1
|
Mohan N, Bosco K, Peter A, Abhitha K, Bhat SG. Bacteriophage entrapment strategies for the treatment of chronic wound infections: a comprehensive review. Arch Microbiol 2024; 206:443. [PMID: 39443305 DOI: 10.1007/s00203-024-04168-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/29/2024] [Accepted: 10/12/2024] [Indexed: 10/25/2024]
Abstract
The growing threat of antimicrobial resistance has made the quest for antibiotic alternatives or synergists one of the most pressing priorities of the 21st century. The emergence of multidrug-resistance in most of the common wound pathogens has amplified the risk of antibiotic-resistant wound infections. Bacteriophages, with their self-replicating ability and targeted specificity, can act as suitable antibiotic alternatives. Nevertheless, targeted delivery of phages to infection sites remains a crucial issue, specifically in the case of topical infections. Hence, different phage delivery systems have been studied in recent years. However, there have been no recent reviews of phage delivery systems focusing exclusively on phage application on wounds. This review provides a compendium of all the major delivery systems that have been used to deliver phages to wound infection sites. Special focus has also been awarded to phage-embedded hydrogels with a discussion on the different aspects to be considered during their preparation.
Collapse
Affiliation(s)
- Nivedya Mohan
- Department of Biotechnology, Cochin University of Science and Technology, Kerala, 682022, India
- Department of Polymer Science and Rubber Technology, Cochin University of Science and Technology, Kerala, 682022, India
| | - Kiran Bosco
- Faculty of Medicine and Health, Sydney Medical School, University of Sydney, Sydney, NSW, Australia
- Center for Infectious Diseases and Microbiology, Westmead, NSW, Australia
| | - Anmiya Peter
- Department of Biotechnology, Cochin University of Science and Technology, Kerala, 682022, India
- Department of Polymer Science and Rubber Technology, Cochin University of Science and Technology, Kerala, 682022, India
| | - K Abhitha
- Department of Polymer Science and Rubber Technology, Cochin University of Science and Technology, Kerala, 682022, India
- Inter University Centre for Nanomaterials and Devices (IUCND), Cochin University of Science and Technology, Kerala, 682022, India
| | - Sarita G Bhat
- Department of Biotechnology, Cochin University of Science and Technology, Kerala, 682022, India.
- Inter University Centre for Nanomaterials and Devices (IUCND), Cochin University of Science and Technology, Kerala, 682022, India.
| |
Collapse
|
2
|
Rodriguez-Gonzalez RA, Balacheff Q, Debarbieux L, Marchi J, Weitz JS. Metapopulation model of phage therapy of an acute Pseudomonas aeruginosa lung infection. mSystems 2024; 9:e0017124. [PMID: 39230264 DOI: 10.1128/msystems.00171-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 07/24/2024] [Indexed: 09/05/2024] Open
Abstract
Infections caused by multidrug resistant (MDR) pathogenic bacteria are a global health threat. Bacteriophages ("phage") are increasingly used as alternative or last-resort therapeutics to treat patients infected by MDR bacteria. However, the therapeutic outcomes of phage therapy may be limited by the emergence of phage resistance during treatment and/or by physical constraints that impede phage-bacteria interactions in vivo. In this work, we evaluate the role of lung spatial structure on the efficacy of phage therapy for Pseudomonas aeruginosa infections. To do so, we developed a spatially structured metapopulation network model based on the geometry of the bronchial tree, including host innate immune responses and the emergence of phage-resistant bacterial mutants. We model the ecological interactions between bacteria, phage, and the host innate immune system at the airway (node) level. The model predicts the synergistic elimination of a P. aeruginosa infection due to the combined effects of phage and neutrophils, given the sufficient innate immune activity and efficient phage-induced lysis. The metapopulation model simulations also predict that MDR bacteria are cleared faster at distal nodes of the bronchial tree. Notably, image analysis of lung tissue time series from wild-type and lymphocyte-depleted mice revealed a concordant, statistically significant pattern: infection intensity cleared in the bottom before the top of the lungs. Overall, the combined use of simulations and image analysis of in vivo experiments further supports the use of phage therapy for treating acute lung infections caused by P. aeruginosa, while highlighting potential limits to therapy in a spatially structured environment given impaired innate immune responses and/or inefficient phage-induced lysis. IMPORTANCE Phage therapy is increasingly employed as a compassionate treatment for severe infections caused by multidrug-resistant (MDR) bacteria. However, the mixed outcomes observed in larger clinical studies highlight a gap in understanding when phage therapy succeeds or fails. Previous research from our team, using in vivo experiments and single-compartment mathematical models, demonstrated the synergistic clearance of acute P. aeruginosa pneumonia by phage and neutrophils despite the emergence of phage-resistant bacteria. In fact, the lung environment is highly structured, prompting the question of whether immunophage synergy explains the curative treatment of P. aeruginosa when incorporating realistic physical connectivity. To address this, we developed a metapopulation network model mimicking the lung branching structure to assess phage therapy efficacy for MDR P. aeruginosa pneumonia. The model predicts the synergistic elimination of P. aeruginosa by phage and neutrophils but emphasizes potential challenges in spatially structured environments, suggesting that higher innate immune levels may be required for successful bacterial clearance. Model simulations reveal a spatial pattern in pathogen clearance where P. aeruginosa are cleared faster at distal nodes of the bronchial tree than in primary nodes. Interestingly, image analysis of infected mice reveals a concordant and statistically significant pattern: infection intensity clears in the bottom before the top of the lungs. The combined use of modeling and image analysis supports the application of phage therapy for acute P. aeruginosa pneumonia while emphasizing potential challenges to curative success in spatially structured in vivo environments, including impaired innate immune responses and reduced phage efficacy.
Collapse
Affiliation(s)
- Rogelio A Rodriguez-Gonzalez
- Interdisciplinary Graduate Program in Quantitative Biosciences,Georgia Institute of Technology, Atlanta, Georgia, USA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Quentin Balacheff
- CHU Félix Guyon, Service des maladies respiratoires, La Réunion, France
| | - Laurent Debarbieux
- Department of Microbiology, Institut Pasteur, Université Paris Cité, CNRS UMR6047, Bacteriophage Bacterium Host, Paris, France
| | - Jacopo Marchi
- Department of Biology, University of Maryland, College Park, Maryland, USA
| | - Joshua S Weitz
- Department of Biology, University of Maryland, College Park, Maryland, USA
- Department of Physics, University of Maryland, College Park, Maryland, USA
- Institut de Biologie, École Normale Supérieure, Paris, France
| |
Collapse
|
3
|
Pérez LM, Havryliuk O, Infante N, Muniesa M, Morató J, Mariychuk R, Tzanov T. Biofilm Prevention and Removal in Non-Target Pseudomonas Strain by Siphovirus-like Coliphage. Biomedicines 2024; 12:2291. [PMID: 39457603 PMCID: PMC11504082 DOI: 10.3390/biomedicines12102291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/04/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives. Bacteriophages have gained significant interest as a potential solution to combat harmful bacteria, especially in the fight against antimicrobial resistance. With the rise in drug-resistant microorganisms, the medical community is increasingly exploring new alternatives to traditional antibiotics, and bacteriophages offer several advantages in this regard. However, phage applications still face some challenges, such as host specificity. Methods. In this study, a somatic Siphovirus-like coliphage (SOM7) was tested for inhibiting the biofilm-forming capacity of the non-target strain Pseudomonas aeruginosa (ATTC 10145). The phage-sensitive strain E. coli WG5 was used as a control. The selected microorganisms were first tested for growth in the presence of SOM7 at three different concentrations (105, 107, and 109 PFU/mL). Results. As expected, the phage-sensitive E. coli WG5 was fully inhibited by the coliphage, and no phage-related affection on the growth rate was observed for the SOM7-resistant P. aeruginosa. More notably, increasing concentrations of SOM7 significantly reduced both the biofilm-forming capacity and the amount of pre-established bacterial biofilm of the phage-insensitive P. aeruginosa (24.9% and 38.8% reduction in the biofilm-forming ability, and 18.8% and 28.0% biofilm degradation for 107 PFU/mL and 109 PFU/mL SOM7, respectively; p < 0.05). These results were supported by transmission electron microscopy (TEM) imaging, providing unprecedent evidence for the interaction of the somatic coliphage with the non-host strain. Conclusions. Although more studies in other biofilm models are necessary, our results show for the very first time that bacteriophages could potentially be used as an alternative to achieve desired anti-biofilm and biofilm-degrading activity in non-host bacterial strains.
Collapse
Affiliation(s)
- Leonardo Martín Pérez
- Laboratory of Sanitary and Environmental Microbiology (MSMLab)-UNESCO Chair on Sustainability, Universitat Politècnica de Catalunya-BarcelonaTech, R/Sant Nebridi, 22, GAIA Building (TR14), 08222 Terrassa, Spain; (L.M.P.); (O.H.); (N.I.); (J.M.)
- Grup de Biotecnologia Molecular i Industrial, Departament d’Enginyeria Química, Universitat Politècnica de Catalunya (UPC-BarcelonaTech), Rambla de Sant Nebridi 22, 08222 Terrassa, Spain;
| | - Olesia Havryliuk
- Laboratory of Sanitary and Environmental Microbiology (MSMLab)-UNESCO Chair on Sustainability, Universitat Politècnica de Catalunya-BarcelonaTech, R/Sant Nebridi, 22, GAIA Building (TR14), 08222 Terrassa, Spain; (L.M.P.); (O.H.); (N.I.); (J.M.)
- Department of Extremophilic Microorganisms Biology, Zabolotny Institute of Microbiology and Virology, National Academy of Sciences of Ukraine, 03143 Kyiv, Ukraine
| | - Nury Infante
- Laboratory of Sanitary and Environmental Microbiology (MSMLab)-UNESCO Chair on Sustainability, Universitat Politècnica de Catalunya-BarcelonaTech, R/Sant Nebridi, 22, GAIA Building (TR14), 08222 Terrassa, Spain; (L.M.P.); (O.H.); (N.I.); (J.M.)
| | - Maite Muniesa
- Departament de Genètica, Microbiologia i Estadística, Universitat de Barcelona, Diagonal 643 (Annex. Floor 0), 08028 Barcelona, Spain;
| | - Jordi Morató
- Laboratory of Sanitary and Environmental Microbiology (MSMLab)-UNESCO Chair on Sustainability, Universitat Politècnica de Catalunya-BarcelonaTech, R/Sant Nebridi, 22, GAIA Building (TR14), 08222 Terrassa, Spain; (L.M.P.); (O.H.); (N.I.); (J.M.)
| | - Ruslan Mariychuk
- Department of Ecology, Faculty of Humanities and Natural Sciences, University of Presov, 08001 Presov, Slovakia
| | - Tzanko Tzanov
- Grup de Biotecnologia Molecular i Industrial, Departament d’Enginyeria Química, Universitat Politècnica de Catalunya (UPC-BarcelonaTech), Rambla de Sant Nebridi 22, 08222 Terrassa, Spain;
| |
Collapse
|
4
|
Martinet MG, Lohde M, Higazy D, Brandt C, Pletz MW, Middelboe M, Makarewicz O, Ciofu O. Diversification of Pseudomonas aeruginosa Biofilm Populations under Repeated Phage Exposures Decreases the Efficacy of the Treatment. Microorganisms 2024; 12:1880. [PMID: 39338555 PMCID: PMC11434582 DOI: 10.3390/microorganisms12091880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Phage therapy has been proposed as a therapeutic alternative to antibiotics for the treatment of chronic, biofilm-related P. aeruginosa infections. To gain a deeper insight into the complex biofilm-phage interactions, we investigated in the present study the effect of three successive exposures to lytic phages of biofilms formed by the reference strains PAO1 and PA14 as well as of two sequential clinical P. aeruginosa isolates from the sputum of a patient with cystic fibrosis (CF). The Calgary device was employed as a biofilm model and the efficacy of phage treatment was evaluated by measurements of the biomass stained with crystal violet (CV) and of the cell density of the biofilm bacterial population (CFU/mL) after each of the three phage exposures. The genetic alterations of P. aeruginosa isolates from biofilms exposed to phages were investigated by whole-genome sequencing. We show here that the anti-biofilm efficacy of the phage treatment decreased rapidly with repeated applications of lytic phages on P. aeruginosa strains with different genetic backgrounds. Although we observed the maintenance of a small subpopulation of sensitive cells after repeated phage treatments, a fast recruitment of mechanisms involved in the persistence of biofilms to the phage attack occurred, mainly by mutations causing alterations of the phage receptors. However, mutations causing phage-tolerant phenotypes such as alginate-hyperproducing mutants were also observed. In conclusion, a decreased anti-biofilm effect occurred after repeated exposure to lytic phages of P. aeruginosa biofilms due to the recruitment of different resistance and tolerance mechanisms.
Collapse
Affiliation(s)
- Mark Grevsen Martinet
- Institute of Infectious Diseases and Infection Control, Jena University Hospital, 07747 Jena, Germany; (M.G.M.); (M.L.); (C.B.); (M.W.P.); (O.M.)
| | - Mara Lohde
- Institute of Infectious Diseases and Infection Control, Jena University Hospital, 07747 Jena, Germany; (M.G.M.); (M.L.); (C.B.); (M.W.P.); (O.M.)
| | - Doaa Higazy
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark;
| | - Christian Brandt
- Institute of Infectious Diseases and Infection Control, Jena University Hospital, 07747 Jena, Germany; (M.G.M.); (M.L.); (C.B.); (M.W.P.); (O.M.)
- Leibniz Center for Photonics in Infection Research (LPI), 07745 Jena, Germany
| | - Mathias W. Pletz
- Institute of Infectious Diseases and Infection Control, Jena University Hospital, 07747 Jena, Germany; (M.G.M.); (M.L.); (C.B.); (M.W.P.); (O.M.)
- Leibniz Center for Photonics in Infection Research (LPI), 07745 Jena, Germany
| | - Mathias Middelboe
- Marine Biological Section, Department of Biology, University of Copenhagen, 3000 Helsingør, Denmark;
- Department of Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Oliwia Makarewicz
- Institute of Infectious Diseases and Infection Control, Jena University Hospital, 07747 Jena, Germany; (M.G.M.); (M.L.); (C.B.); (M.W.P.); (O.M.)
