1
|
de Kretser D, Mora J, Bloomfield M, Campbell A, Cheng MP, Guy S, Hensgens M, Kalimuddin S, Lee TC, Legg A, Mahar RK, Marks M, Marsh J, McGlothin A, Morpeth SC, Sud A, Ten Oever J, Yahav D, Bonten M, Bowen AC, Daneman N, van Hal SJ, Heriot GS, Lewis RJ, Lye DC, McQuilten Z, Paterson DL, Owen Robinson J, Roberts JA, Scarborough M, Webb SA, Whiteway L, Tong SYC, Davis JS, Walls G, Goodman AL. Early Oral Antibiotic Switch in Staphylococcus aureus Bacteraemia: The Staphylococcus aureus Network Adaptive Platform (SNAP) Trial Early Oral Switch Protocol. Clin Infect Dis 2024; 79:871-887. [PMID: 37921609 PMCID: PMC11478773 DOI: 10.1093/cid/ciad666] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 09/07/2023] [Accepted: 10/20/2023] [Indexed: 11/04/2023] Open
Abstract
BACKGROUND Staphylococcus aureus bloodstream infection (bacteremia) is traditionally treated with at least 2 weeks of intravenous (IV) antibiotics in adults, 3-7 days in children, and often longer for those with complicated disease. The current practice of treating S. aureus bacteremia (SAB) with prolonged IV antibiotics (rather than oral antibiotics) is based on historical observational research and expert opinion. Prolonged IV antibiotic therapy has significant disadvantages for patients and healthcare systems, and there is growing interest in whether a switch to oral antibiotics following an initial period of IV therapy is a safe alternative for clinically stable patients. PROTOCOL The early oral switch (EOS) domain of the S. aureus Network Adaptive Platform (SNAP) trial will assess early switch to oral antibiotics compared with continued IV treatment in clinically stable patients with SAB. The primary endpoint is 90-day all-cause mortality. Hospitalised SAB patients are assessed at platform day 7 ±2 (uncomplicated SAB) and day 14 ±2 (complicated SAB) to determine their eligibility for randomization to EOS (intervention) or continued IV treatment (current standard of care). DISCUSSION Recruitment is occurring in the EOS domain of the SNAP trial. As of August 2023, 21% of all SNAP participants had been randomized to the EOS domain, a total of 264 participants across 77 centers, with an aim to recruit at least 1000 participants. We describe challenges and facilitators to enrolment in this domain to aid those planning similar trials.
Collapse
Affiliation(s)
- Dana de Kretser
- Medical Research Council Clinical Trials Unit, University College London, London, United Kingdom
| | - Jocelyn Mora
- Department of Infectious Diseases University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Max Bloomfield
- Department of Infection Services, Wellington Regional Hospital, New Zealand
| | - Anita Campbell
- Telethon Kids Institute, Wesfarmers Center of Infectious Diseases and Vaccines, The University of Western Australia, Perth, Australia
| | - Matthew P Cheng
- Divisions of Infectious Diseases and Medical Microbiology, McGill University Health Center, Montreal, Canada
| | - Stephen Guy
- Department of Infectious Diseases, Eastern Health, Box Hill, Australia
- Monash University (including Australian and New Zealand Intensive Care Research Centre), Clayton, Australia
| | - Marjolein Hensgens
- UMC Utrecht, Utrecht University, Utrecht, The Netherlands
- Julius Center for Health Sciences and Primary Care, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Shirin Kalimuddin
- Department of Infectious Diseases, Singapore General Hospital, Singapore, Singapore
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, Singapore
| | - Todd C Lee
- Clinical Practice Assessment Unit and Division of Infectious Diseases, McGill University, Montreal, Canada
| | - Amy Legg
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
- Herston Infectious Diseases Institute, Herston, Brisbane, Australia
| | - Robert K Mahar
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Parkville, Australia
- Clinical Epidemiology and Biostatistics Unit, Murdoch Children's Research Institute, Parkville, Australia
| | - Michael Marks
- Department of Clinical Research, London School of Hygiene & Tropical Medicine, London, United Kingdom
- Hospital for Tropical Diseases, University College London Hospital, London
- Division of Infection and Immunity, University College London, London
| | - Julie Marsh
- Telethon Kids Institute &/Department of Infectious Diseases &/Wesfarmers Centre for Vaccines and Infectious Diseases, Perth Children's Hospital, Perth, Australia
| | | | - Susan C Morpeth
- Department of Infectious Diseases, Middlemore Hospital, Auckland, New Zealand
| | - Archana Sud
- Department of Infectious Diseases, University of Sydney, Nepean Hospital, Kingswood, New South Wales, Australia
| | - Jaap Ten Oever
- Department of Internal Medicine and Radboud Centre for Infectious Diseases, Radboud University Medical CenterNijmegen, The Netherlands
| | - Dafna Yahav
- Infectious Diseases Unit, Sheba Medical Center, Ramat-Gan, Israel
| | - Marc Bonten
- UMC Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Asha C Bowen
- Telethon Kids Institute &/Department of Infectious Diseases &/Wesfarmers Centre for Vaccines and Infectious Diseases, Perth Children's Hospital, Perth, Australia
| | - Nick Daneman
- Division of Infectious Diseases, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Sebastiaan J van Hal
- Department of Microbiology and Infectious Diseases, Royal Prince Alfred Hospital, Sydney, Australia
- School of Medicine, University of Sydney, Sydney, Australia
| | - George S Heriot
- Department of Infectious Diseases University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | | | - David C Lye
- National Center for Infectious Diseases, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, Singapore
- Yong Loo Lin School of Medicine, Singapore
- Lee Kong Chian School of Medicine, Singapore
| | - Zoe McQuilten
- Monash University (including Australian and New Zealand Intensive Care Research Centre), Clayton, Australia
- Department of Haematology, Monash Health, Melbourne, Australia
| | - David L Paterson
- University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital Campus, Brisbane, Australia
