Phage Therapy: A Potential Novel Therapeutic Treatment of MRSA Download PDF

Journal Name : SunText Review of Virology

DOI : 10.51737/2766-5003.2022.030

Article Type : Research Article

Authors : Gildea L, Ayariga JA, Abugri J, Robertson BK and Villafane R

Keywords : Bacteriophage therapy; MRSA; Antibiotic resistance; Virulence factors

Abstract

The emergence of multi-drug resistant bacterial strains, especially in the clinical setting has renewed interest in alternative therapeutic treatment methods. The utilization of prokaryotic viruses in phage therapy has demonstrated potential as a novel treatment method against multidrug resistant bacterial infections. As the post-antibiotic era quickly approaches the development and standardization of phage therapy is critically relevant to public health. This review serves to highlight the development of phage therapy against methicillin resistant Staphylococcus aureus (MRSA), an antibiotic resistant bacterial strain responsible for severe clinical infections.


Phage Therapy: History and Current Relevance

Phages were discovered independently in 1915 by the British microbiologist Felix Twort and in 1917 by French-Canadian microbiologist Felix d’Herelle. Responsible for the systematic investigation of the nature of bacteriophages, d’Herelle in 1921 first utilized phages for the treatment of dysentery in Paris, France [1]. This treatment resulted in the rapid recovery of patients and brought relevance to phage therapy as a clinical treatment method. Continued study and clinical use led to d’Herelle becoming the leading expert on phage therapy in this period. Throughout the early 20th century d’Herelle and other microbiologist isolated phages for the treatment of pathogenic bacteria such as Shigella dysenteriae, Salmonella typhi, Escherichia coli, Pasteurella multocida, Vibrio cholerae, Yersinia pestis, Pseudomonas aeruginosa, Neisseria meningitis and various strains of Streptococcus [2]. By 1931 d’Herelle had established phage therapy centers across the world, in the United States, France, and Soviet Georgia. While phage therapy showed promise, it was weighed down by a few major problems. These problems included host range, genetic variation, and the inability to consolidate the value of phage in prevention of infectious disease. These problems eventually led to the fall of phage therapy. In this time period, phage therapy was poorly incorporated into the field of medicine, lacking theories that could be integrated with other notions of conventional medicine [3]. The discovery of antibiotics as an efficacious treatment method against bacterial infections led to a decreased interest in the development of phage therapy. While phage therapy maintained some traction in the Eastern world, it was all but forgotten in the Western world. At the turn of the 21st century the field of medicine faced a new challenge. The overuse and eventual abuse of antibiotics over the span of the 20th century led to the prevalence of antibiotic-resistant bacterial strains [4-6]. The inability to treat these bacterial infections with standard antibiotics makes them a significant threat to public health. The continued prevalence of antibiotic-resistant bacterium has led to the need for new novel antimicrobial agents, renewing interest in phage therapy as a potential novel therapeutic treatment. When considering phage therapy in this modern era there are three major characteristics of phages that lead to their consideration as a potential treatment method: 1) Host specificity: Phage targets bacteria with high specificity. This characteristic ensures that phage treatment would only infect the target bacteria while natural micro biota is unaffected. 2) Genetic engineering: In the early stages of phage therapy genetic engineering was not an available option. With current advances in science, we are now able to engineer phages to express traits of potential value. 3) Phages are ideal candidates for co-therapy with antibiotics: Co-therapy involves the use of both antibiotics and phage therapy for the treatment of multidrug resistant bacteria. The advancement of science since the discovery of phages in the early 20th century has led to a greater understanding of phages and an increased ability to utilize them for the benefit of public health [7-11]. As we now enter the 21st century, the prevalence of antimicrobial resistance (AMR) in bacteria has increased on account of the massive and sometimes inappropriate use of antibiotics. Antibiotic-resistant bacterial infections account for over 2.8 million infections and 35,000 deaths annually in the United States alone [5]. The continued occurrence and prevalence of antibiotic-resistant bacterial strains is considered a serious threat to global health and the economy [12-18]. The Institutes of Medicine estimates that the annual cost of antibiotic-resistant bacterial infections in the United States is approximately 4 to 5 billion USD [19,20]. Increased prevalence of antibiotic-resistant bacterial strains as well as a decrease in antibiotic development is a critical issue in the field of medicine [21,22]. Estimates from the United Kingdom project that antibiotic-resistant bacteria could result in losses to approximately 100 trillion USD worldwide by 2050, with a potential death toll up to 10 million per year [19]. Considering this, development of novel treatment methods for antibiotic-resistant bacterial infections is crucial for the preservation of international health and economy. In recent research into antibiotic alternatives, bacteriophages and their components have gained relevance as potential novel therapeutic treatment methods [7-11]. Phage therapy utilizes phage particles that specifically infect and lyse bacterial cells. A major benefit of phage therapy is host specificity; phages only infect prokaryotic cells and cannot infect eukaryotic cells. The development of new alternative treatment methods for bacterial infections are subject to technical and regulatory challenges. Challenges of alternative treatment methods such as phage therapy include activity spectrum, pharmacokinetics, immune response, manufacturing logistics, regulation, quality control, and market acceptance [23]. While these alternative treatments may not be able to replace antibiotics completely, it has been suggested that use in unison with antibiotics could be a potentially viable method for treatment of multidrug resistant bacterial strains [24-26]. This review will focus on the development of phage therapy specifically against methicillin-resistant Staphylococcus aureus (MRSA), a serious threat to public health (Table 1).

Table 1: Distinguishing features between CA-MRSA and HA-MRSA.

Feature

CA-MRSA

HA-MRSA

At-Risk Population

Young, healthy individuals with no exposure to healthcare facilities

Individuals with previous contact to healthcare facilities. (e.g., Hospitals, Nursing Homes)

Risk Factors

Frequent skin-to-skin contact, use of intravenous drugs, HIV, crowded or unsanitary living conditions

Long hospital stays, frequent antibiotic usage, intravenous tubing, compromised immune system, invasive procedures, devices, and surgery

Infection Type

Mild to moderate skin and soft tissue infections

Severe, invasive disease in patients or individuals in frequent contact with healthcare facilities

Infection Locations

Skin and soft tissues, lung

Bloodstream, lung, surgical site, prosthetic implant

Antibiotic Resistance Pattern

Susceptible to many antibiotics except Beta-lactams

Multi-resistant to several antibiotics


Methicillin-resistant S. aureus (MRSA) is one of the most common and clinically relevant examples of an antibiotic resistant bacteria [5,27]. MRSA is the result of a S. aureus infection that has developed resistance to antibiotics commonly used for the treatment of these infections. MRSA is the result of misappropriate use of antibiotics over the span of the 20th century. MRSA is categorized into two general types, healthcare associated MRSA (HA-MRSA) and community associated MRSA (CA-MRSA). Most MRSA cases fall under HA-MRSA infections associated with invasive procedures or devices such as surgeries, intravenous tubing, or artificial joints. Contamination of these devices can lead to deadly MRSA infections and outbreaks in healthcare facilities. In general, those exposed to MRSA in the healthcare setting are typically more susceptible to adverse outcomes as a result of acquiring this infection due to the compromised state of their health [5,27]. CA-MRSA is not as a common and occurs in healthy communities. CA-MRSA infections are commonly associated with skin-to-skin contact among individuals. Conditions that can place individuals at risk for acquiring these infections are group sports that induce skin-to-skin contact, working in child-care, intravenous drug use, or crowded living conditions. The ability of S. aureus to develop resistance paired with the inappropriate use of antibiotics of has created a potentially deadly pathogen (Figure 1).

Figure 1: A chronological map outlining several important aspects of S. aureus treatment, evolution, and impact.

Figure 2: Diagrammatic illustration of the mechanism of inhibition of antibiotic resistance of MRSA.

