1
|
Epperson AB, Awad ME, Gorman M, Loker K, Alfonso NA, Stoneback JW. Clinical practice guidelines for antimicrobial-loaded cements and beads in orthopedic trauma and arthroplasty. EUROPEAN JOURNAL OF ORTHOPAEDIC SURGERY & TRAUMATOLOGY : ORTHOPEDIE TRAUMATOLOGIE 2024; 35:25. [PMID: 39585403 DOI: 10.1007/s00590-024-04132-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 10/26/2024] [Indexed: 11/26/2024]
Abstract
PURPOSE Implants in orthopedic trauma and arthroplasty surgery establish a milieu conducive to biofilm formation. Antimicrobial-loaded cements (ABCs) and beads have become popular in treating acute and chronic orthopedic surgery-related infections. The growing incidence of antimicrobial resistance has necessitated the exploration of alternative antibiotic medications. This review aims to demonstrate meaningful clinical decision-making guidance for orthopedic surgeons in approaching the management of these complex infections. METHODS This study protocol was conducted following the PRISMA checklist and guidelines of the Cochrane Handbook for Systematic Reviews of Interventions. PubMed, Ovid MEDLINE, Web of Science, and other databases were queried using applicable search terms. Relevant dosing, efficacy, and elution profiles were reviewed and compiled from 74 articles published between 1976 and 2019. First-line and targeted therapies were identified against rare and resistant bacteria. Drug therapies not recommended due to excessive cytotoxicity or poor delivery kinetics were also elucidated. RESULTS This compilation describes thirty-two antibiotics and three antifungals that have successfully managed orthopedic surgery-related infections, including infections with numerous recalcitrant and multidrug-resistant species. Optimized ratios of carrier to antimicrobial are provided for each delivery method. The elution and efficacy profiles of the various antibiotics are described when available. DISCUSSION/CONCLUSION These recommendations offer the most up-to-date and comprehensive practice guidelines for using antimicrobials in cements and beads for treating orthopedic hardware-related infections. With the ever-evolving propensity of bacteria to develop antibiotic resistance, these recommendations are dynamic. Collaboration with medicine, infectious disease, and/or pharmacology teams is recommended to create institutional protocols for antibiotic-eluting implants and close comanagement to ensure efficacy and patient safety.
Collapse
Affiliation(s)
- Aaron B Epperson
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, 12631 E. 17th Avenue, Mail Stop B202, Aurora, CO, 80045, USA.
| | - Mohamed E Awad
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, 12631 E. 17th Avenue, Mail Stop B202, Aurora, CO, 80045, USA
| | - Melissa Gorman
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, 12631 E. 17th Avenue, Mail Stop B202, Aurora, CO, 80045, USA
| | - Kristin Loker
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, 12631 E. 17th Avenue, Mail Stop B202, Aurora, CO, 80045, USA
| | - Nicholas A Alfonso
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, 12631 E. 17th Avenue, Mail Stop B202, Aurora, CO, 80045, USA
| | - Jason W Stoneback
- Department of Orthopedics, University of Colorado Anschutz Medical Campus, 12631 E. 17th Avenue, Mail Stop B202, Aurora, CO, 80045, USA
| |
Collapse
|
2
|
Arayici PP, Coksu I, Ozbek T, Acar S. Targeted delivery of rifaximin using P6.2-decorated bifunctional PLGA nanoparticles for combating Staphylococcus aureus infections. BIOMATERIALS ADVANCES 2024; 161:213862. [PMID: 38678666 DOI: 10.1016/j.bioadv.2024.213862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/02/2024] [Accepted: 04/13/2024] [Indexed: 05/01/2024]
Abstract
The emergence of antibiotic resistance makes the treatment of bacterial infections difficult and necessitates the development of alternative strategies. Targeted drug delivery systems are attracting great interest in overcoming the limitations of traditional antibiotics. Here, we aimed for targeted delivery of rifaximin (RFX) by decorating RFX-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles (NPs) with synthetic P6.2 peptide, which was used as a targeting agent for the first time. Our results showed that encapsulation of RFX into NPs increased its antibacterial activity by improving its solubility and providing controlled release, while P6.2 modification allowed targeting of NPs to S. aureus bacterial cells. A promising therapeutic approach for bacterial infections, these P6.2-conjugated RFX-loaded PLGA NPs (TR-NP) demonstrated potent antibacterial activity against both strains of S. aureus. The antibacterial activity of RFX-loaded PLGA NPs (R-NP) showed significant results with an increase of 8 and 16-fold compared to free RFX against S. aureus and MRSA, respectively. Moreover, the activity of targeted nanoparticles was found to be increased 32 or 16-fold with an MBC value of 0.0078 μg/mL. All nanoparticles were found to be biocompatible at doses where they showed antimicrobial activity. Finally, it revealed that P6.2-conjugated targeted nanoparticles extremely accumulated in S. aureus rather than E. coli.
Collapse
Affiliation(s)
- Pelin Pelit Arayici
- Yildiz Technical University, Faculty of Chemical and Metallurgical, Department of Bioengineering, 34210 Esenler, Istanbul, Turkey; Health Biotechnology Joint Research and Application Center of Excellence, 34220 Esenler, Istanbul, Turkey.
| | - Irem Coksu
- Yildiz Technical University, Faculty of Chemical and Metallurgical, Department of Bioengineering, 34210 Esenler, Istanbul, Turkey
| | - Tulin Ozbek
- Yildiz Technical University, Faculty of Arts and Sciences, Department of Molecular Biology and Genetics, 34220 Esenler, Istanbul, Turkey
| | - Serap Acar
- Yildiz Technical University, Faculty of Chemical and Metallurgical, Department of Bioengineering, 34210 Esenler, Istanbul, Turkey
| |
Collapse
|
3
|
Kurnia D, Padilah R, Apriyanti E, Dharsono HDA. Phytochemical Analysis and Anti-Biofilm Potential That Cause Dental Caries from Black Cumin Seeds ( Nigella sativa Linn.). Drug Des Devel Ther 2024; 18:1917-1932. [PMID: 38828022 PMCID: PMC11144408 DOI: 10.2147/dddt.s454217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 04/23/2024] [Indexed: 06/05/2024] Open
Abstract
The oral cavity is an excellent place for various microorganisms to grow. Spectrococcus mutans and Spectrococcus sanguinis are Gram-negative bacteria found in the oral cavity as pioneer biofilm formers on the tooth surface that cause caries. Caries treatment has been done with antibiotics and therapeutics, but the resistance level of S. mutans and S. sanguinis bacteria necessitates the exploration of new drug compounds. Black cumin (Nigella sativa Linn.) is known to contain secondary metabolites that have antioxidant, antibacterial, anti-biofilm, anti-inflammatory and antifungal activities. The purpose of this review article is to present data on the potential of Nigella sativa Linn seeds as anti-biofilm. This article will discuss biofilm-forming bacteria, the resistance mechanism of antibiotics, the bioactivity of N. sativa extracts and seed isolates together with the Structure Activity Relationship (SAR) review of N. sativa compound isolates. We collected data from reliable references that will illustrate the potential of N. sativa seeds as anti-biofilm drug.
Collapse
Affiliation(s)
- Dikdik Kurnia
- Department of Chemistry, Faculty of Mathematics and Natural Science, Universitas Padjadjaran, Sumedang, Indonesia
| | - Rizal Padilah
- Department of Chemistry, Faculty of Mathematics and Natural Science, Universitas Padjadjaran, Sumedang, Indonesia
| | - Eti Apriyanti
- Department of Chemistry, Faculty of Mathematics and Natural Science, Universitas Padjadjaran, Sumedang, Indonesia
| | - Hendra Dian Adhita Dharsono
- Department of Conservative Dentistry, Faculty of Dentistry, Universitas Padjadjaran, Bandung, Jawa Barat, Indonesia
| |
Collapse
|
4
|
Laperche J, Barrett CC, Boduch A, Glasser J, Clippert D, Garcia DR, Antoci V. Mechanically stable rifampin antibiotic cement inhibits Pseudomonas aeruginosa biofilm surface growth. J Orthop Res 2024; 42:547-554. [PMID: 37884321 DOI: 10.1002/jor.25720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/07/2023] [Accepted: 10/16/2023] [Indexed: 10/28/2023]
Abstract
Rifampin has been proven to be effective in the treatment of prosthetic infections due to its ability to intercalate into biofilms. The use of rifampin in antibiotic spacers is not well described, which would be especially important in the local periprosthetic environment where parenteral doses have poor penetration. The null hypothesis tests if rifampin use in polymethyl methacrylate (PMMA) cement will show no clinically significant impact on mechanical strength at antibiotic concentrations that remain bactericidal. Test antibiotic cement samples supplemented with 0, 30, 50, 100, 150, or 200 mg of rifampin into a standard 40 g bag were tested for compression to failure using published ASTM standards. The samples were then inoculated with Pseudomonas aeruginosa and either evaluated for lipopolysaccharide (LPS) presence as a marker of biofilm or tested by elution as the Kirby Bauer assay. Rifampin concentrations of 30 and 50 mg, showed no statistically different mechanical characteristics from control PMMA (p > 0.05). The 100-mg sample fell within the acceptable range of compressive strength and had significantly less LPS and bacterial presence compared to the control at 12 and 24 h. The ability of PMMA with 100 mg of rifampin to maintain its structural integrity and have significant bacterial inhibition at 12 and 24 h makes it a great candidate as an antibiotic bone cement additive. PMMA loaded with up to 100 mg of rifampin shows promise in the treatment and prevention of periprosthetic joint infection for total knee and total hip arthroplasty.
Collapse
Affiliation(s)
- Jacob Laperche
- Joint Replacement Center, University Orthopedics, East Providence, Rhode Island, USA
- Frank H. Netter School of Medicine, Quinnipiac University, North Haven, Connecticut, USA
| | - Caitlin C Barrett
- Joint Replacement Center, University Orthopedics, East Providence, Rhode Island, USA
| | - Abigail Boduch
- Department of Orthopedic Surgery, Rhode Island Hospital, Providence, Rhode Island, USA
| | - Jillian Glasser
- Joint Replacement Center, University Orthopedics, East Providence, Rhode Island, USA
| | - Drew Clippert
- Joint Replacement Center, University Orthopedics, East Providence, Rhode Island, USA
| | - Dioscaris R Garcia
- Department of Orthopedic Surgery, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Valentin Antoci
- Joint Replacement Center, University Orthopedics, East Providence, Rhode Island, USA
- Department of Orthopedic Surgery, Rhode Island Hospital, Providence, Rhode Island, USA
- Department of Orthopedic Surgery, Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| |
Collapse
|
5
|
Ortiz-Fullana JL, Sánchez C, Pagán P, Mulero-Soto P, Dieppa-Barnes G, Santini-Domínguez R, Martinez-Trabal JL. Human gelatin thrombin matrix with rifampin for the treatment of prosthetic vascular graft infections. J Vasc Surg Cases Innov Tech 2024; 10:101365. [PMID: 38130367 PMCID: PMC10731601 DOI: 10.1016/j.jvscit.2023.101365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 10/23/2023] [Indexed: 12/23/2023] Open
Abstract
We aim to describe and report on a novel graft preservation technique using a human gelatin thrombin matrix with rifampin for the treatment of vascular graft infections. Eight patients with vascular graft infections were included, one with bilateral infections, for a total of nine cases from January 2016 through June 2021. All the patients underwent wound exploration and placement of human gelatin thrombin matrix with rifampin. No deaths or allergic reactions had been reported at the 30-day follow-up, with only one major amputation. The graft and limb salvage rates were 77.8% at the 1-year follow-up. The mean time to a major amputation was 122 days, and the mean time to graft excision was 30 days.
