1
|
Ray D, Bose P, Mukherjee S, Roy S, Kaity S. Recent drug delivery systems targeting the gut-brain-microbiome axis for the management of chronic diseases. Int J Pharm 2025; 680:125776. [PMID: 40425058 DOI: 10.1016/j.ijpharm.2025.125776] [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: 02/25/2025] [Revised: 05/14/2025] [Accepted: 05/24/2025] [Indexed: 05/29/2025]
Abstract
In recent years, the study of microorganisms and the brain has become increasingly connected. The gut-brain-microbiome axis (GBMA), a bi-directional communication system, is the key part of how the body's bacteria and the brain interact. This system can influence the brain and behaviour. Changes in this relationship have been linked to various mental and physical health conditions. The immune system, tryptophan metabolism, the vagus nerve, and the enteric nervous system all facilitate connections between the gut and brain. Microbes produce Peptidoglycans, branched-chain amino acids, and short-chain fatty acids, which are involved in this communication. Studies suggest the gut microbiome may be involved in conditions like autism, anxiety, obesity, schizophrenia, Parkinson's disease, and Alzheimer's disease. Researchers are exploring the gut-brain connection to cure a variety of disorders, such as neurological disorders, cancers, metabolic problems, and liver diseases. Developing novel drug delivery systems is a key focus in GBMA for therapeutic targeting at various disease pathways. Notable platforms attracting significant interest include silica nanoparticle-based delivery systems for probiotic spores, composite hydrogels formulated from protein isolates and citrus pectin, and biomimetic nanosystems designed for targeted therapeutic delivery. This review summarizes different methods of delivering drugs and using dietary interventions to target the GBMA and treat these conditions in a less invasive way. By understanding how the gut and brain communicate, scientists aim to develop new and more effective therapies for these complex chronic diseases.
Collapse
Affiliation(s)
- Debjani Ray
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, West Bengal, India
| | - Piyas Bose
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, West Bengal, India
| | - Saptarshi Mukherjee
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, West Bengal, India
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, West Bengal, India; Division of Pharmacology, Department of Pharmaceutical Sciences and Technology, BIT, Mesra, Ranchi, India
| | - Santanu Kaity
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Kolkata, West Bengal, India.
| |
Collapse
|
2
|
Zou H, Huang X, Xiao W, He H, Liu S, Zeng H. Recent advancements in bacterial anti-phage strategies and the underlying mechanisms altering susceptibility to antibiotics. Microbiol Res 2025; 295:128107. [PMID: 40023108 DOI: 10.1016/j.micres.2025.128107] [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: 11/12/2024] [Revised: 02/12/2025] [Accepted: 02/16/2025] [Indexed: 03/04/2025]
Abstract
The rapid spread of multidrug-resistant bacteria and the challenges in developing new antibiotics have brought renewed international attention to phage therapy. However, in bacteria-phage co-evolution, the rapid development of bacterial resistance to phage has limited its clinical application. This review consolidates the latest advancements in research on anti-phage mechanisms, encompassing strategies such as systems associated with reduced nicotinamide adenine dinucleotide (NAD+) to halt the propagation of the phage, symbiotic bacteria episymbiont-mediated modulation of gene expression in host bacteria to resist phage infection, and defence-related reverse transcriptase (DRT) encoded by bacteria to curb phage infections. We conduct an in-depth analysis of the underlying mechanisms by which bacteria undergo alterations in antibiotic susceptibility after developing phage resistance. We also discuss the remaining challenges and promising directions for phage-based therapy in the future.
Collapse
Affiliation(s)
- Huanhuan Zou
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Xiaoyi Huang
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Wenyue Xiao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Haoxuan He
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Shenshen Liu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Haiyan Zeng
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| |
Collapse
|
3
|
Ghosh S, Pradhan S, Ghosh K. Mini-review: Insight of bacteriophage therapy in clinical practice. Virology 2025; 610:110583. [PMID: 40424677 DOI: 10.1016/j.virol.2025.110583] [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: 10/08/2024] [Revised: 05/15/2025] [Accepted: 05/20/2025] [Indexed: 05/29/2025]
Abstract
The emergence of drug-resistant microorganisms requires implementing alternative therapy rather than antibiotics. Phage therapy is a fantastic substitute for antibiotics. Compared to antibiotics, phage therapy has many benefits, such as high specificity for the target bacteria, auto-dosing, biofilm penetration, and a decreased likelihood of resistance development. Regulatory issues, manufacturing barriers, the possibility of phage resistance, and interactions with the human immune system are only a few of the major obstacles that still exist. Various phage-derived enzymes and bioengineered phages may increase the therapeutic potential to combat antibiotic-resistant infections. This mini-review is compiled from research on phage mechanisms in mammalian immune systems, therapeutic uses, regulatory issues, and phage engineering advancements. Thus, it offers a hopeful future in phage therapy by offering a thorough overview of the therapeutic potentiality of phage and the global aspects of phage therapy. In conclusion, phages are expected to become an alternative treatment for antibiotics against multidrug-resistant (MDR) bacteria for medical purposes.
Collapse
Affiliation(s)
- Smita Ghosh
- Department of Biological Sciences, Midnapore City College, Midnapore, West Bengal, India; Biodiversity and Environmental Studies Research Center, Midnapore City College Affiliated to Vidyasagar University, Midnapore, West Bengal, India
| | - Shrabani Pradhan
- Department of Paramedical and Allied Health Sciences, Midnapore City College, Midnapore, West Bengal, India
| | - Kuntal Ghosh
- Department of Biological Sciences, Midnapore City College, Midnapore, West Bengal, India.
| |
Collapse
|
4
|
Morris TC, Reyneke B, Khan S, Khan W. Phage-antibiotic synergy to combat multidrug resistant strains of Gram-negative ESKAPE pathogens. Sci Rep 2025; 15:17235. [PMID: 40383795 PMCID: PMC12086229 DOI: 10.1038/s41598-025-01489-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 05/06/2025] [Indexed: 05/20/2025] Open
Abstract
Bacteriophage-antibiotic-synergy (PAS) was investigated to target Pseudomonas aeruginosa, Klebsiella pneumoniae, Acinetobacter baumannii and Enterobacter cloacae. Whole genome sequencing indicated that bacteriophage KPW17 targeting K. pneumoniae, clustered with genus Webervirus, ECSR5 targeting E. cloacae clustered with Eclunavirus, PAW33 targeting P. aeruginosa clustered with Bruynoghevirus, while ABTW1 targeting A. baumannii clustered with Vieuvirus. PAS analysis showed that the combination of ciprofloxacin (CIP) and levofloxacin (LEV) with PAW33 resulted in the synergistic eradication of all tested P. aeruginosa strains. Similarly, the combined use of doripenem (DOR) and LEV with KPW17 resulted in the synergistic eradication of the environmental and clinical K. pneumoniae strains, while the combined use of DOR and gentamicin (CN) with ECSR5 was synergistic against the clinical E. cloacae NCTC 13406. Gentamicin with ECSR5, however, only exhibited an additive effect for E. cloacae 4L, while ABTW1 with piperacillin-tazobactam (TZP) and imipenem (IPM) resulted in an indifferent interaction between the bacteriophage and tested antibiotics against the clinical A. baumannii AB3, i.e., the activity of the combination is equal to the activity of most active agent. Thus, while the observed PAS may offer an opportunity for the re-introduction or more efficient application of certain antibiotics to combat antibiotic resistance, extensive research is required to determine the optimal phage-antibiotic combinations, dosages and treatment regiments.
Collapse
Affiliation(s)
- Tinta Carmen Morris
- Department of Microbiology, Faculty of Science, Stellenbosch University, Private Bag X1, Stellenbosch, 7602, South Africa
| | - Brandon Reyneke
- Department of Microbiology, Faculty of Science, Stellenbosch University, Private Bag X1, Stellenbosch, 7602, South Africa
| | - Sehaam Khan
- Faculty of Health Science, University of Johannesburg, PO Box 17011, Doornfontein, 2028, South Africa
| | - Wesaal Khan
- Department of Microbiology, Faculty of Science, Stellenbosch University, Private Bag X1, Stellenbosch, 7602, South Africa.
| |
Collapse
|
5
|
Zhou F, Wang K, Ji S, Liao X, Zhang W, Teng T, Wang L, Li Q. The virulent bacteriophage Henu8 as an antimicrobial synergist against Escherichia coli. Microbiol Spectr 2025:e0163324. [PMID: 40377308 DOI: 10.1128/spectrum.01633-24] [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: 07/03/2024] [Accepted: 04/02/2025] [Indexed: 05/18/2025] Open
Abstract
As the overuse of antibiotics has not yet been strictly limited in urban areas, drug-resistant Escherichia coli has become a fatal pressure for bacteremia treatment. Considering the outstanding performance of bacteriophages in vitro, bacteriophages may serve as an alternative to heal chronic refractory infections. In this study, a 49,890 bp double-stranded circular DNA phage, Henu8, was isolated and was able to lyse the group of E. coli strains tested in this study. Prominent biological characterization revealed that the highly adsorbed bacteriophage Henu8 could form a fully transparent plaque with a narrow translucent halo. The optimal multiplicity of infection of the bacteriophage Henu8 was 0.01, with a burst size of 275 PFU/cell. Genomic analysis revealed a G + C content of 44.17% Henu8, in which 65 open reading frames were located, which could be assigned as a new species in the genus Hanrivervirus of the subfamily Tempevirinae. The effective antibacterial ability and the obvious biofilm destruction and inhibition capability of phage Henu8 were observed. The time-killing assay demonstrated the synergetic potential of Henu8 with antibiotics in vitro for E. coli eradication. Henu8 has profound medicinal potential in a mouse bacteremia model. These studies indicate that Henu8 is a novel bacteriophage with therapeutic potential alone or in combination with antibiotics for clinical treatment.IMPORTANCEThe findings described in this study constitute concrete evidence that it is possible to significantly synergize the antimicrobial activity of bacteriophages and antibiotics. We showed that the newly isolated potent bacteriophage Henu8 lyses Escherichia coli rapidly but tends to produce resistant bacteria. The bacteriophage Henu8 has synergistic antimicrobial effects with several antibiotics and is not susceptible to developing resistance. These results provide further evidence that bacterial resistance to phages arises, possibly at an adaptive cost to sensitivity to antibiotics. Therefore, the findings of this study are important for increasing the potential of phages for clinical applications and developing new approaches to improve their therapeutic efficacy against bacterial drug resistance.
Collapse
Affiliation(s)
- Fang Zhou
- Henan Province Engineering Technology Research Center of Rapid-Accuracy Medical Diagnostics, Department of Clinical Laboratory, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, China
| | - Kexiao Wang
- Henan Province Engineering Technology Research Center of Rapid-Accuracy Medical Diagnostics, Department of Clinical Laboratory, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, China
| | - Shuai Ji
- The Joint National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng, China
| | - Xiaochen Liao
- The Joint National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng, China
| | - Wenwen Zhang
- Department of Microbiology, College of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Tieshan Teng
- Henan Province Engineering Technology Research Center of Rapid-Accuracy Medical Diagnostics, Department of Clinical Laboratory, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, China
- Department of Microbiology, College of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Li Wang
- Henan Province Engineering Technology Research Center of Rapid-Accuracy Medical Diagnostics, Department of Clinical Laboratory, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, China
| | - Qiming Li
- Henan Province Engineering Technology Research Center of Rapid-Accuracy Medical Diagnostics, Department of Clinical Laboratory, The First Affiliated Hospital of Henan University, Henan University, Kaifeng, China
- The Joint National Laboratory of Antibody Drug Engineering, Henan University, Kaifeng, China
- Department of Microbiology, College of Basic Medical Sciences, Henan University, Kaifeng, China
| |
Collapse
|
6
|
Chan BK, Stanley GL, Kortright KE, Vill AC, Modak M, Ott IM, Sun Y, Würstle S, Grun CN, Kazmierczak BI, Rajagopalan G, Harris ZM, Britto CJ, Stewart J, Talwalkar JS, Appell CR, Chaudary N, Jagpal SK, Jain R, Kanu A, Quon BS, Reynolds JM, Teneback CC, Mai QA, Shabanova V, Turner PE, Koff JL. Personalized inhaled bacteriophage therapy for treatment of multidrug-resistant Pseudomonas aeruginosa in cystic fibrosis. Nat Med 2025; 31:1494-1501. [PMID: 40301561 PMCID: PMC12092284 DOI: 10.1038/s41591-025-03678-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 03/27/2025] [Indexed: 05/01/2025]
Abstract
Bacteriophage (phage) therapy, which uses lytic viruses as antimicrobials, is a potential strategy to address the antimicrobial resistance crisis. Cystic fibrosis, a disease complicated by recurrent Pseudomonas aeruginosa pulmonary infections, is an example of the clinical impact of antimicrobial resistance. Here, using a personalized phage therapy strategy that selects phages for a predicted evolutionary trade-off, nine adults with cystic fibrosis (eight women and one man) of median age 32 (range 22-46) years were treated with phages on a compassionate basis because their clinical course was complicated by multidrug-resistant or pan-drug-resistant Pseudomonas that was refractory to prior courses of standard antibiotics. The individuals received a nebulized cocktail or single-phage therapy without adverse events. Five to 18 days after phage therapy, sputum Pseudomonas decreased by a median of 104 CFU ml-1, or a mean difference of 102 CFU ml-1 (P = 0.006, two-way analysis of variance with Dunnett's multiple-comparisons test), without altering sputum microbiome, and an analysis of sputum Pseudomonas showed evidence of trade-offs that decreased antibiotic resistance or bacterial virulence. In addition, an improvement of 6% (median) and 8% (mean) predicted FEV1 was observed 21-35 days after phage therapy (P = 0.004, Wilcoxon signed-rank t-test), which may reflect the combined effects of decreased bacterial sputum density and phage-driven trade-offs. These results show that a personalized, nebulized phage therapy trade-off strategy may affect clinical and microbiologic endpoints, which must be evaluated in larger clinical trials.