Figure 1 outlines several notable timepoints in the history of S. aureus of the mid-20th to early 21st centuries. In 1940, the discovery of penicillin as a miracle drug offered an unlimited hope to bacterial control, however, within the space of two years, S. aureus developed resistance to penicillin [7,28-30]. By 1960 over 80% of S. aureus strains had developed resistance to penicillin [28,30]. Methicillin was introduced in 1961 as an alternative treatment of S. aureus. Only a year later, S. aureus developed resistance to this antibiotic as well [29]. The first outbreak of MRSA was recorded in 1968, this was followed with the second and third outbreaks between 1970 and 1980 [29]. By 1980 MRSA had spread worldwide. In 1990, vancomycin became the drug of choice against MRSA, however there was an observed rise in intermediate vancomycin resistance, leading to the occurrence of complete vancomycin resistance in 2002 [29,31]. Since 2002, MRSA prevalence coupled with a decrease in antibiotic development created a serious risk for public health. Several researchers have delved into antibiotics against MRSA; however, none have reached clinical applicability [32,33]. In 2009, a group of researchers set out to examine the safety of bacteriophage-based formulations for the treatment of wounds caused by S. aureus [34]. In a phase I clinical trial they reported that there were no safety concerns with the use of bacteriophage treatment, nonetheless, they encouraged a vigorous test for efficacy of the phage preparations in a phase II trial [34]. As we continue to discuss MRSA and the significant hazard it possesses to public health, it is essential that we discuss and explain what makes this pathogenic bacterium so difficult to treat on a molecular level.


MRSA and Antibiotic Resistance

As previously mentioned, resistance to standard antibiotic treatment is a major obstacle in the treatment of MRSA infections, but it is important to understand the genetic factors that facilitate this resistance. MRSA infections are resistant to beta-lactam antibiotics such as penicillin and semi-synthetic antibiotics such as methicillin that were the standard treatment of S. aureus prior to prevalence of MRSA [30]. To understand the virulence factors that allow for MRSA’s antibiotic resistance it is important to understand the evolution of S. aureus infections. As figure 1 outlines, S. aureus has gradually developed resistance to antibiotics starting with penicillin in the form of penicillin resistant S. aureus (PRSA) which was first reported in 1942 [7,29]. The virulence factor present in PRSA was determined to be the gene blaZ [7,35]. This gene inhibits the binding of penicillin binding proteins (PBPs) that function to disrupt peptidoglycan cross linking during cell wall synthesis [28] (Figure 2).

As shown in Figure 2, inhibition is achieved through the production of beta-lactamase enzymes and structural alteration of the PBP receptor [28]. The development of resistance resulted in methicillin, a semisynthetic derivative of penicillin, becoming the new standard antibiotic treatment. S. aureus and PRSA eventually developed new resistance mechanisms against methicillin resulting in MRSA [7,29]. Methicillin resistance is the result of the development of a mobile genetic element called the staphylococcal cassette chromosome mec (SCCmec) [29]. The SCCmec genetic element contains the gene mecA along with several other functional genetic elements. The gene mecA is fundamentally responsible for inhibition of methicillin binding to the PBP 2a receptor resulting in resistance [29,36]. Methicillin utilizes the PBP 2a receptor for the disruption of peptidoglycan cross linking during cell wall synthesis. This structural change influenced by mecA results in methicillin resistance. There exists an allosteric control of S. aureus penicillin binding protein 2a that allows for methicillin resistance. In ?-lactam susceptible S. aureus, the trans peptidase activity of their PBPs is absent. Consequently ?-lactam permanently acrylates the active site serine [37]. However, MRSA PBP 2a is impervious to ?-lactam acylation, hence the dd-transpeptidation reaction is carried out, thus producing the cell wall of the bacteria. As shown in Figure 3, the PBP 2a enzyme, a dimeric molecule is shown bound to a peptidoglycan moiety during the bacterial cell wall synthesis process (Figure 3).

The PBP 2a structure (3ZG5) was extracted from the RCSB website (https://www.rcsb.org/structure/3ZG5), with the PBD ID, 3ZG535. Molecule - ligand interactions were analyzed using Biovia Discovery Studio 2021 Client (BIOVIA Discovery Studio Visualizer- https://discover.3ds.com/discovery-studio-visualizer). As shown in figure 2A, the dimeric molecule binds to peptidoglycan via a minor cleave found in both monomers (chain A, and chain B). PBP 2a establishes hydrogen bonds with peptidoglycan moiety at the following amino acids in the binding site; ARG151 (bond distance, 2.33Å), THR165 (bond distance, 3.36Å), THR216 (bond distance, 2.75Å), SER240 (bond distance, 2.76Å), ARG241 (bond distance, 2.85Å), TYR373 (bond distance, 2.52Å), GLY166 (bond distance, 3.73Å), HIS293 (bond distance, 3.84Å). It also interacts with the peptidoglycan molecule via alkyl hydrophobic interactions in PRO258 (bond distance, 5.31Å) and MET372 (bond distance, 4.00Å). The ability of MRSA to acquire mobile genetic elements carrying a variety of virulence factors has led to significant variation amongst MRSA strains [29]. Virulence factors that have been highlighted in literature include Panton-Valentine leucocidin (PVL), PSM cytolysins, and toxic shock syndrome toxin-1 [29,39]. These exotoxins are responsible for MRSA’s increased virulence and exceptional ability to evade the immune system [29]. The rapid development of resistance as well as the multifaceted nature of this resistance poses a significant challenge in the development of novel antibacterial agents. While these genetic components serve to develop resistance against several antibiotics, some genetic factors induce physical states that alter the effectiveness of antibiotics such as biofilm development [40]. The formation of biofilms by Staphylococcus spp. is a crucial adaptation for bacterial survival, thus protecting it from harsh environmental factors, antibiotics and even the bacterial host’s immunity [40]. In Staphylococcus epidermidis, discovery of poly-N-acetylglucosamine (PNAG) and polysaccharide intercellular adhesin (PIA) was the first factor shown to mediate biofilm formation [41,42]. The discovery of multiple biofilm formation factors in S. aureus such as the LPXTG-cell wall-anchored biofilm-associated protein (BAP), fibronectin binding protein (FnBP), cell wall anchored clumping factor A (ClfA), cell wall-anchored clumping factor B (ClfB), S. aureus surface protein G (SasG), S. aureus surface protein C (SasC), S. aureus protein A (Spa) as well as other genes such as ebpS, fib, and icaA elucidated the mechanisms of action involved in antibiotic resistance employed by S. aureus via its biofilm formation [42]. Some cytoplasmic proteins have also been implicated in biofilm phenotypes [43-45].

Figure 3: The dimeric PBP 2a binds to peptidoglycan moiety from MRSA.

Table 2: A brief comparative description of antibiotic and potential phage treatments against S. aureus. Chart outlines antibacterial mechanisms as well as bacterial resistance mechanisms.

Antibiotic

Protein

Antimicrobial Mechanism

Bacterial Resistance Mechanism

Resistance Gene

Penicillin

Beta-lactamase

Binding to penicillin binding proteins (PBPs) disrupts peptidoglycan cross linking during cell wall synthesis resulting in lysis.

Production of Beta-lactamase enzymes and alteration of PBP [28]

blaZ [28,35]

Methicillin

Beta-lactamase 2a

Binding to PBP 2a disrupts peptidoglycan cross linking during cell wall synthesis resulting in lysis.

Structural change of PBP 2a [36]

mecA [28]

Vancomycin

(glycopeptides)

D-ala: D-lac ligase

D-ala: D-ser ligase

Interaction with uncross-linked peptidoglycan pentapeptides results in inhibition of the extracellular steps of peptidoglycan synthesis.

Alteration of structure of cell wall peptidoglycan precursors [31]

vanA

vanB [28,31]

 

Phage

Protein

Antimicrobial Mechanism

Bacterial Resistance

S. aureus Phages

Trsa205 [70]

Trsa207 [70]

Trsa220 [70]

Trsa222 [70]

SA003 [71]

MR003 [71]

LS2a [106]

Regional Phages [88,103]

Tailspike Proteins

Phage tailspike proteins specifically target receptors on the membrane to initiate penetration, replication, synthesis, assembly, and release, resulting in lysis of the bacterial host [47-49].

The evolutionary rate of bacteria to develop resistance to phage treatment is significantly slower than bacterial development of antibiotic resistance [7].


