1,079 results on '"Bacteriophage therapy"'
Search Results
2. Alternative approaches for bovine mastitis treatment: A critical review of emerging strategies, their effectiveness and limitations
- Author
-
Debruyn, Ella, Ghumman, Nauman Zaheer, Peng, Jiaxin, Tiwari, Harish Kumar, and Gogoi-Tiwari, Jully
- Published
- 2025
- Full Text
- View/download PDF
3. Rising prevalence and drug resistance of Corynebacterium striatum in lower respiratory tract infections.
- Author
-
Li, Wei, Gao, Mingyue, and Yu, Jinyan
- Subjects
RESPIRATORY infections ,JOINT infections ,CHRONIC obstructive pulmonary disease ,MULTIDRUG resistance ,LUNG infections ,COVID-19 ,DRUG resistance - Abstract
Corynebacterium striatum (C. striatum) is a Gram-positive bacterium commonly colonizing the skin and mucosa in healthy individuals and hospitalized patients. Traditionally regarded as a contaminant, C. striatum is now increasingly recognized as a potential cause of clinical infections, especially after the coronavirus disease pandemic. It has emerged as a pathogen implicated in severe infections, including pneumonia, bacteremia, meningitis, artificial joint infections, abdominal infections, and endocarditis. C. striatum has been reported in lower respiratory tract infections, mostly as a conditioned pathogen in immunocompromised individuals, particularly in those with chronic structural lung diseases, such as chronic obstructive pulmonary disease, leading to severe pneumonia or exacerbation of the existing disease and high mortality. Additionally, C striatum has been implicated in the community-acquired pneumonia among immunocompetent individuals and nosocomial lung infections, with evidence of person-to-person transmission through caregivers. C. striatum may exhibit multidrug resistance. Vancomycin, alone or in combination, is currently considered the most effective treatment for C. striatum. This review highlights the epidemiological characteristics, drug resistance mechanisms, diagnostics approaches, and treatment options for C. striatum lower respiratory tract infections to enhance clinician awareness and improve patient management strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
4. Use of adjuvants to improve antibiotic efficacy and reduce the burden of antimicrobial resistance.
- Author
-
Gil-Gil, Teresa, Laborda, Pablo, Martínez, José Luis, and Hernando-Amado, Sara
- Abstract
Introduction: The increase in antibiotic resistance, together with the absence of novel antibiotics, makes mandatory the introduction of novel strategies to optimize the use of existing antibiotics. Among these strategies, the use of molecules that increase their activity looks promising. Areas covered: Different categories of adjuvants have been reviewed. Anti-resistance adjuvants increase the activity of antibiotics by inhibiting antibiotic resistance determinants. Anti-virulence approaches focus on the infection process itself; reducing virulence in combination with an antibiotic can improve therapeutic efficacy. Combination of phages with antibiotics can also be useful, since they present different mechanisms of action and targets. Finally, combining antibiotics with adjuvants in the same molecule may serve to improve antibiotics' efficacy and to overcome potential problems of differential pharmacokinetics/pharmacodynamics. Expert opinion: The successful combination of inhibitors of β-lactamases with β-lactams has shown that adjuvants can improve the efficacy of current antibiotics. In this sense, novel anti-resistance adjuvants able to inhibit efflux pumps are still needed, as well as anti-virulence compounds that improve the efficacy of antibiotics by interfering with the infection process. Although adjuvants may present different pharmacodynamics/pharmacokinetics than antibiotics, conjugates containing both compounds can solve this problem. Finally, already approved drugs can be a promising source of antibiotic adjuvants. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
5. Alhagi maurorum extract in combination with lytic phage cocktails: a promising therapeutic approach against biofilms of multi-drug resistant P. mirabilis.
- Author
-
Mirzaei, Arezoo, Esfahani, Bahram Nasr, Ghanadian, Mustafa, Wagemans, Jeroen, Lavigne, Rob, and Moghim, Sharareh
- Subjects
BACTERIAL adhesion ,QUORUM sensing ,SCANNING electron microscopy ,PLANT extracts ,DRUG resistance in bacteria ,BIOFILMS - Abstract
Antimicrobial resistance (AMR) poses a significant global threat to public health systems, rendering antibiotics ineffective in treating infectious diseases. Combined use of bio compounds, including bacteriophages and plant extracts, is an attractive approach to controlling antibiotic resistance. In this study, the combination of phage cocktail (Isf-Pm1 and Isf-Pm2) and Alhagi maurorum crude extract (AME) was investigated in controlling biofilm-forming multi-drug resistant P. mirabilis isolates, in vitro and a phantom bladder model. The combination of AME and phage cocktails demonstrated no significant disparity in its ability to inhibit quorum sensing (QS) when compared to the individual control of AME alone. Following treatment with the combination of phage cocktail and AME at a 125 μg/mL concentration, the MDR P. mirabilis biofilm biomass was notably reduced by 73% compared to the control (P< 0.0001). The anti-biofilm effect was confirmed by Scanning Electron Microscopy (SEM). Moreover, in a bladder phantom model, there was a considerable decrease in encrustation levels compared to the control. The combined treatment resulted in a 1.85 logarithmic reduction in bacterial adhesion to Vero cells compared to the control. The real-time PCR results indicated significant downregulation of QS- and adhesion-related gens. The phage therapy, combined with AME, holds promising potential in reducing biofilm formation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Targeted dual-receptor phage cocktail against Cronobacter sakazakii : insights into phage-host interactions and resistance mechanisms.
- Author
-
Kim, Seongok, Son, Bokyung, Kim, Yeran, Kim, Hyeongsoon, Nam, Gahyeon, Shin, Hakdong, and Ryu, Sangryeol
- Subjects
SOIL structure ,PHENOTYPIC plasticity ,PROTEIN receptors ,FOOD pathogens ,CRONOBACTER - Abstract
Introduction: Cronobacter sakazakii is a notorious foodborne pathogen, frequently contaminating powdered infant formula and causing life-threatening diseases in infants. The escalating emergence of antibiotics-resistant mutants has led to increased interest in using bacteriophage as an alternative antimicrobial agent. Methods: Two phages, CR8 and S13, were isolated from feces and soil samples and their morphology, physiology, and genomics were characterized. Phage receptor was determined using deletion mutants lacking flgK, rfaC, fhuA, btuB, lamb , or ompC genes, followed by complementation. Phage-resistant mutants were analyzed for phenotypic changes and fitness trade-offs using motility assays and Caco-2 cell invasion models. Results: CR8 and S13 were identified as members of Caudoviricetes. Phage CR8 and phage S13 utilize flagella and LPS, respectively, to adhere to host cells. Bacterial challenge assay demonstrated delayed emergence of the resistant mutant as well as stronger lytic activity of a phage cocktail consisting of CR8 and S13 than the single phage treatment. Phenotypic analysis of the phage cocktail resistant strain, designated as CSR strain, revealed that the resistance resulted from the impaired receptor proteins for phage, such as defects in motility and alteration in LPS structure. CSR strain exhibited significant attenuation in invading human intestinal epithelial Caco-2 cells compared to WT cells. Conclusion: This study demonstrates that the development of the phage cocktail targeting distinct host receptors can serve as a promising antimicrobial strategy to effectively control C. sakazakii. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Advancing Phage Therapy: A Comprehensive Review of the Safety, Efficacy, and Future Prospects for the Targeted Treatment of Bacterial Infections.
- Author
-
Palma, Marco and Qi, Bowen
- Subjects
- *
DRUG resistance in bacteria , *BACTERIAL diseases , *INDIVIDUALIZED medicine , *FOOD pathogens , *FOOD supply - Abstract
Background: Phage therapy, a treatment utilizing bacteriophages to combat bacterial infections, is gaining attention as a promising alternative to antibiotics, particularly for managing antibiotic-resistant bacteria. This study aims to provide a comprehensive review of phage therapy by examining its safety, efficacy, influencing factors, future prospects, and regulatory considerations. The study also seeks to identify strategies for optimizing its application and to propose a systematic framework for its clinical implementation. Methods: A comprehensive analysis of preclinical studies, clinical trials, and regulatory frameworks was undertaken to evaluate the therapeutic potential of phage therapy. This included an in-depth assessment of key factors influencing clinical outcomes, such as infection site, phage–host specificity, bacterial burden, and immune response. Additionally, innovative strategies—such as combination therapies, bioengineered phages, and phage cocktails—were explored to enhance efficacy. Critical considerations related to dosing, including inoculum size, multiplicity of infection, therapeutic windows, and personalized medicine approaches, were also examined to optimize treatment outcomes. Results: Phage therapy has demonstrated a favorable safety profile in both preclinical and clinical settings, with minimal adverse effects. Its ability to specifically target harmful bacteria while preserving beneficial microbiota underpins its efficacy in treating a range of infections. However, variable outcomes in some studies highlight the importance of addressing critical factors that influence therapeutic success. Innovative approaches, including combination therapies, bioengineered phages, expanded access to diverse phage banks, phage cocktails, and personalized medicine, hold significant promise for improving efficacy. Optimizing dosing strategies remains a key area for enhancement, with critical considerations including inoculum size, multiplicity of infection, phage kinetics, resistance potential, therapeutic windows, dosing frequency, and patient-specific factors. To support the clinical application of phage therapy, a streamlined four-step guideline has been developed, providing a systematic framework for effective treatment planning and implementation. Conclusion: Phage therapy offers a highly adaptable, targeted, and cost-effective approach to addressing antibiotic-resistant infections. While several critical factors must be thoroughly evaluated to optimize treatment efficacy, there remains significant potential for improvement through innovative strategies and refined methodologies. Although phage therapy has yet to achieve widespread approval in the U.S. and Europe, its accessibility through Expanded Access programs and FDA authorizations for food pathogen control underscores its promise. Established practices in countries such as Poland and Georgia further demonstrate its clinical feasibility. To enable broader adoption, regulatory harmonization and advancements in production, delivery, and quality control will be essential. Notably, the affordability and scalability of phage therapy position it as an especially valuable solution for developing regions grappling with escalating rates of antibiotic resistance. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. Prevention and Modern Strategies for Managing Methicillin-Resistant Staphylococcal Infections in Prosthetic Joint Infections (PJIs).