Collapse
Affiliation(s)
- Benjamin K Chan
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
- Center for Phage Biology and Therapy, Yale University, New Haven, CT, USA
| | - Gail L Stanley
- Center for Phage Biology and Therapy, Yale University, New Haven, CT, USA
- Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Kaitlyn E Kortright
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
- Center for Phage Biology and Therapy, Yale University, New Haven, CT, USA
| | - Albert C Vill
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
- Center for Phage Biology and Therapy, Yale University, New Haven, CT, USA
| | - Mrinalini Modak
- Department of Medicine, Division of Pulmonary and Critical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Isabel M Ott
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
- Center for Phage Biology and Therapy, Yale University, New Haven, CT, USA
| | - Ying Sun
- Center for Phage Biology and Therapy, Yale University, New Haven, CT, USA
- Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Silvia Würstle
- Center for Phage Biology and Therapy, Yale University, New Haven, CT, USA
- Department of Internal Medicine, Infectious Diseases, University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Casey N Grun
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT, USA
| | - Barbara I Kazmierczak
- Department of Internal Medicine, Section of Infectious Diseases, Yale School of Medicine, New Haven, CT, USA
| | - Govindarajan Rajagopalan
- Center for Phage Biology and Therapy, Yale University, New Haven, CT, USA
- Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Zachary M Harris
- Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Clemente J Britto
- Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Jill Stewart
- Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Jaideep S Talwalkar
- Department of Internal Medicine, Section General Internal Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Casey R Appell
- Department of Kinesiology & Sports Management, Texas Tech University, Lubbock, TX, USA
| | - Nauman Chaudary
- Department of Medicine, Division of Pulmonary Disease and Critical Care Medicine, Virginia Commonwealth University, Richmond, VA, USA
| | - Sugeet K Jagpal
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Raksha Jain
- Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Adaobi Kanu
- Department of Pediatrics, Texas Tech University Health Sciences Center School of Medicine, Lubbock, TX, USA
| | - Bradley S Quon
- Faculty of Medicine, Centre for Heart Lung Innovation, The University of British Columbia, Vancouver, British Columbia, Canada
| | - John M Reynolds
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Charlotte C Teneback
- Department of Medicine, Division of Pulmonary Disease and Critical Care Medicine, University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Quynh-Anh Mai
- Center for Phage Biology and Therapy, Yale University, New Haven, CT, USA
| | - Veronika Shabanova
- Center for Phage Biology and Therapy, Yale University, New Haven, CT, USA
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Paul E Turner
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, USA
- Center for Phage Biology and Therapy, Yale University, New Haven, CT, USA
- Program in Microbiology, Yale School of Medicine, New Haven, CT, USA
| | - Jonathan L Koff
- Center for Phage Biology and Therapy, Yale University, New Haven, CT, USA.
- Department of Internal Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
7
|
Singh J, Solomon M, Iredell J, Selvadurai H. Overcoming Pseudomonas aeruginosa in Chronic Suppurative Lung Disease: Prevalence, Treatment Challenges, and the Promise of Bacteriophage Therapy. Antibiotics (Basel) 2025; 14:427. [PMID: 40426494 PMCID: PMC12108500 DOI: 10.3390/antibiotics14050427] [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: 02/26/2025] [Revised: 03/19/2025] [Accepted: 04/18/2025] [Indexed: 05/29/2025] Open
Abstract
Pseudomonas aeruginosa, a multidrug-resistant pathogen, significantly impacts patients with chronic respiratory conditions like cystic fibrosis (CF) and non-CF chronic suppurative lung disease (CSLD), contributing to progressive lung damage and poor clinical outcomes. This bacterium thrives in the airway environments of individuals with impaired mucociliary clearance, leading to persistent infections and increased morbidity and mortality. Despite advancements in management of these conditions, treatment failure remains common, emphasising the need for alternative or adjunctive treatment strategies. Bacteriophage therapy, an emerging approach utilising viruses that specifically target bacteria, offers a potential solution to combat P. aeruginosa infections resistant to conventional antibiotics. This review examines the prevalence and disease burden of P. aeruginosa in CF and CSLD, explores the mechanisms behind antibiotic resistance, the promising role of bacteriophage therapy and clinical trials in this sphere.
Collapse
Affiliation(s)
- Jagdev Singh
- Department of Respiratory Medicine, The Children’s Hospital at Westmead, Sydney, NSW 2145, Australia;
- Faculty of Medicine, University of Sydney, Sydney, NSW 2145, Australia;
| | - Melinda Solomon
- Department of Respiratory Medicine, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada;
- Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Jonathan Iredell
- Faculty of Medicine, University of Sydney, Sydney, NSW 2145, Australia;
- Westmead Institute of Medical Research, Sydney, NSW 2145, Australia
| | - Hiran Selvadurai
- Department of Respiratory Medicine, The Children’s Hospital at Westmead, Sydney, NSW 2145, Australia;
- Faculty of Medicine, University of Sydney, Sydney, NSW 2145, Australia;
| |
Collapse
|
8
|
Liu D, Qin K, Hong C, Huang W, Li W, Lian P, Li M, Chen H, Liu X. Isolation and Characterization of a Novel Lytic Phage N22 and Its Effect on Drug-Resistant Klebsiella Pneumoniae. Infect Drug Resist 2025; 18:1807-1818. [PMID: 40231317 PMCID: PMC11995918 DOI: 10.2147/idr.s515363] [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: 01/10/2025] [Accepted: 03/28/2025] [Indexed: 04/16/2025] Open
Abstract
Background Klebsiella pneumoniae (KP) infections present a significant clinical challenge and are frequently associated with elevated drug resistance. The use of phage therapy has resurged in response to escalating antibiotic resistance. This study aimed to address the multidrug resistance crisis in intensive care units by exploring the use of ceftazidime/avibactam (CAZ/AVI), a widely used clinical antimicrobial agent, in conjunction with phage therapy. Materials and Methods We screened a clinical strain of KP from ICU and successfully isolated phage N22 from hospital wastewater. We conducted an in-depth analysis of the physiological and biochemical properties of phage N22 and determined its optimal multiplicity of infection with the clinical KP strain. The inhibitory effects of phage N22 in combination with CAZ/AVI on biofilm formation were investigated. Comparative efficacies of these combinations were evaluated using a Galleria mellonella (G. mellonella) model. Results Phage N22 inhibited KP biofilm formation. The impact of varying phage N22 concentrations when used alongside CAZ/AVI was examined, and the combination of phage N22 and CAZ/AVI was more effective against KP than CAZ/AVI alone. Conclusion This study provides a preliminary investigation into the effects of combining CAZ/AVI with phage therapy, highlighting its potential significance in developing novel therapeutic strategies for bacterial infections resistant to CAZ/AVI. The findings underscore the importance of advancing highly effective phage agents as alternative treatment modalities for patients with infections refractory to conventional antibiotics.
Collapse
Affiliation(s)
- Dongyu Liu
- Department of Critical Care Medicine, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, People’s Republic of China
| | - Kunhao Qin
- Jiangxi Province Key Laboratory of Organ Development and Epigenetics, Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Medical Department of Jinggangshan University, Ji’an, 343009, People’s Republic of China
| | - Chengying Hong
- Department of Critical Care Medicine, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, People’s Republic of China
| | - Wei Huang
- Department of Laboratory Medicine, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, People’s Republic of China
| | - Wei Li
- Department of Critical Care Medicine, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, People’s Republic of China
| | - Puqiao Lian
- Department of Critical Care Medicine, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, People’s Republic of China
| | - Mengyao Li
- Department of Critical Care Medicine, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, People’s Republic of China
| | - Huaisheng Chen
- Department of Critical Care Medicine, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, People’s Republic of China
| | - Xueyan Liu
- Department of Critical Care Medicine, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, 518020, People’s Republic of China
| |
Collapse
|
9
|
Abbas S, Kanwar R, Ullah K, Kanwal R, Tajamal M, Aslam MA, Ahmad A, Qadeer A, Huang HY, Chen CC. Bacteriophage therapy: a possible alternative therapy against antibiotic-resistant strains of Klebsiella pneumoniae. Front Microbiol 2025; 16:1443430. [PMID: 40231234 PMCID: PMC11994585 DOI: 10.3389/fmicb.2025.1443430] [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: 06/04/2024] [Accepted: 03/07/2025] [Indexed: 04/16/2025] Open
Abstract
Klebsiella pneumoniae is a notorious, Gram-negative pathogen and is a leading cause of healthcare settings and community-acquired infections. This is the commensal of human microbiota and can invade and cause infections in different body parts. The global emergence of antibiotic resistance in K. pneumoniae has become a major challenge in the whole medical community. Alternative paths to treat the infections caused by these MDR pathogens are needed as these bacteria become resistant to last-resort antibiotics like colistin. The lytic bacteriophages (phages) are the bacteria's natural predators and can rapidly eliminate the bacterial cells. Phages are abundant in nature and have recently been found to be effective tools in modern biotechnology. They can be used to control the bacterial infectious diseases. They can be manipulated easily and potentially used in therapeutics, biotechnology, and research. Several studies, both in vitro and in vivo, have demonstrated the possible applications of the lytic phages in treating K. pneumoniae superbug strains. Phage endolysins have drawn the scientific world's attention because of their involvement in phage adsorption and bacterial capsules digestion. These phage-encoded enzymes digest the polysaccharide components of bacterial cell walls by recognizing and binding them. Phage lysins, being strong biological agents, are capable of effectively and swiftly eliminating bacteria. This review summarizes the information on phages of K. pneumoniae and phage-based therapies to target their bacterial hosts.
Collapse
Affiliation(s)
- Sadia Abbas
- Institute of Microbiology, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Rabia Kanwar
- Institute of Microbiology, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Kaleem Ullah
- Directorate General (Research) Livestock & Dairy Development Department Peshawar, Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Rimsha Kanwal
- Institute of Microbiology, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Mamoon Tajamal
- Institute of Microbiology, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Muhammad Aamir Aslam
- Institute of Microbiology, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Abid Ahmad
- Department of Animal Nutrition, The University of Agriculture Peshawar, Peshawar, Khyber Pakhtunkhwa, Pakistan
| | - Abdul Qadeer
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, China
| | - Hsun-Yu Huang
- Division of Endodontics, Department of Stomatology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
| | - Chien-Chin Chen
- Department of Pathology, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi, Taiwan
- Department of Cosmetic Science, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
- Doctoral Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung, Taiwan
- Department of Biotechnology and Bioindustry Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
- Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
10
|
Beyoğlu D, Idle JR. The Microbiome and Metabolic Dysfunction-Associated Steatotic Liver Disease. Int J Mol Sci 2025; 26:2882. [PMID: 40243472 PMCID: PMC11988851 DOI: 10.3390/ijms26072882] [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: 02/17/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a condition wherein excessive fat accumulates in the liver, leading to inflammation and potential liver damage. In this narrative review, we evaluate the tissue microbiota, how they arise and their constituent microbes, and the role of the intestinal and hepatic microbiota in MASLD. The history of bacteriophages (phages) and their occurrence in the microbiota, their part in the potential causation of MASLD, and conversely, "phage therapy" for antibiotic resistance, obesity, and MASLD, are all described. The microbiota metabolism of bile acids and dietary tryptophan and histidine is defined, together with the impacts of their individual metabolites on MASLD pathogenesis. Both periodontitis and intestinal microbiota dysbiosis may cause MASLD, and how individual microorganisms and their metabolites are involved in these processes is discussed. Novel treatment opportunities for MASLD involving the microbiota exist and include fecal microbiota transplantation, probiotics, prebiotics, synbiotics, tryptophan dietary supplements, intermittent fasting, and phages or their holins and endolysins. Although FDA is yet to approve phage therapy in clinical use, there are multiple FDA-approved clinical trials, and this may represent a new horizon for the future treatment of MASLD.
Collapse
Affiliation(s)
- Diren Beyoğlu
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA;
| | - Jeffrey R. Idle
- Department of Pharmaceutical and Administrative Sciences, College of Pharmacy and Health Sciences, Western New England University, Springfield, MA 01119, USA;
- Department of Biomedical Research, University of Bern, 3008 Bern, Switzerland
| |
Collapse
|
11
|
Schieferecke AJ, Kuxhausen Ralph N, Schaffer DV. The Application of DNA Viruses to Biotechnology. Viruses 2025; 17:414. [PMID: 40143341 PMCID: PMC11946468 DOI: 10.3390/v17030414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 02/24/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
The delivery of biomolecules to target cells has been a longstanding challenge in biotechnology. DNA viruses naturally evolved the ability to deliver genetic material to cells and modulate cellular processes. As such, they inherently possess requisite characteristics that have led to their extensive study, engineering, and development as biotechnological tools. Here, we overview the application of DNA viruses to biotechnology, with specific implications in basic research, health, biomanufacturing, and agriculture. For each application, we review how an increasing understanding of virology and technological methods to genetically manipulate DNA viruses has enabled advances in these fields. Additionally, we highlight the remaining challenges to unlocking the full biotechnological potential of DNA viral technologies. Finally, we discuss the importance of balancing continued technological progress with ethical and biosafety considerations.
