1
|
Tadesse M, Ali N, White M, Song L, Alberti F, Sagona AP. One-Two Punch: Phage-Antibiotic Synergy Observed against Staphylococcus aureus by Combining Pleurotin and Phage K. ACS OMEGA 2025; 10:12026-12036. [PMID: 40191302 PMCID: PMC11966286 DOI: 10.1021/acsomega.4c09831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/20/2025] [Accepted: 03/10/2025] [Indexed: 04/09/2025]
Abstract
The surge in antibiotic-resistant Staphylococcus aureus infections has been deemed a major public health concern. There is an urgent need for novel antimicrobial therapies, chemical and nonantibiotic. The basidiomycota-derived, secondary metabolite pleurotin has been shown to be effective against Gram-positive bacteria, while bacteriophages could be the ultimate nonantibiotic alternative. In this study, the combination of pleurotin and phage K targeting S. aureus was examined. Pleurotin was isolated from the basidiomycota fungus Hohenbuehelia grisea. The cytotoxicity of pleurotin was assessed in two human cell lines in comparison to pleuromutilin, vancomycin, and phage K. The antibiotics were then tested independently or in combination with phage K against two S. aureus strains. Cytotoxicity of pleurotin in human cells was comparable to vancomycin and pleuromutilin. Results suggest that adding phage K has a synergistic effect and can lower the MIC for pleurotin, pleuromutilin, and vancomycin. This demonstrates that pleurotin could be a viable antistaphylococcal drug.
Collapse
Affiliation(s)
| | - Nala Ali
- School
of Life Sciences, University of Warwick, Coventry CV4 7AL, U.K.
| | - Martha White
- School
of Life Sciences, University of Warwick, Coventry CV4 7AL, U.K.
| | - Lijiang Song
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
| | - Fabrizio Alberti
- School
of Life Sciences, University of Warwick, Coventry CV4 7AL, U.K.
| | - Antonia P. Sagona
- School
of Life Sciences, University of Warwick, Coventry CV4 7AL, U.K.
| |
Collapse
|
2
|
Piechowicz L, Kosznik-Kwaśnicka K, Kaźmierczak N, Grzenkowicz M, Stasiłojć M, Necel A, Werbowy O, Pałubicka A. Efficacy of Three Kayviruses Against Staphylococcus aureus Strains Isolated from COVID-19 Patients. Antibiotics (Basel) 2025; 14:257. [PMID: 40149068 PMCID: PMC11939781 DOI: 10.3390/antibiotics14030257] [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: 02/05/2025] [Revised: 02/21/2025] [Accepted: 03/01/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: The viral pandemic caused by the SARS-CoV-2 virus has affected millions of people. However, it was noticed that high mortality was often a result of bacterial co-infections. One of the main pathogens responsible for secondary infections in patients with viral respiratory tract infections, including COVID-19, is Staphylococcus aureus. In recent years, the number of infections caused by drug-resistant strains of S. aureus has been growing rapidly, often exceeding the number of infections caused by antibiotic-sensitive strains. In addition, biofilm-related infections are more difficult to treat due to the lower sensitivity of biofilm structure to antibiotics. Bacteriophages are seen as alternative treatment of bacterial infections. Therefore, in our work, we have analyzed the efficacy of three Kayviruses against S. aureus strains isolated from COVID-19 patients. Methods: We analyzed the ability of tested phages to remove S. aureus biofilm both from polystyrene plates as well as from the surface of pulmonary epithelial cells. Results: We have observed that tested Kayviruses had a broad host range. Furthermore, phages were able to effectively reduce biofilm biomass and number of viable cells in pure culture. During our research, none of the tested phages was shown to have a negative effect on cell viability and were able to inhibit the negative effect S. aureus had on cell condition. Conclusions: Our results show tested phages were effective in reducing the biofilm of S. aureus strains isolated from COVID-19 patients, had no adverse effect on lung epithelial cell viability. Therefore, it should be recognized that the properties of three studied Kayviruses give them an advantage in the selection of phages for treatment of staphylococcal infections.
Collapse
Affiliation(s)
- Lidia Piechowicz
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdańsk, Dębowa 25, 80-204 Gdansk, Poland; (N.K.); (M.G.); (A.N.)
| | - Katarzyna Kosznik-Kwaśnicka
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdańsk, Dębowa 25, 80-204 Gdansk, Poland; (N.K.); (M.G.); (A.N.)
| | - Natalia Kaźmierczak
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdańsk, Dębowa 25, 80-204 Gdansk, Poland; (N.K.); (M.G.); (A.N.)
| | - Milena Grzenkowicz
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdańsk, Dębowa 25, 80-204 Gdansk, Poland; (N.K.); (M.G.); (A.N.)
- Department of Microbiology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdansk, Poland;
| | - Małgorzata Stasiłojć
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology of University of Gdańsk and Medical University of Gdańsk, Dębinki 1, 80-211 Gdansk, Poland;
| | - Agnieszka Necel
- Department of Medical Microbiology, Faculty of Medicine, Medical University of Gdańsk, Dębowa 25, 80-204 Gdansk, Poland; (N.K.); (M.G.); (A.N.)
| | - Olesia Werbowy
- Department of Microbiology, Faculty of Biology, University of Gdańsk, Wita Stwosza 59, 80-308 Gdansk, Poland;
| | - Anna Pałubicka
- Specialist Hospital in Kościerzyna Sp. z o.o., Department of Laboratory and Microbiological Diagnostics, Kościerzyna, Alojzego Piechowskiego 36, 83-400 Koscierzyna, Poland;
| |
Collapse
|
3
|
Ghatbale P, Sah GP, Dunham S, Khong E, Blanc A, Monsibais A, Garcia A, Schooley RT, Cobián Güemes AG, Whiteson K, Pride DT. In vitro resensitization of multidrug-resistant clinical isolates of Enterococcus faecium and E. faecalis through phage-antibiotic synergy. Antimicrob Agents Chemother 2025; 69:e0074024. [PMID: 39699213 PMCID: PMC11823633 DOI: 10.1128/aac.00740-24] [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: 06/05/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024] Open
Abstract
Bacteriophages are an increasingly attractive option for the treatment of antibiotic-resistant infections, but their efficacy is difficult to discern due to the confounding effects of antibiotics. Phages are generally delivered in conjunction with antibiotics, and thus, when patients improve, it is unclear whether the phages, antibiotics, or both are responsible. This question is particularly relevant for enterococcus infections, as limited data suggest phages might restore antibiotic efficacy against resistant strains. Enterococci can develop high-level resistance to vancomycin, a primary treatment. We assessed clinical and laboratory isolates of Enterococcus faecium and Enterococcus faecalis to determine whether we could observe synergistic interactions between phages and antibiotics. We identified synergy between multiple phages and antibiotics including linezolid, ampicillin, and vancomycin. Notably, antibiotic susceptibility did not predict synergistic interactions with phages. Vancomycin-resistant isolates (n = 6) were eradicated by the vancomycin-phage combination as effectively as vancomycin-susceptible isolates (n = 2). Transcriptome analysis revealed significant gene expression changes under antibiotic-phage conditions, especially for linezolid and vancomycin, with upregulated genes involved in nucleotide and protein biosynthesis and downregulated stress response and prophage-related genes. While our results do not conclusively determine the mechanism of the observed synergistic interactions between antibiotics and phages, they do confirm and build upon previous research that observed these synergistic interactions. Our work highlights how using phages can restore the effectiveness of vancomycin against resistant isolates. This finding provides a promising, although unexpected, strategy for moving forward with phage treatments for vancomycin-resistant Enterococcus infections.
Collapse
Affiliation(s)
- Pooja Ghatbale
- Department of Pathology, University of California, San Diego, California, USA
| | - Govind Prasad Sah
- Department of Pathology, University of California, San Diego, California, USA
| | - Sage Dunham
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
| | - Ethan Khong
- Department of Pathology, University of California, San Diego, California, USA
| | - Alisha Blanc
- Department of Pathology, University of California, San Diego, California, USA
| | - Alisha Monsibais
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
| | - Andrew Garcia
- Department of Pathology, University of California, San Diego, California, USA
| | - Robert T. Schooley
- Department of Medicine, University of California, San Diego, California, USA
| | | | - Katrine Whiteson
- Department of Molecular Biology and Biochemistry, University of California, Irvine, California, USA
| | - David T. Pride
- Department of Pathology, University of California, San Diego, California, USA
- Department of Medicine, University of California, San Diego, California, USA
| |
Collapse
|
4
|
Zellner AA, Wirtz DC, Schildberg FA. In Vitro Efficacy of Phage Therapy Against Common Biofilm-forming Pathogens in Orthopedics and Trauma Surgery. ZEITSCHRIFT FUR ORTHOPADIE UND UNFALLCHIRURGIE 2025. [PMID: 39832775 DOI: 10.1055/a-2436-7394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Formation of biofilms by bacteria is a major challenge in a clinical setting. The importance of these biofilms increases in specialties where foreign bodies and prosthetic material are used. Orthopaedics is such a speciality and phage therapy could offer additional therapeutic options when dealing with biofilm infections.We conducted a systematic literature review using the PubMed database. We searched for phage activity against biofilms of the most common pathogens found in orthopaedics.The results of the systematic review were broken down into different categories and discussed accordingly. We concentrated on the time the biofilms were allowed to mature, and the surface they were grown on. In addition, we checked the efficacy of bacteriophages compared to antibiotics and when applied simultaneously with antibiotics. We also investigated the source of the phages, how they were tested for sensibility against the biofilms, as well the conditions (pH, temperature) under which they remained active and stable.The data suggests that the in vitro efficacy of phages does not change under a wide spectrum of temperature and pH. To further explore the use of bacteriophages in orthopaedics, we need further studies that test biofilms which matured for several weeks on surfaces that are common in arthroplasty and traumatology.
Collapse
Affiliation(s)
- Alberto Alfieri Zellner
- Klinik und Poliklinik für Orthopädie und Unfallchirurgie, Universitätsklinikum Bonn, Bonn, Deutschland
| | - Dieter Christian Wirtz
- Klinik und Poliklinik für Orthopädie und Unfallchirurgie, Universitätsklinikum Bonn, Bonn, Deutschland
| | | |
Collapse
|
5
|
Patra S, Saha S, Singh R, Tomar N, Gulati P. Biofilm battleground: Unveiling the hidden challenges, current approaches and future perspectives in combating biofilm associated bacterial infections. Microb Pathog 2025; 198:107155. [PMID: 39586337 DOI: 10.1016/j.micpath.2024.107155] [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: 07/31/2024] [Revised: 11/09/2024] [Accepted: 11/22/2024] [Indexed: 11/27/2024]
Abstract
A biofilm is a complex aggregation of microorganisms, either of the same or different species, that adhere to a surface and are encased in an extracellular polymeric substances (EPS) matrix. Quorum sensing (QS) and biofilm formation are closely linked, as QS genes regulate the development, maturation, and breakdown of biofilms. Inhibiting QS can be utilized as an effective approach to combat the impacts of biofilm infection. The impact of biofilms includes chronic infections, industrial biofouling, infrastructure corrosion, and environmental contamination as well. Therefore, a deep understanding of biofilms is crucial for enhancing public health, advancing industrial processes, safeguarding the environment, and deepening our knowledge of microbial life as well. This review aims to offer a comprehensive examination of challenges posed by bacterial biofilms, contemporary approaches and strategies for effectively eliminating biofilms, including the inhibition of quorum sensing pathways, while also focusing on emerging technologies and techniques for biofilm treatment. In addition, future research is projected to target the challenges associated with the bacterial biofilms, striving to develop new approaches and improve existing strategies for their effective control and eradication.
Collapse
Affiliation(s)
- Sandeep Patra
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Sumana Saha
- Gujarat Biotechnology University, Gandhinagar, Gujarat, India
| | - Randhir Singh
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| | - Nandini Tomar
- Department of Biotechnology, South Asian University, New Delhi, India
| | - Pallavi Gulati
- Ram Lal Anand College, University of Delhi, New Delhi, India.
| |
Collapse
|
6
|
Abdraimova NK, Shitikov EA, Bespiatykh DA, Gorodnichev RB, Klimina KM, Veselovsky VA, Boldyreva DI, Bogdanova AS, Klinov DV, Kornienko MA. Response of Staphylococcus aureus to combination of virulent bacteriophage vB_SauM-515A1 and linezolid. Front Microbiol 2024; 15:1519312. [PMID: 39760077 PMCID: PMC11695419 DOI: 10.3389/fmicb.2024.1519312] [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: 10/29/2024] [Accepted: 12/06/2024] [Indexed: 01/07/2025] Open
Abstract
The combined use of lytic bacteriophages with antibiotics is currently being explored as a strategy to enhance the effectiveness of infectious disease therapies, including those caused by Staphylococcus aureus. In this study, we investigated the synergistic potential of bacteriophage vB_SauM-515A1 (Herelleviridae family) and the first-line antibiotic linezolid against the methicillin-resistant S. aureus strain SA0413Rev. A checkerboard assay revealed a significant synergistic effect against planktonic cells (FIC = 0.225): a combination of 1/8 MIC of linezolid and 0.01 MOI of the bacteriophage completely inhibited bacterial growth. However, the impact on biofilm-associated cells depended on the treatment sequence. Sequential administration resulted in antagonism, while simultaneous application demonstrated a synergistic effect, as confirmed through scanning electron microscopy. Transcriptomic analysis of S. aureus SA0413Rev under the combined influence of linezolid (1/4 MIC) and bacteriophage vB_SauM-515A1 (10 MOI) predominantly reflected changes associated with productive bacteriophage infection, including alterations in nucleotide metabolism, activation of prophage regions, and virulence factors. Furthermore, both agents affected energy and carbon metabolism. These findings contribute to the development of combination therapy approaches for infections caused by S. aureus and highlight the importance of optimizing treatment conditions for maximal therapeutic efficacy.
