1
|
Wakabayashi H, Hasegawa M, Naito Y, Tone S, Sudo A. Long-Term Outcome of Treating Periprosthetic Hip Joint Infection with Local Antibiotics Delivered Through Antibiotic-Impregnated Calcium Hydroxyapatite. J Clin Med 2024; 13:7469. [PMID: 39685927 DOI: 10.3390/jcm13237469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
Background/Objectives: This study explores the long-term clinical outcomes of antibiotic-impregnated calcium hydroxyapatite (CHA) as an antibiotic delivery system in treating periprosthetic joint infection (PJI) following total hip arthroplasty (THA). Methods: We conducted a retrospective analysis of 12 patients (13 hips) who were treated with antibiotic-impregnated CHA for PJI after THA and followed for more than 10 years at our institution between 1999 and 2011. The study group comprised six men (seven hips) and six women, with a mean age of 61.4 years. Results: The mean follow-up duration was 13.8 years. After irrigation and debridement with modular component exchange, seven hips in six patients underwent revision surgery; however, PJI relapsed in two hips of two patients with a history of diabetes. Two-stage revision surgery was performed on the two relapsed hips and six scheduled hips with antibiotic-impregnated CHA used to treat all cases of PJI. Infection control (100% rate) was achieved in all joints, and revision surgeries were completed. Two patients died 12 years after the initial procedure, and one died 14 years after the first procedure due to unrelated internal diseases; no infection recurrence was observed. No complications related to antibiotic-impregnated CHA were observed. Conclusions: Our results indicate that antibiotic-impregnated CHA is associated with high success rates in treating PJI after THA, even in cases with advanced disease, and yields satisfactory functional outcomes postoperatively.
Collapse
Affiliation(s)
- Hiroki Wakabayashi
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu 514-8507, Japan
| | - Masahiro Hasegawa
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu 514-8507, Japan
| | - Yohei Naito
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu 514-8507, Japan
| | - Shine Tone
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu 514-8507, Japan
| | - Akihiro Sudo
- Department of Orthopaedic Surgery, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu 514-8507, Japan
| |
Collapse
|
2
|
Liu J, Weng Q, Da D, Yao S, Zhang Y, Wu Y. Enhancement of Fluoride's Antibacterial and Antibiofilm Effects against Oral Staphylococcus aureus by the Urea Derivative BPU. Antibiotics (Basel) 2024; 13:930. [PMID: 39452197 PMCID: PMC11505350 DOI: 10.3390/antibiotics13100930] [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: 09/02/2024] [Revised: 09/21/2024] [Accepted: 09/25/2024] [Indexed: 10/26/2024] Open
Abstract
Background: The oral cavity is an important but often overlooked reservoir for Staphylococcus aureus. The effective control and prevention of S. aureus colonization and infection in the oral and maxillofacial regions are crucial for public health. Fluoride is widely used in dental care for its remineralization and antibacterial properties. However, its effectiveness against S. aureus has not been thoroughly investigated. Objectives: This study aimed to evaluate the potential of combining sodium fluoride (NaF) with compounds to enhance its antibacterial and antibiofilm effects against S. aureus. Method: We found that a urea derivative significantly enhances the efficacy of fluoride by promoting the retention of fluoride ions within the cells. The synergistic antibacterial and antibiofilm effects of BPU with NaF were confirmed through various assays, including checkerboard assays, time-kill assays, and growth curve analysis. These findings were further supported by additional methods, including transmission electron microscopy (TEM), in silico simulations, and gene overexpression studies. Results: These findings suggest that targeting fluoride ion membrane exporters could enhance antibacterial efficacy. When combined with fluoride, 1,3-Bis [3,5-bis(trifluoromethyl)phenyl]urea (BPU) showed increased effectiveness in inhibiting S. aureus growth and reducing established biofilms. Conclusions: This novel combination represents a promising therapeutic strategy for treating biofilm-associated S. aureus infections, offering a new strategy in oral healthcare. To fully evaluate the clinical potential of this synergistic therapy, further in vivo studies are essential.
Collapse
Affiliation(s)
- Jia Liu
- Department of Preventive Dentistry, Shanghai Stomatological Hospital, School of Stomatology, Fudan University, Shanghai 200120, China; (J.L.); (Q.W.); (D.D.); (S.Y.)
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai 200120, China
| | - Qingqing Weng
- Department of Preventive Dentistry, Shanghai Stomatological Hospital, School of Stomatology, Fudan University, Shanghai 200120, China; (J.L.); (Q.W.); (D.D.); (S.Y.)
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai 200120, China
| | - Dongxin Da
- Department of Preventive Dentistry, Shanghai Stomatological Hospital, School of Stomatology, Fudan University, Shanghai 200120, China; (J.L.); (Q.W.); (D.D.); (S.Y.)
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai 200120, China
| | - Shuran Yao
- Department of Preventive Dentistry, Shanghai Stomatological Hospital, School of Stomatology, Fudan University, Shanghai 200120, China; (J.L.); (Q.W.); (D.D.); (S.Y.)
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai 200120, China
| | - Ying Zhang
- Department of Preventive Dentistry, Shanghai Stomatological Hospital, School of Stomatology, Fudan University, Shanghai 200120, China; (J.L.); (Q.W.); (D.D.); (S.Y.)
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai 200120, China
| | - Yang Wu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Science, Institutes of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
3
|
Dai T, Ma C, Zhang F, Wang H, Ma Z, Wang H, Wen Y, Chen L. The Efficacy and Safety of an Intra-articular Dual-Acting Antibacterial Agent (TNP-2092) for Implant Infection-Associated Methicillin-Resistant Staphylococcus aureus. J Infect Dis 2024; 229:1658-1668. [PMID: 38147364 DOI: 10.1093/infdis/jiad588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/01/2023] [Accepted: 12/22/2023] [Indexed: 12/27/2023] Open
Abstract
Owing to the presence of microbial biofilm on the implant, the eradication of biofilm-associated infections poses a challenge for antibiotic therapies. The study aimed to investigate the efficacy and safety of the novel antibiotic agent TNP-2092 in the context of implant infections. In vivo, rats with periprosthetic joint infection (PJI) treated with antibiotics showed an increase in body weight and decrease in swelling, temperature, and width of knee, compared with the control group. Meanwhile, inflammatory markers in synovium and serum were decreased in the TNP-2092 group, consistent with the pathological results. Moreover, TNP-2092 was effective in eliminating bacteria and disruption biofilm formation, and further alleviated the abnormal bone absorption and reactive bone changes around the prosthesis. In conclusion, intra-articular injection of TNP-2092 is safe and effective in treating knee PJI in a rat model. The study provides a foundation for the future utilization of TNP-2092 in the management of implant-related infections.
Collapse
Affiliation(s)
- Tianyu Dai
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan
| | - Chi Ma
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan
- Department of Orthopedics, The First Affiliated Hospital of Jishou University, Jishou
| | - Fan Zhang
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan
| | - Zhenkun Ma
- Department of Pharmacology & Toxicology, TenNor Therapeutics, Suzhou
| | - Huan Wang
- Department of Pharmacology & Toxicology, TenNor Therapeutics, Suzhou
| | - Yinxian Wen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan
- Joint Disease Research Center of Wuhan University, Wuhan University, Wuhan, China
| | - Liaobin Chen
- Division of Joint Surgery and Sports Medicine, Department of Orthopedic Surgery, Zhongnan Hospital of Wuhan University, Wuhan
- Joint Disease Research Center of Wuhan University, Wuhan University, Wuhan, China
| |
Collapse
|
4
|
Carbonell-Rosell C, Lakhani K, Lung M, Nadal P, Rodriguez-Pardo D, Corona PS. Etiology and antimicrobial resistance patterns in chronic osteomyelitis of the tibia: an 11-year clinical experience. Arch Orthop Trauma Surg 2024; 144:773-781. [PMID: 38133804 DOI: 10.1007/s00402-023-05095-3] [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: 12/28/2022] [Accepted: 09/27/2023] [Indexed: 12/23/2023]
Abstract
PURPOSE To analyze changes in tendency of etiology and of antimicrobial resistance patterns to most common local and systemic antibiotics in chronic osteomyelitis of the tibia (COM-T) in a Level I trauma center over an 11-year period. METHODS A retrospective review including all patients with COM-T who were surgically treated from January 2009 to December 2019. Patients were divided into two period groups: 2009-2014 and 2015-2019. Microbiologic etiology was analyzed. Bacterial resistance patterns evaluation was based on the Magiorakos et al. classification, including proportions of multidrug-resistant organisms (MDROs, acquired non-susceptibility to at least one agent in three or more antimicrobial categories), extensively drug-resistant (XDR) and pan drug-resistant (PDR) organisms encountered. RESULTS A total of 173 episodes of COM-T were identified. Monomicrobial infections represented 47.4% of all cases, while 28.3% had polymicrobial infections. Negative deep-bone cultures were identified in 24.3% of the patients. The most commonly isolated microorganisms were coagulase-negative Staphylococci (24.5%) and S. aureus (20.5%). No differences were found when comparing Gram-positive infections between periods (58.3% for 2009-2014 vs. 46.7% for 2015-2019; p = 0.10). Findings were similar for Gram-negative infections (37% vs. 33.7%; p = 0.62), although more polymicrobial infections were detected (24.7% vs. 33.3%, respectively; p = 0.359). MDROs were involved in 15% of the cases, with an upward trend when comparing both periods (12.8% vs. 23.6%; p = 0.07). The most-used combination of local antibiotics-glycopeptide (vancomycin) plus aminoglycoside (gentamicin or tobramycin)-was met with low rates of resistance in the most frequently isolated microorganisms. CONCLUSION According to the results of the present study, rates of Gram-positive and Gram-negative infections remained consistent during the two study periods, but with an upward trend in MDRO and polymicrobial infections detected. The local combination of a glycopeptide plus an aminoglycoside was effective in treating the most frequently isolated microorganisms.
Collapse
Affiliation(s)
- Carla Carbonell-Rosell
- Orthopaedic Surgery and Traumatology Department, Hospital Universitari Vall d'Hebron, Passeig Vall d'Hebron, 119-129, 08035, Barcelona, Spain
- Department of Surgery, Universitat Autònoma de Barcelona, 08193, Barcelona, Spain
| | - Kushal Lakhani
- Orthopaedic Surgery and Traumatology Department, Hospital Universitari Vall d'Hebron, Passeig Vall d'Hebron, 119-129, 08035, Barcelona, Spain.
- Septic and Reconstructive Surgery Unit, Orthopaedic Surgery and Traumatology Department, Hospital Universitari Vall d'Hebron, Passeig Vall d'Hebron, 119-129, Barcelona, Spain.
- Department of Surgery, Universitat Autònoma de Barcelona, 08193, Barcelona, Spain.
| | - Mayli Lung
- Microbiology Department, Hospital Universitari Vall d'Hebron, Passeig Vall d'Hebron, 119-129, Barcelona, Spain
- CIBERINFEC, Madrid, Spain
- Department of Surgery, Universitat Autònoma de Barcelona, 08193, Barcelona, Spain
| | - Patricia Nadal
- Microbiology Department, Hospital Universitari Vall d'Hebron, Passeig Vall d'Hebron, 119-129, Barcelona, Spain
- Department of Surgery, Universitat Autònoma de Barcelona, 08193, Barcelona, Spain
| | - Dolors Rodriguez-Pardo
- Infectious Diseases Department, Hospital Universitari Vall d'Hebron, Passeig Vall d'Hebron, 119-129, Barcelona, Spain
- CIBERINFEC, Madrid, Spain
- Department of Surgery, Universitat Autònoma de Barcelona, 08193, Barcelona, Spain
| | - Pablo S Corona
- Orthopaedic Surgery and Traumatology Department, Hospital Universitari Vall d'Hebron, Passeig Vall d'Hebron, 119-129, 08035, Barcelona, Spain
- Septic and Reconstructive Surgery Unit, Orthopaedic Surgery and Traumatology Department, Hospital Universitari Vall d'Hebron, Passeig Vall d'Hebron, 119-129, Barcelona, Spain
- Musculoskeletal Tissue Engineering Group, Vall d'Hebron Research Institute, Barcelona, Spain
- Department of Surgery, Universitat Autònoma de Barcelona, 08193, Barcelona, Spain
| |
Collapse
|
5
|
Yue Y, Chen K, Sun C, Ahmed S, Ojha SC. Antimicrobial peptidase lysostaphin at subinhibitory concentrations modulates staphylococcal adherence, biofilm formation, and toxin production. BMC Microbiol 2023; 23:311. [PMID: 37884887 PMCID: PMC10601153 DOI: 10.1186/s12866-023-03052-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Accepted: 10/09/2023] [Indexed: 10/28/2023] Open
Abstract
BACKGROUND The ability of antimicrobial agents to affect microbial adherence to eukaryotic cell surfaces is a promising antivirulence strategy for combating the global threat of antimicrobial resistance. Inadequate use of antimicrobials has led to widespread instances of suboptimal antibiotic concentrations around infection sites. Therefore, we aimed to examine the varying effect of an antimicrobial peptidase lysostaphin (APLss) on staphylococcal adherence to host cells, biofilm biomass formation, and toxin production as a probable method for mitigating staphylococcal virulence. RESULTS Initially, soluble expression in E. coli and subsequent purification by immobilized-Ni2+ affinity chromatography (IMAC) enabled us to successfully produce a large quantity of highly pure ~ 28-kDa His-tagged mature APLss. The purified protein exhibited potent inhibitory effects against both methicillin-sensitive and methicillin-resistant staphylococcal strains, with minimal inhibitory concentrations (MICs) ranging from 1 to 2 µg/mL, and ultrastructural analysis revealed that APLss-induced concentration-specific changes in the morphological architecture of staphylococcal surface membranes. Furthermore, spectrophotometric and fluorescence microscopy revealed that incubating staphylococcal strains with sub-MIC and MIC of APLss significantly inhibited staphylococcal adherence to human vaginal epithelial cells and biofilm biomass formation. Ultimately, transcriptional investigations revealed that APLss inhibited the expression of agrA (quorum sensing effector) and other virulence genes related to toxin synthesis. CONCLUSIONS Overall, APLss dose-dependently inhibited adhesion to host cell surfaces and staphylococcal-associated virulence factors, warranting further investigation as a potential anti-staphylococcal agent with an antiadhesive mechanism of action using in vivo models of staphylococcal toxic shock syndrome.
