51
|
Saravolatz LD, Pawlak J. In vitro activity of fosfomycin alone and in combination against Staphylococcus aureus with reduced susceptibility or resistance to methicillin, vancomycin, daptomycin or linezolid. J Antimicrob Chemother 2022; 78:238-241. [PMID: 36374572 DOI: 10.1093/jac/dkac380] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVES To evaluate the activity of fosfomycin against a group of MRSA strains, including isolates with reduced susceptibility or resistance to vancomycin, daptomycin, linezolid and ceftaroline and to determine the effect of combining various combinations of antimicrobial agents used in the therapy of serious Gram-positive infections. METHODS Broth microdilution testing was used to determine the MICs of fosfomycin, vancomycin, daptomycin, linezolid, ceftaroline and cefazolin. Isolates were selected for further evaluations to determine in vitro synergy between fosfomycin and select antimicrobial agents using chequerboard broth microdilution testing. Fosfomycin was tested in combination with vancomycin, linezolid, daptomycin, ceftaroline and cefazolin. RESULTS Fosfomycin maintained activity against 100% of strains of vancomycin-resistant Staphylococcus aureus (VRSA) and linezolid-resistant S. aureus (LRSA), 86% of VISA and 95% of daptomycin-resistant S. aureus (DRSA) strains. The combination of fosfomycin with ceftaroline consistently demonstrated synergy among all 18 isolates against the strains tested. The next most potent combination regimen was linezolid with fosfomycin, which demonstrated synergy in 16 of the 18 strains. Daptomycin demonstrated synergy in only 7 of the 18 strains tested when combined with fosfomycin. Cefazolin demonstrated synergy in 6 of 6 strains and vancomycin demonstrated no interaction in 6 of 6 strains tested. CONCLUSIONS Fosfomycin demonstrated excellent activity against MRSA as well as isolates with resistance or reduced activity to other anti-MRSA drugs including vancomycin, daptomycin and linezolid. When combined with linezolid or daptomycin, fosfomycin demonstrated synergy for all or most strains tested. Thus, these combinations may have potential clinical utility when treating patients with serious infections caused by MRSA.
Collapse
Affiliation(s)
- Louis D Saravolatz
- Ascension St. John Hospital, Grosse Pointe Woods, MI, USA.,Thomas Mackey Center for Infectious Disease Research, Grosse Pointe Woods, MI, USA.,Central Michigan University College of Medicine, Mount Pleasant, MI, USA
| | - Joan Pawlak
- Ascension St. John Hospital, Grosse Pointe Woods, MI, USA.,Thomas Mackey Center for Infectious Disease Research, Grosse Pointe Woods, MI, USA.,Central Michigan University College of Medicine, Mount Pleasant, MI, USA
| |
Collapse
|
52
|
Antonello RM, Canetti D, Riccardi N. Daptomycin synergistic properties from in vitro and in vivo studies: a systematic review. J Antimicrob Chemother 2022; 78:52-77. [PMID: 36227704 DOI: 10.1093/jac/dkac346] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 09/21/2022] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION Daptomycin is a bactericidal lipopeptide antibiotic approved for the treatment of systemic infections (i.e. skin and soft tissue infections, bloodstream infections, infective endocarditis) caused by Gram-positive cocci. It is often prescribed in association with a partner drug to increase its bactericidal effect and to prevent the emergence of resistant strains during treatment; however, its synergistic properties are still under evaluation. METHODS We performed a systematic review to offer clinicians an updated overview of daptomycin synergistic properties from in vitro and in vivo studies. Moreover, we reported all in vitro and in vivo data evaluating daptomycin in combination with other antibiotic agents, subdivided by antibiotic classes, and a summary graph presenting the most favourable combinations at a glance. RESULTS A total of 92 studies and 1087 isolates (723 Staphylococcus aureus, 68 Staphylococcus epidermidis, 179 Enterococcus faecium, 105 Enterococcus faecalis, 12 Enterococcus durans) were included. Synergism accounted for 30.9% of total interactions, while indifferent effect was the most frequently observed interaction (41.9%). Antagonistic effect accounted for 0.7% of total interactions. The highest synergistic rates against S. aureus were observed with daptomycin in combination with fosfomycin (55.6%). For S. epidermidis and Enterococcus spp., the most effective combinations were daptomycin plus ceftobiprole (50%) and daptomycin plus fosfomycin (63.6%) or rifampicin (62.8%), respectively. FUTURE PERSPECTIVES We believe this systematic review could be useful for the future updates of guidelines on systemic infections where daptomycin plays a key role.
Collapse
Affiliation(s)
- Roberta Maria Antonello
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50121, Italy
| | - Diana Canetti
- Department of Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Niccolò Riccardi
- Department of Clinical and Experimental Medicine, Infectious Diseases Unit, Azienda Ospedaliera Universitaria Pisana, University of Pisa, Pisa 56124, Italy
| |
Collapse
|
53
|
Chen Y, Ji S, Sun L, Wang H, Zhu F, Chen M, Zhuang H, Wang Z, Jiang S, Yu Y, Chen Y. The novel fosfomycin resistance gene fosY is present on a genomic island in CC1 methicillin-resistant Staphylococcus aureus. Emerg Microbes Infect 2022; 11:1166-1173. [PMID: 35332834 PMCID: PMC9037201 DOI: 10.1080/22221751.2022.2058421] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Fosfomycin has gained attention as a combination therapy for methicillin-resistant Staphylococcus aureus infections. Hence, the detection of novel fosfomycin-resistance mechanisms in S. aureus is important. Here, the minimal inhibitory concentrations (MICs) of fosfomycin in CC1 methicillin-resistant S. aureus were determined. The pangenome analysis and comparative genomics were used to analyse CC1 MRSA. The gene function was confirmed by cloning the gene into pTXΔ. A phylogenetic tree was constructed to determine the clustering of the CC1 strains of S. aureus. We identified a novel gene, designated fosY, that confers fosfomycin resistance in S. aureus. The FosY protein is a putative bacillithiol transferase enzyme sharing 65.9-77.5% amino acid identity with FosB and FosD, respectively. The function of fosY in decreasing fosfomycin susceptibility was confirmed by cloning it into pTXΔ. The pTX-fosY transformant exhibited a 16-fold increase in fosfomycin MIC. The bioinformatic analysis showed that fosY is in a novel genomic island designated RIfosY (for "resistance island carrying fosY") that originated from other species. The global phylogenetic tree of ST1 MRSA displayed this fosY-positive ST1 clone, originating from different regions, in the same clade. The novel resistance gene in the fos family, fosY, and a genomic island, RIfosY, can promote cross-species gene transfer and confer resistance to CC1 MRSA causing the failure of clinical treatment. This emphasises the importance of genetic surveillance of resistance genes among MRSA isolates.
