Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Articles

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
Achim J Kaasch, Luis Eduardo López-Cortés, Jesús Rodríguez-Baño, José Miguel Cisneros, M Dolores Navarro, Gerd Fätkenheuer, Norma Jung,
Siegbert Rieg, Raphaël Lepeule, Laetitia Coutte, Louis Bernard, Adrien Lemaignen, Katrin Kösters, Colin R MacKenzie, Alex Soriano, Stefan Hagel,
Bruno Fantin, Matthieu Lafaurie, Jean-Philippe Talarmin, Aurélien Dinh, Thomas Guimard, David Boutoille, Tobias Welte, Stefan Reuter,
Jan Kluytmans, Maria Luisa Martin, Emmanuel Forestier, Hartmut Stocker, Virginie Vitrat, Pierre Tattevin, Anna Rommerskirchen, Marion Noret,
Anne Adams, Winfried V Kern, Martin Hellmich, Harald Seifert, for the SABATO study group*

Summary
Background Staphylococcus aureus bloodstream infection is treated with at least 14 days of intravenous antimicrobials. Lancet Infect Dis 2024
We assessed the efficacy and safety of an early switch to oral therapy in patients at low risk for complications related Published Online
to S aureus bloodstream infection. January 17, 2024
https://doi.org/10.1016/
S1473-3099(23)00756-9
Methods In this international, open-label, randomised, controlled, non-inferiority trial done in 31 tertiary care
See Online/Comment
hospitals in Germany, France, the Netherlands, and Spain, adult patients with low-risk S aureus bloodstream infection https://doi.org/10.1016/
were randomly assigned after 5–7 days of intravenous antimicrobial therapy to oral antimicrobial therapy or to S1473-3099(24)00032-X
continue intravenous standard therapy. Randomisation was done via a central web-based system, using permuted *Members listed at the end of
blocks of varying length, and stratified by study centre. The main exclusion criteria were signs and symptoms of the Article
complicated S aureus bloodstream infection, non-removable foreign devices, and severe comorbidity. The composite For the German translation of
primary endpoint was the occurrence of any complication related to S aureus bloodstream infection (relapsing the abstract see Online for
appendix 1
S aureus bloodstream infection, deep-seated infection, and mortality attributable to infection) within 90 days, assessed
For the Spanish translation of the
in the intention-to-treat population by clinical assessors who were masked to treatment assignment. Adverse events
abstract see Online for
were assessed in all participants who received at least one dose of study medication (safety population). Due to slow appendix 2
recruitment, the scientific advisory committee decided on Jan 15, 2018, to stop the trial after 215 participants were For the French translation of the
randomly assigned (planned sample size was 430 participants) and to convert the planned interim analysis into the abstract see Online for
final analysis. The decision was taken without knowledge of outcome data, at a time when 126 participants were appendix 3
enrolled. The new sample size accommodated a non-inferiority margin of 10%; to claim non-inferiority, the upper For the Dutch translation of the
bound of the 95% CI for the treatment difference (stratified by centre) had to be below 10 percentage points. The trial abstract see Online for
appendix 4
is closed to recruitment and is registered with ClinicalTrials.gov (NCT01792804), the German Clinical trials register
Institute of Medical
(DRKS00004741), and EudraCT (2013–000577–77). Microbiology and Hospital
Hygiene, Medical Faculty,
Findings Of 5063 patients with S aureus bloodstream infection assessed for eligibility, 213 were randomly assigned to Otto von Guericke University
switch to oral therapy (n=108) or to continue intravenous therapy (n=105). Mean age was 63·5 (SD 17·2) years and Magdeburg, Magdeburg,
Germany (Prof A J Kaasch MD);
148 (69%) participants were male and 65 (31%) were female. In the oral switch group, 14 (13%) participants met the Unidad Clínica de
primary endpoint versus 13 (12%) in the intravenous group, with a treatment difference of 0·7 percentage points Enfermedades Infecciosas y
(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 Microbiología, Hospital
least one serious adverse event compared with 27 (26%) of 103 participants in the intravenous group (p=0·29). Universitario Virgen Macarena
(L E López-Cortés MD,
Prof J Rodríguez-Baño MD) and
Interpretation Oral switch antimicrobial therapy was non-inferior to intravenous standard therapy in participants Unidad Clínica de
with low-risk S aureus bloodstream infection. However, it is necessary to carefully assess patients for signs and Enfermedades Infecciosas,
Microbiología y Parasitología,
symptoms of complicated S aureus bloodstream infection at the time of presentation and thereafter before considering
Hospital Universitario Virgen
early oral switch therapy. del Rocío (Prof J M Cisneros MD,
M Dolores Navarro MD),
Funding Deutsche Forschungsgemeinschaft. Instituto de Biomedicina de
Sevilla (IBiS)/CSIC, Department
of Medicine, Universidad de
Copyright © 2024 Elsevier Ltd. All rights reserved. Sevilla, Seville, Spain; Centro de
Investigación Biomédica en
Introduction metastatic complications, and relapse are common.3 Red de Enfermedades
Infecciosas (CIBERINFEC),
Staphylococcus aureus bloodstream infection affects Low-risk S aureus bloodstream infection occurs in a
Instituto de Salud Carlos III,
20–30 people per 100 000 population annually and has a subset of patients in whom risk factors and the clinical Madrid, Spain (L E López-Cortés,
3-month mortality of 20–30%.1,2 Metastatic foci, late course do not suggest elevated risk for the presence or Prof J Rodríguez-Baño,

www.thelancet.com/infection Published online January 17, 2024 https://doi.org/10.1016/S1473-3099(23)00756-9 1


Descargado para Anonymous User (n/a) en Austral University Faculty of Biomedical Sciences de ClinicalKey.es por Elsevier en enero 30, 2024. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
Articles