Collapse
Affiliation(s)
- Narina K. Abdraimova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Egor A. Shitikov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Dmitry A. Bespiatykh
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Roman B. Gorodnichev
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Ksenia M. Klimina
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Vladimir A. Veselovsky
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Daria I. Boldyreva
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Alexandra S. Bogdanova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Moscow Institute of Physics and Technology, National Research University, Dolgoprudny, Russia
| | - Dmitry V. Klinov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Moscow Institute of Physics and Technology, National Research University, Dolgoprudny, Russia
| | - Maria A. Kornienko
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| |
Collapse
|
7
|
Taner F, Baddal B, Theodoridis L, Petrovski S. Biofilm Production in Intensive Care Units: Challenges and Implications. Pathogens 2024; 13:954. [PMID: 39599508 PMCID: PMC11597785 DOI: 10.3390/pathogens13110954] [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: 09/25/2024] [Revised: 10/23/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
The prevalence of infections amongst intensive care unit (ICU) patients is inevitably high, and the ICU is considered the epicenter for the spread of multidrug-resistant bacteria. Multiple studies have focused on the microbial diversity largely inhabiting ICUs that continues to flourish despite treatment with various antibiotics, investigating the factors that influence the spread of these pathogens, with the aim of implementing sufficient monitoring and infection control methods. Despite joint efforts from healthcare providers and policymakers, ICUs remain a hub for healthcare-associated infections. While persistence is a unique strategy used by these pathogens, multiple other factors can lead to persistent infections and antimicrobial tolerance in the ICU. Despite the recognition of the detrimental effects biofilm-producing pathogens have on ICU patients, overcoming biofilm formation in ICUs continues to be a challenge. This review focuses on various facets of ICUs that may contribute to and/or enhance biofilm production. A comprehensive survey of the literature reveals the apparent need for additional molecular studies to assist in understanding the relationship between biofilm regulation and the adaptive behavior of pathogens in the ICU environment. A better understanding of the interplay between biofilm production and antibiotic resistance within the environmental cues exhibited particularly by the ICU may also reveal ways to limit biofilm production and indivertibly control the spread of antibiotic-resistant pathogens in ICUs.
Collapse
Affiliation(s)
- Ferdiye Taner
- Department of Medical Microbiology and Clinical Microbiology, Faculty of Medicine, Near East University, 99138 Nicosia, Cyprus;
- DESAM Research Institute, Near East University, 99138 Nicosia, Cyprus
| | - Buket Baddal
- Department of Medical Microbiology and Clinical Microbiology, Faculty of Medicine, Near East University, 99138 Nicosia, Cyprus;
- DESAM Research Institute, Near East University, 99138 Nicosia, Cyprus
| | - Liana Theodoridis
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, VIC 3086, Australia; (L.T.); (S.P.)
| | - Steve Petrovski
- Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, VIC 3086, Australia; (L.T.); (S.P.)
| |
Collapse
|
8
|
Costa P, Pereira C, Romalde JL, Almeida A. A game of resistance: War between bacteria and phages and how phage cocktails can be the solution. Virology 2024; 599:110209. [PMID: 39186863 DOI: 10.1016/j.virol.2024.110209] [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: 05/29/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 08/28/2024]
Abstract
While phages hold promise as an antibiotic alternative, they encounter significant challenges in combating bacterial infections, primarily due to the emergence of phage-resistant bacteria. Bacterial defence mechanisms like superinfection exclusion, CRISPR, and restriction-modification systems can hinder phage effectiveness. Innovative strategies, such as combining different phages into cocktails, have been explored to address these challenges. This review delves into these defence mechanisms and their impact at each stage of the infection cycle, their challenges, and the strategies phages have developed to counteract them. Additionally, we examine the role of phage cocktails in the evolving landscape of antibacterial treatments and discuss recent studies that highlight the effectiveness of diverse phage cocktails in targeting essential bacterial receptors and combating resistant strains.
Collapse
Affiliation(s)
- Pedro Costa
- CESAM, Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| | - Carla Pereira
- CESAM, Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| | - Jesús L Romalde
- Department of Microbiology and Parasitology, CRETUS & CIBUS - Faculty of Biology, University of Santiago de Compostela, CP 15782 Santiago de Compostela, Spain.
| | - Adelaide Almeida
- CESAM, Department of Biology, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal.
| |
Collapse
|
9
|
Chen B, Moriarty T, Steenackers H, Vles G, Onsea J, Vackier T, Spriet I, Lavigne R, Richards RG, Metsemakers WJ. Exploring the potential of naturally occurring antimicrobials for managing orthopedic-device-related infections. J Bone Jt Infect 2024; 9:249-260. [PMID: 39539734 PMCID: PMC11555427 DOI: 10.5194/jbji-9-249-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 08/29/2024] [Indexed: 11/16/2024] Open
Abstract
Orthopedic-device-related infections (ODRIs) are challenging clinical complications that are often exacerbated by antibiotic resistance and biofilm formation. This review explores the efficacy of naturally occurring antimicrobials - including agents sourced from bacteria, fungi, viruses, animals, plants and minerals - against pathogens common in ODRIs. The limitations of traditional antibiotic agents are presented, and innovative naturally occurring antimicrobials, such as bacteriophage therapy and antimicrobial peptides, are evaluated with respect to their interaction with conventional antibiotics and antibiofilm efficacy. The integration of these natural agents into clinical practice could revolutionize ODRI treatment strategies, offering effective alternatives to conventional antibiotics and mitigating resistance development. However, the translation of these compounds from research into the clinic may require the substantial investment of intellectual and financial resources.
Collapse
Affiliation(s)
- Baixing Chen
- Department of Trauma Surgery, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | | | - Hans Steenackers
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG), KU Leuven, Leuven, Belgium
| | - Georges F. Vles
- Department of Orthopaedic Surgery, University Hospitals Leuven, Leuven, Belgium
- Institute for Orthopaedic Research and Training (IORT), KU Leuven, Leuven, Belgium
| | - Jolien Onsea
- Department of Trauma Surgery, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Thijs Vackier
- Department of Microbial and Molecular Systems, Centre of Microbial and Plant Genetics (CMPG), KU Leuven, Leuven, Belgium
| | - Isabel Spriet
- Pharmacy Department, University Hospitals Leuven, Leuven, Belgium
- Pharmacotherapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Belgium
| | - Rob Lavigne
- Laboratory of Gene Technology, Department of Biosystems, KU Leuven, Leuven, Belgium
| | | | - Willem-Jan Metsemakers
- Department of Trauma Surgery, University Hospitals Leuven, Leuven, Belgium
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
10
|
Kunz Coyne AJ, Eshaya M, Bleick C, Vader S, Biswas B, Wilson M, Deschenes MV, Alexander J, Lehman SM, Rybak MJ. Exploring synergistic and antagonistic interactions in phage-antibiotic combinations against ESKAPE pathogens. Microbiol Spectr 2024; 12:e0042724. [PMID: 39082827 PMCID: PMC11468199 DOI: 10.1128/spectrum.00427-24] [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: 04/02/2024] [Accepted: 06/14/2024] [Indexed: 09/21/2024] Open
Abstract
In the era of antimicrobial resistance, phage-antibiotic combinations offer a promising therapeutic option, yet research on their synergy and antagonism is limited. This study aims to assess these interactions, focusing on protein synthesis inhibitors and cell envelope-active agents against multidrug-resistant bacterial strains. We evaluated synergistic and antagonistic interactions in multidrug-resistant Staphylococcus aureus, Enterococcus faecium, and Pseudomonas aeruginosa strains. Phages were combined with protein synthesis inhibitors [linezolid (LZD), minocycline (MIN), gentamicin (GEN), and azithromycin (AZM)] or cell envelope-active agents [daptomycin (DAP), ceftaroline (CPT), and cefepime (FEP)]. Modified checkerboard minimum inhibitory concentration assays and 24-h time-kill analyses were conducted, alongside one-step growth curves to analyze phage growth kinetics. Statistical comparisons used one-way analysis of variance (ANOVA) and the Tukey test (P < 0.05). In the checkerboard and 24-h time-kill analyses (TKA) of S. aureus and E. faecium, phage-LZD and phage-MIN combinations were antagonistic (FIC > 4) while phage-DAP and phage-CPT were synergistic (FIC 0.5) (ANOVA range of mean differences 0.52-2.59 log10 CFU/mL; P < 0.001). For P. aeruginosa, phage-AZM was antagonistic (FIC > 4), phage-GEN was additive (FIC = 1), and phage-FEP was synergistic (ANOVA range of mean differences 1.04-1.95 log10 CFU/mL; P < 0.001). Phage growth kinetics were altered in the presence of LZD and MIN against S. aureus and in the presence of LZD against a single E. faecium strain (HOU503). Our findings indicate that select protein synthesis inhibitors may induce phage-antibiotic antagonism. However, this antagonism may not solely stem from changes in phage growth kinetics, warranting further investigation into the complex interplay among strains, phage attributes, and antibiotic mechanisms affecting bacterial inhibition.IMPORTANCEIn the face of escalating antimicrobial resistance, combining phages with antibiotics offers a promising avenue for treating infections unresponsive to traditional antibiotics. However, while studies have explored synergistic interactions, less attention has been given to potential antagonism and its impact on phage growth kinetics. This research evaluates the interplay between phages and antibiotics, revealing both synergistic and antagonistic patterns across various bacterial strains and shedding light on the complex dynamics that influence treatment efficacy. Understanding these interactions is crucial for optimizing combination therapies and advancing phage therapy as a viable solution for combating antimicrobial resistance.
Collapse
Affiliation(s)
- Ashlan J. Kunz Coyne
- Anti-Infective
Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum
College of Pharmacy and Health Sciences, Wayne State
University, Detroit,
Michigan, USA
| | - Mirna Eshaya
- Anti-Infective
Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum
College of Pharmacy and Health Sciences, Wayne State
University, Detroit,
Michigan, USA
| | - Callan Bleick
- Anti-Infective
Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum
College of Pharmacy and Health Sciences, Wayne State
University, Detroit,
Michigan, USA
| | - Samantha Vader
- Anti-Infective
Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum
College of Pharmacy and Health Sciences, Wayne State
University, Detroit,
Michigan, USA
| | - Biswajit Biswas
- Naval Medical Research
Center-Fort Detrick,
Frederick, Maryland,
USA
| | - Melanie Wilson
- Naval Medical Research
Center-Fort Detrick,
Frederick, Maryland,
USA
- Leidos,
Reston, Virginia, USA
| | - Michael V. Deschenes
- Naval Medical Research
Center-Fort Detrick,
Frederick, Maryland,
USA
- Leidos,
Reston, Virginia, USA
| | - Jose Alexander
- Department of
Microbiology, Virology and Immunology, AdventHealth Central
Florida, Orlando,
Florida, USA
| | - Susan M. Lehman
- Center for Biologics
Evaluation and Research, US Food and Drug
Administration, Silver Spring,
Maryland, USA
| | - Michael J. Rybak
- Anti-Infective
Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum
College of Pharmacy and Health Sciences, Wayne State
University, Detroit,
Michigan, USA
- Division of Infectious
Diseases, Department of Medicine, School of Medicine, Wayne State
University, Detroit,
Michigan, USA
- Department of
Pharmacy, Detroit Medical Center,
Detroit, Michigan, USA
| |
Collapse
|
11
|
Khosravi A, Chen Q, Echterhof A, Koff JL, Bollyky PL. Phage Therapy for Respiratory Infections: Opportunities and Challenges. Lung 2024; 202:223-232. [PMID: 38772946 PMCID: PMC11570333 DOI: 10.1007/s00408-024-00700-7] [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: 01/29/2024] [Accepted: 04/13/2024] [Indexed: 05/23/2024]
Abstract
We are entering the post-antibiotic era. Antimicrobial resistance (AMR) is a critical problem in chronic lung infections resulting in progressive respiratory failure and increased mortality. In the absence of emerging novel antibiotics to counter AMR infections, bacteriophages (phages), viruses that infect bacteria, have become a promising option for chronic respiratory infections. However, while personalized phage therapy is associated with improved outcomes in individual cases, clinical trials demonstrating treatment efficacy are lacking, limiting the therapeutic potential of this approach for respiratory infections. In this review, we address the current state of phage therapy for managing chronic respiratory diseases. We then discuss how phage therapy may address major microbiologic obstacles which hinder disease resolution of chronic lung infections with current antibiotic-based treatment practices. Finally, we highlight the challenges that must be addressed for successful phage therapy clinical trials. Through this discussion, we hope to expand on the potential of phages as an adjuvant therapy in chronic lung infections, as well as the microbiologic challenges that need to be addressed for phage therapy to expand beyond personalized salvage therapy.
Collapse
Affiliation(s)
- Arya Khosravi
- Division of Infectious Diseases, School of Medicine, Stanford University, Stanford, CA, USA.