Collapse
Affiliation(s)
- Yiyi Chen
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, People's Republic of China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Shujuan Ji
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, People's Republic of China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Lu Sun
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, People's Republic of China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Haiping Wang
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, People's Republic of China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Feiteng Zhu
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, People's Republic of China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Mengzhen Chen
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, People's Republic of China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Hemu Zhuang
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, People's Republic of China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Zhengan Wang
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, People's Republic of China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Shengnan Jiang
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, People's Republic of China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yunsong Yu
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, People's Republic of China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Yan Chen
- Department of Infectious Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China.,Key Laboratory of Microbial Technology and Bioinformatics of Zhejiang Province, Hangzhou, People's Republic of China.,Regional Medical Center for National Institute of Respiratory Diseases, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| |
Collapse
|
54
|
Ryder JH, Tong SYC, Gallagher JC, McDonald EG, Thevarajan I, Lee TC, Cortés-Penfield NW. Deconstructing the Dogma: Systematic Literature Review and Meta-analysis of Adjunctive Gentamicin and Rifampin in Staphylococcal Prosthetic Valve Endocarditis. Open Forum Infect Dis 2022; 9:ofac583. [PMID: 36408468 PMCID: PMC9669455 DOI: 10.1093/ofid/ofac583] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 10/28/2022] [Indexed: 08/01/2023] Open
Abstract
BACKGROUND Based primarily on in vitro and animal models, with little data directly addressing patient outcomes, current guidelines recommend treating staphylococcal prosthetic valve endocarditis (PVE) with antibiotic combinations including gentamicin and rifampin. Here, we synthesize the clinical data on adjunctive rifampin and gentamicin in staphylococcal PVE. METHODS We conducted a systematic review and meta-analysis of PubMed- and Cochrane-indexed studies reporting outcomes of staphylococcal PVE treated with adjunctive rifampin, gentamicin, both agents, or neither (ie, glycopeptide or β-lactam monotherapy). We recorded outcomes including mortality, relapsed infection, length of stay, nephrotoxicity, hepatotoxicity, and important drug-drug interactions (DDIs). RESULTS Four relevant studies were identified. Two studies (n = 117) suggested that adding gentamicin to rifampin-containing regimens did not reduce clinical failure (odds ratio [OR], 0.98 [95% confidence interval {CI}, .39-2.46]), and 2 studies (n = 201) suggested that adding rifampin to gentamicin-containing regimens did not reduce clinical failure (OR, 1.29 [95% CI, .71-2.33]). Neither gentamicin nor rifampin was associated with reduced infection relapse; 1 study found that rifampin treatment was associated with longer hospitalizations (mean, 31.3 vs 42.3 days; P < .001). Comparative safety outcomes were rarely reported, but 1 study found rifampin to be associated with hepatoxicity, nephrotoxicity, and DDIs, leading to treatment discontinuation in 31% of patients. CONCLUSIONS The existing clinical data do not suggest a benefit of either adjunctive gentamicin or rifampin in staphylococcal PVE. Given that other studies also suggest these agents add nephrotoxicity, hepatoxicity, and risk of DDIs without benefit in staphylococcal endovascular infections, we suggest that recommendations for gentamicin and rifampin in PVE be downgraded and primarily be used within the context of clinical trials.
Collapse
Affiliation(s)
- Jonathan H Ryder
- Correspondence: Jonathan H. Ryder, MD, University of Nebraska Medical Center, 985400 Nebraska Medical Center, Omaha, NE 68198 ()
| | - Steven Y C Tong
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Infectious Diseases, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Jason C Gallagher
- Department of Pharmacy Practice, Temple University, Philadelphia, Pennsylvania, USA
| | - Emily G McDonald
- Clinical Practice Assessment Unit, Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Irani Thevarajan
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Infectious Diseases, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | | | | |
Collapse
|
55
|
Holland TL, Bayer AS, Fowler VG. Persistent Methicilin-Resistant Staphylococcus aureus Bacteremia: Resetting the Clock for Optimal Management. Clin Infect Dis 2022; 75:1668-1674. [PMID: 35535790 PMCID: PMC9617577 DOI: 10.1093/cid/ciac364] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Indexed: 01/25/2023] Open
Abstract
A positive follow-up blood culture for methicillin-resistant Staphylococcus aureus (MRSA) while on seemingly appropriate therapy is a common and ominous development. However, the definition and management of persistent MRSA bacteremia is unstandardized. In this Opinion Paper, we identify the presence of bacteremia for > 1 calendar day as a "worry point" that should trigger an intensive diagnostic evaluation to identify metastatic infection sites. Next, we define the duration of MRSA bacteremia that likely constitutes antibiotic failure and outline a potential management algorithm for such patients. Finally, we propose pragmatic clinical trial designs to test treatment strategies for persistent MRSA bacteremia.
Collapse
Affiliation(s)
- Thomas L Holland
- Department of Medicine, Duke University, Durham, North Carolina, USA
- Duke Clinical Research Institute, Durham, North Carolina, USA
| | - Arnold S Bayer
- The Lundquist Institute for Biomedical Innovation at Harbor-UCLA, Torrance, California, USA
- The Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Vance G Fowler
- Department of Medicine, Duke University, Durham, North Carolina, USA
- Duke Clinical Research Institute, Durham, North Carolina, USA
| |
Collapse
|
56
|
The combination of daptomycin with β-lactam antibiotics is more effective than daptomycin alone for vancomycin-resistant Enterococcus faecium bloodstream infection. J Infect Public Health 2022; 15:1396-1402. [DOI: 10.1016/j.jiph.2022.10.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 10/19/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022] Open
|
57
|
Abstract
PURPOSE OF REVIEW To review recently published evidence relevant to Staphylococcus aureus bacteremia (SAB). RECENT FINDINGS Staphylococcus aureus is the most common pathogen causing co-infections and superinfections in patients with COVID-19. Methicillin-resistant Staphylococcus aureus (MRSA) bacteremia ratios have sharply risen during the pandemic. SAB mortality is 18% at 1 month and 27% at 3 months but has gradually decreased over the last 30 years. Recurrences and reinfections are common (9%). Standardised items to define complicated SAB, and a new cut-off defining persisting bacteremia after 2 days with positive blood cultures have been proposed. Multiple antibiotic combinations have been trialled including vancomycin or daptomycin with β-lactams, fosfomycin, or clindamycin, without significant results. In the recently published guidelines, vancomycin remains the first line of treatment for MRSA bacteremia. For the management of methicillin-susceptible Staphylococcus aureus , cefazolin less frequently causes acute kidney injury than flucloxacillin, and when susceptibility is demonstrated, de-escalation to penicillin G is suggested. SUMMARY Our review confirms that Staphylococcus aureus represents a special aetiology among all causes of bloodstream infections. Pending results of platform and larger trials, its distinct epidemiology and determinants mandate careful integration of clinical variables and best available evidence to optimize patient outcomes.
Collapse
Affiliation(s)
- Alexis Tabah
- Intensive Care Unit, Redcliffe Hospital, Metro North Hospital and Health Services
- Queensland University of Technology
- Faculty of Medicine, University of Queensland
| | - Kevin B Laupland
- Queensland University of Technology
- Department of Intensive Care Services, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| |
Collapse
|
58
|
Tiseo G, Brigante G, Giacobbe DR, Maraolo AE, Gona F, Falcone M, Giannella M, Grossi P, Pea F, Rossolini GM, Sanguinetti M, Sarti M, Scarparo C, Tumbarello M, Venditti M, Viale P, Bassetti M, Luzzaro F, Menichetti F, Stefani S, Tinelli M. Diagnosis and management of infections caused by multidrug-resistant bacteria: guideline endorsed by the Italian Society of Infection and Tropical Diseases (SIMIT), the Italian Society of Anti-Infective Therapy (SITA), the Italian Group for Antimicrobial Stewardship (GISA), the Italian Association of Clinical Microbiologists (AMCLI) and the Italian Society of Microbiology (SIM). Int J Antimicrob Agents 2022; 60:106611. [PMID: 35697179 DOI: 10.1016/j.ijantimicag.2022.106611] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 05/12/2022] [Accepted: 05/29/2022] [Indexed: 02/08/2023]
Abstract
Management of patients with infections caused by multidrug-resistant organisms is challenging and requires a multidisciplinary approach to achieve successful clinical outcomes. The aim of this paper is to provide recommendations for the diagnosis and optimal management of these infections, with a focus on targeted antibiotic therapy. The document was produced by a panel of experts nominated by the five endorsing Italian societies, namely the Italian Association of Clinical Microbiologists (AMCLI), the Italian Group for Antimicrobial Stewardship (GISA), the Italian Society of Microbiology (SIM), the Italian Society of Infectious and Tropical Diseases (SIMIT) and the Italian Society of Anti-Infective Therapy (SITA). Population, Intervention, Comparison and Outcomes (PICO) questions about microbiological diagnosis, pharmacological strategies and targeted antibiotic therapy were addressed for the following pathogens: carbapenem-resistant Enterobacterales; carbapenem-resistant Pseudomonas aeruginosa; carbapenem-resistant Acinetobacter baumannii; and methicillin-resistant Staphylococcus aureus. A systematic review of the literature published from January 2011 to November 2020 was guided by the PICO strategy. As data from randomised controlled trials (RCTs) were expected to be limited, observational studies were also reviewed. The certainty of evidence was classified using the GRADE approach. Recommendations were classified as strong or conditional. Detailed recommendations were formulated for each pathogen. The majority of available RCTs have serious risk of bias, and many observational studies have several limitations, including small sample size, retrospective design and presence of confounders. Thus, some recommendations are based on low or very-low certainty of evidence. Importantly, these recommendations should be continually updated to reflect emerging evidence from clinical studies and real-world experience.
