1
|
Fong SA, Bouras G, Houtak G, Nepal R, Feizi S, Morales S, Psaltis AJ, Wormald PJ, Vreugde S. Genomic variation in Pseudomonas aeruginosa clinical respiratory isolates with de novo resistance to a bacteriophage cocktail. Microbiol Spectr 2025:e0214924. [PMID: 40162801 DOI: 10.1128/spectrum.02149-24] [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: 08/30/2024] [Accepted: 01/20/2025] [Indexed: 04/02/2025] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that can cause sinus infections and pneumonia in cystic fibrosis (CF) patients. Bacteriophage therapy is being investigated as a treatment for antibiotic-resistant P. aeruginosa infections. Although virulent bacteriophages have shown promise in treating P. aeruginosa infections, the development of bacteriophage-insensitive mutants (BIMs) in the presence of bacteriophages has been described. The aim of this study was to examine the genetic changes associated with the BIM phenotype. Biofilms of three genetically distinct P. aeruginosa strains, including PAO1 (ATCC 15692), and two clinical respiratory isolates (one CF and one non-CF) were grown for 7 days and treated with either a cocktail of four bacteriophages or a vehicle control for 7 consecutive days. BIMs isolated from the biofilms were detected by streak assays, and resistance to the phage cocktail was confirmed using spot test assays. Comparison of whole genome sequencing between the recovered BIMs and their respective vehicle control-treated phage-sensitive isolates revealed structural variants in two strains, and several small variants in all three strains. These variations involved a TonB-dependent outer membrane receptor in one strain, and mutations in lipopolysaccharide synthesis genes in two strains. Prophage deletion and induction were also noted in two strains, as well as mutations in several genes associated with virulence factors. Mutations in genes involved in susceptibility to conventional antibiotics were also identified in BIMs, with both decreased and increased antibiotic sensitivity to various antibiotics being observed. These findings may have implications for future applications of lytic phage therapy.IMPORTANCELytic bacteriophages are viruses that infect and kill bacteria and can be used to treat difficult-to-treat bacterial infections, including biofilm-associated infections and multidrug-resistant bacteria. Pseudomonas aeruginosa is a bacterium that can cause life-threatening infections. Lytic bacteriophage therapy has been trialed in the treatment of P. aeruginosa infections; however, sometimes bacteria develop resistance to the bacteriophages. This study sheds light on the genetic mechanisms of such resistance, and how this might be harnessed to restore the sensitivity of multidrug-resistant P. aeruginosa to conventional antibiotics.
Collapse
Affiliation(s)
- Stephanie A Fong
- Department of Surgery - Otolaryngology Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| | - George Bouras
- Department of Surgery - Otolaryngology Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| | - Ghais Houtak
- Department of Surgery - Otolaryngology Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| | - Roshan Nepal
- Department of Surgery - Otolaryngology Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| | - Sholeh Feizi
- Department of Surgery - Otolaryngology Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| | - Sandra Morales
- AmpliPhi Australia, Brookvale, New South Wales, Australia
| | - Alkis J Psaltis
- Department of Surgery - Otolaryngology Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| | - Peter-John Wormald
- Department of Surgery - Otolaryngology Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| | - Sarah Vreugde
- Department of Surgery - Otolaryngology Head and Neck Surgery, University of Adelaide, Adelaide, Australia
| |
Collapse
|
2
|
Michalik M, Podbielska-Kubera A, Dmowska-Koroblewska A. Antibiotic Resistance of Staphylococcus aureus Strains-Searching for New Antimicrobial Agents-Review. Pharmaceuticals (Basel) 2025; 18:81. [PMID: 39861144 PMCID: PMC11768290 DOI: 10.3390/ph18010081] [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: 11/10/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
Inappropriate and excessive use of antibiotics is responsible for the rapid development of antimicrobial resistance, which is associated with increased patient morbidity and mortality. There is an urgent need to explore new antibiotics or alternative antimicrobial agents. S. aureus a commensal microorganism but is also responsible for numerous infections. In addition to innate resistance to β-lactam antibiotics, S. aureus strains resistant to methicillin (MRSA) often show resistance to other classes of antibiotics (multidrug resistance). The advancement of phage therapy against MRSA infections offers a promising alternative in the context of increasing antibiotic resistance. Therapeutic phages are easier to obtain and cheaper to produce than antibiotics. However, there is still a lack of standards to ensure the safe use of phages, including purification, dosage, means of administration, and the quantity of phages used. Some bacteria have developed defense mechanisms against phages. The use of phage cocktails or the combination of antibiotics and phages is preferred. For personalized therapy, it is essential to set up large collections to enable phage selection. In the future, the fight against MRSA strains using phages should be based on a multidisciplinary approach, including molecular biology and medicine. Other therapies in the fight against MRSA strains include the use of endolysin antimicrobial peptides (including defensins and cathelicidins). Researchers' activities also focus on the potential use of plant extracts, honey, propolis, alkaloids, and essential oils. To date, no vaccine has been approved against S. aureus strains.
Collapse
|
3
|
Deng Y, Jiang S, Duan H, Shao H, Duan Y. Bacteriophages and their potential for treatment of metabolic diseases. J Diabetes 2024; 16:e70024. [PMID: 39582431 PMCID: PMC11586638 DOI: 10.1111/1753-0407.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 10/16/2024] [Indexed: 11/26/2024] Open
Abstract
Recent advances highlight the role of gut virome, particularly phageome, in metabolic disorders such as obesity, type 2 diabetes mellitus, metabolic dysfunction-associated fatty liver disease, and cardiovascular diseases, including hypertension, stroke, coronary heart disease, and hyperlipidemia. While alterations in gut bacteria are well-documented, emerging evidence suggests that changes in gut viruses also contribute to these disorders. Bacteriophages, the most abundant gut viruses, influence bacterial populations through their lytic and lysogenic cycles, potentially modulating the gut ecosystem and metabolic pathways. Phage therapy, previously overshadowed by antibiotics, is experiencing renewed interest due to rising antibiotic resistance. It offers a novel approach to precisely edit the gut microbiota, with promising applications in metabolic diseases. In this review, we summarize recent discoveries about gut virome in metabolic disease patients, review preclinical and clinical studies of phage therapy on metabolic diseases as well as the breakthroughs and currently faced problems and concerns.
Collapse
Affiliation(s)
- Youpeng Deng
- Department of Infectious Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Key Laboratory of Immune Response and Immunotherapy, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Shouwei Jiang
- Department of Infectious Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Hanyu Duan
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Key Laboratory of Immune Response and Immunotherapy, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Haonan Shao
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Key Laboratory of Immune Response and Immunotherapy, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Yi Duan
- Department of Infectious Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Key Laboratory of Immune Response and Immunotherapy, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| |
Collapse
|
4
|
Kennewell TL, Haidari H, Mashtoub S, Howarth GS, Wormald PJ, Cowin AJ, Vreugde S, Kopecki Z. Deferiprone and Gallium-Protoporphyrin Chitogel as an antimicrobial treatment: Preclinical studies demonstrating antimicrobial activity for S. aureus infected cutaneous wounds. Int J Biol Macromol 2024; 276:133874. [PMID: 39013511 DOI: 10.1016/j.ijbiomac.2024.133874] [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: 09/18/2023] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 07/18/2024]
Abstract
Staphylococcus aureus (S. aureus) is one of the most common wound pathogens with increased resistance towards currently available antimicrobials. S. aureus biofilms lead to increase wound chronicity and delayed healing. Chitosan-dextran hydrogel (Chitogel) loaded with the hydroxypyridinone-derived iron chelator Deferiprone (Def) and the heme analogue Gallium-Protoporphyrin (GaPP) have previously been shown to have antimicrobial effects in clinical sinusitis. In this study, the efficacy of Chitogel loaded with Def, GaPP and a combination of Def and GaPP, were investigated in an S. aureus biofilm infected wound murine model over 10 days of treatment. Bacterial wound burden was monitored daily showing a significant decrease in bacterial bioburden on days 6 and 8 when treated with Def-GaPP Chitogel (log10 1.0 and 1.2 reduction vs control, respectively). The current study demonstrates that the combination of Def-GaPP delivered in a Chitogel in vivo is not only effective in reducing S. aureus biofilm infection, but also improves cutaneous healing via effects on reduced inflammation, promotion of anti-inflammatory macrophage phenotype and marked early collagen deposition in the wound bed. This delivery platform presents a promising alternative non-toxic, antibacterial, wound-promoting treatment as a novel approach for the management of S. aureus wound infections that warrants further clinical investigation.
