1
|
Yao M, Zhu Y, Duan JA, Xiao P. Phage therapy: A novel approach to combat drug-resistant pathogens. Microbiol Res 2025; 298:128228. [PMID: 40413913 DOI: 10.1016/j.micres.2025.128228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 05/15/2025] [Accepted: 05/18/2025] [Indexed: 05/27/2025]
Abstract
Antibiotic-resistant infections, such as those caused by the overuse of antibiotics, have greatly strained healthcare systems. Among them, drug-resistant bacteria ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) are typical and common. Enterococcus faecalis and Escherichia coli are of equal concern. These pathogens often have higher pathogenicity than the same strains, and resistance has reduced treatment options, so new treatment options are needed to address these pathogens. This review analyzes recent studies related to phage therapy for the treatment of bacterial infections in various parts of the human body (e.g., alcoholic liver disease, skin, and soft tissues, respiratory tract, gastrointestinal tract, urinary system, etc.), to better understand the potential role of phage therapy as a non-antibiotic strategy for the treatment of infections caused by drug-resistant bacteria. In addition, this review introduces a series of products related to phage therapy and points out potential research directions for phage therapy in clinical applications. This paper elucidates the basic mechanism of human infection by some drug-resistant bacteria and the therapeutic effect of phage therapy against drug-resistant bacteria. It popularizes the understanding of phage therapy and provides a reference for research on its use for drug-resistant bacterial infections.
Collapse
Affiliation(s)
- Mengru Yao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Yuan Zhu
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| | - Ping Xiao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| |
Collapse
|
2
|
Blasco L, Bleriot I, Fernández-Grela P, Paño-Pardo JR, Oteo-Iglesias J, Tomás M. Pharmacokinetics and pharmacodynamics studies of phage therapy. FARMACIA HOSPITALARIA 2025:S1130-6343(25)00052-2. [PMID: 40345936 DOI: 10.1016/j.farma.2025.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Accepted: 04/01/2025] [Indexed: 05/11/2025] Open
Abstract
The need for new antimicrobial treatments that work alternatively or synergistically with antibiotics to address the problem of the emergence and transmission of antimicrobial resistance has increased interest in the use of minority therapies such as phage therapy. For safe and widespread application of this therapy, it is necessary to establish the pharmacokinetic and pharmacodynamic parameters for its use in humans. This systematic review analyzes the criteria necessary to establish the PK/PD of this therapy, as well as its current application, based on a review of 66 clinical cases that catch diverse infections and phage administration routes.
Collapse
Affiliation(s)
- Lucía Blasco
- Grupo de Microbiología Traslacional y Multidisciplinar (MicroTM), Instituto de Investigación Biomédica de A Coruña (INIBIC), Departamento de Microbiología, Hospital de A Coruña (CHUAC); Universidad de A Coruña (UDC), A Coruña, España; Grupo de Estudio sobre Mecanismos de Acción y Resistencia a los Antimicrobianos (GEMARA) en Representación de la Sociedad Española de Enfermedades Infecciosas y Microbiología Clínica (SEIMC), A Coruña, España; MePRAM, Proyecto de Medicina de Precisión contra las Resistencias Antimicrobianas, Madrid, España
| | - Inés Bleriot
- Grupo de Microbiología Traslacional y Multidisciplinar (MicroTM), Instituto de Investigación Biomédica de A Coruña (INIBIC), Departamento de Microbiología, Hospital de A Coruña (CHUAC); Universidad de A Coruña (UDC), A Coruña, España; Grupo de Estudio sobre Mecanismos de Acción y Resistencia a los Antimicrobianos (GEMARA) en Representación de la Sociedad Española de Enfermedades Infecciosas y Microbiología Clínica (SEIMC), A Coruña, España; MePRAM, Proyecto de Medicina de Precisión contra las Resistencias Antimicrobianas, Madrid, España
| | - Patricia Fernández-Grela
- Grupo de Microbiología Traslacional y Multidisciplinar (MicroTM), Instituto de Investigación Biomédica de A Coruña (INIBIC), Departamento de Microbiología, Hospital de A Coruña (CHUAC); Universidad de A Coruña (UDC), A Coruña, España; Grupo de Estudio sobre Mecanismos de Acción y Resistencia a los Antimicrobianos (GEMARA) en Representación de la Sociedad Española de Enfermedades Infecciosas y Microbiología Clínica (SEIMC), A Coruña, España; MePRAM, Proyecto de Medicina de Precisión contra las Resistencias Antimicrobianas, Madrid, España
| | - José Ramón Paño-Pardo
- Instituto de Investigación Sanitaria Aragón, Hospital Clínico Universitario Lozano Blesa, Zaragoza, España; CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, España; MePRAM, Proyecto de Medicina de Precisión contra las Resistencias Antimicrobianas, Madrid, España
| | - Jesús Oteo-Iglesias
- Laboratorio de Referencia e Investigación de Resistencias Antibióticas e Infecciones Sanitarias, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Majadahonda, Madrid, España; CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, España; MePRAM, Proyecto de Medicina de Precisión contra las Resistencias Antimicrobianas, Madrid, España
| | - María Tomás
- Grupo de Microbiología Traslacional y Multidisciplinar (MicroTM), Instituto de Investigación Biomédica de A Coruña (INIBIC), Departamento de Microbiología, Hospital de A Coruña (CHUAC); Universidad de A Coruña (UDC), A Coruña, España; Grupo de Estudio sobre Mecanismos de Acción y Resistencia a los Antimicrobianos (GEMARA) en Representación de la Sociedad Española de Enfermedades Infecciosas y Microbiología Clínica (SEIMC), A Coruña, España; MePRAM, Proyecto de Medicina de Precisión contra las Resistencias Antimicrobianas, Madrid, España.
| |
Collapse
|
3
|
Hou X, Zhai L, Fu L, Lu J, Guo P, Zhang Y, Zheng D, Ma G. Advances in Engineered Phages for Disease Treatment. SMALL METHODS 2025; 9:e2401611. [PMID: 39935185 DOI: 10.1002/smtd.202401611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 01/15/2025] [Indexed: 02/13/2025]
Abstract
Phage therapy presents a promising solution for combating multidrug-resistant (MDR) bacterial infections and other bacteria-related diseases, attributed to their innate ability to target and lyse bacteria. Recent clinical successes, particularly in treating MDR-related respiratory and post-surgical infections, validated the therapeutic potential of phage therapy. However, the complex microenvironment within the human body poses significant challenges to phage activity and efficacy in vivo. To overcome these barriers, recent advances in phage engineering have aimed to enhance targeting accuracy, improve stability and survivability, and explore synergistic combinations with other therapeutic modalities. This review provides a comprehensive overview of phage therapy, emphasizing the application of engineered phages in antibacterial therapy, tumor therapy, and vaccine development. Furthermore, the review highlights the current challenges and future trends for advancing phage therapy toward broader clinical applications.
Collapse
Affiliation(s)
- Xiaolin Hou
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
| | - Lin Zhai
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Laiying Fu
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Junna Lu
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
| | - Peilin Guo
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Yu Zhang
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Diwei Zheng
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Guanghui Ma
- State Key Laboratory of Biopharmaceutical Preparation and Delivery Institute of Process Engineering, Chinese Academy of Sciences, Bejing, 100190, P. R. China
- School of Chemical Engineering, University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
4
|
Bleriot I, Blasco L, Fernández-Grela P, Fernández-García L, Armán L, Ibarguren C, Ortiz-Cartagena C, Barrio-Pujante A, Paño JR, Oteo-Iglesias J, Tomás M. Studies in vitro and in vivo of phage therapy medical products (PTMPs) Targeting Clinical Strains of Klebsiella pneumoniae belonging to the clone ST512. Antimicrob Agents Chemother 2025:e0193524. [PMID: 40265927 DOI: 10.1128/aac.01935-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/18/2025] [Indexed: 04/24/2025] Open
Abstract
The widespread incidence of antimicrobial resistance has created renewed interest in the use of alternative antimicrobial treatments such as phage therapy. Phages are viruses that infect bacteria and generally have a narrow bacteria host-range. Combining phages with antibiotics can prevent the emergence of bacterial resistance. The aim of the present study was to develop phage therapy medical products (PTMPs) targeting clinical isolates of carbapenem-producing Klebsiella pneumoniae belonging to the high-risk clone ST512. From a collection of 22 seed of lytic phages sequenced belonging to MePRAM collection (Spanish Health Precision Medicine Project against Antimicrobial Resistance), four were used to generate PTMPs (CAC_Kpn1 and CAC_Kpn2). These PTMPs were partly active against three of the clinical strains of clone ST512 (A, B, and C). The use of Appelmans method in the CAC_Kpn1_ad (adapted CAC_Kpn1) yielded a significant increase in the efficacy against strain A, while adapted CAC_Kpn2 (CAC_Kpn2_ad) only effectively reduced bacterial survival when combined with ½ × MIC ß-lactam antibiotic meropenem for 24 h in clinical strains B and C, showed after this time, resistance to PTMPs. In addition, the amounts of endotoxin released by the PTMPs were quantified and subsequently reduced in preparation for in vivo use of the PTMPs in the Galleria mellonella infection model, confirming the in vitro results from the CAC_Kpn1_ad and CAC_Kpn2_ad. To sum up, the preparation of two PTMPs and their subsequent adaptation can be a good approach to solve part of the occurrence of antimicrobial resistance. In addition, the use of the larval model is an effective method to discriminate the efficacy of in vivo treatment.
Collapse
Affiliation(s)
- Inés Bleriot
- Departamento de Microbiología-Hospital A Coruña (HUAC), Grupo de Microbiología Traslacional y Multidisciplinar (Micro-TM), A Coruña, Spain
- Grupo de Estudio de Mecanismos de Acción y Resistencia a los Antimicrobianos (GEMARA) en nombre de la Sociedad Española de Enfermedades Infecciosas y Microbiología Clínica (SEIMC), Madrid, Spain
| | - Lucía Blasco
- Departamento de Microbiología-Hospital A Coruña (HUAC), Grupo de Microbiología Traslacional y Multidisciplinar (Micro-TM), A Coruña, Spain
- Grupo de Estudio de Mecanismos de Acción y Resistencia a los Antimicrobianos (GEMARA) en nombre de la Sociedad Española de Enfermedades Infecciosas y Microbiología Clínica (SEIMC), Madrid, Spain
| | - Patricia Fernández-Grela
- Departamento de Microbiología-Hospital A Coruña (HUAC), Grupo de Microbiología Traslacional y Multidisciplinar (Micro-TM), A Coruña, Spain
- Grupo de Estudio de Mecanismos de Acción y Resistencia a los Antimicrobianos (GEMARA) en nombre de la Sociedad Española de Enfermedades Infecciosas y Microbiología Clínica (SEIMC), Madrid, Spain
| | - Laura Fernández-García
- Departamento de Microbiología-Hospital A Coruña (HUAC), Grupo de Microbiología Traslacional y Multidisciplinar (Micro-TM), A Coruña, Spain
- Grupo de Estudio de Mecanismos de Acción y Resistencia a los Antimicrobianos (GEMARA) en nombre de la Sociedad Española de Enfermedades Infecciosas y Microbiología Clínica (SEIMC), Madrid, Spain
| | - Lucia Armán
- Departamento de Microbiología-Hospital A Coruña (HUAC), Grupo de Microbiología Traslacional y Multidisciplinar (Micro-TM), A Coruña, Spain
- Grupo de Estudio de Mecanismos de Acción y Resistencia a los Antimicrobianos (GEMARA) en nombre de la Sociedad Española de Enfermedades Infecciosas y Microbiología Clínica (SEIMC), Madrid, Spain
| | - Clara Ibarguren
- Departamento de Microbiología-Hospital A Coruña (HUAC), Grupo de Microbiología Traslacional y Multidisciplinar (Micro-TM), A Coruña, Spain
- Grupo de Estudio de Mecanismos de Acción y Resistencia a los Antimicrobianos (GEMARA) en nombre de la Sociedad Española de Enfermedades Infecciosas y Microbiología Clínica (SEIMC), Madrid, Spain
| | - Concha Ortiz-Cartagena
- Departamento de Microbiología-Hospital A Coruña (HUAC), Grupo de Microbiología Traslacional y Multidisciplinar (Micro-TM), A Coruña, Spain
- Grupo de Estudio de Mecanismos de Acción y Resistencia a los Antimicrobianos (GEMARA) en nombre de la Sociedad Española de Enfermedades Infecciosas y Microbiología Clínica (SEIMC), Madrid, Spain
| | - Antonio Barrio-Pujante
- Departamento de Microbiología-Hospital A Coruña (HUAC), Grupo de Microbiología Traslacional y Multidisciplinar (Micro-TM), A Coruña, Spain
- Grupo de Estudio de Mecanismos de Acción y Resistencia a los Antimicrobianos (GEMARA) en nombre de la Sociedad Española de Enfermedades Infecciosas y Microbiología Clínica (SEIMC), Madrid, Spain
| | - José Ramón Paño
- Hospital Clínico Universitario "Lozano Blesa", Instituto de Investigación Sanitaria Aragón, Zaragoza, Aragon, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Community of Madrid, Spain
- MEPRAM, Proyecto de Medicina de Precisión contra las resistencias Antimicrobianas, Madrid, Spain
| | - Jesús Oteo-Iglesias
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Community of Madrid, Spain
- MEPRAM, Proyecto de Medicina de Precisión contra las resistencias Antimicrobianas, Madrid, Spain
- Laboratorio de Referencia e Investigación de Resistencias Antibióticas e Infecciones Sanitarias, Centro Nacional de Microbiología, Instituto de Salud Carlos III, Madrid, Spain
| | - María Tomás
- Departamento de Microbiología-Hospital A Coruña (HUAC), Grupo de Microbiología Traslacional y Multidisciplinar (Micro-TM), A Coruña, Spain
- MEPRAM, Proyecto de Medicina de Precisión contra las resistencias Antimicrobianas, Madrid, Spain
| |
Collapse
|
5
|
Guo J, Su Z, Zhong J, Li L, An W, Shi B, Xu Y, Qiu C, Chen J, Wang Y, Wen P, Wang J, Li H. Early bacterial pathogen distribution and risk factors for infections after liver transplantation: Retrospective cohort study. Int J Infect Dis 2025; 156:107907. [PMID: 40239757 DOI: 10.1016/j.ijid.2025.107907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025] Open
Abstract
OBJECTIVES To provide a comprehensive analysis of the epidemiological characteristics and clinical features of pathogens and explore the risk factors for post-liver transplant (LT) bacterial infections. METHODS A retrospective analysis was conducted at Xiang'an Hospital of Xiamen University and the First Affiliated Hospital of Zhengzhou University between 2021 and 2024. Binary logistic regression analysis was performed to identify independent risk factors, including clinical characteristics and genetic polymorphisms related to infections within the first month after LT. RESULTS Two hundred forty-two LT donors and recipients were included. Klebsiella pneumoniae (19.0%) was the most common gram-negative bacterium, whereas Staphylococcus aureus (7.1%) was the predominant gram-positive pathogen. A respiratory tract infection was the most common bacterial infection. Binary logistic regression analysis revealed that the recipient SLCO1B1 rs4149015 AA genotype, preoperative hypertension, and low preoperative red blood cell count were independent risk factors for post-LT infection in recipients. CONCLUSION Our analysis revealed the risk factors and clinical manifestations of bacterial infections as well as their polymorphisms. These findings provide valuable insights into the early detection and prevention of bacterial infections, revealing potential avenues for the development of methods to prevent such infections and therefore improve patient prognosis after LT.
Collapse
Affiliation(s)
- Jingjie Guo
- Department of General Surgery, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Organ Transplantation Institute of Xiamen University, Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Zhaojie Su
- Department of General Surgery, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Organ Transplantation Institute of Xiamen University, Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Jianfa Zhong
- Department of General Surgery, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Organ Transplantation Institute of Xiamen University, Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Li Li
- Department of General Surgery, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Organ Transplantation Institute of Xiamen University, Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Wenbin An
- Department of General Surgery, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Organ Transplantation Institute of Xiamen University, Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Baojie Shi
- Department of General Surgery, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Organ Transplantation Institute of Xiamen University, Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Yiran Xu
- Department of General Surgery, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Organ Transplantation Institute of Xiamen University, Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Cheng Qiu
- Department of General Surgery, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Organ Transplantation Institute of Xiamen University, Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Jiajia Chen
- Department of General Surgery, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Organ Transplantation Institute of Xiamen University, Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Ying Wang
- Department of General Surgery, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Organ Transplantation Institute of Xiamen University, Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Peihao Wen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jie Wang
- Department of General Surgery, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Organ Transplantation Institute of Xiamen University, Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China
| | - Hao Li
- Department of General Surgery, Organ Transplantation Clinical Medical Center of Xiamen University, Xiang'an Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China; Organ Transplantation Institute of Xiamen University, Xiamen Human Organ Transplantation Quality Control Center, Xiamen Key Laboratory of Regeneration Medicine, Fujian Provincial Key Laboratory of Organ and Tissue Regeneration, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|
6
|
Kim MK, Suh GA, Cullen GD, Perez Rodriguez S, Dharmaraj T, Chang THW, Li Z, Chen Q, Green SI, Lavigne R, Pirnay JP, Bollyky PL, Sacher JC. Bacteriophage therapy for multidrug-resistant infections: current technologies and therapeutic approaches. J Clin Invest 2025; 135:e187996. [PMID: 40026251 PMCID: PMC11870740 DOI: 10.1172/jci187996] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025] Open
Abstract
Bacteriophage (phage) therapy has emerged as a promising solution to combat the growing crisis of multidrug-resistant (MDR) infections. There are several international centers actively engaged in implementation of phage therapy, and recent case series have reported encouraging success rates in patients receiving personalized, compassionate phage therapy for difficult-to-treat infections. Nonetheless, substantial hurdles remain in the way of more widespread adoption and more consistent success. This Review offers a comprehensive overview of current phage therapy technologies and therapeutic approaches. We first delineate the common steps in phage therapy development, from phage bank establishment to clinical administration, and examine the spectrum of therapeutic approaches, from personalized to fixed phage cocktails. Using the framework of a conventional drug development pipeline, we then identify critical knowledge gaps in areas such as cocktail design, formulation, pharmacology, and clinical trial design. We conclude that, while phage therapy holds promise, a structured drug development pipeline and sustained government support are crucial for widespread adoption of phage therapy for MDR infections.
