Next Article in Journal
A Meta-Analysis of the Analgesic Efficacy of Single-Doses of Ibuprofen Compared to Traditional Non-Opioid Analgesics Following Third Molar Surgery
Next Article in Special Issue
In Vitro and In Vivo Gastrointestinal Survival of Non-Encapsulated and Microencapsulated Salmonella Bacteriophages: Implications for Bacteriophage Therapy in Poultry
Previous Article in Journal
Feasibility of Developing Radiotracers for MDM2: Synthesis and Preliminary Evaluation of an 18F-Labeled Analogue of the MDM2 Inhibitor SP-141
Previous Article in Special Issue
Luminescent Phage-Based Detection of Klebsiella pneumoniae: From Engineering to Diagnostics
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Formulations for Bacteriophage Therapy and the Potential Uses of Immobilization

Fixed Phage Ltd., Glasgow G20 0SP, UK
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2021, 14(4), 359; https://doi.org/10.3390/ph14040359
Submission received: 20 February 2021 / Revised: 9 April 2021 / Accepted: 10 April 2021 / Published: 13 April 2021
(This article belongs to the Special Issue Bacteriophages as Therapeutic Delivery Vehicles)

Abstract

:
The emergence of antibiotic-resistant pathogens is becoming increasingly problematic in the treatment of bacterial diseases. This has led to bacteriophages receiving increased attention as an alternative form of treatment. Phages are effective at targeting and killing bacterial strains of interest and have yielded encouraging results when administered as part of a tailored treatment to severely ill patients as a last resort. Despite this, success in clinical trials has not always been as forthcoming, with several high-profile trials failing to demonstrate the efficacy of phage preparations in curing diseases of interest. Whilst this may be in part due to reasons surrounding poor phage selection and a lack of understanding of the underlying disease, there is growing consensus that future success in clinical trials will depend on effective delivery of phage therapeutics to the area of infection. This can be achieved using bacteriophage formulations instead of purely liquid preparations. Several encapsulation-based strategies can be applied to produce phage formulations and encouraging results have been observed with respect to efficacy as well as long term phage stability. Immobilization-based approaches have generally been neglected for the production of phage therapeutics but could also offer a viable alternative.

1. Introduction

It is estimated that antibiotic-resistant bacterial strains account for approximately 33,000 annual deaths in Europe, emphasizing the urgent need for devising novel strategies to tackle this global challenge [1]. The ability of bacteria to acquire drug resistance through random mutation as well as conjugation-mediated genetic transfer has made bacterial infection and contamination a major concern with far reaching implications. Pseudomonas aeruginosa strains have been shown to become resistant to colistin through cross-species plasmid transfer of the MCR-1 gene from resistant strains of Escherichia coli [2]. Other examples of antibiotic-resistant superbugs include methicillin-resistant Staphylococcus aureus, vancomycin-resistant Enterococcus (VRE) and multi-drug-resistant Mycobacterium tuberculosis [3]. The spread of resistance has resulted in the emergence of ‘superbugs’, responsible for an increase in deaths from illnesses previously treatable with conventional antibiotics [4,5,6,7,8].
The search for alternative strategies to solve these problems has rekindled interest in bacteriophages. These viruses can kill specific bacterial targets, leaving other cells unharmed. Additionally, their ability to propagate to high concentrations at the site of infection reduces the need for continuous application [9,10]. A considerable proportion of bacteriophage-related research now focuses on their practical application for the treatment of diseases including respiratory, gastro-intestinal, wound and skin infections [11,12,13,14,15,16]. While phages can be applied to areas of infection in liquid form, this does not necessarily represent the most effective means of treatment, with the few controlled clinical trials that have been carried out yielding mixed results thus far [17,18,19]. Apart from the difficulty in applying a liquid preparation to a site of infection, adverse conditions brought about by the body’s natural physio-chemical environment as well as its immune response could present a considerable challenge to bacteriophage stability [20]. There has therefore been increased attention towards the development of alternative phage formulations, with a view to improving both their efficiency of application as well as their long-term stability [21,22].
The incorporation of bacteriophages into therapeutic formulations typically involves encapsulating them within a stabilizing substance [23,24]. Through such an approach, various antimicrobial materials such as powders, semisolids and nanofibers can be produced, providing more options for effective delivery at the site of infection and, consequently, improved patient outcomes. To this end, encouraging in vitro and in vivo results have been reported for various encapsulated phage formulations including spray and freeze-dried powders, emulsions and liposomes [25,26,27,28]. A strategy that has received considerably less attention with respect to therapeutic formulations is immobilization. Here, phages are instead bound to substrate surfaces. Whilst more commonly applied to the incorporation of bacteriophages into pathogen biosensors, immobilization represents a broad array of techniques which could potentially also be applied in this area. These are discussed in detail in this review, in addition to an overview of the formulation approaches carried out with respect to phage therapeutics thus far.

2. Stabilization and Formulation of Bacteriophage Therapeutics

As with other protein-based macromolecules, bacteriophages are prone to the effects of protein mis-folding and aggregation as well as denaturization, resulting in subsequent loss of functionality when exposed to adverse conditions [29]. Previous studies have reported on the sensitivity of phages to organic solvents, pH, temperature, and salinity [30,31,32,33,34,35]. Several protocols for the long-term storage of free phage have been established by researchers. In general, phages which commonly exist at ambient temperatures can be stored at 4 °C for extended periods of time with only limited drops in titer observed in most cases [36,37]. It should be noted that survivability is highly variable across bacteriophages, with cases of titers depleting over relatively short amounts of time, even when stored at 4 °C [37]. Additional preservation is often observed by freezing at −80 °C [35]. In cases where quick degradation is observed, stability outcomes can be improved using additives such as gelatine, magnesium ions and glycerol [38].
The preparation of bacteriophage formulations for therapeutic delivery presents additional challenges compared to the storage of free phage lysates in the lab. Unlike the latter, which are stored long term in favorable conditions, phage formulations may ultimately be subjected to extreme conditions which will vary depending on their application. In the case of gastrointestinal infections, for example, a therapeutic phage cocktail needs to survive and carry out its function in a highly acidic environment which could prove to be too adverse for non-formulated phages. Phages existing in dried, non-liquid formulations are generally more stable in the longer-term but are still affected by thermal and other stresses, which can produce a drop in titer. Additionally, the actual process by which a given formulation is produced can result in bacteriophage degradation, as exemplified by the processes of freeze-drying and spray-drying [39,40]. All these factors are considerations in developing stable phage formulations with the development typically focusing on assessing phage delivery to target bacteria, establishing the extent of the stability a given formulation provides in a range of conditions and improving phage survival during the formulation production process.
Common methods used to produce phage formulations typically rely on some form of encapsulation. This is a broad term that is used to describe various techniques including emulsification, freeze-drying, spray-drying, liposome encapsulation and electrospinning, in which bacteriophages are coated/surrounded by certain stabilising agents, providing protection against the external environment (Table 1). Once encapsulated phages need to be released from the material to target bacterial cells.
As discussed previously, the motivation behind investigating alternative formulations for bacteriophages as opposed to pure lysates revolves around the flexibility this can provide clinicians to deliver phages more effectively at the site of infection and improve patient outcomes. In a localized skin infection, for example, several delivery options are made possible. Emulsion encapsulation would allow for the production of a topical cream to be applied directly at the site of infection [44,45]. Freeze-drying and spray-drying techniques can be used to produce phage-coated powders, which can then also be incorporated into a cream for direct application, pill-form for oral application as well as incorporation into an inhaler system [12,22]. Immobilization could be utilized to produce phage-coated patches for direct application onto the skin (bacteriophage immobilization is discussed in more detail later). Additionally, there is also the option of applying the original liquid lysates as either oral drops or as part of a parenteral treatment. Formulations can facilitate viral preservation for longer periods of time in harsher conditions, which facilitates their therapeutic application. A good formulation will also allow for the production of product at large scale in the knowledge that it can be stored easily with minimal periodical drop in phage titer (Table 2).

2.1. Emulsification

Emulsions are mixtures of immiscible liquids in which one liquid acts as the continuous phase with the other(s) dispersed within it with the help of a surfactant. In the case of oil-in-water emulsions, this results in their characteristic cream-like nature, a property that makes the resulting formulation appropriate for topical treatments [45]. Emulsions are thermodynamically stable and are used across various industries including food, health care and chemical synthesis [46]. They can be classified according to the droplet size of the dispersed phase, as well as their thermostability. In conventional emulsions, these measure between 100 nm and 100 µm in diameter and are considered thermodynamically unstable [47]. Micro-emulsions, on the other hand, fall in the 2–50 nm range and are thermodynamically stable [48]. Nano-emulsions represent a different class of emulsion characterised by non-thermodynamically stable droplets (they are, instead, kinetically stable) of diameters measuring 100 nm and below. They are formed through mechanical shear processes [49]. Emulsion-based formulations can be modified to promote percutaneous absorption by varying droplet size, changing the emollient and/or emulsifier and incorporating particulate components into the mixture [50]. This can make them particularly effective in treating deep-rooted infections of the skin.
The dispersal and/or encapsulation of bacteriophages in emulsions has been shown to improve their stability whilst facilitating their bioactivity [51,52]. Esteban et al. (2014) used nano-emulsion encapsulation to stabilize phage K lysates for subsequent in vitro testing against 3 strains of Staphylococcus aureus [27]. Nano-emulsions were prepared from a soybean oil—SM buffer mixture using phase inversion temperature. The resultant droplets were 17 nm in diameter, with the authors concluding that the phages were surrounded by nano-droplets as opposed to being encapsulated inside them. Phages in emulsion demonstrated higher activity than their non-emulsified counterparts over a 10-day period at both 4 °C and room temperature. Furthermore, the phages in emulsion were shown to be effective at killing the three S. aureus strains, resulting in complete clearance in 2–5 h depending on the strain used. This was notably better than the liquid phage control, in which bacterial growth did eventually resume following an initial clearance of two of the strains. The compounds used to make up the phases of an emulsion can directly affect stability of the phage inside it. Dini et al. (2012) compared the ability of aqueous phases prepared from 2% sodium alginate and 3% low methoxylated pectin to stabilize two E. coli 0157:H7 phages in microemulsions containing a 10 % (v/v) oleic acid oil phase [53]. Emulsions were prepared using agitation. There were notable differences between the two aqueous phases, with low methoxylated pectin—oleic acid emulsions proving to be more effective than those containing sodium alginate at stabilising the phages against acidity and high ionic strength. On the other hand, a lower starting number of phages was recorded within the emulsion droplets for the former, suggesting that the pectin may have interacted with the phages to hinder the encapsulation process.
More recent work has focused on understanding the ways in which emulsions can facilitate bacteriophage infectivity. Esteban et al. (2018) found that in nano-emulsion encapsulation, there are typically many more droplets than bacteriophages present within the mixture [54]. The authors postulate that both phages and any bacteria within the emulsion are covered in droplets resulting in electrostatic force shielding, in which the negative charges present across bacterial and phage particles are reduced. This, in turn, would act to reduce repulsion between the particles, facilitating phage adsorption and subsequent infection.
Despite the improved bioavailability and delivery of emulsified bacteriophages, there are challenges to the use of such a technique in an upscaled scenario. Limited work demonstrating long-term viability of phages in this type of formulation has been reported. Cold storage as well as further processing, such as freeze drying of emulsions could be used to increase formulation shelf-life, however this would significantly increase the cost of production. Furthermore, the relatively fragile nature of semi-solid materials as well as the increased risk of bacterial contamination makes them a less attractive option with respect to bulk storage and transport [55].