Collapse
Affiliation(s)
- Yuan Yue
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Shaanxi Normal University, Xi'an, China
| | - Ke Chen
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Southwest Medical University, Jiangyang District, Luzhou, 646000, Sichuan, China
| | - Changfeng Sun
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Southwest Medical University, Jiangyang District, Luzhou, 646000, Sichuan, China
| | - Sarfraz Ahmed
- Wellman Centre for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston MA 02114, USA
| | - Suvash Chandra Ojha
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Southwest Medical University, Jiangyang District, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
6
|
Moore K, Li A, Gupta N, Gupta TT, Delury C, Aiken SS, Laycock PA, Stoodley P. Killing of a Multispecies Biofilm Using Gram-Negative and Gram-Positive Targeted Antibiotic Released from High Purity Calcium Sulfate Beads. Microorganisms 2023; 11:2296. [PMID: 37764142 PMCID: PMC10538001 DOI: 10.3390/microorganisms11092296] [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: 06/28/2023] [Revised: 08/31/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Multispecies biofilm orthopedic infections are more challenging to treat than mono-species infections. In this in-vitro study, we aimed to determine if a multispecies biofilm, consisting of Gram positive and negative species with different antibiotic susceptibilities could be treated more effectively using high purity antibiotic-loaded calcium sulfate beads (HP-ALCSB) containing vancomycin (VAN) and tobramycin (TOB) in combination than alone. METHODS Three sets of species pairs from bioluminescent strains of Pseudomonas aeruginosa (PA) and Staphylococcus aureus (SA) and clinical isolates, Enterococcus faecalis (EF) and Enterobacter cloacae were screened for compatibility. PA + EF developed intermixed biofilms with similar cell concentrations and so were grown on 316L stainless steel coupons for 72 h or as 24 h agar lawn biofilms and then treated with HP-ALCSBs with single or combination antibiotics and assessed by viable count or bioluminescence and light imaging to distinguish each species. Replica plating was used to assess viability. RESULTS The VAN + TOB bead significantly reduced the PA + EF biofilm CFU and reduced the concentration of surviving antibiotic tolerant variants by 50% compared to single antibiotics. CONCLUSIONS The combination of Gram-negative and positive targeted antibiotics released from HP-ALCSBs may be more effective in treating multispecies biofilms than monotherapy alone.
Collapse
Affiliation(s)
- Kelly Moore
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA; (K.M.); (A.L.); (N.G.); (T.T.G.)
| | - Anthony Li
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA; (K.M.); (A.L.); (N.G.); (T.T.G.)
| | - Niraj Gupta
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA; (K.M.); (A.L.); (N.G.); (T.T.G.)
| | - Tripti Thapa Gupta
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA; (K.M.); (A.L.); (N.G.); (T.T.G.)
| | - Craig Delury
- Biocomposites Ltd., Keele Science Park, Keele, Staffordshire ST5 5NL, UK; (C.D.); (S.S.A.); (P.A.L.)
| | - Sean S. Aiken
- Biocomposites Ltd., Keele Science Park, Keele, Staffordshire ST5 5NL, UK; (C.D.); (S.S.A.); (P.A.L.)
| | - Phillip A. Laycock
- Biocomposites Ltd., Keele Science Park, Keele, Staffordshire ST5 5NL, UK; (C.D.); (S.S.A.); (P.A.L.)
| | - Paul Stoodley
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, OH 43210, USA; (K.M.); (A.L.); (N.G.); (T.T.G.)
- Department of Orthopedics, The Ohio State University, Columbus, OH 43203, USA
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
- National Centre for Advanced Tribology at Southampton (nCATS), Department of Mechanical Engineering, University of Southampton, Southampton SO17 1BJ, UK
- National Biofilm Innovation Centre (NBIC), University of Southampton, Southampton SO17 1BJ, UK
| |
Collapse
|
7
|
Oliveira AS, Saraiva LM, Carvalho SM. Staphylococcus epidermidis biofilms undergo metabolic and matrix remodeling under nitrosative stress. Front Cell Infect Microbiol 2023; 13:1200923. [PMID: 37469594 PMCID: PMC10352803 DOI: 10.3389/fcimb.2023.1200923] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/05/2023] [Indexed: 07/21/2023] Open
Abstract
Staphylococcus epidermidis is a commensal skin bacterium that forms host- and antibiotic-resistant biofilms that are a major cause of implant-associated infections. Most research has focused on studying the responses to host-imposed stresses on planktonic bacteria. In this work, we addressed the open question of how S. epidermidis thrives on toxic concentrations of nitric oxide (NO) produced by host innate immune cells during biofilm assembly. We analyzed alterations of gene expression, metabolism, and matrix structure of biofilms of two clinical isolates of S. epidermidis, namely, 1457 and RP62A, formed under NO stress conditions. In both strains, NO lowers the amount of biofilm mass and causes increased production of lactate and decreased acetate excretion from biofilm glucose metabolism. Transcriptional analysis revealed that NO induces icaA, which is directly involved in polysaccharide intercellular adhesion (PIA) production, and genes encoding proteins of the amino sugar pathway (glmM and glmU) that link glycolysis to PIA synthesis. However, the strains seem to have distinct regulatory mechanisms to boost lactate production, as NO causes a substantial upregulation of ldh gene in strain RP62A but not in strain 1457. The analysis of the matrix components of the staphylococcal biofilms, assessed by confocal laser scanning microscopy (CLSM), showed that NO stimulates PIA and protein production and interferes with biofilm structure in a strain-dependent manner, but independently of the Ldh level. Thus, NO resistance is attained by remodeling the staphylococcal matrix architecture and adaptation of main metabolic processes, likely providing in vivo fitness of S. epidermidis biofilms contacting NO-proficient macrophages.
Collapse
|
8
|
Taha M, Arulanandam R, Chen A, Diallo JS, Abdelbary H. Combining povidone-iodine with vancomycin can be beneficial in reducing early biofilm formation of methicillin-resistant Staphylococcus aureus and methicillin-sensitive S. aureus on titanium surface. J Biomed Mater Res B Appl Biomater 2023; 111:1133-1141. [PMID: 36632686 DOI: 10.1002/jbm.b.35220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 12/09/2022] [Accepted: 12/23/2022] [Indexed: 01/13/2023]
Abstract
There is controversial clinical evidence regarding the added antibacterial benefit of locally administering antiseptic solutions or antibiotics to the infected joint space. The objectives of this in vitro study were to test the efficacy of povidone-iodine (PVP-I) and vancomycin in treating premature and developed Staphylococcus aureus biofilms grown on titanium implant surfaces. PVP-I and vancomycin were used to treat immature and developed biofilms formed by two clinical strains of S. aureus (BP043-MRSA, PB011-MSSA). S. aureus strains were grown as immature (3 h-old) or developed (24 h-old) biofilm. These biofilms were grown on titanium plasma sprayed discs. The treatment regimens tested were: 0.8% PVP-I, 500 μg/ml vancomycin as well as a combination of vancomycin and PVP-I. PVP-I was tested at 3 min, as per current clinical practice, versus 1 min treatment times. In addition, the cytotoxicity of the PVP-I and vancomycin was tested using fresh skeletal muscle tissue cores harvested from the rat's abdominal muscles using alamarBlue assay. The combination of PVP-I (3 min) and vancomycin (24 h.) showed synergistic interaction and the best efficacy against immature biofilms formed by both clinical strains. This degree of eradication was statistically significant compared to the untreated control, p < .0001. However, this combination therapy had limited efficiency against developed biofilms. Also, PVP-I alone was more effective when exposure time was 3 min instead of 1 min against immature biofilm for MRSA, p = .02, and MSSA, p = .01. PVP-I and vancomycin were not effective against developed biofilm regardless of exposure time. Also, combining PVP-I and vancomycin was not cytotoxic to muscle tissue. Combining PVP-I with vancomycin is superior in reducing viable S. aureus cells in immature biofilms grown on titanium surface without causing significant cytotoxicity to muscle tissue. Exposure times and biofilm maturity play a role in dictating the efficacy of using local antiseptics and antibiotics to treat biofilms on implant surfaces.
Collapse
Affiliation(s)
- Mariam Taha
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,The Ottawa Hospital, Division of Orthopedic Surgery Ottawa, Ottawa, Ontario, Canada
| | - Rozanne Arulanandam
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Andrew Chen
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Jean-Simon Diallo
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Ontario, Canada
| | - Hesham Abdelbary
- Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,The Ottawa Hospital, Division of Orthopedic Surgery Ottawa, Ottawa, Ontario, Canada.,Centre for Innovative Cancer Research, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| |
Collapse
|
9
|
Daffinee KE, O'Neill ET, Bleick CR, Williams G, Antoci V, Garcia D, LaPlante KL. Staphylococcal Biofilm: Penetration and bioavailability of vancomycin with or without rifampin. Diagn Microbiol Infect Dis 2023; 106:115947. [PMID: 37116243 DOI: 10.1016/j.diagmicrobio.2023.115947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 03/14/2023] [Accepted: 03/25/2023] [Indexed: 03/31/2023]
Abstract
We measured antibiotic penetration and bioavailability in staphylococcus biofilms using simulated humanized concentrations of fluorescent vancomycin plus or minus rifampin. Vancomycin percent recovery across biofilm layers was:upper = 46%, middle = 40%, and lower = 33%. Vancomycin plus rifampin was not significantly different (P = 0.65). Addition of rifampin did not improve vancomycin penetration across biofilm layers.
Collapse
Affiliation(s)
- Kathryn E Daffinee
- Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI, USA
| | - Emily T O'Neill
- Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI, USA; College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Callan R Bleick
- College of Pharmacy, University of Rhode Island, Kingston, RI, USA
| | - Geoff Williams
- Leduc Bioimaging Facility, Brown University, Division of Biology and Medicine, Providence, RI, USA
| | - Valentin Antoci
- Department of Orthopaedics, The Warren Alpert School of Medicine, Brown University, Providence, RI, USA; The Diane N. Weiss Center for Orthopaedic Trauma Research, Rhode Island Hospital, Providence, RI, USA
| | - Dioscaris Garcia
- Department of Orthopaedics, The Warren Alpert School of Medicine, Brown University, Providence, RI, USA; The Diane N. Weiss Center for Orthopaedic Trauma Research, Rhode Island Hospital, Providence, RI, USA
| | - Kerry L LaPlante
- Infectious Diseases Research Program, Providence Veterans Affairs Medical Center, Providence, RI, USA; College of Pharmacy, University of Rhode Island, Kingston, RI, USA; Warren Alpert Medical School of Brown University, Division of Infectious Diseases, Providence, RI, USA.
| |
Collapse
|
10
|
Kumar M, Pandey SK, Lalhall A, Sharma R, Sharma RK, Wangoo N. Targeting bacterial biofilms using vancomycin and multivalent cell-penetrating peptide labeled quantum dots. J Biomed Mater Res B Appl Biomater 2023; 111:284-294. [PMID: 36056808 DOI: 10.1002/jbm.b.35150] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/19/2022] [Accepted: 08/01/2022] [Indexed: 12/15/2022]
Abstract
Bacterial biofilms are highly resilient microbial musters that are difficult to eradicate, driving the development of novel therapeutic strategies. The current study aims to investigate the therapeutic efficacy of cell-penetrating peptide-based targeted delivery of vancomycin functionalized quantum dots in eradicating biofilm formation in gram-positive and gram-negative bacterial strains. The conjugate was characterized using fluorimetry, UV-visible spectroscopy, gel electrophoresis, and zeta potential. The conjugate was then tested for antimicrobial and antibiofilm activity against Staphylococcus aureus, Pseudomonas aeruginosa, and Escherichia coli, and it demonstrated excellent antimicrobial as well as antibiofilm activity against all the tested strains. The findings indicated that the conjugate was capable of overcoming bacterial resistance of bacteria in addition to the eradication of biofilms at effective concentrations.
Collapse
Affiliation(s)
- Munish Kumar
- Department of Chemistry & Centre for Advanced Studies in Chemistry, Panjab University, Chandigarh, India
| | - Satish Kumar Pandey
- Department of Biotechnology, School of Life Sciences, Mizoram University, Aizawl, India
| | - Alisha Lalhall
- Centre for Nanoscience and Nanotechnology, Panjab University, Chandigarh, India.,Department of Applied Sciences, University Institute of Engineering & Technology (U.I.E.T.), Panjab University, Chandigarh, India
| | - Rohit Sharma
- Centre for Stem Cell and Tissue Engineering, Panjab University, Chandigarh, India
| | - Rohit K Sharma
- Department of Chemistry & Centre for Advanced Studies in Chemistry, Panjab University, Chandigarh, India
| | - Nishima Wangoo
- Department of Applied Sciences, University Institute of Engineering & Technology (U.I.E.T.), Panjab University, Chandigarh, India
| |
Collapse
|
11
|
Kuttner H, Pfister S, Moriarty TF, Meier C, Wahl P. Periprosthetic Joint Infection With Actinomyces radingae May Lead to the Identification of a Neglected Source of Intraoperative Contamination. Arthroplast Today 2022; 18:181-184. [PMID: 36405864 PMCID: PMC9672404 DOI: 10.1016/j.artd.2022.08.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/16/2022] Open
Abstract
Periprosthetic joint infection remains a major complication in arthroplasty. We present the first description of a case of periprosthetic joint infection with Actinomyces radingae, microorganism that is mostly found on the skin of the upper body and might cause particular challenges as it is difficult to culture and specify. Furthermore, a thorough microbiologic workup may indicate the source of infection. In this case, it is possible that perspiration from the surgeon was the source of intraoperative contamination. Intraoperative contamination through perspiration may be important and should be avoided by all means.
Collapse
Affiliation(s)
- Hannes Kuttner
- Division of Orthopaedics and Traumatology, Cantonal Hospital Winterthur, Winterthur, Switzerland
- Corresponding author. Division of Orthopaedics and Traumatology, Cantonal Hospital Winterthur, Brauerstrasse 15, 8401 Winterthur, Switzerland.
| | - Stefan Pfister
- Laboratory of Microbiology, HFR Fribourg – Cantonal Hospital, Fribourg, Switzerland
| | | | - Christoph Meier
- Division of Orthopaedics and Traumatology, Cantonal Hospital Winterthur, Winterthur, Switzerland
| | - Peter Wahl
- Division of Orthopaedics and Traumatology, Cantonal Hospital Winterthur, Winterthur, Switzerland
| |
Collapse
|
12
|
Wiesli MG, Livio F, Achermann Y, Gautier E, Wahl P. Wound fluid ceftriaxone concentrations after local application with calcium sulphate as carrier material in the treatment of orthopaedic device-associated hip infections. Bone Joint Res 2022; 11:835-842. [DOI: 10.1302/2046-3758.1111.bjr-2022-0180.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Aims There is a considerable challenge in treating bone infections and orthopaedic device-associated infection (ODAI), partly due to impaired penetration of systemically administrated antibiotics at the site of infection. This may be circumvented by local drug administration. Knowledge of the release kinetics from any carrier material is essential for proper application. Ceftriaxone shows a particular constant release from calcium sulphate (CaSO4) in vitro, and is particularly effective against streptococci and a large portion of Gram-negative bacteria. We present the clinical release kinetics of ceftriaxone-loaded CaSO4 applied locally to treat ODAI. Methods A total of 30 operations with ceftriaxone-loaded CaSO4 had been performed in 28 patients. Ceftriaxone was applied as a single local antibiotic in 21 operations and combined with vancomycin in eight operations, and in an additional operation with vancomycin and amphotericin B. Sampling of wound fluid was performed from drains or aspirations. Ceftriaxone concentrations were measured by liquid chromatography with tandem mass spectrometry (LC-MS/MS). Results A total of 37 wound fluid concentrations from 16 operations performed in 14 patients were collected. The ceftriaxone concentrations remained approximately within a range of 100 to 200 mg/l up to three weeks. The median concentration was 108.9 mg/l (interquartile range 98.8 to 142.5) within the first ten days. No systemic adverse reactions were observed. Conclusion Our study highlights new clinical data of locally administered ceftriaxone with CaSO4 as carrier material. The near-constant release of ceftriaxone from CaSO4 observed in vitro could be confirmed in vivo. The concentrations remained below known local toxicity thresholds. Cite this article: Bone Joint Res 2022;11(11):835–842.
