Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 299
Filter
1.
Arch Virol ; 169(8): 156, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38967872

ABSTRACT

Infections caused by multidrug-resistant (MDR) bacteria are a growing global concern. Enterobacter cloacae complex (ECC) species are particularly adept at developing antibiotic resistance. Phage therapy is proposed as an alternative treatment for pathogens that no longer respond to antibiotics. Unfortunately, ECC phages are understudied when compared to phages of many other bacterial species. In this Ghanaian-Finnish study, we isolated two ECC strains from ready-to-eat food samples and three novel phages from natural waters against these strains. We sequenced the genomic DNA of the novel Enterobacter phages, fGh-Ecl01, fGh-Ecl02, and fGh-Ecl04, and assessed their therapeutic potential. All of the phages were found to be lytic, easy to propagate, and lacking any toxic, integrase, or antibiotic resistance genes and were thus considered suitable for therapy purposes. They all were found to be related to T4-type viruses: fGh-Ecl01 and fGh-Ecl04 to karamviruses and fGh-Ecl02 to agtreviruses. Testing of Finnish clinical ECC strains showed promising susceptibility to these novel phages. As many as 61.1% of the strains were susceptible to fGh-Ecl01 and fGh-Ecl04, and 7.4% were susceptible to fGh-Ecl02. Finally, we investigated the susceptibility of the newly isolated ECC strains to three antibiotics - meropenem, ciprofloxacin, and cefepime - in combination with the novel phages. The use of phages and antibiotics together had synergistic effects. When using an antibiotic-phage combination, even low concentrations of antibiotics fully inhibited the growth of bacteria.


Subject(s)
Anti-Bacterial Agents , Bacteriophages , Enterobacter cloacae , Enterobacter cloacae/virology , Enterobacter cloacae/drug effects , Ghana , Bacteriophages/genetics , Bacteriophages/isolation & purification , Bacteriophages/physiology , Bacteriophages/classification , Anti-Bacterial Agents/pharmacology , Phage Therapy/methods , Genome, Viral , Enterobacteriaceae Infections/therapy , Enterobacteriaceae Infections/microbiology , Drug Resistance, Multiple, Bacterial , Finland , Humans , Microbial Sensitivity Tests , Ciprofloxacin/pharmacology , Meropenem/pharmacology
2.
Arch Virol ; 169(7): 142, 2024 Jun 08.
Article in English | MEDLINE | ID: mdl-38851653

ABSTRACT

Uropathogenic Escherichia coli (UPEC) is the most common causative agent of urinary tract infections, and strains that are resistant to antibiotics are a major problem in treating these infections. Phage therapy is a promising alternative approach that can be used to treat infections caused by polyresistant bacterial strains. In the present study, 16 bacteriophages isolated from sewage and surface water were investigated. Phage host specificity was tested on a collection of 77 UPEC strains. The phages infected 2-44 strains, and 80% of the strains were infected by at least one phage. The susceptible E. coli strains belonged predominantly to the B2 phylogenetic group, including strains of two clones, CC131 and CC73, that have a worldwide distribution. All of the phages belonged to class Caudoviricetes and were identified as members of the families Straboviridae, Autographiviridae, and Drexlerviridae and the genera Kagunavirus, Justusliebigvirus, and Murrayvirus. A phage cocktail composed of six phages - four members of the family Straboviridae and two members of the family Autographiviridae - was prepared, and its antibacterial activity was tested in liquid medium. Complete suppression of bacterial growth was observed after 5-22 hours of cultivation, followed by partial regrowth. At 24 hours postinfection, the cocktail suppressed bacterial growth to 43-92% of control values. Similar results were obtained when testing the activity of the phage cocktail in LB and in artificial urine medium. The results indicate that our phage cocktail has potential to inhibit bacterial growth during infection, and they will therefore be preserved in the national phage bank, serving as valuable resources for therapeutic applications.