Identification of Phages against MRSA

Phages are characterized by a narrow host range and may infect only one species or strain of bacteria within a species. The development of phage therapy for specific bacterial strains requires the identification, isolation, and characterization of phages that exhibit lytic lifestyles in the desired bacterial target. Lytic phages offer the greatest therapeutic potential due to their consistent lethal effects on their host [46]. The lytic lifestyle is comprised of five stages: attachment, penetration, biosynthesis, maturation, and lysis. In the attachment stage, phages utilize their tailspike proteins to interact with specific bacterial surface receptors of the membrane. This interaction has been observed at the molecular level in a variety of phage families [47-49]. As previously mentioned, phages are characterized by a narrow host range and may infect only one species or strain of bacteria within a species [50]. This specificity is unique and can be exploited for targeted treatment of bacterial infections in phage therapy and identification of bacterial pathogens in phage typing [51,52]. Following attachment to the host cell membrane, the phage utilizes its tail machinery to penetrate the cell membrane and inject its viral genome [53,54]. The biosynthesis step of this mechanism is carried out through synthesis of virus encoded endonucleases to degrade the bacterial chromosome. The virus then utilizes the functions of the host cell to replicate, transcribe, and translate viral components for the assembly of a progeny [55]. Assembly of the newly synthesized virions, termed maturation, is followed by the disruption of the host cell membrane by phage proteins holin or lysozyme [56]. This disruption leads to the lysis of the host cell and the release of the progeny to infect other bacterial cells. According to literature all known phages associated with the Staphylococci family of bacteria belong to the order of Caudovirales and are primarily members of the families Siphoviridae and Myoviridae [57-60]. The use of phage typing for the identification of S. aureus infections in the clinical setting served to develop a library of phages specific to this bacterial genus. This library is integral in the screening of phages for lytic activity against MRSA. As the prevalence of MRSA increases, the ability to identify S. aureus phages that carry out lytic lifestyles in MRSA is a vital step in the development of viable therapeutic treatments. Isolation of phages from the order Caudovirales followed by characterization and in vitro testing is a viable method for identification of S. aureus phages with lytic lifestyles within MRSA [61-66]. Phages are known to be abundant in any ecosystem in which their bacterial host is present [64]. Literature has been able to utilize samples, primarily from healthcare facility sewage, for the isolation of S. aureus phages [61-65]. Characterizations of these isolates through double-layer plaque assay (DLA) and electron-microscopy has resulted in the identification of S. aureus phages belonging to the Siphoviridae and Myoviridae families [61-64]. Phages of the order Caudovirales are classified structurally into three families of tailed bacterial viruses: Myoviridae (long contractile tails), Siphoviridae (long non-contractile tails), and Podoviridae (short non-contractile tails) [63]. All three families of Caudovirales feature non-enveloped protein shell heads containing a single linear dsDNA molecule. The dsDNA genomes of these phages encode from 27-600 genes clustered according to function arranged in large operons. Caudovirales are found in over 140 prokaryotic genera representing most branches of the bacterial phylogenetic tree. With a wide variety of host ranges some members of this order can infect members of multiple genera of bacteria while others show high specificity [63]. A major obstacle in the development of phage therapy is the host range of phages. Infection specificity of phages can often lead to difficulties in the development of efficacious phage therapy methods. The host range of S. aureus phages against clinically isolated MRSA strains can be determine through in vitro assays. Against isolates of clinical and community related MRSA infections, phage host ranges have shown wide variation as naturally expected. We contribute this wide variation to the high specificity between phages and their target host. Literature has been able to identify a variety of phages with host ranges suitable for phage therapy against MRSA [67-72]. Phages that are selected for treatment of MRSA infections should exhibit a broad host range against clinically relevant strains. Literature has outlined several polyvalent phages that could be utilized for phage therapy [46]. A phage that has exhibited a broad host range against MRSA is the phage MR003 [71]. This phage, a member of Caudovirales family, has been observed to infect 97% of clinical and community MRSA strains [71]. This host range is significantly higher than other S. aureus phages that typically infect anywhere from 20% to 73% of MRSA strains [70-72]. The host specificity of phage MR003 is hypothesized to be a result of the genomic structure of the tailspike and baseplate structures of the virus. Comparative genomic studies of MR003 to common S. aureus phage SA012 revealed that these two phages share homology in ORF117 and ORF119, responsible for receptor binding to host cells. It was determined that differences in the tailspike and baseplate structures seem to be the key contributing factor to the broad host specificity in MR003 [73]. Another relevant phage is phage 812. In vitro studies have shown this phages ability to kill 95% of 782 clinical S. aureus isolates [74]. Phage 812 is closely related to phage K that is known to demonstrate a large host range against MRSA. Phage K has also been shown to be effective against MRSA strains that are vancomycin resistant and teicoplanin resistant [46]. In vitro study demonstrated that 39 out of 53 clinically isolated strains where sensitive to phage K and that insensitive strains could be treated with variants of phage K [75]. Genomic studies of phages that are potential candidates for phage therapy against MRSA infections could be useful in identifying factors that influence host range [71-73]. A method utilized to increase the host range of phage treatments against MRSA is phage cocktails. Phage cocktails address the challenge of limited host ranges through the incorporation of multiple phages with varying host ranges in solution. This method has been shown to increase the infectivity of phages against MRSA [70]. An experimental phage cocktail of four S. aureus phages that infected 37.5%, 26.7%, 21.4%, and 19.6% of clinical MRSA isolates respectively resulted in a cocktail that had the ability to infect 66% of clinical MRSA isolates [70]. Phage cocktails allow for the lysing of MRSA bacterial strains without the host range limitations associated with individual phage treatments. While phage cocktails provide a greater ease of use a potential downfall of this method is the greater complexity in manufacturing and possible clinical outcomes [76]. While individual phage therapy only requires the isolation of one specific phage, phage cocktails require the isolation and purification of multiple phages which in turn increases the complexity of manufacturing. 


Development of MRSA Phages as Therapeutic Agents

Biological considerations

Phage therapy, first used almost a century ago, is driven by the continued occurrence and prevalence of antibiotic resistant bacterial strains. While the discovery of antibiotics negated the need for new antimicrobial agents in the 20th century, antibiotic resistance in the 21st century has renewed the need for new antimicrobial agents. The rise of phage therapy as a potential novel therapeutic method is facilitated by our improved understanding of phage biology, genetics, immunology, and pharmacology. Aspects of phage therapy that once hindered its efficacy have now been standardized to improve treatment success. Regulatory requirements of phage therapy call for strictly lytic phages, confirmed antimicrobial activity against the target pathogen, and the removal of contaminating bacterial debris and endotoxins [77]. Identification of the bacterial host cell receptor for any therapeutic phage is also key in the long-term success of phage therapy. Identification of these receptors can provide insight into phage resistance, evolutionary trade-offs, and use of co-therapies that are less likely to generate phage resistant hosts. Phages that feature lytic lifestyles are ideal for the success of phage therapy. The use of temperate or lysogenic phages is highly inadvisable in phage therapy as their ability to lysogenize cells is hindered by the rise of homoimmunity in a bacterial population and the possibility of lysogenic conversion [78,79]. Lysogenic conversion can lead to bacterial populations gaining new, often pathogenic genetic traits, such as phage-encoded toxins or antimicrobial resistant determinants [79]. Despite these drawbacks and potential hazards, temperate or lysogenic phages have shown potential to be utilized through genetic manipulation of their life cycle [46]. Research has demonstrated that two distinct mutations, vir and clear plaque, can essentially change temperate phages into obligately lytic phages [46]. Both mutations effect the repressor protein of the phage, inhibiting its ability to become a prophage or carry out lysogenic conversion. A vir mutant has already been successfully utilized in an animal study, showing promise for this method [46].

