Next Article in Journal
Human and Companion Animal Proteus mirabilis Sharing
Previous Article in Journal
Influence of Maternal Microbiome and Inflammatory Response in Preterm Birth: Recent Aspects of the Prevention of Preterm Birth
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The History and Applications of Phage Therapy in Pseudomonas aeruginosa

1
Laboratory of Medical & Industrial Biotechnology (LABMI), Porto Research, Technology & Innovation Center (PORTIC), R. Arquitecto Lobão Vital 172, 4200-374 Porto, Portugal
2
School of Health (ESS), Polytechnic Institute of Porto (IPP), R. Dr. António Bernardino de Almeida 400, 4200-072 Porto, Portugal
3
Hospital Santa Maria Maior, Campo da República 59, 4750-333 Barcelos, Portugal
4
Faculty of Biology, University of Vigo, Lagoas-Marcosende, s/n, 36310 Vigo, Spain
5
School of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
6
School of Medicine and Biomedical Sciences, R. Jorge de Viterbo Ferreira 228, 4050-313 Porto, Portugal
7
School of Medicine, University of Santiago de Compostela, Rúa de San Francisco, s/n, 15782 Santiago de Compostela, Spain
8
School Biotechnology, Portuguese Catholic University, R. de Diogo Botelho 1327, 4169-005 Porto, Portugal
*
Authors to whom correspondence should be addressed.
Microbiol. Res. 2022, 13(1), 14-37; https://doi.org/10.3390/microbiolres13010002
Submission received: 1 October 2021 / Revised: 21 December 2021 / Accepted: 22 December 2021 / Published: 28 December 2021

Abstract

:
The Pseudomonas aeruginosa is one of the bacteria that cause serious infections due to resistance to many antibiotics can be fatal in severe cases. Antimicrobial resistance is a global public health concern. To solve this problem, interest in phage therapy has revived; some studies are being developed to try to prove the effectiveness of this therapy. Thus, in this opinion article, several historical aspects are addressed as well some applications of phage therapy against P. aeruginosa.

1. Introduction

Bacterial resistance to antibiotics is a public health problem that raises concern worldwide, with both clinical and economic implications, and is associated with inappropriate use of antibiotics. The United Nations Organization (UNO) estimates about 10 million deaths per year in the 2050s. For this reason, new therapeutic strategies, beside traditional antibiotics, must be urgently developed. One of these strategies can be the use of bacteriophages (phages).
Phages are viruses that infect bacteria. These are the most ubiquitous and diversified biological group residing on Earth. They have high specificity for the host and high permanency in natural systems [1]. Due to their obligate requirement of a bacterial host, phages are abundantly found distributed essentially anywhere their host exists in the biosphere. Their abundance in nature was estimated as ten to hundreds of millions of phages in every gram of soil, water, and billions on and inside the human body at any moment [2].
Phages can also be used as therapeutic agents [3]. In contrast to antibiotics, phage therapy specifically lyses the host bacteria and does not affect non-host bacteria [4]. After the inactivation of the host pathogenic bacteria, the phage abundance is in proportion to that of the host pathogenic bacteria and thus, when the host bacteria diminish, the phage count also decreases, which maintains the microbial stability and diversity [3].
Due to the high rates of resistance of Pseudomonas aeruginosa to antibiotics, there is a growing concern about finding alternatives to antibiotics, such as phages. As of March 2019, there were about 137 phages sequenced and published in a public bank, targeting P. aeruginosa. Most of these phages belong to the Caudovirales family [5].
The specific phages for Pseudomonas been described in the mid-20th century [6].

2. Pseudomonas Aeruginosa

P. aeruginosa is a Gram-negative bacillus, belong Pseudomonadaceae family, included in the gammaproteobacteria class, mono-flagellated, straight or slightly curved [7]. It has basic nutritional needs, is non-fermenting carbohydrates, and has a high tolerance for stress, physical and chemical factors [7].
High number of virulence factors, such as enzymes that degrade effector molecules of the immune system and elements essential to the host’s cellular and tissue structure, such as proteases and elastases, are present in P. aeruginosa. It also includes phenazine and its derivatives, such as pyocyanin, which are associated with the alteration of states of cellular oxidation, inter-cellular communication, and regulation of gene expression, assuming a crucial role in cell survival, especially in the context of infection. Pyoverdin, a greenish pigment, is a siderophore that mediates the absorption of iron ions, an essential element for the survival and growth of the organism [7]. It is also important to highlight the presence of membrane transport systems, namely type III secretion systems, which, despite not being virulence factors themselves, mediate the efflux of proteins responsible for the destruction of epithelial barriers and host cells, and interfere with the activity of macrophages and neutrophils.
This bacterium is able to alternate between planktonic lifestyle and growth in biofilm, which helps to survive in specific niches, such as catheters and several hospital surfaces, giving it a greater tolerance for disinfectants and antibiotics. It is also associated with chronic colonization of internal devices and prosthetic material, as well as the lungs of patients with cystic fibrosis or bronchiectasis [8]. The polar flagellum and pili type IV are involved in the formation of biofilm, and they mediate the capacity for mobility and are relevant in adherence to host tissues [9].
The genome of P. aeruginosa is relatively large (5.5–7 Mbp) compared to other sequenced bacteria such as Bacillus subtilis (4.2 Mbp), Escherichia coli (4.6 Mbp) and Mycobacterium tuberculosis (4.4 Mbp) and encodes a large proportion of regulatory enzymes important for the metabolism, transport and efflux of organic compounds. This coding capability of P. aeruginosa to the genome allows great metabolic versatility and high adaptability to environmental changes [10]. P. aeruginosa has been recognized as an opportunistic pathogen that is the most common bacterium associated with healthcare-associated infections (HAI) such ventilated-associated pneumonia (VAP), one of the most common HAI [10] It rarely affects healthy individuals, but causes high morbidity and mortality in patients with cystic fibrosis (CF) and immunocompromised individuals [10].
With the increase of antimicrobial resistance and the tendency to evolve to multiple resistance when in the presence of antibiotics for long time, it is necessary to understand the resistance mechanisms at their disposal, the means of evolution and dissemination, and to find alternative methods [11].
When subjected to selective antibiotic pressure, the induced response facilitates bacterial survival and develops antimicrobial resistance [12].

The Problem of Bacterial Resistance in P. aeruginosa

Portugal is a country with a high consumption of antibiotics, and antimicrobial resistance has grown sharply. The development of resistance is a natural process due to the selective pressure exerted by the use of antibiotics, and it has undergone a progression due to the inappropriate use of antibiotics [12].
P. aeruginosa developed the ability to resist to several antibiotics and it became a challenge to treat this type of infection [10]. The WHO recently revealed its antimicrobial resistance to carbapenems, emerging the development of new therapies in the treatment of this type of infections [10].
P. aeruginosa has shown high resistance to a range of antibiotics, including aminoglycosides, quinolones, and β-lactams. The excessive use of antibiotics culminates in the increase of resistance of this strain and consequent inefficiency of the result of the empirical antibiotherapy [13].
The main resistance mechanisms are intrinsic, acquired, and adaptive resistance. Intrinsic resistance is done through the low permeability of the outer membrane, expression of efflux pumps inactivating enzymes and production of antibiotics. Acquired resistance may occur for horizontal transfer of resistance genes or gene mutational changes. Adaptive resistance is the ability of bacteria to adapt to the antibiotic creating mechanisms to protect against it [13].
The most important mechanisms of resistance to β-lactams is the production of β-lactamase enzymes, however, the resistance can also be caused by overexpression of efflux systems by changing the membrane permeability and the synthesis of binding protein to penicillin (PBPs) with low affinity for β-lactams. The chromosomal β-lactamase AmpC is intrinsic to this species and confers resistance to some β-lactamase [14].
Β-lactamases can inactivate β-lactam antimicrobials by breaking the β-lactam ring, by destroying the amide bond. The hydrolysis of β-lactam antibiotics occurs by the formation of an ester bond between the active serine site (or with zinc ions, in the case of metallo-β-lactamases) of the β-lactamase enzyme and the β-lactamic ring of the antimicrobial. In P. aeruginosa these enzymes are present in the periplasmic space and inactivate the β-lactamic after they cross the outer membrane, before binding to PBPs [15].
A large part of the genes encoding the β-lactamases enzymes are in mobile regions of bacterial DNA, such as plasmids and class 1 integrons, which contribute to the increase in bacterial resistance. The most frequently acquired β-lactamases are pseudomonas-specific enzymes (PSEs) PSE-1 and PSE-4.
Transferases are important in resistance to aminoglycosides, since catalyse acetylation, adenylation or phosphorylation of antibiotic inactivating it. Methylases are also relevant in resistance to these pharmacological agents because transform the 30S ribosomal subunit and prevent the binding of clinically relevant compounds such as gentamicin, tobramycin and amikacin [14].
Chromosomal gene mutations such as depression of the ampC gene, loss of the OrpD porin and horizontal gene transfer are extremely important in conferring resistance to antibiotics. The genes responsible for the expression of β-lactamases or aminoglycoside-modifying enzymes are acquired by natural transformation processes. Generally, these genes are inserted in integrations, genetic elements that capture exogenous genes and guarantee their expression, inside plasmids or transposons, which allow their transfer between cells. At P. aeruginosa, these integrations contribute to the development of multidrug resistance, since they rarely include only one element of resistance to antibiotics [16].

