: Human and Animal Health Applications. 2017. pp. 403-436
  136. 136. Hillman JD. Genetically modified Streptococcus mutans for the prevention of dental caries. Antonie Van Leeuwenhoek. 2002;82(1-4):361-366
  137. 137. Turner SR, Love RM, Lyons KM. An in-vitro investigation of the antibacterial effect of nisin in root canals and canal wall radicular dentine. International Endodontic Journal. 2004;37(10):664-671
  138. 138. Gonzalez-Toledo SY, Dominguez-Dominguez J, Garcia-Almendarez BE, Prado-Barragan LA, Regalado-Gonzalez C. Optimization of nisin production by Lactococcus lactis UQ2 using supplemented whey as alternative culture medium. Journal of Food Science. 2010;75(6):M347-M353
  139. 139. Gerke C, Kraft A, Sussmuth R, Schweitzer O, Gotz F. Characterization of the N-acetylglucosaminyltransferase activity involved in the biosynthesis of the Staphylococcus epidermidis polysaccharide intercellular adhesin. The Journal of Biological Chemistry. 1998;273(29):18586-18593
  140. 140. Zoll S, Schlag M, Shkumatov AV, Rautenberg M, Svergun DI, Gotz F, et al. Ligand-binding properties and conformational dynamics of autolysin repeat domains in staphylococcal cell wall recognition. Journal of Bacteriology. 2012;194(15):3789-3802
  141. 141. Chopra L, Singh G, Choudhary V, Sahoo DK. Sonorensin: An antimicrobial peptide, belonging to the heterocycloanthracin subfamily of bacteriocins, from a new marine isolate, Bacillus sonorensis MT93. Applied and Environmental Microbiology. 2014;80(10):2981-2990
  142. 142. Sandiford S, Upton M. Identification, characterization, and recombinant expression of epidermicin NI01, a novel unmodified bacteriocin produced by Staphylococcus epidermidis that displays potent activity against Staphylococci. Antimicrobial Agents and Chemotherapy. 2012;56(3):1539-1547
  143. 143. Arguelles Arias A, Ongena M, Devreese B, Terrak M, Joris B, Fickers P. Characterization of amylolysin, a novel lantibiotic from Bacillus amyloliquefaciens GA1. PLoS One. 2013;8(12):e83037
  144. 144. Zhang C, Occi J, Masurekar P, Barrett JF, Zink DL, Smith S, et al. Isolation, structure, and antibacterial activity of philipimycin, a thiazolyl peptide discovered from Actinoplanes philippinensis MA7347. Journal of the American Chemical Society. 2008;130(36):12102-12110
  145. 145. Gu Y, Xu Y, Xu J, Yu X, Huang X, Liu G, et al. Identification of novel bacteriophage vB_EcoP-EG1 with lytic activity against planktonic and biofilm forms of uropathogenic Escherichia coli. Applied Microbiology and Biotechnology. 2019;103(1):315-326
  146. 146. Pires DP, Melo L, Vilas Boas D, Sillankorva S, Azeredo J. Phage therapy as an alternative or complementary strategy to prevent and control biofilm-related infections. Current Opinion in Microbiology. 2017;39:48-56
  147. 147. Colavecchio A, Goodridge LD. Phage therapy approaches to reducing pathogen persistence and transmission in animal production environments: Opportunities and challenges. Microbiology Spectrum. 2017;5(3):1-14
  148. 148. Villarroel J, Larsen MV, Kilstrup M, Nielsen M. Metagenomic analysis of therapeutic PYO phage cocktails from 1997 to 2014. Viruses. 2017;9(11): pii: E328; 1-22
  149. 149. McCallin S, Alam Sarker S, Barretto C, Sultana S, Berger B, Huq S, et al. Safety analysis of a Russian phage cocktail: From metagenomic analysis to oral application in healthy human subjects. Virology. 2013;443(2):187-196
  150. 150. Pallavali RR, Degati VL, Lomada D, Reddy MC, Durbaka VRP. Isolation and in vitro evaluation of bacteriophages against MDR-bacterial isolates from septic wound infections. PLoS One. 2017;12(7):e0179245
