bims-fagtap Biomed News
on Phage therapies and applications
Issue of 2025–12–21
28 papers selected by
Luca Bolliger, lxBio



  1. Curr Opin Microbiol. 2025 Dec 17. pii: S1369-5274(25)00118-3. [Epub ahead of print]89 102696
      Bacteriophages (phages) are viruses that selectively prey on bacteria. Their use in treating antimicrobial-resistant bacterial infections is steadily increasing due to the need for alternative therapies. The application of phage therapy is not without its challenges, including difficulties associated with isolating phages against a target strain, the limited infectivity of a phage, the cost and complexity of producing well-characterised phage stocks, and the emergence of phage resistance. The directed adaptation of phage to a specific bacterial target, also known as 'phage training', leverages the natural evolutionary capacity of phages and can be used to bolster their bacterial killing abilities. Phage training dates back almost as far as phage therapy itself, being used to expand the therapeutic use of phages. Numerous reports showcase the success and benefits of phage training in vitro and its potential to operate effectively within the framework of phage therapy. However, the time needed to train a given phage, followed by genotypic and phenotypic characterisation of both pre- and post-trained phages, is a major limitation. Here, we explore oversights of the phage training process and propose some considerations and solutions to help drive the field forward to enable its feasible integration into phage therapy.
    DOI:  https://doi.org/10.1016/j.mib.2025.102696
  2. Microbiol Res. 2025 Dec 16. pii: S0944-5013(25)00376-3. [Epub ahead of print]305 128417
      Pseudomonas aeruginosa is a major opportunistic pathogen implicated in a wide range of infections, including chronic respiratory infections, burn wound infections, urinary tract infections, and device-associated infections. Its intrinsic and acquired resistance mechanisms, particularly its capacity for biofilm formation, pose serious challenges to conventional antibiotic therapy. With the continued rise of multidrug-resistant and pan-drug-resistant strains, the need for alternative therapeutic strategies has become increasingly urgent. Phages, viruses that specifically recognize and lyse bacteria, have shown unique advantages in combating antibiotic-resistant infections. This review systematically summarizes recent advances in the application of phage therapy for P. aeruginosa infections, covering in vitro bactericidal activity, biofilm degradation, and synergistic interactions with antibiotics. We further discuss evidence from animal models, including therapeutic efficacy, immunomodulatory effects, and pharmacokinetics. Emphasis is placed on clinical use cases, including different routes of administration, symptom relief, biomarker modulation, pathogen clearance rates, and adverse events. Typical case reports and early-phase clinical trials support the safety and efficacy of phage therapy. Nevertheless, translational barriers persist, such as the need for precise host matching, risks of immune neutralization, and the lack of standardized regulatory frameworks and Good Manufacturing Practice (GMP)-grade production systems. The rapid development of engineered phages and individualized therapeutic approaches offers a feasible path forward. In conclusion, phage therapy holds significant promise for the treatment of drug-resistant P. aeruginosa infections, and future efforts should focus on establishing standardized systems, conducting multicenter clinical studies, and leveraging synthetic biology to accelerate its translation from bench to bedside.
    Keywords:  Antibiotic synergy; Biofilm; Multidrug resistance; Personalized therapy; Phage therapy; Pseudomonas aeruginosa
    DOI:  https://doi.org/10.1016/j.micres.2025.128417
  3. Crit Rev Oncol Hematol. 2025 Dec 13. pii: S1040-8428(25)00473-1. [Epub ahead of print]218 105085
      Bacteriophage therapy has re-emerged as a rapidly advancing field in oncology, bridging antimicrobial precision with tumor-targeted biotherapy. Beyond infection control, phages are now recognized as programmable biological systems capable of eradicating multidrug-resistant (MDR) pathogens, modulating tumor-associated microbiota, activating immune responses, and delivering therapeutic genes or drugs. Preclinical evidence shows that phages can selectively eliminate Fusobacterium nucleatum in oral squamous cell carcinoma, restore microbial balance in colorectal cancer, and enhance immune infiltration via cytokine or antigen display. Engineered constructs including GM-CSF-expressing and MAGE-A1-displaying phages, λ-phage ASPH vaccines, and PEGylated nanocarriers delivering MEG3 or TRAIL have demonstrated strong anti-tumor efficacy across melanoma, hepatocellular, and colorectal cancer models. Additionally, CRISPR-Cas-armed phages precisely remove resistance genes such as bla-CTX-M and mecA, while AI-driven selection pipelines enable data-guided design of personalized phage cocktails. These advances represent a paradigm shift from empirical antibacterial use toward mechanistically engineered, multifunctional phage platforms that integrate microbiome modulation, immune activation, and nanocarrier-mediated gene delivery. Although challenges such as immune clearance, bacterial resistance, and regulatory complexity remain, the convergence of AI, CRISPR, and synthetic biology is accelerating the evolution of phage therapy into a clinically viable precision-oncology strategy. In this context, bacteriophages emerge not merely as antibacterial agents but as intelligent, patient-specific nanomedicines poised to redefine therapeutic boundaries in cancer treatment.
