1
|
Coenye T, Ahonen M, Anderson S, Cámara M, Chundi P, Fields M, Foidl I, Gnimpieba EZ, Griffin K, Hinks J, Loka AR, Lushbough C, MacPhee C, Nater N, Raval R, Slater-Jefferies J, Teo P, Wilks S, Yung M, Webb JS. Global challenges and microbial biofilms: Identification of priority questions in biofilm research, innovation and policy. Biofilm 2024; 8:100210. [PMID: 39221168 PMCID: PMC11364012 DOI: 10.1016/j.bioflm.2024.100210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 09/04/2024] Open
Abstract
Priority question exercises are increasingly used to frame and set future research, innovation and development agendas. They can provide an important bridge between the discoveries, data and outputs generated by researchers, and the information required by policy makers and funders. Microbial biofilms present huge scientific, societal and economic opportunities and challenges. In order to identify key priorities that will help to advance the field, here we review questions from a pool submitted by the international biofilm research community and from practitioners working across industry, the environment and medicine. To avoid bias we used computational approaches to group questions and manage a voting and selection process. The outcome of the exercise is a set of 78 unique questions, categorized in six themes: (i) Biofilm control, disruption, prevention, management, treatment (13 questions); (ii) Resistance, persistence, tolerance, role of aggregation, immune interaction, relevance to infection (10 questions); (iii) Model systems, standards, regulatory, policy education, interdisciplinary approaches (15 questions); (iv) Polymicrobial, interactions, ecology, microbiome, phage (13 questions); (v) Clinical focus, chronic infection, detection, diagnostics (13 questions); and (vi) Matrix, lipids, capsule, metabolism, development, physiology, ecology, evolution environment, microbiome, community engineering (14 questions). The questions presented are intended to highlight opportunities, stimulate discussion and provide focus for researchers, funders and policy makers, informing future research, innovation and development strategy for biofilms and microbial communities.
Collapse
Affiliation(s)
- Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Belgium
- ESCMID Study Group on Biofilms (ESGB), Basel, Switzerland
| | - Merja Ahonen
- Satakunta University of Applied Sciences, Finland
| | - Skip Anderson
- Center for Biofilm Engineering, Montana State University, Bozeman, MT, USA
| | - Miguel Cámara
- National Biofilms Innovation Centre, University of Nottingham Biodiscovery Institute, School of Life Sciences, University of Nottingham, Nottingham, UK
| | | | - Matthew Fields
- Center for Biofilm Engineering, Montana State University, Bozeman, MT, USA
| | - Ines Foidl
- National Biofilms Innovation Centre, School of Physics and Astronomy, University of Edinburgh, Edinburgh, UK
| | | | - Kristen Griffin
- Center for Biofilm Engineering, Montana State University, Bozeman, MT, USA
| | - Jamie Hinks
- Nanyang Technological University, Singapore
- Singapore Centre for Environmental Life Sciences Engineering (SCELSE), Singapore
| | | | | | - Cait MacPhee
- National Biofilms Innovation Centre, School of Physics and Astronomy, University of Edinburgh, Edinburgh, UK
| | - Natasha Nater
- National Biofilms Innovation Centre, School of Biological Sciences, University of Southampton, Southampton, UK
| | - Rasmita Raval
- National Biofilms Innovation Centre, Open Innovation Hub for Antimicrobial Surfaces, Department of Chemistry, University of Liverpool, Liverpool, UK
| | - Jo Slater-Jefferies
- National Biofilms Innovation Centre, School of Biological Sciences, University of Southampton, Southampton, UK
| | - Pauline Teo
- Nanyang Technological University, Singapore
- Singapore Centre for Environmental Life Sciences Engineering (SCELSE), Singapore
| | - Sandra Wilks
- National Biofilms Innovation Centre, School of Biological Sciences, University of Southampton, Southampton, UK
| | - Maria Yung
- Singapore Centre for Environmental Life Sciences Engineering (SCELSE), Singapore
| | | | - Jeremy S. Webb
- National Biofilms Innovation Centre, School of Biological Sciences, University of Southampton, Southampton, UK
| |
Collapse
|
2
|
Voinescu A, Licker M, Muntean D, Musuroi C, Musuroi SI, Izmendi O, Vulpie S, Jumanca R, Munteanu M, Cosnita A. A Comprehensive Review of Microbial Biofilms on Contact Lenses: Challenges and Solutions. Infect Drug Resist 2024; 17:2659-2671. [PMID: 38947374 PMCID: PMC11214797 DOI: 10.2147/idr.s463779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 06/03/2024] [Indexed: 07/02/2024] Open
Abstract
Contact lenses (CL) have become an immensely popular means of vision correction, offering comfort to millions worldwide. However, the persistent issue of biofilm formation on lenses raises significant problems, leading to various ocular complications and discomfort. The aim of this review is to develop safer and more effective strategies for preventing and managing microbial biofilms on CL, improving the eye health and comfort of wearers. Taking these into consideration, the present study investigates the intricate mechanisms of biofilm formation, by exploring the interplay between microbial adhesion, the production of extracellular polymeric substances, and the properties of the lens material itself. Moreover, it emphasizes the diverse range of microorganisms involved, encompassing bacteria, fungi, and other opportunistic pathogens, elucidating their implications within lenses and other medical device-related infections and inflammatory responses. Going beyond the challenges posed by biofilms on CL, this work explores the advancements in biofilm detection techniques and their clinical relevance. It discusses diagnostic tools like confocal microscopy, genetic assays, and emerging technologies, assessing their capacity to identify and quantify biofilm-related infections. Finally, the paper delves into contemporary strategies and innovative approaches for managing and preventing biofilms development on CL. In Conclusion, this review provides insights for eye care practitioners, lens manufacturers, and microbiology researchers. It highlights the intricate interactions between biofilms and CL, serving as a foundation for the development of effective preventive measures and innovative solutions to enhance CL safety, comfort, and overall ocular health. Research into microbial biofilms on CL is continuously evolving, with several future directions being explored to address challenges and improve eye health outcomes as far as CL wearers are concerned.
Collapse
Affiliation(s)
- Adela Voinescu
- Doctoral School “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania
- Department of Microbiology, Multidisciplinary Research Center on Antimicrobial Resistance, “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania
- Clinical Laboratory, “Pius Brinzeu” Emergency Clinical County Hospital, Timisoara, Romania
| | - Monica Licker
- Department of Microbiology, Multidisciplinary Research Center on Antimicrobial Resistance, “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania
- Clinical Laboratory, “Pius Brinzeu” Emergency Clinical County Hospital, Timisoara, Romania
| | - Delia Muntean
- Department of Microbiology, Multidisciplinary Research Center on Antimicrobial Resistance, “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania
- Clinical Laboratory, “Pius Brinzeu” Emergency Clinical County Hospital, Timisoara, Romania
| | - Corina Musuroi
- Department of Microbiology, Multidisciplinary Research Center on Antimicrobial Resistance, “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania
- Clinical Laboratory, “Pius Brinzeu” Emergency Clinical County Hospital, Timisoara, Romania
| | - Silvia Ioana Musuroi
- Doctoral School “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania
| | - Oana Izmendi
- Doctoral School “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania
- Department of Microbiology, Multidisciplinary Research Center on Antimicrobial Resistance, “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania
| | - Silvana Vulpie
- Doctoral School “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania
- Department of Microbiology, Multidisciplinary Research Center on Antimicrobial Resistance, “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania
| | - Romanita Jumanca
- Romanian and Foreign Languages Department, “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania
| | - Mihnea Munteanu
- Department IX, Surgery and Ophthalmology “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania
| | - Andrei Cosnita
- Department IX, Surgery and Ophthalmology “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania
| |
Collapse
|
3
|
Guliy OI, Evstigneeva SS, Shirokov AA, Bunin VD. Sensor system for analysis of biofilm sensitivity to ampicillin. Appl Microbiol Biotechnol 2024; 108:172. [PMID: 38265501 PMCID: PMC10808281 DOI: 10.1007/s00253-023-12831-7] [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: 04/01/2023] [Revised: 11/18/2023] [Accepted: 11/21/2023] [Indexed: 01/25/2024]
Abstract
The resistance of biofilms to antibiotics is a key factor that makes bacterial infections unsusceptible to antimicrobial therapy. The results of classical tests of cell sensitivity to antibiotics cannot be used to predict therapeutic success in infections associated with biofilm formation. We describe a simple and rapid method for the real-time evaluation of bacterial biofilm sensitivity to antibiotics, with Pseudomonas putida and ampicillin as examples. The method uses an electric biosensor to detect the difference between changes in the biofilm electric polarizability, thereby evaluating antibiotic sensitivity. The electric signals showed that P. putida biofilms were susceptible to ampicillin and that at high antibiotic concentrations, the biofilms differed markedly in their susceptibility (dose-dependent effect). The sensor also detected differences between biofilms before and after ampicillin treatment. The electric-signal changes enabled us to describe the physical picture of the processes occurring in bacterial biofilms in the presence of ampicillin. The approach used in this study is promising for evaluating the activity of various compounds against biofilms, because it permits a conclusion about the antibiotic sensitivity of biofilm bacteria to be made in real time and in a short period (analysis time, not longer than 20 min). An added strong point is that analysis can be done directly in liquid, without preliminary sample preparation. KEY POINTS: • Sensor system to analyze biofilm antimicrobial susceptibility is described. • The signal change depended on the ampicillin concentration (dose-dependent effect). • The sensor allows real-time determination of the antibiofilm effect of ampicillin.
Collapse
Affiliation(s)
- Olga I Guliy
- Institute of Biochemistry and Physiology of Plants and Microorganisms - Subdivision of the Federal State Budgetary Research Institution Saratov Federal Scientific Centre of the Russian Academy of Sciences (IBPPM RAS), Saratov, 410049, Russia.
| | - Stella S Evstigneeva
- Institute of Biochemistry and Physiology of Plants and Microorganisms - Subdivision of the Federal State Budgetary Research Institution Saratov Federal Scientific Centre of the Russian Academy of Sciences (IBPPM RAS), Saratov, 410049, Russia
| | - Alexander A Shirokov
- Institute of Biochemistry and Physiology of Plants and Microorganisms - Subdivision of the Federal State Budgetary Research Institution Saratov Federal Scientific Centre of the Russian Academy of Sciences (IBPPM RAS), Saratov, 410049, Russia
| | | |
Collapse
|
4
|
Coenye T. Biofilm antimicrobial susceptibility testing: where are we and where could we be going? Clin Microbiol Rev 2023; 36:e0002423. [PMID: 37812003 PMCID: PMC10732061 DOI: 10.1128/cmr.00024-23] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 07/27/2023] [Indexed: 10/10/2023] Open
Abstract
Our knowledge about the fundamental aspects of biofilm biology, including the mechanisms behind the reduced antimicrobial susceptibility of biofilms, has increased drastically over the last decades. However, this knowledge has so far not been translated into major changes in clinical practice. While the biofilm concept is increasingly on the radar of clinical microbiologists, physicians, and healthcare professionals in general, the standardized tools to study biofilms in the clinical microbiology laboratory are still lacking; one area in which this is particularly obvious is that of antimicrobial susceptibility testing (AST). It is generally accepted that the biofilm lifestyle has a tremendous impact on antibiotic susceptibility, yet AST is typically still carried out with planktonic cells. On top of that, the microenvironment at the site of infection is an important driver for microbial physiology and hence susceptibility; but this is poorly reflected in current AST methods. The goal of this review is to provide an overview of the state of the art concerning biofilm AST and highlight the knowledge gaps in this area. Subsequently, potential ways to improve biofilm-based AST will be discussed. Finally, bottlenecks currently preventing the use of biofilm AST in clinical practice, as well as the steps needed to get past these bottlenecks, will be discussed.
Collapse
Affiliation(s)
- Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
5
|
Rottier W, Seidelman J, Wouthuyzen-Bakker M. Antimicrobial treatment of patients with a periprosthetic joint infection: basic principles. ARTHROPLASTY 2023; 5:10. [PMID: 36864531 PMCID: PMC9979546 DOI: 10.1186/s42836-023-00169-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 01/16/2023] [Indexed: 03/04/2023] Open
Abstract
The antibiotic treatment of periprosthetic joint infections (PJI) is complicated by the presence of biofilm produced by bacteria on the abiotic surface of the implant. Bacteria within the deeper layers of the biofilm become metabolically less active, resulting in antibiotic tolerance due to several mechanisms. This review describes the basic principles of antibiotic treatment in PJI in relation to the behavior of bacteria within the biofilm. The concept of biofilm-active antibiotics will be explained from an in vitro as well as in vivo perspective. Evidence from clinical studies on biofilm-active antibiotics in PJI will be highlighted, mainly focusing on the role of rifampicin for Gram-positive microorganisms and fluoroquinolones for Gram-negative microorganisms. The optimal treatment duration will be discussed as the timing of switching to oral antibiotic therapy.
Collapse
Affiliation(s)
- Wouter Rottier
- grid.4830.f0000 0004 0407 1981Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, 9713 GZ the Netherlands
| | - Jessica Seidelman
- grid.26009.3d0000 0004 1936 7961Division of Infectious Diseases, Duke University School of Medicine, Durham, NC 27708 USA
| | - Marjan Wouthuyzen-Bakker
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, 9713 GZ, the Netherlands.
| |
Collapse
|
6
|
Michaelis C, Grohmann E. Horizontal Gene Transfer of Antibiotic Resistance Genes in Biofilms. Antibiotics (Basel) 2023; 12:antibiotics12020328. [PMID: 36830238 PMCID: PMC9952180 DOI: 10.3390/antibiotics12020328] [Citation(s) in RCA: 68] [Impact Index Per Article: 68.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/08/2023] Open
Abstract
Most bacteria attach to biotic or abiotic surfaces and are embedded in a complex matrix which is known as biofilm. Biofilm formation is especially worrisome in clinical settings as it hinders the treatment of infections with antibiotics due to the facilitated acquisition of antibiotic resistance genes (ARGs). Environmental settings are now considered as pivotal for driving biofilm formation, biofilm-mediated antibiotic resistance development and dissemination. Several studies have demonstrated that environmental biofilms can be hotspots for the dissemination of ARGs. These genes can be encoded on mobile genetic elements (MGEs) such as conjugative and mobilizable plasmids or integrative and conjugative elements (ICEs). ARGs can be rapidly transferred through horizontal gene transfer (HGT) which has been shown to occur more frequently in biofilms than in planktonic cultures. Biofilm models are promising tools to mimic natural biofilms to study the dissemination of ARGs via HGT. This review summarizes the state-of-the-art of biofilm studies and the techniques that visualize the three main HGT mechanisms in biofilms: transformation, transduction, and conjugation.
