Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 288
Filtrar
1.
Nat Microbiol ; 9(6): 1434-1453, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38834776

RESUMEN

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 .


Asunto(s)
Antibacterianos , Infecciones Bacterianas , Bacteriófagos , Terapia de Fagos , Humanos , Estudios Retrospectivos , Terapia de Fagos/métodos , Bacteriófagos/fisiología , Bacteriófagos/genética , Femenino , Masculino , Persona de Mediana Edad , Antibacterianos/uso terapéutico , Adulto , Infecciones Bacterianas/terapia , Resultado del Tratamiento , Anciano , Medicina de Precisión/métodos , Adolescente , Adulto Joven , Bacterias/virología , Bacterias/genética , Niño , Anciano de 80 o más Años , Preescolar , Bélgica , Lactante
4.
Nat Commun ; 15(1): 4764, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38834561

RESUMEN

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.


Asunto(s)
Infecciones por Escherichia coli , Escherichia coli , Mucosa Intestinal , Mucina 2 , Animales , Escherichia coli/virología , Ratones , Mucosa Intestinal/microbiología , Mucosa Intestinal/virología , Mucina 2/metabolismo , Humanos , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/terapia , Terapia de Fagos/métodos , Adhesión Bacteriana , Femenino , Moco/metabolismo , Moco/virología , Colifagos/fisiología , Fucosa/metabolismo , Ratones Endogámicos C57BL
5.
PLoS One ; 19(5): e0303056, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38713691

RESUMEN

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.


Asunto(s)
Terapia de Fagos , Humanos , Reino Unido , Terapia de Fagos/métodos , Femenino , Masculino , Personal de Salud/psicología , Adulto , Encuestas y Cuestionarios , Persona de Mediana Edad , Pautas de la Práctica en Medicina/estadística & datos numéricos , Actitud del Personal de Salud
6.
Front Immunol ; 15: 1398652, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38779682

RESUMEN

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.


Asunto(s)
Bacteriófagos , Microbioma Gastrointestinal , Enfermedades Inflamatorias del Intestino , Terapia de Fagos , Humanos , Terapia de Fagos/métodos , Enfermedades Inflamatorias del Intestino/terapia , Bacteriófagos/fisiología , Animales
7.
Bone Joint J ; 106-B(6): 522-524, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38821504
8.
Int J Mol Sci ; 25(10)2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38791502

RESUMEN

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.


Asunto(s)
Terapia de Fagos , Infección de Heridas , Terapia de Fagos/métodos , Humanos , Infección de Heridas/terapia , Infección de Heridas/microbiología , Cicatrización de Heridas , Infecciones Bacterianas/terapia , Infecciones Bacterianas/microbiología , Bacteriófagos/fisiología , Animales , Antibacterianos/uso terapéutico , Farmacorresistencia Bacteriana Múltiple
9.
Microbiol Spectr ; 12(6): e0042723, 2024 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-38695573

RESUMEN

Escherichia coli O157:H7 is a globally important foodborne pathogen with implications for food safety. Antibiotic treatment for O157 may potentially contribute to the exacerbation of hemolytic uremic syndrome, and the increasing prevalence of antibiotic-resistant strains necessitates the development of new treatment strategies. In this study, the bactericidal effects and resistance development of antibiotic and bacteriophage monotherapy were compared with those of combination therapy against O157. Experiments involving continuous exposure of O157 to phages and antibiotics, along with genetic deletion studies, revealed that the deletion of glpT and uhpT significantly increased resistance to fosfomycin. Furthermore, we found that OmpC functions as a receptor for the PP01 phage, which infects O157, and FhuA functions as a receptor for the newly isolated SP15 phage, targeting O157. In the glpT and uhpT deletion mutants, additional deletion in ompC, the receptor for the PP01 phage, increased resistance to fosfomycin. These findings suggest that specific phages may contribute to antibiotic resistance by selecting the emergence of gene mutations responsible for both phage and antibiotic resistance. While combination therapy with phages and antibiotics holds promise for the treatment of bacterial infections, careful consideration of phage selection is necessary.IMPORTANCEThe combination treatment of fosfomycin and bacteriophages against Escherichia coli O157 demonstrated superior bactericidal efficacy compared to monotherapy, effectively suppressing the emergence of resistance. However, mutations selected by phage PP01 led to enhanced resistance not only to the phage but also to fosfomycin. These findings underscore the importance of exercising caution in selecting phages for combination therapy, as resistance selected by specific phages may increase the risk of developing antibiotic resistance.