Collapse
Affiliation(s)
- Minyoung Kevin Kim
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
- Department of Medicine, Yale University, New Haven, Connecticut, USA
| | - Gina A. Suh
- Division of Public Health, Infectious Diseases and Occupational Health, Mayo Clinic College of Medicine, Rochester, Minnesota, USA
| | - Grace D. Cullen
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Saumel Perez Rodriguez
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Tejas Dharmaraj
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Tony Hong Wei Chang
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Zhiwei Li
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Qingquan Chen
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Sabrina I. Green
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Rob Lavigne
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Jean-Paul Pirnay
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| | - Paul L. Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
| | - Jessica C. Sacher
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University, Stanford, California, USA
- Phage Directory, Atlanta, Georgia, USA
| |
Collapse
|
7
|
Xu J, Chen N, Li Z, Liu Y. Gut microbiome and liver diseases. FUNDAMENTAL RESEARCH 2025; 5:890-901. [PMID: 40242515 PMCID: PMC11997574 DOI: 10.1016/j.fmre.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 04/18/2025] Open
Abstract
Symbiotic microbiota plays a crucial role in the education, development, and maintenance of the host immune system, significantly contributing to overall health. Through the gut-liver axis, the gut microbiota and liver have a bidirectional relationship that is becoming increasingly evident as more research highlights the translocation of the gut microbiota and its metabolites. The focus of this narrative review is to examine and discuss the importance of the gut-liver axis and the enterohepatic barrier in maintaining overall health. Additionally, we emphasize the crucial role of the gut microbiome in liver diseases and explore potential therapeutic strategies for liver diseases by manipulating the microbiota.
Collapse
Affiliation(s)
- Jun Xu
- Department of Gastroenterology, Peking University People's Hospital, Beijing 100044, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing 100044, China
| | - Ning Chen
- Department of Gastroenterology, Peking University People's Hospital, Beijing 100044, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing 100044, China
| | - Zhou Li
- Beijing Key Laboratory of Micro-nano Energy and Sensor, Beijing Institute of Nanoenergy and Nanosystems Chinese Academy of Sciences, Beijing 101400, China
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yulan Liu
- Department of Gastroenterology, Peking University People's Hospital, Beijing 100044, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing 100044, China
| |
Collapse
|
8
|
Wang Q, Li X, Ren Y, Hu Q, Xu L, Chen W, Liu J, Wu N, Tao M, Sun J, Xu Y, Shen F. Rapid and precise treatment selection for antimicrobial-resistant infection enabled by a nano-dilution SlipChip. Biosens Bioelectron 2025; 271:117084. [PMID: 39731822 DOI: 10.1016/j.bios.2024.117084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/10/2024] [Accepted: 12/19/2024] [Indexed: 12/30/2024]
Abstract
Antimicrobial resistance (AMR) has become an increasingly severe threat to global health, and AMR-associated infection is one of the leading causes of death around the world. Due to the long turnaround time and the limited flexibility and availability of current antimicrobial susceptibility testing (AST) methods, a large portion of patients with bacterial infections are still treated empirically, increasing the risk of mistreatment. To address the demand for precision treatment of bacterial infections, we developed a nano-dilution SlipChip (nd-SlipChip)-based systematic evaluation method, which facilitates rapid, logic feedback for the assessment of antibiotics, antibiotic combinations, and phage therapy. The nd-SlipChip can conveniently generate a microdroplet array with serially diluted antibiotics and determine the minimal inhibitory concentration (MIC) within 2 h by monitoring bacterial growth profiles, facilitating timely treatment selection. We demonstrated this method for 24 clinical isolates, including gram-negative bacteria (Escherichia coli, Klebsiella pneumoniae, Acinetobacter baumannii) and gram-positive bacteria (Staphylococcus aureus). Three clinical isolates performed the second-stage antibiotic combinations screening, and effective combination therapies were identified for two of them. The remaining isolate proceeded to third-stage phage screening, where suitable phages were successfully selected. The nd-SlipChip provides a rapid and systematic approach for the precise selection of therapies targeting bacterial infections.
Collapse
Affiliation(s)
- Qi Wang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China; School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Xiang Li
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Yan'an Ren
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Qin Hu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Lei Xu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China
| | - Weiling Chen
- Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China
| | - Jianfang Liu
- CreatiPhage Biotechnology Co., Ltd, Shanghai, 200030, China
| | - Nannan Wu
- Shanghai Institute of Phage, Shanghai Public Health Clinical Center, Fudan University, Shanghai, 200433, China; CreatiPhage Biotechnology Co., Ltd, Shanghai, 200030, China
| | - Meifeng Tao
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jingyong Sun
- Department of Laboratory Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Yumin Xu
- Department of Infectious Diseases, Department of Hospital Infection Management, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Feng Shen
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, 200030, China; Zhengzhou Industrial Technology Research Institute of Shanghai Jiao Tong University, Zhengzhou, 450016, China.
| |
Collapse
|
9
|
Hickson SM, Ledger EL, Wells TJ. Emerging antimicrobial therapies for Gram-negative infections in human clinical use. NPJ ANTIMICROBIALS AND RESISTANCE 2025; 3:16. [PMID: 40016340 PMCID: PMC11868545 DOI: 10.1038/s44259-025-00087-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/13/2025] [Indexed: 03/01/2025]
Abstract
The growing problem of multi-drug resistance (MDR) is prevalent in Gram-negative infections, and the significant decline in antibiotic development poses a critical threat to global public health. Many emerging non-antibiotic therapies have been proposed, including phage therapy, anti-virulence agents, antimicrobial peptides, plasmapheresis, and immunotherapy options. To identify the therapies most likely to be the next immediate step in treatment for MDR Gram-negative infections, this review highlights emerging therapeutics that have either been successfully used for compassionate care or are currently undergoing clinical trials.
Collapse
Affiliation(s)
- Sarah M Hickson
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Emma L Ledger
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Timothy J Wells
- Frazer Institute, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
10
|
Chang TH, Pourtois JD, Haddock NL, Furkuawa D, Kelly KE, Amanatullah DF, Burgener E, Milla C, Banaei N, Bollyky PL. Prophages are Infrequently Associated With Antibiotic Resistance in Pseudomonas aeruginosa Clinical Isolates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.06.02.595912. [PMID: 38895396 PMCID: PMC11185549 DOI: 10.1101/2024.06.02.595912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Lysogenic bacteriophages can integrate their genome into the bacterial chromosome in the form of a prophage and can promote genetic transfer between bacterial strains in vitro . However, the contribution of lysogenic phages to the incidence of antimicrobial resistance (AMR) in clinical settings is poorly understood. Here, in a set of 186 clinical isolates of Pseudomonas aeruginosa collected from respiratory cultures from 82 patients with cystic fibrosis (CF), we evaluate the links between prophage counts and both genomic and phenotypic resistance to six anti-pseudomonal antibiotics: tobramycin, colistin, ciprofloxacin, meropenem, aztreonam, and piperacillin-tazobactam. We identified 239 different prophages in total. We find that P. aeruginosa isolates contain on average 3.06 +/- 1.84 (SD) predicted prophages. We find no significant association between the number of prophages per isolate and the minimum inhibitory concentration (MIC) for any of these antibiotics. We then investigate the relationship between particular prophages and AMR. We identify a single lysogenic phage associated with phenotypic resistance to the antibiotic tobramycin and, consistent with this association, we observe that AMR genes associated with resistance to tobramycin are more likely to be found when this prophage is present. However we find that they are not encoded directly on prophage sequences. Additionally, we identify a single prophage statistically associated with ciprofloxacin resistance but do not identify any genes associated with ciprofloxacin phenotypic resistance. These findings suggest that prophages are only infrequently associated with the AMR genes in clinical isolates of P. aeruginosa . Importance Antibiotic-resistant infections of Pseudomonas aeruginosa , a leading pathogen in patients with Cystic Fibrosis (CF) are a global health threat. While lysogenic bacteriophages are known to facilitate horizontal gene transfer, their role in promoting antibiotic resistance in clinical settings remains poorly understood. In our analysis of 186 clinical isolates of P. aeruginosa from CF patients, we find that prophage abundance does not predict phenotypic resistance to key antibiotics but that specific prophages are infrequently associated with tobramycin resistance genes. In addition, we do not find antimicrobial resistance (AMR) genes encoded directly on prophages. These results highlight that while phages can be associated with AMR, phage-mediated AMR transfer may be rare in clinical isolates and difficult to identify. This work is important for future efforts on mitigating AMR in Cystic Fibrosis and other vulnerable populations affected by Pseudomonas aeruginosa infections and advances our understanding of bacterial-phage dynamics in clinical infections.
Collapse
|
11
|
Rahman MA, Verma KK, Posa MK. Phage Therapy in Bacterial Pneumonia Models: A Systematic Review and Meta-Analysis. Comb Chem High Throughput Screen 2025; 28:447-452. [PMID: 38357944 DOI: 10.2174/0113862073267755240126111628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 01/04/2024] [Accepted: 01/08/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Phage therapy could play an important role in the bacterial pneumonia. However, the exact role of phage therapy in bacterial pneumonia is unclear to date. AIM The current study aims to find out the role of phage therapy in preclinical models of bacterial pneumonia. METHODS The studies were searched in databases with proper MeSH terms along with Boolean operators and selected based on eligibility criteria as per the PRISMA guidelines. The Odd Ratio (OR) was calculated with a 95% confidence interval and the heterogeneity was also calculated. The funnel plot was used to conduct a qualitative examination of publication bias. RESULTS The OR was observed to be 0.11 (0.04, 0.27)] after 24 hrs, 0.11 [0.03, 0.34] after 7 days and 0.04 [0.01, 0.15] after 10 days that showed a significant role of phage therapy in reduction of deaths in the bacterial pneumonia models as compared to the placebo group. However, after 48hrs, a non-significant reduction was observed. CONCLUSION There was a significant role of phage therapy in the reduction of deaths in the bacterial pneumonia models.
Collapse
Affiliation(s)
- Mohammad Akhlaquer Rahman
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Taif University, Taif, 21944, Kingdom of Saudi Arabia
| | | | - Mahesh Kumar Posa
- School of Pharmaceutical Sciences, Jaipur National University, Jagatpura, Jaipur, Rajasthan, 302017, India
| |
Collapse
|
12
|
Gil-Gil T, Laborda P, Martínez JL, Hernando-Amado S. Use of adjuvants to improve antibiotic efficacy and reduce the burden of antimicrobial resistance. Expert Rev Anti Infect Ther 2025; 23:31-47. [PMID: 39670956 DOI: 10.1080/14787210.2024.2441891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/28/2024] [Accepted: 12/10/2024] [Indexed: 12/14/2024]
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.
Collapse
Affiliation(s)
- Teresa Gil-Gil
- Department of Biology, Emory University, Atlanta, GA, USA
| | - Pablo Laborda
- Department of Clinical Microbiology 9301, Rigshospitalet, Copenhagen, Denmark
| | | | | |
Collapse
|
13
|
Yue H, Li Y, Yang T, Wang Y, Bao Q, Xu Y, Liu X, Miao Y, Yang M, Mao C. Filamentous phages as tumour-targeting immunotherapeutic bionanofibres. NATURE NANOTECHNOLOGY 2025; 20:167-176. [PMID: 39468354 DOI: 10.1038/s41565-024-01800-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 09/04/2024] [Indexed: 10/30/2024]
Abstract
Programmed cell death-ligand 1 (PD-L1) blockers have advanced immunotherapy, but their lack of tumour homing capability represents a substantial challenge. Here we show that genetically engineered filamentous phages can be used as tumour-targeting immunotherapeutic agents that reduce the side effects caused by untargeted delivery of PD-L1 blockers. Specifically, we improved biopanning to discover a peptide binding the extracellular domain of PD-L1 and another targeting both melanoma tissues and cancer cells. The two peptides were genetically fused to the sidewall protein and tip protein of fd phages, respectively. The intravenously injected phages homed to tumours and bound PD-L1 on cancer cells, effectively blocking PD-1/PD-L1 recognition to trigger targeted immunotherapy without body weight loss, organ abnormalities and haematological aberrations. The phages, cost-effectively replicated by bacteria, are cancer-targeting immunotherapeutic nanofibres that can be flexibly designed to target different cancer types and immune checkpoints.
Collapse
Affiliation(s)
- Hui Yue
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, P. R. China
| | - Yan Li
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Hangzhou, P. R. China
| | - Tao Yang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, P. R. China
| | - Yecheng Wang
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, P. R. China
| | - Qing Bao
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, P. R. China
| | - Yajing Xu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, P. R. China
| | - Xiangyu Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, P. R. China
| | - Yao Miao
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, P. R. China
| | - Mingying Yang
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Hangzhou, P. R. China
| | - Chuanbin Mao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, P. R. China.
| |
Collapse
|
14
|
Liu H, Gu W, Lu Y, Ding L, Guo Y, Zou G, Wu W, Zheng D, Liu C, Wang C, Cao Y, Li J. Exploration of Phage-Agrochemical Interaction Based on a Novel Potent Phage LPRS20-Targeting Ralstonia solanacearum. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:28005-28018. [PMID: 39360931 DOI: 10.1021/acs.jafc.4c03799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Phage therapy has the potential to alleviate plant bacterial wilt. However, the knowledge gap concerning the phage-agrochemical interaction impedes the broader application of phages in agriculture. This study characterized a phage isolate and investigated its interactions with agrochemicals. A novel species within the Ampunavirus genus was proposed, serving phage LPRS20 as a type phage with a broad lytic range and significant antibacterial activity against Ralstonia solanacearum strains infecting tobacco, chili, or tomato. Sensory evaluation of the morphology of tobacco leaves suggested that phage application resulted in negligible harm to plants. Investigations into phage-agrochemical interactions revealed synergisms when LPRS20 was delivered 4 h before thiodiazole-copper as well as LPRS20 in combination with low-concentration berberine. Overall, our findings reveal that phage LPRS20 represents a novel, effective, and eco-friendly biocontrol agent against tobacco bacterial wilt in vivo and in vitro and contributes to the potential integration of phages and agrochemicals for controlling soil-borne pathogens.
Collapse
Affiliation(s)
- Huai Liu
- Key Laboratory of Plant Nutrition and Fertilizer in South Region, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Nutrient Cycling and Farmland Conservation, Guangdong Engineering Research Center of Soil Microbes and Cultivated Land Conservation, Institute of Agricultural Resources and Environment, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Key Laboratory of Environment Correlative Dietology, College of Biomedicine and Health, College of Food Science and Technology, College of Life Science and Technology, College of Veterinary Medicine, Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan 430070, China
| | - Wenjie Gu
- Key Laboratory of Plant Nutrition and Fertilizer in South Region, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Nutrient Cycling and Farmland Conservation, Guangdong Engineering Research Center of Soil Microbes and Cultivated Land Conservation, Institute of Agricultural Resources and Environment, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Yusheng Lu
- Key Laboratory of Plant Nutrition and Fertilizer in South Region, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Nutrient Cycling and Farmland Conservation, Guangdong Engineering Research Center of Soil Microbes and Cultivated Land Conservation, Institute of Agricultural Resources and Environment, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Lili Ding
- Agricultural Science and Technology Research Center of Chaozhou, Chaozhou 521000, China
| | - Yating Guo
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Key Laboratory of Environment Correlative Dietology, College of Biomedicine and Health, College of Food Science and Technology, College of Life Science and Technology, College of Veterinary Medicine, Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan 430070, China
| | - Geng Zou
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Key Laboratory of Environment Correlative Dietology, College of Biomedicine and Health, College of Food Science and Technology, College of Life Science and Technology, College of Veterinary Medicine, Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan 430070, China
| | - Weiqing Wu
- College of Food Science, Guangdong Pharmaceutical University, Zhongshan 528458, China
| | - Diyuan Zheng
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Key Laboratory of Environment Correlative Dietology, College of Biomedicine and Health, College of Food Science and Technology, College of Life Science and Technology, College of Veterinary Medicine, Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan 430070, China
| | - Chong Liu
- Key Laboratory of Plant Nutrition and Fertilizer in South Region, Ministry of Agriculture and Rural Affairs, Guangdong Key Laboratory of Nutrient Cycling and Farmland Conservation, Guangdong Engineering Research Center of Soil Microbes and Cultivated Land Conservation, Institute of Agricultural Resources and Environment, Guangdong Academy of Agricultural Sciences, Guangzhou 510640, China
| | - Chenyang Wang
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510640, China
| | - Yangrong Cao
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Key Laboratory of Environment Correlative Dietology, College of Biomedicine and Health, College of Food Science and Technology, College of Life Science and Technology, College of Veterinary Medicine, Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan 430070, China
| | - Jinquan Li
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Key Laboratory of Environment Correlative Dietology, College of Biomedicine and Health, College of Food Science and Technology, College of Life Science and Technology, College of Veterinary Medicine, Shenzhen Institute of Nutrition and Health, Huazhong Agricultural University, Wuhan 430070, China
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomes Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, China
| |
Collapse
|
15
|
Washizaki A, Sakiyama A, Ando H. Phage-specific antibodies: are they a hurdle for the success of phage therapy? Essays Biochem 2024; 68:633-644. [PMID: 39254211 PMCID: PMC11652166 DOI: 10.1042/ebc20240024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 09/11/2024]
Abstract
Phage therapy has attracted attention again owing to the increasing number of drug-resistant bacteria. Although the efficacy of phage therapy has been reported, numerous studies have indicated that the generation of phage-specific antibodies resulting from phage administration might have an impact on clinical outcomes. Phage-specific antibodies promote phage uptake by macrophages and contribute to their rapid clearance from the body. In addition, phage-specific neutralizing antibodies bind to the phages and diminish their antibacterial activity. Thus, phage-specific antibody production and its role in phage therapy have been analyzed both in vitro and in vivo. Strategies for prolonging the blood circulation time of phages have also been investigated. However, despite these efforts, the results of clinical trials are still inconsistent, and a consensus on whether phage-specific antibodies influence clinical outcomes has not yet been reached. In this review, we summarize the phage-specific antibody production during phage therapy. In addition, we introduce recently performed clinical trials and discuss whether phage-specific antibodies affect clinical outcomes and what we can do to further improve phage therapy regimens.