- Author
-
Kraus, Karolina, Mikziński, Paweł, Widelski, Jarosław, and Paluch, Emil
- Subjects
PROSTHESIS-related infections ,TOTAL knee replacement ,ARTHROPLASTY ,KNEE surgery ,STAPHYLOCOCCAL diseases - Abstract
Periprosthetic joint infections (PJIs) are a dangerous complication of joint replacement surgeries which have become much more common in recent years (mostly hip and knee replacement surgeries). Such a condition can lead to many health issues and often requires reoperation. Staphylococci is a bacterial group most common in terms of the pathogens causing PJIs. S. aureus and coagulase-negative staphylococci are found in around two-thirds of PJI cases. Recently, the numbers of staphylococci that cause such infections and that are methicillin-resistant are increasing. This trend leads to difficulties in the treatment and prevention of such infections. That is why MRSA and MRSE groups require extraordinary attention when dealing with PJIs in order to successfully treat them. Controlling carriage, using optimal prosthetic materials, and implementing perioperative antimicrobial prophylaxis are crucial strategies in infection prevention and are as essential as quick diagnosis and effective targeted treatment. The comprehensive professional procedures presented in this review show how to deal with such cases. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Exploiting biological tools for post-antibiotic era: novel sustainable strategies against antimicrobial resistance
- Author
-
Omnia Karem Riad
- Subjects
probiotic and predatory bacteria ,bacteriophage therapy ,antimicrobial peptides ,bacterial ghosts ,bacterial outer membrane vesicles (bomvs) ,antibiotic resistance ,Microbiology ,QR1-502 - Abstract
Around the world, one of the biggest risks to public health is antimicrobial resistance (AMR). AMR poses substantial consequences on country economies and health systems. In the post-antibiotic era, searching for new cost-effective approaches is necessary to compensate for the continuous increase in AMR. The aims of this review are to explore the different biological and sustainable approaches that should be exploited to overcome the problem of AMR, discuss the mechanisms and advantages of different sustainable biological strategies, and introduce several strategies, including probiotic bacteria, predatory bacteria, and bacteriophages, which are powerful tools valuable to fighting resistant microorganisms with fewer chances of resistance development. Naturally synthesized products such as antimicrobial peptides and bacteriocins revealed successful treatment options. Additionally, the Clustered regularly interspaced short palindromic repeats (CRISPR-Cas) system is a gene-editing tool that can re-sensitize the resistant bacteria. Vaccination prevents infectious diseases and halts the emergence of resistant pathogens. Meanwhile, bacterial ghosts (BGs) and bacterial outer membrane vesicles (bOMVs) can be used to develop safe vaccines. bOMVs are also used as efficient tools for drug delivery due to their nanosizes. Additionally, antibody therapy and fecal microbial transplants are successful tools. Developing sustainable strategies to combat AMR through biological means looks promising. When compared to conventional antibiotics, these tactics have different mechanisms of action that may slow the emergence of antibiotic resistance.
- Published
- 2024
- Full Text
- View/download PDF
10. Confronting Accelerating Global Antimicrobial Resistance and the Associated Increase in Deaths
- Author
-
Daniel Amsterdam
- Subjects
antimicrobial resistance ,artificial intelligence ,global mortality ,bacteriophage therapy ,antimicrobials with unique mechanisms of action ,Biotechnology ,TP248.13-248.65 ,Medicine - Abstract
Although advances in contemporary medical care have broadened access to healthcare and extended the human life span, deaths resulting from antimicrobial-resistant pathogens continue to increase. This minireview summarizes the evidence that AI and machine learning, coupled with precision medicine and alternative therapies, such as repurposing non-antibiotic drugs and the use of bacteriophages, has promise to halt this advance.
- Published
- 2024
- Full Text
- View/download PDF
11. Periodontitis: etiology, conventional treatments, and emerging bacteriophage and predatory bacteria therapies.
- Author
-
Łasica, Anna, Golec, Piotr, Laskus, Agnieszka, Zalewska, Magdalena, Gędaj, Magdalena, and Popowska, Magdalena
- Subjects
THERAPEUTICS ,PERIODONTAL disease ,DRUG resistance in bacteria ,GINGIVAL diseases ,PERIODONTAL ligament ,CLINDAMYCIN ,TETRACYCLINES - Abstract
Inflammatory periodontal diseases associated with the accumulation of dental biofilm, such as gingivitis and periodontitis, are very common and pose clinical problems for clinicians and patients. Gingivitis is a mild form of gum disease and when treated quickly and properly is completely reversible. Periodontitis is an advanced and irreversible disease of the periodontium with periods of exacerbations, progressions and remission. Periodontitis is a chronic inflammatory condition that damages the tissues supporting the tooth in its socket, i.e., the gums, periodontal ligaments, root cementum and bone. Periodontal inflammation is most commonly triggered by bacteria present in excessive accumulations of dental plaque (biofilm) on tooth surfaces. This disease is driven by disproportionate host inflammatory immune responses induced by imbalance in the composition of oral bacteria and changes in their metabolic activities. This microbial dysbiosis favors the establishment of inflammatory conditions and ultimately results in the destruction of tooth-supporting tissues. Apart microbial shift and host inflammatory response, environmental factors and genetics are also important in etiology In addition to oral tissues destruction, periodontal diseases can also result in significant systemic complications. Conventional methods of periodontal disease treatment (improving oral hygiene, dental biofilm control, mechanical plaque removal, using local or systemic antimicrobial agents) are not fully effective. All this prompts the search for new methods of therapy. Advanced periodontitis with multiple abscesses is often treated with antibiotics, such as amoxicillin, tetracycline, doxycycline, minocycline, clindamycin, or combined therapy of amoxicillin with metronidazole. However, due to the growing problem of antibiotic resistance, treatment does not always achieve the desired therapeutic effect. This review summarizes pathogenesis, current approaches in treatment, limitations of therapy and the current state of research on the possibility of application of bacteriophages and predatory bacteria to combat bacteria responsible for periodontitis. We present the current landscape of potential applications for alternative therapies for periodontitis based on phages and bacteria, and highlight the gaps in existing knowledge that need to be addressed before clinical trials utilizing these therapeutic strategies can be seriously considered. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. A New Approach for Phage Cocktail Design in the Example of Anti-Mastitis Solution.
- Author
-
Królikowska, Daria, Szymańska, Marta, Krzyżaniak, Marta, Guziński, Arkadiusz, Matusiak, Rafał, Kajdanek, Agnieszka, Kaczorek-Łukowska, Edyta, Maszewska, Agnieszka, Wójcik, Ewelina A., and Dastych, Jarosław
- Subjects
ESCHERICHIA coli ,BACTERIAL diseases ,STRUCTURAL models ,ANIMAL welfare ,DRUG resistance in bacteria ,BACTERIOPHAGES - Abstract
The studies on phage therapy have shown an overall protective effect of phages in bacterial infections, thus providing an optimistic outlook on the future benefits of phage-based technologies for treating bacterial diseases. However, the therapeutic effect is highly affected by the proper composition of phage cocktails. The rational approach to the design of bacteriophage cocktails, which is the subject of this study, allowed for development of an effective anti-mastitis solution, composed of virulent bacteriophages acting on Escherichia coli and Staphylococcus aureus. Based on the in-depth bioinformatic characterization of bacteriophages and their in vitro evaluation, the cocktail of five phages against E. coli and three against S. aureus strains was composed. Its testing in the milk model experiment revealed a reduction in the number of S. aureus of 45% and 30% for E. coli strains, and in the study of biofilm prevention, it demonstrated 99% inhibition of biofilm formation for all tested S. aureus strains and a minimum of 50% for 50% of E. coli strains. Such insights justify the need for rational design of cocktails for phage therapy and indicate the potential of the developed cocktail in the treatment of diseased animals, but this requires further investigations to evaluate its in vivo efficacy. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
13. Optimization of bacteriophage therapy for difficult-to-treat musculoskeletal infections: a bench-to-bedside perspective.
- Author
-
Bessems, Laura, Baixing Chen, Uyttebroek, Saartje, Devolder, David, Lood, Ce' dric, Verwimp, Stefaan, De Munter, Paul, Debaveye, Yves, Depypere, Melissa, Spriet, Isabel, Van Gerven, Laura, Dupont, Lieven, Wagemans, Jeroen, van Noort, Vera, Lavigne, Rob, Metsemakers, Willem-Jan, and Onsea, Jolien
- Subjects
MOLECULAR kinetics ,NUCLEOTIDE sequencing ,DRUG resistance in microorganisms ,MOLECULAR interactions ,DATA quality - Abstract
Given the increasing threat of antimicrobial resistance, scientists are urgently seeking adjunct antimicrobial strategies, such as phage therapy (PT). However, despite promising results for the treatment of musculoskeletal infections in our center, crucial knowledge gaps remain. Therefore, a prospective observational study (PHAGEFORCE) and a multidisciplinary approach was set up to achieve and optimize standardized treatment guidelines. At our center, PT is strictly controlled and monitored by a multidisciplinary taskforce. Each phage treatment follows the same pathway to ensure standardization and data quality. Within the PHAGEFORCE framework, we established a testing platform to gain insight in the safety and efficacy of PT, biodistribution, phage kinetics and the molecular interaction between phages and bacteria. The draining fluid is collected to determine the phage titer and bacterial load. In addition, all bacterial isolates are fully characterized by genome sequencing to monitor the emergence of phage resistance. We hereby present a standardized bench-to-bedside protocol to gain more insight in the kinetics and dynamics of PT for musculoskeletal infections. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
14. Confronting Accelerating Global Antimicrobial Resistance and the Associated Increase in Deaths.
- Author
-
Amsterdam, Daniel
- Subjects
DRUG resistance in microorganisms ,DEATH rate ,ARTIFICIAL intelligence ,THERAPEUTIC use of bacteriophages ,ANTI-infective agents - Abstract
Although advances in contemporary medical care have broadened access to healthcare and extended the human life span, deaths resulting from antimicrobial-resistant pathogens continue to increase. This minireview summarizes the evidence that AI and machine learning, coupled with precision medicine and alternative therapies, such as repurposing non-antibiotic drugs and the use of bacteriophages, has promise to halt this advance. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. Safety and Tolerability of ShigActive™, a Shigella spp. Targeting Bacteriophage Preparation, in a Phase 1 Randomized, Double-Blind, Controlled Clinical Trial.
- Author
-
Chen, Wilbur H., Woolston, Joelle, Grant-Beurmann, Silvia, Robinson, Courtney K., Bansal, Garima, Nkeze, Joseph, Permala-Booth, Jasnehta, Fraser, Claire M., Tennant, Sharon M., Shriver, Mallory C., Pasetti, Marcela F., Liang, Yuanyuan, Kotloff, Karen L., Sulakvelidze, Alexander, and Schwartz, Jennifer A.