- Leibniz Center for Photonics in Infection Research (LPI), 07745 Jena, Germany
| | - Oana Ciofu
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark;
| |
Collapse
|
5
|
Tagliaferri TL, Rhode S, Muñoz P, Simon K, Krüttgen A, Stoppe C, Ruhl T, Beier JP, Horz HP, Kim BS. Antiseptic management of critical wounds: differential bacterial response upon exposure to antiseptics and first insights into antiseptic/phage interactions. Int J Surg 2024; 110:5374-5384. [PMID: 38742847 PMCID: PMC11392177 DOI: 10.1097/js9.0000000000001605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/29/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND With the antibiotic crisis, the topical antibacterial control including chronic wounds gains increasing importance. However, little is known regarding tolerance development when bacteria face repetitive exposure to the identical antiseptics as commonly found in clinical practice. MATERIALS AND METHODS Clinical isolates foremost of chronic wounds were exposed in vitro to dilutions of two antiseptics used for wound therapy: polyhexanide or octenidine. Adaptive response was determined by growth/kill curves, minimal inhibitory concentration (MIC), and whole genome sequencing. Antiseptic/bacteriophage combinations were studied by liquid-infection assays and bacterial plating. RESULTS Polyhexanide acted stronger against Escherichia coli and Proteus mirabilis while octenidine was more potent against Staphylococcus aureus . Otherwise, the antiseptic efficacy varied across isolates of Klebsiella pneumoniae , Pseudomonas aeruginosa , and Acinetobacter baumannii . Upon repetitive exposure with constant antiseptic concentrations P. aeruginosa and P. mirabilis adaptation was evident by a reduced lag-phase and a twofold increased MIC. Under increasing octenidine concentrations, P. aeruginosa adapted to an eightfold higher dosage with mutations in smvA , opgH , and kinB affecting an efflux pump, alginate and biofilm formation, respectively. S. aureus adapted to a fourfold increase of polyhexanide with a mutation in the multiple peptide resistance factor MprF, also conferring cross-resistance to daptomycin. Antiseptic/bacteriophage combinations enhanced bacterial inhibition and delayed adaptation. CONCLUSION Different bacterial species/strains respond unequally to low-level antiseptic concentrations. Bacterial adaptation potential at phenotypic and genotypic levels may indicate the necessity for a more nuanced selection of antiseptics. Bacteriophages represent a promising yet underexplored strategy for supporting antiseptic treatment, which may be particularly beneficial for the management of critical wounds.
Collapse
Affiliation(s)
| | - Sophie Rhode
- Department of Plastic Surgery, Hand Surgery - Burn Center, RWTH Aachen University Hospital, Aachen, Germany
- Department of Plastic, Reconstructive and esthetic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg Germany
| | - Priscila Muñoz
- Institute of Medical Microbiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Kevin Simon
- Institute of Medical Microbiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Alex Krüttgen
- Laboratory Diagnostic Center, RWTH Aachen University Hospital, Aachen, Germany
| | - Christian Stoppe
- University Hospital, Würzburg, Department of Anesthesiology, Intensive Care, Emergency and Pain Medicine, Würzburg Germany
- Department of Cardiac Anesthesiology and Intensive Care Medicine, Charité Berlin, Berlin, Germany
| | - Tim Ruhl
- Department of Plastic Surgery, Hand Surgery - Burn Center, RWTH Aachen University Hospital, Aachen, Germany
| | - Justus P Beier
- Department of Plastic Surgery, Hand Surgery - Burn Center, RWTH Aachen University Hospital, Aachen, Germany
| | - Hans-Peter Horz
- Institute of Medical Microbiology, RWTH Aachen University Hospital, Aachen, Germany
| | - Bong-Sung Kim
- Department of Plastic Surgery, Hand Surgery - Burn Center, RWTH Aachen University Hospital, Aachen, Germany
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
6
|
Ziller L, Blum PC, Buhl EM, Krüttgen A, Horz HP, Tagliaferri TL. Newly isolated Drexlerviridae phage LAPAZ is physically robust and fosters eradication of Klebsiella pneumoniae in combination with meropenem. Virus Res 2024; 347:199417. [PMID: 38880333 PMCID: PMC11245953 DOI: 10.1016/j.virusres.2024.199417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/11/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024]
Abstract
Due to the spread of multidrug resistance there is a renewed interest in using bacteriophages (briefly: phages) for controlling bacterial pathogens. The objective of this study was the characterization of a newly isolated phage (i.e. phage LAPAZ, vB_KpnD-LAPAZ), its antimicrobial activity against multidrug resistant Klebsiella pneumoniae and potential synergistic interactions with antibiotics. LAPAZ belongs to the family Drexlerviridae (genus: Webervirus) and lysed 30 % of tested strains, whereby four distinct capsular types can be infected. The genome consists of 51,689 bp and encodes 84 ORFs. The latent period is 30 min with an average burst size of 27 PFU/cell. Long-term storage experiments show that LAPAZ is significantly more stable in wastewater compared to laboratory media. A phage titre of 90 % persists up to 30 min at 50 ˚C and entire phage loss was seen only at temperatures > 66 ˚C. Besides stability against UV-C, antibacterial activity in liquid culture medium was consistent at pH values ranging from 4 to 10. Unlike exposure to phage or antibiotic alone, synergistic interactions and a complete bacterial eradication was achieved when combining LAPAZ with meropenem. In addition, synergism with the co-presence of ciprofloxacin was observed and phage resistance emergence could be delayed. Without co-addition of the antibiotic, phage resistant mutants readily emerged and showed a mixed pattern of drug sensitivity alterations. Around 88 % became less sensitive towards ceftazidime, meropenem and gentamicin. Conversely, around 44 % showed decreased resistance levels against ciprofloxacin. Whole genome analysis of a phage-resistant mutant with a 16-fold increased sensitivity towards ciprofloxacin revealed one de novo frameshift mutation leading to a gene fusion affecting two transport proteins belonging to the major facilitator-superfamily (MFS). Apparently, this mutation compromises ciprofloxacin efflux efficiency and further studies are warranted to understand how the non-mutated protein might be involved in phage-host adsorption.
Collapse
Affiliation(s)
- Leonie Ziller
- Institute of Medical Microbiology, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | | | - Eva Miriam Buhl
- Electron Microscopy Facility, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Alex Krüttgen
- Laboratory Diagnostic Center, RWTH Aachen University Hospital, 52074 Aachen, Germany
| | - Hans-Peter Horz
- Institute of Medical Microbiology, RWTH Aachen University Hospital, 52074 Aachen, Germany.
| | | |
Collapse
|
7
|
Martinho I, Braz M, Duarte J, Brás A, Oliveira V, Gomes NCM, Pereira C, Almeida A. The Potential of Phage Treatment to Inactivate Planktonic and Biofilm-Forming Pseudomonas aeruginosa. Microorganisms 2024; 12:1795. [PMID: 39338470 PMCID: PMC11433742 DOI: 10.3390/microorganisms12091795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 08/18/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
Pseudomonas aeruginosa is a common cause of hospital-acquired infections and exhibits a strong resistance to antibiotics. An alternative treatment option for bacterial infections is the use of bacteriophages (or phages). In this study, two distinct phages, VB_PaD_phPA-G (phPA-G) and VB_PaN_phPA-Intesti (phPA-Intesti), were used as single suspensions or in a phage cocktail to inactivate the planktonic cells and biofilms of P. aeruginosa. Preliminary experiments in culture medium showed that phage phPA-Intesti (reductions of 4.5-4.9 log CFU/mL) outperformed phPA-G (reductions of 0.6-2.6 log CFU/mL) and the phage cocktail (reduction of 4.2 log CFU/mL). Phage phPA-Intesti caused a maximum reduction of 5.5 log CFU/cm2 in the P. aeruginosa biofilm in urine after 4 h of incubation. The combination of phage phPA-Intesti and ciprofloxacin did not improve the efficacy of bacterial inactivation nor reduce the development of resistant mutants. However, the development of resistant bacteria was lower in the combined treatment with the phage and the antibiotic compared to treatment with the antibiotic alone. This phage lacks known toxins, virulence, antibiotic resistance, and integrase genes. Overall, the results suggest that the use of phage phPA-Intesti could be a potential approach to control urinary tract infections (UTIs), namely those caused by biofilm-producing and multidrug-resistant strains of P. aeruginosa.
Collapse
Affiliation(s)
- Inês Martinho
- Department of Biology and CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Márcia Braz
- Department of Biology and CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - João Duarte
- Department of Biology and CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Ana Brás
- Department of Biology and CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Vanessa Oliveira
- Department of Biology and CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Newton C M Gomes
- Department of Biology and CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Carla Pereira
- Department of Biology and CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Adelaide Almeida
- Department of Biology and CESAM, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| |
Collapse
|
8
|
Ikpe F, Williams T, Orok E, Ikpe A. Antimicrobial resistance: use of phage therapy in the management of resistant infections. Mol Biol Rep 2024; 51:925. [PMID: 39167154 DOI: 10.1007/s11033-024-09870-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 08/16/2024] [Indexed: 08/23/2024]
Abstract
The emergence and increase in antimicrobial resistance (AMR) is now widely recognized as a major public health challenge. Traditional antimicrobial drugs are becoming increasingly ineffective, while the development of new antibiotics is waning. As a result, alternative treatments for infections are garnering increased interest. Among these alternatives, bacteriophages, also known as phages, are gaining renewed attention and are reported to offer a promising solution to alleviate the burden of bacterial infections. This review discusses the current successes of phage therapy (PT) against multidrug-resistant organisms (MDROs), such as Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, and Enterobacter spp. The review also compares the efficacy of PT with that of chemical antibiotics, reporting on its benefits and limitations, while highlighting its impact on the human gut microbiome and immune system. Despite its potential, phage therapy is reported to face challenges such as the narrow antibacterial range, the complexity of developing phage cocktails, and the need for precise dosing and duration protocols. Nevertheless, continued research, improved regulatory frameworks, and increased public awareness are essential to realize its full potential and integration into standard medical practice, paving the way for innovative treatments that can effectively manage infections in an era of rising antimicrobial resistance.
Collapse
Affiliation(s)
- Favour Ikpe
- Department of Pharmaceutical Microbiology and Biotechnology, College of Pharmacy, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Tonfamoworio Williams
- Department of Pharmaceutical Microbiology and Biotechnology, College of Pharmacy, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Edidiong Orok
- Department of Clinical Pharmacy and Public Health, College of Pharmacy, Afe Babalola University, Ado-Ekiti, Nigeria.
| | - Augustine Ikpe
- Department of Sciences, Champion Group of Schools, Okene, Kogi State, Nigeria
| |
Collapse
|
9
|
Duarte AC, Fernández L, Jurado A, Campelo AB, Shen Y, Rodríguez A, García P. Synergistic removal of Staphylococcus aureus biofilms by using a combination of phage Kayvirus rodi with the exopolysaccharide depolymerase Dpo7. Front Microbiol 2024; 15:1438022. [PMID: 39171257 PMCID: PMC11335607 DOI: 10.3389/fmicb.2024.1438022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/29/2024] [Indexed: 08/23/2024] Open
Abstract
Introduction Bacteriophages have been shown to penetrate biofilms and replicate if they find suitable host cells. Therefore, these viruses appear to be a good option to tackle the biofilm problem and complement or even substitute more conventional antimicrobials. However, in order to successfully remove biofilms, in particular mature biofilms, phages may need to be administered along with other compounds. Phage-derived proteins, such as endolysins or depolymerases, offer a safer alternative to other compounds in the era of antibiotic resistance. Methods This study examined the interactions between phage Kayvirus rodi with a polysaccharide depolymerase (Dpo7) from another phage (Rockefellervirus IPLA7) against biofilms formed by different Staphylococcus aureus strains, as determined by crystal violet staining, viable cell counts and microscopy analysis. Results and discussion Our results demonstrated that there was synergy between the two antimicrobials, with a more significant decreased in biomass and viable cell number with the combination treatment compared to the phage and enzyme alone. This observation was confirmed by microscopy analysis, which also showed that polysaccharide depolymerase treatment reduced, but did not eliminate extracellular matrix polysaccharides. Activity assays on mutant strains did not identify teichoic acids or PNAG/PIA as the exclusive target of Dpo7, suggesting that may be both are degraded by this enzyme. Phage adsorption to S. aureus cells was not significantly altered by incubation with Dpo7, indicating that the mechanism of the observed synergistic interaction is likely through loosening of the biofilm structure. This would allow easier access of the phage particles to their host cells and facilitate infection progression within the bacterial population.
Collapse
Affiliation(s)
- Ana Catarina Duarte
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Spain
- DairySafe Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Lucía Fernández
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Spain
- DairySafe Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Andrea Jurado
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Spain
- DairySafe Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Ana Belén Campelo
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Spain
| | - Yang Shen
- Laboratory of Food Microbiology, Institute of Food, Nutrition and Health, Zurich, Switzerland
| | - Ana Rodríguez
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Spain
- DairySafe Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Pilar García
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Spain
- DairySafe Group, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| |
Collapse
|
10
|
Parihar A, Vishwakarma P, Khan R. Miniaturized MXene-based electrochemical biosensors for virus detection. Bioelectrochemistry 2024; 158:108700. [PMID: 38582009 DOI: 10.1016/j.bioelechem.2024.108700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 03/29/2024] [Accepted: 03/31/2024] [Indexed: 04/08/2024]
Abstract
The timely control of infectious diseases can prevent the spread of infections and mitigate the significant socio-economic damage witnessed during recent pandemics. Diagnostic methods play a significant role in detecting highly contagious agents, such as viruses, to prevent further transmission. The emergence of advanced point-of-care techniques offers several advantages over conventional approaches for detecting infectious agents. These techniques are highly sensitive, rapid, can be miniaturized, and are cost-effective. Recently, MXene-based 2D nanocomposites have proven beneficial for fabricating electrochemical biosensors due to their suitable electrical, optical, and mechanical properties. This article covers electrochemical biosensors based on MXene nanocomposite for the detection of viruses, along with the associated challenges and future possibilities. Additionally, we highlight various conventional techniques for the detection of infectious agents, discussing their pros and cons. We delve into the challenges faced during the fabrication of MXene-based biosensors and explore future endeavors. It is anticipated that the information presented in this work will pave the way for the development of Point-of-Care (POC) devices capable of sensitive and selective virus detection, enhancing preparedness for ongoing and future pandemics.
Collapse
Affiliation(s)
- Arpana Parihar
- Industrial Waste Utilization, Nano and Biomaterials, CSIR-Advanced Materials and Processes Research Institute (AMPRI), Hoshangabad Road, Bhopal- 462026, MP, India
| | - Preeti Vishwakarma
- Department of Microbiology, Barkatullah University, Hoshangabad Road, Bhopal- 462026, MP, India
| | - Raju Khan
- Industrial Waste Utilization, Nano and Biomaterials, CSIR-Advanced Materials and Processes Research Institute (AMPRI), Hoshangabad Road, Bhopal- 462026, MP, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad- 201002, India.
| |
Collapse
|
11
|
Norton P, Trus P, Wang F, Thornton MJ, Chang C. Understanding and treating diabetic foot ulcers: Insights into the role of cutaneous microbiota and innovative therapies. SKIN HEALTH AND DISEASE 2024; 4:e399. [PMID: 39104636 PMCID: PMC11297444 DOI: 10.1002/ski2.399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/22/2024] [Accepted: 05/18/2024] [Indexed: 08/07/2024]
Abstract
Background Notoriously known as the silent pandemic, chronic, non-healing diabetic foot ulcers (DFUs), pose a significant rate of incidence for amputation and are a major cause of morbidity. Alarmingly, the treatment and management strategies of chronic wounds represent a significant economic and health burden as well as a momentous drain on resources with billions per annum being spent in the US and UK alone. Defective wound healing is a major pathophysiological condition which propagates an acute wound to a chronic wound, further propelled by underlying conditions such as diabetes and vascular complications which are more prevalent amongst the elderly. Chronic wounds are prone to infection, which can exacerbate the condition, occasionally resulting in amputation for the patient, despite the intervention of modern therapies. However, amputation can only yield a 5-year survival rate for 50% of patients, highlighting the need for new treatments for chronic wounds. Findings The dynamic cutaneous microbiota is comprised of diverse microorganisms that often aid wound healing. Conversely, the chronic wound microbiome consists of a combination of common skin commensals such as Staphylococcus aureus and Staphylococcus epidermidis, as well as the opportunistic pathogen Pseudomonas aeruginosa. These bacteria have been identified as the most prevalent bacterial pathogens isolated from chronic wounds and contribute to prolific biofilm formation decreasing the efficiency of antimicrobials and further perpetuating a hyper-inflammatory state. Discussion and Conclusion Here, we review recent advances and provide a new perspective on alternative treatments including phage and microbiome transplant therapies and how the definitive role of the cutaneous microbiota impacts the aetiology of DFUs.