Collapse
Affiliation(s)
- Ayaka Washizaki
- Laboratory of Phage Biologics, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu City, Gifu 501-1194, Japan
| | - Arata Sakiyama
- Laboratory of Phage Biologics, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu City, Gifu 501-1194, Japan
| | - Hiroki Ando
- Laboratory of Phage Biologics, Graduate School of Medicine, Gifu University, 1-1 Yanagido, Gifu City, Gifu 501-1194, Japan
- Center for One Medicine Innovative Translational Research (COMIT), Institute for Advanced Study, Gifu University, 1-1 Yanagido, Gifu City, Gifu 501-1194, Japan
- Venture Unit Engineered Phage Therapy, Discovery Accelerator, Astellas Pharma Inc., Tsukuba City, Ibaraki 305-8585, Japan
| |
Collapse
|
16
|
Pye HV, Krishnamurthi R, Cook R, Adriaenssens EM. Phage diversity in One Health. Essays Biochem 2024; 68:607-619. [PMID: 39475220 PMCID: PMC12055037 DOI: 10.1042/ebc20240012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 12/18/2024]
Abstract
One Health aims to bring together human, animal, and environmental research to achieve optimal health for all. Bacteriophages (phages) are viruses that kill bacteria and their utilisation as biocontrol agents in the environment and as therapeutics for animal and human medicine will aid in the achievement of One Health objectives. Here, we assess the diversity of phages used in One Health in the last 5 years and place them in the context of global phage diversity. Our review shows that 98% of phages applied in One Health belong to the class Caudoviricetes, compared to 85% of sequenced phages belonging to this class. Only three RNA phages from the realm Riboviria have been used in environmental biocontrol and human therapy to date. This emphasises the lack in diversity of phages used commercially and for phage therapy, which may be due to biases in the methods used to both isolate phages and select them for applications. The future of phages as biocontrol agents and therapeutics will depend on the ability to isolate genetically novel dsDNA phages, as well as in improving efforts to isolate ssDNA and RNA phages, as their potential is currently undervalued. Phages have the potential to reduce the burden of antimicrobial resistance, however, we are underutilising the vast diversity of phages present in nature. More research into phage genomics and alternative culture methods is required to fully understand the complex relationships between phages, their hosts, and other organisms in the environment to achieve optimal health for all.
Collapse
Affiliation(s)
- Hannah V Pye
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
- Centre for Microbial Interactions, Norwich Research Park, Norwich, NR4 7UG, UK
| | - Revathy Krishnamurthi
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
- Centre for Microbial Interactions, Norwich Research Park, Norwich, NR4 7UG, UK
| | - Ryan Cook
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
- Centre for Microbial Interactions, Norwich Research Park, Norwich, NR4 7UG, UK
| | - Evelien M Adriaenssens
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
- Centre for Microbial Interactions, Norwich Research Park, Norwich, NR4 7UG, UK
| |
Collapse
|
17
|
Wang Q, Chen R, Liu H, Liu Y, Li J, Wang Y, Jin Y, Bai Y, Song Z, Lu X, Wang C, Hao Y. Isolation and characterization of lytic bacteriophage vB_KpnP_23: A promising antimicrobial candidate against carbapenem-resistant Klebsiella pneumoniae. Virus Res 2024; 350:199473. [PMID: 39332682 PMCID: PMC11474366 DOI: 10.1016/j.virusres.2024.199473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/08/2024] [Accepted: 09/21/2024] [Indexed: 09/29/2024]
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.
Collapse
Affiliation(s)
- Qian Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Ran Chen
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Hui Liu
- Department of Clinical Laboratory, Maternal and Child Health Care Hospital of Zaozhuang, Zaozhuang, Shandong, 277100, China
| | - Yue Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Jinmei Li
- Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo college of Medicine, Shandong University, Jinan, Shandong, China; Department of Clinical Laboratory, Jinan Seventh People's Hospital, Jinan, Shandong, 250021, China
| | - Yueling Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yan Jin
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Yuanyuan Bai
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhen Song
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xinglun Lu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Changyin Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China.
| | - Yingying Hao
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China; Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo college of Medicine, Shandong University, Jinan, Shandong, China.
| |
Collapse
|
18
|
Wen T, Zhao Y, Fu Y, Chen Y, Li X, Shi C, Xian D, Zhao W, Yang D, Lu C, Wu C, Pan X, Quan G. "On-demand" nanosystem-integrated microneedles for amplified triple therapy against recalcitrant bacteria and biofilm growth. Mater Today Bio 2024; 29:101327. [PMID: 39582781 PMCID: PMC11585702 DOI: 10.1016/j.mtbio.2024.101327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/13/2024] [Accepted: 11/05/2024] [Indexed: 11/26/2024] Open
Abstract
Phototherapy has emerged to eradicate recalcitrant bacteria without causing drug resistance, but it is often accompanied by considerable limitations owing to a high tolerance of recalcitrant bacteria to heat and oxidative damage, leading to low efficiency of monotherapy and unwanted side effects. Assuming that employing antimicrobial peptides (AMPs) to disrupt bacterial membranes could reduce bacterial tolerance, a multifunctional "on-demand" nanosystem based on zeolitic imidazolate framework-8 (ZIF-8) with metal ions for intrinsic antibacterial activity was constructed to potently kill methicillin-resistant Staphylococcus aureus (MRSA). Then, microneedles (MNs) were used to transdermally deliver the ZIF-8-based nanosystem for localized skin infection. After MNs insertion, the nanoplatform could specifically deliver the loaded therapeutic components to bacterial infection sites through employing hyaluronic acid (HA) as a capping agent, thus realizing the "on-demand" payload release triggered by excess hyaluronidase secreted by MRSA. The prepared nanosystem and MNs were confirmed to exert an amplified triple therapy originating from membranolytic effect, phototherapy, and ion therapy, thus displaying a powerful bactericidal and MRSA biofilm destruction ability. This intelligent antimicrobial strategy may bring a dawn of hope for eradicating multidrug-resistant bacteria and biofilms.
Collapse
Affiliation(s)
- Ting Wen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yiting Zhao
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yanping Fu
- College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Ying Chen
- College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Xiaodie Li
- College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Chaonan Shi
- College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Dongyi Xian
- College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Wanchen Zhao
- College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Dan Yang
- College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Chao Lu
- College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Chuanbin Wu
- College of Pharmacy, Jinan University, Guangzhou, 511443, China
| | - Xin Pan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Guilan Quan
- College of Pharmacy, Jinan University, Guangzhou, 511443, China
| |
Collapse
|
19
|
Sawa T, Moriyama K, Kinoshita M. Current status of bacteriophage therapy for severe bacterial infections. J Intensive Care 2024; 12:44. [PMID: 39482746 PMCID: PMC11529433 DOI: 10.1186/s40560-024-00759-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 10/23/2024] [Indexed: 11/03/2024] Open
Abstract
The increase in the incidence of antibiotic-resistant bacteria poses a global public health threat. According to a 2019 WHO report, approximately 1.27 million deaths were attributed to antibiotic-resistant bacteria, with many cases linked to specific bacterial species, such as drug-resistant Pseudomonas aeruginosa and Staphylococcus aureus. By 2050, the number of deaths caused by these bacteria is predicted to surpass that caused by cancer. In response to this serious situation, phage therapy, an alternative to antibiotic treatment, has gained attention. Phage therapy involves the use of viruses that target specific bacteria to treat infections. This method has proven effective in multiple clinical cases, particularly for patients with severe infections caused by multidrug-resistant bacteria. For example, there are reports of patients with systemic infections caused by multidrug-resistant Acinetobacter who recovered following phage administration and patients infected with panresistant Pseudomonas aeruginosa who were cured by phage therapy. A key feature of phage therapy is its high specificity. Phages infect only specific bacteria and eliminate them. However, this specificity can also be a disadvantage, as careful selection of the appropriate phage for the target bacteria is needed. Additionally, bacteria can develop resistance to phages, potentially reducing treatment effectiveness over time. Efforts are underway to select, combine, and improve phages to address these challenges. In Belgium, a national phage bank has been established, and in the United States, the University of California, San Diego, has founded Innovative Phage Applications and Therapeutics (IPATH), marking significant progress toward the clinical application of phage therapy in the country. As a result, phage therapy is emerging as a component of personalized medicine, offering a new treatment option against antibiotic-resistant bacteria. The clinical application of phage therapy is particularly important in life-saving treatments for patients with severe bacterial infections, and its use in conjunction with antibiotics could enhance therapeutic outcomes. Continued research and development of this therapy could provide hope for many more patients in the future.
Collapse
Affiliation(s)
- Teiji Sawa
- University Hospital, Kyoto Prefectural University of Medicine, 465 Kajiicho, Kawaramachi-Hirokoji-Agaru, Kamigyo, Kyoto, 602-8566, Japan.
| | - Kiyoshi Moriyama
- Department of Anesthesiology, School of Medicine, Kyorin University, Mitaka, Japan
| | - Mao Kinoshita
- Department of Anesthesiology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
20
|
Köhne M, Hüsch R, Tönissen A, Schmidt M, Müsken M, Böttcher D, Hirnet J, Plötz M, Kittler S, Sieme H. Isolation and characterization of bacteriophages specific to Streptococcus equi subspecies zooepidemicus and evaluation of efficacy ex vivo. Front Microbiol 2024; 15:1448958. [PMID: 39529671 PMCID: PMC11550937 DOI: 10.3389/fmicb.2024.1448958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Streptococcus (S.) equi subspecies (subsp.) zooepidemicus is an important facultative pathogen in horses and can cause severe infections in other species including humans. Facing the post-antibiotic era, novel antimicrobials are needed for fighting bacterial infections. Bacteriophages (phages) are the natural predators of bacteria and discussed as a promising antimicrobial treatment option. The objective of this study was to isolate and characterize S. equi subsp. zooepidemicus-specific phages for the first time and to evaluate their efficacy in vitro and ex vivo. In total, 13 phages with lytic activity were isolated and host ranges were determined. Two phages with broad host ranges and high efficiency of plating (vB_SeqZP_LmqsRe26-2 (lytic activity: 30/37 bacterial isolates) and vB_SeqZP_LmqsRe26-3 (lytic activity: 29/37 bacterial isolates)) and one phage with relatively low efficiency of plating (vB_SeqZP_LmqsRe26-1) were selected for further characterization, including electron microscopy and whole genome sequencing. In in vitro planktonic killing assays at two tested multiplicities of infection (MOI 1 and MOI 10), significant bacterial growth reduction was observed when the phages vB_SeqZP_LmqsRe26-2 and vB_SeqZP_LmqsRe26-3 were added. These phages were subsequently co-incubated with clinical S. equi subsp. zooepidemicus isolates in an equine endometrial explant model but did not achieve bacterial growth reduction at MOI 1 and MOI 10. However, helium ion microscopy revealed presence of particles adherent to the bacteria on the explant after incubation (25 h), suggesting possible phage-bacteria interactions. In conclusion, phages against S. equi subsp. zooepidemicus were successfully isolated and characterized. Promising results were observed in in vitro but no significant reduction was detected in ex vivo experiments, requiring additional investigations. However, after further adaptations (e.g., optimization of MOIs and phage administration or use of phage-antibiotic combination), phages could be a potential antimicrobial tool for future therapeutic use in S. equi subsp. zooepidemicus infections, although the available results do not currently support the therapeutic usage.
Collapse
Affiliation(s)
- Martin Köhne
- Unit for Reproductive Medicine – Clinic for Horses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Ronja Hüsch
- Unit for Reproductive Medicine – Clinic for Horses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Anna Tönissen
- Unit for Reproductive Medicine – Clinic for Horses, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Matthias Schmidt
- Department of Technical Biogeochemistry, Helmholtz Centre for Environmental Research –UFZ, Leipzig, Germany
| | - Mathias Müsken
- Central Facility for Microscopy, Helmholtz Centre for Infection Research – HZI, Braunschweig, Germany
| | - Denny Böttcher
- Institute for Veterinary Pathology, Faculty of Veterinary Medicine, Leipzig University, Leipzig, Germany
| | - Juliane Hirnet
- Institute of Food Quality and Food Safety, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Madeleine Plötz
- Institute of Food Quality and Food Safety, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Sophie Kittler
- Institute of Food Quality and Food Safety, University of Veterinary Medicine, Foundation, Hannover, Germany
| | - Harald Sieme
- Unit for Reproductive Medicine – Clinic for Horses, University of Veterinary Medicine, Foundation, Hannover, Germany
| |
Collapse
|
21
|
Olorundare OO, Zrelovs N, Kabantiyok D, Svanberga K, Jansons J, Kazaks A, Agada GO, Agu CG, Morenikeji OR, Oluwapelumi OA, Dung T, Pewan SB. Isolation and Characterization of a Novel Jumbo Phage HPP-Temi Infecting Pseudomonas aeruginosa Pa9 and Increasing Host Sensitivity to Ciprofloxacin. Antibiotics (Basel) 2024; 13:1006. [PMID: 39596701 PMCID: PMC11591403 DOI: 10.3390/antibiotics13111006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/29/2024] Open
Abstract
Pseudomonas aeruginosa is a bacteria responsible for many hospital-acquired infections. Phages are promising alternatives for treating P. aeruginosa infections, which are often intrinsically resistant. The combination of phage and antibiotics in clearing bacterial infection holds promise due to increasing reports of enhanced effectiveness when both are used together. The aim of the study is to isolate and characterize a novel P. aeruginosa phage and determine its effectiveness in in vitro combination with antibiotics in controlling P. aeruginosa. In this study, a novel jumbo myophage HPP-Temi infecting P. aeruginosa Pa9 (PP334386) was isolated from household sewage. Electron micrographs of the phage were obtained to determine the morphological features of HPP-Temi virions. Complete genome analysis and a combination of Pseudomonas phage HPP-Temi with antibiotics were examined. The phage HPP-Temi was able to productively infect P. aeruginosa ATCC 9027 but was unable to infect a closely related genus. The phage was stable at 4-37 °C, 0.5% NaCl, and pH 8 for at least one hour. The HPP-Temi genome is a 302,719-bp-long dsDNA molecule with a GC content of 46.46%. The genome was predicted to have 436 ORFs and 7 tRNA genes. No virulence factor-related genes, antimicrobial resistance, or temperate lifestyle-associated genes were found in the phage HPP-Temi genome. Phage HPP-Temi is most closely related to the known or tentative representatives of the Pawinskivirus genus and can be proposed as a representative for the creation of a novel phage species in that genus. The phage and antibiotics (Ciprofloxacin) combination at varying phage titers (103, 106, 109) were used against P. aeruginosa Pa9 (PP334386) at 3.0 × 108 CFU/mL, which was carried out in triplicate. The result showed that combining antibiotics with phage significantly reduced the bacteria count at 103 and 106 titers, while no growth was observed at 109 PFU/mL. This suggests that the effect of phage HPP-Temi in combination with antibiotics is a potential and promising agent for the control of P. aeruginosa infections.
Collapse
Affiliation(s)
- Olufunke Olufunmilola Olorundare
- Department of Medical Microbiology, Clinical Sciences, University of Jos, Jos 930105, Nigeria
- Forest Research Institute of Nigeria, Federal College of Forestry, Jos 930105, Nigeria
| | - Nikita Zrelovs
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (N.Z.); (K.S.); (J.J.); (A.K.)
| | - Dennis Kabantiyok
- Fleming Sentinel Lab for AMR, National Veterinary Research Institute NVRI, Vom 930001, Nigeria; (D.K.); (G.O.A.); (O.A.O.); (T.D.)
| | - Karina Svanberga
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (N.Z.); (K.S.); (J.J.); (A.K.)
| | - Juris Jansons
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (N.Z.); (K.S.); (J.J.); (A.K.)
| | - Andris Kazaks
- Latvian Biomedical Research and Study Centre, LV-1067 Riga, Latvia; (N.Z.); (K.S.); (J.J.); (A.K.)
| | - Godwin Ojonugwa Agada
- Fleming Sentinel Lab for AMR, National Veterinary Research Institute NVRI, Vom 930001, Nigeria; (D.K.); (G.O.A.); (O.A.O.); (T.D.)
| | - Chibuzor Gerald Agu
- Bacteria Research National Veterinary Research Institute NVRI, Vom 930001, Nigeria;
| | - Oluwatoyin Ruth Morenikeji
- West African Centre for Emerging Infectious Diseases, Jos University Teaching Hospital, Jos 930241, Nigeria;
| | - Ogundeji Alice Oluwapelumi
- Fleming Sentinel Lab for AMR, National Veterinary Research Institute NVRI, Vom 930001, Nigeria; (D.K.); (G.O.A.); (O.A.O.); (T.D.)
| | - Thomas Dung
- Fleming Sentinel Lab for AMR, National Veterinary Research Institute NVRI, Vom 930001, Nigeria; (D.K.); (G.O.A.); (O.A.O.); (T.D.)
| | | |
Collapse
|
22
|
Echterhof A, Dharmaraj T, Khosravi A, McBride R, Miesel L, Chia JH, Blankenberg PM, Lin KY, Shen CC, Lee YL, Yeh YC, Liao WT, Blankenberg FG, Dąbrowska K, Amanatullah DF, Frymoyer AR, Bollyky PL. The contribution of neutrophils to bacteriophage clearance and pharmacokinetics in vivo. JCI Insight 2024; 9:e181309. [PMID: 39435664 PMCID: PMC11530120 DOI: 10.1172/jci.insight.181309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 09/10/2024] [Indexed: 10/23/2024] Open
Abstract
With the increasing prevalence of antimicrobial-resistant bacterial infections, there is interest in using bacteriophages (phages) to treat such infections. However, the factors that govern bacteriophage pharmacokinetics in vivo remain poorly understood. Here, we have examined the contribution of neutrophils, the most abundant phagocytes in the body, to the pharmacokinetics of i.v. administered bacteriophage in uninfected mice. A single dose of LPS-5, a bacteriophage recently used in human clinical trials to treat drug-resistant Pseudomonas aeruginosa, was administered i.v. to both immunocompetent BALB/c and neutropenic CD1 mice. Phage concentrations were assessed in peripheral blood and spleen at 0.25, 1, 2, 4, 8, 12, and 24 hours after administration by plaque assay and qPCR. We observed that the phage clearance was only minimally affected by neutropenia. Indeed, the half-lives of phages in blood in BALB/c and CD1 mice were 3.45 and 3.66 hours, respectively. These data suggest that neutrophil-mediated phagocytosis is not a major determinant of phage clearance. Conversely, we observed a substantial discrepancy in circulating phage levels over time when measured by qPCR versus plaque assay, suggesting that significant inactivation of circulating phages occurs over time. These data indicate that alternative factors, but not neutrophils, inactivate i.v. administered phages.