- Division of Infectious Diseases, Department of Medicine, Stanford University, 279 Campus Drive, Beckman Center, Room B237, Stanford, CA, 94305, USA.
| | - Qingquan Chen
- Division of Infectious Diseases, School of Medicine, Stanford University, Stanford, CA, USA
| | - Arne Echterhof
- Division of Infectious Diseases, School of Medicine, Stanford University, Stanford, CA, USA
| | - Jonathan L Koff
- Section of Pulmonary, Critical Care & Sleep Medicine, School of Medicine, Yale University, New Haven, CT, USA
| | - Paul L Bollyky
- Division of Infectious Diseases, School of Medicine, Stanford University, Stanford, CA, USA
| |
Collapse
|
12
|
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
|
13
|
Kunz Coyne AJ, Stamper K, Bleick C, Kebriaei R, Lehman SM, Rybak MJ. Synergistic bactericidal effects of phage-enhanced antibiotic therapy against MRSA biofilms. Microbiol Spectr 2024; 12:e0321223. [PMID: 38411110 PMCID: PMC10986480 DOI: 10.1128/spectrum.03212-23] [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: 01/10/2024] [Indexed: 02/28/2024] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) causes biofilm-related medical device infections. Phage-antibiotic combinations offer potential therapy due to proven in vitro antibiofilm efficacy. We evaluated phage-antibiotic synergy against biofilms using modified checkerboard and 24-h time-kill assays. Humanized-simulated daptomycin (DAP) (10, 8, and 6 mg/kg q24h) and ceftaroline (CPT) (600 mg q12h) were combined with Intesti13, Sb-1, and Romulus phages (tMOI 1, q12h). Assays were conducted in 168-h biofilm reactor models against DAP non-susceptible (DNS) vancomycin intermediate S. aureus (VISA) MRSA D712 and DAP-susceptible MRSA 8014. Synergistic activity and bactericidal activity were defined as ≥2log10 CFU/mL reduction from antibiotic-only regimens and ≥3log10 CFU/mL decrease from baseline at 24 h. Differences were analyzed by one-way analysis of variance with Tukey's post hoc test (P ≤ 0.05 is considered significant). Surviving bacteria were examined for antibiotic minimum biofilm inhibitory concentration (MBIC) changes and phage susceptibility. In 168-h biofilm models, humanized DAP 10 mg/kg + CPT, combined with a 2-phage cocktail (Intesti13 + Sb-1) against D712, and a 3-phage cocktail (Intesti13 + Sb-1 + Romulus) against 8014, demonstrated synergistic bactericidal activity. At 168 h, bacteria were minimally detectable [2log10 CFU/cm2 (-Δ4.23 and -Δ4.42 log10 CFU/cm2; both P < 0.001)]. Antibiotic MBIC remained unchanged compared to baseline across various time points. None of the tested bacteria at 168 h exhibited complete phage resistance. This study reveals bactericidal efficacy of DAP + CPT with 2-phage and 3-phage cocktails against DNS VISA and MRSA isolates (D712 and 8014) in biofilm models, maintaining susceptibility. Further research is needed for diverse strains and durations, aligning with infection care. IMPORTANCE The prevalence of biofilm-associated medical device infections caused by methicillin-resistant Staphylococcus aureus (MRSA) presents a pressing medical challenge. The latest research demonstrates the potential of phage-antibiotic combinations (PACs) as a promising solution, notably in vitro antibiofilm efficacy. By adopting modified checkerboard and 24-h time-kill assays, the study investigated the synergistic action of phages combined with humanized-simulated doses of daptomycin (DAP) and ceftaroline (CPT). The results were promising: a combination of DAP, CPT, and either a 2-phage or 3-phage cocktail effectively exhibited bactericidal activity against both DAP non-susceptible vancomycin intermediate S. aureus MRSA and DAP-susceptible MRSA strains within 168-h biofilm models. Moreover, post-treatment evaluations revealed no discernible rise in antibiotic resistance or complete phage resistance. This pioneering work suggests the potential of PACs in addressing MRSA biofilm infections, setting the stage for further expansive research tailored to diverse bacterial strains and treatment durations.
Collapse
Affiliation(s)
- Ashlan J. Kunz Coyne
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Kyle Stamper
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Callan Bleick
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Razieh Kebriaei
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Susan M. Lehman
- Center for Biologics Evaluation and Research, US Food and Drug Administration, Silver Spring, Maryland, USA
| | - Michael J. Rybak
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
- Department of Pharmacy Services, Detroit Receiving Hospital, Detroit Medical Center, Detroit, Michigan, USA
- Department of Medicine, Division of Infectious Diseases, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
14
|
Iaconis A, De Plano LM, Caccamo A, Franco D, Conoci S. Anti-Biofilm Strategies: A Focused Review on Innovative Approaches. Microorganisms 2024; 12:639. [PMID: 38674584 PMCID: PMC11052202 DOI: 10.3390/microorganisms12040639] [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: 03/04/2024] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 04/28/2024] Open
Abstract
Biofilm (BF) can give rise to systemic infections, prolonged hospitalization times, and, in the worst case, death. This review aims to provide an overview of recent strategies for the prevention and destruction of pathogenic BFs. First, the main phases of the life cycle of BF and maturation will be described to identify potential targets for anti-BF approaches. Then, an approach acting on bacterial adhesion, quorum sensing (QS), and the extracellular polymeric substance (EPS) matrix will be introduced and discussed. Finally, bacteriophage-mediated strategies will be presented as innovative approaches against BF inhibition/destruction.
Collapse
Affiliation(s)
- Antonella Iaconis
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (A.I.); (L.M.D.P.); (A.C.)
| | - Laura Maria De Plano
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (A.I.); (L.M.D.P.); (A.C.)
| | - Antonella Caccamo
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (A.I.); (L.M.D.P.); (A.C.)
| | - Domenico Franco
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (A.I.); (L.M.D.P.); (A.C.)
| | - Sabrina Conoci
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences (ChiBioFarAm), University of Messina, Viale F. Stagno d’Alcontres 31, 98166 Messina, Italy; (A.I.); (L.M.D.P.); (A.C.)
- Department of Chemistry “Giacomo Ciamician”, Alma Mater Studiorum—University of Bologna, 40126 Bologna, Italy
- URT Lab Sens Beyond Nano—CNR-DSFTM, Department of Physical Sciences and Technologies of Matter, University of Messina, Viale F. Stagno D’Alcontres 31, 98166 Messina, Italy
| |
Collapse
|
15
|
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
|
16
|
Chen B, Benavente LP, Chittò M, Wychowaniec JK, Post V, D'Este M, Constant C, Zeiter S, Feng W, Moreno MG, Trampuz A, Wagemans J, Onsea J, Richards RG, Lavigne R, Moriarty TF, Metsemakers WJ. Alginate microbeads and hydrogels delivering meropenem and bacteriophages to treat Pseudomonas aeruginosa fracture-related infections. J Control Release 2023; 364:159-173. [PMID: 37866403 DOI: 10.1016/j.jconrel.2023.10.029] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/18/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023]
Abstract
Bacteriophage (phage) therapy has shown promise in treating fracture-related infection (FRI); however, questions remain regarding phage efficacy against biofilms, phage-antibiotic interaction, administration routes and dosing, and the development of phage resistance. The goal of this study was to develop a dual antibiotic-phage delivery system containing hydrogel and alginate microbeads loaded with a phage cocktail plus meropenem and evaluate efficacy against muti-drug resistant Pseudomonas aeruginosa. Two phages (FJK.R9-30 and MK.R3-15) displayed enhanced antibiotic activity against P. aeruginosa biofilms when tested in combination with meropenem. The antimicrobial activity of both antibiotic and phage was retained for eight days at 37 °C in dual phage and antibiotic loaded hydrogel with microbeads (PA-HM). In a mouse FRI model, phages were recovered from all tissues within all treatment groups receiving dual PA-HM. Moreover, animals that received the dual PA-HM either with or without systemic antibiotics had less incidence of phage resistance and less serum neutralization compared to phages in saline. The dual PA-HM could reduce bacterial load in soft tissue when combined with systemic antibiotics, although the infection was not eradicated. The use of alginate microbeads and injectable hydrogel for controlled release of phages and antibiotics, leads to the reduced development of phage resistance and lower exposure to the adaptive immune system, which highlights the translational potential of the dual PA-HM. However, further optimization of phage therapy and its delivery system is necessary to achieve higher bacterial killing activity in vivo in the future.
Collapse
Affiliation(s)
- Baixing Chen
- Department of Trauma Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Development and Regeneration, KU Leuven, Leuven, Belgium; AO Research Institute Davos, Davos, Switzerland
| | - Luis Ponce Benavente
- Center for Musculoskeletal Surgery Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | | | | | | | | | | | | | - Wenli Feng
- AO Research Institute Davos, Davos, Switzerland
| | - Mercedes González Moreno
- Center for Musculoskeletal Surgery Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Andrej Trampuz
- Center for Musculoskeletal Surgery Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | | | - Jolien Onsea
- Department of Trauma Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | | | - Rob Lavigne
- Laboratory of Gene Technology, KU Leuven, Leuven, Belgium
| | | | - Willem-Jan Metsemakers
- Department of Trauma Surgery, University Hospitals Leuven, Leuven, Belgium; Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
17
|
Mukhopadhyay S, Zhang P, To KKW, Liu Y, Bai C, Leung SSY. Sequential treatment effects on phage-antibiotic synergistic application against multi-drug-resistant Acinetobacter baumannii. Int J Antimicrob Agents 2023; 62:106951. [PMID: 37574030 DOI: 10.1016/j.ijantimicag.2023.106951] [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: 03/16/2023] [Revised: 07/05/2023] [Accepted: 08/05/2023] [Indexed: 08/15/2023]
Abstract
Bacteriophage (phage) therapy, exploiting phages which are the natural enemies of bacteria, has been re-introduced to treat multidrug-resistant (MDR) bacterial infections. However, some intrinsic drawbacks of phages are overshadowing their clinical use, particularly the narrow host spectrum and rapid emergence of resistance upon treatment. The use of phage-antibiotic combinations exhibiting synergistic bacterial killing [termed 'phage-antibiotic synergy' (PAS)] has therefore been proposed. It is well reported that the types and doses of phages and antibiotics are critical in achieving PAS. However, the impact of treatment order has received less research attention. As such, this study used an Acinetobacter baumannii phage vB_AbaM-IME-AB2 and colistin as a model PAS combination to elucidate the order effects in-vitro. While application of the phage 8 h before colistin treatment demonstrated the greatest antibacterial synergy, it failed to prevent the development of phage resistance. On the other hand, simultaneous application and antibiotic followed by phage application were able to suppress/delay the development of resistance effectively, and simultaneous application demonstrated superior antibacterial and antibiofilm activities. Further in-vivo investigation is required to confirm the impact of treatment order on PAS.
Collapse
Affiliation(s)
- Subhankar Mukhopadhyay
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region
| | - Pengfei Zhang
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region
| | - Kenneth K W To
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region
| | - Yannan Liu
- Emergency Medicine Clinical Research Centre, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Changqing Bai
- Department of Respiratory, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Guangdong, China
| | - Sharon S Y Leung
- School of Pharmacy, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong Special Administrative Region.
| |
Collapse
|
18
|
Taha M, Arnaud T, Lightly TJ, Peters D, Wang L, Chen W, Cook BWM, Theriault SS, Abdelbary H. Combining bacteriophage and vancomycin is efficacious against MRSA biofilm-like aggregates formed in synovial fluid. Front Med (Lausanne) 2023; 10:1134912. [PMID: 37359001 PMCID: PMC10289194 DOI: 10.3389/fmed.2023.1134912] [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: 12/31/2022] [Accepted: 05/23/2023] [Indexed: 06/28/2023] Open
Abstract
Background Biofilm formation is a major clinical challenge contributing to treatment failure of periprosthetic joint infection (PJI). Lytic bacteriophages (phages) can target biofilm associated bacteria at localized sites of infection. The aim of this study is to investigate whether combination therapy of phage and vancomycin is capable of clearing Staphylococcus aureus biofilm-like aggregates formed in human synovial fluid. Methods In this study, S. aureus BP043, a PJI clinical isolate was utilized. This strain is a methicillin-resistant S. aureus (MRSA) biofilm-former. Phage Remus, known to infect S. aureus, was selected for the treatment protocol. BP043 was grown as aggregates in human synovial fluid. The characterization of S. aureus aggregates was assessed for structure and size using scanning electron microscopy (SEM) and flow cytometry, respectively. Moreover, the formed aggregates were subsequently treated in vitro with: (a) phage Remus [∼108 plaque-forming units (PFU)/ml], (b) vancomycin (500 μg/ml), or (c) phage Remus (∼108 PFU/ml) followed by vancomycin (500 μg/ml), for 48 h. Bacterial survival was quantified by enumeration [colony-forming units (CFU)/ml]. The efficacy of phage and vancomycin against BP043 aggregates was assessed in vivo as individual treatments and in combination. The in vivo model utilized Galleria mellonella larvae which were infected with BP043 aggregates pre-formed in synovial fluid. Results Scanning electron microscopy (SEM) images and flow cytometry data demonstrated the ability of human synovial fluid to promote formation of S. aureus aggregates. Treatment with Remus resulted in significant reduction in viable S. aureus residing within the synovial fluid aggregates compared to the aggregates that did not receive Remus (p < 0.0001). Remus was more efficient in eliminating viable bacteria within the aggregates compared to vancomycin (p < 0.0001). Combination treatment of Remus followed by vancomycin was more efficacious in reducing bacterial load compared to using either Remus or vancomycin alone (p = 0.0023, p < 0.0001, respectively). When tested in vivo, this combination treatment also resulted in the highest survival rate (37%) 96 h post-treatment, compared to untreated larvae (3%; p < 0.0001). Conclusion We demonstrate that combining phage Remus and vancomycin led to synergistic interaction against MRSA biofilm-like aggregates in vitro and in vivo.