Collapse
Affiliation(s)
- Giusy Tiseo
- Infectious Diseases Unit, Department of Clinical and Experimental Medicine, Azienda Ospedaliera Universitaria Pisana, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Gioconda Brigante
- Clinical Pathology Laboratory, ASST Valle Olona, Busto Arsizio, Italy
| | - Daniele Roberto Giacobbe
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy; Clinica Malattie Infettive, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Floriana Gona
- Emerging Bacterial Pathogens Unit, Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Falcone
- Infectious Diseases Unit, Department of Clinical and Experimental Medicine, Azienda Ospedaliera Universitaria Pisana, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy
| | - Maddalena Giannella
- Infectious Diseases Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Paolo Grossi
- Infectious and Tropical Diseases Unit, Department of Medicine and Surgery, University of Insubria-ASST-Sette Laghi, Varese, Italy
| | - Federico Pea
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy; SSD Clinical Pharmacology, Department for Integrated Infectious Risk Management, IRCCS, Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Gian Maria Rossolini
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy, and Microbiology and Virology Unit, Careggi University Hospital, Florence, Italy
| | - Maurizio Sanguinetti
- Microbiology Unit, Fondazione Policlinico Universitario 'A. Gemelli' IRCCS, Università Cattolica del Sacro Cuore, Largo 'A. Gemelli', Rome, Italy
| | - Mario Sarti
- Clinical Microbiology Laboratory, University of Modena and Reggio Emilia, Modena, Italy
| | - Claudio Scarparo
- Clinical Microbiology Laboratory, Angel's Hospital, AULSS3 Serenissima, Mestre, Venice, Italy
| | - Mario Tumbarello
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Mario Venditti
- Policlinico 'Umberto I', Department of Public Health and Infectious Diseases, 'Sapienza' University of Rome, Rome, Italy
| | - Pierluigi Viale
- Infectious Diseases Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy; Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Matteo Bassetti
- Department of Health Sciences (DISSAL), University of Genoa, Genoa, Italy; Clinica Malattie Infettive, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Francesco Luzzaro
- Clinical Microbiology and Virology Unit, A. Manzoni Hospital, Lecco, Italy
| | - Francesco Menichetti
- Infectious Diseases Unit, Department of Clinical and Experimental Medicine, Azienda Ospedaliera Universitaria Pisana, University of Pisa, Via Paradisa 2, 56124 Pisa, Italy.
| | - Stefania Stefani
- Medical Molecular Microbiology and Antibiotic Resistance Laboratory (MMARLab), Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, Catania, Italy
| | - Marco Tinelli
- Infectious Diseases Consultation Service, IRCCS Istituto Auxologico Italiano, Milan, Italy
| |
Collapse
|
59
|
Ledger EVK, Mesnage S, Edwards AM. Human serum triggers antibiotic tolerance in Staphylococcus aureus. Nat Commun 2022; 13:2041. [PMID: 35440121 PMCID: PMC9018823 DOI: 10.1038/s41467-022-29717-3] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/30/2022] [Indexed: 12/13/2022] Open
Abstract
Staphylococcus aureus frequently causes infections that are challenging to treat, leading to high rates of persistent and relapsing infection. Here, to understand how the host environment influences treatment outcomes, we study the impact of human serum on staphylococcal antibiotic susceptibility. We show that serum triggers a high degree of tolerance to the lipopeptide antibiotic daptomycin and several other classes of antibiotic. Serum-induced daptomycin tolerance is due to two independent mechanisms. Firstly, the host defence peptide LL-37 induces tolerance by triggering the staphylococcal GraRS two-component system, leading to increased peptidoglycan accumulation. Secondly, GraRS-independent increases in membrane cardiolipin abundance are required for full tolerance. When both mechanisms are blocked, S. aureus incubated in serum is as susceptible to daptomycin as when grown in laboratory media. Our work demonstrates that host factors can significantly modulate antibiotic susceptibility via diverse mechanisms, and combination therapy may provide a way to mitigate this.
Collapse
Affiliation(s)
- Elizabeth V K Ledger
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Rd, London, SW7 2AZ, UK
| | - Stéphane Mesnage
- School of Biosciences, University of Sheffield, Sheffield, S10 2TN, UK
| | - Andrew M Edwards
- MRC Centre for Molecular Bacteriology and Infection, Imperial College London, Armstrong Rd, London, SW7 2AZ, UK.
| |
Collapse
|
60
|
Giacobbe DR, Dettori S, Corcione S, Vena A, Sepulcri C, Maraolo AE, De Rosa FG, Bassetti M. Emerging Treatment Options for Acute Bacterial Skin and Skin Structure Infections and Bloodstream Infections Caused by Staphylococcus aureus: A Comprehensive Review of the Evidence. Infect Drug Resist 2022; 15:2137-2157. [PMID: 35498629 PMCID: PMC9041368 DOI: 10.2147/idr.s318322] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 04/07/2022] [Indexed: 12/12/2022] Open
Affiliation(s)
- Daniele Roberto Giacobbe
- Clinica Malattie Infettive, Ospedale Policlinico San Martino – IRCCS, Genoa, Italy
- Department of Health Sciences, University of Genoa, Genoa, Italy
- Correspondence: Daniele Roberto Giacobbe, Clinica Malattie Infettive, Ospedale Policlinico San Martino – IRCCS, L.go R. Benzi 10, Genoa, 16132, Italy, Tel +390105554658, Email
| | - Silvia Dettori
- Clinica Malattie Infettive, Ospedale Policlinico San Martino – IRCCS, Genoa, Italy
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Silvia Corcione
- Department of Medical Sciences, Division of Infectious Diseases, Azienda Ospedaliera Universitaria (A.O.U.) Città della Salute e della Scienza di Torino, University of Turin, Turin, Italy
| | - Antonio Vena
- Clinica Malattie Infettive, Ospedale Policlinico San Martino – IRCCS, Genoa, Italy
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Chiara Sepulcri
- Clinica Malattie Infettive, Ospedale Policlinico San Martino – IRCCS, Genoa, Italy
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | | | - Francesco Giuseppe De Rosa
- Department of Medical Sciences, Division of Infectious Diseases, Azienda Ospedaliera Universitaria (A.O.U.) Città della Salute e della Scienza di Torino, University of Turin, Turin, Italy
| | - Matteo Bassetti
- Clinica Malattie Infettive, Ospedale Policlinico San Martino – IRCCS, Genoa, Italy
- Department of Health Sciences, University of Genoa, Genoa, Italy
| |
Collapse
|
61
|
Burgin DJ, Liu R, Hsieh RC, Heinzinger LR, Otto M. Investigational agents for the treatment of methicillin-resistant Staphylococcus aureus (MRSA) bacteremia: progress in clinical trials. Expert Opin Investig Drugs 2022; 31:263-279. [PMID: 35129409 PMCID: PMC10988647 DOI: 10.1080/13543784.2022.2040015] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 02/06/2022] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Bacteremia caused by Staphylococcus aureus is common. Cases caused by methicillin-resistant S. aureus (MRSA) are particularly formidable and often lethal. The mortality associated with MRSA bacteremia has not significantly decreased over the past couple of decades and concerns regarding efficacy and toxicity of standard therapy highlight the need for novel agents and new therapeutic approaches. AREAS COVERED This paper explores clinical trials investigating novel therapeutic approaches to S. aureus bacteremia. There is a special focus on MRSA bacteremia. Monotherapy and combination therapies and novel antimicrobials and adjunctive therapies that are only recently being established for therapeutic use are discussed. EXPERT OPINION The unfavorable safety profile of combination antimicrobial therapy in clinical trials has outweighed its benefits. Therefore, future investigation should focus on optimizing duration and de-escalation protocols. Antibody and bacteriophage lysin-based candidates have mostly been limited to safety trials, but progress with these agents is demonstrated through a lysin-based agent receiving a phase III trial. Antibiotics indicated for use in treating MRSA skin infections see continued investigation as treatments for MRSA bacteremia despite the difficulty of completing trials in this patient population. Promising agents include dalbavancin, ceftobiprole, ceftaroline, and exebacase.