While lytic phages are considered the standard for phage therapy, there are still some concerns about their abilities. Scientific understanding of phage has been greatly advanced since their discovery a century ago. However, our knowledge of phages is still limited. The genomes of lytic phages can contain greater than 50% hypothetical genes with no known function as well as encode auxiliary proteins that alter bacterial physiology in ways that are not fully known [80]. The number of genes and auxiliary proteins that we are currently unaware of makes abortive infections a major concern. Abortive infection is a method of bacterial defense in which the bacterial cell upon infection kills itself to ensure the replication of a phage is stopped. This mechanism could possibly lead to the bacterial host acting as a reservoir inside the human body for phage DNA with unknown function. This concern is also shared with mutant phages such as vir and clear plaque, especially considering that temperate phages typically carry a wide range of virulence factors [46]. Continued research of phage genetics is key in ensuring the safety of phage therapy.

Comparison to Antibiotics

Phages and antibiotics both serve as antibacterial agents functioning to lyse or inhibit the persistence of bacterial infections. While both agents have similar function, they feature several key differences that determine their appropriateness for situational usage. The use of antibiotics has been observed to have adverse health effects in some situations [81]. Adverse health effect of antibiotics includes instances of anaphylaxis, nephrotoxicity, cardiotoxicity, hepatotoxicity, neurotoxicity, and several gastrointestinal and hematological complications [81]. The most common adverse effect of antibiotic treatment is allergic reaction, which is prominent in children. These allergic reactions are most commonly the product of high tissue concentrations [82-84]. The safety of phage therapy has not been as extensively studied, especially in western medicine. However, new studies have deemed phage therapy practices such as oral administration as safe [82-88]. In terms of oral administration, the translocation of phage across the intestinal epithelium into the blood has been suggested as beneficial to the host [89]. The benefit of this translocation is the downregulation of immune response to indigenous gut microbiota antigens through the inhibition of interleukin-2, tumor necrosis factor, and interferon gamma production [89]. This down regulation in addition to phage host specificity protects the natural gut microbiota. The protection of natural gut microbes is a typical criticism of antibiotics. The immunological response to phage therapy may be beneficial in healthy patients however literature disputes the safety of treatment in patients with compromised immune systems [90-92]. Immunological response is especially significant in the context of MRSA infections that are prominent in patients who are immunocompromised. Patient to patient variation in the study of phage therapy has been an area of concern. While transduction may be beneficial to natural gut microbes, there is concern that this characteristic could also be related to disruption of normal intestinal barrier function. This disruption could potentially lead to disorders such as Crohn’s disease, inflammatory bowel disease (IBS), and type 1 diabetes [93]. Literature has determined that there is variation in inflammatory response to phage therapy based on the site of infection [94]. The study of phage therapy is relatively new and there are many characteristics such as immunological response and physiological response that require further study to comprehensively assess the safety of phage therapy. Host specificity is a defining characteristic of phage therapy. The broad use of antibiotics has been documented for its adverse effects on the human gut microbiome that sometimes lead to diarrhea and C. difficile infection [95]. Other potential outcomes of antibiotic perturbations in the gut microbiome include asthma, obesity, and diabetes [96-101]. Phage therapy is highly specific to bacterial species and strain, resulting in less irritation of the natural gut microbes while still effectively reducing presence of pathogens [99,100]. As discussed in the host range section of this review, the specificity of phages can sometimes lead to inability to treat an infection colonized by multiple bacterial species. A common clinical example of this scenario is burn victims who typically suffer infections colonized by more than one singular bacterial strain [101]. The development of phage cocktails that are effective against a range of bacteria present in an infection can increase the host range of treatment, which in turn results in a more effective treatment of the infection. It is important note that the success of phage cocktail treatment is dependent on the ability to identify the pathogens present. While phage cocktails address complex infections and the limitations of host specificity, they result in major logistical challenges [76]. Phage cocktails present limitations in development, large-scale production, and distribution; a distinct advantage of broad-spectrum antibiotics. Another specific advantage of phage therapy in comparison to antibiotics is the rate of resistance development by target species. Phages, as naturally occurring organisms, actively adapt to ensure persistence and survival. However, even with these adaptations, bacteria can often still develop resistance against phages. For this reason, specifically, phage cocktail therapies have been of elevated interest as they make the emergence of a resistant bacterial cell substantially less likely [102]. In addition to the use of cocktail therapies, it is also important to understand that phages, unlike antibiotics, require little to no development and are readily available from the environment. Considering this even in cases of bacterial resistance, there are a multitude of phage species that our target pathogen is likely not resistant to. An interesting characteristic of phage therapy is the relationship between geographic location and phages used for treatment. Studies have shown that phages show high specificity to bacterial targets from their indigenous region [88,103]. These studies utilized Russian E. coli phage cocktails for the treatment of microbiologically determined E. coli diarrhea in Bangladesh [88]. The treatment resulted in no improvement of clinical outcome. Results suggested that phage cocktails are better adapted to local bacteria populations, and that bacterial host range can be restricted both spatially and temporally [104]. A suggested solution to this challenge is the development of phage cocktails with regional specificity for the clinical setting [105]. In the context of MRSA infections, as well as other antibiotic resistant bacterial strains, this means that the phages that can be used to target these bacteria are likely found in the same environment [106]. While this high specificity provides challenges in production that are not common with broad-spectrum antibiotics it does have some benefits. Regions that have limited access to antibiotics would greatly benefit from the ability to isolate phages that could be utilized for specific phage therapy of regionally prevalent pathogens. Utilization of phage therapy in these regions would also positively impact the economic burden that the cost of antibiotic treatment entails. Antibiotics have been a cornerstone of clinical treatment for over a century, but the increased prevalence of antibiotic resistant bacterial strains has required the development of new novel therapeutic treatments. The limited adverse effect, target specificity, and abundance of phages in the natural world make phage therapy a potentially viable therapeutic treatment (Table 2).


Clinical Challenges of Phage Therapy against MRSA

The lack of validated and adequately controlled clinical trials is a current challenge of progressing phage therapy into standard clinical practice [108]. The pharmacological characteristics of phages hinder their standardization in clinical trials. A major pharmacological concern is the self-replicating nature of phages, unlike conventional drug treatments, phage therapy requires awareness of various novel kinetic phenomena [109]. Determining dosage is particularly challenging since phages have the potential to exponentially increase upon infection of the target bacteria. Experimental design of clinical trials utilizing phages requires standardization and guidance using tailored pharmacokinetic models for specific systems. The establishment of these models as standard practice would greatly advance the use of phage therapy in clinical trials [109]. Another challenge in the clinical use of phages for the treatment of bacterial infections is the delivery of phage virions to the location of the infection. Phages require direct contact with the target bacteria to carry out infection and lysis. Broad distribution of phage in the body cannot effectively treat the target infection. Literature has exhaustively examined methods of delivery in animal models, revealing that administration of phages into the intramuscular, subcutaneous, or intraperitoneal have shown significant influence on the success of phage therapy [107,110,111]. Intraperitoneal injection of phage MR11, an S. aureus phage, demonstrated the ability to eradicate MRSA infections in mice models [111]. Animal trials have demonstrated the abilities of phage therapy as a novel therapeutic against MRSA as well as worked towards standardization of dosages for adequate treatment. Dose-response studies in white rabbits have demonstrated the effectiveness of phage therapy against S. aureus via subcutaneous injection. This study concluded that high concentrations of the phage L2Sa, a S. aureus phage, was shown to prevent abscesses caused by infection [107]. While phage immunotherapy has shown promise, combination therapy or phage cocktails also offer a broad range of activity against bacterial infections. Phage cocktails as previously described in this review consist of a combination of several phages with various host ranges. This combination addresses the limitations of monotherapies host range and reduces the potential development of phage resistance in bacteria. While phage cocktails feature a broader host range, it has been shown that they greatly increase the challenge of assessing inflammatory response, potential gene transfer, and development of multi-phage resistance [112]. Further study and standardization of phage cocktail therapy is required to fully determine their effectiveness as well as efficacy. While there are still many questions regarding the effectiveness of phage therapy treatments, groups such as the Center for Innovative Phage Applications and Therapeutics (IPATH) have taken major steps forward in furthering the development of phage-based therapeutics. IPATH utilizes phage therapy to treat patients with life-threatening multi-drug resistant infections through the Food and Drug Administrations (FDA) compassionate use program. Additionally, IPATH serves to rigorously evaluate phage therapy in clinical trials with the eventual goal of combating the global antimicrobial resistance crisis. Hopefully continued study of phages and their clinical efficacy can continue to provide meaningful insight into the progress of phage-based therapy. Utilizing clinical studies from groups such as IPATH will be crucial in the further development of clinical trials for phage therapeutics. 