    Keywords:  Cancer immunotherapy; Engineered bacteriophages; Multidrug-resistant infections; Phage therapy; Tumor microenvironment
    DOI:  https://doi.org/10.1016/j.critrevonc.2025.105085
  4. Sci Rep. 2025 Dec 19.
      Infections caused by multidrug-resistant bacterial pathogens are becoming increasingly frequent, so alternative therapeutic strategies such as phage therapy are gaining in importance. However, one of the main challenges in phage therapy is the rapid emergence of phage-resistant bacteria. Hence, this study investigated the occurrence of phage resistance in Bordetella avium, the causative agent of bordetellosis in poultry of all ages, with a special focus on the association between bacterial phage resistance and antibiotic resistance. Here, we describe the isolation and analysis of four B. avium mutants that showed resistance to B. avium phages, with decreased adsorption leading to phage resistance of the mutants. SDS-PAGE analysis revealed a lipopolysaccharide (LPS) structure with altered nuclear patterns and a missing O-antigen moiety in the phage-resistant B. avium mutants. Whole-genome sequence analysis revealed mutations in the genetic loci BAV2233 and BAV2235, encoding lipopolysaccharide glucosyltransferase family 4 proteins, and BAV0511, encoding Vi polysaccharide biosynthesis UDP-N-acetylglucosamine C-6 dehydrogenase TviB protein, which are involved in LPS biosynthesis. Additionally, antimicrobial susceptibility testing against nine antibiotics using the agar dilution method showed that LPS changes in phage-resistant mutants affected the susceptibility of B. avium to two of the antibiotics examined. Specifically, the MIC values of the polymyxin antibiotics colistin and polymyxin B decreased by up to five two-fold dilutions, depending on the isolate considered. An association was observed between the length of the polysaccharide chain in LPS and the susceptibility of B. avium to colistin and polymyxin B, with shorter chains indicating higher susceptibility. Collective findings illuminate a beneficial aspect that may support the use of B. avium phages. Based on these results, genetically modified phages based on these B. avium phages would be a promising direction for future phage applications as an alternative to the use of antimicrobial agents.
    Keywords:   Bordetella avium ; Antimicrobial susceptibility; Bacterial membrane; LPS; Phage resistance
    DOI:  https://doi.org/10.1038/s41598-025-30405-7
  5. Front Microbiol. 2025 ;16 1672198
      Phage therapy has emerged as a promising alternative to conventional antibiotics for combating intestinal bacterial infections, especially in the era of rising antimicrobial resistance. Despite its therapeutic potential, the clinical translation of phage therapy remains hindered by limited large-scale trial data and incomplete mechanistic understanding. This review systematically evaluates the efficacy of phage therapy in animal models of intestinal diseases, encompassing bacterial infection-induced diarrhea (e.g., cholera, typhoid fever), bacterial enteritis, and sepsis. By synthesizing evidence from bacterial colonization assays, histopathological analyses, and disease severity assessments, we highlight features such as phage-mediated pathogen clearance, changes in inflammatory factors, and intestinal pathology. Furthermore, challenges including phage selection difficulties, host specificity issues, and safety considerations are discussed, along with future research directions aimed at bridging the gap between experimental models and clinical applications.
    Keywords:  antimicrobial resistance; intestinal infections; microbiome modulation; phage therapy; translational medicine
    DOI:  https://doi.org/10.3389/fmicb.2025.1672198
  6. Microbiome Res Rep. 2025 ;4(4): 40
      The gut virome, particularly its viral and phage components, is increasingly recognized as a key modulator of intestinal microbial dynamics in gastrointestinal inflammatory diseases. Beyond well-characterized bacterial dysbiosis, growing evidence suggests that virome alterations contribute to the development and progression of inflammatory bowel disease, metabolic dysfunction-associated steatohepatitis, alcoholic hepatitis, primary sclerosing cholangitis, primary biliary cholangitis, and pancreatitis. As the most abundant viruses in the gut, bacteriophages influence microbial ecosystem stability and host immune responses through lytic and lysogenic interactions with bacterial populations. Amid the growing burden of multidrug-resistant infections and heightened interest in microbiota-based interventions, phage therapy has re-emerged as a viable strategy in both preclinical and translational contexts. This review synthesizes recent insights into bacteriophage dynamics in the context of major gastrointestinal and hepatopancreatic inflammatory diseases, highlighting potential compositional shifts, proposed mechanisms of phage-microbe interactions, and supportive evidence from animal models and early clinical applications. We also discussed the critical challenges that had to be addressed to enable clinical translation, including host range restrictions, resistance and safety concerns, immunogenicity, and delivery limitations, while emphasizing emerging strategies such as phage engineering, encapsulation technologies, and standardized regulatory frameworks.
    Keywords:  Gut virome; bacteriophages; digestive disease; phage therapy
    DOI:  https://doi.org/10.20517/mrr.2025.62
  7. mSystems. 2025 Dec 17. 10(12): e0138425
      Bacteriophages are emerging as promising alternatives to antibiotics for multidrug-resistant (MDR) infections. However, their unique pharmacokinetic and pharmacodynamic (PKPD) properties arising from host-dependent amplification present challenges for dose selection and clinical translation. Here, we present a mechanistic PKPD model informed by in vitro kinetic assays and in vivo mouse studies of phage therapy targeting MDR Pseudomonas aeruginosa. The model extends the classical predator-prey model by addressing dormancy-related bacterial persistence and partitioning bacterial subpopulations based on phage susceptibility profiles. Simulations revealed a non-monotonous dose-exposure curve driven by dose-dependent reduction of phage replication and the importance of cross-resistance in selecting optimal phage cocktails. In vivo, host immunity was identified as a crucial component in inhibiting bacterial regrowth, with bistable outcomes dependent on initial bacterial load and immune competence. Dose-ranging simulations under varying immune statuses suggest that long-term bacterial load is solely determined by host immune function. However, higher doses transiently reduce bacterial load to a greater extent and thereby suppress immune activation. In immunocompetent hosts, phage cocktails can enhance maximal bacterial load reduction when administered at doses higher than a critical threshold. In conclusion, our PKPD framework enables optimal selection of phage cocktails and dosing regimens, supports rational design of first-in-human trials of phage therapy, and potentially advances model-informed drug development for replication-competent biologics.IMPORTANCEIn this study, we construct an integrative model of phage-bacteria dynamics and investigate whether its calibration to in vitro kinetic assay data can inform the rational design of phage therapy regimens and cocktails. Our findings demonstrate a dose range within which lower phage doses yield higher long-term exposure, presenting a fundamentally different framework for dose optimization. Analysis of phage cocktails reveals that combining phages with low cross-resistance delays the regrowth of phage-resistant bacteria in vitro. The extended in vivo model elucidates key differences between in vitro and in vivo outcomes and highlights the importance of the host's immune response in suppressing the growth of phage-resistant bacteria. Phage cocktails to combat phage resistance are therefore of less importance in immune-competent individuals but can enhance bacterial killing when administered at sufficiently high doses. We propose that this modeling framework holds potential for model-informed drug development by quantitatively characterizing bacteria-phage dynamics using preclinical data. Furthermore, it may facilitate the interpretation of in vivo therapeutic outcomes through a mechanistic understanding derived from in vitro observations.
