1
|
Athanasiou V, Ragias D, Tzikopoulou M, Zenios M, Falagas ME. Tedizolid for osteoarticular infections: Evaluation of the published evidence. Eur J Pharmacol 2025; 998:177458. [PMID: 40090537 DOI: 10.1016/j.ejphar.2025.177458] [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: 10/31/2024] [Revised: 02/28/2025] [Accepted: 03/04/2025] [Indexed: 03/18/2025]
Abstract
INTRODUCTION Tedizolid phosphate, an oxazolidinone antibiotic, has been approved for the treatment of acute bacterial skin and skin structure infections (ABSSSIs). However, its off-label use has been reported in various infections, including osteoarticular infections. METHODS A systematic review of data from PubMed, Scopus, and Web of Science was conducted to evaluate the antimicrobial activity, safety, and effectiveness of tedizolid in patients with bone and joint infections, including prosthetic joint infections, osteomyelitis, and septic arthritis. The review encompassed clinical trials, prospective and retrospective studies, and case reports. RESULTS A total of 6 in vitro antimicrobial and 15 clinical studies were included in the review. Tedizolid demonstrated high antimicrobial activity across all in vitro studies. In 106 patients from the included clinical studies, tedizolid showed high effectiveness, with therapy success ranging from 76.5 % to 100 % in 4 cohort studies. Additionally, favorable outcomes were reported in 7 of 9 case reports. Tedizolid exhibited a favorable safety profile, with 11 of 15 clinical studies reporting no adverse events in 37 patients. Adverse events leading to therapy discontinuation were observed in 9 out of 124 patients included in the remaining studies. CONCLUSION The current appraisal suggests that tedizolid is a promising antibiotic for the treatment of bone and joint infections. Nonetheless, its use should be reserved for multi-drug resistant infections when other approved therapeutic options are limited. Further clinical studies are warranted to substantiate the effectiveness and safety of tedizolid in this patient population.
Collapse
Affiliation(s)
| | | | | | - Michalis Zenios
- European University Cyprus School of Medicine, Nicosia, Cyprus
| | - Matthew E Falagas
- Alfa Institute of Biomedical Sciences, Athens, Greece; European University Cyprus School of Medicine, Nicosia, Cyprus; Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
2
|
Al Janabi J, Tevell S, Sieber RN, Stegger M, Söderquist B. Emerging resistance in Staphylococcus epidermidis during dalbavancin exposure: a case report and in vitro analysis of isolates from prosthetic joint infections. J Antimicrob Chemother 2023; 78:669-677. [PMID: 36611258 PMCID: PMC9978592 DOI: 10.1093/jac/dkac434] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 12/07/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Dalbavancin, a semisynthetic lipoglycopeptide with exceptionally long half-life and Gram-positive spectrum, is an attractive option for infections requiring prolonged therapy, including prosthetic joint infections (PJIs). OBJECTIVES To investigate the prevalence of reduced susceptibility to dalbavancin in a strain collection of Staphylococcus epidermidis from PJIs, and to investigate genomic variation in isolates with reduced susceptibility selected during growth under dalbavancin exposure. METHODS MIC determination was performed on S. epidermidis isolates from a strain collection (n = 64) and from one patient with emerging resistance during treatment (n = 4). These isolates were subsequently cultured on dalbavancin-containing agar and evaluated at 48 h; MIC determination was repeated if phenotypical heterogeneity was detected during growth. Population analysis profile (PAP-AUC) was performed in isolates where a ≥ 2-fold increase in MIC was detected, together with corresponding parental isolates (n = 21). Finally, WGS was performed. RESULTS All strains grew at 48 h on agar containing 0.125 mg/L dalbavancin. PAP-AUC demonstrated significant differences between parental and derived strains in four of the eight analysed groups. An amino acid change in the walK gene coinciding with emergence of phenotypic resistance was detected in the patient isolates, whereas no alterations were found in this region in the in vitro derived strains. CONCLUSIONS Exposure to dalbavancin may lead to reduced susceptibility to dalbavancin through either selection of pre-existing subpopulations, epigenetic changes or spontaneous mutations during antibiotic exposure. Source control combined with adequate antibiotic concentrations may be important to prevent emerging reduced susceptibility during dalbavancin treatment.
Collapse
Affiliation(s)
| | | | - Raphael Niklaus Sieber
- Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | - Marc Stegger
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden,Department of Bacteria, Parasites and Fungi, Statens Serum Institut, Copenhagen, Denmark
| | | |
Collapse
|
3
|
Blaskovich MAT, Hansford KA, Butler MS, Ramu S, Kavanagh AM, Jarrad AM, Prasetyoputri A, Pitt ME, Huang JX, Lindahl F, Ziora ZM, Bradford T, Muldoon C, Rajaratnam P, Pelingon R, Edwards DJ, Zhang B, Amado M, Elliott AG, Zuegg J, Coin L, Woischnig AK, Khanna N, Breidenstein E, Stincone A, Mason C, Khan N, Cho HK, Karau MJ, Greenwood-Quaintance KE, Patel R, Wootton M, James ML, Hutton ML, Lyras D, Ogunniyi AD, Mahdi LK, Trott DJ, Wu X, Niles S, Lewis K, Smith JR, Barber KE, Yim J, Rice SA, Rybak MJ, Ishmael CR, Hori KR, Bernthal NM, Francis KP, Roberts JA, Paterson DL, Cooper MA. A lipoglycopeptide antibiotic for Gram-positive biofilm-related infections. Sci Transl Med 2022; 14:eabj2381. [DOI: 10.1126/scitranslmed.abj2381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Drug-resistant Gram-positive bacterial infections are still a substantial burden on the public health system, with two bacteria (
Staphylococcus aureus
and
Streptococcus pneumoniae
) accounting for over 1.5 million drug-resistant infections in the United States alone in 2017. In 2019, 250,000 deaths were attributed to these pathogens globally. We have developed a preclinical glycopeptide antibiotic, MCC5145, that has excellent potency (MIC
90
≤ 0.06 μg/ml) against hundreds of isolates of methicillin-resistant
S. aureus
(MRSA) and other Gram-positive bacteria, with a greater than 1000-fold margin over mammalian cell cytotoxicity values. The antibiotic has therapeutic in vivo efficacy when dosed subcutaneously in multiple murine models of established bacterial infections, including thigh infection with MRSA and blood septicemia with
S. pneumoniae
, as well as when dosed orally in an antibiotic-induced
Clostridioides difficile
infection model. MCC5145 exhibited reduced nephrotoxicity at microbiologically active doses in mice compared to vancomycin. MCC5145 also showed improved activity against biofilms compared to vancomycin, both in vitro and in vivo, and a low propensity to select for drug resistance. Characterization of drug action using a transposon library bioinformatic platform showed a mechanistic distinction from other glycopeptide antibiotics.