Human Clinical Trials

Human clinical trials for phage therapy against MRSA are limited due to the challenges previously mentioned. Standardization of clinical trials requires preliminary studies to determine adequate dosage, delivery, and host response. The use of animal models has been largely beneficial to the progression of standardized phage therapy methods [107,111,112]. As previously mentioned and important to note, while the western worlds use of phages as therapeutics is limited, the eastern world has considered phage therapy a viable option [113]. Not specific to MRSA, phage treatments against drug resistant bacterial species have shown effectiveness with a reported ~85% success rate [114]. The select phage therapy clinical trials that have been conducted show promise for the use of phages against MRSA infections and will be briefly discussed throughout this section. Referenced throughout literature as one of the pioneering clinical trials against MRSA, Rhoads et al in 2009 focused on the treatment of venous leg ulcers in humans [34]. This trial treated ulcers with bacteriophages targeted against Pseudomonas aeruginosa, S. aureus, and Escherichia coli. Results of this phase I trial concluded that there were no adverse events attributed to the phage therapy and that between test and control groups there was no significant difference (p>0.05) in the frequency of adverse events, rate of healing, or frequency of healing [34]. Phase I was successful in demonstrating the safety of phage therapy in humans [34]. While Rhoads et al showed promise, phase II trials of phage therapy must be carried out to determine efficacy. While several clinical trials focused on phage therapy are underway, the results and conclusions of these trials have not been released yet to the public. However, groups such as IPATH, have been utilizing phages for compassionate treatments that have yielded positive outcomes [115-118]. Compassionate treatments, unlike clinical trials, are not designed to provide results to evaluate the efficacy of a treatment but instead to benefit patients who have exhausted all standard medical options. Compassionate treatment cases have provided insight into the potential effectiveness of phage therapy in humans. While compassionate treatment covers a wide variety of diseases, the most frequently reported bacterial infections targeted are resistant S. aureus strains [119]. In 2006, compassionate phage therapy was effective in the treatment of two individuals with radiation burns infected with resistant S. aureus [120]. Following antibiotic treatment, infections continued to persist leading the individuals to seek compassionate alternative treatment. Patients were treated with a phage cocktail preparation called Phage Bio Derm. This treatment utilizes a lytic phage cocktail with various host specificity suspended in a biodegradable polymer mixture. This treatment results in S. aureus inhibition and eradication of the infection. This treatment protocol highlights the non-invasive manner with which phages can be utilized for treatment of persistent infections especially in the case of skin infections. Where antibiotics and other treatment methods rely on sometimes invasive measures, this phage cocktail can simply be applied as a film to the skin where the infected ulcer is present. There are several other notable examples of compassionate phage treatments showing success. In 2016 compassionate phage treatment was carried out on six individuals with perfuse toe ulcers infected with MRSA. These infections that were non-responsive to all antibiotic treatment responded to treatment with staphylococcal phage Sb-1. In all six patients, phage treatment effectively eradicated all MRSA cells [121]. Recent interest in phage therapy has resulted in the increased involvement of pharmaceutical companies in phage research and clinical trials. Novolytics (UK) has recently announced that phage cocktail gels that target MRSA are in the developmental stage [122]. This phage cocktail would serve to treat nasal carriage of MRSA as well as skin infections and indwelling medical devices [122]. In addition to Novolytics several other companies including Armata Pharmaceuticals Inc., Intralytix Inc., Adaptive Phage Therapeutics Inc., Pherecydes Pharma, and Locus Biosciences Inc. have also announced continued development of phage-based therapeutics. While phase II and phase III trials have not been announced for phage therapy treatment of MRSA infections, it can only be assumed that they are on the horizon. Continued research into phage pharmacokinetics, stability, delivery, partnered with the development of novel formulations and exhaustive clinical trials will eventually allow phage therapy to reach widespread clinical application. 


Conclusion

The increased prevalence and occurrence of antibiotic-resistant bacteria is a major threat to public health especially the notorious antibiotic resistant S. aureus. While antibiotic dose-response has been standardized, consideration of MRSA phages varied replication factors is crucial for the determination of standard relative dosage for ‘killing’ titers. Additionally, MRSA phages multiplication is incumbent on host availability, for this reason, an initial “killing titer” might tremendously increase after phage administration through the phage’s replicative process. An added dimension in the phage biology is its ability to co-evolve with its host, this added advantage over antibiotics enhances the need to study MRSA phages as therapeutic tools against the bacteria. Hence, a clearer insight of MRSA phage biology, pharmacokinetics and pharmacodynamics will provide the requisite avenue for broad application of phage therapy. It is undoubtable that an alternative treatment method for these antibiotic-resistant bacteria such as MRSA is essential to counteract human infections as well the economic burden they present. MRSA being one of the most prevalent antibiotic resistant bacterial strains is an immediate and serious threat to public health. The utilization of lytic S. aureus phages for the treatment of MRSA shows potential as a therapeutic treatment method. Literature has outlined the potential benefits of phage therapy against MRSA due to their host specificity, wide diversity, and success in animal and limited clinical trials. While phage therapy against MRSA requires further study, literature to this date suggests that phage therapy shows favorable potential as a novel therapeutic treatment.

Author Contributions: Conceptualization, L.G.; Software, L.G. and J.A.A; Validation, J.A.A, J.A., B.K.; Writing-original draft preparation, L.G.; Writing-review and editing, L.G., J.A.A, J.A., B.K., and R.V.; Project administration, B.K. All authors have read and agreed to the published version of the manuscript.

Funding: This research was funded by National Science Foundation (NSF); HBCU-RISE; grant number: 1646729.

Institutional Review Board: Not Applicable.

Informed Consent Statement: Not Applicable.

Acknowledgements: The authors acknowledge Derrick Dean and Vida Dennis for their NSF-HBCU-RISE grant support. We also acknowledge Alabama State University C-STEM and Microbiology PhD Program for supplies and support.

Conflicts of Interest: The authors declare no conflicts of interest.