Collapse
Affiliation(s)
- Adam J. Schieferecke
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; (N.K.R.); (D.V.S.)
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Nadia Kuxhausen Ralph
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; (N.K.R.); (D.V.S.)
| | - David V. Schaffer
- Department of Molecular & Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA; (N.K.R.); (D.V.S.)
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
12
|
Ranta K, Skurnik M, Kiljunen S. Isolation and characterization of fMGyn-Pae01, a phiKZ-like jumbo phage infecting Pseudomonas aeruginosa. Virol J 2025; 22:55. [PMID: 40033410 PMCID: PMC11877940 DOI: 10.1186/s12985-025-02679-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 02/20/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND Pseudomonas aeruginosa is an opportunistic pathogen that causes a wide variety of infections, and belongs to the group of ESKAPE pathogens that are the leading cause of healthcare-associated infections and have high level of antibiotic resistance. The treatment of infections caused by antibiotic-resistant P. aeruginosa is challenging, which makes it a common target for phage therapy. The successful utilization of phage therapy requires a collection of well characterized phages. METHODS Phage fMGyn-Pae01 was isolated from a commercial phage therapy cocktail. The phage morphology was studied by transmission electron microscopy and the host range was analyzed with a liquid culture method. The phage genome was sequenced and characterized, and the genome was compared to closest phage genomes. Phage resistant bacterial mutants were isolated and whole genome sequencing and motility, phage adsorption and biofilm formation assays were performed to the mutants and host bacterium. RESULTS The genomic analysis revealed that fMGyn-Pae01 is a lytic, phiKZ-like jumbo phage with genome size of 277.8 kb. No genes associated with lysogeny, bacterial virulence, or antibiotic resistance were identified. Phage fMGyn-Pae01 did not reduce biofilm formation of P. aeruginosa, suggesting that it may not be an optimal phage to be used in monophage therapy in conditions where biofilm formation is expected. Host range screening revealed that fMGyn-Pae01 has a wide host range among P. aeruginosa strains and its infection was not dependent on O-serotype. Whole genome sequencing of the host bacterium and phage resistant mutants revealed that the mutations had inactivated either a flagellar or rpoN gene, thereby preventing the biosynthesis of a functional flagellum. The lack of functional flagella was confirmed in motility assays. Additionally, fMGyn-Pae01 failed to adsorb on non-motile mutants indicating that the bacterial flagellum is the phage-binding receptor. CONCLUSION fMGyn-Pae01 is a phiKZ-like jumbo phage infecting P. aeruginosa. fMGyn-Pae01 uses the flagellum as its phage-binding receptor, supporting earlier suggestions that flagellum might be utilized by phiKZ but differs from some other previous findings showing that phiKZ-like phages use the type-IV pili as the phage-binding receptor.
Collapse
Affiliation(s)
- Kira Ranta
- HUS Diagnostic Center, Clinical Microbiology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Human Microbiome Research Program, Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mikael Skurnik
- Human Microbiome Research Program, Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Saija Kiljunen
- Human Microbiome Research Program, Research Program Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
13
|
Kim MK, Suh GA, Cullen GD, Perez Rodriguez S, Dharmaraj T, Chang THW, Li Z, Chen Q, Green SI, Lavigne R, Pirnay JP, Bollyky PL, Sacher JC. Bacteriophage therapy for multidrug-resistant infections: current technologies and therapeutic approaches. J Clin Invest 2025; 135:e187996. [PMID: 40026251 PMCID: PMC11870740 DOI: 10.1172/jci187996] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025] Open
Abstract
Bacteriophage (phage) therapy has emerged as a promising solution to combat the growing crisis of multidrug-resistant (MDR) infections. There are several international centers actively engaged in implementation of phage therapy, and recent case series have reported encouraging success rates in patients receiving personalized, compassionate phage therapy for difficult-to-treat infections. Nonetheless, substantial hurdles remain in the way of more widespread adoption and more consistent success. This Review offers a comprehensive overview of current phage therapy technologies and therapeutic approaches. We first delineate the common steps in phage therapy development, from phage bank establishment to clinical administration, and examine the spectrum of therapeutic approaches, from personalized to fixed phage cocktails. Using the framework of a conventional drug development pipeline, we then identify critical knowledge gaps in areas such as cocktail design, formulation, pharmacology, and clinical trial design. We conclude that, while phage therapy holds promise, a structured drug development pipeline and sustained government support are crucial for widespread adoption of phage therapy for MDR infections.
Collapse
Affiliation(s)
- Minyoung Kevin Kim
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
- Department of Medicine, Yale University, New Haven, Connecticut, USA
| | - Gina A. Suh
- Division of Public Health, Infectious Diseases and Occupational Health, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Grace D. Cullen
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Saumel Perez Rodriguez
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Tejas Dharmaraj
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Tony Hong Wei Chang
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Zhiwei Li
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Qingquan Chen
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Sabrina I. Green
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Rob Lavigne
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Jean-Paul Pirnay
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| | - Paul L. Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Jessica C. Sacher
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
- Phage Directory, Atlanta, Georgia, USA
| |
Collapse
|
14
|
Hickson SM, Ledger EL, Wells TJ. Emerging antimicrobial therapies for Gram-negative infections in human clinical use. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:16. [PMID: 40016340 PMCID: PMC11868545 DOI: 10.1038/s44259-025-00087-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/13/2025] [Indexed: 03/01/2025]
Abstract
The growing problem of multi-drug resistance (MDR) is prevalent in Gram-negative infections, and the significant decline in antibiotic development poses a critical threat to global public health. Many emerging non-antibiotic therapies have been proposed, including phage therapy, anti-virulence agents, antimicrobial peptides, plasmapheresis, and immunotherapy options. To identify the therapies most likely to be the next immediate step in treatment for MDR Gram-negative infections, this review highlights emerging therapeutics that have either been successfully used for compassionate care or are currently undergoing clinical trials.
Collapse
Affiliation(s)
- Sarah M Hickson
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Emma L Ledger
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Timothy J Wells
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
15
|
Cao L, Mi J, He Y, Xuan G, Wang J, Li M, Tong Y. Quorum sensing inhibits phage infection by regulating biofilm formation of P. aeruginosa PAO1. J Virol 2025; 99:e0187224. [PMID: 39745428 PMCID: PMC11853092 DOI: 10.1128/jvi.01872-24] [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: 10/24/2024] [Accepted: 12/13/2024] [Indexed: 02/26/2025] Open
Abstract
Quorum sensing (QS) can regulate diverse critical phenotypic responses in Pseudomonas. aeruginosa (P. aeruginosa), enabling bacterial adaptation to external environmental fluctuations and optimizing population advantages. While there is emerging evidence of QS's involvement in influencing phage infections, our current understanding remains limited, necessitating further investigation. In this study, we isolated and characterized a novel phage designated as BUCT640 that infected P. aeruginosa PAO1. This phage belonged to class Caudoviricetes, genus Bruynoghevirus, with a podovirus morphology, and its adsorption was dependent on Psl polysaccharides, a repeating pentamer used to support biofilm structure. Leveraging phage BUCT640 as a model, we analyzed the role of both rhl QS and las QS in bacteria-phage interactions. Based on its distinctive plaque formation performances on different QS-related mutants, we investigated the variations of phage sensitivity to these strains and ultimately elucidated the mechanism underlying how QS inhibited phage infection to PAO1. Specifically, we unveiled that the las QS could inhibit phage adsorption, which is related to the thickness change caused by biofilm differentiation. Our findings suggest that the inhibition of QS may enhance phage infectivity, potentially facilitating advanced phage therapy combined with QS interference. IMPORTANCE Phage therapy is a powerful solution to combat drug-resistant pathogenic bacterial infections and has earned remarkable success in clinical treatment. However, recent insights underscore the potential impact of bacterial QS on phage infection dynamics. Here, we reported a unique phenomenon wherein QS, particularly in the las QS pathway, showed distinctive plaque formation behaviors by enlarging halos around plaques in mutant strains. In addition to this, we first elucidated the correlation between biofilm formation and phage infection. Notably, the las QS could inhibit phage adsorption, an effect closely related to biofilm thickness. Such research could be the evidence to steer bacterial QS toward favorable therapeutical outcomes. Therefore, our work can extend the comprehension of the interactions between bacteria and phages influenced by QS, thereby providing new perspectives on leveraging QS interference to enhance the efficacy of phage therapy for clinical applications.
Collapse
Affiliation(s)
- Lei Cao
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, Beijing, China
| | - Jinhui Mi
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, Beijing, China
| | - Yile He
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, Beijing, China
| | - Guanhua Xuan
- Food Safety Laboratory, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Jingxue Wang
- Food Safety Laboratory, College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong, China
| | - Mengzhe Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, Beijing, China
- Qinhuangdao Bohai Biological Research Institute, Beijing University of Chemical Technology, Qinhuangdao, Hebei, China
| | - Yigang Tong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, Beijing, China
| |
Collapse
|
16
|
Kabwe M, Tucci J, Darby I, Dashper S. Oral bacteriophages and their potential as adjunctive treatments for periodontitis: a narrative review. J Oral Microbiol 2025; 17:2469890. [PMID: 40013014 PMCID: PMC11864011 DOI: 10.1080/20002297.2025.2469890] [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: 12/23/2024] [Revised: 01/29/2025] [Accepted: 01/31/2025] [Indexed: 02/28/2025] Open
Abstract
Background There is no specific cure for periodontitis and treatment is symptomatic, primarily by physical removal of the subgingival plaque biofilm. Current non-surgical periodontal therapy becomes less effective as the periodontal pocket depth increases and as such new adjunctive treatments are required. The development of antibiotic resistance has driven a recent resurgence of interest in bacteriophage therapy. Methods Here we review the published literature with a focus on the subgingival phageome, key oral pathobionts and the dysbiotic nature of periodontitis leading to the emergence of synergistic, proteolytic and inflammophilic bacterial species in subgingival plaque. We discuss the opportunities available, the barriers and the steps needed to develop bacteriophage therapy as an adjunctive treatment for periodontitis. Results The oral phageome (or virome) is diverse, featuring abundant bacteriophage, that could target key subgingival bacteria. Yet to date few bacteriophages have been isolated and characterised from oral bacterial species, although many more have been predicted by genomic analyses. Bacteriophage therapy has yet to be tested against chronic diseases that are caused by dysbiosis of the endogenous microbial communities. Conclusion To be effective as an adjunctive treatment for periodontitis, bacteriophage therapy must cause the collapse of the dysbiotic bacterial community, thereby resolving inflammation and enabling the reestablishment of a health-associated mutualistic subgingival bacterial community. The isolation and characterisation of novel oral bacteriophage is an essential first step in this process.
Collapse
Affiliation(s)
- Mwila Kabwe
- Department of Rural Clinical Sciences, La Trobe Rural Health School & La Trobe Institute for Molecular Science, La Trobe University, Bendigo, Victoria, Australia
| | - Joseph Tucci
- Department of Rural Clinical Sciences, La Trobe Rural Health School & La Trobe Institute for Molecular Science, La Trobe University, Bendigo, Victoria, Australia
| | - Ivan Darby
- Melbourne Dental School, University of Melbourne, Parkville, Victoria, Australia
| | - Stuart Dashper
- Melbourne Dental School, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
17
|
Kalia VC, Patel SKS, Gong C, Lee JK. Re-Emergence of Bacteriophages and Their Products as Antibacterial Agents: An Overview. Int J Mol Sci 2025; 26:1755. [PMID: 40004222 PMCID: PMC11855700 DOI: 10.3390/ijms26041755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 02/14/2025] [Accepted: 02/15/2025] [Indexed: 02/27/2025] Open
Abstract
Microbes possess diverse genetic and metabolic traits that help them withstand adverse conditions. Microbial pathogens cause significant economic losses and around 7.7 million human deaths annually. While antibiotics have historically been a lifesaving treatment, their effectiveness is declining due to antibiotic-resistant strains, prompting the exploration of bacterial predation as an alternative. Bacteriophages (BPhs) have reemerged as antibacterial agents, offering advantages over antibiotics, such as (i) high specificity, (ii) self-replication, and (iii) strong killing capacity. This review explores BPh- and enzyme-based antibacterial strategies for infectious disease treatment, discussing phage-antibiotic synergy, the risks of BPh resistance, and the role of quorum sensing in BPh therapy.