Collapse
Affiliation(s)
- Mark A. T. Blaskovich
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Karl A. Hansford
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Mark S. Butler
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Soumya Ramu
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Angela M. Kavanagh
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Angie M. Jarrad
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Anggia Prasetyoputri
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Miranda E. Pitt
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Johnny X. Huang
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Fredrik Lindahl
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Zyta M. Ziora
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Tanya Bradford
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Craig Muldoon
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Premraj Rajaratnam
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Ruby Pelingon
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - David J. Edwards
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Bing Zhang
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Maite Amado
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Alysha G. Elliott
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Johannes Zuegg
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Lachlan Coin
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Anne-Kathrin Woischnig
- University and University Hospital of Basel, Division of Infectious Diseases and Infection Biology Laboratory Department of Biomedicine, Hebelstrasse 20, CH-4031 Basel, Switzerland
| | - Nina Khanna
- University and University Hospital of Basel, Division of Infectious Diseases and Infection Biology Laboratory Department of Biomedicine, Hebelstrasse 20, CH-4031 Basel, Switzerland
| | - Elena Breidenstein
- Summit Therapeutics, The Works, Unity Campus, Cambridgeshire, CB22 3FT, UK
| | - Anna Stincone
- Summit Therapeutics, The Works, Unity Campus, Cambridgeshire, CB22 3FT, UK
| | - Clive Mason
- Summit Therapeutics, The Works, Unity Campus, Cambridgeshire, CB22 3FT, UK
| | - Nawaz Khan
- Summit Therapeutics, The Works, Unity Campus, Cambridgeshire, CB22 3FT, UK
| | - Hye-Kyung Cho
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Melissa J. Karau
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Kerryl E. Greenwood-Quaintance
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905, USA
- Division of Public Health, Infectious Diseases and Occupational Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Mandy Wootton
- Specialist Antimicrobial Chemotherapy Unit Public Health Wales, University Hospital of Wales, Heath Park, Cardiff CF14 4XW, Wales
| | - Meagan L. James
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
| | - Melanie L. Hutton
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
| | - Dena Lyras
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Microbiology, Monash University, Clayton, Victoria 3800, Australia
| | - Abiodun D. Ogunniyi
- Australian Centre for Antimicrobial Resistance Ecology, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, South Australia 5371, Australia
| | - Layla K. Mahdi
- Australian Centre for Antimicrobial Resistance Ecology, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, South Australia 5371, Australia
| | - Darren J. Trott
- Australian Centre for Antimicrobial Resistance Ecology, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, South Australia 5371, Australia
| | - Xiaoqian Wu
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Samantha Niles
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Kim Lewis
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA
| | - Jordan R. Smith
- Anti-Infective Research Laboratory, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Katie E. Barber
- Anti-Infective Research Laboratory, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Juwon Yim
- Anti-Infective Research Laboratory, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Seth Alan Rice
- Anti-Infective Research Laboratory, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
| | - Michael J. Rybak
- Anti-Infective Research Laboratory, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48201, USA
- School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Chad R. Ishmael
- Department of Orthopaedic Surgery, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Kellyn R. Hori
- Department of Orthopaedic Surgery, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Nicholas M. Bernthal
- Department of Orthopaedic Surgery, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Kevin P. Francis
- Department of Orthopaedic Surgery, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
- PerkinElmer, 68 Elm Street, Hopkinton, MA 01748, USA
| | - Jason A. Roberts
- University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Queensland 4029, Australia
- Departments of Pharmacy and Intensive Care Medicine, Royal Brisbane and Women’s Hospital, Brisbane, Queensland 4029, Australia
- Division of Anaesthesiology Critical Care Emergency and Pain Medicine, Nîmes University Hospital, University of Montpellier, 30029 Nîmes, France
| | - David L. Paterson
- University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Queensland 4029, Australia
| | - Matthew A. Cooper
- Centre for Superbug Solutions, Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland 4072, Australia
| |
Collapse
|
4
|
Liu F, Rajabi S, Shi C, Afifirad G, Omidi N, Kouhsari E, Khoshnood S, Azizian K. Antibacterial activity of recently approved antibiotics against methicillin-resistant Staphylococcus aureus (MRSA) strains: A systematic review and meta-analysis. Ann Clin Microbiol Antimicrob 2022; 21:37. [PMID: 35978400 PMCID: PMC9382732 DOI: 10.1186/s12941-022-00529-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 08/10/2022] [Indexed: 11/16/2022] Open
Abstract
Background Methicillin-resistant Staphylococcus aureus (MRSA) infections are considered an important public health problem, and treatment options are limited. Accordingly, in this meta-analysis, we analyzed published studies to survey in vitro activity of recently approved antibiotics against MRSA isolates. Methods We searched electronic databases; PubMed, Scopus, and Web of Science to identify relevant studies (until November 30, 2020) that have focused on the in vitro activity of telavancin, dalbavancin, oritavancin, and tedizolid against MRSA isolates. Statistical analyses were conducted using STATA software (version 14.0). Results Thirty-eight studies were included in this meta-analysis. Overall in vitro activity of tedizolid on 12,204 MRSA isolates was 0.250 and 0.5 µg/mL for MIC50 and MIC90, (minimum inhibitory concentration at which 50% and 90% of isolates were inhibited, respectively), respectively. The overall antibacterial activity of dalbavancin on 28539 MRSA isolates was 0.060 and 0.120 µg/mL for MIC50 and MIC90, respectively. The overall antibacterial activity of oritavancin on 420 MRSA isolates was 0.045 and 0.120 µg/mL for MIC50 and MIC90, respectively. The overall antibacterial activity of telavancin on 7353 MRSA isolates was 0.032 and 0.060 µg/mL for MIC50 and MIC90, respectively. The pooled prevalence of tedizolid, telavancin, and dalbavancin susceptibility was 100% (95% CI: 100–100). Conclusion Telavancin, dalbavancin, oritavancin, and tedizolid had potent in vitro activity against MRSA isolates. The low MICs and high susceptibility rates of these antibiotics recommend a hopeful direction to introduce useful antibiotics in treating MRSA infections in the future. Supplementary Information The online version contains supplementary material available at 10.1186/s12941-022-00529-z.
Collapse
Affiliation(s)
- Fei Liu
- Department of Biomedical Engineering, Changzhi Medical College, Changzhi, 046013, Shanxi, China
| | - Sajad Rajabi
- International Medical Campus, Iran University of Medical Sciences, Tehran, Iran
| | - Chunhua Shi
- Department of Biomedical Engineering, Changzhi Medical College, Changzhi, 046013, Shanxi, China.
| | - Ghazale Afifirad
- Department of Microbiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nazanin Omidi
- Clinical Microbiology Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Ebrahim Kouhsari
- Laboratory Sciences Research Center, Golestan University of Medical Sciences, Gorgan, Iran.,Department of Laboratory Sciences, Faculty of Paramedicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Saeed Khoshnood
- Clinical Microbiology Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Khalil Azizian
- Department of Microbiology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran.
| |
Collapse
|
5
|
Cacopardo B, Cattaneo D, Cortese F, Di Luca M, Falcone M, Marchetti G, Tascini C, Tiseo G, Venditti M. Role of dalbavancin as combination therapy: evidences from the literature and clinical scenarios. Expert Rev Anti Infect Ther 2022; 20:997-1004. [PMID: 35353020 DOI: 10.1080/14787210.2022.2060820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The off-label use of dalbavancin in patients with infections other than acute bacterial skin and skin structure infections (ABSSSI) represents an interesting therapeutic option. Its use as monotherapy or in combination with other antibiotics should be better defined. AREAS COVERED The aim of this review is to summarize evidence about the potential role of dalbavancin in combination with other antibiotics and describe clinical scenarios in which combination regimens including dalbavancin are useful. The studies were retrieved from PubMed using different combinations of keywords ("dalbavancin", "combination", "synergy"). EXPERT OPINION Limited data about the use of dalbavancin in monotherapy or combined with other antibiotics are available. In vitro assays showed a synergistic effect of dalbavancin when combined with beta-lactam antibiotics. The use of dalbavancin as combination therapy in patients with ABSSSI did not demonstrate a superiority compared to monotherapy. Conversely, combination regimens including dalbavancin may be useful in specific infection types, such as bone and prosthetic joint infections or subacute/chronic intravascular infections with no possibility of device removal. Potential partner drugs might be rifampin, beta-lactams, fluoroquinolones, doxycycline and trimethoprim/sulfamethoxazole. The choice of the companion drug should be tailored on in vitro results of synergistic tests, patient's profile and type of infection.
