bims-fagtap Biomed News
on Phage therapies and applications
Issue of 2025–11–09
27 papers selected by
Luca Bolliger, lxBio



  1. Int J Antimicrob Agents. 2025 Nov 04. pii: S0924-8579(25)00215-8. [Epub ahead of print] 107660
      Antibiotic resistance is a growing global health crisis, driving the need for alternative antimicrobial strategies. Phage therapy has re-emerged as a promising approach due to its specificity and ability to evolve against bacterial resistance mechanisms. However, therapeutic implementation is often challenged by factors such as phage stability, immune clearance, and site-specific delivery. Encapsulation technologies offer a potential solution, enhancing phage bioavailability, systemic circulation, and therapeutic action at the infection site. This review explores the latest advancements in phage encapsulation within the context of nanomedicine-based delivery systems, including polymeric nanoparticles, liposomes, hydrogels, nanofibers, and inorganic nanoparticles. Each system is analyzed regarding its impact on phage stability, pharmacokinetics, and host-pathogen interactions, with a particular focus on applications in clinical and preclinical models of phage therapy. Liposomal and polymeric nanoparticle encapsulation enhance phage persistence in systemic circulation and protect them from degradation. Hydrogels and nanofibers improve localized phage delivery for wound healing applications, while inorganic nanoparticles and stimuli-responsive nanocarriers facilitate targeted delivery for intracellular and respiratory infections. By leveraging nanotechnology-driven encapsulation, phage therapy can overcome key delivery challenges, expanding its potential for treating multidrug-resistant bacterial infections. This approach opens new possibilities for precision antimicrobial therapies, reinforcing phage therapy as a viable alternative to conventional antibiotics.
    Keywords:  bacteriophages; biological barriers; drug delivery; nanomedicine
    DOI:  https://doi.org/10.1016/j.ijantimicag.2025.107660
  2. Front Cell Infect Microbiol. 2025 ;15 1690404
      A synthetic cell is a membrane-bound vesicle that encapsulates cell-free transcription/translation (TXTL) systems. It represents a transformative platform for advancing bacteriophage therapy. Building on experimental work that demonstrates (i) modular genome assembly, (ii) high-yield phage TXTL systems, and (iii) smart hydrogel encapsulation, we explore how synthetic cells can address major limitations in phage therapy. The promising advances include point-of-care phage manufacturing, logic-responsive antimicrobial biomaterials, and new chassis to dissect the dynamics of phage-host interactions. We also propose a roadmap for the deployment of synthetic cells as programmable and evolvable tools in the context of laboratory research and translational clinical adoption.
    Keywords:  bacteriophages; cell-free protein synthesis; hydrogel; phage therapy; synthetic cells; transcription/translation (TXTL)
    DOI:  https://doi.org/10.3389/fcimb.2025.1690404
  3. J Infect Dev Ctries. 2025 Oct 31. 19(10): 1495-1502
       INTRODUCTION: Phage therapy is a promising alternative for combating multidrug-resistant bacteria, including Acinetobacter baumannii (AB). However, the development of phage-resistant variants after treatment, particularly when using phage cocktails, poses a significant challenge. This resistance can hinder the effectiveness of future phage-based treatments against pathogenic AB.
    METHODOLOGY: Three AB-specific phages-AB-phage 22, AB-phage 27, and AB-phage 32-susceptible to an AB isolate, designated ABU-3, were used as a model to study phage resistance development in AB following phage treatment. This study proposes a strategy to effectively eliminate pathogenic AB using an optimal multiplicity of infection (MOI), referred to as the MOI clearance value.
    RESULTS: The MOI clearance values required for complete elimination of ABU-3 were determined to be 10 for AB-phages 22 and 32 and 100 for AB-phage 27. Surviving ABU-3 colonies from lower MOI treatments were analyzed for phage resistance. ABU-3 treated with AB-phage 27 developed resistance to AB-phage 27 but remained susceptible to AB-phages 22 and 32. ABU-3 treated with AB-phage 22 developed resistance to AB-phage 22 but retained partial susceptibility to the other phages at reduced MOI. In contrast, ABU-3 treated with AB-phage 32 displayed complete resistance to all three phages.
    CONCLUSIONS: These findings highlight a key challenge in phage therapy: insufficient MOI ratio can promote phage resistance. The distinct resistance profiles observed emphasize the importance of optimizing phage combinations and dosages to prevent resistance development during treatment.