Subject(s)
Drug Resistance, Multiple, Bacterial , Host Specificity , Phylogeny , Uropathogenic Escherichia coli , Uropathogenic Escherichia coli/virology , Uropathogenic Escherichia coli/drug effects , Bacteriophages/classification , Bacteriophages/physiology , Bacteriophages/genetics , Bacteriophages/isolation & purification , Sewage/virology , Phage Therapy/methods , Humans , Anti-Bacterial Agents/pharmacology , Escherichia coli Infections/microbiology , Escherichia coli Infections/therapy
3.
Science ; 384(6701): eado0713, 2024 Jun 14.
Article in English | MEDLINE | ID: mdl-38870284

ABSTRACT

Bacteria can repurpose their own bacteriophage viruses (phage) to kill competing bacteria. Phage-derived elements are frequently strain specific in their killing activity, although there is limited evidence that this specificity drives bacterial population dynamics. Here, we identified intact phage and their derived elements in a metapopulation of wild plant-associated Pseudomonas genomes. We discovered that the most abundant viral cluster encodes a phage remnant resembling a phage tail called a tailocin, which bacteria have co-opted to kill bacterial competitors. Each pathogenic Pseudomonas strain carries one of a few distinct tailocin variants that target the variable polysaccharides in the outer membrane of co-occurring pathogenic Pseudomonas strains. Analysis of herbarium samples from the past 170 years revealed that the same tailocin and bacterial receptor variants have persisted in Pseudomonas populations. These results suggest that tailocin genetic diversity can be mined to develop targeted "tailocin cocktails" for microbial control.


Subject(s)
Bacteriocins , Pseudomonas Phages , Pseudomonas , Viral Tail Proteins , Antibiosis , Bacterial Outer Membrane/metabolism , Bacteriocins/genetics , Bacteriocins/metabolism , Genetic Variation , Genome, Bacterial , Polysaccharides, Bacterial/metabolism , Pseudomonas/metabolism , Pseudomonas/virology , Pseudomonas Phages/genetics , Pseudomonas Phages/metabolism , Viral Tail Proteins/metabolism , Viral Tail Proteins/genetics , Phage Therapy/methods
4.
Nat Microbiol ; 9(6): 1434-1453, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38834776

ABSTRACT

In contrast to the many reports of successful real-world cases of personalized bacteriophage therapy (BT), randomized controlled trials of non-personalized bacteriophage products have not produced the expected results. Here we present the outcomes of a retrospective observational analysis of the first 100 consecutive cases of personalized BT of difficult-to-treat infections facilitated by a Belgian consortium in 35 hospitals, 29 cities and 12 countries during the period from 1 January 2008 to 30 April 2022. We assessed how often personalized BT produced a positive clinical outcome (general efficacy) and performed a regression analysis to identify functional relationships. The most common indications were lower respiratory tract, skin and soft tissue, and bone infections, and involved combinations of 26 bacteriophages and 6 defined bacteriophage cocktails, individually selected and sometimes pre-adapted to target the causative bacterial pathogens. Clinical improvement and eradication of the targeted bacteria were reported for 77.2% and 61.3% of infections, respectively. In our dataset of 100 cases, eradication was 70% less probable when no concomitant antibiotics were used (odds ratio = 0.3; 95% confidence interval = 0.127-0.749). In vivo selection of bacteriophage resistance and in vitro bacteriophage-antibiotic synergy were documented in 43.8% (7/16 patients) and 90% (9/10) of evaluated patients, respectively. We observed a combination of antibiotic re-sensitization and reduced virulence in bacteriophage-resistant bacterial isolates that emerged during BT. Bacteriophage immune neutralization was observed in 38.5% (5/13) of screened patients. Fifteen adverse events were reported, including seven non-serious adverse drug reactions suspected to be linked to BT. While our analysis is limited by the uncontrolled nature of these data, it indicates that BT can be effective in combination with antibiotics and can inform the design of future controlled clinical trials. BT100 study, ClinicalTrials.gov registration: NCT05498363 .