Collapse
Affiliation(s)
- Arne Echterhof
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
- Institute of Medical Microbiology, University Hospital of Muenster, Muenster, Germany
| | - Tejas Dharmaraj
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Arya Khosravi
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Robert McBride
- Felix Biotechnology, South San Francisco, California, USA
| | - Lynn Miesel
- Pharmacology Discovery Services, Taipei, Taiwan
| | | | - Patrick M. Blankenberg
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| | | | | | - Yu-Ling Lee
- Pharmacology Discovery Services, Taipei, Taiwan
| | | | | | - Francis G. Blankenberg
- Division of Pediatric Radiology and Nuclear Medicine, Department of Radiology, Lucile Packard Children’s Hospital, Stanford, California, USA
| | - Krystyna Dąbrowska
- Laboratory of Phage Molecular Biology, Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Derek F. Amanatullah
- Department of Orthopedic Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Adam R. Frymoyer
- Division of Neonatal and Developmental Medicine, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, California, USA
| | - Paul L. Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
23
|
Koncz M, Stirling T, Hadj Mehdi H, Méhi O, Eszenyi B, Asbóth A, Apjok G, Tóth Á, Orosz L, Vásárhelyi BM, Ari E, Daruka L, Polgár TF, Schneider G, Zalokh SA, Számel M, Fekete G, Bohár B, Nagy Varga K, Visnyovszki Á, Székely E, Licker MS, Izmendi O, Costache C, Gajic I, Lukovic B, Molnár S, Szőcs-Gazdi UO, Bozai C, Indreas M, Kristóf K, Van der Henst C, Breine A, Pál C, Papp B, Kintses B. Genomic surveillance as a scalable framework for precision phage therapy against antibiotic-resistant pathogens. Cell 2024; 187:5901-5918.e28. [PMID: 39332413 DOI: 10.1016/j.cell.2024.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/15/2024] [Accepted: 09/04/2024] [Indexed: 09/29/2024]
Abstract
Phage therapy is gaining increasing interest in the fight against critically antibiotic-resistant nosocomial pathogens. However, the narrow host range of bacteriophages hampers the development of broadly effective phage therapeutics and demands precision approaches. Here, we combine large-scale phylogeographic analysis with high-throughput phage typing to guide the development of precision phage cocktails targeting carbapenem-resistant Acinetobacter baumannii, a top-priority pathogen. Our analysis reveals that a few strain types dominate infections in each world region, with their geographical distribution remaining stable within 6 years. As we demonstrate in Eastern Europe, this spatiotemporal distribution enables preemptive preparation of region-specific phage collections that target most local infections. Finally, we showcase the efficacy of phage cocktails against prevalent strain types using in vitro and animal infection models. Ultimately, genomic surveillance identifies patients benefiting from the same phages across geographical scales, thus providing a scalable framework for precision phage therapy.
Collapse
Affiliation(s)
- Mihály Koncz
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary; HCEMM-BRC Translational Microbiology Research Group, Budapesti út 9, 6728 Szeged, Hungary
| | - Tamás Stirling
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary; HCEMM-BRC Translational Microbiology Research Group, Budapesti út 9, 6728 Szeged, Hungary; Doctoral School of Biology, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
| | - Hiba Hadj Mehdi
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary; HCEMM-BRC Translational Microbiology Research Group, Budapesti út 9, 6728 Szeged, Hungary; Doctoral School of Biology, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
| | - Orsolya Méhi
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary
| | - Bálint Eszenyi
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary
| | - András Asbóth
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary; HCEMM-BRC Translational Microbiology Research Group, Budapesti út 9, 6728 Szeged, Hungary; Department of Genetics, ELTE Eötvös Loránd University, Pázmány Péter stny. 1/C, 1117 Budapest, Hungary
| | - Gábor Apjok
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary
| | - Ákos Tóth
- National Center for Public Health and Pharmacy, Albert Flórián út 2-6, 1097 Budapest, Hungary
| | - László Orosz
- Department of Medical Microbiology, University of Szeged, Szent-Györgyi Albert Medical School, Dom tér 10, 6720 Szeged, Hungary
| | - Bálint Márk Vásárhelyi
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary
| | - Eszter Ari
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary; Department of Genetics, ELTE Eötvös Loránd University, Pázmány Péter stny. 1/C, 1117 Budapest, Hungary; HCEMM-BRC Metabolic Systems Biology Group, Temesvári Krt. 62, 6726 Szeged, Hungary
| | - Lejla Daruka
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary
| | - Tamás Ferenc Polgár
- Institute of Biophysics, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary; Theoretical Medicine Doctoral School, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
| | - György Schneider
- Department of Medical Microbiology and Immunology, Medical School, University of Pécs, Szigeti út 12, 7624 Pécs, Hungary
| | - Sif Aldin Zalokh
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary
| | - Mónika Számel
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary
| | - Gergely Fekete
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary; HCEMM-BRC Metabolic Systems Biology Group, Temesvári Krt. 62, 6726 Szeged, Hungary
| | - Balázs Bohár
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary; Faculty of Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, 10th Floor Commonwealth Building Hammersmith Campus, Du Cane Road, London W12 0NN, UK
| | - Karolina Nagy Varga
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary
| | - Ádám Visnyovszki
- South-Pest Central Hospital National Institute of Hematology and Infectious Diseases, Nagyvárad tér 1, 1097 Budapest, Hungary; Doctoral School of Interdisciplinary Medical Sciences, University of Szeged, Dugonics tér 13, 6720 Szeged, Hungary
| | - Edit Székely
- George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, Str. Gheorghe Marinescu 38, 540142 Targu Mures, Romania; County Emergency Clinical Hospital of Targu Mures, Str. Dr. Gh. Marinescu 50, 540136 Targu Mures, Romania
| | - Monica-Sorina Licker
- Microbiology Department, Multidisciplinary Research Center on Antimicrobial Resistance, "Victor Babes" University of Medicine and Pharmacy, Str. Eftimie Murgu 2, 300041 Timisoara, Romania; Microbiology Laboratory, "Pius Branzeu" Emergency Clinical County Hospital, Str. Liviu Rebreanu 156, 300723 Timisoara, Romania
| | - Oana Izmendi
- Microbiology Department, Multidisciplinary Research Center on Antimicrobial Resistance, "Victor Babes" University of Medicine and Pharmacy, Str. Eftimie Murgu 2, 300041 Timisoara, Romania; Microbiology Laboratory, "Pius Branzeu" Emergency Clinical County Hospital, Str. Liviu Rebreanu 156, 300723 Timisoara, Romania; Doctoral School, "Victor Babes" University of Medicine and Pharmacy, Str. Eftimie Murgu 2, 300041 Timisoara, Romania
| | - Carmen Costache
- Department of Microbiology, University of Medicine and Pharmacy "Iuliu Hatieganu" Cluj-Napoca, Str. Victor Babes 8, 400347 Cluj-Napoca, Romania
| | - Ina Gajic
- Institute of Microbiology and Immunology, Faculty of Medicine, University of Belgrade, Dr Subotica 8, 11000 Belgrade, Serbia
| | - Bojana Lukovic
- Academy of Applied Studies Belgrade, College of Health Sciences, Bulevar Zorana Djindjica 152a, Belgrade, Serbia
| | - Szabolcs Molnár
- Emergency County Hospital Miercurea-Ciuc, Str. Doctor Dénes László 2, 530173 Miercurea Ciuc, Romania
| | | | - Csilla Bozai
- County Emergency Hospital Satu Mare, Str. Ravensburg 1-3, 440192 Satu Mare, Romania
| | - Marina Indreas
- Bacau County Emergency Hospital, Str. Haret Spiru 2-4, 600114 Bacau, Romania
| | - Katalin Kristóf
- Institute of Laboratory Medicine, Semmelweis University, Üllői út 78/b, 1083 Budapest, Hungary
| | - Charles Van der Henst
- Microbial Resistance and Drug Discovery, VIB-VUB Center for Structural Biology, VIB, Flanders Institute for Biotechnology, Pleinlaan 2, Building E-3, 1050 Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Pleinlaan 2, Elsene, 1050 Brussels, Belgium
| | - Anke Breine
- Microbial Resistance and Drug Discovery, VIB-VUB Center for Structural Biology, VIB, Flanders Institute for Biotechnology, Pleinlaan 2, Building E-3, 1050 Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel (VUB), Pleinlaan 2, Elsene, 1050 Brussels, Belgium
| | - Csaba Pál
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary
| | - Balázs Papp
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary; HCEMM-BRC Metabolic Systems Biology Group, Temesvári Krt. 62, 6726 Szeged, Hungary; National Laboratory for Health Security, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary.
| | - Bálint Kintses
- Synthetic and Systems Biology Unit, Institute of Biochemistry, National Laboratory of Biotechnology, HUN-REN Biological Research Centre, Temesvári Krt. 62, 6726 Szeged, Hungary; HCEMM-BRC Translational Microbiology Research Group, Budapesti út 9, 6728 Szeged, Hungary.
| |
Collapse
|
24
|
Anastassopoulou C, Ferous S, Petsimeri A, Gioula G, Tsakris A. Phage-Based Therapy in Combination with Antibiotics: A Promising Alternative against Multidrug-Resistant Gram-Negative Pathogens. Pathogens 2024; 13:896. [PMID: 39452768 PMCID: PMC11510143 DOI: 10.3390/pathogens13100896] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
The continued rise in antimicrobial resistance poses a serious threat to public health worldwide. The use of phages that can have bactericidal activity without disrupting the normal flora represents a promising alternative treatment method. This practice has been successfully applied for decades, mainly in Eastern Europe, and has recently been used as an emergency therapy for compassionate care in the United States. Here, we provide a comprehensive review of the pre-clinical and clinical applications of phage therapy concerning three major Gram-negative pathogens: Klebsiella pneumoniae, Pseudomonas aeruginosa, and Acinetobacter baumannii. The advantages and the challenges of expanding the usage of phages as an alternative or adjunctive treatment for antimicrobial-resistant bacterial infections are discussed. We emphasize the virologic complexities of using the highly adaptable phage populations as molecular tools, along with antibiotic chemical compounds, to effectively combat rapidly coevolving pathogenic bacteria in the host microenvironment. Pre-clinical studies, isolated clinical reports and a few randomized clinical trials have shown that bacteriophages can be effective in treating multidrug-resistant bacterial infections. The ability of some phages to revert the resistance against antibiotics, and possibly also against the human complement and other phages, appears to be a great advantage of phage therapy despite the inevitable emergence of phage-resistant strains. Bacteriophages (or specific phage-derived products) can enhance antimicrobial efficacy by reducing bacterial virulence via the alteration of basic bacterial structures, primarily of the cellular wall and membrane. Although several issues remain open regarding their effective clinical application, it appears that phage-based therapeutics in combination with antibiotics can provide an effective solution to the spread of antimicrobial resistance.
Collapse
Affiliation(s)
- Cleo Anastassopoulou
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.A.); (S.F.); (A.P.)
| | - Stefanos Ferous
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.A.); (S.F.); (A.P.)
| | - Aikaterini Petsimeri
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.A.); (S.F.); (A.P.)
| | - Georgia Gioula
- Department of Microbiology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Athanasios Tsakris
- Department of Microbiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (C.A.); (S.F.); (A.P.)
| |
Collapse
|
25
|
Hendrix H, Itterbeek A, Longin H, Delanghe L, Vriens E, Vallino M, Lammens EM, Haque F, Yusuf A, Noben JP, Boon M, Koch MD, van Noort V, Lavigne R. PlzR regulates type IV pili assembly in Pseudomonas aeruginosa via PilZ binding. Nat Commun 2024; 15:8717. [PMID: 39379373 PMCID: PMC11461919 DOI: 10.1038/s41467-024-52732-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 09/16/2024] [Indexed: 10/10/2024] Open
Abstract
Type IV pili (T4P) are thin, flexible filaments exposed on the cell surface of gram-negative bacteria and are involved in pathogenesis-related processes, including cell adsorption, biofilm formation, and twitching motility. Bacteriophages often use these filaments as receptors to infect host cells. Here, we describe the identification of a protein that inhibits T4P assembly in Pseudomonas aeruginosa, discovered during a screen for host factors influencing phage infection. We show that expression of PA2560 (renamed PlzR) in P. aeruginosa inhibits adsorption of T4P-dependent phages. PlzR does this by directly binding the T4P chaperone PilZ, which in turn regulates the ATPase PilB and results in disturbed T4P assembly. As the plzR promoter is induced by cyclic di-GMP, PlzR might play a role in coupling T4P function to levels of this second messenger.
Collapse
Affiliation(s)
- Hanne Hendrix
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, Heverlee, Belgium
| | - Annabel Itterbeek
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, Heverlee, Belgium
- Laboratory for Host Pathogen Interactions in Livestock, Department of Biosystems, KU Leuven, Heverlee, Belgium
| | - Hannelore Longin
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, Heverlee, Belgium
- Computational Systems Biology, Department of Microbial and Molecular Systems, KU Leuven, Heverlee, Belgium
| | - Lize Delanghe
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, Heverlee, Belgium
| | - Eveline Vriens
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, Heverlee, Belgium
| | - Marta Vallino
- Institute for Sustainable Plant Protection, National Research Council of Italy, IPSP-CNR Headquarter, Turin, Italy
| | - Eveline-Marie Lammens
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, Heverlee, Belgium
| | - Farhana Haque
- Department of Biology, Texas A&M University, College Station, TX, USA
| | - Ahmed Yusuf
- Department of Biology, Texas A&M University, College Station, TX, USA
| | - Jean-Paul Noben
- Biomedical Research Institute and Transnational University Limburg, School of Life Sciences, Hasselt University, Diepenbeek, Belgium
| | - Maarten Boon
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, Heverlee, Belgium
| | - Matthias D Koch
- Department of Biology, Texas A&M University, College Station, TX, USA
| | - Vera van Noort
- Computational Systems Biology, Department of Microbial and Molecular Systems, KU Leuven, Heverlee, Belgium
- Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
| | - Rob Lavigne
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, Heverlee, Belgium.
| |
Collapse
|
26
|
Pal N, Sharma P, Kumawat M, Singh S, Verma V, Tiwari RR, Sarma DK, Nagpal R, Kumar M. Phage therapy: an alternative treatment modality for MDR bacterial infections. Infect Dis (Lond) 2024; 56:785-817. [PMID: 39017931 DOI: 10.1080/23744235.2024.2379492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 07/07/2024] [Accepted: 07/08/2024] [Indexed: 07/18/2024] Open
Abstract
The increasing global incidence of multidrug-resistant (MDR) bacterial infections threatens public health and compromises various aspects of modern medicine. Recognising the urgency of this issue, the World Health Organisation has prioritised the development of novel antimicrobials to combat ESKAPEE pathogens. Comprising Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, Enterobacter spp. and Escherichia coli, such pathogens represent a spectrum of high to critical drug resistance, accounting for a significant proportion of hospital-acquired infections worldwide. In response to the waning efficacy of antibiotics against these resilient pathogens, phage therapy (PT) has emerged as a promising therapeutic strategy. This review provides a comprehensive summary of clinical research on PT and explores the translational journey of phages from laboratory settings to clinical applications. It examines recent advancements in pre-clinical and clinical developments, highlighting the potential of phages and their proteins, alone or in combination with antibiotics. Furthermore, this review underlines the importance of establishing safe and approved routes of phage administration to patients. In conclusion, the evolving landscape of phage therapy offers a beacon of hope in the fight against MDR bacterial infections, emphasising the imperative for continued research, innovation and regulatory diligence to realise its full potential in clinical practice.
