Next Article in Journal
The Antimicrobial Photoinactivation Effect on Escherichia coli through the Action of Inverted Cationic Porphyrin–Cyclodextrin Conjugates
Previous Article in Journal
Pseudomonas aeruginosa PAO1 Is Attracted to Bovine Bile in a Novel, Cystic Fibrosis-Derived Bronchial Epithelial Cell Model
Previous Article in Special Issue
Exploration of Bacterial Re-Growth as In Vitro Phenomenon Affecting Methods for Analysis of the Antimicrobial Activity of Chimeric Bacteriophage Endolysins
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Essential Topics for the Regulatory Consideration of Phages as Clinically Valuable Therapeutic Agents: A Perspective from Spain

by
Roberto Vázquez
1,†,
Roberto Díez-Martínez
2,†,
Pilar Domingo-Calap
3,†,
Pedro García
4,†,
Diana Gutiérrez
2,†,
Maite Muniesa
5,†,
María Ruiz-Ruigómez
6,†,
Rafael Sanjuán
3,†,
María Tomás
7,8,9,10,†,
María Ángeles Tormo-Mas
11,† and
Pilar García
12,13,*,†
1
Department of Biotechnology, Ghent University, 9000 Ghent, Belgium
2
Telum Therapeutics SL, 31110 Noáin, Spain
3
Institute for Integrative Systems Biology, University of Valencia-CSIC, 46980 Paterna, Spain
4
Center for Biological Research Margarita Salas (CIB-CSIC) and Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), 28040 Madrid, Spain
5
Department of Genetics, Microbiology and Statistics, University of Barcelona, 08028 Barcelona, Spain
6
Internal Medicine, Infectious Diseases Unit, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
7
Department of Microbiology, Hospital Universitario de A Coruña (INIBIC-CHUAC, SERGAS), 15006 A Coruña, Spain
8
Study Group on Mechanisms of Action and Resistance to Antimicrobials (GEMARA) on behalf of the Spanish Society of Infectious Diseases and Clinical Microbiology (SEIMC), 28003 Madrid, Spain
9
Spanish Network for Research in Infectious Diseases (REIPI), 41071 Sevilla, Spain
10
Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain
11
Severe Infection Group, Hospital Universitari i Politècnic La Fe, Health Research Institute Hospital La Fe, IISLaFe, 46026 Valencia, Spain
12
Dairy Research Institute of Asturias, IPLA-CSIC, 33300 Villaviciosa, Spain
13
DairySafe Group, Health Research Institute of Asturias (ISPA), 33011 Oviedo, Spain
*
Author to whom correspondence should be addressed.
Phage Therapy Work Group, Spanish Network of Bacteriophages and Transducing Elements (FAGOMA).
Microorganisms 2022, 10(4), 717; https://doi.org/10.3390/microorganisms10040717
Submission received: 3 March 2022 / Revised: 22 March 2022 / Accepted: 24 March 2022 / Published: 26 March 2022

Abstract

:
Antibiotic resistance is one of the major challenges that humankind shall face in the short term. (Bacterio)phage therapy is a valuable therapeutic alternative to antibiotics and, although the concept is almost as old as the discovery of phages, its wide application was hindered in the West by the discovery and development of antibiotics in the mid-twentieth century. However, research on phage therapy is currently experiencing a renaissance due to the antimicrobial resistance problem. Some countries are already adopting new ad hoc regulations to favor the short-term implantation of phage therapy in clinical practice. In this regard, the Phage Therapy Work Group from FAGOMA (Spanish Network of Bacteriophages and Transducing Elements) recently contacted the Spanish Drugs and Medical Devices Agency (AEMPS) to promote the regulation of phage therapy in Spain. As a result, FAGOMA was asked to provide a general view on key issues regarding phage therapy legislation. This review comes as the culmination of the FAGOMA initiative and aims at appropriately informing the regulatory debate on phage therapy.

1. General Aspects of the Use of Phages as Antimicrobials

The emergence of multi-drug-resistant (MDR) bacteria seriously undermines our ability to control bacterial infectious diseases. A recent study shows that MDR pathogens already cause more than one million deaths a year, and the prospects are even more concerning [1,2]. Therefore, short-term proactive measures are urgently needed. Proposed strategies include controlling the spread of antibiotic resistance, designing new antibiotics, and developing alternative therapies. Bacteriophages (phages), bacterial viruses, were discovered in the second decade of the 20th century and soon used to treat bacterial infectious diseases with encouraging results [3]. However, phage therapy was abandoned due to the discovery of antibiotics, with the exception of the Soviet Union and some Eastern European countries (due to hindered access to antibiotics there). Recently, and in the face of increasing bacterial resistance to antibiotics, phages have re-emerged as alternative or complementary therapies to control bacterial infections [4]. The advantages and disadvantages of phage therapy are discussed below.

1.1. Advantages

(a)
Activity against antibiotic-resistant bacteria. Phages can infect and kill bacteria, including MDR strains [5]. This is the most obvious advantage towards recovering phage therapy to fight antimicrobial resistance today. Moreover, the composition of the phages for therapy may be designed to impose an evolutionary trade-off in which the evolution of bacterial resistance to phage results in increased susceptibility to antibiotics [6]. Under this rationale, the combination therapy of phages plus antibiotics has a remarkable potential to smartly tackle antibiotic resistance by both eliminating resistant strains and preventing the dissemination of resistance genes [7,8]. Many works have been published thus far where phage–antibiotic synergy, known as PAS effect, is reported. This suggests that combined therapy can be the safest and a more advantageous approach, as it minimizes resistance and virulence [9,10,11].
(b)
Specificity. The high specificity of many phages towards their host bacterial strains makes them a highly selective therapy that prevents the dysbiosis of the healthy microbiota. Contrary to antibiotics, only strains of the same genus or species—and often just one or very few strains within a species—are susceptible to infection by a given phage, protecting the normal microbiota and reducing side effects [12]. The specificity of phages lies in the bacterial receptor that the virions recognize through one or more receptor-binding proteins [13], and can be further driven by post-entry anti-phage defense mechanisms [14]. Phages can be polyvalent (i.e., with a broad host range) if they recognize a receptor present in several bacteria or, alternatively, their specificity can be very restricted if they bind receptors exclusive to a single bacterial type. Another possibility is that the phage uses a receptor that is only expressed under certain conditions which therefore restricts its infectivity (e.g., the receptor of phage lambda is the maltose receptor, which is only expressed in the presence of maltose [15]).
(c)
Multiplication at the site of the infection (auto-dosage). Phages can multiply at the site of the infection. Once the phages reach the targeted bacteria, they will replicate and generate progeny. Therefore, if sufficient phage particles are able to reach the infection site, phage therapy can be considered as auto-dosage treatment. Furthermore, once the infection is successfully controlled, phages would be eliminated in the absence of bacterial hosts. Thus, whenever an auto-dosed, “active” treatment is achieved, it can elicit the infection eradication by only a single administration [16].
(d)
Ubiquity and diversity. Phages can be found in virtually any environment [17], and they play an utmost important role in ecosystems by regulating bacterial populations [18], including the human microbiota. The main practical consequence of such ubiquity and the concomitant diversity is the ease of discovery of novel phages, which contrasts with the currently slow antibiotic discovery rate.
(e)
Evolvability. Phages are evolving entities and, therefore, can be optimized using directed evolution techniques. This opens up many possibilities compared to conventional treatments, which are stable chemical compounds. Phage evolvability can be exploited in many ways, such as increasing lytic capability, improving particle stability, expanding the host range, or counteracting bacterial resistance. For instance, the Appelmans’ protocol uses spontaneous mutation and recombination among phages present in a cocktail to produce phage variants capable of infecting initially non-susceptible bacterial strains [19,20].
(f)
Safety. Humans are carriers of many different phages forming the phageome [21,22]. Their biological functions, beyond regulating bacterial populations, are not yet entirely clear [23]. However, their widespread presence in the human body seems to be a good safety indicator. Moreover, there is evidence of phage safety from clinical trials and intake of phage-treated foods [24]. A potential concern of phage therapy could be the release of bacterial endotoxins after lysis of the targeted bacterial cells. It should be noted, however, that similar observations have been made regarding conventional therapy with certain antibiotics [16], and that the current literature does not support detrimental inflammatory reactions upon phage administration. Phages may also enter tissues that are not the specific target of the treatment, but these interactions also do not appear to produce side effects [25].

1.2. Weaknesses

(a)
Phage-resistance. In the same way that resistance to antibiotics emerges, bacteria can become resistant to phage infection. The most common solution to address this involves the use of cocktails of different phages, rather than a single phage, and/or the “à la carte” selection of phages for each particular infectious isolate. This makes it much less likely that the host will become resistant to all phages at the same time [26]. The so-called “step-by-step” technique is an interesting approach in which phages are isolated against phage-resistant bacterial mutants in successive screening steps to obtain other phages capable of infecting resistant variants. By this method, the natural antagonistic co-evolution that would occur upon treatment is mimicked prior to therapy, thus generating a phage cocktail able to infect both the original bacterium and the foreseeable resistant variants [27]. Moreover, the emergence of phage-resistant bacteria is not always a disadvantage, since it sometimes involves a decrease of the fitness or virulence of the bacterial host [28], or may resensitize bacteria to antibiotics [9].
(b)
Specificity. This feature can be a double-edged sword. Phage specificity requires careful susceptibility testing of each bacterial pathogen before treatment, which may be viewed as an issue for certain acute infections that require urgent action. In addition, this specificity may require either the development of large phage libraries and/or extensive sampling and screening efforts to provide sufficient coverage of bacterial diversity. This can be a daunting task and has posed major regulatory issues, since, according to the current framework, each individual phage should undergo review and approval. In addition, the eventual need to develop a different phage preparation for each bacterial pathogen, as a personalized medicine, reduces business profitability and can be viewed as a serious drawback by pharmaceutical companies. Again, phage cocktails targeting different receptors or different bacterial strains would be a potential solution.
(c)
In vivo phage activity. There is not necessarily a correlation between the in vitro and in vivo behavior of a phage, particularly regarding its propagation ability. This is due both to the complexity of body fluids and the ecological in vivo interactions [29,30]. In addition, the phage propagation is dependent on the physiological state of the bacterial host, which may not be optimal for infection in vivo (for example, depending on whether the bacterium is embedded or not within a biofilm, the expression of receptor molecules, etc.) [31]. Moreover, phages are bigger than antibiotics and, therefore, diffuse less efficiently. This limitation is aggravated in vivo, where multiple physical barriers are encountered. Therefore, the probability of infection at low phage and bacterial densities is low, and the threshold densities required to ensure phage infection may often require the administration of very high phage doses [32,33].
(d)
Immune response. Since phages are made of biomacromolecules, they are potentially capable of eliciting an immune reaction upon administration [34]. Generally, the immune reactions against phage components are not considered problematic for the individual under treatment, although they do have a relevant contribution to the outcome of phage therapy [35]. On one hand, the immune response potentially causes the removal of phages from the system [36], although this effect may be overcome by adjusting such parameters as dosing, administration route, etc. On the other hand, synergism with the immune antibacterial response seems relevant for therapeutic success [37], although some evidence suggests that phage therapy can also be successfully applied in immunocompromised patients [38]. To sum up, the interaction of phages with the immune system is complex and not yet well understood, although many unknown implications seem to affect the therapeutic efficacy without contradicting the presumed safety of phage therapy.
(e)
Gene transfer. Phages potentially have the ability to modify the genome of the host bacteria, which may increase their virulence or dissemination of antibiotic resistance genes [39]. Indeed, phages can mobilize large fragments of bacterial genomes at relatively high frequencies [40,41]. To date, it is not known whether the mobilized DNA is randomly selected or whether the transfer of some particular genes is favored, e.g., those related to virulence, survival, or fitness of the host strain. A relevant mechanism for phage-mediated transfer of particular genes is associated with lysogeny (the integration of the phage genetic material into the bacterial chromosome) [42]. Therefore, this issue could be minimized by selecting exclusively virulent phages, as well as by analyzing phage genomes in detail to ensure that they do not contain genes encoding toxins or any other undesirable genes.