2.2. Freeze-Drying

In freeze-drying (lyophilization) the phase change process of sublimation is used to remove all traces of water from a sample [56]. This is achieved by heating up frozen material below the triple point of water (6.12 mbar and 0 °C). In practise, optimal temperature and pressure fall well below the triple point of water, which corresponds to pure water, as opposed to water surrounding and within a material sample together with any additives used. Ice present begins to sublime, resulting in removal of water from the system. Initial freezing is usually carried out by slowly lowering the temperature. This promotes the formation of larger ice crystals which sublime more readily. Larger crystals can alternatively be produced through annealing. To initiate drying, the pressure of the system is lowered below the triple point through formation of a vacuum. Heat is then added gradually, allowing sublimation to occur. The lyophilization process can take several days to complete. The final product is typically a powder of varying grain sizes. This type of formulation can therefore be applied to a range of delivery scenarios, such as incorporation into oral capsules or topical creams. Inhalable freeze-dried powders containing phage cocktails are also being investigated as an alternative treatment for respiratory illnesses [57].
Freeze-drying has been carried out extensively on non-phage viruses and more recently been the subject of increased interest for use in bacteriophage stabilisation [57,58,59,60,61]. As with other viruses, the freeze-drying process can be detrimental to bacteriophages. The sudden change of state coupled with the formation of ice crystals challenges the integrity of the phage capsid and can result in large scale loss of phage viability. For this reason, excipients are usually added to the phage solution prior to lyophilization, resulting in the viruses ultimately becoming encapsulated and stabilized within them. Zhang et al. (2018) carried out a study comparing trehalose, mannitol, PEG6000 and sucrose as excipients for the lyophilization of M13 bacteriophage [41]. All 4 compounds were effective in stabilising the phages during the process, following which negligible titer losses were observed, supporting prior findings concerning the cryoprotective properties of sugars and polymers. From the results, it was concluded that phages were stabilized more effectively by the two disaccharides, which were able to preserve phages in dried powder over the long term at ambient temperature (not more than 1 log drop over 2 months). This agrees with previous findings showing trehalose and sucrose work well as excipients [62,63].
Proteins have also shown promise for stabilising phages during the freeze-drying process [58]. Liang et al. (2020) showed that Campylobacter-targeting bacteriophage CP30A can be lyophilized using tryptone as an excipient with less than a log drop in titer after the process [58]. Larger drops were however recorded following long-term storage in non-refrigerated conditions. Additionally, increased humidity in dried powder adversely affected phage viability. Bacteriophage structure can also influence stability during lyophilization. In studies on a range of animal viruses, Melanovska et al. (2014) concluded that while enveloped virions survived the process in the presence of excipients such as sucrose, gelatine, skimmed milk and sodium glutamate, their non-enveloped counterparts were able to remain viable in culture medium alone [64].
The powder-form product of the freeze-drying process makes it an interesting option with respect to large scale production. Powders are generally light and stable, making them easy to pack and transport. Additionally, the potential for freeze dried phage formulations to retain viability over periods of months would allow for a non-disruptive incorporation into a production chain. These advantages need to be considered against the high costs and long waiting times (between 20 and 40 h per freeze drying freeze-drying cycle) associated with the process.

2.3. Spray-Drying

Spray-drying allows for the transformation of a liquid substance to a dried particulate form through evaporation [65]. This is achieved by spraying concentrated feed droplets (typically between 10–100 µm in diameter) into a hot drying chamber containing hot air. A higher substance concentration facilitates the evaporation by reducing the amount of liquid needing to be removed, as does the spraying of atomized particles due to the increased surface area to volume ratio. Once inside the drying chamber, moisture begins to evaporate until a dried shell of the substance of interest remains. An attractive aspect of spray-drying is the relative simplicity of the process, and for this reason, the technique has received increased attention with respect to bacteriophage formulations. The resulting product of the spray-drying process is a dry powder which much like the product of the freeze-drying process, can be applied to creams, tablets and inhalable formulations [57].
The drying temperature, air flow rate, type of atomizer as well as droplet size can all influence the extent to which phages survive the process and the phage titer achieved for final product [65]. In particular, the drying temperature typically used for spray-drying, which often exceeds 60 °C, can be especially detrimental to phage viability during the processing [35]. The use of lower drying temperatures set to 50 °C or lower is therefore advisable where bacteriophages formulations are concerned. As with freeze-drying, encapsulation in excipients allows for stabilization of phages during and after the process [66]. Trehalose was used in conjunction with trileucine and pullalan by Carrigy et al. (2020) to stabilize bacteriophage CPA30 for spray-drying, with only a 0.6 log drop in titer being recorded following 1 month of storage at ambient room temperature [67]. Stability over 1 year was demonstrated by Leung et al. (2020) who used varying amounts of trehalose and leucine to stabilize the Pseudomonas phages PEV2 and PEV40 during spray-drying [39]. Less than a 1 log drop in titer was observed after the process. The same was true for all formulations, where despite slight differences, all managed to remain within a log of the initial phage load after 1 year at 4 °C and 20 °C in a vacuum.
The advantages with respect to storage, transport and stability associated with freeze-dried powder also hold true for spray dried powder. Additionally, spray drying is considerably less expensive to run than the former. Whilst it has been successfully applied in this context, the relatively high temperatures required for drying cycles may render it incompatible with many bacteriophage species.

2.4. Liposomes

Liposomes have been used to encapsulate a wide variety of substances, including hydrophilic and hydrophobic drugs, proteins, living cells, nanoparticles, quantum dots and plasmid DNAs [68]. Like cell membranes, liposomes vesicles are composed of phospholipid bilayers [69]. The hydrophobic and hydrophilic forces occurring throughout liposomes and cell membranes drive them to fuse readily on contact with one another, allowing for a facilitated means of substance delivery. This, coupled with the protection provided to the encapsulated substance against adverse external factors has led to many successful therapeutic applications and clinical trials [26]. Several procedures for producing liposomes have been described, including conventional methods such as the Bangham method, which involves reverse phase evaporation and phospholipid injection, as well as more novel approaches such as microhydrodynamic focusing [70]. Size, charge and fluidity of liposomes produced directly affect their ability to encapsulate and release a given substance [71].
Liposome encapsulation has been demonstrated to be a viable option for both bacteriophage stabilization as well as the delivery of phage therapeutics [71,72,73,74]. The fact that liposomes can fuse readily with cells they come into contact with broadens their scope of applications by allowing for the potential to target intracellular pathogens. This has been demonstrated by Singla et al. (2016), who encapsulated bacteriophage KPO12 into liposomes with a view to delivering the phage into macrophage cells infected with Klebsiella pneuoniae [74]. Working with a mouse model, the authors reported 100% protection of encapsulated phages against anti-phage antibodies in extracted mouse serum, compared to free phages which did not remain viable for more than three hours following exposure. Differences in encapsulation efficiency have been recorded between bacteriophages. Cinquierrui et al. (2018) carried out liposome encapsulation for T3 podovirus as well as phage K (myovirus) in liposomes measuring up to 300 nm in size [72]. While the titer recorded for T3 phage was 109 PFU/mL after encapsulation, that of the latter was determined to be 105 PFU/mL. This was attributed to interactions arising between phage K’s capsid and the liposome phospholipids. This may have resulted in phages binding to the liposome exterior, obscuring their tail fibers necessary for adsorption onto host cells. It is therefore likely that amino acid constitution of the phage capsids effects their ability to encapsulate within liposomes. This represents a potential drawback of the approach, with liposome encapsulation best considered on a phage-by-phage basis.
The increased protection afforded to bacteriophages by liposomes also increases their retention time in vivo. This was demonstrated by Chadha et al. (2017), who also studied infections of K. pneumoniae using a burn wound model in mice, in which bacterial loads were found to reduce to a greater extent in mouse blood and organs following administration of a phage cocktail in liposomes as opposed to free phage [73]. The effectiveness of the former was further confirmed by measuring mortality outcomes in mice, where all animals treated with the liposome formulation survived and none survived after free phage treatment. This was found to be case even when the treatment was delayed by 24 h. While liposomes have demonstrated effective protection of phages in animal blood, the high acidity conditions encountered when treating gastrointestinal infections present a greater challenge, as demonstrated by Colom et al. (2015) [75]. In their work, bacteriophages UAB_Phi20, UAB_Phi78, and UAB_Phi87 targeting Salmonella strains were encapsulated in positively charged (between +31.6–35.1 mV) liposomes ranging from 309 to 326 nm in diameter. The liposomes were then subjected to gastric fluid conditions (pH 2.8) in which phage titers dropped 3.7 to 5.4 logs. Despite this notable drop, liposomes still performed better than free phage, for which 5.7 to 7.8 log drops were recorded. In subsequent treatment of Salmonella-infected broiler chickens with the liposome and liquid phage formulations, both were able to provide protection to the animals when administered daily, however with encapsulated phages protection remained for up to 1 week after treatment was stopped, by which time all activity of the non-encapsulated phages had disappeared.
As with emulsion-based formulations, the problems that could arise with respect to long-term stability of liposome-encapsulated phage need to be considered. The potential requirement for refrigeration or post-production processing would add to the overall production costs and the liquid nature of the final product would make transportation and storage more challenging. As has been demonstrated in numerous in vivo studies, the main benefits of liposome encapsulation draw from improved delivery of phage to target cells as well increased protection afforded to encapsulated phages against adverse in vivo conditions.

2.5. Electrospinning

The production of nanofibers through electrospinning can be used to stabilize bacteriophages and produce antibacterial fibers [76]. For this technique, a charged, molten polymer solution is drawn onto an electrode of opposite charge. The final, dried nanofibers typically measure 100 nm or less in diameter [76]. The addition of bacteriophages to the liquid polymer prior to carrying out the process results in encapsulated bacteriophages within the nanofibers, which can be applied for both water-soluble and insoluble polymers.
The electrospinning process does present challenges to bacteriophage stability, with the high voltages used during the process resulting in a large proportion of the phages dying [77]. Furthermore, rapid evaporation of water and subsequent osmotic change in the environment surrounding electrospun phages has been identified as a cause of the low storage viability observed in some studies [77,78,79]. Viability during the electrospinning process as well as during subsequent storage may be improved by adding magnesium salts and excipients such as trehalose [80]. Despite these observations, Diaz et al. (2018) demonstrated the applicability of the electrospinning process to a broad range of bacteriophages by integrating Fersis and PhageStaph commercial phage cocktails into nanofibers formed from a soluble (polyethylene glycol) and biodegradable polymer (polyester urea) [81]. Viable phage titers were broadly conserved across samples, with a slight decrease in the case of polyester urea fibers when lyophilized phage was used instead of the original solutions, and with the resultant nanofibers demonstrating antimicrobial activity against corresponding bacterial hosts, inhibiting growth for up to 80 h after exposure. The authors suggested that phages can survive exposure to electric fields up to 40 kVcm−1 for 5 min. These findings agree with observations made by Andriolo et al. (2018) who found that both electric field voltage and solvent had a negligible effect on phage viability [82]. Heat exposure, on the other hand, did adversely affect phages, with temperatures exceeding 55 °C resulting in complete loss of phage viability. It is advisable to use polymers with melting points falling below this value, however, much like spray drying, the higher temperatures associated with this process could make it challenging to avoid significant titer loss for certain bacteriophages.
Electrospinning is unique amongst encapsulation methods in that a final product can take several forms. Fibers can be broken down to form small powders, or molded into specific shapes. This could be particularly relevant to the medical devices field. Furthermore, the process can act as a bridge between encapsulation and immobilization through the act of electrospinning encapsulated phage over substrate surfaces.

