bims-fagtap Biomed News
on Phage therapies and applications
Issue of 2025–08–31
25 papers selected by
Luca Bolliger, lxBio



  1. Viruses. 2025 Aug 08. pii: 1094. [Epub ahead of print]17(8):
      Treatments for bacterial infections can be less effective due to toxicities, bacterial tolerance, or genetic resistance to antibacterial agents. The emphasis here is on combating genetic bacterial resistance to bacteriophages. Commonly described simply as phages, bacteriophages are the viruses of bacteria. As phage therapies, they are one of the oldest clinical treatments for bacterial infections. Thwarting bacterial evolution of resistance to phages, particularly during phage treatments, typically involves targeting more than one bacterial characteristic. This can be achieved serially, involving phage substitution after bacterial resistance has become problematic, something that is used especially during more personalized therapies. Substitution phages can be sourced in various ways. This includes as autophages, from phage banks, or via phage training-all as considered here-as well as through phage engineering. An alternative approach is preventing bacterial mutations from occurring at all. In addition, there is simultaneous targeting of multiple bacterial characteristics. These latter strategies include all of the following: using phages that target bacterial fitness or virulence determinants, employing individual phages that recognize multiple receptors, using phage cocktails, or applying phages in combination with antibiotics. This review discusses these different approaches for combating treatment resistance, highlighting various pros and cons.
    Keywords:  antibiotics; bacteriophage therapy; community resistance; phage breeding; phage library; phage resistance; phage steering; polyphage; treatment resistance
    DOI:  https://doi.org/10.3390/v17081094
  2. Farm Hosp. 2025 Aug 23. pii: S1130-6343(25)00112-6. [Epub ahead of print]
      The need for new antimicrobial treatments that work alternatively or synergistically with antibiotics to address the problem of the emergence and transmission of antimicrobial resistance has increased interest in the use of minority therapies such as phage therapy. For safe and widespread application of this therapy, it is necessary to establish the pharmacokinetic and pharmacodynamic parameters for its use in humans. This systematic review analyzes the criteria necessary to establish the PK/PD of this therapy, as well as its current application, based on a review of 66 clinical cases that catch diverse infections and phage administration routes.
    Keywords:  Efficacy; Eficacia; Fagos; Fagoterapia; Farmacocinéticos y farmacodinámicos; Phage therapy; Phages; Pharmacokinetics and pharmacodynamics; Sinergias; Synergies
    DOI:  https://doi.org/10.1016/j.farma.2025.07.007
  3. Front Bioeng Biotechnol. 2025 ;13 1605263
      Salmonella spp. is the most common pathogen transmitted to humans through contaminated water and food. Due to its ability to infect both animals and humans, as well as the spread of antibiotic-resistant strains, this pathogen has become a priority for food and pharmaceutical industries. Consequently, research and development of treatments to combat infections caused by Salmonella spp. are ongoing. One of the most promising strategies is the phage therapy (PT) which is based on the use of very specific viruses that infect this pathogenic bacterium without any action over the host and which use has shown effectiveness. Now a days, at least 41 companies worldwide market phage therapy products mainly for use in the food sector to reduce the transmission chain of Salmonella spp. to humans. However, the complex production processes required to ensure product quality, stability, safety, and efficacy, as well as the need for regulatory frameworks for phage therapy, present limitations to the global application of this strategy seems to be a limitation to promote its use all over the world as a pharmaceutical product. Thus, this work presents a literature review on state-of-the-art of PT, analysing the opportunities and challenges that are present to consider such a therapy as an emerging treatment for antibiotic resistance of Salmonella enterica.
    Keywords:  Salmonella enterica; bacteriophages; bacteriophages products; phage therapy; regulation of bacteriophages
    DOI:  https://doi.org/10.3389/fbioe.2025.1605263
  4. Viruses. 2025 Aug 04. pii: 1080. [Epub ahead of print]17(8):
      Phage therapy, long overshadowed by antibiotics in Western medicine, has a well-established history in some Eastern European countries and is now being revitalized as a promising strategy against antimicrobial resistance (AMR). This resurgence of phage therapy is driven by the urgent need for innovative countermeasures to AMR, which will cause an estimated 10 million deaths annually by 2050. However, the emergence of phage-resistant variants presents challenges similar to AMR, thus necessitating a deeper understanding of phage resistance mechanisms and control strategies. The highest priority must be to prevent the emergence of phage resistance. Although phage cocktails targeting multiple receptors have demonstrated a certain level of phage resistance suppression, they cannot completely suppress resistance in clinical settings. This highlights the need for strategies beyond simple resistance suppression. Notably, recent studies examining fitness trade-offs associated with phage resistance have opened new avenues in phage therapy that offer the potential of restoring antibiotic susceptibility and attenuating pathogen virulence despite phage resistance. Thus, controlling phage resistance may rely on both its suppression and strategic redirection. This review summarizes key concepts in the control of phage resistance and explores evolutionary engineering as a means of optimizing phage therapy, with a particular focus on Pseudomonas infections. Harnessing evolutionary dynamics by intentionally breaking the spontaneous evolutionary trajectories of target bacterial pathogens could potentially reshape bacterial adaptation by acquisition of phage resistance, unlocking potential in the application of phage therapy.