2. Obtaining Therapeutic Phage Preparations

The main steps for obtaining phage suspensions suitable for use in clinical settings are summarized in Figure 1.

2.1. Selection of Screening Host Strains and Phages Intended for Therapy

The process of phage therapy begins with the identification and selection of phages and bacterial strains suitable for phage production. Phages are found in any environment, even under extreme conditions, being the most abundant biological entities on our planet, with a current total estimate of 1031 particles [17]. Phage “hunting” site selection will depend on the bacteria to be eliminated [43], since phages infecting a certain host can normally be isolated from environments inhabited by such bacterium. However, it may be difficult to find phages active against a bacterial isolate from samples of the same patient, since the resident bacteria (including the pathogenic one) are probably lysogenic, tolerant, or resistant to the accompanying phages. Therefore, alternative sampling places are recommended. Many pathogenic bacteria also present free-living variants in nature, making it possible to find suitable phages for therapy in environmental samples. Human-made sites or infrastructures, such as wastewater treatment plants, usually contain a high diversity of phages active against human pathogens [44].
Once samples are collected, phage screening parameters will depend on the target bacterium. In general, phages can be isolated using reference strains or clinical isolates as hosts for propagation [45]. In some cases, the bacterium of interest may not be amenable to culture, and thus a surrogate host may be used instead (e.g., Mycobacterium smegmatis is strongly preferred for the isolation of mycobacteriophages over the pathogenic, slowly growing Mycobacterium tuberculosis [46]). If a broad-range phage is desired (for example, one that targets several strains), isolation with multiple bacterial strains may be preferred, although single strain enrichment does not necessarily preclude finding broad-range phages [43]. Apart from the search for new phages, it is also possible to use those that are already available in laboratories around the world, or in phage banks [47,48]. In general terms, the isolation of phages does not pose a great difficulty neither methodologically nor economically, as long as its host bacterium is culturable. Indeed, phage isolation and selection methods are as old as the first descriptions of phages and are still considered as solid and useful protocols nowadays.

2.2. Small- and Large-Scale Production Processes

Phage production is typically carried out by the double-layer agar method, which allows isolation of individual phages, and then liquid culture of single plaques [49]. A pure phage lysate (containing a single type of lysis plaques) is obtained by centrifugation, filtration, and/or further purification methods (see below). The process can be scaled up and optimized to an industrial level by using bioreactors of different sizes [50], which allow continuous and semi-continuous production. The latter seems the most advantageous for large-scale standardized production, since it avoids co-evolution of the phage with the bacteria (although it may be operationally complex) [51].

2.3. Purification of Phage Solutions

The main goal of this process is to avoid the presence of bacterial toxins, lipopolysaccharide, or other cellular debris, in the phage suspensions [52]. In addition to centrifugation and membrane filtration, it may be necessary to apply additional steps, such as dialysis, ultrafiltration, or treatment with organic solvents. To verify the absence of toxic components, the corresponding tests should be performed and, if necessary, additional purification methods such as specialized filtration, affinity chromatography, tangential flow filtration, and/or CsCl gradient ultracentrifugation can be used. An alternative may be anion exchange chromatography, as demonstrated using Convective Interaction Media® (CIM) columns [53]. These more specialized approaches often require laborious optimization for each phage (or groups of related phages), but also allow the concentration of viral particles in lysates with low phage titers.

2.4. Storage

Depending on the phage, phages can be stored at different temperatures, commonly at 4 °C, −80 °C, or in liquid nitrogen (−196 °C), or can be freeze-dried [54,55]. Protection of lysates against evaporation or contamination is often sufficient to minimize the decrease in their titer over time. Additives can be added to prevent or delay the loss of infectivity. Mg2+ and Ca2+ ions (around 10 mM in the form of CaCl2 or MgSO4) are the most used supplements since they are added to the culture medium before infection to facilitate adsorption and are then present in the recovered lysate [54,56]. Other common additives are cryopreservants such as disaccharides (lactose, sucrose, trehalose) or polyethyleneglycol, as well as gelatin or Ficoll [57,58]. Adsorption of phages to cell debris can cause a considerable decrease in titer, implying that removing cellular contaminants from a crude lysate is also important for storage. In addition, minimizing the number of passages is an important issue since multiple rounds of propagation combined with the high mutation frequencies shown by phages can lead to genetic differences between the original isolate and its progeny [54].

2.5. Formulation and Administration

Phages are basically protein structures and, therefore, they are susceptible to proteases, certain chemical compounds, high temperatures, pH, and ionic strength. Thus, it is important to use an appropriate formulation to ensure that the phage titer remains stable both during formulation and storage and in the in vivo environment where they are applied. Again, the optimal formulation conditions may vary depending on the specific phage, so this deserves careful consideration in the case of phage cocktails, as each type of phage may require individually tailored conditions [59]. However, the major determinant of phage therapy formulations is the method chosen for delivery, which, in turn, depends on the infection site:
(a)
Oral administration is appropriate for gastrointestinal infectious diseases. In some cases, oral phages given without additional protection, as water-based liquid suspensions, reportedly survived gastric passage and were recovered in the feces [60,61,62]. The formulation efforts for liquid phage suspensions are typically minimal since phages are just prepared in sterile buffers such as phosphate-buffered saline (PBS), the bacterial growth medium, standard saline, or water [61,63,64,65]. More elaborate formulations specifically intended for oral administration can improve phage survival through the extreme conditions of the gastrointestinal tract. For example, encapsulation protects phages from the highly acidic stomach environment and digestive enzymes [66]. Furthermore, their release can be triggered in a controlled manner, for example, pH-dependent release, with capsules programmed to become permeable at different pHs regarding the aimed site of action: from the stomach (pH 1–3) to the small intestine (pH 5.5–6.5) or the colon (pH 6.5–7.2) [67]. A wide range of natural and synthetic polymers are available that offer considerable plasticity for tailoring phage encapsulation and subsequent release to different biomedical applications, including polysaccharides, natural or synthetic plastic polymers, liposomes, and micelles [68,69].
(b)
Topical administration of phages is chosen for skin infections, wounds, burns, ulcers, and osteoarticular infections [70]. Phages have been topically administered in liquid, semi-solid, and liposome-encapsulated formulations, as well as phage-immobilized wound dressings [63]. When using liquid preparations, they may just be dripped onto the infected site or applied in a gauze soaked with the preparation. Alternatively, gel or cream formulations are suitable to overcome some of the limitations of liquid preparations, with a preference for hydrogels over organic solvent-based gels. This is especially relevant for the treatment of burn wounds, since hydrogels help keep the wound hydrated as much as they favor phage stability [63]. Commercial infection-care products can also be used as a formulation basis for topically delivering phages, but care should be taken as to whether the composition of the product reduces phage infectivity [71].
(c)
The local phage treatment of respiratory infections requires preparing phages either as stable liquid formulations for intranasal instillation or nebulization, or as a solid powder in an inhalable form [72]. The most popular formulations for respiratory infections are liquid suspensions, due to the relative simplicity of preparation. Nebulization of liquid phage suspensions has been tested with mixed outcomes, generally suggesting that temperature, relative humidity, the nebulization-induced mechanical stress, delivery efficiency of the system, and the nature of the phage itself greatly influence the outcome. Regarding dry powder inhalation, the methods to obtain solid phage formulations include freeze-drying or spray-drying. In general, both processes subject phages to diverse stresses that may impact their infectivity [73], but the control of key parameters and addition of suitable excipients, including polymers for encapsulation, can enhance phage preservation [74,75].
(d)
Intravenous administration is recommended in the treatment of systemic infections [76]. In this case, liquid phage suspensions are typically used, normally prepared in aqueous buffers suitable for inoculation [64,72].
(e)
Intravesical instillation of liquid phage preparations has also been used to treat genitourinary tract infections [77,78].

3. Quality Criteria for Therapeutic Phage Preparations

As products intended for human therapy, phage preparations must comply with certain quality criteria that ensure their safety for clinical use, as well as exhaustive traceability documentation. The production and delivery parameters must be set up by the phage preparation supplier in accordance with the applicable regulations (see Section 4) and to meet the agreed quality attributes. For phage-based products, these quality controls would typically include:
(a)
Phage identity. The identity of each phage is defined by its specific genomic sequence [79,80]. Metagenomics has already been proposed as a quality control method for some vaccines [81], and thus has also been used to assess the composition of commercial phage products [82,83]. This method allows the detection of biological contaminants while also assuring the active product identity. While random mutations during propagation are inevitable, they need to be as limited as possible by process design (e.g., minimizing subcultivation steps), and functional properties should be regularly tested with validated quality controls, as even single-nucleotide polymorphisms can lead to significant phenotypic changes. However, a highly discriminating PCR-based genotyping technique might be sufficient in some cases [79]. The maximum acceptable level of genomic divergence between the master batch and the phage population in the therapeutic product, as well as the frequency of such quality check, should be nonetheless adjusted on a case-by-case basis [79].
(b)
Phage Titer. The titer of each individual phage is classically assessed by the double-layer agar method. An alternative is lethality curves, in which the kinetics of phage-induced lysis are assessed by measuring the optical density of phage-infected bacterial cultures [84]. Other methods, such as qPCR and ELISA, can be used to quantify phages, but they do not necessarily quantify infectious viral particles, whereas double-layer and lethality assays do determine biological activity [79].
(c)
General Purity. For biopharmaceuticals, the purity and correct composition is classically assessed by high-performance liquid chromatography, combined with mass spectrometry if necessary. These methods can be used to identify phage capsid proteins, toxins, or other bacterial proteins. Because of the potential risk posed by the necessary production with pathogenic bacterial hosts, quality criteria should specify maximum levels for contaminants such as toxins or bacterial DNA, which normally must be tested with specific and appropriate molecular biology methods as specified below.
(d)
Toxins. Several in vitro methods have been developed for endotoxins quantification: gel-clot, turbidimetric, and chromogenic tests. Among the latter, the limulus amebocyte lysate assay is the most widely used [85]. When this assay is not applicable, e.g., due to masking effect, a reporter cell line can be used [86]. In addition, several commercial assays can be used to detect other toxic bacterial proteins, including ELISA or assays based on reporter cell lines.
(e)
Contaminating Nucleic Acids. Quality controls may also be required to determine the concentration of contaminating nucleic acids (i.e., non-phage nucleic acids). The presence and concentration of residual nucleic acids are typically checked by qPCR.
(f)
Other Quality Controls. Current regulations on sterility or general quality parameters in pharmaceutical products should also apply to phage-based pharmaceuticals [87]. Some parameters that may need to be checked are the total microbial load, pH, osmolarity, visual appearance, and/or maximum water content (in lyophilized preparations) [79].