    Keywords:  Pseudomonas aeruginosa; bacteriophage therapy; bacteriophages; computational biology; mathematical modeling; model-informed drug development; phage cocktail optimization; pharmacokinetics pharmacodynamics; predator-prey dynamics; quantitative systems pharmacology
    DOI:  https://doi.org/10.1128/msystems.01384-25
  8. Biofilm. 2025 Dec;10 100333
      Given the high resistance levels in Gram-negative bacteria, phage therapy is receiving increasing attention. In Germany, a clinical study is already underway evaluate a phage cocktail for treating Pseudomonas aeruginosa in cystic fibrosis (CF) patients. In this context, we investigated the prevalence of PF1-like prophages in P. aeruginosa isolates from a local CF cohort, their ability to undergo lytic conversion during biofilm formation, and the resulting impact on the resistance profile of the P. aeruginosa population. Consistent with other studies, prophage Pf4 was the most prevalent in this cohort and became during biofilm formation even in the absence of external triggers. This lytic conversion rapidly generated a subpopulation resistant to the virulent phages, potentially complicating phage therapy. However, this subpopulation also became more susceptible to most antibiotics commonly used in CF, suggesting a potential therapeutic opportunity. Interestingly, this bacterial subset lost its susceptibility to colistin, an important inhaled antibiotic in CF, which could increase the risk of treatment failure. These findings underscore both the challenges and potential strategies for improving treatment outcomes in CF patients.
    Keywords:  Antibiotic resistance; Biofilms; Colistin; Pf1-like; Temperate bacteriophages; Tobramycin
    DOI:  https://doi.org/10.1016/j.bioflm.2025.100333
  9. APMIS. 2025 Dec;133(12): e70122
      Biofilms are microbial communities enclosed in an extracellular polymeric substance (EPS), significantly contributing to antimicrobial resistance (AMR) in medical, industrial, and environmental settings. Their matrix enhances microbial survival, inhibits antibiotic penetration, and facilitates horizontal gene transfer, worsening the AMR crisis. Conventional antimicrobial treatments often fail against biofilms, necessitating novel therapeutic strategies. Emerging biofilm-targeted interventions, such as nanotechnology-based antimicrobials, bacteriophage therapy, and CRISPR-Cas9 gene editing, offer promising solutions. Nanoparticles improve drug delivery, bacteriophages selectively lyse resistant bacterial populations, and CRISPR-Cas9 disrupts AMR-related genes and biofilm virulence factors. Additionally, AI and ML are advancing biofilm prediction models and antimicrobial optimization, paving the way for precision-targeted interventions. This review explores biofilm biology and next-generation biofilm control strategies, with a focus on AI-driven bioinformatics. Future research should focus on clinical translation, regulatory standardization, and scalable implementation in healthcare and industrial settings to combat biofilm-associated AMR.
    Keywords:  AI in biofilms; AMR; CRISPR‐Cas9; biofilms; nanotechnology; synthetic biology
    DOI:  https://doi.org/10.1111/apm.70122
  10. Hum Vaccin Immunother. 2025 Dec;21(1): 2599632
      Bacteriophages (phages) have recently received increased interest as versatile candidates for vaccine development. Their inherent characteristics, such as ease of genetic manipulation, high-density antigen display, intrinsic immunostimulatory properties, demonstrated human safety, and scalability in bacterial hosts, make them attractive as next-generation vaccine platforms. Additionally, their cost-effective production, stability, and existing regulatory approval for food and compassionate phage therapy provide a strong foundation for further development of phage-based vaccines. This commentary summarizes the types of phages, the strategies used, and current advances in phage-based vaccine development for viral and bacterial targets, and discusses the promises and challenges of this platform for novel vaccine development. Phage-based vaccines represent an innovative and promising platform for vaccine development to address significant medical and public health challenges, particularly in antimicrobial resistance, pandemic preparedness, and One Health. Accumulative experimental data have demonstrated that phage-based vaccines induce specific cellular, humoral, and mucosal immune responses at magnitudes comparable to those induced by other vaccine platforms. However, a better understanding of phage biology (interactions with the human immune system and microbiome), more carefully designed preclinical studies, Good Manufacturing Practice production development, the regulatory framework, and ultimately clinical trials are needed before the full potential of this platform is realized.