Subject(s)
Anti-Bacterial Agents , Bacterial Infections , Bacteriophages , Phage Therapy , Humans , Retrospective Studies , Phage Therapy/methods , Bacteriophages/physiology , Bacteriophages/genetics , Female , Male , Middle Aged , Anti-Bacterial Agents/therapeutic use , Adult , Bacterial Infections/therapy , Treatment Outcome , Aged , Precision Medicine/methods , Adolescent , Young Adult , Bacteria/virology , Bacteria/genetics , Child , Aged, 80 and over , Child, Preschool , Belgium , Infant
5.
Eur Respir Rev ; 33(172)2024 Apr.
Article in English | MEDLINE | ID: mdl-38925791

ABSTRACT

Lower respiratory tract infections (LRTIs) present a significant global health burden, exacerbated by the rise in antimicrobial resistance (AMR). The persistence and evolution of multidrug-resistant bacteria intensifies the urgency for alternative treatments. This review explores bacteriophage (phage) therapy as an innovative solution to combat bacterial LRTIs. Phages, abundant in nature, demonstrate specificity towards bacteria, minimal eukaryotic toxicity, and the ability to penetrate and disrupt bacterial biofilms, offering a targeted approach to infection control. The article synthesises evidence from systematic literature reviews spanning 2000-2023, in vitro and in vivo studies, case reports and ongoing clinical trials. It highlights the synergistic potential of phage therapy with antibiotics, the immunophage synergy in animal models, and the pharmacodynamics and pharmacokinetics critical for clinical application. Despite promising results, the article acknowledges that phage therapy is at a nascent stage in clinical settings, the challenges of phage-resistant bacteria, and the lack of comprehensive cost-effectiveness studies. It stresses the need for further research to optimise phage therapy protocols and navigate the complexities of phage-host interactions, particularly in vulnerable populations such as the elderly and immunocompromised. We call for regulatory adjustments to facilitate the exploration of the long-term effects of phage therapy, aiming to incorporate this old-yet-new therapy into mainstream clinical practice to tackle the looming AMR crisis.


Subject(s)
Anti-Bacterial Agents , Bacteriophages , Phage Therapy , Respiratory Tract Infections , Humans , Phage Therapy/methods , Respiratory Tract Infections/therapy , Respiratory Tract Infections/microbiology , Respiratory Tract Infections/virology , Animals , Anti-Bacterial Agents/therapeutic use , Treatment Outcome , Bacterial Infections/therapy , Bacterial Infections/microbiology , Bacteria/virology , Host-Pathogen Interactions
6.
Nat Commun ; 15(1): 4764, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38834561

ABSTRACT

Bacteriophage are sophisticated cellular parasites that can not only parasitize bacteria but are increasingly recognized for their direct interactions with mammalian hosts. Phage adherence to mucus is known to mediate enhanced antimicrobial effects in vitro. However, little is known about the therapeutic efficacy of mucus-adherent phages in vivo. Here, using a combination of in vitro gastrointestinal cell lines, a gut-on-a-chip microfluidic model, and an in vivo murine gut model, we demonstrated that a E. coli phage, øPNJ-6, provided enhanced gastrointestinal persistence and antimicrobial effects. øPNJ-6 bound fucose residues, of the gut secreted glycoprotein MUC2, through domain 1 of its Hoc protein, which led to increased intestinal mucus production that was suggestive of a positive feedback loop mediated by the mucus-adherent phage. These findings extend the Bacteriophage Adherence to Mucus model into phage therapy, demonstrating that øPNJ-6 displays enhanced persistence within the murine gut, leading to targeted depletion of intestinal pathogenic bacteria.


Subject(s)
Escherichia coli Infections , Escherichia coli , Intestinal Mucosa , Mucin-2 , Animals , Escherichia coli/virology , Mice , Intestinal Mucosa/microbiology , Intestinal Mucosa/virology , Mucin-2/metabolism , Humans , Escherichia coli Infections/microbiology , Escherichia coli Infections/therapy , Phage Therapy/methods , Bacterial Adhesion , Female , Mucus/metabolism , Mucus/virology , Coliphages/physiology , Fucose/metabolism , Mice, Inbred C57BL
9.
Biochem Soc Trans ; 52(3): 1373-1384, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38716972

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.