3. Bacteriophages

3.1. History of Phage Therapy

Several visionary scientists contribute over the time to the development of what is now referred of phage therapy (Figure 1). This was a very long history that benefits from the contribution of many.
In 1896, Ernest Hanbury Hankin, a British bacteriologist working as the Chemical Examiner and Bacteriologist to the Government of the United Provinces and of the Central Provinces of India, demonstrated the presence in two rivers in India, the Ganga River and the Yamuna River, of an unidentified substance with high activity against Vibrio cholerae, limiting the spread of the cholera epidemic [17,18,19].
In 1915, Frederick Twort, a British microbiologist, observed and described transparent plaques in Staphylococcus cultures that appeared successively in different strains after appropriate filtration [21]. Twort published an article describing a filterable agent responsible for bacterial lysis, raising the hypothesis that it is a virus [21].
Two years after this report, Felix d’Herelle, a microbiologist at the Pasteur Institute in Paris, was commissioned to investigate a serious outbreak of haemorrhagic dysentery of bacterial origin in French troops. From faecal patient samples, he obtained a bacteria-free filtrate which was later incubated with Shigella spp. He observed a result similar to that described by Twork, which he named lysis plates [18].
In 1917, Felix d’Herelle, proposed that the phenomenon was caused by an obligate intracellular parasite able of parasitizing bacteria, for the first-time naming bacteriophages [22]. During this year, were isolated phages able to lysepathogenic bacteria as Salmonella typhi, Escherichia coli, Pasteurella multocida, Vibrio cholerae, Yersinia pestis, Streptococcus species, Pseudomonas aeruginosa and Neisseria meningitidis [23].
In 1923 was raised in Georgia the Eliava Institute (EIBMV), by d’Herelle e Georgi Elliava.
During World War II, certain regions of the Soviet Union and Eastern Europe had limited access to antibiotics and, consequently, developed phage therapy. The practice of phage therapy in the Soviet Union has been well advised and is still widely used in Russia and Eastern European countries for more than 80 years, especially at the Eliava Institute in Tbilisi, Georgia [2].
In 1952, at the Hirszfed Institute, a program was developed to treat phage patients with suppurative infections [24]. In the 1980s, controlled studies with animals were published in the English scientific literature. In recent years, some Western European countries have started approved therapeutic use [25].
In 2016, Paul Turner and colleagues reported isolating a phage that could restore antibiotic sensitivity in multidrug-resistant P. aeruginosa. This phage was later used to treat a patient with a longstanding aortic graft infection that did not respond to multiple surgical interventions and aggressive antibiotic therapy with a single application of phage [26].

3.2. Phage Structure and Taxonomy

Phage vary in their complexity, structure, genetic material, shape (tail filaments and icosahedral), and size [27]. The genome is composed of single or double stranded DNA or RNA of very variable length, arranged in a linear or circular shape. Its size varies from a few thousand base pairs to 498 kilobase (Kb) pairs in G phage, the largest phage sequenced up to the moment [2,28].
Most bacteriophages are composed of an icosahedral head associated with a helical symmetry protein tail [28]. The head is formed by the capsid, constituted by repeated structural protein subunits that surrounds the nucleic acid. The capsid protects the nucleic acid and possesses proteins which confer specificity for certain bacterial cells [29]. The neck connects the head to the tail, a heteroligomer composed of various proteins that ensure genome release when the virion is bound to the host cell.
The distal portion of the tail has a basal plate to which tail fibers and spicules are attached which have proteins capable of binding to membrane receptors of certain bacteria [28]. The tail may have different contractility and dimensions, may exhibit accessory structures such as spicules, collar, lipid envelope or absence of envelope [28].
Virus taxonomy is currently the responsibility of the International Committee on the Taxonomy of Viruses (ICTV), and the Bacterial and Archaeal Subcommittee (BAVS) within the ICTV that focuses on phages [30] which published its first report in 1971 [29]. The system is based on the evaluation of a variety of phage properties including the molecular composition of the virus genome (ss/ds, DNA, or RNA), the structure of the virus capsid and whether it is enveloped or not, the host range, pathogenicity, and sequence similarity [30].
Phages are classified through a structural and sequence-based taxonomic system (Table 1); initially into families, and each family is further categorized in accordance to the capsid structure, the structural and chemical composition of the genes and the mechanism of their mRNA production [2].
Phages can be classified in to Siphoviridae, Podoviridae, Myoviridae, and Filamentous phages (Table 1), according to morphology and nucleic acid (International Committee on Taxonomy of Viruses, ICTV) [3]. Virulent tailed phages of the Caudovirales order have been the best described for phage therapeutic applications. Within this group, the Myoviridae have a large capsid head and contractile tail, the Siphoviridae have a relatively small capsid and a long flexible non-contractile tail, and the Podoviridae have a small capsid head and short tail [1].

3.3. Phage Infection Process

Phages are mandatory intracellular parasites do not have their own metabolism, for this reason, requiring the metabolism, energy resources and material resources of the hosts to replicate. They need to bind to specific locations on the host’s surface. This connection is always necessary regardless of its propagation cycle, which can be classified as lytic, tempered and chronic phages [2].
The life cycle of the phage may differ only lytic or lysogenic, depending on the type of phage and the physiological state of the bacterial cell.

3.3.1. Nucleic Acid Recognition, Adsorption, and Injection

This stage is common to both life cycles of bacteriophages.
In the first stage of the infection process, the phage recognizes the receptors present on the bacterial surface. A variety of components present in the bacterium, such as flagella, pill, capsule, LPS and proteins, are potential receptors for phages [31,32]. The initial contact with the bacteria occurs through Brownian movements of the viral particle [31].
Subsequently, a reversible bond occurs, mediated by the tail fibres [33]. Then the specific and irreversible binding of the Bio Protein Receptor (RBPs) of the phage with the target receptors of the bacterium, promotes a change in the conformation of the viral particle allowing the release of its genome through the capsid [34], thus transferring the nucleic acid into the bacterial cytoplasm.
Phages that have contractile tails can penetrate the cell wall and inject the nucleic acid directly into the cytoplasm to bacterium [32]. After the Phage introduces its nucleic acid into the bacterial cell, the life cycle may diverge to lithic cycle or lysogenic cycle, depending on the type of phage and the physiological state of the bacterium. If the phage is virulent, it initiates a lytic cycle, which will promote cell lysis. If the phage is temperate, these have genes that regulate the two types of cycles; the type of cycle that will occur may be influenced by various factors [32].

3.3.2. Lytic Cycle

In the lytic cycle (Figure 2), phages bind to specific receptors on the surface of the host cell by injecting its genome. After the entry of the nucleic acid, viral transcription begins, through RNA polymerase [32].
Subsequently, the synthesis of structural and catalytic proteins occurs with the capacity to form the structures of new phages. Genes are expressed to encode the synthesis of holin and lysine, enzymes that have the capacity to change the cytoplasmic membrane and the bacterial cell wall, that is responsible for promoting cell lysis, and consequent release the newly formed viral particles, which has the capacity to infect other bacteria [35].

3.3.3. Lysogenic Cycle

Tempered viruses integrate their genome into the host chromosome or plasmid (Figure 2), which is replicated whenever the cell reproduces. The incorporated genome, also called a prophages, can activate the lytic cycle in the cell. The phage remains in a latent state and replicates its DNA together with that of the bacteria [32] and this state of latency is not irreversible, occasionally these bacteria can enter the lytic cycle.
The factors that lead to the activation of the temperate phage lytic cycle are not well known, but it is known that factors causing stress in the host cell or causing DNA damage, have been shown to induce temperate phage lytic cycle.
In phage infection, the host cell is helpless against the invading virus. However, bacteria have defense mechanisms against phages. In response, phages developed a series of methods to circumvent these mechanisms [36].

3.4. Phage-Host Interactions

Phages attach to the bacterial surface through the adsorption process. Bacteria have resistance mechanisms that impede the phage adsorption process; however, phages are able to adapt their binding proteins to the receptor through a mutation [36].
Pseudomonas spp. has a great capacity for biofilm formation, due to a range of polysaccharides extracellular and other biofilm-forming components. After infection, phages are able to prevent superinfection with other phages, modulating the expression of cell receptors. After adsorption, phages inject their DNA into the host cell. However, many bacteria encode restriction endonucleases to cut foreign DNA and prevent phage infection. In turn, the phages developed anti-restriction strategies to defend their DNA against cleavage by restriction endonucleases [37].
A different antiviral mechanism that protects the host through the degradation of foreign DNA is the CRISPR-Cas System which acts as an adaptive bacterial immune system. Us CRISPR-Cas systems, the system is activated when the virus enters the bacterium: this recognizes the exogenous DNA and enzymes (Cas) cut pieces of this material and introduce them into a specific genomic region of the bacterium, called the CRISPR locus. In the next viral infections, the bacteria that contain these pieces of virus DNA inserted into the CRISPR locus generate an RNA from this sequence. This RNA will associate with the Cas enzyme and then make its way to the viral DNA, which is then cleaved and thus inactivated [38].
Phages have conceived ways to disable CRISPR-Cas systems (Figure 3). In total, 10 different families of anti-CRISPR proteins (Acr) were identified in Pseudomonas temperate phages. Phages appear able to escape CRISPR interference through specific mutations [39].