Collapse
Affiliation(s)
- Vipin Chandra Kalia
- Department of Chemical Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea;
| | - Sanjay K. S. Patel
- Department of Chemical Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea;
| | - Chunjie Gong
- Cooperative Innovation Center of Industrial Fermentation (Ministry of Education & Hubei Province), Key Laboratory of Fermentation Engineering (Ministry of Education), National “111” Center for Cellular Regulation and Molecular Pharmaceutics, Hubei University of Technology, Wuhan 430068, China;
| | - Jung-Kul Lee
- Department of Chemical Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea;
| |
Collapse
|
18
|
Wannasrichan W, Krobthong S, Morgan CJ, Armbruster EG, Gerovac M, Yingchutrakul Y, Wongtrakoongate P, Vogel J, Aonbangkhen C, Nonejuie P, Pogliano J, Chaikeeratisak V. A ribosome-interacting jumbophage protein associates with the phage nucleus to facilitate efficient propagation. PLoS Pathog 2025; 21:e1012936. [PMID: 39992933 PMCID: PMC11849849 DOI: 10.1371/journal.ppat.1012936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 01/24/2025] [Indexed: 02/26/2025] Open
Abstract
Bacteriophages must hijack the gene expression machinery of their bacterial host to efficiently replicate. Recently, we have shown that the early-expressed protein gp014 of Pseudomonas nucleus-forming phage phiKZ forms a stable complex with the host ribosomes and modulates the overall protein expression profile during phage infection. Here, we discover a nucleus-forming phage, designated Churi, that is closely related to phiKZ. Churi encodes gp335, a homolog of gp014-phiKZ, which is expressed during the early stages of infection, and its overexpression in bacterial cells interferes with bacterial growth, suggesting its role in phage-host interplay. We predict experimentally that gp335 also interacts with host ribosomal proteins, similar to its homolog gp014-phiKZ, thereby strengthening its involvement in protein translation during phage infection. We further show that GFP-tagged gp335 specifically localizes by clustering around the phage nucleus and remains associated with it throughout the infection cycle. The CRISPR-Cas13-mediated deletion of gp335 reveals that the mutant phage fails to replicate efficiently, resulting in an extended latent period. Altogether, our study demonstrates that gp335 is an early-expressed protein of the Chimallivirus Churi that localizes in proximity to the phage nucleus, likely serving a role in localized translation to ensure efficient phage propagation.
Collapse
Affiliation(s)
- Wichanan Wannasrichan
- Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Sucheewin Krobthong
- Center of Excellence in Natural Products Chemistry (CENP), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Chase J. Morgan
- School of Biological Sciences, University of California San Diego, La Jolla, California, United States
| | - Emily G. Armbruster
- School of Biological Sciences, University of California San Diego, La Jolla, California, United States
| | - Milan Gerovac
- Institute for Molecular Infection Biology (IMIB), Faculty of Medicine, University of Würzburg, Würzburg, Germany
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
- Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Yodying Yingchutrakul
- National Center for Genetic Engineering and Biotechnology, NSTDA, Pathum Thani, Thailand
| | | | - Jörg Vogel
- Institute for Molecular Infection Biology (IMIB), Faculty of Medicine, University of Würzburg, Würzburg, Germany
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Chanat Aonbangkhen
- Center of Excellence in Natural Products Chemistry (CENP), Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence on Petrochemical and Materials Technology, Chulalongkorn University, Bangkok, Thailand
| | - Poochit Nonejuie
- Center for Advanced Therapeutics, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhon Pathom, Thailand
| | - Joe Pogliano
- School of Biological Sciences, University of California San Diego, La Jolla, California, United States
| | | |
Collapse
|
19
|
Necel A, Dydecka A, Topka-Bielecka G, Wesołowski W, Lewandowska N, Bloch S, Nejman-Faleńczyk B. What, how, and why? - anti-EHEC phages and their application potential in medicine and food industry. J Appl Genet 2025; 66:219-240. [PMID: 39527365 PMCID: PMC11762087 DOI: 10.1007/s13353-024-00918-4] [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: 09/09/2024] [Revised: 10/12/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
Enterohemorrhagic Escherichia coli (EHEC) are pathogens that, only in the United States, cause more than 250,000 foodborne infections a year. Since antibiotics or other antidiarrheal agents may increase the hemolytic-uremic syndrome (HUS) development risk, currently only supportive therapy, including hydration, is used. Therefore, many methods to fight EHEC bacteria focus on their use in food processing to prevent human infection. One of the proposed anti-EHEC agents is bacteriophages, known for their bactericidal effect, host specificity, and lack of cross-resistance with antibiotics. In this review article, we provide an overview of the characteristics like source of isolation, morphology, kinetics of life cycle, and treatment potential of over 130 bacteriophages able to infect EHEC strains. Based on the reviewed literature, we conclude that bacteriophages may play a highly significant role in regulating EHEC propagation. In addition, we also point out the phage features that should be taken into account not only when using bacteriophages but also when examining their properties. This may contribute to accelerating the pace of work on the preventive use of bacteriophages, which is extremely needed in the modern world of the food industry, but also stimulate interest in phages and accelerate regulatory work that would enable the use of bacteriophages also in medicine, to fight the drug-resistant strains.
Collapse
Affiliation(s)
- Agnieszka Necel
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdańsk, Dębowa 25, 80-204, Gdansk, Poland.
| | | | | | - Wojciech Wesołowski
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdansk, Poland
| | - Natalia Lewandowska
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdansk, Poland
| | - Sylwia Bloch
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdansk, Poland
- BNF - New Bio Force sp. z o.o., Kartuska 420a, 80-125, Gdańsk, Poland
| | - Bożena Nejman-Faleńczyk
- Department of Molecular Biology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308, Gdansk, Poland
- BNF - New Bio Force sp. z o.o., Kartuska 420a, 80-125, Gdańsk, Poland
| |
Collapse
|
20
|
Sabzali S, Pazhouhnia S, Shahzamani K, Sedeh PA. Role of phage therapy in acute gastroenteritis. JOURNAL OF RESEARCH IN MEDICAL SCIENCES : THE OFFICIAL JOURNAL OF ISFAHAN UNIVERSITY OF MEDICAL SCIENCES 2025; 30:2. [PMID: 40200968 PMCID: PMC11974603 DOI: 10.4103/jrms.jrms_464_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/30/2024] [Accepted: 11/25/2024] [Indexed: 04/10/2025]
Abstract
The gut ecosystem, comprising the gut microbiota and its interactions, plays a crucial role in human health and disease. This complex ecosystem involves a diverse array of microorganisms such as viruses, fungi, and bacteria, collectively known as the gut microbiota. These microorganisms contribute to various functions, including nutrient metabolism and immune modulation, thereby impacting human health. Dysbiosis, or an imbalance in the gut microbiota, has been associated with the pathogenesis of several diseases, ranging from intestinal disorders such as inflammatory bowel disease to extra-intestinal conditions such as metabolic and neurological disorders. The implications of dysbiosis in the gut ecosystem are far-reaching, affecting not only gastrointestinal health but also contributing to the development and progression of conditions such as autoimmune gastritis and gastric cancer. Furthermore, the burden of antimicrobial use and subsequent side effects, including antibiotic resistance, poses additional challenges in managing gastrointestinal diseases. In light of these complexities, investigating the role of bacteriophages as regulators of the gut ecosystem and their potential clinical applications presents a promising opportunity to tackle antibiotic resistance and fight infectious diseases.
Collapse
Affiliation(s)
- Somaieh Sabzali
- Department of Biology, Faculty of Basic Sciences, Lorestan University, Khorramabad, Iran
| | - Setareh Pazhouhnia
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Kiana Shahzamani
- Hepatitis Research Center, Faculty of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Peyman Adibi Sedeh
- Gastroenterology and Hepatology Research Center, Faculty of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
21
|
Eiferman V, Vion PA, Bleibtreu A. Phage Therapy as a Rescue Treatment for Recurrent Pseudomonas aeruginosa Bentall Infection. Viruses 2025; 17:123. [PMID: 39861912 PMCID: PMC11768548 DOI: 10.3390/v17010123] [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: 12/23/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Phage therapy is experiencing renewed interest, particularly for antibiotic-resistant infections, and may also be useful for difficult-to-treat cases where surgery to remove foreign infected material is deemed too risky. We report a case of recurrent Pseudomonas aeruginosa endocarditis with Bentall infection treated successfully with a combination of antibiotics and phages.
Collapse
Affiliation(s)
- Victor Eiferman
- Service des Maladies Infectieuses et Tropicales, Hôpital Pitié Salpêtrière, APHP Sorbonne Université, 75013 Paris, France;
| | - Pierre-Adrien Vion
- Service de Médecine Nucléaire, Hôpital Pitié Salpêtrière, Assistance Publique des Hôpitaux de Paris (APHP), 75013 Paris, France;
| | - Alexandre Bleibtreu
- Service des Maladies Infectieuses et Tropicales, Hôpital Pitié Salpêtrière, APHP Sorbonne Université, 75013 Paris, France;
| |
Collapse
|
22
|
Sarkodie-Addo P, Osman AH, Aglomasa BC, Donkor ES. Phage therapy in the management of respiratory and pulmonary infections: a systematic review. Ther Adv Infect Dis 2025; 12:20499361241307841. [PMID: 39866829 PMCID: PMC11760135 DOI: 10.1177/20499361241307841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 11/27/2024] [Indexed: 01/28/2025] Open
Abstract
Background Lower respiratory tract infections (LRTIs) pose a significant threat to global health, causing more than 2 million deaths worldwide. This menace is intensified by the alarming increase in drug resistance, which limits the availability of effective antibiotics for bacterial respiratory infections. Consequently, there is an urgent demand for alternative therapeutic options. Phage therapy (PT) has re-emerged as a promising therapeutic approach and as an adjunct to antibiotic treatment. Objective This systematic review synthesises the application of PT for LRTIs in humans, providing unified and updated data on the evaluation of the safety and efficacy of PT for LRTIs. Design Systematic review. Data sources and methods Following the PRISMA guidelines, a comprehensive search strategy was carried out (spanning January 2000 - February 2024) in four databases: PubMed, Scopus, ScienceDirect and Web of Science to retrieve published records of PT for LRTIs in humans only. The reference list of each included study was evaluated for possible inclusion of other relevant articles. Results Among the 18 records that fulfilled the inclusion criteria, 70 patients were administered PT. Microbiologically, 71.42% (n = 50/70) of the patients improved; with either the eradication of the pathogen or a decrease in bacterial load, whilst 15.71% (n = 11/70) did not record any improvement. About 5.71% (n = 4/70) recorded a partial/incomplete improvement, whilst 7.14% (n = 5/70) of the patients microbiological outcomes were unspecified. Clinically, up to 74.29% (n = 52/70) of the patients improved, whilst 10.00% (n = 7/70) of the patients showed no improvement. Another 2.86% (n = 2/70) recorded partial/incomplete improvement, whilst 12.86% (n = 9/70) were uncategorized. Phage titres that yielded positive outcomes ranged from 105 to 1012 PFU/mL. Studies that achieved a substantial phage titre at the site of infection frequently observed notable improvements. Regarding the safety of PT, 77.78% (N = 14/18) of the studies did not record any adverse effects after PT was administered, whilst 16.66% (n = 3/18) of the studies reported adverse effects. Conclusion Based on recently published data originating mainly from observational studies, PT has shown considerable efficacy and safety in the treatment of LRTIs. However, there is a lack of uniform methodologies and protocols across different PT cases in the management of LRTIs. Consequently, there is a need for additional clinical studies to establish standardised pharmacokinetic elements and an overall protocol for PT. By doing so, we can fully unlock the potential of PT in effectively managing clinical bacterial infections, including LRTIs.
Collapse
Affiliation(s)
| | - Abdul-Halim Osman
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Bill Clinton Aglomasa
- Department of Medical Microbiology, University of Ghana Medical School, Accra, Ghana
| | - Eric S. Donkor
- Department of Medical Microbiology, University of Ghana Medical School, P.O. Box KB 4236, Accra, Ghana
| |
Collapse
|
23
|
Parab L, Romeyer Dherbey J, Rivera N, Schwarz M, Gallie J, Bertels F. Chloramphenicol and gentamicin reduce the evolution of resistance to phage ΦX174 by suppressing a subset of E. coli LPS mutants. PLoS Biol 2025; 23:e3002952. [PMID: 39841243 PMCID: PMC11753469 DOI: 10.1371/journal.pbio.3002952] [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: 09/26/2024] [Accepted: 11/25/2024] [Indexed: 01/23/2025] Open
Abstract
Bacteriophages infect gram-negative bacteria by attaching to molecules present on the bacterial surface, often lipopolysaccharides (LPS). Modification of LPS can lead to resistance to phage infection. In addition, LPS modifications can impact antibiotic susceptibility, allowing for phage-antibiotic synergism. The evolutionary mechanism(s) behind such synergistic interactions remain largely unclear. Here, we show that the presence of antibiotics can affect the evolution of resistance to phage infection, using phage ΦX174 and Escherichia coli C. We use a collection of 34 E. coli C LPS strains, each of which is resistant to ΦX174, and has either a "rough" or "deep rough" LPS phenotype. Growth of the bacterial strains with the deep rough phenotype is inhibited at low concentrations of chloramphenicol and, to a much lesser degree, gentamicin. Treating E. coli C wild type with ΦX174 and chloramphenicol eliminates the emergence of mutants with the deep rough phenotype, and thereby slows the evolution of resistance to phage infection. At slightly lower chloramphenicol concentrations, phage resistance rates are similar to those observed at high concentrations; yet, we show that the diversity of possible mutants is much larger than at higher chloramphenicol concentrations. These data suggest that specific antibiotic concentrations can lead to synergistic phage-antibiotic interactions that disappear at higher antibiotic concentrations. Overall, we show that the change in survival of various ΦX174-resistant E. coli C mutants in the presence of antibiotics can explain the observed phage-antibiotic synergism.