Collapse
Affiliation(s)
- T L Kennewell
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
| | - H Haidari
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
| | - S Mashtoub
- School of Biomedicine, The University of Adelaide, Adelaide, SA, Australia; Department of Gastroenterology, Women's and Children's Hospital, North Adelaide, SA, Australia
| | - G S Howarth
- School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, SA, Australia
| | - P J Wormald
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia; Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South, SA, Australia
| | - A J Cowin
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia
| | - S Vreugde
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia; Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville South, SA, Australia
| | - Z Kopecki
- Future Industries Institute, University of South Australia, Mawson Lakes, SA, Australia.
| |
Collapse
|
5
|
Santamaría-Corral G, Aguilera-Correa JJ, Esteban J, García-Quintanilla M. Bacteriophage Therapy on an In Vitro Wound Model and Synergistic Effects in Combination with Beta-Lactam Antibiotics. Antibiotics (Basel) 2024; 13:800. [PMID: 39334975 PMCID: PMC11428794 DOI: 10.3390/antibiotics13090800] [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: 07/20/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
One of the primary opportunistic pathogens that can cause a wide range of diseases is Pseudomonas aeruginosa. This microorganism can become resistant to practically every antibacterial currently in use, including beta-lactam antibiotics. Its ability to proliferate as biofilm has been linked to, among other things, the failure of antimicrobial therapies. Due to a variety of virulence factors and host immune system modifications, P. aeruginosa is one of the most significant and common bacteria that colonize wounds and burns. A novel therapeutic option for treating these multidrug-resistant (MDR) bacterial infections is the combination of antibiotics and bacteriophages. This approach has been linked to improved biofilm penetration, a decreased selection of antibiotic and bacteriophage resistance, and an enhanced antibacterial impact. Combining the F1Pa bacteriophage and beta-lactam antibiotics reduced the viability of the mature biofilm of MDR P. aeruginosa strains and suppressed bacterial growth in vitro. F1Pa critically reduced the amount of biofilm that MDR P. aeruginosa clinical strains formed in the in vitro wound model. These findings highlight the bacteriophage F1Pa's therapeutic potential as a prophylactic topical treatment against MDR pseudomonal infections in wounds and burns.
Collapse
Affiliation(s)
- Guillermo Santamaría-Corral
- Clinical Microbiology Department, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain
| | - John Jairo Aguilera-Correa
- Clinical Microbiology Department, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain
- CIBERINFEC-Consorcio Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas, 28029 Madrid, Spain
| | - Jaime Esteban
- Clinical Microbiology Department, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain
- CIBERINFEC-Consorcio Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas, 28029 Madrid, Spain
| | - Meritxell García-Quintanilla
- Clinical Microbiology Department, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain
- CIBERINFEC-Consorcio Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas, 28029 Madrid, Spain
| |
Collapse
|
6
|
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
|
7
|
Santamaría-Corral G, Pagán I, Aguilera-Correa JJ, Esteban J, García-Quintanilla M. A Novel Bacteriophage Infecting Multi-Drug- and Extended-Drug-Resistant Pseudomonas aeruginosa Strains. Antibiotics (Basel) 2024; 13:523. [PMID: 38927189 PMCID: PMC11200629 DOI: 10.3390/antibiotics13060523] [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: 04/11/2024] [Revised: 05/13/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
The prevalence of carbapenem-resistant P. aeruginosa has dramatically increased over the last decade, and antibiotics alone are not enough to eradicate infections caused by this opportunistic pathogen. Phage therapy is a fresh treatment that can be administered under compassionate use, particularly against chronic cases. However, it is necessary to thoroughly characterize the virus before therapeutic application. Our work describes the discovery of the novel sequenced bacteriophage, vB_PaeP-F1Pa, containing an integrase, performs a phylogenetical analysis, describes its stability at a physiological pH and temperature, latent period (40 min), and burst size (394 ± 166 particles per bacterial cell), and demonstrates its ability to infect MDR and XDR P. aeruginosa strains. Moreover, this novel bacteriophage was able to inhibit the growth of bacteria inside preformed biofilms. The present study offers a road map to analyze essential areas for successful phage therapy against MDR and XDR P. aeruginosa infections, and shows that a phage containing an integrase is also able to show good in vitro results, indicating that it is very important to perform a genomic analysis before any clinical use, in order to prevent adverse effects in patients.
Collapse
Affiliation(s)
- Guillermo Santamaría-Corral
- Clinical Microbiology Department, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (G.S.-C.); (J.J.A.-C.); (M.G.-Q.)
| | - Israel Pagán
- Centro de Biotecnología y Genómica de Plantas UPM-INIA/CSIC and E.T.S. Ingeniería Agronómica, Alimentaria y de Biosistemas, Universidad Politécnica de Madrid, 28223 Madrid, Spain;
| | - John Jairo Aguilera-Correa
- Clinical Microbiology Department, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (G.S.-C.); (J.J.A.-C.); (M.G.-Q.)
- CIBERINFEC-Consorcio Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas, 28029 Madrid, Spain
| | - Jaime Esteban
- Clinical Microbiology Department, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (G.S.-C.); (J.J.A.-C.); (M.G.-Q.)
- CIBERINFEC-Consorcio Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas, 28029 Madrid, Spain
| | - Meritxell García-Quintanilla
- Clinical Microbiology Department, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (G.S.-C.); (J.J.A.-C.); (M.G.-Q.)
- CIBERINFEC-Consorcio Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas, 28029 Madrid, Spain
| |
Collapse
|
8
|
Tayyarcan EK, Evran E, Guven K, Ekiz E, Acar Soykut E, Boyaci IH. Evaluating the efficacy of a phage cocktail against Pseudomonas fluorescens group strains in raw milk: microbiological, physical, and chemical analyses. Arch Microbiol 2024; 206:283. [PMID: 38806864 DOI: 10.1007/s00203-024-04008-1] [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: 04/04/2024] [Revised: 05/02/2024] [Accepted: 05/16/2024] [Indexed: 05/30/2024]
Abstract
The objective of this study was to investigate the effectiveness of a phage cocktail against Pseudomonas fluorescens group and its effect on the microbial, physical and chemical properties of raw milk during different storage conditions. A phage cocktail consisting of Pseudomonas fluorescens, Pseudomonas tolaasii, and Pseudomonas libanensis phages was prepared. As a result, reductions in fluorescent Pseudomonas counts of up to 3.44 log units for the storage at 4 °C and 2.38 log units for the storage at 25 °C were achieved. Following the phage application, it is found that there was no significant difference in the total mesophilic aerobic bacteria and Enterobacteriaceae counts. However, it was observed that the number of lactic acid bacteria was higher in phage-treated groups. The results also showed that pH values in the phage added groups were lower than the others and the highest titratable acidity was obtained only in the bacteria-inoculated group. As a future perspective, this study suggests that, while keeping the number of target microorganisms under control in the milk with the use of phages during storage, the microbiota and accordingly the quality parameters of the milk can be affected. This work contributes to the development of effective strategies for maintaining the quality and extending the shelf life of milk and dairy products.
Collapse
Affiliation(s)
| | - Eylul Evran
- Food Engineering Department, Hacettepe University, Beytepe, Ankara, Turkey
| | - Kubra Guven
- Food Engineering Department, Hacettepe University, Beytepe, Ankara, Turkey
| | - Esra Ekiz
- Food Engineering Department, Hacettepe University, Beytepe, Ankara, Turkey
| | - Esra Acar Soykut
- Food Engineering Department, Bolu Abant Izzet Baysal University, Bolu, Turkey
| | | |
Collapse
|
9
|
Harris EB, Ewool KKK, Bowden LC, Fierro J, Johnson D, Meinzer M, Tayler S, Grose JH. Genomic and Proteomic Analysis of Six Vi01-like Phages Reveals Wide Host Range and Multiple Tail Spike Proteins. Viruses 2024; 16:289. [PMID: 38400064 PMCID: PMC10892097 DOI: 10.3390/v16020289] [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: 12/29/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 02/25/2024] Open
Abstract
Enterobacteriaceae is a large family of Gram-negative bacteria composed of many pathogens, including Salmonella and Shigella. Here, we characterize six bacteriophages that infect Enterobacteriaceae, which were isolated from wastewater plants in the Wasatch front (Utah, United States). These phages are highly similar to the Kuttervirus vB_SenM_Vi01 (Vi01), which was isolated using wastewater from Kiel, Germany. The phages vary little in genome size and are between 157 kb and 164 kb, which is consistent with the sizes of other phages in the Vi01-like phage family. These six phages were characterized through genomic and proteomic comparison, mass spectrometry, and both laboratory and clinical host range studies. While their proteomes are largely unstudied, mass spectrometry analysis confirmed the production of five hypothetical proteins, several of which unveiled a potential operon that suggests a ferritin-mediated entry system on the Vi01-like phage family tail. However, no dependence on this pathway was observed for the single host tested herein. While unable to infect every genus of Enterobacteriaceae tested, these phages are extraordinarily broad ranged, with several demonstrating the ability to infect Salmonella enterica and Citrobacter freundii strains with generally high efficiency, as well as several clinical Salmonella enterica isolates, most likely due to their multiple tail fibers.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Julianne H. Grose
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84604, USA; (E.B.H.); (K.K.K.E.)