| | - J Owen Robinson
- Department of Infectious Diseases, Royal Perth Hospital, Perth, Australia
- Department of Infectious Diseases, Fiona Stanley Hospital, Murdoch, Australia
- PathWest Laboratory Medicine, Perth, Australia
- College of Science, Health, Engineering and Education, Murdoch University, Murdoch, Australia
| | - Jason A Roberts
- Herston Infectious Diseases Institute, Herston, Brisbane, Australia
- University of Queensland Centre for Clinical Research, Royal Brisbane and Women's Hospital Campus, Brisbane, Australia
- Metro North Health, Brisbane, Australia
- Department of Pharmacy and Intensive Care Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia
- Division of Anesthesiology Critical Care Emergency and Pain Medicine, Nîmes University Hospital, University of Montpellier, Nîmes, France
| | - Matthew Scarborough
- Department of Infectious Diseases, Oxford University Hospitals NHS Trust, Oxford, United Kingdom
| | - Steve A Webb
- Department of Infectious Diseases, Oxford University Hospitals NHS Trust, Oxford, United Kingdom
| | | | - Steven Y C Tong
- Department of Infectious Diseases University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Victorian Infectious Diseases Service, The Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Joshua S Davis
- School of Medicine and Public Health and Hunter Medical Research Institute, University of Newcastle, Newcastle, Australia
| | - Genevieve Walls
- Department of Infectious Diseases, Middlemore Hospital, Auckland, New Zealand
| | - Anna L Goodman
- Medical Research Council Clinical Trials Unit, University College London, London, United Kingdom
- Department of Infectious Diseases, Oxford University Hospitals NHS Trust, Oxford, United Kingdom
- Centre for Clinical Infection and Diagnostics Research, Guy's and St Thomas' Foundation NHS Trust, King's College, London, United Kingdom
| |
Collapse
|
2
|
Kaasch AJ, López-Cortés LE, Rodríguez-Baño J, Cisneros JM, Dolores Navarro M, Fätkenheuer G, Jung N, Rieg S, Lepeule R, Coutte L, Bernard L, Lemaignen A, Kösters K, MacKenzie CR, Soriano A, Hagel S, Fantin B, Lafaurie M, Talarmin JP, Dinh A, Guimard T, Boutoille D, Welte T, Reuter S, Kluytmans J, Martin ML, Forestier E, Stocker H, Vitrat V, Tattevin P, Rommerskirchen A, Noret M, Adams A, Kern WV, Hellmich M, Seifert H. Efficacy and safety of an early oral switch in low-risk Staphylococcus aureus bloodstream infection (SABATO): an international, open-label, parallel-group, randomised, controlled, non-inferiority trial. THE LANCET. INFECTIOUS DISEASES 2024; 24:523-534. [PMID: 38244557 DOI: 10.1016/s1473-3099(23)00756-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 01/22/2024]
Abstract
BACKGROUND Staphylococcus aureus bloodstream infection is treated with at least 14 days of intravenous antimicrobials. We assessed the efficacy and safety of an early switch to oral therapy in patients at low risk for complications related to S aureus bloodstream infection. METHODS In this international, open-label, randomised, controlled, non-inferiority trial done in 31 tertiary care hospitals in Germany, France, the Netherlands, and Spain, adult patients with low-risk S aureus bloodstream infection were randomly assigned after 5-7 days of intravenous antimicrobial therapy to oral antimicrobial therapy or to continue intravenous standard therapy. Randomisation was done via a central web-based system, using permuted blocks of varying length, and stratified by study centre. The main exclusion criteria were signs and symptoms of complicated S aureus bloodstream infection, non-removable foreign devices, and severe comorbidity. The composite primary endpoint was the occurrence of any complication related to S aureus bloodstream infection (relapsing S aureus bloodstream infection, deep-seated infection, and mortality attributable to infection) within 90 days, assessed in the intention-to-treat population by clinical assessors who were masked to treatment assignment. Adverse events were assessed in all participants who received at least one dose of study medication (safety population). Due to slow recruitment, the scientific advisory committee decided on Jan 15, 2018, to stop the trial after 215 participants were randomly assigned (planned sample size was 430 participants) and to convert the planned interim analysis into the final analysis. The decision was taken without knowledge of outcome data, at a time when 126 participants were enrolled. The new sample size accommodated a non-inferiority margin of 10%; to claim non-inferiority, the upper bound of the 95% CI for the treatment difference (stratified by centre) had to be below 10 percentage points. The trial is closed to recruitment and is registered with ClinicalTrials.gov (NCT01792804), the German Clinical trials register (DRKS00004741), and EudraCT (2013-000577-77). FINDINGS Of 5063 patients with S aureus bloodstream infection assessed for eligibility, 213 were randomly assigned to switch to oral therapy (n=108) or to continue intravenous therapy (n=105). Mean age was 63·5 (SD 17·2) years and 148 (69%) participants were male and 65 (31%) were female. In the oral switch group, 14 (13%) participants met the primary endpoint versus 13 (12%) in the intravenous group, with a treatment difference of 0·7 percentage points (95% CI -7·8 to 9·1; p=0·013). In the oral switch group, 36 (34%) of 107 participants in the safety population had at least one serious adverse event compared with 27 (26%) of 103 participants in the intravenous group (p=0·29). INTERPRETATION Oral switch antimicrobial therapy was non-inferior to intravenous standard therapy in participants with low-risk S aureus bloodstream infection. However, it is necessary to carefully assess patients for signs and symptoms of complicated S aureus bloodstream infection at the time of presentation and thereafter before considering early oral switch therapy. FUNDING Deutsche Forschungsgemeinschaft. TRANSLATIONS For the German, Spanish, French and Dutch translations of the abstract see Supplementary Materials section.