Collapse
Affiliation(s)
- Paul Norton
- School of Dental SciencesFaculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneUK
- Biosciences InstituteFaculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneUK
- Centre for Skin SciencesFaculty of Life SciencesUniversity of BradfordBradfordUK
| | - Pavlos Trus
- School of Dental SciencesFaculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneUK
- Biosciences InstituteFaculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneUK
| | - Fengyi Wang
- School of Dental SciencesFaculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneUK
- Biosciences InstituteFaculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneUK
| | - M. Julie Thornton
- Centre for Skin SciencesFaculty of Life SciencesUniversity of BradfordBradfordUK
| | - Chien‐Yi Chang
- School of Dental SciencesFaculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneUK
- Biosciences InstituteFaculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneUK
| |
Collapse
|
12
|
Guo Z, Yuan M, Chai J. Mini review advantages and limitations of lytic phages compared with chemical antibiotics to combat bacterial infections. Heliyon 2024; 10:e34849. [PMID: 39148970 PMCID: PMC11324966 DOI: 10.1016/j.heliyon.2024.e34849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/10/2024] [Accepted: 07/17/2024] [Indexed: 08/17/2024] Open
Abstract
The overuse of antibiotics has caused the emergence of antibiotic-resistant strains, such as multidrug-resistant, extensively drug-resistant, and pandrug-resistant bacteria. The treatment of infections caused by such strains has become a formidable challenge. In the post-antibiotic era, phage therapy is an attractive solution for this problem and some successful phase 1 and 2 studies have demonstrated the efficacy and safety of phage therapy over the last decade. It is a form of evolutionary medicine, phages exhibit immunomodulatory and anti-inflammatory properties. However, phage therapy is limited by factors, such as the narrow spectrum of host strains, the special pharmacokinetics and pharmacodynamics in vivo, immune responses, and the development of phage resistance. The aim of this minireview was to compare the potencies of lytic phages and chemical antibiotics to treat bacterial infections. The advantages of phage therapy has fewer side effects, self-replication, evolution, bacterial biofilms eradication, immunomodulatory and anti-inflammatory properties compared with chemical antibiotics. Meanwhile, the disadvantages of phage therapy include the narrow spectrum of available host strains, the special pharmacokinetics and pharmacodynamics in vivo, immune responses, and phage resistance hurdles. Recently, some researchers continue to make efforts to overcome these limitations of phage therapy. Phage therapy will be a welcome addition to the gamut of options available for treating antibiotic-resistant bacterial infections. We focus on the advantages and limitations of phage therapy with the intention of exploiting the advantages and overcoming the limitations.
Collapse
Affiliation(s)
- Zhimin Guo
- Department of Laboratory Medicine, Infectious Diseases and Pathogen Biology Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Mengyao Yuan
- Department of Laboratory Medicine, Infectious Diseases and Pathogen Biology Center, The First Hospital of Jilin University, Changchun, 130021, China
| | - Jiannan Chai
- Department of Laboratory Medicine, The First Hospital of Jilin University, Changchun, 130021, China
| |
Collapse
|
13
|
Shi Z, Hong X, Li Z, Zhang M, Zhou J, Zhao Z, Qiu S, Liu G. Characterization of the novel broad-spectrum lytic phage Phage_Pae01 and its antibiofilm efficacy against Pseudomonas aeruginosa. Front Microbiol 2024; 15:1386830. [PMID: 39091310 PMCID: PMC11292732 DOI: 10.3389/fmicb.2024.1386830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024] Open
Abstract
Introduction Pseudomonas aeruginosa is present throughout nature and is a common opportunistic pathogen in the human body. Carbapenem antibiotics are typically utilized as a last resort in the clinical treatment of multidrug-resistant infections caused by P. aeruginosa. The increase in carbapenem-resistant P. aeruginosa poses an immense challenge for the treatment of these infections. Bacteriophages have the potential to be used as antimicrobial agents for treating antibiotic-resistant bacteria. Methods and Results In this study, a new virulent P. aeruginosa phage, Phage_Pae01, was isolated from hospital sewage and shown to have broad-spectrum antibacterial activity against clinical P. aeruginosa isolates (83.6%). These clinical strains included multidrug-resistant P. aeruginosa and carbapenem-resistant P. aeruginosa. Transmission electron microscopy revealed that the phage possessed an icosahedral head of approximately 80 nm and a long tail about 110 m, indicating that it belongs to the Myoviridae family of the order Caudovirales. Biological characteristic analysis revealed that Phage_Pae01 could maintain stable activity in the temperature range of 4~ 60°C and pH range of 4 ~ 10. According to the in vitro lysis kinetics of the phage, Phage_Pae01 demonstrated strong antibacterial activity. The optimal multiplicity of infection was 0.01. The genome of Phage_Pae01 has a total length of 93,182 bp and contains 176 open reading frames (ORFs). The phage genome does not contain genes related to virulence or antibiotic resistance. In addition, Phage_Pae01 effectively prevented the formation of biofilms and eliminated established biofilms. When Phage_Pae01 was combined with gentamicin, it significantly disrupted established P. aeruginosa biofilms. Conclusion We identified a novel P. aeruginosa phage and demonstrated its effective antimicrobial properties against P. aeruginosa in both the floating and biofilm states. These findings offer a promising approach for the treatment of drug-resistant bacterial infections in clinical settings.
Collapse
Affiliation(s)
- Zhixin Shi
- Department of Laboratory Medicine, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
- National Key Clinical Department of Laboratory Medicine, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Xin Hong
- Department of Laboratory Medicine, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Zexuan Li
- Department of Laboratory Medicine, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Meijuan Zhang
- Department of Laboratory Medicine, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Jun Zhou
- Department of Laboratory Medicine, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Zhe Zhao
- College of Oceanography, Hohai University, Nanjing, China
| | - Shengfeng Qiu
- Department of Laboratory Medicine, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Genyan Liu
- Department of Laboratory Medicine, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
- National Key Clinical Department of Laboratory Medicine, The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| |
Collapse
|
14
|
Subramanian A. Emerging roles of bacteriophage-based therapeutics in combating antibiotic resistance. Front Microbiol 2024; 15:1384164. [PMID: 39035437 PMCID: PMC11257900 DOI: 10.3389/fmicb.2024.1384164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 06/06/2024] [Indexed: 07/23/2024] Open
Abstract
Amid the growing challenge of antibiotic resistance on a global scale, there has been a notable resurgence in bacteriophage-based treatments, signaling a shift in our approach to managing infections. Bacteriophages (BPs), bacterial predators of nature, present a promising alternative for tackling infections caused by antibiotic-resistant pathogens. This review delves into the intricate relationship between bacteriophages and resistant bacteria, exploring various treatment strategies. Drawing upon both preclinical and clinical studies, the review highlights the effectiveness of bacteriophage therapy, particularly when integrated synergistically with conventional antibiotics. It discusses various treatment approaches for systemic and localized infections, demonstrating the adaptability of bacteriophage therapy across different clinical scenarios. Furthermore, the formulation and delivery of bacteriophages shed light on the various methods used to encapsulate and administer them effectively. It also acknowledges the challenge of bacterial resistance to bacteriophages and the ongoing efforts to overcome this hurdle. In addition, this review highlights the importance of the bacteriophage sensitivity profile (phagogram), which helps tailor treatment regimens to individual patients and specific pathogens. By surpassing the limitations of traditional antibiotics, bacteriophage-based therapies offer a personalized and potent solution against antibiotic resistance, promising to reshape the future of infectious disease management.
Collapse
|
15
|
Alipour-Khezri E, Skurnik M, Zarrini G. Pseudomonas aeruginosa Bacteriophages and Their Clinical Applications. Viruses 2024; 16:1051. [PMID: 39066214 PMCID: PMC11281547 DOI: 10.3390/v16071051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Antimicrobial resistance poses a serious risk to contemporary healthcare since it reduces the number of bacterial illnesses that may be treated with antibiotics, particularly for patients with long-term conditions like cystic fibrosis (CF). People with a genetic predisposition to CF often have recurrent bacterial infections in their lungs due to a buildup of sticky mucus, necessitating long-term antibiotic treatment. Pseudomonas aeruginosa infections are a major cause of CF lung illness, and P. aeruginosa airway isolates are frequently resistant to many antibiotics. Bacteriophages (also known as phages), viruses that infect bacteria, are a viable substitute for antimicrobials to treat P. aeruginosa infections in individuals with CF. Here, we reviewed the utilization of P. aeruginosa bacteriophages both in vivo and in vitro, as well as in the treatment of illnesses and diseases, and the outcomes of the latter.
Collapse
Affiliation(s)
- Elaheh Alipour-Khezri
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz 51368, Iran;
| | - Mikael Skurnik
- Human Microbiome Research Program, and Department of Bacteriology and Immunology, Faculty of Medicine, University of Helsinki, 00290 Helsinki, Finland
| | - Gholamreza Zarrini
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz 51368, Iran;
- Microbial Biotechnology Research Group, University of Tabriz, Tabriz 51368, Iran
| |
Collapse
|
16
|
Al-Anany AM, Fatima R, Nair G, Mayol JT, Hynes AP. Temperate phage-antibiotic synergy across antibiotic classes reveals new mechanism for preventing lysogeny. mBio 2024; 15:e0050424. [PMID: 38757974 PMCID: PMC11237771 DOI: 10.1128/mbio.00504-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 04/18/2024] [Indexed: 05/18/2024] Open
Abstract
A recent demonstration of synergy between a temperate phage and the antibiotic ciprofloxacin suggested a scalable approach to exploiting temperate phages in therapy, termed temperate phage-antibiotic synergy, which specifically interacted with the lysis-lysogeny decision. To determine whether this would hold true across antibiotics, we challenged Escherichia coli with the phage HK97 and a set of 13 antibiotics spanning seven classes. As expected, given the conserved induction pathway, we observed synergy with classes of drugs known to induce an SOS response: a sulfa drug, other quinolones, and mitomycin C. While some β-lactams exhibited synergy, this appeared to be traditional phage-antibiotic synergy, with no effect on the lysis-lysogeny decision. Curiously, we observed a potent synergy with antibiotics not known to induce the SOS response: protein synthesis inhibitors gentamicin, kanamycin, tetracycline, and azithromycin. The synergy results in an eightfold reduction in the effective minimum inhibitory concentration of gentamicin, complete eradication of the bacteria, and, when administered at sub-optimal doses, drastically decreases the frequency of lysogens emerging from the combined challenge. However, lysogens exhibit no increased sensitivity to the antibiotic; synergy was maintained in the absence of RecA; and the antibiotic reduced the initial frequency of lysogeny rather than selecting against formed lysogens. Our results confirm that SOS-inducing antibiotics broadly result in temperate-phage-specific synergy, but that other antibiotics can interact with temperate phages specifically and result in synergy. This is the first report of a means of chemically blocking entry into lysogeny, providing a new means for manipulating the key lysis-lysogeny decision.IMPORTANCEThe lysis-lysogeny decision is made by most bacterial viruses (bacteriophages, phages), determining whether to kill their host or go dormant within it. With over half of the bacteria containing phages waiting to wake, this is one of the most important behaviors in all of biology. These phages are also considered unusable for therapy because of this behavior. In this paper, we show that many antibiotics bias this behavior to "wake" the dormant phages, forcing them to kill their host, but some also prevent dormancy in the first place. These will be important tools to study this critical decision point and may enable the therapeutic use of these phages.
Collapse
Affiliation(s)
- Amany M Al-Anany
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Rabia Fatima
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Gayatri Nair
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Jordan T Mayol
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Alexander P Hynes
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
- Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
17
|
Ali S, Aslam MA, Kanwar R, Mehmood Z, Arshad MI, Hussain S. Phage-antibiotic synergism against Salmonella typhi isolated from stool samples of typhoid patients. Ir J Med Sci 2024; 193:1377-1384. [PMID: 38158479 DOI: 10.1007/s11845-023-03599-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/19/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND Typhoid fever is a fatal disease in humans that is caused by Salmonella typhi. S. typhi infections need immediate antibiotic therapy, and their extensive use has led to multidrug-resistant (MDR) pathogens. The use of bacteriophages is becoming a new way to treat these resistant bacteria. This research was directed to bacteriophage isolation against S. typhi and to determine phage-antibiotic synergism. AIMS To isolate bacteriophages targeting S. typhi, the causative agent of typhoid fever, and investigate their potential synergistic effects when combined with antibiotics. STUDY DESIGN A cross-sectional study. METHODS The Widal test was positive; twenty diarrheal stool samples were taken, and for confirmation of S. typhi, different biochemical tests were performed. The disc-diffusion technique was used to determine antimicrobial resistance, and the double agar overlay method was used for bacteriophage isolation from sewage water against S. typhi. To test antibiotic-phage synergism, the S. typhi bacteria was treated by phages together with varying antibiotic concentrations. RESULTS Eleven samples were positive for S. typhi with black colonies on SS-agar. These were catalase and MR positive with alkali butt on TSI. Clear plaques were observed after the agar overlay. Isolated phages were stable at various pH and temperature levels. Synergism was observed on agar plate. The zone was enlarged when phages were combined with bacterial lawn culture and ciprofloxacin disk. Bacterial growth inhibition had a significant p-value of 0.03 in titration plates, with the phage-ciprofloxacin combination being more effective than the phage and antibiotic alone. CONCLUSION The study highlights the synergistic effects of isolated bacteriophages with antibiotics, which are not only effective against S. typhi infection but also decrease antibiotic resistance.
Collapse
Affiliation(s)
- Saqib Ali
- Institute of Microbiology, University of Agriculture Faisalabad, Faisalabad, 38000, Pakistan
| | - Muhammad Aamir Aslam
- Institute of Microbiology, University of Agriculture Faisalabad, Faisalabad, 38000, Pakistan
| | - Rabia Kanwar
- Institute of Microbiology, University of Agriculture Faisalabad, Faisalabad, 38000, Pakistan
| | - Zain Mehmood
- Institute of Microbiology, University of Agriculture Faisalabad, Faisalabad, 38000, Pakistan
| | - Muhammad Imran Arshad
- Institute of Microbiology, University of Agriculture Faisalabad, Faisalabad, 38000, Pakistan
| | - Sarfaraz Hussain
- School of Agricultural Engineering and Food Science, Shandong University of Technology, Zibo, 255049, Shandong, People's Republic of China.
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences, Beijing, 100193, People's Republic of China.
| |
Collapse
|
18
|
Khosravi A, Chen Q, Echterhof A, Koff JL, Bollyky PL. Phage Therapy for Respiratory Infections: Opportunities and Challenges. Lung 2024; 202:223-232. [PMID: 38772946 DOI: 10.1007/s00408-024-00700-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/13/2024] [Indexed: 05/23/2024]
Abstract
We are entering the post-antibiotic era. Antimicrobial resistance (AMR) is a critical problem in chronic lung infections resulting in progressive respiratory failure and increased mortality. In the absence of emerging novel antibiotics to counter AMR infections, bacteriophages (phages), viruses that infect bacteria, have become a promising option for chronic respiratory infections. However, while personalized phage therapy is associated with improved outcomes in individual cases, clinical trials demonstrating treatment efficacy are lacking, limiting the therapeutic potential of this approach for respiratory infections. In this review, we address the current state of phage therapy for managing chronic respiratory diseases. We then discuss how phage therapy may address major microbiologic obstacles which hinder disease resolution of chronic lung infections with current antibiotic-based treatment practices. Finally, we highlight the challenges that must be addressed for successful phage therapy clinical trials. Through this discussion, we hope to expand on the potential of phages as an adjuvant therapy in chronic lung infections, as well as the microbiologic challenges that need to be addressed for phage therapy to expand beyond personalized salvage therapy.