| |
Collapse
|
10
|
Kifelew LG, Warner MS, Morales S, Gordon DL, Thomas N, Mitchell JG, Speck PG. Lytic activity of phages against bacterial pathogens infecting diabetic foot ulcers. Sci Rep 2024; 14:3515. [PMID: 38347019 PMCID: PMC10861545 DOI: 10.1038/s41598-024-53317-4] [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: 04/06/2023] [Accepted: 01/30/2024] [Indexed: 02/15/2024] Open
Abstract
Complications of diabetes, such as diabetic foot ulcers (DFUs), are common, multifactorial in origin, and costly to treat. DFUs are the cause of nearly 90% of limb amputations among persons with diabetes. In most chronic infections such as DFU, biofilms are involved. Bacteria in biofilms are 100-1000 times more resistant to antibiotics than their planktonic counterparts. Multidrug-resistant (MDR) Staphylococcus aureus and Pseudomonas aeruginosa infections in DFUs may require alternative therapeutic agents such as bacteriophages ("phages"). This study describes the lytic activity of phage cocktails AB-SA01 (3-phage cocktail) and AB-PA01 (4-phage cocktail), which target S. aureus and P. aeruginosa, respectively. The host range and lytic effect of AB-SA01 and AB-PA01 on a planktonic culture, single-species biofilm, and mixed-species biofilm were evaluated. In vitro testing showed that 88.7% of S. aureus and 92.7% of P. aeruginosa isolates were susceptible to AB-SA01 and AB-PA01, respectively, in the planktonic state. The component phages of AB-SA01 and AB-PA01 infected 66% to 94.3% of the bacterial isolates tested. Furthermore, AB-SA01 and AB-PA01 treatment significantly (p < 0.05) reduced the biofilm biomass of their hosts, regardless of the antibiotic-resistant characteristics of the isolates and the presence of a non-susceptible host. In conclusion, the strong lytic activity, broad host range, and significant biofilm biomass reduction of AB-SA01 and AB-PA01 suggest the considerable potential of phages in treating antibiotic-resistant S. aureus and P. aeruginosa infections alone or as coinfections in DFUs.
Collapse
Affiliation(s)
- Legesse Garedew Kifelew
- College of Science and Engineering, Flinders University, Bedford Park, SA, 5042, Australia.
- St Paul's Hospital Millennium Medical College, 1271, Addis Ababa, Ethiopia.
| | - Morgyn S Warner
- Infectious Diseases Unit, Queen Elizabeth Hospital, Woodville, SA, 5011, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Sandra Morales
- AmpliPhi Australia Pty Ltd., Brookvale, NSW, 2100, Australia
- Phage Consulting, Sydney, NSW, 2100, Australia
| | - David L Gordon
- Department of Microbiology and Infectious Diseases, College of Medicine and Public Health, Flinders University, Bedford Park, SA, 5042, Australia
| | - Nicky Thomas
- Basil Hetzel Institute for Translational Health Research, Woodville South, SA, 5011, Australia
- Centre for Pharmaceutical Innovation, University of South Australia, North Terrace, Adelaide, SA, 5000, Australia
| | - James G Mitchell
- College of Science and Engineering, Flinders University, Bedford Park, SA, 5042, Australia
| | - Peter G Speck
- College of Science and Engineering, Flinders University, Bedford Park, SA, 5042, Australia.
| |
Collapse
|
11
|
Podlacha M, Węgrzyn G, Węgrzyn A. Bacteriophages-Dangerous Viruses Acting Incognito or Underestimated Saviors in the Fight against Bacteria? Int J Mol Sci 2024; 25:2107. [PMID: 38396784 PMCID: PMC10889324 DOI: 10.3390/ijms25042107] [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: 01/14/2024] [Revised: 02/04/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
The steadily increasing number of drug-resistant bacterial species has prompted the search for alternative treatments, resulting in a growing interest in bacteriophages. Although they are viruses infecting bacterial cells, bacteriophages are an extremely important part of the human microbiota. By interacting with eukaryotic cells, they are able to modulate the functioning of many systems, including the immune and nervous systems, affecting not only the homeostasis of the organism, but potentially also the regulation of pathological processes. Therefore, the aim of this review is to answer the questions of (i) how animal/human immune systems respond to bacteriophages under physiological conditions and under conditions of reduced immunity, especially during bacterial infection; (ii) whether bacteriophages can induce negative changes in brain functioning after crossing the blood-brain barrier, which could result in various disorders or in an increase in the risk of neurodegenerative diseases; and (iii) how bacteriophages can modify gut microbiota. The crucial dilemma is whether administration of bacteriophages is always beneficial or rather if it may involve any risks.
Collapse
Affiliation(s)
- Magdalena Podlacha
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland; (M.P.); (G.W.)
| | - Grzegorz Węgrzyn
- Department of Molecular Biology, University of Gdansk, Wita Stwosza 59, 80-308 Gdansk, Poland; (M.P.); (G.W.)
| | - Alicja Węgrzyn
- Phage Therapy Center, University Center for Applied and Interdisciplinary Research, University of Gdansk, Kładki 24, 80-822 Gdansk, Poland
| |
Collapse
|
12
|
Samson R, Dharne M, Khairnar K. Bacteriophages: Status quo and emerging trends toward one health approach. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 908:168461. [PMID: 37967634 DOI: 10.1016/j.scitotenv.2023.168461] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/30/2023] [Accepted: 11/07/2023] [Indexed: 11/17/2023]
Abstract
The alarming rise in antimicrobial resistance (AMR) among the drug-resistant pathogens has been attributed to the ESKAPEE group (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumanii, Pseudomonas aeruginosa, Enterobacter sp., and Escherichia coli). Recently, these AMR microbes have become difficult to treat, as they have rendered the existing therapeutics ineffective. Thus, there is an urgent need for effective alternatives to lessen or eliminate the current infections and limit the spread of emerging diseases under the "One Health" framework. Bacteriophages (phages) are naturally occurring biological resources with extraordinary potential for biomedical, agriculture/food safety, environmental protection, and energy production. Specific unique properties of phages, such as their bactericidal activity, host specificity, potency, and biocompatibility, make them desirable candidates in therapeutics. The recent biotechnological advancement has broadened the repertoire of phage applications in nanoscience, material science, physical chemistry, and soft-matter research. Herein, we present a comprehensive review, coupling the substantial aspects of phages with their applicability status and emerging opportunities in several interdependent areas under one health concept. Consolidating the recent state-of-the-art studies that integrate human, animal, plant, and environment health, the following points have been highlighted: (i) The biomedical and pharmacological advantages of phages and their antimicrobial derivatives with particular emphasis on in-vivo and clinical studies. (ii) The remarkable potential of phages to be altered, improved, and applied for drug delivery, biosensors, biomedical imaging, tissue engineering, energy, and catalysis. (iii) Resurgence of phages in biocontrol of plant, food, and animal-borne pathogens. (iv) Commercialization of phage-based products, current challenges, and perspectives.
Collapse
Affiliation(s)
- Rachel Samson
- National Collection of Industrial Microorganisms (NCIM), Biochemical Sciences Division, CSIR-National Chemical Laboratory (NCL), Pune 411008, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India
| | - Mahesh Dharne
- National Collection of Industrial Microorganisms (NCIM), Biochemical Sciences Division, CSIR-National Chemical Laboratory (NCL), Pune 411008, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh 201002, India.
| | - Krishna Khairnar
- National Collection of Industrial Microorganisms (NCIM), Biochemical Sciences Division, CSIR-National Chemical Laboratory (NCL), Pune 411008, India; Environmental Virology Cell (EVC), CSIR-National Environmental Engineering Research Institute (NEERI), Nehru Marg, Nagpur 440020, India.
| |
Collapse
|
13
|
Karn SL, Gangwar M, Kumar R, Bhartiya SK, Nath G. Phage therapy: a revolutionary shift in the management of bacterial infections, pioneering new horizons in clinical practice, and reimagining the arsenal against microbial pathogens. Front Med (Lausanne) 2023; 10:1209782. [PMID: 37928478 PMCID: PMC10620811 DOI: 10.3389/fmed.2023.1209782] [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: 04/21/2023] [Accepted: 10/03/2023] [Indexed: 11/07/2023] Open
Abstract
The recent approval of experimental phage therapies by the FDA and other regulatory bodies with expanded access in cases in the United States and other nations caught the attention of the media and the general public, generating enthusiasm for phage therapy. It started to alter the situation so that more medical professionals are willing to use phage therapies with conventional antibiotics. However, more study is required to fully comprehend phage therapy's potential advantages and restrictions, which is still a relatively new field in medicine. It shows promise, nevertheless, as a secure and prosperous substitute for antibiotics when treating bacterial illnesses in animals and humans. Because of their uniqueness, phage disinfection is excellent for ready-to-eat (RTE) foods like milk, vegetables, and meat products. The traditional farm-to-fork method can be used throughout the food chain to employ bacteriophages to prevent food infections at all production stages. Phage therapy improves clinical outcomes in animal models and lowers bacterial burdens in numerous preclinical investigations. The potential of phage resistance and the need to make sure that enough phages are delivered to the infection site are obstacles to employing phages in vivo. However, according to preclinical studies, phages appear to be a promising alternative to antibiotics for treating bacterial infections in vivo. Phage therapy used with compassion (a profound understanding of and empathy for another's suffering) has recently grown with many case reports of supposedly treated patients and clinical trials. This review summarizes the knowledge on the uses of phages in various fields, such as the food industry, preclinical research, and clinical settings. It also includes a list of FDA-approved bacteriophage-based products, commercial phage products, and a global list of companies that use phages for therapeutic purposes.