Collapse
Affiliation(s)
- Matthias G. Wiesli
- Laboratory for Biointerfaces, Empa - Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland
- Clinic for Craniomaxillofacial and Oral Surgery, Cantonal Hospital Aarau, Aarau, Switzerland
| | - Françoise Livio
- Service of Clinical Pharmacology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Yvonne Achermann
- Department of Internal Medicine, Hospital Zollikerberg, Zurich, Switzerland
- Department of Dermatology, University Hospital of Zurich, University of Zurich, Zurich, Switzerland
| | - Emanuel Gautier
- Department of Orthopaedics, HFR Fribourg - Cantonal Hospital, Fribourg, Switzerland
| | - Peter Wahl
- Department of Orthopaedics, HFR Fribourg - Cantonal Hospital, Fribourg, Switzerland
- Division of Orthopaedics and Traumatology, Cantonal Hospital Winterthur, Winterthur, Switzerland
| |
Collapse
|
13
|
Raj DS, Dhamodharan D, Thanigaivel S, Vickram AS, Byun HS. Nanoemulsion as an Effective Inhibitor of Biofilm-forming Bacterial Associated Drug Resistance: An Insight into COVID Based Nosocomial Infections. BIOTECHNOL BIOPROC E 2022; 27:543-555. [PMID: 36092682 PMCID: PMC9449957 DOI: 10.1007/s12257-022-0055-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/30/2022] [Accepted: 04/06/2022] [Indexed: 11/26/2022]
Abstract
Antibiotic overuse has resulted in the microevolution of drug-tolerant bacteria. Understandably it has become one of the most significant obstacles of the current century for scientists and researchers to overcome. Bacteria have a tendency to form biofilm as a survival mechanism. Biofilm producing microorganism become far more resistant to antimicrobial agents and their tolerance to drugs also increases. Prevention of biofilm development and curbing the virulency factors of these multi drug resistant or tolerant bacterial pathogens is a newly recognised tactic for overcoming the challenges associated with such bacterial infections and has become a niche to be addressed. In order to inhibit virulence and biofilm from planktonic bacteria such as, Pseudomonas aeruginosa, Acinetobacter baumannii, and others, stable nanoemulsions (NEs) of essential oils (EOs) and their bioactive compounds prove to be an interesting solution. These NEs demonstrated significantly greater anti-biofilm and anti-virulence activity than commercial antibiotics. The EO reduces disease-causing gene expression, which is required for pathogenicity, biofilm formation and attachment to the surfaces. Essential NE and NE-loaded hydrogel surface coatings demonstrates superior antibiofilm activity which can be employed in healthcare-related equipments like glass, plastic, and metal chairs, hospital beds, ventilators, catheters, and tools used in intensive care units. Thus, anti-virulence and anti-biofilm forming strategies based on NEs-loaded hydrogel may be used as coatings to combat biofilm-mediated infection on solid surfaces.
Collapse
Affiliation(s)
- Deena Santhana Raj
- Department of Biotechnology, Saveetha School of Engineering, SIMATS, Saveetha University, Thandalam, Chennai, Tamil Nadu 602105 India
| | - Duraisami Dhamodharan
- Department of Chemical and Biomolecular Engineering, Chonnam National University, Yeosu, 59626 Korea
| | - S. Thanigaivel
- Department of Biotechnology, Saveetha School of Engineering, SIMATS, Saveetha University, Thandalam, Chennai, Tamil Nadu 602105 India
| | - A. S. Vickram
- Department of Biotechnology, Saveetha School of Engineering, SIMATS, Saveetha University, Thandalam, Chennai, Tamil Nadu 602105 India
| | - Hun-Soo Byun
- Department of Chemical and Biomolecular Engineering, Chonnam National University, Yeosu, 59626 Korea
| |
Collapse
|
14
|
Pijls BG, Sanders IMJG, Kuijper EJ, Nelissen RGHH. Effectiveness of mechanical cleaning, antibiotics, and induction heating on eradication of Staphylococcus aureus in mature biofilms. Bone Joint Res 2022; 11:629-638. [PMID: 36047617 PMCID: PMC9533241 DOI: 10.1302/2046-3758.119.bjr-2022-0010.r1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Aims Here we used a mature seven-day biofilm model of Staphylococcus aureus, exposed to antibiotics up to an additional seven days, to establish the effectiveness of either mechanical cleaning or antibiotics or non-contact induction heating, and which combinations could eradicate S. aureus in mature biofilms. Methods Mature biofilms of S. aureus (ATCC 29213) were grown on titanium alloy (Ti6Al4V) coupons for seven days and were subjected to the following treatments or their combinations: antibiotics, mechanical cleaning, or heat shock by induction heating of 60°C for one minute. Experiments were repeated at least five times. Results In the untreated biofilm, growth up to 1.8×1011 colony-forming units (CFU)/cm2 was observed. Treatment with ciprofloxacin, flucloxacillin, vancomycin, cefuroxime, and amoxicillin all with rifampicin gave 6.0 log, 6.1 log, 1.4 log, 4.8 log, and 3.6 log reduction in CFU/cm2, respectively. Mechanical cleaning alone resulted in 4.9 log reduction and induction heating in 7.3 log reduction. There was an additional effect of ciprofloxacin, flucloxacillin, and induction heating when used in combinations. There was no additional effect for mechanical cleaning. No bacterial growth could be detected after induction heating followed by seven days of ciprofloxacin with rifampicin. Conclusion Mechanical cleaning, antibiotics, and non-contact induction heating reduced the bacterial load of mature S. aureus biofilms with approximately 5 log or more as a single treatment. The effect of mechanical cleaning on mature S. aureus biofilms was limited when used in combination with antibiotics and/or induction heating. Cite this article: Bone Joint Res 2022;11(9):629–638.
Collapse
Affiliation(s)
- B G Pijls
- Department of Orthopaedics, Leiden University Medical Center, Leiden, The Netherlands
| | - Ingrid M J G Sanders
- Department of Medical Microbiology, Section Experimental Bacteriology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ed J Kuijper
- Department of Medical Microbiology, Section Experimental Bacteriology, Leiden University Medical Center, Leiden, The Netherlands
| | - R G H H Nelissen
- Department of Orthopaedics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
15
|
The Incidence of Severe Hypercalcaemia-Induced Mental Status Changes in Patients Treated with Antibiotic-Loaded Calcium Sulphate Depot for Orthopaedic Infections. J Clin Med 2022; 11:jcm11164900. [PMID: 36013138 PMCID: PMC9409894 DOI: 10.3390/jcm11164900] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 11/17/2022] Open
Abstract
Local application of antibiotics with calcium-containing carrier materials (CCCM) might deliver large quantities of calcium, with some cases of hypercalcaemia reported. The incidence of symptomatic hypercalcaemia was estimated retrospectively in a consecutive, prospective series of patients treated between 10/2006 and 02/2019 with antibiotic-loaded CCCM for various orthopaedic infections. Risk factors were analysed. In the study period, 215 CCCM applications were performed. Two patients (0.9%) developed symptomatic hypercalcaemia. In one case, hypercalcaemia occurred 14 days after a second CCCM application during a staged septic hip revision. In the other case, hypercalcaemia became symptomatic six days after application of vancomycin-loaded CCCM in a component-retaining septic revision hip arthroplasty. In both cases, hypercalcemia was not imputable solely to the CCCM. Prolonged immobilization, renal impairment and other specific risk factors were present. Implantation of a CCCM for local application of antibiotics exposes the patient to large quantities of calcium during dissolution. This might induce symptomatic hypercalcaemia, a potentially life-threatening complication. The observed incidence of symptomatic hypercalcaemia remained rare (<1%). In some patients, compensatory mechanisms might be overwhelmed in the presence of other risk factors. Postoperative monitoring of calcaemia as well as elimination of risk factors is mandatory for all patients treated with CCCM.
Collapse
|
16
|
Whitely ME, Helms SM, Muire PJ, Lofgren AL, Lopez RA, Wenke JC. Preclinical evaluation of a commercially available biofilm disrupting wound lavage for musculoskeletal trauma. J Orthop Surg Res 2022; 17:347. [PMID: 35840981 PMCID: PMC9284756 DOI: 10.1186/s13018-022-03199-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 05/16/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Treatment of open fractures remains a significant challenge in trauma care as these fractures are accompanied by extensive soft tissue damage, exposing the wound site to contaminants and increasing infection risk. Formation of biofilm, a capsule-like environment that acts as a barrier to treatment, is a primary mode by which infecting pathogens persist at the wound site. Therefore, a pressing need exists to identify irrigation methods that can disrupt biofilm and expose pathogens to treatment. This study aims to evaluate the antibiofilm wound lavage, Bactisure™, in comparison with saline for care of severe musculoskeletal wounds and elucidate potential effects on antibiotic treatment success. METHODS UAMS-1 Staphylococcus aureus biofilms were formed in vitro and treated with Bactisure™ wound lavage or sterile normal saline, alone, or in combination with sub-biofilm inhibitory levels of vancomycin. Characterization methods included quantification of biofilm biomass, quantification of viable biofilm bacteria, and biofilm matrix imaging. For in vivo assessment, a delayed treatment model of contaminated open fracture was used wherein a critical-sized defect was created in a rat femur and wound site inoculated with UAMS-1. Following a 6 h delay, wounds were debrided, irrigated with lavage of interest, and antibiotic treatments administered. Bacterial enumeration was performed on bone and hardware samples after two weeks. RESULTS An immediate reduction in biofilm biomass was observed in vitro following antibiofilm lavage treatment, with a subsequent 2- to 3- log reduction in viable bacteria achieved after 24 h. Furthermore, biofilms treated with antibiofilm lavage in combination with vancomycin exhibited a minor, but statistically significant, decrease in viable bacteria compared to irrigation alone. In vivo, a minor, not statistically significant, decrease in median bioburden was observed for the antibiofilm lavage compared to saline when used in combination with antibiotics. However, the percentage of bone and hardware samples with detectable bacteria was reduced from 50 to 38%. CONCLUSIONS These results suggest that the antibiofilm wound lavage, Bactisure™, may hold promise in mitigating infection in contaminated musculoskeletal wounds and warrants further investigation. Here, we proposed multiple mechanisms in vitro by which this antibiofilm lavage may help mitigate infection, and demonstrate this treatment slightly outperforms saline in controlling bioburden in vivo.
Collapse
Affiliation(s)
- Michael E Whitely
- Combat Wound Care Department, US Army Institute of Surgical Research, 3698 Chambers Pass, Building 3611, JBSA-Fort Sam Houston, San Antonio, TX, 78234, USA.
| | - Sarah M Helms
- Combat Wound Care Department, US Army Institute of Surgical Research, 3698 Chambers Pass, Building 3611, JBSA-Fort Sam Houston, San Antonio, TX, 78234, USA
| | - Preeti J Muire
- Combat Wound Care Department, US Army Institute of Surgical Research, 3698 Chambers Pass, Building 3611, JBSA-Fort Sam Houston, San Antonio, TX, 78234, USA
| | - Alicia L Lofgren
- Combat Wound Care Department, US Army Institute of Surgical Research, 3698 Chambers Pass, Building 3611, JBSA-Fort Sam Houston, San Antonio, TX, 78234, USA
| | - Rebecca A Lopez
- Combat Wound Care Department, US Army Institute of Surgical Research, 3698 Chambers Pass, Building 3611, JBSA-Fort Sam Houston, San Antonio, TX, 78234, USA
| | - Joseph C Wenke
- Combat Wound Care Department, US Army Institute of Surgical Research, 3698 Chambers Pass, Building 3611, JBSA-Fort Sam Houston, San Antonio, TX, 78234, USA
| |
Collapse
|
17
|
Okae Y, Nishitani K, Sakamoto A, Kawai T, Tomizawa T, Saito M, Kuroda Y, Matsuda S. Estimation of Minimum Biofilm Eradication Concentration (MBEC) on In Vivo Biofilm on Orthopedic Implants in a Rodent Femoral Infection Model. Front Cell Infect Microbiol 2022; 12:896978. [PMID: 35846761 PMCID: PMC9285014 DOI: 10.3389/fcimb.2022.896978] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/01/2022] [Indexed: 12/25/2022] Open
Abstract
The formation of a biofilm on the implant surface is a major cause of intractable implant-associated infection. To investigate the antibiotic concentration needed to eradicate the bacteria inside a biofilm, the minimum biofilm eradication concentration (MBEC) has been used, mostly against in vitro biofilms on plastic surfaces. To produce a more clinically relevant environment, an MBEC assay against biofilms on stainless-steel implants formed in a rat femoral infection model was developed. The rats were implanted with stainless steel screws contaminated by two Staphylococcus aureus strains (UAMS-1, methicillin-sensitive Staphylococcus aureus; USA300LAC, methicillin-resistant Staphylococcus aureus) and euthanized on days 3 and 14. Implants were harvested, washed, and incubated with various concentrations (64–4096 μg/mL) of gentamicin (GM), vancomycin (VA), or cefazolin (CZ) with or without an accompanying systemic treatment dose of VA (20 μg/mL) or rifampicin (RF) (1.5 μg/mL) for 24 h. The implant was vortexed and sonicated, the biofilm was removed, and the implant was re-incubated to determine bacterial recovery. MBEC on the removed biofilm and implant was defined as in vivo MBEC and in vivo implant MBEC, respectively, and the concentrations of 100% and 60% eradication were defined as MBEC100 and MBEC60, respectively. As for in vivo MBEC, MBEC100 of GM was 256–1024 μg/mL, but that of VA and CZ ranged from 2048–4096 μg/mL. Surprisingly, the in vivo implant MBEC was much higher, ranging from 2048 μg/mL to more than 4096 μg/mL. The addition of RF, not VA, as a secondary antibiotic was effective, and MBEC60 on day 3 USA300LAC biofilm was reduced from 1024 μg/mL with GM alone to 128 μg/mL in combination with RF and the MBEC60 on day 14 USA300LAC biofilm was reduced from 2048 μg/mL in GM alone to 256 μg/mL in combination with RF. In conclusion, a novel MBEC assay for in vivo biofilms on orthopedic implants was developed. GM was the most effective against both methicillin-sensitive and methicillin-resistant Staphylococcus aureus, in in vivo biofilms, and the addition of a systemic concentration of RF reduced MBEC of GM. Early initiation of treatment is desired because the required concentration of antibiotics increases with biofilm maturation.