    Keywords:  Acinetobacter baumannii; cocktail phage therapy; multiplicity of infection; phage therapy; phage-resistance variants
    DOI:  https://doi.org/10.3855/jidc.21384
  4. Cancer Pathog Ther. 2025 Nov;3(6): 453-465
      The human oral cavity harbors a diverse and dynamic microbial ecosystem, including bacteriophages (phages), which play a critical role in shaping the microbial community structure. Bacteriophages, viruses that specifically target and infect bacteria, have been increasingly recognized for their potential to influence both microbial balance and disease progression within the oral environment. Recent studies suggest that bacteriophages not only modulate the composition of the oral microbiome but also play an essential role in the pathogenesis and treatment of oral cancer. This review aims to explore the complex crosstalk between bacteriophages and oral health related to oral carcinogenesis, with a particular focus on their emerging roles in oral carcinogenesis and therapeutic interventions. Oral cancer, a major global health concern, is often associated with microbial dysbiosis and chronic inflammation, both of which contribute to tumor progression. Fusobacterium nucleatum, a key bacterial species implicated in oral carcinogenesis, has been shown to promote tumor growth, enhance immune evasion, and exacerbate inflammation within the tumor microenvironment. Bacteriophages offer a promising strategy to selectively target and eliminate such pathogenic bacteria such as Fusobacterium nucleatum, thereby restoring microbial balance and reducing the pro-tumorigenic effects of bacterial infections. Through the disruption of tumor-associated biofilms and modulation of cancer-promoting bacterial populations, phages may help mitigate the inflammatory responses that drive oral cancer progression. Additionally, phage therapy could complement existing treatments by sensitizing cancer cells to chemotherapy and immunotherapy. Beyond their direct antibacterial effects, genetically engineered bacteriophages present novel opportunities for targeted cancer therapy. Advances in synthetic biology have enabled the development of phages capable of delivering therapeutic payloads, such as anti-cancer peptides, cytotoxic agents, and immune modulators. These engineered phages can be designed to selectively target bacterial species that influence tumor progression, offering a highly specific and precision-based approach to oncology. Moreover, phages can serve as vectors for cancer vaccines, facilitating antigen presentation and enhancing immune responses against tumor cells. In addition to therapeutic applications, bacteriophages hold promise in the field of cancer diagnostics. The ability of phages to selectively bind to specific bacterial biomarkers associated with oral cancer could be leveraged for early disease detection and non-invasive screening. Phage-based biosensors, for instance, have shown potential in identifying cancer-associated microbial signatures, paving the way for innovative diagnostic tools that could improve early intervention and patient outcomes. Despite their potential, several challenges must be addressed before phage-based strategies can be fully integrated into clinical practice. Phage resistance, immune system clearance, and regulatory barriers pose significant hurdles to the widespread adoption of phage therapy. Additionally, the complex interactions between bacteriophages, host immunity, and the tumor microenvironment require further investigation to optimize therapeutic efficacy and safety. Advancing research on bacteriophages in the context of oral cancer could revolutionize current treatment paradigms, providing more targeted, efficient, and patient-friendly alternatives to conventional therapies. By harnessing the natural specificity and adaptability of bacteriophages, clinicians and researchers can develop innovative approaches to combat oral cancer, ultimately improving patient care and clinical outcomes.
    Keywords:  Bacteriophages; Biofilms; Microbiota; Mouth neoplasms; Oncolytic Virotherapy; Tumor microenvironment
    DOI:  https://doi.org/10.1016/j.cpt.2025.03.003
  5. Wounds. 2025 Sep;37(9): 363-372
       BACKGROUND: Chronic wounds, which exhibit prolonged inflammation, impaired healing, and vulnerability to infections, remain a global health challenge, largely driven by the persistence of microbial biofilms and escalating antimicrobial resistance (AMR). Biofilm protects pathogens from the host's immune defenses and conventional antibiotic treatments, sustaining wound chronicity and fostering resistance. Due to the inefficacy of traditional antibiotics in penetrating biofilms and mitigating resistant strains, alternative therapeutic strategies are urgently required.
    OBJECTIVE: To review the literature on the use of phytocompounds in chronic wound care.
    METHODS: Various databases such as PubMed, Google Scholar, and Web of Science were thoroughly surveyed by using keywords "phytocompounds, chronic wounds, wound healing, antimicrobial activity, antibiofilm activity and phytotherapy."
    RESULTS: Findings indicate that the compounds, such as flavonoids, terpenoids, and alkaloids, target various biological pathways, disrupt quorum sensing, and suppress virulence factors, making them valuable for disrupting biofilms and managing AMR. Phytocompounds such as coumarin, tannic acid, resveratrol, and berberine have the potential to enhance wound healing by reducing oxidative stress, promoting clotting, stimulating collagen synthesis, and combating infection. This review highlights complications associated with chronic wounds and explores beneficial phytocompounds for their management.
    CONCLUSION: Phytotherapy may play a role in managing wounds by promoting healing, preventing infection, and reducing the need for resistant antibiotics. Combining natural agents with traditional treatments presents a novel and integrative pathway to overcome the limitations posed by antibiotic resistance and biofilm-associated chronicity. While current findings are promising, further validation is needed to fully establish their clinical applications.
  6. J Glob Antimicrob Resist. 2025 Oct 31. pii: S2213-7165(25)00231-0. [Epub ahead of print]
       BACKGROUND: Multidrug resistance Pseudomonas aeruginosa is a global public health issue, and bacteriophages are the effective alternative therapy over antibiotics to combat antibiotic resistance. This study was designed to isolate bacteriophages and determine their antimicrobial potential against P. aeruginosa isolated from burn wounds and the environment of the burn ward.