- Subjects
BACTERIAL diseases ,INFLAMMATORY mediators ,GASTROINTESTINAL diseases ,MORTALITY ,SODIUM bicarbonate ,SHIGELLOSIS - Abstract
Bacterial diseases of the gastrointestinal (GI) tract continue to be a major worldwide cause of human morbidity and mortality. Among various enteric pathogens, Shigella spp. are some of the most common and deadly bacterial pathogens. They are responsible for ~125 million worldwide cases of shigellosis, and ~14,000 deaths annually, the majority in children under the age of 5 and occurring in developing countries. Preventing and treating shigellosis with conventional drugs (e.g., vaccines and antibiotics) has proven to be very difficult. Here, we assessed the safety and tolerability of ShigActive™, a lytic bacteriophage preparation targeting Shigella spp., in a randomized, placebo-controlled, double-blind Phase 1 clinical trial. Ten participants randomized 4:1 received ShigActive™ or placebo co-administered with sodium bicarbonate orally three times daily for 7 days. Solicited and unsolicited adverse events (AEs) were observed for 29 days. Fifty percent of the subjects receiving ShigActive™ reported mild GI-related symptoms, while one participant experienced moderate fatigue. No serious or medically attended AEs occurred through day 90. Additionally, no significant differences in GI-associated inflammatory mediators or fecal microbiome changes were observed between placebo- and ShigActive™-treated subjects, or from a participants' baseline value. The results of this first-in-human (FIH) randomized, controlled Phase 1 trial of ShigActive™ demonstrate that it is safe and well tolerated when orally administered with no significant differences compared to placebo controls. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. Harnessing Bacteriophages to Combat Antibiotic-Resistant Infections in Africa: A Comprehensive Review.
- Author
-
Fabiyi, Kafayath, Sintondji, Kevin, Agbankpe, Jerrold, Assogba, Phenix, Koudokpon, Hornel, Lègba, Boris, Gbotche, Elodie, Baba-Moussa, Lamine, and Dougnon, Victorien
- Subjects
MEDICAL personnel ,DRUG resistance in bacteria ,BACTERIAL diseases ,RESEARCH personnel - Abstract
The conventional treatment of bacterial infections with antibiotics is becoming increasingly ineffective due to the emergence of multidrug-resistant (MDR) pathogens. This literature review explores the potential of bacteriophages as an alternative or adjunctive therapy to antibiotics in combating MDR infections in Africa. This analysis focuses on current research regarding the integration of phage therapy into African healthcare, highlighting its challenges and opportunities. This review begins with the AMR crisis and the need for new treatments, then covers the history, mechanisms, benefits, and limitations of phage therapy. Key African studies are summarized, identifying major obstacles such as regulatory issues, infrastructure, and research standardization. Research efforts in West Africa that have made notable progress in bacteriophage research are highlighted. This review concludes with recommendations for policymakers, researchers, and healthcare professionals to enhance the development and use of phage therapy in Africa, aiming to reduce antibiotic resistance and improve patient outcomes. By addressing the identified challenges and leveraging the unique advantages of phages, there is potential to significantly mitigate the impact of antibiotic resistance and improve patient outcomes in Africa. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. Editorial: Global excellence in bacteriology: Central and South America
- Author
-
Fernando Navarro-Garcia, Marcelo Brocchi, and Angel A. Cataldi
- Subjects
antimicrobial resistance ,complement system lysis ,mucus hypersecretion ,bacterial transmission ,bacteriophage therapy ,Microbiology ,QR1-502 - Published
- 2025
- Full Text
- View/download PDF
18. Phage-mediated virulence loss and antimicrobial susceptibility in carbapenem-resistant Klebsiella pneumoniae
- Author
-
Yanshuang Yu, Mengzhu Wang, Liuying Ju, Minchun Li, Mengshi Zhao, Hui Deng, Christopher Rensing, Qiu E. Yang, and Shungui Zhou
- Subjects
antibiotic resistance ,bacteriophage therapy ,Klebsiella pneumoniae ,phage training ,coevolution ,Microbiology ,QR1-502 - Abstract
ABSTRACT Bacteriophages, known for their ability to kill bacteria, are hampered in their effectiveness because bacteria are able to rapidly develop resistance, thereby posing a significant challenge for the efficacy of phage therapy. The impact of evolutionary trajectories on the long-term success of phage therapy remains largely unclear. Herein, we conducted evolutionary experiments, genomic analysis, and CRISPR-mediated gene editing, to illustrate the evolutionary trajectory occurring between phages and their hosts. Our results illustrate the ongoing “arms race” between a lytic phage and its host, a carbapenem-resistant Klebsiella pneumoniae clinical strain Kp2092, suggesting their respective evolutionary adaptations that shape the efficacy of phage therapy. Specifically, Kp2092 rapidly developed resistance to phages through mutations in a key phage receptor (galU) and bacterial membrane defenses such as LPS synthesis, however, this evolution coincides with unexpected benefits. Evolved bacterial clones not only exhibited increased sensitivity to clinically important antibiotics but also displayed a loss of virulence in an in-vivo model. In contrast, phages evolved under the selection pressure against Kp2092 mutants and exhibited enhanced bacterial killing potency, targeting mutations in phage tail proteins gp12 and gp17. These parallel evolutionary trajectories suggest a common genetic mechanism driving adaptation, ultimately favoring the efficacy of phage therapy. Overall, our findings highlight the potential of phages not only as agents for combating bacterial resistance, but also a driver of evolution outcomes that could lead to more favorable clinical outcomes in the treatment of multidrug resistance pathogens.IMPORTANCECarbapenem-resistant Klebsiella pneumoniae represents one of the leading pathogens for infectious diseases. With traditional antibiotics often being ineffective, phage therapy has emerged as a promising alternative. However, phage predation imposes a strong evolutionary pressure on the rapid evolution of bacteria, challenging treatment efficacy. Our findings illustrate how co-evolution enhances phage lytic capabilities through accumulated mutations in the tail proteins gp12 and gp17, while simultaneously reducing bacterial virulence and antibiotic resistance. These insights advance our understanding of phage-host interactions in clinical settings, potentially inspiring new approaches akin to an “arms race” model to combat multidrug-resistant crises effectively.
- Published
- 2025
- Full Text
- View/download PDF
19. Rising prevalence and drug resistance of Corynebacterium striatum in lower respiratory tract infections
- Author
-
Wei Li, Mingyue Gao, and Jinyan Yu
- Subjects
Corynebacterium striatum ,drug resistance ,lower respiratory tract infections ,vancomycin therapy ,multidrug resistance mechanisms ,bacteriophage therapy ,Microbiology ,QR1-502 - Abstract
Corynebacterium striatum (C. striatum) is a Gram-positive bacterium commonly colonizing the skin and mucosa in healthy individuals and hospitalized patients. Traditionally regarded as a contaminant, C. striatum is now increasingly recognized as a potential cause of clinical infections, especially after the coronavirus disease pandemic. It has emerged as a pathogen implicated in severe infections, including pneumonia, bacteremia, meningitis, artificial joint infections, abdominal infections, and endocarditis. C. striatum has been reported in lower respiratory tract infections, mostly as a conditioned pathogen in immunocompromised individuals, particularly in those with chronic structural lung diseases, such as chronic obstructive pulmonary disease, leading to severe pneumonia or exacerbation of the existing disease and high mortality. Additionally, C striatum has been implicated in the community-acquired pneumonia among immunocompetent individuals and nosocomial lung infections, with evidence of person-to-person transmission through caregivers. C. striatum may exhibit multidrug resistance. Vancomycin, alone or in combination, is currently considered the most effective treatment for C. striatum. This review highlights the epidemiological characteristics, drug resistance mechanisms, diagnostics approaches, and treatment options for C. striatum lower respiratory tract infections to enhance clinician awareness and improve patient management strategies.
- Published
- 2025
- Full Text
- View/download PDF
20. Alhagi maurorum extract in combination with lytic phage cocktails: a promising therapeutic approach against biofilms of multi-drug resistant P. mirabilis
- Author
-
Arezoo Mirzaei, Bahram Nasr Esfahani, Mustafa Ghanadian, Jeroen Wagemans, Rob Lavigne, and Sharareh Moghim
- Subjects
Alhagi maurorum ,bacteriophage therapy ,Proteus mirabilis ,biofilm ,combination therapy ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Antimicrobial resistance (AMR) poses a significant global threat to public health systems, rendering antibiotics ineffective in treating infectious diseases. Combined use of bio compounds, including bacteriophages and plant extracts, is an attractive approach to controlling antibiotic resistance. In this study, the combination of phage cocktail (Isf-Pm1 and Isf-Pm2) and Alhagi maurorum crude extract (AME) was investigated in controlling biofilm-forming multi-drug resistant P. mirabilis isolates, in vitro and a phantom bladder model. The combination of AME and phage cocktails demonstrated no significant disparity in its ability to inhibit quorum sensing (QS) when compared to the individual control of AME alone. Following treatment with the combination of phage cocktail and AME at a 125 μg/mL concentration, the MDR P. mirabilis biofilm biomass was notably reduced by 73% compared to the control (P< 0.0001). The anti-biofilm effect was confirmed by Scanning Electron Microscopy (SEM). Moreover, in a bladder phantom model, there was a considerable decrease in encrustation levels compared to the control. The combined treatment resulted in a 1.85 logarithmic reduction in bacterial adhesion to Vero cells compared to the control. The real-time PCR results indicated significant downregulation of QS- and adhesion-related gens. The phage therapy, combined with AME, holds promising potential in reducing biofilm formation.
- Published
- 2024
- Full Text
- View/download PDF
21. It takes two to tango: Preserving daptomycin efficacy against daptomycin-resistant MRSA using daptomycin-phage co-therapy
- Author
-
Casey L. Madison, Anja S. J. Steinert, Corrin E. Luedeke, Neeka Hajjafar, Prakhar Srivastava, Andrew D. Berti, Arnold S. Bayer, and Razieh Kebriaei
- Subjects
antibiotic resistance ,bacteriophage therapy ,Staphylococcus aureus ,Microbiology ,QR1-502 - Abstract
ABSTRACT Daptomycin (DAP) is a cyclic lipopeptide antibiotic that targets the cell membrane and is commonly used for the treatment of multidrug-resistant Staphylococcus aureus infections, especially methicillin-resistant strains (MRSA). Emergence of DAP resistance during DAP therapy is increasingly reported; however, the mechanisms by which resistance occurs are diverse. Using phages in combination with antibiotics is of high interest to circumvent antibiotic resistance, due to their minimal side effects, accessibility, and synergistic effects with antibiotics. Here, we have investigated the relationship between DAP resistance and sensitivity to phage Sb-1 in a cohort of 14 clinical MRSA strains. This cohort consists of four isogenic pairs (a clinical DAP-susceptible parent and its isogenic DAP-resistant variant isolated during DAP treatment), and three DAP-susceptible/DAP-resistant strain pairs are created by traditional serial passaging in the presence of increasing DAP concentrations. Efficiency of plating (EOP) and bacteriophage plaque sizes were recorded, and synergy between the antibacterial agents was tested using the phage-antibiotic combination (PAC) checkerboard method. Plaque sizes and EOP significantly increased (P = 0.019 and P = 0.008, respectively) as strains developed resistance to DAP. The PAC checkerboards conveyed less antagonistic patterns in DAP-resistant mutant strains compared with their DAP-susceptible counterparts. Antagonism was displayed in 71.4% (5/7) of the DAP-susceptible strains, while it was only present in 14.3% (1/7) of the DAP-resistant mutants. Transmission electron microscopy images showed significantly thicker cell walls in DAP-susceptible strains exposed to DAP monotherapy compared with either growth control or DAP-phage (P = 0.0002 and 0.0007, respectively). These data indicate increased sensitivity to phage Sb-1 concurrent with the emergence of DAP resistance.IMPORTANCEMultidrug-resistant Staphylococcus aureus is a threat to the health care system, especially cross-resistance between daptomycin (DAP) and glycopeptides through various mutations such as mprF (which is involved in the modification of membrane phospholipids in some bacteria) and yycG (part of a two-component regulatory system in bacteria that is important for regulating cell wall biosynthesis and other cellular processes) has been reported previously. Our current study shows adjunctive treatment with phage in DAP-resistant strains will lead to synergistic activity and larger phage plaque sizes, translating to elevated lytic performance. The addition of bacteriophage to standard-of-care antibiotic therapies for multidrug-resistant S. aureus infections has the potential to hinder, and possibly revert, resistance to antibiotics. Applying this strategy can potentially lead to the preservation of the current antibiotics. Verification of this salutary outcome in relevant ex vivo and in vivo models of endovascular infections is required to validate translatability.