Collapse
Affiliation(s)
- Lavisha Parab
- Microbial Molecular Evolution Group, Department of Microbial Population Biology, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Jordan Romeyer Dherbey
- Microbial Molecular Evolution Group, Department of Microbial Population Biology, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Norma Rivera
- Microbial Molecular Evolution Group, Department of Microbial Population Biology, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Michael Schwarz
- Microbial Molecular Evolution Group, Department of Microbial Population Biology, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Jenna Gallie
- Microbial Evolutionary Dynamics Group, Department of Theoretical Biology, Max Planck Institute for Evolutionary Biology, Plön, Germany
| | - Frederic Bertels
- Microbial Molecular Evolution Group, Department of Microbial Population Biology, Max Planck Institute for Evolutionary Biology, Plön, Germany
| |
Collapse
|
24
|
Gil-Gil T, Laborda P, Martínez JL, Hernando-Amado S. Use of adjuvants to improve antibiotic efficacy and reduce the burden of antimicrobial resistance. Expert Rev Anti Infect Ther 2025; 23:31-47. [PMID: 39670956 DOI: 10.1080/14787210.2024.2441891] [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: 06/26/2024] [Revised: 10/28/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
Abstract
INTRODUCTION The increase in antibiotic resistance, together with the absence of novel antibiotics, makes mandatory the introduction of novel strategies to optimize the use of existing antibiotics. Among these strategies, the use of molecules that increase their activity looks promising. AREAS COVERED Different categories of adjuvants have been reviewed. Anti-resistance adjuvants increase the activity of antibiotics by inhibiting antibiotic resistance determinants. Anti-virulence approaches focus on the infection process itself; reducing virulence in combination with an antibiotic can improve therapeutic efficacy. Combination of phages with antibiotics can also be useful, since they present different mechanisms of action and targets. Finally, combining antibiotics with adjuvants in the same molecule may serve to improve antibiotics' efficacy and to overcome potential problems of differential pharmacokinetics/pharmacodynamics. EXPERT OPINION The successful combination of inhibitors of β-lactamases with β-lactams has shown that adjuvants can improve the efficacy of current antibiotics. In this sense, novel anti-resistance adjuvants able to inhibit efflux pumps are still needed, as well as anti-virulence compounds that improve the efficacy of antibiotics by interfering with the infection process. Although adjuvants may present different pharmacodynamics/pharmacokinetics than antibiotics, conjugates containing both compounds can solve this problem. Finally, already approved drugs can be a promising source of antibiotic adjuvants.
Collapse
Affiliation(s)
- Teresa Gil-Gil
- Department of Biology, Emory University, Atlanta, GA, USA
| | - Pablo Laborda
- Department of Clinical Microbiology 9301, Rigshospitalet, Copenhagen, Denmark
| | | | | |
Collapse
|
25
|
Debray R, Conover A, Koskella B. Phages indirectly maintain tomato plant pathogen defense through regulation of the commensal microbiome. ISME COMMUNICATIONS 2025; 5:ycaf065. [PMID: 40356878 PMCID: PMC12066413 DOI: 10.1093/ismeco/ycaf065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 04/14/2025] [Accepted: 04/14/2025] [Indexed: 05/15/2025]
Abstract
As parasites of bacteria, phages can regulate microbiome diversity and composition and may therefore affect susceptibility to pathogens and disease. Many infectious diseases are associated with altered bacteriophage communities, but observational studies alone do not allow us to determine when altered phage community composition is a contributor to disease risk, a response to infection, or simply an indicator of dysbiosis. To address this question directly, we used size-selective filtration to deplete plant-associated microbial communities of phages, then challenged plants with the bacterial pathogen Pseudomonas syringae. Plants with phage-depleted microbiomes were more susceptible to infection, an effect that could not be explained by direct effects of the phage communities on either P. syringae or the plant host. Moreover, the presence of phages was most impactful when the phage communities were isolated from neighboring field locations rather than from the same host plant as the bacteria, possibly suggesting that moderate rates of lysis maintain a community structure that is most resistant to pathogen invasion. Overall, our results support the idea that phage communities contribute to plant defenses by modulating the microbiome.
Collapse
Affiliation(s)
- Reena Debray
- Department of Primate Behavior and Evolution, Max Planck Institute for Evolutionary Anthropology, 04103 Leipzig, Germany
- Department of Integrative Biology, University of California, Berkeley, 94720 Berkeley, CA, United States
| | - Asa Conover
- Department of Integrative Biology, University of California, Berkeley, 94720 Berkeley, CA, United States
| | - Britt Koskella
- Department of Integrative Biology, University of California, Berkeley, 94720 Berkeley, CA, United States
| |
Collapse
|
26
|
Antani JD, Ward T, Emonet T, Turner PE. Microscopic phage adsorption assay: High-throughput quantification of virus particle attachment to host bacterial cells. Proc Natl Acad Sci U S A 2024; 121:e2410905121. [PMID: 39700139 DOI: 10.1073/pnas.2410905121] [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: 06/01/2024] [Accepted: 11/12/2024] [Indexed: 12/21/2024] Open
Abstract
Phages, viruses of bacteria, play a pivotal role in Earth's biosphere and hold great promise as therapeutic and diagnostic tools in combating infectious diseases. Attachment of phages to bacterial cells is a crucial initial step of the interaction. The classic assay to quantify the dynamics of phage attachment involves coculturing and enumeration of bacteria and phages, which is laborious, lengthy, hence low-throughput, and only provides ensemble estimates of model-based adsorption rate constants. Here, we utilized fluorescence microscopy and particle tracking to obtain trajectories of individual virus particles interacting with cells. The trajectory durations quantified the heterogeneity in dwell time, the time that each phage spends interacting with a bacterium. The average dwell time strongly correlated with the classically measured adsorption rate constant. We successfully applied this technique to quantify host-attachment dynamics of several phages including those targeting key bacterial pathogens. This approach should benefit the field of phage biology by providing highly quantitative, model-free readouts at single-virus resolution, helping to uncover single-virus phenomena missed by traditional measurements. Owing to significant reduction in manual effort, our method should enable rapid, high-throughput screening of a phage library against a target bacterial strain for applications such as therapy or diagnosis.
Collapse
Affiliation(s)
- Jyot D Antani
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06520
- Center for Phage Biology & Therapy, Yale University, New Haven, CT 06520
- Quantitative Biology Institute, Yale University, New Haven, CT 06520
| | - Timothy Ward
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06520
| | - Thierry Emonet
- Quantitative Biology Institute, Yale University, New Haven, CT 06520
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520
- Department of Physics, Yale University, New Haven, CT 06520
| | - Paul E Turner
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06520
- Center for Phage Biology & Therapy, Yale University, New Haven, CT 06520
- Quantitative Biology Institute, Yale University, New Haven, CT 06520
- Program in Microbiology, Yale School of Medicine, New Haven, CT 06520
| |
Collapse
|
27
|
Schooley RT. Translational research priorities for bacteriophage therapeutics. Essays Biochem 2024; 68:621-631. [PMID: 39417290 DOI: 10.1042/ebc20240020] [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: 07/26/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 10/19/2024]
Abstract
The growing threat of antimicrobial resistant (AMR) bacterial pathogens coupled with the relative dearth of promising novel antibiotics requires the discovery and development additional medical interventions. Over the past decade bacteriophages have emerged one of the most promising new tools to combat AMR pathogens. Anecdotal clinical experiences under so-called 'compassionate use' regulatory pathways as well as a limited number of clinical trials have provided ample evidence of safety and early evidence of efficacy. For phages to reach their full potential it is critical that rigorous clinical trials be conducted that define their optimal use and that enable regulatory authorities to support the commercialization required to afford global access. The clinical development of phage therapeutics requires the design and execution of clinical trials that take full advantage of lessons learned from a century of antibiotic development and that use clinical investigation as a platform in which aspects of phage biology that are critical to therapeutics are more clearly elucidated. Translational research that elucidates phage biology in the context of clinical trials will promote highly relevant hypothesis-driven work in basic science laboratories and will greatly accelerate the development of the field of phage therapeutics.
Collapse
Affiliation(s)
- Robert T Schooley
- Department of Medicine, Center for Innovative Phage Applications and Therapeutics (IPATH), University of California San Diego, CA, U.S.A
| |
Collapse
|
28
|
Iarema P, Kotovskaya O, Skutel M, Drobiazko A, Moiseenko A, Sokolova O, Samitova A, Korostin D, Severinov K, Isaev A. Sxt1, Isolated from a Therapeutic Phage Cocktail, Is a Broader Host Range Relative of the Phage T3. Viruses 2024; 16:1905. [PMID: 39772213 PMCID: PMC11680406 DOI: 10.3390/v16121905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/02/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Using Escherichia coli BW25113 as a host, we isolated a novel lytic phage from the commercial poly-specific therapeutic phage cocktail Sextaphage® (Microgen, Russia). We provide genetic and phenotypic characterization of the phage and describe its host range on the ECOR collection of reference E. coli strains. The phage, hereafter named Sxt1, is a close relative of classical coliphage T3 and belongs to the Teetrevirus genus, yet its internal virion proteins, forming an ejectosome, differ from those of T3. In addition, the Sxt1 lateral tail fiber (LTF) protein clusters with those of the phages from the Berlinvirus genus. A comparison of T7, T3, and Sxt1 LTFs reveals the presence of insertions leading to the elongation of Sxt1 tail fibers, which, together with the difference in the HRDRs (host range-determining regions), might explain the expanded host specificity for the Sxt1.
Collapse
Affiliation(s)
- Polina Iarema
- Center for Molecular and Cellular Biology, Moscow 121205, Russia; (P.I.); (O.K.); (M.S.); (A.D.)
| | - Oksana Kotovskaya
- Center for Molecular and Cellular Biology, Moscow 121205, Russia; (P.I.); (O.K.); (M.S.); (A.D.)
| | - Mikhail Skutel
- Center for Molecular and Cellular Biology, Moscow 121205, Russia; (P.I.); (O.K.); (M.S.); (A.D.)
| | - Alena Drobiazko
- Center for Molecular and Cellular Biology, Moscow 121205, Russia; (P.I.); (O.K.); (M.S.); (A.D.)
| | - Andrei Moiseenko
- Faculty of Biology, Lomonosov Moscow State University, Moscow 119991, Russia; (A.M.); (O.S.)
| | - Olga Sokolova
- Faculty of Biology, Lomonosov Moscow State University, Moscow 119991, Russia; (A.M.); (O.S.)
| | - Alina Samitova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow 117997, Russia; (A.S.); (D.K.)
| | - Dmitriy Korostin
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow 117997, Russia; (A.S.); (D.K.)
| | | | - Artem Isaev
- Center for Molecular and Cellular Biology, Moscow 121205, Russia; (P.I.); (O.K.); (M.S.); (A.D.)
| |
Collapse
|
29
|
Hameed I, Ahmed A, Gandhi S, Nassiri N, Steinbacher DM, Vallabhajosyula P. Successful Investigational Phage Therapy for Pan-Resistant Bacterial Mediastinitis Following Type II Hybrid Aortic Arch Replacement. JACC Case Rep 2024; 29:102816. [PMID: 39691337 PMCID: PMC11646902 DOI: 10.1016/j.jaccas.2024.102816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 10/10/2024] [Indexed: 12/19/2024]
Abstract
A 47-year-old woman presented to our hospital with Stanford type B aortic dissection with retrograde arch extension. The decision was made to undergo hybrid arch repair with thoracic endovascular aortic repair. The patient underwent surgery, but her intraoperative course was complicated by delayed sternal closure and mediastinitis requiring flap reconstruction. Recurrent pan resistant Klebsiella sternal wound infection (SWI) was observed, for which successful investigational bacteriophage therapy initiated through mediastinal drains. She remains infection-free at 2-year follow-up. SWI is a morbid complication following cardiac surgery, particularly when colonized by pan-resistant bacterium. Phage therapy refers to the therapeutic use of modified bacteriophages and have been previously described for use in mediastinitis. We report the use of phage therapy to treat Klebsiella SWI in our patient. This case report adds to the current evidence supporting the use of phage therapy for treatment of complicated mediastinal wound infection following aortic arch surgery.