Collapse
Affiliation(s)
- Dylan J. Burgin
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ryan Liu
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Roger C. Hsieh
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lauren R. Heinzinger
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Michael Otto
- Pathogen Molecular Genetics Section, Laboratory of Bacteriology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
62
|
Alhadrami HA, Abdulaal WH, Hassan HM, Alhakamy NA, Sayed AM. In Silico-Based Discovery of Natural Anthraquinones with Potential against Multidrug-Resistant E. coli. Pharmaceuticals (Basel) 2022; 15:ph15010086. [PMID: 35056143 PMCID: PMC8778091 DOI: 10.3390/ph15010086] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 01/07/2022] [Accepted: 01/09/2022] [Indexed: 01/21/2023] Open
Abstract
E. coli is a Gram-negative bacterium that causes different human infections. Additionally, it resists common antibiotics due to its outer protective membrane. Natural products have been proven to be efficient antibiotics. However, plant natural products are far less explored in this regard. Accordingly, over 16,000 structures covering almost all African medicinal plants in AfroDb in a structural-based virtual screening were used to find efficient anti-E. coli candidates. These drug-like structures were docked into the active sites of two important molecular targets (i.e., E. coli’s Ddl-B and Gyr-B). The top-scoring hits (i.e., got docking scores < −10 kcal/mol) produced in the initial virtual screening (0.15% of the database structures for Ddl-B and 0.17% of the database structures for Gyr-B in the database) were further refined using molecular dynamic simulation-based binding free energy (ΔG) calculation. Anthraquinones were found to prevail among the retrieved hits. Accordingly, readily available anthraquinone derivatives (10 hits) were selected, prepared, and tested in vitro against Ddl-B, Gyr-B, multidrug-resistant (MDR) E. coli, MRSA, and VRSA. A number of the tested derivatives demonstrated strong micromolar enzyme inhibition and antibacterial activity against E. coli, MRSA, and VRSA, with MIC values ranging from 2 to 64 µg/mL. Moreover, both E. coli’s Ddl-B and Gyr-B were inhibited by emodin and chrysophanol with IC50 values comparable to the reference inhibitors (IC50 = 216 ± 5.6, 236 ± 8.9 and 0.81 ± 0.3, 1.5 ± 0.5 µM for Ddl-B and Gyr-B, respectively). All of the active antibacterial anthraquinone hits showed low to moderate cellular cytotoxicity (CC50 > 50 µM) against human normal fibroblasts (WI-38). Furthermore, molecular dynamic simulation (MDS) experiments were carried out to reveal the binding modes of these inhibitors inside the active site of each enzyme. The findings presented in this study are regarded as a significant step toward developing novel antibacterial agents against MDR strains.
Collapse
Affiliation(s)
- Hani A. Alhadrami
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, P.O. Box 80402, Jeddah 21589, Saudi Arabia;
- Molecular Diagnostic Lab, King Abdulaziz University Hospital, King Abdulaziz University, P.O. Box 80402, Jeddah 21589, Saudi Arabia
- Special Infectious Agent Unit, King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80402, Jeddah 21589, Saudi Arabia
| | - Wesam H. Abdulaal
- Cancer and Mutagenesis Unit, Department of Biochemistry, Faculty of Science, King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80402, Jeddah 21589, Saudi Arabia;
| | - Hossam M. Hassan
- Department of Pharmacognosy, Faculty of Pharmacy, Nahda University, Beni-Suef 62513, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Beni-Suef University, Beni-Suef 62511, Egypt
- Correspondence: (H.M.H.); (A.M.S.)
| | - Nabil A. Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, P.O. Box 80402, Jeddah 21589, Saudi Arabia;
| | - Ahmed M. Sayed
- Department of Pharmacognosy, Faculty of Pharmacy, Nahda University, Beni-Suef 62513, Egypt
- Correspondence: (H.M.H.); (A.M.S.)
| |
Collapse
|
63
|
OUP accepted manuscript. J Antimicrob Chemother 2022; 77:1436-1443. [DOI: 10.1093/jac/dkac023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/31/2021] [Indexed: 11/15/2022] Open
|
64
|
Davis K, Greenstein T, Viau Colindres R, Aldridge BB. Leveraging laboratory and clinical studies to design effective antibiotic combination therapy. Curr Opin Microbiol 2021; 64:68-75. [PMID: 34628295 PMCID: PMC8671129 DOI: 10.1016/j.mib.2021.09.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/28/2021] [Accepted: 09/16/2021] [Indexed: 01/21/2023]
Abstract
Interest in antibiotic combination therapy is increasing due to antimicrobial resistance and a slowing antibiotic pipeline. However, aside from specific indications, combination therapy in the clinic is often not administered systematically; instead, it is used at the physician's discretion as a bet-hedging mechanism to increase the chances of appropriately targeting a pathogen(s) with an unknown antibiotic resistance profile. Some recent clinical trials have been unable to demonstrate superior efficacy of combination therapy over monotherapy. Other trials have shown a benefit of combination therapy in defined circumstances consistent with recent studies indicating that factors including species, strain, resistance profile, and microenvironment affect drug combination efficacy and drug interactions. In this review, we discuss how a careful study design that takes these factors into account, along with the different drug interaction and potency metrics for assessing combination performance, may provide the necessary insight to understand the best clinical use-cases for combination therapy.