    Keywords:  One Health; Vaccine development; bacteriophage; pandemic responses; phage vaccines
    DOI:  https://doi.org/10.1080/21645515.2025.2599632
  11. Macromol Biosci. 2025 Dec 18. e00528
      Implant-associated osteomyelitis (IAO) is a major clinical challenge due to persistent biofilms, antibiotic resistance, and impaired osteogenesis. Hydrogels, with tunable physicochemical properties, biocompatibility, and localized drug delivery capabilities, offer advanced solutions to these problems. This review systematically examines advanced hydrogel-based strategies for IAO treatment, categorized into two primary approaches. Antibiotic-loaded hydrogels leverage nanomaterial integration and hybrid composites to achieve precise, spatiotemporal drug release, thereby minimizing toxicity and resistance. Non-antibiotic approaches, including nanomaterial-based agents such as metals and photothermal nanohybrids, as well as peptides, plant polyphenols, and phage therapy, provide alternative options to circumvent antibiotic resistance. Crucially, we highlight key optimization strategies that encompass controlled cross-linking, stimuli-responsive systems (e.g., pH and temperature), anti-biofilm mechanisms, and biomimicry, synergistically enhancing both antibacterial and osteogenic functions in these platforms. Collectively, these advances signify a shift from passive drug carriers to multifunctional, bioactive platforms that both eradicate resistant bacteria and support bone regeneration. This transformative shift, however, reveals persistent challenges while suggesting promising research avenues for advancing hydrogel-based therapies against IAO.
    Keywords:  antibacterial agent; biofilm eradication; bone regeneration; hydrogels; implant‐associated osteomyelitis
    DOI:  https://doi.org/10.1002/mabi.202500528
  12. Cardiovasc Diabetol Endocrinol Rep. 2025 Dec 19. 11(1): 45
      Diabetic Foot Ulcers (DFUs) are a serious complication of diabetes, often leading to chronic infections, prolonged hospitalization, and, in severe cases, amputations. A major challenge in DFU treatment is the presence of polymicrobial biofilms, complex bacterial communities encased in a protective extracellular matrix, which enhances their survival, evade immune responses, and contributes to antimicrobial resistance (AMR). Conventional treatments, such as systemic antibiotics, antiseptics, and mechanical debridement, often fail to fully eliminate biofilms, highlighting the need for more effective therapeutic strategies. Considering the existing ineffective anti-biofilm treatments, super-oxidized solution (SOS) has emerged as a promising non-antibiotic alternative for DFU infections. As a reactive oxygen species (ROS)-based therapy, SOS effectively disrupts biofilm integrity, enhances bacterial eradication, and promotes wound healing by reducing inflammation and supporting tissue regeneration. Additionally, its mechanism of action could significantly minimize the risk of AMR, making it a valuable adjunct to conventional therapies. The purpose of this review is to explore the mechanisms of biofilm formation in DFUs, evaluate the limitations of current treatment approaches, and highlight the therapeutic potential of SOS. Integrating SOS into DFU treatment protocols could improve infection control, reduce chronic wound burden, and enhance patient outcomes. By advancing non-antibiotic solutions like SOS, DFU management can shift toward more effective and sustainable wound care strategies, ultimately reducing the need for surgical interventions and improving the quality of life for diabetic patients.
    Keywords:  Antimicrobial resistance; Biofilms; Diabetic foot ulcers; Super-oxidized solution; Wound healing
    DOI:  https://doi.org/10.1186/s40842-025-00261-5
  13. mSystems. 2025 Dec 15. e0129025
      Hidradenitis suppurativa (HS) is a chronic inflammatory disease characterized by recurring skin lesions. Despite ongoing research, the exact cause underlying initiation and progression of disease remains unknown. While prior research has linked the skin microbiota to HS pathology, the role of viruses has remained unexplored. To investigate the skin virome, metagenomic sequencing of viral particles was performed on 144 skin samples from 57 individuals (39 HS patients and 18 controls). It was found that the virome is not only linked to BMI, but also to the presence and severity of HS, marking a diverging viral profile in the progression of disease. Despite no differences in alpha-diversity, HS patients exhibited a significantly higher beta-diversity compared to healthy controls, indicating a more personalized virome with reduced viral sharing among patients. We identified distinct groups of commonly shared phages, referred to as the core phageome, associated with either healthy controls or patients. Healthy controls displayed a higher abundance of two core Caudoviricetes phages predicted to infect Corynebacterium and Staphylococcus, comprising normal skin commensals. In contrast, HS patients carried previously uncharacterized phages that were more prevalent in advanced stages of the disease, which likely infect Peptoniphilus and Finegoldia, known HS-associated pathogens. Interestingly, genes involved in superinfection exclusion and antibiotic resistance could be found in phage genomes of healthy controls and HS patients, respectively. In conclusion, we report the existence of distinct core phages that may have clinical relevance in HS pathology by influencing skin bacteria through mechanisms such as superinfection exclusion and antibiotic resistance.IMPORTANCEAn increasing body of research showed that the microbiome has an important role in complex human disease. In line with this, here, we analyzed a longitudinal HS cohort and found a relationship between the skin virome and HS pathology. This relationship was defined by distinct groups of phages associated with either healthy controls or HS patients, yet, in both instances, capable of enhancing bacterial fitness. In healthy individuals, these phages were widely shared, fostering symbiosis by ensuring stability of the commensal skin microbiota. Conversely, in HS patients, these phages revealed a more individualistic nature and could contribute to dysbiosis by providing antibiotic resistance genes to bacterial pathogens. Overall, these findings point to a potential clinical significance of the virome in understanding and addressing HS pathology.
    Keywords:  bacteriophages; hidradenitis suppurativa; metagenomics; microbiome; virome
    DOI:  https://doi.org/10.1128/msystems.01290-25
  14. Front Vet Sci. 2025 ;12 1695255
       Introduction: The rise of multidrug-resistant (MDR) Salmonella enterica poses a significant threat to public health, veterinary medicine, and food safety. Bacteriophages offer a promising alternative to antibiotics due to their host specificity and ability to lyse bacteria without disrupting commensal microbiota.