Collapse
Affiliation(s)
| | - Cristina Sánchez
- School of Medicine, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Pedro Pagán
- School of Medicine, Ponce Health Sciences University, Ponce, Puerto Rico
| | - Patricia Mulero-Soto
- Department of Vascular Surgery, San Luke's Memorial Hospital, Ponce, Puerto Rico
| | | | | | | |
Collapse
|
6
|
Pedroni MA, Ribeiro VST, Cieslinski J, Lopes APDA, Kraft L, Suss PH, Tuon FF. Different concentrations of vancomycin with gentamicin loaded PMMA to inhibit biofilm formation of Staphylococcus aureus and their implications. J Orthop Sci 2024; 29:334-340. [PMID: 36526520 DOI: 10.1016/j.jos.2022.11.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 11/25/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND This study aimed to evaluate different concentrations of vancomycin and/or gentamicin loaded polymethylmethacrylate (PMMA) against biofilm formation of Staphylococcus aureus. METHODS Biofilm production of S. aureus in PMMA loaded with different concentrations of vancomycin and gentamicin were evaluated by quantitative analysis of biofilm cells, scanning electronic microscopy, viability assay, Fourier transform infrared spectroscopy, and checkerboard. Statistical analysis was performed by Mann Whitney test. The difference in colony forming units per mL was significant when p < 0.05. RESULTS All loaded PMMA presented a reduction in the number of colony forming units per mL (p < 0.05). The gentamicin-loaded PMMA could inhibits the grown of sessile cells (p < 0.05), where the group vancomycin 4 g + gentamicin 500 mg presented a better result. The Fourier transform infrared spectra showed no significant differences, and checkerboard of vancomycin and gentamicin showed synergism. CONCLUSION Effects against adherence and bacterial development in PMMA loaded with antibiotics were mainly seen in the group vancomycin 4 g + gentamicin 500 mg, and synergic effect can be applied in antibiotic-loaded cement.
Collapse
Affiliation(s)
- Marco Antonio Pedroni
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, 80215-901, Brazil
| | - Victoria Stadler Tasca Ribeiro
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, 80215-901, Brazil
| | - Juliette Cieslinski
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, 80215-901, Brazil
| | - Ana Paula de Andrade Lopes
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, 80215-901, Brazil
| | - Letícia Kraft
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, 80215-901, Brazil
| | - Paula Hansen Suss
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, 80215-901, Brazil
| | - Felipe Francisco Tuon
- Laboratory of Emerging Infectious Diseases, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Paraná, 80215-901, Brazil.
| |
Collapse
|
7
|
Albac S, Anzala N, Bonnot D, Mirfendereski H, Chavanet P, Croisier D. Efficacy of ceftaroline and rifampin, alone or combined, in a rat model of methicillin-resistant Staphylococcus epidermidis osteomyelitis without implant. Microbiol Spectr 2023; 11:e0015323. [PMID: 37791959 PMCID: PMC10715034 DOI: 10.1128/spectrum.00153-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 07/19/2023] [Indexed: 10/05/2023] Open
Abstract
IMPORTANCE Methicillin-resistant Staphylococcus epidermidis (MRSE) contributes to a high percentage of orthopedic infections, and their treatment represents a huge challenge. The present study aimed to evaluate the efficacy of ceftaroline alone or combined with rifampin in a rat MRSE osteomyelitis model and the bone penetration of ceftaroline. A ceftaroline monotherapy showed a significant bacterial reduction in infected bones after a 7-day period of treatment. The combination ceftaroline plus rifampin leveraged rifampin's bactericidal activity, shortening the duration of positive culture in infected animals. These results suggest that ceftaroline and rifampin combination therapy could represent a valuable therapeutic option for human MRSE osteomyelitis and deserves further preclinical and clinical evaluation.
Collapse
Affiliation(s)
| | | | | | - H. Mirfendereski
- Université de Poitiers, Poitiers, France
- Centre Hospitalier Universitaire de Poitiers, Poitiers, France
| | - P. Chavanet
- Vivexia, Dijon, France
- Département d’Infectiologie, Centre Hospitalier Universitaire, Dijon, France
| | | |
Collapse
|
8
|
Kang YR, Chung DR, Ko JH, Huh K, Cho SY, Kang CI, Peck KR. Comparing the Synergistic and Antagonistic Interactions of Ciprofloxacin and Levofloxacin Combined with Rifampin against Drug-Resistant Staphylococcus aureus: A Time-Kill Assay. Antibiotics (Basel) 2023; 12:antibiotics12040711. [PMID: 37107077 PMCID: PMC10135007 DOI: 10.3390/antibiotics12040711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 03/30/2023] [Accepted: 03/31/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Treatment of device-related infections by drug-resistant Staphylococcus aureus can be challenging, and combination therapy has been proposed as a potential solution. We compared the effectiveness of levofloxacin-rifampin and ciprofloxacin-rifampin combinations in killing methicillin-resistant S. aureus (MRSA) using a time-kill assay. METHODS We randomly selected 15 vancomycin-susceptible S. aureus (VSSA) strains, 3 vancomycin-intermediate S. aureus (VISA) strains, and 12 heterogeneous VISA (hVISA) strains from the Asian Bacterial Bank. Time-kill experiments were performed in duplicate for each isolate. Viable bacterial counts were determined at 0 h, 4 h, 8 h, and 24 h for the ciprofloxacin- and levofloxacin-rifampin combinations at 1× MIC and 0.5× MIC. We compared synergistic and antagonistic interactions between the two combinations. RESULTS The viable bacterial count significantly decreased after 24 h of exposure to ciprofloxacin-rifampin and levofloxacin-rifampin combinations, with synergy observed more frequently in isolates exposed to ciprofloxacin-rifampin (43.3%) than levofloxacin-rifampin (20.0%) (p = 0.0082). The synergistic interactions of both combinations were more frequently observed in resistant strains with high MICs of ciprofloxacin (≥16 mg/L) and levofloxacin (≥8 mg/L). Levofloxacin tended to exhibit more frequent antagonistic interactions with rifampin than ciprofloxacin, although there was no statistical difference in antagonism between the two combinations. CONCLUSIONS Our study demonstrated that ciprofloxacin exhibits superior synergistic activity against MRSA strains, including VISA/hVISA, when combined with rifampin compared with levofloxacin. High MICs of fluoroquinolones were found to predict synergism. Our results suggest that ciprofloxacin may be a more effective choice than levofloxacin for combination therapy with rifampin in the treatment of MRSA infections.
Collapse
Affiliation(s)
- Yu Ri Kang
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
- Asia Pacific Foundation for Infectious Diseases (APFID), Seoul 06351, Republic of Korea
| | - Doo Ryeon Chung
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
- Asia Pacific Foundation for Infectious Diseases (APFID), Seoul 06351, Republic of Korea
- Center for Infection Prevention and Control, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Jae-Hoon Ko
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Kyungmin Huh
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
- Asia Pacific Foundation for Infectious Diseases (APFID), Seoul 06351, Republic of Korea
| | - Sun Young Cho
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
- Center for Infection Prevention and Control, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Cheol-In Kang
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| | - Kyong Ran Peck
- Division of Infectious Diseases, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea
| |
Collapse
|
9
|
Stability of Non-Ionic Surfactant Vesicles Loaded with Rifamycin S. Pharmaceutics 2022; 14:pharmaceutics14122626. [PMID: 36559121 PMCID: PMC9785864 DOI: 10.3390/pharmaceutics14122626] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
These days, the eradication of bacterial infections is more difficult due to the mechanism of resistance that bacteria have developed towards traditional antibiotics. One of the medical strategies used against bacteria is the therapy with drug delivery systems. Non-ionic vesicles are nanomaterials with good characteristics for encapsulating drugs, due to their bioavailability and biodegradability, which allow the drugs to reach the specific target and reduce their side effects. In this work, the antibiotic Rifamycin S was encapsulated. The rifamycin antibiotics family has been widely used against Mycobacterium tuberculosis, but recent studies have also shown that rifamycin S and rifampicin derivatives have bactericidal activity against Staphylococcus epidermidis and Staphylococcus aureus. In this work, a strain of S. aureus was selected to study the antimicrobial activity through Minimum Inhibitory Concentration (MIC) assay. Three formulations of niosomes were prepared using the thin film hydration method by varying the composition of the aqueous phase, which included MilliQ water, glycerol solution, or PEG400 solution. Niosomes with a rifamycin S concentration of 0.13 μg/g were satisfactorily prepared. Nanovesicles with larger size and higher encapsulation efficiency (EE) were obtained when using glycerol and PEG400 in the aqueous media. Our results showed that niosomes consisting of an aqueous glycerol solution have higher stability and EE across a diversity of temperatures and pHs, and a lower MIC of rifamycin S against S. aureus.
Collapse
|
10
|
Zelmer AR, Nelson R, Richter K, Atkins GJ. Can intracellular Staphylococcus aureus in osteomyelitis be treated using current antibiotics? A systematic review and narrative synthesis. Bone Res 2022; 10:53. [PMID: 35961964 PMCID: PMC9374758 DOI: 10.1038/s41413-022-00227-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 05/26/2022] [Accepted: 06/15/2022] [Indexed: 11/09/2022] Open
Abstract
Approximately 40% of treatments of chronic and recurrent osteomyelitis fail in part due to bacterial persistence. Staphylococcus aureus, the predominant pathogen in human osteomyelitis, is known to persist by phenotypic adaptation as small-colony variants (SCVs) and by formation of intracellular reservoirs, including those in major bone cell types, reducing susceptibility to antibiotics. Intracellular infections with S. aureus are difficult to treat; however, there are no evidence-based clinical guidelines addressing these infections in osteomyelitis. We conducted a systematic review of the literature to determine the demonstrated efficacy of all antibiotics against intracellular S. aureus relevant to osteomyelitis, including protein biosynthesis inhibitors (lincosamides, streptogramins, macrolides, oxazolidines, tetracyclines, fusidic acid, and aminoglycosides), enzyme inhibitors (fluoroquinolones and ansamycines), and cell wall inhibitors (beta-lactam inhibitors, glycopeptides, fosfomycin, and lipopeptides). The PubMed and Embase databases were screened for articles related to intracellular S. aureus infections that compared the effectiveness of multiple antibiotics or a single antibiotic together with another treatment, which resulted in 34 full-text articles fitting the inclusion criteria. The combined findings of these studies were largely inconclusive, most likely due to the plethora of methodologies utilized. Therefore, the reported findings in the context of the models employed and possible solutions for improved understanding are explored here. While rifampicin, oritavancin, linezolid, moxifloxacin and oxacillin were identified as the most effective potential intracellular treatments, the scientific evidence for these is still relatively weak. We advocate for more standardized research on determining the intracellular effectiveness of antibiotics in S. aureus osteomyelitis to improve treatments and patient outcomes.
Collapse
Affiliation(s)
- Anja R Zelmer
- Centre for Orthopaedic and Trauma Research, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Renjy Nelson
- Department of Infectious Diseases, Central Adelaide Local Health Network, Adelaide, SA, 5000, Australia.,Royal Adelaide Hospital, Adelaide, SA, 5000, Australia
| | - Katharina Richter
- Richter Lab, Department of Surgery, Basil Hetzel Institute for Translational Health Research, University of Adelaide, Adelaide, SA, 5011, Australia
| | - Gerald J Atkins
- Centre for Orthopaedic and Trauma Research, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA, 5000, Australia.