Collapse
|
18
|
Prospective comparative analysis of efficacy of local debridement with or without vancomycin impregnated calcium sulphate in chronic osteomyelitis. J Orthop 2022; 32:133-138. [DOI: 10.1016/j.jor.2022.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 05/17/2022] [Accepted: 06/02/2022] [Indexed: 11/18/2022] Open
|
19
|
Biofilm Formation by Pathogenic Bacteria: Applying a Staphylococcus aureus Model to Appraise Potential Targets for Therapeutic Intervention. Pathogens 2022; 11:pathogens11040388. [PMID: 35456063 PMCID: PMC9027693 DOI: 10.3390/pathogens11040388] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 03/17/2022] [Accepted: 03/21/2022] [Indexed: 01/02/2023] Open
Abstract
Carried in the nasal passages by up to 30% of humans, Staphylococcus aureus is recognized to be a successful opportunistic pathogen. It is a frequent cause of infections of the upper respiratory tract, including sinusitis, and of the skin, typically abscesses, as well as of food poisoning and medical device contamination. The antimicrobial resistance of such, often chronic, health conditions is underpinned by the unique structure of bacterial biofilm, which is the focus of increasing research to try to overcome this serious public health challenge. Due to the protective barrier of an exopolysaccharide matrix, bacteria that are embedded within biofilm are highly resistant both to an infected individual’s immune response and to any treating antibiotics. An in-depth appraisal of the stepwise progression of biofilm formation by S. aureus, used as a model infection for all cases of bacterial antibiotic resistance, has enhanced understanding of this complicated microscopic structure and served to highlight possible intervention targets for both patient cure and community infection control. While antibiotic therapy offers a practical means of treatment and prevention, the most favorable results are achieved in combination with other methods. This review provides an overview of S. aureus biofilm development, outlines the current range of anti-biofilm agents that are used against each stage and summarizes their relative merits.
Collapse
|
20
|
Periprosthetic Joint Infection (PJI)—Results of One-Stage Revision with Antibiotic-Impregnated Cancellous Allograft Bone—A Retrospective Cohort Study. Antibiotics (Basel) 2022; 11:antibiotics11030310. [PMID: 35326773 PMCID: PMC8944641 DOI: 10.3390/antibiotics11030310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 02/04/2023] Open
Abstract
Controversy exists regarding the optimal treatment of periprosthetic joint infection (PJI), considering control of infection, functional results as well as quality of life. Difficulties in treatment derive from the formation of biofilms within a few days after infection. Biofilms are tolerant to systemically applied antibiotics, requiring extreme concentrations for a prolonged period. Minimum biofilm eradicating concentrations (MBEC) are only feasible by the local application of antibiotics. One established approach is the use of allograft bone as a carrier, granting a sustained release of antibiotics in very high concentrations after appropriate impregnation. The purpose of this study was to determine the rate of reinfection after a one-stage revision of infected hip or knee prostheses, using antibiotic-impregnated allograft bone as the carrier and avoiding cement. Between 1 January 2004 and 31 January 2018, 87 patients with PJI, according to MSIS, underwent a one-stage revision with antibiotic-impregnated cancellous allograft bone. An amount of 17 patients had insufficient follow-ups. There were 70 remaining patients (34 male, 36 female) with a mean follow-up of 5.6 years (range 2–15.6) and with a mean age of 68.2 years (range 31.5–86.9). An amount of 38 hips and 11 knees were implanted without any cement; and 21 knees were implanted with moderate cementing at the articular surface with stems always being uncemented. Within 2 years after surgery, 6 out of 70 patients (8.6%, CI 2–15.1) showed reinfection and after more than 2 years, an additional 6 patients showed late-onset infection. Within 2 years after surgery, 11 out of 70 patients (15.7%, CI 7.2–24.2) had an implant failure for any reason (including infection) and after more than 2 years, an additional 7 patients had an implant failure. Using Kaplan-Meier analysis for all 87 patients, the estimated survival for reinfection was 93.9% (CI 88.8–99.1) at 1 year, 89.9% (CI 83.2–96.6) at 2 years and 81.5% (CI 72.1–90.9) at 5 years. The estimated survival for implant failure for any reason was 90.4% (CI 84.1–96.7) at 1 year, 80.9% (CI 72.2–89.7) at 2 years and 71.1% (CI 60.3–81.8) at 5 years. One-stage revision with antibiotic-impregnated cancellous allograft bone grants comparable results regarding infection control as with multiple stages, while shortening rehabilitation, improving quality of life for the patients and reducing costs for the health care system.
Collapse
|
21
|
Reinisch K, Schläppi M, Meier C, Wahl P. Local antibiotic treatment with calcium sulfate as carrier material improves the outcome of debridement, antibiotics, and implant retention procedures for periprosthetic joint infections after hip arthroplasty – a retrospective study. J Bone Jt Infect 2022; 7:11-21. [PMID: 35111565 PMCID: PMC8795886 DOI: 10.5194/jbji-7-11-2022] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 12/14/2021] [Indexed: 11/11/2022] Open
Abstract
Abstract. Purpose: Debridement, antibiotics, and implant retention (DAIR) is an established treatment modality in periprosthetic joint infections (PJIs), but success rates vary. This study compared the success of DAIR for PJIs after a total hip arthroplasty (THA), with or without local antibiotic delivery with CaSO4 as the carrier material.
Methods: A retrospective review of DAIR for PJIs after THA performed between 2010 and 2018, including 41 patients is conducted. A total of 27 patients were treated by DAIR with local antibiotics with CaSO4 as the carrier material, and 14 patients were treated by a standard DAIR. The endpoints were treatment failure, defined as the need for a reoperation, either a second DAIR or a prosthesis removal or exchange due to persistent or recurrent infection, the initiation of a long-term suppressive antibiotic treatment, or death related to infection.
Results: Considering any reoperation as an outcome, 11 of 14 cases treated without AB-CaSO4 (79 %) and 4 of the 27 cases treated with
AB-CaSO4 failed (15 %). Considering revision as an outcome, 9 out of 14 cases treated without AB-CaSO4 (64 %) and 4 of the 27 cases treated with AB-CaSO4 (15 %) failed. A Kaplan–Meier survival analysis showed that local antibiotic delivery with CaSO4 as the carrier material led to a significantly longer infection-free survival, considering any surgical revision (p<0.0001; hazard ratio 8.9 (95 % CI 2.8–28.2)) or revision with component exchange (p=0.0015; hazard ratio 5.6 (95 % CI 1.7–18.2)) as the endpoint.
Conclusion: The addition of local antibiotics with CaSO4 as the carrier material to DAIR for PJIs after THA significantly increases success rates, such as infection-free survival, any reoperation, and revision with component exchange in particular.
Collapse
|
22
|
Modification of indwelling PVC catheters by ionizing radiation with temperature- and pH-responsive polymers for antibiotic delivery. Radiat Phys Chem Oxf Engl 1993 2022. [DOI: 10.1016/j.radphyschem.2022.110005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
23
|
Jacob B, Makarewicz O, Hartung A, Brodt S, Roehner E, Matziolis G. In vitro additive effects of dalbavancin and rifampicin against biofilm of Staphylococcus aureus. Sci Rep 2021; 11:23425. [PMID: 34873186 PMCID: PMC8648795 DOI: 10.1038/s41598-021-02709-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/08/2021] [Indexed: 02/04/2023] Open
Abstract
Dalbavancin is a novel glycopeptide antibiotic approved for the treatment of acute bacterial skin and skin structure infections (ABSSSIs). It is characterized by a potent activity against numerous Gram-positive pathogens, a long elimination half-life and a favorable safety profile. Most recently, its application for the treatment of periprosthetic joint infections (PJIs) was introduced. The aim of this study was to proof our hypothesis, that dalbavancin shows superior efficacy against staphylococcal biofilms on polyethylene (PE) disk devices compared with vancomycin and additive behavior in combination with rifampicin. Staphylococcus aureus biofilms were formed on PE disk devices for 96 h and subsequently treated with dalbavancin, vancomycin, rifampicin and dalbavancin-rifampicin combination at different concentrations. Quantification of antibacterial activity was determined by counting colony forming units (CFU/ml) after sonification of the PE, serial dilution of the bacterial suspension and plating on agar-plates. Biofilms were additionally life/dead-stained and visualized using fluorescence microscopy. Dalbavancin presented superior anti-biofilm activity compared to vancomycin. Additive effects of the combination dalbavancin and rifampicin were registered. Dalbavancin combined with rifampicin presents promising anti-biofilm activity characteristics in vitro. Further in vivo studies are necessary to establish recommendations for the general use of dalbavancin in the treatment of PJIs.
Collapse
Affiliation(s)
- Benjamin Jacob
- Orthopaedic Professorship of the University Hospital Jena, Campus Eisenberg, Klosterlausnitzer Straße 81, 07607, Eisenberg, Germany.
| | - Oliwia Makarewicz
- Institute for Infectious Diseases and Infection Control, Jena University Hospital, 07747, Jena, Germany
| | - Anita Hartung
- Institute for Infectious Diseases and Infection Control, Jena University Hospital, 07747, Jena, Germany
| | - Steffen Brodt
- Orthopaedic Professorship of the University Hospital Jena, Campus Eisenberg, Klosterlausnitzer Straße 81, 07607, Eisenberg, Germany
| | - Eric Roehner
- Orthopaedic Professorship of the University Hospital Jena, Campus Eisenberg, Klosterlausnitzer Straße 81, 07607, Eisenberg, Germany
| | - Georg Matziolis
- Orthopaedic Professorship of the University Hospital Jena, Campus Eisenberg, Klosterlausnitzer Straße 81, 07607, Eisenberg, Germany
| |
Collapse
|
24
|
New Adapted In Vitro Technology to Evaluate Biofilm Formation and Antibiotic Activity Using Live Imaging under Flow Conditions. Diagnostics (Basel) 2021; 11:diagnostics11101746. [PMID: 34679444 PMCID: PMC8535051 DOI: 10.3390/diagnostics11101746] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/11/2021] [Accepted: 09/19/2021] [Indexed: 12/19/2022] Open
Abstract
The polymicrobial nature of biofilms and bacterial interactions inside chronic wounds are keys for the understanding of bacterial cooperation. The aim of this present study was to develop a technique to study and visualize biofilm in live imaging under flow conditions (Bioflux™ 200, Fluxion Biosciences). The BiofluxTM system was adapted using an in vitro chronic wound-like medium (CWM) that mimics the environment encountered in ulcers. Two reference strains of Staphylococcus aureus (Newman) and Pseudomonas aeruginosa (PAO1) were injected in the BiofluxTM during 24 h to 72 h in mono and coculture (ratio 1:1, bacteria added simultaneously) in the CWM vs. a control medium (BHI). The quantification of biofilm formation at each time was evaluated by inverted microscopy. After 72 h, different antibiotics (ceftazidime, imipenem, linezolid, oxacillin and vancomycin) at 1x MIC, 10x MIC and 100x MIC were administrated to the system after an automatic increase of the flow that mimicked a debridement of the wound surface. Biofilm studies highlighted that the two species, alone or associated, constituted a faster and thicker biofilm in the CWM compared to the BHI medium. The effect of antibiotics on mature or “debrided” biofilm indicated that some of the most clinically used antibiotic such as vancomycin or imipenem were not able to disrupt and reduce the biofilm biomass. The use of a life cell imaging with an in vitro CWM represents a promising tool to study bacterial biofilm and investigate microbial cooperation in a chronic wound context.
Collapse
|
25
|
Tran HA, Tran PA. In Situ Coatings of Silver Nanoparticles for Biofilm Treatment in Implant-Retention Surgeries: Antimicrobial Activities in Monoculture and Coculture. ACS APPLIED MATERIALS & INTERFACES 2021; 13:41435-41444. [PMID: 34448395 DOI: 10.1021/acsami.1c08239] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Bacterial biofilms are indicated in most medical device-associated infections. Treating these biofilms is challenging yet critically important for applications such as in device-retention surgeries, which can have reinfection rates of up to 80%. This in vitro study centered around our new method of treating biofilm and preventing reinfection. Ionic silver (Ag, in the form of silver nitrate) combined with dopamine and a biofilm-lysing enzyme (α-amylase) were applied to model 4-day-old Staphylococcus aureus biofilms on titanium substrates to degrade the extracellular matrix of the biofilm and kill the biofilm bacteria. In this process, the oxidative self-polymerization of dopamine converted Ag ions into Ag nanoparticles that, together with the resultant self-adhering polydopamine (PDA), formed coatings that strongly bound to the treated substrates. Surprisingly, although these Ag/PDA coatings significantly reduced S. aureus growth in standard bacterial monoculture, they showed much lower antimicrobial activity in coculture of the bacteria and osteoblastic MC3T3-E1 cells in which the bacteria were also found attached to the osteoblasts. This S. aureus- osteoblast interaction was also linked to bacterial survival against gentamicin treatment observed in coculture. Our study thus provided clear evidence suggesting that bacteria's interactions with tissue cells surrounding implants may significantly contribute to their resistance to antimicrobial treatment.
Collapse
Affiliation(s)
- Hien A Tran
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), 2 George Street, Brisbane, Queensland 4000, Australia
- Interface Science and Materials Engineering Group, School of Mechanical, Medical and Process Engineering, QUT, 2 George Street, Brisbane, Queensland 4000, Australia
| | - Phong A Tran
- Centre for Biomedical Technologies, Queensland University of Technology (QUT), 2 George Street, Brisbane, Queensland 4000, Australia
- Interface Science and Materials Engineering Group, School of Mechanical, Medical and Process Engineering, QUT, 2 George Street, Brisbane, Queensland 4000, Australia
| |
Collapse
|
26
|
Xia W, Li N, Shan H, Lin Y, Yin F, Yu X, Zhou Z. Gallium Porphyrin and Gallium Nitrate Reduce the High Vancomycin Tolerance of MRSA Biofilms by Promoting Extracellular DNA-Dependent Biofilm Dispersion. ACS Infect Dis 2021; 7:2565-2582. [PMID: 34346692 DOI: 10.1021/acsinfecdis.1c00280] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Biofilms, structured communities of bacterial cells embedded in a self-produced extracellular matrix (ECM) which consists of proteins, polysaccharide intercellular adhesins (PIAs), and extracellular DNA (eDNA), play a key role in clinical infections and are associated with an increased morbidity and mortality by protecting the embedded bacteria against drug and immune response. The high levels of antibiotic tolerance render classical antibiotic therapies impractical for biofilm-related infections. Thus, novel drugs and strategies are required to reduce biofilm tolerance and eliminate biofilm-protected bacteria. Here, we showed that gallium, an iron mimetic metal, can lead to nutritional iron starvation and act as dispersal agent triggering the reconstruction and dispersion of mature methicillin-resistant Staphylococcus aureus (MRSA) biofilms in an eDNA-dependent manner. The extracellular matrix, along with the integral bacteria themselves, establishes the integrated three-dimensional structure of the mature biofilm. The structures and compositions of gallium-treated mature biofilms differed from those of natural or antibiotic-survived mature biofilms but were similar to those of immature biofilms. Similar to immature biofilms, gallium-treated biofilms had lower levels of antibiotic tolerance, and our in vitro tests showed that treatment with gallium agents reduced the antibiotic tolerance of mature MRSA biofilms. Thus, the sequential administration of gallium agents (gallium porphyrin and gallium nitrate) and relatively low concentrations of vancomycin (16 mg/L) effectively eliminated mature MRSA biofilms and eradicated biofilm-enclosed bacteria within 1 week. Our results suggested that gallium agents may represent a potential treatment for refractory biofilm-related infections, such as prosthetic joint infections (PJI) and osteomyelitis, and provide a novel basis for future biofilm treatments based on the disruption of normal biofilm-development processes.
