About the Author(s)


Christoffel J. Opperman Email symbol
National Health Laboratory Service, Green Point Laboratory, Cape Town, South Africa

Department of Pathology, Faculty of Health Science, University of Cape Town, Cape Town, South Africa

Justyna M. Wojno symbol
Lancet Laboratories, Cape Town, South Africa

Adrian J. Brink symbol
Department of Pathology, Faculty of Health Science, University of Cape Town, Cape Town, South Africa

Microbiology Laboratory, National Health Laboratory Service, Groote Schuur Hospital, Cape Town, South Africa

Citation


Opperman CJ, Wojno JM, Brink AJ. Treating bacterial infections with bacteriophages in the 21st century. S Afr J Infect Dis. 2022;37(1), a346. https://doi.org/10.4102/sajid.v37i1.346

Opinion Paper

Treating bacterial infections with bacteriophages in the 21st century

Christoffel J. Opperman, Justyna M. Wojno, Adrian J. Brink

Received: 04 Oct. 2021; Accepted: 24 Jan. 2022; Published: 29 Mar. 2022

Copyright: © 2022. The Author(s). Licensee: AOSIS.
This is an Open Access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Bacteriophages (phages) were discovered in the early part of the 20th century, and their ability to eliminate bacterial infections as bacterial viruses gathered interest almost immediately. Bacteriophage therapy was halted in the Western world due to inconclusive results in early experiments and the concurrent discovery of antibiotics. The spread of antibiotic-resistant bacteria has elicited renewed interest in bacteriophages as a natural alternative to conventional antibiotic therapy. Interest in the application of bacteriophages has also expanded to include the environment, such as wastewater treatment, agriculture and aquaculture. Although the complete phage is important in bacteriophage therapy, the focus is shifting to purified phage enzymes. These enzymes are an attractive option for pharmaceutical companies with their patent potential. They can be bio-engineered for enhanced adjuvant properties, such as a broadened spectrum of activity or binding capability. Enzymes also eliminate the concern that the prophage might integrate resistance genes into the bacterial genome. From a clinical perspective, the first randomised clinical controlled phage therapy trial was conducted with more pioneering phase I/II clinical studies on the horizon. In this opinion paper, the authors outline bacteriophages as naturally occurring bactericidal entities, their therapeutic potential against antibiotic-resistant bacteria and compare them to antibiotics. Their potential multipurpose application in the medical field is also addressed, including the use of bacteriophages for vaccination, and utilisation of the antimicrobial enzymes that they produce.

Keywords: bacteriophage; bacteriophage therapy; phage; non-lytic phage; vaccination; gene-transfer; endolysins; enzybiotics; artilysins.

Introduction

As anticipated by Alexander Fleming, soon after discovering penicillin in the 1920s, 14% of Staphylococci isolated from patients in a London hospital demonstrated resistance to penicillin by 1946.1 Antibiotic resistance has subsequently proliferated at an alarming rate to the level that by 2050, 10 million deaths due to antibiotic-resistant infections may occur.2 The emergence and spread of pan-drug resistant bacteria render the need for new treatment approaches a priority.

Bacteriophages are viruses that kill bacteria following amplification and bacterial lysis to release virion progeny into the environment (lytic-lifecycle). Bacteriophage therapy exploits this lifecycle (Figure 1) without the integration of the bacteriophage (prophage) into the host genome (lysogenic lifecycle).3 Alternative approaches include using non-lytic bacteriophages as a delivery vehicle to transport deoxyribonucleic acid (DNA) vaccines or using purified phage components such as cell wall hydrolases as novel modalities to treat antibiotic-resistant infections. Bacteriophage therapy does have potential limitations that include but are not limited to toxic shock following bacterial lysis or the generation of neutralising bacteriophage antibodies after repeated treatment.4

FIGURE 1: Life cycle of bacteriophages. Adsorption occurs between the bacteriophage (phage) and bacterium cell surface receptors. The phage protein capsule is left behind once the linear DNA enters the infected bacterium to form circular deoxyribonucleic acid molecules. From here, two life cycles can be entered. During the lytic cycle (bacteriophage therapy), tail fibres and protein capsids (heads) are formed during transcription and translation using the biosynthetic apparatus of the bacteria, such as ribosomes. The phage particles are assembled, and the viral genome is packaged within the capsids. Holin proteins and endolysins are involved in cell lysis and phage progeny release. The second pathway is termed as the lysogenic cycle. On this path, the temperate phage DNA is integrated into the bacterium chromosome (prophage), and phage development is halted. Excision of phage DNA from the chromosome can allow the phage to re-enter the lytic life cycle.