Asunto(s)
Antibacterianos , Infecciones por Escherichia coli , Escherichia coli O157 , Fosfomicina , Antibacterianos/farmacología , Escherichia coli O157/virología , Escherichia coli O157/efectos de los fármacos , Escherichia coli O157/genética , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/tratamiento farmacológico , Humanos , Fosfomicina/farmacología , Farmacorresistencia Bacteriana , Bacteriófagos/genética , Bacteriófagos/fisiología , Bacteriófagos/efectos de los fármacos , Terapia de Fagos/métodos , Colifagos/genética , Colifagos/efectos de los fármacos , Colifagos/fisiología , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo
10.
Lung ; 202(3): 223-232, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38772946

RESUMEN

We are entering the post-antibiotic era. Antimicrobial resistance (AMR) is a critical problem in chronic lung infections resulting in progressive respiratory failure and increased mortality. In the absence of emerging novel antibiotics to counter AMR infections, bacteriophages (phages), viruses that infect bacteria, have become a promising option for chronic respiratory infections. However, while personalized phage therapy is associated with improved outcomes in individual cases, clinical trials demonstrating treatment efficacy are lacking, limiting the therapeutic potential of this approach for respiratory infections. In this review, we address the current state of phage therapy for managing chronic respiratory diseases. We then discuss how phage therapy may address major microbiologic obstacles which hinder disease resolution of chronic lung infections with current antibiotic-based treatment practices. Finally, we highlight the challenges that must be addressed for successful phage therapy clinical trials. Through this discussion, we hope to expand on the potential of phages as an adjuvant therapy in chronic lung infections, as well as the microbiologic challenges that need to be addressed for phage therapy to expand beyond personalized salvage therapy.


Asunto(s)
Terapia de Fagos , Infecciones del Sistema Respiratorio , Humanos , Terapia de Fagos/métodos , Infecciones del Sistema Respiratorio/terapia , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/virología , Bacteriófagos , Enfermedad Crónica , Antibacterianos/uso terapéutico
11.
Virology ; 595: 110098, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38705084

RESUMEN

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.


Asunto(s)
Infecciones por Acinetobacter , Acinetobacter baumannii , Bacteriófagos , Farmacorresistencia Bacteriana Múltiple , Terapia de Fagos , Acinetobacter baumannii/virología , Acinetobacter baumannii/efectos de los fármacos , Animales , Infecciones por Acinetobacter/microbiología , Infecciones por Acinetobacter/terapia , Ratones , Bacteriófagos/genética , Bacteriófagos/fisiología , Terapia de Fagos/métodos , Genoma Viral , Biopelículas/efectos de los fármacos , Biopelículas/crecimiento & desarrollo , Humanos , Femenino , Aguas del Alcantarillado/virología
12.
Medicina (Kaunas) ; 60(5)2024 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-38792972

RESUMEN

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.


Asunto(s)
Terapia de Fagos , Infecciones Relacionadas con Prótesis , Humanos , Infecciones Relacionadas con Prótesis/terapia , Terapia de Fagos/métodos , Resultado del Tratamiento
13.
Urologiia ; (1): 114-118, 2024 Mar.
Artículo en Ruso | MEDLINE | ID: mdl-38655779

RESUMEN

Clinical cases of three patients with neurogenic lower urinary tract dysfunction, complicated by chronic urinary tract infection are presented in the article. All patients underwent clean intermittent catheterization and, in order to prevent symptomatic lower urinary tract infections, received bacteriophage therapy with a clinically proven positive effect. During 3 months follow-up, there were no episodes of urinary tract infection. A change in the concentration of uropathogens and restoration of sensitivity to a number of antimicrobial drugs were observed. Although phage therapy in urology requires further clinical research, it provides an additional strategy to treat urinary tract infections considering an increase in antibiotic resistance.