Collapse
Affiliation(s)
- Wenyang Xia
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Niya Li
- Department of Laboratory, Shanghai Sixth People’s Hospital East Affiliated to Shanghai University of Medicine & Health Sciences, Shanghai 200233, China
| | - Haojie Shan
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Yiwei Lin
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Fuli Yin
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Xiaowei Yu
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Zubin Zhou
- Department of Orthopaedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| |
Collapse
|
27
|
Manasherob R, Mooney JA, Lowenberg DW, Bollyky PL, Amanatullah DF. Tolerant Small-colony Variants Form Prior to Resistance Within a Staphylococcus aureus Biofilm Based on Antibiotic Selective Pressure. Clin Orthop Relat Res 2021; 479:1471-1481. [PMID: 33835090 PMCID: PMC8208434 DOI: 10.1097/corr.0000000000001740] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 03/01/2021] [Indexed: 01/31/2023]
Abstract
BACKGROUND The treatment of periprosthetic joint infection (PJI) is focused on the surgical or chemical removal of biofilm. Antibiotics in isolation are typically ineffective against PJI. Bacteria survive after antibiotic administration because of antibiotic tolerance, resistance, and persistence that arise in the resident bacteria of a biofilm. Small-colony variants are typically slow-growing bacterial subpopulations that arise after antibiotic exposure and are associated with persistent and chronic infections such as PJI. The role of biofilm-mediated antibiotic tolerance in the emergence of antibiotic resistance remains poorly defined experimentally. QUESTIONS/PURPOSES We asked: (1) Does prior antibiotic exposure affect how Staphylococcus aureus survives within a developing biofilm when exposed to an antibiotic that penetrates biofilm, like rifampicin? (2) Does exposure to an antibiotic with poor biofilm penetration, such as vancomycin, affect how S. aureus survives within a developing biofilm? (3) Do small-colony variants emerge from antibiotic-tolerant or -resistant bacteria in a S. aureus biofilm? METHODS We used a porous membrane as an in vitro implant model to grow luminescent S. aureus biofilms and simultaneously track microcolony expansion. We evaluated the impact of tolerance on the development of resistance by comparing rifampicin (an antibiotic that penetrates S. aureus biofilm) with vancomycin (an antibiotic that penetrates biofilm poorly). We performed viability counting after membrane dissociation to discriminate among tolerant, resistant, and persistent bacteria. Biofilm quantification and small-colony morphologies were confirmed using scanning electron microscopy. Because of experimental variability induced by the starting bacterial inoculum, relative changes were compared since absolute values may not have been statistically comparable. RESULTS Antibiotic-naïve S. aureus placed under the selective pressure of rifampicin initially survived within an emerging biofilm by using tolerance given that biofilm resident cell viability revealed 1.0 x 108 CFU, of which 7.5 x 106 CFU were attributed to the emergence of resistance and 9.3 x 107 CFU of which were attributed to the development of tolerance. Previous exposure of S. aureus to rifampicin obviated tolerance-mediate survival when rifampicin resistance was present, since the number of viable biofilm resident cells (9.5 x 109 CFU) nearly equaled the number of rifampicin-resistant bacteria (1.1 x 1010 CFU). Bacteria exposed to an antibiotic with poor biofilm penetration, like vancomycin, survive within an emerging biofilm by using tolerance as well because the biofilm resident cell viability for vancomycin-naïve (1.6 x 1010 CFU) and vancomycin-resistant (1.0 x 1010 CFU) S. aureus could not be accounted for by emergence of resistance. Adding rifampicin to vancomycin resulted in a nearly 500-fold reduction in vancomycin-tolerant bacteria from 1.5 x 1010 CFU to 3.3 x 107 CFU. Small-colony variant S. aureus emerged within the tolerant bacterial population within 24 hours of biofilm-penetrating antibiotic administration. Scanning electron microscopy before membrane dissociation confirmed the presence of small, uniform cells with biofilm-related microstructures when unexposed to rifampicin as well as large, misshapen, lysed cells with a small-colony variant morphology [29, 41, 42, 63] and a lack of biofilm-related microstructures when exposed to rifampicin. This visually confirmed the rapid emergence of small-colony variants within the sessile niche of a developing biofilm when exposed to an antibiotic that exerted selective pressure. CONCLUSION Tolerance explains why surgical and nonsurgical modalities that rely on antibiotics to "treat" residual microscopic biofilm may fail over time. The differential emergence of resistance based on biofilm penetration may explain why some suppressive antibiotic therapies that do not penetrate biofilm well may rely on bacterial control while limiting the emergence of resistance. However, this strategy fails to address the tolerant bacterial niche that harbors persistent bacteria with a small-colony variant morphology. CLINICAL RELEVANCE Our work establishes biofilm-mediated antibiotic tolerance as a neglected feature of bacterial communities that prevents the effective treatment of PJI.
Collapse
Affiliation(s)
- Robert Manasherob
- School of Medicine, Stanford University, Palo Alto, CA, USA
- Department of Orthopaedic Surgery, Stanford Medicine, Redwood City, CA, USA
| | - Jake A. Mooney
- School of Medicine, Stanford University, Palo Alto, CA, USA
| | - David W. Lowenberg
- Department of Orthopaedic Surgery, Stanford Medicine, Redwood City, CA, USA
| | - Paul L. Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford Medicine, Palo Alto, CA, USA
| | | |
Collapse
|
28
|
Inhibitory properties of Chinese Herbal Formula SanHuang decoction on biofilm formation by antibiotic-resistant Staphylococcal strains. Sci Rep 2021; 11:7134. [PMID: 33785834 PMCID: PMC8009868 DOI: 10.1038/s41598-021-86647-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Accepted: 03/17/2021] [Indexed: 11/08/2022] Open
Abstract
The aim of this study was to explore the effect of Chinese herbal SanHuang decoction (SH) on biofilm formation of antibiotic-resistant Staphylococci on titanium surface, and to explore its mechanism. Biofilm-forming ATCC 35984, ATCC 43300 and MRSE 287 were used in this study. The MICs of SH and vancomycin against Staphylococci were determined by the broth microdilution method. Six groups were designed, namely control group (bacteria cultured with medium), 1/8MIC SH group (1MIC SH was diluted by 1/8 using TSB or saline), 1/4MIC SH group, 1/2MIC SH group, 1MIC SH group and vancomycin group (bacteria cultured with 1MIC vancomycin). The inhibitory effect on bacterial adhesion and biofilm formation were observed by the spread plate method, CV staining, SEM, and CLSM. Real-time PCR was performed to determine the effect of SH on the expression levels of ica AD and ica R gene in ATCC 35984 during the biofilm formation. The strains were found to be susceptible to SH and vancomycin with MIC of 38.75 mg/ml and 2.5 μg/ml, respectively. SH with 1 MIC and 1/2 MIC could inhibit the bacteria adhesion, showing only scattered adhesion from SEM. CLSM showed that SH with 1 MIC and 1/2 MIC inhibited the biofilm formation. The quantitative results of the spread plate method and CV staining showed that there was significant differences between the SH groups (P < 0.05). Further, with an increase in SH concentration, the inhibitory effect became more obvious when compared with control group. Among the groups, vancomycin had the strongest inhibitory effect on bacterial adhesion and biofilm formation (P < 0.01). With an increase in SH concentration, the expression levels of ica AD decreased, and the expression of ica R increased correspondingly (P < 0.05). In conclusions, SH can inhibit the biofilm formation of antibiotic-resistant Staphylococci. Its probable mechanistic activity may be through the inhibition of polysaccharide intercellular adhesin synthesis by down-regulating the expression of ica AD gene.
Collapse
|
29
|
Wiesli MG, Kaiser JP, Gautier E, Wick P, Maniura-Weber K, Rottmar M, Wahl P. Influence of ceftriaxone on human bone cell viability and in vitro mineralization potential is concentration- and time-dependent. Bone Joint Res 2021; 10:218-225. [PMID: 33739124 PMCID: PMC7998344 DOI: 10.1302/2046-3758.103.bjr-2020-0412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aims In orthopaedic and trauma surgery, implant-associated infections are increasingly treated with local application of antibiotics, which allows a high local drug concentration to be reached without eliciting systematic adverse effects. While ceftriaxone is a widely used antibiotic agent that has been shown to be effective against musculoskeletal infections, high local concentrations may harm the surrounding tissue. This study investigates the acute and subacute cytotoxicity of increasing ceftriaxone concentrations as well as their influence on the osteogenic differentiation of human bone progenitor cells. Methods Human preosteoblasts were cultured in presence of different concentrations of ceftriaxone for up to 28 days and potential cytotoxic effects, cell death, metabolic activity, cell proliferation, and osteogenic differentiation were studied. Results Ceftriaxone showed a cytotoxic effect on human bone progenitor cells at 24 h and 48 h at concentrations above 15,000 mg/l. With a longer incubation time of ten days, subtoxic effects could be observed at concentrations above 500 mg/l. Gene and protein expression of collagen, as well as mineralization levels of human bone progenitor cells, showed a continuous decrease with increasing ceftriaxone concentrations by days 14 and 28, respectively. Notably, mineralization was negatively affected already at concentrations above 250 mg/l. Conclusion This study demonstrates a concentration-dependent influence of ceftriaxone on the viability and mineralization potential of primary human bone progenitor cells. While local application of ceftriaxone is highly established in orthopaedic and trauma surgery, a therapeutic threshold of 250 mg/l or lower should diminish the risk of reduced osseointegration of prosthetic implants. Cite this article: Bone Joint Res 2021;10(3):218–225.
Collapse
Affiliation(s)
- Matthias Guido Wiesli
- Laboratory for Biointerfaces, Empa - Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland
| | - Jean-Pierre Kaiser
- Laboratory for Particles-Biology Interactions, Empa - Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland
| | - Emanuel Gautier
- Department of Orthopaedics, HFR Fribourg - Cantonal Hospital, Fribourg, Switzerland
| | - Peter Wick
- Laboratory for Particles-Biology Interactions, Empa - Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland
| | - Katharina Maniura-Weber
- Laboratory for Biointerfaces, Empa - Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland
| | - Markus Rottmar
- Laboratory for Biointerfaces, Empa - Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland
| | - Peter Wahl
- Division of Orthopaedics and Traumatology, Cantonal Hospital Winterthur, Winterthur, Switzerland
| |
Collapse
|
30
|
Application of 3D Printing-Assisted Articulating Spacer in Two-Stage Revision Surgery for Periprosthetic Infection after Total Knee Arthroplasty: A Retrospective Observational Study. BIOMED RESEARCH INTERNATIONAL 2021; 2021:3948638. [PMID: 33628779 PMCID: PMC7884112 DOI: 10.1155/2021/3948638] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 11/24/2020] [Accepted: 01/27/2021] [Indexed: 12/16/2022]
Abstract
Background Bone cement spacers are widely used in two-stage revision surgeries for periprosthetic joint infection (PJI) after total knee arthroplasty. Current spacer design results in insufficient release of drugs; therefore, current spacers have low efficacy. In this study, we explored a set of alternative articular spacer using 3D printing technology. This novel spacer will increase effectiveness of revision surgery for PJI. Methods The spacer was designed using CAD software and constructed on site using 3D-printed silicone mold during debridement surgery. We carried out a retrospective study among patients undergoing treatment using traditional static and new articular spacers. Infection control rate, bone loss, difficulty of revision surgery, knee joint range of motion, function evaluation, and subjective satisfaction of the patients in the two groups were compared. Results Forty-two patients undergoing knee revision surgery between Jan 2014 and Nov 2019 were included in this study. Twenty-two patients were treated with static antibiotic cement spacers, whereas the other twenty patients were with treated with 3D printing-assisted antibiotic loaded articulating spacers. Patients in the articular group showed significantly lower bone loss on the femur site and tibial site compared with patients in the static group. In addition, patients in the articular group showed significantly less operation time, intraoperative blood loss, and improved knee function and patient overall satisfaction compared with patients in the static group. Conclusions The 3D printing-assisted articular spacer provides satisfactory range of motion during the interim period, prevents bone loss, facilitates second-stage reimplantation and postoperative rehabilitation, and results in low reinfection and complication rates.
Collapse
|
31
|
Foster AL, Boot W, Stenger V, D'Este M, Jaiprakash A, Eglin D, Zeiter S, Richards RG, Moriarty TF. Single-stage revision of MRSA orthopedic device-related infection in sheep with an antibiotic-loaded hydrogel. J Orthop Res 2021; 39:438-448. [PMID: 33305875 DOI: 10.1002/jor.24949] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 02/04/2023]
Abstract
Local antimicrobial therapy is an integral aspect of treating orthopedic device-related infection (ODRI), which is conventionally administered via polymethyl-methacrylate (PMMA) bone cement. PMMA, however, is limited by a suboptimal antibiotic release profile and a lack of biodegradability. In this study, we compare the efficacy of PMMA versus an antibiotic-loaded hydrogel in a single-stage revision for chronic methicillin-resistant Staphylococcus aureus (MRSA) ODRI in sheep. Antibiofilm activity of the antibiotic combination (gentamicin and vancomycin) was determined in vitro. Swiss alpine sheep underwent a single-stage revision of a tibial intramedullary nail with MRSA infection. Local gentamicin and vancomycin therapy was delivered via hydrogel or PMMA (n = 5 per group), in conjunction with systemic antibiotic therapy. In vivo observations included: local antibiotic tissue concentration, renal and liver function tests, and quantitative microbiology on tissues and hardware post-mortem. There was a nonsignificant reduction in biofilm with an increasing antibiotic concentration in vitro (p = 0.12), confirming the antibiotic tolerance of the MRSA biofilm. In the in vivo study, four out of five sheep from each treatment group were culture-negative. Antibiotic delivery via hydrogel resulted in 10-100 times greater local concentrations for the first 2-3 days compared with PMMA and were comparable thereafter. Systemic concentrations of gentamicin were minimal or undetectable in both groups, while renal and liver function tests were within normal limits. This study shows that a single-stage revision with hydrogel or PMMA is equally effective, although the hydrogel offers certain practical benefits over PMMA, which make it an attractive proposition for clinical use.