    Keywords:  antimicrobial resistance; bacteriophage; evolved phages; fitness cost; infection control; trade-offs
    DOI:  https://doi.org/10.3390/v17081080
  5. Microb Pathog. 2025 Aug 21. pii: S0882-4010(25)00728-4. [Epub ahead of print]208 108003
      The escalating crisis of antimicrobial resistance (AMR) has underscored the urgent need for alternative therapeutic strategies. Among these, bacteriophage (phage) therapy has emerged as a promising candidate due to its high specificity, favorable safety profile, and self-replicating nature. Recent studies have unveiled the dual regulatory role of bacterial extracellular vesicles (BEVs) in the context of phage infection. BEVs can inhibit phage activity by acting as decoy particles that adsorb virions, while also facilitating phage propagation through mechanisms such as receptor transfer and protective delivery. This review systematically integrates current findings on the mechanisms by which phages induce BEV production, the bidirectional effects of BEVs on phage-mediated bacterial killing, and the potential of BEVs as bioengineered nanoplatforms to enhance phage delivery. Based on recent advances, we propose that engineering BEVs may overcome key clinical limitations of phage therapy, such as narrow host range and poor in vivo bioavailability. We propose that engineered BEVs represent a promising next-generation delivery platform that could significantly enhance the precision and efficacy of phage therapy against multidrug-resistant bacterial infections.
    Keywords:  Bacterial extracellular vesicles; Dual impact; Phage therapy; Targeted delivery
    DOI:  https://doi.org/10.1016/j.micpath.2025.108003
  6. BMC Microbiol. 2025 Aug 22. 25(1): 528
      Phage therapy has been explored and used compassionately in the post-antibiotic era, though phage resistance might pose a serious challenge. The advent of hypervirulent and hypermucoviscous traits in Klebsiella pneumoniae limits therapeutic choices. This study investigated the phage resistance in hypermucoviscous hypervirulent Klebsiella pneumoniae clinical strain Kleb_53. A Klebsiella phage Disc against the Kleb_53 strain was isolated from sewage. The phage exhibited stability between - 20 °C and 60 °C and within the pH range of 3 to 11. The phage adsorption time was 15 min, with a latent period of 30 min and a burst size of 354 virions. The phage-resistant Kleb_53 variants were screened and examined for their phenotypic variations, antibiotic susceptibility, and biofilm formation. Colony morphotype variants were observed, including smooth, rough, and small colony variants. String, aggregation, and wetness tests confirmed reduced mucoviscosity. The plaque morphology differed between the wild and variants. Additionally, resistance to meropenem and third-generation cephalosporins was reversed, whereas the biofilm-forming ability varied among the recovered variants. This study demonstrates that ongoing phage-host interactions drive phenotypic changes and the emergence of phage-resistant variants with altered antibiotic susceptibility and biofilm-forming capacity. It also underscores the need for further research on phage resistance and strategies to overcome it for the effective application of phage therapy.
    Keywords:   Klebsiella pneumoniae ; Antibiotic resistance; Biofilms; Hypermucoviscous; Phage Therapy; Phage resistance
    DOI:  https://doi.org/10.1186/s12866-025-04268-x
  7. Biotechnol Adv. 2025 Aug 19. pii: S0734-9750(25)00175-2. [Epub ahead of print]84 108689
      Klebsiella pneumoniae represents one of the most concerning ESKAPE pathogens, with multidrug-resistant strains driving urgent clinical interest in phage therapy as a viable alternative to antibiotics. However, the evolutionary arms race between phages and bacteria has equipped K. pneumoniae with sophisticated anti-phage immune defenses, posing a substantial barrier to durable therapeutic success. Through systematic analysis of K. pneumoniae-phage co-evolutionary dynamics, we identify predominant resistance mechanisms and discuss why these mechanisms primarily concentrate on adsorption blocking pathways. We then integrate clinical case studies with preclinical research to evaluate combination strategies against phage resistance, particularly highlighting synergistic approaches using antibiotics-phage or phage cocktails/phage serial therapy that increase selective pressure while reducing bacterial host adaptability and pathogenicity. Finally, we propose a computational roadmap leveraging machine learning for phage characterization, host-interaction prediction and de novo genome engineering, with particular emphasis on minimizing resistance emergence. This interdisciplinary review provides both immediate clinical guidance and a forward-looking vision for rational phage design, applicable beyond not only to K. pneumoniae but also to other high-priority pathogens. We also highlight that integrations of synthetic biology, computational science, and microbiology will be essential for transitioning phage therapy from experimental treatments to standardized interventions addressing antimicrobial resistance.