Collapse
Affiliation(s)
- Namrata Pal
- Department of Microbiology, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, India
- Department of Microbiology, Barkatullah University, Bhopal, Madhya Pradesh, India
| | - Poonam Sharma
- Department of Microbiology, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, India
| | - Manoj Kumawat
- Department of Microbiology, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, India
| | - Samradhi Singh
- Department of Microbiology, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, India
| | - Vinod Verma
- Stem Cell Research Centre, Department of Hematology, Sanjay Gandhi Post-Graduate Institute of Medical Sciences, Lucknow, Uttar Pradesh, India
| | - Rajnarayan R Tiwari
- Department of Microbiology, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, India
| | - Devojit Kumar Sarma
- Department of Microbiology, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, India
| | - Ravinder Nagpal
- Department of Nutrition and Integrative Physiology, College of Health and Human Sciences, Florida State University, Tallahassee, FL, USA
| | - Manoj Kumar
- Department of Microbiology, ICMR-National Institute for Research in Environmental Health, Bhopal, Madhya Pradesh, India
| |
Collapse
|
27
|
Otava UE, Tervo L, Havela R, Vuotari L, Ylänne M, Asplund A, Patpatia S, Kiljunen S. Phage-Antibiotic Combination Therapy against Recurrent Pseudomonas Septicaemia in a Patient with an Arterial Stent. Antibiotics (Basel) 2024; 13:916. [PMID: 39452183 PMCID: PMC11504013 DOI: 10.3390/antibiotics13100916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 09/16/2024] [Accepted: 09/20/2024] [Indexed: 10/26/2024] Open
Abstract
Background: Intravascular stent infections are often associated with high risks of morbidity and mortality. We report here a case of a patient with an arterial stent and recurrent Pseudomonas septicaemias successfully treated with phage-meropenem combination therapy. Methods: A 75-year-old female with arteriosclerosis and comorbidities went through a femoropopliteal bypass with prosthesis in the right inguinal area. After the bypass, she developed a recurring Pseudomonas aeruginosa infection and also neutropenia during different antibiotics. A rapidly growing pseudoaneurysm in the right inguinal area led to an emergency intra-arterial stent placement during blood stream infection, later suspected to host a P. aeruginosa biofilm. Removing the stent was deemed precarious, and phage therapy was considered as a compassionate treatment option. A three-phage cocktail infecting the P. aeruginosa strain was prepared and administered intravenously together with meropenem for two weeks, after which, a ten-month follow-up was carried out. Results: No adverse reactions occurred during the phage therapy treatment, while infection markers were normalized. In addition, recovery was seen in a PET-CT scan. During the 10-month follow-up, no further P. aeruginosa septicaemias occurred. Conclusions: Phage-meropenem combination therapy was thus found safe and effective in the treatment of recurrent Pseudomonas septicaemia in a patient with an arterial stent.
Collapse
Affiliation(s)
- Ulla Elina Otava
- Department of Internal Medicine, Tampere University Hospital, 33520 Tampere, Finland (L.T.); (R.H.)
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland;
| | - Laura Tervo
- Department of Internal Medicine, Tampere University Hospital, 33520 Tampere, Finland (L.T.); (R.H.)
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland;
| | - Riikka Havela
- Department of Internal Medicine, Tampere University Hospital, 33520 Tampere, Finland (L.T.); (R.H.)
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland;
| | - Liisa Vuotari
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland;
- Department of Clinical Physiology and Nuclear Medicine, Tampere University Hospital, 33520 Tampere, Finland
| | - Matti Ylänne
- Human Microbiome Research Program, Research Programs Unit and Medicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland (S.P.)
| | - Annette Asplund
- Human Microbiome Research Program, Research Programs Unit and Medicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland (S.P.)
| | - Sheetal Patpatia
- Human Microbiome Research Program, Research Programs Unit and Medicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland (S.P.)
| | - Saija Kiljunen
- Human Microbiome Research Program, Research Programs Unit and Medicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland (S.P.)
- PrecisionPhage Ltd., 40500 Jyväskylä, Finland
| |
Collapse
|
28
|
Van Nieuwenhuyse B, Balcaen M, Chatzis O, Haenecour A, Derycke E, Detaille T, Clément de Cléty S, Boulanger C, Belkhir L, Yombi JC, De Greef J, Cornu O, Docquier PL, Lentini A, Menten R, Rodriguez-Villalobos H, Verroken A, Djebara S, Merabishvili M, Griselain J, Pirnay JP, Houtekie L, Van der Linden D. Case report: Personalized triple phage-antibiotic combination therapy to rescue necrotizing fasciitis caused by Panton-Valentine leukocidin-producing MRSA in a 12-year-old boy. Front Cell Infect Microbiol 2024; 14:1354681. [PMID: 39355265 PMCID: PMC11442429 DOI: 10.3389/fcimb.2024.1354681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 08/26/2024] [Indexed: 10/03/2024] Open
Abstract
Maximal standard-of-care (SOC) management could not stop the life-threatening progression of a necrotizing fasciitis induced by Panton-Valentine Leukocidin-producing Methicillin-Resistant Staphylococcus aureus (MRSA) in a 12-year-old boy. Multi-route phage therapy was initiated along with antibiotics against Staphylococcus aureus, Pseudomonas aeruginosa and Stenotrophomonas maltophilia, eventually leading to full recovery with no reported adverse events.
Collapse
Affiliation(s)
- Brieuc Van Nieuwenhuyse
- Institute of Experimental and Clinical Research, Pediatric Department (IREC/PEDI), Université catholique de Louvain - UCLouvain, Brussels, Belgium
| | - Mathilde Balcaen
- Pediatric Intensive Care Unit, Cliniques universitaires Saint-Luc, Université catholique de Louvain - UCLouvain, Brussels, Belgium
| | - Olga Chatzis
- Pediatric Infectious Diseases, General Pediatrics Department, Cliniques universitaires Saint-Luc, Université catholique de Louvain - UCLouvain, Brussels, Belgium
| | - Astrid Haenecour
- Pediatric Intensive Care Unit, Cliniques universitaires Saint-Luc, Université catholique de Louvain - UCLouvain, Brussels, Belgium
| | - Emilien Derycke
- Pediatric Intensive Care Unit, Cliniques universitaires Saint-Luc, Université catholique de Louvain - UCLouvain, Brussels, Belgium
| | - Thierry Detaille
- Pediatric Intensive Care Unit, Cliniques universitaires Saint-Luc, Université catholique de Louvain - UCLouvain, Brussels, Belgium
| | - Stéphan Clément de Cléty
- Pediatric Intensive Care Unit, Cliniques universitaires Saint-Luc, Université catholique de Louvain - UCLouvain, Brussels, Belgium
| | - Cécile Boulanger
- Institute of Experimental and Clinical Research, Pediatric Department (IREC/PEDI), Université catholique de Louvain - UCLouvain, Brussels, Belgium
- Genetics of Autoimmune Diseases and Cancer laboratoire (GEDI), de Duve Institute, Université catholique de Louvain - UCLouvain, Brussels, Belgium
- Department of Pediatric Hematology and Oncology, Cliniques Universitaires Saint-Luc, Université catholique de Louvain - UCLouvain, Brussels, Belgium
| | - Leïla Belkhir
- Division of Internal Medicine and Infectious Disease, Cliniques Universitaires Saint-Luc, Université catholique de Louvain - UCLouvain, Brussels, Belgium
- Louvain centre for Toxicology and Applied Pharmacology, Institute of Experimental and Clinical Research (IREC/LTAP), Université catholique de Louvain - UCLouvain, Brussels, Belgium
| | - Jean-Cyr Yombi
- Division of Internal Medicine and Infectious Disease, Cliniques Universitaires Saint-Luc, Université catholique de Louvain - UCLouvain, Brussels, Belgium
| | - Julien De Greef
- Division of Internal Medicine and Infectious Disease, Cliniques Universitaires Saint-Luc, Université catholique de Louvain - UCLouvain, Brussels, Belgium
- Louvain centre for Toxicology and Applied Pharmacology, Institute of Experimental and Clinical Research (IREC/LTAP), Université catholique de Louvain - UCLouvain, Brussels, Belgium
| | - Olivier Cornu
- Department of Orthopedic and Trauma Surgery, Cliniques Universitaires Saint-Luc, Université catholique de Louvain - UCLouvain, Brussels, Belgium
- Institute of Experimental and Clinical Research, Neuromusculoskeletal Lab (IREC/NMSK), Université catholique de Louvain - UCLouvain, Brussels, Belgium
| | - Pierre-Louis Docquier
- Department of Orthopedic and Trauma Surgery, Cliniques Universitaires Saint-Luc, Université catholique de Louvain - UCLouvain, Brussels, Belgium
- Institute of Experimental and Clinical Research, Neuromusculoskeletal Lab (IREC/NMSK), Université catholique de Louvain - UCLouvain, Brussels, Belgium
| | - Audrey Lentini
- Departement of Plastic Surgery, Cliniques universitaires Saint-Luc, Université catholique de Louvain - UCLouvain, Brussels, Belgium
| | - Renaud Menten
- Department of Radiology, Pediatric Radiology Unit, Cliniques universitaires Saint-Luc, Université Catholique de Louvain - UCLouvain, Brussels, Belgium
| | - Hector Rodriguez-Villalobos
- Department of Microbiology, Cliniques universitaires Saint-Luc, Université catholique de Louvain - UCLouvain, Brussels, Belgium
- Institute of Experimental and Clinical Research, Medical Microbiology Department (IREC/MBLG), Université catholique de Louvain - UCLouvain, Brussels, Belgium
| | - Alexia Verroken
- Department of Microbiology, Cliniques universitaires Saint-Luc, Université catholique de Louvain - UCLouvain, Brussels, Belgium
- Institute of Experimental and Clinical Research, Medical Microbiology Department (IREC/MBLG), Université catholique de Louvain - UCLouvain, Brussels, Belgium
| | - Sarah Djebara
- Center for Infectious Diseases, Queen Astrid Military Hospital, Brussels, Belgium
| | - Maya Merabishvili
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| | - Johann Griselain
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| | - Jean-Paul Pirnay
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| | - Laurent Houtekie
- Pediatric Intensive Care Unit, Cliniques universitaires Saint-Luc, Université catholique de Louvain - UCLouvain, Brussels, Belgium
| | - Dimitri Van der Linden
- Institute of Experimental and Clinical Research, Pediatric Department (IREC/PEDI), Université catholique de Louvain - UCLouvain, Brussels, Belgium
- Pediatric Infectious Diseases, General Pediatrics Department, Cliniques universitaires Saint-Luc, Université catholique de Louvain - UCLouvain, Brussels, Belgium
| |
Collapse
|
29
|
Zhao M, Li H, Gan D, Wang M, Deng H, Yang QE. Antibacterial effect of phage cocktails and phage-antibiotic synergy against pathogenic Klebsiella pneumoniae. mSystems 2024; 9:e0060724. [PMID: 39166877 PMCID: PMC11406915 DOI: 10.1128/msystems.00607-24] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/15/2024] [Indexed: 08/23/2024] Open
Abstract
The global rise of antibiotic resistance has renewed interest in phage therapy, as an alternative to antibiotics to eliminate multidrug-resistant (MDR) bacterial pathogens. However, optimizing the broad-spectrum efficacy of phage therapy remains a challenge. In this study, we addressed this issue by employing strategies to improve antimicrobial efficacy of phage therapy against MDR Klebsiella pneumoniae strains, which are notorious for their resistance to conventional antibiotics. This includes the selection of broad host range phages, optimization of phage formulation, and combinations with last-resort antibiotics. Our findings unveil that having a broad host range was a dominant trait of isolated phages, and increasing phage numbers in combination with antibiotics significantly enhanced the suppression of bacterial growth. The decreased incidence of bacterial infection was explained by a reduction in pathogen density and emergence of bacterial resistance. Furthermore, phage-antibiotic synergy (PAS) demonstrated considerable broad-spectrum antibacterial potential against different clades of clinical MDR K. pneumoniae pathogens. The improved treatment outcomes of optimized PAS were also evident in a murine model, where mice receiving optimized PAS therapy demonstrated a reduced bacterial burden in mouse tissues. Taken together, these findings offer an important development in optimizing PAS therapy and its efficacy in the elimination of MDR K. pneumoniae pathogens. IMPORTANCE The worldwide spread of antimicrobial resistance (AMR) has posed a great challenge to global public health. Phage therapy has become a promising alternative against difficult-to-treat pathogens. One important goal of this study was to optimize the therapeutic efficiency of phage-antibiotic combinations, known as phage-antibiotic synergy (PAS). Through comprehensive analysis of the phenotypic and genotypic characteristics of a large number of CRKp-specific phages, we developed a systematic model for phage cocktail combinations. Crucially, our finding demonstrated that PAS treatments not only enhance the bactericidal effects of colistin and tigecycline against multidrug-resistant (MDR) K. pneumoniae strains in in vitro and in vivo context but also provide a robust response when antibiotics fail. Overall, the optimized PAS therapy demonstrates considerable potential in combating diverse K. pneumoniae pathogens, highlighting its relevance as a strategy to mitigate antibiotic resistance threats effectively.
Collapse
Affiliation(s)
- Mengshi Zhao
- College of Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou, China
- Fujian Key Laboratory of Traditional Chinese Veterinary Medicine and Animal Health, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Hongru Li
- Department of Infectious Disease, Shengli Medical College, Fujian Medical University, Fujian Provincial Hospital, Fuzhou University affiliated Provincial Hospital,, Fuzhou, China
| | - Dehao Gan
- Fujian Key Laboratory of Traditional Chinese Veterinary Medicine and Animal Health, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Mengzhu Wang
- College of Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou, China
- Fujian Key Laboratory of Traditional Chinese Veterinary Medicine and Animal Health, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Hui Deng
- Fujian Key Laboratory of Traditional Chinese Veterinary Medicine and Animal Health, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Qiu E Yang
- College of Resources and Environment, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
30
|
Santamaría-Corral G, Aguilera-Correa JJ, Esteban J, García-Quintanilla M. Bacteriophage Therapy on an In Vitro Wound Model and Synergistic Effects in Combination with Beta-Lactam Antibiotics. Antibiotics (Basel) 2024; 13:800. [PMID: 39334975 PMCID: PMC11428794 DOI: 10.3390/antibiotics13090800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 08/19/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
One of the primary opportunistic pathogens that can cause a wide range of diseases is Pseudomonas aeruginosa. This microorganism can become resistant to practically every antibacterial currently in use, including beta-lactam antibiotics. Its ability to proliferate as biofilm has been linked to, among other things, the failure of antimicrobial therapies. Due to a variety of virulence factors and host immune system modifications, P. aeruginosa is one of the most significant and common bacteria that colonize wounds and burns. A novel therapeutic option for treating these multidrug-resistant (MDR) bacterial infections is the combination of antibiotics and bacteriophages. This approach has been linked to improved biofilm penetration, a decreased selection of antibiotic and bacteriophage resistance, and an enhanced antibacterial impact. Combining the F1Pa bacteriophage and beta-lactam antibiotics reduced the viability of the mature biofilm of MDR P. aeruginosa strains and suppressed bacterial growth in vitro. F1Pa critically reduced the amount of biofilm that MDR P. aeruginosa clinical strains formed in the in vitro wound model. These findings highlight the bacteriophage F1Pa's therapeutic potential as a prophylactic topical treatment against MDR pseudomonal infections in wounds and burns.
Collapse
Affiliation(s)
- Guillermo Santamaría-Corral
- Clinical Microbiology Department, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain
| | - John Jairo Aguilera-Correa
- Clinical Microbiology Department, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain
- CIBERINFEC-Consorcio Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas, 28029 Madrid, Spain
| | - Jaime Esteban
- Clinical Microbiology Department, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain
- CIBERINFEC-Consorcio Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas, 28029 Madrid, Spain
| | - Meritxell García-Quintanilla
- Clinical Microbiology Department, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain
- CIBERINFEC-Consorcio Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas, 28029 Madrid, Spain
| |
Collapse
|
31
|
Li HD, Chen YQ, Li Y, Wei X, Wang SY, Cao Y, Wang R, Wang C, Li JY, Li JY, Ding HM, Yang T, Wang JH, Mao C. Harnessing virus flexibility to selectively capture and profile rare circulating target cells for precise cancer subtyping. Nat Commun 2024; 15:5849. [PMID: 38992001 PMCID: PMC11239949 DOI: 10.1038/s41467-024-50064-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 06/26/2024] [Indexed: 07/13/2024] Open
Abstract
The effective isolation of rare target cells, such as circulating tumor cells, from whole blood is still challenging due to the lack of a capturing surface with strong target-binding affinity and non-target-cell resistance. Here we present a solution leveraging the flexibility of bacterial virus (phage) nanofibers with their sidewalls displaying target circulating tumor cell-specific aptamers and their ends tethered to magnetic beads. Such flexible phages, with low stiffness and Young's modulus, can twist and adapt to recognize the cell receptors, energetically enhancing target cell capturing and entropically discouraging non-target cells (white blood cells) adsorption. The magnetic beads with flexible phages can isolate and count target cells with significant increase in cell affinity and reduction in non-target cell absorption compared to magnetic beads having rigid phages. This differentiates breast cancer patients and healthy donors, with impressive area under the curve (0.991) at the optimal detection threshold (>4 target cells mL-1). Immunostaining of captured circulating tumor cells precisely determines breast cancer subtypes with a diagnostic accuracy of 91.07%. Our study reveals the power of viral mechanical attributes in designing surfaces with superior target binding and non-target anti-fouling.