4. Regulation for Phage Preparations

Perhaps the greatest hurdle to the implementation of phage therapy in Western medicine is the lack of appropriate regulation. If phage preparations are considered “classical” medicinal products, they must comply with the corresponding legislation concerning medicinal products production and quality. This essentially means that they should follow GMP (good manufacturing practices) requirements, which has imposed an important problem for the development of medicinal phage preparations in terms of increasing costs or complicating management for developing large-scale production [88].
Although therapeutic phages shall be considered indeed a medicinal product, the nature itself of the phage makes it essentially different from common antimicrobial chemotherapy. Indeed, phage specificity, virus–host co-evolution, or a complex in vivo pharmacological behavior have negatively influenced the outcome of many of the phage therapy clinical trials conducted in contemporary times [33,89,90,91]. Therefore, the clinical trial path to the market still seems rather far away. A provisional conclusion has been that the parameters by which we evaluate phage therapy should perhaps be specifically adapted and may not be the same as those of chemotherapeutics [92]. So far, the most successful approach to applying phage therapy in the clinic has been tailored formulations, i.e., phage preparations specifically designed and developed to tackle the infection present in a specific patient. In fact, the compassionate use of phages intended for specific patients without better treatment options is the current regulatory framework for phage therapy in most countries [4,88]. Within this framework, some of the stricter requirements can be bypassed, although it still requires approval from the competent authorities in every individual case.
A recently adopted strategy in Belgium has allowed a more systematic and practical approach to personalized phage therapy, which also provides production and handling flexibility (e.g., by not forcing strict GMP compliance). The key to this approach is to consider phage products as magistral preparations, since, unlike medicinal products, these are subject to less strict regulation in terms of their production and marketing [93,94]. In this context, phages are regarded as active pharmaceutical ingredients (APIs) (substances used in a finished pharmaceutical product intended to procure a pharmacological effect), which are provided with an external quality assessment according to a dedicated monograph. A practical infrastructure to operate under this regulatory framework would be a dedicated phage bank in which each conserved phage should be certified to be used as an API in such a way that the certification covers all essential quality attributes (including a “genetic passport” issued for each phage in particular) [48,94]. In Spain, magistral formulations are governed by European regulations and by the Royal Spanish Pharmacopoeia (order SSI/23/2015, BOE-A-2015-467) and by the consensus document specifying the requirements for APIs for use in magistral formulations (CTI/FM/150/02/16) and following the rules of correct elaboration and quality control (RD 175/2001, BOE-A-2001-5185). These documents define “magistral formulation” as “the drug intended for an individualized patient to explicitly fulfill a detailed medical prescription of the active ingredients that it includes, prepared by a pharmacist or under his direction according to the rules of proper preparation and quality control established for this purpose, dispensed in a pharmacy or pharmaceutical service and with due information to the user”. In the case of phages, express authorization from the competent agency (in Spain, the AEMPS) would be required to use them in magistral formulations, as well as a dedicated monograph provided by the supplier and stating the “rules of proper preparation and quality control”. The exemplary monograph elaborated in Belgium can be accessed as Supplementary Material in reference [94], and it contains, in a summarized manner, most of the issues covered by this review. While this bold framework may be practical for the time being, it has not yet been adopted by many countries. To our knowledge, there are no examples in Spain of phage therapy application under this regulatory framework, although phages have been administered as compassionate medicines. In any case, it is altogether clear that phage therapy may not fit within the traditional drug regulations. Some calls have already been made to adapt or develop a specific regulation for phage therapy [95,96]. The main objectives of such a regulation should be, at least, (a) developing adequate criteria for assessing the quality, efficacy, and safety of phage products; (b) lightening the formal procedures for administrating personalized phage therapy; and (c) fairly distributing responsibility and compensation among the involved actors.
Regardless of the sub-optimal regulatory status of phage therapy in most countries, including Spain, phage preparations for therapy are available either commercially (e.g., through the Eliava Institute in Georgia, which keeps and promotes the Eastern tradition in phage therapy) or through the request to academic or clinical institutions devoted to phage therapy research and promotion.

5. Clinical Trials and Prospects for Phage Therapy

Although potentially controversial [97], an adapted regulation in support of personalized phage therapy (perhaps following the lead of the Belgian experience) can be a shortcut to bring phage therapy to the clinic and even to the market in Spain. However, as it has been pointed out, once phage therapy becomes increasingly adopted in clinical practice and/or the number of untreatable infections begins to rise, the personalized phage therapy would need such infrastructure dedication that it may become impractical (unless healthcare/research systems are decisively funded to meet the need). This situation would surely favor a more traditional, market-based approach [89]. The latter would still require phage formulations to be developed through the common drug clinical pipeline, i.e., demonstrating efficacy in randomized clinical trials. In fact, one of the major hurdles to the practical prospects of phage therapy is the lack of substantial evidence within the current clinical trial standards. While a good amount of mostly successful case reports has been recently published, the phage research community still fails to deliver convincing randomized clinical trial results supporting phage therapy efficacy. If we consider the two better-known recent phage therapy trials that yielded disappointing results (i.e., the PhagoBurn trial and the Nestlé Bangladesh diarrhea trial), available evidence suggests a poor understanding of the complex phage behavior in vivo, as well as improper assumptions on the pathogenic bacteria susceptibility [33,90]. Both issues greatly complicate standardized, large-scale trials. For example, the abovementioned trials reported a lower phage dose than expected at the infection site and resistance to the cocktails among the infecting bacteria. A posterior reflection on the Bangladesh study by one of its authors pointed out that the polymicrobial nature of some infections makes them a tough target for phage therapy alone, and even more complex for a productive randomized controlled trial [89]. Therefore, a possible suggestion towards successful clinical trials may be to exclusively target well-characterized infections, dedicating extra care to consider the appropriateness of the targeted pathologies [98]. In actuality, the only phase 2 clinical trial that has produced positive efficacy results to date followed this path, by selecting a single-pathogen infection (chronic otitis caused by antibiotic-resistant Pseudomonas aeruginosa) and testing the identity and quantity of bacteria present in every patient [99]. Further issues regarding phage production (according to GMP), storage, product shelf-life, dosage, and proper formulation/administration need to be exhaustively assessed and optimized to ensure the future success of clinical trials. Regardless of the potential hurdles, many phage clinical trials are currently ongoing at different stages, both including predefined phage cocktails and personalized interventions, thus safeguarding a potential future regularization of phage therapy based on clinical trial evidence. Some examples of ongoing clinical trials are included in Table 1.

6. Most Urgent Indications for the Application of Phage Therapy in Spain

In our opinion, the use of phage therapy would be advisable:
(a)
In the case of a severe infection produced by an MDR bacterium.
(b)
When the infection occurs in an area reluctant to the use of antibiotics, such as in prosthetics.
(c)
Or, in general, whenever there is no standard of care option available, such as patients suffering from hypersensitivity to the antibiotic treatment.
In these situations, i.e., in the absence of any other satisfactory therapy and when there is a real risk to the patient’s life or significant deterioration of their quality of life, the use of phages, possibly in combination with standard-of-care antibiotics, would be advisable. Conversely, whenever the patient’s infection can be satisfactorily treated with antibiotics, those will always be the therapy of choice. Additionally, a proper diagnostic of the infection-causing bacteria must be conducted (see Section 5), as well as a susceptibility proof of such bacteria towards the phage preparation to be applied (what is known as a “phagogram”) [100,101]. We provide details below on the most urgent indications in which a regularization of phage therapy could have a greater impact.

6.1. Cystic Fibrosis

Cystic fibrosis (CF) is a rare disease (the prevalence in Spain is estimated to be 0.55 per 10,000 inhabitants [102]). Although not an infectious disease, per se, it is relevant here due to the chronicity of infections in these patients. The lungs of CF patients are generally infected by strains of Haemophilus influenzae, P. aeruginosa, Stenotrophomonas maltophilia, S. aureus, Achromobacter spp., Burkholderia cepacia, and Mycobacterium abscessus, usually resistant to many antimicrobials. Among them, the main pathogens in Spain are S. aureus and P. aeruginosa (chronically present in around half of the total patients), with the latter being significantly associated with worse pulmonary function. According to a Spanish multicenter study, 55% of P. aeruginosa isolates from CF patients were MRD, while 16% were extensively drug-resistant [103]. In addition, these microorganisms can form biofilms, which make antibiotic treatment difficult even when they are susceptible. Prolonged treatment with antimicrobials in adulthood also facilitates the settlement of nontuberculous mycobacteria, yeasts, and filamentous fungi, which further contribute to clinical disease. Colonization is associated with episodes of exacerbation of the respiratory symptoms and progressive deterioration of respiratory function, which is the leading cause of death in CF patients [104,105]. CF is the third most frequent indication for lung transplantation; however, the persistent infections common among CF patients relate to significant post-transplant morbidity and mortality, and in certain circumstances, these infections may even contraindicate transplantation. For this reason, the clearance of MDR infections in CF patients, particularly those caused by P. aeruginosa, is expected to have a remarkable impact on the life quality and expectancy of these patients.

6.2. Osteoarticular Infections

These are a particular form of deep, localized infections with poor response to antibiotic therapy. The diffusion of antibiotics into bone tissue is often low and is negatively affected by the presence of bacterial biofilms at the contact between bone and prosthetic material. Repeated surgeries to eradicate the biofilm mechanically, i.e., removing the implant and/or resecting the infected bone, and sometimes amputation, is the only infection control option [106]. Most of these infections are related to the hospital environment, such as those produced by multidrug-resistant P. aeruginosa and S. aureus resistant to methicillin, fluoroquinolones, and rifampicin. In these cases, treatment options are often limited to antibiotics of last resort, such as polymyxins which have been shown to have higher relapse rates and higher frequency of complications.
There are some recent examples of clinical cases (CF and osteoarticular infections) treated with phages that support the feasibility of this therapy (Table 2).