3. Bacteriophage Immobilization

Immobilization refers to the chemical, physio-chemical or electrostatic binding of bacteriophages to a surface. Most research in the field of phage immobilization has been carried out for the development of pathogen biosensors, as well as for the production of antibacterial food packaging [83,84,85,86,87,88]. Despite the limited number of studies examining the use of immobilized phage for therapeutic applications, it should be considered a credible alternative to the approaches described previously, owing to the success that immobilized phage have shown in killing their bacterial targets in other applications, as well as its relative simplicity compared to some of the other approaches. Despite this, the advantages and drawbacks of the various immobilization techniques that have been described would need to be considered for this application (Table 3 and Table 4). As with encapsulation, immobilized phage can potentially be integrated into various formulations including powders, patches, wound dressings and creams, however the fact that it can technically be carried out on most surfaces increases the potential range of applications.
Immobilized phages are stabilized through the interactions that occur between the virions and the surface. These come in the form of non-specific binding, as well as more permanent covalent bonds [23]. A key difference between immobilization and the encapsulation-based approaches discussed previously is the ultimate location of the phages, as immobilized phages are usually exposed to the external environment. While this has implications with respect to stability, it also allows for more direct contact between the phage and the target bacteria.
Immobilized phages can be rigidly attached to the surface, so the spatial orientation of the phages post immobilization is a factor that needs to be considered. In the case of tailed phages, a ‘tail-up’ orientation is very much preferred to facilitate binding to bacteria and DNA injection, with evidence suggesting that controlling orientation can drastically increase the concentration of infective immobilized phage [86,92]. This does not apply to phages such as PRD1 and PR772, in which receptor binding sites are uniformly distributed on their capsids [93]. Another factor that affects the ability of immobilized phages to infect their target bacteria is coating density and efficiency. Being able to consistently apply phages in a uniform density over the surface is desirable, as it allows for reproducibility, as well as the fact that phage clustering and non-uniform immobilization has been observed to hinder efficient bacteriophage function [94].

3.1. Physical Adsorption

Physical adsorption, or physisorption, refers to the adhesion of particles onto a surface brought about by van der Waal’s forces, dipole-dipole moments, electrostatic forces and steric and hydrophobic interactions [95]. Van der Waals forces, although weak, occur in all molecular species. This makes physical adsorption a ubiquitous occurrence. It represents a quick and relatively simple ways to immobilize a species onto a given surface and, because it occurs through non-chemical interactions, it typically does not result in any chemical alteration of the absorbate. However, because of these physical stresses as well as extremes of acidity, temperature and ionic strength can act to reduce attachment or reverse it post immobilization [23]. For example, Singh et al. (2009) found the density of immobilized phage to decrease by 8 phages/μ2 when the temperature was lowered from 40 °C to ambient [89].
Unaided physical adsorption is a term used to describe the direct application of the adsorbent to the surface. The simplicity of this approach means that complex preparation steps are avoided, with the procedure typically involving the exposure of the surface in question to a high concentration solution of the absorbate. Bennett et al. (1997) studied the unaided adsorption of phages during the development of a technique for the separation of pathogenic Salmonella strains from foodstuffs [96]. Physisorption of the lytic phage Sapphire was achieved by exposing polystyrene strips to high concentration phage solutions and incubating overnight. Similarly, E. coli biosensors have been produced following the immersion of long-period fibres in bacteriophage T4 lysate [97]. Unaided physical adsorption is often used as a control when testing out alternative immobilization strategies [89,90,94].
The use of bandages soaked in bacteriophage lysate to treat topical infections can be considered a form of unaided physical adsorption. Abul–Hassan et al. (1990) made use of such a strategy to treat burn wound sepsis caused by Pseudomonas aeruginosa in 30 patients [98]. Dressings containing adsorbed phage were applied to infected wounds, with positive effects observed in 24 of the patients. Similarly, Kifelew et al. (2020) showed that gauze soaked in purified phage cocktail AB-SA01 was effective in decreasing bacterial load of multidrug resistant Staphylococcus aureus and promoting diabetic wound closure in mice [99]. Application of bacteriophages in this manner might work effectively in cases of immediate application; however, it is unlikely that wound dressing and other medical devices soaked in phage lysates can be stored long term without significant reduction in phage titer and, consequently, overall efficacy.
A drawback of unaided physical adsorption is the fact that the process is chemically undirected. The reliance on weaker, random interactions occurring between the phage and surface leaves limited opportunity to direct the process and spatially arrange the attached phage. In most cases, physical adsorption leads to undesirable disorganized attachment, with adsorbed particles bound in all orientations and with non-uniform spacing and aggregation a common occurrence [100]. The Langmuir–Blodgett technique has been used as a means of depositing bacteriophages as a monolayer on various substrates [92,101,102,103]. This increases uniform spacing between particles, which facilitates the accurate quantification of bound phage and increases targeting efficiency. The organization of phages into single layers could also be advantageous during commercial scale up, as a means of reducing phage wastage and decreasing production costs.
Aided physical adsorption refers to techniques that actively promote the interactions that give rise to physical binding. Dipole-dipole and hydrophobic interactions, as well as electrostatic forces, represent the strongest types of non-covalent binding that occur between a surface and absorbate [95,104]. In the case of hydrophobicity, the occurrence of both polar and non-polar amino acid residues makes it difficult to reliably predict immobilization efficiency, outside of empirical testing. This is illustrated by the contradictory results obtained when measuring the hydrophobicity of MS2 bacteriophage in two separate studies, despite both using the same hydrophobicity assay [105,106,107]. MS2 is one of the only phages tested for its ability to adsorb to surfaces using hydrophobic interactions [104,108,109,110]. One reason for this could be fears that hydrophobic interactions can disrupt protein function by encouraging unfolding, or alternative folding arrangements, a phenomenon that has been reported for the hydrophobic-mediated immobilization of enzymes [111]. Just as hydrophobic bonds have high affinity for each other, the same can be said of regions of high polarity, which can give rise to dipole-dipole interactions. In covalent bonds between atoms of different electronegativities, the electron cloud is distributed unevenly, with either atom assuming a partial positive or negative charge (a dipole) [112]. Opposite partial charges from different covalent bonds are attracted to each other, bringing about dipole-dipole interactions. This phenomenon can be utilized in phage immobilization. Here, polar amino acid sidechains on the surface of the capsid are made to form dipole-dipole interactions with a polar activating layer deposited on the substrate. Singh et al. (2009) improved the immobilization efficiency of T4 on gold surfaces by applying layers of sugars and amino acid coatings, resulting in up to a 7-fold increase in bacteriophage adsorption [89]. Similar loading increases have been observed on gold surfaces with S. aureus phages [113]. Adsorption has also been enhanced through addition of phage to printing ink formulations, with polar molecules in the ink resulting in phage retention on the surface following printing [114,115]. Such an approach demonstrates aspects of both immobilization and encapsulation.

3.2. Charge-Directed Immobilization

In charge-directed immobilization, electrostatic attraction between permanent opposing charges on the surface and adsorbent is used to bring about immobilization. These are considerably stronger than the interactions discussed thus far. Bacteriophages often consist of charged regions; phage heads usually possess a net negative charge with the opposite being true for their tails [60,104]. For this reason, phages have been shown to bind tail-down or tail-up, depending on the net charge present on the surface [116]. It follows that the application of positive charges to surfaces is a favored strategy to bind phages in the desired tail-up orientation. Anany et al. (2011) carried out charge-based immobilization of phage cocktails targeting E. coli and Listeria host strains. Phage cocktails of different concentrations were applied to cellulose disks pre-treated with 0.5% wt/vol polyvinylamine polymer. It was concluded that the net positive charge on treated surfaces lead to increased immobilization efficiency [60]. Similar results with positively charged substrates have been reported in other studies [90,116,117]. Comparative studies have shown more phage binding in charge-based immobilization, when compared to other reversible approaches, which is likely due to the strength of the interactions [90]. This is supported by the observation that it has been shown to perform less efficiently than covalent-based immobilization, which results in an even stronger interaction [82]. The technique can be applied in conjunction with other immobilization approaches as a means of guaranteeing the desired bacteriophage orientation with enhanced attachment strength [118]. A recent study found that the application of alternating current across a gold surface functionalized with polar molecules resulted in a dense, ordered layer of phages in tail-up conformation [91].
As with physical adsorption, electrostatic binding is also influenced by physiochemical properties of the medium, such as ionic strength and pH, which can directly affect protein charge through the isoelectric effect. This was demonstrated by Peng et al. (2011), who increased the pH level in the surrounding medium beyond the isoelectric point of tobacco mosaic virus [119]. The negative charge on the phage heads was increased, leading to increased tail-up orientation on gold surfaces. It is therefore recommended that the isoelectric point of the phage is known prior to carrying out charge-based immobilization, to allow targeted process optimization. The fact that most phage heads possess net negative charges makes this approach broadly applicable across most phage groups. Despite this, one limitation is the resulting attraction/repulsion-based forces that may arise between treated materials. In the case of polymer sheets for example, cationic surfaces may bind to the uncharged side of sheets packed on top of them, making for difficult handling post-production.

3.3. Protein Ligand

The natural tendencies of proteins to adsorb to certain ligands can be exploited for the purpose of bacteriophage immobilization. The surface and absorbate are coupled with a binding protein and its corresponding ligand, respectively, with the interaction and subsequent immobilization occurring once they encounter one another. Streptavidin is a protein that occurs in the bacterium Streptomyces avidinii [120]. It has a strong affinity for biotin, a vitamin involved in several metabolic processes, and binds to it through one of the strongest non-covalent interactions known [121]. Protein-ligand interactions like this have been used for bacteriophage immobilization [86,92,116,122,123,124]. Ligands such as biotin are normally crosslinked to bacteriophages through ester activation with N-hydroxysuccinimide (NHS) and other carboimmides [86,92,122,125]. NHS-biotin reacts with primary amines found in side-chains of amino acids such as lysine, as well as terminal amino groups of polypeptides, resulting in the permanent attachment of biotin. This covalent process is known as biotinylation and there are numerous examples of it being carried out with enzymes [126,127]. Alternatively, the gene coding for the ligand or binding protein of interest can be integrated with the bacteriophage genome. Tolba et al. (2010) fused the genes bccp and cbm, which code for biotin carboxyl carrier protein and cellulose binding domain respectively, with the soc gene of T4 phage [86]. The protein coded for by soc, small outer capsid protein, forms part of the phage capsid, resulting in the expression of biotin carboxyl carrier protein or cellulose binding protein on the T4 head which permitted subsequent affinity immobilization onto respective streptavidin and cellulose-containing surfaces. Coupling a phage with a binding agent this way allows for the strategic positioning of the later. This is especially relevant to tailed phage immobilization, in which the ‘tail-up’ orientation is desired. The major downside to this approach is the complexity of the procedure, which requires a relatively detailed knowledge of the phage, and significant time and resources to plan and effectively execute immobilization. In addition to this, altering a phage’s genome can potentially result in undesired changes to its activity, such as a decreased burst size and an extended latency period [86]. Bacteriophage attachment using protein-ligand interactions has been shown to be effective, with Gervais et al. (2007) reporting on a 15-fold increase in phage binding compared to unaided physisorption [122].