- Published
- 2024
- Full Text
- View/download PDF
22. Effects of the combination of anti-PcrV antibody and bacteriophage therapy in a mouse model of Pseudomonas aeruginosa pneumonia
- Author
-
Ohara Junya, Fujiki Jumpei, Mao Kinoshita, Kazuki Sudo, Ken Kawaguchi, Keita Inoue, Yoshifumi Naito, Kiyoshi Moriyama, Tomohiro Nakamura, Hidetomo Iwano, and Teiji Sawa
- Subjects
bacteriophage therapy ,PcrV ,Pseudomonas aeruginosa ,acute lung injury ,type III secretion system ,Microbiology ,QR1-502 - Abstract
ABSTRACT Acute lung injury caused by Pseudomonas aeruginosa is attributed to the translocation of cytotoxin into pulmonary epithelial cells via the P. aeruginosa type III secretion system. This virulence can be blocked with a specific antibody against PcrV in this secretion system. However, because anti-PcrV antibodies do not have bactericidal activity, the treatment of bacteria depends on the phagocytic system of the host. In this study, we investigated the therapeutic effect of combination therapy with an anti-PcrV antibody and bactericidal bacteriophages on acute lung injury and subsequent death in mice compared with a single treatment. After the mice intratracheally received a lethal dose of the cytotoxic P. aeruginosa strain, a second instillation was performed with saline, anti-PcrV IgG, bacteriophages, or a mixture of anti-PcrV and bacteriophages. The survival rates 24 h after infection were as follows: 7.1% in the saline group, 26.7% in the anti-PcrV group, 41.2% in the phage group, and 66.7% in the anti-PcrV + phage group (P < 0.001 vs saline-treated group). The activity of surviving mice in the anti-PcrV + phage group was significantly greater than that in the saline group. The lung weight in the anti-PcrV + phage group was significantly lower than that in the anti-PcrV group. In conclusion, combination therapy with an anti-PcrV antibody and a bacteriophage reduces acute lung injury and suggests improved survival compared with each treatment alone. This combination therapy, which does not rely on conventional antibiotics, could constitute a new strategy for treating multidrug-resistant P. aeruginosa infections.IMPORTANCECombination therapy with either bacteriophages alone or in combination with anti-PcrV antibodies in a mouse model of Pseudomonas aeruginosa pneumonia may reduce the acute lung injury and improve survival. This combination therapy, which does not rely on conventional antibiotics, may be a new strategy to treat multidrug-resistant Pseudomonas aeruginosa infections.
- Published
- 2024
- Full Text
- View/download PDF
23. Targeted dual-receptor phage cocktail against Cronobacter sakazakii: insights into phage-host interactions and resistance mechanisms
- Author
-
Seongok Kim, Bokyung Son, Yeran Kim, Hyeongsoon Kim, Gahyeon Nam, Hakdong Shin, and Sangryeol Ryu
- Subjects
Cronobacter sakazakii ,phage cocktail ,flagella ,lipopolysaccharides ,bacteriophage therapy ,phage resistance ,Microbiology ,QR1-502 - Abstract
IntroductionCronobacter sakazakii is a notorious foodborne pathogen, frequently contaminating powdered infant formula and causing life-threatening diseases in infants. The escalating emergence of antibiotics-resistant mutants has led to increased interest in using bacteriophage as an alternative antimicrobial agent.MethodsTwo phages, CR8 and S13, were isolated from feces and soil samples and their morphology, physiology, and genomics were characterized. Phage receptor was determined using deletion mutants lacking flgK, rfaC, fhuA, btuB, lamb, or ompC genes, followed by complementation. Phage-resistant mutants were analyzed for phenotypic changes and fitness trade-offs using motility assays and Caco-2 cell invasion models.ResultsCR8 and S13 were identified as members of Caudoviricetes. Phage CR8 and phage S13 utilize flagella and LPS, respectively, to adhere to host cells. Bacterial challenge assay demonstrated delayed emergence of the resistant mutant as well as stronger lytic activity of a phage cocktail consisting of CR8 and S13 than the single phage treatment. Phenotypic analysis of the phage cocktail resistant strain, designated as CSR strain, revealed that the resistance resulted from the impaired receptor proteins for phage, such as defects in motility and alteration in LPS structure. CSR strain exhibited significant attenuation in invading human intestinal epithelial Caco-2 cells compared to WT cells.ConclusionThis study demonstrates that the development of the phage cocktail targeting distinct host receptors can serve as a promising antimicrobial strategy to effectively control C. sakazakii.
- Published
- 2024
- Full Text
- View/download PDF
24. Isolation and characterization of lytic bacteriophage vB_KpnP_23: A promising antimicrobial candidate against carbapenem-resistant Klebsiella pneumoniae
- Author
-
Qian Wang, Ran Chen, Hui Liu, Yue Liu, Jinmei Li, Yueling Wang, Yan Jin, Yuanyuan Bai, Zhen Song, Xinglun Lu, Changyin Wang, and Yingying Hao
- Subjects
Klebsiella pneumoniae ,Bacteriophage therapy ,Carbapenem-resistant ,Genome analysis ,Microbiology ,QR1-502 ,Infectious and parasitic diseases ,RC109-216 - Abstract
The global health threat posed by carbapenem-resistant Klebsiella pneumoniae (CRKP) is exacerbated by the limited availability of effective treatments. Bacteriophages are promising alternatives to conventional antimicrobial agents. However, current phage databases are limited. Thus, identifying and characterizing new phages could provide biological options for the treatment of multi-drug resistant bacterial infections. Here, we report the characterization of a novel lytic phage, vB_KpnP_23, isolated from hospital sewage. This phage exhibited potent activity against carbapenemase-producing CRKP strains and was characterised by an icosahedral head, a retractable tail, and a genome comprising 40,987 base pairs, with a G + C content of 51 %. Capable of targeting and lysing nine different capsule types (K-types) of CRKP, including the clinically relevant ST11-K64, it demonstrated both high bacteriolytic efficiency and stability in various environmental contexts. Crucially, vB_KpnP_23 lacks virulence factors, antimicrobial resistance genes, or tRNA, aligning with the key criteria for therapeutic application. In vitro evaluation of phage-antibiotic combinations revealed a significant synergistic effect between vB_KpnP_23 and meropenem, levofloxacin, or amikacin. This synergy could lead to an 8-fold reduction in the minimum inhibitory concentration (MIC), suggesting that integrated treatments combining this phage with the aforementioned antibiotics may substantially enhance drug effectiveness. This approach not only extends the clinical utility of these antibiotics but also presents a strategic advance in combating antibiotic resistance. Specifically, it underscores the potential of phage-antibiotic combinations as a powerful tool in the treatment of infections caused by CRKP, offering a promising avenue to mitigate the public health challenges of antibiotic-resistant pathogens.
- Published
- 2024
- Full Text
- View/download PDF
25. Novel lytic bacteriophage AhFM11 as an effective therapy against hypervirulent Aeromonas hydrophila
- Author
-
Nithin Muliya Sankappa, Girisha Shivani Kallappa, Kushala Kallihosuru Boregowda, Namrutha Mandrira Ramakrishna, Prithvisagar Kattapuni Suresh, Dheeraj Shriraje Balakrishna, Krishna Kumar Ballamoole, Suresh Thangavel, Lopamudra Sahoo, Miles D. Lange, Michael B. Deshotel, and Jason W. Abernathy
- Subjects
Aquaculture ,Lytic phage ,Hypervirulent Aeromonas hydrophila ,Antibiotic resistance genes and biocontrol ,Bacteriophage therapy ,Medicine ,Science - Abstract
Abstract Several farmed fish species, including carps, tilapia, salmon, and catfish, have experienced significant economic losses in aquaculture due to motile Aeromonas septicemia caused by Aeromonas hydrophila. In the present study, a novel lytic bacteriophage infecting hypervirulent Aeromonas hydrophila (vAh) was isolated and characterized. This is the first report of a phage against vAh. Phage AhFM11 demonstrated lytic activity against both vAh strains and the A. hydrophila reference strain ATCC 35654. The AhFM11 genome was sequenced and assembled, comprising 168,243 bp with an average G + C content of 41.5%. The genome did not harbor any antibiotic resistance genes. Genomic information along with transmission electron microscopy revealed that phage AhFM11 belongs to the Straboviridae family. Therapeutic application of monophage AhFM11 in fish showed 100% survival in injection, 95% in immersion and 93% in oral feeding of phage top-coated feed. Fish and chicken meat spiked with A. hydrophila and phage showed significant reduction of A. hydrophila. These findings support that phage AhFM11 can be used as a biocontrol agent against vAh as an alternative to antibiotics.