    Keywords:  Artificial intelligence; Biological foundation model; Klebsiella pneumoniae; Phage engineering; Phage resistance; Phage therapy
    DOI:  https://doi.org/10.1016/j.biotechadv.2025.108689
  8. Microorganisms. 2025 Aug 11. pii: 1873. [Epub ahead of print]13(8):
      Gram-negative Burkholderia bacteria are known for causing diseases in humans, animals, and plants, and high intrinsic resistance to antibiotics. Phage therapy is a promising alternative to control multidrug-resistant bacterial pathogens. Here, we present an overview of Burkholderia phage characteristics, host specificity, genomic classification, and therapeutic potentials across medical, veterinary, and agricultural systems. We evaluate the efficacy and limitations of current phage candidates, the biological and environmental barriers of phage applications, and the phage cocktail strategy. We highlight the innovations on the development of targeted phage delivery systems and the transition from the exploration of clinical phage therapy to plant disease management, advocating integrated disease control strategies.
    Keywords:  Burkholderia; bacterial panicle blight; bacteriophage; cystic fibrosis; nanocarrier; sustainable disease management
    DOI:  https://doi.org/10.3390/microorganisms13081873
  9. Pharmaceutics. 2025 Aug 20. pii: 1077. [Epub ahead of print]17(8):
      The concerning increase in respiratory infections that are resistant to multiple drugs has led to a growing interest in bacteriophage therapy as a potential alternative to conventional antibiotics. Effective phage delivery to the lungs, however, presents several formulation and stability issues, particularly for inhalation-based methods. This review highlights current developments in the creation of dry powder formulations that can be inhaled for pulmonary phage therapy, with a focus on encapsulation methods based on nanoparticles, such as solid lipid nanoparticles (SLNs) and polymer-based nanoparticles. These carriers enhance the aerodynamic characteristics of phages, making them suitable for deep lung deposition, while also protecting them during processing and storage. Several drying methods have been investigated to create powders with optimal morphologies, porosity, and dispersibility, including spray drying and spray freeze drying. The review also emphasizes how the phage morphotype affects stability, especially when nebulization stress is present. Furthermore, the advantages of nanoparticle matrices are confirmed by the reduced viability loss (usually< 0.5 log PFU) of encapsulated phages. Standardizing production processes, scaling up, and ensuring regulatory compliance remain challenging despite encouraging preclinical results. The combination of phage therapy with nanotechnology creates new avenues for the utilization of inhalable delivery methods to treat multidrug-resistant pulmonary infections. To translate these novel formulations from preclinical development to clinical application, sustained multidisciplinary collaboration across pharmaceutical sciences, microbiology, and clinical pharmacology is essential.
    Keywords:  bacteriophage therapy; inhalable powders; liposomes; multidrug-resistant pathogens; nanoparticles
    DOI:  https://doi.org/10.3390/pharmaceutics17081077
  10. Viruses. 2025 Aug 14. pii: 1118. [Epub ahead of print]17(8):
      We describe the first use of phage therapy in Canada for the treatment of a life-threatening periprosthetic joint infection (PJI), with successful outcome. PJI is a devastating complication of joint replacement surgery, with high morbidity and mortality. Our patient presented with early sepsis from a chronic recalcitrant multidrug-resistant (MDR) Staphylococcus epidermidis hip PJI which had repeatedly failed standard therapy. She had previously undergone 10 operations of the right hip, and only three weeks after completing a prolonged course of daptomycin following her most recent hip revision, she developed a draining sinus tract. Given the high burden of disease, inability to achieve surgical source control, and lack of antibiotic treatment options for long-term suppressive therapy, bacteriophage (phage) therapy was pursued. The patient underwent irrigation and debridement with complex flap reconstruction: intraoperative tissue cultures again yielded MDR S. epidermidis. We developed a novel phage therapy protocol for this patient, with twice daily, intra-articular and intravenous (7 × 109 PFU/dose) phage delivery over a planned 14-day course. Complete healing of the wound with cessation of drainage occurred within one month after treatment. A marked improvement in right hip pain and mobility occurred within three months after treatment. Twelve months following phage treatment, there is normalization of serum inflammatory markers with diminished pain, increased mobility, and no recurrent surgery. Our patient continues to improve and is currently living independently at home, with sustained clinical control of infection.