3.5. Advantages and Disadvantages of Phage Therapy

Phage therapy has several advantages over antibiotics treatment for bacterial infections [40], as shown in Table 2:
Despite the numerous advantages of phage therapy, challenges and limitations exist and must be considered.
Some of the concerns of using this therapy are: (i) Most infections are polymicrobial, and it is necessary to use an effective and efficient phage cocktail, with a greater spectrum of action [2]; (ii) Lack of approval by the Food and Drug Administration (FDA) or the European Medicines Agency (EMA), due to concerns about phage resistance and the role of phage in the evolution of the bacterial genome; (iii) Phages as evolution drivers: unlike antibiotics, they are biological agents based on proteins that contain DNA/RNA that have the potential to interact with the body’s immune system and other microbial cells in the body and can actively replicate and evolve inside the body. This evolution can, in turn, result in the evolution of the bacterial communities of the commensal host and possibly even affect the composition of the niche microbiome; (iv) Phage selection criteria: factors such as the phage’s ability to infect bacteria in stationary phase, phage enzymes, stability to serum inactivation and mutation rate have proved to be important and deserve further investigation; (v) Lack of well-organized public phage libraries [2,41].

3.6. Phage Therapy Applications

For phage therapy to be a viable alternative to antibiotics, clear efficacy data from randomized controlled clinical trials are needed. In recent years, several clinical trials have been carried out, but only a few are concluded [6].
In 2009, the first controlled clinical trial of a therapeutic bacteriophage preparation was carried out, which showed efficacy and safety in antibiotic-resistant P. aeruginosa chronic otitis [42]. Also in 2009, another randomized, double-blind controlled trial addressed the safety of a phage cocktail targeting P. aeruginosa, Staphylococcus aureus and Escherichia coli for the treatment of venous leg ulcers [6].
Between 2013 and 2017 PhagoBurn, funded by the European Commission, was the first clinical trial of phage therapy using good production practices (GMP) [2].
Sextaphage is one such commercial pharmaceutical phage composition from the Russian company Microgen. This phage therapeutic contains phages against six specific pathogens, with the intent of treating urinary tract infections in pregnant women [2].
In 2016, Paul Turner and colleagues reported isolating a phage that could restore antibiotic sensitivity in multidrug-resistant P. aeruginosa. This phage was later used to treat a patient with a longstanding aortic graft infection that did not respond to multiple surgical interventions and aggressive antibiotic therapy with a single application of phage. The same team recently reported the treatment of two cystic fibrosis patients with antibiotic-resistant infections. Aside from the wild type phage isolated from natural sources, genetically engineered phages have also been reported for phage therapy. A young patient with cystic fibrosis and bilateral lung transplantation who had developed a Mycobacterium abscessus infection was reportedly treated with a phage cocktail of genetically engineered lytic phages that were administered intravenously [43].
In the United States, they successfully utilized intravenous phage therapy to treat a patient with a severe systemic infection caused by multidrug-resistant organisms. The phages used in that case were isolated from various environmental samples [43].
In France, the national health regulator has authorized the first treatment of patients with extremely drug-resistant and difficult to treat infections using phage therapy. Since then, six cases with various bacterial infections have been successfully treated [44].
In January 2018, the Federal Government of Belgium, in cooperation with researchers and specialists from the Federal Agency for Medicines and Health Products, took an important step in phage therapy, developing a regulation for the production and clinical application of phages. The procedure is based on the legal authorization of a pharmacist to prepare a medical product with phages individually. The active ingredients used in its preparation must meet the requirements of the European, Belgian or other official Pharmacopoeia [45].
According to several reviews, there are many advantages to using phages as antibacterial therapy [25].

3.7. Phage Therapy and P. aeruginosa, Studies In Vivo and In Vitro

Several in vitro studies have been carried out in recent years to assess the efficacy of phages on P. aeruginosa, including multiresistant strains [5].
Pseudomonas-specific phages were first described in the mid-20th century [5]. There are about 137 sequenced phages targeting P. aeruginosa in public databases, most of them from the Caudovirales order [46].
In 2010, a study was published on the effects of lytic phages in preventing P. aeruginosa biofilm formation in hydrogel-coated catheters [47]. Another study used a combination of phage Podoviridae LUZ7 and streptomycin on P. aeruginosa PAO1 [48].
At the beginning of the year 2019, the result of the first clinical study on phage therapy directed at P. aeruginosa was reported. This study consisted of using an anti-P.aeruginosa phage cocktail for burn patients. The test result showed that pre-determined stutterers might not be the solution. Some strains of P. aeruginosa will not be susceptible or will quickly become resistant to the phages administered. In order to make phage therapy effective, it is necessary to formulate phage cocktail in a personalized way [49].
Some clinical trials were carried out using phages for P. aeruginosa. A study evaluated the efficacy and safety of a therapeutic phage preparation (Biophage-PA) [5].
Another study used a topical application of phage in a burn patient colonized with P. aeruginosa [5].
One study used a BFC-1 phage cocktail to treat nine patients with acute burns colonized by multidrug-resistant P. aeruginosa and Staphylococcus aureus [50].
Table 3, Table 4 and Table 5 describe some in vivo and in vitro and clinical studies carried out in recent years to test the effectiveness of phage therapy against Pseudomonas aeruginosa [46,49,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93].
Regarding in vitro studies, to date, four have been used as models of cystic fibrosis, chronic rhinosinusitis cancer and burn wound infection. The main bacterial strains used were PAO1 with one without pilin (PAO1-NP). Various phages were used, namely Pa193, Pa204, Pa222, Pa223, PEV1, PEV2, PEV20, PEV61, DMS3, vB_PaeM_GUMS6, vB_PaeM_GUMS32, and vB_PaeM_GUMS45 with application ranging from 4 to 48 h and the main result is significant reduction of multidrug resistant (MDR) P. aeruginosa by phages PEV1, PEV20, and PEV61 in cystic fibrosis. DMS3 and PEV2 phages were able to inhibit bacterial growth in chronic rhinosinusitis. It is also known that a single dose of phage is able to significantly reduce biofilms formed in vitro. In burn wound infections, the combination of two phage-antibiotics (vB_PaeM_GUMS6, vB_PaeM_GUMS32, vB_PaeM_GUMS45) had the highest inhibition efficiency against the P. aeruginosa strain.
Regarding in vivo studies, to date the mice has been the most used animal model followed by Galleria mellonella and wax moth. Different pathologies were studied, being CF the most used model followed by COPD and bacterial induced endocarditis. All phages showed positive results, in some cases even after one dose, reducing the bacterial burden and in some cases curing the bacterial infection, all the phages’ cocktails studied showed were capable of curing the infections, in some cases this were complemented with antibiotic therapy. The studies duration ranged from 1 h to 56 days.
The clinical trials showed positive results in every trial. In the trials that phage therapy was complemented with antibiotics all patients healed completely and the bacterial infection were eradicated, when the phage therapy was used alone, not all patients got cured but all showed a reduction in bacterial burden and improvement in the healing process. The age of the patient doesn’t seem to be an important variant in the therapy results, the results were also identical in all pathologies. All studies were done using Pseudomonas clinical isolates and the duration varied from 5 days to 9 months.

4. Conclusions

The significant increase in antibiotic resistant strains and the scarcity of new antibiotics require, in the immediate future, the discovery of new effective therapies, such as phage therapy.
Their diversity, low production cost, low capacity for adverse effects, and their specificity for target cells make phages very attractive.
P. aeruginosa is responsible for serious infections, and over the last few years it has shown high levels of resistance to antibiotics on the market, being one of the emerging bacteria in terms of discovering new effective therapies.
Although there are some studies that demonstrate the clinical success of phage therapy, more studies are needed on the safety, efficacy, and even development of phage resistance as an alternative therapy in P. aeruginosa infections, to solve the serious problem of resistance and lack of alternatives to antibiotics on the market today. Additionally, phages can be used in combination with antibiotics or other antimicrobials for improved performance.
The growing interest in phage therapy by patients and physicians and the consequent increase in phage orders from around the world highlights a growing need for the establishment of phage banks with well-characterized phages that can facilitate access by the international community.

Author Contributions

Conceptualization: C.S.; P.B.; R.F.; Data curation: C.S.; Formal analysis: P.B.; R.F.; Funding acquisition: P.B.; R.F.; Investigation: C.S.; S.S.; C.G.; C.O.; C.L.; M.O.; J.M.; G.N.; P.B.; R.F.; Methodology: C.S.; S.S.; C.G.; C.O.; C.L.; M.O.; J.M.; G.N.; Project administration: P.B.; R.F.; Supervision: P.B.; R.F.; Validation: P.B.; R.F.; Writing—original draft: C.S.; S.S.; C.G.; C.O.; C.L.; M.O.; J.M.; G.N.; Writing—review & editing: P.B.; R.F. All authors have read and agreed to the published version of the manuscript.

Funding

A scholarship with the reference PORTIC/LaBMI/IPP4COVID/01/2020 was endorsed to Carla Guedes and Sara Sá. A scholarship with the reference PORTIC/LaBMI/IPP4COVID/01/2021 was endorsed to Marco Oliveira. A scholarship with the reference PORTIC/LaBMI/PHAGE4TECH/01/2021 was endorsed to João Mendes. A scholarship with the reference PORTIC/LaBMI/CELEGANS4TECH/01/2021 was endorsed to Gonçalo Novais. A grant with the reference FEDER/02/SAICT/2020/072560 was earned by Ruben Fernandes.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

Authors declare no conflict of interest.