Collapse
Affiliation(s)
- Achim J Kaasch
- Institute of Medical Microbiology and Hospital Hygiene, Medical Faculty, Otto von Guericke University Magdeburg, Magdeburg, Germany.
| | - Luis Eduardo López-Cortés
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Department of Medicine, Universidad de Sevilla, Seville, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Rodríguez-Baño
- Unidad Clínica de Enfermedades Infecciosas y Microbiología, Hospital Universitario Virgen Macarena, Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Department of Medicine, Universidad de Sevilla, Seville, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - José Miguel Cisneros
- Unidad Clínica de Enfermedades Infecciosas, Microbiología y Parasitología, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Department of Medicine, Universidad de Sevilla, Seville, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - M Dolores Navarro
- Unidad Clínica de Enfermedades Infecciosas, Microbiología y Parasitología, Hospital Universitario Virgen del Rocío, Instituto de Biomedicina de Sevilla (IBiS)/CSIC, Department of Medicine, Universidad de Sevilla, Seville, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Gerd Fätkenheuer
- Department I of Internal Medicine, University Clinics, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Norma Jung
- Department I of Internal Medicine, University Clinics, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Siegbert Rieg
- Division of Infectious Diseases, Department of Medicine II, Faculty of Medicine, University Medical Centre Freiburg, Freiburg, Germany
| | - Raphaël Lepeule
- Antimicrobial Stewardship Team, Department of Prevention, Diagnosis, and Treatment of Infections, Henri-Mondor University Hospital, Creteil, France
| | - Laetitia Coutte
- Antimicrobial Stewardship Team, Department of Prevention, Diagnosis, and Treatment of Infections, Henri-Mondor University Hospital, Creteil, France
| | - Louis Bernard
- Service de Médecine Interne et Maladies Infectieuses, Centre Hospitalier Régional Universitaire de Tours, Tours, France
| | - Adrien Lemaignen
- Service de Médecine Interne et Maladies Infectieuses, Centre Hospitalier Régional Universitaire de Tours, Tours, France
| | - Katrin Kösters
- Medical Clinic II-Clinic for Gastroenterology, Hepatology, Neurogastroenterology, Infectious Diseases, Hematology, Oncology and Palliative Medicine, Helios Klinikum Krefeld, Krefeld, Germany
| | - Colin R MacKenzie
- Institute of Medical Microbiology and Hospital Hygiene, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Alex Soriano
- Department of Infectious Diseases, Hospital Clínic, Institut d'Investigacions Biomèdiques Agustí Pi i Sunyer (IDIBAPS), CIBERINFEC, Barcelona, Spain
| | - Stefan Hagel
- Institute for Infectious Diseases and Infection Control, Jena University Hospital-Friedrich Schiller University Jena, Jena, Germany
| | - Bruno Fantin
- Internal Medicine Department, Hôpital Beaujon, Assistance Publique Hôpitaux de Paris, Clichy, France
| | | | | | - Aurélien Dinh
- Infectious Diseases Department, Raymond-Poincaré University Hospital, Garches, France
| | - Thomas Guimard
- Infectious Diseases Department, CHD Vendée, La Roche-sur-Yon, France
| | - David Boutoille
- Department of Infectious Diseases, University Hospital of Nantes and CIC 1413, INSERM, Nantes, France
| | - Tobias Welte
- Clinic for Respiratory Medicine and Infectious Diseases, Member of the German Center of Lung Research, Medical School Hannover, Hannover, Germany
| | - Stefan Reuter
- Department of Infectious Diseases and General Internal Medicine, Department of Infection Control, Klinikum Leverkusen, Leverkusen, Germany
| | - Jan Kluytmans
- Department of Medical Microbiology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Maria Luisa Martin
- Infectious Diseases Unit, Internal Medicine Department, Hospital Universitari Son Espases, Fundació Institut d'Investigació Sanitària Illes Balears, Palma de Mallorca, Spain
| | - Emmanuel Forestier
- Infectious Diseases Department, Centre Hospitalier Métropole Savoie, Chambéry, France
| | - Hartmut Stocker
- Klinik für Infektiologie, St Joseph Hospital Berlin Tempelhof, Berlin, Germany
| | - Virginie Vitrat
- Infectious Diseases Unit, Centre Hospitalier d'Annecy Genevois, Epagny Metz-Tessy, France
| | - Pierre Tattevin
- Infectious Diseases and Intensive Care Unit, Pontchaillou University Hospital, Rennes, France
| | - Anna Rommerskirchen
- Institute of Medical Microbiology and Hospital Hygiene, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Marion Noret
- French National Network of Clinical Research in Infectious Diseases, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Anne Adams
- Institute of Medical Statistics and Computational Biology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Winfried V Kern
- Division of Infectious Diseases, Department of Medicine II, Faculty of Medicine, University Medical Centre Freiburg, Freiburg, Germany
| | - Martin Hellmich
- Institute of Medical Statistics and Computational Biology, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Harald Seifert
- Institute for Medical Microbiology, Immunology and Hygiene, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany; German Centre for Infection Research, Partner Site Bonn-Cologne, Cologne, Germany; Institute of Translational Research, CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| |
Collapse
|
3
|
Eleftheriotis G, Marangos M, Lagadinou M, Bhagani S, Assimakopoulos SF. Oral Antibiotics for Bacteremia and Infective Endocarditis: Current Evidence and Future Perspectives. Microorganisms 2023; 11:3004. [PMID: 38138148 PMCID: PMC10745436 DOI: 10.3390/microorganisms11123004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/15/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
Bacteremia and endocarditis are two clinical syndromes that, for decades, were managed exclusively with parenteral antimicrobials, irrespective of a given patient's clinical condition, causative pathogen, or its antibiotic susceptibility profile. This clinical approach, however, was based on low-quality data and outdated expert opinions. When a patient's condition has improved, gastrointestinal absorption is not compromised, and an oral antibiotic regimen reaching adequate serum concentrations is available, a switch to oral antibacterials can be applied. Although available evidence has reduced the timing of the oral switch in bacteremia to three days/until clinical improvement, there are only scarce data regarding less than 10-day intravenous antibiotic therapy in endocarditis. Many standard or studied oral antimicrobial dosages are smaller than the approved doses for parenteral administration, which is a risk factor for treatment failure; in addition, the gastrointestinal barrier may affect drug bioavailability, especially when the causative pathogen has a minimum inhibitory concentration that is close to the susceptibility breakpoint. A considerable number of patients infected by such near-breakpoint strains may not be potential candidates for oral step-down therapy to non-highly bioavailable antibiotics like beta-lactams; different breakpoints should be determined for this setting. This review will focus on summarizing findings about pathogen-specific tailoring of oral step-down therapy for bacteremia and endocarditis, but will also present laboratory and clinical data about antibiotics such as beta-lactams, linezolid, and fosfomycin that should be studied more in order to elucidate their role and optimal dosage in this context.