    Keywords:  antibiotic resistance; periprosthetic joint infection; phage therapy
    DOI:  https://doi.org/10.3390/v17081118
  11. Pharmaceuticals (Basel). 2025 Jul 26. pii: 1115. [Epub ahead of print]18(8):
      The escalating global crisis of antimicrobial resistance, responsible for approximately 1.27 million deaths in 2019, has catalyzed renewed interest in bacteriophage therapy as a viable therapeutic alternative. With projections indicating that drug-resistant bacteria could cause over 39 million deaths worldwide by 2050, developing alternative antimicrobial strategies has become critically urgent. This comprehensive review examines the scientific foundation of bacteriophage therapy, traces its historical development from early Soviet applications through contemporary regulatory frameworks, and provides strategic guidance for developers seeking FDA approval for bacteriophage-based therapeutics. We analyze the current regulatory landscape across major jurisdictions, including manufacturing requirements and clinical development pathways essential for successful market authorization. Approximately 90 clinical trials involving bacteriophages are ongoing worldwide, with 41 studies in the United States demonstrating significant momentum in this field.
    Keywords:  FDA approval; GMP manufacturing; Soviet phage therapy; antimicrobial; antimicrobial resistance; bacteriophage therapy; clinical trials; international regulation; regulatory pathway
    DOI:  https://doi.org/10.3390/ph18081115
  12. Viruses. 2025 Aug 06. pii: 1088. [Epub ahead of print]17(8):
      Antibiotic-resistant Pseudomonas aeruginosa presents a critical global health challenge, particularly in hospital-acquired infections. Bacteriophages offer a promising therapeutic avenue due to their ability to target and lyse resistant strains. This study characterizes Pseudomonas phage Banzai, a newly isolated Pbunavirus (family Lindbergviridae) with lytic activity against multiple P. aeruginosa isolates, including multidrug-resistant strains. Genomic analysis revealed a 66,189 bp genome, lacking antibiotic resistance or virulence factors, and suggested a headful packaging mechanism and the presence of a bidirectional component in the replication. In vivo experiments using Galleria mellonella showed therapeutic potential, significantly improving larval survival (87% at 24 h). Host range analysis revealed activity against 13 of 30 P. aeruginosa isolates, including members of O1, O3, O5 and O6 in silico predicted serogroups. Phylogenomic analyses place phage Banzai within the genus Pbunavirus, sharing 94.8% intergenomic similarity with its closest relatives, supporting its classification as a novel species. These findings highlight phage Banzai as a potential candidate for phage therapy, demonstrating genomic stability, a strictly lytic lifestyle, and in vivo efficacy.
    Keywords:  Lindbergviridae; Pbunavirus; Pseudomonas aeruginosa; antimicrobial resistance; phage; phage evolution; phage genomics; phage therapy
    DOI:  https://doi.org/10.3390/v17081088
  13. Expert Opin Investig Drugs. 2025 Aug 26. 1-12
       INTRODUCTION: Carbapenem-resistant Acinetobacter baumannii (CRAB) infections have become common in healthcare settings worldwide, yet current therapeutic options are limited. A pipeline of new antibiotics and non-traditional antimicrobial agents is being developed to address the urgent need for efficacious therapeutic options for patients with CRAB infections.
    AREAS COVERED: At the time of this writing, 13 traditional antibiotics are in clinical development for CRAB infections, some with a novel mechanism of action. Specifically, 9 antibiotics are in Phase 1 (R-327, xeruborbactam/QPX-7728, upleganan/SPR-206, MRX-8, QPX-9003, zifanocycline/KBP-7072, apramycin/EBL-1003, zosurabalpin/RG-6006, and ANT-3310), two in Phase 2 (BV-100, OMN-6), and two in Phase 3 (zidebactam/WCK-5222, funobactam/XNW-4107) clinical trials. Additionally, there are six non-traditional antimicrobial agents in Phase 1 or 2 clinical trials for treating CRAB infections. In particular, two monoclonal antibodies (TRL-1068, CMTX-101), a phage therapy (Phagebank), an immune-modulating agent (recombinant human plasma gelsolin/Rhu-pGSN), a microbiome-modulating agent (SER-155), and an engineered cationic antibiotic peptide (PLG-0206).
    EXPERT OPINION: Several agents with promising characteristics against CRAB infections are in clinical development (Phases 1, 2, and 3). The urgent need for therapeutic options against CRAB infections necessitates optimizing efforts and time for introducing successfully studied agents into clinical practice.
    Keywords:  Acinetobacter baumannii; Carbapenem-resistant Acinetobacter baumannii; clinical trial; non-traditional antibiotic; pipeline; traditional antibiotic; under development
    DOI:  https://doi.org/10.1080/13543784.2025.2552846
  14. Ann Clin Microbiol Antimicrob. 2025 Aug 20. 24(1): 46
       BACKGROUND: The emergence of MDR K. pneumoniae poses a critical challenge in treating respiratory-associated pneumonia. Bacteriophages are promising antibiotic alternatives with unique features. This study aimed to isolate new bacteriophages from the hospital environment and investigate their therapeutic potential and mechanisms.