7. General Aspects of the Use of Endolysins as Antimicrobials

In addition to phages, their lytic enzymes have also been explored as antimicrobials [117]. Phage lysins are enzymes that mediate enzymatic cleavage of peptidoglycan either in the early stages of phage infection to assist in the injection of phage DNA or the late stages of infection, allowing the release of viral progeny by generalized lysis of the bacterium. Lysins are a promising new class of antibacterial agents that may also offer an answer to the problem of bacterial multidrug resistance. When added exogenously, they induce rapid osmotic lysis of Gram-positive bacteria by degradation of peptidoglycan with subsequent cell death. Their efficacy has also been extended to Gram-negative pathogens, which have a protective outer membrane, by protein engineering [118]. These endolysins are also called “enzybiotics” and have been shown to be safe, effective, fast-acting, and highly specific [119,120]. In addition, they weaken biofilms and have a low likelihood of causing resistance [119,120], and can be used either alone or in combination with traditional antibiotics [121]. Their potential applications are the same as those of phages, but they offer some advantages. For example, enzymes cannot propagate as phages do; therefore, their effect is directly dose-dependent and can be better controlled. In addition, they are not able to mobilize DNA, so horizontal gene transfer is avoided, and they should be easier to produce on an industrial scale given the prior experience with the production of heterologous therapeutic proteins.
Since peptidoglycan is present exclusively in bacteria and not in mammalian cells, the risk of cytotoxic effects to humans and animals is minimized. However, since they are proteins, they may induce an immune response. Indeed, in vitro and in vivo studies have shown that neutralizing antibodies can be generated upon repeated exposure to lysins. However, although these antibodies reduce the antibacterial activity of the enzybiotics, they do not completely neutralize them [122,123], which suggests that enzybiotics could be used repeatedly to treat the same bacterial infection [124]. The situation for the treatment of Gram-negative bacteria with lysins may be slightly different. Testing several non-engineered anti-Gram-negative lysins in vivo resulted in antibody production that did completely inhibit the in vitro bactericidal activity of the enzymes [125]. This suggests that further optimization must be taken into account when translating anti-Gram-negative enzybiotics to the clinic. Engineered endolysins against Gram-negative bacteria, nonetheless, have shown promising results in in vivo models of infection [126]. An additional major problem of many anti-Gram-negative enzybiotics is a lack of activity in the presence of human serum, probably due to their high cationicity, thus perhaps limiting their use for systemic treatment [127]. However, serum-active lysins against Gram-negative bacteria are already being developed by some companies and laboratories [126,128,129]. These advancements point out that, with appropriate experimental development, clinically valuable lysins will also be available in the medium term against Gram-negative pathogens.
Systemic administration of enzybiotics may also release cellular debris from lysed bacteria, which may induce a proinflammatory response. These bacterial cell debris include lipopolysaccharides, (lipo)teichoic acids, and peptidoglycan through membrane fragmentation and can potentially lead to severe complications such as septic shock. However, allergic or severe inflammatory reactions have not been described so far upon administration of enzybiotics, even in clinical trials with human subjects [130,131]. The conclusions from some studies indicate that there should be an optimal dose of enzybiotics that is sufficient to kill the bacterial pathogen without additional fragmentation of the peptidoglycan layer and spreading of proinflammatory factors [122,123].
In recent years, the way has been paved for recombinant lysins to enter different phases in preclinical and clinical trials. Accelerated clinical advances and their high technical feasibility make them a good alternative therapy to replace or to be used in combination with conventional antibiotics in the short term, even with some authors pointing out that lysins may be able to enter the clinic in a shorter term than phages [132]. Some companies are already taking the initiative to conduct phase II and III clinical trials with enzybiotic-based products. An enzybiotic for cosmetic and topical application is already being marketed under the GladSkin brand, marketed by Micreos. Recently, human clinical trials (Phase I) have been conducted with two endolysins, iNtRON-N-Rephasin® SAL200 (Tonabacase) [130] and Exebacase (CF-301) from ContraFect [133], against S. aureus bacteremia without observing any adverse effects. Tonabacase is currently in phase II [134], and Exebacase (CF-301) has completed phase II successfully and is in phase III to test its activity against bacteremia and endocarditis caused by S. aureus [135].

8. Global Phage Therapy Market

As discussed in this review, the application of phage therapy is experiencing a considerable boom in recent years. Since 2017, the global phage therapy market reached USD 567.9 million and growth expectations at compound annual growth rate (CAGR) are of 3.9% for the period from 2018 to 2026 [136]. In 2016, USA had a 37% share of the global market and became the largest market in the use of phages [137]. Europe is the second-largest market due to the wide application of phages in the food and environmental fields. The particular cases of Georgia, Poland, and Belgium are especially relevant because they are pioneer countries in having taken the step from basic research to the market. It seems most probable that this market will experience growth in the short term, supported by heavy investment in biotechnology infrastructures, the evolution of agricultural practices and their corresponding regulation, and foreseeable government initiatives favoring the use of these technologies (or at least discouraging the massive use of antibiotics) in different sectors, including that of human health, due to the antibiotic resistance crisis [138]. In this regard, the most promising market for phage therapy would be the treatment of infections caused by MDR bacteria. Estimates are that this will globally represent more than USD 13.8 billion a year by 2027 [139].
However, some potential hurdles may still be found in the way towards a successful market entrance, such as the already mentioned regulatory issues (Section 4), the quality of the results from future phage therapy clinical trials (Section 5), or questions about phage patentability (although patents granted for phage products do already exist) [132,140]. The more “canonical” nature of enzybiotics, similar to other therapeutic proteins, is an additional reason to think that they may make their market entrance in a shorter term than full phages, following the traditional avenue through intellectual property protection of the molecules and conduction of clinical trials (Section 7). However, provided that reasonable regulatory measures come into place, therapy with full virions, especially in its personalized version, may be available to patients first, although its path to proper marketability may be longer.
In this context and given that large pharmaceutical companies have practically stopped investing in new antimicrobial compounds, many scientific startups are focusing on studying and characterizing phages and their products for their therapeutic application. These new companies are located mainly in the USA, India, Korea, Canada, and some European countries. In Spain, the first company focused on the development of endolysins (Telum Therapeutics) has recently been created [141]. An optimistic short-term future can also be envisioned for these developments if we consider the latest investments taking place in the area of alternatives to antibiotics, such as the BioNtech acquisition of PhagoMed [142], or the high investment rounds to support new endolysins search platforms, as in Micreos [143].

9. Final Conclusions

Bacterial resistance is a major global threat and a leading cause of mortality worldwide. The lack of effective antibiotics against MDR strains urgently requires therapeutic alternatives, and phages or their derivatives can be a promising approach with sufficient scientific and technological know-how in place. However, the lack of both specific regulation for their clinical use and public awareness stands out, in our opinion, as the major hurdles to be presently faced. With this review, we would like to emphasize the advantages of phages as therapeutic tools and how they can be used to combat MDR bacteria. Moreover, despite some foreseeable drawbacks also covered here, phages are nowadays the only short-term solution for many patients. Taking this into account, a suitable regulatory initiative from the competent authorities would be welcomed in Spain in the short term. Both scientific literature and the lead examples of other countries prove that phage therapy is already mature to be translated into a regulation that must ease, protect, and help developing the phage therapy market and infrastructure.

Author Contributions

Conceptualization, R.V., R.D.-M., P.D.-C., P.G. (Pedro García), D.G., M.M., M.R.-R., R.S., M.T., M.Á.T.-M., P.G. (Pilar García); writing—original draft preparation, R.V., R.D.-M., P.D.-C., P.G. (Pedro García), D.G., M.M., M.R.-R., R.S., M.T., M.Á.T.-M., P.G. (Pilar García); writing—review and editing, R.V., R.D.-M., P.D.-C., P.G. (Pedro García), D.G., M.M., M.R.-R., R.S., M.T., M.Á.T.-M., P.G. (Pilar García); visualization, R.V.; funding acquisition, P.G. (Pilar García), P.G. (Pedro García), P.D.-C., M.M., M.Á.T.-M. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by Grant RED2018-102589-T funded by MCIN/AEI/10.13039/501100011033. R.V. and D.G. were supported by the Research Foundation–Flanders (FWO) under grant [G066919N]. Pe. G. and R.V. were supported by the Spanish Ministry of Science and Innovation (Grant number SAF2017-88664-R), with additional funding provided by the Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), an initiative of the Instituto de Salud Carlos III. P.D.-C. was financially supported by a Ramón y Cajal contract RYC2019-028015-I and project PID2020-112835RA-I00 funded by MCIN/AEI/10.13039/501100011033, ESF Invest in your future, project SEJIGENT/2021/014, funded by Conselleria d’Innovació, Universitats, Ciència i Societat Digital (Generalitat Valenciana), and project AP2020-27 funded by VLC-Biomed (UV-La Fe 2020 Programa Acciones Preparatorias). M.M. was supported by project PID2020-113355GB-I00 of Spanish Ministry of Science and Innovation /Agencia Estatal de Investigación (AEI)/European regional fund (ERF). M.Á.T.-M. was supported by Spanish Ministry of Science and Innovation (Grant number SAF2017- 82251-R).

Conflicts of Interest

D.G. is currently employed by Telum Therapeutics SL. R.D.-M. owns stocks from Telum Therapeutics SL.