- Published
- 2024
- Full Text
- View/download PDF
26. The trade-off of Vibrio parahaemolyticus between bacteriophage resistance and growth competitiveness.
- Author
-
Xiuxiu Zeng, Shanyan Liang, Jiayi Dong, Guosheng Gao, Yaoren Hu, and Yuechao Sun
- Subjects
SEDIMENT sampling ,BACTERIOPHAGES ,MICROPLATES ,VIBRIO parahaemolyticus ,BACTERIA ,PATHOGENIC microorganisms - Abstract
Vibrio parahaemolyticus is a food-borne pathogen, which is often isolated from various seafood products. In this study, two kinds of bacteriophages was isolated from the offshore sediments samples. The anti-phage mutant strain were obtained after seventeen rounds of co-culture of Vibrio parahaemolyticus and mixed bacteriophage, multigroup sequencing was carried out on spontaneous the anti-phage mutant strain and the wild-type strain. We used the Sanger sequencing to verify the accuracy of the mutation sites. Biolog GEN III MicroPlates were used to evaluate the metabolic capacity of wild-type strains and the anti-phage mutant strain. In this study, we found that with flaG gene (slight homology to N terminus of multiple flagellins) mutated, making the bacteriophage unable to absorb to the cell surface of the host. And, the growth competitiveness of the anti-phage mutant strain is lower than the wild-type strain. These results indicated that the fitness cost, including loss of the growth competitiveness, constitutes a barrier to the prevalence of these defense mechanisms. And the selection pressure on different anti-phage strategies depends on the trade-off between mortality imposed by bacteriophages and fitness cost of the defense strategy under the given environmental conditions. In conclusion, this study provides valuable insights into the phagehost interaction and phage resistance in Vibrio parahaemolyticus. Our study provided knowledge for the evolutionary adaption of bacteria against the bacteriophage, which could add more information to understand the phage resistance mechanism before applying in the industry. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
27. Evaluation of bacteriophage therapy of Aeromonas hydrophila infection in a freshwater fish, Pangasius buchanani.
- Author
-
Kumari, Ritu, Yadav, Ragini, Kumar, Deepak, Chaube, Radha, and Nath, Gopal
- Subjects
THERAPEUTIC use of bacteriophages ,AEROMONAS hydrophila ,PANGASIUS ,AQUACULTURE ,FISHES - Abstract
Introduction: The present study aimed to optimize the doses and schedule of specific bacteriophage cocktails in freshwater fish infections as prophylactic and therapeutic measures. Methods: The three most active phages against Aeromonas hydrophila (A. hydrophila) (φAHBHU12, φAHBHU16, and φAHBHU19) were characterized phenotypically and genotypically. Intramuscular and water immersion routes were used to calculate the absolute lethal dose of A. hydrophila in Pangasius buchanani. Phage therapy was given simultaneously and after 6, 12, and 24 h of bacterial challenge through intramuscular and water immersion routes. Results: The prophylactic and early phage administration could save the fish. Furthermore, the dose of intramuscular 1.0 × 10
4 plaque-forming unit (PFU)/fish and water immersion 1.0 × 106 PFU mL-1 of the phage cocktail was optimal. Discussion: The efficacy of bacteriophage therapy as preventive or curative measures practical when administered simultaneously or early hours of A. hydrophila infection in aquaculture systems. Phage-based approaches may be used as an alternative to antibiotics in aquaculture to reduce antibiotic use as a part of the "One Health" approach. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
28. Novel lytic bacteriophage AhFM11 as an effective therapy against hypervirulent Aeromonas hydrophila.
- Author
-
Muliya Sankappa, Nithin, Shivani Kallappa, Girisha, Kallihosuru Boregowda, Kushala, Mandrira Ramakrishna, Namrutha, Kattapuni Suresh, Prithvisagar, Shriraje Balakrishna, Dheeraj, Ballamoole, Krishna Kumar, Thangavel, Suresh, Sahoo, Lopamudra, Lange, Miles D., Deshotel, Michael B., and Abernathy, Jason W.
- Subjects
AEROMONAS hydrophila ,BACTERIOPHAGES ,SALMON farming ,CHICKEN as food ,TRANSMISSION electron microscopy ,DRUG resistance in bacteria ,BIOLOGICAL pest control agents - Abstract
Several farmed fish species, including carps, tilapia, salmon, and catfish, have experienced significant economic losses in aquaculture due to motile Aeromonas septicemia caused by Aeromonas hydrophila. In the present study, a novel lytic bacteriophage infecting hypervirulent Aeromonas hydrophila (vAh) was isolated and characterized. This is the first report of a phage against vAh. Phage AhFM11 demonstrated lytic activity against both vAh strains and the A. hydrophila reference strain ATCC 35654. The AhFM11 genome was sequenced and assembled, comprising 168,243 bp with an average G + C content of 41.5%. The genome did not harbor any antibiotic resistance genes. Genomic information along with transmission electron microscopy revealed that phage AhFM11 belongs to the Straboviridae family. Therapeutic application of monophage AhFM11 in fish showed 100% survival in injection, 95% in immersion and 93% in oral feeding of phage top-coated feed. Fish and chicken meat spiked with A. hydrophila and phage showed significant reduction of A. hydrophila. These findings support that phage AhFM11 can be used as a biocontrol agent against vAh as an alternative to antibiotics. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
29. Phage resistance in Klebsiella pneumoniae and bidirectional effects impacting antibiotic susceptibility.
- Author
-
Nang, Sue C., Lu, Jing, Yu, Heidi H., Wickremasinghe, Hasini, Azad, Mohammad A.K., Han, Meiling, Zhao, Jinxin, Rao, Gauri, Bergen, Phillip J., Velkov, Tony, Sherry, Norelle, McCarthy, David T., Aslam, Saima, Schooley, Robert T., Howden, Benjamin P., Barr, Jeremy J., Zhu, Yan, and Li, Jian
- Subjects
- *
KLEBSIELLA pneumoniae , *BACTERIOPHAGES , *DRUG resistance in bacteria , *ANTIBIOTICS , *DRUG resistance in microorganisms , *COLISTIN - Abstract
Bacteriophage (phage) therapy is a promising anti-infective option to combat antimicrobial resistance. However, the clinical utilization of phage therapy has been severely compromised by the potential emergence of phage resistance. Although certain phage resistance mechanisms can restore bacterial susceptibility to certain antibiotics, a lack of knowledge of phage resistance mechanisms hinders optimal use of phages and their combination with antibiotics. Genome-wide transposon screening was performed with a mutant library of Klebsiella pneumoniae MKP103 to identify phage pKMKP103_1-resistant mutants. Phage-resistant phenotypes were evaluated by time-kill kinetics and efficiency of plating assays. Phage resistance mechanisms were investigated with adsorption, one-step growth, and mutation frequency assays. Antibiotic susceptibility was determined with broth microdilution and population analysis profiles. We observed a repertoire of phage resistance mechanisms in K pneumoniae , such as disruption of phage binding (fhuA ::Tn and tonB ::Tn), extension of the phage latent period (mnmE ::Tn and rpoN ::Tn), and increased mutation frequency (mutS ::Tn and mutL ::Tn). Notably, in contrast to the prevailing view that phage resistance re-sensitizes antibiotic-resistant bacteria, we observed a bidirectional steering effect on bacterial antibiotic susceptibility. Specifically, rpoN ::Tn increased susceptibility to colistin while mutS ::Tn and mutL ::Tn increased resistance to rifampicin and colistin. Our findings demonstrate that K pneumoniae employs multiple strategies to overcome phage infection, which may result in enhanced or reduced antibiotic susceptibility. Mechanism-guided phage steering should be incorporated into phage therapy to better inform clinical decisions on phage-antibiotic combinations. [Display omitted] [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
30. Bacteriophage cocktail shows no toxicity and improves the survival of Galleria mellonella infected with Klebsiella spp.
- Author
-
Kelly, Lucy and Jameson, Eleanor
- Subjects
- *
GREATER wax moth , *BACTERIOPHAGES , *KLEBSIELLA , *KLEBSIELLA infections , *KLEBSIELLA oxytoca , *KLEBSIELLA pneumoniae - Abstract
Klebsiella spp. are causative agents of healthcare-associated infections in patients who are immunocompromised and use medical devices. The antibiotic resistance crisis has led to an increase in infections caused by these bacteria, which can develop into potentially life-threatening illnesses if not treated swiftly and effectively. Thus, new treatment options for Klebsiella are urgently required. Phage therapy can offer an alternative to ineffective antibiotic treatments for antibiotic-resistant bacteria infections. The aim of the present study was to produce a safe and effective phage cocktail treatment against Klebsiella pneumoniae and Klebsiella oxytoca, both in liquid in vitro culture and an in vivo Galleria mellonella infection model. The phage cocktail was significantly more effective at killing K. pneumoniae and K. oxytoca strains compared with monophage treatments. Preliminary phage cocktail safety was demonstrated through application in the in vivo G. mellonella model: where the phage cocktail induced no toxic side effects in G. mellonella. In addition, the phage cocktail significantly improved the survival of G. mellonella when administered as a prophylactic treatment, compared with controls. In conclusion, our phage cocktail was demonstrated to be safe and effective against Klebsiella spp. in the G. mellonella infection model. This provides a strong case for future treatment for Klebsiella infections, either as an alternative or adjunct to antibiotics. IMPORTANCE Klebsiella infections are a concern in individuals who are immunocompromised and are becoming increasingly difficult to treat with antibiotics due to their drug-resistant properties. Bacteriophage is one potential alternative therapy that could be used to tackle these infections. The present study describes the design of a non-toxic phage cocktail that improved the survival of Galleria mellonella infected with Klebsiella. This phage cocktail demonstrates potential for the safe and effective treatment of Klebsiella infections, as an adjunct or alternative to antibiotics. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
31. Non-Traditional Antibacterial Therapeutic: Focus on Safety, Manufacturing, and Regulatory Dynamics
- Author
-
Dehari, Deepa, Chaudhuri, Aiswarya, Kumar, Dulla Naveen, Agrawal, Ashish Kumar, Gangwar, Mayank, editor, and Nath, Gopal, editor
- Published
- 2024
- Full Text
- View/download PDF
32. Phage Therapy: Challenges in Its Development and Clinical Implementation
- Author
-
Nag, Vijaya Lakshmi, Bansal, Yashik, Jansari, Mahesh, Gangwar, Mayank, editor, and Nath, Gopal, editor
- Published
- 2024
- Full Text
- View/download PDF
33. Overcoming Challenges and Regulatory Hurdles for Sustainable Bacteriophage Therapy Products
- Author
-
Dave, Rajal, Ahiwale, Sangeeta, Gangwar, Mayank, editor, and Nath, Gopal, editor
- Published
- 2024
- Full Text
- View/download PDF
34. Metapopulation model of phage therapy of an acute Pseudomonas aeruginosa lung infection
- Author
-
Rogelio A. Rodriguez-Gonzalez, Quentin Balacheff, Laurent Debarbieux, Jacopo Marchi, and Joshua S. Weitz
- Subjects
bacteriophage therapy ,microbial ecology ,mathematical modeling ,infectious disease ,lung infection ,Pseudomonas aeruginosa ,Microbiology ,QR1-502 - Abstract
ABSTRACT Infections caused by multidrug resistant (MDR) pathogenic bacteria are a global health threat. Bacteriophages (“phage”) are increasingly used as alternative or last-resort therapeutics to treat patients infected by MDR bacteria. However, the therapeutic outcomes of phage therapy may be limited by the emergence of phage resistance during treatment and/or by physical constraints that impede phage–bacteria interactions in vivo. In this work, we evaluate the role of lung spatial structure on the efficacy of phage therapy for Pseudomonas aeruginosa infections. To do so, we developed a spatially structured metapopulation network model based on the geometry of the bronchial tree, including host innate immune responses and the emergence of phage-resistant bacterial mutants. We model the ecological interactions between bacteria, phage, and the host innate immune system at the airway (node) level. The model predicts the synergistic elimination of a P. aeruginosa infection due to the combined effects of phage and neutrophils, given the sufficient innate immune activity and efficient phage-induced lysis. The metapopulation model simulations also predict that MDR bacteria are cleared faster at distal nodes of the bronchial tree. Notably, image analysis of lung tissue time series from wild-type and lymphocyte-depleted mice revealed a concordant, statistically significant pattern: infection intensity cleared in the bottom before the top of the lungs. Overall, the combined use of simulations and image analysis of in vivo experiments further supports the use of phage therapy for treating acute lung infections caused by P. aeruginosa, while highlighting potential limits to therapy in a spatially structured environment given impaired innate immune responses and/or inefficient phage-induced lysis.IMPORTANCEPhage therapy is increasingly employed as a compassionate treatment for severe infections caused by multidrug-resistant (MDR) bacteria. However, the mixed outcomes observed in larger clinical studies highlight a gap in understanding when phage therapy succeeds or fails. Previous research from our team, using in vivo experiments and single-compartment mathematical models, demonstrated the synergistic clearance of acute P. aeruginosa pneumonia by phage and neutrophils despite the emergence of phage-resistant bacteria. In fact, the lung environment is highly structured, prompting the question of whether immunophage synergy explains the curative treatment of P. aeruginosa when incorporating realistic physical connectivity. To address this, we developed a metapopulation network model mimicking the lung branching structure to assess phage therapy efficacy for MDR P. aeruginosa pneumonia. The model predicts the synergistic elimination of P. aeruginosa by phage and neutrophils but emphasizes potential challenges in spatially structured environments, suggesting that higher innate immune levels may be required for successful bacterial clearance. Model simulations reveal a spatial pattern in pathogen clearance where P. aeruginosa are cleared faster at distal nodes of the bronchial tree than in primary nodes. Interestingly, image analysis of infected mice reveals a concordant and statistically significant pattern: infection intensity clears in the bottom before the top of the lungs. The combined use of modeling and image analysis supports the application of phage therapy for acute P. aeruginosa pneumonia while emphasizing potential challenges to curative success in spatially structured in vivo environments, including impaired innate immune responses and reduced phage efficacy.