Prof J M Cisneros,
M Dolores Navarro); Research in context
Department I of Internal
Medicine, University Clinics Evidence before this study (0·08, 0·03–0·14). However, none of these studies focused on
(Prof G Fätkenheuer MD, The recommended treatment for Staphylococcus aureus S aureus bloodstream infection.
N Jung MD), Institute of bloodstream infection is at least 14 days of antimicrobial
Medical Statistics and Added value of this study
therapy, which is usually administered intravenously. It is
Computational Biology This study is the first randomised controlled trial to show non-
(A Adams MSc, unclear whether an early switch to oral antimicrobials is safe
inferiority of early oral antimicrobial switch therapy compared
Prof M Hellmich PhD), and and effective. We searched PubMed for studies in adult patients
Institute for Medical with standard intravenous treatment in patients with low-risk
with S aureus bloodstream infection from database inception to
Microbiology, Immunology S aureus bloodstream infection. The findings support results
and Hygiene
Nov 10, 2023, using the terms (“oral” OR “per os”) AND
from observational studies that reported similar outcomes
(Prof H Seifert MD), Faculty of “Staphylococcus aureus” AND (“bacteraemia” OR “bloodstream
between groups.
Medicine, University Hospital infection”). Several retrospective observational studies found a
Cologne, University of Cologne, similar or lower mortality in patients with S aureus bloodstream Implications of all the available evidence
Cologne, Germany; Division of
Infectious Diseases,
infection receiving early oral antimicrobial switch therapy than The trial indicates that in the subgroup of patients with low-risk
Department of Medicine II, standard intravenous therapy. However, since these studies are S aureus bloodstream infection, a switch to oral antimicrobial
Faculty of Medicine, University not randomised, they are prone to bias, particularly medication can be considered after 5–7 days of appropriate
Medical Centre Freiburg,
confounding by indication. Metastatic foci, relapse rate, and anti-staphylococcal intravenous therapy. Patients with low-risk
Freiburg, Germany
(Prof S Rieg MD, late metastatic complications are more common in S aureus S aureus bloodstream infection represent a small subgroup of
Prof W V Kern MD); bloodstream infection than in other bloodstream infections. patients with S aureus bloodstream infection, and a careful
Antimicrobial Stewardship Therefore, studies addressing severe infections with other assessment of the individual patient is a prerequisite for
Team, Department of
microorganisms cannot be extrapolated to S aureus. assigning the low-risk category. Our study included only a few
Prevention, Diagnosis, and
Treatment of Infections, Henri- A systematic review identified 21 randomised controlled trials, patients with meticillin-resistant S aureus (MRSA), which might
Mondor University Hospital, published until March, 2021, comparing oral with intravenous limit generalisability of the results to patients with bloodstream
Creteil, France (R Lepeule MD, treatment for osteomyelitis, bacteraemia, and endocarditis. infection caused by MRSA. Future studies should address other
L Coutte MD); Service de
No significant difference was found for overall treatment subgroups of patients with S aureus bloodstream infection,
Médecine Interne et Maladies
Infectieuses, Centre Hospitalier success in ten trials assessing bacteraemia (risk difference 0·07, particularly from other risk categories, and confirm the benefits
Régional Universitaire de Tours, 95% CI 0·01–0·15) and four trials on infective endocarditis of early oral switch.
Tours, France
(Prof L Bernard MD,
A Lemaignen MD); Medical
development of complicated infection (ie, deep-seated the second trial, patients with S aureus bloodstream
Clinic II—Clinic for
Gastroenterology, Hepatology, infection, prolonged bacteraemia, and metastatic foci).4–6 infection were excluded. Retrospective observational
Neurogastroenterology, In a prospective cohort study conducted in 2006–11, studies have found similar or lower 30-day or 90-day
Infectious Diseases, 292 (23%) of 1288 patients with S aureus bloodstream mortality for oral switch therapy compared with
Hematology, Oncology and
Palliative Medicine, Helios
infection met criteria for low-risk infection.4 intravenous therapy in patients with S aureus bloodstream
Klinikum Krefeld, Krefeld, The optimal antimicrobial therapy in patients with infection.5,6,13–19 However, retrospective studies have a high
Germany (K Kösters MD); S aureus bloodstream infection has been a topic of risk of confounding by indication. Two surveys reported
Institute of Medical considerable debate. Guidelines recommend at least that less than 20% of infectious disease physicians from
Microbiology and Hospital
Hygiene, University Hospital
14 days of intravenous antimicrobial therapy for low-risk the USA and Canada felt comfortable with switching to
Düsseldorf, Düsseldorf, S aureus bloodstream infection,7,8 whereas the updated oral medication in patients with S aureus bloodstream
Germany UK guidelines for the treatment of meticillin-resistant infection.20,21
(Prof C R MacKenzie MD, S aureus (MRSA) bacteraemia consider co-trimoxazole as Here, we report the results of the SABATO trial, a
A Rommerskirchen MSc);
Department of Infectious
an oral step-down therapy.9 An early switch to oral randomised non-inferiority trial, which aimed to evaluate
Diseases, Hospital Clínic, antimicrobial therapy might reduce hospital stays and the efficacy and safety of an early switch to oral
Institut d’Investigacions infusion-related complications. However, S aureus antimicrobial therapy in patients with low-risk S aureus
Biomèdiques Agustí Pi i Sunyer bloodstream infection might not be treated successfully bloodstream infection compared with standard
(IDIBAPS), CIBERINFEC,
Barcelona, Spain
if adequate serum concentrations are not reached by oral intravenous antimicrobial therapy.
(A Soriano MD PhD); Institute medication. Furthermore, earlier outpatient management
for Infectious Diseases and could lead to reduced compliance with the prescribed Methods
Infection Control, Jena drug regimen and delayed identification of complications. Study design
University Hospital—Friedrich
Schiller University Jena, Jena,
Randomised controlled trials directly addressing an We conducted an international, open-label, parallel-
Germany (S Hagel MD); Internal early oral switch in S aureus bloodstream infection are group, randomised, controlled, non-inferiority trial in
Medicine Department, Hôpital scarce, and previous trials have included few participants patients with low-risk S aureus bloodstream infection in
Beaujon, Assistance Publique
with S aureus bloodstream infection receiving oral 31 tertiary care hospitals in four European countries
Hôpitaux de Paris, Clichy,
France (Prof B Fantin MD); therapy.10 Two trials have shown non-inferiority of an oral (Germany, France, the Netherlands, and Spain; figure 1;
Infectious Diseases switch in patients with endocarditis11 and bone and joint appendix 5 pp 11–12). The non-inferiority design was
Department, Saint-Louis infection.12 However, the first trial mainly included justified by expected benefits for patients on early oral
Hospital, Paris, France
participants with organisms other than S aureus, and in switch therapy, such as a lower rate of complications

2 www.thelancet.com/infection Published online January 17, 2024 https://doi.org/10.1016/S1473-3099(23)00756-9


Descargado para Anonymous User (n/a) en Austral University Faculty of Biomedical Sciences de ClinicalKey.es por Elsevier en enero 30, 2024. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
Articles