Collapse
Affiliation(s)
- Irbaz Hameed
- Division of Cardiothoracic Surgery, Division of Surgery Yale University School of Medicine, New Haven, Connecticut, USA
| | - Adham Ahmed
- Division of Cardiothoracic Surgery, Division of Surgery Yale University School of Medicine, New Haven, Connecticut, USA
| | - Shiv Gandhi
- Department of Infectious Diseases, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Naiem Nassiri
- Division of Vascular Surgery, Department of Surgery Yale University School of Medicine, New Haven, Connecticut, USA
| | - Derek M. Steinbacher
- Division of Plastic Surgery, Department of Surgery Yale University School of Medicine, New Haven, Connecticut, USA
| | - Prashanth Vallabhajosyula
- Division of Cardiothoracic Surgery, Division of Surgery Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
30
|
Ahmad TA, Houjeiry SE, Kanj SS, Matar GM, Saba ES. From forgotten cure to modern medicine: The resurgence of bacteriophage therapy. J Glob Antimicrob Resist 2024; 39:231-239. [PMID: 39486687 DOI: 10.1016/j.jgar.2024.10.259] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/13/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024] Open
Abstract
OBJECTIVES The unregulated use of antibiotics has led to the rise of antibiotic-resistant bacterial strains. This study explores bacteriophage therapy as an alternative treatment, highlighting its history, significance, and advancements in Europe, the United States, and the Middle East. METHODS A comprehensive literature review on bacteriophage therapy was conducted, focusing on its development, clinical trials, and patient treatment applications. The study also examined challenges, limitations, criteria for ideal phage selection, and manipulation techniques. RESULTS The United States and several European countries have advanced in phage therapy, progressing from clinical trials to patient treatment, whereas Middle Eastern countries are still in the early stages. Bacteriophages offer specificity, abundance, and minimal side effects, but challenges like safety concerns and potential resistance limit their widespread use. CONCLUSION Bacteriophage therapy shows promise as an antibiotic alternative but faces safety and resistance challenges. Continued research and better regulatory frameworks, especially in the Middle East, are needed to realize its potential.
Collapse
Affiliation(s)
- Tasnime Abdo Ahmad
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Samar El Houjeiry
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Souha S Kanj
- Division of Infectious Diseases, Department of Internal Medicine, Center for Infectious Diseases Research, American University of Beirut Medical Center, Beirut, Lebanon
| | - Ghassan M Matar
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Esber S Saba
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
31
|
Kim MK, Chen Q, Echterhof A, Pennetzdorfer N, McBride RC, Banaei N, Burgener EB, Milla CE, Bollyky PL. A blueprint for broadly effective bacteriophage-antibiotic cocktails against bacterial infections. Nat Commun 2024; 15:9987. [PMID: 39609398 PMCID: PMC11604943 DOI: 10.1038/s41467-024-53994-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/29/2024] [Indexed: 11/30/2024] Open
Abstract
Bacteriophage (phage) therapy is a promising therapeutic modality for multidrug-resistant bacterial infections, but its application is mainly limited to personalized therapy due to the narrow host range of individual phages. While phage cocktails targeting all possible bacterial receptors could theoretically confer broad coverage, the extensive diversity of bacteria and the complexity of phage-phage interactions render this approach challenging. Here, using screening protocols for identifying "complementarity groups" of phages using non-redundant receptors, we generate effective, broad-range phage cocktails that prevent the emergence of bacterial resistance. We also discover characteristic interactions between phage complementarity groups and particular antibiotic classes, facilitating the prediction of phage-antibiotic as well as phage-phage interactions. Using this strategy, we create three phage-antibiotic cocktails, each demonstrating efficacy against ≥96% of 153 Pseudomonas aeruginosa clinical isolates, including biofilm cultures, and demonstrate comparable efficacy in an in vivo wound infection model. We similarly develop effective Staphylococcus aureus phage-antibiotic cocktails and demonstrate their utility of combined cocktails against polymicrobial (mixed P. aeruginosa/S. aureus) cultures, highlighting the broad applicability of this approach. These studies establish a blueprint for the development of effective, broad-spectrum phage-antibiotic cocktails, paving the way for off-the-shelf phage-based therapeutics to combat multidrug-resistant bacterial infections.
Collapse
Affiliation(s)
- Minyoung Kevin Kim
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA.
| | - Qingquan Chen
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Arne Echterhof
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Nina Pennetzdorfer
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Robert C McBride
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Niaz Banaei
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Elizabeth B Burgener
- Center for Excellence in Pulmonary Biology, Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Carlos E Milla
- Center for Excellence in Pulmonary Biology, Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Paul L Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
32
|
Gu J, Zhang X, Liu T, Guo Y. Isolation and Characterization of a Lytic Phage PaTJ Against Pseudomonas aeruginosa. Viruses 2024; 16:1816. [PMID: 39772127 PMCID: PMC11680426 DOI: 10.3390/v16121816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 01/11/2025] Open
Abstract
Pseudomonas aeruginosa is a major global threat to human health, and phage therapy has emerged as a promising strategy for treating infections caused by multidrug-resistant pathogens. In this study, we isolated and characterized a Pseudomonas lytic phage, PaTJ, from wastewater. PaTJ belongs to the phage family Mesyanzhinovviridae, and is featured by short latency (30 min) and large burst size (103 PFU per infected cell). Our investigation revealed that PaTJ utilizes the type IV Pili (T4P) as a receptor. Transcriptome analysis of PaTJ infected host at latent stage showed distinct expression patterns of PaTJ encoding genes involved in replication and structure assembly, without expression of the majority of toxic accessory genes responsible for phage release. In addition, host bacteria exhibited specific induction of host metabolism-related genes in response to the PaTJ's infection. Furthermore, our findings demonstrated the PaTJ's potential in degrading biofilms. This work sheds light on the multifaceted impact of this lytic phage PaTJ on P. aeruginosa, presenting potential applications in both gene expression modulation and biofilm management.
Collapse
Affiliation(s)
- Jiayu Gu
- Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510000, China; (J.G.); (T.L.)
- University of Chinese Academy of Sciences, Beijing 100000, China
| | - Xinqiao Zhang
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin 300000, China;
| | - Tianlang Liu
- Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510000, China; (J.G.); (T.L.)
| | - Yunxue Guo
- Key Laboratory of Tropical Marine Bio-Resources and Ecology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510000, China; (J.G.); (T.L.)
- University of Chinese Academy of Sciences, Beijing 100000, China
| |
Collapse
|
33
|
Chaudhary V, Kajla P, Lather D, Chaudhary N, Dangi P, Singh P, Pandiselvam R. Bacteriophages: a potential game changer in food processing industry. Crit Rev Biotechnol 2024; 44:1325-1349. [PMID: 38228500 DOI: 10.1080/07388551.2023.2299768] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 08/16/2023] [Accepted: 10/03/2023] [Indexed: 01/18/2024]
Abstract
In the food industry, despite the widespread use of interventions such as preservatives and thermal and non-thermal processing technologies to improve food safety, incidences of foodborne disease continue to happen worldwide, prompting the search for alternative strategies. Bacteriophages, commonly known as phages, have emerged as a promising alternative for controlling pathogenic bacteria in food. This review emphasizes the potential applications of phages in biological sciences, food processing, and preservation, with a particular focus on their role as biocontrol agents for improving food quality and preservation. By shedding light on recent developments and future possibilities, this review highlights the significance of phages in the food industry. Additionally, it addresses crucial aspects such as regulatory status and safety concerns surrounding the use of bacteriophages. The inclusion of up-to-date literature further underscores the relevance of phage-based strategies in reducing foodborne pathogenic bacteria's presence in both food and the production environment. As we look ahead, new phage products are likely to be targeted against emerging foodborne pathogens. This will further advance the efficacy of approaches that are based on phages in maintaining the safety and security of food.
Collapse
Affiliation(s)
- Vandana Chaudhary
- Department of Dairy Technology, College of Dairy Science and Technology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana, India
| | - Priyanka Kajla
- Department of Food Technology, Guru Jambheshwar University of Science and Technology, Hisar, Haryana, India
| | - Deepika Lather
- Department of Veterinary Pathology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana, India
| | - Nisha Chaudhary
- Department of Food Science and Technology, College of Agriculture, Agriculture University, Jodhpur, Rajasthan, India
| | - Priya Dangi
- Department of Food and Nutrition and Food Technology, Institute of Home Economics, University of Delhi, New Delhi, India
| | - Punit Singh
- Department of Mechanical Engineering, Institute of Engineering and Technology, GLA University Mathura, Mathura, Uttar Pradesh, India
| | - Ravi Pandiselvam
- Physiology, Biochemistry and Post-Harvest Technology Division, ICAR -Central Plantation Crops Research Institute, Kasaragod, Kerala, India
| |
Collapse
|
34
|
Thompson DL, Semersky Z, Feinn R, Huang P, Turner PE, Chan BK, Koff JL, Murray TS. Particle size distribution of viable nebulized bacteriophage for the treatment of multi-drug resistant Pseudomonas aeruginosa. Respir Med Res 2024; 86:101133. [PMID: 39121591 DOI: 10.1016/j.resmer.2024.101133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/05/2024] [Accepted: 07/21/2024] [Indexed: 08/12/2024]
Affiliation(s)
| | | | - Richard Feinn
- Department of Biomedical Sciences, Frank Netter School of Medicine, Quinnipiac University, North Haven, CT, United States
| | - Pamela Huang
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, United States
| | - Paul E Turner
- Program in Microbiology, Yale School of Medicine, New Haven, CT, United States; Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, United States; Center for Phage Biology & Therapy, Yale University, New Haven, CT, United States
| | - Ben K Chan
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT, United States; Center for Phage Biology & Therapy, Yale University, New Haven, CT, United States
| | - Jonathan L Koff
- Center for Phage Biology & Therapy, Yale University, New Haven, CT, United States; Department of Internal Medicine Yale School of Medicine, New Haven, CT, United States
| | - Thomas S Murray
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, United States.
| |
Collapse
|
35
|
Singh A, Tanwar M, Singh TP, Sharma S, Sharma P. An escape from ESKAPE pathogens: A comprehensive review on current and emerging therapeutics against antibiotic resistance. Int J Biol Macromol 2024; 279:135253. [PMID: 39244118 DOI: 10.1016/j.ijbiomac.2024.135253] [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: 05/22/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024]
Abstract
The rise of antimicrobial resistance has positioned ESKAPE pathogens as a serious global health threat, primarily due to the limitations and frequent failures of current treatment options. This growing risk has spurred the scientific community to seek innovative antibiotic therapies and improved oversight strategies. This review aims to provide a comprehensive overview of the origins and resistance mechanisms of ESKAPE pathogens, while also exploring next-generation treatment strategies for these infections. In addition, it will address both traditional and novel approaches to combating antibiotic resistance, offering insights into potential new therapeutic avenues. Emerging research underscores the urgency of developing new antimicrobial agents and strategies to overcome resistance, highlighting the need for novel drug classes and combination therapies. Advances in genomic technologies and a deeper understanding of microbial pathogenesis are crucial in identifying effective treatments. Integrating precision medicine and personalized approaches could enhance therapeutic efficacy. The review also emphasizes the importance of global collaboration in surveillance and stewardship, as well as policy reforms, enhanced diagnostic tools, and public awareness initiatives, to address resistance on a worldwide scale.
Collapse
Affiliation(s)
- Anamika Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Mansi Tanwar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - T P Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Sujata Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India.
| | - Pradeep Sharma
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
36
|
Qin K, Shi X, Yang K, Xu Q, Wang F, Chen S, Xu T, Liu J, Wen W, Chen R, Liu Z, Cui L, Zhou K. Phage-antibiotic synergy suppresses resistance emergence of Klebsiella pneumoniae by altering the evolutionary fitness. mBio 2024; 15:e0139324. [PMID: 39248568 PMCID: PMC11481518 DOI: 10.1128/mbio.01393-24] [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: 05/29/2024] [Accepted: 08/13/2024] [Indexed: 09/10/2024] Open
Abstract
Phage-antibiotic synergy (PAS) represents a superior treatment strategy for pathogen infections with less probability of resistance development. Here, we aim to understand the molecular mechanism by which PAS suppresses resistance in terms of population evolution. A novel hypervirulent Klebsiella pneumoniae (KP) phage H5 was genetically and structurally characterized. The combination of H5 and ceftazidime (CAZ) showed a robust synergistic effect in suppressing resistance emergence. Single-cell Raman analysis showed that the phage-CAZ combination suppressed bacterial metabolic activities, contrasting with the upregulation observed with phage alone. The altered population evolutionary trajectory was found to be responsible for the contrasting metabolic activities under different selective pressures, resulting in pleiotropic effects. A pre-existing wcaJ point mutation (wcaJG949A) was exclusively selected by H5, conferring a fitness advantage and up-regulated activity of carbohydrate metabolism, but also causing a trade-off between phage resistance and collateral sensitivity to CAZ. The wcaJ point mutation was counter-selected by H5-CAZ, inducing various mutations in galU that imposed evolutionary disadvantages with higher fitness costs, and suppressed carbohydrate metabolic activity. H5 and H5-CAZ treatments resulted in opposite effects on the transcriptional activity of the phosphotransferase system and the ascorbate and aldarate metabolism pathway, suggesting potential targets for phage resistance suppression. Our study reveals a novel mechanism of resistance suppression by PAS, highlighting how the complexity of bacterial adaptation to selective pressures drives treatment outcomes. IMPORTANCE Phage-antibiotic synergy (PAS) has been recently proposed as a superior strategy for the treatment of multidrug-resistant pathogens to effectively reduce bacterial load and slow down both phage and antibiotic resistance. However, the underlying mechanisms of resistance suppression by PAS have been poorly and rarely been studied. In this study, we tried to understand how PAS suppresses the emergence of resistance using a hypervirulent Klebsiella pneumoniae (KP) strain and a novel phage H5 in combination with ceftazidime (CAZ) as a model. Our study reveals a novel mechanism by which PAS drives altered evolutionary trajectory of bacterial populations, leading to suppressed emergence of resistance. The findings advance our understanding of how PAS suppresses the emergence of resistance, and are imperative for optimizing the efficacy of phage-antibiotic therapy to further improve clinical outcomes.