Collapse
Affiliation(s)
- Mariam Taha
- The Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Division of Orthopedic Surgery, The Ottawa Hospital, Ottawa, ON, Canada
| | - Tia Arnaud
- Cytophage Technologies Inc., Winnipeg, MB, Canada
- Department of Microbiology, The University of Manitoba, Winnipeg, MB, Canada
| | | | - Danielle Peters
- Human Health Therapeutics Research Center, National Research Council Canada, Ottawa, ON, Canada
| | - Liyuan Wang
- Cell Biology and Image Acquisition, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Wangxue Chen
- Human Health Therapeutics Research Center, National Research Council Canada, Ottawa, ON, Canada
- Department of Biology, Brock University, St. Catharines, ON, Canada
| | | | - Steven S. Theriault
- Cytophage Technologies Inc., Winnipeg, MB, Canada
- Department of Microbiology, The University of Manitoba, Winnipeg, MB, Canada
| | - Hesham Abdelbary
- The Ottawa Hospital Research Institute, Ottawa, ON, Canada
- Division of Orthopedic Surgery, The Ottawa Hospital, Ottawa, ON, Canada
- Department of Surgery, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
19
|
Fungo GBN, Uy JCW, Porciuncula KLJ, Candelario CMA, Chua DPS, Gutierrez TAD, Clokie MRJ, Papa DMD. "Two Is Better Than One": The Multifactorial Nature of Phage-Antibiotic Combinatorial Treatments Against ESKAPE-Induced Infections. PHAGE (NEW ROCHELLE, N.Y.) 2023; 4:55-67. [PMID: 37350995 PMCID: PMC10282822 DOI: 10.1089/phage.2023.0007] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/24/2023]
Abstract
Phage-antibiotic synergy (PAS) has been extensively explored over the past decade, with the aim of developing more effective treatments against multidrug-resistant organisms. However, it remains unclear how to effectively combine these two approaches. To address this uncertainty, we assessed four main aspects of PAS interactions in this review, seeking to identify commonalities of combining treatments within and between bacterial species. We examined all literature on PAS efficacy toward ESKAPE pathogens and present an analysis of the data in papers focusing on: (1) order of treatment, (2) dose of both phage and antibiotics, (3) mechanism of action, and (4) viability of transfer from in vivo or animal model trials to clinical applications. Our analysis indicates that there is little consistency within phage-antibiotic therapy regimens, suggesting that highly individualized treatment regimens should be used. We propose a set of experimental studies to address these research gaps. We end our review with suggestions on how to improve studies on phage-antibiotic combination therapy to advance this field.
Collapse
Affiliation(s)
- Gale Bernice N. Fungo
- Department of Biological Sciences, College of Science, University of Santo Tomas, Manila, Philippines
- Bacteriophage Ecology, Aquaculture, Therapy and Systematics (BEATS) Research Group, University of Santo Tomas, Manila, Philippines
- Research Center for Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
| | - John Christian W. Uy
- Department of Biological Sciences, College of Science, University of Santo Tomas, Manila, Philippines
- Bacteriophage Ecology, Aquaculture, Therapy and Systematics (BEATS) Research Group, University of Santo Tomas, Manila, Philippines
- Research Center for Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
| | - Kristiana Louise J. Porciuncula
- Department of Biological Sciences, College of Science, University of Santo Tomas, Manila, Philippines
- Bacteriophage Ecology, Aquaculture, Therapy and Systematics (BEATS) Research Group, University of Santo Tomas, Manila, Philippines
- Research Center for Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
| | - Chiarah Mae A. Candelario
- Department of Biological Sciences, College of Science, University of Santo Tomas, Manila, Philippines
- Bacteriophage Ecology, Aquaculture, Therapy and Systematics (BEATS) Research Group, University of Santo Tomas, Manila, Philippines
- Research Center for Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
| | - Deneb Philip S. Chua
- Department of Biological Sciences, College of Science, University of Santo Tomas, Manila, Philippines
- Bacteriophage Ecology, Aquaculture, Therapy and Systematics (BEATS) Research Group, University of Santo Tomas, Manila, Philippines
- Research Center for Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
| | - Tracey Antaeus D. Gutierrez
- Bacteriophage Ecology, Aquaculture, Therapy and Systematics (BEATS) Research Group, University of Santo Tomas, Manila, Philippines
- Research Center for Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
| | | | - Donna May D. Papa
- Department of Biological Sciences, College of Science, University of Santo Tomas, Manila, Philippines
- Bacteriophage Ecology, Aquaculture, Therapy and Systematics (BEATS) Research Group, University of Santo Tomas, Manila, Philippines
- Research Center for Natural and Applied Sciences, University of Santo Tomas, Manila, Philippines
| |
Collapse
|
20
|
Martin I, Morales S, Alton EWFW, Davies JC. Lytic Bacteriophage Is a Promising Adjunct to Common Antibiotics across Cystic Fibrosis Clinical Strains and Culture Models of Pseudomonas aeruginosa Infection. Antibiotics (Basel) 2023; 12:antibiotics12030593. [PMID: 36978460 PMCID: PMC10044644 DOI: 10.3390/antibiotics12030593] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/09/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
Bacteriophages (phages) are antimicrobials with resurgent interest that are being investigated for the treatment of antibiotic refractory infection, including for Pseudomonas aeruginosa (Pa) lung infection in cystic fibrosis (CF). In vitro work supports the use of this therapy in planktonic and biofilm culture models; however, consistent data are lacking for efficacy across different clinical Pa strains, culture models, and in combination with antibiotics in clinical use. We first examined the efficacy of a 4-phage cocktail as an adjunct to our CF centre’s first-line systemic combination antibiotic therapy (ceftazidime + tobramycin) for 16 different clinical Pa strains and then determined subinhibitory interactions for a subset of these strains with each antibiotic in planktonic and biofilm culture. When a 4-phage cocktail (4 × 108 PFU/mL) was added to a ceftazidime-tobramycin combination (ceftazidime 16 mg/mL + tobramycin 8 mg/mL), we observed a 1.7-fold and 1.3-fold reduction in biofilm biomass and cell viability, respectively. The four most antibiotic resistant strains in biofilm were very susceptible to phage treatment. When subinhibitory concentrations of antibiotics and phages were investigated, we observed additivity/synergy as well as antagonism/inhibition of effect that varied across the clinical strains and culture model. In general, more additivity was seen with the phage-ceftazidime combination than with phage-tobramycin, particularly in biofilm culture, where no instances of additivity were seen when phages were combined with tobramycin. The fact that different bacterial strains were susceptible to phage treatment when compared to standard antibiotics is promising and these results may be relevant to ongoing clinical trials exploring the use of phages, in particular in the selection of subjects for clinical trials.
Collapse
Affiliation(s)
- Isaac Martin
- National Heart and Lung Institute, Imperial College London, Emmanuel Kaye Building, London SW3 6LY, UK
- Royal Brompton Hospital, Part of Guy’s and St. Thomas’ Trust, Sydney St., London SW3 6NP, UK
- Department of Paediatrics and Translational Medicine, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada
- Correspondence: ; Tel.: +1-647-223-7782
| | | | - Eric W. F. W. Alton
- National Heart and Lung Institute, Imperial College London, Emmanuel Kaye Building, London SW3 6LY, UK
- Royal Brompton Hospital, Part of Guy’s and St. Thomas’ Trust, Sydney St., London SW3 6NP, UK
| | - Jane C. Davies
- National Heart and Lung Institute, Imperial College London, Emmanuel Kaye Building, London SW3 6LY, UK
- Royal Brompton Hospital, Part of Guy’s and St. Thomas’ Trust, Sydney St., London SW3 6NP, UK
| |
Collapse
|
21
|
Fedorov E, Samokhin A, Kozlova Y, Kretien S, Sheraliev T, Morozova V, Tikunova N, Kiselev A, Pavlov V. Short-Term Outcomes of Phage-Antibiotic Combination Treatment in Adult Patients with Periprosthetic Hip Joint Infection. Viruses 2023; 15:v15020499. [PMID: 36851713 PMCID: PMC9964274 DOI: 10.3390/v15020499] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/26/2023] [Accepted: 02/04/2023] [Indexed: 02/17/2023] Open
Abstract
Implant-associated infections are the most costly problem in modern orthopedics due to the continued increase in the occurrence of antibiotic-resistant bacterial strains that requires the development of new effective antimicrobials. A non-randomized, prospective, open-label, with historical control study on the use of combined phage/antibiotic therapy of periprosthetic joint infection (PJI) was carried out. Forty-five adult patients with deep PJI of the hip joint were involved in the study, with a 12-month follow-up after one-stage revision surgery. Patients from a prospective study group (SG, n = 23) were treated with specific phage preparation and etiotropic antibiotics, whereas patients from a retrospective comparator group (CG, n = 22) received antibiotics only. The rate of PJI relapses in the SG was eight times less than that in the CG: one case (4.5%) versus eight cases (36.4%), p = 0.021. The response rate to treatment was 95.5% (95% confidence interval (CI) = 0.7511-0.9976) in the SG and only 63.6% (95% CI = 0.4083-0.8198) in the CG. The odds ratio for PJI relapse in patients of the SG was 0.083 (95% CI = 0.009-0.742), which was almost 12 times lower than that in the CG. The obtained results support the efficacy of the combined phage-antibiotic treatment of PJI.
Collapse
Affiliation(s)
- Eugeny Fedorov
- Orthopedics Department, Novosibirsk Research Institute of Traumatology and Orthopedics, 630091 Novosibirsk, Russia
| | - Alexander Samokhin
- Biotechnology Department, Novosibirsk State Technical University, 630073 Novosibirsk, Russia
- Correspondence: (A.S.); (N.T.)
| | - Yulia Kozlova
- Laboratory of Molecular Microbiology, Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Svetlana Kretien
- Orthopedics Department, Novosibirsk Research Institute of Traumatology and Orthopedics, 630091 Novosibirsk, Russia
| | - Taalai Sheraliev
- Orthopedics Department, Novosibirsk Research Institute of Traumatology and Orthopedics, 630091 Novosibirsk, Russia
| | - Vera Morozova
- Laboratory of Molecular Microbiology, Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - Nina Tikunova
- Laboratory of Molecular Microbiology, Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, 630090 Novosibirsk, Russia
- Correspondence: (A.S.); (N.T.)
| | - Alexey Kiselev
- Biostatistics Department, Bekhterev National Medical Research Center for Psychiatry and Neurology of the Ministry of Health of the Russian Federation, 192019 Saint-Petersburg, Russia
| | - Vitaliy Pavlov
- Orthopedics Department, Novosibirsk Research Institute of Traumatology and Orthopedics, 630091 Novosibirsk, Russia
| |
Collapse
|
22
|
Joo H, Wu SM, Soni I, Wang-Crocker C, Matern T, Beck JP, Loc-Carrillo C. Phage and Antibiotic Combinations Reduce Staphylococcus aureus in Static and Dynamic Biofilms Grown on an Implant Material. Viruses 2023; 15:v15020460. [PMID: 36851674 PMCID: PMC9963128 DOI: 10.3390/v15020460] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/28/2023] [Accepted: 01/31/2023] [Indexed: 02/10/2023] Open
Abstract
Staphylococcus aureus causes the majority of implant-related infections. These infections present as biofilms, in which bacteria adhere to the surface of foreign materials and form robust communities that are resilient to the human immune system and antibiotic drugs. The heavy use of broad-spectrum antibiotics against these pathogens disturbs the host's microbiome and contributes to the growing problem of antibiotic-resistant infections. The use of bacteriophages as antibacterial agents is a potential alternative therapy. In this study, bioluminescent strains of S. aureus were grown to form 48-h biofilms on polyether ether ketone (PEEK), a material used to manufacture orthopaedic implants, in either static or dynamic growth conditions. Biofilms were treated with vancomycin, staphylococcal phage, or a combination of the two. We showed that vancomycin and staph phages were able to independently reduce the total bacterial load. Most phage-antibiotic combinations produced greater log reductions in surviving bacteria compared to single-agent treatments, suggesting antimicrobial synergism. In addition to demonstrating the efficacy of combining vancomycin and staph phage, our results demonstrate the importance of growth conditions in phage-antibiotic combination studies. Dynamic biofilms were found to have a substantial impact on apparent treatment efficacy, as they were more resilient to combination treatments than static biofilms.
Collapse
Affiliation(s)
- Hyonoo Joo
- Micro-Phage Laboratory, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
- Department of Veterans Affairs, Salt Lake City Health Care System, Salt Lake City, UT 84148, USA
| | - Sijia M. Wu
- Department of Veterans Affairs, Salt Lake City Health Care System, Salt Lake City, UT 84148, USA
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84108, USA
| | - Isha Soni
- Micro-Phage Laboratory, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84108, USA
| | - Caroline Wang-Crocker
- Micro-Phage Laboratory, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84108, USA
| | - Tyson Matern
- Micro-Phage Laboratory, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84108, USA
| | - James Peter Beck
- Department of Veterans Affairs, Salt Lake City Health Care System, Salt Lake City, UT 84148, USA
- Department of Orthopaedics, University of Utah, Salt Lake City, UT 84108, USA
| | - Catherine Loc-Carrillo
- Micro-Phage Laboratory, Department of Internal Medicine, University of Utah, Salt Lake City, UT 84132, USA
- Department of Veterans Affairs, Salt Lake City Health Care System, Salt Lake City, UT 84148, USA
- Correspondence:
| |
Collapse
|
23
|
Phage Therapy as an Alternative Treatment Modality for Resistant Staphylococcus aureus Infections. Antibiotics (Basel) 2023; 12:antibiotics12020286. [PMID: 36830196 PMCID: PMC9952150 DOI: 10.3390/antibiotics12020286] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/25/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
The production and use of antibiotics increased significantly after the Second World War due to their effectiveness against bacterial infections. However, bacterial resistance also emerged and has now become an important global issue. Those most in need are typically high-risk and include individuals who experience burns and other wounds, as well as those with pulmonary infections caused by antibiotic-resistant bacteria, such as Pseudomonas aeruginosa, Acinetobacter sp, and Staphylococci. With investment to develop new antibiotics waning, finding and developing alternative therapeutic strategies to tackle this issue is imperative. One option remerging in popularity is bacteriophage (phage) therapy. This review focuses on Staphylococcus aureus and how it has developed resistance to antibiotics. It also discusses the potential of phage therapy in this setting and its appropriateness in high-risk people, such as those with cystic fibrosis, where it typically forms a biofilm.