Collapse
Affiliation(s)
- Andrew L Foster
- AO Research Institute Davos, Davos, Switzerland
- Queensland University of Technology (QUT), Faculty of Science and Engineering, Brisbane, Queensland, Australia
- Department of Orthopaedic Surgery and Jamieson Trauma Institute, Royal Brisbane and Women's Hospital Australia, Brisbane, Queensland, Australia
| | | | | | | | - Anjali Jaiprakash
- Queensland University of Technology (QUT), Faculty of Science and Engineering, Brisbane, Queensland, Australia
| | - David Eglin
- AO Research Institute Davos, Davos, Switzerland
| | | | | | | |
Collapse
|
32
|
Schwarz EM, McLaren AC, Sculco TP, Brause B, Bostrom M, Kates SL, Parvizi J, Alt V, Arnold WV, Carli A, Chen AF, Choe H, Coraça‐Huber DC, Cross M, Ghert M, Hickok N, Jennings JA, Joshi M, Metsemakers W, Ninomiya M, Nishitani K, Oh I, Padgett D, Ricciardi B, Saeed K, Sendi P, Springer B, Stoodley P, Wenke JC. Adjuvant antibiotic-loaded bone cement: Concerns with current use and research to make it work. J Orthop Res 2021; 39:227-239. [PMID: 31997412 PMCID: PMC7390691 DOI: 10.1002/jor.24616] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 01/07/2020] [Accepted: 01/13/2020] [Indexed: 02/04/2023]
Abstract
Antibiotic-loaded bone cement (ALBC) is broadly used to treat orthopaedic infections based on the rationale that high-dose local delivery is essential to eradicate biofilm-associated bacteria. However, ALBC formulations are empirically based on drug susceptibility from routine laboratory testing, which is known to have limited clinical relevance for biofilms. There are also dosing concerns with nonstandardized, surgeon-directed, hand-mixed formulations, which have unknown release kinetics. On the basis of our knowledge of in vivo biofilms, pathogen virulence, safety issues with nonstandardized ALBC formulations, and questions about the cost-effectiveness of ALBC, there is a need to evaluate the evidence for this clinical practice. To this end, thought leaders in the field of musculoskeletal infection (MSKI) met on 1 August 2019 to review and debate published and anecdotal information, which highlighted four major concerns about current ALBC use: (a) substantial lack of level 1 evidence to demonstrate efficacy; (b) ALBC formulations become subtherapeutic following early release, which risks induction of antibiotic resistance, and exacerbated infection from microbial colonization of the carrier; (c) the absence of standardized formulation protocols, and Food and Drug Administration-approved high-dose ALBC products to use following resection in MSKI treatment; and (d) absence of a validated assay to determine the minimum biofilm eradication concentration to predict ALBC efficacy against patient specific micro-organisms. Here, we describe these concerns in detail, and propose areas in need of research.
Collapse
Affiliation(s)
- Edward M. Schwarz
- Department of Orthopaedics, Center for Musculoskeletal Research University of Rochester Rochester New York
| | - Alex C. McLaren
- Department of Orthopaedic Surgery, College of Medicine‐Phoenix University of Arizona Phoenix Arizona
| | - Thomas P. Sculco
- Department of Orthopaedic Surgery, Weill Cornell Medicine Hospital for Special Surgery New York New York
| | - Barry Brause
- Department of Infectious Diseases, Weill Cornell Medicine Hospital for Special Surgery New York New York
| | - Mathias Bostrom
- Department of Orthopaedic Surgery, Weill Cornell Medicine Hospital for Special Surgery New York New York
| | - Stephen L. Kates
- Department of Orthopaedic Surgery Virginia Commonwealth University Richmond Virginia
| | - Javad Parvizi
- Department of Orthopaedics Rothman Institute at Thomas Jefferson University Hospital Philadelphia Pennsylvania
| | - Volker Alt
- Department of Trauma Surgery University Medical Centre Regensburg Regensburg Germany
| | - William V. Arnold
- Department of Orthopaedics Rothman Institute at Thomas Jefferson University Hospital Philadelphia Pennsylvania
| | - Alberto Carli
- Department of Orthopaedic Surgery, Weill Cornell Medicine Hospital for Special Surgery New York New York
| | - Antonia F. Chen
- Department of Orthopaedics, Brigham and Women's Hospital Harvard Medical School Boston Massachusetts
| | - Hyonmin Choe
- Department of Orthopaedic Yokohama City University Yokohama Japan
| | - Débora C. Coraça‐Huber
- Department of Orthopaedic Surgery, Experimental Orthopedics, Research Laboratory for Biofilms and Implant Associated Infections Medical University of Innsbruck Innsbruck Austria
| | - Michael Cross
- Department of Orthopaedic Surgery, Weill Cornell Medicine Hospital for Special Surgery New York New York
| | - Michelle Ghert
- Division of Orthopaedic Surgery, Department of Surgery McMaster University Hamilton Ontario Canada
| | - Noreen Hickok
- Department of Orthopaedic Surgery, Department of Biochemistry & Molecular Biology Thomas Jefferson University Philadelphia Pennsylvania
| | | | - Manjari Joshi
- Division of Infectious Diseases, R Adams Cowley Shock Trauma Center University of Maryland Baltimore Maryland
| | | | - Mark Ninomiya
- Department of Orthopaedics, Center for Musculoskeletal Research University of Rochester Rochester New York
| | - Kohei Nishitani
- Department of Orthopaedic Surgery Graduate School of Medicine, Kyoto University Sakyo Kyoto Japan
| | - Irvin Oh
- Department of Orthopaedics, Center for Musculoskeletal Research University of Rochester Rochester New York
| | - Douglas Padgett
- Department of Orthopaedic Surgery, Weill Cornell Medicine Hospital for Special Surgery New York New York
| | - Benjamin Ricciardi
- Department of Orthopaedics, Center for Musculoskeletal Research University of Rochester Rochester New York
| | - Kordo Saeed
- Southampton University Hospitals NHS Foundation Trust, Department of Microbiology, Microbiology and Innovation Research Unit (MIRU) and University of Southampton, School of Medicine Southampton UK
| | - Parham Sendi
- Institute for Infectious Diseases University of Bern, Bern and Department of Infectious Diseases, Hospital Epidemiology and Department of Orthopaedics and Traumatology, University of Basel Basel Switzerland
- Department of Orthopaedics and Traumatology University Hospital Basel Basel Switzerland
| | - Bryan Springer
- Department of Orthopaedic Surgery, OrthoCarolina Hip and Knee Center Atrium Musculoskeletal Institute Charlotte North Carolina
| | - Paul Stoodley
- Department of Microbial Infection and Immunity and Orthopaedics The Ohio State University Columbus Ohio
| | - Joseph C. Wenke
- Orthopaedic Trauma Department U.S. Army Institute of Surgical Research Fort Sam Houston Texas
| | | |
Collapse
|
33
|
Nguyen HTT, Nguyen TH, Otto M. The staphylococcal exopolysaccharide PIA - Biosynthesis and role in biofilm formation, colonization, and infection. Comput Struct Biotechnol J 2020; 18:3324-3334. [PMID: 33240473 PMCID: PMC7674160 DOI: 10.1016/j.csbj.2020.10.027] [Citation(s) in RCA: 130] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 12/20/2022] Open
Abstract
PIA is a key extracellular matrix component in staphylococci and other bacteria. PIA is a cationic, partially deacetylated N-acetylglucosamine polymer. PIA has a major role in bacterial biofilms and biofilm-associated infection.
Exopolysaccharide is a key part of the extracellular matrix that contributes to important mechanisms of bacterial pathogenicity, most notably biofilm formation and immune evasion. In the human pathogens Staphylococcus aureus and S. epidermidis, as well as in many other staphylococcal species, the only exopolysaccharide is polysaccharide intercellular adhesin (PIA), a cationic, partially deacetylated homopolymer of N-acetylglucosamine, whose biosynthetic machinery is encoded in the ica locus. PIA production is strongly dependent on environmental conditions and controlled by many regulatory systems. PIA contributes significantly to staphylococcal biofilm formation and immune evasion mechanisms, such as resistance to antimicrobial peptides and ingestion and killing by phagocytes, and presence of the ica genes is associated with infectivity. Due to its role in pathogenesis, PIA has raised considerable interest as a potential vaccine component or target.
Collapse
Affiliation(s)
- Hoai T T Nguyen
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, 50 South Drive, Bethesda 20814, MD, USA.,School of Biotechnology, International University, Vietnam National University of Ho Chi Minh City, Khu Pho 6, Thu Duc, Ho Chi Minh City, Viet Nam
| | - Thuan H Nguyen
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, 50 South Drive, Bethesda 20814, MD, USA
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, U.S. National Institutes of Health, 50 South Drive, Bethesda 20814, MD, USA
| |
Collapse
|
34
|
Early application of topical antibiotic powder in open-fracture wounds: A strategy to prevent biofilm formation and infections. OTA Int 2020; 3:e091. [PMID: 33937714 PMCID: PMC8078147 DOI: 10.1097/oi9.0000000000000091] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 07/23/2020] [Indexed: 01/01/2023]
Abstract
Despite meticulous surgical care and systemic antibiotics, open fracture wounds have high rates of infection leading to increased morbidity. To reduce infection rates, orthopaedic surgeons may administer local antibiotics using various carriers that may be ineffective due to poor antibiotic release from carriers, subsequent surgery to remove nondegradable carriers, and mismatch between release kinetics and material degradation. Biofilms form rapidly as bacteria that are within the wound multiply quickly and transform from the antibiotic-susceptible planktonic phenotype to the antibiotic-tolerant biofilm phenotype. This tolerance to antibiotics can occur within hours. Currently, local antibiotics are placed in the wounds using a carrier such as polymethylmethacrylate beads; however, this occurs after surgical debridement that can be hours to even a day after initial injury allowing bacteria enough time to form a biofilm that makes the antibiotic containing polymethylmethacrylate beads less effective. In contrast, emerging practices in elective surgical procedures, such as spine fusion, place antibiotic powder (e.g. vancomycin) in the wound at the time of closure. This has been shown to be extremely effective, presumably because of the very small-time period between potential contamination and local antibiotic application. There is evidence that suggests that the ineffectiveness of local antibiotic use in open fractures is primarily due to the delay in application of local antibiotics from the time of injury and propose a concept of topical antibiotic powder application in the prehospital or emergency department setting.
Collapse
|
35
|
Chen X, Thomsen TR, Winkler H, Xu Y. Influence of biofilm growth age, media, antibiotic concentration and exposure time on Staphylococcus aureus and Pseudomonas aeruginosa biofilm removal in vitro. BMC Microbiol 2020; 20:264. [PMID: 32831025 PMCID: PMC7444035 DOI: 10.1186/s12866-020-01947-9] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 08/13/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Biofilm is known to be tolerant towards antibiotics and difficult to eradicate. Numerous studies have reported minimum biofilm eradication concentration (MBEC) values of antibiotics for many known biofilm pathogens. However, the experimental parameters applied in these studies differ considerably, and often the rationale behind the experimental design are not well described. This makes it difficult to compare the findings. To demonstrate the importance of experimental parameters, we investigated the influence of biofilm growth age, antibiotic concentration and treatment duration, and growth media on biofilm eradication. Additionally, OSTEOmycin™, a clinically used antibiotic containing allograft bone product, was tested for antibiofilm efficacy. RESULTS The commonly used Calgary biofilm device was used to grow 24 h and 72 h biofilms of Staphylococcus aureus and Pseudomonas aeruginosa, which were treated with time-dependent vancomycin (up to 3000 mg L- 1) and concentration-dependent tobramycin (up to 80 mg L- 1), respectively. Two common bacteriological growth media, tryptic soy broth (TSB) and cation-adjusted Mueller Hinton broth (CaMHB), were tested. We found for both species that biofilms were more difficult to kill in TSB than in CaMHB. Furthermore, young biofilms (24 h) were easier to eradicate than old biofilms (72 h). In agreement with vancomycin being time-dependent, extension of the vancomycin exposure increased killing of S. aureus biofilms. Tobramycin treatment of 24 h P. aeruginosa biofilms was found concentration-dependent and time-independent, however, increasing killing was indicated for 72 h P. aeruginosa biofilms. Treatment with tobramycin containing OSTEOmycin T™ removed 72 h and 168 h P. aeruginosa biofilms after 1 day treatment, while few 72 h S. aureus biofilms survived after 2 days treatment with vancomycin containing OSTEOmycin V™. CONCLUSIONS This study demonstrated biofilm removal efficacy was influenced by media, biofilm age and antibiotic concentration and treatment duration. It is therefore necessary to taking these parameters into consideration when designing experiments. The results of OSTEOmycin™ products indicated that simple in vitro biofilm test could be used for initial screening of antibiofilm products. For clinical application, a more clinically relevant biofilm model for the specific biofilm infection in question should be developed to guide the amount of antibiotics used for local antibiofilm treatment.
Collapse
Affiliation(s)
- Xiaofeng Chen
- Center for Microbial Communities, Aalborg University, Aalborg East, Denmark
| | - Trine Rolighed Thomsen
- Center for Microbial Communities, Aalborg University, Aalborg East, Denmark
- Life Science Division, Danish Technological Institute, Aarhus, Denmark
| | - Heinz Winkler
- Osteitis Centre, Privatklinik Döbling, Vienna, Austria
| | - Yijuan Xu
- Center for Microbial Communities, Aalborg University, Aalborg East, Denmark.
- Life Science Division, Danish Technological Institute, Aarhus, Denmark.
| |
Collapse
|
36
|
Zheng JX, Tu HP, Sun X, Xu GJ, Chen JW, Deng QW, Yu ZJ, Qu D. In vitro activities of telithromycin against Staphylococcus aureus biofilms compared with azithromycin, clindamycin, vancomycin and daptomycin. J Med Microbiol 2020; 69:120-131. [PMID: 31916929 DOI: 10.1099/jmm.0.001122] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Introduction. Staphylococcus aureus biofilms are difficult to treat and the effect of telithromycin treatment is still unclear.Aim. This study aimed to explore the effect of telithromycin against Staphylococcus aureus biofilms compared with azithromycin, clindamycin, vancomycin and daptomycin.Methodology. Eight methicillin-susceptible and eight methicillin-resistant S. aureus isolates (MSSA and MRSA, respectively) were used for this study. Biofilm biomasses were detected by crystal violet staining and the adherent cells in the established biofilms were quantified by determination of colony-forming units (c.f.u.). The RNA levels of biofilm formation-related genes were determined by RT-qPCR.Results. Telithromycin [8× minimum inhibitory concentration (MIC)] eradicated more established biofilms than azithromycin or clindamycin in the four MSSA isolates, and eliminated the established biofilms of six MRSA isolates more effectively than vancomycin or daptomycin. Telithromycin (8× MIC) killed more adherent cells in the established biofilms than azithromycin or clindamycin in the six MSSA isolates, and killed more adherent cells than vancomycin in all eight MRSA isolates. Daptomycin also showed an excellent effect on the adherent cells of MRSA isolates, with similarresults to telithromycin. The effect of a subinhibitory concentration of telithromycin (1/4× MIC) was significantly superior to that of azithromycin or clindamycin, inhibiting the biofilm formation of six MSSA isolates and seven MRSA isolates more effectively than vancomycin or daptomycin. The RNA levels of agrA, agrC, clfA, icaA and sigB decreased when treated with telithromycin (1/4× MIC).Conclusions. Telithromycin is more effective than azithromycin, clindamycin, vancomycin, or daptomycin against S. aureus biofilms.