Collapse
Affiliation(s)
- Hui-Da Li
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang, 110819, China
| | - Yuan-Qiang Chen
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou, 215006, China
| | - Yan Li
- Department of Periodontology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Xing Wei
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang, 110819, China
| | - Si-Yi Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang, 110819, China
| | - Ying Cao
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang, 110819, China
| | - Rui Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang, 110819, China
| | - Cong Wang
- Department of Breast Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang, 110042, China
| | - Jing-Yue Li
- Department of Breast Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang, 110042, China
| | - Jian-Yi Li
- Department of Breast Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, Shenyang, 110042, China.
| | - Hong-Ming Ding
- Center for Soft Condensed Matter Physics and Interdisciplinary Research, Soochow University, Suzhou, 215006, China.
| | - Ting Yang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang, 110819, China.
| | - Jian-Hua Wang
- Research Center for Analytical Sciences, Department of Chemistry, College of Sciences, Northeastern University, Box 332, Shenyang, 110819, China
| | - Chuanbin Mao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| |
Collapse
|
32
|
Van Nieuwenhuyse B, Merabishvili M, Goeders N, Vanneste K, Bogaerts B, de Jode M, Ravau J, Wagemans J, Belkhir L, Van der Linden D. Phage-Mediated Digestive Decolonization in a Gut-On-A-Chip Model: A Tale of Gut-Specific Bacterial Prosperity. Viruses 2024; 16:1047. [PMID: 39066209 PMCID: PMC11281504 DOI: 10.3390/v16071047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/17/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Infections due to antimicrobial-resistant bacteria have become a major threat to global health. Some patients may carry resistant bacteria in their gut microbiota. Specific risk factors may trigger the conversion of these carriages into infections in hospitalized patients. Preventively eradicating these carriages has been postulated as a promising preventive intervention. However, previous attempts at such eradication using oral antibiotics or probiotics have led to discouraging results. Phage therapy, the therapeutic use of bacteriophage viruses, might represent a worthy alternative in this context. Taking inspiration from this clinical challenge, we built Gut-On-A-Chip (GOAC) models, which are tridimensional cell culture models mimicking a simplified gut section. These were used to better understand bacterial dynamics under phage pressure using two relevant species: Pseudomonas aeruginosa and Escherichia coli. Model mucus secretion was documented by ELISA assays. Bacterial dynamics assays were performed in GOAC triplicates monitored for 72 h under numerous conditions, such as pre-, per-, or post-bacterial timing of phage introduction, punctual versus continuous phage administration, and phage expression of mucus-binding properties. The potential genomic basis of bacterial phage resistance acquired in the model was investigated by variant sequencing. The bacterial "escape growth" rates under phage pressure were compared to static in vitro conditions. Our results suggest that there is specific bacterial prosperity in this model compared to other in vitro conditions. In E. coli assays, the introduction of a phage harboring unique mucus-binding properties could not shift this balance of power, contradicting previous findings in an in vivo mouse model and highlighting the key differences between these models. Genomic modifications were correlated with bacterial phage resistance acquisition in some but not all instances, suggesting that alternate ways are needed to evade phage predation, which warrants further investigation.
Collapse
Affiliation(s)
- Brieuc Van Nieuwenhuyse
- Institute of Experimental and Clinical Research, Pediatric Department (IREC/PEDI), Université Catholique de Louvain—UCLouvain, 1200 Brussels, Belgium
| | - Maya Merabishvili
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, 1120 Brussels, Belgium
| | - Nathalie Goeders
- Transversal Activities in Applied Genomics, Sciensano, Juliette Wytsmanstraat 14, 1050 Brussels, Belgium (B.B.)
| | - Kevin Vanneste
- Transversal Activities in Applied Genomics, Sciensano, Juliette Wytsmanstraat 14, 1050 Brussels, Belgium (B.B.)
| | - Bert Bogaerts
- Transversal Activities in Applied Genomics, Sciensano, Juliette Wytsmanstraat 14, 1050 Brussels, Belgium (B.B.)
| | - Mathieu de Jode
- Bacterial Diseases, Sciensano, Juliette Wytsmanstraat 14, 1050 Brussels, Belgium
| | - Joachim Ravau
- Institute of Experimental and Clinical Research, Pediatric Department (IREC/PEDI), Université Catholique de Louvain—UCLouvain, 1200 Brussels, Belgium
| | - Jeroen Wagemans
- Laboratory of Gene Technology, KU Leuven, 3000 Leuven, Belgium;
| | - Leïla Belkhir
- Division of Internal Medicine and Infectious Disease, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain—UCLouvain, 1200 Brussels, Belgium
- Louvain Centre for Toxicology and Applied Pharmacology, Institute of Experimental and Clinical Research (IREC/LTAP), Université Catholique de Louvain—UCLouvain, 1200 Brussels, Belgium
| | - Dimitri Van der Linden
- Institute of Experimental and Clinical Research, Pediatric Department (IREC/PEDI), Université Catholique de Louvain—UCLouvain, 1200 Brussels, Belgium
- Pediatric Infectious Diseases, General Pediatrics Department, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain—UCLouvain, 1200 Brussels, Belgium
| |
Collapse
|
33
|
Azam AH, Sato K, Miyanaga K, Nakamura T, Ojima S, Kondo K, Tamura A, Yamashita W, Tanji Y, Kiga K. Selective bacteriophages reduce the emergence of resistant bacteria in bacteriophage-antibiotic combination therapy. Microbiol Spectr 2024; 12:e0042723. [PMID: 38695573 PMCID: PMC11237537 DOI: 10.1128/spectrum.00427-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/09/2024] [Indexed: 06/06/2024] Open
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.
Collapse
Affiliation(s)
- Aa Haeruman Azam
- Therapeutic Drugs and Vaccine Development Research Center, National Institute of Infectious Diseases, Toyama-ku, Shinjuku, Tokyo, Japan
| | - Koji Sato
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsutacho, Yokohama, Japan
| | - Kazuhiko Miyanaga
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsutacho, Yokohama, Japan
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Shimotsukeshi, Tochigi, Japan
| | - Tomohiro Nakamura
- Therapeutic Drugs and Vaccine Development Research Center, National Institute of Infectious Diseases, Toyama-ku, Shinjuku, Tokyo, Japan
| | - Shinjiro Ojima
- Therapeutic Drugs and Vaccine Development Research Center, National Institute of Infectious Diseases, Toyama-ku, Shinjuku, Tokyo, Japan
| | - Kohei Kondo
- Therapeutic Drugs and Vaccine Development Research Center, National Institute of Infectious Diseases, Toyama-ku, Shinjuku, Tokyo, Japan
| | - Azumi Tamura
- Therapeutic Drugs and Vaccine Development Research Center, National Institute of Infectious Diseases, Toyama-ku, Shinjuku, Tokyo, Japan
| | - Wakana Yamashita
- Therapeutic Drugs and Vaccine Development Research Center, National Institute of Infectious Diseases, Toyama-ku, Shinjuku, Tokyo, Japan
| | - Yasunori Tanji
- School of Life Science and Technology, Tokyo Institute of Technology, Nagatsutacho, Yokohama, Japan
| | - Kotaro Kiga
- Therapeutic Drugs and Vaccine Development Research Center, National Institute of Infectious Diseases, Toyama-ku, Shinjuku, Tokyo, Japan
- Division of Bacteriology, Department of Infection and Immunity, Jichi Medical University, Shimotsukeshi, Tochigi, Japan
| |
Collapse
|
34
|
Santamaría-Corral G, Pagán I, Aguilera-Correa JJ, Esteban J, García-Quintanilla M. A Novel Bacteriophage Infecting Multi-Drug- and Extended-Drug-Resistant Pseudomonas aeruginosa Strains. Antibiotics (Basel) 2024; 13:523. [PMID: 38927189 PMCID: PMC11200629 DOI: 10.3390/antibiotics13060523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/13/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
The prevalence of carbapenem-resistant P. aeruginosa has dramatically increased over the last decade, and antibiotics alone are not enough to eradicate infections caused by this opportunistic pathogen. Phage therapy is a fresh treatment that can be administered under compassionate use, particularly against chronic cases. However, it is necessary to thoroughly characterize the virus before therapeutic application. Our work describes the discovery of the novel sequenced bacteriophage, vB_PaeP-F1Pa, containing an integrase, performs a phylogenetical analysis, describes its stability at a physiological pH and temperature, latent period (40 min), and burst size (394 ± 166 particles per bacterial cell), and demonstrates its ability to infect MDR and XDR P. aeruginosa strains. Moreover, this novel bacteriophage was able to inhibit the growth of bacteria inside preformed biofilms. The present study offers a road map to analyze essential areas for successful phage therapy against MDR and XDR P. aeruginosa infections, and shows that a phage containing an integrase is also able to show good in vitro results, indicating that it is very important to perform a genomic analysis before any clinical use, in order to prevent adverse effects in patients.
Collapse
Affiliation(s)
- Guillermo Santamaría-Corral
- Clinical Microbiology Department, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (G.S.-C.); (J.J.A.-C.); (M.G.-Q.)
| | - Israel Pagán
- Centro de Biotecnología y Genómica de Plantas UPM-INIA/CSIC and E.T.S. Ingeniería Agronómica, Alimentaria y de Biosistemas, Universidad Politécnica de Madrid, 28223 Madrid, Spain;
| | - John Jairo Aguilera-Correa
- Clinical Microbiology Department, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (G.S.-C.); (J.J.A.-C.); (M.G.-Q.)
- CIBERINFEC-Consorcio Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas, 28029 Madrid, Spain
| | - Jaime Esteban
- Clinical Microbiology Department, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (G.S.-C.); (J.J.A.-C.); (M.G.-Q.)
- CIBERINFEC-Consorcio Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas, 28029 Madrid, Spain
| | - Meritxell García-Quintanilla
- Clinical Microbiology Department, IIS-Fundación Jiménez Díaz, Universidad Autónoma de Madrid, 28040 Madrid, Spain; (G.S.-C.); (J.J.A.-C.); (M.G.-Q.)
- CIBERINFEC-Consorcio Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Infecciosas, 28029 Madrid, Spain
| |
Collapse
|
35
|
Pirnay JP, Djebara S, Steurs G, Griselain J, Cochez C, De Soir S, Glonti T, Spiessens A, Vanden Berghe E, Green S, Wagemans J, Lood C, Schrevens E, Chanishvili N, Kutateladze M, de Jode M, Ceyssens PJ, Draye JP, Verbeken G, De Vos D, Rose T, Onsea J, Van Nieuwenhuyse B, Soentjens P, Lavigne R, Merabishvili M. Personalized bacteriophage therapy outcomes for 100 consecutive cases: a multicentre, multinational, retrospective observational study. Nat Microbiol 2024; 9:1434-1453. [PMID: 38834776 PMCID: PMC11153159 DOI: 10.1038/s41564-024-01705-x] [Citation(s) in RCA: 96] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 04/19/2024] [Indexed: 06/06/2024]
Abstract
In contrast to the many reports of successful real-world cases of personalized bacteriophage therapy (BT), randomized controlled trials of non-personalized bacteriophage products have not produced the expected results. Here we present the outcomes of a retrospective observational analysis of the first 100 consecutive cases of personalized BT of difficult-to-treat infections facilitated by a Belgian consortium in 35 hospitals, 29 cities and 12 countries during the period from 1 January 2008 to 30 April 2022. We assessed how often personalized BT produced a positive clinical outcome (general efficacy) and performed a regression analysis to identify functional relationships. The most common indications were lower respiratory tract, skin and soft tissue, and bone infections, and involved combinations of 26 bacteriophages and 6 defined bacteriophage cocktails, individually selected and sometimes pre-adapted to target the causative bacterial pathogens. Clinical improvement and eradication of the targeted bacteria were reported for 77.2% and 61.3% of infections, respectively. In our dataset of 100 cases, eradication was 70% less probable when no concomitant antibiotics were used (odds ratio = 0.3; 95% confidence interval = 0.127-0.749). In vivo selection of bacteriophage resistance and in vitro bacteriophage-antibiotic synergy were documented in 43.8% (7/16 patients) and 90% (9/10) of evaluated patients, respectively. We observed a combination of antibiotic re-sensitization and reduced virulence in bacteriophage-resistant bacterial isolates that emerged during BT. Bacteriophage immune neutralization was observed in 38.5% (5/13) of screened patients. Fifteen adverse events were reported, including seven non-serious adverse drug reactions suspected to be linked to BT. While our analysis is limited by the uncontrolled nature of these data, it indicates that BT can be effective in combination with antibiotics and can inform the design of future controlled clinical trials. BT100 study, ClinicalTrials.gov registration: NCT05498363 .
Collapse
Affiliation(s)
- Jean-Paul Pirnay
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium.
- European Society of Clinical Microbiology and Infectious Diseases (ESCMID) Study Group for Non-traditional Antibacterial Therapy (ESGNTA), Basel, Switzerland.
| | - Sarah Djebara
- Center for Infectious Diseases, Queen Astrid Military Hospital, Brussels, Belgium
| | - Griet Steurs
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| | - Johann Griselain
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| | - Christel Cochez
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| | - Steven De Soir
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| | - Tea Glonti
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| | - An Spiessens
- Center for Infectious Diseases, Queen Astrid Military Hospital, Brussels, Belgium
| | - Emily Vanden Berghe
- Center for Infectious Diseases, Queen Astrid Military Hospital, Brussels, Belgium
| | - Sabrina Green
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Jeroen Wagemans
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Cédric Lood
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, Leuven, Belgium
| | | | - Nina Chanishvili
- Eliava Institute of Bacteriophages, Microbiology and Virology, Tbilisi, Georgia
| | - Mzia Kutateladze
- Eliava Institute of Bacteriophages, Microbiology and Virology, Tbilisi, Georgia
| | | | | | - Jean-Pierre Draye
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| | - Gilbert Verbeken
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| | - Daniel De Vos
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| | - Thomas Rose
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| | - Jolien Onsea
- Department of Trauma Surgery, University Hospitals Leuven; Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Brieuc Van Nieuwenhuyse
- Institute of Experimental and Clinical Research, Pediatric Department, UCLouvain, Brussels, Belgium
| | - Patrick Soentjens
- Center for Infectious Diseases, Queen Astrid Military Hospital, Brussels, Belgium
| | - Rob Lavigne
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, Leuven, Belgium
| | - Maya Merabishvili
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| |
Collapse
|
36
|
Kraus S, Fletcher ML, Łapińska U, Chawla K, Baker E, Attrill EL, O'Neill P, Farbos A, Jeffries A, Galyov EE, Korbsrisate S, Barnes KB, Harding SV, Tsaneva-Atanasova K, Blaskovich MAT, Pagliara S. Phage-induced efflux down-regulation boosts antibiotic efficacy. PLoS Pathog 2024; 20:e1012361. [PMID: 38941361 PMCID: PMC11239113 DOI: 10.1371/journal.ppat.1012361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 07/11/2024] [Accepted: 06/21/2024] [Indexed: 06/30/2024] Open
Abstract
The interactions between a virus and its host vary in space and time and are affected by the presence of molecules that alter the physiology of either the host or the virus. Determining the molecular mechanisms at the basis of these interactions is paramount for predicting the fate of bacterial and phage populations and for designing rational phage-antibiotic therapies. We study the interactions between stationary phase Burkholderia thailandensis and the phage ΦBp-AMP1. Although heterogeneous genetic resistance to phage rapidly emerges in B. thailandensis, the presence of phage enhances the efficacy of three major antibiotic classes, the quinolones, the beta-lactams and the tetracyclines, but antagonizes tetrahydrofolate synthesis inhibitors. We discovered that enhanced antibiotic efficacy is facilitated by reduced antibiotic efflux in the presence of phage. This new phage-antibiotic therapy allows for eradication of stationary phase bacteria, whilst requiring reduced antibiotic concentrations, which is crucial for treating infections in sites where it is difficult to achieve high antibiotic concentrations.
Collapse
Affiliation(s)
- Samuel Kraus
- Living Systems Institute and Biosciences, University of Exeter, Exeter, Devon, United Kingdom
| | - Megan L Fletcher
- Living Systems Institute and Biosciences, University of Exeter, Exeter, Devon, United Kingdom
| | - Urszula Łapińska
- Living Systems Institute and Biosciences, University of Exeter, Exeter, Devon, United Kingdom
| | - Krina Chawla
- Living Systems Institute and Biosciences, University of Exeter, Exeter, Devon, United Kingdom
| | - Evan Baker
- Department of Mathematics and Living Systems Institute, University of Exeter, Exeter, Devon, United Kingdom
- EPSRC Hub for Quantitative Modelling in Healthcare, University of Exeter, Exeter, United Kingdom
| | - Erin L Attrill
- Living Systems Institute and Biosciences, University of Exeter, Exeter, Devon, United Kingdom
| | - Paul O'Neill
- Biosciences, University of Exeter, Exeter, Devon, EX4 4QD, United Kingdom
| | - Audrey Farbos
- Biosciences, University of Exeter, Exeter, Devon, EX4 4QD, United Kingdom
| | - Aaron Jeffries
- Biosciences, University of Exeter, Exeter, Devon, EX4 4QD, United Kingdom
| | - Edouard E Galyov
- Department of Genetics and Genome Biology, University of Leicester, University Road, Leicester, United Kingdom
| | - Sunee Korbsrisate
- Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok Thailand
| | - Kay B Barnes
- Defence Science and Technology Laboratory, Porton Down, Salisbury, Wiltshire, United Kingdom
| | - Sarah V Harding
- Defence Science and Technology Laboratory, Porton Down, Salisbury, Wiltshire, United Kingdom
| | - Krasimira Tsaneva-Atanasova
- Department of Mathematics and Living Systems Institute, University of Exeter, Exeter, Devon, United Kingdom
- EPSRC Hub for Quantitative Modelling in Healthcare, University of Exeter, Exeter, United Kingdom
| | - Mark A T Blaskovich
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia
| | - Stefano Pagliara
- Living Systems Institute and Biosciences, University of Exeter, Exeter, Devon, United Kingdom
| |
Collapse
|
37
|
Shaaban MT, Abdel-Raouf M, Zayed M, Emara MA. Microbiological and molecular studies on a multidrug-resistant Pseudomonas aeruginosa from a liver transplant patient with urinary tract infection in Egypt. BMC Microbiol 2024; 24:184. [PMID: 38802754 PMCID: PMC11129433 DOI: 10.1186/s12866-024-03318-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 05/03/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Pseudomonas aeruginosa is an opportunistic pathogen responsible for complicated UTIs and exhibits high antibiotic resistance, leading to increased mortality rates, especially in cases of multidrug-resistant strains. This study aimed to investigate the antibiotic susceptibility patterns and genomic characterization of XDR strains identified in end-stage liver disease patients who underwent liver transplants. METHODS In this study, a number of 30 individuals who underwent liver transplants were registered. Ninety urine and 60 wound site swab samples were collected and processed for culturing, identification, and antimicrobial sensitivity. Extensively drug-resistant strain EMARA01 was confirmed through Sanger sequencing and was then processed for whole genome sequencing to characterize the genomic pattern. Sequencing data were processed for de novo assembly using various tools and databases, including genome annotation, serotype identification, virulence factor genes, and antimicrobial resistance gene. Pangenome analysis of randomly selected 147 reference strains and EMAR01 sequenced strain was performed using the Bacterial Pan Genome Analysis (BPGA) software. RESULTS Of these total examined samples, nosocomial infection due to P. aeruginosa was detected in twelve patients' samples. AST analysis showed that P. aeruginosa strains exhibit resistance to tobramycin, erythromycin, and gentamicin, followed by piperacillin and ofloxacin, and no strains exhibit resistance to meropenem and imipenem. The CARD database identified 59 AMR genes similar to the EMAR01 strain genome and mostly belong to the family involved in the resistance-nodulation-cell division (RND) antibiotic efflux pump. Five genes; nalC, nalD, MexR, MexA, and MexB, exhibit resistance to 14 classes of antibiotics, while two AMR; CpxR, and OprM, exhibit resistance to 15 classes of drugs. Pangenome analysis revealed that the pan-genome remained open, suggesting the potential for acquiring accessory and unique genes. Notably, the genes predominantly involved in amino acid transport metabolism were identified using the KEGG database. CONCLUSIONS This study provides valuable insights into the antimicrobial resistance profile, genetic features, and genomic evolution of P. aeruginosa strains causing UTIs in liver transplant patients. The findings emphasize the significance of comprehending AMR mechanisms and genetic diversity in P. aeruginosa for developing effective treatment strategies and infection control measures.