(M Lafaurie MD); Department of


5063 participants assessed for eligibility Infectious Diseases, Centre
Hospitalier de Cornouaille,
Quimper, France
4850 ineligible (J-P Talarmin MD); Infectious
4694 did not meet eligibility criteria Diseases Department,
154 were not asked to be randomised or declined to participate Raymond-Poincaré University
2 randomised in error Hospital, Garches, France
(A Dinh MD); Infectious
Diseases Department, CHD
213 randomised (intention-to-treat Vendée, La Roche-sur-Yon,
population) France (T Guimard MD);
Department of Infectious
Diseases, University Hospital of
Nantes and CIC 1413, INSERM,
108 assigned to the oral switch group 105 assigned to the intravenous group Nantes, France
(Prof D Boutoille MD PhD); Clinic
for Respiratory Medicine and
1 discontinued (positive follow-up blood culture) 2 discontinued (withdrew informed consent) Infectious Diseases, Member of
the German Center of Lung
Research, Medical School
Hannover, Hannover, Germany
107 received at least one dose of study drug and 103 received at least one dose of study drug and
included in safety population included in safety population (Prof T Welte MD); Department
of Infectious Diseases and
General Internal Medicine,
Department of Infection
2 excluded (insufficient duration of pre-study antimicrobial 2 excluded
therapy) 1 positive follow-up blood culture before randomisation Control, Klinikum Leverkusen,
1 intravascular prosthesis Leverkusen, Germany
(Prof S Reuter MD); Department
of Medical Microbiology,
105 included in modified intention-to-treat 101 included in modified intention-to-treat University Medical Center
population population Utrecht, Utrecht University,
Utrecht, Netherlands
(Prof J Kluytmans MD);
19 excluded 22 excluded Infectious Diseases Unit,
17 protocol violations* 23 protocol violations* Internal Medicine Department,
2 withdrew consent after starting study drug 1 missing data on attributability of death Hospital Universitari Son
2 missing data on attributability of death Espases, Fundació Institut
d’Investigació Sanitària Illes
Balears, Palma de Mallorca,
86 included in clinically evaluable population 79 included in clinically evaluable population Spain (M L Martin MD);
Infectious Diseases
Department, Centre Hospitalier
Figure 1: Trial profile
Métropole Savoie, Chambéry,
Four participants in the oral group and three in the intravenous group were lost to follow-up. *Some participants had more than one protocol violation.
France (E Forestier MD); Klinik
für Infektiologie, St Joseph
Hospital Berlin Tempelhof,
related to intravenous therapy and a shorter hospital stay. present before enrolment, such as a deep-seated focus (eg, Berlin, Germany (H Stocker MD);
Although the study completed in December, 2019, data endocarditis, pneumonia, infected implant, undrained Infectious Diseases Unit, Centre
analysis and sharing were delayed by the COVID-19 abscess, empyema, and osteomyelitis), septic shock within Hospitalier d’Annecy Genevois,
pandemic. The study protocol was published and 4 days before random assignment,24 prolonged Epagny Metz-Tessy, France
(V Vitrat MD); Infectious
approved by the ethics committees of all participating bacteraemia (defined as a positive blood culture obtained Diseases and Intensive Care
centres and the respective competent authorities.22,23 An >72 h after start of adequate antimicrobial therapy, even if Unit, Pontchaillou University
independent data monitoring committee periodically there was a negative follow-up blood culture thereafter), Hospital, Rennes, France
assessed the safety of the trial participants. and a body temperature higher than 38°C measured on (Prof P Tattevin MD); French
National Network of Clinical
both days before random assignment. Patients were Research in Infectious Diseases,
Participants excluded if intravascular catheters were not removed Assistance Publique-Hôpitaux
Adult patients (aged ≥18 years) with S aureus isolated from within 4 days after the first positive blood culture was de Paris, Paris, France
at least one blood culture were eligible if they had received drawn. Patients with a higher a-priori risk for (M Noret MSc); German Centre
for Infection Research, Partner
5–7 days of appropriate intravenous antimicrobial therapy, complications related to S aureus bloodstream infection Site Bonn-Cologne, Cologne,
initiated within 72 h after the first positive blood culture were also excluded—namely, patients with a recent history Germany (Prof H Seifert);
was drawn, and at least one follow-up blood culture of S aureus bloodstream infection within the preceding Institute of Translational
Research, CECAD Cluster of
obtained within 24–96 h after the start of appropriate 3 months, injection drug use, severe immunodeficiency
Excellence, University of
antimicrobial therapy. Blood cultures taken in this period or severe immunosuppression, or presence of a prosthetic Cologne, Cologne, Germany
had to be negative for S aureus for the patient to be heart valve or deep-seated vascular graft. (Prof H Seifert)
included. Patients were excluded if signs and symptoms After a protocol amendment (ethics approval on
of complicated S aureus bloodstream infection were Dec 29, 2014), patients with end-stage renal disease,

www.thelancet.com/infection Published online January 17, 2024 https://doi.org/10.1016/S1473-3099(23)00756-9 3


Descargado para Anonymous User (n/a) en Austral University Faculty of Biomedical Sciences de ClinicalKey.es por Elsevier en enero 30, 2024. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
Articles