Collapse
Affiliation(s)
- Jin-Xin Zheng
- Department of Infectious Diseases and the Key Laboratory of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, PR China.,Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, PR China
| | - Hao-Peng Tu
- Department of Infectious Diseases and the Key Laboratory of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, PR China
| | - Xiang Sun
- Department of Infectious Diseases and the Key Laboratory of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, PR China
| | - Guang-Jian Xu
- Department of Infectious Diseases and the Key Laboratory of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, PR China
| | - Jun-Wen Chen
- Department of Infectious Diseases and the Key Laboratory of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, PR China
| | - Qi-Wen Deng
- Department of Infectious Diseases and the Key Laboratory of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, PR China
| | - Zhi-Jian Yu
- Department of Infectious Diseases and the Key Laboratory of Endogenous Infection, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen 518052, PR China
| | - Di Qu
- Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, Shanghai 200032, PR China
| |
Collapse
|
37
|
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
|
38
|
Peeters A, Putzeys G, Thorrez L. Current Insights in the Application of Bone Grafts for Local Antibiotic Delivery in Bone Reconstruction Surgery. J Bone Jt Infect 2019; 4:245-253. [PMID: 31700774 PMCID: PMC6831806 DOI: 10.7150/jbji.38373] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 08/27/2019] [Indexed: 12/15/2022] Open
Abstract
Introduction: Bone implant related infection is still one of the biggest challenges in bone and joint surgery. Antibiotic impregnated bone grafts seem to be promising in both treatment and prevention of these infections. However, great variance in methodology predominates this field of research. This paper gives an overview of the published literature. Methods: The PRISMA-flowchart was used as protocol for article selection. Medline was searched and articles were selected in accordance with predetermined exclusion criteria. Results: Forty-eight articles were included in the synthesis. Topics including bone graft type, manipulations of the graft, elution profile, bacterial inhibition, osteotoxicity, incorporation, special impregnation methods, clinical use and storage were investigated. Therapeutically, high initial levels seem appropriate for biofilm eradication. A single stage procedure in the treatment of bone implant related infection seems feasible. Prophylactically, the literature indicates a reduction of postoperative infections when using antibiotic impregnated bone grafts. Conclusion: Bone grafts are a suitable carrier for local antibiotic application both therapeutically and prophylactically.
Collapse
Affiliation(s)
| | - Guy Putzeys
- Orthopedic Centre, AZ Groeninge, Kortrijk, Belgium
| | - Lieven Thorrez
- Department of Development and Regeneration, KU Leuven, Kortrijk, Belgium
| |
Collapse
|
39
|
Dudareva M, Hotchen AJ, Ferguson J, Hodgson S, Scarborough M, Atkins BL, McNally MA. The microbiology of chronic osteomyelitis: Changes over ten years. J Infect 2019; 79:189-198. [PMID: 31319142 DOI: 10.1016/j.jinf.2019.07.006] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 05/28/2019] [Accepted: 07/12/2019] [Indexed: 01/03/2023]
Abstract
AIM This study quantified changes in the microbiology of osteomyelitis over a ten year period from a single centre within the UK with regard to infection with multi-drug resistant (MDR) bacteria and susceptibility of antimicrobial regimens. METHOD Patients with chronic osteomyelitis undergoing definitive surgery from 2013-2017 were inluded (n = 223). Microbiology was compared to patients in a cohort from 2001-2004, using the same diagnostic criteria, and same deep tissue sampling technique (n = 157). Clinical features associated with MDR bacterial infection were analysed using logistic regression. RESULTS Both cohorts had similar baseline characteristics. Despite a similar proportion of Staphylococcus aureus in both cohorts, the rate of methicillin resistant Staphylococcus aureus (MRSA) infection was lower in 2013-2017 compared to 2001-2004 (11.4% vs 30.8% of Staphylococcus aureus, p = 0.007). However, the proportion of MDR infections was similar in both cohorts (15.2% versus 17.2%). Metalwork was associated with MDR infection (unadjusted OR 5.0; 95% CI: 1.15 to 22.0). There was no change in resistance to glycopeptide / meropenem combination treatment (2.2% vs 2.5%, p > 0.9). CONCLUSIONS In this centre, rates of MRSA osteomyelitis have fallen by two thirds, over the past 10 years, in line with the reducing rate of MRSA bacteraemia nationally. A history of metalwork may predict MDR infection. A glycopeptide with an anti-pseudomonal carbapenem remains the post-operative empiric systemic regimen of choice. Resistance patterns support the use of a glycopeptide with an aminoglycoside in local antibiotic therapy.
Collapse
Affiliation(s)
- Maria Dudareva
- Bone Infection Unit, Nuffield Orthopaedic Centre, Oxford University Hospitals, Oxford, UK
| | - Andrew James Hotchen
- Bone Infection Unit, Nuffield Orthopaedic Centre, Oxford University Hospitals, Oxford, UK; Department of Trauma and Orthopaedic Surgery, Cambridge University Hospitals, Cambridge, UK.
| | - Jamie Ferguson
- Bone Infection Unit, Nuffield Orthopaedic Centre, Oxford University Hospitals, Oxford, UK
| | - Susanne Hodgson
- The Jenner Institue, University of Oxford, Oxford, UK; Department of Microbiology and Infectious Diseases, Oxford University Hospitals, Oxford UK
| | - Matthew Scarborough
- Bone Infection Unit, Nuffield Orthopaedic Centre, Oxford University Hospitals, Oxford, UK
| | - Bridget L Atkins
- Bone Infection Unit, Nuffield Orthopaedic Centre, Oxford University Hospitals, Oxford, UK; Department of Microbiology and Infectious Diseases, Oxford University Hospitals, Oxford UK
| | - Martin A McNally
- Bone Infection Unit, Nuffield Orthopaedic Centre, Oxford University Hospitals, Oxford, UK
| |
Collapse
|
40
|
Kang J, Dietz MJ, Li B. Antimicrobial peptide LL-37 is bactericidal against Staphylococcus aureus biofilms. PLoS One 2019; 14:e0216676. [PMID: 31170191 PMCID: PMC6553709 DOI: 10.1371/journal.pone.0216676] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 04/26/2019] [Indexed: 11/19/2022] Open
Abstract
Our current challenge in the management of prosthetic joint infection is the eradication of biofilms which has driven the need for improved antimicrobial agents and regimens. In this study, the antimicrobial peptide, LL-37, and silver nanoparticles (AgNPs) were investigated for their antimicrobial efficacies against Staphylococcus aureus (S. aureus), a microorganism commonly implicated in biofilm-related infections. These antimicrobials were compared to conventional antibiotics and combination treatments with rifampin. Using a Centers for Disease Control reactor, 24 h S. aureus biofilms were formed on cobalt-chromium discs and the anti-biofilm activity was determined by quantifying the amount of colony forming units following treatments. We found that LL-37 was the most efficacious antimicrobial agent with a more than 4 log reduction in colony counts. In comparison, silver nanoparticles and conventional antibiotics were not as efficacious, with a less than 1 log reduction in colony counts. Antimicrobial combination treatments with rifampin significantly increased the log reduction for AgNPs and gentamicin, although still significantly less than LL-37 in isolation. Furthermore, kinetic studies revealed the rapid elimination of S. aureus biofilm with LL-37. Collectively, the results of this study demonstrated that LL-37 was an effective agent against S. aureus biofilms and may have potential clinical applications in the eradication of biofilms and treatment of prosthetic joint infection.
Collapse
Affiliation(s)
- Jason Kang
- Department of Orthopaedics, School of Medicine, West Virginia University, Morgantown, WV, United States of America
| | - Matthew J. Dietz
- Department of Orthopaedics, School of Medicine, West Virginia University, Morgantown, WV, United States of America
- * E-mail: (MJD); (BL)
| | - Bingyun Li
- Department of Orthopaedics, School of Medicine, West Virginia University, Morgantown, WV, United States of America
- * E-mail: (MJD); (BL)
| |
Collapse
|
41
|
Stewart PS, Parker AE. Measuring Antimicrobial Efficacy against Biofilms: a Meta-analysis. Antimicrob Agents Chemother 2019; 63:e00020-19. [PMID: 30803974 PMCID: PMC6496104 DOI: 10.1128/aac.00020-19] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 02/21/2019] [Indexed: 12/12/2022] Open
Abstract
Through a statistical meta-analysis of published data on antimicrobial efficacy against biofilms formed by two common bacterial species, it was concluded that the particular experimental method used is the most important factor determining the outcome of the test. An expected dose-response relationship (greater killing with higher doses or longer treatment times) was observed for data sets derived from a single method but was not observed when data from multiple studies using diverse methods were pooled. Method-specific properties such as the surface area/volume ratio, areal biofilm cell density, and microbial species were shown to influence quantitative measurements of biofilm killing. A better appreciation of the method characteristics that affect antibiofilm efficacy tests could aid decision-making related to investment in research and development and regulatory approvals for biofilm control strategies. The following recommendations are offered to those working in research and development related to biofilm control: (i) report the log reduction, surface area/volume ratio, and biofilm areal cell density; (ii) include data for a benchmark agent, making sure that this agent performs competitively at the dose tested; (iii) measure the dose-response relationship, i.e., make measurements at multiple treatment concentrations or dose durations; and (iv) use a standardized method in addition to research methods.
Collapse
Affiliation(s)
- Philip S Stewart
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, USA
- Chemical and Biological Engineering, Montana State University, Bozeman, Montana, USA
| | - Albert E Parker
- Center for Biofilm Engineering, Montana State University, Bozeman, Montana, USA
- Mathematical Sciences, Montana State University, Bozeman, Montana, USA
| |
Collapse
|
42
|
General Assembly, Prevention, Local Antimicrobials: Proceedings of International Consensus on Orthopedic Infections. J Arthroplasty 2019; 34:S75-S84. [PMID: 30352772 DOI: 10.1016/j.arth.2018.09.056] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
43
|
Badha V, Moore R, Heffernan J, Castaneda P, McLaren A, Overstreet D. Determination of Tobramycin and Vancomycin Exposure Required to Eradicate Biofilms on Muscle and Bone Tissue In Vitro. J Bone Jt Infect 2019; 4:1-9. [PMID: 30755841 PMCID: PMC6367195 DOI: 10.7150/jbji.29711] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 11/10/2018] [Indexed: 01/09/2023] Open
Abstract
Background: Bacterial biofilms cause chronic orthopaedic infections. Surgical debridement to remove biofilm can be ineffective without adjuvant local antimicrobials because undetected biofilm fragments may remain in the wound and reestablish the infection if untreated. However, the concentrations and duration of antimicrobial exposure necessary to eradicate bacteria from clinical biofilms remain largely undefined. In this study, we determined the minimum biofilm eradication concentration (MBEC) of tobramycin and vancomycin for bacterial biofilms grown on bone and muscle in vitro. Methods: Biofilms of pathogens found in musculoskeletal infections (S. aureus, S. epidermidis, E. faecalis, P. aeruginosa, and E. coli) were established for 72 hr on rabbit muscle and bone specimens in vitro and characterized by SEM imaging and CFU counts. Biofilm-covered tissue specimens were exposed to serial log2 dilutions (4000-31.25 µg/mL) of tobramycin, vancomycin, or a 1:1 combination of both drugs for 6, 24, or 72 hr. Tissues were subcultured following antimicrobial exposure to determine bacterial survival. The breakpoint concentration with no surviving bacteria was defined as the MBEC for each pathogen-antimicrobial-exposure time combination. Results: All tested pathogens formed biofilm on tissue. Tobramycin/vancomycin (1:1) was the most effective antimicrobial regimen with MBEC on muscle (10/10 pathogens) or bone (7/10 pathogens) generally in the range of 100-750 µg/mL with 24 or 72 hr exposure. MBEC decreased with exposure time for 53.3% of biofilms between 6 and 24 hr, 53.3% of biofilms between 24 and 72 hr, and for 76.7% of biofilms between 6 and 72 hr. MBECs on bone were significantly higher than corresponding MBECs on muscle tissue (p < 0.05). In most cases, tissue MBECs were lower compared to previously published MBECs for the same pathogens on polystyrene tissue-culture plates. Conclusions: The majority of MBECs for orthopaedic infections on bone and muscle are on the order of 100-750 µg/mL of vancomycin+tobramycin when sustained for at least 24 hr, which may be clinically achievable using high-dose antimicrobial-loaded bone cement (ALBC).
Collapse
Affiliation(s)
- Vajra Badha
- School of Biological & Health Systems Engineering, Arizona State University, Tempe, AZ, USA
- Sonoran Biosciences, Chandler, AZ, USA
| | - Rex Moore
- School of Biological & Health Systems Engineering, Arizona State University, Tempe, AZ, USA
- Sonoran Biosciences, Chandler, AZ, USA
| | | | | | - Alex McLaren
- School of Biological & Health Systems Engineering, Arizona State University, Tempe, AZ, USA
- Sonoran Biosciences, Chandler, AZ, USA
- University of Arizona College of Medicine, Phoenix, AZ, USA
| | - Derek Overstreet
- School of Biological & Health Systems Engineering, Arizona State University, Tempe, AZ, USA
- Sonoran Biosciences, Chandler, AZ, USA
| |
Collapse
|
44
|
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
|
45
|
Wahl P, Rönn K, Bohner M, Decosterd LA, Meier C, Schläppi M, Festa S, Gautier E. In vitro study of new combinations for local antibiotic therapy with calcium sulphate - Near constant release of ceftriaxone offers new treatment options. J Bone Jt Infect 2018; 3:212-221. [PMID: 30416946 PMCID: PMC6215989 DOI: 10.7150/jbji.26218] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 09/17/2018] [Indexed: 11/05/2022] Open
Abstract
INTRODUCTION Local application of antibiotics provides high concentrations at the site of interest, with minimal systemic toxicity. Carrier materials might help manage dead space. Calcium sulphate (CaSO4) has a dissolution time that only slightly exceeds the usually recommended duration of systemic antibiotic treatments. This in vitro study evaluates compatibility, release kinetics and antibacterial activity of new combinations of antibiotics with CaSO4 as carrier material. METHODS CaSO4 pellets added with 8% w/w antibiotic powder were exposed once in phosphate-buffered saline (PBS) solution and once in bovine plasma, in an elution experiment run over 6 weeks at 37 °C. Antibiotic elution was examined at various time points. Concentration was measured by liquid chromatography with tandem mass spectrometry. Antimicrobial activity was checked with an agar diffusion test. RESULTS Piperacillin-tazobactam, ceftazidime, cefepime, and meropenem showed fast reduction of concentration and activity. Flucloxacillin and cefuroxime remained present in relevant concentrations for 4 weeks. Ciprofloxacin, levofloxacin and clindamycin lasted for 6 weeks, but also at cell toxic concentrations. Ceftriaxone showed a near-constant release with only a small reduction of concentration from 130 to 75 mg/l. Elution profiles from PBS and plasma were comparable. CONCLUSION CaSO4 provides new possibilities in the local treatment of bone and joint infections. Ceftriaxone appears to be of particular interest in combination with CaSO4. Release persists at clinically promising concentrations, and appears to have a depot-like slow release from CaSO4, with only a small reduction in activity and concentration over 6 weeks. To the best of our knowledge, such a particular persistent release never was described before, for any antibiotic in combination with a carrier material for local application.