3.4. Covalent

All of the approaches discussed so far have not involved the alteration of substances through the formation of new chemical bonds. Covalent immobilization represents the most permanent and irreversible form of attachment, demonstrated by the ability of covalently immobilized phages to remain bound to substrate even after prolonged exposure to sonication forces [128]. The ability to withstand mechanical stresses could play a role in future development of robust therapeutic products. Studies have also found covalent-based approaches allow for increased binding efficiency, with one study reporting a 37-fold increase in binding efficiency compared to unaided physical adsorption [89,100]. Covalent immobilization can be achieved by crosslinking phage to the substrate. Bacteriophages react covalently through amino acid residues protruding from their viral capsids. These include carboxylic groups from glutamine and aspartic acid, amines from lysine, sulfide groups from cysteine and phenols from tyrosine [129]. In some cases, phages have been observed to bond on mere exposure to certain substrates. M13 phage, for example, readily binds to sulfur particles covalently through carboxylic acid functional groups on glutamine and aspartic acid residues [130]. The technique employed here is similar to the biotinylation technique described previously except in this case, the phage is cross-linked directly to the surface as opposed to an affinity-binding protein. As with protein-ligand immobilization, carboiimide-based cross-linking is often favored as a means of bioconjugation in covalent-based immobilization [82,91,93,128,131]. Janczuk et al. (2017) used EDC to activate carboxylic groups on magnetic fluorescent beads. Subsequent phage attachment via lysine residues resulted in the formation of amide linkages to EDC. Non-carbodiimide-based cross-linkers that have been applied to bacteriophage immobilization include glutaraldehyde and maleic anhydride [89,132]. The main drawbacks to linker-based covalent attachment arise from the potential disruptions to phage activity after bond formation. If a bonding occurs with a residue near the phage adsorption site, this can potentially obscure it, limiting or eliminating the ability of the phage to bind to its target. Additionally, the process is relatively complicated, and could present significant challenges with respect to scale-up.
The direct covalent binding of bacteriophages to surfaces in the absence of cross-linkers has also been reported [133]. Here, the authors attached phage vB_Pae_Kakheti25 onto polycaprolactone fibers. These were subjected to acidic conditions for activation prior to bacteriophage attachment. Effective killing of the host was observed even after 25 rinses of the substrate, demonstrating the robustness of the attachment. Simpler processes such as the one described are appealing as a means of producing bioactive surface on a large scale, due to the lower costs associated with the process. An area of increasing interest concerns the use of plasma treatment to achieve immobilization.
The ionisation of gaseous particles through electron bombardment results in the formation of plasma [134]. Gas in this state typically consists of a mixture of ionized particles, free electrons and radicals, whose application results in chemical changes to treated surfaces [135]. These can be exploited to permanently coat surfaces with substances of interest. Initial studies into plasma-mediated immobilization found that it allowed for substances to be deposited consistently, resulting in production of layers of uniform thickness and limited damage to the adsorbate [136]. In their covalent immobilization of phages T1 and ɸ11, Pearson et al. (2013) first grafted maleic anhydride onto polyethylene and polytetrafluoroethylene surface using microwave plasma [132]. This acted as a linker to allow amide bonding with amine residues on the phages. Wang et al. (2016) used reactive ion etching on polyhydroxyalkanoate surfaces for EDC/sulfo-NHS linker-based immobilization of T4 phage [137]. The linker was attached to the surface either through reaction with plasma generated carboxylate groups on the surface, or through graft polymerisation of acrylic acid with subsequent addition of the linker. T4 bacteriophage was then covalently bound to the surface. The authors of this study compared immobilization efficiency of plasma-treatment with and without linkers. An interesting observation was the higher efficiencies for immobilization performed in the absence of linkers. Considering the increased costs and complications of using linkers for the attachment of biological entities, this suggests that plasma-based techniques may have an important role to play in developing stable formulations for therapeutic phage applications. Linker-free immobilization is believed to occur through direct reaction of the adsorbate with free radicals generated on the treated surface, resulting in covalent bond formation [138]. Therefore, conditions which stabilize radicals on the surface, increase the efficiency of covalent bond formation. Tropoelastin was covalently immobilized onto polyethersulfone (PES) treated with plasma-immersion ion implantation, yielding permanent biofunctionality to the material [139].
Plasma-based immobilization of bacteriophages has already started being commercialized. At Fixed Phage Ltd (Glasgow, UK), corona discharge is used to bind bacteriophages covalently to a range of substrates, such as food packaging, wound dressings, animal feed and powders, for formulation into creams and gels [140,141]. In agreement with other studies, promising results have been generated demonstrating substantially enhanced phage stability after immobilization. For example, bacteriophages specific for Vibrio parahaemolyticus were immobilized onto shrimp feed and shown to retain titers sufficient to treat disease for more than 250 days after storage at 30 °C [142]. and manuscript in preparation. In the same study, phages applied to feed without prior corona treatment lost all activity after 21 days. In a subsequent tank trial, phage-treated feed was shown to protect Thor amboinensis model shrimps against a V. parahaemolyticus challenge [143]. 5 days following exposure to the pathogen, 80% of shrimps receiving regular feed died whilst 90% of the phage-treated group were still alive. The technology has also been applied to demonstrate extensions of shelf life in bagged spinach. A phage cocktail targeting Pseudomonas was developed and immobilized onto plastic inserts. Bags containing the inserts demonstrated a 1 day increase in shelf life compared to their untreated counterparts [manuscript in preparation]. Corona discharge is a well-established industrial process, is relatively cheap to operate and can be applied onto most materials to activate them. These factors make it, along with other plasma-based immobilization processes, ideally suited for use in formulating effective, stable therapeutic phage formulations.

4. Conclusions and Future Prospects

Bacteriophages represent a viable treatment alternative for bacterial-borne diseases. Their application in clinical settings as a last resort treatment has demonstrated their potential in individual patients but for the widespread therapeutic use of licensed phage products to be achieved in the future, two conditions will need to be satisfied: the successful completion of clinical trials proving their efficacy in a significant portion of cases, and an economically and qualitatively viable means of mass production.
The application of specialized formulations will be key to any future clinical trial successes in bacteriophage therapy. The results of recent studies strongly suggest that phage formulations can act to stabilize phages against adverse in vivo conditions while also offering a more pragmatic route of administration compared to liquid phage preparations. While substantial progress has been made with encapsulation-based approaches, another promising approach to the formulation of phage therapeutics is through bacteriophage immobilization. Bacteriophage diversity can affect the extent to which a given formulation process will be successful, emphasizing the importance of selecting the most appropriate approach for the phage(s) being considered. This has increased the need for a comparative analysis of the different strategies currently available and better understanding of the role phage diversity plays in this regard. Another point of consideration is the potential variability of pathogenic strains across patients, which will require the stable and cost-effective formulation of large phage cocktails offering maximum coverage to offset any differences.
In addition to the other considerations, formulations will need to be considered in terms of the ease at which their production can be scaled up. In cases such as linker-based immobilization, multiple processing steps would be involved, while higher running cost and increased lead times would be associated with other processes such as freeze-drying. The most attractive phage products from a production point of view will therefore be those that are relatively straightforward to produce consistently. Formulations which can be stored for extended periods of time at ambient temperatures are also more likely to be favored.
It is likely that different formulation methods will be required for different applications and that further research is needed in this area to facilitate the widespread use of phages as genuine viable alternatives to other antibiotics in human therapy.

Author Contributions

Writing—original draft preparation, D.R.; writing—review and editing, D.R. and J.C.; supervision, J.C.; All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors of this review paper are both employees of Fixed Phage Ltd (UK), a company actively involved in the development of bacteriophage formulations.