Collapse
Affiliation(s)
- Gerasimos Eleftheriotis
- Division of Infectious Diseases, Department of Internal Medicine, Medical School, University of Patras, University Hospital of Patras, Rion, 26504 Patras, Greece; (G.E.); (M.M.); (M.L.)
| | - Markos Marangos
- Division of Infectious Diseases, Department of Internal Medicine, Medical School, University of Patras, University Hospital of Patras, Rion, 26504 Patras, Greece; (G.E.); (M.M.); (M.L.)
| | - Maria Lagadinou
- Division of Infectious Diseases, Department of Internal Medicine, Medical School, University of Patras, University Hospital of Patras, Rion, 26504 Patras, Greece; (G.E.); (M.M.); (M.L.)
| | - Sanjay Bhagani
- Department of Infectious Diseases and HIV Medicine, Royal Free London NHS Foundation Trust, London NW3 2QG, UK;
| | - Stelios F. Assimakopoulos
- Division of Infectious Diseases, Department of Internal Medicine, Medical School, University of Patras, University Hospital of Patras, Rion, 26504 Patras, Greece; (G.E.); (M.M.); (M.L.)
| |
Collapse
|
4
|
Yetmar ZA, Chesdachai S, Lahr BD, Challener DW, Arensman Hannan KN, Epps K, Stevens RW, Seville MT, Tande AJ, Virk A. Comparison of Oral and Intravenous Definitive Antibiotic Therapy for Beta-Hemolytic Streptococcus Species Bloodstream Infections from Soft Tissue Sources: a Propensity Score-Matched Analysis. Antimicrob Agents Chemother 2023; 67:e0012023. [PMID: 37191533 PMCID: PMC10269088 DOI: 10.1128/aac.00120-23] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 04/26/2023] [Indexed: 05/17/2023] Open
Abstract
Beta-hemolytic streptococci are common causes of bloodstream infection (BSI). There is emerging data regarding oral antibiotics for BSI but limited for beta-hemolytic streptococcal BSI. We conducted a retrospective study of adults with beta-hemolytic streptococcal BSI from a primary skin/soft tissue source from 2015 to 2020. Patients transitioned to oral antibiotics within 7 days of treatment initiation were compared to those who continued intravenous therapy, after propensity score matching. The primary outcome was 30-day treatment failure (composite of mortality, infection relapse, and hospital readmission). A prespecified 10% noninferiority margin was used for the primary outcome. We identified 66 matched pairs of patients treated with oral and intravenous antibiotics as definitive therapy. Based on an absolute difference in 30-day treatment failure of 13.6% (95% confidence interval 2.4 to 24.8%), the noninferiority of oral therapy was not confirmed (P = 0.741); on the contrary, the superiority of intravenous antibiotics is suggested by this difference. Acute kidney injury occurred in two patients who received intravenous treatment and zero who received oral therapy. No patients experienced deep vein thrombosis or other vascular complications related to treatment. In patients treated for beta-hemolytic streptococcal BSI, those who transitioned to oral antibiotics by day 7 showed higher rates of 30-day treatment failure than propensity-matched patients. This difference may have been driven by underdosing of oral therapy. Further investigation into optimal antibiotic choice, route, and dosing for definitive therapy of BSI is needed.
Collapse
Affiliation(s)
- Zachary A. Yetmar
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Supavit Chesdachai
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Brian D. Lahr
- Division of Clinical Trials and Biostatistics, Mayo Clinic, Rochester, Minnesota, USA
| | - Douglas W. Challener
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Kevin Epps
- Department of Pharmacy, Mayo Clinic, Jacksonville, Florida, USA
| | - Ryan W. Stevens
- Department of Pharmacy, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Aaron J. Tande
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Abinash Virk
- Division of Public Health, Infectious Diseases, and Occupational Medicine, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
5
|
Caniff KE, Rebold N, Rybak MJ. Oral stepdown in Gram-positive bloodstream infections: A step in the right direction. Pharmacotherapy 2023; 43:247-256. [PMID: 36727285 DOI: 10.1002/phar.2775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/13/2023] [Accepted: 01/16/2023] [Indexed: 02/03/2023]
Abstract
Bloodstream infections (BSIs) due to Gram-positive organisms have traditionally been treated with prolonged courses of intravenous antimicrobials. However, this dogma is associated with substantial burden to the patient and health care system. Consequently, there is growing interest in the utilization of oral stepdown therapy, defined as the transition of intravenous therapy to an active oral agent, for this indication. This review highlights available literature examining oral stepdown in adult patients with BSI due to commonly encountered Gram-positive pathogens, including Staphylococcus aureus, Streptococcus spp., and Enterococcus spp. Support for oral stepdown in this setting is primarily derived from observational studies subject to selection bias. Nevertheless, this treatment strategy exhibits promising potential in carefully selected patients as it is consistently associated with reductions in hospital length of stay without jeopardizing clinical cure or survivability. Prospective, randomized trials are needed for validation of oral stepdown in Gram-positive BSI and to identify the optimal patient population and regimen.