Collapse
Affiliation(s)
- Peter Wahl
- Department of Orthopaedic Surgery, HFR Fribourg - Cantonal Hospital, Fribourg, Switzerland.,Division of Orthopaedics and Traumatology, Cantonal Hospital Winterthur, Winterthur, Switzerland
| | - Karolin Rönn
- Department of Orthopaedic Surgery, HFR Fribourg - Cantonal Hospital, Fribourg, Switzerland.,Schulthess Clinic, Zürich, Switzerland
| | | | - Laurent A Decosterd
- Division of Clinical Pharmacology, University Hospital Centre CHUV, Lausanne, Switzerland
| | - Christoph Meier
- Division of Orthopaedics and Traumatology, Cantonal Hospital Winterthur, Winterthur, Switzerland
| | - Michel Schläppi
- Division of Orthopaedics and Traumatology, Cantonal Hospital Winterthur, Winterthur, Switzerland
| | - Sandrine Festa
- Department of Orthopaedic Surgery, HFR Fribourg - Cantonal Hospital, Fribourg, Switzerland
| | - Emanuel Gautier
- Department of Orthopaedic Surgery, HFR Fribourg - Cantonal Hospital, Fribourg, Switzerland
| |
Collapse
|
46
|
Broussou DC, Lacroix MZ, Toutain PL, Woehrlé F, El Garch F, Bousquet-Melou A, Ferran AA. Differential Activity of the Combination of Vancomycin and Amikacin on Planktonic vs. Biofilm-Growing Staphylococcus aureus Bacteria in a Hollow Fiber Infection Model. Front Microbiol 2018; 9:572. [PMID: 29636741 PMCID: PMC5880918 DOI: 10.3389/fmicb.2018.00572] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 03/13/2018] [Indexed: 11/13/2022] Open
Abstract
Combining currently available antibiotics to optimize their use is a promising strategy to reduce treatment failures against biofilm-associated infections. Nevertheless, most assays of such combinations have been performed in vitro on planktonic bacteria exposed to constant concentrations of antibiotics over only 24 h and the synergistic effects obtained under these conditions do not necessarily predict the behavior of chronic clinical infections associated with biofilms. To improve the predictivity of in vitro combination assays for bacterial biofilms, we first adapted a previously described Hollow-fiber (HF) infection model by allowing a Staphylococcus aureus biofilm to form before drug exposure. We then mimicked different concentration profiles of amikacin and vancomycin, similar to the free plasma concentration profiles that would be observed in patients treated daily over 5 days. We assessed the ability of the two drugs, alone or in combination, to reduce planktonic and biofilm-embedded bacterial populations, and to prevent the selection of resistance within these populations. Although neither amikacin nor vancomycin exhibited any bactericidal activity on S. aureus in monotherapy, the combination had a synergistic effect and significantly reduced the planktonic bacterial population by -3.0 to -6.0 log10 CFU/mL. In parallel, no obvious advantage of the combination, as compared to amikacin alone, was demonstrated on biofilm-embedded bacteria for which the addition of vancomycin to amikacin only conferred a further maximum reduction of 0.3 log10 CFU/mL. No resistance to vancomycin was ever found whereas a few bacteria less-susceptible to amikacin were systematically detected before treatment. These resistant bacteria, which were rapidly amplified by exposure to amikacin alone, could be maintained at a low level in the biofilm population and even suppressed in the planktonic population by adding vancomycin. In conclusion, by adapting the HF model, we were able to demonstrate the different bactericidal activities of the vancomycin and amikacin combination on planktonic and biofilm-embedded bacterial populations, suggesting that, for biofilm-associated infections, the efficacy of this combination would not be much greater than with amikacin monotherapy. However, adding vancomycin could reduce possible resistance to amikacin and provide a relevant strategy to prevent the selection of antibiotic-resistant bacteria during treatments.
Collapse
Affiliation(s)
- Diane C Broussou
- INTHERES, INRA, ENVT, Université de Toulouse, Toulouse, France.,Vétoquinol, Global Drug Development, Lure, France
| | | | - Pierre-Louis Toutain
- Department of Veterinary Basic Sciences, Royal Veterinary College, London, United Kingdom
| | | | | | | | - Aude A Ferran
- INTHERES, INRA, ENVT, Université de Toulouse, Toulouse, France
| |
Collapse
|
47
|
Wahl P, Guidi M, Benninger E, Rönn K, Gautier E, Buclin T, Magnin JL, Livio F. The levels of vancomycin in the blood and the wound after the local treatment of bone and soft-tissue infection with antibiotic-loaded calcium sulphate as carrier material. Bone Joint J 2017; 99-B:1537-1544. [PMID: 29092996 DOI: 10.1302/0301-620x.99b11.bjj-2016-0298.r3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 06/08/2017] [Indexed: 11/05/2022]
Abstract
AIMS Calcium sulphate (CaSO4) is a resorbable material that can be used simultaneously as filler of a dead space and as a carrier for the local application of antibiotics. Our aim was to describe the systemic exposure and the wound fluid concentrations of vancomycin in patients treated with vancomycin-loaded CaSO4 as an adjunct to the routine therapy of bone and joint infections. PATIENTS AND METHODS A total of 680 post-operative blood and 233 wound fluid samples were available for analysis from 94 implantations performed in 87 patients for various infective indications. Up to 6 g of vancomycin were used. Non-compartmental pharmacokinetic analysis was performed on the data from 37 patients treated for an infection of the hip. RESULTS The overall systemic exposure remained within a safe range, even in patients with post-operative renal failure, none requiring removal of the pellets. Local concentrations were approximately ten times higher than with polymethylmethacrylate (PMMA) as a carrier, but remained below reported cell toxicity thresholds. Decreasing concentrations in wound fluid were observed over several weeks, but remained above the common minimum inhibitory concentrations for Staphylococcus up to three months post-operatively. CONCLUSION This study provides the first pharmacokinetic description of the local application of vancomycin with CaSO4 as a carrier, documenting slow release, systemic safety and a release profile far more interesting than from PMMA. In particular, considering in vitro data, concentrations of vancomycin active against staphylococcal biofilm were seen for several weeks. Cite this article: Bone Joint J 2017;99-B:1537-44.
Collapse
Affiliation(s)
- P Wahl
- HFR Fribourg - Cantonal Hospital, 1708 Fribourg, Switzerland, and Division of Orthopaedics and Traumatology, Cantonal Hospital Winterhur, 8401 Winterthur, Switzerland
| | - M Guidi
- Pharmacometrician, Division of Clinical Pharmacology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland, and School of Pharmaceutical Sciences, University of Geneva and University of Lausanne, 1205 Geneva, Switzerland
| | - E Benninger
- Cantonal Hospital Winterhur, 8401 Winterthur, Switzerland
| | - K Rönn
- HFR Fribourg - Cantonal Hospital, 1708 Fribourg, Switzerland and Schulthess Clinic, 8008 Zürich, Switzerland
| | - E Gautier
- HFR Fribourg - Cantonal Hospital, 1708 Fribourg, Switzerland
| | - T Buclin
- Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland
| | - J-L Magnin
- HFR Fribourg - Cantonal Hospital, 1708 Fribourg, Switzerland
| | - F Livio
- Centre Hospitalier Universitaire Vaudois, 1011 Lausanne, Switzerland
| |
Collapse
|
48
|
Krishnaiah M, de Almeida NR, Udumula V, Song Z, Chhonker YS, Abdelmoaty MM, do Nascimento VA, Murry DJ, Conda-Sheridan M. Synthesis, biological evaluation, and metabolic stability of phenazine derivatives as antibacterial agents. Eur J Med Chem 2017; 143:936-947. [PMID: 29227933 DOI: 10.1016/j.ejmech.2017.11.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 11/06/2017] [Accepted: 11/08/2017] [Indexed: 11/27/2022]
Abstract
Drug-resistant pathogens are a major cause of hospital- and community-associated bacterial infections in the United States and around the world. These infections are increasingly difficult to treat due to the development of antibiotic resistance and the formation of bacterial biofilms. In the paper, a series of phenazines were synthesized and evaluated for their in vitro antimicrobial activity against Gram positive (methicillin resistant staphylococcus aureus, MRSA) and Gram negative (Escherichia coli, E. coli) bacteria. The compound 6,9-dichloro-N-(methylsulfonyl)phenazine-1-carboxamide (18c) proved to be the most active molecule (MIC = 16 μg/mL) against MRSA whereas 9-methyl-N-(methylsulfonyl)phenazine-1-carboxamide (30e) showed good activity against both MRSA (MIC = 32 μg/mL) and E. coli (MIC = 32 μg/mL). Molecule 18c also demonstrated significant biofilm dispersion and inhibition against S. aureus. Preliminary studies indicate the molecules do not disturb bacterial membranes and there activity is not directly linked to the generation of reactive oxygen species. Compound 18c displayed minor toxicity against mammalian cells. Metabolic stability studies of the most promising compounds indicate stability towards phase I and phase II metabolizing enzymes.
Collapse
Affiliation(s)
- Maddeboina Krishnaiah
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Nathalia Rodrigues de Almeida
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Venkatareddy Udumula
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Zhongcheng Song
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA; School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, People's Republic of China
| | - Yashpal Singh Chhonker
- Department of Pharmacy Practice, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mai M Abdelmoaty
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA; Therapeutic Chemistry Department, Pharmaceutical and Drug Industries, Research Division, National Research Centre, Giza, Egypt
| | - Valter Aragao do Nascimento
- Group of Spectroscopy and Bioinformatics Applied to Biodiversity and Health, School of Medicine, Federal University of Mato Grosso Do Sul, Campo Grande, MS 79070900, Brazil
| | - Daryl J Murry
- Department of Pharmacy Practice, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Martin Conda-Sheridan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
49
|
Working ZM, Frederiksen H, Drew A, Loc-Carrillo C, Kubiak EN. Bone penetrance of locally administered vancomycin powder in a rat femur fracture model. Injury 2017; 48:1459-1465. [PMID: 28456368 DOI: 10.1016/j.injury.2017.04.040] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 04/18/2017] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Locally delivered, crystalline vancomycin has been suggested as a potential prophylactic measure against the development of deep and superficial surgical site infection. Clinical expectations regarding the duration and peak of drug concentration in local tissues following administration are unknown. Our goal was to develop concentration vs time curves for locally administered vancomycin powder in a high-energy, open femur fracture rat model in local tissues and to compare that data to two well performed similar, systemic administration studies. METHODS After approval for animal research, 24 adult Sprague-Dawley rats sustained closed, midshaft femoral fracture under anesthesia. Fractures were caused via blunt guillotine with 750g metal rod dropped 50cm. Injured hindlimbs were surgically opened at fracture to simulate open injury and stabilized using 0.054 Kirschner wires. Vancomycin powder was administered using weight-based protocol (goal: 25mg/kg). Rats were sacrificed in groups of 4 at 4, 8, 24, 48, 72, 96h. Samples harvested included rat-tail venous blood prior to sacrifice, and femoral bone and anterior thigh soft-tissue were harvested post-mortem. High Performance Liquid Chromatography (HPLC) was performed on all samples. RESULTS Concentration vs. time curves demonstrated that the surrounding soft-tissues demonstrated highest maximum concentration (1.5mg vancomycin/g muscle). Bone reached maximum average of 199μg vancomycin/g femur: approximately 13% of maximal soft-tissue absorption. Plasma reached maximum concentration of 1.8μg/mL plasma. All peaks at t=4h. Within 48h, average muscle vancomycin concentration dropped to 3μg/g muscle (0.2% maximum muscle concentration) and the average bone concentration dropped to 1.9μg/g femur (0.9% maximum bone concentration). Vancomycin was undetectable on all samples at 96h. Comparison to classical animal studies suggest local delivery to bone exceeds that of IV dosing for approximately 48h and may peak near concentrations of 102 multiples. CONCLUSIONS Locally administered vancomycin provides drug delivery in excess of IV dosing for approximately 48h after intervention. Exponential decay demonstrates rapid removal of drug to near undetectable levels in bone, plasma, and local soft tissue thereafter in a rat model. Local delivery may generate concentrations exceeding that achievable by steady state systemic dosing for 48h.
Collapse
Affiliation(s)
- Zachary M Working
- University of Utah Department of Orthopaedic Surgery, Salt Lake City, UT, USA.
| | | | - Alex Drew
- University of Utah Department of Orthopaedic Surgery, Salt Lake City, UT, USA
| | | | - Erik N Kubiak
- University of Utah Department of Orthopaedic Surgery, Salt Lake City, UT, USA
| |
Collapse
|
50
|
Abstract
Chronic infections are one of the major challenges in orthopaedic surgery, both for surgeons and patients. They are characterised by obstinate persistency of the causing microorganisms and resulting long-term disablement of the patients, associated with remarkable costs for the health care system. Difficulties derive from the biofilm-mode of living of pathogens with resistances against immunological defence and antimicrobial substances, and osseous defects resulting from the disease itself and surgical interventions. Established techniques usually require multiple costly operations with extended periods of disablement and impairment of the patients, sometimes making the therapy worse than the disease. Better understanding of the backgrounds of the conditions has led to new surgical techniques and differentiated application of antibiotics, aiming in improved quality of life for our patients.
Cite this article: EFORT Open Rev 2017;2. DOI: 10.1302/2058-5241.2.160063. Originally published online at www.efortopenreviews.org
Collapse
Affiliation(s)
- Heinz Winkler
- Osteitis Centre, Privatklinik Döbling Wien, Heiligenstaedter Strasse 57-63, A-1190 Wien, Austria
| |
Collapse
|