Subject(s)
Bacteriophages , Genetic Engineering , Phage Therapy , Phage Therapy/methods , Humans , Bacteriophages/genetics , Bacterial Infections/therapy , Bacteria/virology , Bacteria/genetics , Animals , Anti-Bacterial Agents/therapeutic use
10.
Life Sci ; 350: 122749, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38821215

ABSTRACT

Emergence of antimicrobial-resistant bacteria (AMR) is one of the health major problems worldwide. The scientists are looking for a novel method to treat infectious diseases. Phage therapy is considered a suitable approach for treating infectious diseases. However, there are different challenges in this way. Some biological aspects can probably influence on therapeutic results and further investigations are necessary to reach a successful phage therapy. Bacteriophage activity can influence by bacterial defense system. Bacterial extracellular vesicles (BEVs) are one of the bacterial defense mechanisms which can modify the results of bacteriophage activity. BEVs have the significant roles in the gene transferring, invasion, escape, and spreading of bacteriophages. In this review, the defense mechanisms of bacteria against bacteriophages, especially BEVs secretion, the hidden linkage of BEVs and bacteriophages, and its possible consequences on the bacteriophage activity as well phage therapy will be discussed.


Subject(s)
Bacteria , Bacteriophages , Extracellular Vesicles , Phage Therapy , Bacteriophages/physiology , Bacteria/virology , Humans , Phage Therapy/methods , Bacterial Infections/therapy , Bacterial Infections/microbiology , Animals
11.
STAR Protoc ; 5(2): 102949, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38691464

ABSTRACT

Phage therapy has re-emerged as a promising treatment for non-resolving infections. Given the lack of approved phage treatments, there is a need to establish a compassionate use pipeline. Here, we present a protocol for phage matching, treatment, and monitoring for compassionate bacteriophage use in non-resolving infections. We describe steps for consultation and request implementation, evaluating and comparing different aspects of phage activity, and phage production. We then detail procedures for multidisciplinary meetings, ethics approvals, phage therapy, and follow-up. For complete details on the use and execution of this protocol, please refer to Onallah et al.1,2.


Subject(s)
Bacteriophages , Compassionate Use Trials , Phage Therapy , Humans , Bacteriophages/physiology , Phage Therapy/methods , Bacterial Infections/therapy
12.
Virology ; 595: 110098, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38705084

ABSTRACT

Acinetobacter baumannii is one of the most important pathogens of healthcare-associated infections. The rising prevalence of multidrug-resistant A. baumannii (MRAB) strains and biofilm formation impact the outcome of conventional treatment. Phage-related therapy is a promising strategy to tame troublesome multidrug-resistant bacteria. Here, we isolated and evaluated a highly efficient lytic phage called MRABP9 from hospital sewage. The phage was a novel species within the genus Friunavirus and exhibited lytic activity against 2 other identified MRAB strains. Genomic analysis revealed it was a safe virulent phage and a pectate lyase domain was identified within its tail spike protein. MRABP9 showed potent bactericidal and anti-biofilm activity against MRAB, significantly delaying the time point of bacterial regrowth in vitro. Phage administration could rescue the mice from acute lethal MRAB infection. Considering its features, MRABP9 has the potential as an efficient candidate for prophylactic and therapeutic use against acute infections caused by MRAB strains.


Subject(s)
Acinetobacter Infections , Acinetobacter baumannii , Bacteriophages , Drug Resistance, Multiple, Bacterial , Phage Therapy , Acinetobacter baumannii/virology , Acinetobacter baumannii/drug effects , Animals , Acinetobacter Infections/microbiology , Acinetobacter Infections/therapy , Mice , Bacteriophages/genetics , Bacteriophages/physiology , Phage Therapy/methods , Genome, Viral , Biofilms/drug effects , Biofilms/growth & development , Humans , Female , Sewage/virology
13.
J Virol ; 98(6): e0027224, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38771043