Collapse
Affiliation(s)
- Subhash Lal Karn
- Department of Microbiology, Faculty of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Mayank Gangwar
- Department of Microbiology, Faculty of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Rajesh Kumar
- Department of Microbiology, Faculty of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Satyanam Kumar Bhartiya
- Department of General Surgery, Faculty of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Gopal Nath
- Department of Microbiology, Faculty of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
14
|
Singh J, Fitzgerald DA, Jaffe A, Hunt S, Barr JJ, Iredell J, Selvadurai H. Single-arm, open-labelled, safety and tolerability of intrabronchial and nebulised bacteriophage treatment in children with cystic fibrosis and Pseudomonas aeruginosa. BMJ Open Respir Res 2023; 10:e001360. [PMID: 37160359 PMCID: PMC10173968 DOI: 10.1136/bmjresp-2022-001360] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 04/21/2023] [Indexed: 05/11/2023] Open
Abstract
INTRODUCTION Cystic fibrosis (CF) is a multisystem condition that is complicated by recurrent pulmonary infections requiring aggressive antibiotic treatment. This predisposes the patient to complications such as sensorineural hearing loss, renal impairment, hypersensitivity and the development of antibiotic resistance. Pseudomonas aeruginosa is one of the more common organisms which cause recurrent infections and result in greater morbidity and mortality in people living with CF. Bacteriophages have been identified as a potential alternative or adjunct to antibiotics. We hypothesise that bacteriophage therapy is a safe and well-tolerated treatment in children with CF infected with P. aeruginosa infection in their airways. METHODS This single-arm, open-labelled, non-randomised trial will run for a maximum period of 36 months with up to 10 participants. Adolescents (≥12 years and <18 years of age) who continue to shed P.aeruginosa (within 3 months of enrolment) despite undergoing eradication therapy previously, will be considered for this trial. Non-genetically modified bacteriophages that have demonstrated obligate lytic activity against each of the study participants' P. aeruginosa strains will be selected and prepared according to a combination of established protocols (isolation, purification, sterility testing and packaging) to achieve close to good manufacturing practice recommendations. The selected bacteriophage will be administered endo-bronchially first under direct vision, followed by two times a day nebulisation for 7 days in addition to standard CF treatment (intravenous antibiotics, physiotherapy to be completed as inpatient for 10-14 days). Safety and tolerability will be defined as the absence of (1) fever above 38.5°C occurring within 1 hour of the administration of the nebulised bacteriophage, (2) a 10% decline in spirometry (forced expiratory volume in 1 s %) measured preadministration and postadministration of the first dose of nebulised bacteriophage. Clinical reviews including repeat sputum cultures and spirometry will be performed at 3, 6, 9 and 12 months following bacteriophage treatment. ETHICS AND DISSEMINATION Our clinical trial is conducted in accordance with (1) good clinical practice, (2) Australian legislation, (3) National Health and Medical Research Council guidelines for the ethical conduct of research. TRIAL REGISTRATION NUMBER Australia and New Zealand Clinical Trial Registry (ACTRN12622000767707).
Collapse
Affiliation(s)
- Jagdev Singh
- Department of Respiratory Medicine, The Children's Hospital at Westmead, Sydney, New South Wales, Australia
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Dominic A Fitzgerald
- Department of Respiratory Medicine, The Children's Hospital at Westmead, Sydney, New South Wales, Australia
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Adam Jaffe
- School of Women's and Children's Health, Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Sharon Hunt
- Department of Respiratory Medicine, The Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | | | - Jonathan Iredell
- Westmead Institute for Medical Research, Westmead, New South Wales, Australia
- Department of Infectious Diseases, Westmead Hospital, Westmead, New South Wales, Australia
| | - Hiran Selvadurai
- Department of Respiratory Medicine, The Children's Hospital at Westmead, Sydney, New South Wales, Australia
- Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
15
|
Hale SJM, Wagner Mackenzie B, Lux CA, Biswas K, Kim R, Douglas RG. Topical Antibiofilm Agents With Potential Utility in the Treatment of Chronic Rhinosinusitis: A Narrative Review. Front Pharmacol 2022; 13:840323. [PMID: 35770097 PMCID: PMC9234399 DOI: 10.3389/fphar.2022.840323] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
The role of bacterial biofilms in chronic and recalcitrant diseases is widely appreciated, and the treatment of biofilm infection is an increasingly important area of research. Chronic rhinosinusitis (CRS) is a complex disease associated with sinonasal dysbiosis and the presence of bacterial biofilms. While most biofilm-related diseases are associated with highly persistent but relatively less severe inflammation, the presence of biofilms in CRS is associated with greater severity of inflammation and recalcitrance despite appropriate treatment. Oral antibiotics are commonly used to treat CRS but they are often ineffective, due to poor penetration of the sinonasal mucosa and the inherently antibiotic resistant nature of bacteria in biofilms. Topical non-antibiotic antibiofilm agents may prove more effective, but few such agents are available for sinonasal application. We review compounds with antibiofilm activity that may be useful for treating biofilm-associated CRS, including halogen-based compounds, quaternary ammonium compounds and derivatives, biguanides, antimicrobial peptides, chelating agents and natural products. These include preparations that are currently available and those still in development. For each compound, antibiofilm efficacy, mechanism of action, and toxicity as it relates to sinonasal application are summarised. We highlight the antibiofilm agents that we believe hold the greatest promise for the treatment of biofilm-associated CRS in order to inform future research on the management of this difficult condition.
Collapse
Affiliation(s)
- Samuel J M Hale
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Brett Wagner Mackenzie
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Christian A Lux
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Kristi Biswas
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Raymond Kim
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard G Douglas
- Department of Surgery, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
16
|
Chang RYK, Nang SC, Chan HK, Li J. Novel antimicrobial agents for combating antibiotic-resistant bacteria. Adv Drug Deliv Rev 2022; 187:114378. [PMID: 35671882 DOI: 10.1016/j.addr.2022.114378] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/26/2022] [Accepted: 05/26/2022] [Indexed: 12/16/2022]
Abstract
Antibiotic therapy has become increasingly ineffective against bacterial infections due to the rise of resistance. In particular, ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) have caused life-threatening infections in humans and represent a major global health threat due to a high degree of antibiotic resistance. To respond to this urgent call, novel strategies are urgently needed, such as bacteriophages (or phages), phage-encoded enzymes, immunomodulators and monoclonal antibodies. This review critically analyses these promising antimicrobial therapies for the treatment of multidrug-resistant bacterial infections. Recent advances in these novel therapeutic strategies are discussed, focusing on preclinical and clinical investigations, as well as combinatorial approaches. In this 'Bad Bugs, No Drugs' era, novel therapeutic strategies can play a key role in treating deadly infections and help extend the lifetime of antibiotics.
Collapse
|
17
|
Liu S, Hon K, Bouras GS, Psaltis AJ, Shearwin K, Wormald PJ, Vreugde S. APTC-C-SA01: A Novel Bacteriophage Cocktail Targeting Staphylococcus aureus and MRSA Biofilms. Int J Mol Sci 2022; 23:ijms23116116. [PMID: 35682794 PMCID: PMC9181636 DOI: 10.3390/ijms23116116] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 02/06/2023] Open
Abstract
The high infection and mortality rate of methicillin-resistant Staphylococcus aureus (MRSA) necessitates the urgent development of new treatment strategies. Bacteriophages (phages) have several advantages compared to antibiotics for the treatment of multi-drug-resistant bacterial infections, and thus provide a promising alternative to antibiotics. Here, S. aureus phages were isolated from patients and environmental sources. Phages were characterized for stability, morphology and genomic sequence and their bactericidal activity against the biofilm form of methicillin-susceptible Staphylococcus aureus (MSSA) and MRSA was investigated. Four S. aureus phages were isolated and tested against 51 MSSA and MRSA clinical isolates and reference strains. The phages had a broad host range of 82−94% individually and of >98% when combined and could significantly reduce the viability of S. aureus biofilms. The phages had a latent period of ≤20 min and burst size of >11 plaque forming units (PFU)/infected cell. Transmission electron microscopy (TEM) identified phages belonging to the family of Myoviridae. Genomic sequencing indicated the lytic nature of all four phages, with no identified resistance or virulence genes. The 4 phages showed a high complementarity with 49/51 strains (96%) sensitive to at least 2/4 phages tested. Furthermore, the frequency of bacteriophage insensitive mutant (BIM) generation was lower when the phages were combined into the phage cocktail APTC-C-SA01 than for bacteria exposed to each of the phages alone. In conclusion, APTC-C-SA01, containing four lytic S. aureus phages has the potential for further development as a treatment against MSSA and MRSA infections.