Collapse
Affiliation(s)
- Kathleen Davis
- Department of Molecular Biology & Microbiology, Tufts University School of Medicine, United States; Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance, United States
| | - Talia Greenstein
- Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance, United States; Graduate School of Biomedical Sciences, Tufts University School of Medicine, United States
| | - Roberto Viau Colindres
- Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance, United States; Department of Geographic Medicine and Infectious Diseases, Tufts Medical Center, United States
| | - Bree B Aldridge
- Department of Molecular Biology & Microbiology, Tufts University School of Medicine, United States; Stuart B. Levy Center for Integrated Management of Antimicrobial Resistance, United States; Graduate School of Biomedical Sciences, Tufts University School of Medicine, United States
| |
Collapse
|
65
|
Bian L, Liang J, Zhao H, Ye K, Li Z, Liu T, Peng J, Wu Y, Lin G. Rapid Monitoring of Vancomycin Concentration in Serum Using Europium (III) Chelate Nanoparticle-Based Lateral Flow Immunoassay. Front Chem 2021; 9:763686. [PMID: 34733823 PMCID: PMC8558538 DOI: 10.3389/fchem.2021.763686] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 09/29/2021] [Indexed: 11/21/2022] Open
Abstract
Establishing personalized medication plans for patients to maximize therapeutic efficacy and minimize the toxicity of vancomycin (VAN) requires rapid, simple, and accurate monitoring of VAN concentration in body fluid. In this study, we have developed a simple and rapid analytical method by integrating Eu (III) chelate nanoparticles (CN-EUs) and lateral flow immunoassay (LFIA) to achieve the real-time monitoring of VAN concentration in serum within 15 min. This approach was performed on nitrocellulose (NC) membrane assembled LFIA strips via indirect competitive immunoassay and exhibited a wide linear range of detection (0.1–80 μg*ml−1) with a low limit of detection (69.2 ng*ml−1). The coefficients of variation (CV) of the intra- and inter-assay in the detection of VAN were 7.12–8.53% and 8.46–11.82%, respectively. The dilution test and specificity indicated this method had a stability that was not affected by the serum matrix and some other antibiotics. Furthermore, the applicability of the proposed method was assessed by comparing the determined results with those measured by LC-MS/MS, showing a satisfactory correlation (R2 = 0.9713). The proposed CN-EUs-based LFIA manifested promising analytical performance, which showed potential value in the real-time monitoring of VAN and could help optimize the clinical use of more antibiotics.
Collapse
Affiliation(s)
- Lun Bian
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Junyu Liang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hui Zhao
- Department of Plastic and Aesthetic Surgery, Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ke Ye
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Zhaoyue Li
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Tiancai Liu
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Jie Peng
- Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yingsong Wu
- Key Laboratory of Antibody Engineering of Guangdong Higher Education Institutes, Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Guanfeng Lin
- Experimental Center of Teaching and Scientific Research, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| |
Collapse
|
66
|
Davis JS, Petersiel N, Tong SYC. How I manage a patient with MRSA bacteraemia. Clin Microbiol Infect 2021; 28:190-194. [PMID: 34757117 DOI: 10.1016/j.cmi.2021.10.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/13/2021] [Accepted: 10/23/2021] [Indexed: 11/29/2022]
Abstract
BACKGROUND Staphylococcus aureus bloodstream infections are common and associated with a high mortality of 15-25%. Methicillin-resistant S. aureus (MRSA) bloodstream infection accounts for 10-40% of cases, and has an even higher mortality. Despite being the 'bread and butter' of clinical infectious diseases practice, robust evidence to guide optimal management is often lacking and there is wide variation in practice. OBJECTIVES To provide a real-world example of a case of MRSA bacteraemia and the thought processes of the authors as key management decision points are reached. SOURCES The discussion is based on recent literature searches of relevant topics. In making recommendations, randomized clinical trial data have been prioritized and highlighted, and where these are not available recommendations are based on the experience and opinions of the authors. CONTENT For a patient with MRSA bacteraemia and a primary bone and joint infection the following points are discussed: empirical antibiotic choice for suspected S. aureus bacteraemia; directed antibiotic choice for MRSA; monitoring and dosing of vancomycin; the role of combination therapy when bacteraemia is persistent; and the duration of therapy and role of switching to oral antibiotics. IMPLICATIONS While broad principles of aggressive source control and appropriate choice and duration of antibiotics are important, the heterogeneity of S. aureus bacteraemia means that a tailored rather than algorithmic approach to management is often required. Further randomized controlled trials are needed to strengthen the evidence base for the management of MRSA bacteraemia.
Collapse
Affiliation(s)
- Joshua S Davis
- Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia; Department of Infectious Diseases, John Hunter Hospital, Newcastle, NSW, Australia.
| | - Neta Petersiel
- Victorian Infectious Diseases Service, The Royal Melbourne and Hospital, At the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia; Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Steven Y C Tong
- Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia; Victorian Infectious Diseases Service, The Royal Melbourne and Hospital, At the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia; Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| |
Collapse
|
67
|
Synergy Mechanisms of Daptomycin-Fosfomycin Combinations in Daptomycin-Susceptible and -Resistant Methicillin-Resistant S. aureus: In vitro, Ex vivo and In vivo Metrics. Antimicrob Agents Chemother 2021; 66:e0164921. [PMID: 34694870 DOI: 10.1128/aac.01649-21] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Increased usage of daptomycin (DAP) for methicillin-resistant Staphylococcus aureus (MRSA) infections has led to emergence of DAP-resistant (DAP-R) strains, resulting in treatment failures. DAP-fosfomycin (Fosfo) combinations are synergistically active against MRSA, although the mechanism(s) of this interaction are not fully understood. The current study explores four unique, but likely interrelated activities of DAP-Fosfo combinations: i) synergistic killing; ii) prevention of evolution of DAP-R; iii) resensitization of already DAP-R subpopulations to a DAP-susceptible (DAP-S) phenotype; and iv) perturbations of specific cell envelope phenotypes known to correlate with DAP-R in MRSA. Using an isogenic DAP-S (CB1483) / DAP-R (CB185) clinical MRSA strain-pair, we demonstrated that DAP + Fosfo combinations: i) enhanced killing of both strains in vitro and ex vivo; ii) increased target tissue clearances of the DAP-R strain in an in vivo model of experimental infective endocarditis (IE); iii) prevented emergence of DAP-R in the DAP-S parental strain both in vitro and ex vivo; and iv) resensitized the DAP-R strain to a DAP-S phenotype ex vivo. Phenotypically, following exposure to sub-MIC Fosfo, the DAP-S/ DAP-R strain-pair exhibited distinct modifications in: i) net positive surface charge (p<0.0001); ii) quantity (p<0.0001) and localization of cell membrane cardiolipin (CL); iii) DAP surface binding; and iv) membrane fluidity (p <0.0001). Furthermore, pre-conditioning to this strain-pair to DAP +/- Fosfo sensitized these organisms to killing by the human host defense peptide, LL37. These data underscore the notion that DAP-Fosfo combinations can impact MRSA clearances within multiple microenvironments, likely based on specific phenotypic adaptations.
Collapse
|
68
|
The Effect of Combination Therapy on Mortality and Adverse Events in Patients with Staphylococcus aureus Bacteraemia: A Systematic Review and Meta-analysis of Randomized Controlled Trials. Infect Dis Ther 2021; 10:2643-2660. [PMID: 34596881 PMCID: PMC8572899 DOI: 10.1007/s40121-021-00539-y] [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] [Received: 08/19/2021] [Accepted: 09/15/2021] [Indexed: 01/16/2023] Open
Abstract
Introduction The findings of randomized controlled trials (RCTs), observational studies, and meta-analyses vary regarding the effectiveness and safety of combination therapy for patients with Staphylococcus aureus bacteraemia (SAB). We aimed to identify the effectiveness and safety of combination therapy in patients with SAB compared with those of monotherapy. Methods We performed a systematic review and meta-analysis to compare combination therapy versus monotherapy in patients with SAB. Two authors independently searched PubMed, Embase, and the Cochrane Library of clinical trials until 17 February 2021. Any RCT comparing mortality or adverse events (AEs) of combination therapy versus monotherapy for patients with SAB was eligible. Summary risk ratios (RRs) and 95% confidence intervals (CIs) were evaluated using a random-effects model. The primary outcome was all-cause mortality at any time point. This meta-analysis is registered with the PROSPERO database (CRD42020188176) and reported according to PRISMA guidelines. Results In total, 1906 articles were identified and screened, and 14 studies (2367 patients) were included in the meta-analysis. There was no significant difference in the risk of all-cause mortality between the two groups (RR = 1.00; 95% CI 0.83–1.20; P = 0.99; I2 = 0%). Similar results were obtained by subgroup analysis of mortality recording time, endocarditis, pathogen resistance, article publication time, number of patients, and adjuvant antibiotics. Notably, combination treatment might significantly increase the risk of drug-related AEs (RR = 1.68; 95% CI 1.06–2.66; P = 0.03; I2 = 67%) and nephrotoxicity (RR = 2.30; 95% CI 1.68–3.16; P < 0.00001; I2 = 0%), although the occurrences of AEs leading to treatment discontinuation and serious AEs were not significantly different between the two groups. Conclusions The meta-analysis suggested that combination therapy could not reduce mortality but might increase the risk of drug-related AEs and nephrotoxicity and should be applied very cautiously. Future studies on combined drug therapy for SAB need careful and rigorous design for specific antibiotic combinations. Supplementary Information The online version contains supplementary material available at 10.1007/s40121-021-00539-y.