    METHODS: We employed plaque assays, transmission electron microscopy, and whole-genome sequencing to systematically characterize the biological properties, morphology, and genomic profiles of the phages in parallel. The bacteriostatic curve, biofilm staining quantification, and biofilm inhibition rate assay were employed to evaluate the in vitro lytic efficacy of the phage. More importantly, we established the murine pneumonia infection models through nasal instillation, assessed the therapeutic potential of the phage in vivo by observing pathological morphology via HE staining, detecting pro-inflammatory cytokine levels via qPCR and ELISA, and monitoring bacterial load changes in lung tissue through PCR analysis.
    RESULTS: Phages vB_KpnP_XY3 and vB_KpnP_XY4, taxonomically classified as Siphoviridae, demonstrated broad temperature (4-60 °C), pH (4-11) tolerance, chloroform resistance, latent periods of 40/35 min, and burst sizes of 340/126 PFU/cell. Both genomes contained circular dsDNA genomes (47,466 bp/50,036 bp) without virulence or antibiotic resistance genes. The bacterial concentration markedly decreased at 2 h post-treatment, reaching its biological nadir by 6 h. Concurrent biofilm assays demonstrated 80% biofilm inhibition and rapid bacterial clearance. In murine pneumonia models, both phage monotherapy and phage-antibiotic combinations significantly reduced bacterial loads compared with antibiotics alone (P < 0.05), concurrently attenuating inflammation (IL-1β/IL-6/TNF-a. P < 0.0001) and restoring alveolar architecture with reduced necrosis.
    CONCLUSION: The phages vB_KpnP_XY3 and vB_KpnP_XY4 demonstrated robust environmental adaptability. Its antibacterial effect is related to its specific biofilm dissolution performance in vivo and in vitro. These findings provide strong evidence for the precise phage treatment of MDR K. pneumoniae infections.
    Keywords:  Bacteriophage therapy; Biofilm degradation; Histopathology; Lytic bacteriophage; Multidrug-resistant Klebsiella pneumoniae
    DOI:  https://doi.org/10.1186/s12941-025-00812-9
  15. Microb Pathog. 2025 Aug 25. pii: S0882-4010(25)00730-2. [Epub ahead of print] 108005
      The therapeutic challenges caused by multidrug-resistant (MDR) and extensively drug-resistant (XDR) bacteria have necessitated the development of alternative treatment strategies. Phage-antibiotic synergy (PAS) has recently emerged to replace or possibly supplement antibiotics. We isolated the lytic phage NTNC80A from hospital sewage in Chandigarh, India, and it belongs to the class Caudoviricetes of viruses. We tested multiple strategies to investigate the synergistic effects of phage and antibiotic combinations against highly drug-resistant uropathogenic Escherichia coli isolates. The maximum synergistic effects (ΣFIC ≤ 0.5) were observed when phage NTNC80A was combined with ciprofloxacin simultaneously. Cell filamentation, characterised by elongated and interconnected cells, was observed at the sub-inhibitory ciprofloxacin doses (1/4,1/10 of MIC). Ciprofloxacin could accelerate cell lysis by stimulating filamentation, disrupting the peptidoglycan layer and enhancing susceptibility to phage enzymes. Phenotypic changes in phage and antibiotic-resistant mutants indicate that bacteria become susceptible to antibiotics or phages when their growth rate, metabolism, or resistance gene expression is altered. The simultaneous strategy outperformed the sequential strategy by enhancing synergistic effects and minimising the risk of resistance development. A significant reduction (p<0.01-0.001) in bacterial loads in urine, bladder, and kidneys was observed in mice treated with a combination of phage (106 PFU) and sub-inhibitory concentrations of ciprofloxacin. The optimized phage-antibiotic combination reduced tissue bacterial burden and enhanced animal survival rate, indicating improved PAS therapeutic effects. These findings have significant implications for the future of UTI treatment strategies, particularly in the context of growing antibiotic resistance.
    Keywords:  Phage therapy; multidrug-resistance; synergy; urinary tract infections; uropathogenic Escherichia coli
    DOI:  https://doi.org/10.1016/j.micpath.2025.108005
  16. Microorganisms. 2025 Aug 21. pii: 1960. [Epub ahead of print]13(8):
      The escalating global threat of antimicrobial resistance (AMR) underscores the urgent need for innovative therapeutics. Bacteriophages (phages), natural bacterial predators, offer promising solutions, especially when harnessed through advances in artificial intelligence (AI). This review explores how AI-driven innovations are transforming phage biology, with an emphasis on three pivotal areas: (1) AI-enhanced structural prediction (e.g., AlphaFold); (2) deep learning functional annotation; (3) bioengineering strategies, including CRISPR-Cas. We further discuss applications extending to medical therapy, biosensing, agricultural biocontrol, and environmental remediation. Despite progress, critical challenges persist-including high false-positive rates, difficulties in modeling disordered protein regions, and biosafety concerns remain. Overcoming these requires experimental validation, robust computational frameworks, and global regulatory oversight. AI integration in phage research is accelerating the development of next-generation therapeutics to combat AMR and advance engineered living therapeutics.