Comparison: Bacteriophages versus antibiotics

Lytic bacteriophages offer several potential advantages over antibiotic therapy (Table 1). Most importantly, unlike antibiotics, they are pathogen-specific, thus reducing the extensive disruption of microbial flora due to high specificity. Maura et al., in 2011, constructed a stable mouse intestine model to study the possible long-term effect of self-replicating bacteriophages within a mammalian poly-microbial niche. The study concluded that no change in the faecal bacteria levels was observed.5

TABLE 1: A comparison in the therapeutic use of bacteriophage and antibiotics.

Another major advantage of using therapeutic phages is their self-limiting amplification capability, as they only persist as long as the bacteria are present. Notably, the use of localised phage therapy leads to deep penetration as long as the infection is present, as opposed to antibiotics, where from a pharmacokinetic point of view specific targets need to be maintained with repetitive doses to maintain concentrations.6 As auto-replication occurs after one dose, less phage is required to achieve the optimum therapeutic effect.

Most bacteriophages promote a strong antibody response and are highly immunogenic.7 It is unclear if this response is because of the phage itself or the lysed bacterial components. Zimecki et al. demonstrated that prophylactic phage administration in an immunosuppressed mouse model could overcome neutrophil deficit in the clearance of Staphylococcus aureus infection and mobilise cells from both lymphocytic as well as myelocytic lineages. It was postulated in this study that phages could promote the recruitment of immature neutrophil cell types from bone marrow precursors and trigger a rapid output of functional mature neutrophils into the peripheral circulation to clear the bacterial infection.8

Endotoxin released from bacteria lysed in vivo may lead to potential health concerns in patients. A particularly useful strategy may be to use lysis deficient bacteriophages, which decreases the release of pro-inflammatory cytokines.9 However, considering the minor side effects reported for bacteriophage therapy, such as occasional serum-like illness after intravenous administration, multiple side effects have been observed with antibiotics. Anaphylaxis following bacteriophage therapy has never been described in the literature. The initial reporting of ‘shock’ was most likely because of a low level of purification and contaminants in phage preparations.10 In this regard, protein toxins produced by pathogenic species and gram-negative bacteria endotoxins are impurities in phage lysates that are harmful to humans. Therefore, similar to antibiotic purification, procedures should be rapid, scalable and efficient in removing impurities to ensure patient safety.11

Bacteriophage therapy is a promising alternative to antibiotics in vulnerable population groups such as pregnant population and neonates, where numerous antibiotic contraindications pertain. Minimal disruption of the host microbiota during the antenatal and perinatal periods is beneficial. In addition, the transplacental transfer of phages has been observed in guinea pigs following intravenous maternal phage administration. Further studies are required to explore in utero bacteriophage treatment strategies.12

Phage-antibiotic combination therapy

There is a clear potential to use phage therapy concurrently with antibiotics, especially with increasingly antibiotic-resistant pathogens.13 This is especially true when the phage component is applied at high titres topically and therefore not easily removed by the immune system. Furthermore, phages encapsulated in hydrogels can result in a long-term release of active particles, whilst liposome preparations applied topically protect against immune system components and enzymatic hydrolysis.14,15 It is unlikely; however that bacteriophage therapy will replace antibiotics but combining antibiotics with phage therapy should reduce bacterial resistance because the pathogen must acquire two separate mechanisms to survive. Altering binding targets for both antibiotics and phages may come at a fitness cost for the bacteria because mutations can influence essential biological functions. The concept of a trade-off of antibiotic resistance mechanisms and phage resistance is referred to as ‘phage steering’.16,17 Further research is required to explore the evolutionary response of bacterial strains to the phage-antibiotic combinations. The argument supports the idea that a phage can be selected against every antibiotic or phage-resistant bacterium from an evolutionary standpoint. The ongoing process of natural selection achieves this.18

A strategy to prevent phage resistance would be to use complex mixtures of bacteriophages or ‘phage cocktails’ to circumvent phage resistance.19 Alternatively, the non-lytic bacteriophage discussed in the following sections may be used to deliver DNA encoding phage-proteins that affect bacterial cell division, DNA replication, transcription and translation.20

Bacteriophage mediated gene and antimicrobial agent transfer

Many novel antibiotic agents are excluded from clinical use because of their inability to be selected explicitly for bacterial versus human cells. This may result in toxicity. During ‘phage display’, an antigen gene is fused with a gene coding for a coat-surface protein and, when expressed, it is displayed as a fusion protein on the capsule of the bacteriophage.21 In targeted drug delivery, a targeting moiety is displayed on the filamentous phage coat with phage display. The cytotoxic drug is then chemically linked with an ester bond to the bacteriophage. The drug is a prodrug with no cytotoxic activity in the conjugated state. Once the prodrug is dissociated from the phage at the target site in a controlled manner, it is activated. Targeted drug delivery has been shown in vitro between S. aureus and chloramphenicol.22 Adapted bacteriophage vectors have been used to restore antibiotic sensitivity by reversing antibiotic resistance in pathogens. For example, a proof-of-concept model exploited the temperate phage to introduce the rpsL and gyrA genes and confer sensitivity to streptomycin and nalidixic acid, respectively, where these antibiotics were resistant.22,23 Unlike conventional bacteriophage therapy, the authors proposed a scenario where these phages can deliver genetic constructs and render bacteria sensitive to previously resistant antibiotics. Rather than administering phages to the patient, they may also be applied to hospital surfaces and, in the process, potentially reduce environmental contamination by antibiotic-resistant pathogens.