References

  1. Antimicrobial Resistance Collaborators. Global burden of bacterial antimicrobial resistance in 2019: A systematic analysis. Lancet 2022, 399, 629–655. [Google Scholar] [CrossRef]
  2. O’Neill, J. Tackling Drug-Resistant Infections Globally: Final Report and Recommendations; HM Government: London, UK, 2016. [Google Scholar]
  3. Sulakvelidze, A.; Alavidze, Z.; Morris, J.G., Jr. Bacteriophage therapy. Antimicrob. Agents Chemother. 2001, 45, 649–659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. McCallin, S.; Sacher, J.C.; Zheng, J.; Chan, B.K. Current state of compassionate phage therapy. Viruses 2019, 11, 343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Chen, L.K.; Kuo, S.C.; Chang, K.C.; Cheng, C.C.; Yu, P.Y.; Chang, C.H.; Chen, T.Y.; Tseng, C.C. Clinical Antibiotic-resistant Acinetobacter baumannii Strains with Higher Susceptibility to Environmental Phages than Antibiotic-sensitive Strains. Sci. Rep. 2017, 7, 6319. [Google Scholar] [CrossRef] [PubMed]
  6. Chan, B.K.; Sistrom, M.; Wertz, J.E.; Kortright, K.E.; Narayan, D.; Turner, P.E. Phage selection restores antibiotic sensitivity in MDR Pseudomonas aeruginosa. Sci. Rep. 2016, 6, 26717. [Google Scholar] [CrossRef] [Green Version]
  7. North, O.I.; Brown, E.D. Phage-antibiotic combinations: A promising approach to constrain resistance evolution in bacteria. Ann. N. Y. Acad. Sci. 2021, 1496, 23–34. [Google Scholar] [CrossRef]
  8. Jansen, M.; Wahida, A.; Latz, S.; Kruttgen, A.; Hafner, H.; Buhl, E.M.; Ritter, K.; Horz, H.P. Enhanced antibacterial effect of the novel T4-like bacteriophage KARL-1 in combination with antibiotics against multi-drug resistant Acinetobacter baumannii. Sci. Rep. 2018, 8, 14140. [Google Scholar] [CrossRef] [Green Version]
  9. Gordillo Altamirano, F.; Forsyth, J.H.; Patwa, R.; Kostoulias, X.; Trim, M.; Subedi, D.; Archer, S.K.; Morris, F.C.; Oliveira, C.; Kielty, L.; et al. Bacteriophage-resistant Acinetobacter baumannii are resensitized to antimicrobials. Nat. Microbiol. 2021, 6, 157–161. [Google Scholar] [CrossRef]
  10. Manohar, P.; Madurantakam Royam, M.; Loh, B.; Bozdogan, B.; Nachimuthu, R.; Leptihn, S. Synergistic Effects of Phage-Antibiotic Combinations against Citrobacter amalonaticus. ACS Infect. Dis. 2022, 8, 59–65. [Google Scholar] [CrossRef]
  11. Blasco, L.; Ambroa, A.; Lopez, M.; Fernandez-Garcia, L.; Bleriot, I.; Trastoy, R.; Ramos-Vivas, J.; Coenye, T.; Fernandez-Cuenca, F.; Vila, J.; et al. Combined Use of the Ab105-2phiDeltaCI Lytic Mutant Phage and Different Antibiotics in Clinical Isolates of Multi-Resistant Acinetobacter baumannii. Microorganisms 2019, 7, 556. [Google Scholar] [CrossRef] [Green Version]
  12. Langdon, A.; Crook, N.; Dantas, G. The effects of antibiotics on the microbiome throughout development and alternative approaches for therapeutic modulation. Genome Med. 2016, 8, 39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Boeckaerts, D.; Stock, M.; Criel, B.; Gerstmans, H.; De Baets, B.; Briers, Y. Predicting bacteriophage hosts based on sequences of annotated receptor-binding proteins. Sci. Rep. 2021, 11, 1467. [Google Scholar] [CrossRef] [PubMed]
  14. Hampton, H.G.; Watson, B.N.J.; Fineran, P.C. The arms race between bacteria and their phage foes. Nature 2020, 577, 327–336. [Google Scholar] [CrossRef] [PubMed]
  15. Gehring, K.; Charbit, A.; Brissaud, E.; Hofnung, M. Bacteriophage lambda receptor site on the Escherichia coli K-12 LamB protein. J. Bacteriol. 1987, 169, 2103–2106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Loc-Carrillo, C.; Abedon, S.T. Pros and cons of phage therapy. Bacteriophage 2011, 1, 111–114. [Google Scholar] [CrossRef] [Green Version]
  17. Mushegian, A.R. Are There 10(31) Virus Particles on Earth, or More, or Fewer? J. Bacteriol. 2020, 202, e00052-20. [Google Scholar] [CrossRef]
  18. Chibani-Chennoufi, S.; Bruttin, A.; Dillmann, M.L.; Brussow, H. Phage-host interaction: An ecological perspective. J. Bacteriol. 2004, 186, 3677–3686. [Google Scholar] [CrossRef] [Green Version]
  19. Burrowes, B.H.; Molineux, I.J.; Fralick, J.A. Directed in Vitro Evolution of Therapeutic Bacteriophages: The Appelmans Protocol. Viruses 2019, 11, 241. [Google Scholar] [CrossRef] [Green Version]
  20. Eskenazi, A.; Lood, C.; Wubbolts, J.; Hites, M.; Balarjishvili, N.; Leshkasheli, L.; Askilashvili, L.; Kvachadze, L.; van Noort, V.; Wagemans, J.; et al. Combination of pre-adapted bacteriophage therapy and antibiotics for treatment of fracture-related infection due to pandrug-resistant Klebsiella pneumoniae. Nat. Commun. 2022, 13, 302. [Google Scholar] [CrossRef]
  21. Navarro, F.; Muniesa, M. Phages in the Human Body. Front. Microbiol. 2017, 8, 566. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Manrique, P.; Bolduc, B.; Walk, S.T.; van der Oost, J.; de Vos, W.M.; Young, M.J. Healthy human gut phageome. Proc. Natl. Acad. Sci. USA 2016, 113, 10400–10405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Lawrence, D.; Baldridge, M.T.; Handley, S.A. Phages and Human Health: More Than Idle Hitchhikers. Viruses 2019, 11, 587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. EFSA Panel on Biological Hazards (BIOHAZ). Evaluation of the safety and efficacy of ListexTM P100 for reduction of pathogens on different ready-to-eat (RTE) food products. EFSA J. 2016, 14, 4565–4594. [Google Scholar] [CrossRef]
  25. Abedon, S.T.; Kuhl, S.J.; Blasdel, B.G.; Kutter, E.M. Phage treatment of human infections. Bacteriophage 2011, 1, 66–85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Chan, B.K.; Abedon, S.T.; Loc-Carrillo, C. Phage cocktails and the future of phage therapy. Future Microbiol. 2013, 8, 769–783. [Google Scholar] [CrossRef]
  27. Gu, J.; Liu, X.; Li, Y.; Han, W.; Lei, L.; Yang, Y.; Zhao, H.; Gao, Y.; Song, J.; Lu, R.; et al. A method for generation phage cocktail with great therapeutic potential. PLoS ONE 2012, 7, e31698. [Google Scholar] [CrossRef] [Green Version]
  28. Oechslin, F. Resistance Development to Bacteriophages Occurring during Bacteriophage Therapy. Viruses 2018, 10, 351. [Google Scholar] [CrossRef] [Green Version]
  29. Dabrowska, K. Phage therapy: What factors shape phage pharmacokinetics and bioavailability? Systematic and critical review. Med. Res. Rev. 2019, 39, 2000–2025. [Google Scholar] [CrossRef] [Green Version]
  30. Dabrowska, K.; Abedon, S.T. Pharmacologically Aware Phage Therapy: Pharmacodynamic and Pharmacokinetic Obstacles to Phage Antibacterial Action in Animal and Human Bodies. Microbiol. Mol. Biol. Rev. 2019, 83, e00012–e00019. [Google Scholar] [CrossRef]
  31. Attrill, E.L.; Claydon, R.; Lapinska, U.; Recker, M.; Meaden, S.; Brown, A.T.; Westra, E.R.; Harding, S.V.; Pagliara, S. Individual bacteria in structured environments rely on phenotypic resistance to phage. PLoS Biol. 2021, 19, e3001406. [Google Scholar] [CrossRef]
  32. Abedon, S.T.; Thomas-Abedon, C. Phage therapy pharmacology. Curr. Pharm. Biotechnol. 2010, 11, 28–47. [Google Scholar] [CrossRef] [PubMed]
  33. 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, 35–45. [Google Scholar] [CrossRef]
  34. Perea, L.; Rodriguez-Rubio, L.; Nieto, J.C.; Zamora, C.; Canto, E.; Soriano, G.; Poca, M.; Blanco-Picazo, P.; Navarro, F.; Muniesa, M.; et al. Bacteriophages immunomodulate the response of monocytes. Exp. Biol. Med. 2021, 246, 1263–1268. [Google Scholar] [CrossRef] [PubMed]
  35. Krut, O.; Bekeredjian-Ding, I. Contribution of the Immune Response to Phage Therapy. J. Immunol. 2018, 200, 3037–3044. [Google Scholar] [CrossRef] [PubMed]
  36. Hodyra-Stefaniak, K.; Miernikiewicz, P.; Drapala, J.; Drab, M.; Jonczyk-Matysiak, E.; Lecion, D.; Kazmierczak, Z.; Beta, W.; Majewska, J.; Harhala, M.; et al. Mammalian Host-Versus-Phage immune response determines phage fate in vivo. Sci. Rep. 2015, 5, 14802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Roach, D.R.; Leung, C.Y.; Henry, M.; Morello, E.; Singh, D.; Di Santo, J.P.; Weitz, J.S.; Debarbieux, L. Synergy between the Host Immune System and Bacteriophage Is Essential for Successful Phage Therapy against an Acute Respiratory Pathogen. Cell Host Microbe 2017, 22, 38–47.e4. [Google Scholar] [CrossRef] [PubMed]
  38. Borysowski, J.; Gorski, A. Is phage therapy acceptable in the immunocompromised host. Int. J. Infect. Dis. 2008, 12, 466–471. [Google Scholar] [CrossRef] [Green Version]
  39. Brown-Jaque, M.; Calero-Caceres, W.; Muniesa, M. Transfer of antibiotic-resistance genes via phage-related mobile elements. Plasmid 2015, 79, 1–7. [Google Scholar] [CrossRef]
  40. Chen, J.; Quiles-Puchalt, N.; Chiang, Y.N.; Bacigalupe, R.; Fillol-Salom, A.; Chee, M.S.J.; Fitzgerald, J.R.; Penades, J.R. Genome hypermobility by lateral transduction. Science 2018, 362, 207–212. [Google Scholar] [CrossRef] [Green Version]
  41. Humphrey, S.; Fillol-Salom, A.; Quiles-Puchalt, N.; Ibarra-Chavez, R.; Haag, A.F.; Chen, J.; Penades, J.R. Bacterial chromosomal mobility via lateral transduction exceeds that of classical mobile genetic elements. Nat. Commun. 2021, 12, 6509. [Google Scholar] [CrossRef]
  42. Ramisetty, B.C.M.; Sudhakari, P.A. Bacterial ‘Grounded’ Prophages: Hotspots for Genetic Renovation and Innovation. Front. Genet. 2019, 10, 65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Hyman, P. Phages for Phage Therapy: Isolation, Characterization, and Host Range Breadth. Pharmaceuticals 2019, 12, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Cantalupo, P.G.; Calgua, B.; Zhao, G.; Hundesa, A.; Wier, A.D.; Katz, J.P.; Grabe, M.; Hendrix, R.W.; Girones, R.; Wang, D.; et al. Raw sewage harbors diverse viral populations. mBio 2011, 2, e00180-11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Domingo-Calap, P.; Beamud, B.; Vienne, J.; Gonzalez-Candelas, F.; Sanjuan, R. Isolation of Four Lytic Phages Infecting Klebsiella pneumoniae K22 Clinical Isolates from Spain. Int. J. Mol. Sci. 2020, 21, 425. [Google Scholar] [CrossRef] [Green Version]