Collapse
|
24
|
Mitropoulou G, Koutsokera A, Csajka C, Blanchon S, Sauty A, Brunet JF, von Garnier C, Resch G, Guery B. Phage therapy for pulmonary infections: lessons from clinical experiences and key considerations. Eur Respir Rev 2022; 31:31/166/220121. [PMID: 36198417 DOI: 10.1183/16000617.0121-2022] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Accepted: 07/27/2022] [Indexed: 11/05/2022] Open
Abstract
Lower respiratory tract infections lead to significant morbidity and mortality. They are increasingly caused by multidrug-resistant pathogens, notably in individuals with cystic fibrosis, hospital-acquired pneumonia and lung transplantation. The use of bacteriophages (phages) to treat bacterial infections is gaining growing attention, with numerous published cases of compassionate treatment over the last few years. Although the use of phages appears safe, the lack of standardisation, the significant heterogeneity of published studies and the paucity of robust efficacy data, alongside regulatory hurdles arising from the existing pharmaceutical legislation, are just some of the challenges phage therapy has to overcome. In this review, we discuss the lessons learned from recent clinical experiences of phage therapy for the treatment of pulmonary infections. We review the key aspects, opportunities and challenges of phage therapy regarding formulations and administration routes, interactions with antibiotics and the immune system, and phage resistance. Building upon the current knowledge base, future pre-clinical studies using emerging technologies and carefully designed clinical trials are expected to enhance our understanding and explore the therapeutic potential of phage therapy.
Collapse
Affiliation(s)
- Georgia Mitropoulou
- Division of Pulmonology, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland .,University of Lausanne, Lausanne, Switzerland.,Shared first authorship
| | - Angela Koutsokera
- Division of Pulmonology, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland.,University of Lausanne, Lausanne, Switzerland.,Shared first authorship
| | - Chantal Csajka
- Centre for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital, Lausanne, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, University of Lausanne, Geneva, Switzerland.,School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland
| | - Sylvain Blanchon
- University of Lausanne, Lausanne, Switzerland.,Paediatric Pulmonology and Cystic Fibrosis Unit, Division of Paediatrics, Department of Woman-Mother-Child, Lausanne University Hospital, Lausanne, Switzerland
| | - Alain Sauty
- University of Lausanne, Lausanne, Switzerland.,Division of Pulmonology, Neuchâtel Hospital Network, Neuchâtel, Switzerland
| | - Jean-Francois Brunet
- University of Lausanne, Lausanne, Switzerland.,Cell Production Centre, Dept of Interdisciplinary Centres, Lausanne University Hospital, Lausanne, Switzerland
| | - Christophe von Garnier
- Division of Pulmonology, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland.,University of Lausanne, Lausanne, Switzerland
| | - Grégory Resch
- University of Lausanne, Lausanne, Switzerland.,Centre for Research and Innovation in Clinical Pharmaceutical Sciences, Lausanne University Hospital, Lausanne, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, University of Lausanne, Geneva, Switzerland.,Shared last authorship
| | - Benoit Guery
- University of Lausanne, Lausanne, Switzerland.,Service of Infectious Diseases, Department of Medicine, Lausanne University Hospital, Lausanne, Switzerland.,Shared last authorship
| |
Collapse
|
25
|
Leclerc QJ, Lindsay JA, Knight GM. Modelling the synergistic effect of bacteriophage and antibiotics on bacteria: Killers and drivers of resistance evolution. PLoS Comput Biol 2022; 18:e1010746. [PMID: 36449520 PMCID: PMC9744316 DOI: 10.1371/journal.pcbi.1010746] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 12/12/2022] [Accepted: 11/17/2022] [Indexed: 12/05/2022] Open
Abstract
Bacteriophage (phage) are bacterial predators that can also spread antimicrobial resistance (AMR) genes between bacteria by generalised transduction. Phage are often present alongside antibiotics in the environment, yet evidence of their joint killing effect on bacteria is conflicted, and the dynamics of transduction in such systems are unknown. Here, we combine in vitro data and mathematical modelling to identify conditions where phage and antibiotics act in synergy to remove bacteria or drive AMR evolution. We adapt a published model of phage-bacteria dynamics, including transduction, to add the pharmacodynamics of erythromycin and tetracycline, parameterised from new in vitro data. We simulate a system where two strains of Staphylococcus aureus are present at stationary phase, each carrying either an erythromycin or tetracycline resistance gene, and where multidrug-resistant bacteria can be generated by transduction only. We determine rates of bacterial clearance and multidrug-resistant bacteria appearance, when either or both antibiotics and phage are present at varying timings and concentrations. Although phage and antibiotics act in synergy to kill bacteria, by reducing bacterial growth antibiotics reduce phage production. A low concentration of phage introduced shortly after antibiotics fails to replicate and exert a strong killing pressure on bacteria, instead generating multidrug-resistant bacteria by transduction which are then selected for by the antibiotics. Multidrug-resistant bacteria numbers were highest when antibiotics and phage were introduced simultaneously. The interaction between phage and antibiotics leads to a trade-off between a slower clearing rate of bacteria (if antibiotics are added before phage), and a higher risk of multidrug-resistance evolution (if phage are added before antibiotics), exacerbated by low concentrations of phage or antibiotics. Our results form hypotheses to guide future experimental and clinical work on the impact of phage on AMR evolution, notably for studies of phage therapy which should investigate varying timings and concentrations of phage and antibiotics.
Collapse
Affiliation(s)
- Quentin J. Leclerc
- Centre for Mathematical Modelling of Infectious Diseases, Department of Infectious Disease Epidemiology, Faculty of Epidemiology & Population Health, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Antimicrobial Resistance Centre, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Institute for Infection & Immunity, St George’s University of London, London, United Kingdom
- * E-mail: ,
| | - Jodi A. Lindsay
- Institute for Infection & Immunity, St George’s University of London, London, United Kingdom
| | - Gwenan M. Knight
- Centre for Mathematical Modelling of Infectious Diseases, Department of Infectious Disease Epidemiology, Faculty of Epidemiology & Population Health, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Antimicrobial Resistance Centre, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
26
|
Lu M, Liu B, Xiong W, Liu X. The Combination of Salmonella Phage ST-3 and Antibiotics to Prevent Salmonella Typhimurium In Vitro. Curr Microbiol 2022; 79:371. [PMID: 36269452 DOI: 10.1007/s00284-022-03073-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 10/03/2022] [Indexed: 11/28/2022]
Abstract
The novel phage ST-3, capable of infecting the foodborne pathogen Salmonella Typhimurium, was isolated from wastewater. The Biological characters and genome information of ST-3 were analyzed. In the in vitro assay, the phage ST-3 with a MOI of 10 effectively inhibited the growth of Salmonella Typhimurium CGMCC 1.1174 in 6 h. The inhibitory effect of combination phage ST-3 and antibiotics was also studied, the removal rate of planktonic host exposed to ST-3 and levofloxacin hydrochloride at the same time, or to ciprofloxacin followed by ST-3, is higher than that exposed to antibiotic dosing group alone and antibiotic + phage dosing group. The phage ST-3 combined with 0.5 µg/mL levofloxacin hydrochloride resulted in the largest decrease in biofilm biomass at 54%. The phage ST-3 could be a potential agent to control Salmonella Typhimurium growth and provide instruction for use it and antibiotics together.
Collapse
Affiliation(s)
- Min Lu
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Bingxin Liu
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Wenbin Xiong
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 101408, China
| | - Xinchun Liu
- College of Resources and Environment, University of Chinese Academy of Sciences, Beijing, 101408, China.
| |
Collapse
|
27
|
Coyne AJK, Stamper K, Kebriaei R, Holger DJ, El Ghali A, Morrisette T, Biswas B, Wilson M, Deschenes MV, Canfield GS, Duerkop BA, Arias CA, Rybak MJ. Phage Cocktails with Daptomycin and Ampicillin Eradicates Biofilm-Embedded Multidrug-Resistant Enterococcus faecium with Preserved Phage Susceptibility. Antibiotics (Basel) 2022; 11:1175. [PMID: 36139953 PMCID: PMC9495159 DOI: 10.3390/antibiotics11091175] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/23/2022] [Accepted: 08/26/2022] [Indexed: 02/03/2023] Open
Abstract
Multidrug-resistant (MDR) Enterococcus faecium is a challenging nosocomial pathogen known to colonize medical device surfaces and form biofilms. Bacterio (phages) may constitute an emerging anti-infective option for refractory, biofilm-mediated infections. This study evaluates eight MDR E. faecium strains for biofilm production and phage susceptibility against nine phages. Two E. faecium strains isolated from patients with bacteremia and identified to be biofilm producers, R497 (daptomycin (DAP)-resistant) and HOU503 (DAP-susceptible dose-dependent (SDD), in addition to four phages with the broadest host ranges (ATCC 113, NV-497, NV-503-01, NV-503-02) were selected for further experiments. Preliminary phage-antibiotic screening was performed with modified checkerboard minimum biofilm inhibitory concentration (MBIC) assays to efficiently screen for bacterial killing and phage-antibiotic synergy (PAS). Data were compared by one-way ANOVA and Tukey (HSD) tests. Time kill analyses (TKA) were performed against R497 and HOU503 with DAP at 0.5× MBIC, ampicillin (AMP) at free peak = 72 µg/mL, and phage at a multiplicity of infection (MOI) of 0.01. In 24 h TKA against R497, phage-antibiotic combinations (PAC) with DAP, AMP, or DAP + AMP combined with 3- or 4-phage cocktails demonstrated significant killing compared to the most effective double combination (ANOVA range of mean differences 2.998 to 3.102 log10 colony forming units (CFU)/mL; p = 0.011, 2.548 to 2.868 log10 colony forming units (CFU)/mL; p = 0.023, and 2.006 to 2.329 log10 colony forming units (CFU)/mL; p = 0.039, respectively), with preserved phage susceptibility identified in regimens with 3-phage cocktails containing NV-497 and the 4-phage cocktail. Against HOU503, AMP combined with any 3- or 4-phage cocktail and DAP + AMP combined with the 3-phage cocktail ATCC 113 + NV-497 + NV-503-01 demonstrated significant PAS and bactericidal activity (ANOVA range of mean differences 2.251 to 2.466 log10 colony forming units (CFU)/mL; p = 0.044 and 2.119 to 2.350 log10 colony forming units (CFU)/mL; p = 0.028, respectively), however, only PAC with DAP + AMP maintained phage susceptibility at the end of 24 h TKA. R497 and HOU503 exposure to DAP, AMP, or DAP + AMP in the presence of single phage or phage cocktail resulted in antibiotic resistance stabilization (i.e., no antibiotic MBIC elevation compared to baseline) without identified antibiotic MBIC reversion (i.e., lowering of antibiotic MBIC compared to baseline in DAP-resistant and DAP-SDD isolates) at the end of 24 h TKA. In conclusion, against DAP-resistant R497 and DAP-SDD HOU503 E. faecium clinical blood isolates, the use of DAP + AMP combined with 3- and 4-phage cocktails effectively eradicated biofilm-embedded MDR E. faecium without altering antibiotic MBIC or phage susceptibility compared to baseline.
Collapse
Affiliation(s)
- Ashlan J. Kunz Coyne
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Kyle Stamper
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Razieh Kebriaei
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Dana J. Holger
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
- Department of Pharmacy Practice, College of Pharmacy, Nova Southeastern University, Davie, FL 33328, USA
| | - Amer El Ghali
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Taylor Morrisette
- Department of Pharmacy and Clinical Services, College of Pharmacy, Medical University of South Carolina, Charleston, SC 29208, USA
- Department of Pharmacy Services, Shawn Jenkins Children’s Hospital, Medical University of South Carolina, Charleston, SC 29208, USA
| | | | - Melanie Wilson
- Naval Medical Research Center, Fort Detrick, MD 21702, USA
- Leidos, Reston, VA 20190, USA
| | - Michael V. Deschenes
- Naval Medical Research Center, Fort Detrick, MD 21702, USA
- Leidos, Reston, VA 20190, USA
| | - Gregory S. Canfield
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO 80045, USA
- Department of Infectious Diseases, School of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Breck A. Duerkop
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Cesar A. Arias
- Division of Infectious Diseases, Houston Methodist Hospital, Houston, TX 77030, USA
- Center for Infectious Diseases, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Michael J. Rybak
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
- School of Medicine, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
28
|
In Vitro Evaluation of Five Newly Isolated Bacteriophages against E. faecalis Biofilm for Their Potential Use against Post-Treatment Apical Periodontitis. Pharmaceutics 2022; 14:pharmaceutics14091779. [PMID: 36145527 PMCID: PMC9503355 DOI: 10.3390/pharmaceutics14091779] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/22/2022] [Accepted: 08/22/2022] [Indexed: 11/17/2022] Open
Abstract
State-of-the-art treatment of root canal infection includes the use of mechanical debridement and chemical agents. This disinfection method is limited, and microorganisms can remain in the canal system. Enterococcus faecalis appears with a high prevalence in secondary and persistent root canal infections and can be linked to endodontic treatment failure due to its various resistance mechanisms. Here, we evaluated the activity of newly isolated bacteriophages against clinical isolates of E. faecalis (including one vancomycin- and gentamicin-resistant strain) as a single treatment or in combination with gentamicin and vancomycin. For the resistant strain, daptomycin and fosfomycin were tested. Sixteen E. faecalis strains were used to screen for the presence of bacteriophages in sewage. Five different bacteriophages were characterized in terms of virion morphology, host range and killing-kinetics against each E. faecalis host strain. To investigate the antibiofilm effect of antibiotic and phages, E. faecalis biofilm was grown on porous glass beads and treated with different antibiotic concentrations and with isolated bacteriophages alone or in staggered combinations. A strong biofilm reduction was observed when phages were combined with antibiotic, where combinations with gentamicin showed a better outcome compared to vancomycin. Regarding the resistant strain, daptomycin had a superior antibiofilm effect than fosfomycin.
Collapse
|
29
|
Benefits of Combined Phage–Antibiotic Therapy for the Control of Antibiotic-Resistant Bacteria: A Literature Review. Antibiotics (Basel) 2022; 11:antibiotics11070839. [PMID: 35884092 PMCID: PMC9311689 DOI: 10.3390/antibiotics11070839] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 01/27/2023] Open
Abstract
With the increase in bacterial resistance to antibiotics, more and more therapeutic failures are being reported worldwide. The market for antibiotics is now broken due to the high cost of developing new molecules. A promising solution to bacterial resistance is combined phage–antibiotic therapy, a century-old method that can potentiate existing antibiotics by prolonging or even restoring their activity against specific bacteria. The aim of this literature review was to provide an overview of different phage–antibiotic combinations and to describe the possible mechanisms of phage–antibiotic synergy.