Collapse
Affiliation(s)
- Sha Liu
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville, SA 5011, Australia; (S.L.); (K.H.); (G.S.B.); (A.J.P.); (P.-J.W.)
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Karen Hon
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville, SA 5011, Australia; (S.L.); (K.H.); (G.S.B.); (A.J.P.); (P.-J.W.)
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - George Spyro Bouras
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville, SA 5011, Australia; (S.L.); (K.H.); (G.S.B.); (A.J.P.); (P.-J.W.)
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Alkis James Psaltis
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville, SA 5011, Australia; (S.L.); (K.H.); (G.S.B.); (A.J.P.); (P.-J.W.)
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Keith Shearwin
- School of Biological Sciences, The University of Adelaide, Adelaide, SA 5000, Australia;
| | - Peter-John Wormald
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville, SA 5011, Australia; (S.L.); (K.H.); (G.S.B.); (A.J.P.); (P.-J.W.)
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Sarah Vreugde
- Department of Surgery-Otolaryngology Head and Neck Surgery, Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Woodville, SA 5011, Australia; (S.L.); (K.H.); (G.S.B.); (A.J.P.); (P.-J.W.)
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
- Correspondence: ; Tel.: +61-8-8222-6928
| |
Collapse
|
18
|
Bhargava K, Nath G, Bhargava A, Aseri GK, Jain N. Phage therapeutics: from promises to practices and prospectives. Appl Microbiol Biotechnol 2021; 105:9047-9067. [PMID: 34821965 PMCID: PMC8852341 DOI: 10.1007/s00253-021-11695-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 01/09/2023]
Abstract
The rise in multi-drug resistant bacteria and the inability to develop novel antibacterial agents limits our arsenal against infectious diseases. Antibiotic resistance is a global issue requiring an immediate solution, including the development of new antibiotic molecules and other alternative modes of therapy. This article highlights the mechanism of bacteriophage treatment that makes it a real solution for multidrug-resistant infectious diseases. Several case reports identified phage therapy as a potential solution to the emerging challenge of multi-drug resistance. Bacteriophages, unlike antibiotics, have special features, such as host specificity and do not impact other commensals. A new outlook has also arisen with recent advancements in the understanding of phage immunobiology, where phages are repurposed against both bacterial and viral infections. Thus, the potential possibility of phages in COVID-19 patients with secondary bacterial infections has been briefly elucidated. However, significant obstacles that need to be addressed are to design better clinical studies that may contribute to the widespread use of bacteriophage therapy against multi-drug resistant pathogens. In conclusion, antibacterial agents can be used with bacteriophages, i.e. bacteriophage-antibiotic combination therapy, or they can be administered alone in cases when antibiotics are ineffective.Key points• AMR, a consequence of antibiotic generated menace globally, has led to the resurgence of phage therapy as an effective and sustainable solution without any side effects and high specificity against refractory MDR bacterial infections.• Bacteriophages have fewer adverse reactions and can thus be used as monotherapy as well as in conjunction with antibiotics.• In the context of the COVID-19 pandemic, phage therapy may be a viable option.
Collapse
Affiliation(s)
- Kanika Bhargava
- Amity Institute of Microbial Technology, Amity University Rajasthan, Jaipur, 303 002 India
- Department of Microbiology, IMS, Banaras Hindu University, Varanasi, 221005 India
| | - Gopal Nath
- Department of Microbiology, IMS, Banaras Hindu University, Varanasi, 221005 India
| | - Amit Bhargava
- Department of Medicine, Hayes Memorial Hospital, SHUATS, Allahabad, 211007 India
| | - G. K. Aseri
- Amity Institute of Microbial Technology, Amity University Rajasthan, Jaipur, 303 002 India
| | - Neelam Jain
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, 303 002 India
| |
Collapse
|
19
|
Liu S, Zhao Y, Hayes A, Hon K, Zhang G, Bennett C, Hu H, Finnie J, Morales S, Shearwin L, Psaltis AJ, Shearwin K, Wormald P, Vreugde S. Overcoming bacteriophage insensitivity in Staphylococcus aureus using clindamycin and azithromycinat subinhibitory concentrations. Allergy 2021; 76:3446-3458. [PMID: 33930199 DOI: 10.1111/all.14883] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 03/02/2021] [Accepted: 03/18/2021] [Indexed: 01/21/2023]
Abstract
BACKGROUND Staphylococcus aureus is a pathogen of major concern in both acute infections and chronic conditions such as chronic rhinosinusitis (CRS). Bacteriophage (phage) therapy has recently regained interest for its potential to treat infections caused by antibiotic resistant strains including Methicillin Resistant Staphylococcus aureus (MRSA). However, bacteria can adapt and become resistant to phages. The aim of this study is to determine the potential for antibiotics to overcome phage resistance. METHODS The susceptibility of S. aureus clinical isolates (CIs) to phages J-Sa36, Sa83 and Sa87 alone or in combination with protein synthesis inhibitor (PSI) antibiotics clindamycin, azithromycin and erythromycin was assessed using plaque spot assays, minimum inhibitory concentration (MIC) assays, double layer spot assays and resazurin assays. The safety and efficacy of subinhibitory PSI antibiotics in combination with phage was tested in a Sprague Dawley rat model of sinusitis infected with a phage resistant S. aureus CI. RESULTS All three antibiotics at subinhibitory concentrations showed synergy when combined with all 3 phages against S. aureus CIs in planktonic and biofilm form and could sensitize phage-resistant S. aureus to promote phage infection. The combination of topical subinhibitory clindamycin or azithromycin and phage was safe and could eradicate S. aureus sinonasal biofilms in vivo. CONCLUSION Subinhibitory concentrations of PSI antibiotics could sensitize phage-resistant S. aureus and MRSA strains to phages in vitro and in vivo. This data supports the potential use of phage-PSI antibiotic combination therapies, in particular for difficult-to-treat infections with phage-resistant S. aureus and MRSA strains.
Collapse
Affiliation(s)
- Sha Liu
- Department of Surgery‐Otolaryngology Head and Neck Surgery Basil Hetzel Institute for Translational Health Research Central Adelaide Local Health Network Woodville South SA Australia
- Adelaide Medical School The University of Adelaide Adelaide SA Australia
| | - Yin Zhao
- Department of Surgery‐Otolaryngology Head and Neck Surgery Basil Hetzel Institute for Translational Health Research Central Adelaide Local Health Network Woodville South SA Australia
- Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Department of Otolaryngology, Head and Neck Surgery The Second Hospital of Jilin University Changchun China
| | - Andrew Hayes
- Department of Surgery‐Otolaryngology Head and Neck Surgery Basil Hetzel Institute for Translational Health Research Central Adelaide Local Health Network Woodville South SA Australia
- Adelaide Medical School The University of Adelaide Adelaide SA Australia
| | - Karen Hon
- Department of Surgery‐Otolaryngology Head and Neck Surgery Basil Hetzel Institute for Translational Health Research Central Adelaide Local Health Network Woodville South SA Australia
- Adelaide Medical School The University of Adelaide Adelaide SA Australia
| | - Guimin Zhang
- Department of Surgery‐Otolaryngology Head and Neck Surgery Basil Hetzel Institute for Translational Health Research Central Adelaide Local Health Network Woodville South SA Australia
- Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Department of Otolaryngology‐Head and Neck Surgery Tianjin First Center Hospital Tianjin China
| | - Catherine Bennett
- Department of Surgery‐Otolaryngology Head and Neck Surgery Basil Hetzel Institute for Translational Health Research Central Adelaide Local Health Network Woodville South SA Australia
- Adelaide Medical School The University of Adelaide Adelaide SA Australia
| | - Hua Hu
- Department of Surgery‐Otolaryngology Head and Neck Surgery Basil Hetzel Institute for Translational Health Research Central Adelaide Local Health Network Woodville South SA Australia
- Adelaide Medical School The University of Adelaide Adelaide SA Australia
- Department of Otolaryngology, Head and Neck Surgery Shanghai General Hospital Shanghai Jiaotong University Shanghai China
| | - John Finnie
- Discipline of Anatomy and Pathology Adelaide Medical School University of Adelaide Adelaide SA Australia
| | | | - Linda Shearwin
- Department of Molecular and Biomedical Science Adelaide University Adelaide SA Australia
| | - Alkis J. Psaltis
- Department of Surgery‐Otolaryngology Head and Neck Surgery Basil Hetzel Institute for Translational Health Research Central Adelaide Local Health Network Woodville South SA Australia
- Adelaide Medical School The University of Adelaide Adelaide SA Australia
| | - Keith Shearwin
- Department of Molecular and Biomedical Science Adelaide University Adelaide SA Australia
| | - Peter‐John Wormald
- Department of Surgery‐Otolaryngology Head and Neck Surgery Basil Hetzel Institute for Translational Health Research Central Adelaide Local Health Network Woodville South SA Australia
- Adelaide Medical School The University of Adelaide Adelaide SA Australia
| | - Sarah Vreugde
- Department of Surgery‐Otolaryngology Head and Neck Surgery Basil Hetzel Institute for Translational Health Research Central Adelaide Local Health Network Woodville South SA Australia
- Adelaide Medical School The University of Adelaide Adelaide SA Australia
| |
Collapse
|
20
|
Preclinical Development of a Bacteriophage Cocktail for Treating Multidrug Resistant Pseudomonas aeruginosa Infections. Microorganisms 2021; 9:microorganisms9092001. [PMID: 34576896 PMCID: PMC8464757 DOI: 10.3390/microorganisms9092001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/16/2021] [Accepted: 09/16/2021] [Indexed: 01/21/2023] Open
Abstract
A Pseudomonas aeruginosa (P. aeruginosa) airway infection is one of the predominant causes contributing to the high morbidity and mortality rates in cystic fibrosis (CF) patients. The emergence of antibiotic resistant P. aeruginosa strains has led to an urgent need for new therapeutic approaches. Bacteriophages (phages) are viruses that can infect and lyse specific bacteria, providing a potential alternative approach in targeting antibiotic-resistant strains. We aim to isolate and characterise novel P. aeruginosa phages for combination in a cocktail to kill P. aeruginosa. One particular phage, PA4, could lyse 14/20 clinical isolates as observed through spot assays. This phage could significantly reduce the growth of bacteria in vitro, as determined through planktonic adsorption and inhibition assays as well as crystal violet- and LIVE/DEAD-stained biofilm assays. A morphological and genomic analysis revealed that PA4 belongs to the Myoviridae family and contained 66,450 bp. The broad infectivity profile, good stability in various pH and temperature conditions, lytic ability and the absence of the absences of antibiotic resistance, toxic and lysogenic genes suggest that PA4 is a good candidate for clinical grade use. Overall, phage therapy represents a promising alternative treatment option to antibiotics when treating a P. aeruginosa infection.