| |
Collapse
|
11
|
Higashihira S, Simpson SJ, Collier CD, Natoli RM, Kittaka M, Greenfield EM. Halicin Is Effective Against Staphylococcus aureus Biofilms In Vitro. Clin Orthop Relat Res 2022; 480:1476-1487. [PMID: 35583504 PMCID: PMC9278916 DOI: 10.1097/corr.0000000000002251] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 04/28/2022] [Indexed: 01/31/2023]
Abstract
BACKGROUND Biofilms protect bacteria from the host immune system and many antibiotics, making the treatment of orthopaedic infections difficult. Halicin, a recently discovered antibiotic, has potent activity against nonorthopaedic infections in mice and the planktonic, free-living forms of many bacterial species, including Staphylococcus aureus , a common cause of orthopaedic infections. Importantly, halicin did not induce resistance in vitro and was effective against drug-resistant bacteria and proliferating and quiescent bacteria. Quiescence is an important cause of antibiotic tolerance in biofilms. However, whether halicin acts on biofilms has not been tested. QUESTIONS/PURPOSES (1) Does halicin reduce the viability of S. aureus in less mature and more mature biofilms as it does in planktonic cultures? (2) How do the relative effects of halicin on S. aureus biofilms and planktonic cultures compare with those of conventional antibiotics (tobramycin, cefazolin, vancomycin, or rifampicin) that are commonly used in clinical orthopaedic infections? METHODS To measure minimal biofilm eradication concentrations (MBECs) with less mature 3-day and more mature 7-day biofilms, we used 96-well peg plates that provided high throughput and excellent reproducibility. After S. aureus -Xen36 biofilm formation, planktonic bacteria were removed from the cultures, and the biofilms were exposed to various concentrations of halicin, tobramycin, cefazolin, vancomycin, or rifampicin for 20 hours. Biofilm viability was determined by measuring resazurin reduction or by counting colony-forming units after sonication. To determine effects of halicin and the conventional antibiotics on biofilm viability, we defined MBEC 75 as the lowest concentration that decreased viability by 75% or more. To determine effects on bacterial viability in planktonic cultures, minimum inhibitory concentrations (MICs) were determined with the broth dilution method. Each result was measured in four to 10 independent experiments. RESULTS We found no differences between halicin's effectiveness against planktonic S. aureus and 3-day biofilms (MIC and MBEC 75 for 3-day biofilms was 25 μM [interquartile range 25 to 25 and 25 to 25, respectively]; p > 0.99). Halicin was eightfold less effective against more mature 7-day biofilms (MBEC 75 = 200 μM [100 to 200]; p < 0.001). Similarly, tobramycin was equally effective against planktonic culture and 3-day biofilms (MIC and MBEC 75 for 3-day biofilms was 20 μM [20 to 20 and 10 to 20, respectively]; p > 0.99). Tobramycin's MBEC 75 against more mature 7-day biofilms was 320 μM (320 to 480), which is 16-fold greater than its planktonic MIC (p = 0.03). In contrast, the MBEC 75 for cefazolin, vancomycin, and rifampicin against more mature 7-day biofilms were more than 1000-fold (> 1000; p < 0.001), 500-fold (500 to 875; p < 0.001), and 3125-fold (3125 to 5469; p = 0.004) greater than their planktonic MICs, respectively, consistent with those antibiotics' relative inactivity against biofilms. CONCLUSION Halicin was as effective against S. aureus in less mature 3-day biofilms as those in planktonic cultures, but eightfold higher concentrations were needed for more mature 7-day biofilms. Tobramycin, an antibiotic whose effectiveness depends on biofilm maturity, was also as effective against S. aureus in less mature 3-day biofilms as those in planktonic cultures, but 16-fold higher concentrations were needed for more mature 7-day biofilms. In contrast, cefazolin, vancomycin, and rifampicin were substantially less active against both less and more mature biofilms than against planktonic cultures. CLINICAL RELEVANCE Halicin is a promising antibiotic that may be effective against S. aureus osteomyelitis and infections on orthopaedic implants. Future studies should assess the translational value of halicin by testing its effects in animal models of orthopaedic infections; on the biofilms of other bacterial species, including multidrug-resistant bacteria; and in combination therapy with conventional antibiotics.
Collapse
Affiliation(s)
- Shota Higashihira
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Orthopaedic Surgery, Yokohama City University, Yokohama, Japan
| | - Stefanie Jan Simpson
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Christopher David Collier
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Roman Michael Natoli
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mizuho Kittaka
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, USA
| | - Edward Michael Greenfield
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
12
|
Rifampicin-loaded PLGA nanoparticles for local treatment of musculoskeletal infections: Formulation and characterization. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
13
|
Monk M, Elshaboury R, Tatara A, Nelson S, Bidell MR. A Case Series of Rifabutin Use in Staphylococcal Prosthetic Infections. Microbiol Spectr 2022; 10:e0038422. [PMID: 35543561 PMCID: PMC9241794 DOI: 10.1128/spectrum.00384-22] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/12/2022] [Indexed: 01/24/2023] Open
Abstract
This case series describes seven patients who received rifabutin in place of rifampin combined with conventional antimicrobial therapy for treatment of hardware-associated staphylococcal infections. Infection recurrence, defined as need for unplanned surgical intervention within the evaluable follow up period after starting rifabutin, occurred in two patients. Two patients experienced possible treatment-associated adverse effects. Findings support future work to examine rifabutin use, when rifampin is not suitable, for adjunctive treatment of staphylococcal hardware infections. IMPORTANCE This work evaluates real-world data and clinical outcomes when rifabutin is used in place of rifampin for adjunctive management of staphylococcal hardware-associated infections. This is the second case study looking at this specific use of rifabutin, signifying the current lack of clinical data in this area. Assessing use of rifabutin in this capacity is clinically important given its lower propensity for drug interactions compared to rifampin.
Collapse
Affiliation(s)
- Miranda Monk
- Department of Pharmacy, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ramy Elshaboury
- Department of Pharmacy, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alexander Tatara
- Department of Medicine, Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sandra Nelson
- Department of Medicine, Division of Infectious Diseases, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Monique R. Bidell
- Department of Pharmacy, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
14
|
Thill P, Robineau O, Roosen G, Patoz P, Gachet B, Lafon-Desmurs B, Tetart M, Nadji S, Senneville E, Blondiaux N. Rifabutin versus rifampicin bactericidal and antibiofilm activities against clinical strains of Staphylococcus spp. isolated from bone and joint infections. J Antimicrob Chemother 2022; 77:1036-1040. [PMID: 35028671 DOI: 10.1093/jac/dkab486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/08/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Staphylococci account for approximately 60% of periprosthetic joint infections (PJIs). Rifampicin (RMP) combination therapy is generally considered to be the treatment of choice for staphylococcal PJIs but carries an important risk of adverse events and drug-drug interactions. Rifabutin (RFB) shares many of the properties of rifampicin but causes fewer adverse events. OBJECTIVES To compare the minimal inhibitory concentration (MIC), the minimum bactericidal concentrations (MBC), and the minimum biofilm eradication concentrations (MBEC) of rifabutin and rifampicin for staphylococcal clinical strains isolated from PJIs. METHODS 132 clinical strains of rifampicin-susceptible staphylococci [51 Staphylococcus aureus (SA), 48 Staphylococcus epidermidis (SE) and 33 other coagulase-negative staphylococci (CoNS)] were studied. The MBC and the MBEC were determined using the MBEC® Assay for rifabutin and rifampicin and were compared. RESULTS When compared with the rifampicin MIC median value, the rifabutin MIC median value was significantly higher for SA (P < 0.05), but there was no statistically significant difference for SE (P = 0.25) and CoNS (P = 0.29). The rifabutin MBC median value was significantly higher than that of rifampicin for SA (P = 0.003) and was lower for SE (P = 0.003) and CoNS (P = 0.03). Rifabutin MBEC median value was statistically lower than that of rifampicin for all strains tested. CONCLUSIONS Using the determination of MBEC values, our study suggests that rifabutin is more effective than rifampicin against clinical strains of Staphylococcus spp. obtained from PJIs. Using MBECs instead of MICs seems to be of interest when considering biofilms. In vivo higher efficacy of rifabutin when compared with rifampicin needs to be confirmed.
Collapse
Affiliation(s)
- Pauline Thill
- Department of Infectious Diseases, Hospital of Tourcoing, Tourcoing, France
| | - Olivier Robineau
- Department of Infectious Diseases, Hospital of Tourcoing, Tourcoing, France.,EA2694, Univ Lille, Centre Hospitalier de Tourcoing, Tourcoing, France
| | - Gabrielle Roosen
- Department of Bacteriology, Hospital of Tourcoing, Tourcoing, France
| | - Pierre Patoz
- Department of Bacteriology, Hospital of Tourcoing, Tourcoing, France
| | - Benoit Gachet
- Department of Infectious Diseases, Hospital of Tourcoing, Tourcoing, France.,EA2694, Univ Lille, Centre Hospitalier de Tourcoing, Tourcoing, France
| | | | - Macha Tetart
- Department of Infectious Diseases, Hospital of Tourcoing, Tourcoing, France
| | - Safia Nadji
- Department of Bacteriology, Hospital of Tourcoing, Tourcoing, France
| | - Eric Senneville
- Department of Infectious Diseases, Hospital of Tourcoing, Tourcoing, France.,EA2694, Univ Lille, Centre Hospitalier de Tourcoing, Tourcoing, France
| | - Nicolas Blondiaux
- Department of Bacteriology, Hospital of Tourcoing, Tourcoing, France.,Univ. Lille, CNRS, Inserm, Institut Pasteur de Lille, U1019 - UMR9017 Center for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
15
|
López-Torres II, Vaquero-Martín J, Torres-Suárez AI, Navarro-García F, Fraguas-Sánchez AI, León-Román VE, Sanz-Ruíz P. The tale of microencapsulated rifampicin: is it useful for the treatment of periprosthetic joint infection? INTERNATIONAL ORTHOPAEDICS 2022; 46:677-685. [PMID: 34988621 PMCID: PMC8930928 DOI: 10.1007/s00264-021-05290-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 12/17/2021] [Indexed: 11/17/2022]
Abstract
Purpose Microencapsulation techniques have allowed the addition of rifampicin to bone cement, but its in vivo efficacy has not been proven. The aim of our study is to determine the superiority of cement containing gentamicin and rifampicin microcapsules in the treatment of PJI versus cement exclusively containing gentamicin. Methods An S. aureus PJI was induced in 15 NZW rabbits. A week after inoculation, the first stage of replacement was carried out, and the animals were divided into two groups: group R received a spacer containing gentamicin and rifampicin microcapsules, and group C received a spacer containing gentamicin. Intra-articular release curve of rifampicin and infection and toxicity markers were monitored for four weeks post-operatively, when microbiological analysis was performed. Results The microbiological cultures showed a significantly lower growth of S. aureus in soft tissue (2.3·104 vs 0; p = 0.01) and bone (5.7·102 vs 0; p = 0.03) in the group with rifampicin microcapsules. No differences were found in systemic toxicity markers. Rifampicin release from the cement spacer showed higher concentrations than the staphylococcal MIC throughout the analysis. Conclusion The in vivo analyses demonstrated the superiority of cement containing gentamicin and rifampicin microcapsules versus the isolated use of gentamicin in the treatment of PJI in the rabbit model without serious side effects due to the systemic absorption of rifampicin. Given the increasing incidence of staphylococci-related PJI, the development of new strategies for intra-articular administration of rifampicin for its treatment has a high clinical impact.
Collapse
Affiliation(s)
- Irene Isabel López-Torres
- Traumatology and Orthopaedic Surgery department, Fundación Jiménez Díaz Hospital, Av. Reyes Católicos 2, 28040, Madrid, Spain.
| | - Javier Vaquero-Martín
- Traumatology and Orthopaedic Surgery department, Gregorio Marañón General Hospital, C/Doctor Esquerdo 46, 28007, Madrid, Spain.,Surgery Department, Faculty of Medicine, Complutense University of Madrid, Plaza Ramón Y Cajal S/N, 28040, Madrid, Spain
| | - Ana-Isabel Torres-Suárez
- Galenic Department, Faculty of Pharmacy, Complutense University of Madrid, Plaza Ramón Y Cajal S/N, 28040, Madrid, Spain
| | - Federico Navarro-García
- Microbiology and Parasitology Department, Faculty of Pharmacy, Complutense University of Madrid, Plaza Ramón Y Cajal S/N, 28040, Madrid, Spain
| | - Ana-Isabel Fraguas-Sánchez
- Galenic Department, Faculty of Pharmacy, Complutense University of Madrid, Plaza Ramón Y Cajal S/N, 28040, Madrid, Spain
| | - Víctor Estuardo León-Román
- Traumatology and Orthopaedic Surgery department, Villalba General Hospital, Carretera de Alpedrete a Moralzarzal, M-608 km 41, Collado Villalba, Madrid, Spain
| | - Pablo Sanz-Ruíz
- Traumatology and Orthopaedic Surgery department, Gregorio Marañón General Hospital, C/Doctor Esquerdo 46, 28007, Madrid, Spain.,Surgery Department, Faculty of Medicine, Complutense University of Madrid, Plaza Ramón Y Cajal S/N, 28040, Madrid, Spain
| |
Collapse
|
16
|
Marro FC, Abad L, Blocker AJ, Laurent F, Josse J, Valour F. In vitro antibiotic activity against intraosteoblastic Staphylococcus aureus: a narrative review of the literature. J Antimicrob Chemother 2021; 76:3091-3102. [PMID: 34459881 PMCID: PMC8598303 DOI: 10.1093/jac/dkab301] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Staphylococcus aureus – a major aetiological agent of bone and joint infection (BJI) – is associated with a high risk of relapse and chronicity, in part due to its ability to invade and persist in non-professional phagocytic bone cells such as osteoblasts. This intracellular reservoir protects S. aureus from the action of the immune system and most antibiotics. To date, the choice of antimicrobial strategies for BJI treatment mostly relies on standard susceptibility testing, bone penetration of antibiotics and their ‘antibiofilm’ activity. Despite the role of intracellular persistent S. aureus in the development of chronic infection, the ability of antibiotics to target the S. aureus intraosteoblastic reservoir is not considered in therapeutic choices but might represent a key determinant of treatment outcome. This review provides an overview of the intracellular pharmacokinetics of antistaphylococcal drugs used in the treatment of BJI and of their ability to target intraosteoblastic S. aureus. Thirteen studies focusing on the intraosteoblastic activity of antibiotics against S. aureus were reviewed, all relying on in vitro models of osteoblast infection. Despite varying incubation times, multiplicities of infection, bacterial strains, and the types of infected cell lines, rifamycins and fluoroquinolones remain the two most potent antimicrobial classes for intraosteoblastic S. aureus eradication, consistent with clinical data showing a superiority of this combination therapy in S. aureus orthopaedic device-related infections.