Correspondence to: severe liver disease, a prosthetic joint, or a pacemaker follow-up visits. Adverse events were graded according to
Prof Achim J Kaasch, Institute of were eligible if the infective focus was a removable the Common Terminology Criteria for Adverse Events,
Medical Microbiology and
Hospital Hygiene, Medical
intravascular catheter or a skin or soft-tissue infection.23 version 4.0.25 Only events graded 3 or worse were
Faculty, Otto von Guericke Prosthetic joints and pacemakers needed to be implanted recorded and reported from the start of treatment to the
University Magdeburg, more than 6 months before enrolment. Patients with end of the study. C difficile infection was determined via a
39120 Magdeburg, Germany end-stage renal disease or a pacemaker required an positive test result during a diarrhoea episode.
achim.kaasch@med.ovgu.de
echocardiogram to rule out endocarditis (appendix 5
See Online for appendix 5
p 25). All participants gave written informed consent Outcomes
before enrolment. The composite primary endpoint was complications
related to S aureus bloodstream infection within 90 days,
Randomisation and masking defined as relapsing S aureus bloodstream infection,
Participants were randomly allocated to treatment (1:1) no deep-seated infection with S aureus, or death attributable
earlier than 1 day before starting the study drug through a to S aureus bloodstream infection. This outcome was
central internet randomisation service (TENALEA), centrally assessed. Relapsing S aureus bloodstream
stratified by study centre with permuted blocks of varying infection was defined as a positive blood culture for
length. Participants and investigators were unmasked to S aureus within the intervention or follow-up period.
treatment assignment. An independent clinical review Deep-seated infection was defined as any deep-seated
committee, who made the final adjudication, was masked focus of S aureus infection resulting from haematogenous
to group assignment (drug names were removed and dissemination. Diagnosis of deep-seated infection
only masked information was provided to the committee). required a positive culture from the respective site or a
blood culture positive with S aureus plus imaging studies
Procedures indicating a deep-seated focus. Death was attributed to
Participants either switched to oral therapy or continued S aureus infection when at least one of the following
intravenous standard therapy after 5–7 days of conditions was present: positive blood culture for
intravenous standard therapy, with a total duration of S aureus taken within 72 h before death, persistent focus
antimicrobial therapy of 14 days. Oral antimicrobials of deep-seated S aureus infection at time of death,
were selected by the study physician according to persistent signs and symptoms of systemic infection at
susceptibility results and suspected allergy or intolerance time of death as judged by the study physician, or post-
in the following order: co-trimoxazole (160 mg mortem analysis proving S aureus-related complication
trimethoprim plus 800 mg sulfamethoxazole) every 12 h as the cause of death. All other deaths were classified as
for meticillin-susceptible S aureus (MSSA) and MRSA, unrelated to S aureus.
then clindamycin 600 mg every 8 h for MSSA or linezolid Secondary outcomes were the length of hospital stay
600 mg every 12 h for MRSA in the oral switch group; from the date of first positive blood culture to hospital
and intravenous flucloxacillin 2 g every 6 h (cloxacillin discharge or death; complications of intravenous
2 g every 6 h in Spain and France), cefazolin 2 g every 8 h, administration (caused by study medication or not) from
or vancomycin 1 g every 12 h for MSSA or vancomycin 1 g the start of study drug to the end of follow-up; C difficile
every 12 h or daptomycin 6–10 mg/kg every 24 h for infection; 14-day, 30-day, and up to 90-day survival;
MRSA in the intravenous standard therapy group. Dose adverse events; and serious adverse events.
adjustments in individual patients were performed as
judged appropriate by the local investigator. Therapeutic Statistical analysis
drug monitoring for vancomycin was recommended The trial was originally designed with a 5% non-
to be performed once weekly starting before the inferiority margin, which yielded a total sample size of
fourth vancomycin dose, and the target vancomycin 430 participants (one-sided α=0·05, β=0·2; one interim
trough concentration was 10–20 µg/mL. The local analysis at an information fraction of 0·5 [ie,
hospital pharmacy provided the study drugs and standard 215 participants] using the O’Brien-Fleming bound
drugs from the pharmacy were used. of 2·373), with 2·5% of participants reaching the
Data on participants’ sex were collected by the study endpoint plus adjustments for deaths unrelated to
teams and defined as recorded in the hospital information S aureus blood infection (10%), protocol violations (10%),
system. Ethnicity data were not collected. and stratification (5%; appendix 5 pp 5–6). To
Follow-up started at the end of therapy and ended accommodate conflicting views in the review board of
90 days from the first positive blood culture. A 30-day the German Research Foundation about the size of the
follow-up visit was added as a protocol amendment non-inferiority margin (ie, 5% or 10%) we planned a
(Dec 29, 2014). Antimicrobial treatment, S aureus sequentially rejective testing procedure (hierarchical
bloodstream-related complications, length of admission testing stratified by centre) based on Zhao’s test from
to hospital, survival, adverse events, Clostridium difficile weaker to stronger assertions.26 Due to slow recruitment,
infection, and complications of intravenous therapy were the scientific advisory committee decided on Jan 15, 2018,
assessed during the follow-up period and during to convert the interim analysis to the final analysis. The

4 www.thelancet.com/infection Published online January 17, 2024 https://doi.org/10.1016/S1473-3099(23)00756-9


Descargado para Anonymous User (n/a) en Austral University Faculty of Biomedical Sciences de ClinicalKey.es por Elsevier en enero 30, 2024. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
Articles

decision was taken without knowledge of outcome data, to S aureus bloodstream infection that occurred more than
at a time when 126 participants were enrolled. The new 7 days after starting the study drug were counted as
sample size still accommodated a 10% non-inferiority meeting the primary endpoint and missing or
margin (one-sided α=0·05, β=0·2, expected event rate indeterminate outcomes were disregarded (thus,
of 2·5%, hierarchical testing stratified by centre with complications that could have been identified with a more
Zhao’s test). stringent work-up when starting the study drug were
According to recommendations from the European excluded; post-hoc analysis); (3) only complications related
Medicines Agency, we initially selected the clinically to S aureus bloodstream infection that were confirmed
evaluable population (ie, per-protocol population) for the with a microbiological result were counted as events and
primary analysis and the intention-to-treat population for missing or indeterminate outcomes were disregarded
the coprimary analysis.27,28 However, updated guidance (prespecified analysis); and (4) the primary endpoint was
recommends the intention-to-treat population for the modified to include all-cause mortality (prespecified
primary analysis,29 and we therefore report both analysis).
populations side by side, with a focus on the intention-to- The difference in median lengths of stay between
treat population. MSSA and MRSA was evaluated by the Mann-Whitney
We also assessed the primary endpoint in the modified U test and corresponding Hodges-Lehman 95% CI.
intention-to-treat population, which comprised parti­ Kaplan-Meier curves were calculated to assess differences
cipants who were randomly assigned and received any in time to S aureus bloodstream infection-related
study drug, excluding participants in whom an inclusion complications and the length of hospital stay (prespecified
criterion incompatible with low-risk S aureus bloodstream analyses) and compared with an unstratified log-rank
infection was violated before starting the study drug. For test. For time-to-event analyses, hazard ratios were
example, participants were excluded from the modified estimated from Cox regression. To assess the effect of
intention-to-treat population when follow-up blood non-attributable mortality, competing risks were
cultures (obtained before the start of study drug) turned calculated according to Fine and Gray, considering non-
positive after the start of study drug. In both intention-to- attributable mortality as a competing event (post-hoc
treat populations, data were analysed as assigned, with sensitivity analysis).30 Safety endpoints were assessed
indeterminate and missing outcomes counted as the with Fisher’s exact test. Rates and rate ratios of adverse
primary outcome being met. events were calculated by Poisson regression. The p value
The clinically evaluable population included all for the primary endpoint is for non-inferiority; all other
participants who were treated according to the protocol p values are for superiority. Statistical calculations were
with study drug, were observed until the end of follow- done using SAS (version 9.4) and Stata (version 17.0).
up, or they reached the primary endpoint and did not Reporting of the trial followed the extension of the For CONSORT extensions see
receive antimicrobial therapy during follow-up that could CONSORT statement to non-inferiority trials. A masked, https://www.equator-network.
org/reporting-guidelines/
have masked the primary endpoint. Study drug was independent clinical review committee carefully
consort/
considered according to the protocol when at least 5 days evaluated study outcomes and treatment. This trial is
of initial intravenous therapy was given and the total registered with ClinicalTrials.gov (NCT01792804), the
duration of antimicrobial therapy was 12–16 days, thus German clinical trials register (DRKS00004741), and
allowing a variation of 15% around the 14-day target EudraCT (2013–000577–77).
duration. The safety population included all participants
who received any study drug. Specifically, participants Role of the funding source
who ever received an oral antimicrobial agent were The funder of the study had no role in study design, data
compared with participants who never received an oral collection, data analysis, data interpretation, or writing of
antimicrobial agent. the report.
Prespecified subgroups with at least ten events were
formed according to baseline data of age, sex, meticillin Results
susceptibility, focus of infection, specific comorbidities From Dec 20, 2013, to Dec 22, 2019, 5063 participants
(pacemaker, prosthetic joint, moderate or severe liver were assessed for eligibility and 213 were enrolled in the
disease, end-stage renal disease, and immuno­ intention-to-treat population (figure 1; appendix 5
suppression), Charlson Comorbidity Index, country, and pp 17–19). 108 participants were assigned to the oral
whether any echocardiography was performed. switch group and 105 to the intravenous group.
In sensitivity analyses, we estimated the treatment The clinically evaluable population consisted of
difference using alternative definitions for endpoints in 165 participants, with 86 participants in the oral switch
the intention-to-treat, modified intention-to-treat, and group and 79 participants in the intravenous standard
clinically evaluable populations. We did sensitivity analyses therapy group. The safety population comprised
wherein: (1) missing or indeterminate outcomes were not 210 participants who received at least one dose of study
considered to indicate that the primary endpoint had been drug, including 107 participants in the oral switch group
met (prespecified analysis); (2) only complications related and 103 in the intravenous group.