References

  1. Romero-Calle, D.; Guimarães Benevides, R.; Góes-Neto, A.; Billington, C. Bacteriophages as Alternatives to Antibiotics in Clinical Care. Antibiotics 2019, 8, 138. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Divya Ganeshan, S.; Hosseinidoust, Z. Phage Therapy with a Focus on the Human Microbiota. Antibiotics 2019, 8, 131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Ye, M.; Sun, M.; Huang, D.; Zhang, Z.; Zhang, H.; Zhang, S.; Hu, F.; Jiang, X.; Jiao, W. A review of bacteriophage therapy for pathogenic bacteria inactivation in the soil environment. Environ. Int. 2019, 129, 488–496. [Google Scholar] [CrossRef]
  4. Kakasis, A.; Panitsa, G. Bacteriophage therapy as an alternative treatment for human infections. A comprehensive review. Int. J. Antimicrob. Agents 2019, 53, 16–21. [Google Scholar] [CrossRef]
  5. Pires, D.P.; Vilas Boas, D.; Sillankorva, S.; Azeredo, J. Phage Therapy: A Step Forward in the Treatment of Pseudomonas aeruginosa Infections. J. Virol. 2015, 89, 7449–7456. [Google Scholar] [CrossRef] [Green Version]
  6. Pires, D.P.; Costa, A.R.; Pinto, G.; Meneses, L.; Azeredo, J. Current challenges and future opportunities of phage therapy. FEMS Microbiol. Rev. 2020, 44, 684–700. [Google Scholar] [CrossRef] [PubMed]
  7. Marques, D.R.A. Prevalência e Susceptibilidade de Isolados Clínicos de Pseudomonas aeruginosa numa unidade hospitalar de Portugal. Ph.D. Thesis, University of Minho, Braga, Portugal, 2015. [Google Scholar]
  8. Kerr, K.G.; Snelling, A.M. Pseudomonas aeruginosa: A formidable and ever-present adversary. J. Hosp. Infect. 2009, 73, 338–344. [Google Scholar] [CrossRef]
  9. Hernandez, E.; Segundo, N.V. Los bacteriófagos como una alternativa en el tratamiento de enfermedades infecciosas Bacterianas (Fagoterapia). Rev. Mex. Ciencias Farm. 2010, 41, 17–26. [Google Scholar]
  10. Pang, Z.; Raudonis, R.; Glick, B.R.; Lin, T.J.; Cheng, Z. Antibiotic resistance in Pseudomonas aeruginosa: Mechanisms and alternative therapeutic strategies. Biotechnol. Adv. 2019, 37, 177–192. [Google Scholar] [CrossRef]
  11. Pereira, S.G.; Rosa, A.; Ferreira, A.; Moreira, L.; Proença, D.; Morais, P.V.; Cardoso, O. Virulence factors and infection ability of Pseudomonas aeruginosa isolates from a hydropathic facility and respiratory infections. J. Appl. Microbiol. 2014, 116, 1359–1368. [Google Scholar] [CrossRef] [Green Version]
  12. Loureiro, R.J.; Roque, F.; Teixeira Rodrigues, A.; Herdeiro, M.T.; Ramalheira, E. Use of antibiotics and bacterial resistances: Brief notes on its evolution. Rev. Port. Saúde Publica 2016, 34, 77–84. [Google Scholar]
  13. Pachori, P.; Gothalwal, R.; Gandhi, P. Emergence of antibiotic resistance Pseudomonas aeruginosa in intensive care unit; a critical review. Genes Dis. 2019, 6, 109–119. [Google Scholar] [CrossRef] [PubMed]
  14. Moradali, M.F.; Ghods, S.; Rehm, B.H.A. Pseudomonas aeruginosa Lifestyle: A Paradigm for Adaptation, Survival, and Persistence. Front. Cell Infect. Microbiol. 2017, 7, 39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Fernandes, R.; Amador, P.; Prudencio, C. β-lactams: Chemical structure, mode of action and mechanisms of resistance. Rev. Med. Microbiol. 2013, 24, 7–17. [Google Scholar] [CrossRef] [Green Version]
  16. Tagliaferri, T.L.; Jansen, M.; Horz, H.-P. Fighting Pathogenic Bacteria on Two Fronts: Phages and Antibiotics as Combined Strategy. Front. Cell Infect. Microbiol. 2019, 9, 2. [Google Scholar] [CrossRef]
  17. Wittebole, X.; De Roock, S.; Opal, S.M. A historical overview of bacteriophage therapy as an alternative to antibiotics for the treatment of bacterial pathogens. Virulence 2014, 5, 226–235. [Google Scholar] [CrossRef]
  18. Sulakvelidze, A.; Alavidze, Z.; Morris, J.G., Jr. Bacteriophage therapy. Antimicrob Agents Chemother. 2001, 45, 649–659. [Google Scholar] [CrossRef] [Green Version]
  19. Haq, I.U.; Chaudhry, W.N.; Akhtar, M.N.; Andleeb, S.; Qadri, I. Bacteriophages and their implications on future biotechnology: A review. Virol. J. 2012, 9, 9. [Google Scholar] [CrossRef] [Green Version]
  20. Green, S.; Ma, L.; Maresso, A. Phage therapy. Encycl. Microbiol. 2019, 1915, 485–495. [Google Scholar]
  21. Twort, F.W. An investigation on the nature of ultra-microscopic viruses. Lancet. 1915, 186, 1241–1243. [Google Scholar] [CrossRef] [Green Version]
  22. Lawrence, D.; Baldridge, M.T.; Handley, S.A. Phages and Human Health: More Than Idle Hitchhikers. Viruses 2019, 11, 587. [Google Scholar] [CrossRef] [Green Version]
  23. Fruciano, D.E.; Bourne, S. Phage as an antimicrobial agent: d’Herelle’s heretical theories and their role in the decline of phage prophylaxis in the West. Can. J. Infect. Dis. Med. Microbiol. 2007, 18, 19–26. [Google Scholar] [CrossRef] [PubMed]
  24. Hyman, P.; Abedon, S.T. Bacteriophage: Overview. In Schmidt TMBT-E of M; Fourth, E., Ed.; Oxford Academic Press: Oxford, UK, 2019; pp. 441–457. [Google Scholar] [CrossRef]
  25. Nikolich, M.P.; Filippov, A.A. Bacteriophage Therapy: Developments and Directions. Antibiotics 2020, 9, 135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. 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]
  27. Keen, E.C. A century of phage research: Bacteriophages and the shaping of modern biology. Bioessays 2015, 37, 6–9. [Google Scholar] [CrossRef] [PubMed]
  28. White, H.E. Bacteriophages: Their Structural Organisation and Function. In Bacteriophages-Perspectives and Future; Savva, E.-R., Ed.; IntechOpen: Rijeka, Croatia, 2020. [Google Scholar] [CrossRef] [Green Version]
  29. Adriaenssens, E.M.; Rodney Brister, J. How to name and classify your phage: An informal guide. Viruses 2017, 9, 70. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Chibani, C.M.; Farr, A.; Klama, S.; Dietrich, S.; Liesegang, H. Classifying the Unclassified: A Phage Classification Method. Viruses 2019, 11, 195. [Google Scholar] [CrossRef] [Green Version]
  31. Koskella, B.; Meaden, S. Understanding bacteriophage specificity in natural microbial communities. Viruses 2013, 5, 806–823. [Google Scholar] [CrossRef] [Green Version]
  32. Rakhuba, D.V.; Kolomiets, E.I.; Dey, E.; Novik, G.I. Bacteriophage Receptors, Mechanisms of Phage Adsorption and Penetration into Host Cell. Pol. J. Microbiol. 2010, 59, 145–155. [Google Scholar] [CrossRef]
  33. Chiaruttini, N.; de Frutos, M.; Augarde, E.; Boulanger, P.; Letellier, L.; Viasnoff, V. Is the In Vitro Ejection of Bacteriophage DNA Quasistatic? A Bulk to Single Virus Study. Biophys J. 2010, 99, 447–455. [Google Scholar] [CrossRef] [Green Version]
  34. Kropinski, A.M. Phage Therapy-Everything Old is New Again. Can. J. Infect. Dis. Med. Microbiol. 2006, 17, 297–306. [Google Scholar] [CrossRef] [PubMed]
  35. De Smet, J.; Hendrix, H.; Blasdel, B.G.; Danis-Wlodarczyk, K.; Lavigne, R. Pseudomonas predators: Understanding and exploiting phage-host interactions. Nat. Rev. Microbiol. 2017, 15, 517–530. [Google Scholar] [CrossRef] [PubMed]
  36. Weigele, P.; Raleigh, E.A. Biosynthesis and Function of Modified Bases in Bacteria and Their Viruses. Chem. Rev. 2016, 116, 12655–12687. [Google Scholar] [CrossRef] [PubMed]
  37. Garneau, J.E.; Dupuis, M.-È.; Villion, M.; Romero, D.A.; Barrangou, R.; Boyaval, P.; Fremaux, C.; Horvath, P.; Magadan, A.H.; Moineau, S. The CRISPR/cas bacterial immune system cleaves bacteriophage and plasmid DNA. Nature 2010, 468, 67–71. [Google Scholar] [CrossRef] [PubMed]
  38. Bondy-Denomy, J.; Pawluk, A.; Maxwell, K.L.; Davidson, A.R. Bacteriophage genes that inactivate the CRISPR/Cas bacterial immune system. Nature 2013, 493, 429–432. [Google Scholar] [CrossRef] [Green Version]
  39. Le Rhun, A.; Escalera-Maurer, A.; Bratovič, M.; Charpentier, E. CRISPR-Cas in Streptococcus pyogenes. RNA Biol. 2019, 16, 380–389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  40. Principi, N.; Silvestri, E.; Esposito, S. Advantages and limitations of bacteriophages for the treatment of bacterial infections. Front. Pharmacol. 2019, 10, 513. [Google Scholar] [CrossRef] [Green Version]