  46. Hatfull, G.F. The secret lives of mycobacteriophages. Adv. Virus Res. 2012, 82, 179–288. [Google Scholar] [CrossRef] [PubMed]
  47. Onsea, J.; Soentjens, P.; Djebara, S.; Merabishvili, M.; Depypere, M.; Spriet, I.; De Munter, P.; Debaveye, Y.; Nijs, S.; Vanderschot, P.; et al. Bacteriophage Application for Difficult-to-treat Musculoskeletal Infections: Development of a Standardized Multidisciplinary Treatment Protocol. Viruses 2019, 11, 891. [Google Scholar] [CrossRef] [Green Version]
  48. Lin, R.C.; Sacher, J.C.; Ceyssens, P.J.; Zheng, J.; Khalid, A.; Iredell, J.R.; Australian Phage Biobanking Network. Phage Biobank: Present Challenges and Future Perspectives. Curr. Opin. Biotechnol. 2021, 68, 221–230. [Google Scholar] [CrossRef] [PubMed]
  49. Kropinski, A.M.; Mazzocco, A.; Waddell, T.E.; Lingohr, E.; Johnson, R.P. Enumeration of bacteriophages by double agar overlay plaque assay. Methods Mol. Biol. 2009, 501, 69–76. [Google Scholar] [CrossRef] [PubMed]
  50. Joao, J.; Lampreia, J.; Prazeres, D.M.F.; Azevedo, A.M. Manufacturing of bacteriophages for therapeutic applications. Biotechnol. Adv. 2021, 49, 107758. [Google Scholar] [CrossRef]
  51. Jurac, K.; Nabergoj, D.; Podgornik, A. Bacteriophage production processes. Appl. Microbiol. Biotechnol. 2019, 103, 685–694. [Google Scholar] [CrossRef]
  52. Luong, T.; Salabarria, A.C.; Edwards, R.A.; Roach, D.R. Standardized bacteriophage purification for personalized phage therapy. Nat. Protoc. 2020, 15, 2867–2890. [Google Scholar] [CrossRef] [PubMed]
  53. Adriaenssens, E.M.; Lehman, S.M.; Vandersteegen, K.; Vandenheuvel, D.; Philippe, D.L.; Cornelissen, A.; Clokie, M.R.; Garcia, A.J.; De Proft, M.; Maes, M.; et al. CIM(R) monolithic anion-exchange chromatography as a useful alternative to CsCl gradient purification of bacteriophage particles. Virology 2012, 434, 265–270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Fortier, L.C.; Moineau, S. Phage production and maintenance of stocks, including expected stock lifetimes. Methods Mol. Biol. 2009, 501, 203–219. [Google Scholar] [CrossRef] [PubMed]
  55. Zierdt, C.H. Stabilities of lyophilized Staphylococcus aureus typing bacteriophages. Appl. Environ. Microbiol. 1988, 54, 2590. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Bourdin, G.; Schmitt, B.; Marvin Guy, L.; Germond, J.E.; Zuber, S.; Michot, L.; Reuteler, G.; Brussow, H. Amplification and purification of T4-like Escherichia coli phages for phage therapy: From laboratory to pilot scale. Appl. Environ. Microbiol. 2014, 80, 1469–1476. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Puapermpoonsiri, U.; Ford, S.J.; van der Walle, C.F. Stabilization of bacteriophage during freeze drying. Int. J. Pharm. 2010, 389, 168–175. [Google Scholar] [CrossRef] [PubMed]
  58. Tovkach, F.I.; Zhuminska, G.I.; Kushkina, A.I. Long-term preservation of unstable bacteriophages of enterobacteria. Mikrobiol. Z. 2012, 74, 60–66. [Google Scholar]
  59. 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]
  60. Sarker, S.A.; Brussow, H. From bench to bed and back again: Phage therapy of childhood Escherichia coli diarrhea. Ann. N. Y. Acad. Sci. 2016, 1372, 42–52. [Google Scholar] [CrossRef] [PubMed]
  61. Bruttin, A.; Brussow, H. Human volunteers receiving Escherichia coli phage T4 orally: A safety test of phage therapy. Antimicrob. Agents Chemother. 2005, 49, 2874–2878. [Google Scholar] [CrossRef] [Green Version]
  62. Sarker, S.A.; McCallin, S.; Barretto, C.; Berger, B.; Pittet, A.C.; Sultana, S.; Krause, L.; Huq, S.; Bibiloni, R.; Bruttin, A.; et al. Oral T4-like phage cocktail application to healthy adult volunteers from Bangladesh. Virology 2012, 434, 222–232. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  63. Chang, R.Y.K.; Morales, S.; Okamoto, Y.; Chan, H.K. Topical application of bacteriophages for treatment of wound infections. Transl. Res. 2020, 220, 153–166. [Google Scholar] [CrossRef] [PubMed]
  64. Ryan, E.M.; Gorman, S.P.; Donnelly, R.F.; Gilmore, B.F. Recent advances in bacteriophage therapy: How delivery routes, formulation, concentration and timing influence the success of phage therapy. J. Pharm. Pharmacol. 2011, 63, 1253–1264. [Google Scholar] [CrossRef] [PubMed]
  65. Eliava BioPreparations. Available online: https://phage.ge/products/?lang=en (accessed on 16 February 2022).
  66. Loh, B.; Gondil, V.S.; Manohar, P.; Khan, F.M.; Yang, H.; Leptihn, S. Encapsulation and Delivery of Therapeutic Phages. Appl. Environ. Microbiol. 2020, 87, e01979-20. [Google Scholar] [CrossRef] [PubMed]
  67. McConnell, E.L.; Fadda, H.M.; Basit, A.W. Gut instincts: Explorations in intestinal physiology and drug delivery. Int. J. Pharm. 2008, 364, 213–226. [Google Scholar] [CrossRef]
  68. Colom, J.; Cano-Sarabia, M.; Otero, J.; Arinez-Soriano, J.; Cortes, P.; Maspoch, D.; Llagostera, M. Microencapsulation with alginate/CaCO3: A strategy for improved phage therapy. Sci. Rep. 2017, 7, 41441. [Google Scholar] [CrossRef]
  69. 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]
  70. Duplessis, C.A.; Biswas, B. A Review of Topical Phage Therapy for Chronically Infected Wounds and Preparations for a Randomized Adaptive Clinical Trial Evaluating Topical Phage Therapy in Chronically Infected Diabetic Foot Ulcers. Antibiotics 2020, 9, 377. [Google Scholar] [CrossRef]
  71. Merabishvili, M.; Monserez, R.; van Belleghem, J.; Rose, T.; Jennes, S.; De Vos, D.; Verbeken, G.; Vaneechoutte, M.; Pirnay, J.P. Stability of bacteriophages in burn wound care products. PLoS ONE 2017, 12, e0182121. [Google Scholar] [CrossRef] [Green Version]
  72. Chang, R.Y.K.; Wallin, M.; Lin, Y.; Leung, S.S.Y.; Wang, H.; Morales, S.; Chan, H.K. Phage therapy for respiratory infections. Adv. Drug Deliv. Rev. 2018, 133, 76–86. [Google Scholar] [CrossRef]
  73. Bodier-Montagutelli, E.; Morello, E.; L’Hostis, G.; Guillon, A.; Dalloneau, E.; Respaud, R.; Pallaoro, N.; Blois, H.; Vecellio, L.; Gabard, J.; et al. Inhaled phage therapy: A promising and challenging approach to treat bacterial respiratory infections. Expert Opin. Drug Deliv. 2017, 14, 959–972. [Google Scholar] [CrossRef] [PubMed]
  74. Malik, D.J. Bacteriophage Encapsulation Using Spray Drying for Phage Therapy. Curr. Issues Mol. Biol. 2021, 40, 303–316. [Google Scholar] [CrossRef] [PubMed]
  75. Golshahi, L.; Lynch, K.H.; Dennis, J.J.; Finlay, W.H. In vitro lung delivery of bacteriophages KS4-M and PhiKZ using dry powder inhalers for treatment of Burkholderia cepacia complex and Pseudomonas aeruginosa infections in cystic fibrosis. J. Appl. Microbiol. 2011, 110, 106–117. [Google Scholar] [CrossRef] [PubMed]
  76. Speck, P.; Smithyman, A. Safety and efficacy of phage therapy via the intravenous route. FEMS Microbiol. Lett. 2016, 363, fnv242. [Google Scholar] [CrossRef] [PubMed]
  77. Ujmajuridze, A.; Chanishvili, N.; Goderdzishvili, M.; Leitner, L.; Mehnert, U.; Chkhotua, A.; Kessler, T.M.; Sybesma, W. Adapted Bacteriophages for Treating Urinary Tract Infections. Front. Microbiol. 2018, 9, 1832. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Letkiewicz, S.; Lusiak-Szelachowska, M.; Miedzybrodzki, R.; Zaczek, M.; Weber-Dabrowska, B.; Gorski, A. Low Immunogenicity of Intravesical Phage Therapy for Urogenitary Tract Infections. Antibiotics 2021, 10, 627. [Google Scholar] [CrossRef]
  79. Pirnay, J.P.; Blasdel, B.G.; Bretaudeau, L.; Buckling, A.; Chanishvili, N.; Clark, J.R.; Corte-Real, S.; Debarbieux, L.; Dublanchet, A.; De Vos, D.; et al. Quality and safety requirements for sustainable phage therapy products. Pharm. Res. 2015, 32, 2173–2179. [Google Scholar] [CrossRef] [Green Version]
  80. Philipson, C.W.; Voegtly, L.J.; Lueder, M.R.; Long, K.A.; Rice, G.K.; Frey, K.G.; Biswas, B.; Cer, R.Z.; Hamilton, T.; Bishop-Lilly, K.A. Characterizing Phage Genomes for Therapeutic Applications. Viruses 2018, 10, 188. [Google Scholar] [CrossRef] [Green Version]
  81. Hoper, D.; Freuling, C.M.; Muller, T.; Hanke, D.; von Messling, V.; Duchow, K.; Beer, M.; Mettenleiter, T.C. High definition viral vaccine strain identity and stability testing using full-genome population data--The next generation of vaccine quality control. Vaccine 2015, 33, 5829–5837. [Google Scholar] [CrossRef]
  82. McCallin, S.; Sarker, S.A.; Sultana, S.; Oechslin, F.; Brussow, H. Metagenome analysis of Russian and Georgian Pyophage cocktails and a placebo-controlled safety trial of single phage versus phage cocktail in healthy Staphylococcus aureus carriers. Environ. Microbiol. 2018, 20, 3278–3293. [Google Scholar] [CrossRef]
  83. Villarroel, J.; Larsen, M.V.; Kilstrup, M.; Nielsen, M. Metagenomic Analysis of Therapeutic PYO Phage Cocktails from 1997 to 2014. Viruses 2017, 9, 328. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Rajnovic, D.; Munoz-Berbel, X.; Mas, J. Fast phage detection and quantification: An optical density-based approach. PLoS ONE 2019, 14, e0216292. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Abate, W.; Sattar, A.A.; Liu, J.; Conway, M.E.; Jackson, S.K. Evaluation of recombinant factor C assay for the detection of divergent lipopolysaccharide structural species and comparison with Limulus amebocyte lysate-based assays and a human monocyte activity assay. J. Med. Microbiol. 2017, 66, 888–897. [Google Scholar] [CrossRef] [PubMed]