Collapse
Affiliation(s)
- Kaylee E Caniff
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA
| | - Nicholas Rebold
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA.,Department of Clinical & Administrative Pharmacy Sciences, College of Pharmacy, Howard University, Washington, District of Columbia, USA
| | - Michael J Rybak
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan, USA.,Division of Infectious Diseases, School of Medicine, Wayne State University, Detroit, Michigan, USA.,Department of Pharmacy, Detroit Receiving Hospital, Detroit, Michigan, USA
| |
Collapse
|
6
|
Waked R, Craig WY, Mercuro NJ, Wungwattana M, Wood E, Rokas KE. Uncomplicated Streptococcal Bacteremia: The Era of Oral Antibiotic Step-down Therapy? Int J Antimicrob Agents 2023; 61:106736. [PMID: 36690120 PMCID: PMC10023366 DOI: 10.1016/j.ijantimicag.2023.106736] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/28/2022] [Accepted: 01/16/2023] [Indexed: 01/22/2023]
Abstract
BACKGROUND The purpose of this study was to compare the clinical outcomes of adults with uncomplicated streptococcal bacteremia who received either oral (PO) step-down or continued intravenous (IV) therapy. METHODS This was a retrospective, single-center, cohort study, including adults admitted with Streptococcal bloodstream infection between January 1, 2013, and December 31, 2020. Only patients with uncomplicated Streptococcal bloodstream infections were included. Patients who transitioned to PO therapy within 5 days from bacteremia onset were compared to patients receiving continued IV therapy. The primary outcome was clinical failure, defined by either 90-day hospital readmission or mortality. Secondary outcomes included hospital length of stay (LOS) and antibiotic-related adverse events (AAEs). RESULTS Of the 264 patients included, 42% were transitioned to PO therapy. Group B Streptococcus (22.7%) was the most common isolate. The most common sources of infection were skin and soft tissue (35%) and pulmonary (25%). Intensive care unit (ICU) stay was more common in the continued IV therapy group (22.2%) than in the PO step-down group (5.4%). The frequency of clinical failure was similar in the IV and PO groups (24.2% vs. 18.0%, P=0.23). The IV group had longer hospital LOS (median, [interquartile range (IQR)]) compared with the PO group (7 [5-13.5] vs. 4 [3-5] days, P<0.001). The incidence of AAEs was similar in the IV and PO groups (1.3% vs. 1.8%, P=0.74). CONCLUSION Oral antibiotic step-down therapy may be appropriate for the treatment of uncomplicated Streptococcal bacteremia, with consideration of factors such as patient comorbidities, type of infection, source control and clinical progress.
Collapse
Affiliation(s)
- Rami Waked
- Infectious Diseases, Maine Medical Center, Portland, Maine, USA
| | - Wendy Y Craig
- Maine Health Institute for Research, Scarborough, Maine, USA
| | | | | | - Emily Wood
- Infectious Diseases, Maine Medical Center, Portland, Maine, USA
| | | |
Collapse
|
7
|
Sequential oral antibiotic in uncomplicated Staphylococcus aureus bacteraemia: a propensity-matched cohort analysis. Clin Microbiol Infect 2023:S1198-743X(23)00054-X. [PMID: 36773773 DOI: 10.1016/j.cmi.2023.02.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 02/01/2023] [Accepted: 02/04/2023] [Indexed: 02/11/2023]
Abstract
OBJECTIVES We aimed to analyse the efficacy and safety of oral sequential therapy (OST) in uncomplicated Staphylococcus aureus bacteraemia (SAB). METHODS Single-centre observational cohort at a tertiary hospital in Spain, including all patients with the first SAB episode from January 2015 to December 2020. We excluded patients with complicated SAB and those who died during the first week. Patients were classified into the OST group (patients who received oral therapy after initial intravenous antibiotic therapy [IVT]), and IVT group (patients who received exclusively IVT). We performed a propensity-score matching to balance baseline differences. The primary composite endpoint was 90-day mortality or microbiological failure. Secondary endpoints included 90-day SAB relapse. RESULTS Out of 407 SAB first episodes, 230 (56.5%) were included. Of these, 112 (n = 48.7%) received OST and 118 (51.3%) IVT exclusively. Transition to oral therapy was performed after 7 days (interquartile range, 4-11). The primary endpoint occurred in 10.7% (11/112) in OST vs. 30.5% (36/118) in IVT (p < 0.001). SAB relapses occurred in 3.6% (4/112) vs. 1.7% (2/118) (p 0.436). None of the deaths in OST were related to SAB or its complications. After propensity-score matching, the primary endpoint was not more frequent in the OST group (relative risk, 0.42; 95% CI, 0.22-0.79). Ninety-day relapses occurred similarly in both groups (relative risk, 1.35; 95% CI, 0.75-2.39). DISCUSSION After an initial intravenous antibiotic, patients with uncomplicated SAB can safely be switched to oral antibiotics without apparent adverse outcomes. This strategy could save costs and complications of prolonged hospital stays. Prospective randomized studies are needed.
Collapse
|
8
|
Impact of specialty on the self-reported practice of using oral antibiotic therapy for definitive treatment of bloodstream infections. ANTIMICROBIAL STEWARDSHIP & HEALTHCARE EPIDEMIOLOGY 2023; 3:e48. [PMID: 36970426 PMCID: PMC10031584 DOI: 10.1017/ash.2023.132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/02/2023] [Accepted: 02/03/2023] [Indexed: 03/11/2023]
Abstract
Abstract
Background:
No established guidelines exist regarding the role of oral antibiotic therapy (OAT) to treat bloodstream infections (BSIs), and practices may vary depending on clinician specialty and experience.
Objective:
To assess practice patterns regarding oral antibiotic use for treatment of bacteremia in infectious diseases clinicians (IDCs, including physicians and pharmacists and trainees in these groups) and non–infectious diseases clinicians (NIDCs).
Design:
Open-access survey.
Participants:
Clinicians caring for hospitalized patients receiving antibiotics.
Methods:
An open-access, web-based survey was distributed to clinicians at a Midwestern academic medical center using e-mail and to clinicians outside the medical center using social media. Respondents answered questions regarding confidence prescribing OAT for BSI in different scenarios. We used χ2 analysis for categorical data evaluated association between responses and demographic groups.
Results:
Of 282 survey responses, 82.6% of respondents were physicians, 17.4% pharmacists, and IDCs represented 69.2% of all respondents. IDCs were more likely to select routine use of OAT for BSI due to gram-negative anaerobes (84.6% vs 59.8%; P < .0001), Klebsiella spp (84.5% vs 69.0%; P < .009), Proteus spp (83.6% vs 71.3%; P < .027), and other Enterobacterales (79.5% vs 60.9%; P < .004). Our survey results revealed significant differences in selected treatment of Staphylococcus aureus syndromes. Fewer IDCs than NIDCs selected OAT to complete treatment for methicillin-resistant S. aureus (MRSA) BSI due to gluteal abscess (11.9% vs 25.6%; P = .012) and methicillin-susceptible S. aureus (MSSA) BSI due to septic arthritis (13.9% vs 20.9%; P = .219).