  41. Wright, A.; Hawkins, C.H.; Änggård, E.E.; Harper, D.R. A controlled clinical trial of a therapeutic bacteriophage preparation in chronic otitis due to antibiotic-resistant Pseudomonas aeruginosa: A preliminary report of efficacy. Clin. Otolaryngol. 2009, 34, 349–357. [Google Scholar] [CrossRef]
  42. Chan, B.K.; Turner, P.E.; Kim, S.; Mojibian, H.R.; Elefteriades, J.A.; Narayan, D. Phage treatment of an aortic graft infected with Pseudomonas aeruginosa. Evol. Med. Public Health 2018, 2018, 60–66. [Google Scholar] [CrossRef] [Green Version]
  43. Kutter, E.; De Vos, D.; Gvasalia, G.; Alavidze, Z.; Gogokhia, L.; Kuhl, S.; Abedon, S.T. Phage Therapy in Clinical Practice: Treatment of Human Infections. Curr. Pharm. Biotechnol. 2010, 11, 69–86. [Google Scholar] [CrossRef]
  44. Górski, A.; Międzybrodzki, R.; Łobocka, M.; Głowacka-Rutkowska, A.; Bednarek, A.; Borysowski, J.; Jończyk-Matysiak, E.; Łusiak-Szelachowska, M.; Weber-Dąbrowska, B.; Bagińska, N.; et al. Phage therapy: What have we learned? Viruses 2018, 10, 288. [Google Scholar] [CrossRef] [Green Version]
  45. Hoggarth, A.; Weaver, A.; Pu, Q.; Huang, T.; Schettler, J.; Chen, F.; Yuan, X.; Wu, M. Mechanistic research holds promise for bacterial vaccines and phage therapies for Pseudomonas aeruginosa. Drug Des. Dev. Ther. 2019, 13, 909–924. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Fu, W.; Forster, T.; Mayer, O.; Curtin, J.J.; Lehman, S.M.; Donlan, R.M. Bacteriophage cocktail for the prevention of biofilm formation by Pseudomonas aeruginosa on catheters in an in vitro model system. Antimicrob. Agents Chemother. 2010, 54, 397–404. [Google Scholar] [CrossRef] [Green Version]
  47. Torres-Barceló, C.; Arias-Sánchez, F.I.; Vasse, M.; Ramsayer, J.; Kaltz, O.; Hochberg, M.E. A window of opportunity to control the bacterial pathogen Pseudomonas aeruginosa combining antibiotics and phages. PLoS ONE 2014, 9, e106628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  48. Tümmler, B. Emerging therapies against infections with Pseudomonas aeruginosa [version 1; peer review: 2 approved]. F1000Research 2019, 8, 1–14. [Google Scholar] [CrossRef]
  49. Rose, T.; Verbeken, G.; De Vos, D.; Merabishvili, M.; Vaneechoutte, M.; Lavigne, R.; Jennes, S.; Zizi, M.; Pirnay, J.-P. Experimental phage therapy of burn wound infection: Difficult first steps. Int. J. Burns Trauma. 2014, 4, 66–73. [Google Scholar]
  50. Markwitz, P.; Olszak, T.; Gula, G.; Kowalska, M.; Arabski, M.; Drulis-Kawa, Z. Emerging Phage Resistance in Pseudomonas aeruginosa PAO1 Is Accompanied by an Enhanced Heterogeneity and Reduced Virulence. Viruses 2021, 13, 1332. [Google Scholar] [CrossRef]
  51. Tkhilaishvili, T.; Merabishvili, M.; Pirnay, J.P.; Starck, C.; Potapov, E.; Falk, V.; Schoenrath, F. Successful case of adjunctive intravenous bacteriophage therapy to treat left ventricular assist device infection. J. Infect. 2021, 83, e1–e3. [Google Scholar] [CrossRef]
  52. Antoine, C.; Laforêt, F.; Blasdel, B.; Glonti, T.; Kutter, E.; Pirnay, J.; Mainil, J.; Delcenserie, V.; Thiry, D. Efficacy assessment of PEV2 phage on Galleria mellonella larvae infected with a Pseudomonas aeruginosa dog otitis isolate. Res. Vet. Sci. 2021, 136, 598–601. [Google Scholar] [CrossRef]
  53. Aghaee, B.L.; Khan Mirzaei, M.; Alikhani, M.Y.; Mojtahedi, A.; Maurice, C.F. Improving the Inhibitory Effect of Phages against Pseudomonas aeruginosa Isolated from a Burn Patient Using a Combination of Phages and Antibiotics. Viruses 2021, 13, 334. [Google Scholar] [CrossRef] [PubMed]
  54. Krylov, V.; Bourkaltseva, M.; Pleteneva, E.; Shaburova, O.; Krylov, S.; Karaulov, A.; Zhavoronok, S.; Svitich, O.; Zverev, V. Phage phikz—The first of giants. Viruses 2021, 13, 149. [Google Scholar] [CrossRef] [PubMed]
  55. Alvi, I.A.; Asif, M.; ur Rehman, S. A single dose of a virulent bacteriophage vB PaeP-SaPL, rescues bacteremic mice infected with multi drug resistant Pseudomonas aeruginosa. Virus. Res. 2021, 292, 198250. [Google Scholar] [CrossRef] [PubMed]
  56. Ibrahim, D.; Jabbour, J.F.; Kanj, S.S. Current choices of antibiotic treatment for Pseudomonas aeruginosa infections. Curr. Opin. Infect. Dis. 2020, 33, 464–473. [Google Scholar] [CrossRef] [PubMed]
  57. Chow, M.Y.; Chang, R.Y.K.; Li, M.; Wang, Y.; Lin, Y.; Morales, S.; McLachlan, A.J.; Kutter, E.; Li, J.; Chan, H.K. Pharmacokinetics and time-kill study of inhaled antipseudomonal bacteriophage therapy in mice. Antimicrob. Agents Chemother. 2021, 65, 1–12. [Google Scholar] [CrossRef]
  58. Chegini, Z.; Khoshbayan, A.; Taati Moghadam, M.; Farahani, I.; Jazireian, P.; Shariati, A. Bacteriophage therapy against Pseudomonas aeruginosa biofilms: A review. Ann. Clin. Microbiol. Antimicrob. 2020, 19, 1–17. [Google Scholar] [CrossRef]
  59. McCallin, S.; Sarker, S.A.; Sultana, S.; Oechslin, F.; Brüssow, 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]
  60. Fujiki, J.; Furusawa, T.; Munby, M.; Kawaguchi, C.; Matsuda, Y.; Shiokura, Y.; Nakamura, K.; Nakamura, T.; Sasaki, M.; Usui, M.; et al. Susceptibility of Pseudomonas aeruginosa veterinary isolates to Pbunavirus PB1-like phages. Microbiol. Immunol. 2020, 64, 778–782. [Google Scholar] [CrossRef]
  61. Shao, X.; Xie, Y.; Zhang, Y.; Liu, J.; Ding, Y.; Wu, M.; Wang, X.; Deng, X. Novel therapeutic strategies for treating Pseudomonas aeruginosa infection. Expert Opin. Drug Discov. 2020, 15, 1403–1423. [Google Scholar] [CrossRef]
  62. Aslam, S. Bacteriophage therapy as a treatment option for transplant infections. Curr. Opin. Infect. Dis. 2020, 33, 298–303. [Google Scholar] [CrossRef]
  63. Lin, Y.-W.; Chang, R.Y.K.; Rao, G.G.; Jermain, B.; Han, M.-L.; Zhao, J.; Chen, K.; Wang, J.; Barr, J.J.; Schooley, R.T.; et al. Pharmacokinetics/pharmacodynamics of antipseudomonal bacteriophage therapy in rats: A proof-of-concept study. Clin. Microbiol. Infect. 2020, 26, 1229–1235. [Google Scholar] [CrossRef]
  64. Peng, H.; Borg, R.E.; Dow, L.P.; Pruitt, B.L.; Chen, I.A. Controlled phage therapy by photothermal ablation of specific bacterial species using gold nanorods targeted by chimeric phages. Proc. Natl. Acad. Sci. USA 2020, 117, 1951–1961. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Olszak, T.; Danis-Wlodarczyk, K.; Arabski, M.; Gula, G.; Maciejewska, B.; Wasik, S.; Lood, C.; Higgins, G.; Harvey, B.J.; Lavigne, R.; et al. Pseudomonas aeruginosa PA5oct Jumbo Phage Impacts Planktonic and Biofilm Population and Reduces Its Host Virulence. Viruses 2019, 11, 1089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Nilsson, A.S. Pharmacological limitations of phage therapy. Upsala J. Med. Sci. 2019, 124, 218–227. [Google Scholar] [CrossRef] [PubMed]
  67. Tkhilaishvili, T.; Winkler, T.; Müller, 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. 2020, 64, 1–5. [Google Scholar] [CrossRef] [Green Version]
  68. Maddocks, S.; Fabijan, A.P.; Ho, J.; Lin, R.; Ben Zakour, N.L.; Dugan, C.; Kliman, I.; Branston, S.; Morales, S.; Iredell, J.R. Bacteriophage therapy of ventilator-associated pneumonia and empyema caused by Pseudomonas aeruginosa. Am. J. Respir. Crit. Care Med. 2019, 200, 1179–1181. [Google Scholar] [CrossRef]
  69. Law, N.; Logan, C.; Yung, G.; Furr, C.-L.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]
  70. Gupta, P.; Singh, H.S.; Shukla, V.K.; Nath, G.; Bhartiya, S.K. Bacteriophage Therapy of Chronic Nonhealing Wound: Clinical Study. Int. J. Low. Extrem. Wounds 2019, 18, 171–175. [Google Scholar] [CrossRef]
  71. Yuan, Y.; Qu, K.; Tan, D.; Li, X.; Wang, L.; Cong, C.; Xiu, Z.; Xu, Y. Isolation and characterization of a bacteriophage and its potential to disrupt multi-drug resistant Pseudomonas aeruginosa biofilms. Microb. Pathog. 2019, 128, 329–336. [Google Scholar] [CrossRef]
  72. Abd El-Aziz, A.M.; Elgaml, A.; Ali, Y.M. Bacteriophage Therapy Increases Complement-Mediated Lysis of Bacteria and Enhances Bacterial Clearance after Acute Lung Infection with Multidrug-Resistant Pseudomonas aeruginosa. J. Infect. Dis. 2019, 219, 1439–1447. [Google Scholar] [CrossRef]