  86. Schwarz, H.; Gornicec, J.; Neuper, T.; Parigiani, M.A.; Wallner, M.; Duschl, A.; Horejs-Hoeck, J. Biological Activity of Masked Endotoxin. Sci. Rep. 2017, 7, 44750. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Shintani, H. Validation Study of Rapid Assays of Bioburden, Endotoxins and Other Contamination. Biocontrol Sci. 2016, 21, 63–72. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Patey, O.; McCallin, S.; Mazure, H.; Liddle, M.; Smithyman, A.; Dublanchet, A. Clinical Indications and Compassionate Use of Phage Therapy: Personal Experience and Literature Review with a Focus on Osteoarticular Infections. Viruses 2018, 11, 18. [Google Scholar] [CrossRef] [Green Version]
  89. Brussow, H. Hurdles for Phage Therapy to Become a Reality-An Editorial Comment. Viruses 2019, 11, 557. [Google Scholar] [CrossRef] [Green Version]
  90. 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] [PubMed] [Green Version]
  91. Leitner, L.; Ujmajuridze, A.; Chanishvili, N.; Goderdzishvili, M.; Chkonia, I.; Rigvava, S.; Chkhotua, A.; Changashvili, G.; McCallin, S.; Schneider, M.P.; et al. Intravesical bacteriophages for treating urinary tract infections in patients undergoing transurethral resection of the prostate: A randomised, placebo-controlled, double-blind clinical trial. Lancet Infect. Dis. 2021, 21, 427–436. [Google Scholar] [CrossRef]
  92. Pirnay, J.P.; Kutter, E. Bacteriophages: It’s a medicine, Jim, but not as we know it. Lancet Infect. Dis. 2021, 21, 309–311. [Google Scholar] [CrossRef]
  93. Pirnay, J.P.; Merabishvili, M.; Van Raemdonck, H.; De Vos, D.; Verbeken, G. Bacteriophage Production in Compliance with Regulatory Requirements. Methods Mol. Biol. 2018, 1693, 233–252. [Google Scholar] [CrossRef] [PubMed]
  94. Pirnay, J.P.; Verbeken, G.; Ceyssens, P.J.; Huys, I.; De Vos, D.; Ameloot, C.; Fauconnier, A. The Magistral Phage. Viruses 2018, 10, 64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Fauconnier, A. Regulating phage therapy: The biological master file concept could help to overcome regulatory challenge of personalized medicines. EMBO Rep. 2017, 18, 198–200. [Google Scholar] [CrossRef] [PubMed]
  96. Verbeken, G.; Pirnay, J.P.; Lavigne, R.; Jennes, S.; De Vos, D.; Casteels, M.; Huys, I. Call for a dedicated European legal framework for bacteriophage therapy. Arch. Immunol. Ther. Exp. 2014, 62, 117–129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Pelfrene, E.; Sebris, Z.; Cavaleri, M. Comment on Fauconnier, A. Phage Therapy Regulation: From Night to Dawn. Viruses 2019, 11, 771. [Google Scholar] [CrossRef] [Green Version]
  98. Harper, D.R. Criteria for Selecting Suitable Infectious Diseases for Phage Therapy. Viruses 2018, 10, 177. [Google Scholar] [CrossRef] [Green Version]
  99. Wright, A.; Hawkins, C.H.; Anggard, 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]
  100. Djebara, S.; Maussen, C.; De Vos, D.; Merabishvili, M.; Damanet, B.; Pang, K.W.; De Leenheer, P.; Strachinaru, I.; Soentjens, P.; Pirnay, J.P. Processing Phage Therapy Requests in a Brussels Military Hospital: Lessons Identified. Viruses 2019, 11, 265. [Google Scholar] [CrossRef] [Green Version]
  101. Lood, C.; Boeckaerts, D.; Stock, M.; De Baets, B.; Lavigne, R.; van Noort, V.; Briers, Y. Digital phagograms: Predicting phage infectivity through a multilayer machine learning approach. Curr. Opin. Virol. 2021, 52, 174–181. [Google Scholar] [CrossRef]
  102. Farrell, P.M. The prevalence of cystic fibrosis in the European Union. J. Cyst. Fibros. 2008, 7, 450–453. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Lopez-Causape, C.; de Dios-Caballero, J.; Cobo, M.; Escribano, A.; Asensio, O.; Oliver, A.; Del Campo, R.; Canton, R.; Sole, A.; Cortell, I.; et al. Antibiotic resistance and population structure of cystic fibrosis Pseudomonas aeruginosa isolates from a Spanish multi-centre study. Int. J. Antimicrob. Agents 2017, 50, 334–341. [Google Scholar] [CrossRef] [PubMed]
  104. Furukawa, B.S.; Flume, P.A. Nontuberculous Mycobacteria in Cystic Fibrosis. Semin. Respir. Crit. Care Med. 2018, 39, 383–391. [Google Scholar] [CrossRef] [PubMed]
  105. Osmani, M.; Sotello, D.; Alvarez, S.; Odell, J.A.; Thomas, M. Mycobacterium abscessus infections in lung transplant recipients: 15-year experience from a single institution. Transpl. Infect. Dis. 2018, 20, e12835. [Google Scholar] [CrossRef]
  106. Beam, E.; Osmon, D. Prosthetic Joint Infection Update. Infect. Dis. Clin. N. Am. 2018, 32, 843–859. [Google Scholar] [CrossRef]
  107. Hoyle, N.; Zhvaniya, P.; Balarjishvili, N.; Bolkvadze, D.; Nadareishvili, L.; Nizharadze, D.; Wittmann, J.; Rohde, C.; Kutateladze, M. Phage therapy against Achromobacter xylosoxidans lung infection in a patient with cystic fibrosis: A case report. Res. Microbiol. 2018, 169, 540–542. [Google Scholar] [CrossRef] [PubMed]
  108. Dedrick, R.M.; Guerrero-Bustamante, C.A.; Garlena, R.A.; Russell, D.A.; Ford, K.; Harris, K.; Gilmour, K.C.; Soothill, J.; Jacobs-Sera, D.; Schooley, R.T.; et al. Engineered bacteriophages for treatment of a patient with a disseminated drug-resistant Mycobacterium abscessus. Nat. Med. 2019, 25, 730–733. [Google Scholar] [CrossRef] [PubMed]
  109. Law, N.; Logan, C.; Yung, G.; Furr, C.L.; Lehman, S.M.; Morales, S.; Rosas, F.; Gaidamaka, A.; Bilinsky, I.; Grint, P.; et al. Successful adjunctive use of bacteriophage therapy for treatment of multidrug-resistant Pseudomonas aeruginosa infection in a cystic fibrosis patient. Infection 2019, 47, 665–668. [Google Scholar] [CrossRef]
  110. Lebeaux, D.; Merabishvili, M.; Caudron, E.; Lannoy, D.; Van Simaey, L.; Duyvejonck, H.; Guillemain, R.; Thumerelle, C.; Podglajen, I.; Compain, F.; et al. A Case of Phage Therapy against Pandrug-Resistant Achromobacter xylosoxidans in a 12-Year-Old Lung-Transplanted Cystic Fibrosis Patient. Viruses 2021, 13, 60. [Google Scholar] [CrossRef]
  111. Aslam, S.; Courtwright, A.M.; Koval, C.; Lehman, S.M.; Morales, S.; Furr, C.L.; Rosas, F.; Brownstein, M.J.; Fackler, J.R.; Sisson, B.M.; et al. Early clinical experience of bacteriophage therapy in 3 lung transplant recipients. Am. J. Transpl. 2019, 19, 2631–2639. [Google Scholar] [CrossRef]
  112. Tan, X.; Chen, H.; Zhang, M.; Zhao, Y.; Jiang, Y.; Liu, X.; Huang, W.; Ma, Y. Clinical Experience of Personalized Phage Therapy Against Carbapenem-Resistant Acinetobacter baumannii Lung Infection in a Patient with Chronic Obstructive Pulmonary Disease. Front. Cell. Infect. Microbiol 2021, 11, 631585. [Google Scholar] [CrossRef] [PubMed]
  113. Ferry, T.; Boucher, F.; Fevre, C.; Perpoint, T.; Chateau, J.; Petitjean, C.; Josse, J.; Chidiac, C.; L’Hostis, G.; Leboucher, G.; et al. Innovations for the treatment of a complex bone and joint infection due to XDR Pseudomonas aeruginosa including local application of a selected cocktail of bacteriophages. J. Antimicrob. Chemother. 2018, 73, 2901–2903. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  114. Ferry, T.L.G.; Fevre, C.; Herry, Y.; Conrad, A.; Josse, J.; Batailler, C.; Chidiac, C.; Medina, M.; Lustig, S.; Laurent, F.; et al. Salvage Debridement, Antibiotics and Implant Retention (“DAIR”) with Local Injection of a Selected Cocktail of Bacteriophages: Is It an Option for an Elderly Patient with Relapsing Staphylococcus aureus Prosthetic-Joint Infection. Open Forum Infect. Dis. 2018, 5, ofy269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Tkhilaishvili, T.; Winkler, T.; Muller, M.; Perka, C.; Trampuz, A. Bacteriophages as Adjuvant to Antibiotics for the Treatment of Periprosthetic Joint Infection Caused by Multidrug-Resistant Pseudomonas aeruginosa. Antimicrob. Agents Chemother. 2019, 64, e00924-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Fish, R.; Kutter, E.; Bryan, D.; Wheat, G.; Kuhl, S. Resolving Digital Staphylococcal Osteomyelitis Using Bacteriophage-A Case Report. Antibiotics 2018, 7, 87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Vázquez, R.; García, E.; García, P. Phage lysins for fighting bacterial respiratory infections: A new generation of antimicrobials. Front. Immunol. 2018, 9, 2252. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Gutierrez, D.; Briers, Y. Lysins breaking down the walls of Gram-negative bacteria, no longer a no-go. Curr. Opin. Biotechnol. 2021, 68, 15–22. [Google Scholar] [CrossRef] [PubMed]
  119. Pastagia, M.; Schuch, R.; Fischetti, V.A.; Huang, D.B. Lysins: The arrival of pathogen-directed anti-infectives. J. Med. Microbiol. 2013, 62, 1506–1516. [Google Scholar] [CrossRef] [PubMed]
  120. Dams, D.; Briers, Y. Enzybiotics: Enzyme-based antibacterials as therapeutics. Adv. Exp. Med. Biol. 2019, 1148, 233–253. [Google Scholar] [CrossRef]
  121. Schuch, R.; Lee, H.M.; Schneider, B.C.; Sauve, K.L.; Law, C.; Khan, B.K.; Rotolo, J.A.; Horiuchi, Y.; Couto, D.E.; Raz, A.; et al. Combination therapy with lysin CF-301 and antibiotic is superior to antibiotic alone for treating methicillin-resistant Staphylococcus aureus-induced murine bacteremia. J. Infect. Dis. 2014, 209, 1469–1478. [Google Scholar] [CrossRef]
  122. Loeffler, J.M.; Djurkovic, S.; Fischetti, V.A. Phage lytic enzyme Cpl-1 as a novel antimicrobial for pneumococcal bacteremia. Infect. Immun. 2003, 71, 6199–6204. [Google Scholar] [CrossRef] [Green Version]
  123. Rashel, M.; Uchiyama, J.; Ujihara, T.; Uehara, Y.; Kuramoto, S.; Sugihara, S.; Yagyu, K.; Muraoka, A.; Sugai, M.; Hiramatsu, K.; et al. Efficient elimination of multidrug-resistant Staphylococcus aureus by cloned lysin derived from bacteriophage phi MR11. J. Infect. Dis. 2007, 196, 1237–1247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Fischetti, V.A. Bacteriophage endolysins: A novel anti-infective to control Gram-positive pathogens. Int. J. Med. Microbiol. 2010, 300, 357–362. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Vasina, D.V.; Antonova, N.P.; Grigoriev, I.V.; Yakimakha, V.S.; Lendel, A.M.; Nikiforova, M.A.; Pochtovyi, A.A.; Remizov, T.A.; Usachev, E.V.; Shevlyagina, N.V.; et al. Discovering the Potentials of Four Phage Endolysins to Combat Gram-Negative Infections. Front. Microbiol. 2021, 12, 748718. [Google Scholar] [CrossRef] [PubMed]