Collapse
Affiliation(s)
- Bruno Cacopardo
- Department of Clinical and Molecular Biomedicine, Chair of Infectious Diseases, University of Catania, 95124 Catania, Italy
| | - Dario Cattaneo
- Unit of Clinical Pharmacology, Fatebenefratelli Sacco University Hospital, Milan, Italy
| | | | | | - Marco Falcone
- Division of Infectious Diseases, Department of Clinical and Experimental Medicine, University of Pisa
| | - Giulia Marchetti
- Clinic of Infectious Disease and Tropical Medicine, Department of Health Sciences, University of Milan, ASST Santi Paolo e Carlo, Milan, Italy
| | - Carlo Tascini
- Department of Medicine, University of Udine, Udine, Italy
| | - Giusy Tiseo
- Division of Infectious Diseases, Department of Clinical and Experimental Medicine, University of Pisa
| | - Mario Venditti
- Department of Public Health and Infectious Diseases, 'Sapienza' University of Rome, Italy
| |
Collapse
|
6
|
Gatti M, Barnini S, Guarracino F, Parisio EM, Spinicci M, Viaggi B, D’Arienzo S, Forni S, Galano A, Gemmi F. Orthopaedic Implant-Associated Staphylococcal Infections: A Critical Reappraisal of Unmet Clinical Needs Associated with the Implementation of the Best Antibiotic Choice. Antibiotics (Basel) 2022; 11:antibiotics11030406. [PMID: 35326869 PMCID: PMC8944676 DOI: 10.3390/antibiotics11030406] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 12/24/2022] Open
Abstract
Infections associated with orthopaedic implants represent a major health concern characterized by a remarkable incidence of morbidity and mortality. The wide variety of clinical scenarios encountered in the heterogeneous world of infections associated with orthopaedic implants makes the implementation of an optimal and standardized antimicrobial treatment challenging. Antibiotic bone penetration, anti-biofilm activity, long-term safety, and drug choice/dosage regimens favouring outpatient management (i.e., long-acting or oral agents) play a major role in regards to the chronic evolution of these infections. The aim of this multidisciplinary opinion article is to summarize evidence supporting the use of the different anti-staphylococcal agents in terms of microbiological and pharmacological optimization according to bone penetration, anti-biofilm activity, long-term safety, and feasibility for outpatient regimens, and to provide a useful guide for clinicians in the management of patients affected by staphylococcal infections associated with orthopaedic implants Novel long-acting lipoglycopeptides, and particularly dalbavancin, alone or in combination with rifampicin, could represent the best antibiotic choice according to real-world evidence and pharmacokinetic/pharmacodynamic properties. The implementation of a multidisciplinary taskforce and close cooperation between microbiologists and clinicians is crucial for providing the best care in this scenario.
Collapse
Affiliation(s)
- Milo Gatti
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40138 Bologna, Italy;
- SSD Clinical Pharmacology, Department for Integrated Infectious Risk Management, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Simona Barnini
- Bacteriology Unit, Azienda Ospedaliero Universitaria Pisana, 56126 Pisa, Italy;
| | - Fabio Guarracino
- Department of Anaesthesia and Critical Care Medicine, Azienda Ospedaliero Universitaria Pisana, 56126 Pisa, Italy;
| | - Eva Maria Parisio
- UOSD Microbiologia Arezzo PO San Donato, Azienda Usl Toscana Sud Est, 52100 Arezzo, Italy;
| | - Michele Spinicci
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy;
- Infectious and Tropical Diseases Unit, Careggi University Hospital, 50134 Florence, Italy
| | - Bruno Viaggi
- Neurointensive Care Unit, Department of Anesthesiology, Careggi University Hospital, 50134 Florence, Italy;
| | - Sara D’Arienzo
- Agenzia Regionale di Sanità della Toscana, 50141 Florence, Italy; (S.D.); (S.F.)
| | - Silvia Forni
- Agenzia Regionale di Sanità della Toscana, 50141 Florence, Italy; (S.D.); (S.F.)
| | - Angelo Galano
- SOD Microbiologia e Virologia, Careggi University Hospital, 50134 Florence, Italy;
| | - Fabrizio Gemmi
- Agenzia Regionale di Sanità della Toscana, 50141 Florence, Italy; (S.D.); (S.F.)
- Correspondence:
| |
Collapse
|
7
|
Thill P, Robineau O, Roosen G, Patoz P, Gachet B, Lafon-Desmurs B, Tetart M, Nadji S, Senneville E, Blondiaux N. Rifabutin versus rifampicin bactericidal and antibiofilm activities against clinical strains of Staphylococcus spp. isolated from bone and joint infections. J Antimicrob Chemother 2022; 77:1036-1040. [PMID: 35028671 DOI: 10.1093/jac/dkab486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 12/08/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Staphylococci account for approximately 60% of periprosthetic joint infections (PJIs). Rifampicin (RMP) combination therapy is generally considered to be the treatment of choice for staphylococcal PJIs but carries an important risk of adverse events and drug-drug interactions. Rifabutin (RFB) shares many of the properties of rifampicin but causes fewer adverse events. OBJECTIVES To compare the minimal inhibitory concentration (MIC), the minimum bactericidal concentrations (MBC), and the minimum biofilm eradication concentrations (MBEC) of rifabutin and rifampicin for staphylococcal clinical strains isolated from PJIs. METHODS 132 clinical strains of rifampicin-susceptible staphylococci [51 Staphylococcus aureus (SA), 48 Staphylococcus epidermidis (SE) and 33 other coagulase-negative staphylococci (CoNS)] were studied. The MBC and the MBEC were determined using the MBEC® Assay for rifabutin and rifampicin and were compared. RESULTS When compared with the rifampicin MIC median value, the rifabutin MIC median value was significantly higher for SA (P < 0.05), but there was no statistically significant difference for SE (P = 0.25) and CoNS (P = 0.29). The rifabutin MBC median value was significantly higher than that of rifampicin for SA (P = 0.003) and was lower for SE (P = 0.003) and CoNS (P = 0.03). Rifabutin MBEC median value was statistically lower than that of rifampicin for all strains tested. CONCLUSIONS Using the determination of MBEC values, our study suggests that rifabutin is more effective than rifampicin against clinical strains of Staphylococcus spp. obtained from PJIs. Using MBECs instead of MICs seems to be of interest when considering biofilms. In vivo higher efficacy of rifabutin when compared with rifampicin needs to be confirmed.
Collapse
Affiliation(s)
- Pauline Thill
- Department of Infectious Diseases, Hospital of Tourcoing, Tourcoing, France
| | - Olivier Robineau
- Department of Infectious Diseases, Hospital of Tourcoing, Tourcoing, France.,EA2694, Univ Lille, Centre Hospitalier de Tourcoing, Tourcoing, France
| | - Gabrielle Roosen
- Department of Bacteriology, Hospital of Tourcoing, Tourcoing, France
| | - Pierre Patoz
- Department of Bacteriology, Hospital of Tourcoing, Tourcoing, France
| | - Benoit Gachet
- Department of Infectious Diseases, Hospital of Tourcoing, Tourcoing, France.,EA2694, Univ Lille, Centre Hospitalier de Tourcoing, Tourcoing, France
| | | | - Macha Tetart
- Department of Infectious Diseases, Hospital of Tourcoing, Tourcoing, France
| | - Safia Nadji
- Department of Bacteriology, Hospital of Tourcoing, Tourcoing, France
| | - Eric Senneville
- Department of Infectious Diseases, Hospital of Tourcoing, Tourcoing, France.,EA2694, Univ Lille, Centre Hospitalier de Tourcoing, Tourcoing, France
| | - Nicolas Blondiaux
- Department of Bacteriology, Hospital of Tourcoing, Tourcoing, France.,Univ. Lille, CNRS, Inserm, Institut Pasteur de Lille, U1019 - UMR9017 Center for Infection and Immunity of Lille, Lille, France
| |
Collapse
|
8
|
Lee Y, Kim SS, Choi SM, Bae CJ, Oh TH, Kim SE, Kim UJ, Kang SJ, Jung SI, Park KH. Rifamycin resistance, rpoB gene mutation, and clinical outcomes of Staphylococcus species isolated from prosthetic joint infections in Korea. J Glob Antimicrob Resist 2021; 28:43-48. [PMID: 34920175 DOI: 10.1016/j.jgar.2021.12.005] [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] [Received: 09/30/2021] [Revised: 11/11/2021] [Accepted: 12/06/2021] [Indexed: 11/16/2022] Open
Abstract
PURPOSE We conducted an in vitro investigation of rifamycin activity against planktonic and biofilm states of Staphylococcus aureus and S. epidermidis isolated from patients with prosthetic join infection (PJIs), characterized their rpoB mutations, and analyzed the clinical outcomes of rifampin-resistant isolates. METHODS A total of 110 staphylococcal isolates were collected from patients with PJIs. Antimicrobials tested using the broth microdilution method included rifampin, rifabutin, rifapentine, and rifaximin. We evaluated rpoB gene mutations to identify rifampin resistance mechanisms. Clinical outcomes were assessed in rifampin-resistant isolates. RESULTS Among the 110 staphylococcal isolates, we detected 85 S. aureus (methicillin-resistant, 55%) and 25 S. epidermidis (methicillin-resistant, 100%) isolates. Seven S. aureus isolates and two S. epidermidis isolates were rifampin resistant (minimum inhibitory concentration [MIC], ≥ 2 μg/mL); these exhibited rpoB gene mutations. Among 78 rifampin-susceptible S. aureus isolates and 23 S. epidermidis isolates, 76 S. aureus isolates and all S. epidermidis isolates were highly susceptible (MIC ≤ 0.06 μg/mL) to other rifamycins. The rifampin, rifabutin, rifapentine, and rifaximin minimum biofilm bactericidal concentrations for ≥ 50% of isolates were 4, 1, 2, and 4 μg/mL for S. aureus and 1, 0.125, 0.25, and 0.5 μg/mL for S. epidermidis, respectively, among rifampin-susceptible isolates. Among nine patients bearing rifampin-resistant isolates, only three (33%) had successful outcomes. CONCLUSIONS Rifamycins other than rifampin show promising staphylococcal activity, including antibiofilm activity. Rifamycin-resistant staphylococci exhibit rpoB gene mutations.