  73. 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]
  74. Breederveld, R.S. Phage therapy 2.0: Where do we stand? Lancet Infect. Dis. 2019, 19, 2–3. [Google Scholar] [CrossRef]
  75. Forti, F.; Roach, D.R.; Cafora, M.; Pasini, M.E.; Horner, D.S.; Fiscarelli, E.V.; Rossitto, M.; Cariani, L.; Briani, F.; Debarbieux, L.; et al. Design of a broad-range bacteriophage cocktail that reduces Pseudomonas aeruginosa biofilms and treats acute infections in two animal models. Antimicrob. Agents Chemother. 2018, 62, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Uchiyama, J.; Shigehisa, R.; Nasukawa, T.; Mizukami, K.; Takemura-Uchiyama, I.; Ujihara, T.; Murakami, H.; Imanishi, I.; Nishifuji, K.; Sakaguchi, M.; et al. Piperacillin and ceftazidime produce the strongest synergistic phage–antibiotic effect in Pseudomonas aeruginosa. Arch. Virol. 2018, 163, 1941–1948. [Google Scholar] [CrossRef]
  77. Międzybrodzki, R.; Borysowski, J.; Kłak, M.; Jończyk-Matysiak, E.; Obmińska-Mrukowicz, B.; Suszko-Pawłowska, A.; Bubak, B.; Weber-Dąbrowska, B.; Górski, A. In Vivo Studies on the Influence of Bacteriophage Preparations on the Autoimmune Inflammatory Process. BioMed Res. Int. 2017, 2017, 3612015. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. 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] [Green Version]
  79. Fong, S.A.; Drilling, A.; Morales, S.; Cornet, M.E.; Woodworth, B.A.; Fokkens, W.J.; Psaltis, A.J.; Vreugde, S.; Wormald, P.-J. Activity of Bacteriophages in Removing Biofilms of Pseudomonas aeruginosa Isolates from Chronic Rhinosinusitis Patients. Front. Cell Infect. Microbiol. 2017, 7, 418. [Google Scholar] [CrossRef] [Green Version]
  80. Shiley, J.R.; Comfort, K.K.; Robinson, J.B. Immunogenicity and antimicrobial effectiveness of Pseudomonas aeruginosa specific bacteriophage in a human lung in vitro model. Appl. Microbiol. Biotechnol. 2017, 101, 7977–7985. [Google Scholar] [CrossRef] [PubMed]
  81. Trend, S.; Fonceca, A.M.; Ditcham, W.G.; Kicic, A.C.F.; Cf, A. The potential of phage therapy in cystic fibrosis: Essential human-bacterial-phage interactions and delivery considerations for use in Pseudomonas aeruginosa-infected airways. J. Cyst. Fibros. 2017, 16, 663–670. [Google Scholar] [CrossRef] [Green Version]
  82. 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]
  83. Waters, E.M.; Neill, D.; Kaman, B.; Sahota, J.S.; Clokie, M.R.J.; Winstanley, C.; Kadioglu, A. Phage therapy is highly effective against chronic lung infections with Pseudomonas aeruginosa. Thorax 2017, 72, 666–667. [Google Scholar] [CrossRef] [Green Version]
  84. Al-Wrafy, F.; Brzozowska, E.; Górska, S.; Gamian, A. Pathogenic factors of Pseudomonas aeruginosa-the role of biofilm in pathogenicity and as a target for phage therapy. Adv. Hyg. Exp. Med. Hig. Med. Dosw. 2017, 71. [Google Scholar] [CrossRef] [PubMed]
  85. Stratton, C.W. Phages, fitness, virulence, and synergy: A novel approach for the therapy of infections caused by Pseudomonas aeruginosa. J. Infect. Dis. 2017, 215, 668–670. [Google Scholar] [CrossRef] [PubMed]
  86. Oechslin, F.; Piccardi, P.; Mancini, S.; Gabard, J.; Moreillon, P.; Entenza, J.M.; Resch, G.; Que, Y.-A. Synergistic interaction between phage therapy and antibiotics clears Pseudomonas aeruginosa infection in endocarditis and reduces virulence. J. Infect. Dis. 2017, 215, 703–712. [Google Scholar] [CrossRef] [Green Version]
  87. Høyland-Kroghsbo, N.M.; Paczkowski, J.; Mukherjee, S.; Broniewski, J.; Westra, E.; Bondy-Denomy, J.; Bassler, B.L. Quorum sensing controls the Pseudomonas aeruginosa CRISPR-Cas adaptive immune system. Proc. Natl. Acad. Sci. USA 2017, 114, 131–135. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Furusawa, T.; Iwano, H.; Hiyashimizu, Y.; Matsubara, K.; Higuchi, H.; Nagahata, H.; Niwa, H.; Katayama, Y.; Kinoshita, Y.; Hagiwara, K.; et al. Phage therapy is effective in a mouse model of bacterial equine keratitis. Appl. Environ. Microbiol. 2016, 82, 5332–5339. [Google Scholar] [CrossRef] [Green Version]
  89. Danis-Wlodarczyk, K.; Vandenheuvel, D.; Jang, H.B.; Briers, Y.; Olszak, T.; Arabski, M.; Wąsik, S.; Drabik, M.; Higgins, G.; Tyrrell, J.; et al. A proposed integrated approach for the preclinical evaluation of phage therapy in Pseudomonas infections. Sci Rep. 2016, 6, 1–13. [Google Scholar]
  90. Latz, S.; Wahida, A.; Arif, A.; Häfner, H.; Hoß, M.; Ritter, K.; Horz, H.P. Preliminary survey of local bacteriophages with lytic activity against multi-drug resistant bacteria. J. Basic Microbiol. 2016, 56, 1117–1123. [Google Scholar] [CrossRef]
  91. Khawaja, K.A.; Rauf, M.; Abbas, Z.; Rehman, S. A virulent phage JHP against Pseudomonas aeruginosa showed infectivity against multiple genera. J. Basic Microbiol. 2016, 56, 1090–1097. [Google Scholar] [CrossRef] [PubMed]
  92. Sagona, A.P.; Grigonyte, A.M.; MacDonald, P.R.; Jaramillo, A. Genetically modified bacteriophages. Integr. Biol. 2016, 8, 465–474. [Google Scholar] [CrossRef] [Green Version]
  93. Cafora, M.; Deflorian, G.; Forti, F.; Ferrari, L.; Binelli, G.; Briani, F.; Ghisotti, D.; Pistocchi, A. Phage therapy against Pseudomonas aeruginosa infections in a cystic fibrosis zebrafish model. Sci. Rep. 2019, 9, 1527. [Google Scholar] [CrossRef] [Green Version]
  94. Jang, H.-J.; Bae, H.-W.; Cho, Y.-H. Exploitation of Drosophila Infection Models to Evaluate Antibacterial Efficacy of Phages BT-Bacteriophages: Methods and Protocols; Clokie, M.R.J., Kropinski, A., Lavigne, R., Eds.; Springer: New York, NY, USA, 2019; pp. 183–190. [Google Scholar] [CrossRef]
  95. Chang, R.Y.K.; Wallin, M.; Kutter, E.; Morales, S.; Britton, W.; Li, J.; Chan, H.-K. Storage stability of inhalable phage powders containing lactose at ambient conditions. Int. J. Pharm. 2019, 560, 11–18. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Phage therapy timeline. Adapted from Green et al. [20].
Figure 1. Phage therapy timeline. Adapted from Green et al. [20].
Microbiolres 13 00002 g001
Figure 2. Lytic cycle and Lysogenic cycle. Lytic cycle: Immediately following injection into the host cell, the phage genome synthesizes early proteins that break down the host DNA, allowing the phage to take control of the cellular machinery. The phage then uses the host cell to synthesize the remaining proteins required to build new phage particles. The heads and sheaths are assembled separately, the new genetic material packed into the head and new daughter phage particles constructed. During this process, the host cells gradually become weakened by phage enzymes and eventually burst, releasing on average 100–200 new phage progeny into the surrounding environment. Lysogenic cycle: Following the injection of the phage DNA into the host cell, it integrates itself into the host genome, with the help of phage-encoded integrases, where it is then termed a prophage. The prophage genome is then replicated passively along with the host genome as the host cell divides for as long as it remains there and does not form the proteins required to produce progeny.
Figure 2. Lytic cycle and Lysogenic cycle. Lytic cycle: Immediately following injection into the host cell, the phage genome synthesizes early proteins that break down the host DNA, allowing the phage to take control of the cellular machinery. The phage then uses the host cell to synthesize the remaining proteins required to build new phage particles. The heads and sheaths are assembled separately, the new genetic material packed into the head and new daughter phage particles constructed. During this process, the host cells gradually become weakened by phage enzymes and eventually burst, releasing on average 100–200 new phage progeny into the surrounding environment. Lysogenic cycle: Following the injection of the phage DNA into the host cell, it integrates itself into the host genome, with the help of phage-encoded integrases, where it is then termed a prophage. The prophage genome is then replicated passively along with the host genome as the host cell divides for as long as it remains there and does not form the proteins required to produce progeny.