- Published
- 2024
- Full Text
- View/download PDF
35. Optimization of bacteriophage therapy for difficult-to-treat musculoskeletal infections: a bench-to-bedside perspective
- Author
-
Laura Bessems, Baixing Chen, Saartje Uyttebroek, David Devolder, Cédric Lood, Stefaan Verwimp, Paul De Munter, Yves Debaveye, Melissa Depypere, Isabel Spriet, Laura Van Gerven, Lieven Dupont, Jeroen Wagemans, Vera van Noort, Rob Lavigne, Willem-Jan Metsemakers, and Jolien Onsea
- Subjects
bacteriophages ,bacteriophage therapy ,treatment optimization ,bench-to-bedside ,musculoskeletal infections ,Microbiology ,QR1-502 - Abstract
Given the increasing threat of antimicrobial resistance, scientists are urgently seeking adjunct antimicrobial strategies, such as phage therapy (PT). However, despite promising results for the treatment of musculoskeletal infections in our center, crucial knowledge gaps remain. Therefore, a prospective observational study (PHAGEFORCE) and a multidisciplinary approach was set up to achieve and optimize standardized treatment guidelines. At our center, PT is strictly controlled and monitored by a multidisciplinary taskforce. Each phage treatment follows the same pathway to ensure standardization and data quality. Within the PHAGEFORCE framework, we established a testing platform to gain insight in the safety and efficacy of PT, biodistribution, phage kinetics and the molecular interaction between phages and bacteria. The draining fluid is collected to determine the phage titer and bacterial load. In addition, all bacterial isolates are fully characterized by genome sequencing to monitor the emergence of phage resistance. We hereby present a standardized bench-to-bedside protocol to gain more insight in the kinetics and dynamics of PT for musculoskeletal infections.
- Published
- 2024
- Full Text
- View/download PDF
36. Periodontitis: etiology, conventional treatments, and emerging bacteriophage and predatory bacteria therapies
- Author
-
Anna Łasica, Piotr Golec, Agnieszka Laskus, Magdalena Zalewska, Magdalena Gędaj, and Magdalena Popowska
- Subjects
antibiotic resistance ,bacterial biofilm ,bacteriophage therapy ,inflammation bacteria ,oral microbiome ,periodontal diseases ,Microbiology ,QR1-502 - Abstract
Inflammatory periodontal diseases associated with the accumulation of dental biofilm, such as gingivitis and periodontitis, are very common and pose clinical problems for clinicians and patients. Gingivitis is a mild form of gum disease and when treated quickly and properly is completely reversible. Periodontitis is an advanced and irreversible disease of the periodontium with periods of exacerbations, progressions and remission. Periodontitis is a chronic inflammatory condition that damages the tissues supporting the tooth in its socket, i.e., the gums, periodontal ligaments, root cementum and bone. Periodontal inflammation is most commonly triggered by bacteria present in excessive accumulations of dental plaque (biofilm) on tooth surfaces. This disease is driven by disproportionate host inflammatory immune responses induced by imbalance in the composition of oral bacteria and changes in their metabolic activities. This microbial dysbiosis favors the establishment of inflammatory conditions and ultimately results in the destruction of tooth-supporting tissues. Apart microbial shift and host inflammatory response, environmental factors and genetics are also important in etiology In addition to oral tissues destruction, periodontal diseases can also result in significant systemic complications. Conventional methods of periodontal disease treatment (improving oral hygiene, dental biofilm control, mechanical plaque removal, using local or systemic antimicrobial agents) are not fully effective. All this prompts the search for new methods of therapy. Advanced periodontitis with multiple abscesses is often treated with antibiotics, such as amoxicillin, tetracycline, doxycycline, minocycline, clindamycin, or combined therapy of amoxicillin with metronidazole. However, due to the growing problem of antibiotic resistance, treatment does not always achieve the desired therapeutic effect. This review summarizes pathogenesis, current approaches in treatment, limitations of therapy and the current state of research on the possibility of application of bacteriophages and predatory bacteria to combat bacteria responsible for periodontitis. We present the current landscape of potential applications for alternative therapies for periodontitis based on phages and bacteria, and highlight the gaps in existing knowledge that need to be addressed before clinical trials utilizing these therapeutic strategies can be seriously considered.
- Published
- 2024
- Full Text
- View/download PDF
37. Phage therapy: From biological mechanisms to future directions
- Author
-
Strathdee, Steffanie A, Hatfull, Graham F, Mutalik, Vivek K, and Schooley, Robert T
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,5.1 Pharmaceuticals ,Phage Therapy ,Bacteriophages ,Bacteria ,bacteriophage ,bacteriophage therapy ,phage ,phage therapy ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
Increasing antimicrobial resistance rates have revitalized bacteriophage (phage) research, the natural predators of bacteria discovered over 100 years ago. In order to use phages therapeutically, they should (1) preferably be lytic, (2) kill the bacterial host efficiently, and (3) be fully characterized to exclude side effects. Developing therapeutic phages takes a coordinated effort of multiple stakeholders. Herein, we review the state of the art in phage therapy, covering biological mechanisms, clinical applications, remaining challenges, and future directions involving naturally occurring and genetically modified or synthetic phages.
- Published
- 2023
38. Bacteriophage therapy in musculoskeletal infections: from basic science to clinical application
- Author
-
Tristan Ferry, Jolien Onsea, Tiphaine Roussel-Gaillard, Cécile Batailler, Thomas Fintan Moriarty, and Willem-Jan Metsemakers
- Subjects
fracture-related infection ,periprosthetic joint infection ,antimicrobial resistance ,bacteriophage therapy ,Orthopedic surgery ,RD701-811 - Abstract
The treatment of musculoskeletal infections (MSIs), including periprosthetic joint infection (PJI) and fracture-related infection (FRI), is often complicated by biofilm-related challenges necessitating multiple revision surgeries and incurring substantial costs. The emergence of antimicrobial resistance (AMR) adds to the complexity of the problem, leading to increased morbidity and healthcare expenses. There is an urgent need for novel antibacterial strategies, with the World Health Organization endorsing non-traditional approaches like bacteriophage (phage) therapy. Phage therapy, involving the targeted application of lytic potent phages, shows promise in the treatment of MSIs. Although historical clinical trials and recent case studies present significant milestones in the evolution of phage therapy over the past century, challenges persist, including variability in study designs, administration protocols and phage selection. Efforts to enhance treatment efficacy consist of personalized phage therapy and combination with antibiotics. Future perspectives entail addressing regulatory barriers, standardizing treatment protocols, and conducting high-quality clinical trials to establish phage therapy’s efficacy for the treatment of MSIs. Initiatives like the PHAGEFORCE study and the PHAGEinLYON Clinic programme aim to streamline phage therapy, facilitating personalized treatment approaches and systematic data collection to advance its clinical utility in these challenging infections.
- Published
- 2024
- Full Text
- View/download PDF
39. Prevention and Modern Strategies for Managing Methicillin-Resistant Staphylococcal Infections in Prosthetic Joint Infections (PJIs)
- Author
-
Karolina Kraus, Paweł Mikziński, Jarosław Widelski, and Emil Paluch
- Subjects
Staphylococcus aureus ,coagulase-negative staphylococci ,endoprostheses ,periprosthetic joint infections (PJIs) ,antimicrobial treatment ,bacteriophage therapy ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Periprosthetic joint infections (PJIs) are a dangerous complication of joint replacement surgeries which have become much more common in recent years (mostly hip and knee replacement surgeries). Such a condition can lead to many health issues and often requires reoperation. Staphylococci is a bacterial group most common in terms of the pathogens causing PJIs. S. aureus and coagulase-negative staphylococci are found in around two-thirds of PJI cases. Recently, the numbers of staphylococci that cause such infections and that are methicillin-resistant are increasing. This trend leads to difficulties in the treatment and prevention of such infections. That is why MRSA and MRSE groups require extraordinary attention when dealing with PJIs in order to successfully treat them. Controlling carriage, using optimal prosthetic materials, and implementing perioperative antimicrobial prophylaxis are crucial strategies in infection prevention and are as essential as quick diagnosis and effective targeted treatment. The comprehensive professional procedures presented in this review show how to deal with such cases.