ABSTRACT

Klebsiella spp. are causative agents of healthcare-associated infections in patients who are immunocompromised and use medical devices. The antibiotic resistance crisis has led to an increase in infections caused by these bacteria, which can develop into potentially life-threatening illnesses if not treated swiftly and effectively. Thus, new treatment options for Klebsiella are urgently required. Phage therapy can offer an alternative to ineffective antibiotic treatments for antibiotic-resistant bacteria infections. The aim of the present study was to produce a safe and effective phage cocktail treatment against Klebsiella pneumoniae and Klebsiella oxytoca, both in liquid in vitro culture and an in vivo Galleria mellonella infection model. The phage cocktail was significantly more effective at killing K. pneumoniae and K. oxytoca strains compared with monophage treatments. Preliminary phage cocktail safety was demonstrated through application in the in vivo G. mellonella model: where the phage cocktail induced no toxic side effects in G. mellonella. In addition, the phage cocktail significantly improved the survival of G. mellonella when administered as a prophylactic treatment, compared with controls. In conclusion, our phage cocktail was demonstrated to be safe and effective against Klebsiella spp. in the G. mellonella infection model. This provides a strong case for future treatment for Klebsiella infections, either as an alternative or adjunct to antibiotics.IMPORTANCEKlebsiella infections are a concern in individuals who are immunocompromised and are becoming increasingly difficult to treat with antibiotics due to their drug-resistant properties. Bacteriophage is one potential alternative therapy that could be used to tackle these infections. The present study describes the design of a non-toxic phage cocktail that improved the survival of Galleria mellonella infected with Klebsiella. This phage cocktail demonstrates potential for the safe and effective treatment of Klebsiella infections, as an adjunct or alternative to antibiotics.


Subject(s)
Bacteriophages , Klebsiella Infections , Klebsiella oxytoca , Klebsiella pneumoniae , Phage Therapy , Animals , Klebsiella Infections/therapy , Klebsiella Infections/microbiology , Bacteriophages/physiology , Phage Therapy/methods , Klebsiella pneumoniae/virology , Klebsiella oxytoca/virology , Moths/microbiology , Moths/virology , Klebsiella/virology , Disease Models, Animal , Larva/microbiology , Larva/virology , Lepidoptera/microbiology , Lepidoptera/virology
14.
Curr Opin Microbiol ; 79: 102482, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38714140

ABSTRACT

Phage therapy holds much promise as an alternative to antibiotics for fighting infection. However, this approach is no panacea as recent results show that a small fraction of cells survives lytic phage infection due to both dormancy (i.e. formation of persister cells) and resistance (genetic change). In this brief review, we summarize evidence suggesting phages induce the persister state. Therefore, it is predicted that phage cocktails should be combined with antipersister compounds to eradicate bacterial infections.


Subject(s)
Bacteria , Bacterial Infections , Bacteriophages , Phage Therapy , Bacteriophages/physiology , Bacteriophages/genetics , Phage Therapy/methods , Bacteria/virology , Bacteria/drug effects , Bacteria/genetics , Bacterial Infections/microbiology , Bacterial Infections/therapy , Anti-Bacterial Agents/pharmacology , Humans
15.
Mil Med ; 189(7-8): e1488-e1496, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38780999