Collapse
Affiliation(s)
- Kunhao Qin
- Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
- Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
- Jiangxi Province Key Laboratory of Organ Development and Epigenetics, Clinical Medical Research Center, Affiliated Hospital of Jinggangshan University, Health Science Center, Medical Department of Jinggangshan University, Ji'an, China
| | - Xing Shi
- Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Kai Yang
- Key Laboratory of Urban Environment and Health, Fujian Key Laboratory of Watershed Ecology, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, China
| | - Qiuqing Xu
- Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Fuxing Wang
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, Chinese University of Hong Kong, Shenzhen, China
| | - Senxiong Chen
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, Chinese University of Hong Kong, Shenzhen, China
| | - Tingting Xu
- Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Jinquan Liu
- Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Wangrong Wen
- Clinical Laboratory, The Affiliated Shunde Hospital of Jinan University, Foshan, China
- Clinical Laboratory Centre, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Rongchang Chen
- Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Zheng Liu
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, Chinese University of Hong Kong, Shenzhen, China
| | - Li Cui
- Key Laboratory of Urban Environment and Health, Fujian Key Laboratory of Watershed Ecology, Institute of Urban Environment, Chinese Academy of Sciences, Xiamen, China
| | - Kai Zhou
- Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
- Shenzhen Institute of Respiratory Diseases, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| |
Collapse
|
37
|
Anastassopoulou C, Ferous S, Petsimeri A, Gioula G, Tsakris A. Phage-Based Therapy in Combination with Antibiotics: A Promising Alternative against Multidrug-Resistant Gram-Negative Pathogens. Pathogens 2024; 13:896. [PMID: 39452768 PMCID: PMC11510143 DOI: 10.3390/pathogens13100896] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
The continued rise in antimicrobial resistance poses a serious threat to public health worldwide. The use of phages that can have bactericidal activity without disrupting the normal flora represents a promising alternative treatment method. This practice has been successfully applied for decades, mainly in Eastern Europe, and has recently been used as an emergency therapy for compassionate care in the United States. Here, we provide a comprehensive review of the pre-clinical and clinical applications of phage therapy concerning three major Gram-negative pathogens: Klebsiella pneumoniae, Pseudomonas aeruginosa, and Acinetobacter baumannii. The advantages and the challenges of expanding the usage of phages as an alternative or adjunctive treatment for antimicrobial-resistant bacterial infections are discussed. We emphasize the virologic complexities of using the highly adaptable phage populations as molecular tools, along with antibiotic chemical compounds, to effectively combat rapidly coevolving pathogenic bacteria in the host microenvironment. Pre-clinical studies, isolated clinical reports and a few randomized clinical trials have shown that bacteriophages can be effective in treating multidrug-resistant bacterial infections. The ability of some phages to revert the resistance against antibiotics, and possibly also against the human complement and other phages, appears to be a great advantage of phage therapy despite the inevitable emergence of phage-resistant strains. Bacteriophages (or specific phage-derived products) can enhance antimicrobial efficacy by reducing bacterial virulence via the alteration of basic bacterial structures, primarily of the cellular wall and membrane. Although several issues remain open regarding their effective clinical application, it appears that phage-based therapeutics in combination with antibiotics can provide an effective solution to the spread of antimicrobial resistance.
Collapse
Affiliation(s)
- Cleo Anastassopoulou
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.A.); (S.F.); (A.P.)
| | - Stefanos Ferous
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.A.); (S.F.); (A.P.)
| | - Aikaterini Petsimeri
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.A.); (S.F.); (A.P.)
| | - Georgia Gioula
- Department of Microbiology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Athanasios Tsakris
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.A.); (S.F.); (A.P.)
| |
Collapse
|
38
|
Antani JD, Ward T, Emonet T, Turner PE. Microscopic Phage Adsorption Assay: High-throughput quantification of virus particle attachment to host bacterial cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.09.617072. [PMID: 39416219 PMCID: PMC11482966 DOI: 10.1101/2024.10.09.617072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Phages, viruses of bacteria, play a pivotal role in Earth's biosphere and hold great promise as therapeutic and diagnostic tools in combating infectious diseases. Attachment of phages to bacterial cells is a crucial initial step of the interaction. The classic assay to quantify the dynamics of phage attachment involves co-culturing and enumeration of bacteria and phages, which is laborious, lengthy, hence low-throughput, and only provides ensemble estimates of model-based adsorption rate constants. Here, we utilized fluorescence microscopy and particle tracking to obtain trajectories of individual virus particles interacting with cells. The trajectory durations quantified the heterogeneity in dwell time, the time that each phage spends interacting with a bacterium. The average dwell time strongly correlated with the classically-measured adsorption rate constant. We successfully applied this technique to quantify host-attachment dynamics of several phages including those targeting key bacterial pathogens. This approach should benefit the field of phage biology by providing highly quantitative, model-free readouts at single-virus resolution, helping to uncover single-virus phenomena missed by traditional measurements. Owing to significant reduction in manual effort, our method should enable rapid, high-throughput screening of a phage library against a target bacterial strain for applications such as therapy or diagnosis.
Collapse
Affiliation(s)
- Jyot D. Antani
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06520, USA
- Center for Phage Biology & Therapy, Yale University, New Haven, CT 06520, USA
- Quantitative Biology Institute, Yale University, New Haven, CT 06520, USA
| | - Timothy Ward
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06520, USA
| | - Thierry Emonet
- Quantitative Biology Institute, Yale University, New Haven, CT 06520, USA
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06520, USA
- Department of Physics, Yale University, New Haven, CT 06520, USA
| | - Paul E. Turner
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, CT 06520, USA
- Center for Phage Biology & Therapy, Yale University, New Haven, CT 06520, USA
- Quantitative Biology Institute, Yale University, New Haven, CT 06520, USA
- Program in Microbiology, Yale School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
39
|
Otava UE, Tervo L, Havela R, Vuotari L, Ylänne M, Asplund A, Patpatia S, Kiljunen S. Phage-Antibiotic Combination Therapy against Recurrent Pseudomonas Septicaemia in a Patient with an Arterial Stent. Antibiotics (Basel) 2024; 13:916. [PMID: 39452183 PMCID: PMC11504013 DOI: 10.3390/antibiotics13100916] [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: 08/27/2024] [Revised: 09/16/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
Background: Intravascular stent infections are often associated with high risks of morbidity and mortality. We report here a case of a patient with an arterial stent and recurrent Pseudomonas septicaemias successfully treated with phage-meropenem combination therapy. Methods: A 75-year-old female with arteriosclerosis and comorbidities went through a femoropopliteal bypass with prosthesis in the right inguinal area. After the bypass, she developed a recurring Pseudomonas aeruginosa infection and also neutropenia during different antibiotics. A rapidly growing pseudoaneurysm in the right inguinal area led to an emergency intra-arterial stent placement during blood stream infection, later suspected to host a P. aeruginosa biofilm. Removing the stent was deemed precarious, and phage therapy was considered as a compassionate treatment option. A three-phage cocktail infecting the P. aeruginosa strain was prepared and administered intravenously together with meropenem for two weeks, after which, a ten-month follow-up was carried out. Results: No adverse reactions occurred during the phage therapy treatment, while infection markers were normalized. In addition, recovery was seen in a PET-CT scan. During the 10-month follow-up, no further P. aeruginosa septicaemias occurred. Conclusions: Phage-meropenem combination therapy was thus found safe and effective in the treatment of recurrent Pseudomonas septicaemia in a patient with an arterial stent.
Collapse
Affiliation(s)
- Ulla Elina Otava
- Department of Internal Medicine, Tampere University Hospital, 33520 Tampere, Finland (L.T.); (R.H.)
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland;
| | - Laura Tervo
- Department of Internal Medicine, Tampere University Hospital, 33520 Tampere, Finland (L.T.); (R.H.)
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland;
| | - Riikka Havela
- Department of Internal Medicine, Tampere University Hospital, 33520 Tampere, Finland (L.T.); (R.H.)
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland;
| | - Liisa Vuotari
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland;
- Department of Clinical Physiology and Nuclear Medicine, Tampere University Hospital, 33520 Tampere, Finland
| | - Matti Ylänne
- Human Microbiome Research Program, Research Programs Unit and Medicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland (S.P.)
| | - Annette Asplund
- Human Microbiome Research Program, Research Programs Unit and Medicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland (S.P.)
| | - Sheetal Patpatia
- Human Microbiome Research Program, Research Programs Unit and Medicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland (S.P.)
| | - Saija Kiljunen
- Human Microbiome Research Program, Research Programs Unit and Medicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland (S.P.)
- PrecisionPhage Ltd., 40500 Jyväskylä, Finland
| |
Collapse
|
40
|
Uchechukwu CF, Shonekan A. Current status of clinical trials for phage therapy. J Med Microbiol 2024; 73:001895. [PMID: 39320361 PMCID: PMC11423923 DOI: 10.1099/jmm.0.001895] [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: 01/05/2024] [Accepted: 08/30/2024] [Indexed: 09/26/2024] Open
Abstract
Recently, bacteriophages have been considered alternatives to antibacterial treatments. Infectious diseases continue to plague the world because bacteria can adapt and develop defence mechanisms against antibiotics. The growing incidence of antibiotic-resistant bacterial infections necessitated the development of new techniques for treating bacterial infections worldwide. Clinical trials have shown efficiency against antibiotic-resistant bacteria. However, scientists in future clinical trials should scrutinize phage resistance implications, assess combination strategies with antimicrobial agents and address challenges in phage therapy delivery for effective implementation.
Collapse
Affiliation(s)
- Chidiebere F. Uchechukwu
- Warwick Medical School, University of Warwick, Coventry, UK
- University of Birmingham, Birmingham, UK
| | | |
Collapse
|
41
|
Chen L, Wu MY, Chen SL, Hu R, Wang Y, Zeng W, Feng S, Ke M, Wang L, Chen S, Gu M. The Guardian of Vision: Intelligent Bacteriophage-Based Eyedrops for Clinical Multidrug-Resistant Ocular Surface Infections. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407268. [PMID: 39091071 DOI: 10.1002/adma.202407268] [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: 05/21/2024] [Revised: 07/22/2024] [Indexed: 08/04/2024]
Abstract
Clinical multidrug-resistant Pseudomonas aeruginosa (MDR-PA) is the leading cause of refractory bacterial keratitis (BK). However, the reported BK treatment methods lack biosecurity and bioavailability, which usually causes irreversible visual impairment and even blindness. Herein, for BK caused by clinically isolated MDR-PA infection, armed phages are modularized with the type I photosensitizer (PS) ACR-DMT, and an intelligent phage eyedrop is developed for combined phagotherapy and photodynamic therapy (PDT). These eyedrops maximize the advantages of bacteriophages and ACR-DMT, enabling more robust and specific targeting killing of MDR-PA under low oxygen-dependence, penetrating and disrupting biofilms, and efficiently preventing biofilm reformation. Altering the biofilm and immune microenvironments alleviates inflammation noninvasively, promotes corneal healing without scar formation, protects ocular tissues, restores visual function, and prevents long-term discomfort and pain. This strategy exhibits strong scalability, enables at-home treatment of ocular surface infections with great patient compliance and a favorable prognosis, and has significant potential for clinical application.
Collapse
Affiliation(s)
- Luojia Chen
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Ming-Yu Wu
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Si-Ling Chen
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Rui Hu
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Yifei Wang
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Weijuan Zeng
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Shun Feng
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, 610031, China
| | - Min Ke
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Lianrong Wang
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
- Department of Respiratory Diseases, Institute of Pediatrics, Shenzhen Children's Hospital, Shenzhen, 518026, China
| | - Shi Chen
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
- Department of Burn and Plastic Surgery, Shenzhen Key Laboratory of Microbiology in Genomic Modification & Editing and Application, Shenzhen Institute of Translational Medicine, Shenzhen University Medical School, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, China
| | - Meijia Gu
- Department of Ophthalmology, Ministry of Education Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, TaiKang Center for Life and Medical Sciences, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
42
|
Lin YH, Dharmaraj T, Chen Q, Echterhof A, Manasherob R, Zhang LJ, de Leeuw C, Peterson NA, Stannard W, Li Z, Hajfathalian M, Hargil A, Martinez HA, Pourtois J, Chang THW, Blankenberg FG, Amanatullah D, Chaudhuri O, Bollyky PL. Optimized Dosing and Delivery of Bacteriophage Therapy for Wound Infections. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.07.593005. [PMID: 38766200 PMCID: PMC11100690 DOI: 10.1101/2024.05.07.593005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Lytic bacteriophages, viruses that lyse (kill) bacteria, hold great promise for treating infections, including wound infections caused by antimicrobial-resistant Pseudomonas aeruginosa. However, the optimal dosing and delivery strategies for phage therapy remain unclear. In a mouse wound infection model, we investigated the impact of dose, frequency, and administration route on the efficacy of phage therapy. We find that topical but not intravenous delivery is effective in this model. High-doses of phage reduces bacterial burden more effectively than low-doses, and repeated dosing achieves the highest eradication rates. Building on these insights, we developed "HydroPhage", a hyaluronan-based hydrogel system that uses dynamic covalent crosslinking to deliver high-titre phages over one week. HydroPhage eradicates infections five times more effectively than intravenous injection. We conclude that hydrogel-based sustained phage delivery enhances the efficacy of phage therapy and offers a practical, well-tolerated option for topical application.