- Published
- 2024
- Full Text
- View/download PDF
40. Advances and optimization strategies in bacteriophage therapy for treating inflammatory bowel disease.
- Author
-
Yang Li, Xiao-meng Li, Hao-yu Duan, Kai-di Yang, and Jun-feng Ye
- Subjects
INFLAMMATORY bowel diseases ,BACTERIOPHAGES ,GUT microbiome - Abstract
In the advancement of Inflammatory Bowel Disease (IBD) treatment, existing therapeutic methods exhibit limitations; they do not offer a complete cure for IBD and can trigger adverse side effects. Consequently, the exploration of novel therapies and multifaceted treatment strategies provides patients with a broader range of options. Within the framework of IBD, gut microbiota plays a pivotal role in disease onset through diverse mechanisms. Bacteriophages, as natural microbial regulators, demonstrate remarkable specificity by accurately identifying and eliminating specific pathogens, thus holding therapeutic promise. Although clinical trials have affirmed the safety of phage therapy, its efficacy is prone to external influences during storage and transport, which may affect its infectivity and regulatory roles within the microbiota. Improving the stability and precise dosage control of bacteriophages--ensuring robustness in storage and transport, consistent dosing, and targeted delivery to infection sites --is crucial. This review thoroughly explores the latest developments in IBD treatment and its inherent challenges, focusing on the interaction between the microbiota and bacteriophages. It highlights bacteriophages' potential as microbiome modulators in IBD treatment, offering detailed insights into research on bacteriophage encapsulation and targeted delivery mechanisms. Particular attention is paid to the functionality of various carrier systems, especially regarding their protective properties and ability for colon-specific delivery. This review aims to provide a theoretical foundation for using bacteriophages as microbiome modulators in IBD treatment, paving the way for enhanced regulation of the intestinal microbiota. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
41. Phage P2-71 against multi-drug resistant Proteus mirabilis: isolation, characterization, and non-antibiotic antimicrobial potential.
- Author
-
Zhiyou Dong, Ruihu Wu, Lijuan Liu, Shengquan Ai, Jinpeng Yang, Qianlan Li, Keyi Fu, Yunian Zhou, Hualin Fu, Ziyao Zhou, Haifeng Liu, Zhijun Zhong, Xianmeng Qiu, and Guangneng Peng
- Subjects
CATHETER-associated urinary tract infections ,VETERINARY public health ,BACTERIOPHAGES ,EMERGING infectious diseases ,GENOMICS ,MORPHOGENESIS - Abstract
Proteus mirabilis, a prevalent urinary tract pathogen and formidable biofilm producer, especially in Catheter-Associated Urinary Tract Infection, has seen a worrying rise in multidrug-resistant (MDR) strains. This upsurge calls for innovative approaches in infection control, beyond traditional antibiotics. Our research introduces bacteriophage (phage) therapy as a novel non-antibiotic strategy to combat these drug-resistant infections. We isolated P2-71, a lytic phage derived from canine feces, demonstrating potent activity against MDR P. mirabilis strains. P2-71 showcases a notably brief 10-minute latent period and a significant burst size of 228 particles per infected bacterium, ensuring rapid bacterial clearance. The phage maintains stability over a broad temperature range of 30-50°C and within a pH spectrum of 4-11, highlighting its resilience in various environmental conditions. Our host range assessment solidifies its potential against diverse MDR P. mirabilis strains. Through killing curve analysis, P2-71's effectiveness was validated at various MOI levels against P. mirabilis 37, highlighting its versatility. We extended our research to examine P2-71's stability and bactericidal kinetics in artificial urine, affirming its potential for clinical application. A detailed genomic analysis reveals P2-71's complex genetic makeup, including genes essential for morphogenesis, lysis, and DNA modification, which are crucial for its therapeutic action. This study not only furthers the understanding of phage therapy as a promising non-antibiotic antimicrobial but also underscores its critical role in combating emerging MDR infections in both veterinary and public health contexts. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
42. A Novel and Effective Therapeutic Method for Treating Aeromonas schubertii Infection in Channa maculata.
- Author
-
Luo, Xia, Liao, Guoli, Fu, Xiaozhe, Liang, Hongru, Niu, Yinjie, Lin, Qiang, Liu, Lihui, Ma, Baofu, and Li, Ningqiu
- Subjects
- *
AEROMONAS , *BACTERIOPHAGES , *INTRAPERITONEAL injections , *CYTOSKELETAL proteins , *BACTERIAL diseases , *BACTERIAL growth - Abstract
Simple Summary: Aeromonas schubertii is one of the most significant opportunistic pathogen threats to humans, other mammals, and aquatic animals. Lytic bacteriophages have been recognized as effective alternatives to antibiotics for controlling bacterial infections. In this study, a lytic bacteriophage, which could effectively infect A. schubertii, was isolated for the first time. We report some of the characteristics of the phage, including its morphology, host range, storage stability, and structural proteins. Furthermore, in vivo experimental infections included intraperitoneal injection and immersion administration to evaluate the effects of phage therapy for A. schubertii infection. Our results suggest a potential use for phage SD04 in controlling this disease. Current experimental data indicate the value of using phage SD04 to prevent A. schubertii infection in snakehead. Aeromonas schubertii is a pathogen that severely affects aquatic animals, including the snakehead, Channa maculata. Lytic bacteriophages have been recognized as effective alternatives to antibiotics for controlling bacterial infections. However, there have been no reports of A. schubertii phages as far as we know. In this study, a lytic bacteriophage SD04, which could effectively infect A. schubertii, was isolated from pond water cultured with diseased snakehead. The SD04 phage formed small, round plaques on Petri dishes. Electron microscopy revealed a hexagonal head and a contractile tail. Based on its morphology, it may belong to the Myoviridae family. Two major protein bands with molecular weights of 50 and 38 kilodaltons were observed after the phage was subjected to SDS-PAGE. The phage showed a large average burst size, high specificity, and a broad host range. When stored at 4 °C, phage SD04 had high stability over 12 months and showed almost no variation within the first six months. All fish were healthy after both intraperitoneal injection and immersion administration of SD04, indicating the safety of the phage. After treatment with SD04, Channa maculata in both phage therapy groups and prevention groups showed high survival rates (i.e., 83.3 ± 3.3% and 100 ± 1.3%, respectively). Phage therapy inhibits bacterial growth in the liver, the target organ of the infected Channa maculat. The experimental results indicate the potential use of phage SD04 for preventing A. schubertii infection in Channa maculata. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
43. The stability of Staphylococcal bacteriophages with commonly used prosthetic joint infection lavage solutions.
- Author
-
Doub, James B., Fogel, Jessa, and Urish, Ken L.
- Subjects
- *
PROSTHESIS-related infections , *BACTERIOPHAGES , *IRRIGATION (Medicine) , *POVIDONE-iodine , *ACETIC acid - Abstract
The aim of this study was to assess the viability of four Staphylococcal bacteriophages when exposed to different concentrations of commonly used lavage solutions in the surgical treatment of prosthetic joint infections (PJI). Four tailed Staphylococcal bacteriophages and six different lavage solutions (chlorhexidine 4%, hydrogen peroxide 3%, acetic acid 3%, povidone iodine 10%, sodium hypochlorite 0.5%, and Vashe solution) at 100%, 1%, and 0.01% concentrations were used in this experiment. In addition, the temporal impact of exposing bacteriophages to these lavage solutions was also evaluated at 5‐min exposures and 24‐h exposures. The results show that the titers of the four bacteriophages were statistically significantly decreased for all lavage solutions (100% and 1%) at 5‐min exposures and 24‐h exposures. However, with 0.01% concentrations of the lavage solutions, only acetic acid caused a statistically significant decrease in bacteriophage titers compared to normal saline control. Our findings suggest that tailed Staphylococcal bacteriophages do not remain stable in high concentrations of the most commonly used lavage solutions. However, at very dilute concentrations the bacteriophages do remain viable. This has important clinical ramifications in that it shows when using bacteriophage therapy for PJI it is critical to thoroughly wash out any lavage solutions before the introduction of therapeutic bacteriophages especially when acetic acid is used. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
44. Corrigendum: The trade-off of Vibrio parahaemolyticus between bacteriophage resistance and growth competitiveness
- Author
-
Xiuxiu Zeng, Shanyan Liang, Jiayi Dong, Guosheng Gao, Yaoren Hu, and Yuechao Sun
- Subjects
vibriophage ,bacteriophage therapy ,anti-phage mutant ,bacteriophage resistance ,trade-off ,Microbiology ,QR1-502 - Published
- 2024
- Full Text
- View/download PDF
45. Characterization, genome analysis, and therapeutic evaluation of a novel Salmonella phage vB_SalS_JNS02: a candidate bacteriophage for phage therapy
- Author
-
Lulu Li, Ruining Fan, Yibao Chen, Qing Zhang, Xiaonan Zhao, Ming Hu, Qianghua Lv, Yanbo Luo, Xiaohui Xu, Yumei Cai, and Yuqing Liu
- Subjects
Salmonella Enteritidis, Galleria mellonella ,bacteriophage therapy ,intestinal microbiome ,Animal culture ,SF1-1100 - Abstract
ABSTRACT: Phage therapy is gaining momentum as an alternative to antibiotics in the treatment of salmonellosis caused by Salmonella. In this study, a novel Salmonella phage, vB_SalS_JNS02, was isolated successfully from poultry farms in Shandong, China. The biological characteristics of vB_SalS_JNS02 were analysed, which revealed a short latent period of approximately 10 min and a burst size of 110 PFU/cell. Moreover, vB_SalS_JNS02 exhibited remarkable stability across a wide pH range (pH 3–12) and temperatures ranging from 30 to 80°C. Genome sequencing analysis provided valuable insights into the genetic composition of vB_SalS_JNS02, which consists of a double-stranded DNA genome that spans 42,450 base pairs and has a G + C content of 49.4%. Of significant importance, the genomic sequence of vB_SalS_JNS02 did not contain any genes related to lysogenicity, virulence, or antibiotic resistance. The phage's efficacy was evaluated in a larval challenge study. Treatment with the phage resulted in increased survival of Galleria mellonella larvae (100, 70, and 85%) (MOI 0.1) in the prophylactic treatment, co-infection treatment, and remedial treatment experiments, respectively. Another in vivo experiment investigated the potential application of the phage in broiler chickens and revealed that a single oral dose of vB_SalS_JNS02 (108 PFU/mL, 100 µL/chick) administered 3 h after S. enteritidis oral administration provided effective protection. The introduction of bacteriophage not only enhances the production of secretory immunoglobulin A (sIgA), but also induces alterations in the composition of the gut microbial community. Phage therapy increases the relative abundance of beneficial bacteria, which helps to maintain intestinal barrier homeostasis. However, it is unable to fully restore the disrupted intestinal microbiome caused by S. enteritidis infection. Importantly, no significant adverse effects were observed in the animal subjects following oral administration of the phage, and our findings highlight vB_SalS_JNS02 is a hopeful candidate as a promising tool to target Salmonella infections in poultry.