Various methods have been employed to increase phage uptake and stability to target intracellular pathogens. In this Trojan horse approach, pages are loaded onto a carrier vehicle during receptor-mediated uptake. These novel technologies include nanocrystal, liposome, polymer encapsulation and delivery, genetically engineered phage, nanofibre entrapment, nanoparticle absorbed and hydrogel embedded phages.24

Bacteriophage vaccination

Bacteriophages vaccination is an innovative and novel vaccine system.25,26 This is a bio-engineered structure that entails a DNA vaccine contained within a phage capsule under the expression of a eukaryotic promoter.21 Once the phage transporting the DNA vaccine is engulfed into a eukaryotic antigen-presenting cell, the vaccine gene is expressed. Following this, major histocompatibility complexes (MHC) present the processed antigens to the immune system. Antigens presented to MHC II generate an antibody response, while a cellular response is activated through cross-priming with the MHC I pathway. Phage display vaccines can also directly activate B cells to produce antibodies.26 Haq et al. reported on studies proposing a hybrid phage with phage display and DNA vaccine capability.27 The advantage of an engineered DNA phage vaccine is to treat and prevent infection. Bacteriophage vaccines are also suitable to immunise large communities because they are relatively easy to apply and inexpensive to produce. Jepson and March, in 2004 postulated the concept of bacteriophage DNA vaccines administered orally via drinking water.28 Notably, during such a proposed vaccination programme, a high phage titre would be required as it appears that gut transit and phage penetration after oral administration are dose-dependent.29

Bacteriophage enzymes

Purified lysins are promising antibacterial phage components currently under development. They consist of a class of cell wall hydrolases that translocate into the peptidoglycan wall of bacteria and cleave the bonds, which results in the release of viral progeny during a viral infection cycle. Endolysins have a characteristic structure with multiple cell wall binding domains. They degrade the peptidoglycan wall with lytic transglycosylase, amidase glycosidase or endopeptidase activities.30 Their synergistic mechanism of action with a range of other antimicrobials or fellow peptidoglycan hydrolases is of interest. Endolysins are less likely to invoke resistance because of the coevolution of bacteriophages and bacteria.31 The purified lysins can also be applied directly to sensitive bacteria. Enzyme preparations include aerosols for inhalation, formulations for systematic injection, topical ointments and creams.32 The lytic activity spectrum of lysins is usually narrow because lysin receptors are distributed in a strain-specific manner. Furthermore, the binding affinity is equivalent to mature antibodies, making them an irreversible inhibitor.33 This alone promises that lysins could be highly efficacious when compared to antibiotics. An anti-staphylococcal lysin (exebacase or formally known as CF-301) demonstrated a 42.8% higher clinical response rate when given alongside the standard of care antibiotic compared to the antibiotic alone when treating methicillin-resistant Staphylococcus aureus (MRSA) bacteremia, including infective endocarditis. As a result, this is the first lysin to have entered a phase III human trial to treat S. aureus.34,35,36

An interesting feature of these ‘enzybiotics’ is their structural versatility.37 An enzybiotic is a bacteriophage enzyme with cell wall degrading capability against bacteria or fungi. It is a hybrid term derived from the words enzyme and antibiotic. The cell wall hydrolysis and receptor binding domains of the structure can be separated while maintaining their activity. These domains can be fused in a complementary fashion to other lysins to alter catalytic function, redirect binding or both. These bioengineered lysins are the so-called next-generation lysins.38 Artilysins are a group of next-generation lysins with an additional lipopolysaccharide disrupter to increase outer membrane penetration of gram-negative bacteria. A limited number of lysins can be recombined to generate a highly variable number to avoid bacterial resistance. So far, no neutralising antibodies or lysine resistance has been detected under the same conditions that favour antibiotic resistance and similarly no toxic side effects have been demonstrated.39 Lysins have shown promising results to clear pathogens on the mucous membranes in animal models and in vitro. The chimeric ectolysin P128 was manufactured to clear nasal MRSA infections. In situ efficacy was seen with a P128 hydrogel formulation against Staphylococci recovered from the nares of 31 healthy individuals, including mupirocin-resistant isolates. In addition, an MRSA nasal decolonisation rat model compared P128 formulated as a hydrogel to 2% mupirocin ointment. P128 cleared colonisation twofold, whereas the mupirocin alone was ineffective in the rat nares.37,40,41 A randomised, double-blind placebo-controlled study to determine the efficacy and safety of P128 applied intra-nasally to healthy volunteers has been completed (results not publicly available) to evaluate S. aureus clearance and re-colonisation rates.42