Collapse
Affiliation(s)
- Arya Khosravi
- Division of Infectious Diseases, School of Medicine, Stanford University, Stanford, CA, USA.
- Division of Infectious Diseases, Department of Medicine, Stanford University, 279 Campus Drive, Beckman Center, Room B237, Stanford, CA, 94305, USA.
| | - Qingquan Chen
- Division of Infectious Diseases, School of Medicine, Stanford University, Stanford, CA, USA
| | - Arne Echterhof
- Division of Infectious Diseases, School of Medicine, Stanford University, Stanford, CA, USA
| | - Jonathan L Koff
- Section of Pulmonary, Critical Care & Sleep Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Paul L Bollyky
- Division of Infectious Diseases, School of Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
19
|
Zhen J, Liu R, Man C, Xu S, Zhang W, Zou L, Liu W, Ni HB, Zou M, He T, Wang R, Zhang XX, Zhang C. Bacteriophage LHE83 targeting OmpA as a receptor exhibited synergism with spectinomycin against Escherichia coli. Poult Sci 2024; 103:103643. [PMID: 38537406 PMCID: PMC10987938 DOI: 10.1016/j.psj.2024.103643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 04/07/2024] Open
Abstract
Understanding the characteristics of bacteriophages is crucial for the optimization of phage therapy. In this study, the biological and genomic characteristics of coliphage LHE83 were determined and its synergistic effects with different types of antibiotics against E. coli E82 were investigated. Phage LHE83 displayed a contractile tail morphology and had a titer of 3.02 × 109 pfu/mL at an optimal MOI of 0.01. Meanwhile, phage LHE83 exhibited good physical and chemical factors tolerance. The 1-step growth analysis revealed a latent period of approx. 10 min with a burst size of 87 pfu/infected cell. Phage LHE83 belongs to the genus Dhakavirus. Its genome consists of 170,464 bp with a 40% GC content, and a total of 268 Open Reading Frames (ORF) were predicted with no detected virulent or resistant genes. ORF 213 was predicted to encode the receptor binding protein (RBP) and confirmed by the antibody-blocking assay. Furthermore, a phage-resistant strain E. coli E82R was generated by co-culturing phage LHE83 with E. coli E82. Genomic analysis revealed that OmpA served as the receptor for phage LHE83, which was further confirmed by phage adsorption assay using E. coli BL21ΔOmpA, E. coli BL21ΔOmpA: OmpA and E. coli BL21:OmpA strains. Additionally, a synergistic effect was observed between phage LHE83 and spectinomycin against the drug-resistant strain E. coli E82. These results provide a theoretical basis for understanding the interactions between phages, antibiotics, and host bacteria, which can assist in the clinical application of phages and antibiotics against drug-resistant bacteria.
Collapse
Affiliation(s)
- Jianyu Zhen
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Rui Liu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Cheng Man
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Shijie Xu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Wenxiu Zhang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Ling Zou
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Wenhua Liu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Hong-Bo Ni
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Ming Zou
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Tao He
- Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Ran Wang
- Institute of Food Safety and Nutrition, Jiangsu Academy of Agricultural Sciences, Nanjing, China
| | - Xiao-Xuan Zhang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China
| | - Can Zhang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, China.
| |
Collapse
|
20
|
Alves D, Pereira MO, Lopes SP. Co-immobilization of Ciprofloxacin and Chlorhexidine as a Broad-Spectrum Antimicrobial Dual-Drug Coating for Poly(vinyl chloride) (PVC)-Based Endotracheal Tubes. ACS APPLIED MATERIALS & INTERFACES 2024; 16:16861-16879. [PMID: 38507790 PMCID: PMC10995906 DOI: 10.1021/acsami.4c01334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 02/23/2024] [Accepted: 03/05/2024] [Indexed: 03/22/2024]
Abstract
The endotracheal tube (ETT) affords support for intubated patients, but the increasing incidence of ventilator-associated pneumonia (VAP) is jeopardizing its application. ETT surfaces promote (poly)microbial colonization and biofilm formation, with a heavy burden for VAP. Devising safe, broad-spectrum antimicrobial materials to tackle the ETT bioburden is needful. Herein, we immobilized ciprofloxacin (CIP) and/or chlorhexidine (CHX), through polydopamine (pDA)-based functionalization, onto poly(vinyl chloride) (PVC) surfaces. These surfaces were characterized regarding physicochemical properties and challenged with single and polymicrobial cultures of VAP-relevant bacteria (Pseudomonas aeruginosa, Acinetobacter baumannii, Klebsiella pneumoniae, Staphylococcus aureus, Staphylococcus epidermidis) and fungi (Candida albicans). The coatings imparted PVC surfaces with a homogeneous morphology, varied wettability, and low roughness. The antimicrobial immobilization via pDA chemistry was still evidenced by infrared spectroscopy. Coated surfaces exhibited sustained CIP/CHX release, retaining prolonged (10 days) activity. CIP/CHX-coated surfaces evidencing no A549 lung cell toxicity displayed better antibiofilm outcomes than CIP or CHX coatings, preventing bacterial attachment by 4.1-7.2 Log10 CFU/mL and modestly distressingC. albicans. Their antibiofilm effectiveness was endured toward polymicrobial consortia, substantially inhibiting the adhesion of the bacterial populations (up to 8 Log10 CFU/mL) within the consortia in dual- and even inP. aeruginosa/S. aureus/C. albicans triple-species biofilms while affecting fungal adhesion by 2.7 Log10 CFU/mL (dual consortia) and 1 Log10 CFU/mL (triple consortia). The potential of the dual-drug coating strategy in preventing triple-species adhesion and impairing bacterial viability was still strengthened by live/dead microscopy. The pDA-assisted CIP/CHX co-immobilization holds a safe and robust broad-spectrum antimicrobial coating strategy for PVC-ETTs, with the promise laying in reducing VAP incidence.
Collapse
Affiliation(s)
- Diana
Filipa Alves
- CEB
- Centre of Biological Engineering, University
of Minho, 4710-057 Braga, Portugal
- LABBELS—Associate
Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Maria Olívia Pereira
- CEB
- Centre of Biological Engineering, University
of Minho, 4710-057 Braga, Portugal
- LABBELS—Associate
Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Susana Patrícia Lopes
- CEB
- Centre of Biological Engineering, University
of Minho, 4710-057 Braga, Portugal
- LABBELS—Associate
Laboratory, 4710-057 Braga/Guimarães, Portugal
| |
Collapse
|
21
|
Lu H, Ni SQ. Review on sterilization techniques, and the application potential of phage lyase and lyase immobilization in fighting drug-resistant bacteria. J Mater Chem B 2024; 12:3317-3335. [PMID: 38380677 DOI: 10.1039/d3tb02366d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Many human health problems and property losses caused by pathogenic contamination cannot be underestimated. Bactericidal techniques have been extensively studied to address this issue of public health and economy. Bacterial resistance develops as a result of the extensive use of single or multiple but persistent usage of sterilizing drugs, and the emergence of super-resistant bacteria brings new challenges. Therefore, it is crucial to control pathogen contamination by applying innovative and effective sterilization techniques. As organisms that exist in nature and can specifically kill bacteria, phages have become the focus as an alternative to antibacterial agents. Furthermore, phage-encoded lyases are proteins that play important roles in phage sterilization. The in vitro sterilization of phage lyase has been developed as a novel biosterilization technique to reduce bacterial resistance and is more environmentally friendly than conventional sterilization treatments. For the shortcomings of enzyme applications, this review discusses the enzyme immobilization methods and the application potential of immobilized lyases for sterilization. Although some techniques provide effective solutions, immobilized lyase sterilization technology has been proven to be a more effective innovation for efficient pathogen killing and reducing bacterial resistance. We hope that this review can provide new insights for the development of sterilization techniques.
Collapse
Affiliation(s)
- Han Lu
- School of Environmental Science and Engineering, Shandong University, Qingdao, Shandong 266237, China.
| | - Shou-Qing Ni
- School of Environmental Science and Engineering, Shandong University, Qingdao, Shandong 266237, China.
| |
Collapse
|
22
|
Vaezi A, Healy T, Ebrahimi G, Rezvankhah S, Hashemi Shahraki A, Mirsaeidi M. Phage therapy: breathing new tactics into lower respiratory tract infection treatments. Eur Respir Rev 2024; 33:240029. [PMID: 38925791 PMCID: PMC11216685 DOI: 10.1183/16000617.0029-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 04/16/2024] [Indexed: 06/28/2024] Open
Abstract
Lower respiratory tract infections (LRTIs) present a significant global health burden, exacerbated by the rise in antimicrobial resistance (AMR). The persistence and evolution of multidrug-resistant bacteria intensifies the urgency for alternative treatments. This review explores bacteriophage (phage) therapy as an innovative solution to combat bacterial LRTIs. Phages, abundant in nature, demonstrate specificity towards bacteria, minimal eukaryotic toxicity, and the ability to penetrate and disrupt bacterial biofilms, offering a targeted approach to infection control. The article synthesises evidence from systematic literature reviews spanning 2000-2023, in vitro and in vivo studies, case reports and ongoing clinical trials. It highlights the synergistic potential of phage therapy with antibiotics, the immunophage synergy in animal models, and the pharmacodynamics and pharmacokinetics critical for clinical application. Despite promising results, the article acknowledges that phage therapy is at a nascent stage in clinical settings, the challenges of phage-resistant bacteria, and the lack of comprehensive cost-effectiveness studies. It stresses the need for further research to optimise phage therapy protocols and navigate the complexities of phage-host interactions, particularly in vulnerable populations such as the elderly and immunocompromised. We call for regulatory adjustments to facilitate the exploration of the long-term effects of phage therapy, aiming to incorporate this old-yet-new therapy into mainstream clinical practice to tackle the looming AMR crisis.
Collapse
Affiliation(s)
- Atefeh Vaezi
- Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine-Jacksonville, University of Florida, Jacksonville, FL, USA
| | - Thomas Healy
- Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine-Jacksonville, University of Florida, Jacksonville, FL, USA
| | | | | | - Abdolrazagh Hashemi Shahraki
- Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine-Jacksonville, University of Florida, Jacksonville, FL, USA
| | - Mehdi Mirsaeidi
- Division of Pulmonary, Critical Care and Sleep Medicine, College of Medicine-Jacksonville, University of Florida, Jacksonville, FL, USA
| |
Collapse
|
23
|
Hong Q, Chang RYK, Assafiri O, Morales S, Chan HK. Optimizing in vitro phage-ciprofloxacin combination formulation for respiratory therapy of multi-drug resistant Pseudomonas aeruginosa infections. Int J Pharm 2024; 652:123853. [PMID: 38280500 DOI: 10.1016/j.ijpharm.2024.123853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 01/21/2024] [Accepted: 01/23/2024] [Indexed: 01/29/2024]
Abstract
Respiratory infection caused by multi-drug resistant (MDR) Pseudomonas aeruginosa is challenging to treat. In this study, we investigate the optimal dose of anti-pseudomonas phage PEV31 (103, 105, and 108 PFU/mL) combined with ciprofloxacin (ranging from 1/8× MIC to 8× MIC) to treat the MDR P. aeruginosa strain FADD1-PA001 using time-kill studies. We determined the impact of phage growth kinetics in the presence of ciprofloxacin through one-step growth analysis. Single treatments with either phage PEV31 or ciprofloxacin (except at 8× MIC) showed limited bactericidal efficiency, with bacterial regrowth observed at 48 h. The most effective treatments were PEV31 at multiplicity of infection (MOI) of 0.1 and 100 combined with ciprofloxacin at concentrations above 1× MIC, resulting in a >4 log10 reduction in bacterial counts. While the burst size of phage PEV31 was decreased with increasing ciprofloxacin concentration, robust antimicrobial effects were still maintained in the combination treatment. Aerosol samples collected from vibrating mesh nebulization of the combination formulation at phage MOI of 100 with 2× MIC effectively inhibited bacterial density. In summary, our combination treatments eradicated in vitro bacterial growth and sustained antimicrobial effects for 48 h. These results indicated the potential application of nebulization-based strategies for the combination treatment against MDR lung infections.
Collapse
Affiliation(s)
- Qixuan Hong
- Advanced Drug Delivery Group, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Rachel Yoon Kyung Chang
- Advanced Drug Delivery Group, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Omar Assafiri
- Advanced Drug Delivery Group, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | | | - Hak-Kim Chan
- Advanced Drug Delivery Group, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
24
|
Ipoutcha T, Racharaks R, Huttelmaier S, Wilson CJ, Ozer EA, Hartmann EM. A synthetic biology approach to assemble and reboot clinically relevant Pseudomonas aeruginosa tailed phages. Microbiol Spectr 2024; 12:e0289723. [PMID: 38294230 PMCID: PMC10913387 DOI: 10.1128/spectrum.02897-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 12/17/2023] [Indexed: 02/01/2024] Open
Abstract
The rise in the frequency of antibiotic resistance has made bacterial infections, specifically Pseudomonas aeruginosa, a cause for greater concern. Phage therapy is a promising solution that uses naturally isolated phages to treat bacterial infections. Ecological limitations, which stipulate a discrete host range and the inevitable evolution of resistance, may be overcome through a better understanding of phage biology and the utilization of engineered phages. In this study, we developed a synthetic biology approach to construct tailed phages that naturally target clinically relevant strains of Pseudomonas aeruginosa. As proof of concept, we successfully cloned and assembled the JG024 and DMS3 phage genomes in yeast using transformation-associated recombination cloning and rebooted these two phage genomes in two different strains of P. aeruginosa. We identified factors that affected phage reboot efficiency like the phage species or the presence of antiviral defense systems in the bacterial strain. We have successfully extended this method to two other phage species and observed that the method enables the reboot of phages that are naturally unable to infect the strain used for reboot. This research represents a critical step toward the construction of clinically relevant, engineered P. aeruginosa phages.IMPORTANCEPseudomonas aeruginosa is a bacterium responsible for severe infections and a common major complication in cystic fibrosis. The use of antibiotics to treat bacterial infections has become increasingly difficult as antibiotic resistance has become more prevalent. Phage therapy is an alternative solution that is already being used in some European countries, but its use is limited by the narrow host range due to the phage receptor specificity, the presence of antiviral defense systems in the bacterial strain, and the possible emergence of phage resistance. In this study, we demonstrate the use of a synthetic biology approach to construct and reboot clinically relevant P. aeruginosa tailed phages. This method enables a significant expansion of possibilities through the construction of engineered phages for therapy applications.