Collapse
|
69
|
Suarez JF, Ong’uti S, Holubar M. Select controversies in the management of methicillin-resistant Staphylococcus aureus bacteremia: answers and remaining questions from recent evidence. Fac Rev 2021; 10:66. [PMID: 34557870 PMCID: PMC8441996 DOI: 10.12703/r/10-66] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) bacteremia continues to cause significant morbidity and mortality despite advances in medical therapy. Vancomycin therapy remains the standard of care for most cases of MRSA bacteremia but has pharmacokinetic and pharmacodynamic limitations, dosing complications, and known toxicity. Welcomed clinical trials have recently addressed some of the controversies that plague this field, including optimization of vancomycin dosing and use of combination therapy. In this review, we discuss these trials and their implications for clinical care and future research.
Collapse
Affiliation(s)
- Jose F Suarez
- Jackson Memorial Hospital/University of Miami Miller School of Medicine, Division of Infectious Diseases, Miami, FL, USA
| | - Sharon Ong’uti
- Stanford University School of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford, CA, USA
| | - Marisa Holubar
- Stanford University School of Medicine, Division of Infectious Diseases and Geographic Medicine, Stanford, CA, USA
| |
Collapse
|
70
|
Khatri AM, Anderson AD, Camargo JF. Addition Of Oral Fosfomycin To Antimicrobial Salvage Therapy For Persistent Vancomycin-Resistant Enterococcal Bacteremia. Clin Infect Dis 2021; 74:1710-1711. [PMID: 34536272 DOI: 10.1093/cid/ciab832] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Affiliation(s)
- Akshay M Khatri
- Immunocompromised Host Service, Division of Infectious Diseases, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Anthony D Anderson
- Department of Pharmacy, Sylvester Comprehensive Cancer Center, Miami, FL, USA
| | - Jose F Camargo
- Immunocompromised Host Service, Division of Infectious Diseases, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
71
|
Exploratory Cost-Effectiveness Analysis for Treatment of Methicillin-Resistant Staphylococcus aureus Bloodstream Infections: Is Linezolid or Daptomycin Favored Over Vancomycin? Clin Drug Investig 2021; 41:885-894. [PMID: 34480725 DOI: 10.1007/s40261-021-01077-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/15/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND AND OBJECTIVE Methicillin-resistant Staphylococcus aureus bloodstream infections (MRSAB) cause significant mortality, and often require extended antibiotic therapy. Vancomycin, the most common initial MRSAB treatment, carries significant monitoring burden and nephrotoxicity risks. Our objective was to compare the cost-effectiveness of vancomycin and other antibiotic regimens against MRSAB. METHODS We estimated the cost-effectiveness of intravenous antibiotics (vancomycin, daptomycin, linezolid, ceftaroline/daptomycin) for Veterans Health Administration patients with MRSAB using an exploratory decision-tree model. Primary effectiveness outcome was composite of microbiological failure at 7 days and adverse drug event (ADE)-related discontinuation after at least 7 days. RESULTS In base-case analyses, intravenous linezolid was the least expensive regimen at 4 and 6 weeks. Daptomycin was more expensive and more effective than linezolid, with an incremental cost-effectiveness ratio (ICER) of ~$13,000 (4 weeks) per composite failure avoided. With 6 weeks of treatment, daptomycin was more expensive and more effective than vancomycin (ICER ~$21,000 per composite failure avoided). Vancomycin and ceftaroline/daptomycin were dominated strategies at both 4 and 6 weeks. In one-way sensitivity analyses, vancomycin was favored when its microbiological failure risk was less than 20.1% (base-case: 27.2%), assuming a willingness to pay (WTP) threshold of $40,000/composite treatment failure avoided. In two-way sensitivity analyses, intravenous linezolid was favored if linezolid microbiological failure and ADE-related discontinuation rates were < 22.5% and < 17.3%, respectively. Daptomycin, vancomycin, and linezolid were favored in 50%, 31%, and 17% of 4-week probabilistic iterations, respectively, at $40,000 WTP. CONCLUSION Daptomycin is likely less expensive and more effective than vancomycin or other initial regimens for MRSAB. More data are needed on the safety of linezolid against MRSAB.
Collapse
|
72
|
Grillo S, Cuervo G, Carratala J, San-Juan R, Aguado JM, Morata L, Gomez-Zorrilla S, López-Contreras J, Gasch O, Gomila-Grange A, Iftimie S, Garcia-Pardo G, Calbo E, Boix-Palop L, Oriol I, Jover-Sáenz A, López-Cortés LE, Euba G, Aguirregabiria M, Garcia-Pais MJ, Gioia F, Paño JR, Pedro-Botet ML, Benítez RM, Pérez-Rodríguez MT, Meije Y, Loeches-Yagüe MB, Horna G, Berbel D, Domínguez MÁ, Padullés A, Cobo S, Hereu P, Videla S, Tebe C, Pallarés N, Miro JM, Pujol M. Multicentre, randomised, open-label, phase IV-III study to evaluate the efficacy of cloxacillin plus fosfomycin versus cloxacillin alone in adult patients with methicillin-susceptible Staphylococcus aureus bacteraemia: study protocol for the SAFO trial. BMJ Open 2021; 11:e051208. [PMID: 34353808 PMCID: PMC8344278 DOI: 10.1136/bmjopen-2021-051208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
INTRODUCTION Methicillin-susceptible Staphylococcus aureus (MSSA) bacteraemia is a frequent condition, with high mortality rates. There is a growing interest in identifying new therapeutic regimens able to reduce therapeutic failure and mortality observed with the standard of care of beta-lactam monotherapy. In vitro and small-scale studies have found synergy between cloxacillin and fosfomycin against S. aureus. Our aim is to test the hypothesis that cloxacillin plus fosfomycin achieves higher treatment success than cloxacillin alone in patients with MSSA bacteraemia. METHODS We will perform a superiority, randomised, open-label, phase IV-III, two-armed parallel group (1:1) clinical trial at 20 Spanish tertiary hospitals. Adults (≥18 years) with isolation of MSSA from at least one blood culture ≤72 hours before inclusion with evidence of infection, will be randomly allocated to receive either cloxacillin 2 g/4-hour intravenous plus fosfomycin 3 g/6-hour intravenous or cloxacillin 2 g/4-hour intravenous alone for 7 days. After the first week, sequential treatment and total duration of antibiotic therapy will be determined according to clinical criteria by the attending physician.Primary endpoints: (1) Treatment success at day 7, a composite endpoint comprising all the following criteria: patient alive, stable or with improved quick-Sequential Organ Failure Assessment score, afebrile and with negative blood cultures for MSSA at day 7. (2) Treatment success at test of cure (TOC) visit: patient alive and no isolation of MSSA in blood culture or at another sterile site from day 8 until TOC (12 weeks after randomisation).We assume a rate of treatment success of 74% in the cloxacillin group. Accepting alpha risk of 0.05 and beta risk of 0.2 in a two-sided test, 183 subjects will be required in each of the control and experimental groups to obtain statistically significant difference of 12% (considered clinically significant). ETHICS AND DISSEMINATION Ethical approval has been obtained from the Ethics Committee of Bellvitge University Hospital (AC069/18) and from the Spanish Medicines and Healthcare Product Regulatory Agency (AEMPS, AC069/18), and is valid for all participating centres under existing Spanish legislation. The results will be presented at international meetings and will be made available to patients and funders. TRIAL REGISTRATION NUMBER The protocol has been approved by AEMPS with the Trial Registration Number EudraCT 2018-001207-37. ClinicalTrials.gov Identifier: NCT03959345; Pre-results.