Collapse
Affiliation(s)
- Yunjung Lee
- Department of Infectious Diseases, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Soo Sung Kim
- Department of Infectious Diseases, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Su-Mi Choi
- Department of Infectious Diseases, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Chan-Joong Bae
- Department of Infectious Diseases, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Tae-Hoon Oh
- Department of Infectious Diseases, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Seong Eun Kim
- Department of Infectious Diseases, Chonnam National University Hospital, Gwangju, Republic of Korea; Department of Infectious Diseases, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Uh Jin Kim
- Department of Infectious Diseases, Chonnam National University Hospital, Gwangju, Republic of Korea; Department of Infectious Diseases, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Seung-Ji Kang
- Department of Infectious Diseases, Chonnam National University Hospital, Gwangju, Republic of Korea; Department of Infectious Diseases, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Sook-In Jung
- Department of Infectious Diseases, Chonnam National University Hospital, Gwangju, Republic of Korea; Department of Infectious Diseases, Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Kyung-Hwa Park
- Department of Infectious Diseases, Chonnam National University Hospital, Gwangju, Republic of Korea; Department of Infectious Diseases, Chonnam National University Medical School, Gwangju, Republic of Korea.
| |
Collapse
|
9
|
Activity of Omadacycline in Rat Methicillin-Resistant Staphylococcus aureus Osteomyelitis. Antimicrob Agents Chemother 2021; 66:e0170321. [PMID: 34723626 PMCID: PMC8765317 DOI: 10.1128/aac.01703-21] [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] [Indexed: 11/20/2022] Open
Abstract
Omadacycline, vancomycin, and rifampin, as well as rifampin combination therapies, were evaluated in an experimental rat model of methicillin-resistant Staphylococcus aureus (MRSA) osteomyelitis. All treatment groups had less MRSA recovered than saline-treated animals. The emergence of rifampin resistance was observed in 3 of 16 animals with rifampin monotherapy and none with rifampin combination therapy. After treatment, the median tibial bacterial loads were 6.04, 0.1, 4.81, and 5.24 log10 CFU/g for saline-, rifampin-, vancomycin-, and omadacycline-treated animals, respectively. Omadacycline or vancomycin administered with rifampin yielded no detectable MRSA. Omadacycline administered with rifampin deserves evaluation in humans as a potential treatment for osteomyelitis.
Collapse
|
10
|
In Vitro Activity of Rifampin, Rifabutin, and Rifapentine against Enterococci and Streptococci from Periprosthetic Joint Infection. Microbiol Spectr 2021; 9:e0007121. [PMID: 34259553 PMCID: PMC8552655 DOI: 10.1128/spectrum.00071-21] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
After staphylococci, streptococci and enterococci are the most frequent causes of periprosthetic joint infection (PJI). MICs and minimum biofilm bactericidal concentrations of rifampin, rifabutin, and rifapentine were determined for 67 enterococcal and 59 streptococcal PJI isolates. Eighty-eight isolates had rifampin MICs of ≤1 μg/ml, among which rifabutin and rifapentine MICs were ≤ 8 and ≤4 μg/ml, respectively. There was low rifamycin in vitro antibiofilm activity except for a subset of Streptococcus mitis group isolates. IMPORTANCE Rifampin is an antibiotic with antistaphylococcal biofilm activity used in the management of staphylococcal periprosthetic joint infection with irrigation and debridement with component retention; some patients are unable to receive rifampin due to drug interactions or intolerance. We recently showed rifabutin and rifapentine to have in vitro activity against planktonic and biofilm states of rifampin-susceptible periprosthetic joint infection-associated staphylococci. After staphylococci, streptococci and enterococci combined are the most common causes of periprosthetic joint infection. Here, we investigated the in vitro antibiofilm activity of rifampin, rifabutin, and rifapentine against 126 Streptococcus and Enterococcus periprosthetic joint infection isolates. In contrast to our prior findings with staphylococcal biofilms, there was low antibiofilm activity of rifampin, rifabutin, and rifapentine against PJI-associated streptococci and enterococci, apart from some Streptococcus mitis group isolates.
Collapse
|
11
|
Dalbavancin for the Treatment of Prosthetic Joint Infections: A Narrative Review. Antibiotics (Basel) 2021; 10:antibiotics10060656. [PMID: 34072670 PMCID: PMC8227288 DOI: 10.3390/antibiotics10060656] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 05/24/2021] [Accepted: 05/26/2021] [Indexed: 01/18/2023] Open
Abstract
Dalbavancin (DAL) is a lipoglycopeptide with bactericidal activity against a very wide range of Gram-positive microorganisms. It also has unique pharmacokinetic properties, namely a prolonged half-life (around 181 h), which allows a convenient weekly dosing regimen, and good diffusion in bone tissue. These features have led to off-label use of dalbavancin in the setting of bone and joint infection, including prosthetic joint infections (PJI). In this narrative review, we go over the pharmacokinetic and pharmacodynamic characteristics of DAL, along with published in vitro and in vivo experimental models evaluating its activity against biofilm-embedded bacteria. We also examine published experience of osteoarticular infection with special attention to DAL and PJI.
Collapse
|
12
|
In Vitro Activity of Vancapticin MCC5145 against Methicillin-Resistant Staphylococcus aureus from Periprosthetic Joint Infection. Antimicrob Agents Chemother 2021; 65:AAC.02443-20. [PMID: 33685894 PMCID: PMC8092875 DOI: 10.1128/aac.02443-20] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
MRSA periprosthetic 1 joint infection (PJI) can be challenging to treat due to biofilm formation, alongside sometimes limited vancomycin activity (1-3).….