Microbiolres 13 00002 g002
Figure 3. CRISPR-Cas9 Adapative Immune System of Pseudomonas aeruginosa against bacteriophage. After infection, the Cas1-Cas2 complex recognizes the invading DNA and integrates a portion of it into the CRISPR array, giving rise to a new spacer. The bacterium incorporates a fragment of the invading phage DNA into its genome as a spacer into the CRISPR array, this spacer will serve as memory allowing the bacterium to recognize the same threat upon reinfection. The CRISPR array is transcribed as a long precursor CRISPR RNA (pre-crRNA). The host RNase III recognizes the tracrRNA:crRNA-Cas9 complex and cleaves both tracrRNA and crRNA. The Cas9 protein and the gRNA form a ribonucleoprotein complex through interactions between the gRNA scaffold and surface-exposed positively-charged grooves on Cas9. Cas9 undergoes a conformational change upon gRNA binding that shifts the molecule from an inactive, non-DNA binding conformation into an active DNA-binding conformation.
Figure 3. CRISPR-Cas9 Adapative Immune System of Pseudomonas aeruginosa against bacteriophage. After infection, the Cas1-Cas2 complex recognizes the invading DNA and integrates a portion of it into the CRISPR array, giving rise to a new spacer. The bacterium incorporates a fragment of the invading phage DNA into its genome as a spacer into the CRISPR array, this spacer will serve as memory allowing the bacterium to recognize the same threat upon reinfection. The CRISPR array is transcribed as a long precursor CRISPR RNA (pre-crRNA). The host RNase III recognizes the tracrRNA:crRNA-Cas9 complex and cleaves both tracrRNA and crRNA. The Cas9 protein and the gRNA form a ribonucleoprotein complex through interactions between the gRNA scaffold and surface-exposed positively-charged grooves on Cas9. Cas9 undergoes a conformational change upon gRNA binding that shifts the molecule from an inactive, non-DNA binding conformation into an active DNA-binding conformation.
Microbiolres 13 00002 g003
Table 1. Phage classification according to ICTV [3].
Table 1. Phage classification according to ICTV [3].
FormNucleic
Acid
OrderFamilyDetailsExamples
With taildsDNA (L)Caudovi-ralesMyoviridaeContractile tailT4, Um, P1, P2
Siphoviri-daeLong tail, not contractileΛ, T5, HK97, N15
Podoviri-daeShort tailT7, T3, φ29, P22
PolyhedralssDNA (C)Petitvira-lesMicroviri-daeCapsomers conspicuousPM2
dsDNA (C,S)Linavira-lesCorticovi-ridaeComplex capsid, lipidsPRD1
dsDNA (L)Kalamavi-ralesTectiviri-daeDouble capsid, lipids, pseudo-tailPRD1
dsDNA (L) SH1 *Double capsid, lipidsSH1
dsDNA (C) STIV *Tower-shaped protrusionsSTIV
ssRNA (L)Levivira-lesLeviviridae Like poliovirusMS2
dsRNA (L, M)Mindivi-ralesCystoviri-daeEnvelope, lipidsΦ6
FilamentousssDNA (C)Tubulavi-ralesInoviridae Long filamentous, short stemM13
dsDNA (L)Ligamen-viralesLipothixvi-ridaeEnvelope, lipidsTTV1
dsDNA (L)Rudiviro-daeRigid rods type, TMVSIRV-1
PleomorphicdsDNA (C,S) Plasmavi-ridaeEnvelope, without lipid capsid L2
dsDNA (C, S)Fusellovi-ridaeLemon shape, envelopeSSV1
dsDNA (L,S)**Lemon shape, envelopeHis1
dsDNA (C, S)Guttaviri-dae Drop shapeSNDV
dsDNA (L)Ampulla-viridae * Bottle shape, NC helicalABV
Legend: * unnamed; ** unclassified; dsDNA—double strand DNA; ssDNA—single strand DNA; C, circular; L, linear; M, multiparty; NC, nucleocapsid, S, supercoiled.
Table 2. Phages therapy advantages.
Table 2. Phages therapy advantages.
AdvantagesDescription
SpecificityVery specific to the host. infections with more than one host, it is necessary to use a phage cocktail
Bactericidal versus bacteriostaticLytic phages infect target host bacteria and cause cell death, compared to certain bacteriostatic antibiotics
Active on-site propagationPhages increase the concentration in the host as they replicate, theoretically requiring only one therapeutic dose
Low inherent toxicityPhages exist in large amounts in the biosphere, it is possible to isolate and purify the phage required to achieve certain bacteria.
Formulation and application versatilityVarious phages can be converted in a cocktail to target several bacteria simultaneously. The type of administration can also vary, liquid, powder, ointment, tablets.
Narrow potential for antibiotic cross-resistanceSince the phage and bacterial resistance mechanisms are different, bacteria resistant to certain antibiotics can be treated with the use of phage therapy.
Biofilm clearancePhages can penetrate through biofilms. Part of this capacity is due to the presence of depolymerases and lysins
Relatively low discovery and production costThe costs associated with discovering phage isolation and purification are relatively low
Low environmental impactPhages are natural components of the environment
Font: Adapted from [40].
Table 3. In vitro studies about phage therapy for Pseudomonas aeruginosa.
Table 3. In vitro studies about phage therapy for Pseudomonas aeruginosa.
StudySubjectPathologyBact StrainPhageDurationOutcome
Chang, R.Y.K. [93]Lung alveolar macrophage and epithelial cellsCystic fibrosisPAV237PEV1, PEV20 and PEV6124 hSignificantly reduces the multidrug-resistant (MDR) P. aeruginosa burden in mouse lungs.
Fong, S.A. [73]44 isolates Pseudomonas aeruginosa from humansChronic rhinosinusi-tisPAO1Pa 193, Pa 204, Pa 222, Pa 22348 hA single dose of phages is able to significantly reduce biofilms formed in vitro.
Shiley, J.R. [74]Lung cell A549 and U937 monocyteCancerPAO1-WT, pilin lacking PAO1-NPDMS3, PEV224 hDMS3 and PEV2 were able to inhibit bacterial growth in a PAO1-WT and PAO1-NP infection model, respectively.
Aghaee, B.L. [88]HumanBurn wound infectionP. aeruginosavB_PaeM_GUMS6, vB_PaeM_GUMS32, vB_PaeM_GUMS454, 8, 12, 24 and 48 hThe combination of two phage-antibiotics had the highest inhibition efficiency against the P. aeruginosa strain. The phages tested showed low stability at high temperatures, acid pH values, and in both lotions.
Legend: PAO1—Pseudomonas aeruginosa serotype O1; WT—wild type; NP—pilin-lacking; MDR—multidrug-resistant.
Table 4. In vivo studies about phage therapy for Pseudomonas aeruginosa.
Table 4. In vivo studies about phage therapy for Pseudomonas aeruginosa.
StudySubjectParticipantsAgePathologyBacterial StrainPhageDurationOutcome
Antoine, C. [82]Galleria mellonella450 LarvaeN.D.Canine otitisPAV237PEV224, 48 and 72 hDespite the ineffectiveness as life-saving antimicrobials in this model, PEV2 is active against P. aeruginosa in G. mellonella, even if this did not result in the complete elimination of bacteria at the tested MOIs.
Olszak, T. [60]Galleria mellonella LarvaeN.D.N.D.PAO1PA5oct Jumbo phage8, 18, 24, 48 and 72 hThe use of a phage mixture targeting different extracellular structures as receptors causes a huge impact on bacterial fitness, virulence, and pathogenicity.
Cafora, M. [94]Danio rerio EmbryosN.D.Cystic fibrosis (CF)PAO1vB_PaeP_PYO2, vB_PaeP_DEV, vB_PaeM_E215, vB_PaeM_E21720, 24, 26, 48 hpf (to block the embryo pigmentation from 24 h post fertilization)The lethality and bacterial burden were both decreased following phage therapy application.
Jang, H.J. [95]Drosophila melanogaster15–30 M/F5–7 daysPA infectionPAO1MPK1,MPK6,D3112, and PP748 hN.D.
Alvi, I.A. [90]Mice81 F6–8 weeksCFPAO1vB PaeP- SaPL1, 6, 12, 24, 48 and 96 hThe majority of tested MDR P. aeruginosa isolates are highly susceptible to SaPL, which inhibits their growth for longer time. SaPL is a potential candidate that can be used in phage therapy against MDR P. aeruginosa infections.
Lin, Y. [92]MiceF8–10 weeksCF, bronchiectasis and chronic obstructive pulmonary disease (COPD)FADD1-PA001PEV20N.D.Co-spray dried phage PEV20 and ciprofloxacin combination powder showed synergistic bacterial killing effect compared with single PEV20 or ciprofloxacin powders in an acute mouse lung infection model caused by P. aeruginosa.