www.thelancet.com/infection Published online January 17, 2024 https://doi.org/10.1016/S1473-3099(23)00756-9 5


Descargado para Anonymous User (n/a) en Austral University Faculty of Biomedical Sciences de ClinicalKey.es por Elsevier en enero 30, 2024. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
Articles

In the intention-to-treat population, baseline charac­ infection. There were numerically more participants in
teristics were largely balanced between groups and the oral switch group than the intravenous group with
participants were overall representative of low-risk moderate or severe liver disease (11 [10%] of 108 vs
S aureus infection (table 1). Mean age was 63·5 years four [4%] of 105) and diabetes (44 [41%] vs 28 [27%]).
(SD 17·5), and 148 (69%) participants were male and Before starting study medication, participants received a
65 (31%) were female. Overall, catheter-related infection median of 6 days of intravenous antimicrobials in both
and skin and soft-tissue infection were the most prevalent groups. Baseline characteristics in the clinically evaluable
foci, and 16 (8%) participants had an MRSA bloodstream population were similar to the intention-to-treat
population.
Intention-to-treat population Clinically evaluable Participants in the intention-to-treat population were
population treated with study medication for a median duration of
Oral switch Intravenous Oral switch Intravenous 8 days (IQR 7–9) in both groups, resulting in a full
group (n=108) group (n=105) group (n=86) group (n=79) treatment course with antimicrobials of a median of
Age, years 64·4 (16·8) 62·6 (17·6) 62·4 (17·1) 62·1 (17·8) 14 days (IQR 14–15; appendix 5 p 20). Of 108 participants
Sex in the oral switch group, 63 (58%) received co-
Male 71 (66%) 77 (73%) 59 (69%) 58 (73%) trimoxazole, 35 (32%) received clindamycin, nine (8%)
Female 37 (34%) 28 (27%) 27 (31%) 21 (27%) received linezolid, and one (1%) did not receive any study
BMI, kg/m2 27·6 (6·7) 25·6 (5·4) 27·4 (6·5) 26·1 (5·3)
medication. Of 105 participants in the intravenous group,
Time in hospital before 10 (7–14) 11 (7–15) 10 (7–13) 10 (7–13)
46 (44%) received cefazolin, 45 (43%) received intravenous
randomisation, days flucloxacillin or cloxacillin, seven (7%) received
Intervention to remove or drain 9 (8%) 14 (13%) 5 (6%) 10 (13%) vancomycin, five (5%) received daptomycin, and two (2%)
infective focus did not receive any study medication. Nine (4%) of
Echocardiography (TTE or TEE, or 69 (64%) 60 (57%); 1 55 (64%) 46 (58%) 213 participants who initially started study medication
both) performed within 7 days before switched to an alternative study medication (four oral
or after randomisation
switch and five intravenous standard therapy).
CRP at baseline visit, mg/L* 35 (13–70); 13 23 (10–59); 15 35 (14–64); 11 20 (11–52); 13
In the intention-to-treat population, the primary
Resistance of Staphylococcus aureus isolate
composite outcome (ie, complication related to S aureus
Meticillin 6 (6%) 10 (10%) 4 (5%) 5 (6%)
bloodstream infection or a missing outcome within
Clindamycin 12 (12%); 4 11 (11%); 5 10 (12%); 4 7 (10%); 5
90 days) occurred in 14 (13%) of 108 participants in the
Co-trimoxazole 1 (1%); 2 3 (3%); 1 0 (0%); 2 2 (3%)
oral switch group and 13 (12%) of 105 in the intravenous
Focus of infection
standard therapy group (table 2). The treatment
Peripheral venous catheter 47 (44%) 46 (44%) 41 (48%) 35 (44%) difference was 0·7 percentage points (95% CI –7·8 to 9·1;
Central venous catheter 24 (22%) 25 (24%) 18 (21%) 16 (20%) p=0·013). In the clinically evaluable population,
Skin and soft-tissue infection 26 (24%) 22 (21%) 19 (22%) 19 (24%) complications related to S aureus bloodstream infection
Other† 5 (5%) 4 (4%) 4 (5%) 3 (4%) occurred in three (4%) of 86 participants in the oral
Not identified 6 (6%) 8 (8%) 4 (5%) 6 (8%) switch group versus four (5%) of 79 participants in the
Comorbidities intravenous group (treatment difference –2·9 percentage
Moderate or severe liver disease 11 (10%) 4 (4%) 8 (10%) 2 (3%) points, 95% CI –9·6 to 3·9; p<0·0001). Non-inferiority
Chronic renal failure 17 (16%); 1 18 (17%) 10 (12%); 1 12 (15%) was achieved in both populations (upper bound of the
End-stage renal disease 9 (8%); 1 5 (5%) 5 (6%); 1 4 (5%) 95% CI was <10 percentage points for the intention-to-
Chronic lung disease 14 (13%); 1 17 (16%) 10 (12%) 11 (14%) treat population and <5 percentage points for the
Diabetes without end-organ 25 (23%) 18 (17%) 21 (24%) 11 (14%) clinically evaluable population). The results showed
damage stability in sensitivity analyses when addressing missing
Diabetes with end-organ damage 19 (18%) 10 (10%) 13 (15%) 9 (11%) or indeterminate outcomes, the contribution of
Any immunosuppression‡ 16 (15%) 16 (15%); 1 12 (14%) 14 (18%); 1 complications related to S aureus bloodstream infection
Charlson Comorbidity Index 3 (1–5) 3 (1–4) 2 (1–4) 3 (1–4) that occurred more than 7 days after random assignment,
Intravenous antimicrobials before 6 (6–7) 6 (5–7) 7 (6–7) 6 (5–7) and whether complications were microbiologically
randomisation, days
confirmed as being related to S aureus infection
Data are mean (SD), n (%), median (IQR), n (%); missing, or median (IQR); missing. TEE=transoesophageal (appendix 5 pp 13–14). However, when non-attributable
echocardiography. TTE=transthoracic echocardiography. *Laboratory results were from the day of randomisation;
if unavailable, the closest result within 3 days before the start of study drug was accepted. †In the intention-to-treat
mortality was included in the primary endpoint, the
population, the sites of other focus were urinary tract (n=2), suspected pulmonary focus (n=2), and cholecystitis (n=1) 95% CI crossed the 10% non-inferiority margin in the
in the oral switch group and urinary tract (n=4) in the intravenous group. ‡Any immunosuppression comprises intention-to-treat population but not in the clinically
systemic corticosteroid therapy for more than 1 week, current antineoplastic chemotherapy, any other
evaluable population (appendix 5 pp 13–14). Cumulative
immunosuppressive therapy, and organ transplant. A transplanted organ (kidney) was present in one participant,
which would have been an exclusion criterion. The clinical review committee decided to include the participant in the incidence plots for the primary outcome with (figure 2A)
clinically evaluable population since the transplant was present for more than 10 years. and without non-attributable death considered as a
competing risk factor (figure 2B) show a higher
Table 1: Baseline characteristics of the intention-to-treat and clinically evaluable populations
cumulative incidence in the intravenous group; however,