Collapse
Affiliation(s)
- Florian C Marro
- CIRI-Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France.,Evotec ID Lyon, In Vitro Biology, Infectious Diseases and Antibacterials Unit, Gerland, 69007 Lyon, France.,Université Claude Bernard Lyon 1, Lyon, France
| | - Lélia Abad
- CIRI-Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France.,Université Claude Bernard Lyon 1, Lyon, France.,Laboratoire de bactériologie, Institut des Agents Infectieux, French National Reference Center for Staphylococci, Hospices Civils de Lyon, Lyon, France
| | - Ariel J Blocker
- Evotec ID Lyon, In Vitro Biology, Infectious Diseases and Antibacterials Unit, Gerland, 69007 Lyon, France
| | - Frédéric Laurent
- CIRI-Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France.,Université Claude Bernard Lyon 1, Lyon, France.,Laboratoire de bactériologie, Institut des Agents Infectieux, French National Reference Center for Staphylococci, Hospices Civils de Lyon, Lyon, France.,Centre de Référence pour la prise en charge des Infections ostéo-articulaires complexes (CRIOAc) Lyon, Hospices Civils de Lyon, Lyon, France
| | - Jérôme Josse
- CIRI-Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France.,Université Claude Bernard Lyon 1, Lyon, France.,Centre de Référence pour la prise en charge des Infections ostéo-articulaires complexes (CRIOAc) Lyon, Hospices Civils de Lyon, Lyon, France
| | - Florent Valour
- CIRI-Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France.,Université Claude Bernard Lyon 1, Lyon, France.,Centre de Référence pour la prise en charge des Infections ostéo-articulaires complexes (CRIOAc) Lyon, Hospices Civils de Lyon, Lyon, France.,Service des maladies infectieuses et tropicales, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
17
|
Rifamycin antibiotics and the mechanisms of their failure. J Antibiot (Tokyo) 2021; 74:786-798. [PMID: 34400805 DOI: 10.1038/s41429-021-00462-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/21/2021] [Accepted: 07/26/2021] [Indexed: 02/07/2023]
Abstract
Rifamycins are a class of antibiotics that were first discovered in 1957 and are known for their use in treating tuberculosis (TB). Rifamycins exhibit bactericidal activity against many Gram-positive and Gram-negative bacteria by inhibiting RNA polymerase (RNAP); however, resistance is prevalent and the mechanisms range from primary target modification and antibiotic inactivation to cytoplasmic exclusion. Further, phenotypic resistance, in which only a subpopulation of bacteria grow in concentrations exceeding their minimum inhibitory concentration, and tolerance, which is characterized by reduced rates of bacterial cell death, have been identified as additional causes of rifamycin failure. Here we summarize current understanding and recent developments regarding this critical antibiotic class.
Collapse
|
18
|
Surur AS, Sun D. Macrocycle-Antibiotic Hybrids: A Path to Clinical Candidates. Front Chem 2021; 9:659845. [PMID: 33996753 PMCID: PMC8120311 DOI: 10.3389/fchem.2021.659845] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 04/15/2021] [Indexed: 12/21/2022] Open
Abstract
The tale of abate in antibiotics continued defense mechanisms that chaperone the rise of drug-defying superbugs—on the other hand, the astray in antibacterial drug discovery and development. Our salvation lies in circumventing the genesis of resistance. Considering the competitive advantages of antibacterial chemotherapeutic agents equipped with multiple warheads against resistance, the development of hybrids has rejuvenated. The adoption of antibiotic hybrid paradigm to macrocycles has advanced novel chemical entities to clinical trials. The multi-targeted TD-1792, for instance, retained potent antibacterial activities against multiple strains that are resistant to its constituent, vancomycin. Moreover, the antibiotic conjugation of rifamycins has provided hybrid clinical candidates with desirable efficacy and safety profiles. In 2020, the U.S. FDA has granted an orphan drug designation to TNP-2092, a conjugate of rifamycin and fluoroquinolone, for the treatment of prosthetic joint infections. DSTA4637S is a pioneer antibacterial agent under clinical development and represents a novel class of bacterial therapy, that is, antibody–antibiotic conjugates. DSTA4637S is effective against the notorious persistent S. aureus bacteremia, a revelation of the abracadabra potential of antibiotic hybrid approaches.
Collapse
Affiliation(s)
- Abdrrahman Shemsu Surur
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai'i at Hilo, Hilo, HI, United States
| | - Dianqing Sun
- Department of Pharmaceutical Sciences, The Daniel K. Inouye College of Pharmacy, University of Hawai'i at Hilo, Hilo, HI, United States
| |
Collapse
|
19
|
Karau MJ, Schmidt-Malan SM, Albano M, Mandrekar JN, Rivera CG, Osmon DR, Oravec CP, Berry DJ, Abdel MP, Patel R. Novel Use of Rifabutin and Rifapentine to Treat Methicillin-Resistant Staphylococcus aureus in a Rat Model of Foreign Body Osteomyelitis. J Infect Dis 2021; 222:1498-1504. [PMID: 32914837 DOI: 10.1093/infdis/jiaa401] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 07/05/2020] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Owing to patient intolerance or drug interactions, alternative agents to rifampin are needed for management of staphylococcal periprosthetic joint infection. In the current study, we evaluated rifabutin, rifapentine and rifampin, with and without vancomycin, in a rat model of foreign body osteomyelitis. METHODS Proximal tibiae were inoculated with methicillin-resistant Staphylococcus aureus (MRSA) and a Kirschner wire (K-wire) implanted in each. After 4 weeks of infection, rifampin, rifabutin, or rifapentine were administered, alone or with vancomycin. Tibiae and K-wires were cultured, and medians were reported as log10 colony-forming units (CFUs) per gram of bone or log10 CFUs per K-wire, respectively. RESULTS Rifampin, rifabutin or rifapentine administered with vancomycin yielded less MRSA from bones (0.10, 3.02, and 0.10 log10 CFUs/g, respectively) than did no treatment (4.36 log10 CFUs/g) or vancomycin alone (4.64 log10 CFUs/g) (both P ≤ .02). The K-wires of animals receiving no treatment or vancomycin monotherapy recovered medians of 1.76 and 2.91 log10 CFUs/g per K-wire, respectively. In contrast, rifampin, rifabutin and rifapentine administered with vancomycin yielded medians of 0.1 log10 CFUs per K-wire, respectively. Rifampin resistance was detected in a single animal in the rifampin monotherapy group. CONCLUSIONS Rifabutin or rifapentine with vancomycin were as active as rifampin with vancomycin against MRSA in rat foreign body osteomyelitis, suggesting that rifabutin and/or rifapentine may be alternatives to rifampin in the clinical management of staphylococcal periprosthetic joint infections.
Collapse
Affiliation(s)
- Melissa J Karau
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Rochester, Minnesota, USA
| | - Suzannah M Schmidt-Malan
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Rochester, Minnesota, USA
| | - Mariana Albano
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Rochester, Minnesota, USA
| | | | | | - Douglas R Osmon
- Division of Infectious Diseases, Rochester, Minnesota, USA.,Department of Orthopaedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Daniel J Berry
- Department of Orthopaedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Matthew P Abdel
- Department of Orthopaedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Rochester, Minnesota, USA.,Department of Pharmacy, Rochester, Minnesota, USA
| |
Collapse
|
20
|
Rifampin, Rifapentine, and Rifabutin Are Active against Intracellular Periprosthetic Joint Infection-Associated Staphylococcus epidermidis. Antimicrob Agents Chemother 2021; 65:AAC.01275-20. [PMID: 33199387 DOI: 10.1128/aac.01275-20] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 11/10/2020] [Indexed: 11/20/2022] Open
Abstract
Staphylococcus epidermidis is a major cause of periprosthetic joint infection (PJI); its intracellular persistence within osteoblasts may compromise therapy if that therapy is not intracellularly active. The intracellular activity of rifampin, rifapentine, and rifabutin was assessed against five rifampin-susceptible and two rifampin-resistant S. epidermidis isolates. Compared to no treatment, treatment resulted in a ≥2-fold log10 reduction of intracellular rifampin-susceptible, but not rifampin-resistant, S. epidermidis These findings show activity of rifampin, rifapentine, and rifabutin against intraosteoblast PJI-associated S. epidermidis.
Collapse
|
21
|
Development of a heat labile antibiotic eluting 3D printed scaffold for the treatment of osteomyelitis. Sci Rep 2020; 10:7554. [PMID: 32371998 PMCID: PMC7200676 DOI: 10.1038/s41598-020-64573-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 04/02/2020] [Indexed: 12/17/2022] Open
Abstract
In general, osteomyelitis is treated with antibiotics, and in severe cases, the inflammatory bone tissue is removed and substituted with poly (methyl methacrylate) (PMMA) beads containing antibiotics. However, this treatment necessitates re-surgery to remove the inserted PMMA beads. Moreover, rifampicin, a primary heat-sensitive antibiotic used for osteomyelitis, is deemed unsuitable in this strategy. Three-dimensional (3D) printing technology has gained popularity, as it facilitates the production of a patient-customized implantable structure using various biodegradable biomaterials as well as controlling printing temperature. Therefore, in this study, we developed a rifampicin-loaded 3D scaffold for the treatment of osteomyelitis using 3D printing and polycaprolactone (PCL), a biodegradable polymer that can be printed at low temperatures. We successfully fabricated rifampicin-loaded PCL 3D scaffolds connected with all pores using computer-aided design and manufacturing (CAD/CAM) and printed them at a temperature of 60 °C to prevent the loss of the antibacterial activity of rifampicin. The growth inhibitory activity against Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus), the representative causative organisms of osteomyelitis, was confirmed. In addition, we optimized the rifampicin-loading capacity that causes no damage to the normal bone tissues in 3D scaffold with toxicity evaluation using human osteoblasts. The rifampicin-releasing 3D scaffold developed herein opens new possibilities of the patient-customized treatment of osteomyelitis.
Collapse
|
22
|
Abad L, Josse J, Tasse J, Lustig S, Ferry T, Diot A, Laurent F, Valour F. Antibiofilm and intraosteoblastic activities of rifamycins against Staphylococcus aureus: promising in vitro profile of rifabutin. J Antimicrob Chemother 2020; 75:1466-1473. [DOI: 10.1093/jac/dkaa061] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/28/2020] [Accepted: 01/30/2020] [Indexed: 12/14/2022] Open
Abstract
Abstract
Background
Targeting biofilm-embedded and intraosteoblastic Staphylococcus aureus, rifampicin gained a pivotal role in bone and joint infection (BJI) treatment. Two other rifamycins, rifabutin and rifapentine, may represent better-tolerated alternatives, but their activity against bacterial reservoirs associated with BJI chronicity has never been evaluated.
Objectives
To evaluate the activities of rifampicin, rifabutin and rifapentine in osteoblast infection models.