Collapse
Affiliation(s)
- Yung-Hao Lin
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Tejas Dharmaraj
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Qingquan Chen
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Arne Echterhof
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
- Institute of Medical Microbiology, University Hospital of Muenster, Muenster, Germany
| | - Robert Manasherob
- Department of Orthopaedic Surgery, Stanford Hospital and Clinics, Redwood City, CA, USA
| | - Lucy J. Zhang
- Department of Material Science and Engineering, Stanford University, Stanford, CA, USA
| | - Cas de Leeuw
- Institute for Molecules and Materials, Radboud University, Nijmegen, Netherlands
| | - Nana A. Peterson
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Whitney Stannard
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Zhiwei Li
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Maryam Hajfathalian
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
- Department of Biomedical Engineering, New Jersey Institute of Technology, University Heights, Newark, NJ
| | - Aviv Hargil
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Hunter A. Martinez
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Julie Pourtois
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Tony H. W. Chang
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| | - Francis G. Blankenberg
- Division of Pediatric Radiology and Nuclear Medicine, Department of Radiology, Lucile Packard Children’s Hospital, Stanford, CA, USA
| | - Derek Amanatullah
- Department of Orthopaedic Surgery, Stanford Hospital and Clinics, Redwood City, CA, USA
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Paul L. Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
43
|
Bucher MJ, Czyż DM. Phage against the Machine: The SIE-ence of Superinfection Exclusion. Viruses 2024; 16:1348. [PMID: 39339825 PMCID: PMC11436027 DOI: 10.3390/v16091348] [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: 05/28/2024] [Revised: 08/10/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
Prophages can alter their bacterial hosts to prevent other phages from infecting the same cell, a mechanism known as superinfection exclusion (SIE). Such alterations are facilitated by phage interactions with critical bacterial components involved in motility, adhesion, biofilm production, conjugation, antimicrobial resistance, and immune evasion. Therefore, the impact of SIE extends beyond the immediate defense against superinfection, influencing the overall fitness and virulence of the bacteria. Evaluating the interactions between phages and their bacterial targets is critical for leading phage therapy candidates like Pseudomonas aeruginosa, a Gram-negative bacterium responsible for persistent and antibiotic-resistant opportunistic infections. However, comprehensive literature on the mechanisms underlying SIE remains scarce. Here, we provide a compilation of well-characterized and potential mechanisms employed by Pseudomonas phages to establish SIE. We hypothesize that the fitness costs imposed by SIE affect bacterial virulence, highlighting the potential role of this mechanism in the management of bacterial infections.
Collapse
Affiliation(s)
- Michael J Bucher
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Daniel M Czyż
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
44
|
Pathak V, Chan HK, Zhou QT. Formulation of Bacteriophage for Inhalation to Treat Multidrug-Resistant Pulmonary Infections. KONA : POWDER SCIENCE AND TECHNOLOGY IN JAPAN 2024; 42:200-212. [PMID: 40114780 PMCID: PMC11925536 DOI: 10.14356/kona.2025016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Rapid development of antibiotic resistance in pathogenic bacteria and a decline in the pharmaceutical development of new antibiotics are pushing the research community to explore alternative antimicrobials that can replace or complement antibiotics. Bacteriophages (or, phages) are naturally occurring viruses that can kill bacteria with high specificity and can evolve to target resistant bacteria. Phages have been historically employed as antimicrobial agents, but they were overshadowed by the emergence of antibiotics. With a renewed focus on phages, it is important to study their clinical efficacy, safety, and formulation. Pulmonary infections have a large burden of global morbidity and frequently involve multidrug-resistant pathogens such as Acinetobacter baumannii, Klebsiella pneumoniae, Mycobacterium tuberculosis, and Pseudomonas aeruginosa. Therefore, this can be an important area of application of phages. Dry powder inhalers can be an effective strategy to deliver phages to the lungs because they are easy-to-use, portable, and capable of delivering a higher lung dose than oral or intravenous route. They also have longer shelf life and lower cold storage requirements than solutions. Therefore, the aim of the current review is to summarize recent findings on bacteriophage dry powder formulations, particularly focusing on the effect of various excipients and manufacturing factors on phage titer preservation.
Collapse
Affiliation(s)
- Vaibhav Pathak
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, USA
| | - Hak-Kim Chan
- Advanced Drug Delivery Group, School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Australia
| | - Qi Tony Zhou
- Department of Industrial and Molecular Pharmaceutics, College of Pharmacy, Purdue University, USA
| |
Collapse
|
45
|
Ssekatawa K, Ntulume I, Byarugaba DK, Michniewski S, Jameson E, Wampande EM, Nakavuma J. Isolation and Characterization of Novel Lytic Bacteriophages Infecting Carbapenem-Resistant Pathogenic Diarrheagenic and Uropathogenic Escherichia Coli. Infect Drug Resist 2024; 17:3367-3384. [PMID: 39135747 PMCID: PMC11317518 DOI: 10.2147/idr.s466101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 07/31/2024] [Indexed: 08/15/2024] Open
Abstract
Background The evolution of antimicrobial resistance has dramatically reduced the efficacy of the first-choice and last-resort antibiotics used to treat E. coli infections. Thus, searching for novel therapeutics to treat and control the emergence of antibiotic resistance is urgent. Therefore, this study aimed to illustrate the lytic effect of phages against carbapenem-resistant pathogenic E. coli. Methods Phages were isolated from hospital effluents by the enrichment assay. This was followed by the evaluation of the host range of the phages by the spot assay. The time taken by phages to bind to the host bacterial cells was determined by the adsorption assay. The phage latent period and burst size were determined using a one-step growth experiment. Phage morphology was determined by the Transmission Electron Microscopy. Molecular characterization of phages was done by whole genome sequencing. Results Two phages named UGKSEcP1 and UGKSEcP2 were isolated from hospital effluents. The phages were professionally lytic with a broad host range. The two phages recorded an average adsorption time of 11.25 minutes, an adsorption rate of 99.3%, a latency period of 20 minutes, and a burst size of approximately 528 phages/infected cell. Phages UGKSEcP1 and UGKSEcP2 had genome lengths of 167433bp, and 167221bp with 277 and 276 predicted genes, respectively, and no undesirable genes were detected. Phylogenetic analysis revealed the two phages belonged genus Tequatrovirus. TEM micrograph showed that the two phages had a similar morphotype with icosahedral heads and contractile tails; thus, classified as members of the Myoviridae phage family. Conclusion The findings demonstrate that the study isolated two novel professionally lytic phages with a broad host range and thus, are candidates for phage-mediated biocontrol.
Collapse
Affiliation(s)
- Kenneth Ssekatawa
- Department of Science Technical and Vocational Education, Makerere University, Kampala, Uganda
- Africa Center of Excellence in Materials, Product Development and Nanotechnology (MAPRONANO ACE), Makerere University, Kampala, Uganda
| | - Ibrahim Ntulume
- Department of Biotechnical and Diagnostic Sciences, Makerere University, Kampala, Uganda
| | | | | | - Eleanor Jameson
- School of Environmental and Natural Sciences, Bangor University, Gwynedd, UK
| | - Eddie M Wampande
- Department of Veterinary Pharmacy, Clinical and Comparative Medicine, Makerere University, Kampala, Uganda
| | - Jesca Nakavuma
- Department of Biotechnical and Diagnostic Sciences, Makerere University, Kampala, Uganda
| |
Collapse
|
46
|
Kakkar A, Kandwal G, Nayak T, Jaiswal LK, Srivastava A, Gupta A. Engineered bacteriophages: A panacea against pathogenic and drug resistant bacteria. Heliyon 2024; 10:e34333. [PMID: 39100447 PMCID: PMC11295868 DOI: 10.1016/j.heliyon.2024.e34333] [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: 04/11/2024] [Revised: 06/18/2024] [Accepted: 07/08/2024] [Indexed: 08/06/2024] Open
Abstract
Antimicrobial resistance (AMR) is a major global concern; antibiotics and other regular treatment methods have failed to overcome the increasing number of infectious diseases. Bacteriophages (phages) are viruses that specifically target/kill bacterial hosts without affecting other human microbiome. Phage therapy provides optimism in the current global healthcare scenario with a long history of its applications in humans that has now reached various clinical trials. Phages in clinical trials have specific requirements of being exclusively lytic, free from toxic genes with an enhanced host range that adds an advantage to this requisite. This review explains in detail the various phage engineering methods and their potential applications in therapy. To make phages more efficient, engineering has been attempted using techniques like conventional homologous recombination, Bacteriophage Recombineering of Electroporated DNA (BRED), clustered regularly interspaced short palindromic repeats (CRISPR)-Cas, CRISPY-BRED/Bacteriophage Recombineering with Infectious Particles (BRIP), chemically accelerated viral evolution (CAVE), and phage genome rebooting. Phages are administered in cocktail form in combination with antibiotics, vaccines, and purified proteins, such as endolysins. Thus, phage therapy is proving to be a better alternative for treating life-threatening infections, with more specificity and fewer detrimental consequences.
Collapse
Affiliation(s)
- Anuja Kakkar
- Molecular Microbiology Laboratory, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, UP, 221005, India
| | - Garima Kandwal
- Molecular Microbiology Laboratory, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, UP, 221005, India
| | - Tanmayee Nayak
- Molecular Microbiology Laboratory, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, UP, 221005, India
| | - Lav Kumar Jaiswal
- Molecular Microbiology Laboratory, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, UP, 221005, India
| | - Amit Srivastava
- University of Jyväskylä, Nanoscience Centre, Department of Biological and Environmental Science, 40014, Jyväskylä, Finland
| | - Ankush Gupta
- Molecular Microbiology Laboratory, Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, UP, 221005, India
| |
Collapse
|
47
|
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
|
48
|
Yang S, Meng X, Zhen Y, Baima Q, Wang Y, Jiang X, Xu Z. Strategies and mechanisms targeting Enterococcus faecalis biofilms associated with endodontic infections: a comprehensive review. Front Cell Infect Microbiol 2024; 14:1433313. [PMID: 39091674 PMCID: PMC11291369 DOI: 10.3389/fcimb.2024.1433313] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/05/2024] [Indexed: 08/04/2024] Open
Abstract
Enterococcus faecalis is one of the main microorganisms that infects root canals, ranking among the most prevalent microorganisms associated with endodontic treatment failure. Given its pervasive presence in persistent endodontic infections, the successful elimination of Enterococcus faecalis is crucial for effective endodontic treatment and retreatment. Furthermore, Enterococcus faecalis can form biofilms - defense structures that microbes use to fight environmental threats. These biofilms confer resistance against host immune system attacks and antibiotic interventions. Consequently, the presence of biofilms poses a significant challenge in the complete eradication of Enterococcus faecalis and its associated disease. In response, numerous scholars have discovered promising outcomes in addressing Enterococcus faecalis biofilms within root canals and undertaken endeavors to explore more efficacious approaches in combating these biofilms. This study provides a comprehensive review of strategies and mechanisms for the removal of Enterococcus faecalis biofilms.
Collapse
Affiliation(s)
- Shipeng Yang
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Xiuping Meng
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yuqi Zhen
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Quzhen Baima
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yu Wang
- Department of Dental Implantology, Hospital of Stomatology, Jilin University, Changchun, China
| | - Xinmiao Jiang
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| | - Zhibo Xu
- Department of Endodontics, Hospital of Stomatology, Jilin University, Changchun, China
| |
Collapse
|
49
|
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] [Download PDF] [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
|
50
|
Lewis JM, Williams J, Sagona AP. Making the leap from technique to treatment - genetic engineering is paving the way for more efficient phage therapy. Biochem Soc Trans 2024; 52:1373-1384. [PMID: 38716972 PMCID: PMC11346441 DOI: 10.1042/bst20231289] [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: 01/16/2024] [Revised: 03/30/2024] [Accepted: 04/29/2024] [Indexed: 06/27/2024]
Abstract
Bacteriophages (phages) are viruses specific to bacteria that target them with great efficiency and specificity. Phages were first studied for their antibacterial potential in the early twentieth century; however, their use was largely eclipsed by the popularity of antibiotics. Given the surge of antimicrobial-resistant strains worldwide, there has been a renaissance in harnessing phages as therapeutics once more. One of the key advantages of phages is their amenability to modification, allowing the generation of numerous derivatives optimised for specific functions depending on the modification. These enhanced derivatives could display higher infectivity, expanded host range or greater affinity to human tissues, where some bacterial species exert their pathogenesis. Despite this, there has been a noticeable discrepancy between the generation of derivatives in vitro and their clinical application in vivo. In most instances, phage therapy is only used on a compassionate-use basis, where all other treatment options have been exhausted. A lack of clinical trials and numerous regulatory hurdles hamper the progress of phage therapy and in turn, the engineered variants, in becoming widely used in the clinic. In this review, we outline the various types of modifications enacted upon phages and how these modifications contribute to their enhanced bactericidal function compared with wild-type phages. We also discuss the nascent progress of genetically modified phages in clinical trials along with the current issues these are confronted with, to validate it as a therapy in the clinic.
Collapse
Affiliation(s)
| | - Joshua Williams
- School of Life Sciences, University of Warwick, Coventry, U.K
| | | |
Collapse
|