    METHODS: A total of 100 random swab samples were collected from the burn ward, including 50 from the burn wounds and 50 from the environment and proceeded for further investigations at the University of Agriculture Faisalabad. P. aeruginosa was confirmed by genomic detection of OprL gene. Bacteriophages were isolated from sewage water through the agar overlay method, and the lytic spectrum was determined. Genome sequencing and analysis was performed. Phage stability and synergism between phage and antibiotics were analyzed.
    RESULTS: Overall, 50 samples were positive for P. aeruginosa, in which 35/50 were positive from wounds of burn patients and 15/50 were positive from the hospital environment. OprL-positive strains were preceded for bacteriophage isolation. Two phages, PhPa-6 and PhPa-4, with a lytic spectrum of 88% and 60%, respectively, were selected. The genome sizes of the PhPa-6 and PhPa-4 were 186.978 kb and 73.3 kb, respectively. A cocktail of these phages has shown an active in vitro synergistic effect with the ciprofloxacin (0.125 µg/ml).
    CONCLUSION: Cocktail of PhPa-6 and PhPa-4 with ciprofloxacin displayed significant lytic activity under in vitro conditions and a broad host range against P. aeruginosa isolated from burn patients.
    Keywords:  Burn injuries; Ciprofloxacin; Lytic bacteriophage; P. aeruginosa; Synergism
    DOI:  https://doi.org/10.1016/j.jgar.2025.10.012
  7. Adv Colloid Interface Sci. 2025 Oct 27. pii: S0001-8686(25)00304-5. [Epub ahead of print]347 103693
      Bacteriophage (phage) colloidal bionanoparticles garnered significant interest in colloid and interface science for their unique structural, interfacial, and self-assembly properties, as well as their potential for biomedical applications. The global rise of drug-resistant bacteria presented an urgent global health challenge, contributing to millions of deaths annually. Phages offered a promising alternative to antibiotics due to their high specificity and minimal side effects, and they also found diverse applications in environmental disinfection, food safety, and biomaterial engineering. However, large-scale phage manufacture remained a considerable challenge due to the complexity of production pipelines and the need to control contaminants that could compromise both safety and efficacy. In this review, we provided a comprehensive overview of phage acquisition, identification, and propagation methods, followed by an in-depth examination of advanced approaches for phage concentration and purification. We particularly emphasized how colloidal and interfacial phenomena could be leveraged to optimize the stability and functionality of phage colloidal bionanoparticles. We further presented an integrated framework for enhancing phage purification processes to ensure high bioactivity and broad applicability in clinical and industrial contexts. Lastly, we highlighted the need for tailoring propagation and purification protocols to specific phages to meet increasing demands for tissue regeneration, disease therapy, and beyond. By addressing these interdisciplinary challenges, this review underscored the substantial promise of phage colloidal bionanoparticles in generating significant economic and societal benefits.
    Keywords:  Drug-resistance; Phage biomaterials; Phage purification; Phages colloidal bionanoparticles
    DOI:  https://doi.org/10.1016/j.cis.2025.103693
  8. Microbiol Spectr. 2025 Nov 03. e0227725
      Infections with nontuberculous mycobacterium (NTM) strains are frequently nonresponsive to antibiotic regimens, even after multiple drugs are used for extensive periods of months or years. Therapy with bacteriophages specific for NTM clinical isolates shows considerable promise in compassionate-use single-patient cases but, like antibiotics, must be administered over long periods of time and typically intravenously. It is thus important that the phage preparations are highly purified and stable to avoid adverse reactions or decay of viability. Here, we describe methods for purification of mycobacteriophages including CsCl equilibrium gradient ultracentrifugation, desalting by either dialysis or column chromatography, and stabilization for storage by lyophilization. These phage preparations have low levels of contaminating Mycobacterium smegmatis host proteins and host genomic DNA, and the lack of host DNA strongly indicates that these phages are not competent for generalized transduction. Desalting by column chromatography is both rapid and efficient for removal of CsCl after equilibrium density ultracentrifugation, and the preparations maintain viability well in lyophilization using trehalose as an excipient. Once lyophilized, the phage preparations typically maintain viability over extended periods of time, although once reconstituted at working dose concentrations and distributed into syringes, they experience about a 10-fold loss in viability over a 2-week period.IMPORTANCEBacteriophages specific for Mycobacterium hosts show promise as potential therapeutic agents for controlling nontuberculosis Mycobacterium infections. Phage administration for compassionate-use cases frequently involves intravenous twice-daily doses, but over a period of many months or years; the biosafety profile is, therefore, of substantial importance. Here, we describe the detailed methods we have used to grow mycobacteriophages to high titers, to concentrate and purify them so that they are devoid of major contaminants, and methods for stable long-term storage.