Although further attention is needed in preclinical testing with lysins, applications could include the treatment of antibiotic-resistant bacterial biofilms on medical devices such as catheters.43 LysGH15 inhibits biofilm formation in vitro, but at higher doses, it distorts the integrity of the biofilm by dislodging the bacteria instead of destroying the biofilm matrix.44 The bacteriophage-derived lysin PlySs2 reduced colony-forming units by 99% and biofilm by 75% relative to vancomycin in vitro.45 Lastly, killing slow or non-growing organisms associated with bacterial infections of a chronic nature that may be naturally resistant to phage treatment or antibiotics could also be susceptible to lysins.39,46

Clinical case studies and trials using bacteriophages

Several case reports highlight the potential use of phages in various clinical syndromes. Case studies illustrating the successful eradication of multidrug-resistant (MDR) Pseudomonas aeruginosa in ventilated patients and Klebsiella pneumoniae lung infections are promising.47,48 Furthermore, phage therapy co-administered with antibiotics has been successfully utilised in a refractory case of a complicated intra-abdominal infection because of MDR Acinetobacter baumannii.49 Even though phage resistance emerged in this patient with a pancreatic pseudocyst infection, it was rapidly circumvented with the selection and purification of a salvage phage that was subsequently added to the treatment regime.49 Notably, a case series in critically ill patients with prosthetic or native valve endocarditis because of S. aureus treated with a bacteriophage displayed reduced bacterial counts, clinical improvement and corresponding decreases in inflammatory markers.50 Other applications include gut decolonisation strategies, such as eradicating MDR, carbapenemase-producing K. pneumoniae isolates, following intra-rectal and oral therapy with a lytic bacteriophage preparation.51

Although several phase I/II clinical studies on bacteriophage therapy have been published with many case reports that include different routes of phage administration, sources of infection and targeted pathogens,52 only two have progressed to phase III to our knowledge. The first is an active open-label, single-group assigned intervention of a nebulised liquid pyobacteriophage complex that entails the irrigation of mucous membranes in children with confirmed acute tonsillitis.53 A phase III randomised, placebo-controlled, double-blind trial was recently conducted in patients undergoing transurethral resection of the prostate with urinary tract infections. Treatment with intravesicular phage administration was safe and non-inferior in outcome compared to standard antibiotic treatment.54,55

The poor clinical efficacy of phage therapy in many previous phase I/II trials could be related to design and technical issues.56 Possible factors for phage failure include sub-optimal titres and quality of phage preparations, the production of serum anti-phage antibodies, insufficient phage coverage of targeted pathogens, phage-eukaryotic cell interactions that might modify the effect of phage therapy and external factors such as medication (proton pump inhibitors, antibiotics) as well as food and drinks (yoghurt, alcohol).57,58 A better focus and understanding of these confounding factors during trial design might lead to more favourable outcomes in the future.

Bacteriophages for laboratory identification of bacteria

Although not the focus of this overview, mention should be made to bacteriophage’s unique capability to detect specific bacteria for pathogen diagnostic purposes. Fluorescently stained phages that bind to targeted bacteria can be visualised under a fluorescence microscope as halo structures. Alternatively, genetically engineered phages express reporter genes during phage-bacteria binding. Quantum dots (nanocrystals) bioconjugated to the capsid surface can be visualised using a microscope during phage-bacteria interaction. In optical and electrochemical transducer systems, phage heads are immobilised onto electrodes exposing their tails to capture specific bacteria that can identify microorganisms.59

Conclusion and future direction

Phage research is gaining interest on the African continent, but few groups have entered phage product design and development stages. As a result, in vivo studies, phage safety, efficacy and phage biobanking in the African context remain a priority.60 Although no single phage manufacturing centre has been established in Africa yet, research groups have been initiated to foster collaboration with Phages for Global Health.60 This initiative is focused on initially teaching scientists in African countries to isolate and characterise phages.

It remains unclear if phages should be classified as biological or chemical agents (lysins), food additives or pharmaceutical drugs. This uncertainty complicates legislation, regulatory processes for bacteriophage therapy and investment.60 Current regulatory frameworks should support phage research and academic institutions to promote development, be flexible in compassionate use cases and be open to possible alternative pathways for the approval of phage therapy.

In this regard, a pertinent question for the future of phage therapy is whether manufacturers are willing to invest in the development of a product with limited patent protection. Intellectual property status is one of the major contributing factors to the general reluctance to develop phage therapy in developed countries. Pharmaceutical companies will not undergo expensive and rigorous clinical trials for drug approval and registration without patent protection.61,62 The possibility for an individual phage to be patented is unlikely; however, firms have adopted strategies to commercialise phage therapy. This includes patenting specific phage sequences or phage components mentioned above. Establishing regional phage banks across Africa that have already isolated, characterised, sequenced and purified phages could contribute to sustainable and cost-effective phage therapy.60,63 In addition, inaugurating networks of phage banks across the globe between reference phage laboratories and low-income regions could further help transfer knowledge and lower the cost of bacteriophage therapy.