Collapse
|
21
|
Liu D, Van Belleghem JD, de Vries CR, Burgener E, Chen Q, Manasherob R, Aronson JR, Amanatullah DF, Tamma PD, Suh GA. The Safety and Toxicity of Phage Therapy: A Review of Animal and Clinical Studies. Viruses 2021; 13:1268. [PMID: 34209836 PMCID: PMC8310247 DOI: 10.3390/v13071268] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/23/2021] [Accepted: 06/23/2021] [Indexed: 12/13/2022] Open
Abstract
Increasing rates of infection by antibiotic resistant bacteria have led to a resurgence of interest in bacteriophage (phage) therapy. Several phage therapy studies in animals and humans have been completed over the last two decades. We conducted a systematic review of safety and toxicity data associated with phage therapy in both animals and humans reported in English language publications from 2008-2021. Overall, 69 publications met our eligibility criteria including 20 animal studies, 35 clinical case reports or case series, and 14 clinical trials. After summarizing safety and toxicity data from these publications, we discuss potential approaches to optimize safety and toxicity monitoring with the therapeutic use of phage moving forward. In our systematic review of the literature, we found some adverse events associated with phage therapy, but serious events were extremely rare. Comprehensive and standardized reporting of potential toxicities associated with phage therapy has generally been lacking in the published literature. Structured safety and tolerability endpoints are necessary when phages are administered as anti-infective therapeutics.
Collapse
Affiliation(s)
- Dan Liu
- Department of Burn, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China;
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (J.D.V.B.); (C.R.d.V.); (Q.C.); (J.R.A.)
| | - Jonas D. Van Belleghem
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (J.D.V.B.); (C.R.d.V.); (Q.C.); (J.R.A.)
| | - Christiaan R. de Vries
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (J.D.V.B.); (C.R.d.V.); (Q.C.); (J.R.A.)
| | - Elizabeth Burgener
- Center for Excellence in Pulmonary Biology, Department of Pediatrics, Stanford University, Stanford, CA 94305, USA;
| | - Qingquan Chen
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (J.D.V.B.); (C.R.d.V.); (Q.C.); (J.R.A.)
| | - Robert Manasherob
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA 94305, USA; (R.M.); (D.F.A.)
| | - Jenny R. Aronson
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA; (J.D.V.B.); (C.R.d.V.); (Q.C.); (J.R.A.)
| | - Derek F. Amanatullah
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA 94305, USA; (R.M.); (D.F.A.)
| | - Pranita D. Tamma
- Division of Pediatric Infectious Diseases, Department of Pediatrics, School of Medicine, Johns Hopkins University, Baltimore, MD 21287, USA;
| | - Gina A. Suh
- Division of Infectious Diseases, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
22
|
Phages in Food Industry Biocontrol and Bioremediation. Antibiotics (Basel) 2021; 10:antibiotics10070786. [PMID: 34203362 PMCID: PMC8300737 DOI: 10.3390/antibiotics10070786] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 12/25/2022] Open
Abstract
Bacteriophages are ubiquitous in nature and their use is a current promising alternative in biological control. Multidrug resistant (MDR) bacterial strains are present in the livestock industry and phages are attractive candidates to eliminate them and their biofilms. This alternative therapy also reduces the non-desirable effects produced by chemicals on food. The World Health Organization (WHO) estimates that around 420,000 people die due to a foodborne illness annually, suggesting that an improvement in food biocontrol is desirable. This review summarizes relevant studies of phage use in biocontrol focusing on treatments in live animals, plants, surfaces, foods, wastewaters and bioremediation.
Collapse
|
23
|
Cristobal-Cueto P, García-Quintanilla A, Esteban J, García-Quintanilla M. Phages in Food Industry Biocontrol and Bioremediation. Antibiotics (Basel) 2021; 10:antibiotics10070786. [PMID: 34203362 DOI: 10.3390/antibiotic6as10070786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 05/28/2023] Open
Abstract
Bacteriophages are ubiquitous in nature and their use is a current promising alternative in biological control. Multidrug resistant (MDR) bacterial strains are present in the livestock industry and phages are attractive candidates to eliminate them and their biofilms. This alternative therapy also reduces the non-desirable effects produced by chemicals on food. The World Health Organization (WHO) estimates that around 420,000 people die due to a foodborne illness annually, suggesting that an improvement in food biocontrol is desirable. This review summarizes relevant studies of phage use in biocontrol focusing on treatments in live animals, plants, surfaces, foods, wastewaters and bioremediation.
Collapse
Affiliation(s)
- Pablo Cristobal-Cueto
- Department of Clinical Microbiology, IIS-Fundación Jiménez Díaz, Av. Reyes Católicos, 2, 28040 Madrid, Spain
| | - Alberto García-Quintanilla
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Seville, Calle Profesor García Gonzalez, 2, 41012 Seville, Spain
| | - Jaime Esteban
- Department of Clinical Microbiology, IIS-Fundación Jiménez Díaz, Av. Reyes Católicos, 2, 28040 Madrid, Spain
| | | |
Collapse
|
24
|
Uyttebroek S, Onsea J, Metsemakers WJ, Dupont L, Devolder D, Wagemans J, Lavigne R, Spriet I, Van Gerven L. The Potential Role of Bacteriophages in the Treatment of Recalcitrant Chronic Rhinosinusitis. Antibiotics (Basel) 2021; 10:antibiotics10060675. [PMID: 34198741 PMCID: PMC8229554 DOI: 10.3390/antibiotics10060675] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/26/2021] [Accepted: 06/02/2021] [Indexed: 01/28/2023] Open
Abstract
Chronic rhinosinusitis is a common condition affecting 5-12% of the general population worldwide. In a limited number of cases, the disease is recalcitrant to medical and surgical interventions, causing a major impact on physical, social and emotional well-being and increasing pressure on healthcare systems. Biofilm formation and dysbiosis caused by Staphylococcus aureus and Pseudomonas aeruginosa play a role in the pathogenesis of recalcitrant chronic rhinosinusitis. In these cases, a promising treatment alternative is the application of bacteriophages, which are viruses that infect and lyse bacteria. In this review, we appraise the evidence for the use of bacteriophages in the treatment of recalcitrant chronic rhinosinusitis. Additionally, (dis)advantages of bacteriophages and considerations for implementation of phage therapy in otorhinolaryngology practice will be discussed.
Collapse
Affiliation(s)
- Saartje Uyttebroek
- Department of Otorhinolaryngology, UZ Leuven, 3000 Leuven, Belgium;
- Department of Neurosciences, Experimental Otorhinolaryngology, Rhinology Research, KU Leuven, 3000 Leuven, Belgium
| | - Jolien Onsea
- Department of Trauma Surgery, UZ Leuven, 3000 Leuven, Belgium; (J.O.); (W.-J.M.)