Collapse
|
30
|
Chang RYK, Nang SC, Chan HK, Li J. Novel antimicrobial agents for combating antibiotic-resistant bacteria. Adv Drug Deliv Rev 2022; 187:114378. [PMID: 35671882 DOI: 10.1016/j.addr.2022.114378] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 05/26/2022] [Accepted: 05/26/2022] [Indexed: 12/16/2022]
Abstract
Antibiotic therapy has become increasingly ineffective against bacterial infections due to the rise of resistance. In particular, ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) have caused life-threatening infections in humans and represent a major global health threat due to a high degree of antibiotic resistance. To respond to this urgent call, novel strategies are urgently needed, such as bacteriophages (or phages), phage-encoded enzymes, immunomodulators and monoclonal antibodies. This review critically analyses these promising antimicrobial therapies for the treatment of multidrug-resistant bacterial infections. Recent advances in these novel therapeutic strategies are discussed, focusing on preclinical and clinical investigations, as well as combinatorial approaches. In this 'Bad Bugs, No Drugs' era, novel therapeutic strategies can play a key role in treating deadly infections and help extend the lifetime of antibiotics.
Collapse
|
31
|
Silva V, Correia E, Pereira JE, González-Machado C, Capita R, Alonso-Calleja C, Igrejas G, Poeta P. Biofilm Formation of Staphylococcus aureus from Pets, Livestock, and Wild Animals: Relationship with Clonal Lineages and Antimicrobial Resistance. Antibiotics (Basel) 2022; 11:antibiotics11060772. [PMID: 35740178 PMCID: PMC9219840 DOI: 10.3390/antibiotics11060772] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 12/14/2022] Open
Abstract
This study aimed to compare the biofilm formation ability of Staphylococcus aureus isolated from a wide range of animals and study the association between biofilm formation and antimicrobial resistance and genetic lineages. A total of 214 S. aureus strains isolated from pets, livestock, and wild animals were evaluated regarding their ability to form biofilms by the microtiter biofilm assay and their structure via confocal scanning laser microscopy. Statistical analysis was used to find an association between biofilm formation and antimicrobial resistance, multidrug resistance, sequence types (STs), spa and agr-types of the isolates. The antimicrobial susceptibility of 24 h-old biofilms was assessed against minimum inhibitory concentrations (MIC) and 10× MIC of amikacin and tetracycline, and the biomass reduction was measured. The metabolic activity of biofilms after antimicrobial treatment was evaluated by the XTT assay. All isolates were had the ability to form biofilms. Yet, significant differences in biofilm biomass production were detected among animal species. Multidrug resistance had a positive association with biofilm formation as well as methicillin-resistance. Significant differences were also detected among the clonal lineages of the isolates. Both tetracycline and amikacin were able to significantly reduce the biofilm mass. However, none of the antimicrobials were able to eradicate the biofilm at the maximum concentration used. Our results provide important information on the biofilm-forming capacity of animal-adapted S. aureus isolates, which may have potential implications for the development of new biofilm-targeted therapeutics.
Collapse
Affiliation(s)
- Vanessa Silva
- Microbiology and Antibiotic Resistance Team (MicroART), Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal;
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal;
- Functional Genomics and Proteomics Unit, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - Elisete Correia
- Center for Computational and Stochastic Mathematics (CEMAT), Department of Mathematics, University of Trás-os-Montes and Alto Douro (UTAD), 5001-801 Vila Real, Portugal;
| | - José Eduardo Pereira
- CECAV—Veterinary and Animal Research Centre, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
| | - Camino González-Machado
- Department of Food Hygiene and Technology, Veterinary Faculty, University of León, E-24071 León, Spain; (C.G.-M.); (R.C.); (C.A.-C.)
- Institute of Food Science and Technology, University of León, E-24071 León, Spain
| | - Rosa Capita
- Department of Food Hygiene and Technology, Veterinary Faculty, University of León, E-24071 León, Spain; (C.G.-M.); (R.C.); (C.A.-C.)
- Institute of Food Science and Technology, University of León, E-24071 León, Spain
| | - Carlos Alonso-Calleja
- Department of Food Hygiene and Technology, Veterinary Faculty, University of León, E-24071 León, Spain; (C.G.-M.); (R.C.); (C.A.-C.)
- Institute of Food Science and Technology, University of León, E-24071 León, Spain
| | - Gilberto Igrejas
- Department of Genetics and Biotechnology, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal;
- Functional Genomics and Proteomics Unit, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- LAQV-REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, Universidade Nova de Lisboa, 2829-516 Caparica, Portugal
| | - Patrícia Poeta
- Microbiology and Antibiotic Resistance Team (MicroART), Department of Veterinary Sciences, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal;
- CECAV—Veterinary and Animal Research Centre, University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal;
- Associate Laboratory for Animal and Veterinary Science (AL4AnimalS), University of Trás-os-Montes and Alto Douro (UTAD), 5000-801 Vila Real, Portugal
- Correspondence:
| |
Collapse
|
32
|
Synergistic Effects of Bacteriophage vB_Eco4-M7 and Selected Antibiotics on the Biofilm Formed by Shiga Toxin-Producing Escherichia coli. Antibiotics (Basel) 2022; 11:antibiotics11060712. [PMID: 35740119 PMCID: PMC9219966 DOI: 10.3390/antibiotics11060712] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 02/04/2023] Open
Abstract
Apart from antibiotic resistance of pathogenic bacteria, the formation of biofilms is a feature that makes bacterial infections especially difficulty to treat. Shiga toxin-producing Escherichia coli (STEC) strains are dangerous pathogens, causing severe infections in humans, and capable of biofilm production. We have reported previously the identification and characterization of the vB_Eco4-M7 bacteriophage, infecting various STEC strains. It was suggested that this phage might be potentially used in phage therapy against these bacteria. Here, we tested the effects of vB_Eco4-M7 alone or in a phage cocktail with another STEC-infecting phage, and/or in a combination with different antibiotics (ciprofloxacin and rifampicin) on biofilm formed by a model STEC strain, named E. coli O157:H7 (ST2-8624). The vB_Eco4-M7 phage appeared effective in anti-biofilm action in all these experimental conditions (2–3-fold reduction of the biofilm density, and 2–3 orders of magnitude reduction of the number of bacterial cells). However, the highest efficiency in reducing a biofilm’s density and number of bacterial cells was observed when phage infection preceded antibiotic treatment (6-fold reduction of the biofilm density, and 5–6 orders of magnitude reduction of the number of bacterial cells). Previous reports indicated that the use of antibiotics to treat STEC-caused infections might be dangerous due to the induction of Shiga toxin-converting prophages from bacterial genomes under stress conditions caused by antibacterial agents. We found that ciprofloxacin was almost as efficient in inducing prophages from the E. coli O15:H7 (ST2-8624) genome as a classical inducer, mitomycin C, while no detectable prophage induction could be observed in rifampicin-treated STEC cells. Therefore, we conclude the latter antibiotic or similarly acting compounds might be candidate(s) as effective and safe drug(s) when used in combination with phage therapy to combat STEC-mediated infections.
Collapse
|
33
|
Tarín-Pelló A, Suay-García B, Pérez-Gracia MT. Antibiotic resistant bacteria: current situation and treatment options to accelerate the development of a new antimicrobial arsenal. Expert Rev Anti Infect Ther 2022; 20:1095-1108. [PMID: 35576494 DOI: 10.1080/14787210.2022.2078308] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Antibiotic resistance is one of the biggest public health threats worldwide. Currently, antibiotic-resistant bacteria kill 700,000 people every year. These data represent the near future in which we find ourselves, a "post-antibiotic era" where the identification and development of new treatments are key. This review is focused on the current and emerging antimicrobial therapies which can solve this global threat. AREAS COVERED Through a literature search using databases such as Medline and Web of Science, and search engines such as Google Scholar, different antimicrobial therapies were analyzed, including pathogen-oriented therapy, phagotherapy, microbiota and antivirulent therapy. Additionally, the development pathways of new antibiotics were described, emphasizing on the potential advantages that the combination of a drug repurposing strategy with the application of mathematical prediction models could bring to solve the problem of AMRs. EXPERT OPINION This review offers several starting points to solve a single problem: reducing the number of AMR. The data suggest that the strategies described could provide many benefits to improve antimicrobial treatments. However, the development of new antimicrobials remains necessary. Drug repurposing, with the application of mathematical prediction models, is considered to be of interest due to its rapid and effective potential to increase the current therapeutic arsenal.
Collapse
Affiliation(s)
- Antonio Tarín-Pelló
- Área de Microbiología, Departamento de Farmacia, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud
| | - Beatriz Suay-García
- ESI International Chair@CEU-UCH, Departamento de Matemáticas, Física y Ciencias Tecnológicas, Universidad Cardenal Herrera-CEU, CEU Universities, C/ Santiago Ramón y Cajal, 46115 Alfara del Patriarca, Valencia, Spain
| | - María-Teresa Pérez-Gracia
- Área de Microbiología, Departamento de Farmacia, Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud
| |
Collapse
|
34
|
Understanding the Mechanisms That Drive Phage Resistance in Staphylococci to Prevent Phage Therapy Failure. Viruses 2022; 14:v14051061. [PMID: 35632803 PMCID: PMC9146914 DOI: 10.3390/v14051061] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 02/07/2023] Open
Abstract
Despite occurring at the microscopic scale, the armed race between phages and their bacterial hosts involves multiple mechanisms, some of which are just starting to be understood. On the one hand, bacteria have evolved strategies that can stop the viral infection at different stages (adsorption, DNA injection and replication, biosynthesis and assembly of the viral progeny and/or release of the newly formed virions); on the other, phages have gradually evolved counterattack strategies that allow them to continue infecting their prey. This co-evolutionary process has played a major role in the development of microbial populations in both natural and man-made environments. Notably, understanding the parameters of this microscopic war will be paramount to fully benefit from the application of phage therapy against dangerous, antibiotic-resistant human pathogens. This review gathers the current knowledge regarding the mechanisms of phage resistance in the Staphylococcus genus, which includes Staphylococcus aureus, one of the most concerning microorganisms in terms of antibiotic resistance acquisition. Some of these strategies involve permanent changes to the bacterial cell via mutations, while others are transient, adaptive changes whose expression depends on certain environmental cues or the growth phase. Finally, we discuss the most plausible strategies to limit the impact of phage resistance on therapy, with a special emphasis on the importance of a rational design of phage cocktails in order to thwart therapeutic failure.
Collapse
|
35
|
Phage–Antibiotic Therapy as a Promising Strategy to Combat Multidrug-Resistant Infections and to Enhance Antimicrobial Efficiency. Antibiotics (Basel) 2022; 11:antibiotics11050570. [PMID: 35625214 PMCID: PMC9137994 DOI: 10.3390/antibiotics11050570] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/20/2022] [Accepted: 04/24/2022] [Indexed: 02/06/2023] Open
Abstract
Infections caused by multidrug-resistant (MDR) bacteria have highlighted the importance of the development of new antimicrobial agents. While bacteriophages (phages) are widely studied as alternative agents to antibiotics, combined treatments using phages and antibiotics have exhibited Phage–Antibiotic Synergy (PAS), in which antibiotics promote phage replication and extraordinary antimicrobial efficacy with reduced development of bacterial resistance. This review paper on the current progress of phage–antibiotic therapy includes aspects of the mechanisms of PAS and the therapeutic performance of PAS in combating multidrug-resistant bacterial infections. The choice of phages and antibiotics, the administration time and sequence, and the concentrations of the two agents impact the bacterial inhibitory effects to different extents.
Collapse
|
36
|
Roszak M, Dołęgowska B, Cecerska-Heryć E, Serwin N, Jabłońska J, Grygorcewicz B. Bacteriophage–Ciprofloxacin Combination Effectiveness Depends on Staphylococcus aureus– Candida albicans Dual-Species Communities’ Growth Model. Microb Drug Resist 2022; 28:613-622. [DOI: 10.1089/mdr.2021.0324] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Marta Roszak
- Department of Laboratory Medicine, Chair of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Barbara Dołęgowska
- Department of Laboratory Medicine, Chair of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Elżbieta Cecerska-Heryć
- Department of Laboratory Medicine, Chair of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Natalia Serwin
- Department of Laboratory Medicine, Chair of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Joanna Jabłońska
- Department of Chemical and Process Engineering, Faculty of Chemical Technology and Engineering, West Pomeranian University of Technology in Szczecin, Szczecin, Poland
| | - Bartłomiej Grygorcewicz
- Department of Laboratory Medicine, Chair of Microbiology, Immunology and Laboratory Medicine, Pomeranian Medical University in Szczecin, Szczecin, Poland
| |
Collapse
|
37
|
Łusiak-Szelachowska M, Międzybrodzki R, Drulis-Kawa Z, Cater K, Knežević P, Winogradow C, Amaro K, Jończyk-Matysiak E, Weber-Dąbrowska B, Rękas J, Górski A. Bacteriophages and antibiotic interactions in clinical practice: what we have learned so far. J Biomed Sci 2022; 29:23. [PMID: 35354477 PMCID: PMC8969238 DOI: 10.1186/s12929-022-00806-1] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/24/2022] [Indexed: 01/04/2023] Open
Abstract
Bacteriophages (phages) may be used as an alternative to antibiotic therapy for combating infections caused by multidrug-resistant bacteria. In the last decades, there have been studies concerning the use of phages and antibiotics separately or in combination both in animal models as well as in humans. The phenomenon of phage–antibiotic synergy, in which antibiotics may induce the production of phages by bacterial hosts has been observed. The potential mechanisms of phage and antibiotic synergy was presented in this paper. Studies of a biofilm model showed that a combination of phages with antibiotics may increase removal of bacteria and sequential treatment, consisting of phage administration followed by an antibiotic, was most effective in eliminating biofilms. In vivo studies predominantly show the phenomenon of phage and antibiotic synergy. A few studies also describe antagonism or indifference between phages and antibiotics. Recent papers regarding the application of phages and antibiotics in patients with severe bacterial infections show the effectiveness of simultaneous treatment with both antimicrobials on the clinical outcome.