Methods
Using three S. aureus isolates, rifamycins were compared regarding: (i) their intracellular activity in ‘acute’ (24 h) and ‘chronic’ (7 days) osteoblast infection models at 0.1× MIC, 1× MIC, 10× MIC and 100× MIC, while impacting infection-induced cytotoxicity (MTT assay), intracellular phenol-soluble modulin (PSM) secretion (RT–PCR), resistance selection and small colony variant (SCV) emergence; and (ii) their minimal biofilm eradication concentration (MBEC) and their MIC to prevent biofilm formation (bMIC).
Results
At 0.1× MIC, only rifabutin significantly reduced intracellular inoculum and PSM secretion. All rifamycins allowed a 50% reduction of intraosteoblastic inoculum at higher concentrations, with no difference between acute and chronic infection models, while reducing infection-induced cytotoxicity and PSM secretion. Dose-dependent emergence of intracellular SCVs was observed for all molecules. No intracellular emergence of resistance was detected. bMICs were equivalent for all molecules, but MBEC90s of rifapentine and rifabutin were 10- to 100-fold lower than those of rifampicin, respectively.
Conclusions
All rifamycins are efficient in reducing the S. aureus intraosteoblastic reservoir while limiting infection-induced cytotoxicity, with a higher activity of rifabutin at low concentrations. All molecules prevent biofilm formation, but only rifapentine and rifabutin consistently reduce formed biofilm-embedded bacteria for all isolates. The activity of rifabutin at lower doses highlights its therapeutic potential.
Collapse
Affiliation(s)
- Lélia Abad
- CIRI – Centre International de Recherche en Infectiologie, Inserm, U1111, Université´ Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France
- Université Claude Bernard Lyon 1, Lyon, France
- Laboratoire de bactériologie, Institut des Agents Infectieux, French National Reference Center for Staphylococci, Hospices Civils de Lyon, Lyon, France
| | - Jérôme Josse
- CIRI – Centre International de Recherche en Infectiologie, Inserm, U1111, Université´ Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France
| | - Jason Tasse
- CIRI – Centre International de Recherche en Infectiologie, Inserm, U1111, Université´ Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France
| | - Sébastien Lustig
- Université Claude Bernard Lyon 1, Lyon, France
- Centres de Référence pour la prise en charge des Infections ostéoarticulaires complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France
- Department de chirurgie orthopédique, Hospices Civils de Lyon, Lyon, France
| | - Tristan Ferry
- CIRI – Centre International de Recherche en Infectiologie, Inserm, U1111, Université´ Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France
- Université Claude Bernard Lyon 1, Lyon, France
- Centres de Référence pour la prise en charge des Infections ostéoarticulaires complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France
- Département maladies infectieuses et tropicales, Hospices Civils de Lyon, Lyon, France
| | - Alan Diot
- CIRI – Centre International de Recherche en Infectiologie, Inserm, U1111, Université´ Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France
| | - Frédéric Laurent
- Laboratoire de bactériologie, Institut des Agents Infectieux, French National Reference Center for Staphylococci, Hospices Civils de Lyon, Lyon, France
- CIRI – Centre International de Recherche en Infectiologie, Inserm, U1111, Université´ Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France
- Université Claude Bernard Lyon 1, Lyon, France
- Laboratoire de bactériologie, Institut des Agents Infectieux, French National Reference Center for Staphylococci, Hospices Civils de Lyon, Lyon, France
- Centres de Référence pour la prise en charge des Infections ostéoarticulaires complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France
| | - Florent Valour
- CIRI – Centre International de Recherche en Infectiologie, Inserm, U1111, Université´ Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, F-69007 Lyon, France
- Université Claude Bernard Lyon 1, Lyon, France
- Centres de Référence pour la prise en charge des Infections ostéoarticulaires complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France
- Département maladies infectieuses et tropicales, Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
23
|
Douthit C, Gudenkauf B, Hamood A, Mudaliar N, Caroom C, Jenkins M. Effects of powdered rifampin and vancomycin solutions on biofilm production of staphylococcus aureus on orthopedic implants. J Clin Orthop Trauma 2020; 11:S113-S117. [PMID: 31992930 PMCID: PMC6977531 DOI: 10.1016/j.jcot.2019.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 10/05/2019] [Accepted: 10/08/2019] [Indexed: 11/26/2022] Open
Abstract
PURPOSE Hardware infections in orthopedic surgery, specifically those involving biofilm producing bacteria, are troublesome and are highly resistant to systemic antibiotics. The purpose of this study was to demonstrate the power of rifampin and vancomycin solutions in inhibiting as well as eliminating in vitro on staphylococcus aureus (S. aureus) biofilm in vitro on stainless-steel implants. METHODS A suspension of either S. aureus or a S. aureus containing a plasmid that cods for the green fluorescence protein containing fluorescent protein plasmid was applied to 1 × 1cm sterile stainless steel orthopedic plating material (coupon). Biofilm development was confirmed by; the quantitative assay (colony forming unit [CFU/coupon]) and visualized using confocal laser scanning microscopy. With this established method of biofilm development, we determined the minimum biofilm inhibitory concentration (MBIC) and the minimum biofilm eradication concertation (MBEC) of Rifampicin and Vancomycin. To determine the MBIC, stainless steel plates were subjected to different concentrations of antibiotic solution and inoculated with overnight cultures of S. aureus. After 24 h of incubation at 37 °C, the biofilms on the untreated and antibiotic-treated coupons were quantified. To determine the MBEC, partial S. aureus biofilms were developed on the coupons and then treated with the different concentrations of each antibiotic for 24 h. The number of bacteria within the control untreated as well as treated coupons was determined. RESULTS Both rifampin and vancomycin solutions inhibited biofilm production of S. aureus on stainless steel mediums; the MBIC for rifampin and vancomycin were 80 ng/mL and 1 μg/mL respectively. The MBEC for Rifampicin was similar to the MBIC. However, the MBEC for Vancomycin was 6 mg/ml. CONCLUSIONS When applied to orthopedic stainless steel hardware in vitro, solutions of rifampin and vancomycin powder separately or in combination can completely prevent and eliminate biofilm produced by S. aureus. LEVEL OF EVIDENCE II.
Collapse
Affiliation(s)
- Christian Douthit
- Corresponding author. Texas Tech University Health Sciences Center, Department of Orthopedic Surgery and Rehabilitation, 3601 4th St, Lubbock, TX, 79430, USA.
| | | | | | | | | | | |
Collapse
|
24
|
In Vitro Activity of Rifampin, Rifabutin, Rifapentine, and Rifaximin against Planktonic and Biofilm States of Staphylococci Isolated from Periprosthetic Joint Infection. Antimicrob Agents Chemother 2019; 63:AAC.00959-19. [PMID: 31451499 DOI: 10.1128/aac.00959-19] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/17/2019] [Indexed: 11/20/2022] Open
Abstract
The in vitro activities of rifampin, rifabutin, rifapentine, and rifaximin were tested against 200 periprosthetic joint infection (PJI)-associated staphylococci. Seven rifampin-resistant isolates had MICs of ≥4 μg/ml. Three isolates had rifampin MICs of 0.25 to 1 μg/ml and harbored an Asp471Gly RpoB variant, suggesting that the CLSI rifampin-susceptible staphylococcal breakpoint of ≤1 μg/ml may be too high. The remaining isolates had rifampin MICs of ≤0.016 μg/ml, and the rifampin, rifabutin, rifapentine, and rifaximin minimum biofilm bactericidal concentrations (MBBC) for ≥50% of isolates were 8, 1, 2, and 4 μg/ml (for S. aureus) and 2, 0.06, 0.25, and 0.5 μg/ml (for S. epidermidis), respectively, for rifampin-susceptible isolates. Nonrifampin rifamycins have promising staphylococcal activity, including antibiofilm activity.
Collapse
|
25
|
van Vugt TAG, Arts JJ, Geurts JAP. Antibiotic-Loaded Polymethylmethacrylate Beads and Spacers in Treatment of Orthopedic Infections and the Role of Biofilm Formation. Front Microbiol 2019; 10:1626. [PMID: 31402901 PMCID: PMC6671866 DOI: 10.3389/fmicb.2019.01626] [Citation(s) in RCA: 121] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 07/02/2019] [Indexed: 02/06/2023] Open
Abstract
Polymethylmethacrylate (PMMA) also referred as (acrylic) bone cement is a non-degradable biomaterial that has been used in clinical orthopedic practice for several decades. PMMA can be used in a plain formulation, but is often used in an antibiotic-loaded formulation in (primary and revision) arthroplasty and in treatment of orthopedic infections as prosthetic joint infections (PJI) and chronic osteomyelitis. In treatment of PJIs antibiotic-loaded PMMA is often used as a carrier material for local antibiotic delivery in addition to treatment with systemic antibiotics. In this case, the antibiotic-loaded PMMA is often used as a spacer or as a bead chain. Since the introduction of PMMA as an antibiotic carrier there is a tremendous amount of scientific and clinical papers published, which studied numerous different aspects of antibiotic-loaded PMMA. This paper will review the research regarding basic principles of antibiotic-loaded PMMA as mechanism of action, antibiotic-release capacities, choice of antibiotics and influences on mechanical properties of PMMA. Subsequently, concerns regarding the application of antibiotic-loaded PMMA, biofilm formation, antibiotic resistance and local or systemic toxicity will be discussed. In addition to these subjects, the role of antibiotic loaded PMMA in clinical treatment of PJIs and chronic osteomyelitis is discussed in the final part of this paper.
Collapse
|
26
|
Funk GA, Menuey EM, Cole KA, Schuman TP, Kilway KV, McIff TE. Radical scavenging of poly(methyl methacrylate) bone cement by rifampin and clinically relevant properties of the rifampin-loaded cement. Bone Joint Res 2019; 8:81-89. [PMID: 30915214 PMCID: PMC6397418 DOI: 10.1302/2046-3758.82.bjr-2018-0170.r2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Objectives The objective of this study was to characterize the effect of rifampin incorporation into poly(methyl methacrylate) (PMMA) bone cement. While incompatibilities between the two materials have been previously noted, we sought to identify and quantify the cause of rifampin’s effects, including alterations in curing properties, mechanical strength, and residual monomer content. Methods Four cement groups were prepared using commercial PMMA bone cement: a control; one with 1 g of rifampin; and one each with equimolar amounts of ascorbic acid or hydroquinone relative to the amount of rifampin added. The handling properties, setting time, exothermic output, and monomer loss were measured throughout curing. The mechanical strength of each group was tested over 14 days. A radical scavenging assay was used to assess the scavenging abilities of rifampin and its individual moieties. Results Compared with control, the rifampin-incorporated cement had a prolonged setting time and a reduction in exothermic output during polymerization. The rifampin cement showed significantly reduced strength and was below the orthopaedic weight-bearing threshold of 70 MPa. Based on the radical scavenging assay and strength tests, the hydroquinone structure within rifampin was identified as the polymerization inhibitor. Conclusion The incorporation of rifampin into PMMA bone cement interferes with the cement’s radical polymerization. This interference is due to the hydroquinone moiety within rifampin. This combination alters the cement’s handling and curing properties, and lowers the strength below the threshold for weight-bearing applications. Additionally, the incomplete polymerization leads to increased toxic monomer output, which discourages its use even in non-weight-bearing applications. Cite this article: G. A. Funk, E. M. Menuey, K. A. Cole, T. P. Schuman, K. V. Kilway, T. E. McIff. Radical scavenging of poly(methyl methacrylate) bone cement by rifampin and clinically relevant properties of the rifampin-loaded cement. Bone Joint Res 2019;8:81–89. DOI: 10.1302/2046-3758.82.BJR-2018-0170.R2.