Collapse
Affiliation(s)
- Thomas Ipoutcha
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, Illinois, USA
| | - Ratanachat Racharaks
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, Illinois, USA
| | - Stefanie Huttelmaier
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, Illinois, USA
| | - Cole J. Wilson
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, Illinois, USA
| | - Egon A. Ozer
- Division of Infectious Diseases, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Erica M. Hartmann
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, Illinois, USA
- Center for Synthetic Biology, Northwestern University, Evanston, Illinois, USA
| |
Collapse
|
25
|
Rodriguez-Gonzalez RA, Balacheff Q, Debarbieux L, Marchi J, Weitz JS. Metapopulation model of phage therapy of an acute Pseudomonas aeruginosa lung infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.31.578251. [PMID: 38352502 PMCID: PMC10862780 DOI: 10.1101/2024.01.31.578251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
Infections caused by multi-drug resistant (MDR) pathogenic bacteria are a global health threat. Phage therapy, which uses phage to kill bacterial pathogens, is increasingly used to treat patients infected by MDR bacteria. However, the therapeutic outcome of phage therapy may be limited by the emergence of phage resistance during treatment and/or by physical constraints that impede phage-bacteria interactions in vivo. In this work, we evaluate the role of lung spatial structure on the efficacy of phage therapy for Pseudomonas aeruginosa infection. To do so, we developed a spatially structured metapopulation network model based on the geometry of the bronchial tree, and included the emergence of phage-resistant bacterial mutants and host innate immune responses. We model the ecological interactions between bacteria, phage, and the host innate immune system at the airway (node) level. The model predicts the synergistic elimination of a P. aeruginosa infection due to the combined effects of phage and neutrophils given sufficiently active immune states and suitable phage life history traits. Moreover, the metapopulation model simulations predict that local MDR pathogens are cleared faster at distal nodes of the bronchial tree. Notably, image analysis of lung tissue time series from wild-type and lymphocyte-depleted mice (n=13) revealed a concordant, statistically significant pattern: infection intensity cleared in the bottom before the top of the lungs. Overall, the combined use of simulations and image analysis of in vivo experiments further supports the use of phage therapy for treating acute lung infections caused by P. aeruginosa while highlighting potential limits to therapy given a spatially structured environment, such as impaired innate immune responses and low phage efficacy.
Collapse
Affiliation(s)
- Rogelio A. Rodriguez-Gonzalez
- Interdisciplinary Graduate Program in Quantitative Biosciences, Georgia Institute of Technology, Atlanta, Georgia, USA
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, Georgia, USA
| | - Quentin Balacheff
- CHU Felix Guyon, Service des maladies respiratoires, La Réunion, France
| | | | - Jacopo Marchi
- Department of Biology, University of Maryland, College Park, Maryland, USA
| | - Joshua S. Weitz
- Department of Biology, University of Maryland, College Park, Maryland, USA
- Department of Physics, University of Maryland, College Park, Maryland, USA
- Institut de Biologie de l’École Normale Supérieure, Paris, France
| |
Collapse
|
26
|
Gordon M, Ramirez P. Efficacy and Experience of Bacteriophages in Biofilm-Related Infections. Antibiotics (Basel) 2024; 13:125. [PMID: 38391511 PMCID: PMC10886175 DOI: 10.3390/antibiotics13020125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 01/16/2024] [Accepted: 01/19/2024] [Indexed: 02/24/2024] Open
Abstract
Bacterial infection has always accompanied human beings, causing suffering and death while also contributing to the advancement of medical science. However, the treatment of infections has become more complex in recent times. The increasing resistance of bacterial strains to antibiotics has diminished the effectiveness of the therapeutic arsenal, making it less likely to find the appropriate empiric antibiotic option. Additionally, the development and persistence of bacterial biofilms have become more prevalent, attributed to the greater use of invasive devices that facilitate biofilm formation and the enhanced survival of chronic infection models where biofilm plays a crucial role. Bacteria within biofilms are less susceptible to antibiotics due to physical, chemical, and genetic factors. Bacteriophages, as biological weapons, can overcome both antimicrobial resistance and biofilm protection. In this review, we will analyze the scientific progress achieved in vitro to justify their clinical application. In the absence of scientific evidence, we will compile publications of clinical cases where phages have been used to treat infections related to biofilm. The scientific basis obtained in vitro and the success rate and safety observed in clinical practice should motivate the medical community to conduct clinical trials establishing a protocol for the proper use of bacteriophages.
Collapse
Affiliation(s)
- Monica Gordon
- Critical Care Department, Hospital Universitario y Politécnico la Fe, Av. Vicente Abril Martorell 106, 46026 Valencia, Spain
| | - Paula Ramirez
- Critical Care Department, Hospital Universitario y Politécnico la Fe, Av. Vicente Abril Martorell 106, 46026 Valencia, Spain
| |
Collapse
|
27
|
De Soir S, Parée H, Kamarudin NHN, Wagemans J, Lavigne R, Braem A, Merabishvili M, De Vos D, Pirnay JP, Van Bambeke F. Exploiting phage-antibiotic synergies to disrupt Pseudomonas aeruginosa PAO1 biofilms in the context of orthopedic infections. Microbiol Spectr 2024; 12:e0321923. [PMID: 38084971 PMCID: PMC10783084 DOI: 10.1128/spectrum.03219-23] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/20/2023] [Indexed: 01/13/2024] Open
Abstract
IMPORTANCE Biofilm-related infections are among the most difficult-to-treat infections in all fields of medicine due to their antibiotic tolerance and persistent character. In the field of orthopedics, these biofilms often lead to therapeutic failure of medical implantable devices and urgently need novel treatment strategies. This forthcoming article aims to explore the dynamic interplay between newly isolated bacteriophages and routinely used antibiotics and clearly indicates synergetic patterns when used as a dual treatment modality. Biofilms were drastically more reduced when both active agents were combined, thereby providing additional evidence that phage-antibiotic combinations lead to synergism and could potentially improve clinical outcome for affected patients.
Collapse
Affiliation(s)
- Steven De Soir
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
- Laboratory for Molecular and Cellular Technology (LabMCT), Queen Astrid Military Hospital, Neder-over-Heembeek, Belgium
| | - Hortence Parée
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Nur Hidayatul Nazirah Kamarudin
- Department of Materials Engineering, Biomaterials and Tissue Engineering Research Group, KU Leuven, Leuven, Belgium
- Department of Chemical and Process Engineering, Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | | | - Rob Lavigne
- Laboratory of Gene Technology, KU Leuven, Leuven, Belgium
| | - Annabel Braem
- Department of Materials Engineering, Biomaterials and Tissue Engineering Research Group, KU Leuven, Leuven, Belgium
| | - Maya Merabishvili
- Laboratory for Molecular and Cellular Technology (LabMCT), Queen Astrid Military Hospital, Neder-over-Heembeek, Belgium
| | - Daniel De Vos
- Laboratory for Molecular and Cellular Technology (LabMCT), Queen Astrid Military Hospital, Neder-over-Heembeek, Belgium
| | - Jean-Paul Pirnay
- Laboratory for Molecular and Cellular Technology (LabMCT), Queen Astrid Military Hospital, Neder-over-Heembeek, Belgium
| | - Françoise Van Bambeke
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
28
|
Gliźniewicz M, Miłek D, Olszewska P, Czajkowski A, Serwin N, Cecerska-Heryć E, Dołęgowska B, Grygorcewicz B. Advances in bacteriophage-mediated strategies for combating polymicrobial biofilms. Front Microbiol 2024; 14:1320345. [PMID: 38249486 PMCID: PMC10797108 DOI: 10.3389/fmicb.2023.1320345] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/04/2023] [Indexed: 01/23/2024] Open
Abstract
Bacteria and fungi tend to coexist within biofilms instead of in planktonic states. Usually, such communities include cross-kingdom microorganisms, which make them harder to remove from abiotic surfaces or infection sites. Additionally, the produced biofilm matrix protects embedded microorganisms from antibiotics, disinfectants, or the host immune system. Therefore, classic therapies based on antibiotics might be ineffective, especially when multidrug-resistant bacteria are causative factors. The complexities surrounding the eradication of biofilms from diverse surfaces and the human body have spurred the exploration of alternative therapeutic modalities. Among these options, bacteriophages and their enzymatic counterparts have emerged as promising candidates, either employed independently or in synergy with antibiotics and other agents. Phages are natural bacteria killers because of mechanisms of action that differ from antibiotics, phages might answer worldwide problems with bacterial infections. In this review, we report the attempts to use bacteriophages in combating polymicrobial biofilms in in vitro studies, using different models, including the therapeutical use of phages. In addition, we sum up the advantages, disadvantages, and perspectives of phage therapy.
Collapse
Affiliation(s)
- Marta Gliźniewicz
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Dominika Miłek
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Patrycja Olszewska
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Artur Czajkowski
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Natalia Serwin
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Elżbieta Cecerska-Heryć
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Barbara Dołęgowska
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Bartłomiej Grygorcewicz
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
- Department of Chemical Technology and Engineering, Institute of Chemical Engineering and Environmental Protection Processes, West Pomeranian University of Technology, Szczecin, Poland
| |
Collapse
|
29
|
Kulshrestha M, Tiwari M, Tiwari V. Bacteriophage therapy against ESKAPE bacterial pathogens: Current status, strategies, challenges, and future scope. Microb Pathog 2024; 186:106467. [PMID: 38036110 DOI: 10.1016/j.micpath.2023.106467] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 10/19/2023] [Accepted: 11/24/2023] [Indexed: 12/02/2023]
Abstract
The ESKAPE pathogens are the primary threat due to their constant spread of drug resistance worldwide. These pathogens are also regarded as opportunistic pathogens and could potentially cause nosocomial infections. Most of the ESKAPE pathogens have developed resistance to almost all the antibiotics that are used against them. Therefore, to deal with antimicrobial resistance, there is an urgent requirement for alternative non-antibiotic strategies to combat this rising issue of drug-resistant organisms. One of the promising alternatives to this scenario is implementing bacteriophage therapy. This under-explored mode of treatment in modern medicine has posed several concerns, such as preferable phages for the treatment, impact on the microbiome (or gut microflora), dose optimisation, safety, etc. The review will cover a rationale for phage therapy, clinical challenges, and propose phage therapy as an effective therapeutic against bacterial coinfections during pandemics. This review also addresses the expected uncertainties for administering the phage as a treatment against the ESKAPE pathogens and the advantages of using lytic phage over temperate, the immune response to phages, and phages in combinational therapies. The interaction between bacteria and bacteriophages in humans and countless animal models can also be used to design novel and futuristic therapeutics like personalised medicine or bacteriophages as anti-biofilm agents. Hence, this review explores different aspects of phage therapy and its potential to emerge as a frontline therapy against the ESKAPE bacterial pathogen.
Collapse
Affiliation(s)
- Mukta Kulshrestha
- Department of Biochemistry, Central University of Rajasthan, Ajmer, 305817, India
| | - Monalisa Tiwari
- Department of Biochemistry, Central University of Rajasthan, Ajmer, 305817, India
| | - Vishvanath Tiwari
- Department of Biochemistry, Central University of Rajasthan, Ajmer, 305817, India.
| |
Collapse
|
30
|
Akturk E, Melo LD, Oliveira H, Crabbé A, Coenye T, Azeredo J. Combining phages and antibiotic to enhance antibiofilm efficacy against an in vitro dual species wound biofilm. Biofilm 2023; 6:100147. [PMID: 37662851 PMCID: PMC10474582 DOI: 10.1016/j.bioflm.2023.100147] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 07/31/2023] [Accepted: 08/01/2023] [Indexed: 09/05/2023] Open
Abstract
Chronic wound management is extremely challenging because of the persistence of biofilm-forming pathogens, such as Pseudomonas aeruginosa and Staphylococcus aureus, which are the prevailing bacterial species that co-infect chronic wounds. Phage therapy has gained an increased interest to treat biofilm-associated infections, namely when combined with antibiotics. Here, we tested the effect of gentamicin as a co-adjuvant of phages in a dual species-biofilm wound model formed on artificial dermis. The biofilm-killing capacity of the tested treatments was significantly increased when phages were combined with gentamicin and applied multiple times as multiple dose (three doses, every 8 h). Our results suggest that gentamycin is an effective adjuvant of phage therapy particularly when applied simultaneously with phages and in three consecutive doses. The multiple and simultaneous dose treatment seems to be essential to avoid bacterial resistance development to each of the antimicrobial agents.
Collapse
Affiliation(s)
- Ergun Akturk
- CEB - Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
- LABBELS – Associate Laboratory, Braga, Guimarães, Portugal
| | - Luís D.R. Melo
- CEB - Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
- LABBELS – Associate Laboratory, Braga, Guimarães, Portugal
- ESCMID Study Group for Biofilms (ESGB), Switzerland
| | - Hugo Oliveira
- CEB - Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
- LABBELS – Associate Laboratory, Braga, Guimarães, Portugal
- ESCMID Study Group for Biofilms (ESGB), Switzerland
| | - Aurélie Crabbé
- Laboratory of Pharmaceutical Microbiology (LPM), Ghent University, Ghent, Belgium
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology (LPM), Ghent University, Ghent, Belgium
- ESCMID Study Group for Biofilms (ESGB), Switzerland
| | - Joana Azeredo
- CEB - Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, Campus de Gualtar, 4710-057, Braga, Portugal
- LABBELS – Associate Laboratory, Braga, Guimarães, Portugal
- ESCMID Study Group for Biofilms (ESGB), Switzerland
| |
Collapse
|
31
|
Chen B, Benavente LP, Chittò M, Wychowaniec JK, Post V, D'Este M, Constant C, Zeiter S, Feng W, Moreno MG, Trampuz A, Wagemans J, Onsea J, Richards RG, Lavigne R, Moriarty TF, Metsemakers WJ. Alginate microbeads and hydrogels delivering meropenem and bacteriophages to treat Pseudomonas aeruginosa fracture-related infections. J Control Release 2023; 364:159-173. [PMID: 37866403 DOI: 10.1016/j.jconrel.2023.10.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/18/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023]
Abstract
Bacteriophage (phage) therapy has shown promise in treating fracture-related infection (FRI); however, questions remain regarding phage efficacy against biofilms, phage-antibiotic interaction, administration routes and dosing, and the development of phage resistance. The goal of this study was to develop a dual antibiotic-phage delivery system containing hydrogel and alginate microbeads loaded with a phage cocktail plus meropenem and evaluate efficacy against muti-drug resistant Pseudomonas aeruginosa. Two phages (FJK.R9-30 and MK.R3-15) displayed enhanced antibiotic activity against P. aeruginosa biofilms when tested in combination with meropenem. The antimicrobial activity of both antibiotic and phage was retained for eight days at 37 °C in dual phage and antibiotic loaded hydrogel with microbeads (PA-HM). In a mouse FRI model, phages were recovered from all tissues within all treatment groups receiving dual PA-HM. Moreover, animals that received the dual PA-HM either with or without systemic antibiotics had less incidence of phage resistance and less serum neutralization compared to phages in saline. The dual PA-HM could reduce bacterial load in soft tissue when combined with systemic antibiotics, although the infection was not eradicated. The use of alginate microbeads and injectable hydrogel for controlled release of phages and antibiotics, leads to the reduced development of phage resistance and lower exposure to the adaptive immune system, which highlights the translational potential of the dual PA-HM. However, further optimization of phage therapy and its delivery system is necessary to achieve higher bacterial killing activity in vivo in the future.