References

  1. Cassini, A.; Högberg, L.D.; Plachouras, D.; Quattrocchi, A.; Hoxha, A.; Simonsen, G.S.; Colomb-Cotinat, M.; Kretzschmar, M.E.; Devleesschauwer, B.; Cecchini, M.; et al. Attributable deaths and disability-adjusted life-years caused by infections with antibiotic-resistant bacteria in the EU and the european economic area in 2015: A population-level modelling analysis. Lancet Infect. Dis. 2019, 19, 56–66. [Google Scholar] [CrossRef] [Green Version]
  2. Willmann, M.; Steglich, M.; Bunk, B.; Peter, S.; Neher, R.A. Rapid and consistent evolution of colistin resistance in extensively drug-resistant Pseudomonas aeruginosa during morbidostat culture. Antomicrob. Agents Chemother. 2017, 61, 1–16. [Google Scholar] [CrossRef] [Green Version]
  3. Azam, A.H.; Tanji, Y. Bacteriophage-host arm race: An update on themechanism of phage resistance in bacteria and revenge of the phage with the perspective for phage therapy. Appl. Microbiol. Biotechnol. 2019, 103, 2121–2131. [Google Scholar] [CrossRef]
  4. Zaman, S.B.; Hussain, M.A.; Nye, R.; Mehta, V.; Mamun, K.T.; Hossain, N. A review on antibiotic resistance: Alarm bells are ringing. Cureus 2017, 9, e1403. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Hume, P.J.; Singh, V.; Davidson, A.C.; Koronakis, V. Swiss army pathogen: The salmonella entry toolkit. Front. Cell. Infect. Microbiol. 2017, 7. [Google Scholar] [CrossRef] [PubMed]
  6. Valilis, E.; Ramsey, A.; Sidiq, S.; DuPont, H.L. Non-o157 shiga toxin-producing escherichia coli—a poorly appreciated enteric pathogen: Systematic review. Int. J. Infect. Dis. 2018, 76, 82–87. [Google Scholar] [CrossRef] [Green Version]
  7. Gao, W.; Howden, B.P.; Stinear, T.P. Evolution of virulence in enterococcus faecium, a hospital-adapted opportunistic pathogen. Curr. Opin. Microbiol. 2018, 41, 76–82. [Google Scholar] [CrossRef]
  8. Navon-Venezia, S.; Kondratyeva, K.; Carattoli, A. Klebsiella pneumoniae: A major worldwide source and shuttle for antibiotic resistance. FEMS Microbiol. Rev. 2017, 41, 252–275. [Google Scholar] [CrossRef]
  9. Curtright, A.J.; Abedon, S. Phage therapy: Emergent property pharmacology. J. Bioanal. Biomed. 2012, S6. [Google Scholar] [CrossRef] [Green Version]
  10. Abedon, S. Phage therapy pharmacology: Calculating phage dosing. Adv. Appl. Microbiol. 2011, 77, 1–40. [Google Scholar] [CrossRef]
  11. Seo, B.-J.; Song, E.-T.; Lee, K.; Kim, J.-W.; Jeong, C.-G.; Moon, S.-H.; Son, J.S.; Kang, S.H.; Cho, H.-S.; Jung, B.Y.; et al. Evaluation of the broad-spectrum lytic capability of bacteriophage cocktails against various salmonella serovars and their effects on weaned pigs infected with salmonella typhimurium. J. Vet. Med. Sci. 2018, 80, 851–860. [Google Scholar] [CrossRef] [Green Version]
  12. Chang, R.Y.K.; Chen, K.; Wang, J.; Wallin, M.; Britton, W.; Morales, S.; Kutter, E.; Li, J.; Chan, H.-K. Proof-of-principle study in a murine lung infection model of antipseudomonal activity of phage pev20 in a dry-powder formulation. Antimicrob. Agents Chemother. 2018, 62, e01714-17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Nabil, N.M.; Tawakol, M.M.; Hassan, H.M. Assessing the impact of bacteriophages in the treatment of salmonella in broiler chickens. Infect. Ecol. Epidemiol. 2018, 8, 1539056. [Google Scholar] [CrossRef] [PubMed]
  14. Wright, A.; Hawkins, C.H.; Änggård, E.E.; Harper, D.R. A controlled clinical trial of a therapeutic bacteriophage preparation in chronic otitis due to antibiotic-resistant Pseudomonas aeruginosa; a preliminary report of efficacy. Clin. Otolaryngol. 2009, 34, 349–357. [Google Scholar] [CrossRef] [PubMed]
  15. Kortright, K.E.; Chan, B.K.; Koff, J.L.; Turner, P.E. Phage therapy: A renewed approach to combat antibiotic-resistant bacteria. Cell Host Microbe 2019, 25, 219–232. [Google Scholar] [CrossRef] [Green Version]
  16. Aminov, R.; Caplin, B.J.; Chanishvili, N.; Coffey, A.; Cooper, I.; De Vos, D.; Rí Doška, J.; Friman, V.-P.; Kurtböke, I.; Pantucek, R.; et al. Application of bacteriophages in focus. Microbiol. Aust. 2017, 38, 63–66. [Google Scholar] [CrossRef] [Green Version]
  17. Sarker, S.A.; Sultana, S.; Reuteler, G.; Moine, D.; Descombes, P.; Charton, F.; Bourdin, G.; McCallin, S.; Ngom-Bru, C.; Neville, T.; et al. Oral phage therapy of acute bacterial diarrhea with two coliphage preparations: A randomized trial in children from bangladesh. EBioMedicine 2016, 4, 124–137. [Google Scholar] [CrossRef] [Green Version]
  18. Jault, P.; Leclerc, T.; Jennes, S.; Pirnay, J.P.; Que, Y.-A.; Resch, G.; Rousseau, A.F.; Ravat, F.; Carsin, H.; Le Floch, R.; et al. Efficacy and tolerability of a cocktail of bacteriophages to treat burn wounds infected by Pseudomonas aeruginosa (PhagoBurn): A randomised, controlled, double-blind phase 1/2 trial. Lancet Infect. Dis. 2019, 19. [Google Scholar] [CrossRef]
  19. Górski, A.; Borysowski, J.; Międzybrodzki, R. Phage therapy: Towards a successful clinical trial. Antibiotics 2020, 9, 827. [Google Scholar] [CrossRef]
  20. Hodyra-Stefaniak, K.; Miernikiewicz, P.; Drapała, J.; Drab, M.; Jończyk-Matysiak, E.; Lecion, D.; Kaźmierczak, Z.; Beta, W.; Majewska, J.; Harhala, M.; et al. Mammalian host-versus-phage immune response determines phage fate in vivo. Sci. Rep. 2015, 5. [Google Scholar] [CrossRef] [Green Version]
  21. Vandenheuvel, D.; Lavigne, R.; Brüssow, H. Bacteriophage therapy: Advances in formulation strategies and human clinical trials. Annu. Rev. Virol. 2015, 2, 599–618. [Google Scholar] [CrossRef]
  22. Malik, D.J.; Sokolov, I.J.; Vinner, G.K.; Mancuso, F.; Cinquerrui, S.; Vladisavljevic, G.T.; Clokie, M.R.J.; Garton, N.J.; Stapley, A.G.F.; Kirpichnikova, A. Formulation, stabilisation and encapsulation of bacteriophage for phage therapy. Adv. Colloid Interface Sci. 2017, 249, 100–133. [Google Scholar] [CrossRef] [Green Version]
  23. Hosseinidoust, Z.; Olsson, A.L.J.; Tufenkji, N. Going viral: Designing bioactive surfaces with bacteriophage. Colloids Surf. 2014, 124, 2–16. [Google Scholar] [CrossRef]
  24. Choińska-Pulit, A.; Mituła, P.; Śliwka, P.; Łaba, W.; Skaradzińska, A. Bacteriophage encapsulation: Trends and potential applications. Trends Food Sci. Technol. 2015, 45, 212–221. [Google Scholar] [CrossRef]
  25. Lin, Y.; Chang, R.Y.K.; Britton, W.J.; Morales, S.; Kutter, E.; Chan, H.-K. Synergy of nebulized phage pev20 and ciprofloxacin combination against pseudomonas aeruginosa. Int. J. Pharm. 2018, 551, 158–165. [Google Scholar] [CrossRef] [PubMed]
  26. Bulbake, U.; Doppalapudi, S.; Kommineni, N.; Khan, W. Liposomal formulations in clinical use: An updated review. Pharmaceutics 2017, 9, 12. [Google Scholar] [CrossRef] [PubMed]
  27. Esteban, P.P.; Alves, D.R.; Enright, M.C.; Bean, J.E.; Gaudion, A.; Jenkins, A.T.A.; Young, A.E.R.; Arnot, T.C. Enhancement of the antimicrobial properties of bacteriophage-k via stabilization using oil-in-water nano-emulsions. Biotechnol. Prog. 2014, 30, 932–944. [Google Scholar] [CrossRef] [Green Version]
  28. Dryakhlov, V.O.; Nikitina, M.Y.; Shaikhiev, I.G.; Galikhanov, M.F.; Shaikhiev, T.I.; Bonev, B.S. Effect of parameters of the corona discharge treatment of the surface of polyacrylonitrile membranes on the separation efficiency of oil-in-water emulsions. Surf. Eng. Appl. Electrochem. 2015, 51, 406–411. [Google Scholar] [CrossRef]
  29. Vagenende, V.; Yap, M.G.S.; Trout, B.L. Mechanisms of protein stabilization and prevention of protein aggregation by glycerol. Biochemistry 2009, 48, 11084–11096. [Google Scholar] [CrossRef]
  30. Olofsson, L.; Ankarloo, J.; Andersson, P.O.; Nicholls, I.A. Filamentous bacteriophage stability in non-aqueous media. Chem. Biol. 2001, 8, 661–671. [Google Scholar] [CrossRef] [Green Version]
  31. Taj, M.K.; Ling, J.X.; Bing, L.L.; Qi, Z.; Taj, I.; Hassani, T.M.; Samreen, Z.; Yunlin, W. Effect of dilution, temperature and ph on the lysis activity of t4 phage against e. coli bl21. J. Anim. Plant Sci. 2014, 24, 1252–1255. [Google Scholar]
  32. Litt, P.K.; Jaroni, D. Isolation and physiomorphological characterization of escherichia coli o157:h7-infecting bacteriophages recovered from beef cattle operations. Int. J. Microbiol. 2017, 2017, 7013236. [Google Scholar] [CrossRef] [Green Version]
  33. Silva, Y.J.; Costa, L.; Pereira, C.; Cunha, Â.; Calado, R.; Gomes, N.C.M.; Almeida, A. Influence of environmental variables in the efficiency of phage therapy in aquaculture. Microb. Biotechnol. 2014, 7, 401–413. [Google Scholar] [CrossRef] [PubMed]
  34. Moghimian, P.; Srot, V.; Pichon, B.P.; Facey, S.J.; van Aken, P.A. Stability of m13 phage in organic solvents. J. Biomater. Nanobiotechnol. 2016, 7, 72–77. [Google Scholar] [CrossRef] [Green Version]
  35. Jończyk, E.; Kłak, M.; Międzybrodzki, R.; Górski, A. The influence of external factors on bacteriophages—review. Folia Microbiol. 2011, 56, 191–200. [Google Scholar] [CrossRef] [Green Version]
  36. Łobocka, M.B.; Głowacka, A.; Golec, P. Methods for bacteriophage preservation. In Bacteriophage Therapy; Azaredo, J., Sillankorva, S., Eds.; Humana Press: New York, NY, USA, 2018; Volume 1693. [Google Scholar] [CrossRef]
  37. Ackermann, H.-W.; Tremblay, D.; Moineau, S. Long-term bacteriophage preservation. WFCC Newsl. 2004, 38, 35–40. [Google Scholar]
  38. Tovkach, F.; Zhuminska, G.; Khushkina, A. Long-term preservation of unstable bacteriophages of enterobacteria. Mīkrobiol. Zh. 2012, 74, 60–66. [Google Scholar]
  39. Leung, S.S.Y.; Parumasivam, T.; Gao, F.G.; Carter, E.A.; Carrigy, N.B.; Vehring, R.; Finlay, W.H.; Morales, S.; Britton, W.J.; Kutter, E.; et al. Effects of storage conditions on the stability of spray dried, inhalable bacteriophage powders. Int. J. Pharm. 2017, 521, 141–149. [Google Scholar] [CrossRef] [Green Version]