Collapse
Affiliation(s)
- G A Funk
- Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - E M Menuey
- Department of Chemistry, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - K A Cole
- Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - T P Schuman
- Department of Chemistry, Missouri University of Science and Technology, Rolla, Missouri, USA
| | - K V Kilway
- Department of Chemistry, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - T E McIff
- Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
27
|
Trombetta RP, Ninomiya MJ, El-Atawneh IM, Knapp EK, de Mesy Bentley KL, Dunman PM, Schwarz EM, Kates SL, Awad HA. Calcium Phosphate Spacers for the Local Delivery of Sitafloxacin and Rifampin to Treat Orthopedic Infections: Efficacy and Proof of Concept in a Mouse Model of Single-Stage Revision of Device-Associated Osteomyelitis. Pharmaceutics 2019; 11:E94. [PMID: 30813284 PMCID: PMC6410209 DOI: 10.3390/pharmaceutics11020094] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/12/2019] [Accepted: 02/17/2019] [Indexed: 12/15/2022] Open
Abstract
Osteomyelitis is a chronic bone infection that is often treated with adjuvant antibiotic-impregnated poly(methyl methacrylate) (PMMA) cement spacers in multi-staged revisions. However, failure rates remain substantial due to recurrence of infection, which is attributed to the poor performance of the PMMA cement as a drug release device. Hence, the objective of this study was to design and evaluate a bioresorbable calcium phosphate scaffold (CaPS) for sustained antimicrobial drug release and investigate its efficacy in a murine model of femoral implant-associated osteomyelitis. Incorporating rifampin and sitafloxacin, which are effective against bacterial phenotypes responsible for bacterial persistence, into 3D-printed CaPS coated with poly(lactic co-glycolic) acid, achieved controlled release for up to two weeks. Implantation into the murine infection model resulted in decreased bacterial colonization rates at 3- and 10-weeks post-revision for the 3D printed CaPS in comparison to gentamicin-laden PMMA. Furthermore, a significant increase in bone formation was observed for 3D printed CaPS incorporated with rifampin at 3 and 10 weeks. The results of this study demonstrate that osteoconductive 3D printed CaPS incorporated with antimicrobials demonstrate more efficacious bacterial colonization outcomes and bone growth in a single-stage revision in comparison to gentamicin-laden PMMA requiring a two-stage revision.
Collapse
Affiliation(s)
- Ryan P Trombetta
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA.
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY 14642, USA.
| | - Mark J Ninomiya
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY 14642, USA.
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA.
| | - Ihab M El-Atawneh
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA.
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY 14642, USA.
| | - Emma K Knapp
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY 14642, USA.
- Department of Orthopedics, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Karen L de Mesy Bentley
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY 14642, USA.
- Department of Pathology & Laboratory Medicine, University of Rochester, Rochester, NY 14642, USA.
- Department of Orthopedics, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Paul M Dunman
- Department of Microbiology and Immunology, University of Rochester, Rochester, NY 14642, USA.
| | - Edward M Schwarz
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA.
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY 14642, USA.
- Department of Pathology & Laboratory Medicine, University of Rochester, Rochester, NY 14642, USA.
- Department of Orthopedics, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | - Stephen L Kates
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY 14642, USA.
- Department of Orthopaedic Surgery, Virginia Commonwealth University School of Medicine, Richmond, VA 0153, USA.
| | - Hani A Awad
- Department of Biomedical Engineering, University of Rochester, Rochester, NY 14642, USA.
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY 14642, USA.
| |
Collapse
|
28
|
Rifampin and tobramycin combination with PMMA antibiotic cement. EUROPEAN JOURNAL OF ORTHOPAEDIC SURGERY & TRAUMATOLOGY : ORTHOPEDIE TRAUMATOLOGIE 2019; 29:499-500. [PMID: 30232565 DOI: 10.1007/s00590-018-2302-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 09/10/2018] [Indexed: 10/28/2022]
Abstract
Rifampin is a powerful antibiotic used in the treatment of biofilm-forming bacteria with studies supporting its use in PMMA cement. However, it has not been widely accepted for its use in PMMA cement due to a perception that it is unable to solidify in a timely manner. The technique described consists of ratio of aminoglycoside and rifampin that reduces time to solidification to approximately 15-20 min.
Collapse
|
29
|
Role of Rifampin against Staphylococcal Biofilm Infections In Vitro, in Animal Models, and in Orthopedic-Device-Related Infections. Antimicrob Agents Chemother 2019; 63:AAC.01746-18. [PMID: 30455229 DOI: 10.1128/aac.01746-18] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Rifampin has been used as an agent in combination therapy in orthopedic device-related infections (ODRI) for almost three decades. The aim of this review is to provide data regarding the role of rifampin against biofilm infection in vitro, in animal models, and in clinical ODRI. Available data are gathered in order to present the rational use of rifampin combinations in patients with periprosthetic joint infection (PJI). The role of rifampin is well defined in patients with PJI and is indicated in those who fulfill the Infectious Diseases Society of America criteria for debridement and implant retention or one-stage exchange. It should be used with care because of the danger of rapid emergence of resistance. Potential drug interactions should be considered.
Collapse
|
30
|
Shiels SM, Tennent DJ, Wenke JC. Topical rifampin powder for orthopedic trauma part I: Rifampin powder reduces recalcitrant infection in a delayed treatment musculoskeletal trauma model. J Orthop Res 2018; 36:3136-3141. [PMID: 29781552 DOI: 10.1002/jor.24055] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 05/15/2018] [Indexed: 02/04/2023]
Abstract
Open fractures become infected despite meticulous debridement and care. Locally applied antibiotics, commonly embedded in polymethylmethacrylate, deliver high doses of drug directly to the fracture site. Direct application of antibiotic powder, which is being applied prophylactically in spine surgery, is a recent interest in the trauma sector, where bacterial biofilms are more prevalent. Traditional antibiotics, such as vancomycin, are poor performers against bacterial biofilms thus are ineffective in delayed treatment. Rifampin is an effective eradicator of Staphylococcal biofilms. Here, a rat model of musculoskeletal trauma was used to evaluate the utility of locally applied rifampin powder for reducing established orthopedic Staphylococcal infections in a delayed treatment scenario that previously indicated the limited use of local vancomycin. By applying rifampin powder directly to the contaminated segmental defect, the number of bacteria, as well as clinical indications of infection, were significantly reduced compared to vancomycin and daptomycin. Considering the Infectious Disease Society of America's recommendation to use rifampin in combination with another antibiotic to reduce the onset of rifampin resistance, rifampin powder was also applied in combination with vancomycin or daptomycin with insignificant changes in eradication performance. No indications of rifampin resistance were identified. Clinical Significance: The use of locally applied rifampin is a promising therapy for mature and tolerant musculoskeletal infections. Published 2018. This article is a U.S. Government work and is in the public domain in the USA. J Orthop Res 36:3136-3141, 2018.
Collapse
Affiliation(s)
- Stefanie M Shiels
- US Army Institute of Surgical Research, 3698 Chambers Pass, Fort Sam Houston 78234, Texas
| | - David J Tennent
- US Army Institute of Surgical Research, 3698 Chambers Pass, Fort Sam Houston 78234, Texas.,Department of Orthopaedics, Brooke Army Medical Center, Fort Sam Houston, Texas
| | - Joseph C Wenke
- US Army Institute of Surgical Research, 3698 Chambers Pass, Fort Sam Houston 78234, Texas
| |
Collapse
|
31
|
Shiels SM, Tennent DJ, Lofgren AL, Wenke JC. Topical rifampin powder for orthopaedic trauma part II: Topical rifampin allows for spontaneous bone healing in sterile and contaminated wounds. J Orthop Res 2018; 36:3142-3150. [PMID: 30270538 DOI: 10.1002/jor.24155] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 09/24/2018] [Indexed: 02/04/2023]
Abstract
Infectious complications can reduce fracture healing rate. Broad spectrum antibiotics are commonly administered to prevent and treat musculoskeletal infections. Local antibiotics are applied to the wound site to increase therapeutic concentrations without increasing systemic toxicity, however, may hinder local tissue recovery. Rifampin has been shown to eradicate mature Staphylococcal biofilms and its use proven for treating musculoskeletal infections. In this study, a spontaneously healing defect model in a rat was used to investigate the impact rifampin powder has on endogenous bone healing in both a sterile and contaminated wound. No significant differences were identified in bone volume fraction via microcomputed tomography, radiological scoring, or histology between an empty defect and animals that received vancomycin or rifampin powder in a sterile wound. When applied to a contaminated musculoskeletal wound, the rifampin powder had significantly greater bone formation compared to the control, as measured by microcomputed tomography, plain radiology, and histology. In addition, the animals treated with rifampin powder had reduced bacteria, reduced white blood cell count and reduced number of clinical indications of infection. Interestingly, while the vancomycin group still displayed signs of infection via quantitative microbiology, plain radiology, and histology, there was significant bone formation within the defect and reduction of systemic signs of infection. We demonstrated that the use of rifampin powder allows bone to heal in both a sterile and contaminated model of musculoskeletal infection. To our knowledge, this is the first time the direct impact of local antibiotics on bone healing has been investigated. Published 2018. This article is a U.S. Government work and is in the public domain in the USA. J Orthop Res 36:3142-3150, 2018.
Collapse
Affiliation(s)
- Stefanie M Shiels
- U.S. Army Institute of Surgical Research, 3698 Chambers Pass, Fort Sam Houston, Texas, 78234
| | - David J Tennent
- US Army Institute of Surgical Research, Fort Sam Houston, Department of Orthopaedics, Brooke Army Medical Center, Fort Sam Houston, Texas
| | - Alicia L Lofgren
- U.S. Army Institute of Surgical Research, 3698 Chambers Pass, Fort Sam Houston, Texas, 78234
| | - Joseph C Wenke
- U.S. Army Institute of Surgical Research, 3698 Chambers Pass, Fort Sam Houston, Texas, 78234
| |
Collapse
|
32
|
Blanchette KA, Wenke JC. Current therapies in treatment and prevention of fracture wound biofilms: why a multifaceted approach is essential for resolving persistent infections. J Bone Jt Infect 2018; 3:50-67. [PMID: 29761067 PMCID: PMC5949568 DOI: 10.7150/jbji.23423] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/16/2018] [Indexed: 12/13/2022] Open
Abstract
Traumatic orthopedic injuries, particularly extremity wounds, are a significant cause of morbidity. Despite prophylactic antibiotic treatment and surgical intervention, persistent infectious complications can and do occur. Persistent bacterial infections are often caused by biofilms, communities of antibiotic tolerant bacteria encased within a matrix. The structural and metabolic differences in this mode of growth make treatment difficult. Herein, we describe both established and novel, experimental treatments targeted at various stages of wound healing that are specifically aimed at reducing and eliminating biofilm bacteria. Importantly, the highly tolerant nature of these bacterial communities suggests that most singular approaches could be circumvented and a multifaceted, combinatorial approach will be the most effective strategy for treating these complicated infections.
Collapse
Affiliation(s)
| | - Joseph C Wenke
- US Army Institute of Surgical Research, Ft Sam Houston, TX
| |
Collapse
|
33
|
Wang J, Nong XH, Amin M, Qi SH. Hygrocin C from marine-derived Streptomyces sp. SCSGAA 0027 inhibits biofilm formation in Bacillus amyloliquefaciens SCSGAB0082 isolated from South China Sea gorgonian. Appl Microbiol Biotechnol 2017; 102:1417-1427. [PMID: 29189900 DOI: 10.1007/s00253-017-8672-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/19/2017] [Accepted: 11/22/2017] [Indexed: 11/25/2022]
Abstract
Several ansamycins have been reported to inhibit bacterial biofilm formation and accelerate the eradication of developed biofilms, but little is known about the effect of hygrocin C, an ansamycin, on bacterial biofilm formation. Here, hygrocin C was isolated from the marine-derived Streptomyces sp. SCSGAA 0027 and reported for the first time to be capable of inhibiting the biofilm formation of Staphylococcus aureus and Bacillus amyloliquefaciens SCSGAB0082 with the production of anti-microbial lipopeptides from South China Sea gorgonian Subergorgia suberosa at concentrations of less than minimum inhibitory concentrations. Moreover, hygrocin C also promoted the eradication of developed biofilms, affected the biofilm architecture, and lowered the extracellular polymeric matrix formation, cell motility, and surface hydrophobicity in B. amyloliquefaciens, which was in accordance with the inhibition of biofilm formation. Furthermore, transcriptome analysis revealed that hygrocin C altered the transcripts of several genes associated with bacterial chemotaxis and flagellar, two-component system and the synthesis of arginine and histidine, which are important for bacterial biofilm formation. In conclusion, hygrocin C could be used as a potential biofilm inhibitor against S. aureus and B. amyloliquefaciens. But further genetic investigations are needed to provide more details for elucidation of the molecular mechanisms responsible for the effects of hygrocin C on B. amyloliquefaciens biofilm formation.