ABSTRACT

INTRODUCTION: Antibacterial resistance is an emerging problem in military medicine. Disruptions to the health care systems in war-torn countries that result from ongoing conflict can potentially exacerbate this problem and increase the risk to U.S. forces in the deployed environment. Therefore, novel therapies are needed to mitigate the impact of these potentially devastating infections on military operations. Bacteriophages are viruses that infect and kill bacteria. They can be delivered as therapeutic agents and offer a promising alternative to traditional antibiotic chemotherapy. There are several potential benefits to their use, including high specificity and comparative ease of use in the field setting. However, the process of engineering phages for military medical applications can be a laborious and time-consuming endeavor. This review examines available techniques and compares their efficacy. MATERIALS AND METHODS: This review evaluates the scientific literature on the development and application of four methods of bacteriophage genome engineering and their consideration in the context of military applications. Preffered Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were followed for a systematic review of available literature that met criteria for analysis and inclusion. The research completed for this review article originated from the United States Military Academy's library "Scout" search engine, which compiles results from 254 available databases (including PubMed, Google Scholar, and SciFinder). Particular attention was focused on identifying useful mechanistic insight into the nature of the engineering technique, the ease of use, and the applicability of the technique to countering the problem of antimicrobial resistance in the military setting. RESULTS: A total of 52 studies were identified that met inclusion criteria following PRISMA guidelines. The bioengineering techniques analyzed included homologous recombination (12 articles), in vivo recombineering (9 articles), bacteriophage recombineering of electroporated DNA (7 articles), and the CRISPR-Cas system (10 articles). Rates of success and fidelity varied across each platform, and comparative benefits and drawbacks are considered. CONCLUSIONS: Each of the phage engineering techniques addressed herein varies in amount of effort and overall success rate. CRISPR-Cas-facilitated modification of phage genomes presents a highly efficient method that does not require a lengthy purification and screening process. It therefore appears to be the method best suited for military medical applications.


Subject(s)
Bacteriophages , Genetic Engineering , Bacteriophages/genetics , Humans , Genetic Engineering/methods , Host Specificity , Phage Therapy/methods
16.
PLoS One ; 19(5): e0303056, 2024.
Article in English | MEDLINE | ID: mdl-38713691

ABSTRACT

With the global challenge of antimicrobial resistance (AMR), interest in the development of antibiotic alternatives has surged worldwide. While phage therapy is not a new phenomenon, technological and socio-economic factors have limited its implementation in the Western world. There is now a resurged effort, especially in the UK, to address these challenges. In this study, we collect survey data on UK general practitioners (n = 131) and other healthcare professionals (n = 103), as well as interviews with medical professionals (n = 4) and a focus group with medical students (n = 6) to explore factors associated with their willingness to prescribe phage therapy to patients. The interviews with medical professionals show support for the expansion of bacteriophage clinical trials and highlight their role as a viable alternative to antibiotics. A conjoint experiment reveals that success rate, side effect rate, and patient attitude to treatment are the decisive factors when it comes to phage therapy prescription; in contrast, the effects of administration route, type of treatment, and severity of infection were not statistically significant. Moreover, we show that general practitioners overall are more likely to recommend phage treatment to patients, compared to other healthcare professionals. The results of the study suggest that phage therapy has a potential to be widely accepted and used by healthcare workers in the UK.


Subject(s)
Phage Therapy , Humans , United Kingdom , Phage Therapy/methods , Female , Male , Health Personnel/psychology , Adult , Surveys and Questionnaires , Middle Aged , Practice Patterns, Physicians'/statistics & numerical data , Attitude of Health Personnel
17.
Medicina (Kaunas) ; 60(5)2024 May 09.
Article in English | MEDLINE | ID: mdl-38792972

ABSTRACT

Background and Objectives: Despite the promise of phage therapy (PT), its efficacy in prosthetic joint infection (PJI) management is unknown. Much of the current literature is largely limited to case reports and series. Materials and Methods: In order to help inform power calculations for future clinical trials and comparative analyses, we performed a systematic review and proportional meta-analysis of early PT outcomes to provide a preliminary assessment of early phage therapy treatment outcomes for cases of PJI. Results: In a search of available literature across MEDLINE (Ovid, Wolters Kluwer, Alphen aan den Rijn, The Netherlands), Embase (Elsevier, Amsterdam, The Netherlands), the Web of Science Core Collection (Clarivate, London, UK), and Cochrane Central (Wiley, Hoboken, NJ, USA) up to 23 September 2023, we identified 37 patients with PJIs receiving adjunctive PT. Patients most frequently reported Staphylococcal species infection (95%) and intraarticular phage delivery (73%). Phage cocktail (65%) and antibiotic co-administration (97%) were common. A random-effects proportional meta-analysis suggested infection remission in 78% of patients (95% CI: 39%, 95%) (I2 = 55%, p = 0.08) and 83% with a minimum 12-month follow-up (95% CI: 53%, 95%) (I2 = 26%, p = 0.26). Conclusions: Our study provides a preliminary estimate of PT's efficacy in PJIs and informs future comparative studies.