Collapse
Affiliation(s)
- Baixing Chen
- Department of Trauma Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Development and Regeneration, KU Leuven, Leuven, Belgium; AO Research Institute Davos, Davos, Switzerland
| | - Luis Ponce Benavente
- Center for Musculoskeletal Surgery Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | | | | | | | | | | | | | - Wenli Feng
- AO Research Institute Davos, Davos, Switzerland
| | - Mercedes González Moreno
- Center for Musculoskeletal Surgery Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Andrej Trampuz
- Center for Musculoskeletal Surgery Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | | | - Jolien Onsea
- Department of Trauma Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | | | - Rob Lavigne
- Laboratory of Gene Technology, KU Leuven, Leuven, Belgium
| | | | - Willem-Jan Metsemakers
- Department of Trauma Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
32
|
Holger DJ, El Ghali A, Bhutani N, Lev KL, Stamper K, Kebriaei R, Kunz Coyne AJ, Morrisette T, Shah R, Alexander J, Lehman SM, Rojas LJ, Marshall SH, Bonomo RA, Rybak MJ. Phage-antibiotic combinations against multidrug-resistant Pseudomonas aeruginosa in in vitro static and dynamic biofilm models. Antimicrob Agents Chemother 2023; 67:e0057823. [PMID: 37855639 PMCID: PMC10648846 DOI: 10.1128/aac.00578-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 07/18/2023] [Indexed: 10/20/2023] Open
Abstract
Biofilm-producing Pseudomonas aeruginosa infections pose a severe threat to public health and are responsible for high morbidity and mortality. Phage-antibiotic combinations (PACs) are a promising strategy for combatting multidrug-resistant (MDR), extensively drug-resistant (XDR), and difficult-to-treat P. aeruginosa infections. Ten MDR/XDR P. aeruginosa strains and five P. aeruginosa-specific phages were genetically characterized and evaluated based upon their antibiotic susceptibilities and phage sensitivities. Two selected strains, AR351 (XDR) and I0003-1 (MDR), were treated singly and in combination with either a broad-spectrum or narrow-spectrum phage, phage EM-T3762627-2_AH (EM), or 14207, respectively, and bactericidal antibiotics of five classes in biofilm time-kill analyses. Synergy and/or bactericidal activity was demonstrated with all PACs against one or both drug-resistant P. aeruginosa strains (average reduction: -Δ3.32 log10 CFU/cm2). Slightly improved ciprofloxacin susceptibility was observed in both strains after exposure to phages (EM and 14207) in combination with ciprofloxacin and colistin. Based on phage cocktail optimization with four phages (EM, 14207, E20050-C (EC), and 109), we identified several effective phage-antibiotic cocktails for further analysis in a 4-day pharmacokinetic/pharmacodynamic in vitro biofilm model. Three-phage cocktail, EM + EC + 109, in combination with ciprofloxacin demonstrated the greatest biofilm reduction against AR351 (-Δ4.70 log10 CFU/cm2 from baseline). Of remarkable interest, the addition of phage 109 prevented phage resistance development to EM and EC in the biofilm model. PACs can demonstrate synergy and offer enhanced eradication of biofilm against drug-resistant P. aeruginosa while preventing the emergence of resistance.
Collapse
Affiliation(s)
- Dana J. Holger
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Amer El Ghali
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Natasha Bhutani
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Katherine L. Lev
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Kyle Stamper
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Razieh Kebriaei
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Ashlan J. Kunz Coyne
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Taylor Morrisette
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Rahi Shah
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Jose Alexander
- Department of Microbiology, Virology, and Immunology, AdventHealth Central Florida, Orlando, Florida, USA
| | - Susan M. Lehman
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, Maryland, USA
| | - Laura J. Rojas
- Department of Molecular Biology and Microbiology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Center for Antimicrobial Resistance and Epidemiology, Veterans Affairs Medical Center, Case Western Reserve University, Cleveland, Ohio, USA
- Research Service, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio, USA
| | - Steven H. Marshall
- Research Service, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio, USA
| | - Robert A. Bonomo
- Department of Molecular Biology and Microbiology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Center for Antimicrobial Resistance and Epidemiology, Veterans Affairs Medical Center, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Michael J. Rybak
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
- Department of Pharmacy Services, Detroit Receiving Hospital, Detroit Medical Center, Detroit, Michigan, USA
- Department of Medicine, Division of Infectious Diseases, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
33
|
Fajardo-Lubian A, Venturini C. Use of Bacteriophages to Target Intracellular Pathogens. Clin Infect Dis 2023; 77:S423-S432. [PMID: 37932114 DOI: 10.1093/cid/ciad515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023] Open
Abstract
Bacteriophages (phages) have shown great potential as natural antimicrobials against extracellular pathogens (eg, Escherichia coli or Klebsiella pneumoniae), but little is known about how they interact with intracellular targets (eg, Shigella spp., Salmonella spp., Mycobacterium spp.) in the mammalian host. Recent research has demonstrated that phages can enter human cells. However, for the design of successful clinical applications, further investigation is required to define their subcellular behavior and to understand the complex biological processes that underlie the interaction with their bacterial targets. In this review, we summarize the molecular evidence of phage internalization in eucaryotic cells, with specific focus on proof of phage activity against their bacterial targets within the eucaryotic host, and the current proposed strategies to overcome poor penetrance issues that may impact therapeutic use against the most clinically relevant intracellular pathogens.
Collapse
Affiliation(s)
- Alicia Fajardo-Lubian
- Faculty of Medicine and Health, Sydney ID Institute, University of Sydney, Sydney, New South Wales, Australia
- Centre for Infectious Diseases and Microbiology, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
| | - Carola Venturini
- Centre for Infectious Diseases and Microbiology, Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Faculty of Science, Sydney School of Veterinary Science, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
34
|
Mukhopadhyay S, Zhang P, To KKW, Liu Y, Bai C, Leung SSY. Sequential treatment effects on phage-antibiotic synergistic application against multi-drug-resistant Acinetobacter baumannii. Int J Antimicrob Agents 2023; 62:106951. [PMID: 37574030 DOI: 10.1016/j.ijantimicag.2023.106951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 07/05/2023] [Accepted: 08/05/2023] [Indexed: 08/15/2023]
Abstract
Bacteriophage (phage) therapy, exploiting phages which are the natural enemies of bacteria, has been re-introduced to treat multidrug-resistant (MDR) bacterial infections. However, some intrinsic drawbacks of phages are overshadowing their clinical use, particularly the narrow host spectrum and rapid emergence of resistance upon treatment. The use of phage-antibiotic combinations exhibiting synergistic bacterial killing [termed 'phage-antibiotic synergy' (PAS)] has therefore been proposed. It is well reported that the types and doses of phages and antibiotics are critical in achieving PAS. However, the impact of treatment order has received less research attention. As such, this study used an Acinetobacter baumannii phage vB_AbaM-IME-AB2 and colistin as a model PAS combination to elucidate the order effects in-vitro. While application of the phage 8 h before colistin treatment demonstrated the greatest antibacterial synergy, it failed to prevent the development of phage resistance. On the other hand, simultaneous application and antibiotic followed by phage application were able to suppress/delay the development of resistance effectively, and simultaneous application demonstrated superior antibacterial and antibiofilm activities. Further in-vivo investigation is required to confirm the impact of treatment order on PAS.
Collapse
Affiliation(s)
- Subhankar Mukhopadhyay
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region
| | - Pengfei Zhang
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region
| | - Kenneth K W To
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region
| | - Yannan Liu
- Emergency Medicine Clinical Research Centre, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Changqing Bai
- Department of Respiratory, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Guangdong, China
| | - Sharon S Y Leung
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region.
| |
Collapse
|
35
|
Xiao G, Li J, Sun Z. The Combination of Antibiotic and Non-Antibiotic Compounds Improves Antibiotic Efficacy against Multidrug-Resistant Bacteria. Int J Mol Sci 2023; 24:15493. [PMID: 37895172 PMCID: PMC10607837 DOI: 10.3390/ijms242015493] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 10/29/2023] Open
Abstract
Bacterial antibiotic resistance, especially the emergence of multidrug-resistant (MDR) strains, urgently requires the development of effective treatment strategies. It is always of interest to delve into the mechanisms of resistance to current antibiotics and target them to promote the efficacy of existing antibiotics. In recent years, non-antibiotic compounds have played an important auxiliary role in improving the efficacy of antibiotics and promoting the treatment of drug-resistant bacteria. The combination of non-antibiotic compounds with antibiotics is considered a promising strategy against MDR bacteria. In this review, we first briefly summarize the main resistance mechanisms of current antibiotics. In addition, we propose several strategies to enhance antibiotic action based on resistance mechanisms. Then, the research progress of non-antibiotic compounds that can promote antibiotic-resistant bacteria through different mechanisms in recent years is also summarized. Finally, the development prospects and challenges of these non-antibiotic compounds in combination with antibiotics are discussed.
Collapse
Affiliation(s)
| | | | - Zhiliang Sun
- College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China; (G.X.); (J.L.)
| |
Collapse
|
36
|
Grygorcewicz B, Gliźniewicz M, Olszewska P, Miłek D, Czajkowski A, Serwin N, Cecerska-Heryć E, Rakoczy R. Response Surface Methodology Application for Bacteriophage-Antibiotic Antibiofilm Activity Optimization. Microorganisms 2023; 11:2352. [PMID: 37764196 PMCID: PMC10536537 DOI: 10.3390/microorganisms11092352] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 09/13/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Phage-antibiotic combination-based protocols are presently under heightened investigation. This paradigm extends to engagements with bacterial biofilms, necessitating novel computational approaches to comprehensively characterize and optimize the outcomes achievable via these combinations. This study aimed to explore the Response Surface Methodology (RSM) in optimizing the antibiofilm activity of bacteriophage-antibiotic combinations. We employ a combination of antibiotics (gentamicin, meropenem, amikacin, ceftazidime, fosfomycin, imipenem, and colistin) alongside the bacteriophage vB_AbaP_AGC01 to combat Acinetobacter baumannii biofilm. Based on the conducted biofilm challenge assays analyzed using the RSM, the optimal points of antibiofilm activity efficacy were effectively selected by applying this methodology, enabling the quantifiable mathematical representations. Subsequent optimization showed the synergistic potential of the anti-biofilm that arises when antibiotics are judiciously combined with the AGC01 bacteriophage, reducing biofilm biomass by up to 80% depending on the antibiotic used. The data suggest that the phage-imipenem combination demonstrates the highest efficacy, with an 88.74% reduction. Notably, the lower concentrations characterized by a high maximum reduction in biofilm biomass were observed in the phage-amikacin combination at cA = 0.00195 and cP = 0.38 as the option that required minimum resources. It is worth noting that only gentamicin antagonism between the phage and the antibiotic was detected.
Collapse
Affiliation(s)
- Bartłomiej Grygorcewicz
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (M.G.); (P.O.); (D.M.); (A.C.); (N.S.); (E.C.-H.)
- Department of Chemical and Process Engineering, Faculty of Chemical Technology and Engineering, West Pomeranian University of Technology in Szczecin, Piastów Ave. 42, 71-065 Szczecin, Poland;
| | - Marta Gliźniewicz
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (M.G.); (P.O.); (D.M.); (A.C.); (N.S.); (E.C.-H.)
| | - Patrycja Olszewska
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (M.G.); (P.O.); (D.M.); (A.C.); (N.S.); (E.C.-H.)
| | - Dominika Miłek
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (M.G.); (P.O.); (D.M.); (A.C.); (N.S.); (E.C.-H.)
| | - Artur Czajkowski
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (M.G.); (P.O.); (D.M.); (A.C.); (N.S.); (E.C.-H.)
| | - Natalia Serwin
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (M.G.); (P.O.); (D.M.); (A.C.); (N.S.); (E.C.-H.)
| | - Elżbieta Cecerska-Heryć
- Faculty of Pharmacy, Medical Biotechnology and Laboratory Medicine, Pomeranian Medical University, Powstańców Wielkopolskich 72, 70-111 Szczecin, Poland; (M.G.); (P.O.); (D.M.); (A.C.); (N.S.); (E.C.-H.)
| | - Rafał Rakoczy
- Department of Chemical and Process Engineering, Faculty of Chemical Technology and Engineering, West Pomeranian University of Technology in Szczecin, Piastów Ave. 42, 71-065 Szczecin, Poland;
| |
Collapse
|
37
|
Suresh S, Saldanha J, Bhaskar Shetty A, Premanath R, Akhila DS, Mohan Raj JR. Comparison of Antibiofilm Activity of Pseudomonas aeruginosa Phages on Isolates from Wounds of Diabetic and Non-Diabetic Patients. Microorganisms 2023; 11:2230. [PMID: 37764074 PMCID: PMC10536433 DOI: 10.3390/microorganisms11092230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/03/2023] [Accepted: 08/15/2023] [Indexed: 09/29/2023] Open
Abstract
The persistence of organisms as biofilms and the increase in antimicrobial resistance has raised the need for alternative strategies. The study objective was to compare the ability of isolated bacteriophages to remove in vitro biofilms formed by Pseudomonas aeruginosa isolated from the environment with those isolated from diabetic and non-diabetic wounds. P. aeruginosa were isolated from clinical and environmental sites, and antimicrobial susceptibility was tested. Bacteriophages were isolated and characterized based on plaque morphology and host range. A reduction in the viable count assayed the lytic ability of candidate phages. The crystal violet method was used to determine the residual biofilm after 24 h of phage treatment on 72-h-old biofilms. The statistical significance of phage treatment was tested by one-way ANOVA. Of 35 clinical isolates, 17 showed resistance to 1 antibiotic at least, and 7 were multidrug resistant. Nineteen environmental isolates and 11 clinical isolates were drug-sensitive. Nine phages showed 91.2% host coverage, including multidrug-resistant isolates. Phages eradicated 85% of biofilms formed by environmental isolates compared to 58% of biofilms of diabetic isolates and 56% of biofilms of non-diabetic isolates. Clinical isolates are susceptible to phage infection in planktonic form. Biofilms of P. aeruginosa isolated from diabetic wounds and non-diabetic wounds resist removal by phages compared to biofilms formed by environmental isolates. All phages were efficient in dispersing PAO1 biofilms. However, there was a significant difference in their ability to disperse PAO1 biofilms across the different surfaces tested. Partial eradication of biofilm by phages can aid in complementing antibiotics that are unable to penetrate biofilms in a clinical set-up.
Collapse
Affiliation(s)
| | | | | | | | | | - Juliet Roshini Mohan Raj
- Division of Infectious Diseases, Nitte University Center for Science Education and Research, Paneer Campus, Nitte (Deemed to be University), Derelakatte, Mangaluru 575018, India (R.P.); (D.S.A.)
| |
Collapse
|
38
|
Bulssico J, PapukashvilI I, Espinosa L, Gandon S, Ansaldi M. Phage-antibiotic synergy: Cell filamentation is a key driver of successful phage predation. PLoS Pathog 2023; 19:e1011602. [PMID: 37703280 PMCID: PMC10519598 DOI: 10.1371/journal.ppat.1011602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 09/25/2023] [Accepted: 08/07/2023] [Indexed: 09/15/2023] Open
Abstract
Phages are promising tools to fight antibiotic-resistant bacteria, and as for now, phage therapy is essentially performed in combination with antibiotics. Interestingly, combined treatments including phages and a wide range of antibiotics lead to an increased bacterial killing, a phenomenon called phage-antibiotic synergy (PAS), suggesting that antibiotic-induced changes in bacterial physiology alter the dynamics of phage propagation. Using single-phage and single-cell techniques, each step of the lytic cycle of phage HK620 was studied in E. coli cultures treated with either ceftazidime, cephalexin or ciprofloxacin, three filamentation-inducing antibiotics. In the presence of sublethal doses of antibiotics, multiple stress tolerance and DNA repair pathways are triggered following activation of the SOS response. One of the most notable effects is the inhibition of bacterial division. As a result, a significant fraction of cells forms filaments that stop dividing but have higher rates of mutagenesis. Antibiotic-induced filaments become easy targets for phages due to their enlarged surface areas, as demonstrated by fluorescence microscopy and flow cytometry techniques. Adsorption, infection and lysis occur more often in filamentous cells compared to regular-sized bacteria. In addition, the reduction in bacterial numbers caused by impaired cell division may account for the faster elimination of bacteria during PAS. We developed a mathematical model to capture the interaction between sublethal doses of antibiotics and exposition to phages. This model shows that the induction of filamentation by sublethal doses of antibiotics can amplify the replication of phages and therefore yield PAS. We also use this model to study the consequences of PAS on the emergence of antibiotic resistance. A significant percentage of hyper-mutagenic filamentous bacteria are effectively killed by phages due to their increased susceptibility to infection. As a result, the addition of even a very low number of bacteriophages produced a strong reduction of the mutagenesis rate of the entire bacterial population. We confirm this prediction experimentally using reporters for bacterial DNA repair. Our work highlights the multiple benefits associated with the combination of sublethal doses of antibiotics with bacteriophages.