    Keywords:  CRISPR; artificial intelligence; bacteriophages; biocontrol; phage therapy; phage-based biosensor
    DOI:  https://doi.org/10.3390/microorganisms13081960
  17. Viruses. 2025 Jul 25. pii: 1039. [Epub ahead of print]17(8):
      Phage therapy shows promise as an adjunct to antibiotics for treating Staphylococcus aureus infections. We previously reported a combined flucloxacillin/two-phage cocktail treatment selected for resistance to podovirus phage 66 in a rodent model of methicillin-susceptible S. aureus (MSSA) endocarditis. Here we show that resistant clones harbor mutations in tarS, which encodes a glycosyltransferase essential for β-GlcNAcylation of wall teichoic acid (WTA). This WTA modification has been described in vitro as critical for podoviruses adsorption. Transcriptomics confirmed continued tarS expression in resistant clones, supporting a loss-of-function mechanism. Accordingly, phage 66 binding and killing were restored by WT tarS complementation. In addition, we investigated the counterintuitive innate susceptibility to phage 66 of the tarM + Laus102 strain used in the endocarditis model. We show that it likely results from a significant lower tarM expression, in contrast to the innate resistant strain RN4220. Our findings demonstrate that tarS-mediated WTA β-GlcNAcylation is critical for podovirus infection also in vivo and identify tarM transcriptional defect as a new mechanism of podoviruses susceptibility in S. aureus. Moreover, and since tarS disruption has been previously shown to enhance β-lactam susceptibility, our results support the development of combined podovirus/antibiotic strategies for the management of MRSA infections.
    Keywords:  Staphylococcus aureus; bacteriophage; experimental infective endocarditis; flucloxacillin; phage cocktail; phage therapy; phage-antibiotic synergism
    DOI:  https://doi.org/10.3390/v17081039
  18. Antibiotics (Basel). 2025 Aug 06. pii: 801. [Epub ahead of print]14(8):
       BACKGROUND/OBJECTIVES: Mycobacterium abscessus is an opportunistic pathogen causing infections mainly in patients with immunosuppression and chronic pulmonary pathologies. Extended treatment periods are needed to tackle this pathogen, bacterial eradication is rare, and recurrence can take place with time. New alternative treatments are being investigated, such as bacteriophage therapy. This work describes the characterization of the mycobacteriophage P3MA, showing its ability to infect clinical and standard M. abscessus strains.
    METHODS: Phylogenetic analysis, electron microscopy, growth curves, biofilm assays, checkerboard, and granuloma-like medium studies were performed.
    RESULTS: P3MA inhibited the growth of clinical samples in both planktonic and biofilm states as well as in a granuloma-like model. The study of the interaction with antibiotics revealed that P3MA exhibited an antagonistic effect combined with clarithromycin, indifference with amikacin, and synergy with imipenem.
    CONCLUSIONS: All these results suggest that, after genetic engineering, P3MA could be a promising candidate for phage therapy in combination with imipenem, including lung infections.
    Keywords:  Mycobacterium abscessus; antibiotic; biofilm; granuloma; mycobacteriophage; phage therapy
    DOI:  https://doi.org/10.3390/antibiotics14080801
  19. Microbiol Res. 2025 Aug 20. pii: S0944-5013(25)00279-4. [Epub ahead of print]301 128320
      Due to the rapidly increasing problem of pathogen resistance to drugs, including bacterial resistance to antibiotics, there is a growing need to explore new therapeutic solutions. One of the multidrug-resistant pathogens responsible for many difficult-to-treat infections is Acinetobacter baumannii. The clinical relevance of another species, A. johnsonii, is also increasingly being recognized. As the development pipeline for new antibiotics remains limited, alternative antimicrobial approaches are urgently required. Among these, the therapeutic use of bacteriophages against multidrug-resistant pathogens is currently gaining renewed interest. In this study, we describe the comprehensive characterization of three novel Acinetobacter-specific bacteriophages (Acba_19, Acjo_20 and Acba_21) isolated from sewage samples. The research were conducted using clinical strains obtained from various infection sites of patients in healthcare facilities in Poland. Genomic analysis revealed no markers of temperate phages in any of the isolates, allowing their classification as strictly lytic viruses. In addition to determining basic parameters of phages - such as lytic range, species specificity, virion morphology, adsorption dynamics and infection kinetics - we also conducted stability studies of phage preparations under various conditions. These studies included assessments of pH and temperature stability, evaluation of different cryoprotectants (such as trehalose, glycerol, and pluronic), as well as the effects of storage containers made of glass and plastic materials.