    Keywords:  Mycobacterium; bacteriophages; phage preparation
    DOI:  https://doi.org/10.1128/spectrum.02277-25
  9. Discov Nano. 2025 Nov 07. 20(1): 201
      Wound healing is a complex biological process involving multiple stages that require interactions between several cells, growth factors, and cytokines. The lack of a suitable environment, along with microbial infection, impairs the normal wound-healing process. Burns, trauma, diabetic injuries, and chronic ulcers are the most prevalent types of wounds on the skin. Conventional wound treatments, with their focus on a single aspect, often fail to consider the complex conditions fully surrounding the pathogens that cause infections. Nanotechnology, a rapidly developing and cutting-edge field, facilitates tissue engineering and drug delivery and has proven effective in tackling several wound healing challenges by delivering anti-inflammatory, antibacterial, and angiogenic benefits. This review explores the wound healing process, chronic wounds, and current wound care challenges, highlighting how nanotechnology, through the use of metallic nanoparticles (such as silver, gold, and zinc oxide), polymeric systems, lipid-based carriers, and carbon-based materials, offers significant advantages and has become a crucial part of modern wound treatment. It further explores the role of nanotechnology in skin wound repair, its advantages over conventional treatment techniques, its biocompatibility, and it also highlights clinical trials using nanotech-based approaches and outlines future directions in wound healing research.
    Keywords:  Chronic wounds; Nanostructure; Nanotechnology; Tissue regeneration; Wound healing
    DOI:  https://doi.org/10.1186/s11671-025-04381-w
  10. BMC Microbiol. 2025 Nov 06. 25(1): 718
      Enterococcus faecium is a Gram-positive bacterium commonly found in the intestines of humans and animals. While certain strains exhibit probiotic properties, others have become significant pathogens due to their high level of antibiotic resistance. Bacteriophages, as natural bacterial predators, have emerged as promising alternatives for controlling bacterial infections, especially antibiotic-resistant strains. In this study, we isolated and characterized a novel bacteriophage, Enterococcus phage XWef1, which targets E. faecium, from waste materials of a feed additive production industry. The phage displayed optimal activity at pH levels between 6 and 8, with high stability at temperatures below 40 °C. It exhibited a narrow host range. Genomic analysis revealed 63 open reading frames (ORFs), including genes related to structural proteins, DNA replication, and host lysis. The comparative genomic analysis showed that XWef1 has less than 70% intergenomic similarities with its most similar phages. Thus, we propose that XWef1 is a novel Enterococcus phage belonging to a new genus. The discovery of XWef1 not only enriches our understanding of phage diversity but also offers new perspectives for phage research and development, with promising implications for microbial control and therapeutic applications.
    Keywords:  Biological features; Enterococcus faecium; New genus; Phage
    DOI:  https://doi.org/10.1186/s12866-025-04428-z
  11. Nat Rev Gastroenterol Hepatol. 2025 Nov 05.
      The gut virome is a complex ecosystem characterized by the interplay of diverse viral entities, predominantly bacteriophages and eukaryotic viruses. The gut virome has a critical role in human health by shaping microbial community profiles, modulating host immunity and influencing metabolic processes. Different viral metagenomics approaches have revealed the remarkable diversity of the gut virome, showing individual-specific patterns that evolve over time and adapt dynamically to environmental factors. Perturbations in this community are increasingly associated with chronic immune and inflammatory conditions, metabolic disorders and neurological conditions, highlighting its potential as a diagnostic biomarker and therapeutic target. The early-life gut virome is particularly influential in establishing lifelong health trajectories through its interactions with diet, immune pathways and others, thereby contributing to inflammatory and metabolic regulation. This Review synthesizes current knowledge of gut virome composition, dynamics and functional relevance, critically evaluating evidence distinguishing causal from correlative roles in disease pathogenesis. The interactions of the virome with other microbiome components and host immunity are examined, and emerging translational applications, including phage therapy and biomarker development, are discussed. Integrating these insights while acknowledging methodological challenges provides a comprehensive framework for understanding the complex roles of the gut virome in health and disease.
    DOI:  https://doi.org/10.1038/s41575-025-01134-z
  12. J Biomater Sci Polym Ed. 2025 Nov 02. 1-24
      Dressings are essential medical devices in the treatment of wounds. The great diversity of skin lesions has led to the development of different therapeutic approaches involved in optimizing re-epithelialization. Advances in the field's technologies have led to an increase in therapeutic patents with various applications, including complex lesions that are difficult to heal. This article presents an analysis of patents and scientific publications on the development of innovative dressings for the treatment of complex dermal lesions, using Derwent World Patents Index (DWPI) database. The study provides an overview of materials, drugs, and nanotechnologies used in dressings, as well as their applications. Finally, the patent analysis enabled a comprehensive examination of trends, places of protection, market dynamics, and technological domains, providing valuable information on the advances and therapeutic potential of dressings. The development of new technologies associated with wound dressings applied to the treatment of injuries demonstrates the expressive interest of the scientific community, indicating advances in the sector. The incorporation and combination of diverse technologies employed in currently developed and investigational dressings demonstrate the emergence of increasingly efficient products, enhancing accessibility to therapies and contributing to the population's quality of life.