References

  1. Dublanchet A, Fruciano E. A short history of phage therapy. Med Mal Infect. 2008; 38: 415-420. 
  2. Abedon ST. Ecology of Anti-Biofilm Agents I: Antibiotics versus Bacteriophages. Pharmaceuticals. 2015; 8: 525-558.
  3. Fruciano DE, Bourne S. Phage as an antimicrobial agent: d'Herelle's heretical theories and their role in the decline of phage prophylaxis in the West. Can J Infect Dis Med Microbiol. 2007; 18: 19-26.
  4. WHO. Antimicrobial Resistance. 2020. 
  5. CDC. Antibiotic Resistance threats in the United States. 2019.
  6. DM L. Has the era of untreatable infections arrived. J Antimicrobial Chemotherapy. 2009; 64: 29-36.  
  7. Lin DM, Koskella B, Lin HC. Phage therapy: An alternative to antibiotics in the age of multi-drug resistance. World J Gastrointest Pharmacol Ther. 2017; 8: 162-173.
  8. D'Accolti M, Soffritti I, Mazzacane S, Caselli E. Bacteriophages as a Potential 360-Degree Pathogen Control Strategy. Microorganisms. 2021; 9.
  9. Lukacik P, Barnard TJ, Buchanan SK. Using a bacteriocin structure to engineer a phage lysin that targets Yersinia pestis. Biochem Soc Trans. 2012; 40: 1503-1506.  
  10. Lukacik P, Barnard TJ, Keller PW, Chaturvedi KS, Seddiki N, Fairman JW, et al. Structural engineering of a phage lysin that targets gram-negative pathogens. Proc Natl Acad Sci. 2012; 109: 9857-9862.  
  11. Ghosh C, Sarkar P, Issa R, Haldar J. Alternatives to conventional antibiotics in the era of antimicrobial resistance. Trends Microbiol. 2019; 27: 323-338.  
  12. Gildea L, Ayariga J, Boakai KR. Bacteriophages as Biocontrol Agents in Food Microbiology. 2002.
  13. Ayariga JA, Villafane R. Proteolytic analysis of Epsilon 34 Phage Tailspike protein indicates partial Sensitivity to Proteinase K. SunText Rev Virol. 2022; 3: 129.
  14. Gildea L, Ayariga JA, Boakai K. Villafane RR. P22 Phage shows promising antibacterial activity under pathophysiological conditions. Archives Microbiology Immunol. 2022; 6: 81-100.
  15. Ayariga JA, Gildea L, Villafane R. ?34 Phage Tailspike protein is resistant to trypsin and inhibits salmonella biofilm formation. Enliven: Microb Microbial Tech. 2022; 9: 2.
  16. Ayariga JA, Gildea L, Villafane R. Extended ?34 Phage TSP renatures after urea-acid unfolding. Enliven: Microb Microbial Tech. 2022; 9: 1.
  17. Ayariga JA, Gildea L, Wu H, Villafane R. The ?34 Phage Tailspike Protein: an in vitro characterization, structure prediction, potential interaction with s. Newington lps and cytotoxicity assessment to animal Cell Line. J Clin Trials. 2022; 14: 2.
  18. Atia AJ, Azumah AD, Deepa B, Dean D. Tuning phage for cartilage regeneration. In Bacteriophages in Therapeutics. Intech Open. 2021.
  19. Morehead MS. Emergence of global antibiotic resistance. Prime Care. 2018. 45: 467-484. 
  20. Neill JO. Tackling drug-resistant infections globally: Final report and recommendations. Review Antimicrobial Resistance. 2016.
  21. Ardal CBM, Laxminarayan R, McAdams D, Outterson K. Antibiotic Development- Economic, regulatory, and societal challenges. National Review Microbiol. 2019.
  22. Sharland HB, Moja L, Pulcini C, Zeng M. Eml expert committee and antibiotic Working Group. Essential Medicines list becomes a global antibiotic stewardship tool. Lancet Infectious Dis. 2019; 19: 1278-1280. 
  23. Reindel R, Fiore CR. Phage Therapy: considerations and challenges for development. Clin Infect Dis. 2017; 64: 1589-1590.
  24. Chan BK, Abedon ST, Loc-Carrillo C. Phage cocktails and the future of phage therapy. Future Microbiol. 2013; 8: 769-783.
  25. McCallin S, Sarker SA, Sultana S, Oechslin F, Brussow H. Metagenome analysis of Russian and Georgian Pyophage cocktails and a placebo-controlled safety trial of single phage versus phage cocktail in healthy Staphylococcus aureus carriers. Environ Microbiol. 2018; 20: 3278-3293.
  26. Chen L, Yuan S, Liu Q, Mai G, Yang J, Deng D, et al. In vitro design and evaluation of phage cocktails against aeromonas salmonicida. Front Microbiol. 2018; 9: 1476.  
  27. CDC. Methicillin-resistant Staphylococcus aureus (MRSA). 2019. 
  28. Rasmussen G, Monecke S, Brus O, Ehricht R, Soderquist B. Long term molecular epidemiology of methicillin-susceptible Staphylococcus aureus bacteremia isolates in Sweden. PLoS One. 2014; 9: 114276.
  29. Otto M. MRSA virulence and spread. Cell Microbiol. 2012; 14: 1513-1521.
  30. DeLeo FR, Chambers HF. Reemergence of antibiotic-resistant Staphylococcus aureus in the genomics era. J Clin Invest. 2009; 119: 2464-2474.
  31. Cherif DM, Haouz A, Courvalin P. Structural and functional characterization of VanG D-Ala:D-Ser ligase assocaited with vancomycin resistance in enterococcus faecalis. J Biological Chemistry. 2012; 45: 287.
  32. Nicolas I, Bordeau V, Bondon A, Baudy-Floc'h M, Felden B. Novel antibiotics effective against gram-positive and -negative multi-resistant bacteria with limited resistance. PLoS boil. 2019; 17: 3000337.
  33. Geitani R, Moubareck CA, Touqui L, Sarkis DK. Cationic antimicrobial peptides: alternatives and/or adjuvants to antibiotics active against methicillin-resistant Staphylococcus aureus and multidrug-resistant Pseudomonas aeruginosa. BMC microbial. 2019; 19: 54.
  34. Rhoads DD, Wolcott RD, Kuskowski MA, Wolcott BM, Ward LS, Sulakvelidze A, et al. Bacteriophage therapy of venous leg ulcers in humans: results of a phase I safety trial. J Wound Care. 2009; 18: 237-238.
  35. Demir C, Demirci M, Yigin A, Tokman HB, Yildiz SC. Presence of biofilm and adhesin genes in Staphylococcus aureus strains taken from chronic wound infections and their genotypic and phenotypic antimicrobial sensitivity patterns. Photodiagnosis photodynamic therapy. 2020; 29: 101584.
  36. Stapleton PD, Shah S, Ehlert K, Hara Y, Taylor PW. The beta-lactam-resistance modifier (-)-epicatechin gallate alters the architecture of the cell wall of Staphylococcus aureus. Microbiol. 2007; 153: 2093-2103.
  37. Otero LH, Altuve A, Llarrull LI, Lopez C, Kumarasiri M, Lastochkin E, et al. How allosteric control of Staphylococcus aureus penicillin binding protein 2a enables methicillin resistance and physiological function. Proceedings National Academy Sciences United States America. 2013; 110: 16808-16813.
  38. Hoppe PA, Holzhauer S, Lala B, Buhrer C, Gratopp A, Hanitsch LG, et al. Severe infections of Panton-Valentine leukocidin positive Staphylococcus aureus in children. Med. 2019; 98: 17185.
  39. McDevitt D, Nanavaty T, Pompeo K, Bell E, Turner N, McIntire L, et al. Characterization of the interaction between the Staphylococcus aureus clumping factor (ClfA) and fibrinogen. Eur J Biochem. 1997; 247: 416-424.
  40. Mari RT, Langsrud S, Holck A, Moretro T. Different patterns of biofilm formation in Staphylococcus aureus under food-related stress conditions. Int J food microbial. 2019; 116: 372-383.
  41. Gara JP. ica and beyond: biofilm mechanisms and regulation in Staphylococcus epidermidis and Staphylococcus aureus. FEMS Microbiol Lett. 2007; 270: 179-188.
  42. Zapotoczna M, Neill EO, OGara JP. Untangling the diverse and redundant mechanisms of Staphylococcus aureus biofilm formation. PLoS pathogens. 2016; 12: 1005671.
  43. Cucarella C, Solano C, Valle J, Amorena B, Lasa I, Penades JR. Bap, a Staphylococcus aureus surface protein involved in biofilm formation. J Bacteriol. 2001; 183: 2888-2896.