  151. 151. Lin DM, Koskella B, Lin HC. Phage therapy: An alternative to antibiotics in the age of multi-drug resistance. World Journal of Gastrointestinal Pharmacology and Therapeutics. 2017;8(3):162-173
  152. 152. Twort FW. An investigation on the nature of ultra-microscopic viruses. The Lancet. 1915;186(4814):1241-1243
  153. 153. Schofield DA, Wray DJ, Molineux IJ. Isolation and development of bioluminescent reporter phages for bacterial dysentery. European Journal of Clinical Microbiology & Infectious Diseases. 2015;34(2):395-403
  154. 154. Abd, El-Aziz AM, Elgaml A, Ali YM. Bacteriophage therapy increases complement-mediated lysis of bacteria and enhances bacterial clearance after acute lung infection with multidrug-resistant Pseudomonas aeruginosa. The Journal of Infectious Diseases. 2019;219(9):1439-1447
  155. 155. Orlova ZN, Garnova NA. Use of tablet-form polyvalent bacteriophage with acid resistant coating in the treatment of dysentery in children. Voprosy Okhrany Materinstva i Detstva. 1970;15(3):25-29
  156. 156. Vinner GK, Rezaie-Yazdi Z, Leppanen M. Microencapsulation of salmonella-specific bacteriophage Felix O1 using spray-drying in a pH-responsive formulation and direct compression tableting of powders into a solid oral dosage form. 2019;12(1): pii: E43; 1-14
  157. 157. Endersen L, O’Mahony J, Hill C, Ross RP, McAuliffe O, Coffey A. Phage therapy in the food industry. Annual Review of Food Science and Technology. 2014;5:327-349
  158. 158. Janez N, Loc-Carrillo C. Use of phages to control Campylobacter spp. Journal of Microbiological Methods. 2013;95(1):68-75
  159. 159. Svircev A, Roach D, Castle A. Framing the future with bacteriophages in agriculture. Viruses. 2018;10(5): pii: E218; 1-13
  160. 160. Doss J, Culbertson K, Hahn D, Camacho J, Barekzi N. A review of phage therapy against bacterial pathogens of aquatic and terrestrial organisms. Viruses. 2017;9(3): pii: E50; 1-10
  161. 161. Eyer L, Pantucek R, Ruzickova V, Doskar J. New perspectives of the phage therapy. Klinická Mikrobiologie a Infekc̆ní Lékar̆ství. 2007;13(6):231-235
  162. 162. Kutateladze M, Adamia R. Bacteriophages as potential new therapeutics to replace or supplement antibiotics. Trends in Biotechnology. 2010;28(12):591-595
  163. 163. Yacoby I, Bar H, Benhar I. Targeted drug-carrying bacteriophages as antibacterial nanomedicines. Antimicrobial Agents and Chemotherapy. 2007;51(6):2156-2163
  164. 164. Yacoby I, Shamis M, Bar H, Shabat D, Benhar I. Targeting antibacterial agents by using drug-carrying filamentous bacteriophages. Antimicrobial Agents and Chemotherapy. 2006;50(6):2087-2097
  165. 165. Bar H, Yacoby I, Benhar I. Killing cancer cells by targeted drug-carrying phage nanomedicines. BMC Biotechnology. 2008;8:37
  166. 166. Borysowski J, Weber-Dabrowska B, Gorski A. Bacteriophage endolysins as a novel class of antibacterial agents. Experimental Biology and Medicine (Maywood, NJ). 2006;231(4):366-377
  167. 167. Pohane AA, Jain V. Insights into the regulation of bacteriophage endolysin: Multiple means to the same end. Microbiology (Reading, England). 2015;161(12):2269-2276
  168. 168. Schmelcher M, Loessner MJ. Bacteriophage endolysins: Applications for food safety. Current Opinion in Biotechnology. 2016;37:76-87
  169. 169. Wright A, Hawkins CH, Anggard EE, Harper DR. A controlled clinical trial of a therapeutic bacteriophage preparation in chronic otitis due to antibiotic-resistant Pseudomonas aeruginosa; a preliminary report of efficacy. Clinical Otolaryngology. 2009;34(4):349-357