    Keywords:  Cutaneous; complex wounds; dressings; healing; nanotechnology; skin lesions
    DOI:  https://doi.org/10.1080/09205063.2025.2580792
  13. RSC Chem Biol. 2025 Oct 27.
      Bacteriophages have emerged as important factors in human health and disease, with elevated phage levels associated with exacerbated inflammatory bowel disease, type 2 diabetes and poor outcomes in skin and lung infections. The mechanisms linking phages to these pathologies remain largely unknown, partly because specific chemical tools inhibiting bacteriophage replication (phage blockers) are lacking. Here, we identify benzimidazylpyrazoles as novel bacteriophage antivirals. Unlike existing synthetic antiphage compounds benzimidazylpyrazoles do not intercalate DNA and target an early stage of phage infection after adsorption. An optimized derivative reduced phage titer up to 105-fold and demonstrated activity against different phage morphotypes and bacterial hosts, establishing it as a valuable chemical tool for the study of disease-related phage-host interactions.
    DOI:  https://doi.org/10.1039/d5cb00120j
  14. Int Wound J. 2025 Nov;22(11): e70771
      This study aimed to gain clinician consensus on which signs/symptoms reported to be indicative of biofilm in chronic wounds are likely to be so. An international, two-round eDelphi process including wound care clinicians ran from December 2023 to February 2024. Participants rated 26 items on a 9-point Likert scale. Consensus to include: ≥ 70% of respondents rate an item 7-9, ≤ 15% rate it 1-3. Consensus to exclude: ≥ 70% of respondents rate an item 1-3, ≤ 15% rate it 7-9. Eleven items (visual indicators [a shiny, slimy, persistent layer, easily removed, returns quickly without frequent intervention]; failure to respond to antimicrobials; infection > 30 days duration; poor quality granulation tissue; stalled wound despite optimal management; persistent/prolonged inflammation; wound > 6 weeks duration; soft tissue deterioration despite antimicrobials/debridement; signs of local infection; tunnelling/undermining; presence of slough) achieved consensus to include status. To our knowledge, consensus work on this topic has not previously been performed on such a wide scale. When examined alongside similar work, clinical opinion on the matter lacks coherence. We hope that these findings will help direct us toward greater cohesiveness. The work supports a need for research to quantify the predictive abilities of signs and symptoms reported to be indicative of biofilm in chronic wounds.
    Keywords:  biofilms; chronic wounds; consensus; electronic Delphi; signs and symptoms
    DOI:  https://doi.org/10.1111/iwj.70771
  15. Clin Microbiol Rev. 2025 Nov 04. e0012125
      SUMMARYCoagulase-negative staphylococci (CoNS) are a group of bacteria commonly found on human skin and mucous membranes. Traditionally regarded as low-virulence microorganisms, they have gained recognition as significant pathogens in healthcare-associated infections, particularly among immunocompromised individuals and patients with indwelling medical devices. In diabetic foot infections (DFIs), CoNS can play a substantial role, particularly following previous antibiotic treatments or in the presence of indwelling devices. DFIs are usually polymicrobial, involving a mixture of aerobic and anaerobic bacteria. Although Staphylococcus aureus is recognized as the major pathogen, especially in Western countries, CoNS are increasingly emerging as significant pathogens in DFIs, including osteomyelitis. Their presence may complicate treatment by increasing the microbial burden and harboring antibiotic resistance mechanisms. The treatment of DFIs involving CoNS often requires a combination of antibiotics targeting both gram-positive and gram-negative bacteria, selected according to the severity of the infection and results of antibiotic susceptibility testing. This review aims to highlight the growing importance of CoNS in DFIs, discussing their pathogenic mechanisms, clinical implications, and the necessity for healthcare providers to consider their involvement in order to ensure effective treatment and successful patient outcomes. This narrative review aims to underscore the growing clinical relevance of CoNS in DFIs by exploring their pathogenic mechanisms, diagnostic challenges, and therapeutic implications. It emphasizes the need for clinicians to recognize CoNS as potential pathogens rather than mere contaminants and highlights the ongoing difficulty in distinguishing true infection from colonization. This distinction is critical for accurate diagnosis, appropriate antimicrobial stewardship, and the development of improved treatment strategies.
    Keywords:  coagulase-negative staphylococci; diabetic foot infections; diversity; epidemiology; management; pathophysiology; resistance; virulence
    DOI:  https://doi.org/10.1128/cmr.00121-25
  16. Ann Med Surg (Lond). 2025 Oct;87(10): 6868-6869
      Autoimmune hepatitis (AIH) is a chronic inflammatory liver disease with unclear etiology but likely results from a complex interplay of genetic susceptibility and environmental triggers. Emerging evidence highlights the role of gut microbiota in AIH pathogenesis, with specific genera such as Veillonella, Lactobacillus, and Oscillospira demonstrating diagnostic value. Dysbiosis-associated biomarkers like lipopolysaccharide and aspartate aminotransferase further support a microbial role in disease onset and progression. Despite these promising developments, rare AIH variants remain poorly characterized due to methodological and population-level limitations. Moving forward, large-scale, longitudinal studies integrating metagenomics, metabolomics, and host genomic data are needed to establish subtype-specific microbial markers and assess the efficacy of targeted interventions such as probiotics and bacteriophage therapy.