  44. Neill EO, Pozzi C, Houston P, Humphreys H, Robinson DA, Loughman A, et al. A novel Staphylococcus aureus biofilm phenotype mediated by the fibronectin-binding proteins, FnBPA and FnBPB. J Bacteriol. 2008; 190: 3835-3850.
  45. Foulston L, Elsholz AKW, DeFrancesco AS, Losick R. The extracellular matrix of staphylococcus aureus biofilms comprises cytoplasmic proteins that associate with the cell surface in response to decreasing pH. Mbio. 2014; 5.
  46. Nicholas H. The potential of phages to prevent MRSA infections, Res Microbiol. 2008; 159: 400-405.
  47. Steinbacher S, Miller S, Baxa U, Weintraub A, Seckler R. Interaction of Salmonella phage P22 with its O-antigen receptor studied by X-ray crystallography. Biol Chem. 1997; 378: 337-343.
  48. Nilsson N, Malmborg AC, Borrebaeck CA. The phage infection process: a functional role for the distal linker region of bacteriophage protein 3. J Virol. 2000; 74: 4229-4235.
  49. Susskind MM, Botstein D. Molecular genetics of bacteriophage P22. Microbiol Rev. 1978; 42:413. 
  50. Joseph A, Karthikeya V, Robert W, Hongzhuan W, Doba J, Robert V, et al. Initiation of P22 Infection at the Phage Centennial. Frontiers Sci, Technol, Engg Mathematics: Peer-Reviewed J. 2018; 2.
  51. 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.
  52. Mehndiratta PL, Bhalla P. Typing of Methicillin resistant Staphylococcus aureus: a technical review. Indian J Med Microbiol. 2012; 30: 16-23.
  53. Andres D, Hanke C, Baxa U, Seul A, Barbirz S, Seckler R, et al. Tailspike interactions with lipopolysaccharide effect DNA ejection from phage P22 particles in vitro. J Biol Chem. 2010; 285: 36768-36775.  
  54. Prokhorov NS, Riccio C, Zdorovenko EL, Shneider MM, Browning C, Knirel YA, et al. Function of bacteriophage G7C esterase tailspike in host cell adsorption. Mol Microbiol. 2017; 105: 385-398. 
  55. Levinthal C. The Mechanism of DNA replication and genetic recombination in phage. Proc Natl Acad Sci. 1956; 42: 394-404.
  56. Levinthal C, Fisher HW. Maturation of phage and the evidence of phage precursors. Cold Spring Harb Symp Quant Biol. 1953; 18: 29-33.  
  57. Deghorain M, Melderen LV. The Staphylococci phages family: an overview. Viruses. 2012; 4: 3316-3335.
  58. Lindsay JA. Genomic variation and evolution of Staphylococcus aureus. Int J Med Microbiol. 2010; 300: 98-103. 
  59. Brussow H, Canchaya C, Hardt WD. Phages and the evolution of bacterial pathogens: From genomic rearrangements to lysogenic conversion. Microbiol Mol Biol Rev. 2004; 68: 560-602. 
  60. Kwan T, Liu J, DuBow M, Gros P, Pelletier J. The complete genomes and proteomes of 27 Staphylococcus aureus bacteriophages. Proc Natl Acad Sci. 2005; 102: 5174-5179. 
  61. Wentworth BB. Bacteriophage typing of the Staphylococci. Bacteriol Rev. 1963; 27: 253-272. 
  62. Mahmoud HAHA, Senna ASM, Riad OKM, Al MM, Shadi RA. Isolation and characterization of six gamma- irradiated bacteriophages specific for MRSA and VRSA isolated from skin infections. J Radiation Res Applied Sci. 2021; 14: 34-43.
  63. Andrew MQ, King MJA, Carstens EB, Lefkowitz EJ. Order - Caudovirales. Virus Taxonomy Elsevier. 2012; 39-45. 
  64. Kitamura N, Sasabe E, Matsuzaki S, Daibata M, Yamamoto T. Characterization of two newly isolated Staphylococcus aureus bacteriophages from Japan belonging to the genus Silviavirus. Arch Virol. 2020; 165: 2355-2359. 
  65. Nasser A, Azizian R, Tabasi M, Khezerloo JK, Heravi FS, Kalani MT, et al. Specification of bacteriophage isolated against clinical methicillin-resistant staphylococcus aureus. Osong Public Health Res Perspect. 2019; 10: 20-24. 
  66. Rahimzadeh PG, Rezai MS. Characterization of Methicillin-Resistant Staphylococcus aureus (MRSA) phages from sewage at a tertiary pediatric hospital. Archives Pediatric Infectious Dis. 2016; 5.
  67. Mariem N, Mohammed-Ali NMJ. Isolation and characterization of bacteriophage against methicillin resistant staphylococcus aureus. J Med Microbiol Diagnosis. 2015; 5: 213.  
  68. O'Flaherty S, Ross RP, Flynn J, Meaney WJ, Fitzgerald GF, Coffey A, et al. Isolation and characterization of two anti-staphylococcal bacteriophages specific for pathogenic Staphylococcus aureus associated with bovine infections. Lett Appl Microbiol. 2005; 41: 482-486.
  69. Danovaro  R, Corinaldesi C, Dell'anno A, Fuhrman JA, Middelburg JJ, Noble RT, et al. Marine viruses and global climate change. FEMS Microbiol Rev. 2011; 35: 993-1034.
  70. Abdurahman MI, Durukan I. Four temperate bacteriophages from methicillin-resistant staphylococcus aureus show broad bactericidal and biofilm removal activity. Kafkas Universitesi Veteriner Fakultesi Dergisi. 2021; 27: 29-36.
  71. Jeon J, D'Souza R, Hong SK, Lee Y, Yong D, Choi J, et al. Complete genome sequence of the siphoviral Bacteriophage YMC/09/04/R1988 MRSA BP: A lytic phage from a methicillin-resistant Staphylococcus aureus isolate. FEMS Microbiol Lett. 2015; 359: 144-146.
  72. Peng C, Hanawa T, Azam AH, LeBlanc C, Ung P, Matsuda T, et al. Silviavirus phage MR003 displays a broad host range against methicillin-resistant Staphylococcus aureus of human origin. Appl Microbiol Biotechnol. 2019; 103: 7751-7765.
  73. Dakheel KH, Rahim RA, Neela VK, Al-Obaidi JR, Hun TG, Isa M NM, et al. Genomic analyses of two novel biofilm-degrading methicillin-resistant Staphylococcus aureus phages. BMC Microbiol. 2019; 19: 114.
  74. Abatangelo V, Bacci NP, Boncompain CA, Amadio AA, Carrasco S, et al. Broad-range lytic bacteriophages that kill Staphylococcus aureus local field strains. PLOS ONE. 2017; 12: 0181671.
  75. Stephen C. Becker, Frey JF, Donovan DM. The phage K lytic enzyme LysK and lysostaphin act synergistically to kill MRSA. FEMS Microbiol Letters. 2008; 287: 185-191.
  76. Chan BK, Abedon ST, Loc-Carrillo C. Phage cocktails and the future of phage therapy. Future Microbiol. 2013; 8: 769-783.
  77. Young R, Gill JJ. Microbiology. Phage therapy redux-What is to be done. Sci. 2015; 350: 1163-1164.
  78. Fortier LC, Sekulovic O. Importance of prophages to evolution and virulence of bacterial pathogens. Virulence. 2013; 4: 354-365.
  79. Haaber J, Leisner JJ, Cohn MT, Catalan-Moreno A, Nielsen JB, Westh H, et al. Bacterial viruses enable their host to acquire antibiotic resistance genes from neighbouring cells. Nat Commun. 2016; 7: 13333.
  80. Philipson CW, Voegtly LJ, Lueder MR, Long KA, Rice GK, Frey KG, et al. Characterizing Phage Genomes for Therapeutic Applications. Viruses. 2018; 10.
  81. Granowitz EV, Brown RB. Antibiotic adverse reactions and drug interactions. Crit Care Clin. 2008; 24: 421-442.
  82. Rouveix B. Antibiotic safety assessment. Int J Antimicrob Agents. 2003; 21: 215-221.  
  83. Shehab N, Patel PR, Srinivasan A, Budnitz DS. Emergency department visits for antibiotic-associated adverse events. Clin Infect Dis. 2008; 47: 735-743.
  84. Naqi SA, Sahin N, Wagner G, Williams J. Adverse effects of antibiotics on the development of gut-associated lymphoid tissues and the serum immunoglobulins in chickens. Am J Veterinary Res. 1984; 45: 1425-1429.
  85. Bruttin A, Brussow H. Human volunteers receiving Escherichia coli phage T4 orally: a safety test of phage therapy. Antimicrob Agents Chemother. 2005; 49: 2874-2878.  