Chow, M.Y.T. [93]MiceF6–8 weeksAcute respiratory infectionFADDI-PA001PEV3126 hPulmonary delivery of phage PEV31 in mice can reduce the MDR bacterial burden.
Lin, Y.W. [55]Rats17 (14F; 3M)8 weeksCFPandrug-resistant (PDR) isolate P. aeruginosa 112 and P. aeruginosa ADDG.øPEV2048–72 hThis studied showed positive outcomes in the utilization of PEV20 phage, in Rats, with simullation results predicting an infection erradication 12 h after the treatment, but some inconsistencie in in vivo models.
Abd El-Aziz, A.M. [65]MiceThree groups of F mice (15 each)10-week-oldLung infectionP. aeruginosa clinical strainsMMI-Ps172 h post infectionIn vitro and animal model studies showed that MMI-Ps1 effectively killed different serotypes of P. aeruginosa, including mucoid and nonmucoid strains. In addition, phage therapy significantly protected mice from lung and bloodstream infection caused by mucoid strains of P. aeruginosa.
Międzybrodzki, R. [70]MiceN.D.8–16 weeks at the begin-ningMurine collagen-induced arthritisP. aeruginosa 119x119x phage56 daysPreliminary results do not suggest that phages may aggravate the symptoms of rheumatoid arthritis
Chang, R.Y.K. [95]Mice48 to 10 weeksLung infectionFADDI-PA001Phage PEV2024 hThe study showed that intratracheally delivered aerosolized phage powder can significantly reduce MDR P. aeruginosa load in mouse lungs.
Roach, D.R. [76]Mice6N.D.CF, acute pneumoniaP. aeruginosa PAKlumiPAK_P114 daysThe study shows that successful phage curative and pro-phylactic therapies of acute respiratory infections depend on an essential synergy between phage lysis and the host’s own immune defenses.
Waters, E.M. [77]Mice606–8 weeksCF, COPDP. aeruginosa LESB65 and NP22_2Phage PELP2048–72 h and 7 days depending on the treatment.The study shows that phage administered intranasally up to 6 days after establishment of chronic lung infection were efficient in reducing bacterial numbers in the lungs of mice infected with P. aeruginosa, demonstrating the potential for phage therapy in the treatment of established and recalcitrant chronic respiratory tract infections.
Stratton, C.W. [79]MiceRodentsN.D.Catheter-induced rat aortic valve endocarditisP. aeruginosa strain CHACocktail of 12 phages agaisnt P. aeruginosa6 hThis study provide a strong proof of concept for the use of phage cocktails combined with antimicrobial agents for therapy of deep-seated and systemic infections.
Oechslin, F. [51]MiceRodentsN.D.Induced rat aortic valve endocarditis33 strains of P. aeruginosa reference strains and clinical isolates12 phages contained in the PP1131 cocktail18 hSingle-dose phage therapy was active against P. aeruginosa EE and highly synergistic with ciprofloxacin. Phage-resistant mutants had impaired infectivity.
Furusawa, T. [83]Mice, horseRodentsN.D.Bacterial keratitisPseudomonas sp. (Pa12, Pa18, Pa26, and Pa50) and P. aeruginosa strain NE-126ΦR18 and ΦS12-13 hThe phages used in this study rapidly adsorbed to P. aeruginosa and widely killed these isolates from horse lesions. One of the phages was shown to completely prevent keratitis in a keratitis mouse model.
Danis-Wlodarczyk, K. [84]Wax mothLarvaeN.D.InfectionPseudomonas aeruginosa (PAO1)KTN4 phage36 hConsidering all these characteristics, KTN4 phage is a suitable and promising for applications in treatment and prophylaxis in lung infections.
Forti, F. [94]Mice and wax mothF8–10 weeksAcute respiratory infection (mice) and bacteremia (wax moth)PAO1; PA14; PAK-lumi; LESB58; AG5; AG3; AA10; GJY9; CL1; CL2; VR8; AG6; DV4; CH7; AA2; AA43; AA44; TR1;TR66; TR67; E1 to E9 and PaPh1 a PaPh33.Cocktail composed of four novel phages (PYO2, DEV, E215 and E217) and two previously characterized phages (PAK_P1 and PAK_P4)After two hours post-infection (P.I.) (mice); 1 h P.I. (wax moth)Our findings indicate that in mice, lethal acute respiratory infection can be cured by treatment with the cocktail. Compared to our previous data obtained with a single phage, the cocktail showed the advantage of having more rapid efficacy in reducing the bacterial load.
Legend: F—female; M—male; N.D.—Not determined; PA—Pseudomonas aeruginosa; PAO1—Pseudomonas aeruginosa serotype O1; CF—Cystic fibrosis; COPD—chronic obstructive pulmonary disease; MDR—multidrug-resistant; PDR—pandrug-resistant.
Table 5. Clinical studies about phage therapy for Pseudomonas aeruginosa.
Table 5. Clinical studies about phage therapy for Pseudomonas aeruginosa.
StudySubjectParticipantsAgePathologyP. aeruginosa StrainPhageDurationOutcome
Tkhila-ishvili [71]Human1F83 years oldPeriprosthetic joint infectionClinical isolate of P. aeruginosaN.D.5 daysThe wound healed and the patient was discharged after 14 days with oral antibiotics for six weeks. The patient died four months after bacteriophage treatment due to left ventricular assist device pump thrombosis.
Madd-ocks, S. [61]Human1F77 years oldAdenocarcinomaClinical isolate of P. aeruginosaLytic anti-P.aeruginosa phages7 daysIntravenous antibiotics, bacteriophage therapy was well tolerated, with no adverse events detected either during therapy or subsequently. In combination with antimicrobials, bacteriophage therapy was associated with resolution of infection and with apparent eradication of Pseudomonas colonization. The successful decolonization would have occurred in the absence of phage treatment is unlikely, given the extent of disease and the sequential development of antimicrobial resistance.
Aslam, S. [62]Human3 (2F;1M)51 years oldLung transplantClinical isolate of P. aeruginosa among other strainsPA193; Pa204;Pa222;Pa223;Pa176; Paϕ1; PaSKWϕ17,PaSKWϕ22; PaATFϕ1; PaATFϕ3; BdPF16phi428.60 daysBacteriophage therapy was well tolerated and associated with clinical improvement when used as an adjunct to antibiotics in lung transplant recipients with against multidrug-resistant respiratory infections otherwise not responsive to antibiotics alone.
Law, N. [69]Human1M26 years oldCystic fibrosisClinical isolate of P. aeruginosaCocktail AB-PAo19 monthsGiven the concern for multidrug-resistant P. aeruginosa infections in cystic fibrosis patients, bacteriophage therapy may offer a viable anti-infective adjunct to traditional antibiotic therapy.
Gupta, P. [63]Human2036 years oldNonhealing woundClinical isolate of P. aeruginosa among other strainsCocktail from water sources21 daysA significant improvement was observed in the wound healing, and there were no signs of infection clinically and microbiologically after 3 to 5 doses of topical bacteriophage therapy. Seven patients achieved complete healing on day 21 during follow up while in others healthy margins and healthy granulation tissue were observed.
Jault, P. [66]Human2718 years or olderBurn wound infectionClinical isolate of P. aeruginosaPP11318 daysClinically relevant reduction in bacterial burden was observed in the phage group, with numerically fewer serious adverse events seen in those treated with the phage cocktail than the standard of care, indicating a favourable potential of phage therapy.
Legend: F—female; M—male; N.D.—Not determined; PA—Pseudomonas aeruginosa.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Silva, C.; Sá, S.; Guedes, C.; Oliveira, C.; Lima, C.; Oliveira, M.; Mendes, J.; Novais, G.; Baylina, P.; Fernandes, R. The History and Applications of Phage Therapy in Pseudomonas aeruginosa. Microbiol. Res. 2022, 13, 14-37. https://doi.org/10.3390/microbiolres13010002

AMA Style

Silva C, Sá S, Guedes C, Oliveira C, Lima C, Oliveira M, Mendes J, Novais G, Baylina P, Fernandes R. The History and Applications of Phage Therapy in Pseudomonas aeruginosa. Microbiology Research. 2022; 13(1):14-37. https://doi.org/10.3390/microbiolres13010002

Chicago/Turabian Style

Silva, Carina, Sara Sá, Carla Guedes, Carla Oliveira, Cláudio Lima, Marco Oliveira, João Mendes, Gonçalo Novais, Pilar Baylina, and Ruben Fernandes. 2022. "The History and Applications of Phage Therapy in Pseudomonas aeruginosa" Microbiology Research 13, no. 1: 14-37. https://doi.org/10.3390/microbiolres13010002

Article Metrics

Back to TopTop