    Keywords:  autoimmune hepatitis; dysbiosis; gut microbiota; microbial biomarkers
    DOI:  https://doi.org/10.1097/MS9.0000000000003709
  17. Expert Opin Drug Deliv. 2025 Nov 07.
       INTRODUCTION: Cystic fibrosis (CF) is a genetic disease caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene, leading to impaired ion transport by the CFTR protein and accumulation of thick, sticky mucus, resulting in chronic lung disease and other organ complications. Messenger ribonucleic acid (mRNA) therapies hold great potential for CF, as they can be used for both CFTR mRNA replacement and editing of the CFTR gene, which could mitigate the pathophysiological and clinical symptoms of CF.
    AREAS COVERED: We provide an overview of non-viral mRNA therapies for CF and their ongoing clinical trials. We begin with a discussion of the pathophysiological functions of CFTR and clinical symptoms of CF. We provide a summary of conventional treatments, modulator therapies, and potential gene therapies for CF. We discuss the pertinence of mRNA therapies for CF as well as the challenges associated with mRNA delivery. We conclude with an overview of non-viral vectors for mRNA delivery and a summary of clinical trials for CFTR mRNA therapies. Literature searches for this review were performed using databases (Google Scholar and PubMed).
    EXPERT OPINION: With continued innovation and overcoming of delivery challenges, mRNA therapies hold great potential for the treatment of all people with CF, regardless of mutation.
    Keywords:  Cystic fibrosis; gene therapy; mRNA lung delivery; non-viral delivery vectors; nucleic acid therapeutics
    DOI:  https://doi.org/10.1080/17425247.2025.2586172
  18. NPJ Antimicrob Resist. 2025 Nov 03. 3(1): 90
      Antibiotic resistance represents a growing global health crisis, diminishing the effectiveness of existing treatments and accelerating the emergence of multidrug-resistant bacterial strains. In this study, we present a mathematical framework for systematically characterizing data sets of collateral sensitivity patterns in evolving drug-resistant bacterial populations. This formalization is implemented in an open-source computational platform providing an intuitive and accessible in silico tool for data-driven antibiotic selection. By leveraging this approach, we can rapidly identify a therapeutic regimen that minimizes the risk of resistance evolution. The utility of this framework is demonstrated by highlighting the failure of antibiotic therapy in chronic Pseudomonas aeruginosa infections. Our approach offers a scalable strategy for navigating bacterial evolutionary landscapes and delineates key conditions under which sequential antibiotic therapies are prone to failure.
    DOI:  https://doi.org/10.1038/s44259-025-00160-w
  19. Microbiol Spectr. 2025 Nov 05. e0229924
    Amsterdam Mucociliary Clearance Disease Research Group
      Lung infections in people with cystic fibrosis (CF) cause lung damage, which is the leading factor in the morbidity and mortality of CF. Prescription of antibiotics to treat these infections is essential to maintain a higher quality of life and increase life expectancy. Determination of antibiotic susceptibility (ABS) is done by culture-dependent, phenotypic methods. These procedures take several days, while timely intervention is key. The analysis of antibiotic resistance genes by use of shotgun metagenomics might offer a time-sensitive alternative. Twenty people with CF with a homozygous Phe508del mutation provided 68 sputum samples during different visits over a period of roughly a year. After shotgun sequencing, the samples were analyzed using the deep learning tool deepARG. These results were compared with the results from routine ABS testing. The performance was determined by area under the curve-receiver operating characteristics (AUCROC) and sensitivity. Significant results were obtained for the following antibiotic classes: aminoglycoside (AUCROC = 0.81 [95% CI: 0.67-0.95, sensitivity = 73%]), cephalosporin (AUCROC = 0.70 [95% CI: 0.54-0.86, sensitivity = 95%]), and fluoroquinolone (AUCROC = 0.73 [95% CI: 0.56-0.89, sensitivity = 88%]). For other antibiotic classes, results were not significant. Using antibiotic class-specific cut-offs for positive reads of ARGs, a metagenomics approach potentially offers a culture-independent and more time-efficient manner to predict ABS for commonly prescribed antibiotic classes for sputum samples of adult people with CF. The use of metagenomics and artificial intelligence in clinical care will add to more personalized care for people with CF as well as better antibiotic stewardship.
    IMPORTANCE: Damage induced by lung infections in people with cystic fibrosis (CF) is the leading factor to the mortality and morbidity of CF. To treat bacterial infections, people with CF are prescribed antibiotics. Routine antibiotic susceptibility (ABS) testing relies on culture-dependent, phenotypic techniques. These take several days up to more than a week, while timely intervention is key. To bridge this time gap, physicians in CF care use patient history of ABS data to start antibiotics, with risk of resistance to it. This pilot study explores a time saving alternative: the possibility to predict antibiotic resistance genes using shotgun metagenomics and artificial intelligence. By quicker prediction of ABS, people with CF can receive more adequate care, which results in the possible prevention of chronic infections and contributes to antibiotic stewardship.