Collapse
Affiliation(s)
- Marzanna Łusiak-Szelachowska
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114, Wrocław, Poland.
| | - Ryszard Międzybrodzki
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114, Wrocław, Poland.,Phage Therapy Unit, Medical Center of the Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114, Wrocław, Poland.,Department of Clinical Immunology, Transplantation Institute, Medical University of Warsaw, 02-006, Warsaw, Poland
| | - Zuzanna Drulis-Kawa
- Department of Pathogen Biology and Immunology, University of Wrocław, 51-148, Wrocław, Poland
| | - Kathryn Cater
- Rush University Medical Center, 1620 W. Harrison St., Chicago, IL, 60612, USA
| | - Petar Knežević
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, 21000, Novi Sad, Republic of Serbia
| | - Cyprian Winogradow
- Faculty of Life Sciences, University College London, London, WC1E 6BT, UK
| | | | - Ewa Jończyk-Matysiak
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114, Wrocław, Poland
| | - Beata Weber-Dąbrowska
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114, Wrocław, Poland.,Phage Therapy Unit, Medical Center of the Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114, Wrocław, Poland
| | - Justyna Rękas
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114, Wrocław, Poland
| | - Andrzej Górski
- Bacteriophage Laboratory, Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114, Wrocław, Poland.,Phage Therapy Unit, Medical Center of the Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114, Wrocław, Poland.,Infant Jesus Hospital, Medical University of Warsaw, 02-005, Warsaw, Poland
| |
Collapse
|
38
|
Lev K, Kunz Coyne AJ, Kebriaei R, Morrisette T, Stamper K, Holger DJ, Canfield GS, Duerkop BA, Arias CA, Rybak MJ. Evaluation of Bacteriophage-Antibiotic Combination Therapy for Biofilm-Embedded MDR Enterococcus faecium. Antibiotics (Basel) 2022; 11:antibiotics11030392. [PMID: 35326855 PMCID: PMC8944492 DOI: 10.3390/antibiotics11030392] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/22/2022] [Accepted: 03/05/2022] [Indexed: 02/03/2023] Open
Abstract
Multidrug-resistant (MDR) Enterococcus faecium is a challenging pathogen known to cause biofilm-mediated infections with limited effective therapeutic options. Lytic bacteriophages target, infect, and lyse specific bacterial cells and have anti-biofilm activity, making them a possible treatment option. Here, we examine two biofilm-producing clinical E. faecium strains, daptomycin (DAP)-resistant R497 and DAP-susceptible dose-dependent (SDD) HOU503, with initial susceptibility to E. faecium bacteriophage 113 (ATCC 19950-B1). An initial synergy screening was performed with modified checkerboard MIC assays developed by our laboratory to efficiently screen for antibiotic and phage synergy, including at very low phage multiplicity of infection (MOI). The data were compared by one-way ANOVA and Tukey (HSD) tests. In 24 h time kill analyses (TKA), combinations with phage-DAP-ampicillin (AMP), phage-DAP-ceftaroline (CPT), and phage-DAP-ertapenem (ERT) were synergistic and bactericidal compared to any single agent (ANOVA range of mean differences 3.34 to 3.84 log10 CFU/mL; p < 0.001). Furthermore, phage-DAP-AMP and phage-DAP-CPT prevented the emergence of DAP and phage resistance. With HOU503, the combination of phage-DAP-AMP showed the best killing effect, followed closely by phage-DAP-CPT; both showed bactericidal and synergistic effects compared to any single agent (ANOVA range of mean differences 3.99 to 4.08 log10 CFU/mL; p < 0.001).
Collapse
Affiliation(s)
- Katherine Lev
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA; (K.L.); (A.J.K.C.); (R.K.); (K.S.); (D.J.H.)
| | - Ashlan J. Kunz Coyne
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA; (K.L.); (A.J.K.C.); (R.K.); (K.S.); (D.J.H.)
| | - Razieh Kebriaei
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA; (K.L.); (A.J.K.C.); (R.K.); (K.S.); (D.J.H.)
| | - Taylor Morrisette
- Department of Pharmacy and Clinical Services, Medical University of South Carolina College of Pharmacy, Charleston, SC 29208, USA;
| | - Kyle Stamper
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA; (K.L.); (A.J.K.C.); (R.K.); (K.S.); (D.J.H.)
| | - Dana J. Holger
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA; (K.L.); (A.J.K.C.); (R.K.); (K.S.); (D.J.H.)
| | - Gregory S. Canfield
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA; (G.S.C.); (B.A.D.)
- Department of Infectious Diseases, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Breck A. Duerkop
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA; (G.S.C.); (B.A.D.)
| | - Cesar A. Arias
- Division of Infectious Diseases, Houston Methodist Hospital, Houston, TX 77030, USA;
- Center for Infectious Diseases Research, Houston Methodist Research Institute, Houston, TX 77030, USA
| | - Michael J. Rybak
- Anti-Infective Research Laboratory, College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA; (K.L.); (A.J.K.C.); (R.K.); (K.S.); (D.J.H.)
- School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Correspondence:
| |
Collapse
|
39
|
Abstract
Increasing antimicrobial resistance and medical device-related infections have led to a renewed interest in phage therapy as an alternative or adjunct to conventional antimicrobials. Expanded access and compassionate use cases have risen exponentially but have varied widely in approach, methodology, and clinical situations in which phage therapy might be considered. Large gaps in knowledge contribute to heterogeneity in approach and lack of consensus in many important clinical areas. The Antibacterial Resistance Leadership Group (ARLG) has convened a panel of experts in phage therapy, clinical microbiology, infectious diseases, and pharmacology, who worked with regulatory experts and a funding agency to identify questions based on a clinical framework and divided them into three themes: potential clinical situations in which phage therapy might be considered, laboratory testing, and pharmacokinetic considerations. Suggestions are provided as answers to a series of questions intended to inform clinicians considering experimental phage therapy for patients in their clinical practices.
Collapse
|
40
|
Liu S, Lu H, Zhang S, Shi Y, Chen Q. Phages against Pathogenic Bacterial Biofilms and Biofilm-Based Infections: A Review. Pharmaceutics 2022; 14:pharmaceutics14020427. [PMID: 35214158 PMCID: PMC8875263 DOI: 10.3390/pharmaceutics14020427] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 02/01/2023] Open
Abstract
Bacterial biofilms formed by pathogens are known to be hundreds of times more resistant to antimicrobial agents than planktonic cells, making it extremely difficult to cure biofilm-based infections despite the use of antibiotics, which poses a serious threat to human health. Therefore, there is an urgent need to develop promising alternative antimicrobial therapies to reduce the burden of drug-resistant bacterial infections caused by biofilms. As natural enemies of bacteria, bacteriophages (phages) have the advantages of high specificity, safety and non-toxicity, and possess great potential in the defense and removal of pathogenic bacterial biofilms, which are considered to be alternatives to treat bacterial diseases. This work mainly reviews the composition, structure and formation process of bacterial biofilms, briefly discusses the interaction between phages and biofilms, and summarizes several strategies based on phages and their derivatives against biofilms and drug-resistant bacterial infections caused by biofilms, serving the purpose of developing novel, safe and effective treatment methods against biofilm-based infections and promoting the application of phages in maintaining human health.
Collapse
Affiliation(s)
| | | | | | - Ying Shi
- Correspondence: (Y.S.); (Q.C.); Tel.: +86-139-6717-1522 (Y.S.)
| | - Qihe Chen
- Correspondence: (Y.S.); (Q.C.); Tel.: +86-139-6717-1522 (Y.S.)
| |
Collapse
|
41
|
Comparative Assessment of Bacteriophage and Antibiotic Activity against Multidrug-Resistant Staphylococcus aureus Biofilms. Int J Mol Sci 2022; 23:ijms23031274. [PMID: 35163197 PMCID: PMC8836238 DOI: 10.3390/ijms23031274] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 02/01/2023] Open
Abstract
Problems connected with biofilm-related infections and antibiotic resistance necessitate the investigation and development of novel treatment strategies. Given their unique characteristics, one of the most promising alternatives to conventional antibiotics are bacteriophages. In the in vitro and in vivo larva model study, we demonstrate that phages vB_SauM-A, vB_SauM-C, and vB_SauM-D are effective antibiofilm agents. The exposure of biofilm to phages vB_SauM-A and vB_SauM-D led to 2-3 log reductions in the colony-forming unit number in most of the multidrug-resistant S. aureus strains. It was found that phage application reduced the formed biofilms independently of the used titer. Moreover, the study demonstrated that bacteriophages are more efficient in biofilm biomass removal and reduction in staphylococci count when compared to the antibiotics used. The scanning electron microscopy analysis results are in line with colony forming unit (CFU) counting but not entirely consistent with crystal violet (CV) staining. Additionally, phages vB_SauM-A, vB_SauM-C, and vB_SauM-D can significantly increase the survival rate and extend the survival time of Galleria mellonella larvae.
Collapse
|
42
|
Puah SM, Fong SP, Kee BP, Puthucheary SD, Chua KH. Molecular identification and biofilm-forming ability of Elizabethkingia species. Microb Pathog 2022; 162:105345. [PMID: 34896547 DOI: 10.1016/j.micpath.2021.105345] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Revised: 11/30/2021] [Accepted: 12/01/2021] [Indexed: 11/17/2022]
Abstract
Recently, Elizabethkingia species have gained attention as a cause of life-threatening infections. The identification via phenotypic methods of three important species- Elizabethkingia meningoseptica, E. anophelis and E. miricola is difficult. Our objectives were to re-assess 30 archived Flavobacterium meningosepticum isolates using 16S rRNA gene sequencing, ERIC-PCR, and biofilm formation assay. Twenty-four isolates were re-identified as E. anophelis and 6 as E. miricola. All of them had the ability to form biofilm as shown in microtiter plate assay based on crystal violet staining. Overall, E. anophelis had a higher specific biofilm formation index compared to E. miricola. A total of 42% (10 out of 24) of E. anophelis were classified as strong, 29% (7 out of 24) as moderate and 29% (7 out of 24) as weak biofilm producers. E. miricola, 17% (1 out of 6) isolates were strong biofilm producers, 50% (3 out of 6) moderate and 33% (2 out of 6) were weak producers. E. anophelis from tracheal secretions were significantly associated with (p = 0.0361) strong biofilm formation. In summary, this study showed that the isolates originally identified as F. meningosepticum were re-classified using the 16S rRNA gene as one of two Elizabethkingia species. The ability of E. anophelis to form strong biofilm in endotracheal tubes indicates their probable role in the pathogenesis of Elizabethkingia infections.
Collapse
Affiliation(s)
- Suat Moi Puah
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Sam Pei Fong
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Boon Pin Kee
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - S D Puthucheary
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - Kek Heng Chua
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia.
| |
Collapse
|
43
|
Danis-Wlodarczyk KM, Cai A, Chen A, Gittrich MR, Sullivan MB, Wozniak DJ, Abedon ST. Friends or Foes? Rapid Determination of Dissimilar Colistin and Ciprofloxacin Antagonism of Pseudomonas aeruginosa Phages. Pharmaceuticals (Basel) 2021; 14:1162. [PMID: 34832944 PMCID: PMC8624478 DOI: 10.3390/ph14111162] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 12/14/2022] Open
Abstract
Phage therapy is a century-old technique employing viruses (phages) to treat bacterial infections, and in the clinic it is often used in combination with antibiotics. Antibiotics, however, interfere with critical bacterial metabolic activities that can be required by phages. Explicit testing of antibiotic antagonism of phage infection activities, though, is not a common feature of phage therapy studies. Here we use optical density-based 'lysis-profile' assays to assess the impact of two antibiotics, colistin and ciprofloxacin, on the bactericidal, bacteriolytic, and new-virion-production activities of three Pseudomonas aeruginosa phages. Though phages and antibiotics in combination are more potent in killing P. aeruginosa than either acting alone, colistin nevertheless substantially interferes with phage bacteriolytic and virion-production activities even at its minimum inhibitory concentration (1× MIC). Ciprofloxacin, by contrast, has little anti-phage impact at 1× or 3× MIC. We corroborate these results with more traditional measures, particularly colony-forming units, plaque-forming units, and one-step growth experiments. Our results suggest that ciprofloxacin could be useful as a concurrent phage therapy co-treatment especially when phage replication is required for treatment success. Lysis-profile assays also appear to be useful, fast, and high-throughput means of assessing antibiotic antagonism of phage infection activities.
Collapse
Affiliation(s)
| | - Alice Cai
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA; (A.C.); (A.C.); (M.R.G.); (M.B.S.)
| | - Anna Chen
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA; (A.C.); (A.C.); (M.R.G.); (M.B.S.)
| | - Marissa R. Gittrich
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA; (A.C.); (A.C.); (M.R.G.); (M.B.S.)
| | - Matthew B. Sullivan
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA; (A.C.); (A.C.); (M.R.G.); (M.B.S.)