Conclusions:
Practice variation and discordance with evidence for the use of OAT for BSIs exists among IDCs versus NIDCs, highlighting opportunities for education in both clinician groups.
Collapse
|
9
|
Platts S, Payne BA, Price DA, Pareja-Cebrian L, Schwab U. Oral step-down for Staphylococcus aureus bacteraemia: An opportunity for antimicrobial stewardship? CLINICAL INFECTION IN PRACTICE 2022; 16:100202. [DOI: 10.1016/j.clinpr.2022.100202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
10
|
Sanchez MJ, Patel K, Lindsay EA, Tareen NG, Jo C, Copley LA, Sue PK. Early Transition to Oral Antimicrobial Therapy Among Children With Staphylococcus aureus Bacteremia and Acute Hematogenous Osteomyelitis. Pediatr Infect Dis J 2022; 41:690-695. [PMID: 35703303 DOI: 10.1097/inf.0000000000003594] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Staphylococcus aureus bacteremia (SAB) is a frequent complication of acute hematogenous osteomyelitis (AHO) in children, but data on the optimal duration of parenteral antibiotics prior to transition to oral antibiotics remains sparse. We examined clinical outcomes associated with early transition to oral antimicrobial therapy among children admitted to our institution with AHO and SAB, and evaluated the utility of a severity of illness score (SIS) to guide treatment decisions in this setting. METHODS Children with AHO and SAB admitted to our institution between January 1, 2009, and December 31, 2018, were retrospectively reviewed and stratified according to a previously validated SIS into mild (0-3), moderate (4-7) and severe (8-10) cohorts. Groups were assessed for differences in treatment (eg, parenteral and oral antibiotic durations, surgeries) and clinical response (eg, bacteremia duration, acute kidney injury, length of stay and treatment failure). RESULTS Among 246 children identified with AHO and SAB, median parenteral antibiotic duration differed significantly between mild (n = 80), moderate (n = 98) and severe (n = 68) cohorts (3.6 vs. 6.5 vs. 14.3 days; P ≤ 0.001). SIS cohorts also differed with regard to number of surgeries (0.4 vs. 1.0 vs. 2.1; P ≤ 0.001), duration of bacteremia (1.0 vs. 2.0 vs. 4.0 days; P ≤ 0.001), acute kidney injury (0.0% vs. 3.0% vs. 20.5%; P ≤ 0.001), hospital length of stay (4.8 vs. 7.4 vs. 16.4 days; P ≤ 0.001) and total duration of antibiotics (34.5 vs. 44.7 vs. 60.7 days; P ≤ 0.001). Early transition to oral antimicrobial therapy among mild or moderate SIS cohorts was not associated with treatment failure despite SAB. CONCLUSIONS SAB is associated with a wide range of illness among children with AHO, and classification of severity may be useful for guiding treatment decisions. Early transition to oral antimicrobial therapy appears safe in children with mild or moderate AHO despite the presence of SAB.
Collapse
Affiliation(s)
- Maria J Sanchez
- From the Department of Pediatrics, Children's Health System of Texas
| | - Karisma Patel
- Department of Pharmacy, University of Texas Southwestern Medical Center, Children's Health System of Texas, Dallas, Texas
| | - Eduardo A Lindsay
- Department of Family Medicine, Mayagüez Medical Center, Mayagüez, Puerto Rico
| | - Naureen G Tareen
- From the Department of Pediatrics, Children's Health System of Texas
| | - Chanhee Jo
- Research Department, Texas Scottish Rite Hospital for Children
| | - Lawson A Copley
- Department of Orthopaedic Surgery and Pediatrics, University of Texas Southwestern Medical Center, Children's Health System of Texas, Texas Scottish Rite Hospital for Children, Dallas, Texas
| | - Paul K Sue
- Department of Pediatrics, Division of Pediatric Infectious Diseases, University of Texas Southwestern Medical Center, Dallas
| |
Collapse
|
11
|
Molina KC, Lunowa C, Lebin M, Segerstrom Nunez A, Azimi SF, Krsak M, Mueller SW, Miller MA. Comparison of Sequential Dalbavancin to Standard-of-Care Treatment for Staphylococcus aureus Bloodstream Infections. Open Forum Infect Dis 2022; 9:ofac335. [PMID: 35899276 PMCID: PMC9314917 DOI: 10.1093/ofid/ofac335] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 07/12/2022] [Indexed: 11/20/2022] Open
Abstract
Background Dalbavancin (DAL) is a long-acting lipoglycopeptide with activity against Staphylococcus aureus, including methicillin-resistant S. aureus (MRSA). This study investigates DAL as sequential therapy in S. aureus bloodstream infections (BSIs). Methods We conducted a retrospective cohort study from 2014 to 2021 comparing sequential DAL with standard-of-care therapy (SoC) for S. aureus BSI. The primary outcome was 90-day clinical failure (90-day all-cause mortality or 90-day recurrence). Secondary outcomes were incidence of acute kidney injury, creatinine phosphokinase elevations, catheter-related thrombosis, and hospital-acquired infections. Analyses were adjusted using inverse probability of treatment weighting (IPTW). Results Overall, 225 patients (45 DAL, 180 SoC) were included. DAL patients had a higher incidence of community-acquired infection and persons who use drugs; SoC patients had more comorbidities and a longer duration of bacteremia. MRSA incidence was similar between the DAL and SoC groups. The median length of stay was 16 days among DAL recipients compared with 24 days among SoC recipients. Central catheter placement was 17.8% compared with 57.2% in the SoC group. Ninety-day clinical failure occurred in 13.3% and 18.3% of participants in the DAL and SOC groups, respectively. In IPTW-adjusted analysis, sequential DAL was not associated with 90-day clinical failure (adjusted odds ratio, 0.94; 95% CI, 0.333–2.32). Conclusions This study provides preliminary evidence that select patients with S. aureus BSI treated with sequential DAL have similar clinical failure rates, with significant reductions in catheter placement and hospital length of stay compared with SoC. Further prospective evaluation is needed.