Collapse
|
7
|
Tuon FF, Suss PH, Telles JP, Dantas LR, Borges NH, Ribeiro VST. Antimicrobial Treatment of Staphylococcus aureus Biofilms. Antibiotics (Basel) 2023; 12:87. [PMID: 36671287 PMCID: PMC9854895 DOI: 10.3390/antibiotics12010087] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/01/2023] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Staphylococcus aureus is a microorganism frequently associated with implant-related infections, owing to its ability to produce biofilms. These infections are difficult to treat because antimicrobials must cross the biofilm to effectively inhibit bacterial growth. Although some antibiotics can penetrate the biofilm and reduce the bacterial load, it is important to understand that the results of routine sensitivity tests are not always valid for interpreting the activity of different drugs. In this review, a broad discussion on the genes involved in biofilm formation, quorum sensing, and antimicrobial activity in monotherapy and combination therapy is presented that should benefit researchers engaged in optimizing the treatment of infections associated with S. aureus biofilms.
Collapse
Affiliation(s)
- Felipe Francisco Tuon
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil
| | - Paula Hansen Suss
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil
| | - Joao Paulo Telles
- AC Camargo Cancer Center, Infectious Diseases Department, São Paulo 01525-001, São Paulo, Brazil
| | - Leticia Ramos Dantas
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil
| | - Nícolas Henrique Borges
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil
| | - Victoria Stadler Tasca Ribeiro
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba 80215-901, Paraná, Brazil
| |
Collapse
|
8
|
Neuhaus S, Feßler AT, Dieckmann R, Thieme L, Pletz MW, Schwarz S, Al Dahouk S. Towards a Harmonized Terminology: A Glossary for Biocide Susceptibility Testing. Pathogens 2022; 11:1455. [PMID: 36558789 PMCID: PMC9780826 DOI: 10.3390/pathogens11121455] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/03/2022] Open
Abstract
Disinfection is a key strategy to reduce the burden of infections. The contact of bacteria to biocides-the active substances of disinfectants-has been linked to bacterial adaptation and the development of antimicrobial resistance. Currently, there is no scientific consensus on whether the excessive use of biocides contributes to the emergence and spread of multidrug resistant bacteria. The comprehensive analysis of available data remains a challenge because neither uniform test procedures nor standardized interpretive criteria nor harmonized terms are available to describe altered bacterial susceptibility to biocides. In our review, we investigated the variety of criteria and the diversity of terms applied to interpret findings in original studies performing biocide susceptibility testing (BST) of field isolates. An additional analysis of reviews summarizing the knowledge of individual studies on altered biocide susceptibility provided insights into currently available broader concepts for data interpretation. Both approaches pointed out the urgent need for standardization. We, therefore, propose that the well-established and approved concepts for interpretation of antimicrobial susceptibility testing data should serve as a role model to evaluate biocide resistance mechanisms on a single cell level. Furthermore, we emphasize the adaptations necessary to acknowledge the specific needs for the evaluation of BST data. Our approach might help to increase scientific awareness and acceptance.
Collapse
Affiliation(s)
- Szilvia Neuhaus
- German Federal Institute for Risk Assessment, 10589 Berlin, Germany
| | - Andrea T. Feßler
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, 14163 Berlin, Germany
- Veterinary Centre for Resistance Research (TZR), Freie Universität Berlin, 14163 Berlin, Germany
| | - Ralf Dieckmann
- German Federal Institute for Risk Assessment, 10589 Berlin, Germany
| | - Lara Thieme
- Institute of Infectious Diseases and Infection Control, Jena University Hospital, Friedrich-Schiller-University Jena, 07747 Jena, Germany
- Leibniz Center for Photonics in Infection Research, Jena University Hospital, Friedrich Schiller University Jena, 07747 Jena, Germany
| | - Mathias W. Pletz
- Institute of Infectious Diseases and Infection Control, Jena University Hospital, Friedrich-Schiller-University Jena, 07747 Jena, Germany
| | - Stefan Schwarz
- Centre for Infection Medicine, Department of Veterinary Medicine, Institute of Microbiology and Epizootics, Freie Universität Berlin, 14163 Berlin, Germany
- Veterinary Centre for Resistance Research (TZR), Freie Universität Berlin, 14163 Berlin, Germany
| | - Sascha Al Dahouk
- German Federal Institute for Risk Assessment, 10589 Berlin, Germany
- Department of Internal Medicine, RWTH Aachen University Hospital, 52074 Aachen, Germany
| |
Collapse
|
9
|
Eckert JA, Rosenberg M, Rhen M, Choong FX, Richter-Dahlfors A. An optotracer-based antibiotic susceptibility test specifically targeting the biofilm lifestyle of Salmonella. Biofilm 2022; 4:100083. [PMID: 36117547 PMCID: PMC9474290 DOI: 10.1016/j.bioflm.2022.100083] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/25/2022] Open
Abstract
Antimicrobial resistance is a medical threat of global dimensions. Proper antimicrobial susceptibility testing (AST) for drug development, patient diagnosis and treatment is crucial to counteract ineffective drug use and resistance development. Despite the important role of bacterial biofilms in chronic and device-associated infections, the efficacy of antibiotics is determined using planktonic cultures. To address the need for antibiotics targeting bacteria in the biofilm lifestyle, we here present an optotracing-based biofilm-AST using Salmonella as model. Our non-disruptive method enables real-time recording of the extracellular matrix (ECM) components, providing specific detection of the biofilm lifestyle. Biofilm formation prior to antibiotic challenge can thus be confirmed and pre-treatment data collected. By introducing Kirby-Bauer discs, we performed a broad screen of the effects of antibiotics representing multiple classes, and identified compounds with ECM inhibitory as well as promoting effects. These compounds were further tested in agar-based dose-response biofilm-AST assays. By quantifying the ECM based on the amount of curli, and by visualizing the biofilm size and morphology, we achieved new information directly reflecting the treated biofilm. This verified the efficacy of several antibiotics that were effective in eradicating pre-formed biofilms, and it uncovered intriguing possible resistance mechanisms initiated in response to treatments. By providing deeper insights into the resistances and susceptibilities of microbes, expanded use of the biofilm-AST will contribute to more effective treatments of infections and reduced resistance development.
Collapse
Affiliation(s)
- Johannes A Eckert
- AIMES - Center for the Advancement of Integrated Medical and Engineering Sciences, Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, Sweden.,Department of Neuroscience, Karolinska Institutet, Solnavägen 9, SE-171 77, Stockholm, Sweden
| | - Ming Rosenberg
- AIMES - Center for the Advancement of Integrated Medical and Engineering Sciences, Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, Sweden.,Department of Neuroscience, Karolinska Institutet, Solnavägen 9, SE-171 77, Stockholm, Sweden
| | - Mikael Rhen
- AIMES - Center for the Advancement of Integrated Medical and Engineering Sciences, Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, Sweden.,Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Solnavägen 9, SE-171 77, Stockholm, Sweden
| | - Ferdinand X Choong
- AIMES - Center for the Advancement of Integrated Medical and Engineering Sciences, Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, Sweden.,Department of Neuroscience, Karolinska Institutet, Solnavägen 9, SE-171 77, Stockholm, Sweden
| | - Agneta Richter-Dahlfors
- AIMES - Center for the Advancement of Integrated Medical and Engineering Sciences, Karolinska Institutet and KTH Royal Institute of Technology, Stockholm, Sweden.,Department of Neuroscience, Karolinska Institutet, Solnavägen 9, SE-171 77, Stockholm, Sweden
| |
Collapse
|
10
|
Tetz G, Tetz V. Overcoming Antibiotic Resistance with Novel Paradigms of Antibiotic Selection. Microorganisms 2022; 10:2383. [PMID: 36557636 PMCID: PMC9781420 DOI: 10.3390/microorganisms10122383] [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: 11/05/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/04/2022] Open
Abstract
Conventional antimicrobial susceptibility tests, including phenotypic and genotypic methods, are insufficiently accurate and frequently fail to identify effective antibiotics. These methods predominantly select therapies based on the antibiotic response of only the lead bacterial pathogen within pure bacterial culture. However, this neglects the fact that, in the majority of human infections, the lead bacterial pathogens are present as a part of multispecies communities that modulate the response of these lead pathogens to antibiotics and that multiple pathogens can contribute to the infection simultaneously. This discrepancy is a major cause of the failure of antimicrobial susceptibility tests to detect antibiotics that are effective in vivo. This review article provides a comprehensive overview of the factors that are missed by conventional antimicrobial susceptibility tests and it explains how accounting for these methods can aid the development of novel diagnostic approaches.
Collapse
Affiliation(s)
- George Tetz
- Human Microbiology Institute, New York, NY 100141, USA
| | | |
Collapse
|
11
|
Meléndez-Carmona MÁ, Mancheño-Losa M, Ruiz-Sorribas A, Muñoz-Gallego I, Viedma E, Chaves F, Van Bambeke F, Lora-Tamayo J. Strain-to-strain variability among Staphylococcus aureus causing prosthetic joint infection drives heterogeneity in response to levofloxacin and rifampicin. J Antimicrob Chemother 2022; 77:3265-3269. [PMID: 36124848 DOI: 10.1093/jac/dkac311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 08/19/2022] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Levofloxacin and rifampicin are the preferred treatment for prosthetic joint infection (PJI) caused by Staphylococcus aureus, especially when managed with implant retention (DAIR). However, a significant variability of success has been reported, which could be related to intrinsic characteristics of the microorganism. Our aim was to evaluate the variability in the anti-biofilm response to levofloxacin and rifampicin in a clinical collection of S. aureus. MATERIAL AND METHODS Eleven levofloxacin- and rifampicin-susceptible S. aureus isolates causing PJI managed with DAIR were included. Levofloxacin, rifampicin and levofloxacin + rifampicin were tested in an in vitro static biofilm model in microtitre plates, where 48 h biofilms were challenged with antimicrobials during 24 h. Additionally, two genetically similar strains were tested in the CDC Biofilm Reactor, where 48 h biofilms were treated during 56 h. Antimicrobial activity was assessed by viable biofilm-embedded cells recount, and by crystal violet staining. RESULTS All antimicrobial regimens showed significant anti-biofilm activity, but a notable scattering in the response was observed across all strains (inter-strain coefficient of variation for levofloxacin, rifampicin and levofloxacin + rifampicin of 22.8%, 35.8% and 34.5%, respectively). This variability was tempered with the combination regimen when tested in the biofilm reactor. No correlation was observed between the minimal biofilm eradicative concentration and the antimicrobial activity. Recurrent S. aureus isolates exhibited higher biofilm-forming ability compared with strains from resolved infections (7.6 log10 cfu/cm2±0.50 versus 9.0 log10 cfu±0.07). CONCLUSIONS Significant variability may be expected in response to levofloxacin and rifampicin among biofilm-embedded S. aureus. A response in the lower range, together with other factors of bad prognosis, could be responsible of treatment failure.
Collapse
Affiliation(s)
- María Ángeles Meléndez-Carmona
- Department of Clinical Microbiology, Hospital Universitario 12 de Octubre, Instituto de Investigación Biomédica 'i+12' Hospital 12 de Octubre, Madrid, Spain
| | - Mikel Mancheño-Losa
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, Instituto de Investigación Biomédica 'i+12' Hospital 12 de Octubre, Madrid, Spain
| | - Albert Ruiz-Sorribas
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Irene Muñoz-Gallego
- Department of Clinical Microbiology, Hospital Universitario 12 de Octubre, Instituto de Investigación Biomédica 'i+12' Hospital 12 de Octubre, Madrid, Spain
| | - Esther Viedma
- Department of Clinical Microbiology, Hospital Universitario 12 de Octubre, Instituto de Investigación Biomédica 'i+12' Hospital 12 de Octubre, Madrid, Spain
| | - Fernando Chaves
- Department of Clinical Microbiology, Hospital Universitario 12 de Octubre, Instituto de Investigación Biomédica 'i+12' Hospital 12 de Octubre, Madrid, Spain
| | - Françoise Van Bambeke
- Pharmacologie cellulaire et moléculaire, Louvain Drug Research Institute, Université Catholique de Louvain, Brussels, Belgium
| | - Jaime Lora-Tamayo
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, Instituto de Investigación Biomédica 'i+12' Hospital 12 de Octubre, Madrid, Spain.,CIBER Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
12
|
Lora-Tamayo J, Meléndez-Carmona MÁ. The clinical meaning of biofilm formation ability: The importance of context. ENFERMEDADES INFECCIOSAS Y MICROBIOLOGIA CLINICA (ENGLISH ED.) 2022; 40:415-417. [PMID: 36195404 DOI: 10.1016/j.eimce.2022.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 02/22/2022] [Indexed: 06/16/2023]
Affiliation(s)
- Jaime Lora-Tamayo
- Department of Internal Medicine, Hospital Universitario 12 de Octubre, Spain; Instituto de Investigación Biomédica "imas12" Hospital 12 de Octubre, Spain; CIBER de Enfermedades Infecciosas CIBERINFEC, Instituto de Salud Carlos III, Spain.
| | - Mª Ángeles Meléndez-Carmona
- Department of Microbiology, Hospital Universitario 12 de Octubre, Spain; Instituto de Investigación Biomédica "imas12" Hospital 12 de Octubre, Spain
| |
Collapse
|
13
|
Tillander JAN, Rilby K, Svensson Malchau K, Skovbjerg S, Lindberg E, Rolfson O, Trobos M. Treatment of periprosthetic joint infections guided by minimum biofilm eradication concentration (MBEC) in addition to minimum inhibitory concentration (MIC): protocol for a prospective randomised clinical trial. BMJ Open 2022; 12:e058168. [PMID: 36109038 PMCID: PMC9478849 DOI: 10.1136/bmjopen-2021-058168] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
INTRODUCTION Prosthetic joint infections (PJIs) are disastrous complications for patients and costly for healthcare organisations. They may promote bacterial resistance due to the extensive antibiotic use necessary in the PJI treatment. The PJI incidence is estimated to be 1%-3%, but the absolute numbers worldwide are high and increasing as large joint arthroplasties are performed by the millions each year. Current treatment algorithms, based on implant preserving surgery or full revision followed by a semitailored antibiotic regimen for no less than 2-3 months, lead to infection resolution in approximately 60% and 90%, respectively. Antibiotic choice is currently guided by minimum inhibitory concentrations (MICs) of free-living bacteria and not of bacteria in biofilm growth mode. Biofilm assays with relatively rapid output for the determination of minimum biofilm eradication concentrations (MBECs) have previously been developed but their clinical usefulness have not been established. METHODS AND ANALYSIS This single-blinded, two-arm randomised study of hip or knee staphylococcal PJI will evaluate 6-week standard of care (MIC guided), or an alternative antibiotic regimen according to an MBEC-guided-based decision algorithm. Sixty-four patients with a first-time PJI treated according to the debridement, antibiotics, and implant retention principle will be enrolled at a single tertiary orthopaedic centre (Sahlgrenska University Hospital). Patients will receive 14 days of standard parenteral antibiotics before entering the comparative study arms. The primary outcome measurement is the proportion of changes in antimicrobial regimen from first-line treatment dependent on randomisation arm. Secondary endpoints are unresolved infection, how microbial properties including biofilm abilities and emerging antimicrobial resistance correlate to infection outcomes, patient reported outcomes and costs with a 12-month follow-up. ETHICS AND DISSEMINATION Approval is received from the Swedish Ethical Review Authority, no 2020-01471 and the Swedish Medical Products Agency, EudraCT, no 2020-003444-80. TRIAL REGISTRATION NUMBER ClinicalTrials.gov ID: NCT04488458.
Collapse
Affiliation(s)
- Jonatan A N Tillander
- Department of Infectious Diseases, Sahlgrenska University Hospital, Goteborg, Sweden
- Department of Infectious Diseases, Institute of Biomedicine, University of Gothenburg, Goteborg, Sweden
| | - Karin Rilby
- Department of Orthopaedics, Sahlgrenska University Hospital, Goteborg, Sweden
| | - Karin Svensson Malchau
- Department of Orthopaedics, Institute of Clinical Sciences, Sahlgrenska University Hospital, Goteborg, Sweden
| | - Susann Skovbjerg
- Department of Clinical Microbiology, Institute of Biomedicine, University of Gothenburg, Goteborg, Sweden
- Department of Clinical Microbiology, Sahlgrenska University Hospital, Goteborg, Sweden
| | - Erika Lindberg
- Department of Clinical Microbiology, Institute of Biomedicine, University of Gothenburg, Goteborg, Sweden
| | - Ola Rolfson
- Department of Orthopaedics, Institute of Clinical Sciences, Sahlgrenska University Hospital, Goteborg, Sweden
| | - Margarita Trobos
- Department of Biomaterials, University of Gothenburg Sahlgrenska Academy, Goteborg, Sweden
| |
Collapse
|
14
|
Villarreal-Salazar V, Bocanegra-Ibarias P, Villarreal-Treviño L, Salas-Treviño D, Morfin-Otero R, Camacho-Ortiz A, Flores-Treviño S. Improvement of antimicrobial susceptibility testing in biofilm-growingcoagulase-negative Staphylococcus hominis. J Microbiol Methods 2022; 198:106493. [DOI: 10.1016/j.mimet.2022.106493] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/20/2022] [Accepted: 05/21/2022] [Indexed: 11/30/2022]
|
15
|
De Waele JJ, Boelens J, Van De Putte D, Huis In ‘t Veld D, Coenye T. The Role of Abdominal Drain Cultures in Managing Abdominal Infections. Antibiotics (Basel) 2022; 11:697. [PMID: 35625341 PMCID: PMC9137968 DOI: 10.3390/antibiotics11050697] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 05/05/2022] [Accepted: 05/17/2022] [Indexed: 02/01/2023] Open
Abstract
Intra-abdominal infections (IAI) are common in hospitalized patients, both in and outside of the intensive care unit. Management principles include antimicrobial therapy and source control. Typically, these infections are polymicrobial, and intra-operative samples will guide the targeted antimicrobial therapy. Although the use of prophylactic abdominal drains in patients undergoing abdominal surgery is decreasing, the use of drains to treat IAI, both in surgical and non-surgical strategies for abdominal infection, is increasing. In this context, samples from abdominal drains are often used to assist in antimicrobial decision making. In this narrative review, we provide an overview of the current role of abdominal drains in surgery, discuss the importance of biofilm formation in abdominal drains and the mechanisms involved, and review the clinical data on the use of sampling these drains for diagnostic purposes. We conclude that biofilm formation and the colonization of abdominal drains is common, which precludes the use of abdominal fluid to reliably diagnose IAI and identify the pathogens involved. We recommend limiting the use of drains and, when present, avoiding routine microbiological sampling.
Collapse
Affiliation(s)
- Jan J. De Waele
- Department of Intensive Care Medicine, Ghent University Hospital, 9000 Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Jerina Boelens
- Department of Medical Microbiology, Ghent University Hospital, 9000 Ghent, Belgium;
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, Belgium
| | - Dirk Van De Putte
- Department of Gastrointestinal Surgery, Ghent University Hospital, 9000 Ghent, Belgium;
| | - Diana Huis In ‘t Veld
- Department of Internal Medicine and Infectious Diseases, Ghent University Hospital, 9000 Ghent, Belgium;
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, 9000 Ghent, Belgium;
| |
Collapse
|
16
|
Efficacy of Surgical/Wound Washes against Bacteria: Effect of Different In Vitro Models. MATERIALS 2022; 15:ma15103630. [PMID: 35629656 PMCID: PMC9145943 DOI: 10.3390/ma15103630] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 05/08/2022] [Accepted: 05/17/2022] [Indexed: 01/03/2023]
Abstract
Topical antiseptics are often used to treat chronic wounds with biofilm infections and during salvage of biofilm contaminated implants, but their antibacterial efficacy is frequently only tested against non-aggregated planktonic or free-swimming organisms. This study evaluated the antibacterial and antibiofilm efficacy of four commercial surgical washes Bactisure, TorrenTX, minimally invasive lavage (MIS), and Betadine against six bacterial species: Staphylococcus epidermidis, Staphylococcus aureus, Streptococcus pyogenes, Acinetobacter baumannii, Pseudomonas aeruginosa, and Escherichia coli, which are commonly isolated from surgical site infections and chronic wound infections using different in vitro models. We determined minimum planktonic inhibitory and eradication concentration and minimum 1-day-old biofilm inhibition and eradication concentration of antiseptics in 96-well plates format with 24 h contact time. We also tested the efficacy of antiseptics at in-use concentration and contact time in the presence of biological soil against 3-day-old biofilm grown on coupons with shear in a bioreactor, such that the results are more applicable to the clinical biofilm situations. In the 96-well plate model, the minimum concentration required to inhibit or kill planktonic and biofilm bacteria was lower for Bactisure and TorrenTX than for MIS and Betadine. However, Betadine and Bactisure showed better antibiofilm efficacy than TorrenTX and MIS in the 3-day-old biofilm bioreactor model at in-use concentration. The minimal concentration of surgical washes required to inhibit or kill planktonic bacterial cells and biofilms varies, suggesting the need for the development and use of biofilm-based assays to assess antimicrobial therapies, such as topical antiseptics and their effective concentrations. The antibiofilm efficacy of surgical washes against different bacterial species also varies, highlighting the importance of testing against various bacterial species to achieve a thorough understanding of their efficacy.
Collapse
|
17
|
Pouget C, Dunyach-Remy C, Bernardi T, Provot C, Tasse J, Sotto A, Lavigne JP. A Relevant Wound-Like in vitro Media to Study Bacterial Cooperation and Biofilm in Chronic Wounds. Front Microbiol 2022; 13:705479. [PMID: 35464992 PMCID: PMC9019750 DOI: 10.3389/fmicb.2022.705479] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 02/28/2022] [Indexed: 11/23/2022] Open
Abstract
Biofilm on the skin surface of chronic wounds is an important factor in the pathology, inhibiting wound healing. The polymicrobial nature of these infected wounds and bacterial interactions inside this pathogenic biofilm are the keys for understanding chronic infection. The aim of our work was to develop an innovative in vitro medium that closely mimics the chronic wound emphasizing the microbiological, cellular, and inflammatory environment of chronic wounds but also focusing on the pH found at the wound level. This new medium, called chronic wound medium (CWM), will thus facilitate the study of pathogenic biofilm organization. Clinical Staphylococcus aureus and Pseudomonas aeruginosa strains coisolated from diabetic foot infection were collected and cultivated in this new medium for 24 h in monoculture and coculture. Bacterial growth (growth curves), presence of small colony variant (SCV), biofilm formation (BioFilm Ring Test® assay, biofilm biomass quantification), and virulence (survival curve in a Caenorhabditis elegans model) were evaluated. After 24 h in the in vitro conditions, we observed that P. aeruginosa growth was not affected, compared with a control bacterial medium, whereas for S. aureus, the stationary phase was reduced by two logs. Interestingly, S. aureus growth increased when cocultured with P. aeruginosa in CWM. In coculture with P. aeruginosa, SCV forms of S. aureus were detected. Biofilm studies showed that bacteria, alone and in combination, formed biofilm faster (as soon as 3 h) than the bacteria exposed in a control medium (as soon as 5 h). The virulence of all strains decreased in the nematode model when cultivated in our new in vitro medium. Taken together, our data confirmed the impact of the chronic wound environment on biofilm formation and bacteria virulence. They indicated that P. aeruginosa and S. aureus cooperated in coinfected wounds. Therefore, this in vitro model provides a new tool for bacterial cooperation investigation and polymicrobial biofilm formation.
Collapse
Affiliation(s)
- Cassandra Pouget
- Virulence Bactérienne et Infections Chroniques, INSERM U1047, Université de Montpellier, Nîmes, France.,Biofilm Pharma SAS, Saint-Beauzire, France
| | - Catherine Dunyach-Remy
- Virulence Bactérienne et Infections Chroniques, INSERM U1047, Université de Montpellier, Department of Microbiology and Hospital Hygiene, CHU Nîmes, Nîmes, France
| | | | | | | | - Albert Sotto
- Virulence Bactérienne et Infections Chroniques, INSERM U1047, Université de Montpellier, Department of Infectious and Tropical Diseases, CHU Nîmes, Nîmes, France
| | - Jean-Philippe Lavigne
- Virulence Bactérienne et Infections Chroniques, INSERM U1047, Université de Montpellier, Department of Microbiology and Hospital Hygiene, CHU Nîmes, Nîmes, France
| |
Collapse
|
18
|
Biofilm Formation by Pathogenic Bacteria: Applying a Staphylococcus aureus Model to Appraise Potential Targets for Therapeutic Intervention. Pathogens 2022; 11:pathogens11040388. [PMID: 35456063 PMCID: PMC9027693 DOI: 10.3390/pathogens11040388] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 01/02/2023] Open
Abstract
Carried in the nasal passages by up to 30% of humans, Staphylococcus aureus is recognized to be a successful opportunistic pathogen. It is a frequent cause of infections of the upper respiratory tract, including sinusitis, and of the skin, typically abscesses, as well as of food poisoning and medical device contamination. The antimicrobial resistance of such, often chronic, health conditions is underpinned by the unique structure of bacterial biofilm, which is the focus of increasing research to try to overcome this serious public health challenge. Due to the protective barrier of an exopolysaccharide matrix, bacteria that are embedded within biofilm are highly resistant both to an infected individual’s immune response and to any treating antibiotics. An in-depth appraisal of the stepwise progression of biofilm formation by S. aureus, used as a model infection for all cases of bacterial antibiotic resistance, has enhanced understanding of this complicated microscopic structure and served to highlight possible intervention targets for both patient cure and community infection control. While antibiotic therapy offers a practical means of treatment and prevention, the most favorable results are achieved in combination with other methods. This review provides an overview of S. aureus biofilm development, outlines the current range of anti-biofilm agents that are used against each stage and summarizes their relative merits.
Collapse
|
19
|
Lora-Tamayo J, Meléndez-Carmona MÁ. The clinical meaning of biofilm formation ability: The importance of context. Enferm Infecc Microbiol Clin 2022. [DOI: 10.1016/j.eimc.2022.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
20
|
Thöming JG, Häussler S. Pseudomonas aeruginosa Is More Tolerant Under Biofilm Than Under Planktonic Growth Conditions: A Multi-Isolate Survey. Front Cell Infect Microbiol 2022; 12:851784. [PMID: 35295755 PMCID: PMC8920030 DOI: 10.3389/fcimb.2022.851784] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 01/24/2022] [Indexed: 01/14/2023] Open
Abstract
Biofilm-associated bacteria exhibit profound changes in bacterial physiology. They thrive in the environment but also in the human host in protected sessile communities. Antimicrobial therapy usually fails, despite the absence of genotypic resistance, and it is commonly accepted that biofilm-grown bacteria are up to 1,000-fold more resistant than planktonic cells. We are only at the beginning to understand the reasons for biofilm recalcitrance, and systematic approaches to describe biofilm-induced tolerance phenotypes are lacking. In this study, we investigated a large and highly diverse collection of 352 clinical Pseudomonas aeruginosa isolates for their antimicrobial susceptibility profiles under biofilm growth conditions towards the antibiotics ciprofloxacin, tobramycin, and colistin. We discovered characteristic patterns of drug-specific killing activity and detected conditional tolerance levels far lower (in the range of the minimal inhibitory concentration (MIC)), but also far higher (up to 16,000-fold increase compared to planktonic cells) than generally believed. This extremely broad distribution of biofilm-induced tolerance phenotypes across the clinical isolates was greatly influenced by the choice of the antibiotic. We furthermore describe cross-tolerance against ciprofloxacin and tobramycin, but not colistin, and observed an additive activity between biofilm-induced tolerance and genetically determined resistance. This became less evident when the biofilm-grown cells were exposed to very high antibiotic concentrations. Although much more remains to be learned on the molecular mechanisms underlying biofilm-induced tolerance, our data on intra-species variations in tolerance profiles provide valuable new insights. Furthermore, our observation that colistin appears to act independently of the tolerance mechanisms of individual clinical strains could make colistin a valuable therapeutic option in chronic biofilm-associated infections characterized by the presence of particularly tolerant strains.
Collapse
Affiliation(s)
- Janne G. Thöming
- Department of Clinical Microbiology, University Hospital Copenhagen, Rigshospitalet, Copenhagen, Denmark
- Molecular Bacteriology, Twincore Center for Experimental and Clinical Infection Research GmbH, Hannover, Germany
| | - Susanne Häussler
- Department of Clinical Microbiology, University Hospital Copenhagen, Rigshospitalet, Copenhagen, Denmark
- Molecular Bacteriology, Twincore Center for Experimental and Clinical Infection Research GmbH, Hannover, Germany
- Molecular Bacteriology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- *Correspondence: Susanne Häussler,
| |
Collapse
|
21
|
Sultan AR, Tavakol M, Lemmens-den Toom NA, Croughs PD, Verkaik NJ, Verbon A, van Wamel WJB. Real time monitoring of Staphylococcus aureus biofilm sensitivity towards antibiotics with isothermal microcalorimetry. PLoS One 2022; 17:e0260272. [PMID: 35171906 PMCID: PMC8849495 DOI: 10.1371/journal.pone.0260272] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 11/05/2021] [Indexed: 12/14/2022] Open
Abstract
Biofilm-associated infections with Staphylococcus aureus are difficult to treat even after administration of antibiotics that according to the standard susceptibility assays are effective. Currently, the assays used in the clinical laboratories to determine the sensitivity of S. aureus towards antibiotics are not representing the behaviour of biofilm-associated S. aureus, since these assays are performed on planktonic bacteria. In research settings, microcalorimetry has been used for antibiotic susceptibility studies. Therefore, in this study we investigated if we can use isothermal microcalorimetry to monitor the response of biofilm towards antibiotic treatment in real-time. We developed a reproducible method to generate biofilm in an isothermal microcalorimeter setup. Using this system, the sensitivity of 5 methicillin-sensitive S. aureus (MSSA) and 5 methicillin-resistant S. aureus (MRSA) strains from different genetic lineages were determined towards: flucloxacillin, cefuroxime, cefotaxime, gentamicin, rifampicin, vancomycin, levofloxacin, clindamycin, erythromycin, linezolid, fusidic acid, co-trimoxazole, and doxycycline. In contrast to conventional assays, our calorimetry-based biofilm susceptibility assay showed that S. aureus biofilms, regardless MSSA or MRSA, can survive the exposure to the maximum serum concentration of all tested antibiotics. The only treatment with a single antibiotic showing a significant reduction in biofilm survival was rifampicin, yet in 20% of the strains, emerging antibiotic resistance was observed. Furthermore, the combination of rifampicin with flucloxacillin, vancomycin or levofloxacin was able to prevent S. aureus biofilm from becoming resistant to rifampicin. Isothermal microcalorimetry allows real-time monitoring of the sensitivity of S. aureus biofilms towards antibiotics in a fast and reliable way.
Collapse
Affiliation(s)
- Andi Rofian Sultan
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
- Department of Microbiology, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Mehri Tavakol
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Nicole A. Lemmens-den Toom
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Peter D. Croughs
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Nelianne J. Verkaik
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Annelies Verbon
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Willem J. B. van Wamel
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Center, Rotterdam, The Netherlands
- * E-mail:
| |
Collapse
|
22
|
Ekkelenkamp MB, Díez-Aguilar M, Tunney MM, Elborn JS, Fluit AC, Cantón R. Establishing antimicrobial susceptibility testing methods and clinical breakpoints for inhaled antibiotic therapy. Open Forum Infect Dis 2022; 9:ofac082. [PMID: 35265731 PMCID: PMC8900927 DOI: 10.1093/ofid/ofac082] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/10/2022] [Indexed: 12/05/2022] Open
Abstract
Inhaled antibiotics are a common and valuable therapy for patients suffering from chronic lung infection, with this particularly well demonstrated for patients with cystic fibrosis. However, in vitro tests to predict patient response to inhaled antibiotic therapy are currently lacking. There are indications that antimicrobial susceptibility testing (AST) may have a role in guidance of therapy, but which tests would correlate best still needs to be researched in clinical studies or animal models. Applying the principles of European Committee on Antimicrobial Susceptibility Testing methodology, the analysis of relevant and reliable data correlating different AST tests to patients’ outcomes may yield clinical breakpoints for susceptibility, but these data are currently unavailable. At present, we believe that it is unlikely that standard determination of minimum inhibitory concentration will prove the best predictor.
Collapse
Affiliation(s)
- Miquel B Ekkelenkamp
- University Medical Center Utrecht, Department of Medical Microbiology, Utrecht, The Netherlands
| | - María Díez-Aguilar
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Servicio de Microbiología y Parasitología, Hospital Universitario La Princesa, Madrid, Spain
| | - Michael M Tunney
- Queen’s University Belfast, Department of Pulmonology, Belfast, United Kingdom
| | - J Stuart Elborn
- Queen’s University Belfast, Department of Pulmonology, Belfast, United Kingdom
| | - Ad C Fluit
- University Medical Center Utrecht, Department of Medical Microbiology, Utrecht, The Netherlands
| | - Rafael Cantón
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
- CIBER de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| |
Collapse
|
23
|
Thieme L, Hartung A, Tramm K, Graf J, Spott R, Makarewicz O, Pletz MW. Adaptation of the Start-Growth-Time Method for High-Throughput Biofilm Quantification. Front Microbiol 2021; 12:631248. [PMID: 34512560 PMCID: PMC8428173 DOI: 10.3389/fmicb.2021.631248] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 07/29/2021] [Indexed: 11/29/2022] Open
Abstract
Colony forming unit (CFU) determination by agar plating is still regarded as the gold standard for biofilm quantification despite being time- and resource-consuming. Here, we propose an adaption of the high-throughput Start-Growth-Time (SGT) method from planktonic to biofilm analysis, which indirectly quantifies CFU/mL numbers by evaluating regrowth curves of detached biofilms. For validation, the effect of dalbavancin, rifampicin and gentamicin against mature biofilms of Staphylococcus aureus and Enterococcus faecium was measured by accessing different features of the viability status of the cell, i.e., the cultivability (conventional agar plating), growth behavior (SGT) and metabolic activity (resazurin assay). SGT correlated well with the resazurin assay for all tested antibiotics, but only for gentamicin and rifampicin with conventional agar plating. Dalbavancin treatment-derived growth curves showed a compared to untreated controls significantly slower increase with reduced cell doubling times and reduced metabolic rate, but no change in CFU numbers was observed by conventional agar plating. Here, unspecific binding of dalbavancin to the biofilm interfered with the SGT methodology since the renewed release of dalbavancin during detachment of the biofilms led to an unintended antimicrobial effect. The application of the SGT method for anti-biofilm testing is therefore not suited for antibiotics which stick to the biofilm and/or to the bacterial cell wall. Importantly, the same applies for the well-established resazurin method for anti-biofilm testing. However, for antibiotics which do not bind to the biofilm as seen for gentamicin and rifampicin, the SGT method presents a much less labor-intensive method suited for high-throughput screening of anti-biofilm compounds.
Collapse
Affiliation(s)
- Lara Thieme
- Institute for Infectious Diseases and Infection Control, Jena University Hospital/Friedrich-Schiller-University, Jena, Germany.,Leibniz Center for Photonics in Infection Research, Jena University Hospital/Friedrich-Schiller-University, Jena, Germany
| | - Anita Hartung
- Institute for Infectious Diseases and Infection Control, Jena University Hospital/Friedrich-Schiller-University, Jena, Germany
| | - Kristina Tramm
- Institute for Infectious Diseases and Infection Control, Jena University Hospital/Friedrich-Schiller-University, Jena, Germany
| | - Julia Graf
- Institute for Infectious Diseases and Infection Control, Jena University Hospital/Friedrich-Schiller-University, Jena, Germany
| | - Riccardo Spott
- Institute for Infectious Diseases and Infection Control, Jena University Hospital/Friedrich-Schiller-University, Jena, Germany
| | - Oliwia Makarewicz
- Institute for Infectious Diseases and Infection Control, Jena University Hospital/Friedrich-Schiller-University, Jena, Germany
| | - Mathias W Pletz
- Institute for Infectious Diseases and Infection Control, Jena University Hospital/Friedrich-Schiller-University, Jena, Germany
| |
Collapse
|
24
|
Bjarnsholt T, Whiteley M, Rumbaugh KP, Stewart PS, Jensen PØ, Frimodt-Møller N. The importance of understanding the infectious microenvironment. THE LANCET. INFECTIOUS DISEASES 2021; 22:e88-e92. [PMID: 34506737 DOI: 10.1016/s1473-3099(21)00122-5] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 01/25/2021] [Accepted: 02/12/2021] [Indexed: 12/12/2022]
Abstract
Standard doses of antibiotics do not efficiently treat chronic infections of the soft tissue and bone. In this Personal View, we advocate for improving treatment of these infections by taking the infectious microenvironment into account. The infectious microenvironment can cause sensitive bacteria to lose their susceptibility to antibiotics that are effective in standard laboratory susceptibility testing. We propose that bacteria behave substantially different in standard laboratory conditions than they do in actual infections. The infectious microenvironment could impose changes in growth and metabolic activity that result in increased protection against antibiotics. Therefore, we advocate that improved antibiotic treatment of chronic infection is achievable when antibiotics are recommended on the basis of susceptibility testing in relevant in vitro conditions that resemble actual infectious microenvironments. We recommend establishing knowledge of the relevant conditions of the chemical and physical composition of the infectious microenvironment. Recent advances in RNA sequencing, metabolomics, and microscopy have made it possible for the characterisation of the microenvironment of infections and to validate the clinical relevance of in vitro conditions to actual infections.
Collapse
Affiliation(s)
- Thomas Bjarnsholt
- Department of Clinical Microbiology, Copenhagen University Hospital, Copenhagen, Denmark; Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark.
| | - Marvin Whiteley
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA; Center for Microbial Dynamics and Infection, Georgia Institute of Technology, Atlanta, GA, USA; Emory-Children's Cystic Fibrosis Center, Atlanta, GA, USA
| | - Kendra P Rumbaugh
- Department of Surgery, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Philip S Stewart
- Center for Biofilm Engineering, Montana State University, Bozeman, MT, USA
| | - Peter Ø Jensen
- Department of Clinical Microbiology, Copenhagen University Hospital, Copenhagen, Denmark; Costerton Biofilm Center, Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Niels Frimodt-Møller
- Department of Clinical Microbiology, Copenhagen University Hospital, Copenhagen, Denmark
| |
Collapse
|
25
|
Blanco-Cabra N, López-Martínez MJ, Arévalo-Jaimes BV, Martin-Gómez MT, Samitier J, Torrents E. A new BiofilmChip device for testing biofilm formation and antibiotic susceptibility. NPJ Biofilms Microbiomes 2021; 7:62. [PMID: 34344902 PMCID: PMC8333102 DOI: 10.1038/s41522-021-00236-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 07/15/2021] [Indexed: 12/15/2022] Open
Abstract
Currently, three major circumstances threaten the management of bacterial infections: increasing antimicrobial resistance, expansion of chronic biofilm-associated infections, and lack of an appropriate approach to treat them. To date, the development of accelerated drug susceptibility testing of biofilms and of new antibiofouling systems has not been achieved despite the availability of different methodologies. There is a need for easy-to-use methods of testing the antibiotic susceptibility of bacteria that form biofilms and for screening new possible antibiofilm strategies. Herein, we present a microfluidic platform with an integrated interdigitated sensor (BiofilmChip). This new device allows an irreversible and homogeneous attachment of bacterial cells of clinical origin, even directly from clinical specimens, and the biofilms grown can be monitored by confocal microscopy or electrical impedance spectroscopy. The device proved to be suitable to study polymicrobial communities, as well as to measure the effect of antimicrobials on biofilms without introducing disturbances due to manipulation, thus better mimicking real-life clinical situations. Our results demonstrate that BiofilmChip is a straightforward tool for antimicrobial biofilm susceptibility testing that could be easily implemented in routine clinical laboratories.
Collapse
Affiliation(s)
- Núria Blanco-Cabra
- Bacterial Infections and Antimicrobial Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Maria José López-Martínez
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN) Monforte de Lemos 3-5, Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| | - Betsy Verónica Arévalo-Jaimes
- Bacterial Infections and Antimicrobial Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | | | - Josep Samitier
- Nanobioengineering Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Networking Biomedical Research Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN) Monforte de Lemos 3-5, Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| | - Eduard Torrents
- Bacterial Infections and Antimicrobial Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain.
| |
Collapse
|
26
|
Jensen LK. Implant-associated osteomyelitis: Development, characterisation, and application of a porcine model. APMIS 2021; 129 Suppl 141:1-44. [PMID: 34129250 DOI: 10.1111/apm.13125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Louise Kruse Jensen
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
27
|
Redfern J, Wallace J, van Belkum A, Jaillard M, Whittard E, Ragupathy R, Verran J, Kelly P, Enright MC. Biofilm associated genotypes of multiple antibiotic resistant Pseudomonas aeruginosa. BMC Genomics 2021; 22:572. [PMID: 34311706 PMCID: PMC8314537 DOI: 10.1186/s12864-021-07818-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 06/14/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Pseudomonas aeruginosa is a ubiquitous environmental microorganism and also a common cause of infection. Its ability to survive in many different environments and persistently colonize humans is linked to its presence in biofilms formed on indwelling device surfaces. Biofilm promotes adhesion to, and survival on surfaces, protects from desiccation and the actions of antibiotics and disinfectants. RESULTS We examined the genetic basis for biofilm production on polystyrene at room (22 °C) and body temperature (37 °C) within 280 P. aeruginosa. 193 isolates (69 %) produced more biofilm at 22 °C than at 37 °C. Using GWAS and pan-GWAS, we found a number of accessory genes significantly associated with greater biofilm production at 22 °C. Many of these are present on a 165 kb region containing genes for heavy metal resistance (arsenic, copper, mercury and cadmium), transcriptional regulators and methytransferases. We also discovered multiple core genome SNPs in the A-type flagellin gene and Type II secretion system gene xpsD. Analysis of biofilm production of isolates of the MDR ST111 and ST235 lineages on stainless-steel revealed several accessory genes associated with enhanced biofilm production. These include a putative translocase with homology to a Helicobacter pylori type IV secretion system protein, a TA system II toxin gene and the alginate biosynthesis gene algA, several transcriptional regulators and methytransferases as well as core SNPs in genes involved in quorum sensing and protein translocation. CONCLUSIONS Using genetic association approaches we discovered a number of accessory genes and core-genome SNPs that were associated with enhanced early biofilm formation at 22 °C compared to 37 °C. These included a 165 kb genomic island containing multiple heavy metal resistance genes, transcriptional regulators and methyltransferases. We hypothesize that this genomic island may be associated with overall genotypes that are environmentally adapted to survive at lower temperatures. Further work to examine their importance in, for example gene-knockout studies, are required to confirm their relevance. GWAS and pan-GWAS approaches have great potential as a first step in examining the genetic basis of novel bacterial phenotypes.
Collapse
Affiliation(s)
- James Redfern
- Faculty of Science and Engineering, Manchester Metropolitan University, Chester Street, Manchester, United Kingdom
| | - Janine Wallace
- Faculty of Science and Engineering, Manchester Metropolitan University, Chester Street, Manchester, United Kingdom
| | | | | | - Elliot Whittard
- Faculty of Science and Engineering, Manchester Metropolitan University, Chester Street, Manchester, United Kingdom
| | - Roobinidevi Ragupathy
- Faculty of Science and Engineering, Manchester Metropolitan University, Chester Street, Manchester, United Kingdom
| | - Joanna Verran
- Faculty of Science and Engineering, Manchester Metropolitan University, Chester Street, Manchester, United Kingdom
| | - Peter Kelly
- Faculty of Science and Engineering, Manchester Metropolitan University, Chester Street, Manchester, United Kingdom
| | - Mark Charles Enright
- Faculty of Science and Engineering, Manchester Metropolitan University, Chester Street, Manchester, United Kingdom.
| |
Collapse
|
28
|
Díez-Aguilar M, Ekkelenkamp M, Morosini MI, Huertas N, Del Campo R, Zamora J, Fluit AC, Tunney MM, Obrecht D, Bernardini F, Cantón R. Anti-biofilm activity of murepavadin against cystic fibrosis Pseudomonas aeruginosa isolates. J Antimicrob Chemother 2021; 76:2578-2585. [PMID: 34283223 DOI: 10.1093/jac/dkab222] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/04/2021] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES To determine the activity of murepavadin in comparison with tobramycin, colistin and aztreonam, against cystic fibrosis (CF) Pseudomonas aeruginosa isolates growing in biofilms. The biofilm-epidemiological cut-off (ECOFF) values that include intrinsic resistance mechanisms present in biofilms were estimated. METHODS Fifty-three CF P. aeruginosa isolates from respiratory samples were tested using the Calgary (closed system) device, while 4 [2 clinical (one smooth, one mucoid) and 2 reference strains] were tested using the BioFlux, a microfluidic open model of biofilm testing. Biofilm was stained with SYTO9® and propidium iodide. The minimal biofilm inhibitory concentration (MBIC) and the minimal biofilm eradication concentration (MBEC) were determined. The MBIC-ECOFF and the MBEC-ECOFF were calculated. RESULTS Colistin, tobramycin and murepavadin presented similar MBIC50/MBIC90 values (4/32, 8/64 and 2/32, respectively). Murepavadin exhibited the lowest MBEC90 (64 mg/L). Aztreonam MBIC and MBEC values were higher than those of the other antibiotics tested. Tobramycin and murepavadin had the lowest MBEC-ECOFF (64 and 128 mg/L, respectively), while those of aztreonam and colistin exceeded 512 mg/L. Using the BioFlux, for the PAO1, PAO mutS and the smooth clinical strain, a significant difference (P < 0.0125) was observed when comparing the fluorescence of treated and untreated biofilms. For the mucoid strain, only the biofilm treated with aztreonam (MBIC and MBEC) and tobramycin (MBEC) showed differences with respect to the untreated biofilm. CONCLUSIONS Murepavadin demonstrated good activity against P. aeruginosa biofilms both in open and closed systems. The MBIC-ECOFF and the MBEC-ECOFF are proposed as new parameters to estimate the activity of antibiotics on biofilms.
Collapse
Affiliation(s)
- María Díez-Aguilar
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Red Española de Investigación en Patología Infecciosa (REIPI), Madrid, Spain
| | - Miquel Ekkelenkamp
- Unidad de Bioestadística Clínica, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - María-Isabel Morosini
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Red Española de Investigación en Patología Infecciosa (REIPI), Madrid, Spain
| | - Natalia Huertas
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Rosa Del Campo
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Red Española de Investigación en Patología Infecciosa (REIPI), Madrid, Spain
| | - Javier Zamora
- Unidad de Bioestadística Clínica, Hospital Universitario Ramón y Cajal, IRYCIS, Madrid, Spain
| | - Ad C Fluit
- University Medical Center Utrecht, Department of Medical Microbiology, Utrecht, The Netherlands
| | - Michael M Tunney
- Queen's University Belfast, School of Pharmacy, Belfast, United Kingdom
| | | | | | - Rafael Cantón
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Red Española de Investigación en Patología Infecciosa (REIPI), Madrid, Spain
| |
Collapse
|
29
|
Prevalence and Impact of Biofilms on Bloodstream and Urinary Tract Infections: A Systematic Review and Meta-Analysis. Antibiotics (Basel) 2021; 10:antibiotics10070825. [PMID: 34356749 PMCID: PMC8300799 DOI: 10.3390/antibiotics10070825] [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: 06/08/2021] [Revised: 06/29/2021] [Accepted: 07/02/2021] [Indexed: 02/07/2023] Open
Abstract
This study sought to assess the prevalence and impact of biofilms on two commonly biofilm-related infections, bloodstream and urinary tract infections (BSI and UTI). Separated systematic reviews and meta-analyses of observational studies were carried out in PubMed and Web of Sciences databases from January 2005 to May 2020, following PRISMA protocols. Studies were selected according to specific and defined inclusion/exclusion criteria. The obtained outcomes were grouped into biofilm production (BFP) prevalence, BFP in resistant vs. susceptible strains, persistent vs. non-persistent BSI, survivor vs. non-survivor patients with BSI, and catheter-associated UTI (CAUTI) vs. non-CAUTI. Single-arm and two-arm analyses were conducted for data analysis. In vitro BFP in BSI was highly related to resistant strains (odds ratio-OR: 2.68; 95% confidence intervals-CI: 1.60–4.47; p < 0.01), especially for methicillin-resistant Staphylococci. BFP was also highly linked to BSI persistence (OR: 2.65; 95% CI: 1.28–5.48; p < 0.01) and even to mortality (OR: 2.05; 95% CI: 1.53–2.74; p < 0.01). Candida spp. was the microorganism group where the highest associations were observed. Biofilms seem to impact Candida BSI independently from clinical differences, including treatment interventions. Regarding UTI, multi-drug resistant and extended-spectrum β-lactamase-producing strains of Escherichia coli, were linked to a great BFP prevalence (OR: 2.92; 95% CI: 1.30–6.54; p < 0.01 and OR: 2.80; 95% CI: 1.33–5.86; p < 0.01). More in vitro BFP was shown in CAUTI compared to non-CAUTI, but with less statistical confidence (OR: 2.61; 95% CI: 0.67–10.17; p < 0.17). This study highlights that biofilms must be recognized as a BSI and UTI resistance factor as well as a BSI virulence factor.
Collapse
|
30
|
Interlaboratory study for the evaluation of three microtiter plate-based biofilm quantification methods. Sci Rep 2021; 11:13779. [PMID: 34215805 PMCID: PMC8253847 DOI: 10.1038/s41598-021-93115-w] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 06/03/2021] [Indexed: 11/08/2022] Open
Abstract
Microtiter plate methods are commonly used for biofilm assessment. However, results obtained with these methods have often been difficult to reproduce. Hence, it is important to obtain a better understanding of the repeatability and reproducibility of these methods. An interlaboratory study was performed in five different laboratories to evaluate the reproducibility and responsiveness of three methods to quantify Staphylococcus aureus biofilm formation in 96-well microtiter plates: crystal violet, resazurin, and plate counts. An inter-lab protocol was developed for the study. The protocol was separated into three steps: biofilm growth, biofilm challenge, biofilm assessment. For control experiments participants performed the growth and assessment steps only. For treatment experiments, all three steps were performed and the efficacy of sodium hypochlorite (NaOCl) in killing S. aureus biofilms was evaluated. In control experiments, on the log10-scale, the reproducibility SD (SR) was 0.44 for crystal violet, 0.53 for resazurin, and 0.92 for the plate counts. In the treatment experiments, plate counts had the best responsiveness to different levels of efficacy and also the best reproducibility with respect to responsiveness (Slope/SR = 1.02), making it the more reliable method to use in an antimicrobial efficacy test. This study showed that the microtiter plate is a versatile and easy-to-use biofilm reactor, which exhibits good repeatability and reproducibility for different types of assessment methods, as long as a suitable experimental design and statistical analysis is applied.
Collapse
|
31
|
Guzmán-Soto I, McTiernan C, Gonzalez-Gomez M, Ross A, Gupta K, Suuronen EJ, Mah TF, Griffith M, Alarcon EI. Mimicking biofilm formation and development: Recent progress in in vitro and in vivo biofilm models. iScience 2021; 24:102443. [PMID: 34013169 PMCID: PMC8113887 DOI: 10.1016/j.isci.2021.102443] [Citation(s) in RCA: 109] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Biofilm formation in living organisms is associated to tissue and implant infections, and it has also been linked to the contribution of antibiotic resistance. Thus, understanding biofilm development and being able to mimic such processes is vital for the successful development of antibiofilm treatments and therapies. Several decades of research have contributed to building the foundation for developing in vitro and in vivo biofilm models. However, no such thing as an "all fit" in vitro or in vivo biofilm models is currently available. In this review, in addition to presenting an updated overview of biofilm formation, we critically revise recent approaches for the improvement of in vitro and in vivo biofilm models.
Collapse
Affiliation(s)
- Irene Guzmán-Soto
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
| | - Christopher McTiernan
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
| | - Mayte Gonzalez-Gomez
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
| | - Alex Ross
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, K1H8M5, Canada
| | - Keshav Gupta
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
| | - Erik J. Suuronen
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
| | - Thien-Fah Mah
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, K1H8M5, Canada
| | - May Griffith
- Centre de Recherche Hôpital Maisonneuve-Rosemont, Montréal, QC, H1T 2M4, Canada
- Département d'ophtalmologie, Université de Montréal, Montréal, QC, H3T1J4, Canada
| | - Emilio I. Alarcon
- Division of Cardiac Surgery, University of Ottawa Heart Institute, Ottawa, ON, K1Y4W7, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, K1H8M5, Canada
| |
Collapse
|
32
|
Silva NBS, Marques LA, Röder DDB. Diagnosis of biofilm infections: current methods used, challenges and perspectives for the future. J Appl Microbiol 2021; 131:2148-2160. [PMID: 33629487 DOI: 10.1111/jam.15049] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 02/01/2021] [Accepted: 02/23/2021] [Indexed: 12/16/2022]
Abstract
The diagnosis of biofilms continues to be a challenge, and there is no standardized protocol for such a diagnosis in clinical practice. In addition, some proposed methodologies are expensive to require significant amounts of time and a high number of trained staff, making them impracticable for clinical practice. In recent years, mass spectrophotometry/matrix-assisted laser desorption ionization time of flight (MALDI-TOF) has been applied it in biofilm studies. However, due to several problems and limitations of the technique, MALDI-TOF is far from being the gold standard for identifying biofilm formation. The omics analysis may prove to be a promising strategy for the diagnosis of biofilms in clinical laboratories since it allows the identification of pathogens in less time than needed for conventional techniques and in a more specific manner. However, omic tools are expensive and require qualified technical expertise, and an analysis of the data obtained needs to be careful not to neglect subpopulations in the biofilm. More studies must therefore be developed for creating a protocol that guarantees rapid biofilm identification, ensuring greater chances of success in infection control. This review discusses the current methods of microbial biofilm detection and future perspectives for its diagnosis in clinical practice.
Collapse
Affiliation(s)
- N B S Silva
- Applied Immunology and Parasitology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - L A Marques
- Health Sciences, Medical School, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| | - D D B Röder
- Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Minas Gerais, Brazil
| |
Collapse
|
33
|
Muñoz-Gallego I, Viedma E, Esteban J, Mancheño-Losa M, García-Cañete J, Blanco-García A, Rico A, García-Perea A, Ruiz Garbajosa P, Escudero-Sánchez R, Sánchez Somolinos M, Marín Arriaza M, Romanyk J, Barbero JM, Arribi Vilela A, González Romo F, Pérez-Jorge C, M. Arana D, Monereo A, Domingo D, Cordero J, Sánchez Romero MI, García Viejo MÁ, Lora-Tamayo J, Chaves F. Genotypic and Phenotypic Characteristics of Staphylococcus aureus Prosthetic Joint Infections: Insight on the Pathogenesis and Prognosis of a Multicenter Prospective Cohort. Open Forum Infect Dis 2020; 7:ofaa344. [PMID: 33005695 PMCID: PMC7519778 DOI: 10.1093/ofid/ofaa344] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Accepted: 08/08/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Staphylococcus aureus is the leading cause of prosthetic joint infection (PJI). Beyond the antibiogram, little attention has been paid to the influence of deep microbiological characteristics on patient prognosis. Our aim was to investigate whether microbiological genotypic and phenotypic features have a significant influence on infection pathogenesis and patient outcome. METHODS A prospective multicenter study was performed, including all S. aureus PJIs (2016-2017). Clinical data and phenotypic (agr functionality, β-hemolysis, biofilm formation) and genotypic characteristics of the strains were collected. Biofilm susceptibility to antimicrobials was investigated (minimal biofilm eradication concentration [MBEC] assay). RESULTS Eighty-eight patients (39.8% men, age 74.7 ± 14.1 years) were included. Forty-five had early postoperative infections (EPIs), 21 had chronic infections (CPIs), and 19 had hematogenous infections (HIs). Twenty (22.7%) were caused by methicillin-resistant S. aureus. High genotypic diversity was observed, including 16 clonal complexes (CCs), with CC5 being the most frequent (30.7%). agr activity was greater in EPI than CPI (55.6% vs 28.6%; P = .041). Strains causing EPI were phenotypically and genotypically similar, regardless of symptom duration. Treatment failure (36.5%) occurred less frequently among cases treated with implant removal. In cases treated with debridement and implant retention, there were fewer failures among those who received combination therapy with rifampin. No genotypic or phenotypic characteristics predicted failure, except vancomycin minimal inhibitory concentration ≥1.5 mg/L (23.1% failure vs 3.4%; P = .044). MBEC50 was >128 mg/L for all antibiotics tested and showed no association with prognosis. CONCLUSIONS S. aureus with different genotypic backgrounds is capable of causing PJI, showing slight differences in clinical presentation and pathogenesis. No major microbiological characteristics were observed to influence the outcome, including MBEC.
Collapse
Affiliation(s)
- Irene Muñoz-Gallego
- Servicio de Microbiología, Hospital Universitario 12 de Octubre, Universidad Complutense, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Esther Viedma
- Servicio de Microbiología, Hospital Universitario 12 de Octubre, Universidad Complutense, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Red Española de Investigación en Patología Infecciosa (REIPI), Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Sevilla, Spain
| | - Jaime Esteban
- Servicio de Microbiología, IIS-Fundación Jiménez Díaz, Madrid, Spain
| | - Mikel Mancheño-Losa
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Servicio de Medicina Interna, Hospital Universitario 12 de Octubre, Universidad Complutense, Madrid, Spain
| | - Joaquín García-Cañete
- Servicio de Medicina Interna-Urgencias, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - Antonio Blanco-García
- Servicio de Medicina Interna-Urgencias, Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - Alicia Rico
- Servicio de Medicina Interna, Hospital Universitario La Paz, Madrid, Spain
| | | | - Patricia Ruiz Garbajosa
- Red Española de Investigación en Patología Infecciosa (REIPI), Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Sevilla, Spain
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Rosa Escudero-Sánchez
- Red Española de Investigación en Patología Infecciosa (REIPI), Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Sevilla, Spain
- Servicio de Enfermedades Infecciosas, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Mar Sánchez Somolinos
- Servicio de Microbiología y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Mercedes Marín Arriaza
- Servicio de Microbiología y Enfermedades Infecciosas, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- CIBER de Enfermedades Respiratorias (CIBERES CB06/06/0058), Madrid, Spain
| | - Juan Romanyk
- Servicio de Microbiología, Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid, Spain
| | - José María Barbero
- Servicio de Medicina Interna, Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Ana Arribi Vilela
- Servicio de Microbiología, Hospital Clínico San Carlos, Madrid, Spain
| | | | - Conchita Pérez-Jorge
- Servicio de Microbiología, Hospital Universitario Rey Juan Carlos, Móstoles, Madrid, Spain
| | - David M. Arana
- Servicio de Microbiología, Hospital Universitario de Getafe, Getafe, Madrid, Spain
| | - Alfonso Monereo
- Servicio de Medicina Interna, Hospital Universitario de Getafe, Getafe, Madrid, Spain
| | - Diego Domingo
- Servicio de Microbiología, Hospital Universitario de La Princesa, Madrid, Spain
| | - José Cordero
- Servicio de Traumatología, Hospital Universitario de La Princesa, Madrid, Spain
| | | | | | - Jaime Lora-Tamayo
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Red Española de Investigación en Patología Infecciosa (REIPI), Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Sevilla, Spain
- Servicio de Medicina Interna, Hospital Universitario 12 de Octubre, Universidad Complutense, Madrid, Spain
| | - Fernando Chaves
- Servicio de Microbiología, Hospital Universitario 12 de Octubre, Universidad Complutense, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
- Red Española de Investigación en Patología Infecciosa (REIPI), Instituto de Salud Carlos III, Ministerio de Ciencia e Innovación, Sevilla, Spain
| |
Collapse
|
34
|
Stoffel JJ, Kohler Riedi PL, Hadj Romdhane B. A multimodel regime for evaluating effectiveness of antimicrobial wound care products in microbial biofilms. Wound Repair Regen 2020; 28:438-447. [PMID: 32175636 PMCID: PMC7540695 DOI: 10.1111/wrr.12806] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 02/20/2020] [Accepted: 03/10/2020] [Indexed: 01/04/2023]
Abstract
Microbial biofilms have become increasingly recognized as a cause of wound chronicity. There are several topical antimicrobial wound care products available for use; however, their effectiveness has routinely been demonstrated with planktonic microorganisms. There is no target reference value for antimicrobial effectiveness of wound care products in biofilm models. In addition, data on antimicrobial activity of products in biofilm models are scattered across many test methods in a variety of studies. The aim of this work is to directly compare commercial products containing the commonly used topical antimicrobial agents iodine, silver, polyhexamethylene biguanide, octenidine, hypochlorous acid, benzalkonium chloride, and a surfactant-based topical containing poloxamer 188. Five different in vitro biofilm models of varied complexity were used, incorporating several bacterial pathogens such as Staphylococcus, Enterococcus, Streptococcus, Pseudomonas, Acinetobacter, Klebsiella, and Enterobacter. The fungal pathogens Candida albicans and Candida auris were also evaluated. A multispecies bacterial biofilm model was also used to evaluate the products. Additionally, C. albicans was used in combination with S. aureus and P. aeruginosa in a multikingdom version of the polymicrobial biofilm model. Statistically significant differences in antimicrobial performance were observed between treatments in each model and changing microbial growth conditions or combinations of organisms resulted in significant performance differences for some treatments. The iodine and benzalkonium chloride-containing products were overall the most effective in vitro and were then selected for in vivo evaluation in an infected immunocompromised murine model. Unexpectedly, the iodine product was statistically (P > .05) no different than the untreated control, while the benzalkonium chloride containing product significantly (P < .05) reduced the biofilm compared to untreated control. This body of work demonstrates the importance of not only evaluating antimicrobial wound care products in biofilm models but also the importance of using several different models to gain a comprehensive understanding of products' effectiveness.
Collapse
|
35
|
Francolini I, Hall-Stoodley L, Stoodley P. Biofilms, Biomaterials, and Device-Related Infections. Biomater Sci 2020. [DOI: 10.1016/b978-0-12-816137-1.00054-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
36
|
Coenye T, Kjellerup B, Stoodley P, Bjarnsholt T. The future of biofilm research - Report on the '2019 Biofilm Bash'. Biofilm 2019; 2:100012. [PMID: 33447799 PMCID: PMC7798458 DOI: 10.1016/j.bioflm.2019.100012] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 11/25/2019] [Accepted: 11/25/2019] [Indexed: 12/22/2022] Open
Abstract
In May 2019, 29 scientists with expertise in various subdisciplines of biofilm research got together in Leavenworth (WA, USA) at an event designated as the ‘2019 Biofilm Bash’. The goal of this informal two-day meeting was first to identify gaps in our knowledge, and then to come up with ways how the biofilm community can fill these gaps. The meeting was organized around six questions that covered the most important items brought forward by the organizers and participants. The outcome of these discussions is summarized in the present paper. We are aware that these views represent a small subset of our field, and that inevitably we will have inadvertently overlooked important developing research areas and ideas. We are nevertheless hopeful that this report will stimulate discussions and help create new ways of how we can advance our field.
Collapse
Affiliation(s)
- Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium.,ESCMID Study Group on Biofilms, Basel, Switzerland
| | - Birthe Kjellerup
- Department of Civil and Environmental Engineering, University of Maryland, College Park, MD, USA.,Fischell Department of Bioengineering, University of Maryland, College Park, MD, USA
| | - Paul Stoodley
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH, USA.,Department of Orthopaedics, The Ohio State University, Columbus, OH, USA.,National Biofilms Innovation Centre (NBIC), UK.,National Centre for Advanced Tribology at Southampton, University of Southampton, Southampton, UK
| | - Thomas Bjarnsholt
- ESCMID Study Group on Biofilms, Basel, Switzerland.,Costerton Biofilm Center, University of Copenhagen, Copenhagen, Denmark.,Department of Microbiology, Copenhagen University Hospital, Copenhagen, Denmark
| | | |
Collapse
|
37
|
Jensen P, Møller S, Lerche C, Moser C, Bjarnsholt T, Ciofu O, Faurholt-Jepsen D, Høiby N, Kolpen M. Improving antibiotic treatment of bacterial biofilm by hyperbaric oxygen therapy: Not just hot air. Biofilm 2019; 1:100008. [PMID: 33447795 PMCID: PMC7798444 DOI: 10.1016/j.bioflm.2019.100008] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 10/22/2019] [Accepted: 10/23/2019] [Indexed: 10/26/2022] Open
Abstract
Bacteria and fungi show substantial increased recalcitrance when growing as infectious biofilms. Chronic infections caused by biofilm growing microorganisms is considered a major problem of modern medicine. New strategies are needed to improve antibiotic treatment of biofilms. We have improved antibiotic treatment of bacterial biofilms by reviving the dormant bacteria and thereby make them susceptible to antibiotics by means of reoxygenation. Here we review the rationale for associating lack of oxygen with low susceptibility in infectious biofilm, and how hyperbaric oxygen therapy may result in reoxygenation leading to enhanced bactericidal activity of antibiotics. We address issues of feasibility and potential adverse effects regarding patient safety and development of resistance. Finally, we propose means for supplying reoxygenation to antibiotic treatment of infectious biofilm with the potential to benefit large groups of patients.
Collapse
Affiliation(s)
- P.Ø. Jensen
- Department of Clinical Microbiology, Rigshospitalet, 2100, Copenhagen, Denmark
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health Sciences University of Copenhagen, 2200, Copenhagen, Denmark
| | - S.A. Møller
- Department of Clinical Microbiology, Rigshospitalet, 2100, Copenhagen, Denmark
| | - C.J. Lerche
- Department of Clinical Microbiology, Rigshospitalet, 2100, Copenhagen, Denmark
| | - C. Moser
- Department of Clinical Microbiology, Rigshospitalet, 2100, Copenhagen, Denmark
| | - T. Bjarnsholt
- Department of Clinical Microbiology, Rigshospitalet, 2100, Copenhagen, Denmark
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health Sciences University of Copenhagen, 2200, Copenhagen, Denmark
| | - O. Ciofu
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health Sciences University of Copenhagen, 2200, Copenhagen, Denmark
| | - D. Faurholt-Jepsen
- Copenhagen Cystic Fibrosis Center, Department of Infectious Diseases, Rigshospitalet, Copenhagen University Hospital, Blegdamsvej 9, DK2100, Copenhagen, Denmark
| | - N. Høiby
- Department of Clinical Microbiology, Rigshospitalet, 2100, Copenhagen, Denmark
- Costerton Biofilm Center, Department of Immunology and Microbiology, Faculty of Health Sciences University of Copenhagen, 2200, Copenhagen, Denmark
| | - M. Kolpen
- Department of Clinical Microbiology, Rigshospitalet, 2100, Copenhagen, Denmark
| |
Collapse
|
38
|
Thieme L, Hartung A, Tramm K, Klinger-Strobel M, Jandt KD, Makarewicz O, Pletz MW. MBEC Versus MBIC: the Lack of Differentiation between Biofilm Reducing and Inhibitory Effects as a Current Problem in Biofilm Methodology. Biol Proced Online 2019; 21:18. [PMID: 31528123 PMCID: PMC6743098 DOI: 10.1186/s12575-019-0106-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 08/08/2019] [Indexed: 12/11/2022] Open
Abstract
Background Biofilms are communities of aggregated, matrix-embedded microbial cells showing a high tolerance to an in principle adequate antibiotic therapy, often resulting in treatment failure. A major challenge in the management of biofilm-associated infections is the development of adequate, standardized biofilm susceptibility testing assays that are clinically meaningful, i.e. that their results correlate with treatment outcome. Different biofilm susceptibility endpoint parameters like the minimal biofilm eradication concentration (MBEC) or the minimal biofilm inhibitory concentration (MBIC) have been suggested as a guide for treatment of biofilm-associated infections, however with inconsistent perception and use among biofilm researchers, leading to confusion and contradictions among different anti-biofilm component studies and clinical trials. Findings Evaluation of anti-biofilm effects is mostly based on the untreated reference growth control biofilm measured at the same endpoint as the treated biofilm, neglecting the possible change of the untreated reference biofilm from the time point of pre-antimicrobial exposure to the measured endpoint. In this commentary, we point out the importance of individual quantification of mature, established biofilms before antimicrobial treatment for each biofilm model in order to draw conclusions on the measured biofilm effect size, i.e. biofilm reducing (MBEC) or biofilm inhibitory (MBIC) effects. Conclusion The assessment of pre-treatment biofilms contributes to a standardized use of biofilm susceptibility endpoint parameters, which is urgently needed to improve the clinical validity of future anti-biofilm assays.
Collapse
Affiliation(s)
- Lara Thieme
- 1Institute for Infectious Diseases and Infection Control, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Anita Hartung
- 1Institute for Infectious Diseases and Infection Control, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Kristina Tramm
- 1Institute for Infectious Diseases and Infection Control, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Mareike Klinger-Strobel
- 1Institute for Infectious Diseases and Infection Control, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Klaus D Jandt
- 2Otto Schott Institute of Materials Research, Friedrich Schiller University Jena, Löbdergraben 32, Jena, 07743 Germany
| | - Oliwia Makarewicz
- 1Institute for Infectious Diseases and Infection Control, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Mathias W Pletz
- 1Institute for Infectious Diseases and Infection Control, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| |
Collapse
|
39
|
Brown JL, Johnston W, Delaney C, Short B, Butcher MC, Young T, Butcher J, Riggio M, Culshaw S, Ramage G. Polymicrobial oral biofilm models: simplifying the complex. J Med Microbiol 2019; 68:1573-1584. [PMID: 31524581 DOI: 10.1099/jmm.0.001063] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Over the past century, numerous studies have used oral biofilm models to investigate growth kinetics, biofilm formation, structure and composition, antimicrobial susceptibility and host-pathogen interactions. In vivo animal models provide useful models of some oral diseases; however, these are expensive and carry vast ethical implications. Oral biofilms grown or maintained in vitro offer a useful platform for certain studies and have the advantages of being inexpensive to establish and easy to reproduce and manipulate. In addition, a wide range of variables can be monitored and adjusted to mimic the dynamic environmental changes at different sites in the oral cavity, such as pH, temperature, salivary and gingival crevicular fluid flow rates, or microbial composition. This review provides a detailed insight for early-career oral science researchers into how the biofilm models used in oral research have progressed and improved over the years, their advantages and disadvantages, and how such systems have contributed to our current understanding of oral disease pathogenesis and aetiology.
Collapse
Affiliation(s)
- Jason L Brown
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK.,Institute of Biomedical and Environmental Health Research, School of Science and Sport, University of the West of Scotland, Paisley, PA1 2BE, UK
| | - William Johnston
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Christopher Delaney
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Bryn Short
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Mark C Butcher
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Tracy Young
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - John Butcher
- Department of Life Sciences, School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, G4 0BA, UK.,Institute of Biomedical and Environmental Health Research, School of Science and Sport, University of the West of Scotland, Paisley, PA1 2BE, UK
| | - Marcello Riggio
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Shauna Culshaw
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| | - Gordon Ramage
- Oral Sciences Research Group, Glasgow Dental School, School of Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
| |
Collapse
|
40
|
Cattò C, Cappitelli F. Testing Anti-Biofilm Polymeric Surfaces: Where to Start? Int J Mol Sci 2019; 20:E3794. [PMID: 31382580 PMCID: PMC6696330 DOI: 10.3390/ijms20153794] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 08/02/2019] [Indexed: 12/11/2022] Open
Abstract
Present day awareness of biofilm colonization on polymeric surfaces has prompted the scientific community to develop an ever-increasing number of new materials with anti-biofilm features. However, compared to the large amount of work put into discovering potent biofilm inhibitors, only a small number of papers deal with their validation, a critical step in the translation of research into practical applications. This is due to the lack of standardized testing methods and/or of well-controlled in vivo studies that show biofilm prevention on polymeric surfaces; furthermore, there has been little correlation with the reduced incidence of material deterioration. Here an overview of the most common methods for studying biofilms and for testing the anti-biofilm properties of new surfaces is provided.
Collapse
Affiliation(s)
- Cristina Cattò
- Department of Food Environmental and Nutritional Sciences, Università degli Studi di Milano, via Celoria 2, 20133 Milano, Italy
| | - Francesca Cappitelli
- Department of Food Environmental and Nutritional Sciences, Università degli Studi di Milano, via Celoria 2, 20133 Milano, Italy.
| |
Collapse
|
41
|
Kreth J, Ferracane J, Pfeifer C, Khajotia S, Merritt J. At the Interface of Materials and Microbiology: A Call for the Development of Standardized Approaches to Assay Biomaterial-Biofilm Interactions. J Dent Res 2019; 98:850-852. [PMID: 31173560 PMCID: PMC6616120 DOI: 10.1177/0022034519854685] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Affiliation(s)
- J. Kreth
- Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, OR, USA
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| | - J.L. Ferracane
- Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, OR, USA
| | - C.S. Pfeifer
- Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, OR, USA
| | - S. Khajotia
- Department of Restorative Sciences, College of Dentistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - J. Merritt
- Department of Restorative Dentistry, School of Dentistry, Oregon Health and Science University, Portland, OR, USA
- Department of Molecular Microbiology and Immunology, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
42
|
Activity of Antibiotics against Staphylococcus aureus in an In Vitro Model of Biofilms in the Context of Cystic Fibrosis: Influence of the Culture Medium. Antimicrob Agents Chemother 2019; 63:AAC.00602-19. [PMID: 31036685 DOI: 10.1128/aac.00602-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 04/20/2019] [Indexed: 01/27/2023] Open
Abstract
Staphylococcus aureus is a highly prevalent pathogen in the respiratory tract of young patients with cystic fibrosis (CF) and causes biofilm-related infections. Here, we set up an in vitro model of a biofilm grown in Trypticase soy broth supplemented with glucose and NaCl (TGN) or in artificial sputum medium (ASM) and used it to evaluate on a pharmacodynamic basis the activity of antibiotics used in CF patients and active on staphylococci (meropenem, vancomycin, azithromycin, linezolid, rifampin, ciprofloxacin, tobramycin). Rheological studies showed that ASM was more elastic than viscous, as was also observed for sputa from CF patients, with elastic and viscous moduli being, respectively, similar to and slightly lower than those of CF sputa. Biofilms formed by methicillin-sensitive S. aureus strain ATCC 25923 and methicillin-resistant S. aureus strain ATCC 33591 reached maturity after 24 h, with biomass (measured by crystal violet staining) and metabolic activity (assessed by following resazurin metabolization) being lower in ASM than in TGN and viability (assessed by bacterial counts) being similar in both media. Full concentration-response curves of antibiotics obtained after 24 h of incubation of biofilms showed that all antibiotics were drastically less potent and less efficient in ASM than in TGN toward viability, metabolic activity, and biomass. Tobramycin selected for small-colony variants, specifically in biofilms grown in ASM; the auxotrophism of these variants could not be established. These data highlight the major influence exerted by the culture medium on S. aureus responsiveness to antibiotics in biofilms. The use of ASM may help to determine effective drug concentrations or to evaluate new therapeutic options against biofilms in CF patients.
Collapse
|
43
|
Stewart PS, Parker AE. Measuring Antimicrobial Efficacy against Biofilms: a Meta-analysis. Antimicrob Agents Chemother 2019; 63:e00020-19. [PMID: 30803974 PMCID: PMC6496104 DOI: 10.1128/aac.00020-19] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 02/21/2019] [Indexed: 12/12/2022] Open
Abstract
Through a statistical meta-analysis of published data on antimicrobial efficacy against biofilms formed by two common bacterial species, it was concluded that the particular experimental method used is the most important factor determining the outcome of the test. An expected dose-response relationship (greater killing with higher doses or longer treatment times) was observed for data sets derived from a single method but was not observed when data from multiple studies using diverse methods were pooled. Method-specific properties such as the surface area/volume ratio, areal biofilm cell density, and microbial species were shown to influence quantitative measurements of biofilm killing. A better appreciation of the method characteristics that affect antibiofilm efficacy tests could aid decision-making related to investment in research and development and regulatory approvals for biofilm control strategies. The following recommendations are offered to those working in research and development related to biofilm control: (i) report the log reduction, surface area/volume ratio, and biofilm areal cell density; (ii) include data for a benchmark agent, making sure that this agent performs competitively at the dose tested; (iii) measure the dose-response relationship, i.e., make measurements at multiple treatment concentrations or dose durations; and (iv) use a standardized method in addition to research methods.
Collapse
Affiliation(s)
- Philip S Stewart
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, USA
- Chemical and Biological Engineering, Montana State University, Bozeman, Montana, USA
| | - Albert E Parker
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, USA
- Mathematical Sciences, Montana State University, Bozeman, Montana, USA
| |
Collapse
|
44
|
Roberts AEL, Powell LC, Pritchard MF, Thomas DW, Jenkins RE. Anti-pseudomonad Activity of Manuka Honey and Antibiotics in a Specialized ex vivo Model Simulating Cystic Fibrosis Lung Infection. Front Microbiol 2019; 10:869. [PMID: 31105667 PMCID: PMC6491927 DOI: 10.3389/fmicb.2019.00869] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 04/04/2019] [Indexed: 01/22/2023] Open
Abstract
Pseudomonas aeruginosa causes problematic chronic lung infections in those suffering from cystic fibrosis. This is due to its antimicrobial resistance mechanisms and its ability to form robust biofilm communities with increased antimicrobial tolerances. Using novel antimicrobials or repurposing current ones is required in order to overcome these problems. Manuka honey is a natural antimicrobial agent that has been used for many decades in the treatment of chronic surface wounds with great success, particularly those infected with P. aeruginosa. Here we aim to determine whether the antimicrobial activity of manuka honey could potentially be repurposed to inhibit pulmonary P. aeruginosa infections using two ex vivo models. P. aeruginosa isolates (n = 28) from an international panel were tested for their susceptibility to manuka honey and clinically relevant antibiotics (ciprofloxacin, ceftazidime, and tobramycin), alone and in combination, using conventional antimicrobial susceptibility testing (AST). To increase clinical applicability, two ex vivo porcine lung (EVPL) models (using alveolar and bronchiolar tissue) were used to determine the anti-biofilm effects of manuka honey alone and in combination with antibiotics. All P. aeruginosa isolates were susceptible to manuka honey, however, varying incidences of resistance were seen against antibiotics. The combination of sub-inhibitory manuka honey and antibiotics using conventional AST had no effect on activity against the majority of isolates tested. Using the two ex vivo models, 64% (w/v) manuka honey inhibited many of the isolates where abnormally high concentrations of antibiotics could not. Typically, combinations of both manuka honey and antibiotics had increased antimicrobial activity. These results highlight the potential of manuka honey as a future antimicrobial for the treatment of pulmonary P. aeruginosa isolates, clearing potential infection reservoirs within the upper airway.
Collapse
Affiliation(s)
- Aled E L Roberts
- Department of Biomedical Sciences, Cardiff School of Health Sciences, Cardiff Metropolitan University, Cardiff, United Kingdom
- Swansea University Medical School, Swansea University, Swansea, United Kingdom
| | - Lydia C Powell
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | - Manon F Pritchard
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | - David W Thomas
- School of Dentistry, College of Biomedical and Life Sciences, Cardiff University, Cardiff, United Kingdom
| | - Rowena E Jenkins
- Department of Biomedical Sciences, Cardiff School of Health Sciences, Cardiff Metropolitan University, Cardiff, United Kingdom
- Swansea University Medical School, Swansea University, Swansea, United Kingdom
| |
Collapse
|
45
|
Sendi P, Wustmann K, Büchi AE, Noti F, Klaeser B, Sonderegger B, Auf der Maur C, Mercier T, Schwerzmann M, Ruppen C. Cardiac Implantable Electronic Device-Related Infection Due to Granulicatella adiacens. Open Forum Infect Dis 2019; 6:ofz130. [PMID: 31008142 PMCID: PMC6467393 DOI: 10.1093/ofid/ofz130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 03/12/2019] [Indexed: 12/21/2022] Open
Abstract
Cardiac implantable electronic device–related infection is clinically challenging. Curative treatment commonly includes system removal. A case caused by Granulicatella adiacens occurred in a 32-year-old woman. Clinical course, literature review, and biofilm investigations enabled successful antibiotic management without system removal.
Collapse
Affiliation(s)
- Parham Sendi
- Institute for Infectious Diseases, University of Bern.,Department of Infectious Diseases, University of Bern
| | - Kerstin Wustmann
- Center for Congenital Heart Disease, Inselspital, University Hospital Bern, University of Bern.,Department of Cardiology, Inselspital, University Hospital Bern, University of Bern
| | - Annina E Büchi
- Department of General Internal Medicine, Inselspital, University Hospital Bern, University of Bern
| | - Fabian Noti
- Department of Cardiology, Inselspital, University Hospital Bern, University of Bern
| | - Bernd Klaeser
- Department of Nuclear Medicine, Inselspital, University Hospital Bern, University of Bern
| | - Beat Sonderegger
- Department of Infectious Diseases and Hospital Epidemiology, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Christoph Auf der Maur
- Paediatric and Adult Cardiology, Kantonsspital Lucerne, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Thomas Mercier
- Service and Laboratory of Clinical Pharmacology, Department of Laboratories, Lausanne University Hospital and University of Lausanne, Switzerland
| | - Markus Schwerzmann
- Center for Congenital Heart Disease, Inselspital, University Hospital Bern, University of Bern.,Department of Cardiology, Inselspital, University Hospital Bern, University of Bern
| | | |
Collapse
|
46
|
Grassi L, Batoni G, Ostyn L, Rigole P, Van den Bossche S, Rinaldi AC, Maisetta G, Esin S, Coenye T, Crabbé A. The Antimicrobial Peptide lin-SB056-1 and Its Dendrimeric Derivative Prevent Pseudomonas aeruginosa Biofilm Formation in Physiologically Relevant Models of Chronic Infections. Front Microbiol 2019; 10:198. [PMID: 30800115 PMCID: PMC6376900 DOI: 10.3389/fmicb.2019.00198] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 01/24/2019] [Indexed: 12/11/2022] Open
Abstract
Antimicrobial peptides (AMPs) are promising templates for the development of novel antibiofilm drugs. Despite the large number of studies on screening and optimization of AMPs, only a few of these evaluated the antibiofilm activity in physiologically relevant model systems. Potent in vitro activity of AMPs often does not translate into in vivo effectiveness due to the interference of the host microenvironment with peptide stability/availability. Hence, mimicking the complex environment found in biofilm-associated infections is essential to predict the clinical potential of novel AMP-based antimicrobials. In the present study, we examined the antibiofilm activity of the semi-synthetic peptide lin-SB056-1 and its dendrimeric derivative (lin-SB056-1)2-K against Pseudomonas aeruginosa in an in vivo-like three-dimensional (3-D) lung epithelial cell model and an in vitro wound model (consisting of an artificial dermis and blood components at physiological levels). Although moderately active when tested alone, lin-SB056-1 was effective in reducing P. aeruginosa biofilm formation in association with 3-D lung epithelial cells in combination with the chelating agent EDTA. The dimeric derivative (lin-SB056-1)2-K demonstrated an enhanced biofilm-inhibitory activity as compared to both lin-SB056-1 and the lin-SB056-1/EDTA combination, reducing the number of biofilm-associated bacteria up to 3-Log units at concentrations causing less than 20% cell death. Biofilm inhibition by (lin-SB056-1)2-K was reported both for the reference strain PAO1 and cystic fibrosis lung isolates of P. aeruginosa. In addition, using fluorescence microscopy, a significant decrease in biofilm-like structures associated with 3-D cells was observed after peptide exposure. Interestingly, effectiveness of (lin-SB056-1)2-K was also demonstrated in the wound model with a reduction of up to 1-Log unit in biofilm formation by P. aeruginosa PAO1 and wound isolates. Overall, combination treatment and peptide dendrimerization emerged as promising strategies to improve the efficacy of AMPs, especially under challenging host-mimicking conditions. Furthermore, the results of the present study underlined the importance of evaluating the biological properties of novel AMPs in in vivo-like model systems representative of specific infectious sites in order to make a more realistic prediction of their therapeutic success, and avoid the inclusion of unpromising peptides in animal studies and clinical trials.
Collapse
Affiliation(s)
- Lucia Grassi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Giovanna Batoni
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Lisa Ostyn
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Petra Rigole
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | | | - Andrea C Rinaldi
- Department of Biomedical Sciences, University of Cagliari, Cagliari, Italy
| | - Giuseppantonio Maisetta
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Semih Esin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| | - Aurélie Crabbé
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
47
|
Moriarty TF, Harris LG, Mooney RA, Wenke JC, Riool M, Zaat SAJ, Moter A, Schaer TP, Khanna N, Kuehl R, Alt V, Montali A, Liu J, Zeiter S, Busscher HJ, Grainger DW, Richards RG. Recommendations for design and conduct of preclinical in vivo studies of orthopedic device-related infection. J Orthop Res 2019; 37:271-287. [PMID: 30667561 DOI: 10.1002/jor.24230] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 12/06/2018] [Indexed: 02/04/2023]
Abstract
Orthopedic device-related infection (ODRI), including both fracture-related infection (FRI) and periprosthetic joint infection (PJI), remain among the most challenging complications in orthopedic and musculoskeletal trauma surgery. ODRI has been convincingly shown to delay healing, worsen functional outcome and incur significant socio-economic costs. To address this clinical problem, ever more sophisticated technologies targeting the prevention and/or treatment of ODRI are being developed and tested in vitro and in vivo. Among the most commonly described innovations are antimicrobial-coated orthopedic devices, antimicrobial-loaded bone cements and void fillers, and dual osteo-inductive/antimicrobial biomaterials. Unfortunately, translation of these technologies to the clinic has been limited, at least partially due to the challenging and still evolving regulatory environment for antimicrobial drug-device combination products, and a lack of clarity in the burden of proof required in preclinical studies. Preclinical in vivo testing (i.e. animal studies) represents a critical phase of the multidisciplinary effort to design, produce and reliably test both safety and efficacy of any new antimicrobial device. Nonetheless, current in vivo testing protocols, procedures, models, and assessments are highly disparate, irregularly conducted and reported, and without standardization and validation. The purpose of the present opinion piece is to discuss best practices in preclinical in vivo testing of antimicrobial interventions targeting ODRI. By sharing these experience-driven views, we aim to aid others in conducting such studies both for fundamental biomedical research, but also for regulatory and clinical evaluation. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:271-287, 2019.
Collapse
Affiliation(s)
- T Fintan Moriarty
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos Platz, Switzerland
| | - Llinos G Harris
- Microbiology and Infectious Diseases, Institute of Life Science, Swansea University Medical School, Swansea, United Kingdom
| | - Robert A Mooney
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York
| | - Joseph C Wenke
- Extremity Trauma and Regenerative Medicine Task Area, US Army Institute of Surgical Research, JBSA-Fort Sam Houston, Texas
| | - Martijn Riool
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology, Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands
| | - Sebastian A J Zaat
- Amsterdam UMC, University of Amsterdam, Department of Medical Microbiology, Amsterdam Infection and Immunity Institute, Amsterdam, The Netherlands
| | - Annette Moter
- Institute of Microbiology and Infection Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas P Schaer
- Department of Clinical Studies New Bolton Center, University of Pennsylvania, Kennett Square, Pennsylvania
| | - Nina Khanna
- Infection Biology Laboratory, Department of Biomedicine, University Hospital of Basel, Basel, Switzerland.,Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Basel, Basel, Switzerland
| | - Richard Kuehl
- Infection Biology Laboratory, Department of Biomedicine, University Hospital of Basel, Basel, Switzerland.,Division of Infectious Diseases and Hospital Epidemiology, University Hospital of Basel, Basel, Switzerland
| | - Volker Alt
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Giessen-Marburg, GmbH, Campus Giessen, Germany
| | | | - Jianfeng Liu
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, P.R. China
| | - Stephan Zeiter
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos Platz, Switzerland
| | - Henk J Busscher
- Department of Biomedical Engineering, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - David W Grainger
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA.,Department of Pharmaceutics and Pharmaceutical Chemistry, University of Utah, Salt Lake City, Utah, USA
| | - R Geoff Richards
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos Platz, Switzerland
| |
Collapse
|
48
|
In Vivo Gentamicin Susceptibility Test for Prevention of Bacterial Biofilms in Bone Tissue and on Implants. Antimicrob Agents Chemother 2019; 63:AAC.01889-18. [PMID: 30455228 DOI: 10.1128/aac.01889-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 11/12/2018] [Indexed: 12/22/2022] Open
Abstract
The objective of this study was to set up an in vivo gentamicin susceptibility test for biofilm prevention in bone tissue and on implants. Twenty-five pigs were allocated to six groups. Pigs in group A (n = 6) were inoculated with saline. Pigs in groups B (n = 6), C (n = 3), D (n = 3), E (n = 3), and F (n = 4) were inoculated with 10 μl saline containing 104 CFU of Staphylococcus aureus Different concentrations based on the MIC of gentamicin for the specific strain were added to the 10-μl inoculum for groups C (160× MIC), D (1,600× MIC), E (16,000× MIC), and F (160,000× MIC). The inocula were injected into a predrilled tibial implant cavity, followed by insertion of a steel implant (2 by 15 mm). The pigs were euthanized after 5 days. In vitro, all the doses used were found to be bactericidal after up to 6 h. All implant cavities of pigs inoculated with bacteria and bacteria plus 160× MIC or 1,600× MIC of gentamicin were positive for S. aureus In animals in each of groups E (16,000× MIC) and F (160,000× MIC), 2/3 and 1/4 of the implant cavities were S. aureus positive, respectively. By grouping groups C and D (<10,000× MIC) and groups E and F (>10,000× MIC), a significant decrease in the number of implant-attached bacteria was seen only between the high-MIC-value group and group B. Histologically, it was demonstrated that 1,600×, 16,000×, and 160,000× MIC resulted in a peri-implant tissue reaction comparable to that in saline-inoculated animals. In vivo, the antimicrobial tolerance of the inoculated planktonic bacteria was increased by in vivo-specific factors of acute inflammation. This resulted in bacterial aggregation and biofilm formation, which further increased the gentamicin tolerance. Thus, susceptibility patterns in vitro might not reflect the actual in vivo susceptibility locally within a developing infectious area.
Collapse
|
49
|
Van Dijck P, Sjollema J, Cammue BPA, Lagrou K, Berman J, d’Enfert C, Andes DR, Arendrup MC, Brakhage AA, Calderone R, Cantón E, Coenye T, Cos P, Cowen LE, Edgerton M, Espinel-Ingroff A, Filler SG, Ghannoum M, Gow NA, Haas H, Jabra-Rizk MA, Johnson EM, Lockhart SR, Lopez-Ribot JL, Maertens J, Munro CA, Nett JE, Nobile CJ, Pfaller MA, Ramage G, Sanglard D, Sanguinetti M, Spriet I, Verweij PE, Warris A, Wauters J, Yeaman MR, Zaat SA, Thevissen K. Methodologies for in vitro and in vivo evaluation of efficacy of antifungal and antibiofilm agents and surface coatings against fungal biofilms. MICROBIAL CELL (GRAZ, AUSTRIA) 2018; 5:300-326. [PMID: 29992128 PMCID: PMC6035839 DOI: 10.15698/mic2018.07.638] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 05/24/2018] [Indexed: 12/13/2022]
Abstract
Unlike superficial fungal infections of the skin and nails, which are the most common fungal diseases in humans, invasive fungal infections carry high morbidity and mortality, particularly those associated with biofilm formation on indwelling medical devices. Therapeutic management of these complex diseases is often complicated by the rise in resistance to the commonly used antifungal agents. Therefore, the availability of accurate susceptibility testing methods for determining antifungal resistance, as well as discovery of novel antifungal and antibiofilm agents, are key priorities in medical mycology research. To direct advancements in this field, here we present an overview of the methods currently available for determining (i) the susceptibility or resistance of fungal isolates or biofilms to antifungal or antibiofilm compounds and compound combinations; (ii) the in vivo efficacy of antifungal and antibiofilm compounds and compound combinations; and (iii) the in vitro and in vivo performance of anti-infective coatings and materials to prevent fungal biofilm-based infections.
Collapse
Affiliation(s)
- Patrick Van Dijck
- VIB-KU Leuven Center for Microbiology, Leuven, Belgium
- KU Leuven Laboratory of Molecular Cell Biology, Leuven, Belgium
| | - Jelmer Sjollema
- University of Groningen, University Medical Center Groningen, Department of BioMedical Engineering, Groningen, The Netherlands
| | - Bruno P. A. Cammue
- Centre for Microbial and Plant Genetics, KU Leuven, Leuven, Belgium
- Department of Plant Systems Biology, VIB, Ghent, Belgium
| | - Katrien Lagrou
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium
- Clinical Department of Laboratory Medicine and National Reference Center for Mycosis, UZ Leuven, Belgium
| | - Judith Berman
- School of Molecular Cell Biology and Biotechnology, Faculty of Life Sciences, Tel Aviv University, Ramat Aviv, Israel
| | - Christophe d’Enfert
- Institut Pasteur, INRA, Unité Biologie et Pathogénicité Fongiques, Paris, France
| | - David R. Andes
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Maiken C. Arendrup
- Unit of Mycology, Statens Serum Institut, Copenhagen, Denmark
- Department of Clinical Microbiology, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Axel A. Brakhage
- Leibniz Institute for Natural Product Research and Infection Biology - Hans Knoell Institute (HKI), Dept. Microbiology and Molecular Biology, Friedrich Schiller University Jena, Institute of Microbiology, Jena, Germany
| | - Richard Calderone
- Department of Microbiology & Immunology, Georgetown University Medical Center, Washington DC, USA
| | - Emilia Cantón
- Severe Infection Research Group: Medical Research Institute La Fe (IISLaFe), Valencia, Spain
| | - Tom Coenye
- Laboratory of Pharmaceutical Microbiology, Ghent University, Ghent, Belgium
- ESCMID Study Group for Biofilms, Switzerland
| | - Paul Cos
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Belgium
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Mira Edgerton
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, Buffalo, NY USA
| | | | - Scott G. Filler
- Division of Infectious Diseases, Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Mahmoud Ghannoum
- Center for Medical Mycology, Department of Dermatology, University Hospitals Cleveland Medical Center and Case Western Re-serve University, Cleveland, OH, USA
| | - Neil A.R. Gow
- MRC Centre for Medical Mycology, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Hubertus Haas
- Biocenter - Division of Molecular Biology, Medical University Innsbruck, Innsbruck, Austria
| | - Mary Ann Jabra-Rizk
- Department of Oncology and Diagnostic Sciences, School of Dentistry; Department of Microbiology and Immunology, School of Medicine, University of Maryland, Baltimore, USA
| | - Elizabeth M. Johnson
- National Infection Service, Public Health England, Mycology Reference Laboratory, Bristol, UK
| | | | | | - Johan Maertens
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium and Clinical Department of Haematology, UZ Leuven, Leuven, Belgium
| | - Carol A. Munro
- MRC Centre for Medical Mycology, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Jeniel E. Nett
- University of Wisconsin-Madison, Departments of Medicine and Medical Microbiology & Immunology, Madison, WI, USA
| | - Clarissa J. Nobile
- Department of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, Merced, USA
| | - Michael A. Pfaller
- Departments of Pathology and Epidemiology, University of Iowa, Iowa, USA
- JMI Laboratories, North Liberty, Iowa, USA
| | - Gordon Ramage
- ESCMID Study Group for Biofilms, Switzerland
- College of Medical, Veterinary and Life Sciences, University of Glasgow, UK
| | - Dominique Sanglard
- Institute of Microbiology, University of Lausanne and University Hospital, CH-1011 Lausanne
| | - Maurizio Sanguinetti
- Institute of Microbiology, Università Cattolica del Sacro Cuore, IRCCS-Fondazione Policlinico "Agostino Gemelli", Rome, Italy
| | - Isabel Spriet
- Pharmacy Dpt, University Hospitals Leuven and Clinical Pharmacology and Pharmacotherapy, Dpt. of Pharmaceutical and Pharma-cological Sciences, KU Leuven, Belgium
| | - Paul E. Verweij
- Center of Expertise in Mycology Radboudumc/CWZ, Radboud University Medical Center, Nijmegen, the Netherlands (omit "Nijmegen" in Radboud University Medical Center)
| | - Adilia Warris
- MRC Centre for Medical Mycology, Aberdeen Fungal Group, University of Aberdeen, Foresterhill, Aberdeen, UK
| | - Joost Wauters
- KU Leuven-University of Leuven, University Hospitals Leuven, Department of General Internal Medicine, Herestraat 49, B-3000 Leuven, Belgium
| | - Michael R. Yeaman
- Geffen School of Medicine at the University of California, Los Angeles, Divisions of Molecular Medicine & Infectious Diseases, Har-bor-UCLA Medical Center, LABioMed at Harbor-UCLA Medical Center
| | - Sebastian A.J. Zaat
- Department of Medical Microbiology, Amsterdam Infection and Immunity Institute, Academic Medical Center, University of Am-sterdam, Netherlands
| | - Karin Thevissen
- Centre for Microbial and Plant Genetics, KU Leuven, Leuven, Belgium
| |
Collapse
|
50
|
Gilmore BF, Flynn PB, O'Brien S, Hickok N, Freeman T, Bourke P. Cold Plasmas for Biofilm Control: Opportunities and Challenges. Trends Biotechnol 2018; 36:627-638. [PMID: 29729997 DOI: 10.1016/j.tibtech.2018.03.007] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 03/21/2018] [Accepted: 03/23/2018] [Indexed: 02/07/2023]
Abstract
Bacterial biofilm infections account for a major proportion of chronic and medical device associated infections in humans, yet our ability to control them is compromised by their inherent tolerance to antimicrobial agents. Cold atmospheric plasma (CAP) represents a promising therapeutic option. CAP treatment of microbial biofilms represents the convergence of two complex phenomena: the production of a chemically diverse mixture of reactive species and intermediates, and their interaction with a heterogeneous 3D interface created by the biofilm extracellular polymeric matrix. Therefore, understanding these interactions and physiological responses to CAP exposure are central to effective management of infectious biofilms. We review the unique opportunities and challenges for translating CAP to the management of biofilms.
Collapse
Affiliation(s)
- Brendan F Gilmore
- Biofilm and Pharmaceutical Microbiology Research Group, School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK. http://twitter.com/@BrendanFGilmore
| | - Padrig B Flynn
- Biofilm and Pharmaceutical Microbiology Research Group, School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Séamus O'Brien
- Biofilm and Pharmaceutical Microbiology Research Group, School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast, BT9 7BL, UK
| | - Noreen Hickok
- Department of Orthopaedic Research, Sidney Kimmel Medical College of Thomas Jefferson University, Jefferson Medical College, 1015 Walnut Street, Suite 501, Philadelphia, PA 19107, USA
| | - Theresa Freeman
- Department of Orthopaedic Research, Sidney Kimmel Medical College of Thomas Jefferson University, Jefferson Medical College, 1015 Walnut Street, Suite 501, Philadelphia, PA 19107, USA
| | - Paula Bourke
- Plasma Research Group, School of Food Science and Environmental Health, Dublin Institute of Technology, Marlborough Street, Dublin 1, Ireland
| |
Collapse
|