Asunto(s)
Terapia de Fagos , Infecciones Urinarias , Humanos , Infecciones Urinarias/terapia , Infecciones Urinarias/etiología , Masculino , Terapia de Fagos/métodos , Persona de Mediana Edad , Femenino , Bacteriófagos , Vejiga Urinaria Neurogénica/terapia , Adulto , Anciano , Recurrencia
14.
Antimicrob Agents Chemother ; 68(5): e0143923, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38591854

RESUMEN

Phage therapy has (re)emerged as a serious possibility for combating multidrug-resistant bacterial infections, including those caused by vancomycin-resistant Enterococcus faecium strains. These opportunistic pathogens belong to a specific clonal complex 17, against which relatively few phages have been screened. We isolated a collection of 21 virulent phages growing on these vancomycin-resistant isolates. Each of these phages harbored a typical narrow plaquing host range, lysing at most 5 strains and covering together 10 strains of our panel of 14 clinical isolates. To enlarge the host spectrum of our phages, the Appelmans protocol was used. We mixed four out of our most complementary phages in a cocktail that we iteratively grew on eight naive strains from our panel, of which six were initially refractory to at least three of the combined phages. Fifteen successive passages permitted to significantly improve the lytic activity of the cocktail, from which phages with extended host ranges within the E. faecium species could be isolated. A single evolved phage able to kill up to 10 of the 14 initial E. faecium strains was obtained, and it barely infected nearby species. All evolved phages had acquired point mutations or a recombination event in the tail fiber genetic region, suggesting these genes might have driven phage evolution by contributing to their extended host spectra.


Asunto(s)
Bacteriófagos , Enterococcus faecium , Especificidad del Huésped , Enterococos Resistentes a la Vancomicina , Enterococcus faecium/efectos de los fármacos , Bacteriófagos/genética , Enterococos Resistentes a la Vancomicina/efectos de los fármacos , Terapia de Fagos/métodos , Infecciones por Bacterias Grampositivas/microbiología , Resistencia a la Vancomicina , Vancomicina/farmacología , Humanos , Antibacterianos/farmacología
15.
Sci Rep ; 14(1): 9399, 2024 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-38658654

RESUMEN

Edwardsiella piscicida causes significant economic losses to the aquaculture industry worldwide. Phage-based biocontrol methods are experiencing a renaissance because of the spread of drug-resistant genes and bacteria resulting from the heavy use of antibiotics. Here, we showed that the novel Edwardsiella phage EPP-1 could achieve comparable efficacy to florfenicol using a zebrafish model of Edwardsiella piscicida infection and could reduce the content of the floR resistance gene in zebrafish excreta. Specifically, phage EPP-1 inhibited bacterial growth in vitro and significantly improved the zebrafish survival rate in vivo (P = 0.0035), achieving an efficacy comparable to that of florfenicol (P = 0.2304). Notably, integrating the results of 16S rRNA sequencing, metagenomic sequencing, and qPCR, although the effects of phage EPP-1 converged with those of florfenicol in terms of the community composition and potential function of the zebrafish gut microbiota, it reduced the floR gene content in zebrafish excreta and aquaculture water. Overall, our study highlights the feasibility and safety of phage therapy for edwardsiellosis control, which has profound implications for the development of antibiotic alternatives to address the antibiotic crisis.


Asunto(s)
Antibacterianos , Bacteriófagos , Edwardsiella , Infecciones por Enterobacteriaceae , Tianfenicol/análogos & derivados , Pez Cebra , Animales , Pez Cebra/microbiología , Edwardsiella/genética , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/veterinaria , Infecciones por Enterobacteriaceae/terapia , Bacteriófagos/genética , Bacteriófagos/fisiología , Antibacterianos/farmacología , Farmacorresistencia Bacteriana/genética , Microbioma Gastrointestinal , Terapia de Fagos/métodos , ARN Ribosómico 16S/genética , Enfermedades de los Peces/microbiología , Enfermedades de los Peces/terapia , Enfermedades de los Peces/prevención & control , Tianfenicol/farmacología , Acuicultura/métodos
16.
Sci Rep ; 14(1): 6026, 2024 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-38472239

RESUMEN

The continuing and rapid emergence of antibiotic resistance (AMR) calls for innovations in antimicrobial therapies. A promising, 're-emerging' approach is the application of bacteriophage viruses to selectively infect and kill pathogenic bacteria, referred to as phage therapy. In practice, phage therapy is personalized and requires companion diagnostics to identify efficacious phages, which are then formulated into a therapeutic cocktail. The predominant means for phage screening involves optical-based assays, but these methods cannot be carried out in complex media, such as colored solutions, inhomogeneous mixtures, or high-viscosity samples, which are often conditions encountered in vivo. Moreover, these assays cannot distinguish phage binding and lysis parameters, which are important for standardizing phage cocktail formulation. To address these challenges, we developed Phage-layer Interferometry (PLI) as a companion diagnostic. Herein, PLI is assessed as a quantitative phage screening method and prototyped as a bacterial detection platform. Importantly, PLI is amenable to automation and is functional in complex, opaque media, such as baby formula. Due to these newfound capabilities, we foresee immediate and broad impact of PLI for combating AMR and protecting against foodborne illnesses.