  170. 170. Kutateladze Á, Adamia R. Phage therapy experience at the Eliava Institute. Médecine et Maladies Infectieuses. 2008;38(8):426-430
  171. 171. Fish R, Kutter E, Wheat G, Blasdel B, Kutateladze M, Kuhl S. Bacteriophage treatment of intransigent diabetic toe ulcers: A case series. Journal of Wound Care. 2016;25(Sup7):S27-S33
  172. 172. Jun JW, Shin TH, Kim JH, Shin SP, Han JE, Heo GJ, et al. Bacteriophage therapy of a Vibrio parahaemolyticus infection caused by a multiple-antibiotic–resistant O3: K6 pandemic clinical strain. The Journal of Infectious Diseases. 2014;210(1):72-78
  173. 173. Markoishvili K, Tsitlanadze G, Katsarava R, Glenn J, Morris M Jr, Sulakvelidze A. A novel sustained-release matrix based on biodegradable poly (ester amide) s and impregnated with bacteriophages and an antibiotic shows promise in management of infected venous stasis ulcers and other poorly healing wounds. International Journal of Dermatology. 2002;41(7):453-458
  174. 174. Pouillot F, Chomton M, Blois H, Courroux C, Noelig J, Bidet P, et al. Efficacy of bacteriophage therapy in experimental sepsis and meningitis caused by a clone O25b: H4-ST131 Escherichia coli strain producing CTX-M-15. Antimicrobial Agents and Chemotherapy. 2012;56(7):3568-3575
  175. 175. Soothill J. Treatment of experimental infections of mice with bacteriophages. Journal of Medical Microbiology. 1992;37(4):258-261
  176. 176. Biswas B, Adhya S, Washart P, Paul B, Trostel AN, Powell B, et al. Bacteriophage therapy rescues mice bacteremic from a clinical isolate of vancomycin-resistant Enterococcus faecium. Infection and Immunity. 2002;70(1):204-210
  177. 177. Ramesh V, Fralick JA, Rolfe RD. Prevention of Clostridium difficile-induced ileocecitis with bacteriophage. Anaerobe. 1999;5(2):69-78
  178. 178. Chanishvili N. Phage therapy—History from Twort and d’Herelle through Soviet experience to current approaches. Advances in Virus Research. 2012;83:3-40
  179. 179. Gilmer DB, Schmitz JE, Euler CW, Fischetti VA. Novel bacteriophage lysin with broad lytic activity protects against mixed infection by Streptococcus pyogenes and methicillin-resistant Staphylococcus aureus. Antimicrobial Agents and Chemotherapy. 2013;57(6):2743-2750
  180. 180. Yang H, Wang D-B, Dong Q , Zhang Z, Cui Z, Deng J, et al. Existence of separate domains in lysin PlyG for recognizing Bacillus anthracis spores and vegetative cells. Antimicrobial Agents and Chemotherapy. 2012;56(10):5031-5039
  181. 181. Singh PK, Donovan DM, Kumar A. Intravitreal injection of the chimeric phage endolysin Ply187 protects mice from Staphylococcus aureus endophthalmitis. Antimicrobial Agents and Chemotherapy. 2014;58(8):4621-4629
  182. 182. Lood R, Winer BY, Pelzek AJ, Diez-Martinez R, Thandar M, Euler CW, et al. Novel phage lysin capable of killing the multidrug-resistant gram-negative bacterium Acinetobacter baumannii in a mouse bacteremia model. Antimicrobial Agents and Chemotherapy. 2015;59(4):1983-1991
  183. 183. Oliveira H, Vilas Boas D, Mesnage S, Kluskens LD, Lavigne R, Sillankorva S, et al. Structural and enzymatic characterization of ABgp46, a novel phage endolysin with broad anti-gram-negative bacterial activity. Frontiers in Microbiology. 2016;7:208

Written By

Mohammed El-Mowafy, Abdelaziz Elgaml and Mona Shaaban

Submitted: 24 July 2019 Reviewed: 05 November 2019 Published: 06 December 2019