- Published
- 2024
- Full Text
- View/download PDF
46. Selective bacteriophages reduce the emergence of resistant bacteria in bacteriophage-antibiotic combination therapy
- Author
-
Aa Haeruman Azam, Koji Sato, Kazuhiko Miyanaga, Tomohiro Nakamura, Shinjiro Ojima, Kohei Kondo, Azumi Tamura, Wakana Yamashita, Yasunori Tanji, and Kotaro Kiga
- Subjects
bacteriophage therapy ,O157:H7 ,antimicrobials ,fosfomycin ,drug resistance evolution ,drug resistance mechanisms ,Microbiology ,QR1-502 - Abstract
ABSTRACT Escherichia coli O157:H7 is a globally important foodborne pathogen with implications for food safety. Antibiotic treatment for O157 may potentially contribute to the exacerbation of hemolytic uremic syndrome, and the increasing prevalence of antibiotic-resistant strains necessitates the development of new treatment strategies. In this study, the bactericidal effects and resistance development of antibiotic and bacteriophage monotherapy were compared with those of combination therapy against O157. Experiments involving continuous exposure of O157 to phages and antibiotics, along with genetic deletion studies, revealed that the deletion of glpT and uhpT significantly increased resistance to fosfomycin. Furthermore, we found that OmpC functions as a receptor for the PP01 phage, which infects O157, and FhuA functions as a receptor for the newly isolated SP15 phage, targeting O157. In the glpT and uhpT deletion mutants, additional deletion in ompC, the receptor for the PP01 phage, increased resistance to fosfomycin. These findings suggest that specific phages may contribute to antibiotic resistance by selecting the emergence of gene mutations responsible for both phage and antibiotic resistance. While combination therapy with phages and antibiotics holds promise for the treatment of bacterial infections, careful consideration of phage selection is necessary.IMPORTANCEThe combination treatment of fosfomycin and bacteriophages against Escherichia coli O157 demonstrated superior bactericidal efficacy compared to monotherapy, effectively suppressing the emergence of resistance. However, mutations selected by phage PP01 led to enhanced resistance not only to the phage but also to fosfomycin. These findings underscore the importance of exercising caution in selecting phages for combination therapy, as resistance selected by specific phages may increase the risk of developing antibiotic resistance.
- Published
- 2024
- Full Text
- View/download PDF
47. Increased Innate Immune Susceptibility in Hyperpigmented Bacteriophage-Resistant Mutants of Pseudomonas aeruginosa
- Author
-
Menon, Nitasha D, Penziner, Samuel, Montaño, Elizabeth T, Zurich, Raymond, Pride, David T, Nair, Bipin G, Kumar, Geetha B, and Nizet, Victor
- Subjects
Microbiology ,Medical Microbiology ,Biomedical and Clinical Sciences ,Biological Sciences ,Antimicrobial Resistance ,Emerging Infectious Diseases ,Lung ,Biodefense ,Infectious Diseases ,Genetics ,5.1 Pharmaceuticals ,2.2 Factors relating to the physical environment ,Infection ,Animals ,Anti-Bacterial Agents ,Bacteriophages ,Colistin ,Humans ,Hydrogen Peroxide ,Immunity ,Innate ,Mice ,Pseudomonas Infections ,Pseudomonas Phages ,Pseudomonas aeruginosa ,bacteriophage therapy ,bacteriophage resistance ,pyomelanin ,innate immunity ,Pharmacology and Pharmaceutical Sciences ,Medical microbiology ,Pharmacology and pharmaceutical sciences - Abstract
Bacteriophage (phage) therapy is an alternative to traditional antibiotic treatments that is particularly important for multidrug-resistant pathogens, such as Pseudomonas aeruginosa. Unfortunately, phage resistance commonly arises during treatment as bacteria evolve to survive phage predation. During in vitro phage treatment of a P. aeruginosa-type strain, we observed the emergence of phage-resistant mutants with brown pigmentation that was indicative of pyomelanin. As increased pyomelanin (due to hmgA gene mutation) was recently associated with enhanced resistance to hydrogen peroxide and persistence in experimental lung infection, we questioned if therapeutic phage applications could inadvertently select for hypervirulent populations. Pyomelanogenic phage-resistant mutants of P. aeruginosa PAO1 were selected for upon treatment with three distinct phages. Phage-resistant pyomelanogenic mutants did not possess increased survival of pyomelanogenic ΔhmgA in hydrogen peroxide. At the genomic level, large (~300 kb) deletions in the phage-resistant mutants resulted in the loss of ≥227 genes, many of which had roles in survival, virulence, and antibiotic resistance. Phage-resistant pyomelanogenic mutants were hypersusceptible to cationic peptides LL-37 and colistin and were more easily cleared in human whole blood, serum, and a murine infection model. Our findings suggest that hyperpigmented phage-resistant mutants that may arise during phage therapy are markedly less virulent than their predecessors due to large genomic deletions. Thus, their existence does not present a contraindication to using anti-pseudomonal phage therapy, especially considering that these mutants develop drug susceptibility to the familiar FDA-approved antibiotic, colistin.
- Published
- 2022
48. Safety and Tolerability of ShigActive™, a Shigella spp. Targeting Bacteriophage Preparation, in a Phase 1 Randomized, Double-Blind, Controlled Clinical Trial
- Author
-
Wilbur H. Chen, Joelle Woolston, Silvia Grant-Beurmann, Courtney K. Robinson, Garima Bansal, Joseph Nkeze, Jasnehta Permala-Booth, Claire M. Fraser, Sharon M. Tennant, Mallory C. Shriver, Marcela F. Pasetti, Yuanyuan Liang, Karen L. Kotloff, Alexander Sulakvelidze, and Jennifer A. Schwartz
- Subjects
bacteriophage ,shigella ,bacteriophage therapy ,alternative antibacterials ,microbiome ,safety ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Bacterial diseases of the gastrointestinal (GI) tract continue to be a major worldwide cause of human morbidity and mortality. Among various enteric pathogens, Shigella spp. are some of the most common and deadly bacterial pathogens. They are responsible for ~125 million worldwide cases of shigellosis, and ~14,000 deaths annually, the majority in children under the age of 5 and occurring in developing countries. Preventing and treating shigellosis with conventional drugs (e.g., vaccines and antibiotics) has proven to be very difficult. Here, we assessed the safety and tolerability of ShigActive™, a lytic bacteriophage preparation targeting Shigella spp., in a randomized, placebo-controlled, double-blind Phase 1 clinical trial. Ten participants randomized 4:1 received ShigActive™ or placebo co-administered with sodium bicarbonate orally three times daily for 7 days. Solicited and unsolicited adverse events (AEs) were observed for 29 days. Fifty percent of the subjects receiving ShigActive™ reported mild GI-related symptoms, while one participant experienced moderate fatigue. No serious or medically attended AEs occurred through day 90. Additionally, no significant differences in GI-associated inflammatory mediators or fecal microbiome changes were observed between placebo- and ShigActive™-treated subjects, or from a participants’ baseline value. The results of this first-in-human (FIH) randomized, controlled Phase 1 trial of ShigActive™ demonstrate that it is safe and well tolerated when orally administered with no significant differences compared to placebo controls.
- Published
- 2024
- Full Text
- View/download PDF
49. A New Approach for Phage Cocktail Design in the Example of Anti-Mastitis Solution
- Author
-
Daria Królikowska, Marta Szymańska, Marta Krzyżaniak, Arkadiusz Guziński, Rafał Matusiak, Agnieszka Kajdanek, Edyta Kaczorek-Łukowska, Agnieszka Maszewska, Ewelina A. Wójcik, and Jarosław Dastych
- Subjects
bacteriophage therapy ,bioinformatics-based design ,structural analyses of phages ,antibiotic resistance ,mastitis ,biofilm ,Medicine - Abstract
The studies on phage therapy have shown an overall protective effect of phages in bacterial infections, thus providing an optimistic outlook on the future benefits of phage-based technologies for treating bacterial diseases. However, the therapeutic effect is highly affected by the proper composition of phage cocktails. The rational approach to the design of bacteriophage cocktails, which is the subject of this study, allowed for development of an effective anti-mastitis solution, composed of virulent bacteriophages acting on Escherichia coli and Staphylococcus aureus. Based on the in-depth bioinformatic characterization of bacteriophages and their in vitro evaluation, the cocktail of five phages against E. coli and three against S. aureus strains was composed. Its testing in the milk model experiment revealed a reduction in the number of S. aureus of 45% and 30% for E. coli strains, and in the study of biofilm prevention, it demonstrated 99% inhibition of biofilm formation for all tested S. aureus strains and a minimum of 50% for 50% of E. coli strains. Such insights justify the need for rational design of cocktails for phage therapy and indicate the potential of the developed cocktail in the treatment of diseased animals, but this requires further investigations to evaluate its in vivo efficacy.
- Published
- 2024
- Full Text
- View/download PDF
50. Influence of inhaled bacteriophage therapy on oral mucosal immunity in children with acute tonsillitis
- Author
-
Shokhida T. Turdieva, Durdona K. Ganieva, and Gulmira R. Nasirova
- Subjects
children ,acute tonsillitis ,bacteriophage therapy ,secretory immunoglobulin a ,proinflammatory cytokine ,oral cavity ,Infectious and parasitic diseases ,RC109-216 - Abstract
The study aimed to assess the effect of inhaled bacteriophage therapy on oral mucosal immunity in children with acute tonsillitis. Materials and methods.We examined 212 patients aged 4 to 15 years old with acute tonsillitis and 110 age-matched apparently healthy children. Research methods: calculating the Neutrophil to lymphocyte ratio (NLR), saliva diagnostics — secretory immunoglobulin A (sIgA) and the pro-inflammatory cytokine (TNFα). Taking into account the scheme of the treatment, the patients were divided into mutually comparable groups: the first group included patients with acute tonsillitis who received the standard generally accepted treatment depending on the clinical form of the pathology, without using bacteriophage therapy — n = 107 (50.5%), the second group — patients receiving a course of bacteriophage therapy — n = 105 (49.95%), nebulizer bacteriophage therapy using liquid complex pyobacteriophage (PCL) (Microgen, Russia) from the first days of the disease along with standard treatment.Results.During bacteriophage therapy, on the 6th day of treatment, an increased sIgA level up to 97.2% was observed particularly in younger and adolescent patients up to 97.2% (p ≤ 0.05). At the same time, this parameter reached 75.8% and 81.6%, respectively (p ≤ 0.05), in patients who received only standard treatment. The following difference between the two study groups was observed: between patients in the younger age subgroup — 21.4%, in the older age subgroup -16.1% (p ≤ 0.05 relative to control group), which indicates a more effective drug-related effect in patients from the younger age group groups. Similar changes are observed while analyzing level of the pro-inflammatory cytokine (TNFα). Conclusion.The use of inhaled bacteriophage therapy in the combination treatment of children with acute tonsillitis helps to shorten the period of general and local clinical manifestations of the disease by 1.4-fold and improve mean local immunity from 5.7% up to 16.1% (p ≤ 0.05).
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.