  86. Merabishvili M, Pirnay JP, Verbeken G, Chanishvili N, Tediashvili M, Lashkhi N, et al. Quality-controlled small-scale production of a well-defined bacteriophage cocktail for use in human clinical trials. PLoS One. 2009; 4: 4944.
  87. McCallin S, Sarker SA, Sultana S, Berger B, Huq S, Krause L, et al. Safety analysis of a Russian phage cocktail: from metagenomic analysis to oral application in healthy human subjects. Virol. 2013; 443: 187-196.
  88. Sarker SA, Sultana S, Reuteler G, Moine D, Descombes P, Charton F, et al. Oral Phage therapy of acute bacterial diarrhea with two coliphage preparations: a randomized trial in children from Bangladesh. EBio Med. 2016; 4: 124-137.  
  89. Gorski A, Wazna E, Dabrowska BW, Dabrowska K, Switala-Jelen K, Miedzybrodzki R, et al. Bacteriophage translocation. FEMS Immunol Med Microbiol. 2016; 46: 313-319.
  90. Stefaniak K, Miernikiewicz P, Drapala J, Drab M, Jonczyk-Matysiak E, Lecion D, et al. Mammalian Host-Versus-Phage immune response determines phage fate in vivo. Sci Rep. 2015; 5: 14802.
  91. Park K, Cha KE, Myung H. Observation of inflammatory responses in mice orally fed with bacteriophage T7. J Appl Microbiol. 2014; 117: 627-633.
  92. Borysowski J, Gorski A. Is phage therapy acceptable in the immune compromised host. Int J Infect Dis. 2008; 12: 466-471. 
  93. Tetz G, Tetz V. Bacteriophage infections of microbiota can lead to leaky gut in an experimental rodent model. Gut Pathog. 2016; 8: 33.
  94. Pincus NB, Reckhow JD, Saleem D, Jammeh ML, Datta SK, Myles IA, et al. Strain specific phage treatment for staphylococcus aureus. infection is influenced by host immunity and site of infection. PLoS One. 2015; 10: 0124280.
  95. Rea K, Dinan TG, Cryan J F. The microbiome: A key regulator of stress and neuro inflammation. Neurobiol Stress. 2016; 4: 23-33.
  96. Metsala J, Lundqvist A, Virta LJ, Kaila M, Gissler M, Virtanen SM, et al. Prenatal and post-natal exposure to antibiotics and risk of asthma in childhood. Clin Exp Allergy. 2015; 45: 137-145.
  97. Cox LM, Blaser MJ. Antibiotics in early life and obesity. Nat Rev Endocrinol. 2015; 11: 182-190.
  98. Mikkelsen KH, Allin KH, Knop FK. Effect of antibiotics on gut microbiota, glucose metabolism and body weight regulation: a review of the literature. Diabetes Obes Metab. 2016; 18: 444-453.
  99. Mai V, Ukhanova M, Reinhard MK, Li M, Sulakvelidze A. Bacteriophage administration significantly reduces Shigella colonization and shedding by Shigella-challenged mice without deleterious side effects and distortions in the gut microbiota. Bacteriophage. 2015; 5: 1088124.
  100. Galtier M, Sordi L, Maura D, Arachchi S, Volant S, Dillies MA, et al. Bacteriophages to reduce gut carriage of antibiotic resistant uropathogens with low impact on microbiota composition. Environ Microbiol. 2016; 18: 2237-2245.
  101. Servick K.  DRUG DEVELOPMENT. Beleaguered phage therapy trial presses on. Sci. 2010; 352: 1506.
  102. Gu J, Liu X, Li Y, Han W, Lei L, Yang Y, et al. A method for generation phage cocktail with great therapeutic potential. PLoS One 2012; 7: 31698.
  103. Bourdin G, Navarro A, Sarker SA, Pittet AC, Qadri F, Sultana S, et al. Coverage of diarrhoea-associated Escherichia coli isolates from different origins with two types of phage cocktails. Microb Biotechnol. 2014; 7: 165-176.
  104. Koskella B. Bacteria-phage interactions across time and space: merging local adaptation and time-shift experiments to understand phage evolution. Am Nat. 2014; 184: 9-21.
  105. Niu YD, Johnson RP, Xu Y, McAllister TA, Sharma R, Louie M, et al. Host range and lytic capability of four bacteriophages against bovine and clinical human isolates of Shiga toxin-producing Escherichia coli O157:H7. J Appl Microbiol. 2019; 107: 646-656.
  106. Latz S, Wahida A, Arif A, Hafner H, Hoss M, Ritter K, et al. Preliminary survey of local bacteriophages with lytic activity against multidrug resistant bacteria. J Basic Microbiol. 2016; 56: 1117-1123.
  107. Wills QF, Kerrigan C, Soothill JS. Experimental bacteriophage protection against Staphylococcus aureus abscesses in a rabbit model. Antimicrob Agents Chemother. 2005; 49: 1220-1221.
  108. Furfaro LL, Payne MS, Chang BJ. Bacteriophage Therapy: clinical trials and regulatory hurdles. Front Cell Infect Microbiol. 2019; 8: 376.  
  109. Payne RJ, Jansen VA. Pharmacokinetic principles of bacteriophage therapy. Clin Pharmacokinet. 2003; 42: 315-325.
  110. Ryan EM, Gorman SP, Donnelly RF, Gilmore BF. Recent advances in bacteriophage therapy: how delivery routes, formulation, concentration, and timing influence the success of phage therapy. J Pharm Pharmacol. 2011; 63: 1253-1264.  
  111. Matsuzaki S, Yasuda M, Nishikawa H, Kuroda M, Ujihara T, Shuin T, et al. Experimental protection of mice against lethal Staphylococcus aureus infection by novel bacteriophage phi MR11. J Infect Dis. 2003; 187: 613-624.
  112. Parracho HM, Burrowes BH, Enright MC, McConville ML, Harper DR. The role of regulated clinical trials in the development of bacteriophage therapeutics. J Mol Genet Med. 2012; 6: 279-286. 
  113. Walsh L, Johnson CN, Hill C, Ross RP. Efficacy of Phage- and Bacteriocin-Based therapies in combatting nosocomial mrsa infections. Frontiers Molecular Biosci. 2021; 8.
  114. O’Flaherty S, Ross RP, Meaney W, Fitzgerald GF, Elbreki MF, Coffey A, et al. Potential of the polyvalent anti-Staphylococcus bacteriophage K for control of antibiotic-resistant staphylococci from hospitals. Appl. Environ. Microbiol. 2005; 71: 1836-1842.
  115. Ooi ML, Drilling AJ, Morales S, Fong S, Moraitis S, Macias-Valle L, et al. Safety and tolerability of bacteriophage therapy for chronic rhinosinusitis due to Staphylococcus aureus. JAMA Otolaryngol. 2019; 145: 723-729.
  116. Leszczynski P, Dabrowska B, Kohutnicka M. Successful eradication of methicillin-resistantStaphylococcus aureus (MRSA) intestinal carrier status in a healthcare worker Case report. Folia Microbio. 2006; 51: 236-238.
  117. Tuomala H, Verkola M, Meller A, Auwera JV, Patpatia S, Jarvinen A, et al. Phage Treatment Trial to Eradicate LA-MRSA from Healthy Carrier Pigs. Viruses. 2021; 13: 1888.
  118. Aslam S, Lampley E, Wooten D, Karris M, Benson C, Strathdee S, et al. Lessons learned from the first 10 consecutive cases of intravenous bacteriophage therapy to treat multidrug-resistant bacterial infections at a single center in the United States. Open Forum Infectious Dis. 2020; 7: 389.
  119. McCallin S, Sacher JC, Zheng J, Chan BK. Current state of compassionate phage therapy. Viruses. 2019; 11: 343.
  120. Jikia D, Chkhaidze N, Imedashvili E, Mgaloblishvili I, Tsitlanadze G, Katsarava R, et al. The use of a novel biodegradable preparation capable of the sustained release of bacteriophages and ciprofloxacin, in the complex treatment of multidrug-resistant Staphylococcus aureus-infected local radiation injuries caused by exposure to Sr90. Clin Exp Dermatol Clin Dermatol. 2005; 30: 23-26.
  121. Fish R, Kutter E, Wheat G, Blasdel B, Kutateladze M, Kuhl S, et al. Bacteriophage treatment of intransigent diabetic toe ulcers: a case series. J Wound Care. 2016; 25: 27-33.
  122. Monk AB, Rees CD, Barrow P, Hagens S, Harper DR.  Bacteriophage applications: where are we now. Lett Appl Microbiol. 2010; 51: 363369.