6 www.thelancet.com/infection Published online January 17, 2024 https://doi.org/10.1016/S1473-3099(23)00756-9


Descargado para Anonymous User (n/a) en Austral University Faculty of Biomedical Sciences de ClinicalKey.es por Elsevier en enero 30, 2024. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
Articles

Intention-to-treat population Clinically evaluable population


Oral switch Intravenous Percentage-point Oral switch Intravenous Percentage-point
group (n=108) group (n=105) difference (95% CI) group (n=86) group (n=79) difference (95% CI)
Primary endpoint
SAB-related complication within 14 (13%) 13 (12%) 0·7 (–7·8 to 9·1) 3 (4%) 4 (5%) –2·9 (–9·6 to 3·9)
90 days
Reason primary outcome was met
SAB-related complication 6 (6%) 8 (8%) –2·1 (–9·7 to 5·5) 3 (4%) 4 (5%) –1·6 (–9·0 to 5·8)
Relapsing SAB 3 (3%) 4 (4%) –1·0 (–6·8 to 4·7) 2 (2%) 2 (3%) –0·2 (–5·1 to 4·7)
Deep-seated infection with 5 (5%) 8 (8%) –3·0 (–10·4 to 4·4) 3 (4%) 4 (5%) –1·6 (–9·0 to 5·8)
S aureus
Death attributable to SAB 2 (2%) 0 1·9 (–1·6 to 5·3) 1 (1%) 0 1·2 (–2·3 to 4·6)
Missing outcome data 8 (7%) 5 (5%) 2·7 (–4·7 to 10·0) ∙∙ ∙∙ ∙∙
Attributability of death 3 (3%) 1 (1%) 1·8 (–2·7 to 6·4) ∙∙ ∙∙ ∙∙
non-evaluable
Secondary endpoints
Length of hospital stay from SAB 12 (9–19) 16 (10–19) –2 (–4 to 0); p=0·043* 11 (9–16) 15 (10–18) –2 (–5 to 0); p=0·020*
onset, days
Participants with complications of intravenous administration†
Any complication 9 (9%); 11 17 (17%); 5 –7·9 (–17·6 to 1·9) 6 (7%); 3 13 (17%); 2 –9·5 (–20·5 to 1·5)
Chemical phlebitis 7 9 –2·1 (–10·1 to 5·9) 5 8 –4·3 (–13·8 to 5·2)
Infectious thrombophlebitis or 0 2 –1·9 (–5·5 to 1·7) 0 2 –2·5 (–7·2 to 2·2)
phlebitis
Other‡ 2 6 –3·9 (–9·9 to 2·2) 1 3 –2·6 (–8·6 to 3·4)
Participants with Clostridium 2 (2%); 8 2 (2%); 7 –0·1 (–3·8 to 3·6) 2 (2%); 7 1 (1%); 5 1·1 (–4·0 to 6·1)
difficile infection§
Survival
14 days 98·1% (1·3); 2 100·0% (0); 0 –1·9 (–4·5 to 0·7) 98·8% (1·2); 1 100·0% (0); 0 –1·2 (–3·4 to 1·1)
30 days 94·3% (2·3); 6 96·0% (2·0); 4 –1·7 (–7·6 to 4·2) 98·8% (1·2); 1 98·7% (1·3); 1 0·1 (–3·3 to 3·5)
90 days 83·6% (3·6); 17 89·0% (3·1); 11 –5·4 (–14·8 to 4·0) 92·9% (2·8); 6 94·8% (2·5); 4 –1·9 (–9·3 to 5·5)
Data are n (%), median (IQR), n (%); missing, or Kaplan-Meier estimate (SE); number of deaths, unless specified otherwise. SAB=Staphylococcus aureus bloodstream infection.
*Data are median difference (95% CI). †Complications of intravenous administrations might be caused by study drug or other intravenous medications. ‡In the intention-to-
treat population, other complications of intravenous therapy were haematoma (n=1) and inflammation at a previous peripheral catheter insertion site (n=1) in the oral
switch group and extravasation (n=1), refusal of renewed catheter placement (n=3), and clogged intravenous access (n=2) in the intravenous group. §Missing participants
were those with diarrhoea that did not undergo C difficile testing and were considered negative for statistical analysis.