    Methods: In this study, twelve Salmonella-specific phages were isolated from diverse environmental sources across China. Specifically, DN01 originated from livestock manure in Shanxi Province (Northwest China), DN03 from poultry wastewater in Suizhou, Hubei Province (Central China), DN19 from slaughterhouse effluent in Guangdong Province (South China), and DN28 from hospital sewage in Zhejiang Province (Eastern China). This geographic and ecological diversity underscores the broad natural distribution of Salmonella phages, providing a representative foundation for subsequent morphological, genomic, and therapeutic analyses. The host bacterium Salmonella Enteritidis SE006, was used for phage propagation. Four lytic phages, which exhibited broad host ranges, were selected for in-depth characterization. Their biological properties, including optimal multiplicity of infection (MOI), latent period, burst size, pH, and thermal stability, and anti-biofilm activity, were systematically evaluated. Morphology was analyzed via transmission electron microscopy (TEM), and whole-genome sequencing, functional annotation, and phylogenetic analysis were conducted to assess genetic safety and taxonomic placement.
    Results: All phages exhibited potent lytic activity across multiple Salmonella serovars, including MDR strains, with MOIs as low as 0.00001 and short latent periods (10-20 min). They remained stable over a broad pH range (3-11) and exhibited thermal stability from 4 °C up to 50 °C, with partial loss of activity observed at 60 °C for some phages. Genomes ranged from 5,563 to 86,377 bp and lacked genes related to lysogeny, virulence, or antibiotic resistance. TEM and phylogenetic analyses classified the phages within distinct families of the Caudoviricetes class. In vitro assays demonstrated significant inhibition of bacterial growth and disruption of mature biofilms. Phage treatment significantly improved the survival of S. Enteritidis-infected mice over a 12-day period., survival rates were 0% (PBS), 25% (DN01), 33% (DN03), 75% (DN19), 85% (DN28), and 100% (cocktail).
    Conclusion: These results highlight the promise of DN01, DN03, DN19, and DN28, particularly in cocktail form, as safe, stable, and effective agents for phage-based control of multidrug-resistant Salmonella enterica.
    Keywords:  Salmonella enterica; biofilm; multidrug-resistant (MDR); phage; phage cocktails; phage therapy
    DOI:  https://doi.org/10.3389/fvets.2025.1695255
  15. JAC Antimicrob Resist. 2025 Dec;7(6): dlaf233
       Background: The global rise of antimicrobial resistance requires innovative and affordable wound care solutions. Moreover, managing wounds infected with priority pathogens remains a challenge. Despite the widespread availability of over-the-counter (OTC) antiseptics in wound care, comparative studies on their efficacy against biofilms or multidrug-resistant pathogens are limited.
    Objectives: This study compares the ability of different OTC antiseptics to disrupt biofilms of multidrug-resistant clinical isolates of Acinetobacter baumannii and Pseudomonas aeruginosa.
    Methods: The antimicrobial activity of seven antiseptics (polyhexanide, octenidine, chloroxylenol, chlorhexidine, ethanol, cetrimide, phenol) against early-stage (3-hour) and mature (24-hour) biofilms was determined by measuring reductions in colony-forming units (cfu)/biofilm while varying treatment exposure time.
    Results: All OTC products significantly reduced early-stage biofilms of both pathogens below detectable limits within 5 minutes (P < 0.0001, n = 3, LOD = 100 cfu/biofilm). In mature biofilms, significant differences emerged. Polyhexanide, octenidine and cetrimide yielded modest reductions in cfu count/biofilm (0.55-0.64-log) after 5 minutes, while chloroxylenol and phenol achieved ∼2.5-log reductions; notably, chlorhexidine reduced cfu/mature biofilms below detectable limits within 5 minutes. Extended exposure (60 minutes) enhanced the efficacy of phenol and ethanol, with chloroxylenol and octenidine reducing cfu/biofilm below detectable limits.
    Conclusion: OTC antiseptics are effective in eliminating early-stage biofilms; however, mature biofilms require either prolonged exposure, which may increase their toxicity and delay wound healing, or the use of potent formulations. Chlorhexidine gluconate, chloroxylenol and phenol offer an optimal balance between antibiofilm potency and tissue safety, offering promise for acute and chronic wound management particularly in low-resource settings.
    DOI:  https://doi.org/10.1093/jacamr/dlaf233
  16. Front Endocrinol (Lausanne). 2025 ;16 1689232
       Background: Diabetic foot ulcers impair quality of life and prognosis in diabetes. IL-6 and TNF-α regulate wound healing through inflammation. Although negative pressure wound therapy and silver dressings aid chronic wound repair, their combined efficacy in DFUs remains understudied.
    Objective: This study aimed to assess how negative pressure occlusion drainage combined with silver ion dressing affects inflammatory cytokine levels (IL-6, TNF-α) and wound healing in patients with diabetic foot ulcer.
    Methods: This study included 78 DFU patients treated with NPWT plus silver dressings (February 2023-April 2025). Serum IL-6 and TNF-α levels were measured at baseline, day 14, and day 30 post-treatment.
    Results: The findings indicated that IL-6 and TNF-α levels were significantly reduced at 14- and 30-days post-treatment compared to pre-treatment levels (P < 0.001), and the findings indicated a notable reduction in the wound area, with a healing duration of 19.49 ± 4.18 days and granulation tissue appearing in 6.83 ± 1.85 days. After 30 days of treatment, the healing rate reached 93.59%. The incidence of adverse reactions was low, 6.41%, and most of them were mild skin itching, nausea and vomiting, and a small amount of diarrhea.