In the future, treatment of antibiotic-resistant bacterial infections with phage therapy might be preceded by bacteriophage control of pathogens from an ecological perspective. To prevent infection with phage rather than to treat it is the focus for future research in agriculture, water treatment and food-related industries.64,65,66 Until bacteriophage bio-control becomes the next frontier against pathogens, focusing on research that could provide regulatory bodies with the evidence to justify phage commercialisation and clinical use remains essential. This includes research on their complimentary bactericidal activity against antibiotic-resistant infections and their ability to reduce the gastro-intestinal carriage of MDR pathogens, from an infection prevention control point of view. In addition, designing phage formulations specifically in the African context that are easy to mass administer, have a long shelf life, are robust for transportation at different temperatures, and obtaining large-scale production skills should not be ignored.67 Finally, clinical data on bacteriophage and phage-derived products remain limited, with more rigorous clinical trials and reference standards needed to substantiate phage efficacy, determine appropriate treatment regimens and confirm its safety profile.

Acknowledgements

Competing interests

The authors declare that they have no financial or personal relationships that may have inappropriately influenced them in writing this article.

Authors’ contributions

C.J.O., J.M.W. and A.J.B. contributed equally to the design and implementation of the research, to the analysis of the results and to the writing of the manuscript.

Ethical considerations

This article followed all ethical standards for research without direct contact with human or animal subjects.

Funding information

This research received no specific grant from any funding agency in the public, commercial or not-for-profit sectors.

Data availability

Data sharing is not applicable to this article as no new data were created or analysed in this study.

Disclaimer

The views and opinions expressed in this article are those of the authors and do not necessarily reflect the official policy or position of any affiliated agency of the authors.