Collapse
Affiliation(s)
- Julián Bulssico
- Laboratoire de Chimie Bactérienne, UMR7283, Centre National de la Recherche Scientifique, Aix-Marseille Université, Marseille, France
| | - Irina PapukashvilI
- Laboratoire de Chimie Bactérienne, UMR7283, Centre National de la Recherche Scientifique, Aix-Marseille Université, Marseille, France
- Faculty of Exact and Natural Sciences, Ivane Javakhishvili Tbilisi State University, Tbilisi, Georgia
| | - Leon Espinosa
- Laboratoire de Chimie Bactérienne, UMR7283, Centre National de la Recherche Scientifique, Aix-Marseille Université, Marseille, France
| | - Sylvain Gandon
- CEFE, CNRS, Univ Montpellier, EPHE, IRD, Montpellier, France
| | - Mireille Ansaldi
- Laboratoire de Chimie Bactérienne, UMR7283, Centre National de la Recherche Scientifique, Aix-Marseille Université, Marseille, France
| |
Collapse
|
39
|
Osman AH, Kotey FCN, Odoom A, Darkwah S, Yeboah RK, Dayie NTKD, Donkor ES. The Potential of Bacteriophage-Antibiotic Combination Therapy in Treating Infections with Multidrug-Resistant Bacteria. Antibiotics (Basel) 2023; 12:1329. [PMID: 37627749 PMCID: PMC10451467 DOI: 10.3390/antibiotics12081329] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/04/2023] [Accepted: 08/05/2023] [Indexed: 08/27/2023] Open
Abstract
The growing threat of antibiotic resistance is a significant global health challenge that has intensified in recent years. The burden of antibiotic resistance on public health is augmented due to its multifaceted nature, as well as the slow-paced and limited development of new antibiotics. The threat posed by resistance is now existential in phage therapy, which had long been touted as a promising replacement for antibiotics. Consequently, it is imperative to explore the potential of combination therapies involving antibiotics and phages as a feasible alternative for treating infections with multidrug-resistant bacteria. Although either bacteriophage or antibiotics can potentially treat bacterial infections, they are each fraught with resistance. Combination therapies, however, yielded positive outcomes in most cases; nonetheless, a few combinations did not show any benefit. Combination therapies comprising the synergistic activity of phages and antibiotics and combinations of phages with other treatments such as probiotics hold promise in the treatment of drug-resistant bacterial infections.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Eric S. Donkor
- Department of Medical Microbiology, University of Ghana Medical School, Korle Bu, Accra P.O. Box KB 4236, Ghana; (A.-H.O.); (F.C.N.K.); (A.O.); (S.D.); (R.K.Y.); (N.T.K.D.D.)
| |
Collapse
|
40
|
Xiao J, Su L, Huang S, Liu L, Ali K, Chen Z. Epidemic Trends and Biofilm Formation Mechanisms of Haemophilus influenzae: Insights into Clinical Implications and Prevention Strategies. Infect Drug Resist 2023; 16:5359-5373. [PMID: 37605758 PMCID: PMC10440118 DOI: 10.2147/idr.s424468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/10/2023] [Indexed: 08/23/2023] Open
Abstract
Haemophilus influenzae (H. influenzae) is a significant pathogen responsible for causing respiratory tract infections and invasive diseases, leading to a considerable disease burden. The Haemophilus influenzae type b (Hib) conjugate vaccine has notably decreased the incidence of severe infections caused by Hib strains, and other non-typable H. influenzae (NTHi) serotypes have emerged as epidemic strains worldwide. As a result, the global epidemic trends and antibiotic resistance characteristics of H. influenzae have been altered. Researches on the virulence factors of H. influenzae, particularly the mechanisms underlying biofilm formation, and the development of anti-biofilm strategies hold significant clinical value. This article provides a summary of the epidemic trends, typing methods, virulence factors, biofilm formation mechanisms, and prevention strategies of H. influenzae. The increasing prevalence of NTHi strains and antibiotic resistance among H. influenzae, especially the high β-lactamase positivity and the emergence of BLNAR strains have increased clinical difficulties. Understanding its virulence factors, especially the formation mechanism of biofilm, and formulating effective anti-biofilm strategies may help to reduce the clinical impact. Therefore, future research efforts should focus on developing new approaches to prevent and control H. influenzae infections.
Collapse
Affiliation(s)
- Jiying Xiao
- Department of Pulmonology, Hangzhou Children’s Hospital, Hangzhou, Zhejiang, 310015, People’s Republic of China
| | - Lin Su
- Department of Pulmonology, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310052, People’s Republic of China
- National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, Zhejiang, 310052, People’s Republic of China
| | - Shumin Huang
- Department of Pulmonology, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310052, People’s Republic of China
- National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, Zhejiang, 310052, People’s Republic of China
| | - Lingyue Liu
- Department of Pulmonology, Hangzhou Children’s Hospital, Hangzhou, Zhejiang, 310015, People’s Republic of China
| | - Kamran Ali
- Department of Oncology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang, 322000, People’s Republic of China
| | - Zhimin Chen
- Department of Pulmonology, Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310052, People’s Republic of China
- National Clinical Research Center for Child Health, National Children’s Regional Medical Center, Hangzhou, Zhejiang, 310052, People’s Republic of China
| |
Collapse
|
41
|
Nicholls P, Clark JR, Gu Liu C, Terwilliger A, Maresso AW. Class-Driven Synergy and Antagonism between a Pseudomonas Phage and Antibiotics. Infect Immun 2023; 91:e0006523. [PMID: 37404162 PMCID: PMC10429645 DOI: 10.1128/iai.00065-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/19/2023] [Indexed: 07/06/2023] Open
Abstract
The ubiquitous bacterial pathogen Pseudomonas aeruginosa is responsible for severe infections in patients with burns, cystic fibrosis, and neutropenia. Biofilm formation gives physical refuge and a protected microenvironment for sessile cells, rendering cure by antibiotics a challenge. Bacteriophages have evolved to prey on these biofilms over millions of years, using hydrolases and depolymerases to penetrate biofilms and reach cellular targets. Here, we assessed how a newly discovered KMV-like phage (ΦJB10) interacts with antibiotics to treat P. aeruginosa more effectively in both planktonic and biofilm forms. By testing representatives of four classes of antibiotics (cephalosporins, aminoglycosides, fluoroquinolones, and carbapenems), we demonstrated class-dependent interactions between ΦJB10 and antibiotics in both biofilm clearance and P. aeruginosa killing. Despite identifying antagonism between some antibiotic classes and ΦJB10 at early time points, all classes showed neutral to favorable interactions with the phage at later time points. In one notable example where the antibiotic alone had poor activity against both biofilm and high-density planktonic cells, we found that addition of ΦJB10 demonstrated synergy and resulted in effective treatment of both. Further, ΦJB10 seemed to act as an adjuvant to several antibiotics, reducing the concentration of antibiotics required to ablate the biofilm. This report shows that phages such as ΦJB10 may be valuable additions to the armamentarium against difficult-to-treat biofilm-based infections.
Collapse
Affiliation(s)
- Paul Nicholls
- Section of Infectious Diseases, Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Justin R. Clark
- TAILΦR LABS, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Carmen Gu Liu
- TAILΦR LABS, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Austen Terwilliger
- TAILΦR LABS, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Anthony W. Maresso
- TAILΦR LABS, Baylor College of Medicine, Houston, Texas, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
42
|
Kalpana S, Lin WY, Wang YC, Fu Y, Wang HY. Alternate Antimicrobial Therapies and Their Companion Tests. Diagnostics (Basel) 2023; 13:2490. [PMID: 37568853 PMCID: PMC10417861 DOI: 10.3390/diagnostics13152490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 07/14/2023] [Indexed: 08/13/2023] Open
Abstract
New antimicrobial approaches are essential to counter antimicrobial resistance. The drug development pipeline is exhausted with the emergence of resistance, resulting in unsuccessful trials. The lack of an effective drug developed from the conventional drug portfolio has mandated the introspection into the list of potentially effective unconventional alternate antimicrobial molecules. Alternate therapies with clinically explicable forms include monoclonal antibodies, antimicrobial peptides, aptamers, and phages. Clinical diagnostics optimize the drug delivery. In the era of diagnostic-based applications, it is logical to draw diagnostic-based treatment for infectious diseases. Selection criteria of alternate therapeutics in infectious diseases include detection, monitoring of response, and resistance mechanism identification. Integrating these diagnostic applications is disruptive to the traditional therapeutic development. The challenges and mitigation methods need to be noted. Applying the goals of clinical pharmacokinetics that include enhancing efficacy and decreasing toxicity of drug therapy, this review analyses the strong correlation of alternate antimicrobial therapeutics in infectious diseases. The relationship between drug concentration and the resulting effect defined by the pharmacodynamic parameters are also analyzed. This review analyzes the perspectives of aligning diagnostic initiatives with the use of alternate therapeutics, with a particular focus on companion diagnostic applications in infectious diseases.
Collapse
Affiliation(s)
- Sriram Kalpana
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan;
| | - Wan-Ying Lin
- Department of Medicine, University of California San Diego, San Diego, CA 92093, USA;
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA;
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Yu-Chiang Wang
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA;
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Yiwen Fu
- Department of Medicine, Kaiser Permanente Santa Clara Medical Center, Santa Clara, CA 95051, USA;
| | - Hsin-Yao Wang
- Department of Laboratory Medicine, Linkou Chang Gung Memorial Hospital, Taoyuan 333423, Taiwan;
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA;
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
| |
Collapse
|
43
|
Tsunemoto H, Sugie J, Enustun E, Pogliano K, Pogliano J. Bacterial cytological profiling reveals interactions between jumbo phage φKZ infection and cell wall active antibiotics in Pseudomonas aeruginosa. PLoS One 2023; 18:e0280070. [PMID: 37418366 PMCID: PMC10328376 DOI: 10.1371/journal.pone.0280070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/20/2022] [Indexed: 07/09/2023] Open
Abstract
The emergence of antibiotic resistance in bacteria has led to the investigation of alternative treatments, such as phage therapy. In this study, we examined the interactions between the nucleus-forming jumbo phage ФKZ and antibiotic treatment against Pseudomonas aeruginosa. Using the fluorescence microscopy technique of bacterial cytological profiling, we identified mechanism-of-action-specific interactions between antibiotics that target different biosynthetic pathways and ФKZ infection. We found that certain classes of antibiotics strongly inhibited phage replication, while others had no effect or only mildly affected progression through the lytic cycle. Antibiotics that caused an increase in host cell length, such as the cell wall active antibiotic ceftazidime, prevented proper centering of the ФKZ nucleus via the PhuZ spindle at midcell, leading us to hypothesize that the kinetic parameters of the PhuZ spindle evolved to match the average length of the host cell. To test this, we developed a computational model explaining how the dynamic properties of the PhuZ spindle contribute to phage nucleus centering and why some antibiotics affect nucleus positioning while others do not. These findings provide an understanding of the molecular mechanisms underlying the interactions between antibiotics and jumbo phage replication.
Collapse
Affiliation(s)
- Hannah Tsunemoto
- Division of Biological Sciences, University of California, San Diego, San Diego, CA, United States of America
| | - Joseph Sugie
- Division of Biological Sciences, University of California, San Diego, San Diego, CA, United States of America
| | - Eray Enustun
- Division of Biological Sciences, University of California, San Diego, San Diego, CA, United States of America
| | - Kit Pogliano
- Division of Biological Sciences, University of California, San Diego, San Diego, CA, United States of America
| | - Joe Pogliano
- Division of Biological Sciences, University of California, San Diego, San Diego, CA, United States of America
| |
Collapse
|
44
|
Erol HB, Kaskatepe B, Yildiz S, Altanlar N. The effect of phage-antibiotic combination strategy on multidrug-resistant Acinetobacter baumannii biofilms. J Microbiol Methods 2023; 210:106752. [PMID: 37268109 DOI: 10.1016/j.mimet.2023.106752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 05/18/2023] [Accepted: 05/28/2023] [Indexed: 06/04/2023]
Abstract
Acinetobacter baumannii (A. baumannii) is considered a critical human pathogen due to multi-drug resistance and increased infections. As a result of the resistance of A. baumannii biofilms to antimicrobial agents, it is necessary to develop new biofilm control strategies. In the present study, we evaluated the efficacy of two previously isolated bacteriophage C2 phage, K3 phage and phage cocktail (C2 + K3 phage) as a therapeutic agent in combination with antibiotic (colistin) against biofilm of multidrug-resistant A. baumannii strains (n = 24). The effects of phage and antibiotics on mature biofilm were investigated simultaneously and sequentially in 24 and 48 h. The combination protocol was more effective than antibiotics alone in 54.16% of the strains in 24 h. The sequential application was more effective than the simultaneous protocol compared with the 24 h single applications. When the application of antibiotics and phages alone was compared with their combined administration in 48 h. The sequential and simultaneous applications were more effective than single applications in all strains except two. We observed that combination of phage and antibiotics could increase biofilm eradication and provides new insights into the use of bacteriophages and antibiotics in the treatment of biofilm-associated infections caused by antibiotic-resistant bacteria.