Collapse
Affiliation(s)
- Sara Grillo
- Department of Infectious Diseases, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain
- Bellvitge Institute for Biomedical Research, IDIBELL, Barcelona, Spain
| | - Guillermo Cuervo
- Department of Infectious Diseases, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain
- Bellvitge Institute for Biomedical Research, IDIBELL, Barcelona, Spain
| | - Jordi Carratala
- Department of Infectious Diseases, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain
- University of Barcelona, Barcelona, Spain
| | - Rafael San-Juan
- Department of Infectious Diseases, Hospital Universitario 12 de Octubre, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Jose M Aguado
- Department of Infectious Diseases, Hospital Universitario 12 de Octubre, Madrid, Spain
- Complutense University of Madrid, Madrid, Spain
| | - Laura Morata
- Department of Infectious Diseases, Hospital Clinic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, IDIBAPS, Barcelona, Spain
| | - Silvia Gomez-Zorrilla
- Department of Infectious Diseases, Consorci Parc de Salut MAR de Barcelona, Barcelona, Spain
- Institut de Recerca Hospital del Mar, IMIM, Barcelona, Spain
| | - Joaquín López-Contreras
- Department of Infectious diseases, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Institut d'Investigació Biomèdica Sant Pau IIB Sant Pau, Barcelona, Spain
| | - Oriol Gasch
- Infectious Diseases Department, Consorcio Corporacion Sanitaria Parc Tauli, Sabadell, Spain
- Institut d'Investigació i Innovació Parc Taulí, I3PT, Sabadell, Spain
| | - Aina Gomila-Grange
- Institut d'Investigació i Innovació Parc Taulí, I3PT, Sabadell, Spain
- Consorcio Corporación Sanitaria Parc Taulí, Sabadell, Spain
| | - Simona Iftimie
- Department of Infection and Immunity, Hospital Universitari Sant Joan de Reus, Reus, Spain
| | - Graciano Garcia-Pardo
- Departament of Preventive Medicine, Hospital Universitari de Tarragona Joan XXIII, Tarragona, Spain
| | - Esther Calbo
- Infectious Diseases Unit, Hospital Universitari MutuaTerrassa, Terrassa, Spain
- Fundació per la Docència i Recerca MútuaTerrassa, Terrassa, Spain
| | - Lucía Boix-Palop
- Infectious Diseases Unit, Hospital Universitari MutuaTerrassa, Terrassa, Spain
- Fundació per la Docència i Recerca MútuaTerrassa, Terrassa, Spain
| | - Isabel Oriol
- Department of Internal Medicine, Hospital de Sant Joan Despi Moises Broggi, Sant Joan Despi, Spain
| | - Alfredo Jover-Sáenz
- Territorial Unit of Nosocomial Infection, Hospital Universitari Arnau de Vilanova, Lleida, Spain
- Institut de Recerca Biomèdica de Lleida, IRBLLEIDA, Lleida, Spain
| | - Luis Eduardo López-Cortés
- Department of Infectious diseases, Hospital Universitario Virgen Macarena, Seville, Spain
- Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | - Gorane Euba
- Department of Infectious Diseases, Hospital Universitario Cruces, Barakaldo, Spain
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
| | - Malen Aguirregabiria
- Biocruces Bizkaia Health Research Institute, Barakaldo, Spain
- Microbiology Department, Hospital Universitario Cruces, Barakaldo, Spain
| | - Maria Jose Garcia-Pais
- Internal Medicine, Hospital Universitario Lucus Augusti, Lugo, Spain
- Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Francesca Gioia
- Department of Infectious diseases, Hospital Universitario Ramon y Cajal, Madrid, Spain
- Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
| | - Jose Ramón Paño
- Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón, Zaragoza, Spain
| | - Maria Luisa Pedro-Botet
- Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Rosa Maria Benítez
- Hospital Universitari Germans Trias i Pujol, Badalona, Spain
- Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | | | - Yolanda Meije
- Hospital de Barcelona, Barcelona, Spain
- Societat Cooperativa d'Instal·lacions Assistencials Sanitàries, Barcelona, Spain
| | | | - Gertrudis Horna
- Bellvitge Institute for Biomedical Research, IDIBELL, Barcelona, Spain
| | - Damaris Berbel
- Bellvitge Institute for Biomedical Research, IDIBELL, Barcelona, Spain
- Department of Microbiology and Parassitology, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain
| | - Maria Ángeles Domínguez
- Bellvitge Institute for Biomedical Research, IDIBELL, Barcelona, Spain
- Department of Microbiology and Parassitology, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain
| | - Ariadna Padullés
- Bellvitge Institute for Biomedical Research, IDIBELL, Barcelona, Spain
- Pharmacy Department, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain
| | - Sara Cobo
- Bellvitge Institute for Biomedical Research, IDIBELL, Barcelona, Spain
- Pharmacy Department, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain
| | - Pilar Hereu
- Bellvitge Institute for Biomedical Research, IDIBELL, Barcelona, Spain
- Department of Clinical Pharmacology, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain
| | - Sebastian Videla
- Bellvitge Institute for Biomedical Research, IDIBELL, Barcelona, Spain
- Department of Clinical Pharmacology, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain
| | - Cristian Tebe
- University of Barcelona, Barcelona, Spain
- Biostatistics Unit, Institut d'Investigacio Biomedica de Bellvitge, L'Hospitalet de Llobregat, Spain
| | - Natàlia Pallarés
- University of Barcelona, Barcelona, Spain
- Biostatistics Unit, Institut d'Investigacio Biomedica de Bellvitge, L'Hospitalet de Llobregat, Spain
| | - Josep M Miro
- Department of Infectious Diseases, Hospital Clinic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, IDIBAPS, Barcelona, Spain
| | - Miquel Pujol
- Department of Infectious Diseases, Hospital Universitari de Bellvitge, L'Hospitalet de Llobregat, Spain
- Bellvitge Institute for Biomedical Research, IDIBELL, Barcelona, Spain
| |
Collapse
|
73
|
Pujol M, Tebé C, Pallarès N, Miró JM, Carratalà J. Reply to Pacios-Martínez and García-Monzón. Clin Infect Dis 2021; 72:e923. [PMID: 33005953 DOI: 10.1093/cid/ciaa1498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Miquel Pujol
- Department of Infectious Diseases, Hospital Universitari de Bellvitge, Institut Investigacions Biomèdiques de Bellvitge (IDIBELL), University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Cristian Tebé
- Biostatistics Unit, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Natalia Pallarès
- Biostatistics Unit, IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Jose M Miró
- Department of Infectious Diseases, Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Jordi Carratalà
- Department of Infectious Diseases, Hospital Universitari de Bellvitge, Institut Investigacions Biomèdiques de Bellvitge (IDIBELL), University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
74
|
Pacios-Martínez E, García-Monzón C. Daptomycin Plus Fosfomycin Versus Daptomycin Alone for Methicillin-resistant Staphylococcus aureus Severe Infection: Post Hoc Analysis Is Warranted. Clin Infect Dis 2021; 72:e922. [PMID: 33005937 DOI: 10.1093/cid/ciaa1497] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Enrique Pacios-Martínez
- Internal Medicine Department, Santa Cristina University Hospital, Instituto de Investigación Sanitaria Princesa, Madrid, Spain
| | - Carmelo García-Monzón
- Internal Medicine Department, Santa Cristina University Hospital, Instituto de Investigación Sanitaria Princesa, Madrid, Spain
| |
Collapse
|
75
|
Rose W, Fantl M, Geriak M, Nizet V, Sakoulas G. Current Paradigms of Combination therapy in Methicillin-Resistant Staphylococcus aureus (MRSA) Bacteremia: Does it Work, Which Combination and For Which Patients? Clin Infect Dis 2021; 73:2353-2360. [PMID: 33993226 DOI: 10.1093/cid/ciab452] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Indexed: 11/13/2022] Open
Abstract
The last several years have seen an emergence of literature documenting the utility of combination antimicrobial therapy, particularly in the salvage of refractory MRSA bacteremia. Recent clinical data are shaping conundrums of which regimens may be more beneficial, which can be potentially harmful, and which subset of patients stand to benefit from more aggressive treatment regimens than called for by current standards. In addition, the incorporation of combination therapy for MRSA bacteremia should be accompanied by the reminder that antimicrobial therapy does not need to be uniform for the entire duration, with an early intensive phase in high inoculum infections (e.g. with combination therapy), followed by a consolidation phase (i.e. monotherapy). This review and perspective consolidates the recent data on this subject and directs future goals in filling the knowledge gaps to methodically move forward towards improving patient outcomes.