References

  1. Smith JT, Hamilton-Miller JMT, Knox R. Bacterial resistance to penicillins and cephalosporins. J Pharm Pharmacol. 1969;21(6):337–358. https://doi.org/10.1111/j.2042-7158.1969.tb08267.x
  2. Vázquez R, García E, García P. Phage lysins for fighting bacterial respiratory infections: A new generation of antimicrobials. Front Immunol. 2018;9:2252. https://doi.org/10.3389/fimmu.2018.02252
  3. Mathieu J, Yu P, Zuo P, Da Silva MLB, Alvarez PJJ. Going viral: Emerging opportunities for phage-based bacterial control in water treatment and reuse. Acc Chem Res. 2019;52(4):849–857. https://doi.org/10.1021/acs.accounts.8b00576
  4. Principi N, Silvestri E, Esposito S. Advantages, and limitations of bacteriophages for the treatment of bacterial infections. Front Pharmacol. 2019;10:513. https://doi.org/10.3389/fphar.2019.00513
  5. Maura D, Morello E, Du Merle L, Bomme P, Le Bouguénec C, Debarbieux L. Intestinal colonization by enteroaggregative Escherichia coli supports long-term bacteriophage replication in mice. Environ Microbiol. 2012;14(8):1844–1854. https://doi.org/10.1111/j.1462-2920.2011.02644.x
  6. Gu Liu C, Green SI, Min L, et al. Phage-antibiotic synergy is driven by a unique combination of antibacterial mechanism of action and stoichiometry. mBio. 2020;11(4):e01462-20. https://doi.org/10.1128/mBio.01462-20
  7. Van Belleghem JD, Dąbrowska K, Vaneechoutte M, Barr JJ, Bollyky PL. Interactions between bacteriophage, bacteria, and the mammalian immune system. Viruses. 2018;11(1):10. https://doi.org/10.3390/v11010010
  8. Zimecki M, Artym J, Kocięba M, Weber-Dąbrowska B, Borysowski J, Górski A. Effects of prophylactic administration of bacteriophages to immunosuppressed mice infected with Staphylococcus aureus. BMC Microbiol. 2009;9(1):169. https://doi.org/10.1186/1471-2180-9-169
  9. Matsuda T, Freeman TA, Hilbert DW, et al. Lysis-deficient bacteriophage therapy decreases endotoxin and inflammatory mediator release and improves survival in a murine peritonitis model. Surgery. 2005;137(6):639–646. https://doi.org/10.1016/j.surg.2005.02.012
  10. Speck P, Smithyman A. Safety, and efficacy of phage therapy via the intravenous route. FEMS Microbiol Lett. 2016;363(3):242. https://doi.org/10.1093/femsle/fnv242
  11. Hietala V, Horsma-Heikkinen J, Carron A, Skurnik M, Kiljunen S. The removal of endo- and enterotoxins from bacteriophage preparations. Front Microbiol. 2019;10:1674. https://doi.org/10.3389/fmicb.2019.01674
  12. Furfaro LL, Chang BJ, Payne MS. Applications for bacteriophage therapy during pregnancy and the perinatal period. Front Microbiol. 2018;8:2660. https://doi.org/10.3389/fmicb.2017.02660
  13. Tagliaferri TL, Jansen M, Horz HP. Fighting pathogenic bacteria on two fronts: Phages and antibiotics as combined strategy. Front Cell Infect Microbiol. 2019;9:22. https://doi.org/10.3389/fcimb.2019.00022
  14. Rosner D, Clark J. Formulations for bacteriophage therapy and the potential uses of immobilization. Pharmaceuticals (Basel). 2021;14(4):359. https://doi.org/10.3390/ph14040359
  15. Loh B, Gondil VS, Manohar P, Khan FM, Yang H, Leptihn S. Encapsulation and delivery of therapeutic phages. Appl Environ Microbiol. 2020;87(5):e01979-20. https://doi.org/10.1128/AEM.01979-20
  16. Melnyk AH, Wong A, Kassen R. The fitness costs of antibiotic resistance mutations. Evol Appl. 2015;8(3):273–283. https://doi.org/10.1111/eva.12196
  17. Gurney J, Pradier L, Griffin JS, et al. Phage steering of antibiotic-resistance evolution in the bacterial pathogen, Pseudomonas aeruginosa. Evol Med Public Health. 2020;1:148–157. https://doi.org/10.1093/emph/eoaa026
  18. Dhar N, McKinney JD. Microbial phenotypic heterogeneity and antibiotic tolerance. Curr Opin Microbiol. 2007;10(1):30–38. https://doi.org/10.1016/j.mib.2006.12.007
  19. Haines MEK, Hodges FE, Nale JY, et al. Analysis of selection methods to develop novel phage therapy cocktails against antimicrobial resistant clinical isolates of bacteria. Front Microbiol. 2021;12:613529. https://doi.org/10.3389/fmicb.2021.613529
  20. Drulis-Kawa Z, Majkowska-Skrobek G, Maciejewska B, Delattre AS, Lavigne R. Learning from bacteriophages – Advantages and limitations of phage and phage-encoded protein applications. Curr Protein Pept Sci. 2012;13(8):699–722. https://doi.org/10.2174/138920312804871193
  21. Aghebati-Maleki L, Bakhshinejad B, Baradaran B, et al. Phage display as a promising approach for vaccine development. J Biomed Sci. 2016;23:66. https://doi.org/10.1186/s12929-016-0285-9
  22. Yacoby I, Shamis M, Bar H, Shabat D, Benhar I. Targeting antibacterial agents by using drug-carrying filamentous bacteriophages. Antimicrob Agents Chemother. 2006;50(6):2087–2097. https://doi.org/10.1128/AAC.00169-06