    Keywords:  Acinetobacter spp.; ESKAPE group; antimicrobial resistance; clinical strain; environmental samples; lytic bacteriophage; phage stability
    DOI:  https://doi.org/10.1016/j.micres.2025.128320
  20. Microlife. 2025 ;6 uqaf017
      Bacteriophages are a promising tool for treating bacterial infections, given the rise and spread of antibiotic resistances. However, phage-resistant bacteria can emerge during treatment, jeopardizing the success of therapy. In vitro studies with model organisms have provided valuable insights into the mechanisms by which phage resistance can evolve. However, the relevance of these findings often remains unclear. Here, we investigate the selection of phage-resistant variants and the cost of phage resistance in vitro and in the murine gut using a clinically relevant Escherichia coli K1 strain and a strain-specific phage cocktail. By performing experimental evolution studies in both settings, we obtained different phage-resistant E. coli mutants. Genome resequencing identified lipopolysaccharide (LPS) and the K1 capsule as bacterial surface structures altered in phage-resistant mutants. Targeted deletions of waaO, encoding an ɑ-1,3 glucosyltransferase, involved in the synthesis of the R core of LPS, a gene encoding a predicted O-antigen ligase and emrR involved in capsule gene regulation were generated and confirmed their role in phage-resistance. Escherichia coli mutants deficient in LPS or capsule showed a growth advantage in vitro when exposed to phages but LPS-deficient mutants exhibited severely attenuated growth in the murine gut, even in the presence of phages. Our observations add to the evidence that bacteria in the intestinal environment face a high cost of phage resistance conferred by cell surface alteration, which is not apparent in nutrient-rich culture media. Therefore, it is crucial to carefully consider the context in which phage cocktails are tested, particularly when studying phage efficacy and evolution of phage resistance.
    Keywords:  ESKAPE; Phage therapy; diarrhea; intestinal infection; mutations; phage efficacy; within host evolution
    DOI:  https://doi.org/10.1093/femsml/uqaf017
  21. Scand J Prim Health Care. 2025 Aug 21. 1-9
       BACKGROUND: An updated overview of the antibiotic prescribing pattern in Danish general practice is needed to help inform continued efforts for rational antibiotic prescribing.
    OBJECTIVE: To investigate clinical indications for antibiotic prescriptions issued in general practice in Denmark.
    MATERIALS AND METHODS: This register-based study included all redeemed antibiotic prescriptions issued in Danish general practice between 1 January 2023 and 31 December 2023. Data were extracted from 'Antibiotikastatistik', a publicly available register maintained by the Danish Health Data Authority. Descriptive statistics were used to analyze the distribution of the clinical indications. Furthermore, the distribution of the antibiotics prescriptions was analyzed by age across different clinical indication groups, stratified by gender.
    RESULTS: A total of 1,916,910 antibiotic prescriptions were issued from Danish general practice in 2023. More than half of these prescriptions were used for treatment of either a respiratory tract infection (28.4%) or a urinary tract infection (26.7%). Throat infection and pneumonia comprised about 70% of indications for treatment of a respiratory tract infection. Prophylactic treatment was mainly used for elderly patients - and most often for urinary tract infections. Some 23.3% of the prescriptions either contained an 'unspecific indication' or had no indication stated.
    CONCLUSIONS: This study provides a solid overview of indications used for antibiotic prescriptions in Danish general practice. This information might be used for development of future antibiotic stewardship interventions.
    Keywords:  Antibiotics; clinical indications; community-acquired infections; general practice; prescriptions
    DOI:  https://doi.org/10.1080/02813432.2025.2546415
  22. Polymers (Basel). 2025 Aug 19. pii: 2244. [Epub ahead of print]17(16):
      The progress in biopolymers and their composites as advanced materials for wound healing has revolutionized therapeutic approaches for skin regeneration. These materials can effectively integrate their inherent biocompatibility and biodegradability with the enhanced mechanical strength and customizable properties of polymers and functional additives. This review presents a detailed investigation of the design principles, classifications, and biomedical applications of biopolymeric composites, focusing on their capabilities to promote angiogenesis, exhibit antimicrobial activities, and facilitate controlled drug delivery. By overcoming the challenges of conventional wound dressings, such as inadequate exudate management, mechanical fragility, and cytotoxicity, these composites provide dynamic, stimuli-responsive platforms that can adapt to the wound microenvironment. This study further highlights innovative advances in nanoparticle-assisted reinforcement, fiber-based scaffolds, and multi-stimuli responsive smart delivery systems. Finally, the future perspective illustrates how the challenges related to long-term physiological stability, scalable manufacturing, and clinical implementation can be addressed. Overall, this article delivers a comprehensive framework for understanding the transformative impact of biopolymeric composites in next-generation wound care.