Collapse
|
13
|
Karau MJ, Schmidt-Malan SM, Albano M, Mandrekar JN, Rivera CG, Osmon DR, Oravec CP, Berry DJ, Abdel MP, Patel R. Novel Use of Rifabutin and Rifapentine to Treat Methicillin-Resistant Staphylococcus aureus in a Rat Model of Foreign Body Osteomyelitis. J Infect Dis 2021; 222:1498-1504. [PMID: 32914837 DOI: 10.1093/infdis/jiaa401] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 07/05/2020] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Owing to patient intolerance or drug interactions, alternative agents to rifampin are needed for management of staphylococcal periprosthetic joint infection. In the current study, we evaluated rifabutin, rifapentine and rifampin, with and without vancomycin, in a rat model of foreign body osteomyelitis. METHODS Proximal tibiae were inoculated with methicillin-resistant Staphylococcus aureus (MRSA) and a Kirschner wire (K-wire) implanted in each. After 4 weeks of infection, rifampin, rifabutin, or rifapentine were administered, alone or with vancomycin. Tibiae and K-wires were cultured, and medians were reported as log10 colony-forming units (CFUs) per gram of bone or log10 CFUs per K-wire, respectively. RESULTS Rifampin, rifabutin or rifapentine administered with vancomycin yielded less MRSA from bones (0.10, 3.02, and 0.10 log10 CFUs/g, respectively) than did no treatment (4.36 log10 CFUs/g) or vancomycin alone (4.64 log10 CFUs/g) (both P ≤ .02). The K-wires of animals receiving no treatment or vancomycin monotherapy recovered medians of 1.76 and 2.91 log10 CFUs/g per K-wire, respectively. In contrast, rifampin, rifabutin and rifapentine administered with vancomycin yielded medians of 0.1 log10 CFUs per K-wire, respectively. Rifampin resistance was detected in a single animal in the rifampin monotherapy group. CONCLUSIONS Rifabutin or rifapentine with vancomycin were as active as rifampin with vancomycin against MRSA in rat foreign body osteomyelitis, suggesting that rifabutin and/or rifapentine may be alternatives to rifampin in the clinical management of staphylococcal periprosthetic joint infections.
Collapse
Affiliation(s)
- Melissa J Karau
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Rochester, Minnesota, USA
| | - Suzannah M Schmidt-Malan
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Rochester, Minnesota, USA
| | - Mariana Albano
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Rochester, Minnesota, USA
| | | | | | - Douglas R Osmon
- Division of Infectious Diseases, Rochester, Minnesota, USA.,Department of Orthopaedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Daniel J Berry
- Department of Orthopaedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Matthew P Abdel
- Department of Orthopaedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Rochester, Minnesota, USA.,Department of Pharmacy, Rochester, Minnesota, USA
| |
Collapse
|
14
|
Senneville E, Dinh A, Ferry T, Beltrand E, Blondiaux N, Robineau O. Tolerance of Prolonged Oral Tedizolid for Prosthetic Joint Infections: Results of a Multicentre Prospective Study. Antibiotics (Basel) 2020; 10:antibiotics10010004. [PMID: 33374817 PMCID: PMC7824147 DOI: 10.3390/antibiotics10010004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 12/18/2020] [Accepted: 12/21/2020] [Indexed: 12/12/2022] Open
Abstract
Objectives: Data on clinical and biological tolerance of tedizolid (TZD) prolonged therapy are lacking. Methods: We conducted a prospective multicentre study including patients with prosthetic joint infections (PJIs) who were treated for at least 6 weeks but not more than 12 weeks. Results: Thirty-three adult patients of mean age 73.3 ± 10.5 years, with PJI including hip (n = 19), knee (n = 13) and shoulder (n = 1) were included. All patients were operated, with retention of the infected implants and one/two stage-replacements in 11 (33.3%) and 17/5 (51.5%/15.2%), respectively. Staphylococci and enterococci were the most prevalent bacteria identified. The mean duration of TZD therapy was 8.0 ± 3.27 weeks (6–12). TZD was associated with another antibiotic in 18 patients (54.5%), including rifampicin in 16 cases (48.5). Six patients (18.2%) had to stop TZD therapy prematurely because of intolerance which was potentially attributable to TZD (n = 2), early failure of PJI treatment (n = 2) or severe anaemia due to bleeding (n = 2). Regarding compliance with TZD therapy, no cases of two or more omissions of medication intake were recorded during the whole TZD treatment duration. Conclusions: These results suggest good compliance and a favourable safety profile of TZD, providing evidence of the potential benefit of the use of this agent for the antibiotic treatment of PJIs.
Collapse
Affiliation(s)
- Eric Senneville
- Infectious Diseases Department, Gustave Dron Hospital, 59200 Tourcoing, France;
- Faculty of Medicine Henri Warembourg, Lille University, 59000 Lille, France
- French National Referent Centre for Complex Bone and Joint Infections, CRIOAC Lille-Tourcoing, 59000 Lille, France; (E.B.); (N.B.)
- Correspondence: ; Tel.: +33-0-32694848; Fax: +33-0-32694496
| | - Aurélien Dinh
- Infectious Diseases Department, Ambroise Paré Hospital, 92100 Boulogne-Billancourt, France;
- French National Referent Centre for Complex Bone and Joint Infections, CRIOAC Paris-Ambroise, 75000 Paré, France
| | - Tristan Ferry
- Infectious Diseases Department, Croix-Rousse Hospital, 69004 Lyon, France;
- French National Referent Centre for Complex Bone and Joint Infections, CRIOAC Lyon, 69004 Lyon, France
| | - Eric Beltrand
- French National Referent Centre for Complex Bone and Joint Infections, CRIOAC Lille-Tourcoing, 59000 Lille, France; (E.B.); (N.B.)
- Orthopaedic Surgery Department, G. Dron Hospital Tourcoing, 59200 Tourcoing, France
| | - Nicolas Blondiaux
- French National Referent Centre for Complex Bone and Joint Infections, CRIOAC Lille-Tourcoing, 59000 Lille, France; (E.B.); (N.B.)
- Microbiology Laboratory, G. Dron Hospital Tourcoing, 59200 Tourcoing, France
| | - Olivier Robineau
- Infectious Diseases Department, Gustave Dron Hospital, 59200 Tourcoing, France;
- Faculty of Medicine Henri Warembourg, Lille University, 59000 Lille, France
- French National Referent Centre for Complex Bone and Joint Infections, CRIOAC Lille-Tourcoing, 59000 Lille, France; (E.B.); (N.B.)
| |
Collapse
|
15
|
Fisher CR, Schmidt-Malan SM, Ma Z, Yuan Y, He S, Patel R. In vitro activity of TNP-2092 against periprosthetic joint infection-associated staphylococci. Diagn Microbiol Infect Dis 2020; 97:115040. [PMID: 32354459 DOI: 10.1016/j.diagmicrobio.2020.115040] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 02/07/2020] [Accepted: 03/16/2020] [Indexed: 11/26/2022]
Abstract
Staphylococci are the most common causes of periprosthetic joint infection (PJI). TNP-2092 is an investigational hybrid drug composed of rifamycin and quinolizinone pharmacophores conjugated via a covalent linker. We determined minimum inhibitory concentration (MIC), minimum bactericidal concentration (MBC), and minimum biofilm bactericidal concentration (MBBC) values of TNP-2092 against 80 PJI-associated Staphylococcus aureus and Staphylococcus epidermidis isolates compared to ciprofloxacin and rifampin alone and in combination, alongside daptomycin and vancomycin. TNP-2092 exhibited the following activity against S. aureus: MIC50/MIC90, ≤0.0075/0.015 μg/mL; MBC50/MBC90, 0.5/4 μg/mL; and MBBC50/MBBC90, 0.5/2 μg/mL, and the following activity against S. epidermidis: MIC50/MIC90, ≤0.0075/0.015 μg/mL; MBC50/MBC90, 0.015/0.125 μg/mL; and MBBC50/MBBC90, 0.06/0.25 μg/mL. TNP-2092 MIC, MBC, and MBBC values were >8 μg/mL for 1 isolate, while MIC values were ≤0.25 μg/mL and MBC and MBBC values were ≤4 μg/mL for all other isolates. Results of this study show that TNP-2092 has promising in vitro activity against PJI-associated staphylococci.