Collapse
Affiliation(s)
- Hilal Basak Erol
- Department of Pharmaceutical Microbiology, Ankara University Faculty of Pharmacy, Ankara, Turkey.
| | - Banu Kaskatepe
- Department of Pharmaceutical Microbiology, Ankara University Faculty of Pharmacy, Ankara, Turkey.
| | - Sulhiye Yildiz
- Department of Pharmaceutical Microbiology, Lokman Hekim University Faculty of Pharmacy, Ankara, Turkey
| | - Nurten Altanlar
- Department of Pharmaceutical Microbiology, Ankara University Faculty of Pharmacy, Ankara, Turkey
| |
Collapse
|
45
|
Shariati A, Noei M, Chegini Z. Bacteriophages: The promising therapeutic approach for enhancing ciprofloxacin efficacy against bacterial infection. J Clin Lab Anal 2023:e24932. [PMID: 37377167 PMCID: PMC10388223 DOI: 10.1002/jcla.24932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/14/2023] [Accepted: 06/11/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND The emergence of ciprofloxacin-resistant bacteria is a serious challenge worldwide, bringing the need to find new approaches to manage this bacterium. Bacteriophages (phages) have been shown inhibitory effects against ciprofloxacin-resistance bacteria; thus, ciprofloxacin resistance or tolerance may not affect the phage's infection ability. Additionally, researchers used phage-ciprofloxacin combination therapy for the inhibition of multidrug-resistant bacteria. RESULTS The sublethal concentrations of ciprofloxacin could lead to an increase in progeny production. Antibiotic treatments could enhance the release of progeny phages by shortening the lytic cycle and latent period. Thus, sublethal concentrations of antibiotics combined with phages can be used for the management of bacterial infections with high antibiotic resistance. In addition, combination therapy exerts various selection pressures that can mutually decrease phage and antibiotic resistance. Moreover, phage ciprofloxacin could significantly reduce bacterial counts in the biofilm community. Immediate usage of phages after the attachment of bacteria to the surface of the flow cells, before the development of micro-colonies, could lead to the best effect of phage therapy against bacterial biofilm. Noteworthy, phage should be used before antibiotics usage because this condition may have allowed phage replication to occur first before ciprofloxacin interrupted the bacterial DNA replication process, thereby interfering with the activity of the phages. Furthermore, the phage-ciprofloxacin combination showed a promising result for the management of Pseudomonas aeruginosa infections in mouse models. Nevertheless, low data are existing about the interaction between phages and ciprofloxacin in combination therapies, especially regarding the emergence of phage-resistant mutants. Additionally, there is a challenging and important question of how the combined ciprofloxacin with phages can increase antibacterial functions. Therefore, more examinations are required to support the clinical usage of phage-ciprofloxacin combination therapy.
Collapse
Affiliation(s)
- Aref Shariati
- Molecular and Medicine Research Centre, Khomein University of Medical Sciences, Khomein, Iran
| | - Milad Noei
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Zahra Chegini
- Department of Microbiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
46
|
Lauman P, Dennis JJ. Synergistic Interactions among Burkholderia cepacia Complex-Targeting Phages Reveal a Novel Therapeutic Role for Lysogenization-Capable Phages. Microbiol Spectr 2023; 11:e0443022. [PMID: 37195168 PMCID: PMC10269493 DOI: 10.1128/spectrum.04430-22] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 04/17/2023] [Indexed: 05/18/2023] Open
Abstract
Antimicrobial resistance is a danger to global public health and threatens many aspects of modern medicine. Bacterial species such as those of the Burkholderia cepacia complex (Bcc) cause life-threatening respiratory infections and are highly resistant to antibiotics. One promising alternative being explored to combat Bcc infections is phage therapy (PT): the use of phages to treat bacterial infections. Unfortunately, the utility of PT against many pathogenic species is limited by its prevailing paradigm: that only obligately lytic phages should be used therapeutically. It is thought that 'lysogenic' phages do not lyse all bacteria and can transfer antimicrobial resistance or virulence factors to their hosts. We argue that the tendency of a lysogenization-capable (LC) phage to form stable lysogens is not predicated exclusively on its ability to do so, and that the therapeutic suitability of a phage must be evaluated on a case-by-case basis. Concordantly, we developed several novel metrics-Efficiency of Phage Activity, Growth Reduction Coefficient, and Stable Lysogenization Frequency-and used them to evaluate eight Bcc-specific phages. Although these parameters vary considerably among Bcc phages, a strong inverse correlation (R2 = 0.67; P < 0.0001) exists between lysogen formation and antibacterial activity, indicating that certain LC phages with low frequency of stable lysogenization may be therapeutically efficacious. Moreover, we show that many LC Bcc phages interact synergistically with other phages in the first reported instance of mathematically defined polyphage synergy, and that these interactions result in the eradication of in vitro bacterial growth. Together, these findings reveal a novel therapeutic role for LC phages and challenge the current paradigm of PT. IMPORTANCE The spread of antimicrobial resistance is an imminent threat to public health around the world. Particularly concerning are species of the Burkholderia cepacia complex (Bcc), which cause life-threatening respiratory infections and are notoriously resistant to antibiotics. Phage therapy is a promising alternative being explored to combat Bcc infections and antimicrobial resistance in general, but its utility against many pathogenic species, including the Bcc, is restricted by the currently prevailing paradigm of exclusively using rare obligately lytic phages due to the perception that 'lysogenic' phages are therapeutically unsuitable. Our findings show that many lysogenization-capable phages exhibit powerful in vitro antibacterial activity both alone and through mathematically defined synergistic interactions with other phages, demonstrating a novel therapeutic role for LC phages and therefore challenging the currently prevailing paradigm of PT.
Collapse
Affiliation(s)
- Philip Lauman
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Jonathan J. Dennis
- Department of Biological Sciences, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
47
|
Marshall K, Marsella R. Topical Bacteriophage Therapy for Staphylococcal Superficial Pyoderma in Horses: A Double-Blind, Placebo-Controlled Pilot Study. Pathogens 2023; 12:828. [PMID: 37375518 DOI: 10.3390/pathogens12060828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/07/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
Increased antimicrobial resistance highlights the need for alternatives to antibiotics. Bacteriophages, which are benign viruses that kill bacteria, are promising. We studied the efficacy of topical bacteriophages for treating equine staphylococcal superficial pyodermas. Eight Staphylococcus aureus isolates were tested against a bacteriophage bank, and a cocktail consisting of two bacteriophages was prepared. Twenty horses with clinical and cytological evidence of superficial pyoderma and confirmed S. aureus infection based on swabbed culture were enrolled in the study. Each horse received both the bacteriophage cocktail and the placebo at two different infection sites, once daily for four weeks. Clinical lesions and cytology were evaluated weekly by an investigator who was unaware of the treatment sites. All infection sites were swabbed and cultured at the end of the study. A linear mixed model showed no significant differences between the placebo and treatment sites in terms of clinical signs, cytological scores of inflammation, and bacterial counts at the end of the study. It is possible that the bacteriophage cocktail killed S. aureus, but cytology scores did not change as new populations of cocci took over. The study limitations included a small sample size and inconsistent control of the underlying causes of pyodermas.
Collapse
Affiliation(s)
- Kalie Marshall
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, 2015 SW 16th Avenue, Gainesville, FL 32610, USA
| | - Rosanna Marsella
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, 2015 SW 16th Avenue, Gainesville, FL 32610, USA
| |
Collapse
|
48
|
Taha M, Arnaud T, Lightly TJ, Peters D, Wang L, Chen W, Cook BWM, Theriault SS, Abdelbary H. Combining bacteriophage and vancomycin is efficacious against MRSA biofilm-like aggregates formed in synovial fluid. Front Med (Lausanne) 2023; 10:1134912. [PMID: 37359001 PMCID: PMC10289194 DOI: 10.3389/fmed.2023.1134912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
Background Biofilm formation is a major clinical challenge contributing to treatment failure of periprosthetic joint infection (PJI). Lytic bacteriophages (phages) can target biofilm associated bacteria at localized sites of infection. The aim of this study is to investigate whether combination therapy of phage and vancomycin is capable of clearing Staphylococcus aureus biofilm-like aggregates formed in human synovial fluid. Methods In this study, S. aureus BP043, a PJI clinical isolate was utilized. This strain is a methicillin-resistant S. aureus (MRSA) biofilm-former. Phage Remus, known to infect S. aureus, was selected for the treatment protocol. BP043 was grown as aggregates in human synovial fluid. The characterization of S. aureus aggregates was assessed for structure and size using scanning electron microscopy (SEM) and flow cytometry, respectively. Moreover, the formed aggregates were subsequently treated in vitro with: (a) phage Remus [∼108 plaque-forming units (PFU)/ml], (b) vancomycin (500 μg/ml), or (c) phage Remus (∼108 PFU/ml) followed by vancomycin (500 μg/ml), for 48 h. Bacterial survival was quantified by enumeration [colony-forming units (CFU)/ml]. The efficacy of phage and vancomycin against BP043 aggregates was assessed in vivo as individual treatments and in combination. The in vivo model utilized Galleria mellonella larvae which were infected with BP043 aggregates pre-formed in synovial fluid. Results Scanning electron microscopy (SEM) images and flow cytometry data demonstrated the ability of human synovial fluid to promote formation of S. aureus aggregates. Treatment with Remus resulted in significant reduction in viable S. aureus residing within the synovial fluid aggregates compared to the aggregates that did not receive Remus (p < 0.0001). Remus was more efficient in eliminating viable bacteria within the aggregates compared to vancomycin (p < 0.0001). Combination treatment of Remus followed by vancomycin was more efficacious in reducing bacterial load compared to using either Remus or vancomycin alone (p = 0.0023, p < 0.0001, respectively). When tested in vivo, this combination treatment also resulted in the highest survival rate (37%) 96 h post-treatment, compared to untreated larvae (3%; p < 0.0001). Conclusion We demonstrate that combining phage Remus and vancomycin led to synergistic interaction against MRSA biofilm-like aggregates in vitro and in vivo.
Collapse
Affiliation(s)
- Mariam Taha
- The Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Division of Orthopedic Surgery, The Ottawa Hospital, Ottawa, ON, Canada
| | - Tia Arnaud
- Cytophage Technologies Inc., Winnipeg, MB, Canada
- Department of Microbiology, The University of Manitoba, Winnipeg, MB, Canada
| | | | - Danielle Peters
- Human Health Therapeutics Research Center, National Research Council Canada, Ottawa, ON, Canada
| | - Liyuan Wang
- Cell Biology and Image Acquisition, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Wangxue Chen
- Human Health Therapeutics Research Center, National Research Council Canada, Ottawa, ON, Canada
- Department of Biology, Brock University, St. Catharines, ON, Canada
| | | | - Steven S. Theriault
- Cytophage Technologies Inc., Winnipeg, MB, Canada
- Department of Microbiology, The University of Manitoba, Winnipeg, MB, Canada
| | - Hesham Abdelbary
- The Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Division of Orthopedic Surgery, The Ottawa Hospital, Ottawa, ON, Canada
- Department of Surgery, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
49
|
Fungo GBN, Uy JCW, Porciuncula KLJ, Candelario CMA, Chua DPS, Gutierrez TAD, Clokie MRJ, Papa DMD. "Two Is Better Than One": The Multifactorial Nature of Phage-Antibiotic Combinatorial Treatments Against ESKAPE-Induced Infections. PHAGE (NEW ROCHELLE, N.Y.) 2023; 4:55-67. [PMID: 37350995 PMCID: PMC10282822 DOI: 10.1089/phage.2023.0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/24/2023]
Abstract
Phage-antibiotic synergy (PAS) has been extensively explored over the past decade, with the aim of developing more effective treatments against multidrug-resistant organisms. However, it remains unclear how to effectively combine these two approaches. To address this uncertainty, we assessed four main aspects of PAS interactions in this review, seeking to identify commonalities of combining treatments within and between bacterial species. We examined all literature on PAS efficacy toward ESKAPE pathogens and present an analysis of the data in papers focusing on: (1) order of treatment, (2) dose of both phage and antibiotics, (3) mechanism of action, and (4) viability of transfer from in vivo or animal model trials to clinical applications. Our analysis indicates that there is little consistency within phage-antibiotic therapy regimens, suggesting that highly individualized treatment regimens should be used. We propose a set of experimental studies to address these research gaps. We end our review with suggestions on how to improve studies on phage-antibiotic combination therapy to advance this field.
Collapse
Affiliation(s)
- Gale Bernice N. Fungo
- Department of Biological Sciences, College of Science, University of Santo Tomas, Manila, Philippines
- Bacteriophage Ecology, Aquaculture, Therapy and Systematics (BEATS) Research Group, University of Santo Tomas, Manila, Philippines
- Research Center for Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
| | - John Christian W. Uy
- Department of Biological Sciences, College of Science, University of Santo Tomas, Manila, Philippines
- Bacteriophage Ecology, Aquaculture, Therapy and Systematics (BEATS) Research Group, University of Santo Tomas, Manila, Philippines
- Research Center for Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
| | - Kristiana Louise J. Porciuncula
- Department of Biological Sciences, College of Science, University of Santo Tomas, Manila, Philippines
- Bacteriophage Ecology, Aquaculture, Therapy and Systematics (BEATS) Research Group, University of Santo Tomas, Manila, Philippines
- Research Center for Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
| | - Chiarah Mae A. Candelario
- Department of Biological Sciences, College of Science, University of Santo Tomas, Manila, Philippines
- Bacteriophage Ecology, Aquaculture, Therapy and Systematics (BEATS) Research Group, University of Santo Tomas, Manila, Philippines
- Research Center for Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
| | - Deneb Philip S. Chua
- Department of Biological Sciences, College of Science, University of Santo Tomas, Manila, Philippines
- Bacteriophage Ecology, Aquaculture, Therapy and Systematics (BEATS) Research Group, University of Santo Tomas, Manila, Philippines
- Research Center for Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
| | - Tracey Antaeus D. Gutierrez
- Bacteriophage Ecology, Aquaculture, Therapy and Systematics (BEATS) Research Group, University of Santo Tomas, Manila, Philippines
- Research Center for Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
| | | | - Donna May D. Papa
- Department of Biological Sciences, College of Science, University of Santo Tomas, Manila, Philippines
- Bacteriophage Ecology, Aquaculture, Therapy and Systematics (BEATS) Research Group, University of Santo Tomas, Manila, Philippines
- Research Center for Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
| |
Collapse
|
50
|
Zia S, Alkheraije KA. Recent trends in the use of bacteriophages as replacement of antimicrobials against food-animal pathogens. Front Vet Sci 2023; 10:1162465. [PMID: 37303721 PMCID: PMC10247982 DOI: 10.3389/fvets.2023.1162465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/06/2023] [Indexed: 06/13/2023] Open
Abstract
A major public health impact is associated with foodborne illnesses around the globe. Additionally, bacteria are becoming more resistant to antibiotics, which pose a global threat. Currently, many scientific efforts have been made to develop and implement new technologies to combat bacteria considering the increasing emergence of multidrug-resistant bacteria. In recent years, there has been considerable interest in using phages as biocontrol agents for foodborne pathogens in animals used for food production and in food products themselves. Foodborne outbreaks persist, globally, in many foods, some of which lack adequate methods to control any pathogenic contamination (like fresh produce). This interest may be attributed both to consumers' desire for more natural food and to the fact that foodborne outbreaks continue to occur in many foods. Poultry is the most common animal to be treated with phage therapy to control foodborne pathogens. A large number of foodborne illnesses worldwide are caused by Salmonella spp. and Campylobacter, which are found in poultry and egg products. Conventional bacteriophage-based therapy can prevent and control humans and animals from various infectious diseases. In this context, describing bacteriophage therapy based on bacterial cells may offer a breakthrough for treating bacterial infections. Large-scale production of pheasants may be economically challenging to meet the needs of the poultry market. It is also possible to produce bacteriophage therapy on a large scale at a reduced cost. Recently, they have provided an ideal platform for designing and producing immune-inducing phages. Emerging foodborne pathogens will likely be targeted by new phage products in the future. In this review article, we will mainly focus on the Bacteriophages (phages) that have been proposed as an alternative strategy to antibiotics for food animal pathogens and their use for public health and food safety.
Collapse
Affiliation(s)
- Sana Zia
- Department of Zoology, Government Sadiq College Women University Bahawalpur, Bahawalpur, Pakistan
| | - Khalid A. Alkheraije
- Department of Veterinary Medicine College of Agriculture and Veterinary Medicine, Qassim University, Buraidah, Saudi Arabia
| |
Collapse
|