Asunto(s)
Bacteriófagos , Enfermedades Transmitidas por los Alimentos , Terapia de Fagos , Humanos , Terapia de Fagos/métodos , Bacterias , Antibacterianos
17.
Lancet Microbe ; 5(5): e489-e499, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38452780

RESUMEN

Colonisation by bacterial pathogens typically precedes invasive infection and seeds transmission. Thus, effective decolonisation strategies are urgently needed. The literature reports attempts to use phages for decolonisation. To assess the in-vivo efficacy and safety of phages for bacterial decolonisation, we performed a systematic review by identifying relevant studies to assess the in-vivo efficacy and safety of phages for bacterial decolonisation. We searched PubMed, Embase (Ovid), MEDLINE (Ovid), Web of Science, and the Cochrane Library to identify relevant articles published between Jan 1, 1990, and May 12, 2023, without language restrictions. We included studies that assessed the efficacy of phage for bacterial decolonisation in humans or vertebrate animal models. This systematic review is registered with PROSPERO, CRD42023457637. We identified 6694 articles, of which 56 (51 animal studies and five clinical reports) met the predetermined selection criteria and were included in the final analysis. The gastrointestinal tract (n=49, 88%) was the most studied bacterial colonisation site, and other sites were central venous catheters, lung, nose, skin, and urinary tract. Of the 56 included studies, the bacterial load at the colonisation site was reported to decrease significantly in 45 (80%) studies, but only five described eradication of the target bacteria. 15 studies reported the safety of phages for decolonisation. No obvious adverse events were reported in both the short-term and long-term observation period. Given the increasing life-threatening risks posed by bacteria that are difficult to treat, phages could be an alternative option for bacterial decolonisation, although further optimisation is required before their application to meet clinical needs.


Asunto(s)
Infecciones Bacterianas , Bacteriófagos , Humanos , Infecciones Bacterianas/terapia , Animales , Bacterias/virología , Terapia de Fagos/métodos
18.
Microbiol Spectr ; 12(5): e0322123, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38526142

RESUMEN

The emergence of antibiotic-resistant bacteria (ARB) has necessitated the development of alternative therapies to deal with this global threat. Bacteriophages (viruses that target bacteria) that kill ARB are one such alternative. Although phages have been used clinically for decades with inconsistent results, a number of recent advances in phage selection, propagation, and purification have enabled a reevaluation of their utility in contemporary clinical medicine. In most phage therapy cases, phages are administered in combination with antibiotics to ensure that patients receive the standard-of-care treatment. Some phages may work cooperatively with antibiotics to eradicate ARB, as often determined using non-standardized broth assays. We sought to develop a solid media-based assay to assess cooperativity between antibiotics and phages to offer a standardized platform for such testing. We modeled the interactions that occur between antibiotics and phages on solid medium to measure additive, antagonistic, and synergistic interactions. We then tested the method using different bacterial isolates and identified a number of isolates where synergistic interactions were identified. These interactions were not dependent on the specific organism, phage family, or antibiotic used. A priori susceptibility to the antibiotic or the specific phage were not requirements to observe synergistic interactions. Our data also confirm the potential for the restoration of vancomycin to treat vancomycin-resistant Enterococcus (VRE) when used in combination with phages. Solid media assays for the detection of cooperative interactions between antibiotics and phages can be an accessible technique adopted by clinical laboratories to evaluate antibiotic and phage choices in phage therapy.IMPORTANCEBacteriophages have become an important alternative treatment for individuals with life-threatening antibiotic-resistant bacteria (ARB) infections. Because antibiotics represent the standard-of-care for treatment of ARB, antibiotics and phages often are delivered together without evidence that they work cooperatively. Testing for cooperativity can be difficult due to the equipment necessary and a lack of standardized means for performing the testing in liquid medium. We developed an assay using solid medium to identify interactions between antibiotics and phages for gram-positive and gram-negative bacteria. We modeled the interactions between antibiotics and phages on solid medium, and then tested multiple replicates of vancomycin-resistant Enterococcus (VRE) and Stenotrophomonas in the assay. For each organism, we identified synergy between different phage and antibiotic combinations. The development of this solid media assay for assessing synergy between phages and antibiotics will better inform the use of these combinations in the treatment of ARB infections.