Subject(s)
Phage Therapy , Prosthesis-Related Infections , Humans , Prosthesis-Related Infections/therapy , Phage Therapy/methods , Treatment Outcome
18.
Int J Mol Sci ; 25(10)2024 May 17.
Article in English | MEDLINE | ID: mdl-38791502

ABSTRACT

Wound infection is one of the most important factors affecting wound healing, so its effective control is critical to promote the process of wound healing. However, with the increasing prevalence of multi-drug-resistant (MDR) bacterial strains, the prevention and treatment of wound infections are now more challenging, imposing heavy medical and financial burdens on patients. Furthermore, the diminishing effectiveness of conventional antimicrobials and the declining research on new antibiotics necessitate the urgent exploration of alternative treatments for wound infections. Recently, phage therapy has been revitalized as a promising strategy to address the challenges posed by bacterial infections in the era of antibiotic resistance. The use of phage therapy in treating infectious diseases has demonstrated positive results. This review provides an overview of the mechanisms, characteristics, and delivery methods of phage therapy for combating pathogenic bacteria. Then, we focus on the clinical application of various phage therapies in managing refractory wound infections, such as diabetic foot infections, as well as traumatic, surgical, and burn wound infections. Additionally, an analysis of the potential obstacles and challenges of phage therapy in clinical practice is presented, along with corresponding strategies for addressing these issues. This review serves to enhance our understanding of phage therapy and provides innovative avenues for addressing refractory infections in wound healing.


Subject(s)
Phage Therapy , Wound Infection , Phage Therapy/methods , Humans , Wound Infection/therapy , Wound Infection/microbiology , Wound Healing , Bacterial Infections/therapy , Bacterial Infections/microbiology , Bacteriophages/physiology , Animals , Anti-Bacterial Agents/therapeutic use , Drug Resistance, Multiple, Bacterial
19.
Front Immunol ; 15: 1398652, 2024.
Article in English | MEDLINE | ID: mdl-38779682

ABSTRACT

In the advancement of Inflammatory Bowel Disease (IBD) treatment, existing therapeutic methods exhibit limitations; they do not offer a complete cure for IBD and can trigger adverse side effects. Consequently, the exploration of novel therapies and multifaceted treatment strategies provides patients with a broader range of options. Within the framework of IBD, gut microbiota plays a pivotal role in disease onset through diverse mechanisms. Bacteriophages, as natural microbial regulators, demonstrate remarkable specificity by accurately identifying and eliminating specific pathogens, thus holding therapeutic promise. Although clinical trials have affirmed the safety of phage therapy, its efficacy is prone to external influences during storage and transport, which may affect its infectivity and regulatory roles within the microbiota. Improving the stability and precise dosage control of bacteriophages-ensuring robustness in storage and transport, consistent dosing, and targeted delivery to infection sites-is crucial. This review thoroughly explores the latest developments in IBD treatment and its inherent challenges, focusing on the interaction between the microbiota and bacteriophages. It highlights bacteriophages' potential as microbiome modulators in IBD treatment, offering detailed insights into research on bacteriophage encapsulation and targeted delivery mechanisms. Particular attention is paid to the functionality of various carrier systems, especially regarding their protective properties and ability for colon-specific delivery. This review aims to provide a theoretical foundation for using bacteriophages as microbiome modulators in IBD treatment, paving the way for enhanced regulation of the intestinal microbiota.


Subject(s)
Bacteriophages , Gastrointestinal Microbiome , Inflammatory Bowel Diseases , Phage Therapy , Humans , Phage Therapy/methods , Inflammatory Bowel Diseases/therapy , Bacteriophages/physiology , Animals
20.
Bone Joint J ; 106-B(6): 522-524, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38821504
SELECTION OF CITATIONS
SEARCH DETAIL
...