  126. Lysando. Available online: https://www.lysando.com/systemic-applications.html (accessed on 16 February 2022).
  127. Ghose, C.; Euler, C.W. Gram-Negative Bacterial Lysins. Antibiotics 2020, 9, 74. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Gerstmans, H.; Grimon, D.; Gutierrez, D.; Lood, C.; Rodriguez, A.; van Noort, V.; Lammertyn, J.; Lavigne, R.; Briers, Y. A VersaTile-driven platform for rapid hit-to-lead development of engineered lysins. Sci. Adv. 2020, 6, eaaz1136. [Google Scholar] [CrossRef] [PubMed]
  129. ContraFect. Contrafect Announces First Gram-Negative Product Candidate CF_370, a Direct Lytic Agent Targeting Pseudomonas aeruginosa. Globe Newswire, 18 December 2019. [Google Scholar]
  130. Jun, S.Y.; Jang, I.J.; Yoon, S.; Jang, K.; Yu, K.S.; Cho, J.Y.; Seong, M.W.; Jung, G.M.; Yoon, S.J.; Kang, S.H. Pharmacokinetics and tolerance of the phage endolysin-based candidate drug SAL200 after a single intravenous administration among healthy volunteers. Antimicrob. Agents Chemother. 2017, 61, e02629-02616. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Fowler, V.G., Jr.; Das, A.F.; Lipka-Diamond, J.; Schuch, R.; Pomerantz, R.; Jauregui-Peredo, L.; Bressler, A.; Evans, D.; Moran, G.J.; Rupp, M.E.; et al. Exebacase for patients with Staphylococcus aureus bloodstream infection and endocarditis. J. Clin. Investig. 2020, 130, 3750–3760. [Google Scholar] [CrossRef] [Green Version]
  132. Abdelkader, K.; Gerstmans, H.; Saafan, A.; Dishisha, T.; Briers, Y. The preclinical and clinical progress of bacteriophages and their lytic enzymes: The parts are easier than the whole. Viruses 2019, 11, 96. [Google Scholar] [CrossRef] [Green Version]
  133. ContraFect. Available online: https://www.contrafect.com/pipeline/exebacase (accessed on 21 February 2022).
  134. Intron Biotechnology. Phase IIa Clinical Study of N-Rephasin® SAL200. Available online: https://clinicaltrials.gov/ct2/show/NCT03089697 (accessed on 21 February 2022).
  135. ContraFect. Direct Lysis of Staph Aureus Resistant Pathogen Trial of Exebacase (DISRUPT). Available online: https://www.clinicaltrials.gov/ct2/show/NCT04160468?term=CF-301&draw=1 (accessed on 21 February 2022).
  136. Palaniappan, R.; Dayanithi, G. Therapeutic Efficacy of Bacteriophages. In Bacteriophages in Therapeutics; IntechOpen: London, UK, 2021. [Google Scholar]
  137. Keswani, C. Agri-Based Bioeconomy: Reintegrating Trans-Disciplinary Research and Sustainable Development Goals, 1st ed.; CRC Press: Boca Raton, FL, USA, 2021. [Google Scholar]
  138. Bacteriophage Market by Product Type (dsDNA Bacteriophage, ssDNA Bacteriophage, ssRNA Bacteriophage), by Application (Clinical Application, Food & Beverages, Phage Display, Phage Therapy, Environmental Application, Veterinary)-Growth, Future Prospects & Competitive Analysis, 2018–2026. ID: 16969; 2018. Available online: https://www.credenceresearch.com/report/bacteriophage-market (accessed on 21 February 2022).
  139. Global Antimicrobial Resistance Market Size, Trends & Growth Opportunity, by Disease, by Pathogen, by Drug Class, by Region and Forecast till 2027. ID: 5398519; August 2021. Available online: https://www.researchandmarkets.com/reports/5398519/global-antimicrobial-resistance-market-size (accessed on 21 February 2022).
  140. Fauconnier, A. Phage Therapy Regulation: From Night to Dawn. Viruses 2019, 11, 352. [Google Scholar] [CrossRef] [Green Version]
  141. Telum Therapeutics. Available online: https://telumtherapeutics.com/our-science/pipeline/ (accessed on 20 January 2022).
  142. EIT Health. Available online: https://eithealth.eu/news-article/phagomed-acquired-by-biontech/ (accessed on 21 February 2022).
  143. PR Newswire. Available online: https://www.prnewswire.com/news-releases/micreos-secures-32-million-for-its-endolysin-based-platform-as-sustainable-alternative-to-antibiotics-301388573.html (accessed on 21 February 2022).
Figure 1. Steps in preparation of phage suspensions suitable for phage therapy, including screening, propagations, purification, storage, and formulation.
Figure 1. Steps in preparation of phage suspensions suitable for phage therapy, including screening, propagations, purification, storage, and formulation.
Microorganisms 10 00717 g001
Table 1. Ongoing clinical trial examples involving phage therapy.
Table 1. Ongoing clinical trial examples involving phage therapy.
DiseasePathogen(s)TreatmentStatusReferences
Diabetic foot ulcersStaphylococcus aureusTopical phage cocktailNot yet recruiting (expected start date: June 2022)NCT02664740
Invasive infection in patients with inactive Crohn’s diseaseE. coliOral phage cocktailRecruiting (estimated completion: June 2023)NCT03808103
Chronic airway infection in cystic fibrosis patientsP. aeruginosaNebulized phage therapyRecruiting (estimated completion: December 2022)NCT04684641
Diabetic foot ulcersP. aeruginosa, S. aureus and/or Acinetobacter baumanniiTopical phage cocktailRecruiting (estimated completion: December 2021NCT04803708
Prosthetic joint infectionsSeveral pathogensCombined antibiotic/personalized phage therapyNot yet recruiting (estimated start date: October 2022)NCT04787250
Chronic airway infection in cystic fibrosis patientsP. aeruginosaNebulized phage cocktailNot yet recruitingNCT05010577
Wound infections in burned patientsS. aureus, P. aeruginosa or Klebsiella pneumoniaeTopical phage cocktailNot yet recruiting (estimated start date: January 2022)NCT04323475
Pressure injury infectionsS. aureus, P. aeruginosa, K. pneumoniaeTopical phage cocktail in combination with antibioticsNot yet recruiting (estimated start date: January 2022)NCT04815798
Urinary tract infectionsE. coli or K. pneumoniaePersonalized phage therapy administered through intravenous or intravesical routeRecruiting (estimated completion: September 2023)NCT04287478
TonsillitisSeveral pathogensNebulized phage cocktailPhase 3. Active, not recruiting (estimated completion: December 2024)NCT04682964
Chronic airway infection in cystic fibrosis patientsP. aeruginosaInhaled phage cocktailRecruiting (estimated completion: March 2022)NCT04596319
Table 2. Examples of clinical cases treated with phage therapy.
Table 2. Examples of clinical cases treated with phage therapy.
DiseasePathogen(s)TreatmentOutcomeReferences
CF with chronic MDR lung infectionAchromobacter
xylosoxidans
Inhalation, orallyDyspnea resolved and cough reduced. Lung function improved[107]
CF with disseminated infection, lung transplantationM. abscessusIntravenousSternal wound closure, improved liver function, substantial resolution of infected skin nodules[108]
CF with MDR pneumonia, persistent respiratory failure, and colistin-induced renal failureP. aeruginosaIntravenousPneumonia clinically resolved, no sputum production, return to baseline renal function, white blood cell count normalized[109]
CF with persistent lung infection, lung transplantationA. xylosoxidansInhalationRespiratory condition improved; sputum cultures positive but with low bacteria concentration[110]
Lung transplant recipient patients with MRD resistant infectionsP. aeruginosa and Burkholderia dolosaIntravenous, inhalationTwo patients were discharged from the hospital off ventilator support. A third patient infection relapsed and died[111]
COPD with drug-resistant pneumoniaA. baumanniiInhalationSputum/ blood and bronchoalveolar lavage fluid negative, restoration sinus rhythm, lung function improved[112]
Prosthesis infectionS. aureusLocalBacteria removed, rapid healing[113]
OsteomyelitisP. aeruginosaLocalNo clinical signs of persistent infection[114]
Infection of the right knee and chronic osteomyelitis of the femur after injuryP. aeruginosaLocalNo pain, soft tissue at the surgical site unremarkable, mobility satisfactory[115]
Osteomyelitis of the distal phalanxS. aureusLocalThe ulcer healed, re-ossification of the distal phalanx, erythema and edema decreased[116]
Fracture-related infectionK. pneumoniaeLocalSkin graft vascularized and viable, the sinus tract closed and dry, pus no longer discharged from the pin sites of the external fixator, restored muscle function[20]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Vázquez, R.; Díez-Martínez, R.; Domingo-Calap, P.; García, P.; Gutiérrez, D.; Muniesa, M.; Ruiz-Ruigómez, M.; Sanjuán, R.; Tomás, M.; Tormo-Mas, M.Á.; et al. Essential Topics for the Regulatory Consideration of Phages as Clinically Valuable Therapeutic Agents: A Perspective from Spain. Microorganisms 2022, 10, 717. https://doi.org/10.3390/microorganisms10040717

AMA Style

Vázquez R, Díez-Martínez R, Domingo-Calap P, García P, Gutiérrez D, Muniesa M, Ruiz-Ruigómez M, Sanjuán R, Tomás M, Tormo-Mas MÁ, et al. Essential Topics for the Regulatory Consideration of Phages as Clinically Valuable Therapeutic Agents: A Perspective from Spain. Microorganisms. 2022; 10(4):717. https://doi.org/10.3390/microorganisms10040717

Chicago/Turabian Style

Vázquez, Roberto, Roberto Díez-Martínez, Pilar Domingo-Calap, Pedro García, Diana Gutiérrez, Maite Muniesa, María Ruiz-Ruigómez, Rafael Sanjuán, María Tomás, María Ángeles Tormo-Mas, and et al. 2022. "Essential Topics for the Regulatory Consideration of Phages as Clinically Valuable Therapeutic Agents: A Perspective from Spain" Microorganisms 10, no. 4: 717. https://doi.org/10.3390/microorganisms10040717

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