Table 2: Primary and secondary outcome variables for the intention-to-treat and clinically evaluable populations

this difference was not significant. Details of all points (95% CI –14·8 to 4·0) at 90 days (see figure 2D).
participants with complications related to S aureus Differences between these secondary endpoints were not
bloodstream infection are shown in appendix 5 significant.
(pp 21–22). In subgroup analyses, the treatment difference for the
The length of hospital stay after the first positive blood primary endpoint was similar for female and male
culture was shorter in the oral switch group than in the participants (appendix 5 pp 11–12, 27). However, a higher
intravenous group, with a median stay of 12 days rate of complications related to S aureus bloodstream
(IQR 9–19) versus 16 days (10–19; median difference infection occurred in female participants than male
–2 days, 95% CI –4 to 0; p=0·043; table 2). The shorter participants (11 [17%] of 65 female participants had a
hospital stay in the oral switch group corresponds to the complication related to S aureus bloodstream infection vs
separation of the curves in the second week, as shown in 16 [11%] of 148 male participants). Yet, these differences
the Kaplan-Meier plot of the cumulative incidence of were not significant (p=0·26; appendix 5 p 27). The
discharge or death (figure 2C). Orally treated participants 90-day survival was similar between sexes, with nine
had numerically fewer complications of intravenous deaths (Kaplan-Meier estimate 85·4%, 95% CI
therapy (treatment difference –7·9 percentage points, 76·6–94·2) in female participants and 19 deaths (86·6%,
95% CI –17·6 to 1·9). The incidence of C difficile infection 80·9–92·3) in male participants. Demographic variables
was similar in both groups. Survival was numerically were also similar between female and male participants,
lower in the oral switch group than in the intravenous except for renal failure, which was present in seven (11%)
group, with a treatment difference of –1·7 percentage of 65 female participants and 42 (28%) of 148 male
points (95% CI –7·6 to 4·2) at 30 days and –5·4 percentage participants (appendix 5 pp 28–29).

www.thelancet.com/infection Published online January 17, 2024 https://doi.org/10.1016/S1473-3099(23)00756-9 7


Descargado para Anonymous User (n/a) en Austral University Faculty of Biomedical Sciences de ClinicalKey.es por Elsevier en enero 30, 2024. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
Articles

A B
0·10 Oral switch group 0·10
Intravenous group

0·08 0·08
Cumulative incidence

0·06 0·06

0·04 0·04

0·02 0·02
HR 0·74 SHR 0·74
95% CI (0·26–2·13) 95% CI (0·26–2·12)
p=0·58 p=0·57
0 0
0 30 60 90 0 30 60 90
Time to event (days) Time to event (days)
Number at risk
(censored)
Oral switch group 108 94 (10) 87 (16) 82 (21)
Intravenous group 105 93 (6) 86 (13) 82 (15)

C D
1·0 1·0

0·8 0·8
Cumulative incidence

Cumulative survival

0·6 0·6

0·4 0·4

0·2 0·2
HR 1·15 HR 1·54
95% CI (0·88–1·5) 95% CI (0·72–3·28)
p=0·32 p=0·27
0 0
0 30 60 90 0 30 60 90
Time to discharge or death (days) Time to event (days)
Number at risk
(censored)
Oral switch group 108 14 (0) 4 (0) 2 (0) 108 98 (4) 91 (5) 86 (5)
Intravenous group 105 13 (0) 2 (0) 2 (0) 105 97 (5) 90 (6) 88 (6)

Figure 2: Estimates of cumulative incidence from the date of the first positive blood culture for the intention-to-treat population
(A) Cumulative incidence of the composite primary endpoint, complications related to Staphylococcus aureus bloodstream infection, with complications defined as
relapsing S aureus infection, deep-seated S aureus infection, or death attributable to S aureus and participants with deaths not attributable to S aureus bloodstream
infection censored (Kaplan-Meier method). (B) Cumulative incidence of the composite primary endpoint, with mortality not attributable to S aureus bloodstream
infection considered a competing event (post-hoc sensitivity analysis, Fine and Gray model). (C) Cumulative incidence of hospital discharge or death. (D) Cumulative
survival (all-cause mortality, Kaplan-Meier method) with HRs estimated from Cox regression. HR=hazard ratio. SHR=subdistribution hazard ratio.

Regarding other subgroups, analyses of the primary infection than in participants with peripheral venous
outcome were similar regardless of whether the bacterial catheter infection, as well as in participants with a
isolate was meticillin susceptible or resistant, the specific comorbidity than in those with no specific
participants had different foci at baseline, they had a comorbidity (appendix 5 pp 11–12).
Charlson Comorbidity Index less than 3 or 3 or more, The length of hospital stay was longer in the
an echocardiography was performed, or they were 16 participants with MRSA bacteraemia (median 19 days
enrolled in different countries (appendix 5 pp 11–12, 26). [IQR 14–28]) than in the 197 participants with MSSA
Although the numbers of participants in the subgroups (median 15 days [9–18]; p=0·016). The Kaplan-Meier
were small, in the oral switch therapy group, higher estimate for 90-day survival in participants with MRSA
numerical rates of complications related to S aureus was lower (74·5%, SD 11·0) than in participants with
bloodstream infection occurred in participants with MSSA (87·2%, 2·4; p=0·15; appendix 5 p 26). The
central venous catheter infection or skin or soft-tissue incidence of complications of intravenous administration

8 www.thelancet.com/infection Published online January 17, 2024 https://doi.org/10.1016/S1473-3099(23)00756-9


Descargado para Anonymous User (n/a) en Austral University Faculty of Biomedical Sciences de ClinicalKey.es por Elsevier en enero 30, 2024. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
Articles

was lower in patients with MRSA (one [6%] of 16 with


Oral switch group Intravenous group p value*
MRSA vs 25 (14%) of 197 with MSSA; p=0·70; appendix 5 (n=107) (n=103)
p 26).
Participants Total Participants Total
Overall, 52 (49%) of 107 participants in the oral switch with events events with events events
group and 42 (41%) of 103 in the intravenous group
Any adverse event 52 (49%) 89 42 (41%) 61 0·27
reported an adverse event (p=0·27); 36 (34%) and
Any serious adverse event 36 (34%) 60 27 (26%) 40 0·29
27 (26%), respectively, were serious adverse events
Drug related† 3 (3%) 3 0 0 ··
(p=0·29; table 3, appendix 5 pp 23–24). The most
Infections and infestations 23 (22%) 27 17 (17%) 18 0·38
common adverse events were infections, with 45 events
Sepsis, septic shock, bacteraemia, and ∙∙ 7 ∙∙ 5 ∙∙
recorded in 40 (19%) participants (table 3, appendix 5 endocarditis
pp 30–32). The rate of serious adverse events per person
Urinary tract infection and cystitis ∙∙ 5 ∙∙ 3 ∙∙
per month was 0·21 (95% CI 0·15–0·29) in the oral
Respiratory tract infection, pneumonia, ∙∙ 4 ·· 2 ∙∙
switch group and 0·14 (0·10–0·20) in the intravenous and bronchitis
group, with a rate ratio of 1·54 (95% CI 0·95–2·50; Wound infection and skin infection ∙∙ 4 ∙∙ 2 ∙∙
p=0·079). Arthritis and osteomyelitis ∙∙ 2 ∙∙ 2 ∙∙
C difficile infection ∙∙ 1 ∙∙ 1 ∙∙
Discussion General disorders and administration site 11 (10%) 13 4 (4%) 4 0·11
In this multicentre, randomised trial in patients with conditions
low-risk S aureus bloodstream infection, an early switch Fever ∙∙ 5 ∙∙ 1 ∙∙
to oral antimicrobial therapy was non-inferior to Oedema ∙∙ 3 ∙∙ 0 ∙∙
continuing standard intravenous therapy regarding the Asthenia and general physical ∙∙ 2 ∙∙ 1 ∙∙
primary endpoint. Participants who received oral deterioration
medication were discharged from hospital earlier than Severe pain ∙∙ 2 ·· 0 ··
participants who received intravenous medication, as Cardiac disorders 9 (8%) 9 5 (5%) 5 0·41
expected from previous trials on oral switch antimicrobial Cardiac and cardiorespiratory arrest ∙∙ 4 ·· 1 ∙∙
therapy in patients with various infections.11,31 We further Cardiac failure ∙∙ 1 ·· 3 ∙∙
found that the rate of complications of intravenous Myocardial ischaemia and myocardial ∙∙ 2 ·· 0 ∙∙
administration was lower in the oral switch group, infarction
although the difference was not significant. We consider Respiratory, thoracic, and mediastinal 4 (4%) 4 10 (10%) 10 0·10
disorders
this outcome to be a true treatment effect rather than due
Pulmonary embolism ∙∙ 1 ∙∙ 2 ∙∙
to chance since with the switch to oral therapy, the
number of intravenously administered drugs that can Dyspnoea ∙∙ 1 ∙∙ 1 ∙∙