    Keywords:  biopolymers; composites; polymers; skin regeneration; wound healing
    DOI:  https://doi.org/10.3390/polym17162244
  23. Microbiology (Reading). 2025 Aug;171(8):
      Honey bee (Apis mellifera) larvae are susceptible to the bacterial pathogen Paenibacillus larvae, which causes severe damage to bee colonies. Antibiotic treatment requires veterinary supervision in the USA, is not used in many parts of the world, perpetuates problems associated with antibiotic resistance and may necessitate residual testing in bee products. There is interest in using bacteriophages to treat infected colonies (bacteriophage therapy), and several trials are promising. Nevertheless, the safety of using biological agents in the environment must be scrutinized. In this study, we analysed the ability of P. larvae to evolve resistance to several different bacteriophages. We found that bacteriophage resistance rapidly developed in culture but often results in growth defects. Mutations in the bacteriophage-resistant isolates are concentrated in genes encoding potential surface receptors but are also observed in genes controlling general cellular functions and in two cases - lysogeny. Testing one of these isolates in bee larvae, we found it to have reduced virulence compared to the parental P. larvae strain. We also found that bacteriophages are likely able to counteract resistance evolution. This work suggests that while bacteriophage resistance may arise, its impact will likely be mitigated by reduced pathogenicity and secondary bacteriophage mutations that overcome resistance.
    Keywords:  Paenibacillus larvae; bacteriophage resistance; bacteriophage therapy; honey bees
    DOI:  https://doi.org/10.1099/mic.0.001595
  24. J Microbiol Methods. 2025 Aug 20. pii: S0167-7012(25)00148-4. [Epub ahead of print]237 107232
      Artificial intelligence (AI) is revolutionizing antimicrobial drug discovery by delivering major improvements in precision, innovation, and efficiency for combating bacterial, fungal, and viral pathogens. Traditional approaches to developing treatments for microbial infections are often hampered by high costs, lengthy timelines, and frequent failures. Modern AI technologies, particularly deep learning, machine learning, computational biology, and big data analytics, provide robust solutions to these challenges by analyzing large-scale biological datasets to predict molecular interactions, identify promising treatment candidates, and expedite both preclinical and clinical development. Innovative techniques such as generative adversarial networks for novel compound discovery, reinforcement learning for optimizing antimicrobial candidates, and natural language processing for extracting knowledge from biomedical literature are now vital to infectious disease research. These approaches facilitate early toxicity prediction, microbial target identification, virtual screening, and the development of more individualized therapies. Notwithstanding these advances, challenges remain, including inconsistent data quality, limited interpretability, and unresolved ethical or legal concerns. This review examines recent advancements in AI applications for microbial drug discovery, with a focus on de novo molecular design, ligand- and structure-based screening, and AI-enabled biomarker identification. Remaining application barriers and promising future directions in AI-driven antimicrobial drug development are also elucidated. Collectively, these innovations are poised to accelerate the discovery of new therapies, reduce costs, and enhance patient outcomes in the fight against infectious diseases.
    Keywords:  Antimicrobial resistance; Artificial intelligence; Drug discovery; Genomics; Microbial pathogens
    DOI:  https://doi.org/10.1016/j.mimet.2025.107232
  25. Microorganisms. 2025 Aug 07. pii: 1850. [Epub ahead of print]13(8):
      The Mycoplasmataceae are a family of bacteria that typically cause respiratory, arthritic, and genitourinary disease in humans. Mycoplasma spp. of animal origin are also the causative agents of porcine wheezing disease, chronic respiratory disease and arthritis in chickens and other conditions. These diseases have a significant impact on public health and the economic development of livestock breeding. Clinical prevention and treatment of mycoplasma infections is primarily dependent on the use of antibiotics. However, inappropriate and excessive use of antimicrobials has enabled resistance development that has become a significant clinical concern. Mycoplasma are also robust biofilm producers, and this process is a major factor for the persistence of these infections, especially in conjunction with common antibiotic resistance mechanisms, including target gene mutations and the action of efflux pumps. A mycoplasma biofilm refers to a structured and stable microbial community formed by Mycoplasma spp. adhering to biological or non-biological surfaces under suitable conditions and secreting extracellular polymers (EPS) such as polysaccharides. This process allows the microorganisms to adapt to their surrounding environment and survive during the growth process. These biofilms render bacteria more resistant to antimicrobials than planktonic bacteria, resulting in biofilm-associated infections that are more challenging to eradicate and more likely to recur. The current study reviews progress from the fields of biofilm formation, structure and identification, correlations between biofilms and drug resistance and virulence as well as methods of biofilm prevention and control. Our aim was to provide a reference basis for the subsequent in-depth understanding of the research of mycoplasma biofilms.
    Keywords:  Mycoplasma; biofilm; drug resistance; virulence
    DOI:  https://doi.org/10.3390/microorganisms13081850