  23. Gibb B, Hyman P, Schneider CL. The many applications of engineered bacteriophages – An overview. Pharmaceuticals (Basel). 2021;14(7):634. https://doi.org/10.3390/ph14070634
  24. Goswami A, Sharma PR, Agarwal R. Combatting intracellular pathogens using bacteriophage delivery. Crit Rev Microbiol. 2021;47(4):461–478. https://doi.org/10.1080/1040841X.2021.1902266
  25. Staquicini DI, Tang FHF, Markosian C, et al. Design and proof-of-concept for targeted phage-based COVID-19 vaccination strategies with a streamlined cold-free supply chain. Proc Natl Acad Sci USA. 2021;118(30):e2105739118. https://doi.org/10.1073/pnas.2105739118
  26. Adhya S, Merril CR, Biswas B. Therapeutic and prophylactic applications of bacteriophage components in modern medicine. Cold Spring Harb Perspect Med. 2014;4(1):a012518. https://doi.org/10.1101/cshperspect.a012518
  27. Haq IU, Chaudhry WN, Akhtar MN, Andleeb S, Qadr I. Bacteriophages, and their implications on future biotechnology: A review. Virol J. 2012;9:9. https://doi.org/10.1186/1743-422X-9-9
  28. Jepson CD, March JB. Bacteriophage lambda is a highly stable DNA vaccine delivery vehicle. Vaccine. 2004;22(19):2413–2419. https://doi.org/10.1016/j.vaccine.2003.11.065
  29. Dąbrowska K. Phage therapy: What factors shape phage pharmacokinetics and bioavailability? Systematic and critical review. Med Res Rev. 2019;39(5):2000–2025. https://doi.org/10.1002/med.21572
  30. Fischetti VA. Development of phage lysins as novel therapeutics: A historical perspective. Viruses. 2018;10(6):310. https://doi.org/10.3390/v10060310
  31. Nelson DC, Schmelcher M, Rodriguez-Rubio L, et al. Endolysins as antimicrobials. Adv Virus Res. 2012;83:299–365. https://doi.org/10.1016/B978-0-12-394438-2.00007-4
  32. Maciejewska B, Olszak T, Drulis-Kawa Z. Applications of bacteriophages versus phage enzymes to combat and cure bacterial infections: An ambitious and also a realistic application? Appl Microbiol Biotechnol. 2018;102(6):2563–2581. https://doi.org/10.1007/s00253-018-8811-1
  33. São-José C. Engineering of phage-derived lytic enzymes: Improving their potential as antimicrobials. Antibiotics (Basel). 2018;7(2):29. https://doi.org/10.3390/antibiotics7020029
  34. Murray E, Draper LA, Ross RP, Hill C. The advantages and challenges of using endolysins in a clinical setting. Viruses. 2021;13(4):680. https://doi.org/10.3390/v13040680
  35. De Maesschalck V, Gutiérrez D, Paeshuyse J, Lavigne R, Briers Y. Advanced engineering of third-generation lysins and formulation strategies for clinical applications. Crit Rev Microbiol. 2020;46(5):548–564. https://doi.org/10.1080/1040841X.2020.1809346
  36. Watson A, Oh JT, Sauve K, Bradford PA, Cassino C, Schuch R. Antimicrobial activity of exebacase (Lysin CF-301) against the most common causes of infective endocarditis. Antimicrob Agents Chemother. 2019;63(10):e01078-19. https://doi.org/10.1128/AAC.01078-19
  37. Dams D, Briers Y. Enzybiotics: Enzyme-based antibacterials as therapeutics. Adv Exp Med Biol. 2019;1148:233–253. https://doi.org/10.1007/978-981-13-7709-9_11
  38. Yang H, Yu J, Wei H. Engineered bacteriophage lysins as novel anti-infectives. Front Microbiol. 2014;5:542. https://doi.org/10.3389/fmicb.2014.00542
  39. Fischetti VA, Nelson D, Schuch R. Reinventing phage therapy: Are the parts greater than the sum? Nat Biotechnol. 2006;24(12):1508–1511. https://doi.org/10.1038/nbt1206-1508
  40. Vipra AA, Desai SN, Roy P, et al. Antistaphylococcal activity of bacteriophage derived chimeric protein P128. BMC Microbiol. 2012;12:41. https://doi.org/10.1186/1471-2180-12-41
  41. Roach DR, Donovan DM. Antimicrobial bacteriophage-derived proteins and therapeutic applications. Bacteriophage. 2015;5(3):e1062590. https://doi.org/10.1080/21597081.2015.1062590
  42. Abdelrahman F, Easwaran M, Daramola OI, et al. Phage-encoded endolysins. Antibiotics (Basel). 2021;10(2):124. https://doi.org/10.3390/antibiotics10020124
  43. Ferriol-González C, Domingo-Calap P. Phages for biofilm removal. Antibiotics (Basel). 2020;9(5):268. https://doi.org/10.3390/antibiotics9050268
  44. Li X, Sun L, Zhang P, Wang Y. Novel approaches to combat medical device-associated biofilms. Coatings. 2021;11(3):294. https://doi.org/10.3390/coatings11030294
  45. Sosa BR, Niu Y, Turajane K, et al. The antimicrobial potential of bacteriophage-derived lysin in a murine debridement, antibiotics, and implant retention model of prosthetic joint infection. Bone Joint J. 2020;102-B(7 Suppl B):3–10. https://doi.org/10.1302/0301-620X.102B7.BJJ-2019-1590.R1
  46. Fenton M, Ross P, McAuliffe O, O’Mahony J, Coffey A. Recombinant bacteriophage lysins as antibacterials. Bioeng Bugs. 2010;1(1):9–16. https://doi.org/10.4161/bbug.1.1.9818
  47. Aslam S, Courtwright AM, Koval C, et al. Early clinical experience of bacteriophage therapy in 3 lung transplant recipients. Am J Transplant. 2019;19(9):2631–2639. https://doi.org/10.1111/ajt.15503