  40. Dini, C.; de Urraza, P.J. Effect of buffer systems and disaccharides concentration on podoviridae coliphage stability during freeze drying and storage. Cryobiology 2013, 66, 339–342. [Google Scholar] [CrossRef]
  41. Zhang, Y.; Peng, X.; Zhang, H.; Watts, A.B.; Ghosh, D. Manufacturing and ambient stability of shelf freeze dried bacteriophage powder formulations. Int. J. Pharm. 2018, 542, 1–7. [Google Scholar] [CrossRef]
  42. Singla, S.; Harjai, K.; Raza, K.; Wadhwa, S.; Katare, O.P.; Chhibber, S. Phospholipid vesicles encapsulated bacteriophage: A novel approach to enhance phage biodistribution. J. Virol. Methods 2016, 236, 68–76. [Google Scholar] [CrossRef]
  43. Costa, M.; Milho, C.; Teixeira, J.; Sillankova, S.; Cerqueira, M. Electrospun nanofibres as a novel encapsulation vehicle for felix o1 bacteriophage for new food packaging applications. IUFoST World Congr. Food Sci. Technol. 2018, 755, 23–27. [Google Scholar]
  44. Bean, J.E.; Alves, D.R.; Laabei, M.; Esteban, P.P.; Thet, N.T.; Enright, M.C.; Jenkins, A.T.A. Triggered Release of Bacteriophage K from Agarose/Hyaluronan Hydrogel Matrixes by Staphylococcus Aureus Virulence Factors. Chem. Mater. 2014, 26, 7201–7208. [Google Scholar] [CrossRef] [Green Version]
  45. Brown, T.L.; Thomas, T.; Odgers, J.; Petrovski, S.; Spark, M.J.; Tucci, J. Bacteriophage formulated into a range of semisolid and solid dosage forms maintain lytic capacity against isolated cutaneous and opportunistic oral bacteria. J. Pharm. Pharmacol. 2017, 69. [Google Scholar] [CrossRef]
  46. Ravera, F.; Dziza, K.; Santini, E.; Cristofolini, L.; Liggieri, L. Emulsification and emulsion stability: The role of the interfacial properties. Adv. Colloid Interface Sci. 2021, 288, 102344. [Google Scholar] [CrossRef] [PubMed]
  47. Jintapattanakit, A. Preparation of nanoemulsions by phase inversion temperature (pit). Pharm. Sci. Asia 2018, 42, 1–12. [Google Scholar] [CrossRef]
  48. Callender, S.P.; Mathews, J.A.; Kobernyk, K.; Wettig, S.D. Microemulsion utility in pharmaceuticals: Implications for multi-drug delivery. Int. J. Pharm. 2017, 526, 425–442. [Google Scholar] [CrossRef] [PubMed]
  49. Singh, Y.; Meher, J.G.; Raval, K.; Khan, F.A.; Chaurasia, M.; Jain, N.K.; Chourasia, M.K. Nanoemulsion: Concepts, development and applications in drug delivery. J. Control. Release 2017, 252, 28–49. [Google Scholar] [CrossRef]
  50. Otto, A.; du Plessis, J.; Wiechers, J.W. Formulation effects of topical emulsions on transdermal and dermal delivery. Int. J. Cosmet. Sci. 2009, 31. [Google Scholar] [CrossRef]
  51. Puapermpoonsiri, U.; Spencer, J.; van der Walle, C.F. A freeze-dried formulation of bacteriophage encapsulated in biodegradable microspheres. Eur. J. Pharm. Biopharm. 2009, 72, 26–33. [Google Scholar] [CrossRef] [PubMed]
  52. Balcao, V.M.; Azevedo, A.F.; Castro, C.I.; Santos, S.; Matos, C.M.; Moutinho, C.; Texeira, J.A.; Azaredo, J. Design of a lipid nanovesicle system encapsulating bacteriophages integrated in a multiple emulsion formulation: A proof-of-concept. NSTI Nanotechnol. Conf. Expo 2010, 459–462. [Google Scholar]
  53. Dini, C.; Islan, G.A.; de Urraza, P.J.; Castro, G.R. Novel biopolymer matrices for microencapsulation of phages: Enhanced protection against acidity and protease activity. Macromol. Biosci. 2012, 12, 1200–1208. [Google Scholar] [CrossRef] [PubMed]
  54. Esteban, P.P.; Jenkins, A.T.A.; Arnot, T.C. Elucidation of the mechanisms of action of bacteriophage k/nano-emulsion formulations against s. aureus via measurement of particle size and zeta potential. Colloids Surf. B Biointerfaces 2016, 139, 87–94. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Dao, H.; Lakhani, P.; Police, A.; Kallakunta, V.; Ajjarapu, S.S.; Wu, K.-W.; Ponkshe, P.; Repka, M.A.; Narasimha Murthy, S. Microbial stability of pharmaceutical and cosmetic products. AAPS PharmSciTech 2018, 19. [Google Scholar] [CrossRef] [PubMed]
  56. Nail, S.L.; Jiang, S.; Chongprasert, S.; Knopp, S.A. Fundamentals of freeze-drying. Pharm. Biotechnol. 2002, 14, 281–360. [Google Scholar] [CrossRef] [PubMed]
  57. Leung, S.S.Y.; Parumasivam, T.; Gao, F.G.; Carrigy, N.B.; Vehring, R.; Finlay, W.H.; Morales, S.; Britton, W.J.; Kutter, E.; Chan, H.-K. Production of inhalation phage powders using spray freeze drying and spray drying techniques for treatment of respiratory infections. Pharm. Res. 2016, 33, 1486–1496. [Google Scholar] [CrossRef] [PubMed]
  58. Liang, L.; Carrigy, N.B.; Kariuki, S.; Muturi, P.; Onsare, R.; Nagel, T.; Vehring, R.; Connerton, P.L.; Connerton, I.F. Development of a lyophilization process for campylobacter bacteriophage storage and transport. Microorganisms 2020, 8, 282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Gonzalez-Menendez, E.; Fernandez, L.; Gutierrez, D.; Rodríguez, A.; Martínez, B.; GarcíaI, P. Comparative analysis of different preservation techniques for the storage of staphylococcus phages aimed for the industrial development of phage-based antimicrobial products. PLoS ONE 2018, 13, e0205728. [Google Scholar] [CrossRef]
  60. Anany, H.; Chen, W.; Pelton, R.; Griffiths, M.W. Biocontrol of listeria monocytogenes and escherichia coli o157:h7 in meat by using phages immobilized on modified cellulose membranes. Appl. Environ. Microbiol. 2011, 77, 6379–6387. [Google Scholar] [CrossRef] [Green Version]
  61. Merabishvili, M.; Vervaet, C.; Pirnay, J.-P.; De Vos, D.; Verbeken, G.; Mast, J.; Chanishvili, N.; Vaneechoutte, M. Stability of staphylococcus aureus phage isp after freeze-drying (lyophilization). PLoS ONE 2013, 8, e68797. [Google Scholar] [CrossRef] [Green Version]
  62. Chang, R.Y.; Wong, J.; Mathai, A.; Morales, S.; Kutter, E.; Britton, W.; Li, J.; Chan, H.K. Production of highly stable spray dried phage formulations for treatment of pseudomonas aeruginosa lung infection. Eur. J. Pharm. Biopharm. 2017, 121, 1–13. [Google Scholar] [CrossRef]
  63. Ly, A.; Carrigy, N.B.; Wang, H.; Harrison, M.; Sauvageau, D.; Martin, A.R.; Vehring, R.; Finlay, W.H. Atmospheric spray freeze drying of sugar solution with phage d29. Front. Microbiol. 2019, 10, 488. [Google Scholar] [CrossRef]
  64. Calliste, C.A.; Trouillas, P.; Allais, D.P.; Simon, A.; Duroux, J.L. Free Radical scavenging activities measured by electron spin resonance spectroscopy and b16 cell antiproliferative behaviors of seven plants. J. Agric. Food Chem. 2001, 49, 3321–3327. [Google Scholar] [CrossRef]
  65. Malik, D.J. Bacteriophage encapsulation using spray drying for phage therapy. Curr. Issues Mol. Biol. 2021, 40, 303–316. [Google Scholar] [CrossRef] [PubMed]
  66. Leung, V.; Szewczyk, A.; Chau, J.; Hosseinidoust, Z.; Groves, L.; Hawsawi, H.; Anany, H.; Griffiths, M.W.; Ali, M.M.; Filipe, C.D.M. Long-term preservation of bacteriophage antimicrobials using sugar glasses. ACS Biomater. Sci. Eng. 2017, 4, 3802–3808. [Google Scholar] [CrossRef] [PubMed]
  67. Carrigy, N.B.; Liang, L.; Wang, H.; Kariuki, S.; Nagel, T.E.; Connerton, I.F.; Vehring, R. Trileucine and pullulan improve anti-campylobacter bacteriophage stability in engineered spray-dried microparticles. Ann. Biomed. Eng. 2020, 48, 1169–1180. [Google Scholar] [CrossRef]
  68. Pattni, B.S.; Chupin, V.V.; Torchilin, V.P. New developments in liposomal drug delivery. Chem. Rev. 2015, 115, 10938–10966. [Google Scholar] [CrossRef] [PubMed]
  69. Li, M.; Du, C.; Guo, N.; Teng, Y.; Meng, X.; Sun, H.; Li, S.; Yu, P.; Galons, H. Composition design and medical application of liposomes. Eur. J. Med. Chem. 2019, 164, 640–653. [Google Scholar] [CrossRef] [PubMed]
  70. Trucillo, P.; Campardelli, R.; Reverchon, E. Liposomes: From bangham to supercritical fluids. Processes 2020, 8, 1022. [Google Scholar] [CrossRef]
  71. Leung, S.S.Y.; Morales, S.; Britton, W.; Kutter, E.; Chan, H.-K. Microfluidic-assisted bacteriophage encapsulation into liposomes. Int. J. Pharm. 2018, 545, 176–182. [Google Scholar] [CrossRef]
  72. Cinquerrui, S.; Mancuso, F.; Vladisavljević, G.T.; Bakker, S.E.; Malik, D.J. Nanoencapsulation of bacteriophages in liposomes prepared using microfluidic hydrodynamic flow focusing. Front. Microbiol. 2018, 9, 2172. [Google Scholar] [CrossRef] [Green Version]
  73. Chadha, P.; Katare, O.P.; Chhibber, S. Liposome loaded phage cocktail: Enhanced therapeutic potential in resolving klebsiella pneumoniae mediated burn wound infections. Burns 2017, 43, 1532–1543. [Google Scholar] [CrossRef] [PubMed]
  74. Singla, S.; Harjai, K.; Katare, O.P.; Chhibber, S. Encapsulation of bacteriophage in liposome accentuates its entry in to macrophage and shields it from neutralizing antibodies. PLoS ONE 2016, 11, e0153777. [Google Scholar] [CrossRef] [Green Version]
  75. Colom, J.; Cano-Sarabia, M.; Otero, J.; Cortés, P.; Maspoch, D.; Llagostera, M. Liposome-encapsulated bacteriophages for enhanced oral phage therapy against salmonella spp. Appl. Environ. Microbiol. 2015, 81, 4841–4849. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Bhardwaj, N.; Kundu, S.C. Electrospinning: A fascinating fiber fabrication technique. Biotechnol. Adv. 2010, 28, 325–347. [Google Scholar] [CrossRef] [PubMed]
  77. Salalha, W.; Kuhn, J.; Dror, Y.; Zussman, E. Encapsulation of bacteria and viruses in electrospun nanofibres. Nanotechnology 2006, 17, 4675–4681. [Google Scholar] [CrossRef]
  78. Korehei, R.; Kadla, J. Incorporation of T4 bacteriophage in electrospun fibres. J. Appl. Microbiol. 2013, 114, 1425–1434. [Google Scholar] [CrossRef] [PubMed]
  79. Korehei, R.; Kadla, J.F. Encapsulation of T4 bacteriophage in electrospun poly(ethylene oxide)/cellulose diacetate fibers. Carbohydr. Polym. 2014, 100, 150–157. [Google Scholar] [CrossRef]
  80. Dai, M.; Senecal, A.; Nugen, S.R. Electrospun water-soluble polymer nanofibers for the dehydration and storage of sensitive reagents. Nanotechnology 2014, 25. [Google Scholar] [CrossRef]