    Keywords:  antibiotic resistance; antibiotic susceptibility testing; artificial intelligence; cystic fibrosis; metagenomics; next-generation sequencing
    DOI:  https://doi.org/10.1128/spectrum.02299-24
  20. Br J Nurs. 2025 Nov 06. 34(20): S4-S10
      Wound care presents a significant, yet often under-prioritised, challenge within the UK healthcare system. Despite affecting millions of patients and costing the NHS an estimated £8.3 billion annually (based on data primarily from England), wound management remains highly variable across regions, specialties, and care settings throughout the UK. This article explores the impact of inconsistent clinical practice, the economic implications of chronic and non-healing wounds, and the inequities experienced by patients, especially those with complex care needs. It also highlights the absence of a UK wound care database, which would enable the systemic and standardised monitoring of patient numbers, wound type prevalence, healing rates, and complications. A national wound care database would support quality assurance through the monitoring of outcomes, allow for benchmarking among healthcare providers as well as supporting the provision of robust data for research. Furthermore, it would strengthen healthcare policy, guideline development and support more efficient allocation of resources and commissioning across the four nations of the UK. This lack of coordinated data hinders meaningful research and service planning, quality improvement, equitable evidence-based care, and the opportunity to inform appropriate commissioning and targeted investment. Key strategies are proposed including the adoption of standardised pathways, investment in education, and the development of a UK-wide data infrastructure to support a more coordinated, evidence-based, and cost-effective approach to wound care in the UK.
    Keywords:  Cost-benefit; Evidence-based nursing; Health inequalities; Outcomes; Policy
    DOI:  https://doi.org/10.12968/bjon.2025.0428
  21. Front Immunol. 2025 ;16 1681037
      Periodontitis (PD) is a chronic inflammatory disease linked to microbial dysbiosis, while rheumatoid arthritis (RA) is an autoimmune disorder characterized by anti-citrullinated protein antibodies (ACPA). Despite their distinct etiologies, a clinical and serological association between PD and RA has been observed. Oral microorganisms, especially Porphyromonas gingivalis (P. gingivalis), may contribute to RA onset or progression through dissemination to joints or systemic inflammation. This review explores a: the role of oral microbiota and immune responses in RA b; clinical pathogenic pathways from oral pathogens to the joints c; mechanistic studies on the impact of periodontal pathogens on RA; and d. preventive and therapeutic strategies. P. gingivalis and other periodontal pathogens have been detected in synovial tissues and fluids of RA patients. Microbiome analyses show a more diverse oral microbiota with elevated periodontal disease-associated bacteria in RA patients. Studies demonstrate that P. gingivalis can induce citrullination, autoantibody production, and inflammation, exacerbating joint damage. Future research should investigate the impact of periodontal therapy and RA treatments on the oral microbiota, while large-scale clinical trials are needed to validate the causal relationship between periodontal pathogens and RA.
    Keywords:  citrullinated antigens; oral pathogens; periodontitis; porphyromonas gingivalis; rheumatoid arthritis
    DOI:  https://doi.org/10.3389/fimmu.2025.1681037
  22. Front Bioeng Biotechnol. 2025 ;13 1678136
      The global rise of antimicrobial resistance has driven the search for novel antimicrobial strategies with higher effectiveness than common antibiotics. Among various solutions, polydopamine nanoparticles (PDA NPs) have gained widespread attention owing to their biocompatibility, functional versatility, and responsiveness to environmental stimuli. This review summarizes recent advances in the synthesis, functionalization, and antimicrobial applications of PDA NPs, highlighting their potential as smart nanomaterials. PDA NPs exhibit intrinsic antimicrobial activity, large drug delivery capabilities, and excellent photothermal properties. Moreover, they can potentially eradicate biofilms; can be synergistically combined with other entities such as metal ions, antimicrobial peptides, and Fenton-like catalysts; and can provide in vivo models of bacterial infection. Despite these advantages, the widespread use of PDA NPs is limited by low synthesis reproducibility, insufficient accurate characterization, and lack of comprehensive biocompatibility assessment. Resolving these challenges is essential for fully comprehending and using the potential of PDA-based antimicrobial platforms. This review aims to explain the current landscape of PDA-based nanoformulations and to inspire future research toward clinically viable PDA-based nanoformulations.
    Keywords:  antimicrobial resistance; biofilm eradication; photothermal therapy; polydopamine nanoparticles; smart nanomaterials
    DOI:  https://doi.org/10.3389/fbioe.2025.1678136
  23. Ther Adv Infect Dis. 2025 Jan-Dec;12:12 20499361251388382
      Pseudomonas aeruginosa is one of the most clinically important Gram-negative bacteria and is related to many severe and life-threatening infections worldwide. It presents intrinsic resistance against many antibiotics and has the ability to acquire or develop additional mechanisms to overcome the action of all anti-pseudomonal drugs. Formerly abandoned antibiotics and new compounds such as cefiderocol and combinations of β-lactams with new β-lactamase inhibitors are considered for the treatment of infections due to multi- or extensively-drug-resistant strains. In the present review, the antimicrobial resistance mechanisms of P. aeruginosa and the potential treatment options for the difficult to treat P. aeruginosa infections are discussed in an attempt to correlate microbiological and laboratory data to the choice of optimal treatment in everyday clinical practice.