Collapse
Affiliation(s)
- Jie Wang
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Material Medical, RNAM Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou, 510301, China
- College of Food Science, South China Agricultural University, Guangzhou, 510642, China
| | - Xu-Hua Nong
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Material Medical, RNAM Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou, 510301, China
| | - Muhammad Amin
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Material Medical, RNAM Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou, 510301, China
| | - Shu-Hua Qi
- Key Laboratory of Tropical Marine Bio-resources and Ecology, Guangdong Key Laboratory of Marine Material Medical, RNAM Center for Marine Microbiology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou, 510301, China.
| |
Collapse
|
34
|
Shiels SM, Tennent DJ, Akers KS, Wenke JC. Determining potential of PMMA as a depot for rifampin to treat recalcitrant orthopaedic infections. Injury 2017; 48:2095-2100. [PMID: 28842287 DOI: 10.1016/j.injury.2017.08.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 07/17/2017] [Accepted: 08/10/2017] [Indexed: 02/02/2023]
Abstract
BACKGROUND Open fractures are often complicated by infection. In cases of severe soft tissue and vascular injury, systemic antibiotics may be ineffective due to their inability to reach and provide direct antimicrobial activity to the zone of injury. High antibiotic concentrations within the wound can be achieved with reduced systemic toxicity by using local antibiotic delivery. As bacteria associated with musculoskeletal injuries frequently form biofilms, antibiotic selection is important. Herein, the use of rifampin, an antibiotic with activity against biofilms, delivered via polymethylmethacrylate (PMMA) beads is evaluated for use in a traumatic musculoskeletal wound model. METHODS PMMA beads loaded with rifampin, or combinations of rifampin and vancomycin, were prepared and evaluated for time to curing, drug release kinetics in vitro, and infection prevention in vivo using a well-established rat model of musculoskeletal infection. A segmental bone defect was created and contaminated with methicillin susceptible Staphylococcus aureus (UAMS-1). Wounds were debrided, irrigated, and treated with PMMA beads, containing rifampin or combinations of rifampin plus vancomycin, following a 6-h (early) or 24-h (delayed) treatment. After 14days, tissue, implants, and beads were removed for bacterial quantification and assessed for rifampin resistance. RESULTS There was a direct association between loaded concentration and release kinetics of the rifampin and vancomycin from PMMA beads. Higher rifampin concentrations delayed PMMA curing times. The addition of vancomycin to PMMA resulted in more rapid release of rifampin from beads. However, the highest concentration of rifampin loaded PMMA beads (10% wt/wt) was the only treatment to significantly reduce bacterial counts. No rifampin resistance was observed. CONCLUSION Although higher concentrations of rifampin resulted in significant reductions of bacteria, these levels extended PMMA curing times and transformed PMMA material characteristics. While these characteristics make the material unsuitable for weight-bearing applications, such as total joint arthroplasty, the use of rifampin-loaded PMMA beads may be an effective intervention in a contaminated traumatic extremity wound due to its ability to eradicate biofilms.
Collapse
Affiliation(s)
- Stefanie M Shiels
- Extremity Trauma and Regenerative Medicine Task Area, US Army Institute of Surgical Research, JBSA-Fort Sam Houston, TX, United States.
| | - David J Tennent
- Extremity Trauma and Regenerative Medicine Task Area, US Army Institute of Surgical Research, JBSA-Fort Sam Houston, TX, United States; Department of Orthopaedics, San Antonio Military Medical Center (Brooke Army Medical Center), JBSA-Fort Sam Houston, TX, United States
| | - Kevin S Akers
- US Army Institute of Surgical Research, JBSA-Fort Sam Houston, TX, United States; Infectious Disease Service, San Antonio Military Medical Center (Brooke Army Medical Center), JBSA-Fort Sam Houston, TX, United States
| | - Joseph C Wenke
- Extremity Trauma and Regenerative Medicine Task Area, US Army Institute of Surgical Research, JBSA-Fort Sam Houston, TX, United States
| |
Collapse
|
35
|
Blanchette KA, Prabhakara R, Shirtliff ME, Wenke JC. Inhibition of fracture healing in the presence of contamination by Staphylococcus aureus: Effects of growth state and immune response. J Orthop Res 2017; 35:1845-1854. [PMID: 28387956 DOI: 10.1002/jor.23573] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 03/29/2017] [Indexed: 02/04/2023]
Abstract
Extremity injuries comprise a significant portion of trauma, affecting quality of life, financial burden, and return to duty. Bacterial contamination is commonly associated with failure to heal, despite antibiotic treatment, suggesting that additional therapies must be developed to combat these complications. Treatment failure is likely due to the presence of resistant microbial communities known as biofilms. Biofilm bacteria are able to elicit a direct inhibition of healing through a multitude of known factors. However, they likely also inhibit healing through alteration of the inflammatory response. As inflammation is a critical step in fracture healing, how the presence of biofilm bacteria shifts this response to one that is suboptimal for healing is an important consideration that is currently understudied. The profile of inflammatory factors in response to biofilm bacteria is unique and distinct from those induced during normal healing or by planktonic bacteria alone. This review will examine the presence of inflammatory factors during normal healing and those induced by contaminating bacteria, and will discuss how these differences may ultimately lead to nonunion. Specifically, this review will focus on the Th1/Th2/Th17 type inflammatory responses and how shifts in the balance of these responses during infection can lead to both ineffective clearance and disruption of fracture healing. © 2017 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:1845-1854, 2017.
Collapse
Affiliation(s)
- Krystle A Blanchette
- US Army Institute of Surgical Research, 3698 Chambers Pass STE B, JBSA Ft Sam, Houston 78234-7767, Texas
| | | | | | - Joseph C Wenke
- US Army Institute of Surgical Research, 3698 Chambers Pass STE B, JBSA Ft Sam, Houston 78234-7767, Texas
| |
Collapse
|
36
|
Zimmerli W, Sendi P. Orthopaedic biofilm infections. APMIS 2017; 125:353-364. [PMID: 28407423 DOI: 10.1111/apm.12687] [Citation(s) in RCA: 205] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 01/10/2017] [Indexed: 02/01/2023]
Abstract
Many infections of the musculoskeletal system are biofilm infections that develop on non-living surfaces. Microorganisms adhere either on dead bone (sequesters) or implants. As a rule for a curative concept, chronic osteomyelitis or implant-associated bone infection must be treated with a combination of surgery and antimicrobial therapy. If an implant is kept in place, or a new device is implanted before complete healing of infection, a biofilm-active antibiotic should be used. Rifamycins are active against biofilms of staphylococci, and fluoroquinolones against those of Gram-negative bacilli. In this review, the management of chronic osteomyelitis, periprosthetic joint infection and implant-associated osteomyelitis of long bones is presented.
Collapse
Affiliation(s)
- Werner Zimmerli
- Kantonsspital Baselland, Basel University Medical Clinic and Interdisciplinary Unit for Orthopaedic Infections, Liestal, Switzerland
| | - Parham Sendi
- Department of Infectious Diseases, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
37
|
Post V, Harris LG, Morgenstern M, Geoff Richards R, Sheppard SK, Fintan Moriarty T. Characterization of nasal methicillin-resistant Staphylococcus aureus isolated from international human and veterinary surgeons. J Med Microbiol 2017; 66:360-370. [PMID: 28005521 DOI: 10.1099/jmm.0.000415] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Nasal colonization with methicillin-resistant Staphylococcus aureus (MRSA) is poorly described for surgeons, despite the increased exposure to nosocomial pathogens and at-risk patients. This study investigated the molecular epidemiology and antimicrobial resistance of 26 MRSA isolates cultured from the nares of an international cross-sectional study of 1166 human and 60 veterinary surgeons. METHODOLOGY All isolates were subjected to agr, spa and multilocus sequence typing, and the presence of 22 virulence factors was screened for by PCR. Additionally, biofilm-forming ability, haemolytic activity, staphyloxanthin production and antibiotic resistance were determined. The genome of a rifampicin-resistant MRSA was sequenced. RESULTS Approximately half of the isolates belonged to well-described clonal lineages, ST1, ST5, ST8, ST45 and ST59, that have previously been associated with severe infections and increased patient mortality. Two of the three veterinarian MRSA belonged to epidemic livestock-associated MRSA clonal lineages (ST398 and ST8) previously associated with high transmission potential between animals and humans. The isolates did not display any consistent virulence gene pattern, and 35 % of the isolates carried at least one of the Panton-Valentine leukocidin (lukFS-PV), exfoliative toxin (eta) or toxic shock syndrome (tst) genes. Resistance to rifampicin was detected in one veterinarian isolate and was found to be due to three mutations in the rpoB gene. CONCLUSION Surgeons occupy a critical position in the healthcare profession due to their close contact with patients. In this study, surgeons were found to be colonized with MRSA at low rates, similar to those of the general population, and the colonizing strains were often common clonal lineages.
Collapse
Affiliation(s)
| | - Llinos G Harris
- Department of Microbiology and Infectious Diseases, Swansea University Medical School, Swansea, UK
| | - Mario Morgenstern
- AO Research Institute Davos, Davos, Switzerland.,Department of Orthopedic and Trauma Surgery, University Hospital Basel, Basel, Switzerland
| | | | | | | |
Collapse
|
38
|
Optimizing the treatment of osteomyelitis with antimicrobial drugs: current concepts. CURRENT ORTHOPAEDIC PRACTICE 2017. [DOI: 10.1097/bco.0000000000000477] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
39
|
Post V, Wahl P, Richards RG, Moriarty TF. Vancomycin displays time-dependent eradication of mature Staphylococcus aureus biofilms. J Orthop Res 2017; 35:381-388. [PMID: 27175462 DOI: 10.1002/jor.23291] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Accepted: 04/29/2016] [Indexed: 02/04/2023]
Abstract
UNLABELLED This study was carried out to determine the time and concentration profile required to achieve vancomycin-mediated eradication of Staphylococcus aureus biofilm. This information is critical for the identification of performance targets for local antibiotic delivery vehicles that target biofilm infections. S. aureus UAMS-1 biofilms were grown for 7 days on titanium-aluminium-niobium discs in Mueller Hinton broth. After 7 days, the discs were then incubated in Mueller Hinton broth containing vancomycin at concentrations of 100, 200, 500, 1,000, and 2,000 mg/L. Biofilm eradication was assessed under both static and shaking conditions. Samples were retrieved at regular intervals for up to 28 days for quantification of residual biofilm. One additional disc was processed per time point for scanning electron microscopy. Progressive and significant reduction of viable bacteria was observed over time at all concentrations compared to unexposed controls. After 28 days under static conditions, the S. aureus biofilm was completely eradicated at 200 mg/L vancomycin and higher concentrations, but not at 100 mg/L. In contrast, bacterial biofilm could not be eradicated under shaking conditions at any concentration. CLINICAL SIGNIFICANCE The present study shows that it is possible to eradicate mature S. aureus biofilm from metal implants by vancomycin alone although the time concentration profile required cannot be achieved by systemic administration or any of the local delivery vehicles currently available. Identifying targets for antibiotic delivery is the first step in developing fit for purpose local antibiotic delivery vehicles that will successfully and predictably treat established biofilm infection. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 35:381-388, 2017.