  81. Díaz, A.; del Valle, L.; Rodrigo, N.; Casas, M.; Chumburidze, G.; Katsarava, R.; Puiggalí, J. Antimicrobial activity of poly(ester urea) electrospun fibers loaded with bacteriophages. Fibers 2018, 6, 33. [Google Scholar] [CrossRef] [Green Version]
  82. Andriolo, J.M.; Sutton, N.J.; Murphy, J.P.; Huston, L.G.; Kooistra-Manning, E.A.; West, R.F.; Pedulla, M.L.; Hailer, M.K.; Skinner, J.L. Electrospun fibers for controlled release of nanoparticle-assisted phage therapy treatment of topical wounds. MRS Adv. 2018, 3. [Google Scholar] [CrossRef]
  83. Jabrane, T.; Dubé, M.; Griffiths, M.; Mangin, P.J. Towards a Commercial Production of Phage-Based Bioactive Paper. J. Sci. Technol. For. Prod. Process 2011, 1, 6–13. [Google Scholar]
  84. Olsson, A.L.J.; Wargenau, A.; Tufenkji, N. Optimizing bacteriophage surface densities for bacterial capture and sensing in quartz crystal microbalance with dissipation monitoring. ACS Appl. Mater. Interfaces 2016, 8, 13698–13706. [Google Scholar] [CrossRef] [PubMed]
  85. Richter, Ł.; Bielec, K.; Leśniewski, A.; Łoś, M.; Paczesny, J.; Hołyst, R. Dense layer of bacteriophages ordered in alternating electric field and immobilized by surface chemical modification as sensing element for bacteria detection. ACS Appl. Mater. Interfaces 2017, 9, 19622–19629. [Google Scholar] [CrossRef]
  86. Tolba, M.; Minikh, O.; Brovko, L.Y.; Evoy, S.; Griffiths, M.W. Oriented immobilization of bacteriophages for biosensor applications. Appl. Environ. Microbiol. 2010, 76, 528–535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Sorokulova, I.; Olsen, E.; Vodyanoy, V. Bacteriophage biosensors for antibiotic-resistant bacteria. Expert Rev. Med. Dev. 2014, 11, 175–186. [Google Scholar] [CrossRef]
  88. Farooq, U.; Yang, Q.; Ullah, M.W.; Wang, S. Bacterial biosensing: Recent advances in phage-based bioassays and biosensors. Biosens. Bioelectron. 2018, 118, 204–216. [Google Scholar] [CrossRef]
  89. Singh, A.; Glass, N.; Tolba, M.; Brovko, L.; Griffiths, M.; Evoy, S. Immobilization of bacteriophages on gold surfaces for the specific capture of pathogens. Biosens. Bioelectron. 2009, 24, 3645–3651. [Google Scholar] [CrossRef]
  90. Vonasek, E.; Lu, P.; Hsieh, Y.L.; Nitin, N. Bacteriophages immobilized on electrospun cellulose microfibers by non-specific adsorption, protein–ligand binding, and electrostatic interactions. Cellulose 2017, 24, 4581–4589. [Google Scholar] [CrossRef]
  91. Janczuk, M.; Richter, Ł.; Hoser, G.; Kawiak, J.; Łoś, M.; Niedziółka-Jönsson, J.; Paczesny, J.; Hołyst, R. Bacteriophage-based bioconjugates as a flow cytometry probe for fast bacteria detection. Bioconjug Chem. 2017, 28, 419–425. [Google Scholar] [CrossRef]
  92. Ashiani, D.; Keihan, A.H.; Rashidiani, J.; Dashtestani, F.; Eskandari, K. Oriented t4 bacteriophage immobilization for recognition of escherichia coli in capacitance method. Int. J. Electrochem. Sci. 2016, 11, 10087–10095. [Google Scholar] [CrossRef]
  93. Hosseinidoust, Z.; Van De Ven, T.G.M.; Tufenkji, N. Bacterial capture efficiency and antimicrobial activity of phage-functionalized model surfaces. Langmuir 2011, 27, 5472–5480. [Google Scholar] [CrossRef]
  94. Naidoo, R.; Singh, A.; Arya, S.K.; Beadle, B.; Glass, N.; Tanha, J.; Szymanski, C.M.; Evoy, S. Surface-Immobilization of chromatographically purified bacteriophages for the optimized capture of bacteria. Bacteriophage 2012, 2, 15–24. [Google Scholar] [CrossRef] [Green Version]
  95. Cerofolini, G.F.; Meda, L.; Bandosz, T.J. Adsorption with soft adsorbents or adsorbates. theory and practice. In Adsorption and its Applications in Industry and Environmental Protection Studies in Surface Science and Catalysis; Dabrawski, A., Ed.; Elsevier: Amsterdam, The Netherland, 1998; Volume 120, pp. 227–272. [Google Scholar] [CrossRef]
  96. Bennett, A.R.; Davids, F.G.C.; Vlahodimou, S.; Banks, J.G.; Betts, R.P. The use of bacteriophage-based systems for the separation and concentration of salmonella. J. Appl. Microbiol. 1997, 83, 259–265. [Google Scholar] [CrossRef] [PubMed]
  97. Smietana, M.; Bock, W.J.; Mikulic, P.; Ng, A.; Chinnappan, R.; Zourob, M.; Zourob, M.; Mohr, S.; Brown, B.J.; Fielden, P.R.; et al. Detection of bacteria using bacteriophages as recognition elements immobilized on long-period fiber gratings. Sens. Actuators B Chem. 2005, 107, 202–208. [Google Scholar] [CrossRef] [PubMed]
  98. Abul-Hassan, H.S.; El-Tahan k Massoud, B.; Gomaa, R. Bacteriophage Therapy of Pseudomonas Burn Wound Sepsis. Ann. MBC 1991, 3, 4. [Google Scholar]
  99. Kifelew, L.G.; Warner, M.S.; Morales, S.; Vaughan, L.; Woodman, R.; Fitridge, R.; Mitchell, J.G.; Speck, P. Efficacy of Phage Cocktail AB-SA01 Therapy in Diabetic Mouse Wound Infections Caused by Multidrug-Resistant Staphylococcus Aureus. BMC Microbiol. 2020, 20. [Google Scholar] [CrossRef] [PubMed]
  100. Hirsh, S.L.; Bilek, M.M.M.; Nosworthy, N.J.; Kondyurin, A.; Dos Remedios, C.G.; McKenzie, D.R. A Comparison of covalent immobilization and physical adsorption of a cellulase enzyme mixture. Langmuir 2010, 26, 14380–14388. [Google Scholar] [CrossRef]
  101. Szot-Karpińska, K.; Leśniewski, A.; Jönsson-Niedziółka, M.; Marken, F.; Niedziółka-Jönsson, J. Electrodes modified with bacteriophages and carbon nanofibres for cysteine detection. Sens. Actuators B Chem. 2019, 287, 78–85. [Google Scholar] [CrossRef]
  102. Guntupalli, R.; Sorokulova, I.; Olsen, E.; Globa, L.; Pustovyy, O.; Vodyanoy, V. Biosensor for detection of antibiotic resistant staphylococcus bacteria. J. Vis. Exp. 2013, 75, e50474. [Google Scholar] [CrossRef] [Green Version]
  103. Guntupalli, R.; Sorokulova, I.; Krumnow, A.; Pustovyy, O.; Olsen, E.; Vodyanoy, V. Real-time optical detection of methicillin-resistant staphylococcus aureus using lytic phage probes. Biosens. Bioelectron. 2008, 24, 151–154. [Google Scholar] [CrossRef] [PubMed]
  104. Van Voorthuizen, E.M.; Ashbolt, N.J.; Schäfer, A.I. Role of hydrophobic and electrostatic interactions for initial enteric virus retention by mf membranes. J. Memb. Sci. 2001, 194, 69–79. [Google Scholar] [CrossRef] [Green Version]
  105. Farkas, K.; Varsani, A.; Pang, L. Adsorption of Rotavirus, MS2 Bacteriophage and surface-modified silica nanoparticles to hydrophobic matter. Food Environ. Virol. 2015, 7, 261–268. [Google Scholar] [CrossRef]
  106. Pinto, F.; Maillard, J.Y.; Denyer, S.P.; McGeechan, P. Polyhexamethylene biguanide exposure leads to viral aggregation. J. Appl. Microbiol. 2010, 108, 1880–1888. [Google Scholar] [CrossRef] [PubMed]
  107. Rosenberg, M. Microbial adhesion to hydrocarbons: Twenty-five years of doing MATH. FEMS Microbiol. Lett. 2006, 262, 129–134. [Google Scholar] [CrossRef] [PubMed]
  108. Aronino, R.; Dlugy, C.; Arkhangelsky, E.; Shandalov, S.; Oron, G.; Brenner, A.; Gitis, V. Removal of viruses from surface water and secondary effluents by sand filtration. Water Res. 2009, 43, 87–96. [Google Scholar] [CrossRef]
  109. Bales, R.C.; Li, S.; Maguire, K.M.; Yahya, M.T.; Gerba, C.P. MS-2 and poliovirus transport in porous media: Hydrophobic effects and chemical perturbations. Water Resour. Res. 1993, 29, 957–963. [Google Scholar] [CrossRef]
  110. Han, J.; Jin, Y.; Willson, C.S. Virus Retention and transport in chemically heterogeneous porous media under saturated and unsaturated flow conditions. Environ. Sci. Technol. 2006, 40, 1547–1555. [Google Scholar] [CrossRef]
  111. Wu, S.; Lan, X.; Zhao, W.; Li, Y.; Zhang, L.; Wang, H.; Han, M.; Tao, S. Biosensors and bioelectronics controlled immobilization of acetylcholinesterase on improved hydrophobic gold nanoparticle/prussian blue modified surface for ultra-trace organophosphate pesticide detection. Biosens. Bioelectron. 2011, 27, 82–87. [Google Scholar] [CrossRef]
  112. Su, P.; Li, H. Energy decomposition analysis of covalent bonds and intermolecular interactions. J. Chem. Phys. 2009, 131, 014102. [Google Scholar] [CrossRef] [Green Version]
  113. Tawil, N.; Sacher, E.; Mandeville, R.; Meunier, M. Strategies for the immobilization of bacteriophages on gold surfaces monitored by surface plasmon resonance and surface morphology. J. Phys. Chem. C 2013, 117, 6689–6691. [Google Scholar] [CrossRef]
  114. Jabrane, T.; Dubé, M.; Mangin, P.J. Bacteriophage immobilization on paper surface: Effect of cationic pre-coat layer. In Proceedings of the Canadian PAPTAC 95th Annual Meeting, Montreal, QC, Canada, 3–4 February 2009. [Google Scholar]
  115. Mangin, P.J.; Jabrane, T.; Jeaidi, J.; Dubé, M. Gravure printing of enzymes and phages. Adv. Print. Media Technol. 2008, 35, 351–358. [Google Scholar]
  116. Cademartiri, R.; Anany, H.; Gross, I.; Bhayani, R.; Griffiths, M.; Brook, M.A. Immobilization of bacteriophages on modified silica particles. Biomaterials 2010, 31, 1904–1910. [Google Scholar] [CrossRef]
  117. Liana, A.E.; Marquis, C.P.; Gunawan, C.; Gooding, J.J.; Amal, R. T4 bacteriophage conjugated magnetic particles for e. coli capturing: Influence of bacteriophage loading, temperature and tryptone. Colloids Surf. B Biointerfaces 2017, 151, 47–57. [Google Scholar] [CrossRef]
  118. Zhou, Y.; Marar, A.; Kner, P.; Ramasamy, R.P. Charge-directed immobilization of bacteriophage on nanostructured electrode for whole-cell electrochemical biosensors. Anal. Chem. 2017, 89, 5734–5741. [Google Scholar] [CrossRef]
  119. Peng, B.; Liu, N.; Lin, Y.; Wang, L.; Zhang, W.; Niu, Z.; Wang, Q.; Su, Z. Self-assembly of anisotropic tobacco mosaic virus nanoparticles on gold substrate. Sci. China Chem. 2011, 54, 137–143. [Google Scholar] [CrossRef]
  120. Tytgat, H.L.P.; Schoofs, G.; Driesen, M.; Proost, P.; Van Damme, E.J.M.; Vanderleyden, J.; Lebeer, S. Endogenous biotin-binding proteins: An overlooked factor causing false positives in streptavidin-based protein detection. Microb. Biotechnol. 2015, 8, 164–168. [Google Scholar] [CrossRef]