Collapse
Affiliation(s)
- Cody R Fisher
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Suzannah M Schmidt-Malan
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Zhenkun Ma
- TenNor Therapeutics Limited, Suzhou, China
| | - Ying Yuan
- TenNor Therapeutics Limited, Suzhou, China
| | - Shijie He
- TenNor Therapeutics Limited, Suzhou, China
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN; Division of Infectious Diseases, Department of Medicine, Mayo Clinic, Rochester, MN.
| |
Collapse
|
16
|
Hasannejad-Bibalan M, Mojtahedi A, Biglari H, Halaji M, Sedigh Ebrahim-Saraie H. Antibacterial Activity of Tedizolid, a Novel Oxazolidinone Against Methicillin-Resistant Staphylococcus aureus: A Systematic Review and Meta-Analysis. Microb Drug Resist 2019; 25:1330-1337. [DOI: 10.1089/mdr.2018.0457] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
| | - Ali Mojtahedi
- Department of Microbiology, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Haniyeh Biglari
- Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Mehrdad Halaji
- Department of Microbiology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | |
Collapse
|
17
|
In Vitro Activity of Rifampin, Rifabutin, Rifapentine, and Rifaximin against Planktonic and Biofilm States of Staphylococci Isolated from Periprosthetic Joint Infection. Antimicrob Agents Chemother 2019; 63:AAC.00959-19. [PMID: 31451499 DOI: 10.1128/aac.00959-19] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 08/17/2019] [Indexed: 11/20/2022] Open
Abstract
The in vitro activities of rifampin, rifabutin, rifapentine, and rifaximin were tested against 200 periprosthetic joint infection (PJI)-associated staphylococci. Seven rifampin-resistant isolates had MICs of ≥4 μg/ml. Three isolates had rifampin MICs of 0.25 to 1 μg/ml and harbored an Asp471Gly RpoB variant, suggesting that the CLSI rifampin-susceptible staphylococcal breakpoint of ≤1 μg/ml may be too high. The remaining isolates had rifampin MICs of ≤0.016 μg/ml, and the rifampin, rifabutin, rifapentine, and rifaximin minimum biofilm bactericidal concentrations (MBBC) for ≥50% of isolates were 8, 1, 2, and 4 μg/ml (for S. aureus) and 2, 0.06, 0.25, and 0.5 μg/ml (for S. epidermidis), respectively, for rifampin-susceptible isolates. Nonrifampin rifamycins have promising staphylococcal activity, including antibiofilm activity.
Collapse
|
18
|
Exebacase in Addition to Daptomycin Is More Active than Daptomycin or Exebacase Alone in Methicillin-Resistant Staphylococcus aureus Osteomyelitis in Rats. Antimicrob Agents Chemother 2019; 63:AAC.01235-19. [PMID: 31358593 DOI: 10.1128/aac.01235-19] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 07/24/2019] [Indexed: 12/14/2022] Open
Abstract
Bacteriophage-derived lysins are being developed as anti-infective agents. In an acute osteomyelitis methicillin-resistant Staphylococcus aureus (MRSA) model, rats receiving no treatment or treatment with daptomycin, exebacase (CF-301), or daptomycin plus exebacase had means of 5.13, 4.09, 4.65, and 3.57 log10 CFU/gram of bone, respectively. All treated animals had fewer bacteria than did untreated animals (P ≤ 0.0001), with daptomycin plus exebacase being more active than daptomycin (P = 0.0042) or exebacase (P < 0.001) alone.
Collapse
|
19
|
Abstract
PURPOSE OF REVIEW Tedizolid is a second-generation oxazolidinone with activity against Gram-positive bacteria, including MRSA isolates resistant to linezolid. Pivotal clinical trials showed that tedizolid at 200 mg once-daily for 6 days is not inferior to linezolid 600 mg twice daily for 10 days in patients with SSTI. The comparison of adverse events is favorable to tedizolid under the circumstances of the clinical trials. This is a review of recent literature on tedizolid, its use in special populations and potential adverse effects. RECENT FINDINGS Findings suggest that tedizolid can be used in SSTI in adolescents, those older than 65 years, obese individuals and patients with diabetic foot infections. Forthcoming research to determine the future uses of this drug in other clinical syndromes requires demonstration of tolerance whenever tedizolid is administered for longer than 6 days.We also speculate on missing data and potential future indications of tedizolid in the highly competitive field of the treatment of severe Gram-positive infections other than SSTI. SUMMARY Tedizolid is a second-generation oxazolidinone, very convenient for treatment of SSTI, in search for other indications including nosocomial pneumonia and bone and joint infections. VIDEO ABSTRACT.
Collapse
|
20
|
Azrad M, Baum M, Rokney A, Levi Y, Peretz A. In vitro activity of Tedizolid and Dalbavancin against MRSA strains is dependent on infection source. Int J Infect Dis 2019; 78:107-112. [DOI: 10.1016/j.ijid.2018.11.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 11/07/2018] [Accepted: 11/15/2018] [Indexed: 01/18/2023] Open
|
21
|
Bolia IK, Tsiodras S, Chloros GD, Kaspiris A, Sarlikiotis T, Savvidou OD, Papagelopoulos PJ. A Review of Novel Antibiotic Regimens for the Treatment of Orthopedic Infections. Orthopedics 2018; 41:323-328. [PMID: 30452066 DOI: 10.3928/01477447-20181024-02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
As a result of the increasing numbers of joint replacement surgeries and other implant-associated procedures performed, the incidences of periprosthetic joint infections and osteomyelitis are on the rise. Antibiotic resistance to gram-positive species, which are mostly isolated from such infections, is a significant obstacle in clinical practice. Promising clinical outcomes have been reported with the use of novel antibiotics for patients with periprosthetic joint infections and osteomyelitis. Further research is necessary for the establishment of these novel antibiotic therapies in routine clinical practice. [Orthopedics. 2018; 41(6):323-328.].
Collapse
|
22
|
Yan Q, Karau MJ, Patel R. In vitro activity of oritavancin against biofilms of staphylococci isolated from prosthetic joint infection. Diagn Microbiol Infect Dis 2018; 92:155-157. [PMID: 29885758 DOI: 10.1016/j.diagmicrobio.2018.05.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 05/07/2018] [Accepted: 05/14/2018] [Indexed: 11/27/2022]
Abstract
We tested the in vitro activity of oritavancin against 185 staphylococci associated with prosthetic joint infection, including 37 methicillin-resistant S. aureus, 67 methicillin-susceptible S. aureus, 59 methicillin-resistant S. epidermidis (MRSE), and 22 methicillin-susceptible S. epidermidis (MSSE) isolates. The oritavancin MIC50 for S. aureus and MSSE was 0.03 μg/mL, and for MRSE, it was 0.06 μg/mL; MIC90 for S. aureus and S. epidermidis was 0.12 μg/mL for both the methicillin-resistant and -susceptible subgroups. The oritavancin MBBC50 for S. aureus and S. epidermidis was 2 μg/mL for both the methicillin-resistant and -susceptible subgroups; the MBBC90 for S. aureus and MSSE was 4 μg/mL, and for MRSE, it was 8 μg/mL.
Collapse
Affiliation(s)
- Qun Yan
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN; Department of Clinical Laboratory, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Melissa J Karau
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN; Division of Infectious Diseases, Department of Medicine, Mayo Clinic, Rochester, MN.
| |
Collapse
|
23
|
Yan Q, Karau MJ, Patel R. In vitro activity of oritavancin against planktonic and biofilm states of vancomycin-susceptible and vancomycin-resistant enterococci. Diagn Microbiol Infect Dis 2018; 91:348-350. [PMID: 29678300 DOI: 10.1016/j.diagmicrobio.2018.03.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/08/2018] [Accepted: 03/11/2018] [Indexed: 11/20/2022]
Abstract
We tested the in vitro activity of oritavancin against 60 vancomycin-susceptible enterococci (VSE) and 27 vancomycin-resistant enterococci (VRE). The oritavancin MIC ranged from ≤0.002 to 0.5μg/mL; the minimum biofilm bactericidal concentration ranged from ≤0.002 to 2μg/mL. Oritavancin has promising in vitro activity against VSE and VRE in both planktonic and biofilm states.