    Conclusion: Negative pressure occlusion drainage combined with silver ion dressing has potential advantages in reducing inflammatory response and promoting the healing of diabetic foot ulcers, which is worthy of clinical application. In the future, multicenter randomized controlled trials are needed to further verify its efficacy and safety.
    Keywords:  diabetic foot; inflammatory factors; negative pressure closed drainage; silver ion dressings; ulcers
    DOI:  https://doi.org/10.3389/fendo.2025.1689232
  17. Sci Rep. 2025 Dec 16.
      Ulcerative colitis (UC) is a chronic inflammatory disease of the colon, associated with gut microbiota dysbiosis. While global studies have explored this link, region-specific microbial profiles remain underreported. This pilot study aimed to characterize and compare, for the first time, the gut microbiota of Lebanese UC patients and healthy controls using 16 S rRNA gene sequencing (V3-V4 region). Fecal samples from 11 UC patients and 11 healthy individuals were analyzed. Alpha and beta diversity metrics were computed, and gut microbial composition was assessed across taxonomic levels. Statistical comparisons used Mann-Whitney and Fisher's exact tests. UC patients showed significantly reduced microbial diversity based on Faith's Phylogenetic Diversity and Shannon index (p < 0.05), though evenness was unaffected. Beta diversity also revealed significant group-level dissimilarities (p < 0.05). At the phylum level, Bacteroidota was elevated in UC, while Bacillota and Actinomycetota were reduced. Genera such as Ruminococcus, Bacteroides, and Coprococcus were depleted in UC. Faecalibacterium, commonly reduced in UC, showed no significant difference. This first analysis of gut microbiota in Lebanese UC patients reveals a distinct microbial signature that partially diverges from global trends, supporting the need for region-specific microbiome studies and personalized microbiota-targeted therapies.
    Keywords:  Amplicon sequencing; Faecalibacterium; Gut microbiota; Lebanese gut microbiota.; Ulcerative colitis
    DOI:  https://doi.org/10.1038/s41598-025-31435-x
  18. Int J Food Microbiol. 2025 Dec 08. pii: S0168-1605(25)00519-7. [Epub ahead of print]448 111574
    TÜBİTAK-121Z447 project scholars and consultants
      The rise of multidrug-resistant Salmonella poses a significant threat to food safety and public health, necessitating novel antimicrobial strategies. The primary objective of this study was to characterize novel bacteriophages and assess their biocontrol potential against predominant Salmonella serotypes. A total of 84 lytic bacteriophages specific to various Salmonella enterica serotypes were isolated from wastewater sources across Türkiye. Five phages (S.Hadar 4-5-1, S.Inf 5-2, S.Typ Adana, S.Ent 1-35-3, and S.Kent 1-2-1) demonstrating broad lytic activity to tested major serotypes (S. Enteritidis, S. Typhimurium, S. Infantis, S. Kentucky, S. Newport, S. Hadar, S. Gallinarum and S. Pullorum) and genetic diversity were selected for detailed phenotypic and genomic analysis. These phages, four from Siphoviridae and one from Podoviridae, exhibited tolerance to thermal (up to 60 °C) and mildly acidic conditions (pH 4), as well as 12-month stability when stored in Tris-Buffered Saline (TBS) with 20 % (v/v) glycerol at -20 °C and - 80 °C. Whole-genome sequencing confirmed their novelty and the absence of antimicrobial resistance and virulence genes. A cocktail formulated from these phages was applied against Salmonella Enteritidis both in-vitro (at 37 °C) and on artificially contaminated chicken wings (at 4 °C). The phage cocktail effectively reduced Salmonella counts in both environments, keeping levels below the detection limit (< 1 log CFU/g) over 24 h. For chicken wings food model, bacterial reductions reached 3.30 log CFU/g and 4.86 log CFU/g. These results underscore the potential of the newly characterized Salmonella phages as effective tools for controlling bacterial contamination on chicken meat, supporting their use as a natural, and antibiotic-free strategy in modern food safety management.
    Keywords:  Multidrug-resistant Salmonella; Phage biocontrol; Phage genome analysis; Phage storage conditions; Poultry meat safety; Salmonella reduction
    DOI:  https://doi.org/10.1016/j.ijfoodmicro.2025.111574
  19. Arch Virol. 2025 Dec 18. 171(1): 26
      Vibrio alginolyticus is a zoonotic pathogenic bacterium that causes various diseases in marine animals, including Penaeus vannamei. Phage therapy, as a natural antibiotic, offers a novel approach to the prevention and control of vibriosis. In this study, we isolated, identified, and characterized a novel bacteriophage targeting V. alginolyticus, designated as PV48. PV48, a short-tailed bacteriophage isolated from the Ningbo aquaculture farm, possesses the ability to lyse V. alginolyticus strains that affect shrimp. It displays stability over a range of temperatures (30-50°C) and pH values (4-10). The latent period and burst size for phage PV48 were determined to be 20 minutes and 15.6 PFU/cell, respectively. Whole-genome sequencing revealed that PV48 has a dsDNA genome with a length of 43,398 bp and a G + C content of 49.54%. The phage genome contains 44 open reading frames (ORFs). Comparative genomic analysis suggested that PV48 might represent a new species within the genus Maculvirus. Phylogenetic analysis indicated a close evolutionary link between PV48 and other Vibrio-targeting phages, suggesting a shared viral lineage. This study lays the groundwork for the application of V. alginolyticus bacteriophage in aquaculture.