- Department of Development and Regeneration, Locomotor and Neurological Disorders, KU Leuven, 3000 Leuven, Belgium
| | - Willem-Jan Metsemakers
- Department of Trauma Surgery, UZ Leuven, 3000 Leuven, Belgium; (J.O.); (W.-J.M.)
- Department of Development and Regeneration, Locomotor and Neurological Disorders, KU Leuven, 3000 Leuven, Belgium
| | - Lieven Dupont
- Department of Pneumology, UZ Leuven, 3000 Leuven, Belgium;
- Department of Chronic Diseases and Metabolism, Respiratory Diseases and Thoracic Surgery, KU Leuven, 3000 Leuven, Belgium
| | - David Devolder
- Pharmacy Department, UZ Leuven, 3000 Leuven, Belgium; (D.D.); (I.S.)
| | - Jeroen Wagemans
- Department of Biosystems, Laboratory of Gene Technology, KU Leuven, 3000 Leuven, Belgium; (J.W.); (R.L.)
| | - Rob Lavigne
- Department of Biosystems, Laboratory of Gene Technology, KU Leuven, 3000 Leuven, Belgium; (J.W.); (R.L.)
| | - Isabel Spriet
- Pharmacy Department, UZ Leuven, 3000 Leuven, Belgium; (D.D.); (I.S.)
- Department of Pharmaceutical and Pharmacological Sciences, Clinical Pharmacology and Pharmacotherapy, KU Leuven, 3000 Leuven, Belgium
| | - Laura Van Gerven
- Department of Otorhinolaryngology, UZ Leuven, 3000 Leuven, Belgium;
- Department of Neurosciences, Experimental Otorhinolaryngology, Rhinology Research, KU Leuven, 3000 Leuven, Belgium
- Department of Microbiology, Immunology and Transplantation, Allergy and Clinical Immunology Research Unit, KU Leuven, 3000 Leuven, Belgium
- Correspondence: ; Tel.: +32-16-33-63-41
| |
Collapse
|
25
|
Pinto AM, Silva MD, Pastrana LM, Bañobre-López M, Sillankorva S. The clinical path to deliver encapsulated phages and lysins. FEMS Microbiol Rev 2021; 45:6204673. [PMID: 33784387 DOI: 10.1093/femsre/fuab019] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 03/26/2021] [Indexed: 02/06/2023] Open
Abstract
The global emergence of multidrug-resistant pathogens is shaping the current dogma regarding the use of antibiotherapy. Many bacteria have evolved to become resistant to conventional antibiotherapy, representing a health and economic burden for those afflicted. The search for alternative and complementary therapeutic approaches has intensified and revived phage therapy. In recent decades, the exogenous use of lysins, encoded in phage genomes, has shown encouraging effectiveness. These two antimicrobial agents reduce bacterial populations; however, many barriers challenge their prompt delivery at the infection site. Encapsulation in delivery vehicles provides targeted therapy with a controlled compound delivery, surpassing chemical, physical and immunological barriers that can inactivate and eliminate them. This review explores phages and lysins' current use to resolve bacterial infections in the respiratory, digestive, and integumentary systems. We also highlight the different challenges they face in each of the three systems and discuss the advances towards a more expansive use of delivery vehicles.
Collapse
Affiliation(s)
- Ana Mafalda Pinto
- Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, 4710-057 Braga, Portugal.,INL - International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, Braga 4715-330, Portugal
| | - Maria Daniela Silva
- Centre of Biological Engineering, LIBRO - Laboratório de Investigação em Biofilmes Rosário Oliveira, University of Minho, 4710-057 Braga, Portugal.,INL - International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, Braga 4715-330, Portugal
| | - Lorenzo M Pastrana
- INL - International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, Braga 4715-330, Portugal
| | - Manuel Bañobre-López
- INL - International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, Braga 4715-330, Portugal
| | - Sanna Sillankorva
- INL - International Iberian Nanotechnology Laboratory, Avenida Mestre José Veiga, Braga 4715-330, Portugal
| |
Collapse
|
26
|
Tian F, Li J, Nazir A, Tong Y. Bacteriophage - A Promising Alternative Measure for Bacterial Biofilm Control. Infect Drug Resist 2021; 14:205-217. [PMID: 33505163 PMCID: PMC7829120 DOI: 10.2147/idr.s290093] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 12/23/2020] [Indexed: 01/09/2023] Open
Abstract
Bacterial biofilms can enhance bacteria's viability by providing resistance against antibiotics and conventional disinfectants. The existence of biofilm is a serious threat to human health, causing incalculable loss. Therefore, new strategies to deal with bacterial biofilms are needed. Bacteriophages are unique due to their activity on bacteria and do not pose a threat to humans. Consequently, they are considered safe alternatives to drugs for the treatment of bacterial diseases. They can effectively obliterate bacterial biofilms and have great potential in medical treatment, the food industry, and pollution control. There are intricate mechanisms of interaction between phages and biofilms. Biofilms may prevent the invasion of phages, and phages can kill bacteria for biofilm control purposes or influence the formation of biofilms. At present, there are various measures for the prevention and control of biofilms through phages, including the combined use of drugs and the application of phage cocktails. This article mainly reviews the function and formation process of bacterial biofilms, summarizes the different mechanisms between phages and biofilms, briefly explains the phage usage for the control of bacterial biofilms, and promotes phage application maintenance human health and the protection of the natural environment.
Collapse
Affiliation(s)
- Fengjuan Tian
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, People’s Republic of China
| | - Jing Li
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, People’s Republic of China
| | - Amina Nazir
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, People’s Republic of China
| | - Yigang Tong
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, People’s Republic of China
| |
Collapse
|
27
|
Comprehensive Evaluation of the Safety and Efficacy of BAFASAL ® Bacteriophage Preparation for the Reduction of Salmonella in the Food Chain. Viruses 2020; 12:v12070742. [PMID: 32664206 PMCID: PMC7412135 DOI: 10.3390/v12070742] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/06/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
Bacteriophages are bacterial predators, which are garnering much interest nowadays vis-à-vis the global phenomenon of antimicrobial resistance. Bacteriophage preparations seem to be an alternative to antibiotics, which can be used at all levels of the food production chain. Their safety and efficacy, however, are of public concern. In this study, a detailed evaluation of BAFASAL® preparation was performed. BAFASAL® is a bacteriophage cocktail that reduces Salmonella in poultry farming. In vivo acute and sub-chronic toxicity studies on rats and tolerance study on targeted animals (chicken broiler) conducted according to GLP and OECD guidelines did not reveal any signs of toxicity, which could be associated with BAFASAL® administration. In addition, no evidences of genotoxicity were observed. The tolerance study with 100-times concentrated dose also did not show any statistically significant differences in the assessed parameters. The in vitro crop assay, mimicking normal feed storage and feed application conditions showed that BAFASAL® reduced the number of Salmonella bacteria in experimentally contaminated feed. Moreover, reductions were observed for all examined forms (liquid, powder, spray). Furthermore, the in vivo efficacy study showed that treatment with BAFASAL® significantly decreased Salmonella content in caeca of birds infected with Salmonella Enteritidis. Detailed examination of BAFASAL® in terms of safety and efficacy, adds to the body of evidence that bacteriophages are harmless to animals and effective in the struggle against bacteria.