Collapse
Affiliation(s)
- Warren Rose
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael Fantl
- School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Matthew Geriak
- Pharmacy Department, Sharp Memorial Hospital, San Diego, CA, USA
| | - Victor Nizet
- Division of Host-Microbe Systems & Therapeutics, Center for Immunity, Infection & Inflammation, University of California-San Diego School of Medicine, La Jolla, CA, USA
| | - George Sakoulas
- Division of Host-Microbe Systems & Therapeutics, Center for Immunity, Infection & Inflammation, University of California-San Diego School of Medicine, La Jolla, CA, USA
| |
Collapse
|
76
|
New-Generation Antibiotics for Treatment of Gram-Positive Infections: A Review with Focus on Endocarditis and Osteomyelitis. J Clin Med 2021; 10:jcm10081743. [PMID: 33920526 PMCID: PMC8074169 DOI: 10.3390/jcm10081743] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/24/2021] [Accepted: 04/03/2021] [Indexed: 01/03/2023] Open
Abstract
Infective endocarditis, osteomyelitis, and osteosynthesis-associated infections are mostly caused by Gram-positive bacteria. They are often difficult to treat and are associated with a poor prognosis. In the past 20 years, nine antibiotic drugs with predominant activity against Gram-positive bacteria have been introduced and approved by the Food and Drug Administration or the European Medicines Agency: ceftaroline, daptomycin, telavancin, dalbavancin, oritavancin, linezolid, tedizolid, delafloxacin, and omadacycline. This narrative review aims to provide an overview on these antibiotics with a special focus on their use in infective endocarditis, osteomyelitis, and osteosynthesis-associated infections. Although some of these approved antibiotics are promising, they should not be used as first- or second-line therapy, awaiting more clinical data.
Collapse
|
77
|
Fisher JF, Mobashery S. β-Lactams against the Fortress of the Gram-Positive Staphylococcus aureus Bacterium. Chem Rev 2021; 121:3412-3463. [PMID: 33373523 PMCID: PMC8653850 DOI: 10.1021/acs.chemrev.0c01010] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The biological diversity of the unicellular bacteria-whether assessed by shape, food, metabolism, or ecological niche-surely rivals (if not exceeds) that of the multicellular eukaryotes. The relationship between bacteria whose ecological niche is the eukaryote, and the eukaryote, is often symbiosis or stasis. Some bacteria, however, seek advantage in this relationship. One of the most successful-to the disadvantage of the eukaryote-is the small (less than 1 μm diameter) and nearly spherical Staphylococcus aureus bacterium. For decades, successful clinical control of its infection has been accomplished using β-lactam antibiotics such as the penicillins and the cephalosporins. Over these same decades S. aureus has perfected resistance mechanisms against these antibiotics, which are then countered by new generations of β-lactam structure. This review addresses the current breadth of biochemical and microbiological efforts to preserve the future of the β-lactam antibiotics through a better understanding of how S. aureus protects the enzyme targets of the β-lactams, the penicillin-binding proteins. The penicillin-binding proteins are essential enzyme catalysts for the biosynthesis of the cell wall, and understanding how this cell wall is integrated into the protective cell envelope of the bacterium may identify new antibacterials and new adjuvants that preserve the efficacy of the β-lactams.
Collapse
Affiliation(s)
- Jed F Fisher
- Department of Chemistry and Biochemistry, McCourtney Hall, University of Notre Dame, Notre Dame Indiana 46556, United States
| | - Shahriar Mobashery
- Department of Chemistry and Biochemistry, McCourtney Hall, University of Notre Dame, Notre Dame Indiana 46556, United States
| |
Collapse
|
78
|
Cuervo G, Escrihuela-Vidal F, Gudiol C, Carratalà J. Current Challenges in the Management of Infective Endocarditis. Front Med (Lausanne) 2021; 8:641243. [PMID: 33693021 PMCID: PMC7937698 DOI: 10.3389/fmed.2021.641243] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 02/01/2021] [Indexed: 12/17/2022] Open
Abstract
Infective endocarditis is a relatively rare, but deadly cause of sepsis, with an overall mortality ranging from 20 to 25% in most series. Although the classic clinical classification into syndromes of acute or subacute endocarditis have not completely lost their usefulness, current clinical forms have changed according to the profound epidemiological changes observed in developed countries. In this review, we aim to address the changing epidemiology of endocarditis, several recent advances in the understanding of the pathophysiology of endocarditis and endocarditis-triggered sepsis, new useful diagnostic tools as well as current concepts in the medical and surgical management of this disease. Given its complexity, the management of infective endocarditis requires the close collaboration of multidisciplinary endocarditis teams that must decide on the diagnostic approach; the appropriate initial treatment in the critical phase; the detection of patients needing surgery and the timing of this intervention; and finally the accurate selection of patients for out-of-hospital treatment, either at home hospitalization or with oral antibiotic treatment.
Collapse
Affiliation(s)
- Guillermo Cuervo
- Infectious Diseases Department, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Bellvitge University Hospital, University of Barcelona, Barcelona, Spain.,Spanish Network for Research in Infectious Diseases (REIPI), Instituto de Salud Carlos III, Madrid, Spain
| | - Francesc Escrihuela-Vidal
- Infectious Diseases Department, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Bellvitge University Hospital, University of Barcelona, Barcelona, Spain
| | - Carlota Gudiol
- Infectious Diseases Department, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Bellvitge University Hospital, University of Barcelona, Barcelona, Spain.,Spanish Network for Research in Infectious Diseases (REIPI), Instituto de Salud Carlos III, Madrid, Spain.,Insitut Català d'Oncologia, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, Barcelona, Spain
| | - Jordi Carratalà
- Infectious Diseases Department, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Bellvitge University Hospital, University of Barcelona, Barcelona, Spain.,Spanish Network for Research in Infectious Diseases (REIPI), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|