Collapse
Affiliation(s)
- Virginia Post
- AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz, 7270, Switzerland
| | - Peter Wahl
- Division for Orthopaedic and Trauma Surgery, Cantonal Hospital Winterthur, Winterthur, Switzerland
| | - R Geoff Richards
- AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz, 7270, Switzerland
| | - T Fintan Moriarty
- AO Research Institute Davos, Clavadelerstrasse 8, Davos Platz, 7270, Switzerland
| |
Collapse
|
40
|
Time-Dependent Effectiveness of Locally Applied Vancomycin Powder in a Contaminated Traumatic Orthopaedic Wound Model. J Orthop Trauma 2016; 30:531-7. [PMID: 27124826 DOI: 10.1097/bot.0000000000000617] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVES To evaluate the effectiveness of locally applied vancomycin powder at different times postinfection in a contaminated traumatic animal model. METHODS This study used an established segmental defect rat femur model contaminated with Staphylococcus aureus UAMS-1 followed by treatment at 6 or 24 hours postinfection. Three treatments were evaluated: debridement and irrigation alone (control group) or in combination with either vancomycin powder or vancomycin-impregnated poly(methyl methacrylate) beads. Serum vancomycin levels were determined at scheduled time points over 14 days; bone, surrounding muscle, and implants were harvested for bacterial and inflammatory analyses. RESULTS Locally applied vancomycin powder and impregnated beads significantly reduced bacteria both within the bone and implant when treatment was performed at 6 hours. Delaying treatment to 24 hours significantly reduced the therapeutic efficacy of locally applied vancomycin of both groups. Serum vancomycin levels were detectable in all animals treated with vancomycin powder at 24 hours, but absorption was negligible from beads. At 14 days, vancomycin was detectable in the surrounding musculature of all animals and in serum of 20% of animals treated with vancomycin powder. CONCLUSIONS This study suggests that vancomycin powder is a promising adjunctive therapy for preventing infection in traumatic wounds when treatment is performed early. This time-dependent effectiveness of vancomycin powder is similar to that observed with systemic and other local delivery adjuncts, which is likely attributable to biofilm formation after contamination, conferring intrinsic recalcitrance to antimicrobials.
Collapse
|
41
|
Kluin OS, Busscher HJ, Neut D, van der Mei HC. Poly(trimethylene carbonate) as a carrier for rifampicin and vancomycin to target therapy-recalcitrant staphylococcal biofilms. J Orthop Res 2016; 34:1828-1837. [PMID: 26876651 DOI: 10.1002/jor.23194] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 02/08/2016] [Indexed: 02/04/2023]
Abstract
Standard antibiotic therapy in osteomyelitis patients is of limited value when methicillin-resistant Staphylococcus aureus (MRSA), Staphylococcus epidermidis (MRSE), or small-colony variants (SCV) are present. Far better results are obtained by local drug delivery of antibiotic combinations including rifampicin, using a suitable carrier. We therefore investigated release kinetics of antibiotics from biodegradable poly(trimethylene carbonate) (PTMC) and in vitro biofilm inhibition of MRSA, MRSE, and S. aureus SCV strains in the course of 24, 72, and 168 h treatment by PTMC, either unloaded, gentamicin-loaded, loaded with rifampicin and fosfomycin, or rifampicin and vancomycin. PTMC appeared to be a suitable carrier for rifampicin alone or in combination with other antibiotics. Biofilm colony forming units and metabolic activity measurement (MTT assay) demonstrated significant (p < 0.05) inhibition for all strains when PTMC loaded with rifampicin and vancomycin was employed, especially after 168 h treatment. Confocal laser scanning microscopy images showed similar qualitative results. PTMC loaded with only gentamicin did not show any inhibition. This exemplifies that PTMC loaded with rifampicin and vancomycin holds promise for the treatment of recalcitrant osteomyelitis. © 2016 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 34:1828-1837, 2016.
Collapse
Affiliation(s)
- Otto S Kluin
- Department of Biomedical Engineering, University of Groningen and University Medical Center Groningen, Antonius Deusinglaan 1, 9713, AV Groningen, The Netherlands
| | - Henk J Busscher
- Department of Biomedical Engineering, University of Groningen and University Medical Center Groningen, Antonius Deusinglaan 1, 9713, AV Groningen, The Netherlands
| | - Daniëlle Neut
- Department of Biomedical Engineering, University of Groningen and University Medical Center Groningen, Antonius Deusinglaan 1, 9713, AV Groningen, The Netherlands.
| | - Henny C van der Mei
- Department of Biomedical Engineering, University of Groningen and University Medical Center Groningen, Antonius Deusinglaan 1, 9713, AV Groningen, The Netherlands
| |
Collapse
|
42
|
Ma Z, Lynch AS. Development of a Dual-Acting Antibacterial Agent (TNP-2092) for the Treatment of Persistent Bacterial Infections. J Med Chem 2016; 59:6645-57. [PMID: 27336583 DOI: 10.1021/acs.jmedchem.6b00485] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The clinical management of prosthetic joint infections and other persistent bacterial infections represents a major unmet medical need. The rifamycins are one of the most potent antibiotic classes against persistent bacterial infections, but bacteria can develop resistance to rifamycins rapidly and the clinical utility of the rifamycin class is typically limited to antibiotic combinations to minimize the development of resistance. To develop a better therapy against persistent bacterial infections, a series of rifamycin based bifunctional molecules were designed, synthesized, and evaluated with the goal to identify a dual-acting drug that maintains the potent activity of rifamycins against persistent pathogens and at the same time minimize the development of rifamycin resistance. TNP-2092 was identified as a drug candidate and is currently in an early stage of clinical development for the treatment of prosthetic joint infections.
Collapse
Affiliation(s)
- Zhenkun Ma
- TenNor Therapeutics Ltd. , 218 Xinghu Street, Suzhou Industrial Park, Suzhou 215123, China
| | - Anthony Simon Lynch
- Janssen Research & Development LLC. , 1400 McKean Road, Spring House, Pennsylvania 18940, United States
| |
Collapse
|
43
|
Update on the Management of Pediatric Acute Osteomyelitis and Septic Arthritis. Int J Mol Sci 2016; 17:ijms17060855. [PMID: 27258258 PMCID: PMC4926389 DOI: 10.3390/ijms17060855] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 05/26/2016] [Accepted: 05/26/2016] [Indexed: 12/15/2022] Open
Abstract
Acute osteomyelitis and septic arthritis are two infections whose frequencies are increasing in pediatric patients. Acute osteomyelitis and septic arthritis need to be carefully assessed, diagnosed, and treated to avoid devastating sequelae. Traditionally, the treatment of acute osteoarticular infection in pediatrics was based on prolonged intravenous anti-infective therapy. However, results from clinical trials have suggested that in uncomplicated cases, a short course of a few days of parenteral antibiotics followed by oral therapy is safe and effective. The aim of this review is to provide clinicians an update on recent controversies and advances regarding the management of acute osteomyelitis and septic arthritis in children. In recent years, the emergence of bacterial species resistant to commonly used antibiotics that are particularly aggressive highlights the necessity for further research to optimize treatment approaches and to develop new molecules able to fight the war against acute osteoarticular infection in pediatric patients.
Collapse
|
44
|
Crabol Y, Catherinot E, Veziris N, Jullien V, Lortholary O. Rifabutin: where do we stand in 2016? J Antimicrob Chemother 2016; 71:1759-71. [PMID: 27009031 DOI: 10.1093/jac/dkw024] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Rifabutin is a spiro-piperidyl-rifamycin structurally closely related to rifampicin that shares many of its properties. We attempted to address the reasons why this drug, which was recently recognized as a WHO Essential Medicine, still had a far narrower range of indications than rifampicin, 24 years after its launch. In this comprehensive review of the classic and more recent rifabutin experimental and clinical studies, the current state of knowledge about rifabutin is depicted, relying on specific pharmacokinetics, pharmacodynamics, antimicrobial properties, resistance data and side effects compared with rifampicin. There are consistent in vitro data and clinical studies showing that rifabutin has at least equivalent activity/efficacy and acceptable tolerance compared with rifampicin in TB and non-tuberculous mycobacterial diseases. Clinical studies have emphasized the clinical benefits of low rifabutin liver induction in patients with AIDS under PIs, in solid organ transplant patients under immunosuppressive drugs or in patients presenting intolerable side effects related to rifampicin. The contribution of rifabutin for rifampicin-resistant, but rifabutin-susceptible, Mycobacterium tuberculosis isolates according to the present breakpoints has been challenged and is now controversial. Compared with rifampicin, rifabutin's lower AUC is balanced by higher intracellular penetration and lower MIC for most pathogens. Clinical studies are lacking in non-mycobacterial infections.
Collapse
Affiliation(s)
- Yoann Crabol
- APHP-Hôpital Necker-Enfants malades, Service de Maladies Infectieuses et Tropicales, Centre d'Infectiologie Necker-Pasteur, Paris, France
| | | | - Nicolas Veziris
- AP-HP, Hôpital Pitié-Salpêtrière, Laboratoire de Bactériologie-Hygiène, Centre National de Référence des Mycobactéries et de la Résistance des Mycobactéries aux Antituberculeux, Paris, France UPMC, INSERM, Centre d'Immunologie et des Maladies Infectieuses, E13, Paris, France
| | - Vincent Jullien
- AP-HP, Hôpital Européen Georges-Pompidou, Pharmacology Department, Paris, France Université Paris Descartes, Sorbonne Paris Cité, Inserm U1129, Paris, France
| | - Olivier Lortholary
- APHP-Hôpital Necker-Enfants malades, Service de Maladies Infectieuses et Tropicales, Centre d'Infectiologie Necker-Pasteur, Paris, France Université Paris Descartes, Sorbonne Paris Cité, Paris, France IHU Imagine, Paris, France
| |
Collapse
|
45
|
Activity of Gallium Meso- and Protoporphyrin IX against Biofilms of Multidrug-Resistant Acinetobacter baumannii Isolates. Pharmaceuticals (Basel) 2016; 9:ph9010016. [PMID: 26999163 PMCID: PMC4812380 DOI: 10.3390/ph9010016] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Revised: 03/12/2016] [Accepted: 03/15/2016] [Indexed: 11/17/2022] Open
Abstract
Acinetobacter baumannii is a challenging pathogen due to antimicrobial resistance and biofilm development. The role of iron in bacterial physiology has prompted the evaluation of iron-modulation as an antimicrobial strategy. The non-reducible iron analog gallium(III) nitrate, Ga(NO3)3, has been shown to inhibit A. baumannii planktonic growth; however, utilization of heme-iron by clinical isolates has been associated with development of tolerance. These observations prompted the evaluation of iron-heme sources on planktonic and biofilm growth, as well as antimicrobial activities of gallium meso- and protoporphyrin IX (Ga-MPIX and Ga-PPIX), metal heme derivatives against planktonic and biofilm bacteria of multidrug-resistant (MDR) clinical isolates of A. baumannii in vitro. Ga(NO3)3 was moderately effective at reducing planktonic bacteria (64 to 128 µM) with little activity against biofilms (≥512 µM). In contrast, Ga-MPIX and Ga-PPIX were highly active against planktonic bacteria (0.25 to 8 µM). Cytotoxic effects in human fibroblasts were observed following exposure to concentrations exceeding 128 µM of Ga-MPIX and Ga-PPIX. We observed that the gallium metal heme conjugates were more active against planktonic and biofilm bacteria, possibly due to utilization of heme-iron as demonstrated by the enhanced effects on bacterial growth and biofilm formation.
Collapse
|
46
|
An In Vitro Comparison of PMMA and Calcium Sulfate as Carriers for the Local Delivery of Gallium(III) Nitrate to Staphylococcal Infected Surgical Sites. BIOMED RESEARCH INTERNATIONAL 2016; 2016:7078989. [PMID: 26885514 PMCID: PMC4739006 DOI: 10.1155/2016/7078989] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 12/24/2015] [Indexed: 11/17/2022]
Abstract
Antibiotic-loaded bone cements, including poly(methyl methacrylate) (PMMA) and calcium sulfate (CaSO4), are often used for treatment of orthopaedic infections involving Staphylococcus spp., although the effectiveness of this treatment modality may be limited due to the emergence of antimicrobial resistance and/or the development of biofilms within surgical sites. Gallium(III) is an iron analog capable of inhibiting essential iron-dependent pathways, exerting broad antimicrobial activity against multiple microorganisms, including Staphylococcus spp. Herein, we evaluated PMMA and CaSO4 as carriers for delivery of gallium(III) nitrate (Ga(NO3)3) to infected surgical sites by assessing the release kinetics subsequent to incorporation and antimicrobial activity against S. aureus and S. epidermidis. PMMA and to a lesser extent CaSO4 were observed to be compatible as carriers for Ga(NO3)3, eluting concentrations with antimicrobial activity against planktonic bacteria, inhibiting bacterial growth, and preventing bacterial colonization of beads, and effective against established bacterial biofilms of S. aureus and S. epidermidis. Collectively, our in vitro results indicate that PMMA is a more suitable carrier compared to CaSO4 for delivery of Ga(NO3)3; moreover they provide evidence for the potential use of Ga(NO3)3 with PMMA as a strategy for the prevention and/or treatment for orthopaedic infections.
Collapse
|