  48. Rubalskii E, Ruemke S, Salmoukas C, et al. Bacteriophage therapy for critical infections related to cardiothoracic surgery. Antibiotics (Basel). 2020;9(5):232. https://doi.org/10.3390/antibiotics9050232
  49. Schooley RT, Biswas B, Gill JJ, et al. Development and use of personalized bacteriophage-based therapeutic cocktails to treat a patient with a disseminated resistant Acinetobacter baumannii Infection. Antimicrob Agents Chemother. 2017;61(10):e00954-17. https://doi.org/10.1128/AAC.00954-17
  50. Petrovic Fabijan A, Lin RCY, Ho J, Maddocks S, Ben Zakour NL, Iredell JR. Safety of bacteriophage therapy in severe Staphylococcus aureus infection. Nat Microbiol. 2020(3):465–472. https://doi.org/10.1038/s41564-019-0634-z
  51. Corbellino M, Kieffer N, Kutateladze M, et al. Eradication of a multidrug-resistant, carbapenemase-producing Klebsiella pneumoniae isolate following oral and intra-rectal therapy with a custom made, lytic bacteriophage preparation. Clin Infect Dis. 2020;70(9):1998–2001. https://doi.org/10.1093/cid/ciz782
  52. Luong T, Salabarria AC, Roach DR. Phage therapy in the resistance era: Where do we stand and where are we going? Clin Ther. 2020;42(9):1659–1680. https://doi.org/10.1016/j.clinthera.2020.07.014
  53. Tolkunovna TS. Bacteriophage therapy in tonsillitis [homepage on the Internet]. Trial: NCT04682964. [cited 2021 Sep 1]. Availabe from: https://clinicaltrials.gov/ct2/
  54. Leitner L, Ujmajuridze A, Chanishvili N, 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(3):427–436. https://doi.org/10.1016/S1473-3099(20)30330-3
  55. Leitner L, Sybesma W, Chanishvili N, et al. Bacteriophages for treating urinary tract infections in patients undergoing transurethral resection of the prostate: A randomized, placebo-controlled, double-blind clinical trial. BMC Urol. 2017;17(1):90. https://doi.org/10.1186/s12894-017-0283-6
  56. 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(2):96. https://doi.org/10.3390/v11020096
  57. Górski A, Borysowski J, Międzybrodzki R. Phage therapy: Towards a successful clinical trial. Antibiotics (Basel). 2020;9(11):827. https://doi.org/10.3390/antibiotics9110827
  58. Valente L, Prazak J, Que YA, Cameron DR. Progress and pitfalls of bacteriophage therapy in critical care: A concise definitive review. Crit Care Explor. 2021;3(3):e0351. https://doi.org/10.1097/CCE.0000000000000351
  59. Hussain W, Ullah MW, Farooq U, Aziz A, Wang S. Bacteriophage-based advanced bacterial detection: Concept, mechanisms, and applications. Biosens Bioelectron. 2021;177:112973. https://doi.org/10.1016/j.bios.2021.112973
  60. Makumi A, Mhone AL, Odaba J, Guantai L, Svitek N. Phages for Africa: The potential benefit and challenges of phage therapy for the livestock sector in sub-Saharan Africa. Antibiotics. 2021;10(9):1085. https://doi.org/10.3390/antibiotics10091085
  61. Cooper CJ, Khan Mirzaei M, Nilsson AS. Adapting drug approval pathways for bacteriophage-based therapeutics. Front Microbiol. 2016;7:1209. https://doi.org/10.3389/fmicb.2016.01209
  62. Clark JR, March JB. Bacteriophages, and biotechnology: Vaccines, gene therapy and antibacterials. Trends Biotechnol. 2006;24(5):212–218. https://doi.org/10.1016/j.tibtech.2006.03.003
  63. Kassa T. Bacteriophages against pathogenic bacteria and possibilities for future application in Africa. Infect Drug Resist. 2021;14:17–31. https://doi.org/10.2147/IDR.S284331
  64. Huss P, Raman S. Engineered bacteriophages as programmable biocontrol agents. Curr Opin Biotechnol. 2020;61:116–121. https://doi.org/10.1016/j.copbio.2019.11.013
  65. Jassim SA, Limoges RG, El-Cheikh H. Bacteriophage biocontrol in wastewater treatment. World J Microbiol Biotechnol. 2016;32(4):70. https://doi.org/10.1007/s11274-016-2028-1
  66. Dy RL, Rigano LA, Fineran PC. Phage-based biocontrol strategies and their application in agriculture and aquaculture. Biochem Soc Trans. 2018;46(6):1605–1613. https://doi.org/10.1042/BST20180178
  67. García R, Latz S, Romero J, Higuera G, García K, Bastías R. Bacteriophage production models: An Overview. Front Microbiol. 2019;10:1187. https://doi.org/10.3389/fmicb.2019.01187
  68. Romero-Calle D, GuimarãesBenevides R, Góes-Neto A, Billington C. Bacteriophages as alternatives to antibiotics in clinical care. Antibiotics (Basel). 2019;8(3):138. https://doi.org/10.3390/antibiotics8030138
  69. Abdelsattar AS, Dawooud A, Rezk N, et al. How to train your phage: The recent efforts in phage training. Biologics. 2021;1:70–88. https://doi.org/10.3390/biologics1020005
  70. Koskella B, Brockhurst MA. Bacteriaphage coevolution as a driver of ecological and evolutionary processes in microbial communities. FEMS Microbiol Rev. 2014;38(5):916–931. https://doi.org/10.1111/1574-6976.12072


Crossref Citations

No related citations found.