    Keywords:   Penaeus vannamei ; Vibrio alginolyticus ; Vibrio phage; Biological characterization; Genomic analysis
    DOI:  https://doi.org/10.1007/s00705-025-06482-4
  20. Lancet Reg Health West Pac. 2025 Dec;65 101754
    KIDS DOSE group
      Antimicrobial resistance poses a significant threat to children's health, with up to 20% of 1.27 million deaths attributable to bacterial AMR annually, occurring in children <5 years. The WHO 2024 Bacterial Priority Pathogens List identifies methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant Enterococcus faecium (VRE) as critical pathogens. This review examines the epidemiology, treatment recommendations, dosing strategies, efficacy, and safety data for antibiotics targeting MRSA and VRE infections in children in Oceania. Paediatric MRSA infections are prevalent (13-43%) across Oceania, while VRE infections remain uncommon (3-5%). Disparate access to recommended treatments, particularly in Pacific Island Countries and Territories, highlights the need for paediatric licensing. Paediatric trials primarily assess safety, with efficacy data limited to vancomycin, teicoplanin, and daptomycin. Pharmacokinetic/pharmacodynamic studies show standard dosing in children under 12 years often fails to achieve therapeutic targets, highlighting the need for dedicated dosing studies. Addressing these gaps is essential to advancing paediatric access to optimal treatment for drug-resistant infections in the region.
    Keywords:  Antimicrobial resistance; Dosing; Epidemiology; Gram-positive infection; Methicillin-resistant Staphylococcus aureus (MRSA); Oceania; Paediatrics; Pharmacokinetic-pharmacodynamic data; Vancomycin-resistant Enterococcus faecium (VRE)
    DOI:  https://doi.org/10.1016/j.lanwpc.2025.101754
  21. Recent Adv Inflamm Allergy Drug Discov. 2025 ;19(3): 343-358
      This extensive analysis explores the dynamic interface between precision medicine and diabetes mellitus treatment, with a specific emphasis on wound healing in diabetic populations. Beginning with an insightful introduction, the article underscores the critical importance of effective wound healing within the broader context of diabetes mellitus, while tracing the evolutionary trajectory of precision medicine in healthcare. By elucidating the pathophysiological intricacies of diabetic wound healing, the review unveils the complex molecular mechanisms that drive this multifaceted process. Subsequently, a meticulous exploration follows into the application of precision medicine paradigms in diabetic wound care, delineating fundamental principles and diverse avenues through which precision medicine strategies can optimize diabetes management. Through a nuanced discussion of targeted therapies and interventions, the review highlights burgeoning approaches tailored to individual patient needs, accentuating the transformative potential of precision medicine in reshaping treatment paradigms. Drawing upon clinical trials and compelling case studies, the article offers valuable insights into the real-world efficacy of precision treatment modalities, elucidating successful applications and their profound implications for diabetic wound healing outcomes. Moreover, the review anticipates and addresses emerging challenges and future trajectories within the field, including the pivotal roles of biomarkers and diagnostic modalities, the integration of telemedicine platforms, and the increasing influence of artificial intelligence on diabetic wound healing endeavours. By synthesizing contemporary knowledge and delineating prospective pathways, this review underscores the catalytic potential of precision medicine in heralding a new era of enhanced outcomes for diabetic patients grappling with impaired wound healing.
    Keywords:  Artificial intelligence; biomarkers; diabetes mellitus; precision medicine; targeted therapy.; wound healing
    DOI:  https://doi.org/10.2174/0127722708335238240920035556
  22. Curr Opin Otolaryngol Head Neck Surg. 2025 Dec 12.
       PURPOSE OF REVIEW: In this review, we summarize the current state of medical treatment for cystic fibrosis (CF) patients and how cystic fibrosis transmembrane conductance regulator (CFTR) modulator therapy has revolutionalized the landscape of CF management, including from a sinonasal perspective. We describe the indications for endoscopic sinus surgery (ESS), perioperative decisions otolaryngologists must consider, and the most effective surgical treatment approaches.
    RECENT FINDINGS: Effective CFTR modulator therapy reduces SNOT-22 scores as well as endoscopic and radiographic scores. In this era, surgical indications for people with CF and chronic rhinosinsusitis (CRS) includes persistent, recalcitrant symptoms despite medical interventions, including CFTR therapy. The decision to pursue surgery should incorporate sinus symptoms, as ESS solely for pulmonary function improvement remains controversial. When ESS is performed, extended approaches to the sinuses may be beneficial, although evidence in this area is limited.
    SUMMARY: Despite prominent advances in disease modifying therapies, ESS remains a treatment option to manage refractory sinonasal symptoms and CRS in people with CF who do not respond to medical therapy. The decision to pursue ESS remains individualized and should involve a multidisciplinary discussion between clinicians to optimize patient selection, surgical goals, and perioperative medical management.
    Keywords:  chronic rhinosinusitis; cystic fibrosis; endoscopic sinus surgery
    DOI:  https://doi.org/10.1097/MOO.0000000000001103
  23. J Tissue Viability. 2025 Dec 16. pii: S0965-206X(25)00131-7. [Epub ahead of print]35(1): 100982
      Negative pressure wound therapy (NPWT) has demonstrated substantial advantages in a variety of clinical settings, including the management of acute and chronic wounds, open injuries with exposed bone, nerves, or tendons, and implant-associated infections in orthopedics. Although numerous studies have reported the beneficial effects of NPWT in controlling wound infections and infections related to orthopedic implants, the underlying molecular and cellular mechanisms remain incompletely understood and require further investigation. Given its widespread clinical use, a comprehensive understanding of the biological effects of NPWT is essential to ensuring its rational and optimized application in clinical practice. This review summarizes the potential mechanisms through which NPWT facilitates wound healing and provides insights to inform both clinical use and future research.