Collapse
Affiliation(s)
- Kyle C Molina
- Department of Pharmacy, University of Colorado Hospital , Aurora, CO , USA
- Department of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences , Aurora, CO , USA
- Department of Medicine, Division of Infectious Diseases, University of Colorado School of Medicine , Aurora, CO , USA
| | - Cali Lunowa
- Department of Pharmacy, University of Colorado Hospital , Aurora, CO , USA
- Department of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences , Aurora, CO , USA
| | - Madelyn Lebin
- Department of Pharmacy, University of Colorado Hospital , Aurora, CO , USA
- Department of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences , Aurora, CO , USA
| | - Andrea Segerstrom Nunez
- Department of Pharmacy, University of Colorado Hospital , Aurora, CO , USA
- Department of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences , Aurora, CO , USA
| | - Sara F Azimi
- Department of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences , Aurora, CO , USA
| | - Martin Krsak
- Department of Medicine, Division of Infectious Diseases, University of Colorado School of Medicine , Aurora, CO , USA
| | - Scott W Mueller
- Department of Pharmacy, University of Colorado Hospital , Aurora, CO , USA
- Department of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences , Aurora, CO , USA
| | - Matthew A Miller
- Department of Pharmacy, University of Colorado Hospital , Aurora, CO , USA
- Department of Clinical Pharmacy, University of Colorado Skaggs School of Pharmacy and Pharmaceutical Sciences , Aurora, CO , USA
- Department of Medicine, Division of Infectious Diseases, University of Colorado School of Medicine , Aurora, CO , USA
| |
Collapse
|
12
|
Mun SJ, Kim SH, Huh K, Cho SY, Kang CI, Chung DR, Peck KR. Oral step-down therapy in patients with uncomplicated Staphylococcus aureus primary bacteremia and catheter-related bloodstream infections. J Chemother 2022; 34:319-325. [PMID: 35100939 DOI: 10.1080/1120009x.2022.2031469] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Patients with uncomplicated Staphylococcus aureus primary bacteremia and catheter-related bloodstream infection (CRBSI) should be treated for at least 14 days. However, evidence for oral step-down therapy is lacking in these patients. A retrospective cohort was identified from 2013 to 2018 in a 1,950-bed tertiary hospital. An oral antimicrobial therapy (OAT) group was defined as patients treated with oral antibiotics following less than 10 days of intravenous antimicrobial therapy (IAT). Treatment failure was defined as any case of recurrence or death within 90 days. A total of 103 patients were included in the analysis, including 32 patients treated with OAT. Rates of treatment failure were 3.2% and 12.7% in the OAT and IAT groups (P = 0.113). The length of hospital stay was shorter in the OAT group. OAT was not an independent risk factor for treatment failure. OAT may reduce the duration of hospitalization without adverse effects in these patients.
Collapse
Affiliation(s)
- Seok Jun Mun
- Division of Infectious Diseases, Department of Internal Medicine, Inje University Busan Paik Hospital, Inje University College of Medicine, Busan, South Korea
| | - Si-Ho Kim
- Division of Infectious Diseases, Samsung Changwon Hospital, Sungkyunkwan University School of Medicine, Changwon, South Korea
| | - Kyungmin Huh
- Division of Infectious Diseases, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Sun Young Cho
- Division of Infectious Diseases, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Cheol-In Kang
- Division of Infectious Diseases, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Doo Ryeon Chung
- Division of Infectious Diseases, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Kyong Ran Peck
- Division of Infectious Diseases, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| |
Collapse
|
13
|
Intravenous versus Oral Step-Down for the Treatment of Staphylococcus aureus Bacteremia in a Pediatric Population. PHARMACY 2022; 10:pharmacy10010016. [PMID: 35076616 PMCID: PMC8788527 DOI: 10.3390/pharmacy10010016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/07/2022] [Accepted: 01/12/2022] [Indexed: 01/18/2023] Open
Abstract
Limited data are available regarding optimal antimicrobial therapy for Staphylococcus aureus bacteremia (SAB) in pediatric patients. The purpose of this study was to assess clinical characteristics and outcomes associated with intravenous (IV) versus oral step-down treatment of pediatric SAB. This study evaluated patients aged 3 months to 18 years that received at least 72 h of inpatient treatment for SAB. The primary endpoint was 30-day readmission. Secondary endpoints included hospital length of stay and inpatient mortality. One hundred and one patients were included in this study. The median age was 7.9 years. Patients who underwent oral step-down were less likely to be immunocompromised and more likely to have community-acquired SAB from osteomyelitis or skin and soft tissue infection (SSTI). More patients in the IV therapy group had a 30-day readmission (10 (25.6%) vs. 3 (5.3%), p = 0.006). Mortality was low (5 (5%)) and not statistically different between groups. Length of stay was greater in patients receiving IV therapy only (11 vs. 7 days, p = 0.001). In this study, over half of the patients received oral step-down therapy and 30-day readmission was low for this group. Oral therapy appears to be safe and effective for patients with SAB from osteomyelitis or SSTIs.
Collapse
|
14
|
Florman K, Jones HT, Moores R. How to investigate and manage a patient with a Staphylococcus aureus bacteraemia. Br J Hosp Med (Lond) 2021; 82:1-7. [PMID: 34601929 DOI: 10.12968/hmed.2021.0077] [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: 11/11/2022]
Abstract
Staphylococcus aureus bacteraemia is common, and associated with significant morbidity and mortality as a result of its high relapse rate and the risk of complicated infection. A positive blood culture for S. aureus should prompt a thorough patient assessment to identify a potential focus of infection, and the risk factors for the development or presence of complicated infection. Clinical management depends on the patient's characteristics and presenting features. This article gives a systematic approach to the patient with S. aureus bacteraemia, including points to look for on history and examination, the markers of complicated infection, and when to request transoesophageal echocardiography and further imaging. Treatment principles outlined include the rationale for choice of antibiotic treatment and need to involve infection specialists.