Collapse
Affiliation(s)
- Mohamed T Shaaban
- Department of Botany and Microbiology, Faculty of Science, Menoufia University, Shebin El- Kom, Egypt
| | | | - Muhammad Zayed
- Department of Botany and Microbiology, Faculty of Science, Menoufia University, Shebin El- Kom, Egypt.
| | - Mahmoud A Emara
- Department of Botany and Microbiology, Faculty of Science, Menoufia University, Shebin El- Kom, Egypt
| |
Collapse
|
38
|
Greenrod STE, Cazares D, Johnson S, Hector TE, Stevens EJ, MacLean RC, King KC. Warming alters life-history traits and competition in a phage community. Appl Environ Microbiol 2024; 90:e0028624. [PMID: 38624196 PMCID: PMC11107170 DOI: 10.1128/aem.00286-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 03/26/2024] [Indexed: 04/17/2024] Open
Abstract
Host-parasite interactions are highly susceptible to changes in temperature due to mismatches in species thermal responses. In nature, parasites often exist in communities, and responses to temperature are expected to vary between host-parasite pairs. Temperature change thus has consequences for both host-parasite dynamics and parasite-parasite interactions. Here, we investigate the impact of warming (37°C, 40°C, and 42°C) on parasite life-history traits and competition using the opportunistic bacterial pathogen Pseudomonas aeruginosa (host) and a panel of three genetically diverse lytic bacteriophages (parasites). We show that phages vary in their responses to temperature. While 37°C and 40°C did not have a major effect on phage infectivity, infection by two phages was restricted at 42°C. This outcome was attributed to disruption of different phage life-history traits including host attachment and replication inside hosts. Furthermore, we show that temperature mediates competition between phages by altering their competitiveness. These results highlight phage trait variation across thermal regimes with the potential to drive community dynamics. Our results have important implications for eukaryotic viromes and the design of phage cocktail therapies.IMPORTANCEMammalian hosts often elevate their body temperatures through fevers to restrict the growth of bacterial infections. However, the extent to which fever temperatures affect the communities of phages with the ability to parasitize those bacteria remains unclear. In this study, we investigate the impact of warming across a fever temperature range (37°C, 40°C, and 42°C) on phage life-history traits and competition using a bacterium (host) and bacteriophage (parasite) system. We show that phages vary in their responses to temperature due to disruption of different phage life-history traits. Furthermore, we show that temperature can alter phage competitiveness and shape phage-phage competition outcomes. These results suggest that fever temperatures have the potential to restrict phage infectivity and drive phage community dynamics. We discuss implications for the role of temperature in shaping host-parasite interactions more widely.
Collapse
Affiliation(s)
| | - Daniel Cazares
- Department of Biology, University of Oxford, Oxford, United Kingdom
| | - Serena Johnson
- Department of Biology, University of Oxford, Oxford, United Kingdom
- Department of Zoology, University of British Columbia, Vancouver, Canada
| | - Tobias E. Hector
- Department of Biology, University of Oxford, Oxford, United Kingdom
| | - Emily J. Stevens
- Department of Biology, University of Oxford, Oxford, United Kingdom
| | - R. Craig MacLean
- Department of Biology, University of Oxford, Oxford, United Kingdom
| | - Kayla C. King
- Department of Biology, University of Oxford, Oxford, United Kingdom
- Department of Zoology, University of British Columbia, Vancouver, Canada
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada
| |
Collapse
|
39
|
Wang B, Du L, Dong B, Kou E, Wang L, Zhu Y. Current Knowledge and Perspectives of Phage Therapy for Combating Refractory Wound Infections. Int J Mol Sci 2024; 25:5465. [PMID: 38791502 PMCID: PMC11122179 DOI: 10.3390/ijms25105465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 05/09/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
Wound infection is one of the most important factors affecting wound healing, so its effective control is critical to promote the process of wound healing. However, with the increasing prevalence of multi-drug-resistant (MDR) bacterial strains, the prevention and treatment of wound infections are now more challenging, imposing heavy medical and financial burdens on patients. Furthermore, the diminishing effectiveness of conventional antimicrobials and the declining research on new antibiotics necessitate the urgent exploration of alternative treatments for wound infections. Recently, phage therapy has been revitalized as a promising strategy to address the challenges posed by bacterial infections in the era of antibiotic resistance. The use of phage therapy in treating infectious diseases has demonstrated positive results. This review provides an overview of the mechanisms, characteristics, and delivery methods of phage therapy for combating pathogenic bacteria. Then, we focus on the clinical application of various phage therapies in managing refractory wound infections, such as diabetic foot infections, as well as traumatic, surgical, and burn wound infections. Additionally, an analysis of the potential obstacles and challenges of phage therapy in clinical practice is presented, along with corresponding strategies for addressing these issues. This review serves to enhance our understanding of phage therapy and provides innovative avenues for addressing refractory infections in wound healing.
Collapse
Affiliation(s)
- Bo Wang
- Department of Dermatology, Naval Medical Center, Naval Medical University, Shanghai 200052, China
| | - Lin Du
- Department of Dermatology, Naval Medical Center, Naval Medical University, Shanghai 200052, China
| | - Baiping Dong
- Department of Dermatology, Naval Medical Center, Naval Medical University, Shanghai 200052, China
| | - Erwen Kou
- Department of Dermatology, Naval Medical Center, Naval Medical University, Shanghai 200052, China
| | - Liangzhe Wang
- Department of Dermatology, Naval Medical Center, Naval Medical University, Shanghai 200052, China
| | - Yuanjie Zhu
- Department of Dermatology, Naval Medical Center, Naval Medical University, Shanghai 200052, China
| |
Collapse
|
40
|
Khong E, Oh JJ, Jimenez JM, Liu R, Dunham S, Monsibais A, Rhoads A, Ghatbale P, Garcia A, Cobián Güemes AG, Blanc AN, Chiu M, Kuo P, Proost M, Kline A, Aslam S, Schooley RT, Whiteson K, Fraley SI, Pride DT. A simple solid media assay for detection of synergy between bacteriophages and antibiotics. Microbiol Spectr 2024; 12:e0322123. [PMID: 38526142 PMCID: PMC11064537 DOI: 10.1128/spectrum.03221-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 02/24/2024] [Indexed: 03/26/2024] Open
Abstract
The emergence of antibiotic-resistant bacteria (ARB) has necessitated the development of alternative therapies to deal with this global threat. Bacteriophages (viruses that target bacteria) that kill ARB are one such alternative. Although phages have been used clinically for decades with inconsistent results, a number of recent advances in phage selection, propagation, and purification have enabled a reevaluation of their utility in contemporary clinical medicine. In most phage therapy cases, phages are administered in combination with antibiotics to ensure that patients receive the standard-of-care treatment. Some phages may work cooperatively with antibiotics to eradicate ARB, as often determined using non-standardized broth assays. We sought to develop a solid media-based assay to assess cooperativity between antibiotics and phages to offer a standardized platform for such testing. We modeled the interactions that occur between antibiotics and phages on solid medium to measure additive, antagonistic, and synergistic interactions. We then tested the method using different bacterial isolates and identified a number of isolates where synergistic interactions were identified. These interactions were not dependent on the specific organism, phage family, or antibiotic used. A priori susceptibility to the antibiotic or the specific phage were not requirements to observe synergistic interactions. Our data also confirm the potential for the restoration of vancomycin to treat vancomycin-resistant Enterococcus (VRE) when used in combination with phages. Solid media assays for the detection of cooperative interactions between antibiotics and phages can be an accessible technique adopted by clinical laboratories to evaluate antibiotic and phage choices in phage therapy.IMPORTANCEBacteriophages have become an important alternative treatment for individuals with life-threatening antibiotic-resistant bacteria (ARB) infections. Because antibiotics represent the standard-of-care for treatment of ARB, antibiotics and phages often are delivered together without evidence that they work cooperatively. Testing for cooperativity can be difficult due to the equipment necessary and a lack of standardized means for performing the testing in liquid medium. We developed an assay using solid medium to identify interactions between antibiotics and phages for gram-positive and gram-negative bacteria. We modeled the interactions between antibiotics and phages on solid medium, and then tested multiple replicates of vancomycin-resistant Enterococcus (VRE) and Stenotrophomonas in the assay. For each organism, we identified synergy between different phage and antibiotic combinations. The development of this solid media assay for assessing synergy between phages and antibiotics will better inform the use of these combinations in the treatment of ARB infections.
Collapse
Affiliation(s)
- Ethan Khong
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Joseph J. Oh
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Julian M. Jimenez
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - Roland Liu
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Sage Dunham
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, California, USA
| | - Alisha Monsibais
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, California, USA
| | - Alison Rhoads
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Pooja Ghatbale
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Andrew Garcia
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | | | - Alisha N. Blanc
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Megan Chiu
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Peiting Kuo
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Marissa Proost
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Ahnika Kline
- Department of Pathology, University of California San Diego, La Jolla, California, USA
| | - Saima Aslam
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Robert T. Schooley
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| | - Katrine Whiteson
- Department of Molecular Biology and Biochemistry, University of California Irvine, Irvine, California, USA
| | - Stephanie I. Fraley
- Department of Bioengineering, University of California San Diego, La Jolla, California, USA
| | - David T. Pride
- Department of Pathology, University of California San Diego, La Jolla, California, USA
- Department of Medicine, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
41
|
Loganathan A, Bozdogan B, Manohar P, Nachimuthu R. Phage-antibiotic combinations in various treatment modalities to manage MRSA infections. Front Pharmacol 2024; 15:1356179. [PMID: 38659581 PMCID: PMC11041375 DOI: 10.3389/fphar.2024.1356179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 03/22/2024] [Indexed: 04/26/2024] Open
Abstract
Introduction: The emergence of antibiotic resistance is a significant challenge in the treatment of bacterial infections, particularly in patients in the intensive care unit (ICU). Phage-antibiotic combination therapy is now being utilized as a preferred therapeutic option for infections that are multi-drug resistant in nature. Methods: In this study, we examined the combined impact of the staph phage vB_Sau_S90 and four antibiotics on methicillin-resistant Staphylococcus aureus (MRSA). We conducted experiments on three different treatment sequences: a) administering phages before antibiotics, b) administering phages and antibiotics simultaneously, and c) administering antibiotics before phages. Results: When the media was supplemented with sub-inhibitory concentrations of 0.25 μg/mL and 1 μg/mL, the size of the plaque increased from 0.5 ± 0.1 mm (in the control group with only the phage) to 4 ± 0.2 mm, 1.6 ± 0.1 mm, and 1.6 ± 0.4 mm when fosfomycin, ciprofloxacin, and oxacillin were added, respectively. The checkerboard analysis revealed a synergistic effect between the phages and antibiotics investigated, as indicated by a FIC value of less than 0.5. The combination treatment of phages and antibiotics demonstrated universal efficacy across all treatments. Nevertheless, the optimal effectiveness was demonstrated when the antibiotics were delivered subsequent to the phages. Utilizing the Galleria mellonella model, in vivo experiments showed that the combination of phage-oxacillin effectively eliminated biofilm-infected larvae, resulting in a survival rate of up to 80% in the treated groups. Discussion: Our findings highlight the advantages of using a combination of phage and antibiotic over using phages alone in the treatment of MRSA infections.
Collapse
Affiliation(s)
- Archana Loganathan
- School of Bioscience and Technology, Vellore Institute of Technology (VIT), Vellore, India
| | - Bulent Bozdogan
- Medical Microbiology Department, Adnan Menderes University, Aydin, Türkiye
| | - Prasanth Manohar
- School of Bioscience and Technology, Vellore Institute of Technology (VIT), Vellore, India
| | - Ramesh Nachimuthu
- School of Bioscience and Technology, Vellore Institute of Technology (VIT), Vellore, India
| |
Collapse
|
42
|
Aslam S, Roach D, Nikolich MP, Biswas B, Schooley RT, Lilly-Bishop KA, Rice GK, Cer RZ, Hamilton T, Henry M, Luong T, Salabarria AC, Sisk-Hackworth L, Filippov AA, Lebreton F, Hall L, Nir-Paz R, Onallah H, Livni G, Shostak E, Wieder-Finesod A, Yahav D, Yerushalmy O, Alkalay-Oren S, Braunstein R, Khalifa L, Rimon A, Gelman D, Hazan R. Pseudomonas aeruginosa ventricular assist device infections: findings from ineffective phage therapies in five cases. Antimicrob Agents Chemother 2024; 68:e0172823. [PMID: 38470133 PMCID: PMC10989018 DOI: 10.1128/aac.01728-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 02/06/2024] [Indexed: 03/13/2024] Open
Abstract
Left ventricular assist devices (LVAD) are increasingly used for management of heart failure; infection remains a frequent complication. Phage therapy has been successful in a variety of antibiotic refractory infections and is of interest in treating LVAD infections. We performed a retrospective review of four patients that underwent five separate courses of intravenous (IV) phage therapy with concomitant antibiotic for treatment of endovascular Pseudomonas aeruginosa LVAD infection. We assessed phage susceptibility, bacterial strain sequencing, serum neutralization, biofilm activity, and shelf-life of phage preparations. Five treatments of one to four wild-type virulent phage(s) were administered for 14-51 days after informed consent and regulatory approval. There was no successful outcome. Breakthrough bacteremia occurred in four of five treatments. Two patients died from the underlying infection. We noted a variable decline in phage susceptibility following three of five treatments, four of four tested developed serum neutralization, and prophage presence was confirmed in isolates of two tested patients. Two phage preparations showed an initial titer drop. Phage biofilm activity was confirmed in two. Phage susceptibility alone was not predictive of clinical efficacy in P. aeruginosa endovascular LVAD infection. IV phage was associated with serum neutralization in most cases though lack of clinical effect may be multifactorial including presence of multiple bacterial isolates with varying phage susceptibility, presence of prophages, decline in phage titers, and possible lack of biofilm activity. Breakthrough bacteremia occurred frequently (while the organism remained susceptible to administered phage) and is an important safety consideration.