    Keywords:  Healing; Negative pressure wound therapy; Wound
    DOI:  https://doi.org/10.1016/j.jtv.2025.100982
  24. J Comp Eff Res. 2026 Jan;15(1): e250196
      In this update, we explore a review on transportability methods to enable the use of cross-jurisdictional evidence when local data are limited, a review of clinical trials that use pragmatic elements and finally, we discuss a study highlighting the potential transformative role of large language models in disease progression modeling.
    Keywords:  artificial intelligence; clinical trials; health technology assessment; large language models; pragmatic trial design; real-world data; real-world evidence; transportability
    DOI:  https://doi.org/10.57264/cer-2025-0196
  25. Small. 2025 Dec 15. e11542
      Topical chronic wound dressings offer personalized management but have limited efficacy in sensing and delivering therapeutics due to their reliance on the wound surface. Herein, a theranostic hydrogel-forming microneedles (HFMNs) dressing system is developed that penetrates the epidermis to continuously sample dermal wound interstitial fluid (ISF), providing high-fidelity diagnostics and active therapy. The dressing is fabricated from a polyvinyl alcohol/chitosan hydrogel incorporating MXene for intrinsic antibacterial and pro-healing properties. Central to the platform's design is a laser-scribed phase separation process that converts a poly(3,4-ethylenedioxythiophene): polystyrene sulfate/graphene oxide coating into highly conductive (384 S/m) and water-stable electrodes directly on the HFMNs. This enables a multiplexed, replaceable array for continuously monitoring seven key wound biomarkers: glucose, uric acid, Na⁺, K⁺, Cl-, pH, and temperature. In vitro studies confirmed the dressing's cytocompatibility and antimicrobial efficacy against E. coli and S. aureus, while in vivo rat models showed accelerated wound closure. Critically, the HFMN system captured the biochemical dynamics of wound infection and healing from wound ISF with significantly greater fidelity and more distinct responses compared to conventional surface sensors sampling exudate. This work validates a robust platform that directly links deep-tissue biomarkers to wound state, paving the way for personalized, proactive chronic wound management.
    Keywords:  chronic wounds; conductive hydrogel‐forming microneedles array; correlation between wound exudate and interstitial fluid; diagnostics and therapy; laser‐scribed phase separation; transdermal dressing
    DOI:  https://doi.org/10.1002/smll.202511542
  26. J Gerontol Nurs. 2025 Dec 18. 1-5
       PURPOSE: To describe older adults' lived experience with a chronic wound in a long-term care (LTC) facility, with the ultimate goal of enhancing well-being and overall quality of life (QoL).
    METHOD: A descriptive phenomenological design guided by Colaizzi's method was used. Seven cognitively intact LTC residents with chronic wounds were interviewed using a semi-structured guide. Interviews were analyzed inductively through multiple coding cycles, consensus-building, and member checking.
    RESULTS: Four interrelated themes were constructed: Lack of Independence, Confinement and Limitations, Interruptions to Daily Life, and Emotional Sequelae. Participants described physical and emotional burdens, a reliance on staff, and disruptions to routines and relationships. Despite challenges, most expressed appreciation for the wound care received in the facility.
    CONCLUSION: Chronic wounds deeply affect quality of life and identity. Clinicians should recognize the emotional and social dimensions of wound care. Results can help guide clinicians in LTC facilities to improve residents' wound healing and promote overall QoL.
    DOI:  https://doi.org/10.3928/00989134-20251211-01
  27. J Infect Dis. 2025 Dec 17. pii: jiaf626. [Epub ahead of print]
      Cystic fibrosis (CF) lung disease is a result of defective CFTR-mediated ion transport, producing dehydrated mucus, impaired mucociliary clearance and an opportune environment for chronic airway infection. CF airway infections are polymicrobial airway ecosystems often dominated by CF pathogens such as Pseudomonas aeruginosa, Staphylococcus aureus, Burkholderia, Stenotrophomonas, Achromobacter, and nontuberculous mycobacteria that drive cycles of infection, inflammation, and bronchiectasis. Highly effective CFTR modulators, including elexacaftor/tezacaftor/ivacaftor, improve airway hydration and mucociliary clearance and reduce pathogen CF acquisition and density. However, even with CFTR modulator treatment, most individuals with established infection remain chronically infected, and long-term impacts of CFTR modulators on airway infection dynamics and associated clinical outcomes remain unclear. In this review, we address key gaps in understanding chronic infection in the CFTR modulator era, including changes in infection-related lung disease pathogenesis, airway-gut microbiome interactions, approaches to airway infection sampling, and implications for infection management.
    Keywords:  CFTR modulators; Cystic fibrosis; airway microbiome; chronic airway infection; gastrointestinal microbiome
    DOI:  https://doi.org/10.1093/infdis/jiaf626
  28. Pol J Vet Sci. 2025 ;28(4): 691-700
      The intestinal microbiome is essential for the proper functioning of the immune system and the course of metabolic processes in the living organism. Intestinal bacteria produce a variety of metabolites that affect the health of many organs, especially the intestines. Disturbances in the composition of the intestinal microflora are referred to as dysbiosis. Dysbiosis occurring in chronic enteropathies may exacerbate intestinal inflammation. Therefore, effective methods of treating enteropathy are still being sought, which involve restoring the proper composition of the intestinal microbiome. In recent years, many scientific centers have drawn attention to the possibility of treating enteropathy by transplanting intestinal contents from a healthy donor. This review presents the advantages and disadvantages of this therapeutic method, described in the latest available literature and the newest guidelines regarding the donor and transplant administration methods.
    Keywords:  cats; dogs; intestinal microbiome; intestinal microbiome transplantation
    DOI:  https://doi.org/10.24425/pjvs.2025.157285