Collapse
Affiliation(s)
- Qun Yan
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN; Department of Clinical Laboratory, Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Melissa J Karau
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN; Division of Infectious Diseases, Department of Medicine, Mayo Clinic, Rochester, MN.
| |
Collapse
|
24
|
Karlowsky JA, Hackel MA, Bouchillon SK, Alder J, Sahm DF. In Vitro activities of Tedizolid and comparator antimicrobial agents against clinical isolates of Staphylococcus aureus collected in 12 countries from 2014 to 2016. Diagn Microbiol Infect Dis 2017; 89:151-157. [DOI: 10.1016/j.diagmicrobio.2017.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Revised: 06/26/2017] [Accepted: 07/03/2017] [Indexed: 11/29/2022]
|
25
|
Neudorfer K, Schmidt-Malan SM, Patel R. Dalbavancin is active in vitro against biofilms formed by dalbavancin-susceptible enterococci. Diagn Microbiol Infect Dis 2017; 90:58-63. [PMID: 29195766 DOI: 10.1016/j.diagmicrobio.2017.09.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/18/2017] [Accepted: 09/23/2017] [Indexed: 11/16/2022]
Abstract
We tested the in vitro activity of dalbavancin, vancomycin and daptomycin against 83 enterococcal isolates in planktonic and biofilm states. The MIC90 for vancomycin-susceptible Enterococcus faecalis was 0.125 and 4μg/mL for dalbavancin and daptomycin, respectively. For vancomycin-resistant Enterococcus faecium, the MIC90 was >16 and 2μg/mL for dalbavancin and daptomycin, respectively. Dalbavancin minimum biofilm inhibitory concentrations (MBICs) for vancomycin-susceptible and -resistant isolates were ≤0.25 and >16μg/mL, respectively. The daptomycin MBIC90 for all isolates was 4μg/mL. For E. faecalis and E. faecium, dalbavancin minimum biofilm bactericidal concentrations (MBBCs) for vancomycin-susceptible and -resistant isolates were ≤4 and >16μg/mL, respectively, whereas vancomycin MBBCs were >128μg/mL for all isolates, and daptomycin MBBC90 values for both species were 128μg/mL. In summary, dalbavancin exhibited in vitro activity against all tested isolates of vancomycin-susceptible, but not against vancomycin-resistant enterococci; activity was observed in both the planktonic and biofilm states.
Collapse
Affiliation(s)
- Katharina Neudorfer
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905
| | - Suzannah M Schmidt-Malan
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905
| | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN 55905; Division of Infectious Diseases, Department of Medicine, Mayo Clinic, Rochester, MN 55905.
| |
Collapse
|
26
|
Ract P, Piau-Couapel C, Compain F, Auzou M, Michon J, Cattoir V. In vitro activity of tedizolid and comparator agents against Gram-positive pathogens responsible for bone and joint infections. J Med Microbiol 2017; 66:1374-1378. [PMID: 28920854 DOI: 10.1099/jmm.0.000595] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Tedizolid, a second-generation oxazolidinone that displays a potent activity against Gram-positive pathogens, could be an interesting option for the treatment of bone and joint infections (BJIs). The aim of the study was to determine minimal inhibitory concentration (MIC) of tedizolid against a collection of 359 clinical isolates involved in clinically-documented BJIs and to compare them to those of comparator agents used in Gram-positive infections. Of the 104 Staphylococcusaureus and 102 coagulase-negative staphylococci (CoNS) isolates, 99 and 92 % were categorized as susceptible to tedizolid, respectively (MIC25=0.12/0.25 µg ml-1 and MIC90=0.25/0.5 µg ml-1), regardless of their methicillin resistance. MIC50 and MIC90 for the 51 enterococci, the 50 Corynebacterium spp. and the 52 Propionibacterium spp. were either equal or inferior to 0.5 µg ml-1. Altogether, tedizolid possessed a potent in vitro activity against most of the BJI Gram-positive pathogens with 95 % of them exhibiting a MIC ≤0.5 µg ml-1.
Collapse
Affiliation(s)
- Pauline Ract
- CHU de Caen, Service de Microbiologie, Caen, France.,Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France
| | | | - Fabrice Compain
- Hôpital Européen Georges Pompidou, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Michel Auzou
- CHU de Caen, Service de Microbiologie, Caen, France
| | - Jocelyn Michon
- CHU de Caen, Service de Maladies Infectieuses et Tropicales, Caen, France
| | - Vincent Cattoir
- CNR de la Résistance aux Antibiotiques (laboratoire associé "Entérocoques"), Rennes, France.,CHU de Rennes, Service de Bactériologie-Hygiène Hospitalière, Rennes, France
| |
Collapse
|
27
|
In vitro activity of tedizolid and linezolid against Staphylococcus epidermidis isolated from prosthetic joint infections. Eur J Clin Microbiol Infect Dis 2017; 36:1549-1552. [PMID: 28326447 PMCID: PMC5554274 DOI: 10.1007/s10096-017-2966-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 03/09/2017] [Indexed: 12/31/2022]
Abstract
Prosthetic joint infections (PJIs) are rare but long-lasting and are serious complications without any spontaneous resolution, requiring additional surgery and long-term treatment with antibiotics. Staphylococci are the most important aetiological agents of PJIs, and among the coagulase-negative staphylococci Staphylococcus epidermidis is the most common. However, S. epidermidis often displays multidrug resistance (MDR), demanding additional treatment options. The objective was to examine the effectiveness of tedizolid and linezolid against S. epidermidis isolated from PJIs. The standard antibiotic susceptibility pattern of S. epidermidis (n = 183) obtained from PJIs was determined by disc diffusion test, and MIC was determined by Etest for tedizolid, linezolid, and vancomycin. Tedizolid displayed MIC values ranging from 0.094 to 0.5 mg/L (MIC50: 0.19 mg/L, MIC90: 0.38 mg/L), linezolid MIC values ranging from 0.25 to 2 mg/L (MIC50: 0.75 mg/L, MIC90: 1 mg/L), and vancomycin MIC values ranging from 0.5 to 3 mg/L (MIC50 and MIC90 both 2 mg/L). According to the disc diffusion test, 153/183 (84%) isolates were resistant to ≥3 antibiotic groups, indicating MDR. In conclusion, S. epidermidis isolates from PJIs were fully susceptible, and the MIC50 and MIC90 values for tedizolid were two- to four-fold dilution steps lower compared with linezolid. Tedizolid is not approved, and there are no reports of long-term treatment, but it may display better tolerability and fewer adverse effects than linezolid; it thus could be a possible treatment option for PJIs, alone or in combination with rifampicin.
Collapse
|
28
|
Si S, Durkin MJ, Mercier MM, Yarbrough ML, Liang SY. Successful Treatment of Prosthetic Joint Infection due to Vancomycin-resistant Enterococci with Tedizolid. INFECTIOUS DISEASES IN CLINICAL PRACTICE 2017; 25:105-107. [PMID: 28428726 PMCID: PMC5393450 DOI: 10.1097/ipc.0000000000000469] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Few antibiotic options exist for the management of infections due to vancomycin-resistant enterococci (VRE). We describe a case involving the safe and successful use of tedizolid, a new oxazolidinone, to treat VRE prosthetic joint infection.