    Keywords:  MDR; PDR; Pseudomonas aeruginosa; XDR; antibiotic resistance
    DOI:  https://doi.org/10.1177/20499361251388382
  24. Infection. 2025 Nov 07.
      Chronic infections are a persistent global health problem and are frequently sustained by polymicrobial communities rather than by a single pathogen. This review brings together current evidence for the infectome concept, defined as the dynamic set of pathogenic or pathobiont taxa in the host, their shared functional capacities, and the interactions that connect them. We analyze how community-level processes promote persistence, cause diagnostic failure, and drive therapeutic resistance, with emphasis on multispecies biofilms, quorum sensing, horizontal gene transfer, metabolic cooperation, and immune modulation. We also highlight advances in multi-omics and computational integration that now permit high-resolution infectome profiling and reveal taxa and interspecies networks that are not captured by routine culture. Clinical examples such as periodontitis, bacterial vaginosis, chronic rhinosinusitis, device-associated infections, and recurrent urinary tract infections show the translational value of this shift. On the therapeutic side, we discuss infectome-informed options including antivirulence agents, biofilm-disrupting enzymes, bacteriophages and lysins, community-wide susceptibility-guided regimens, and microbiome-restoration strategies. Finally, we identify the main requirements for the field: standardized sampling and analytic workflows, reproducible infectome signatures linked to clinical outcomes, and trial designs able to capture ecological dynamics and meet regulatory expectations for community-targeted interventions. Adopting an infectome perspective can enable precision infectiology and reshape the management of chronic and recurrent infections.
    Keywords:  Biofilm; Infectome; Metagenomics; Polymicrobial infections; Precision infectiology
    DOI:  https://doi.org/10.1007/s15010-025-02687-6
  25. Ann Med Surg (Lond). 2025 Nov;87(11): 7753-7754
      Artificial intelligence (AI) is transforming drug discovery by enabling the rapid identification of existing drugs with potential anti-cancer properties, a process known as drug repurposing. This approach offers a cost-effective and time-efficient alternative to traditional drug development, particularly in oncology, where the need for novel therapies is urgent. AI-driven methods, including machine learning (ML) and deep learning (DL), analyze vast datasets to uncover hidden drug-disease relationships, predict drug efficacy, and optimize therapeutic combinations. This letter highlights the transformative potential of AI in drug repurposing for cancer therapies, supported by successful examples from recent research. We also discuss the challenges and ethical considerations of integrating AI into drug repurposing efforts, emphasizing the need for robust validation and regulatory oversight to ensure patient safety and efficacy.
    Keywords:  artificial intelligence; cancer research; clinical trials; data analysis; drug repurposing; patient recruitment
    DOI:  https://doi.org/10.1097/MS9.0000000000003893
  26. Adv Wound Care (New Rochelle). 2025 Nov 05.
      Significance: Wound healing in pediatric patients is affected by physiology, growth, and development considerations unique from those in adults. In the following report, we review the primary literature on aging and wound healing and highlight clinical wound healing applications for the pediatric patient across age ranges from neonates and infants in the first year of life to adolescents (aged 10-19 years by World Health Organization definition). Recent Advances: We characterize the differences in wound healing biology between infants, adolescents, and adults and discuss wound care strategies for pediatric surgical patients, highlighting evidence-based guidelines for wound management. We discuss relevant animal models and review the multidisciplinary aspects of providing wound care for children. Critical Issues: Pediatric surgical patients have specialized wound care needs. Optimizing wound care outcomes for infants, children, and adolescents relies on an understanding of their wound-healing biology and unique physiological, psychological, and social considerations. Future Directions: Future directions in pediatric wound care will focus on validating and optimizing emerging technologies through pediatric-specific clinical trials, while also addressing key knowledge gaps in topical agent pharmacokinetics and advancing regenerative approaches like mesenchymal stem cell therapies tailored to the unique biology of infants and children.
    Keywords:  fibrosis; pediatric; stoma care; wound care; wound healing
    DOI:  https://doi.org/10.1177/21621918251387640
  27. Front Cell Infect Microbiol. 2025 ;15 1668573
      Vulvovaginal candidiasis (VVC) is an infection caused by Candida albicans that presents an escalating threat to humans. Lactobacilli may play a critical role in maintaining microbiome balance in the gut and vagina as well as limiting fungal colonization, including C. albicans. Certain Lactobacilli, classified as nomadic groups is gaining immense popularity in antifungal defense due to its unique morphological adaptations. One significant adaptation is the V-shaped cell chaining observed under low pH conditions governed by the LuxS-mediated quorum-sensing system. This structural adaptation potentiates altered secondary metabolite secretion. These geometric forms are not solely survival responses but reflect a structurally coordinated strategy that enhances both antibiofilm and antihyphal activities. In this perspective, we argue that morphology-driven transitions identify nomadic Lactobacilli as a promising frontier in probiotic therapy. By shifting from conventional probiotic formulations to structured microbial interventions, we propose the development of novel sustainable therapeutics for anticandidal therapy.
    Keywords:  C. albicans; Lactobacilli; V-shaped structure; cellular heterogeneity; nomadic; probiotic biofilm
    DOI:  https://doi.org/10.3389/fcimb.2025.1668573