Collapse
Affiliation(s)
- Katia Florman
- Department of Acute Medicine, Royal Free Hospital, London, UK
| | - Howell T Jones
- Department of Geriatric Medicine, UCL, Royal Free Hospital, London, UK
| | - Rachel Moores
- Department of Infectious Diseases, Royal Free Hospital, London, UK
| |
Collapse
|
15
|
Drennan PG, Green JK, Gardiner SJ, Metcalf SCL, Kirkpatrick CMJ, Everts RJ, Zhang M, Chambers ST. Population pharmacokinetics of free flucloxacillin in patients treated with oral flucloxacillin plus probenecid. Br J Clin Pharmacol 2021; 87:4681-4690. [PMID: 33963595 DOI: 10.1111/bcp.14887] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 04/22/2021] [Accepted: 05/02/2021] [Indexed: 12/27/2022] Open
Abstract
Oral flucloxacillin may be coadministered with probenecid to reduce flucloxacillin clearance and increase attainment of pharmacokinetic-pharmacodynamic (PK/PD) targets. The aims of this study were to develop a population PK model of free flucloxacillin when administered orally with probenecid, and to identify optimal dosing regimens for this combination. METHODS We performed a prospective observational study of adults (45 participants) treated with oral flucloxacillin 1000 mg and probenecid 500 mg 8-hourly for proven or probable staphylococcal infections. Steady-state mid-dose-interval flucloxacillin measurements (45 concentrations) were combined with existing data from a crossover study of healthy participants receiving flucloxacillin with and without probenecid (11 participants, 363 concentrations). We developed a population pharmacokinetic model of free flucloxacillin concentrations within Monolix, and used Monte Carlo simulation to explore optimal dosing regimens to attain PK/PD targets proposed in the literature (free drug time above minimum inhibitory concentration). RESULTS Flucloxacillin disposition was best described by a 1-compartment model with a lag time and first-order absorption. Free flucloxacillin clearance depended on probenecid, allometrically-scaled fat free mass (FFM) and estimated glomerular filtration rate (eGFR). Predicted PK/PD target attainment was suboptimal with standard dosing regimens with flucloxacillin alone, but substantially improved in the presence of probenecid. CONCLUSION The simulation results reported can be used to identify dose regimens that optimise flucloxacillin exposure according to eGFR and FFM. Patients with higher FFM and eGFR may require the addition of probenecid and 6-hourly dosing to achieve PK/PD targets. The regimen was well-tolerated, suggesting a potential for further evaluation in controlled clinical trials to establish efficacy.
Collapse
Affiliation(s)
- Philip G Drennan
- Department of Microbiology and Infectious Diseases, Royal Prince Alfred Hospital, Sydney, Australia.,Department of Microbiology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Jared K Green
- Department of Infectious Diseases, Christchurch Hospital, Christchurch, New Zealand
| | - Sharon J Gardiner
- Department of Infectious Diseases, Christchurch Hospital, Christchurch, New Zealand.,Department of Clinical Pharmacology, Christchurch Hospital, Christchurch, New Zealand.,Department of Pharmacy, Christchurch Hospital, Christchurch, New Zealand
| | - Sarah C L Metcalf
- Department of Infectious Diseases, Christchurch Hospital, Christchurch, New Zealand
| | - Carl M J Kirkpatrick
- Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Melbourne, Australia
| | | | - Mei Zhang
- Department of Medicine, University of Otago, Christchurch, New Zealand.,Toxicology, Canterbury Health Laboratories, Christchurch, New Zealand
| | - Stephen T Chambers
- Department of Infectious Diseases, Christchurch Hospital, Christchurch, New Zealand.,Department of Pathology, University of Otago, Christchurch, New Zealand
| |
Collapse
|
16
|
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: 17.3] [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
|
17
|
Early Oral Antibiotic Switch for Staphylococcus aureus Bacteremia: Many Are Called, but Few Are Chosen. Antimicrob Agents Chemother 2020; 64:AAC.00317-20. [PMID: 32393495 DOI: 10.1128/aac.00317-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Staphylococcus aureus bacteremia (SAB) is a complicated, high-risk disease. For selected low-risk SAB, the role of oral antibiotic stepdown therapy is unknown. Bupha-Intr et al. report a retrospective cohort of low-risk SAB patients who did well with a short duration of intravenous antibiotics, followed by an additional ∼10 days of oral antibiotics, primarily using beta-lactams. Prospective trials will help further define the efficacy of this approach.
Collapse
|
18
|
Dagher M, Fowler VG, Wright PW, Staub MB. A Narrative Review of Early Oral Stepdown Therapy for the Treatment of Uncomplicated Staphylococcus aureus Bacteremia: Yay or Nay? Open Forum Infect Dis 2020; 7:ofaa151. [PMID: 32523971 PMCID: PMC7270708 DOI: 10.1093/ofid/ofaa151] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 04/28/2020] [Indexed: 02/06/2023] Open
Abstract
Historically, intravenous (IV) antibiotics have been the cornerstone of treatment for uncomplicated Staphylococcus aureus bacteremia (SAB). However, IV antibiotics are expensive, increase the rates of hospital readmission, and can be associated with catheter-related complications. As a result, the potential role of oral antibiotics in the treatment of uncomplicated SAB has become a subject of interest. This narrative review article aims to summarize key arguments for and against the use of oral antibiotics to complete treatment of uncomplicated SAB and evaluates the available evidence for specific oral regimens. We conclude that evidence suggests that oral step-down therapy can be an alternative for select patients who meet the criteria for uncomplicated SAB and will comply with medical treatment and outpatient follow-up. Of the currently studied regimens discussed in this article, linezolid has the most support, followed by fluoroquinolone plus rifampin.
Collapse
Affiliation(s)
- Michael Dagher
- Division of Infectious Diseases, Duke University School of Medicine, Durham, North Carolina, USA
| | - Vance G Fowler
- Division of Infectious Diseases, Duke University School of Medicine, Durham, North Carolina, USA
| | - Patty W Wright
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Milner B Staub
- Division of Infectious Diseases, Vanderbilt University Medical Center, Nashville, Tennessee, USA.,Geriatric Research, Education and Clinical Center (GRECC), Tennessee Valley Healthcare System, Veterans Health Administration, Nashville, Tennessee, USA
| |
Collapse
|