Collapse
Affiliation(s)
- Sheng Si
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Michael J. Durkin
- Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO
| | - Maureen M. Mercier
- Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO
| | - Melanie L. Yarbrough
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO
| | - Stephen Y. Liang
- Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO
- Division of Emergency Medicine, Washington University School of Medicine, St. Louis, MO
| |
Collapse
|
29
|
Activity of Tedizolid in Methicillin-Resistant Staphylococcus epidermidis Experimental Foreign Body-Associated Osteomyelitis. Antimicrob Agents Chemother 2017; 61:AAC.01644-16. [PMID: 27855069 DOI: 10.1128/aac.01644-16] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Accepted: 10/22/2016] [Indexed: 12/19/2022] Open
Abstract
We developed a rat model of methicillin-resistant Staphylococcus epidermidis (MRSE) foreign body-associated osteomyelitis and used it to compare tedizolid alone and in combination with rifampin against rifampin alone, vancomycin plus rifampin, and vancomycin alone. A clinical strain of MRSE was inoculated into the proximal tibia, and a stainless steel wire with a precolonized MRSE biofilm was implanted. Following a 1-week infection period, 92 rats received either no treatment (n = 17) or 14 days of intraperitoneal tedizolid (n = 15), tedizolid plus rifampin (n = 15), rifampin (n = 15), vancomycin plus rifampin (n = 15), or vancomycin (n = 15). Quantitative bone and wire cultures were performed after treatment completion and also 1 week after infection in a separate group of five rats. The median quantity of staphylococci in bone after the 1-week infection period was 4.89 log10 CFU/g bone (interquartile range, 3.83 to 5.33 log10 CFU/g bone); staphylococci were recovered from all associated wires. A median quantity of staphylococci of 3.70 log10 CFU/g bone was detected in bones of untreated control rats after 3 weeks. Quantities of staphylococci in bones of all treatment groups except the group receiving vancomycin alone (2.78 log10 CFU/g) were significantly lower than those for untreated controls, with no staphylococci being detected in the groups receiving rifampin monotherapy, tedizolid-plus-rifampin combination therapy, and vancomycin-plus-rifampin combination therapy. Quantities of staphylococci on wires from all treatment groups that included rifampin were significantly lower than those for untreated controls. No resistance to rifampin, tedizolid, or vancomycin was detected. Tedizolid combined with rifampin was active in a rat model of MRSE foreign body-associated osteomyelitis.
Collapse
|
30
|
Ferrández O, Urbina O, Grau S. Critical role of tedizolid in the treatment of acute bacterial skin and skin structure infections. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 11:65-82. [PMID: 28053508 PMCID: PMC5191846 DOI: 10.2147/dddt.s84667] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tedizolid phosphate has high activity against the Gram-positive microorganisms mainly involved in acute bacterial skin and skin structure infections, such as strains of Staphylococcus aureus (including methicillin-resistant S. aureus strains and methicillin-sensitive S. aureus strains), Streptococcus pyogenes, Streptococcus agalactiae, the Streptococcus anginosus group, and Enterococcus faecalis, including those with some mechanism of resistance limiting the use of linezolid. The area under the curve for time 0-24 hours/minimum inhibitory concentration (MIC) pharmacodynamic ratio has shown the best correlation with the efficacy of tedizolid, versus the time above MIC ratio and the maximum drug concentration/minimum inhibitory concentration ratio. Administration of this antibiotic for 6 days has shown its noninferiority versus administration of linezolid for 10 days in patients with skin and skin structure infections enrolled in two Phase III studies (ESTABLISH-1 and ESTABLISH-2). Tedizolid's more favorable safety profile and dosage regimen, which allow once-daily administration, versus linezolid, position it as a good therapeutic alternative. However, whether or not the greater economic cost associated with this antibiotic is offset by its shorter treatment duration and possibility of oral administration in routine clinical practice has yet to be clarified.
Collapse
Affiliation(s)
- Olivia Ferrández
- Hospital Pharmacy, Hospital Universitari del Mar, Barcelona, Spain; Nursing Department, Universitat Pompeu Fabra, Barcelona, Spain
| | - Olatz Urbina
- Hospital Pharmacy, Hospital Universitari del Mar, Barcelona, Spain
| | - Santiago Grau
- Hospital Pharmacy, Hospital Universitari del Mar, Barcelona, Spain; Medicine Department, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
31
|
Activity of Tedizolid in Methicillin-Resistant Staphylococcus aureus Experimental Foreign Body-Associated Osteomyelitis. Antimicrob Agents Chemother 2016; 60:6568-6572. [PMID: 27550347 DOI: 10.1128/aac.01248-16] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 08/12/2016] [Indexed: 01/24/2023] Open
Abstract
We compared tedizolid alone and tedizolid with rifampin to rifampin and vancomycin plus rifampin in a rat model of methicillin-resistant Staphylococcus aureus (MRSA) foreign body-associated osteomyelitis. The study strain was a prosthetic joint infection-associated isolate. Steady-state pharmacokinetics for intraperitoneal administration of tedizolid, vancomycin, and rifampin were determined in uninfected rats. MRSA was inoculated into the proximal tibia, and a wire was implanted. Four weeks later, the rats were treated intraperitoneally for 21 days with tedizolid (n = 14), tedizolid plus rifampin (n = 11), rifampin (n = 16), or vancomycin plus rifampin (n = 13). Seventeen rats received no treatment. After treatment, quantitative bone cultures were performed. Blood was obtained for determination of drug trough concentrations in the tedizolid and tedizolid plus rifampin groups. The mean peak plasma concentration and mean area under the concentration-time curve from time zero to 24 h for tedizolid were 12 μg/ml and 60 μg · h/ml, respectively. The bacterial loads in all treatment groups were significantly lower than those in the control group; those in the tedizolid- plus rifampin-treated animals were not significantly different from those in the vancomycin- plus rifampin-treated animals. The range of mean plasma trough concentrations in the tedizolid group was 0.44 to 0.73 μg/ml. Although neither tedizolid nor vancomycin resistance was detected in isolates recovered from bones, rifampin resistance was detected in 10 animals (63%) in the rifampin group, 8 animals (73%) in the tedizolid plus rifampin group, and a single animal (8%) in the vancomycin plus rifampin group. Tedizolid alone or tedizolid combined with rifampin was active in a rat model of MRSA foreign body-associated osteomyelitis. The emergence of rifampin resistance was noted in animals receiving tedizolid plus rifampin.
Collapse
|
32
|
Comparative Efficacies of Tedizolid Phosphate, Linezolid, and Vancomycin in a Murine Model of Subcutaneous Catheter-Related Biofilm Infection Due to Methicillin-Susceptible and -Resistant Staphylococcus aureus. Antimicrob Agents Chemother 2016; 60:5092-6. [PMID: 27297485 DOI: 10.1128/aac.00880-16] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 06/07/2016] [Indexed: 12/17/2022] Open
Abstract
Tedizolid, a novel oxazolidinone, exhibits bacteriostatic activity through inhibition of protein synthesis. The efficacies of tedizolid, linezolid, and vancomycin were compared in a murine catheter-related biofilm infection caused by methicillin-susceptible and -resistant Staphylococcus aureus (MSSA and MRSA, respectively) strains engineered for bioluminescence. We observed significantly improved efficacy in terms of decreased S. aureus densities and bioluminescent signals in the tedizolid-treated group versus the linezolid- and vancomycin-treated groups in the model of infection caused by the MSSA and MRSA strains.
Collapse
|
33
|
Fernández J, Greenwood-Quaintance KE, Patel R. In vitro activity of dalbavancin against biofilms of staphylococci isolated from prosthetic joint infections. Diagn Microbiol Infect Dis 2016; 85:449-51. [PMID: 27241369 DOI: 10.1016/j.diagmicrobio.2016.05.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 05/09/2016] [Accepted: 05/11/2016] [Indexed: 10/21/2022]
Abstract
The in vitro activity of dalbavancin was tested against biofilms of 171 staphylococci associated with prosthetic joint infection. Dalbavancin minimum biofilm bactericidal concentration (MBBC) values were: MBBC50 for Staphylococcus aureus and Staphylococcus epidermidis, 1μg/mL; MBBC90 for S. aureus, 2μg/mL; MBBC90 for S. epidermidis, 4μg/mL.
Collapse
Affiliation(s)
- Javier Fernández
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA; Department of Functional Biology, Section of Microbiology, University of Oviedo, Oviedo, Spain; Service of Microbiology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | - Robin Patel
- Division of Clinical Microbiology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA; Division of Infectious Diseases, Department of Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|