- Department of Civil, Environmental and Geodetic Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel J. Wozniak
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA;
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA; (A.C.); (A.C.); (M.R.G.); (M.B.S.)
| | - Stephen T. Abedon
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA; (A.C.); (A.C.); (M.R.G.); (M.B.S.)
| |
Collapse
|
44
|
Abstract
Mediators of the initiation, development, and recurrence of periodontitis include the oral microbiome embedded in subgingival plaque and the host immune response to a dysbiosis within this dynamic and complex microbial community. Although mediators have been studied extensively, researchers in the field have been unable to fully ascribe certain clinical presentations of periodontitis to their nature. Emergence of high-throughput sequencing technologies has resulted in better characterization of the microbial oral dysbiosis that extends beyond the extensively studied putative bacterial periodontopathogens to a shift in the oral virome composition during disease conditions. Although the biological dark matter inserted by retroviruses was once believed to be nonfunctional, research has revealed that it encodes historical viral-eukaryotic interactions and influences host development. The objective of this review is to evaluate the proposed association of herpesviruses to the etiology and pathogenesis of periodontal disease and survey the highly abundant prokaryotic viruses to delineate their potential roles in biofilm dynamics, as well as their interactions with putative bacterial periodontopathogens and eukaryotic cells. The findings suggest that potential novel periodontal therapies targeting or utilizing the oral virome can alleviate certain clinical presentations of periodontitis. Perhaps it is time to embrace the viral dark matter within the periodontal environment to fully comprehend the pathogenesis and systemic implications of periodontitis.
Collapse
Affiliation(s)
- April Martínez
- Orofacial Sciences DepartmentSchool of DentistryUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Ryutaro Kuraji
- Orofacial Sciences DepartmentSchool of DentistryUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
- Department of Life Science DentistryThe Nippon Dental UniversityTokyoJapan
- Department of PeriodontologyThe Nippon Dental University School of Life Dentistry at TokyoTokyoJapan
| | - Yvonne L. Kapila
- Orofacial Sciences DepartmentSchool of DentistryUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| |
Collapse
|
45
|
A Case of In Situ Phage Therapy against Staphylococcus aureus in a Bone Allograft Polymicrobial Biofilm Infection: Outcomes and Phage-Antibiotic Interactions. Viruses 2021; 13:v13101898. [PMID: 34696328 PMCID: PMC8539586 DOI: 10.3390/v13101898] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/14/2021] [Accepted: 09/16/2021] [Indexed: 01/10/2023] Open
Abstract
Phage therapy (PT) shows promising potential in managing biofilm infections, which include refractory orthopedic infections. We report the case of a 13-year-old girl who developed chronic polymicrobial biofilm infection of a pelvic bone allograft after Ewing's sarcoma resection surgery. Chronic infection by Clostridium hathewayi, Proteus mirabilis and Finegoldia magna was worsened by methicillin-susceptible Staphylococcus aureus exhibiting an inducible Macrolides-Lincosamides-Streptogramin B resistance phenotype (iMLSB). After failure of conventional conservative treatment, combination of in situ anti-S. aureus PT with surgical debridement and intravenous antibiotic therapy led to marked clinical and microbiological improvement, yet failed to prevent a recurrence of infection on the midterm. This eventually led to surgical graft replacement. Multiple factors can explain this midterm failure, among which incomplete coverage of the polymicrobial infection by PT. Indeed, no phage therapy against C. hathewayi, P. mirabilis or F. magna could be administered. Phage-antibiotic interactions were investigated using OmniLog® technology. Our results suggest that phage-antibiotic interactions should not be considered "unconditionally synergistic", and should be assessed on a case-by-case basis. Specific pharmacodynamics of phages and antibiotics might explain these differences. More than two years after final graft replacement, the patient remains cured of her sarcoma and no further infections occurred.
Collapse
|
46
|
Amankwah S, Abdella K, Kassa T. Bacterial Biofilm Destruction: A Focused Review On The Recent Use of Phage-Based Strategies With Other Antibiofilm Agents. Nanotechnol Sci Appl 2021; 14:161-177. [PMID: 34548785 PMCID: PMC8449863 DOI: 10.2147/nsa.s325594] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 08/20/2021] [Indexed: 12/11/2022] Open
Abstract
Biofilms are bacterial communities that live in association with biotic or abiotic surfaces and enclosed in an extracellular polymeric substance. Their formation on both biotic and abiotic surfaces, including human tissue and medical device surfaces, pose a major threat causing chronic infections. In addition, current antibiotics and antiseptic agents have shown limited ability to completely remove biofilms. In this review, the authors provide an overview on the formation of bacterial biofilms and its characteristics, burden and evolution with phages. Moreover, the most recent possible use of phages and phage-derived enzymes to combat bacteria in biofilm structures is elucidated. From the emerging results, it can be concluded that despite successful use of phages and phage-derived products in destroying biofilms, they are mostly not adequate to eradicate all bacterial cells. Nevertheless, a combined therapy with the use of phages and/or phage-derived products with other antimicrobial agents including antibiotics, nanoparticles, and antimicrobial peptides may be effective approaches to remove biofilms from medical device surfaces and to treat their associated infections in humans.
Collapse
Affiliation(s)
- Stephen Amankwah
- School of Medical Laboratory Sciences, Institute of Health, Jimma University, Jimma, Ethiopia
- Accra Medical Centre, Accra, Ghana
| | - Kedir Abdella
- School of Medical Laboratory Sciences, Institute of Health, Jimma University, Jimma, Ethiopia
| | - Tesfaye Kassa
- School of Medical Laboratory Sciences, Institute of Health, Jimma University, Jimma, Ethiopia
| |
Collapse
|
47
|
Horiuk YV, Kukhtyn MD, Horiuk VV, Sytnik VA, Dashkovskyy OO. Effect of Phage SAvB14 combined with antibiotics on Staphylococcus aureus variant bovis. REGULATORY MECHANISMS IN BIOSYSTEMS 2021. [DOI: 10.15421/022173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Because using antimicrobial drugs leads to development of resistance among bacterial isolates, the treatment with antimicrobial drugs in human and veterinary medicine in general should be reduced. Currently, therapeutic use of bacteriophages may be an alternative or addition to the treatment of bacterial infections of animals. The article presents the results of studying the effect of bacteriophage Phage SAvB14 on microbial biofilms of Staphylococcus aureus variant bovis both alone and in complex with antibiotics. For this purpose, we used strain S. aureus var. bovis 1491 f and bacteriophage Phage SAvB14, isolated at dairy farms. The effect of combined application of phage and antibiotics (gentamicin, tetracycline, сeftriaxone and enrofloxacin) were assessed after simultaneous and subsequent introduction of Phage SAvB14 in the dose of 105 plaque-forming units per milliliter (PFU/mL) and corresponding concentrations of antibiotics to 24h biofilms. We determined that of the tested antibiotics, only gentamicin and ceftriazone exerted synergic effects in combinations with Phage SAvB14. Combination treatment using gentamicin and the phage decreased the amount of S. aureus in biofilm by 39.81 times compared with the phage-only treatment. Significant synergic effect was also taken by ceftriaxone – it killed 1.26 times more bacteria in combination with the phage than alone. Other antibiotics did not increase antibiotic activity of the phage. Specifically, 1.11 and 1.26 times more vital cells remained after the actions of tetracycline and enrofloxacin than after the exposure to the bacteriophage only. Therefore, the obtained results indicate that biofilm of S. aureus var. bovis may be eliminated using Phage SAvB14 as an individual antibacterial agent, as well as in complex with antibiotics. However, complex treatment would imply introducing the phage and then antibiotic some time later.
Collapse
|
48
|
Successful Intratracheal Treatment of Phage and Antibiotic Combination Therapy of a Multi-Drug Resistant Pseudomonas aeruginosa Murine Model. Antibiotics (Basel) 2021; 10:antibiotics10080946. [PMID: 34438996 PMCID: PMC8388862 DOI: 10.3390/antibiotics10080946] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/29/2021] [Accepted: 07/31/2021] [Indexed: 01/21/2023] Open
Abstract
Background: Pseudomonas aeruginosa (PsA) is a common etiology of bacteria-mediated lower respiratory tract infections, including pneumonia, hospital acquired pneumonia (HAP), and ventilator-associated pneumonia (VAP). Given the paucity of novel antibiotics in our foreseeable pipeline, developing novel non-antibiotic antimicrobial therapies saliently targeting drug resistant PsA isolates remains a priority. Lytic bacteriophages (or phages) have come under scrutiny as a potential antimicrobial for refractory bacterial infections. We evaluated intratracheally and intraperitoneally (IP) administered phage therapy (with/without meropenem) in an acute immunocompromised mouse model of multi-drug resistant (MDR) PsA pulmonary infection. The MDR P. aeruginosa respiratory disease model used in these studies was developed to investigate novel therapies that might have efficacy as either monotherapies or as combination therapy with meropenem. Methods: We utilized eight-week-old, 18 g BALB/cJ female mice and an MDR strain of PsA (UNC-D). Mice were immunosuppressed with cyclophosphamide. We employed a three-phage cocktail targeting PsA (PaAH2ΦP (103), PaBAP5Φ2 (130), and PaΦ (134)), confirmed to exhibit in vitro suppression of the infecting isolate out to 45 h. Suppression was confirmed with phages acting in isolation and in combination with meropenem. Results: IP administration of phage did not protect mice from death. A one-time delivery of phage directly to the lungs via a single intubation-mediated, intratracheal (IMIT) instillation protected mice from lethal infection. Protection was observed despite delaying therapy out to 6 h. Finally, we observed that, by slowing the progression of infection by treatment with a sub-efficacious dose of meropenem, we could protect the mice from lethal infection via IP phage administration coupled to meropenem, observing partial additive effects of phage–antibiotic combination therapy. Conclusions: A personalized phage cocktail administered via IMIT exhibits high therapeutic efficacy, despite delayed treatment of 6 h in a lethal MDR PsA pneumonia model. IP phage alone did not forestall mortality, but exhibited efficacy when combined with meropenem and IMIT-administered phage. These additive effects of combined IP phage and meropenem confirm that phage may indeed reach the lung bed via the systemic circulation and protect mice if the infection is not too acute. Therefore, adjunctive phage therapy with concerted attention to identifying optimal phage targeting of the infecting isolate in vitro may exhibit transformative potential for combating the specter of MDR bacterial infections. Phage should serve as an integral component of a four-pronged approach coupled with antibiotics, source control, and immune optimization.
Collapse
|
49
|
Bacteriophage treatment before chemical disinfection can enhance removal of plastic surface-associated Pseudomonas aeruginosa. Appl Environ Microbiol 2021; 87:e0098021. [PMID: 34347517 PMCID: PMC8478462 DOI: 10.1128/aem.00980-21] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Opportunistic pathogens can linger on surfaces in hospital and building plumbing environments, leading to infections in at-risk populations. Further, biofilm-associated bacteria are protected from removal and inactivation protocols, such as disinfection. Bacteriophages show promise as tools to treat antibiotic resistant infections. As such, phages may also be useful in environmental applications to prevent newly acquired infections. In the current study, the potential of synergies between bacteriophage and chemical disinfection of the opportunistic pathogen Pseudomonas aeruginosa was assessed under various conditions. Specifically, surface-associated P. aeruginosa was treated with various concentrations of phages (P1 or JG004), chemical disinfectant (sodium hypochlorite or benzalkonium chloride), or combined sequential treatments under three distinct attachment models (spot inoculations, dry biofilms, and wet biofilms). Phages were very effective at removing bacteria in spot inoculation (>3.2 log10 removal) and wet biofilms (up to 2.6 log10 removal), while phages prevented regrowth of dry biofilms in the application time. In addition, phage treatment followed by chemical disinfection inactivated more P. aeruginosa under wet biofilm conditions better than either treatment alone. This effect was hindered when chemical disinfection was applied first, followed by phage treatment, suggesting additive benefits of combination treatments are lost when phage is applied last. Further, we confirm prior evidence of greater phage tolerance to benzalkonium chloride relative to sodium hypochlorite, informing choices for combination phage-disinfectant approaches. Overall, this paper further supports the potential of using combination phage and chemical disinfectant treatments to improve inactivation of surface-associated P. aeruginosa. Importance Phages are already utilized in the healthcare industry to treat antibiotic resistant infections, such as on implant-associated biofilms and in compassionate care cases. Phage treatment could also be a promising new tool to control pathogens in the built environment, preventing infections from occurring. This study shows that phage can be combined effectively with chemical disinfectants to improve removal of wet biofilms and bacteria spotted onto surfaces while preventing regrowth in dry biofilms. This has the potential to improve pathogen containment within the built environment and drinking water infrastructure to prevent infections of opportunistic pathogens.
Collapse
|
50
|
Synergy between Phage Sb-1 and Oxacillin against Methicillin-Resistant Staphylococcus aureus. Antibiotics (Basel) 2021; 10:antibiotics10070849. [PMID: 34356770 PMCID: PMC8300854 DOI: 10.3390/antibiotics10070849] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 12/21/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is a notorious pathogen responsible for not only a number of difficult-to-treat hospital-acquired infections, but also for infections that are community- or livestock-acquired. The increasing lack of efficient antibiotics has renewed the interest in lytic bacteriophages (briefly phages) as additional antimicrobials against multi-drug resistant bacteria, including MRSA. The aim of this study was to test the hypothesis that a combination of the well-known and strictly lytic S. aureus phage Sb-1 and oxacillin, which as sole agent is ineffective against MRSA, exerts a significantly stronger bacterial reduction than either antimicrobial alone. Eighteen different MRSA isolates and, for comparison, five MSSA and four reference strains were included in this study. The bacteria were challenged with a combination of varying dosages of the phage and the antibiotic in liquid medium using five different antibiotic levels and four different viral titers (i.e., multiplicity of infections (MOIs) ranging from 10-5 to 10). The dynamics of the cell density changes were determined via time-kill assays over 16 h. Positive interactions between both antimicrobials in the form of facilitation, additive effects, or synergism were observed for most S. aureus isolates. These enhanced antibacterial effects were robust with phage MOIs of 10-1 and 10 irrespective of the antibiotic concentrations, ranging from 5 to 100 µg/mL. Neutral effects between both antimicrobials were seen only with few isolates. Importantly, antagonism was a rare exception. As a conclusion, phage Sb-1 and oxacillin constitute a robust heterologous antimicrobial pair which extends the efficacy of a phage-only approach for controlling MRSA.
Collapse
|