Collapse
Affiliation(s)
- Saima Aslam
- Division of Infectious Diseases and Global Public Health and the Center for Innovative Phage Applications and Therapeutics, University of California San Diego, La Jolla, California, USA
| | - Dwayne Roach
- Department of Biology, San Diego State University, San Diego, California, USA
| | - Mikeljon P. Nikolich
- Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Biswajit Biswas
- Naval Medical Research Command – Frederick, Fort Detrick, Maryland, USA
| | - Robert T. Schooley
- Division of Infectious Diseases and Global Public Health and the Center for Innovative Phage Applications and Therapeutics, University of California San Diego, La Jolla, California, USA
| | | | - Gregory K. Rice
- Naval Medical Research Command – Frederick, Fort Detrick, Maryland, USA
- Leidos, Inc, Reston, Virginia, USA
| | - Regina Z. Cer
- Naval Medical Research Command – Frederick, Fort Detrick, Maryland, USA
| | - Theron Hamilton
- Naval Medical Research Command – Frederick, Fort Detrick, Maryland, USA
| | - Matthew Henry
- Naval Medical Research Command – Frederick, Fort Detrick, Maryland, USA
- The Geneva Foundation, Tacoma, Washington, USA
| | - Tiffany Luong
- Department of Biology, San Diego State University, San Diego, California, USA
| | | | | | - Andrey A. Filippov
- Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Francois Lebreton
- Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Lindsey Hall
- Bacterial Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Ran Nir-Paz
- Department of Clinical Microbiology and Infectious Diseases, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Hadil Onallah
- Department of Clinical Microbiology and Infectious Diseases, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Gilat Livni
- Schneider Children’s Medical Center, Petah Tikva, Israel
| | - Eran Shostak
- Schneider Children’s Medical Center, Petah Tikva, Israel
| | - Anat Wieder-Finesod
- The Infectious Diseases Unit, Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel
| | - Dafna Yahav
- The Infectious Diseases Unit, Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel
| | - Ortal Yerushalmy
- Institute of Biomedical and Oral Research (IBOR), Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Sivan Alkalay-Oren
- Institute of Biomedical and Oral Research (IBOR), Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ron Braunstein
- Institute of Biomedical and Oral Research (IBOR), Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Leron Khalifa
- Institute of Biomedical and Oral Research (IBOR), Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Amit Rimon
- Institute of Biomedical and Oral Research (IBOR), Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Daniel Gelman
- Institute of Biomedical and Oral Research (IBOR), Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ronen Hazan
- Institute of Biomedical and Oral Research (IBOR), Faculty of Dental Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
43
|
Moghadam MT, Mojtahedi A, Salamy S, Shahbazi R, Satarzadeh N, Delavar M, Ashoobi MT. Phage therapy as a glimmer of hope in the fight against the recurrence or emergence of surgical site bacterial infections. Infection 2024; 52:385-402. [PMID: 38308075 DOI: 10.1007/s15010-024-02178-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 01/05/2024] [Indexed: 02/04/2024]
Abstract
PURPOSE Over the last decade, surgery rates have risen alarmingly, and surgical-site infections are expanding these concerns. In spite of advances in infection control practices, surgical infections continue to be a significant cause of death, prolonged hospitalization, and morbidity. As well as the presence of bacterial infections and their antibiotic resistance, biofilm formation is one of the challenges in the treatment of surgical wounds. METHODS This review article was based on published studies on inpatients and laboratory animals receiving phage therapy for surgical wounds, phage therapy for tissue and bone infections treated with surgery to prevent recurrence, antibiotic-resistant wound infections treated with phage therapy, and biofilm-involved surgical wounds treated with phage therapy which were searched without date restrictions. RESULTS It has been shown in this review article that phage therapy can be used to treat surgical-site infections in patients and animals, eliminate biofilms at the surgical site, prevent infection recurrence in wounds that have been operated on, and eradicate antibiotic-resistant infections in surgical wounds, including multi-drug resistance (MDR), extensively drug resistance (XDR), and pan-drug resistance (PDR). A cocktail of phages and antibiotics can also reduce surgical-site infections more effectively than phages alone. CONCLUSION In light of these encouraging results, clinical trials and research with phages will continue in the near future to treat surgical-site infections, biofilm removal, and antibiotic-resistant wounds, all of which could be used to prescribe phages as an alternative to antibiotics.
Collapse
Affiliation(s)
- Majid Taati Moghadam
- Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ali Mojtahedi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shakiba Salamy
- Department of Microbiology, Faculty of Pharmacy, Islamic Azad University, Tehran, Iran
| | - Razieh Shahbazi
- Department of Microbiology, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Naghmeh Satarzadeh
- Student Research Committee, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Majid Delavar
- Vice President of Health and Executive Vice President, Rey Health Center, Tehran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
44
|
Turner PE, Azeredo J, Buurman ET, Green S, Haaber JK, Haggstrom D, Kameda de Figueiredo Carvalho K, Kirchhelle C, Gonzalez Moreno M, Pirnay JP, Portillo MA. Addressing the Research and Development Gaps in Modern Phage Therapy. PHAGE (NEW ROCHELLE, N.Y.) 2024; 5:30-39. [PMID: 40114805 PMCID: PMC11920706 DOI: 10.1089/phage.2023.0045] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Antimicrobial resistance is on the rise globally, prompting increased research and development (R&D) of phage therapy as a strategy to address difficult-to-treat bacterial infections. We review the current state of phage therapy research, including major operational, epistemic, and biological challenges for phage R&D, and discuss some new approaches to developing the technology motivated by recent breakthroughs such as artificial intelligence and synthetic phage production. In addition, we contextualize these R&D challenges and opportunities in light of the ongoing predicament of commercial antimicrobial innovation and current public-private efforts to reinvigorate the pipeline of antimicrobial drug discovery. We conclude with reflections on the potential for new phage therapies to be readily accessible across all income contexts to better ensure broad patient access, and consider possible alternatives to current public and public-private solutions for phage therapy and production.
Collapse
Affiliation(s)
- Paul E Turner
- Department of Ecology and Evolutionary Biology, Yale University, New Haven, Connecticut, USA
- Microbiology Program, Yale University School of Medicine, New Haven, Connecticut, USA
- Center for Phage Biology and Therapy, Yale University, New Haven, Connecticut, USA
| | - Joana Azeredo
- Centre of Biological Engineering, University of Minho, Braga, Portugal
- LABBELS-Associate Laboratory, Braga, Guimarães, Portugal
| | - Ed T Buurman
- CARB-X, Boston University, Boston, Massachusetts, USA
| | - Sabrina Green
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, Leuven, Belgium
| | | | - Douglas Haggstrom
- INCubator for Antibiotic Therapies Europe (INCATE), Basel, Switzerland
- Innovation Office, University of Basel, Basel, Switzerland
| | | | | | - Mercedes Gonzalez Moreno
- INCubator for Antibiotic Therapies Europe (INCATE), Basel, Switzerland
- Leibniz Institute for Natural Product Research and Infection Biology, Hans Knoll Institute, Jena, Germany
| | - Jean-Paul Pirnay
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| | | |
Collapse
|
45
|
Desmecht S, Latka A, Ceyssens PJ, Garcia-Pino A, Gillis A, Lavigne R, Lima-Mendez G, Matthijnssens J, Vázquez R, Venneman J, Wagemans J, Briers Y, Thiry D. Meeting Report of the Second Symposium of the Belgian Society for Viruses of Microbes and Launch of the Phage Valley. Viruses 2024; 16:299. [PMID: 38400074 PMCID: PMC10891784 DOI: 10.3390/v16020299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
The second symposium of the Belgian Society for Viruses of Microbes (BSVoM) took place on 8 September 2023 at the University of Liège with 141 participants from 10 countries. The meeting program covered three thematic sessions opened by international keynote speakers: two sessions were devoted to "Fundamental research in phage ecology and biology" and the third one to the "Present and future applications of phages". During this one day symposium, four invited keynote lectures, nine selected talks and eight student pitches were given along with thirty presented posters. The president of the Belgian Society for Viruses of Microbes, Prof. Yves Briers, took advantage of this symposium to launch the Phage Valley concept that will put the spotlight on the exceptionally high density of researchers investigating viruses of microbes as well as the successful triple helix approach between academia, industry and government in Belgium.
Collapse
Affiliation(s)
- Salomé Desmecht
- Veterinary Bacteriology, Department of Infectious and Parasitic Diseases, Fundamental and Applied Research for Animals and Health, Faculty of Veterinary Medicine, University of Liège (ULiège), 4000 Liège, Belgium;
| | - Agnieszka Latka
- Laboratory of Applied Biotechnology, Department of Biotechnology, Faculty of Bioscience Engineering, University of Ghent (UGent), 9000 Gent, Belgium; (A.L.); (R.V.)
- Department of Pathogen Biology and Immunology, Faculty of Biological Sciences, University of Wroclaw, 51-148 Wroclaw, Poland
| | | | - Abel Garcia-Pino
- Cellular and Molecular Microbiology, Faculty of Sciences, Université Libre de Bruxelles (ULB), 1050 Brussels, Belgium;
| | - Annika Gillis
- Laboratory of Food and Environmental Microbiology, Earth and Life Institute, Catholic University of Louvain (UCLouvain), 1348 Louvain-la-Neuve, Belgium;
| | - Rob Lavigne
- Laboratory of Gene Technology, Department of Biosystems, Faculty of Bioscience Engineering, KU Leuven, 3001 Leuven, Belgium; (R.L.); (J.W.)
| | - Gipsi Lima-Mendez
- Biology of Microorganisms Research Unit (URBM), Namur Research Institute for Life Sciences (NARILIS), University of Namur (UNamur), 5000 Namur, Belgium;
| | - Jelle Matthijnssens
- Laboratory of Viral Metagenomics, Department of Microbiology, Immunology and Transplantation, Rega Institute, Division of Clinical and Epidemiological Virology, KU Leuven, 3000 Leuven, Belgium;
| | - Roberto Vázquez
- Laboratory of Applied Biotechnology, Department of Biotechnology, Faculty of Bioscience Engineering, University of Ghent (UGent), 9000 Gent, Belgium; (A.L.); (R.V.)
| | - Jolien Venneman
- Flanders Research Institute for Agriculture, Fisheries and Food (ILVO), 9820 Merelbeke, Belgium;
| | - Jeroen Wagemans
- Laboratory of Gene Technology, Department of Biosystems, Faculty of Bioscience Engineering, KU Leuven, 3001 Leuven, Belgium; (R.L.); (J.W.)
| | - Yves Briers
- Laboratory of Applied Biotechnology, Department of Biotechnology, Faculty of Bioscience Engineering, University of Ghent (UGent), 9000 Gent, Belgium; (A.L.); (R.V.)
| | - Damien Thiry
- Veterinary Bacteriology, Department of Infectious and Parasitic Diseases, Fundamental and Applied Research for Animals and Health, Faculty of Veterinary Medicine, University of Liège (ULiège), 4000 Liège, Belgium;
| |
Collapse
|
46
|
Bogun K, Peh E, Meyer-Kühling B, Hartmann J, Hirnet J, Plötz M, Kittler S. Investigating bacteriophages as a novel multiple-hurdle measure against Campylobacter: field trials in commercial broiler plants. Sci Rep 2024; 14:3182. [PMID: 38326411 PMCID: PMC10850366 DOI: 10.1038/s41598-024-53365-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 01/31/2024] [Indexed: 02/09/2024] Open
Abstract
Campylobacter mitigation along the food production chain is considered effective for minimizing the public health burden of human campylobacteriosis. This study is the first combining different measures in a multiple-hurdle approach, using drinking water additives and feed additives in single and combined application schemes in commercial broiler plants. Broiler chickens in the study groups were naturally contaminated with Campylobacter. Application of an organic acid blend via drinking water, consisting of sodium propionate, potassium sorbate, and sodium diacetate, resulted in significant reductions of up to 4.9 log10 CFU/mL in fecal samples and in cecal samples at slaughter. The application of a phage mixture, consisting of Fletchervirus phage NCTC 12673 and Firehammervirus phage vB_CcM-LmqsCPL1/1, resulted in reductions of up to 1.1 log10 CFU/mL in fecal samples 1 day after dosing. The sole administration of curcumin via feed resulted in small and inconsistent reductions. In the group receiving a combination of all tested measures, reductions of up to 1.1 log10 CFU/mL were observed. Based on the results of our field trials, it was shown that both the sole application and the combined application of mitigation measures in primary production can reduce the Campylobacter load in broiler chickens, while no synergism could be observed.
Collapse
Affiliation(s)
- Katrin Bogun
- Institute for Food Quality and Food Safety, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Elisa Peh
- Institute for Food Quality and Food Safety, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | | | | | - Juliane Hirnet
- Institute for Food Quality and Food Safety, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Madeleine Plötz
- Institute for Food Quality and Food Safety, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Sophie Kittler
- Institute for Food Quality and Food Safety, University of Veterinary Medicine Hannover, Foundation, Hannover, Germany.
| |
Collapse
|
47
|
Echterhof A, Dharmaraj T, McBride R, Berry J, Hopkins M, Selvakumar H, Miesel L, Chia JH, Lin KY, Shen CC, Lee YL, Yeh YC, Liao WT, Suh G, Blankenberg FG, Frymoyer AR, Bollyky PL. The contribution of neutrophils to bacteriophage clearance and pharmacokinetics in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.25.577154. [PMID: 38328123 PMCID: PMC10849746 DOI: 10.1101/2024.01.25.577154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
With the increasing prevalence of antimicrobial-resistant bacterial infections, there is great interest in using lytic bacteriophages (phages) to treat such infections. However, the factors that govern bacteriophage pharmacokinetics in vivo remain poorly understood. Here, we have examined the contribution of neutrophils, the most abundant phagocytes in the body, to the pharmacokinetics of intravenously administered bacteriophage in uninfected mice. A single dose of LPS-5, an antipseudomonal bacteriophage recently used in human clinical trials, was administered intravenously to both wild-type BALB/c and neutropenic ICR mice. Phage concentrations were assessed in peripheral blood and spleen at 0.5, 1, 2, 4, 8, 12, and 24 hours after administration by plaque assay and qPCR. We observed that the phage clearance is only minimally affected by neutropenia. Indeed, the half-life of phages in blood in BALB/c and ICR mice is 3.45 and 3.66 hours, respectively. These data suggest that neutrophil-mediated phagocytosis is not a major determinant of phage clearance. Conversely, we observed a substantial discrepancy in circulating phage levels over time when measured by qPCR versus plaque assay, suggesting that substantial functional inactivation of circulating phages occurs over time. These data indicate that circulating factors, but not neutrophils, inactivate intravenously administered phages.
Collapse
|
48
|
De Soir S, Parée H, Kamarudin NHN, Wagemans J, Lavigne R, Braem A, Merabishvili M, De Vos D, Pirnay JP, Van Bambeke F. Exploiting phage-antibiotic synergies to disrupt Pseudomonas aeruginosa PAO1 biofilms in the context of orthopedic infections. Microbiol Spectr 2024; 12:e0321923. [PMID: 38084971 PMCID: PMC10783084 DOI: 10.1128/spectrum.03219-23] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/20/2023] [Indexed: 01/13/2024] Open
Abstract
IMPORTANCE Biofilm-related infections are among the most difficult-to-treat infections in all fields of medicine due to their antibiotic tolerance and persistent character. In the field of orthopedics, these biofilms often lead to therapeutic failure of medical implantable devices and urgently need novel treatment strategies. This forthcoming article aims to explore the dynamic interplay between newly isolated bacteriophages and routinely used antibiotics and clearly indicates synergetic patterns when used as a dual treatment modality. Biofilms were drastically more reduced when both active agents were combined, thereby providing additional evidence that phage-antibiotic combinations lead to synergism and could potentially improve clinical outcome for affected patients.
Collapse
Affiliation(s)
- Steven De Soir
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
- Laboratory for Molecular and Cellular Technology (LabMCT), Queen Astrid Military Hospital, Neder-over-Heembeek, Belgium
| | - Hortence Parée
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Nur Hidayatul Nazirah Kamarudin
- Department of Materials Engineering, Biomaterials and Tissue Engineering Research Group, KU Leuven, Leuven, Belgium
- Department of Chemical and Process Engineering, Faculty of Engineering and Built Environment, Universiti Kebangsaan Malaysia, Bangi, Selangor, Malaysia
| | | | - Rob Lavigne
- Laboratory of Gene Technology, KU Leuven, Leuven, Belgium
| | - Annabel Braem
- Department of Materials Engineering, Biomaterials and Tissue Engineering Research Group, KU Leuven, Leuven, Belgium
| | - Maya Merabishvili
- Laboratory for Molecular and Cellular Technology (LabMCT), Queen Astrid Military Hospital, Neder-over-Heembeek, Belgium
| | - Daniel De Vos
- Laboratory for Molecular and Cellular Technology (LabMCT), Queen Astrid Military Hospital, Neder-over-Heembeek, Belgium
| | - Jean-Paul Pirnay
- Laboratory for Molecular and Cellular Technology (LabMCT), Queen Astrid Military Hospital, Neder-over-Heembeek, Belgium
| | - Françoise Van Bambeke
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
49
|
Pirnay JP, Merabishvili M, De Vos D, Verbeken G. Bacteriophage Production in Compliance with Regulatory Requirements. Methods Mol Biol 2024; 2734:89-115. [PMID: 38066364 DOI: 10.1007/978-1-0716-3523-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
In this chapter, we discuss production requirements for therapeutic bacteriophage preparations. We review the current regulatory expectancies and focus on pragmatic production processes, implementing relevant controls to ensure the quality, safety, and efficacy of the final products. The information disclosed in this chapter can also serve as a basis for discussions with competent authorities regarding the implementation of expedited bacteriophage product development and licensing pathways, taking into account some peculiarities of bacteriophages (as compared to conventional medicines), such as their specificity for, and co-evolution with, their bacterial hosts. To maximize the potential of bacteriophages as natural controllers of bacterial populations, the implemented regulatory frameworks and manufacturing processes should not only cater to defined bacteriophage products. But, they should also facilitate personalized approaches in which bacteriophages are selected ad hoc and even trained to target the patient's infecting bacterial strain(s), whether or not in combination with other antimicrobials such as antibiotics.
Collapse
Affiliation(s)
- Jean-Paul Pirnay
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium.
| | - Maia Merabishvili
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| | - Daniel De Vos
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| | - Gilbert Verbeken
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| |
Collapse
|
50
|
Merabishvili M, Pirnay JP, De Vos D. Guidelines to Compose an Ideal Bacteriophage Cocktail. Methods Mol Biol 2024; 2734:49-66. [PMID: 38066362 DOI: 10.1007/978-1-0716-3523-0_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Properly designed bacteriophage therapeutics are the cornerstone for a successful outcome of bacteriophage therapy. Here we present an overview of the different strategies and steps that can be taken to develop a bacteriophage cocktail that complies with relevant quality and safety requirements. It is based on empirical bacteriophage therapy knowledge from over a century of experience, more recently performed studies, and emerging technologies. We emphasize the selection of adequate bacteriophages and describe a modified Appelmans' method to improve the overall performance of therapeutic bacteriophages individually and collectively in the cocktail. We present two versions of the method, which differ from each other by the employed techniques to evaluate phage activity and synergy: photometric assessment of bacterial growth versus measurement of bacterial respiration via the Omnilog® system.
Collapse
Affiliation(s)
- Maia Merabishvili
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium.
| | - Jean-Paul Pirnay
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| | - Daniel De Vos
- Laboratory for Molecular and Cellular Technology, Queen Astrid Military Hospital, Brussels, Belgium
| |
Collapse
|