  121. Houk, K.N.; Leach, A.G.; Kim, S.P.; Zhang, X. Binding affinities of host-guest, protein-ligand, and protein-transition-state complexes. Angewandye Chem. 2003, 42, 4872–4897. [Google Scholar] [CrossRef]
  122. Gervais, L.; Gel, M.; Allain, B.; Tolba, M.; Brovko, L.; Zourob, M.; Mandeville, R.; Griffiths, M.; Evoy, S. Immobilization of biotinylated bacteriophages on biosensor surfaces. Sens. Actuators B Chem. 2007, 125, 615–621. [Google Scholar] [CrossRef]
  123. Horikawa, S.; Bedi, D.; Li, S.; Shen, W.; Huang, S.; Chen, I.H.; Chai, Y.; Auad, M.L.; Bozack, M.J.; Barbaree, J.M.; et al. Effects of surface functionalization on the surface phage coverage and the subsequent performance of phage-immobilized magnetoelastic biosensors. Biosens. Bioelectron. 2011, 26, 2361–2367. [Google Scholar] [CrossRef]
  124. Ong, E.; Gilkes, N.R.; Antony, R.; Warren, J.; Miller, R.C.; Kilburn, D.G. Enzyme immobilization using the cellulose-binding domain of a cellulomonas fimiexoglucanase. Bio/Technology 1989, 7, 604–607. [Google Scholar] [CrossRef] [Green Version]
  125. DeChancie, J.; Houk, K.N. Biotin-streptavidin binding constant. J. Am. Chem. Soc. 2007, 129, 5419–5429. [Google Scholar] [CrossRef] [Green Version]
  126. Sassolas, A.; Blum, L.J.; Leca-Bouvier, B.D. Immobilization strategies to develop enzymatic biosensors. Biotechnol. Adv. 2012. [Google Scholar] [CrossRef]
  127. Tran, C.T.H.; Nosworthy, N.; Bilek, M.M.M.; McKenzie, D.R. Covalent immobilization of enzymes and yeast: Towards a continuous simultaneous saccharification and fermentation process for cellulosic ethanol. Biomass Bioenergy 2015, 81, 234–241. [Google Scholar] [CrossRef]
  128. Choi, I.; Yoo, D.S.; Chang, Y.; Kim, S.Y.; Han, J. Polycaprolactone film functionalized with bacteriophage t4 promotes antibacterial activity of food packaging toward escherichia coli. Food Chem. 2021, 346, 128883. [Google Scholar] [CrossRef]
  129. Lee, L.A.; Wang, Q. Adaptations of nanoscale viruses and other protein cages for medical applications. Nanomed. Nanotechnol. Biol. Med. 2006, 2, 137–149. [Google Scholar] [CrossRef] [PubMed]
  130. Chen, P.; Konarov, A.; Dong, D.; Zhang, Y.; Sutaria, S. Binding mechanism and electrochemical properties of m13 phage-sulfur composite. PLoS ONE 2013, 8, e82332. [Google Scholar] [CrossRef]
  131. Niyomdecha, S.; Limbut, W.; Numnuam, A.; Kanatharana, P.; Charlermroj, R.; Karoonuthaisiri, N.; Thavarungkul, P. Phage-based capacitive biosensor for salmonella detection. Talanta 2018, 188, 658–664. [Google Scholar] [CrossRef]
  132. Pearson, H.A.; Sahukhal, G.S.; Elasri, M.O.; Urban, M.W. Phage-bacterium war on polymeric surfaces: Can surface-anchored bacteriophages eliminate microbial infections? Biomacromolecules 2013, 14, 1257–1261. [Google Scholar] [CrossRef] [Green Version]
  133. Nogueira, F.; Karumidze, N.; Kusradze, I.; Goderdzishvili, M.; Teixeira, P.; Gouveia, I.C. Immobilization of bacteriophage in wound-dressing nanostructure. Nanomed. Nanotechnol. Biol. Med. 2017, 13, 2475–2484. [Google Scholar] [CrossRef] [Green Version]
  134. Bitterncourt, J.A. Fundamentals of Plasma Physics, 3rd ed.; Springer: New York, NY, USA, 2004. [Google Scholar] [CrossRef] [Green Version]
  135. Collaud, M.; Groening, P.; Nowak, S.; Schlapbach, L. Plasma treatment of polymers: The effect of the plasma parameters on the chemical, physical, and morphological states of the polymer surface and on the metal-polymer interface. J. Adhes. Sci. Technol. 1994, 8, 1115–1127. [Google Scholar] [CrossRef]
  136. Favia, P.; D’Agostino, R. Plasma treatments and plasma deposition of polymers for biomedical applications. Surf. Coatings Technol. 1998, 98, 1102–1106. [Google Scholar] [CrossRef]
  137. Gao, R.; Wang, Y.; Tong, J.; Zhou, P.; Yang, Z. Strategies for the Immobilization of Bacteriophages Applied in the Biosensors. In Proceedings 2015 International Conference on Computational Intelligence and Communication Networks, CICN 2015; Institute of Electrical and Electronics Engineers Inc.: New York, NY, USA, 2016; pp. 178–182. [Google Scholar] [CrossRef]
  138. Bilek, M.M.; McKenzie, D.R. Plasma modified surfaces for covalent immobilization of functional biomolecules in the absence of chemical linkers: Towards better biosensors and a new generation of medical implants. Biophys. Rev. 2010, 2, 55–65. [Google Scholar] [CrossRef] [Green Version]
  139. Yeo, G.C.; Kondyurin, A.; Kosobrodova, E.; Weiss, A.S.; Bilek, M.M.M. A sterilizable, biocompatible, tropoelastin surface coating immobilized by energetic ion activation. J. R. Soc. Interface 2017, 14. [Google Scholar] [CrossRef] [Green Version]
  140. Hugh, S.; Mattey, M. Immobilisation and Stabilisation of Virus. U.S. Patent 7,482,115 B2, 27 January 2009. [Google Scholar]
  141. Mattey, M.; Bell, E. Treatment of Topical and Systemic Bacterial Infections. U.S. Patent 2021/0038658 A1, 11 February 2021. [Google Scholar]
  142. Mattey, M. Treatment of Bacterial Infections in Aquaculture. U.S. Patent 10,849,942 B2, 1 December 2020. [Google Scholar]
  143. Fixed Phage Ltd. Case Studies. Available online: https://www.fixed-phage.com/case-studies/ (accessed on 10 March 2021).
Table 1. Examples of the various encapsulation approaches that have been carried out with bacteriophages.
Table 1. Examples of the various encapsulation approaches that have been carried out with bacteriophages.
Encapsulation MethodBacteriophage (Host Genus)FormulationObservationsReference
EmulsificationK (Staphylococcus)Semi-solidUp to 10 days of activity at 20 °C[27]
Freeze-DryingM13 (Escherichia)Powder<1 log drop in titer after 2 months at ambient temperature[41]
Spray-DryingPEV2, PEV40 (Pseudomonas)Powder<1 log drop in titer after 1 year at 20 °C[39]
Liposome EntrapmentKP01K2 (Klebsiella)LiquidUp to 14 days of activity in vivo[42]
ElectrospinningFelix O1 (Salmonella)NanofibersPhage activity of equivalent to 105–106 PFU/mL after fiber preparation[43]
Table 2. Summary of the benefits and limitations associated with the mass production of encapsulated therapeutic phage formulations.
Table 2. Summary of the benefits and limitations associated with the mass production of encapsulated therapeutic phage formulations.
Encapsulation MethodBenefitsLimitations
EmulsificationMaterial produced ideal for cream-type treatments
Promote absorption when applied topically
Difficult to transport/store at large scale
Prone to bacterial contamination
Only stable when refrigerated
Freeze-DryingFinal product easy to store/transport
High stability post-production
Variety of applications
Time-consuming, costly process
Ice crystal formation can decrease phage viability
Spray-DryingFinal product easy to store/transport
High stability post-production
Variety of applications
Energy-consuming process
Temperature can decrease phage viability during process
Liposome EntrapmentProtection of phages against in vivo conditions
Extensive studies demonstrating therapeutic effect compared free phage
Encapsulation yield of phages in liposomes difficult to control
Difficult to transport/store at large scaleOnly stable when refrigerated
ElectrospinningDiverse array of materials can be produced.
Easy deposition of fiber-encapsulated phage onto other substrates
Fiber-spinning process can damage phages
Table 3. Examples of studies involving immobilization of bacteriophages onto surfaces.
Table 3. Examples of studies involving immobilization of bacteriophages onto surfaces.
Immobilization ApproachBacteriophage
(Host Genus)
SurfaceObservationsReference
Physical AdsorptionT4 (Escherichia)Gold surface modified with cysteine and glutaraldehydePhage surface concentration of 18 ± 0.15 phages per um2[89]
Protein-LigandT4 (Escherichia)Magnetic beads, microcrystalline cellulose beadsUp to 81% improved binding efficiency compared to physical adsorption[86]
ElectrostaticT7 (Escherichia)Cellulose microfibers15–25% phage loading efficiency on surface[90]
Covalent LinkageAG10 (Escherichia)
CG4 (Salmonella)
Magnetic-fluorescent beadsPhage activity equivalent to 108 PFU/mL observed in material[91]
Table 4. Summary of benefits and limitations associated with various bacteriophage immobilization techniques for the production of therapeutic phage formulations.
Table 4. Summary of benefits and limitations associated with various bacteriophage immobilization techniques for the production of therapeutic phage formulations.
Immobilization ApproachBenefitsLimitations
Physical AdsorptionSimple process
Inexpensive
Undirected, inconsistent
Phage not strongly bound to substrate
Protein-LigandStrongly bound phage
High binding efficiency
Tail-up orientation
Complicated process
Expensive
Electrostatic High binding efficiency
Applicable to most tailed phages
Tail-up Orientation
Electrostatically charged surface may not be desirable
Covalent LinkageStrongly bound phage
Potentially longer shelf life
Can be a costly and complex process (in the case of linker-based immobilization)
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Rosner, D.; Clark, J. Formulations for Bacteriophage Therapy and the Potential Uses of Immobilization. Pharmaceuticals 2021, 14, 359. https://doi.org/10.3390/ph14040359

AMA Style

Rosner D, Clark J. Formulations for Bacteriophage Therapy and the Potential Uses of Immobilization. Pharmaceuticals. 2021; 14(4):359. https://doi.org/10.3390/ph14040359

Chicago/Turabian Style

Rosner, Daniel, and Jason Clark. 2021. "Formulations for Bacteriophage Therapy and the Potential Uses of Immobilization" Pharmaceuticals 14, no. 4: 359. https://doi.org/10.3390/ph14040359

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

Article Metrics

Back to TopTop