Collapse
|
28
|
Maddocks S, Fabijan AP, Ho J, Lin RCY, Ben Zakour NL, Dugan C, Kliman I, Branston S, Morales S, Iredell JR. Bacteriophage Therapy of Ventilator-associated Pneumonia and Empyema Caused by Pseudomonas aeruginosa. Am J Respir Crit Care Med 2020; 200:1179-1181. [PMID: 31437402 DOI: 10.1164/rccm.201904-0839le] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Susan Maddocks
- Westmead HospitalWestmead, New South Wales, Australia.,Westmead Institute for Medical ResearchSydney, New South Wales, Australia.,University of SydneySydney, New South Wales, Australia
| | | | - Josephine Ho
- Westmead Institute for Medical ResearchSydney, New South Wales, Australia.,University of New South WalesSydney, New South Wales, Australiaand
| | - Ruby C Y Lin
- Westmead Institute for Medical ResearchSydney, New South Wales, Australia.,University of SydneySydney, New South Wales, Australia.,AmpliPhi Biosciences CorporationSydney, New South Wales, Australia
| | - Nouri L Ben Zakour
- Westmead Institute for Medical ResearchSydney, New South Wales, Australia.,University of SydneySydney, New South Wales, Australia
| | - Chris Dugan
- Westmead HospitalWestmead, New South Wales, Australia
| | - Ivana Kliman
- Westmead HospitalWestmead, New South Wales, Australia
| | - Steven Branston
- AmpliPhi Biosciences CorporationSydney, New South Wales, Australia
| | - Sandra Morales
- AmpliPhi Biosciences CorporationSydney, New South Wales, Australia
| | - Jonathan R Iredell
- Westmead HospitalWestmead, New South Wales, Australia.,Westmead Institute for Medical ResearchSydney, New South Wales, Australia.,University of SydneySydney, New South Wales, Australia
| |
Collapse
|
29
|
Melo LDR, Oliveira H, Pires DP, Dabrowska K, Azeredo J. Phage therapy efficacy: a review of the last 10 years of preclinical studies. Crit Rev Microbiol 2020; 46:78-99. [DOI: 10.1080/1040841x.2020.1729695] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Luís D. R. Melo
- CEB – Centre of Biological Engineering, University of Minho, Braga, Portugal
| | - Hugo Oliveira
- CEB – Centre of Biological Engineering, University of Minho, Braga, Portugal
| | - Diana P. Pires
- CEB – Centre of Biological Engineering, University of Minho, Braga, Portugal
| | - Krystyna Dabrowska
- Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Joana Azeredo
- CEB – Centre of Biological Engineering, University of Minho, Braga, Portugal
| |
Collapse
|
30
|
Latest developments on topical therapies in chronic rhinosinusitis. Curr Opin Otolaryngol Head Neck Surg 2020; 28:25-30. [DOI: 10.1097/moo.0000000000000598] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
31
|
Caflisch KM, Suh GA, Patel R. Biological challenges of phage therapy and proposed solutions: a literature review. Expert Rev Anti Infect Ther 2019; 17:1011-1041. [PMID: 31735090 DOI: 10.1080/14787210.2019.1694905] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: In light of the emergence of antibiotic-resistant bacteria, phage (bacteriophage) therapy has been recognized as a potential alternative or addition to antibiotics in Western medicine for use in humans.Areas covered: This review assessed the scientific literature on phage therapy published between 1 January 2007 and 21 October 2019, with a focus on the successes and challenges of this prospective therapeutic.Expert opinion: Efficacy has been shown in animal models and experimental findings suggest promise for the safety of human phagotherapy. Significant challenges remain to be addressed prior to the standardization of phage therapy in the West, including the development of phage-resistant bacteria; the pharmacokinetic complexities of phage; and any potential human immune response incited by phagotherapy.
Collapse
Affiliation(s)
- Katherine M Caflisch
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Gina A Suh
- Division of Infectious Diseases, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Robin Patel
- Division of Infectious Diseases, Department of Medicine, Mayo Clinic, Rochester, MN, USA.,Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
32
|
Affiliation(s)
- Juliet Roshini Mohan Raj
- Nitte University Centre for Science Education and Research, Nitte (Deemed to be University), Mangaluru, India
| | - Indrani Karunasagar
- Nitte University Centre for Science Education and Research, Nitte (Deemed to be University), Mangaluru, India
| |
Collapse
|
33
|
Szaleniec J, Gibała A, Pobiega M, Parasion S, Składzień J, Stręk P, Gosiewski T, Szaleniec M. Exacerbations of Chronic Rhinosinusitis-Microbiology and Perspectives of Phage Therapy. Antibiotics (Basel) 2019; 8:antibiotics8040175. [PMID: 31590369 PMCID: PMC6963383 DOI: 10.3390/antibiotics8040175] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/01/2019] [Accepted: 10/01/2019] [Indexed: 11/16/2022] Open
Abstract
The chronically inflamed mucosa in patients with chronic rhinosinusitis (CRS) can additionally be infected by bacteria, which results in an acute exacerbation of the disease (AECRS). Currently, AECRS is universally treated with antibiotics following the guidelines for acute bacterial rhinosinusitis (ABRS), as our understanding of its microbiology is insufficient to establish specific treatment recommendations. Unfortunately, antibiotics frequently fail to control the symptoms of AECRS due to biofilm formation, disruption of the natural microbiota, and arising antibiotic resistance. These issues can potentially be addressed by phage therapy. In this study, the endoscopically-guided cultures were postoperatively obtained from 50 patients in order to explore the microbiology of AECRS, evaluate options for antibiotic treatment, and, most importantly, assess a possibility of efficient phage therapy. Staphylococcus aureus and coagulase-negative staphylococci were the most frequently isolated bacteria, followed by Haemophilus influenzae, Pseudomonas aeruginosa, and Enterobacteriaceae. Alarmingly, mechanisms of antibiotic resistance were detected in the isolates from 46% of the patients. Bacteria not sensitive to amoxicillin were carried by 28% of the patients. The lowest rates of resistance were noted for fluoroquinolones and aminoglycosides. Fortunately, 60% of the patients carried bacterial strains that were sensitive to bacteriophages from the Biophage Pharma collection and 81% of the antibiotic-resistant strains turned out to be sensitive to bacteriophages. The results showed that microbiology of AECRS is distinct from ABRS and amoxicillin should not be the antibiotic of first choice. Currently available bacteriophages could be used instead of antibiotics or as an adjunct to antibiotics in the majority of patients with AECRS.
Collapse
Affiliation(s)
- Joanna Szaleniec
- Department of Otolaryngology, Faculty of Medicine, Jagiellonian University Medical College, Sniadeckich 2, 31-531 Krakow, Poland.
| | - Agnieszka Gibała
- Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, Niezapominajek 8, 30-239 Krakow, Poland.
- Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Cracow Branch, Garncarska 11, 31-115 Krakow, Poland.
| | - Monika Pobiega
- Biophage Pharma S.A., Mogilska 40, 31-546 Krakow, Poland.
| | | | - Jacek Składzień
- Department of Otolaryngology, Faculty of Medicine, Jagiellonian University Medical College, Sniadeckich 2, 31-531 Krakow, Poland.
| | - Paweł Stręk
- Department of Otolaryngology, Faculty of Medicine, Jagiellonian University Medical College, Sniadeckich 2, 31-531 Krakow, Poland.
| | - Tomasz Gosiewski
- Chair of Microbiology, Department of Molecular Medical Microbiology Unit, Faculty of Medicine, Jagiellonian University Medical College, Czysta 18, 31-121 Krakow, Poland.
| | - Maciej Szaleniec
- Jerzy Haber Institute of Catalysis and Surface Chemistry Polish Academy of Sciences, Niezapominajek 8, 30-239 Krakow, Poland.
| |
Collapse
|
34
|
Dapgh AN, Hakim AS, Abouelhag HA, Abdou AM, Elgabry EA. Detection of virulence and multidrug resistance operons in Pseudomonas aeruginosa isolated from Egyptian Baladi sheep and goat. Vet World 2019; 12:1524-1528. [PMID: 31849411 PMCID: PMC6868267 DOI: 10.14202/vetworld.2019.1524-1528] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/26/2019] [Indexed: 11/24/2022] Open
Abstract
Background: Pseudomonas aeruginosa is a pit of an enormous group of free-living bacteria that are able to live everywhere and suggested to be the causative agent of great scope of acute and chronic animal infections. Aim: The current study was carried out to illustrate the prevalence of P. aeruginosa in small ruminants and existence of some virulence operons as well as its antimicrobial resistance. Materials and Methods: A total of 155 samples from sheep and 105 samples from goats (mouth abscesses, fecal swabs, nasal, tracheal swabs, and lung tissue) were collected for bacteriological study, existence of some virulence expression operons with the study of their sensitivity to the antimicrobials using disc diffusion and presence of mexR operon which is responsible for multidrug resistance (MDR). Results: The bacteriological examination revealed that P. aeruginosa was isolated from nine out of 155 samples from sheep (5.8%) and four isolates out of 105 samples from goat (3.8%). It is found that 12 (92.3%), 10 (76.9 %), and 8 (61.5%) of P. aeruginosa isolates harbored hemolysin phospholipase gene (pcl H), gene (exo S), and enterotoxin gene (tox A), respectively. The results of antibiotic sensitivity test showed that all tested isolates were resistant to ampicillin, bacitracin, erythromycin, streptomycin, tetracycline, trimethoprim-sulfamethoxazole, and tobramycin but sensitive to ciprofloxacin and norfloxacin. The MDR (mex R) operon was existed in all isolates. Conclusion: There is a growing risk for isolation of virulent MDR P. aeruginosa from sheep and goat illness cases, and this should be regarded in the efficient control programs.
Collapse
Affiliation(s)
- A N Dapgh
- Department of Bacteriology, Animal Health Research Institute, Dokki, Giza, Egypt
| | - A S Hakim
- Department of Microbiology and Immunology, National Research Centre, 33 Bohouth Street, 12622 Dokki, Cairo, Egypt
| | - H A Abouelhag
- Department of Microbiology and Immunology, National Research Centre, 33 Bohouth Street, 12622 Dokki, Cairo, Egypt
| | - A M Abdou
- Department of Microbiology and Immunology, National Research Centre, 33 Bohouth Street, 12622 Dokki, Cairo, Egypt
| | - E A Elgabry
- Department of Microbiology and Immunology, National Research Centre, 33 Bohouth Street, 12622 Dokki, Cairo, Egypt
| |
Collapse
|