Asunto(s)
Antibacterianos , Bacteriófagos , Terapia de Fagos , Bacteriófagos/fisiología , Bacteriófagos/aislamiento & purificación , Antibacterianos/farmacología , Terapia de Fagos/métodos , Humanos , Medios de Cultivo/química , Pruebas de Sensibilidad Microbiana/métodos , Bacterias/virología , Bacterias/efectos de los fármacos , Farmacorresistencia Bacteriana
19.
Microbiol Spectr ; 12(5): e0378423, 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38511948

RESUMEN

Clostridium perfringens is a prevalent bacterial pathogen in poultry, and due to the spread of antimicrobial resistance, alternative treatments are needed to prevent and treat infection. Bacteriophages (phages), viruses that kill bacteria, offer a viable option and can be used therapeutically to treat C. perfringens infections. The aim of this study was to isolate phages against C. perfringens strains currently circulating on farms across the world and establish their virulence and development potential using host range screening, virulence assays, and larva infection studies. We isolated 32 phages of which 19 lysed 80%-92% of our global C. perfringens poultry strain collection (n = 97). The virulence of these individual phages and 32 different phage combinations was quantified in liquid culture at multiple doses. We then developed a multi-strain C. perfringens larva infection model, to mimic an effective poultry model used by the industry. We tested the efficacy of 16/32 phage cocktails in the larva model. From this, we identified that our phage cocktail consisting of phages CPLM2, CPLM15, and CPLS41 was the most effective at reducing C. perfringens colonization in infected larvae when administered before bacterial challenge. These data suggest that phages do have significant potential to prevent and treat C. perfringens infection in poultry. IMPORTANCE: Clostridium perfringens causes foodborne illness worldwide, and 95% of human infections are linked to the consumption of contaminated meat, including chicken products. In poultry, C. perfringens infection causes necrotic enteritis, and associated mortality rates can be up to 50%. However, treating infections is difficult as the bacterium is becoming antibiotic-resistant. Furthermore, the poultry industry is striving toward reduced antibiotic usage. Bacteriophages (phages) offer a promising alternative, and to progress this approach, robust suitable phages and laboratory models that mimic C. perfringens infections in poultry are required. In our study, we isolated phages targeting C. perfringens and found that many lyse C. perfringens strains isolated from chickens worldwide. Consistent with other published studies, in the model systems we assayed here, when some phages were combined as cocktails, the infection was cleared most effectively compared to individual phage use.


Asunto(s)
Bacteriófagos , Infecciones por Clostridium , Clostridium perfringens , Especificidad del Huésped , Enfermedades de las Aves de Corral , Clostridium perfringens/virología , Animales , Bacteriófagos/fisiología , Infecciones por Clostridium/microbiología , Infecciones por Clostridium/terapia , Infecciones por Clostridium/veterinaria , Enfermedades de las Aves de Corral/microbiología , Enfermedades de las Aves de Corral/virología , Virulencia , Pollos , Aves de Corral/microbiología , Terapia de Fagos/métodos , Larva/microbiología , Larva/virología , Modelos Animales de Enfermedad
20.
Burns ; 50(5): 1045-1052, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38472000

RESUMEN

Infections are a major cause of morbidity and mortality in burn patients, and the rise of multidrug-resistant organisms (MDROs) has made it more challenging to manage and prevent infections. This review examines the available treatment options for MDROs in burn patients and anticipates the future challenges posed by their increasing prevalence. The review covers new antibiotics, such as Eravacycline and Plazomicin, as well as non-antibiotic therapies, such as bacteriophages and nanoparticles. Future research should focus on examining the long-term efficacy, cost-effectiveness, and in vivo efficacy of different treatment modalities. The potential of alternative therapies, such as probiotics and low-frequency magnetic fields, should also be explored. Accurate and rapid diagnostic and monitoring tools for detecting MDROs in burn patients should be developed. The emergence of MDROs in burn care is a challenge and a new beginning in infection innovation and novel treatments.


Asunto(s)
Antibacterianos , Quemaduras , Farmacorresistencia Bacteriana Múltiple , Humanos , Quemaduras/complicaciones , Quemaduras/terapia , Quemaduras/microbiología , Antibacterianos/uso terapéutico , Infección de Heridas/microbiología , Infección de Heridas/tratamiento farmacológico , Probióticos/uso terapéutico , Bacteriófagos , Infecciones Bacterianas/tratamiento farmacológico , Terapia de Fagos/métodos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA