1
|
Barman S, Kurnaz LB, Leighton R, Hossain MW, Decho AW, Tang C. Intrinsic antimicrobial resistance: Molecular biomaterials to combat microbial biofilms and bacterial persisters. Biomaterials 2024; 311:122690. [PMID: 38976935 PMCID: PMC11298303 DOI: 10.1016/j.biomaterials.2024.122690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 05/13/2024] [Accepted: 06/26/2024] [Indexed: 07/10/2024]
Abstract
The escalating rise in antimicrobial resistance (AMR) coupled with a declining arsenal of new antibiotics is imposing serious threats to global public health. A pervasive aspect of many acquired AMR infections is that the pathogenic microorganisms exist as biofilms, which are equipped with superior survival strategies. In addition, persistent and recalcitrant infections are seeded with bacterial persister cells at infection sites. Together, conventional antibiotic therapeutics often fail in the complete treatment of infections associated with bacterial persisters and biofilms. Novel therapeutics have been attempted to tackle AMR, biofilms, and persister-associated complex infections. This review focuses on the progress in designing molecular biomaterials and therapeutics to address acquired and intrinsic AMR, and the fundamental microbiology behind biofilms and persisters. Starting with a brief introduction of AMR basics and approaches to tackling acquired AMR, the emphasis is placed on various biomaterial approaches to combating intrinsic AMR, including (1) semi-synthetic antibiotics; (2) macromolecular or polymeric biomaterials mimicking antimicrobial peptides; (3) adjuvant effects in synergy; (4) nano-therapeutics; (5) nitric oxide-releasing antimicrobials; (6) antimicrobial hydrogels; (7) antimicrobial coatings. Particularly, the structure-activity relationship is elucidated in each category of these biomaterials. Finally, illuminating perspectives are provided for the future design of molecular biomaterials to bypass AMR and cure chronic multi-drug resistant (MDR) infections.
Collapse
Affiliation(s)
- Swagatam Barman
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, United States; Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, 29208, United States
| | - Leman Buzoglu Kurnaz
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, United States
| | - Ryan Leighton
- Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, 29208, United States
| | - Md Waliullah Hossain
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, United States
| | - Alan W Decho
- Department of Environmental Health Sciences, University of South Carolina, Columbia, SC, 29208, United States.
| | - Chuanbing Tang
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, 29208, United States.
| |
Collapse
|
2
|
Huang Z, Li Y, Yin W, Raby RBN, Liang H, Yu B. A magnetic-guided nano-antibacterial platform for alternating magnetic field controlled vancomycin release in staphylococcus aureus biofilm eradication. Drug Deliv Transl Res 2024:10.1007/s13346-024-01667-x. [PMID: 39020245 DOI: 10.1007/s13346-024-01667-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/01/2024] [Indexed: 07/19/2024]
Abstract
Bacterial resilience within biofilms, rendering them up to 1000 times more resistant to antibiotic drugs, poses a formidable challenge. This study introduces a targeted biofilm eradication strategy, termed "target-penetration-killing-eradication", implemented through magnetic micro-robotic technology. Specifically, we present the development of a magnetic-guided nano-antibacterial platform designed for alternating magnetic field (AMF) controlled vancomycin release in the eradication of Staphylococcus aureus biofilms. To address the issue of premature vancomycin release in physiological conditions, we employed a temperature-sensitive linking agent, 4,4'-azobis(4-cyano valeric acid), facilitating the conjugation of vancomycin onto Fe3O4/CS nanocomposites, resulting in the novel construct Fe3O4@CS-ACVA-VH. The release mechanism adheres to first-order kinetics and Fickian diffusion, with each 10-min AMF treatment releasing approximately 8.4 ± 1.1% of vancomycin. The potency of vancomycin in the release solution was similar to that of the original drug (MIC: 7.4 ± 3.5 vs. 5.6 μg/mL). Fe3O4@CS-ACVA-VH exhibited sustained antibacterial efficacy, inhibiting bacterial growth for four consecutive days and preventing the formation of bacterial biofilms on its surface. Contact-inhibition bacterial activity of Fe3O4@CS-ACVA-VH against S. aureus was 0.046875 mg/mL. Conceptually validating our approach, we emphasize Fe3O4@CS-ACVA-VH's exceptional ability to penetrate S. aureus biofilms under static magnetic field attraction. Furthermore, the nano-platform offers the unique advantage of on-demand vancomycin release through alternating magnetic field stimulation, effectively clearing a larger biofilm area. This multifunctional nano-platform demonstrates magnetic-guided biofilm penetration followed by controlled vancomycin release, presenting a promising strategy for enhanced biofilm eradication.
Collapse
Affiliation(s)
- Zhi Huang
- Institute of Biomedical Engineering, School of Basic Medical Sciences, Central South University, Changsha, 410083, China
| | - Yuankai Li
- Institute of Biomedical Engineering, School of Basic Medical Sciences, Central South University, Changsha, 410083, China
| | - Wang Yin
- Institute of Biomedical Engineering, School of Basic Medical Sciences, Central South University, Changsha, 410083, China
| | - Randy Bachelard Nziengui Raby
- Institute of Biomedical Engineering, School of Basic Medical Sciences, Central South University, Changsha, 410083, China
| | - Haifeng Liang
- Orthopedic and Traumatology Department, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
- Department of Orthopedics, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, 510150, China.
| | - Bo Yu
- Orthopedic and Traumatology Department, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, China.
| |
Collapse
|
3
|
Shah KN, Shah PN, Agobe FO, Lovato K, Gao H, Ogun O, Hoffman C, Yabe-Gill M, Chen Q, Sweatt J, Chirra B, Muñoz-Medina R, Farmer DE, Kürti L, Cannon CL. Antimicrobial activity of a natural compound and analogs against multi-drug-resistant Gram-positive pathogens. Microbiol Spectr 2024; 12:e0151522. [PMID: 38289721 PMCID: PMC10913730 DOI: 10.1128/spectrum.01515-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 11/06/2023] [Indexed: 02/01/2024] Open
Abstract
The increasing prevalence of methicillin-resistant Staphylococcus aureus (MRSA) has sparked global concern due to the dwindling availability of effective antibiotics. To increase our treatment options, researchers have investigated naturally occurring antimicrobial compounds and have identified MC21-A (C58), which has potent antimicrobial activity against MRSA. Recently, we have devised total synthesis schemes for C58 and its chloro-analog, C59. Here, we report that both compounds eradicate 90% of the 39 MRSA isolates tested [MIC90 and minimum bactericidal concentration (MBC90)] at lower or comparable concentrations compared to several standard-of-care (SoC) antimicrobials including daptomycin, vancomycin, and linezolid. Furthermore, a stable, water-soluble sodium salt of C59, C59Na, demonstrates antimicrobial activity comparable to C59. C59, unlike vancomycin, kills stationary-phase MRSA in a dose-dependent manner and completely eradicates MRSA biofilms. In contrast to vancomycin, exposing MRSA to sub-MIC concentrations of C59 does not result in the emergence of spontaneous resistance. Similarly, in a multi-step study, C59 demonstrates a low propensity of resistance acquisition when compared to SoC antimicrobials, such as linezolid and clindamycin. Our findings suggest C58, C59, and C59Na are non-toxic to mammalian cells at concentrations that exert antimicrobial activity; the lethal dose at median cell viability (LD50) is at least fivefold higher than the MBC90 in the two mammalian cell lines tested. A morphological examination of the effects of C59 on a MRSA isolate suggests the inhibition of the cell division process as a mechanism of action. Our results demonstrate the potential of this naturally occurring compound and its analogs as non-toxic next-generation antimicrobials to combat MRSA infections. IMPORTANCE The rapid emergence of methicillin-resistant Staphylococcus aureus (MRSA) isolates has precipitated a critical need for novel antibiotics. We have developed a one-pot synthesis method for naturally occurring compounds such as MC21-A (C58) and its chloro-analog, C59. Our findings demonstrate that these compounds kill MRSA isolates at lower or comparable concentrations to standard-of-care (SoC) antimicrobials. C59 eradicates MRSA cells in biofilms, which are notoriously difficult to treat with SoC antibiotics. Additionally, the lack of resistance development observed with C59 treatment is a significant advantage when compared to currently available antibiotics. Furthermore, these compounds are non-toxic to mammalian cell lines at effective concentrations. Our findings indicate the potential of these compounds to treat MRSA infections and underscore the importance of exploring natural products for novel antibiotics. Further investigation will be essential to fully realize the therapeutic potential of these next-generation antimicrobials to address the critical issue of antimicrobial resistance.
Collapse
Affiliation(s)
- Kush N. Shah
- Department of Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Parth N. Shah
- Department of Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Francesca O. Agobe
- Department of Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Kaitlyn Lovato
- Department of Chemistry, Rice University, Houston, Texas, USA
| | - Hongyin Gao
- Department of Chemistry, Rice University, Houston, Texas, USA
| | - Oluwadara Ogun
- Department of Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Cason Hoffman
- Department of Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Marium Yabe-Gill
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Qingquan Chen
- Department of Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Jordan Sweatt
- Department of Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Bhagath Chirra
- Department of Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Ricardo Muñoz-Medina
- Department of Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - Delaney E. Farmer
- Department of Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| | - László Kürti
- Department of Chemistry, Rice University, Houston, Texas, USA
| | - Carolyn L. Cannon
- Department of Microbial Pathogenesis & Immunology, College of Medicine, Texas A&M University Health Science Center, Bryan, Texas, USA
| |
Collapse
|
4
|
Yue Y, Chen K, Sun C, Ahmed S, Ojha SC. Antimicrobial peptidase lysostaphin at subinhibitory concentrations modulates staphylococcal adherence, biofilm formation, and toxin production. BMC Microbiol 2023; 23:311. [PMID: 37884887 PMCID: PMC10601153 DOI: 10.1186/s12866-023-03052-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND The ability of antimicrobial agents to affect microbial adherence to eukaryotic cell surfaces is a promising antivirulence strategy for combating the global threat of antimicrobial resistance. Inadequate use of antimicrobials has led to widespread instances of suboptimal antibiotic concentrations around infection sites. Therefore, we aimed to examine the varying effect of an antimicrobial peptidase lysostaphin (APLss) on staphylococcal adherence to host cells, biofilm biomass formation, and toxin production as a probable method for mitigating staphylococcal virulence. RESULTS Initially, soluble expression in E. coli and subsequent purification by immobilized-Ni2+ affinity chromatography (IMAC) enabled us to successfully produce a large quantity of highly pure ~ 28-kDa His-tagged mature APLss. The purified protein exhibited potent inhibitory effects against both methicillin-sensitive and methicillin-resistant staphylococcal strains, with minimal inhibitory concentrations (MICs) ranging from 1 to 2 µg/mL, and ultrastructural analysis revealed that APLss-induced concentration-specific changes in the morphological architecture of staphylococcal surface membranes. Furthermore, spectrophotometric and fluorescence microscopy revealed that incubating staphylococcal strains with sub-MIC and MIC of APLss significantly inhibited staphylococcal adherence to human vaginal epithelial cells and biofilm biomass formation. Ultimately, transcriptional investigations revealed that APLss inhibited the expression of agrA (quorum sensing effector) and other virulence genes related to toxin synthesis. CONCLUSIONS Overall, APLss dose-dependently inhibited adhesion to host cell surfaces and staphylococcal-associated virulence factors, warranting further investigation as a potential anti-staphylococcal agent with an antiadhesive mechanism of action using in vivo models of staphylococcal toxic shock syndrome.
Collapse
Affiliation(s)
- Yuan Yue
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Shaanxi Normal University, Xi'an, China
| | - Ke Chen
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Southwest Medical University, Jiangyang District, Luzhou, 646000, Sichuan, China
| | - Changfeng Sun
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Southwest Medical University, Jiangyang District, Luzhou, 646000, Sichuan, China
| | - Sarfraz Ahmed
- Wellman Centre for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston MA 02114, USA
| | - Suvash Chandra Ojha
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Southwest Medical University, Jiangyang District, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
5
|
Daffinee KE, Piehl EC, Bleick C, LaPlante KL. Eradication of Staphylococcus epidermidis within Biofilms: Comparison of Systemic versus Supratherapeutic Concentrations of Antibiotics. Antimicrob Agents Chemother 2023; 67:e0010823. [PMID: 37154699 PMCID: PMC10269123 DOI: 10.1128/aac.00108-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/15/2023] [Indexed: 05/10/2023] Open
Abstract
Biofilm-forming bacterial infections result in clinical failure, recurring infections, and high health care costs. The antibiotic concentrations needed to eradicate biofilm require further research. We aimed to model an in vitro prosthetic joint infection (PJI) to elucidate the activity of traditional systemic concentrations versus supratherapeutic concentrations to eradicate a Staphylococcus epidermidis biofilm PJI. We evaluated S. epidermidis high-biofilm-forming (ATCC 35984) and low-biofilm-forming (ATCC 12228) isolates in an in vitro pharmacodynamic biofilm reactor model with chromium cobalt coupons to simulate prosthetic joint infection. Vancomycin, daptomycin, levofloxacin, and minocycline were used alone and combined with rifampin to evaluate the effect of biofilm eradication. We simulated three exposures: (i) humanized systemic dosing alone, (ii) supratherapeutic doses (1,000× MIC), and (iii) and dosing in combination with rifampin. Resistance development was monitored throughout the study. Simulated humanized systemic doses of a lipoglycopeptide (daptomycin), a fluoroquinolone (levofloxacin), a tetracycline (minocycline), and a glycopeptide (vancomycin) alone failed to eradicate a formed S. epidermidis biofilm. Supratherapeutic doses of vancomycin (2,000 μg/mL) and minocycline (15 μg/mL) with or without rifampin (15 μg/mL) failed to eradicate biofilms. However, a levofloxacin supratherapeutic dose (125 μg/mL) with rifampin eradicated the high-biofilm-producing isolate by 48 h. Interestingly, supratherapeutic-dose exposures of daptomycin (500 μg/mL) alone eradicated high- and low-biofilm-forming isolates in established biofilms. The concentrations needed to eradicate biofilms on foreign materials are not obtained with systemic dosing regimens. The failure of systemic dosing regimens to eradicate biofilms validates clinical findings with recurring infections. The addition of rifampin to supratherapeutic dosing regimens does not result in synergy. Supratherapeutic daptomycin dosing may be effective at the site of action to eradicate biofilms. Further studies are needed.
Collapse
Affiliation(s)
- K. E. Daffinee
- Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, Rhode Island, USA
| | - E. C. Piehl
- College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
- Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, Rhode Island, USA
| | - C. Bleick
- College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
| | - K. L. LaPlante
- College of Pharmacy, University of Rhode Island, Kingston, Rhode Island, USA
- Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, Rhode Island, USA
- Warren Alpert Medical School of Brown University, Division of Infectious Diseases, Providence, Rhode Island, USA
| |
Collapse
|
6
|
Daffinee KE, O'Neill ET, Bleick CR, Williams G, Antoci V, Garcia D, LaPlante KL. Staphylococcal Biofilm: Penetration and bioavailability of vancomycin with or without rifampin. Diagn Microbiol Infect Dis 2023; 106:115947. [PMID: 37116243 DOI: 10.1016/j.diagmicrobio.2023.115947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 03/14/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023]
Abstract
We measured antibiotic penetration and bioavailability in staphylococcus biofilms using simulated humanized concentrations of fluorescent vancomycin plus or minus rifampin. Vancomycin percent recovery across biofilm layers was:upper = 46%, middle = 40%, and lower = 33%. Vancomycin plus rifampin was not significantly different (P = 0.65). Addition of rifampin did not improve vancomycin penetration across biofilm layers.
Collapse
Affiliation(s)
- Kathryn E Daffinee
- Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Emily T O'Neill
- Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI, USA; College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Callan R Bleick
- College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Geoff Williams
- Leduc Bioimaging Facility, Brown University, Division of Biology and Medicine, Providence, RI, USA
| | - Valentin Antoci
- Department of Orthopaedics, The Warren Alpert School of Medicine, Brown University, Providence, RI, USA; The Diane N. Weiss Center for Orthopaedic Trauma Research, Rhode Island Hospital, Providence, RI, USA
| | - Dioscaris Garcia
- Department of Orthopaedics, The Warren Alpert School of Medicine, Brown University, Providence, RI, USA; The Diane N. Weiss Center for Orthopaedic Trauma Research, Rhode Island Hospital, Providence, RI, USA
| | - Kerry L LaPlante
- Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI, USA; College of Pharmacy, University of Rhode Island, Kingston, RI, USA; Warren Alpert Medical School of Brown University, Division of Infectious Diseases, Providence, RI, USA.
| |
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
|
Sebastian S, Tandberg F, Liu Y, Raina DB, Tägil M, Collin M, Lidgren L. Extended local release and improved bacterial eradication by adding rifampicin to a biphasic ceramic carrier containing gentamicin or vancomycin. Bone Joint Res 2022; 11:787-802. [DOI: 10.1302/2046-3758.1111.bjr-2022-0101.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Aims There is a lack of biomaterial-based carriers for the local delivery of rifampicin (RIF), one of the cornerstone second defence antibiotics for bone infections. RIF is also known for causing rapid development of antibiotic resistance when given as monotherapy. This in vitro study evaluated a clinically used biphasic calcium sulphate/hydroxyapatite (CaS/HA) biomaterial as a carrier for dual delivery of RIF with vancomycin (VAN) or gentamicin (GEN). Methods The CaS/HA composites containing RIF/GEN/VAN, either alone or in combination, were first prepared and their injectability, setting time, and antibiotic elution profiles were assessed. Using a continuous disk diffusion assay, the antibacterial behaviour of the material was tested on both planktonic and biofilm-embedded forms of standard and clinical strains of Staphylococcus aureus for 28 days. Development of bacterial resistance to RIF was determined by exposing the biofilm-embedded bacteria continuously to released fractions of antibiotics from CaS/HA-antibiotic composites. Results Following the addition of RIF to CaS/HA-VAN/GEN, adequate injectability and setting of the CaS/HA composites were noted. Sustained release of RIF above the minimum inhibitory concentrations of S. aureus was observed until study endpoint (day 35). Only combinations of CaS/HA-VAN/GEN + RIF exhibited antibacterial and antibiofilm effects yielding no viable bacteria at study endpoint. The S. aureus strains developed resistance to RIF when biofilms were subjected to CaS/HA-RIF alone but not with CaS/HA-VAN/GEN + RIF. Conclusion Our in vitro results indicate that biphasic CaS/HA loaded with VAN or GEN could be used as a carrier for RIF for local delivery in clinically demanding bone infections. Cite this article: Bone Joint Res 2022;11(11):787–802.
Collapse
Affiliation(s)
- Sujeesh Sebastian
- Department of Clinical Sciences, Orthopedics, Lund University Faculty of Medicine, Lund, Sweden
| | - Felix Tandberg
- Department of Clinical Sciences, Orthopedics, Lund University Faculty of Medicine, Lund, Sweden
| | - Yang Liu
- Department of Clinical Sciences, Orthopedics, Lund University Faculty of Medicine, Lund, Sweden
| | - Deepak B. Raina
- Department of Clinical Sciences, Orthopedics, Lund University Faculty of Medicine, Lund, Sweden
| | - Magnus Tägil
- Department of Clinical Sciences, Orthopedics, Lund University Faculty of Medicine, Lund, Sweden
| | - Mattias Collin
- Division of Infection Medicine, Department of Clinical Sciences, Lund University Faculty of Medicine, Lund, Sweden
| | - Lars Lidgren
- Department of Clinical Sciences, Orthopedics, Lund University Faculty of Medicine, Lund, Sweden
| |
Collapse
|
9
|
Venkateswaran P, Lakshmanan PM, Muthukrishnan S, Bhagavathi H, Vasudevan S, Neelakantan P, Solomon AP. Hidden agenda of Enterococcus faecalis lifestyle transition: planktonic to sessile state. Future Microbiol 2022; 17:1051-1069. [PMID: 35899477 DOI: 10.2217/fmb-2021-0212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Enterococcus faecalis, a human gastrointestinal tract commensal, is known to cause nosocomial infections. Interestingly, the pathogen's host colonization and persistent infections are possibly linked to its lifestyle changes from planktonic to sessile state. Also, the multidrug resistance and survival fitness acquired in the sessile stage of E. faecalis has challenged treatment regimes. This situation exists because of the critical role played by several root genes and their molecular branches, which are part of quorum sensing, aggregation substance, surface adhesions, stress-related response and sex pheromones in the sessile state. It is therefore imperative to decode the hidden agenda of E. faecalis and understand the significant factors influencing biofilm formation. This would, in turn, augment the development of novel strategies to tackle E. faecalis infections.
Collapse
Affiliation(s)
- Parvathy Venkateswaran
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur, 613401, India
| | - Priya M Lakshmanan
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur, 613401, India
| | - Sudhiksha Muthukrishnan
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur, 613401, India
| | - Hema Bhagavathi
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur, 613401, India
| | - Sahana Vasudevan
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur, 613401, India
| | | | - Adline P Solomon
- Quorum Sensing Laboratory, Centre for Research in Infectious Diseases (CRID), School of Chemical & Biotechnology, SASTRA Deemed to be University, Thanjavur, 613401, India
| |
Collapse
|
10
|
Fiallos NDM, Aguiar ALR, Nascimento da Silva B, Rocha MFG, Sidrim JJC, Castelo Branco de Souza Collares Maia D, Cordeiro RDA. Enterococcus faecalis and Candida albicans dual-species biofilm: establishment of an in vitro protocol and characterization. BIOFOULING 2022; 38:401-413. [PMID: 35655421 DOI: 10.1080/08927014.2022.2084612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 05/25/2022] [Accepted: 05/26/2022] [Indexed: 06/15/2023]
Abstract
Enterococcus faecalis is the most important agent of persistent apical periodontitis, and recently, Candida albicans has also been implicated in periapical infections. This study aimed to optimize an in vitro E. faecalis and C. albicans dual-species biofilm protocol for endodontic research. Different physicochemical conditions for biofilm formation were tested. Susceptibility assays to antimicrobials, biochemical composition and an ultra-morphological structure analyses were performed. Reproducible dual-species biofilms were established in BHI medium at 35 °C, for 48 h and in a microaerophilic atmosphere. An increase in biomass and chitin content was detected after vancomycin treatment. Structural analysis revealed that the dual-species biofilm was formed by both microorganisms adhered to the substrate. The proposed protocol could be useful for the study of interkingdom relationships and help to find new strategies against periapical infections.
Collapse
|
11
|
Gatti M, Barnini S, Guarracino F, Parisio EM, Spinicci M, Viaggi B, D’Arienzo S, Forni S, Galano A, Gemmi F. Orthopaedic Implant-Associated Staphylococcal Infections: A Critical Reappraisal of Unmet Clinical Needs Associated with the Implementation of the Best Antibiotic Choice. Antibiotics (Basel) 2022; 11:antibiotics11030406. [PMID: 35326869 PMCID: PMC8944676 DOI: 10.3390/antibiotics11030406] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 12/24/2022] Open
Abstract
Infections associated with orthopaedic implants represent a major health concern characterized by a remarkable incidence of morbidity and mortality. The wide variety of clinical scenarios encountered in the heterogeneous world of infections associated with orthopaedic implants makes the implementation of an optimal and standardized antimicrobial treatment challenging. Antibiotic bone penetration, anti-biofilm activity, long-term safety, and drug choice/dosage regimens favouring outpatient management (i.e., long-acting or oral agents) play a major role in regards to the chronic evolution of these infections. The aim of this multidisciplinary opinion article is to summarize evidence supporting the use of the different anti-staphylococcal agents in terms of microbiological and pharmacological optimization according to bone penetration, anti-biofilm activity, long-term safety, and feasibility for outpatient regimens, and to provide a useful guide for clinicians in the management of patients affected by staphylococcal infections associated with orthopaedic implants Novel long-acting lipoglycopeptides, and particularly dalbavancin, alone or in combination with rifampicin, could represent the best antibiotic choice according to real-world evidence and pharmacokinetic/pharmacodynamic properties. The implementation of a multidisciplinary taskforce and close cooperation between microbiologists and clinicians is crucial for providing the best care in this scenario.
Collapse
Affiliation(s)
- Milo Gatti
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy;
- SSD Clinical Pharmacology, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Simona Barnini
- Bacteriology Unit, Azienda Ospedaliero Universitaria Pisana, 56126 Pisa, Italy;
| | - Fabio Guarracino
- Department of Anaesthesia and Critical Care Medicine, Azienda Ospedaliero Universitaria Pisana, 56126 Pisa, Italy;
| | - Eva Maria Parisio
- UOSD Microbiologia Arezzo PO San Donato, Azienda Usl Toscana Sud Est, 52100 Arezzo, Italy;
| | - Michele Spinicci
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy;
- Infectious and Tropical Diseases Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Bruno Viaggi
- Neurointensive Care Unit, Department of Anesthesiology, Careggi University Hospital, 50134 Florence, Italy;
| | - Sara D’Arienzo
- Agenzia Regionale di Sanità della Toscana, 50141 Florence, Italy; (S.D.); (S.F.)
| | - Silvia Forni
- Agenzia Regionale di Sanità della Toscana, 50141 Florence, Italy; (S.D.); (S.F.)
| | - Angelo Galano
- SOD Microbiologia e Virologia, Careggi University Hospital, 50134 Florence, Italy;
| | - Fabrizio Gemmi
- Agenzia Regionale di Sanità della Toscana, 50141 Florence, Italy; (S.D.); (S.F.)
- Correspondence:
| |
Collapse
|
12
|
Guchhait KC, Manna T, Barai M, Karmakar M, Nandi SK, Jana D, Dey A, Panda S, Raul P, Patra A, Bhattacharya R, Chatterjee S, Panda AK, Ghosh C. Antibiofilm and anticancer activities of unripe and ripe Azadirachta indica (neem) seed extracts. BMC Complement Med Ther 2022; 22:42. [PMID: 35152903 PMCID: PMC8843028 DOI: 10.1186/s12906-022-03513-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 01/07/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Antibiotic resistances of pathogens and breast cancer warrant the search for new alternative strategies. Phytoextracts can eradicate microbe-borne diseases as well as cancer with lower side effects compared to conventional antibiotics. AIM Unripe and ripe Azadirachta indica (neem) seed extracts were explored as potential antibiofilm and anticancer agents in combating multidrug-resistant infectious bacteria as well as anticancer agents against the MDR breast cancer cell lines. METHODS Shed-dried neem seeds (both unripe and ripe) were pulverized and extracted using methanol. The chemical components were identified with FTIR and gas chromatography - mass spectrometry. Antibiofilm activity of neem seed extracts were assessed in terms of minimum biofilm inhibitory concentration (MBIC), minimum biofilm eradication concentration (MBEC), and fluorescence microscopic studies on Staphylococcus aureus and Vibrio cholerae. Bacterial cells were studied by fluorescence microscopy using acridine orange/ethidium bromide as the staining agents. Minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) values were evaluated to observe the antibacterial activities. Cytotoxicity of the extracts against human blood lymphocytes and the anticancer activity against drug-resistant breast cancer cell lines were assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and fluorescence-activated cell sorting (FACS) studies. RESULTS 4-Ethyl-2-hydroxy-2-cyclopentene-1-one, phthalic acid, and 2-hexyl-tetrahydro thiophane were the major compounds in unripe neem seed, whereas 3,5-dihydroxy-6-methyl-2,3-dihydro-4-H-pyran-4-one and 4-ethylbenzamide were predominant in ripe neem seed. Triazine derivatives were also common for both the extracts. MBIC values of unripe and ripe neem seed extracts for S. aureus are 75 and 100 µg/mL, respectively, and for V. cholerae, they are 100 and 300 µg/mL, respectively. MBEC values of unripe and ripe seed extracts are 500 and 300 µg/mL, respectively for S. aureus and for V. cholerae the values are 700 and 500 µg/mL, respectively. Fluorescence microscopic studies at 16 and 24 h, after bacterial culture, demonstrate enhanced antibiofilm activity for the ripe seed extract than that of the unripe seeds for both the bacteria. MTT assay reveals lower cytotoxicity of both the extracts towards normal blood lymphocytes, and anticancer activity against breast cancer cell line (MDA-MB-231) with superior activity of ripe seed extract. FACS studies further supported higher anticancer activity for ripe seed extract. CONCLUSIONS Methanolic extract of neem seeds could substantially inhibit and eradicate biofilm along with their potent antibacterial and anticancer activities. Both the extracts showed higher antibiofilm and antibacterial activity against S. aureus (gram-positive) than V. cholerae (gram-negative). Moreover, ripe seed extract showed higher antibiofilm and anticancer activity than unripe extracts.
Collapse
Affiliation(s)
- Kartik Chandra Guchhait
- Department of Human Physiology, Vidyasagar University, Midnapore, 721102, West Bengal, India
| | - Tuhin Manna
- Department of Human Physiology, Vidyasagar University, Midnapore, 721102, West Bengal, India
| | - Manas Barai
- Department of Chemistry, Vidyasagar University, Midnapore, 721102, West Bengal, India
| | - Monalisha Karmakar
- Department of Human Physiology, Vidyasagar University, Midnapore, 721102, West Bengal, India
| | - Sourav Kumar Nandi
- Department of Molecular Biology, Netaji Subhas Chandra Bose Cancer Research Institute, 3081 Nayabad, Kolkata, 700094, West Bengal, India
| | - Debarati Jana
- Department of Human Physiology, Vidyasagar University, Midnapore, 721102, West Bengal, India
| | - Aditi Dey
- Department of Human Physiology, Vidyasagar University, Midnapore, 721102, West Bengal, India
| | - Suman Panda
- Department of Biophysics, Bose Institute, P-1/12 CIT Road, Scheme VIIM, Kankurgachi, Kolkata, 700054, West Bengal, India
| | - Priyanka Raul
- Department of Human Physiology, Vidyasagar University, Midnapore, 721102, West Bengal, India
| | - Anuttam Patra
- Chemistry of Interfaces Group, Luleå University of Technology, SE- 97187, Luleå, Sweden
| | - Rittwika Bhattacharya
- Department of Molecular Biology, Netaji Subhas Chandra Bose Cancer Research Institute, 3081 Nayabad, Kolkata, 700094, West Bengal, India
| | - Subhrangsu Chatterjee
- Department of Biophysics, Bose Institute, P-1/12 CIT Road, Scheme VIIM, Kankurgachi, Kolkata, 700054, West Bengal, India
| | - Amiya Kumar Panda
- Department of Chemistry, Vidyasagar University, Midnapore, 721102, West Bengal, India
- Sadhu Ram Chand Murmu University of Jhargram, Jhargram, 721507, West Bengal, India
| | - Chandradipa Ghosh
- Department of Human Physiology, Vidyasagar University, Midnapore, 721102, West Bengal, India.
| |
Collapse
|
13
|
Song ZM, Zhang JL, Zhou K, Yue LM, Zhang Y, Wang CY, Wang KL, Xu Y. Anthraquinones as Potential Antibiofilm Agents Against Methicillin-Resistant Staphylococcus aureus. Front Microbiol 2021; 12:709826. [PMID: 34539607 PMCID: PMC8446625 DOI: 10.3389/fmicb.2021.709826] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 08/04/2021] [Indexed: 12/01/2022] Open
Abstract
Biofilms formed by methicillin-resistant Staphylococcus aureus (MRSA) are one of the contributing factors to recurrent nosocomial infection in humans. There is currently no specific treatment targeting on biofilms in clinical trials approved by FDA, and antibiotics remain the primary therapeutic strategy. In this study, two anthraquinone compounds isolated from a rare actinobacterial strain Kitasatospora albolonga R62, 3,8-dihydroxy-l-methylanthraquinon-2-carboxylic acid (1) and 3,6,8-trihydroxy-1-methylanthraquinone-2-carboxylic acid (2), together with their 10 commercial analogs 3-12 were evaluated for antibacterial and antibiofilm activities against MRSA, which led to the discovery of two potential antibiofilm anthraquinone compounds anthraquinone-2-carboxlic acid (6) and rhein (12). The structure-activity relationship analysis of these anthraquinones indicated that the hydroxyl group at the C-2 position of the anthraquinone skeleton played an important role in inhibiting biofilm formation at high concentrations, while the carboxyl group at the same C-2 position had a great influence on the antibacterial activity and biofilm eradication activity. The results of crystal violet and methyl thiazolyl tetrazolium staining assays, as well as scanning electron microscope and confocal scanning laser microscopy imaging of compounds 6 and 12 treatment groups showed that both compounds could disrupt preformed MRSA biofilms possibly by killing or dispersing biofilm cells. RNA-Seq was subsequently used for the preliminary elucidation of the mechanism of biofilm eradication, and the results showed upregulation of phosphate transport-related genes in the overlapping differentially expressed genes of both compound treatment groups. Herein, we propose that anthraquinone compounds 6 and 12 could be considered promising candidates for the development of antibiofilm agents.
Collapse
Affiliation(s)
- Zhi-Man Song
- Shenzhen Key Laboratory of Marine Bioresource and Eco-Environmental Science, Shenzhen Engineering Laboratory for Marine Algal Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
- Department of Chemistry, The University of Hong Kong, Pokfulam, Hong Kong, China
- College of Pharmacy, Institute of Materia Medica, Dali University, Dali, China
| | - Jun-Liang Zhang
- Shenzhen Key Laboratory of Marine Bioresource and Eco-Environmental Science, Shenzhen Engineering Laboratory for Marine Algal Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Kun Zhou
- Shenzhen Key Laboratory of Marine Bioresource and Eco-Environmental Science, Shenzhen Engineering Laboratory for Marine Algal Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Lu-Ming Yue
- Shenzhen Key Laboratory of Marine Bioresource and Eco-Environmental Science, Shenzhen Engineering Laboratory for Marine Algal Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Yu Zhang
- Shenzhen Key Laboratory of Marine Bioresource and Eco-Environmental Science, Shenzhen Engineering Laboratory for Marine Algal Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Chang-Yun Wang
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
- Laboratory for Marine Drugs and Bioproducts, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
- Institute of Evolution and Marine Biodiversity, Ocean University of China, Qingdao, China
| | - Kai-Ling Wang
- College of Pharmacy, Institute of Materia Medica, Dali University, Dali, China
| | - Ying Xu
- Shenzhen Key Laboratory of Marine Bioresource and Eco-Environmental Science, Shenzhen Engineering Laboratory for Marine Algal Biotechnology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| |
Collapse
|
14
|
Xia W, Li N, Shan H, Lin Y, Yin F, Yu X, Zhou Z. Gallium Porphyrin and Gallium Nitrate Reduce the High Vancomycin Tolerance of MRSA Biofilms by Promoting Extracellular DNA-Dependent Biofilm Dispersion. ACS Infect Dis 2021; 7:2565-2582. [PMID: 34346692 DOI: 10.1021/acsinfecdis.1c00280] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Biofilms, structured communities of bacterial cells embedded in a self-produced extracellular matrix (ECM) which consists of proteins, polysaccharide intercellular adhesins (PIAs), and extracellular DNA (eDNA), play a key role in clinical infections and are associated with an increased morbidity and mortality by protecting the embedded bacteria against drug and immune response. The high levels of antibiotic tolerance render classical antibiotic therapies impractical for biofilm-related infections. Thus, novel drugs and strategies are required to reduce biofilm tolerance and eliminate biofilm-protected bacteria. Here, we showed that gallium, an iron mimetic metal, can lead to nutritional iron starvation and act as dispersal agent triggering the reconstruction and dispersion of mature methicillin-resistant Staphylococcus aureus (MRSA) biofilms in an eDNA-dependent manner. The extracellular matrix, along with the integral bacteria themselves, establishes the integrated three-dimensional structure of the mature biofilm. The structures and compositions of gallium-treated mature biofilms differed from those of natural or antibiotic-survived mature biofilms but were similar to those of immature biofilms. Similar to immature biofilms, gallium-treated biofilms had lower levels of antibiotic tolerance, and our in vitro tests showed that treatment with gallium agents reduced the antibiotic tolerance of mature MRSA biofilms. Thus, the sequential administration of gallium agents (gallium porphyrin and gallium nitrate) and relatively low concentrations of vancomycin (16 mg/L) effectively eliminated mature MRSA biofilms and eradicated biofilm-enclosed bacteria within 1 week. Our results suggested that gallium agents may represent a potential treatment for refractory biofilm-related infections, such as prosthetic joint infections (PJI) and osteomyelitis, and provide a novel basis for future biofilm treatments based on the disruption of normal biofilm-development processes.
Collapse
Affiliation(s)
- Wenyang Xia
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Niya Li
- Department of Laboratory, Shanghai Sixth People’s Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai 200233, China
| | - Haojie Shan
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Yiwei Lin
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Fuli Yin
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Xiaowei Yu
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Zubin Zhou
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| |
Collapse
|
15
|
Development and Preclinical Evaluation of New Inhaled Lipoglycopeptides for the Treatment of Persistent Pulmonary Methicillin-Resistant Staphylococcus aureus Infections. Antimicrob Agents Chemother 2021; 65:e0031621. [PMID: 33941518 PMCID: PMC8373216 DOI: 10.1128/aac.00316-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Chronic pulmonary methicillin-resistant Staphylococcus aureus (MRSA) disease in cystic fibrosis (CF) has a high probability of recurrence following treatment with standard-of-care antibiotics and represents an area of unmet need associated with reduced life expectancy. We developed a lipoglycopeptide therapy customized for pulmonary delivery that not only demonstrates potent activity against planktonic MRSA, but also against protected colonies of MRSA in biofilms and within cells, the latter of which have been linked to clinical antibiotic failure. A library of next-generation potent lipoglycopeptides was synthesized with an emphasis on attaining superior pharmacokinetics (PK) and pharmacodynamics to similar compounds of their class. Our strategy focused on hydrophobic modification of vancomycin, where ester and amide functionality were included with carbonyl configuration and alkyl length as key variables. Candidates representative of each carbonyl attachment chemistry demonstrated potent activity in vitro, with several compounds being 30 to 60 times more potent than vancomycin. Selected compounds were advanced into in vivo nose-only inhalation PK evaluations in rats, where RV94, a potent lipoglycopeptide that utilizes an inverted amide linker to attach a 10-carbon chain to vancomycin, demonstrated the most favorable lung residence time after inhalation. Further in vitro evaluation of RV94 showed superior activity to vancomycin against an expanded panel of Gram-positive organisms, cellular accumulation and efficacy against intracellular MRSA, and MRSA biofilm killing. Moreover, in vivo efficacy of inhaled nebulized RV94 in a 48 h acute model of pulmonary MRSA (USA300) infection in neutropenic rats demonstrated statistically significant antibacterial activity that was superior to inhaled vancomycin.
Collapse
|
16
|
Mumtaz S, Behera S, Mukhopadhyay K. Lipidated Short Analogue of α-Melanocyte Stimulating Hormone Exerts Bactericidal Activity against the Stationary Phase of Methicillin-Resistant Staphylococcus aureus and Inhibits Biofilm Formation. ACS OMEGA 2020; 5:28425-28440. [PMID: 33195893 PMCID: PMC7658953 DOI: 10.1021/acsomega.0c01462] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 09/04/2020] [Indexed: 05/20/2023]
Abstract
Stationary phase Staphylococcus aureus, especially methicillin-resistant S. aureus (MRSA), has been widely associated with many persistent infections as well as biofilm-associated infections, which are challenging due to their increasing antibiotic resistance. α-Melanocyte stimulating hormone (α-MSH) is an antimicrobial peptide (AMP) with well-established potent activity against S. aureus , but little is known about its antimicrobial efficacy against the stationary phase of the bacteria. We investigated the in vitro activities of two palmitoylated analogues, Pal-α-MSH(6-13) and Pal-α-MSH(11-13), of the C-terminal fragments of α-MSH against biofilm-producing strains of methicillin-sensitive S. aureus (MSSA) and MRSA. While both the peptides demonstrated anti-staphylococcal efficacy, Pal-α-MSH(11-13) emerged as the most effective AMP as palmitoylation led to a remarkable enhancement in its activity against stationary phase bacteria. Similar to α-MSH, both the designed analogues were membrane-active and exhibited improved bacterial membrane depolarization and permeabilization, as further confirmed via electron microscopy studies. Of the two peptides, Pal-α-MSH(11-13) was able to retain its activity in the presence of standard microbiological media, which otherwise is a major limiting factor toward the therapeutic use of α-MSH-based peptides. More importantly, Pal-α-MSH(11-13) was also highly effective in inhibiting the formation of biofilms. Furthermore, it did not lead to resistance development in MRSA cells even upon 18 serial passages at sub-MIC concentrations. These observations support the potential use of Pal-α-MSH(11-13) in the treatment of planktonic as well as sessile S. aureus infections.
Collapse
|
17
|
Chen X, Thomsen TR, Winkler H, Xu Y. Influence of biofilm growth age, media, antibiotic concentration and exposure time on Staphylococcus aureus and Pseudomonas aeruginosa biofilm removal in vitro. BMC Microbiol 2020; 20:264. [PMID: 32831025 PMCID: PMC7444035 DOI: 10.1186/s12866-020-01947-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 08/13/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Biofilm is known to be tolerant towards antibiotics and difficult to eradicate. Numerous studies have reported minimum biofilm eradication concentration (MBEC) values of antibiotics for many known biofilm pathogens. However, the experimental parameters applied in these studies differ considerably, and often the rationale behind the experimental design are not well described. This makes it difficult to compare the findings. To demonstrate the importance of experimental parameters, we investigated the influence of biofilm growth age, antibiotic concentration and treatment duration, and growth media on biofilm eradication. Additionally, OSTEOmycin™, a clinically used antibiotic containing allograft bone product, was tested for antibiofilm efficacy. RESULTS The commonly used Calgary biofilm device was used to grow 24 h and 72 h biofilms of Staphylococcus aureus and Pseudomonas aeruginosa, which were treated with time-dependent vancomycin (up to 3000 mg L- 1) and concentration-dependent tobramycin (up to 80 mg L- 1), respectively. Two common bacteriological growth media, tryptic soy broth (TSB) and cation-adjusted Mueller Hinton broth (CaMHB), were tested. We found for both species that biofilms were more difficult to kill in TSB than in CaMHB. Furthermore, young biofilms (24 h) were easier to eradicate than old biofilms (72 h). In agreement with vancomycin being time-dependent, extension of the vancomycin exposure increased killing of S. aureus biofilms. Tobramycin treatment of 24 h P. aeruginosa biofilms was found concentration-dependent and time-independent, however, increasing killing was indicated for 72 h P. aeruginosa biofilms. Treatment with tobramycin containing OSTEOmycin T™ removed 72 h and 168 h P. aeruginosa biofilms after 1 day treatment, while few 72 h S. aureus biofilms survived after 2 days treatment with vancomycin containing OSTEOmycin V™. CONCLUSIONS This study demonstrated biofilm removal efficacy was influenced by media, biofilm age and antibiotic concentration and treatment duration. It is therefore necessary to taking these parameters into consideration when designing experiments. The results of OSTEOmycin™ products indicated that simple in vitro biofilm test could be used for initial screening of antibiofilm products. For clinical application, a more clinically relevant biofilm model for the specific biofilm infection in question should be developed to guide the amount of antibiotics used for local antibiofilm treatment.
Collapse
Affiliation(s)
- Xiaofeng Chen
- Center for Microbial Communities, Aalborg University, Aalborg East, Denmark
| | - Trine Rolighed Thomsen
- Center for Microbial Communities, Aalborg University, Aalborg East, Denmark
- Life Science Division, Danish Technological Institute, Aarhus, Denmark
| | - Heinz Winkler
- Osteitis Centre, Privatklinik Döbling, Vienna, Austria
| | - Yijuan Xu
- Center for Microbial Communities, Aalborg University, Aalborg East, Denmark.
- Life Science Division, Danish Technological Institute, Aarhus, Denmark.
| |
Collapse
|
18
|
Ballén V, Ratia C, Cepas V, Soto SM. Enterococcus faecalis inhibits Klebsiella pneumoniae growth in polymicrobial biofilms in a glucose-enriched medium. BIOFOULING 2020; 36:846-861. [PMID: 32972252 DOI: 10.1080/08927014.2020.1824272] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 09/03/2020] [Accepted: 09/09/2020] [Indexed: 06/11/2023]
Abstract
Catheter-related urinary tract infections are one of the most common biofilm-associated diseases. Within biofilms, bacteria cooperate, compete, or have neutral interactions. This study aimed to investigate the interactions in polymicrobial biofilms of Klebsiella pneumoniae and Enterococcus faecalis, two of the most common uropathogens. Although K. pneumoniae was the most adherent strain, it could not maintain dominance in the polymicrobial biofilm due to the lactic acid produced by E. faecalis in a glucose-enriched medium. This result was supported by the use of E. faecalis V583 ldh-1/ldh-2 double mutant (non-producer of lactic acid), which did not inhibit the growth of K. pneumoniae. Lyophilized cell-free supernatants obtained from E. faecalis biofilms also showed antimicrobial/anti-biofilm activity against K. pneumoniae. Conversely, there were no significant differences in planktonic polymicrobial cultures. In summary, E. faecalis modifies the pH by lactic acid production in polymicrobial biofilms, which impairs the growth of K. pneumoniae.
Collapse
Affiliation(s)
- Victoria Ballén
- ISGlobal, Barcelona Center for International Health Research (CRESIB), Hospital Clinic - Universitat de Barcelona, Barcelona, Spain
| | - Carlos Ratia
- ISGlobal, Barcelona Center for International Health Research (CRESIB), Hospital Clinic - Universitat de Barcelona, Barcelona, Spain
| | - Virginio Cepas
- ISGlobal, Barcelona Center for International Health Research (CRESIB), Hospital Clinic - Universitat de Barcelona, Barcelona, Spain
| | - Sara M Soto
- ISGlobal, Barcelona Center for International Health Research (CRESIB), Hospital Clinic - Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
19
|
Zheng JX, Tu HP, Sun X, Xu GJ, Chen JW, Deng QW, Yu ZJ, Qu D. In vitro activities of telithromycin against Staphylococcus aureus biofilms compared with azithromycin, clindamycin, vancomycin and daptomycin. J Med Microbiol 2020; 69:120-131. [PMID: 31916929 DOI: 10.1099/jmm.0.001122] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Introduction. Staphylococcus aureus biofilms are difficult to treat and the effect of telithromycin treatment is still unclear.Aim. This study aimed to explore the effect of telithromycin against Staphylococcus aureus biofilms compared with azithromycin, clindamycin, vancomycin and daptomycin.Methodology. Eight methicillin-susceptible and eight methicillin-resistant S. aureus isolates (MSSA and MRSA, respectively) were used for this study. Biofilm biomasses were detected by crystal violet staining and the adherent cells in the established biofilms were quantified by determination of colony-forming units (c.f.u.). The RNA levels of biofilm formation-related genes were determined by RT-qPCR.Results. Telithromycin [8× minimum inhibitory concentration (MIC)] eradicated more established biofilms than azithromycin or clindamycin in the four MSSA isolates, and eliminated the established biofilms of six MRSA isolates more effectively than vancomycin or daptomycin. Telithromycin (8× MIC) killed more adherent cells in the established biofilms than azithromycin or clindamycin in the six MSSA isolates, and killed more adherent cells than vancomycin in all eight MRSA isolates. Daptomycin also showed an excellent effect on the adherent cells of MRSA isolates, with similarresults to telithromycin. The effect of a subinhibitory concentration of telithromycin (1/4× MIC) was significantly superior to that of azithromycin or clindamycin, inhibiting the biofilm formation of six MSSA isolates and seven MRSA isolates more effectively than vancomycin or daptomycin. The RNA levels of agrA, agrC, clfA, icaA and sigB decreased when treated with telithromycin (1/4× MIC).Conclusions. Telithromycin is more effective than azithromycin, clindamycin, vancomycin, or daptomycin against S. aureus biofilms.
Collapse
Affiliation(s)
- Jin-Xin Zheng
- Department of Infectious Diseases and the Key Laboratory of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, PR China.,Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, PR China
| | - Hao-Peng Tu
- Department of Infectious Diseases and the Key Laboratory of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, PR China
| | - Xiang Sun
- Department of Infectious Diseases and the Key Laboratory of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, PR China
| | - Guang-Jian Xu
- Department of Infectious Diseases and the Key Laboratory of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, PR China
| | - Jun-Wen Chen
- Department of Infectious Diseases and the Key Laboratory of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, PR China
| | - Qi-Wen Deng
- Department of Infectious Diseases and the Key Laboratory of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, PR China
| | - Zhi-Jian Yu
- Department of Infectious Diseases and the Key Laboratory of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, PR China
| | - Di Qu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, PR China
| |
Collapse
|
20
|
Britt NS, Hazlett DS, Horvat RT, Liesman RM, Steed ME. Activity of pulmonary vancomycin exposures versus planktonic and biofilm isolates of methicillin-resistant Staphylococcus aureus from cystic fibrosis sputum. Int J Antimicrob Agents 2020; 55:105898. [PMID: 31931147 DOI: 10.1016/j.ijantimicag.2020.105898] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 11/30/2019] [Accepted: 01/04/2020] [Indexed: 11/30/2022]
Abstract
Vancomycin is commonly used to treat methicillin-resistant Staphylococcus aureus (MRSA) infections in patients with cystic fibrosis (CF) lung disease. However, there are limited data to support the in vitro activity of this agent against MRSA isolated from CF sputum. The primary objective of this study was to evaluate the activity of vancomycin at pulmonary concentrations (intravenous and inhaled) against four clinical MRSA CF sputum isolates in planktonic and biofilm time-kill (TK) experiments. Vancomycin minimum inhibitory concentrations (MICs) were determined for these isolates at standard inoculum (SI) (~106 CFU/mL) and high inoculum (HI) (~108 CFU/mL) as well as in biofilms cultivated using physiological medium representing the microenvironment of the CF lung. Vancomycin concentrations of 10, 25, 100 and 275 µg/mL were evaluated in TK experiments against planktonic MRSA at varying inocula and versus biofilm MRSA. Vancomycin MICs increased from 0.5 µg/mL when tested at SI to 8-16 µg/mL at HI. Vancomycin MICs were further increased to 16-32 µg/mL in biofilm studies. In TK experiments, vancomycin displayed bactericidal activity (≥3 log10 killing at 24 h) against 1/4 and 0/4 planktonic MRSA isolates at SI and HI, respectively, whereas vancomycin was bactericidal against 0/4 isolates against MRSA biofilms. Based on these findings, vancomycin monotherapy appears unlikely to eradicate MRSA from the respiratory tract of patients with CF, even at high concentrations similar to those observed with inhaled therapy. Novel vancomycin formulations with enhanced biofilm penetration or combination therapy with other potentially synergistic agents should be explored.
Collapse
Affiliation(s)
- Nicholas S Britt
- Department of Pharmacy Practice, University of Kansas School of Pharmacy, Lawrence, Kansas, USA; Department of Internal Medicine, Division of Infectious Diseases, University of Kansas School of Medicine, Kansas City, Kansas, USA.
| | - Daniel S Hazlett
- Department of Pharmacy Practice, University of Kansas School of Pharmacy, Lawrence, Kansas, USA
| | - Rebecca T Horvat
- Department of Pathology and Laboratory Medicine, University of Kansas School of Medicine, Kansas City, Kansas, USA
| | - Rachael M Liesman
- Department of Pathology and Laboratory Medicine, University of Kansas School of Medicine, Kansas City, Kansas, USA
| | - Molly E Steed
- Department of Pharmacy Practice, University of Kansas School of Pharmacy, Lawrence, Kansas, USA
| |
Collapse
|
21
|
Santos R, Ruza D, Cunha E, Tavares L, Oliveira M. Diabetic foot infections: Application of a nisin-biogel to complement the activity of conventional antibiotics and antiseptics against Staphylococcus aureus biofilms. PLoS One 2019; 14:e0220000. [PMID: 31339915 PMCID: PMC6655664 DOI: 10.1371/journal.pone.0220000] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 07/05/2019] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Diabetic foot infections (DFIs) are a frequent complication of Diabetes mellitus and a major cause of nontraumatic limb amputations. The Gram-positive bacterium Staphylococcus aureus, known for its resilient biofilms and antibiotic resistant profile, is the most frequent DFI pathogen. It is urgent to develop innovative treatments for these infections, being the antimicrobial peptide (AMP) nisin a potential candidate. We have previously proposed the use of a guar gum biogel as a delivery system for nisin. Here, we evaluated the potential of the nisin-biogel to enhance the efficacy of conventional antibiotics and antiseptics against DFIs S. aureus clinical isolates. METHODS A collection of 23 S. aureus strains isolated from DFI patients, including multidrug- and methicillin-resistant strains, was used. The antimicrobial activity of the nisin-biogel was tested alone and in different combinations with the antiseptic chlorhexidine and the antibiotics clindamycin, gentamicin and vancomycin. Isolates' in vitro susceptibility to the different protocols was assessed using broth microdilution methods in order to determine their ability to inhibit and/or eradicate established S. aureus biofilms. Antimicrobials were added to the 96-well plates every 8 h to simulate a typical DFI treatment protocol. Statistical analysis was conducted using RCBD ANOVA in SPSS. RESULTS The nisin-biogel showed a high antibacterial activity against biofilms formed by DFI S. aureus. The combined protocol using nisin-biogel and chlorhexidine presented the highest efficacy in biofilm formation inhibition, significantly higher (p<0.05) than the ones presented by the antibiotics-based protocols tested. Regarding biofilm eradication, there were no significant differences (p>0.05) between the activity of the combination nisin-biogel plus chlorhexidine and the conventional antibiotic-based protocols. CONCLUSIONS Results provide a valuable contribution for the development of complementary strategies to conventional antibiotics protocols. A combined protocol including chlorhexidine and nisin-biogel could be potentially applied in medical centres, contributing for the reduction of antibiotic administration, selection pressure on DFI pathogens and resistance strains dissemination.
Collapse
Affiliation(s)
- Raquel Santos
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisboa, Portugal
| | - Diana Ruza
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisboa, Portugal
| | - Eva Cunha
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisboa, Portugal
| | - Luís Tavares
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisboa, Portugal
| | - Manuela Oliveira
- CIISA-Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
22
|
Luther MK, Mermel LA, LaPlante KL. Comparison of linezolid and vancomycin lock solutions with and without heparin against biofilm-producing bacteria. Am J Health Syst Pharm 2019; 74:e193-e201. [PMID: 28438824 DOI: 10.2146/ajhp150804] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
PURPOSE The activity of linezolid and vancomycin lock solutions against biofilm-producing strains of Staphylococcus aureus, S. epidermidis, and Enterococcus faecalis was studied. METHODS Two strains each of methicillin-susceptible S. aureus (MSSA), methicillin-resistant S. aureus (MRSA), and S. epidermidis, and 1 strain of vancomycin-susceptible E. faecalis and vancomycin-resistant E. faecalis were tested against vancomycin and linezolid to assess prevention of biofilm formation and eradication of these pathogens within a formed biofilm. Activity was also tested in a 72-hour in vitro central venous catheter (CVC) model. After 24 hours of biofilm growth in a CVC, a lock solution containing vancomycin (2 or 5 mg/mL) or linezolid (1 or 2 mg/mL) alone or in combination with heparin sodium (5,000 units/mL with benzyl alcohol 0.45%) was instilled and incubated at 35 °C for 72 hr. Heparin and 0.9% sodium chloride injection were also tested. RESULTS Linezolid and vancomycin prevented biofilm formation below the minimum inhibitory concentration for 88% and 25% of isolates tested, respectively. The addition of preservative-containing heparin decreased the activity of vancomycin and linezolid lock solutions against all strains. Vancomycin 2- and 5-mg/mL lock solutions had the most activity against MSSA and E. faecalis strains (p < 0.01). Linezolid 2 mg/mL was the most active lock solution against the MRSA strains tested (p < 0.01). There were no significant differences in vancomycin or linezolid lock solution activity against S. epidermidis. CONCLUSION Heparin reduced activity of vancomycin and linezolid lock solutions against S. aureus, S. epidermidis, and E. faecalis biofilms. While linezolid or vancomycin lock solution reduced overall biofilm burden, it did not completely eradicate the bacteria at tested concentrations.
Collapse
Affiliation(s)
- Megan K Luther
- Rhode Island Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI.,Department of Pharmacy Practice, University of Rhode Island, Kingston, RI
| | - Leonard A Mermel
- Department of Medicine, Warren Alpert Medical School, Brown University, Providence, RI.,Division of Infectious Diseases, Rhode Island Hospital, Providence, RI
| | - Kerry L LaPlante
- Department of Pharmacy Practice, University of Rhode Island, Kingston, RI .,Rhode Island Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI.
| |
Collapse
|
23
|
Eco-friendly synthesized spherical ZnO materials: Effect of the core-shell to solid morphology transition on antimicrobial activity. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 97:438-450. [DOI: 10.1016/j.msec.2018.12.063] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 11/11/2018] [Accepted: 12/18/2018] [Indexed: 11/24/2022]
|
24
|
Beganovic M, Luther MK, Daffinee KE, LaPlante KL. Biofilm prevention concentrations (BPC) of minocycline compared to polymyxin B, meropenem, and amikacin against Acinetobacter baumannii. Diagn Microbiol Infect Dis 2019; 94:223-226. [PMID: 30827805 DOI: 10.1016/j.diagmicrobio.2019.01.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 12/03/2018] [Accepted: 01/21/2019] [Indexed: 11/29/2022]
Abstract
Infections caused by Acinetobacter baumannii are difficult to treat as they are often multidrug resistant (MDR) and frequently form biofilms. We investigated the activities of minocycline, polymyxin B, meropenem, and amikacin against diverse Acinetobacter baumannii strains with biofilm formation classified as weak versus moderate/strong. At clinically achievable concentrations, minocycline prevented biofilm formation for 96% of isolates versus 54% for polymyxin B, 29% for meropenem and 29% for amikacin. Minocycline and polymyxin B demonstrated highest in vitro activity against A. baumannii and prevented biofilm formation for a majority of isolates.
Collapse
Affiliation(s)
- Maya Beganovic
- Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI, United States; College of Pharmacy, University of Rhode Island, Kingston, RI, United States
| | - Megan K Luther
- Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI, United States; College of Pharmacy, University of Rhode Island, Kingston, RI, United States; Center of Innovation in Long-Term Support Services, Providence Veterans Affairs Medical Center, Providence, RI, United States
| | - Kathryn E Daffinee
- Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI, United States
| | - Kerry L LaPlante
- Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI, United States; College of Pharmacy, University of Rhode Island, Kingston, RI, United States; Center of Innovation in Long-Term Support Services, Providence Veterans Affairs Medical Center, Providence, RI, United States; Warren Alpert Medical School of Brown University, Division of Infectious Diseases, Providence, RI.
| |
Collapse
|
25
|
Suresh MK, Biswas R, Biswas L. An update on recent developments in the prevention and treatment of Staphylococcus aureus biofilms. Int J Med Microbiol 2019; 309:1-12. [DOI: 10.1016/j.ijmm.2018.11.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 11/19/2018] [Accepted: 11/26/2018] [Indexed: 12/17/2022] Open
|
26
|
Visinescu D, Hussien MD, Moreno JC, Negrea R, Birjega R, Somacescu S, Ene CD, Chifiriuc MC, Popa M, Stan MS, Carp O. Zinc Oxide Spherical-Shaped Nanostructures: Investigation of Surface Reactivity and Interactions with Microbial and Mammalian Cells. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2018; 34:13638-13651. [PMID: 30340439 DOI: 10.1021/acs.langmuir.8b02528] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Two ZnO materials of spherical hierarchical morphologies, with hollow (ZnOHS) and solid cores (ZnOSS), were obtained through the hydrolysis of zinc acetylacetonate in 1,4-butanediol. The nature of the defects and surface reactivity for the two ZnO materials were investigated through photoluminescence, X-ray photoelectron spectroscopy, and electron paramagnetic resonance (EPR) spectroscopy proving the coexistence of shallow and deep defects and, also, the presence of polyol byproducts adsorbed on the outer layers of the ZnO samples. The EPR spectroscopy coupled with the spin-trapping technique showed that the surface of the ZnO samples generates reactive oxygen species (ROS) like hydroxyl (•OH) and singlet oxygen (1O2) as well as carbon-centered radicals. The ZnO materials exhibited a wide spectrum of antimicrobial activity, being active against Gram-positive, Gram-negative, and fungi strains, both in planktonic and, more importantly, adherent growth states. The decrease of antimicrobial efficiency in the presence of a ROS scavenger (mannitol) and the decrease of the cell viability with the ROS level suggest that one of the mechanisms that governs both the antimicrobial and cytotoxic activities on human liver cells is ROS-mediated. However, at active antimicrobial concentrations, the biocompatibility of the tested materials is very good.
Collapse
Affiliation(s)
- Diana Visinescu
- "Ilie Murgulescu" Institute of Physical Chemistry , Romanian Academy , 202 Splaiul Independentei , 060021 Bucharest , Romania
| | | | - Jose Calderon Moreno
- "Ilie Murgulescu" Institute of Physical Chemistry , Romanian Academy , 202 Splaiul Independentei , 060021 Bucharest , Romania
| | - Raluca Negrea
- National Institute of Materials Physics , Atomistilor 105bis , 77125 Magurele , Ilfov , Romania
| | - Ruxandra Birjega
- National Institute for Lasers, Plasma and Radiation Physics , 409 Atomistilor , P.O. Box MG-36, 077125 Bucharest , Romania
| | - Simona Somacescu
- "Ilie Murgulescu" Institute of Physical Chemistry , Romanian Academy , 202 Splaiul Independentei , 060021 Bucharest , Romania
| | - Cristian D Ene
- "Ilie Murgulescu" Institute of Physical Chemistry , Romanian Academy , 202 Splaiul Independentei , 060021 Bucharest , Romania
| | | | | | | | - Oana Carp
- "Ilie Murgulescu" Institute of Physical Chemistry , Romanian Academy , 202 Splaiul Independentei , 060021 Bucharest , Romania
| |
Collapse
|
27
|
Dhanda G, Sarkar P, Samaddar S, Haldar J. Battle against Vancomycin-Resistant Bacteria: Recent Developments in Chemical Strategies. J Med Chem 2018; 62:3184-3205. [DOI: 10.1021/acs.jmedchem.8b01093] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Geetika Dhanda
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| | - Paramita Sarkar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| | - Sandip Samaddar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| | - Jayanta Haldar
- Antimicrobial Research Laboratory, New Chemistry Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Jakkur, Bengaluru 560064, Karnataka, India
| |
Collapse
|
28
|
Bhandari V, Chakraborty S, Brahma U, Sharma P. Identification of Anti-staphylococcal and Anti-biofilm Compounds by Repurposing the Medicines for Malaria Venture Pathogen Box. Front Cell Infect Microbiol 2018; 8:365. [PMID: 30406042 PMCID: PMC6206229 DOI: 10.3389/fcimb.2018.00365] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 10/01/2018] [Indexed: 11/13/2022] Open
Abstract
There has been an alarming increase in infections caused by antimicrobial-resistant pathogens. These infections are responsible for more than half a million deaths globally each year. Staphylococcus aureus is one of the deadliest bacterial pathogen responsible for nosocomial and community acquired infections. The open-access Pathogen Box (PBox) provides a potential platform to identify new treatment options against antibiotic-resistant bacteria by repurposing it. In this study, we have screened the PBox library comprised of ~400 compounds to identify novel anti-staphylococcal compounds. in vitro antimicrobial screening using S. aureus isolates, ATCC 29213 (methicillin-sensitive) and ATCC 700699 (methicillin-resistant) revealed 13 compounds which showed highly potent antibacterial activity against both planktonic and biofilm state. The 13 compounds were not found cytotoxic to mouse macrophage cell line, RAW264.7. Out of the 13 compounds, only MMV687251 and MMV676477 revealed structural similarity with vancomycin by comparing their atomic pair fingerprints using Tanimoto coefficient method. The structural similarities may indicate similar mode of action like vancomycin for the two compounds. Our result showed that PBox compounds offer a promising lead for the development of new anti-staphylococcal treatment options.
Collapse
Affiliation(s)
| | | | - Umarani Brahma
- National Institute of Animal Biotechnology, Hyderabad, India
| | - Paresh Sharma
- National Institute of Animal Biotechnology, Hyderabad, India
| |
Collapse
|
29
|
Jahanbakhsh S, Singh NB, Yim J, Rose WE, Rybak MJ. Evaluation of Telavancin Alone and Combined with Ceftaroline or Rifampin against Methicillin-Resistant Staphylococcus aureus in an In Vitro Biofilm Model. Antimicrob Agents Chemother 2018; 62:e00567-18. [PMID: 29784849 PMCID: PMC6105779 DOI: 10.1128/aac.00567-18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 05/15/2018] [Indexed: 12/11/2022] Open
Abstract
Infections caused by biofilm-producing methicillin-resistant Staphylococcus aureus (MRSA) bacteria are challenging due to increasing antibiotic resistance. Synergistic activities of lipopeptides and lipoglycopeptides with β-lactams have been demonstrated for MRSA, but little is known about biofilm-embedded organisms. Our objective was to evaluate two telavancin (TLV) dosage regimens (7.5 mg/kg of body weight and 10 mg/kg every 24 h [q24h]) alone and in combination with ceftaroline (CPT) (600 mg every 8 h [q8h]) or rifampin (RIF) (450 mg every 12 h [q12h]) against two biofilm-producing MRSA strains (494 and N315). Pharmacokinetic/pharmacodynamic CDC biofilm reactor models with polyurethane coupons were used to evaluate the efficacies of the antibiotic combinations over 72 h. Overall, there were no significant differences observed between the two TLV dosing regimens either alone or in combination with RIF or CPT against these strains. Both TLV dosing regimens and CPT alone demonstrated killing but did not reach bactericidal reduction at 72 h. However, both TLV regimens in combination with RIF demonstrated enhanced activity against both strains, with a rapid decrease in CFU/ml at 4 h that was bactericidal and maintained over the 72-h experiment (-Δ3.75 log10 CFU/ml from baseline; P < 0.0001). Of interest, no enhanced activity was observed for TLV combined with CPT. No development of resistance was observed in any of the combination models. However, resistance to RIF developed as early as 24 h, with MIC values exceeding 32 mg/liter. Our results show that TLV plus RIF displayed therapeutic improvement against biofilm-producing MRSA. These results suggest that TLV at 7.5 and 10 mg/kg q24h are equally effective in eradicating biofilm-associated MRSA strains in vitro.
Collapse
Affiliation(s)
- Seyedehameneh Jahanbakhsh
- Anti-Infective Research Laboratory, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Nivedita B Singh
- Anti-Infective Research Laboratory, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Juwon Yim
- Anti-Infective Research Laboratory, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Warren E Rose
- School of Pharmacy and Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Michael J Rybak
- Anti-Infective Research Laboratory, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
- School of Medicine, Wayne State University, Detroit, Michigan, USA
| |
Collapse
|
30
|
Abstract
The molecular and clinical factors associated with biofilm-forming methicillin-resistant Staphylococcus aureus (MRSA) are incompletely understood. Biofilm production for 182 MRSA isolates obtained from clinical culture sites (2004 to 2013) was quantified. Microbiological toxins, pigmentation, and genotypes were evaluated, and patient demographics were collected. Logistic regression was used to quantify the effect of strong biofilm production (versus weak biofilm production) on clinical outcomes and independent predictors of a strong biofilm. Of the isolates evaluated, 25.8% (47/182) produced strong biofilms and 40.7% (74/182) produced weak biofilms. Strong biofilm-producing isolates were more likely to be from multilocus sequence typing (MLST) clonal complex 8 (CC8) (34.0% versus 14.9%; P = 0.01) but less likely to be from MLST CC5 (48.9% versus 73.0%; P = 0.007). Predictors for strong biofilms were spa type t008 (adjusted odds ratio [aOR], 4.54; 95% confidence interval [CI], 1.21 to 17.1) and receipt of chemotherapy or immunosuppressants in the previous 90 days (aOR, 33.6; 95% CI, 1.68 to 673). Conversely, patients with high serum creatinine concentrations (aOR, 0.33; 95% CI, 0.15 to 0.72) or who previously received vancomycin (aOR, 0.03; 95% CI, 0.002 to 0.39) were less likely to harbor strong biofilm-producing MRSA. Beta-toxin-producing isolates (aOR, 0.31; 95% CI, 0.11 to 0.89) and isolates with spa type t895 (aOR, 0.02 95% CI, <0.001 to 0.47) were less likely to produce strong biofilms. Patient outcomes also varied between the two groups. Specifically, patients with strong biofilm-forming MRSA were significantly more likely to be readmitted within 90 days (aOR, 5.43; 95% CI, 1.69 to 17.4) but tended to have decreased 90-day mortality (aOR, 0.36; 95% CI, 0.12 to 1.06). Patients that harbored t008 and received immunosuppressants were more likely to have strong biofilm-producing MRSA isolates. Clinically, patients with strong biofilm-forming MRSA were less likely to die at 90 days but five times more likely to be readmitted.
Collapse
|
31
|
Yan Q, Karau MJ, Patel R. In vitro activity of oritavancin against planktonic and biofilm states of vancomycin-susceptible and vancomycin-resistant enterococci. Diagn Microbiol Infect Dis 2018; 91:348-350. [PMID: 29678300 DOI: 10.1016/j.diagmicrobio.2018.03.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/08/2018] [Accepted: 03/11/2018] [Indexed: 11/20/2022]
Abstract
We tested the in vitro activity of oritavancin against 60 vancomycin-susceptible enterococci (VSE) and 27 vancomycin-resistant enterococci (VRE). The oritavancin MIC ranged from ≤0.002 to 0.5μg/mL; the minimum biofilm bactericidal concentration ranged from ≤0.002 to 2μg/mL. Oritavancin has promising in vitro activity against VSE and VRE in both planktonic and biofilm states.
Collapse
Affiliation(s)
- Qun Yan
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN; Department of Clinical Laboratory, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Melissa J Karau
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN; Division of Infectious Diseases, Department of Medicine, Mayo Clinic, Rochester, MN.
| |
Collapse
|
32
|
Sarkar P, Yarlagadda V, Ghosh C, Haldar J. A review on cell wall synthesis inhibitors with an emphasis on glycopeptide antibiotics. MEDCHEMCOMM 2017; 8:516-533. [PMID: 30108769 DOI: 10.1039/c6md00585c] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/18/2017] [Indexed: 01/24/2023]
Abstract
Cell wall biosynthesis inhibitors (CBIs) have historically been one of the most effective classes of antibiotics. They are the most extensively used class of antibiotics and their importance is exemplified by the β-lactams and glycopeptide antibiotics. However, this class of antibiotics has not received impunity from resistance development. In the wake of this predicament, this review presents the progress of CBIs, especially glycopeptide derivatives as antibiotics to confront antibacterial resistance. The various strategies used for the development of CBIs, their clinical status and possible directions in which this field can evolve have also been discussed.
Collapse
Affiliation(s)
- Paramita Sarkar
- Chemical Biology and Medicinal Chemistry Laboratory , New Chemistry Unit , Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) , Jakkur , Bengaluru 5600064 , Karnataka , India .
| | - Venkateswarlu Yarlagadda
- Chemical Biology and Medicinal Chemistry Laboratory , New Chemistry Unit , Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) , Jakkur , Bengaluru 5600064 , Karnataka , India .
| | - Chandradhish Ghosh
- Chemical Biology and Medicinal Chemistry Laboratory , New Chemistry Unit , Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) , Jakkur , Bengaluru 5600064 , Karnataka , India .
| | - Jayanta Haldar
- Chemical Biology and Medicinal Chemistry Laboratory , New Chemistry Unit , Jawaharlal Nehru Centre for Advanced Scientific Research (JNCASR) , Jakkur , Bengaluru 5600064 , Karnataka , India .
| |
Collapse
|
33
|
Streptokinase Treatment Reverses Biofilm-Associated Antibiotic Resistance in Staphylococcus aureus. Microorganisms 2016; 4:microorganisms4030036. [PMID: 27681928 PMCID: PMC5039596 DOI: 10.3390/microorganisms4030036] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 09/07/2016] [Accepted: 09/14/2016] [Indexed: 11/24/2022] Open
Abstract
Biofilms formed by Staphylococcus aureus is a serious complication to the use of medical implants. A central part of the pathogenesis relies on S. aureus’ ability to adhere to host extracellular matrix proteins, which adsorb to medical implants and stimulate biofilm formation. Being coagulase positive, S. aureus furthermore induces formation of fibrin fibers from fibrinogen in the blood. Consequently, we hypothesized that fibrin is a key component of the extracellular matrix of S. aureus biofilms under in vivo conditions, and that the recalcitrance of biofilm infections can be overcome by combining antibiotic treatment with a fibrinolytic drug. We quantified S. aureus USA300 biofilms grown on peg-lids in brain heart infusion (BHI) broth with 0%–50% human plasma. Young (2 h) and mature (24 h) biofilms were then treated with streptokinase to determine if this lead to dispersal. Then, the minimal biofilm eradication concentration (MBEC) of 24 h old biofilms was measured for vancomycin and daptomycin alone or in combination with 10 µg/mL rifampicin in the presence or absence of streptokinase in the antibiotic treatment step. Finally, biofilms were visualized by confocal laser scanning microscopy. Addition of human plasma stimulated biofilm formation in BHI in a dose-dependent manner, and biofilms could be partially dispersed by streptokinase. The biofilms could be eradicated with physiologically relevant concentrations of streptokinase in combination with rifampicin and vancomycin or daptomycin, which are commonly used antibiotics for treatment of S. aureus infections. Fibronolytic drugs have been used to treat thromboembolic events for decades, and our findings suggest that their use against biofilm infections has the potential to improve the efficacy of antibiotics in treatment of S. aureus biofilm infections.
Collapse
|
34
|
da Costa Luciano C, Olson N, DeGagne P, Franca R, Tipple AFV, Alfa M. A new buildup biofilm model that mimics accumulation of material in flexible endoscope channels. J Microbiol Methods 2016; 127:224-229. [DOI: 10.1016/j.mimet.2016.06.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 06/20/2016] [Accepted: 06/20/2016] [Indexed: 01/22/2023]
|
35
|
Liapikou A, Dimakou K, Toumbis M. Telavancin in the treatment of Staphylococcus aureus hospital-acquired and ventilator-associated pneumonia: clinical evidence and experience. Ther Adv Respir Dis 2016; 10:368-78. [PMID: 27340253 DOI: 10.1177/1753465816651594] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Telavancin (TLV) is a lipoglycopeptide derivative of vancomycin (VAN), which has activity against Gram-positive aerobic bacteria, and is especially effective against methicillin-resistant Staphylococcus aureus (MRSA) and Gram-positive bacteria resistant to VAN. Comparative clinical studies of TLV have demonstrated noninferiority compared with VAN in the treatment of hospital-acquired Gram-positive pneumonia, with high cure rates for TLV-treated patients with monomicrobial S. aureus infection, including isolates with reduced VAN susceptibility. The results based on the patients' clinical response were supported by supplemental post-hoc analyses of 28-day mortality. In Europe and the USA, TLV is approved as a useful alternative for patients with difficult-to-treat, hospital-acquired MRSA pneumonia when there are very few alternatives. The present article reviews TLV's pharmacological characteristics and clinical efficacy resulting from clinical trials giving a detailed picture of its properties and position in the management of hospital-acquired pneumonia.
Collapse
|
36
|
Mottola C, Matias CS, Mendes JJ, Melo-Cristino J, Tavares L, Cavaco-Silva P, Oliveira M. Susceptibility patterns of Staphylococcus aureus biofilms in diabetic foot infections. BMC Microbiol 2016; 16:119. [PMID: 27339028 PMCID: PMC4918071 DOI: 10.1186/s12866-016-0737-0] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 06/08/2016] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Foot infections are a major cause of morbidity in people with diabetes and the most common cause of diabetes-related hospitalization and lower extremity amputation. Staphylococcus aureus is by far the most frequent species isolated from these infections. In particular, methicillin-resistant S. aureus (MRSA) has emerged as a major clinical and epidemiological problem in hospitals. MRSA strains have the ability to be resistant to most β-lactam antibiotics, but also to a wide range of other antimicrobials, making infections difficult to manage and very costly to treat. To date, there are two fifth-generation cephalosporins generally efficacious against MRSA, ceftaroline and ceftobripole, sharing a similar spectrum. Biofilm formation is one of the most important virulence traits of S. aureus. Biofilm growth plays an important role during infection by providing defence against several antagonistic mechanisms. In this study, we analysed the antimicrobial susceptibility patterns of biofilm-producing S. aureus strains isolated from diabetic foot infections. The antibiotic minimum inhibitory concentration (MIC) was determined for ten antimicrobial compounds, along with the minimum biofilm inhibitory concentration (MBIC) and minimum biofilm eradication concentration (MBEC), followed by PCR identification of genetic determinants of biofilm production and antimicrobial resistance. RESULTS Results demonstrate that very high concentrations of the most used antibiotics in treating diabetic foot infections (DFI) are required to inhibit S. aureus biofilms in vitro, which may explain why monotherapy with these agents frequently fails to eradicate biofilm infections. In fact, biofilms were resistant to antibiotics at concentrations 10-1000 times greater than the ones required to kill free-living or planktonic cells. The only antibiotics able to inhibit biofilm eradication on 50 % of isolates were ceftaroline and gentamicin. CONCLUSIONS The results suggest that the antibiotic susceptibility patterns cannot be applied to biofilm established infections. Selection of antimicrobial therapy is a critical step in DFI and should aim at overcoming biofilm disease in order to optimize the outcomes of this complex pathology.
Collapse
Affiliation(s)
- Carla Mottola
- />Centro de Investigação Interdisciplinar em Sanidade Animal (CIISA), Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal
| | - Carina S. Matias
- />Centro de Investigação Interdisciplinar em Sanidade Animal (CIISA), Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal
| | - João J. Mendes
- />Departamento de Medicina Interna, Hospital de Santa Marta/Centro Hospitalar de Lisboa Central, EPE, Lisbon, Portugal
| | - José Melo-Cristino
- />Faculdade de Medicina, Universidade de Lisboa, Instituto de Microbiologia, Lisbon, Portugal
| | - Luís Tavares
- />Centro de Investigação Interdisciplinar em Sanidade Animal (CIISA), Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal
| | - Patrícia Cavaco-Silva
- />TechnoPhage, S.A., Lisbon, Portugal
- />Centro de Investigação Interdisciplinar Egas Moniz (CiiEM), Instituto Superior de Ciências da Saúde Egas Moniz, Monte de Caparica, Portugal
| | - Manuela Oliveira
- />Centro de Investigação Interdisciplinar em Sanidade Animal (CIISA), Faculdade de Medicina Veterinária, Universidade de Lisboa, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal
| |
Collapse
|
37
|
Adamantia L, Antoni T. Pharmacodynamics, pharmacokinetics and clinical efficacy of telavancin in the treatment of pneumonia. Expert Opin Drug Metab Toxicol 2016; 12:803-12. [DOI: 10.1080/17425255.2016.1187599] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Affiliation(s)
| | - Torres Antoni
- Department of Pneumology, Hospital Clinic of Barcelona, Barcelona, Spain
| |
Collapse
|
38
|
Siala W, Van Bambeke F, Taresco V, Piozzi A, Francolini I. Synergistic activity between an antimicrobial polyacrylamide and daptomycin versusStaphylococcus aureusbiofilm. Pathog Dis 2016; 74:ftw042. [DOI: 10.1093/femspd/ftw042] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2016] [Indexed: 11/13/2022] Open
|
39
|
Revest M, Jacqueline C, Boudjemaa R, Caillon J, Le Mabecque V, Breteche A, Steenkeste K, Tattevin P, Potel G, Michelet C, Fontaine-Aupart MP, Boutoille D. New in vitro and in vivo models to evaluate antibiotic efficacy in Staphylococcus aureus prosthetic vascular graft infection. J Antimicrob Chemother 2016; 71:1291-9. [PMID: 26851611 DOI: 10.1093/jac/dkv496] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 12/19/2015] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE Prosthetic vascular graft infection (PVGI) is an emerging disease, mostly caused by staphylococci, with limited data regarding efficacy of current antistaphylococcal agents. We aimed to assess the efficacy of different antibiotic regimens. METHODS Six different strains of MSSA and MRSA were used. We compared results of minimal biofilm inhibitory and eradicating concentrations (MBICs and MBECs) obtained with a Calgary Biofilm Pin Lid Device (CBPD) with those yielded by an original Dacron(®)-related minimal inhibitory and eradicating concentration measure model. We then used a murine model of Staphylococcus aureus vascular prosthetic material infection to evaluate efficacy of different antibiotic regimens: vancomycin and daptomycin combined or not with rifampicin for MRSA and the same groups with cloxacillin and cloxacillin combined with rifampicin for MSSA. RESULTS We demonstrated that classical measures of MBICs and MBECs obtained with a CPBD could overestimate the decrease in antibiotic susceptibility in material-related infections and that the nature of the support used might influence the measure of biofilm susceptibility, since results yielded by our Dacron(®)-related minimal eradicating assay were lower than those found with a plastic device. In our in vivo model, we showed that daptomycin was significantly more bactericidal than comparators for some strains of MRSA or MSSA but not for all. For the majority of strains, it was as efficient as comparators. The addition of rifampicin to daptomycin did not enhance daptomycin efficacy. CONCLUSIONS Despite the heterogeneity of results according to bacterial strains, these innovative models represent an option to better evaluate the in vitro efficacy of antibiotics on Dacron(®)-related biofilm S. aureus infections, and to screen different antibiotic regimens in a mouse model of PVGIs.
Collapse
Affiliation(s)
- M Revest
- Université Nantes, Faculté Médecine EA3826 Nantes, France CHU Rennes Infectious Diseases and Intensive Care Unit, Pontchaillou Hospital, 35033 Rennes Cedex, France CIC Inserm 1414, Rennes 1 University, Pontchaillou Hospital, 35033 Rennes Cedex, France
| | - C Jacqueline
- Université Nantes, Faculté Médecine EA3826 Nantes, France
| | - R Boudjemaa
- Institut des Sciences Moléculaires Orsay, CNRS, Université Paris-Sud, 91405 Orsay, France
| | - J Caillon
- Université Nantes, Faculté Médecine EA3826 Nantes, France
| | - V Le Mabecque
- Université Nantes, Faculté Médecine EA3826 Nantes, France
| | - A Breteche
- Université Nantes, Faculté Médecine EA3826 Nantes, France
| | - K Steenkeste
- Institut des Sciences Moléculaires Orsay, CNRS, Université Paris-Sud, 91405 Orsay, France
| | - P Tattevin
- CHU Rennes Infectious Diseases and Intensive Care Unit, Pontchaillou Hospital, 35033 Rennes Cedex, France CIC Inserm 1414, Rennes 1 University, Pontchaillou Hospital, 35033 Rennes Cedex, France
| | - G Potel
- Université Nantes, Faculté Médecine EA3826 Nantes, France
| | - C Michelet
- CHU Rennes Infectious Diseases and Intensive Care Unit, Pontchaillou Hospital, 35033 Rennes Cedex, France CIC Inserm 1414, Rennes 1 University, Pontchaillou Hospital, 35033 Rennes Cedex, France
| | - M P Fontaine-Aupart
- Institut des Sciences Moléculaires Orsay, CNRS, Université Paris-Sud, 91405 Orsay, France
| | - D Boutoille
- Université Nantes, Faculté Médecine EA3826 Nantes, France CHU Nantes, Infectious Diseases Unit, Hôtel Dieu, Nantes, France
| |
Collapse
|
40
|
Van Bambeke F. Lipoglycopeptide Antibacterial Agents in Gram-Positive Infections: A Comparative Review. Drugs 2015; 75:2073-95. [DOI: 10.1007/s40265-015-0505-8] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
41
|
Wang X, Leung AW, Hua H, Xu C, Ip M. Sonodynamic action of hypocrellin B on biofilm-producing Staphylococcus epidermidis in planktonic condition. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2015; 138:2548-2553. [PMID: 26520337 DOI: 10.1121/1.4932014] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Staphylococcus epidermidis is an opportunistic pathogen causing biofilm-associated infections. To investigate sonodynamic action of hypocrellin B on biofilm-producing Staphylococcus epidermidis in planktonic culture, a biofilm-producing strain Staphylococcus epidermidis (ATCC 35984) was incubated with hypocrellin B and then exposed to ultrasound at intensity (ISATA) of 1.56 W/cm(2) with a frequency of 1 MHz in continuous mode for 5 min. After sonodynamic treatment of hypocrellin B, the bacterial growth was measured using the colony counting method. Bacterial membrane integrity was investigated using a flow cytometry with propidium iodide staining. Intracellular reactive oxygen species (ROS) level was measured using a flow cytometry with DCFH-DA staining. The results showed that sonodynamic action of hypocrellin B significantly induced survival reduction of Staphylococcus epidermidis in a hypocrellin B dose-dependent manner, and a 4-log reduction was observed after the combined treatment of hypcorellin B (40 μM) and ultrasound sonication with the intensity of 1.56 W/cm(2) for 5 min. Bacterial membrane integrity was notably damaged and the level of intracellular ROS level was remarkably increased after sonodynamic treatment. The findings demonstrated that sonodynamic action of hypocrellin B had significant antibacterial activity on biofilm-producing Staphylococcus epidermidis in planktonic condition probably through increasing intracellular ROS level to cause damage to bacterial membrane integrity.
Collapse
Affiliation(s)
- Xinna Wang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Albert Wingnang Leung
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Heyu Hua
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Chuanshan Xu
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Margaret Ip
- Department of Microbiology, Faculty of Medicine, The Prince of Wales Hospital, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| |
Collapse
|
42
|
|
43
|
Chan C, Hardin TC, Smart JI. A review of telavancin activity in in vitro biofilms and animal models of biofilm-associated infections. Future Microbiol 2015; 10:1325-38. [DOI: 10.2217/fmb.15.53] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
ABSTRACT Tissue- and device-associated biofilm infections are important medical problems. These infections are difficult to treat due to a high-level of tolerance to antibiotics. Telavancin has been studied in several in vitro biofilm models and has demonstrated efficacy against staphylococcal and enterococcal-associated biofilm infections, including those formed by methicillin-resistant Staphylococcus aureus. Telavancin was effective against the difficult-to-treat vancomycin- and glycopeptide-intermediate strains of S. aureus in these models. Furthermore, the efficacy of telavancin has been evaluated in several biofilm-related in vivo models, including osteomyelitis, endocarditis and device-associated infections in rabbits. Overall, telavancin exhibited similar or greater efficacy than vancomycin and other comparators in these animal models and maintained activity against vancomycin-intermediate and daptomycin nonsusceptible strains of S. aureus.
Collapse
Affiliation(s)
- Cynthia Chan
- Theravance Biopharma US, Inc. 901 Gateway Blvd, South San Francisco, CA 94080, USA
| | - Thomas C Hardin
- Theravance Biopharma US, Inc. 901 Gateway Blvd, South San Francisco, CA 94080, USA
| | - Jennifer I Smart
- Theravance Biopharma US, Inc. 901 Gateway Blvd, South San Francisco, CA 94080, USA
| |
Collapse
|
44
|
Luther MK, Bilida S, Mermel LA, LaPlante KL. Ethanol and Isopropyl Alcohol Exposure Increases Biofilm Formation in Staphylococcus aureus and Staphylococcus epidermidis. Infect Dis Ther 2015; 4:219-26. [PMID: 25935134 PMCID: PMC4471055 DOI: 10.1007/s40121-015-0065-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Indexed: 11/27/2022] Open
Abstract
Introduction Alcohols, including ethanol and isopropyl alcohol, are used in clinical practice for disinfection and infection prevention. Recent studies, however, demonstrate that alcohols may enhance biofilm production in Staphylococci. Methods We quantified biofilm formation in the presence of ethanol and isopropyl alcohol in six different, well-characterized strains of Staphylococcus epidermidis and Staphylococcus aureus. After 24 h of biofilm development, each strain was exposed to normal saline (NS), ethanol, or isopropyl alcohol (40%, 60%, 80% and 95%) for additional 24 h incubation. Adherent biofilms were stained and optical density was determined. Viability of strains was also determined after alcohol exposure. Results Ethanol increased biofilm formation in all six strains compared to normal saline (p < 0.05). There was increased biofilm formation with increasing ethanol concentration. Isopropyl alcohol also increased biofilm formation with increasing alcohol concentration in all six strains (p < 0.01 vs NS). The slime-negative, chemical mutant strain of S. epidermidis increased biofilm formation after exposure to both alcohols, likely reverting back its primary phenotype through modulation of the intercellular adhesin repressor. All strains demonstrated viability after exposure to each alcohol concentration, though viability was decreased. Conclusion Ethanol and isopropyl alcohol exposure increases biofilm formation of S. aureus and S. epidermidis at concentrations used in clinical settings. Ethanol and isopropyl alcohol did not eradicate viable Staphylococci from formed biofilm. Electronic supplementary material The online version of this article (doi:10.1007/s40121-015-0065-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Megan K. Luther
- Department of Pharmacy Practice, University of Rhode Island, 7 Greenhouse Road, Kingston, RI 02881 USA
- Rhode Island Infectious Diseases (RIID) Research Program Laboratory, Veterans Affairs Medical Center, Providence, RI USA
| | | | - Leonard A. Mermel
- Department of Pharmacy Practice, University of Rhode Island, 7 Greenhouse Road, Kingston, RI 02881 USA
- Warren Alpert Medical School of Brown University, Providence, RI USA
- Rhode Island Hospital, Providence, RI USA
| | - Kerry L. LaPlante
- Department of Pharmacy Practice, University of Rhode Island, 7 Greenhouse Road, Kingston, RI 02881 USA
- Rhode Island Infectious Diseases (RIID) Research Program Laboratory, Veterans Affairs Medical Center, Providence, RI USA
- Warren Alpert Medical School of Brown University, Providence, RI USA
| |
Collapse
|
45
|
Jacqueline C, Caillon J. Impact of bacterial biofilm on the treatment of prosthetic joint infections. J Antimicrob Chemother 2015; 69 Suppl 1:i37-40. [PMID: 25135088 DOI: 10.1093/jac/dku254] [Citation(s) in RCA: 105] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Microbial biofilm contributes to chronic infection and is involved in the pathogenesis of prosthetic joint infections. Biofilms are structurally complex and should be considered a dynamic system able to protect the bacteria from host defence mechanisms and from antibacterial agents. Despite the use of antibiotics recognized as effective against acute infections, prosthetic joint infections require long-term suppressive treatment acting on adherent bacteria. Conventional in vitro susceptibility testing methods are not suitable for biofilm-associated infections given that these tests do not take into account the physiological parameters of bacterial cells in vivo. Most anti-staphylococcal drugs are able to inhibit in vitro the adhesion of bacteria to a surface, considered to be the first step in biofilm formation. Recent studies suggest that the lack of activity of antibiotics against biofilm-embedded bacteria seems to be more related to the decreased effect of the drug on the pathogen than to the poor penetration of the drug into the biofilm. Eradication of biofilm-embedded bacteria is a very difficult task and combination therapy is required in the treatment of persistent infections involving biofilm. Although several combinations demonstrate potent efficacy, rifampicin is the most common partner drug of effective combinations against staphylococcal biofilms. Considering the complexity of biofilm-related infections, further studies are needed to assess the activity of new therapeutic agents in combination with antibiotics (quorum-sensing inhibitors, biofilm disruptors and specific anti-biofilm molecules).
Collapse
Affiliation(s)
- Cédric Jacqueline
- Université de Nantes, Faculté de Médecine, UPRES EA 3826, 1 rue Gaston Veil, Nantes, F-44000 France
| | - Jocelyne Caillon
- Université de Nantes, Faculté de Médecine, UPRES EA 3826, 1 rue Gaston Veil, Nantes, F-44000 France
| |
Collapse
|
46
|
Nnedu ON, Pankey GA. Update on the emerging role of telavancin in hospital-acquired infections. Ther Clin Risk Manag 2015; 11:605-10. [PMID: 25926737 PMCID: PMC4403509 DOI: 10.2147/tcrm.s57376] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Telavancin is a lipoglycopeptide that has activity against Gram-positive aerobic and anaerobic bacteria. It has activity against methicillin-resistant Staphylococcus aureus, vancomycin-intermediate S. aureus and non-Van-A strains of vancomycin-resistant enterococci. It has been approved by the US Food and Drug Administration (FDA) for complicated skin and skin structure infections and hospital-acquired pneumonia. There is a need for more clinical studies to determine the role of telavancin in treating bacteremia and prosthetic device infections. In this review, we discuss the published data on the use of telavancin in treating hospital-acquired infections and provide an update on new research.
Collapse
Affiliation(s)
- Obinna N Nnedu
- Infectious Disease Department, Ochsner Clinic Foundation, New Orleans, LA, USA
| | - George A Pankey
- Infectious Disease Research, Ochsner Clinic Foundation, New Orleans, LA, USA
| |
Collapse
|
47
|
Masterton R, Cornaglia G, Courvalin P, Lode HM, Rello J, Torres A. The clinical positioning of telavancin in Europe. Int J Antimicrob Agents 2015; 45:213-20. [DOI: 10.1016/j.ijantimicag.2014.12.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 12/03/2014] [Indexed: 11/16/2022]
|
48
|
Loo CY, Lee WH, Young PM, Cavaliere R, Whitchurch CB, Rohanizadeh R. Implications and emerging control strategies for ventilator-associated infections. Expert Rev Anti Infect Ther 2015; 13:379-93. [DOI: 10.1586/14787210.2015.1007045] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
49
|
Hegde SS, Janc JW. Efficacy of telavancin, a lipoglycopeptide antibiotic, in experimental models of Gram-positive infection. Expert Rev Anti Infect Ther 2014; 12:1463-75. [DOI: 10.1586/14787210.2014.979789] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
50
|
Dosler S, Mataraci E. In vitro pharmacokinetics of antimicrobial cationic peptides alone and in combination with antibiotics against methicillin resistant Staphylococcus aureus biofilms. Peptides 2013; 49:53-8. [PMID: 23988790 DOI: 10.1016/j.peptides.2013.08.008] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Revised: 08/07/2013] [Accepted: 08/07/2013] [Indexed: 12/15/2022]
Abstract
Antibiotic therapy for methicillin-resistant Staphylococcus aureus (MRSA) infections is becoming more difficult in hospitals and communities because of strong biofilm-forming properties and multidrug resistance. Biofilm-associated MRSA is not affected by therapeutically achievable concentrations of antibiotics. Therefore, we investigated the in vitro pharmacokinetic activities of antimicrobial cationic peptides (AMPs; indolicidin, cecropin [1-7]-melittin A [2-9] amide [CAMA], and nisin), either alone or in combination with antibiotics (daptomycin, linezolid, teicoplanin, ciprofloxacin, and azithromycin), against standard and 2 clinically obtained MRSA biofilms. The minimum inhibitory concentrations (MIC) and minimum biofilm-eradication concentrations (MBEC) were determined by microbroth dilution technique. The time-kill curve (TKC) method was used to determine the bactericidal activities of the AMPs alone and in combination with the antibiotics against standard and clinically obtained MRSA biofilms. The MIC values of the AMPs and antibiotics ranged between 2 to 16 and 0.25 to 512 mg/L, and their MBEC values were 640 and 512 to 5120 mg/L, respectively. The TKC studies demonstrated that synergistic interactions occurred most frequently when using nisin+daptomycin/ciprofloxacin, indolicidin+teicoplanin, and CAMA+ciprofloxacin combinations. No antagonism was observed with any combination. AMPs appear to be good candidates for the treatment of MRSA biofilms, as they act as both enhancers of anti-biofilm activities and help to prevent or delay the emergence of resistance when used either alone or in combination with antibiotics.
Collapse
Affiliation(s)
- Sibel Dosler
- Department of Pharmaceutical Microbiology Faculty of Pharmacy, Istanbul University, 34116, Beyazit, Istanbul, Turkiye.
| | | |
Collapse
|