potentially cause a complication is reduced. Exacerbated chronic obstructive ∙∙ 1 ∙∙ 1 ∙∙


pulmonary disease
Survival at 90 days was numerically lower in the oral
switch group than in the intravenous group in both the Common Terminology Criteria for Adverse Events were recorded from grade 3 and above and listed by System Organ
Class and preferred term. System Organ Class terms are shown when at least 5% of all participants had a serious
intention-to-treat and clinically evaluable analyses adverse event in the respective class. Preferred terms are shown when at least two serious adverse events occurred in
(table 2), albeit not significantly. The two participants the preferred term. Preferred terms were aggregated as denoted. *Fisher’s exact test compared the number of
with death attributable to S aureus infection were in the participants with at least one event. †Drug-related serious adverse events (classified as certain or probable) were
cardiac failure, Clostridium difficile colitis, and fever. Some participants had more than one adverse event so numbers
oral switch group. Both participants had catheter-related might not add up.
S aureus infection and developed disseminated infection
within 3 weeks after starting study medication. Table 3: Adverse events in the safety population occurring within 90 days
One participant was discharged and declined readmission
when the general condition deteriorated. This finding
highlights the importance of monitoring patients with that the oral medication affects outcome, although oral
S aureus bloodstream infection closely for developing regimens are generally considered to have a favourable
complications. safety profile. Nevertheless, future trials should confirm
We further observed a higher incidence of adverse equivalence of oral therapy regarding all-cause mortality.
events and serious adverse events in the oral switch When comparing subgroups of male and female
group than in the intravenous group, although the participants, we found a similar treatment difference for
difference was not significant and the overall incidence the primary outcome and similar 90-day survival.
was in the range of previous trials.32,33 A large proportion However, the observed higher rate of complications
of adverse events were infections not caused by S aureus, related to S aureus bloodstream infection in female
which underscores that the participant population is participants than male participants might warrant
generally at risk for infections. The numerically lower further studies.
survival and the concordant higher incidence of adverse Assigning the low-risk category to patients with
events in the oral switch group were not significant and, S aureus bloodstream infection can be challenging. In
thus, might be due to chance alone. We cannot exclude a randomised controlled trial in patients with

www.thelancet.com/infection Published online January 17, 2024 https://doi.org/10.1016/S1473-3099(23)00756-9 9


Descargado para Anonymous User (n/a) en Austral University Faculty of Biomedical Sciences de ClinicalKey.es por Elsevier en enero 30, 2024. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
Articles

uncomplicated staphylococcal bacteraemia, a third of Due to the relatively low rates of MRSA in participating
patients without suspected metastatic infection at countries, we enrolled only 16 participants with MRSA
enrolment were later diagnosed with complicated infection. This raises the question of whether the results
S aureus bloodstream infection.33 14 (7%) of 213 patients are generalisable to patients with low-risk bloodstream
who met our criteria for low-risk S aureus bloodstream infection due to MRSA. As in many studies, we found a
infection at enrolment had complications related to lower 90-day survival in participants with MRSA than
S aureus infection (table 2), of which more than half MSSA.2 It is important to note that the interpretation of
became apparent during the intervention phase (ie, the primary endpoint did not change when restricting
during the second week of illness). These early the analysis to MSSA only.
complications might have been present at baseline and, The results should not be generalised to other oral
unlike in our study, it has been proposed to consider medications or to other populations, such as injection
complications occurring within 5–7 days post-enrolment drug users, who were not included in the study due to an
as a baseline condition.34 When we excluded early elevated risk for the presence or development of
complications from the primary endpoint in a sensitivity complicated infection, although these populations might
analysis, we showed non-inferiority (appendix 5 be perceived as good candidates for oral antimicrobial
pp 13–14). therapy. It is important to note that we included more
The strengths of this study are the pragmatic approach patients with peripheral venous catheter infection than
with a globally available oral medication as the first with central venous catheter infection, although central
choice, the strict definition of low-risk S aureus venous catheter infection is usually a more frequent
bloodstream infection, and the choice of a composite focus in S aureus bloodstream infection.37 We assume
primary endpoint specific to S aureus bloodstream that this difference was driven by our selection of a low-
infection. The trial has several limitations. It was open risk population, since patients that require central
label since intravenous administration of placebo in the venous access will have a higher risk of comorbidities
oral switch group would have interfered with the aim of that might lead to exclusion.
facilitating early discharge. This study design poses a risk Misclassification of low-risk S aureus bloodstream
of bias from investigators when assessing outcomes and infection might impede safe implementation of the
safety endpoints. To mitigate this bias, the clinical review findings into clinical practice. A more aggressive
committee, who were masked to treatment assignment, diagnostic approach (eg, involving fluorodeoxyglucose-
assessed the outcomes. In an open-label design, patients PET or CT),38 a more stringent evaluation of the need for
might also introduce bias (eg, by withdrawing informed echocardiography (eg, the VIRSTA study score),39 or
consent when not randomised into the desired study novel biomarkers40 might identify and predict deep-
group). However, as two participants from each group seated infections more accurately. Nevertheless, careful
withdrew informed consent, the risk of bias was low. assessment of the individual, preferably by consulting an
The large number of patients screened raises the infectious diseases expert, is necessary to correctly assign
question of whether low-risk S aureus bloodstream a patient to the low-risk group, and close monitoring
infection is clinically relevant. However, screening-to- for early detection of complications and potential
enrolment ratios above 1:25 are typical for trials on prolongation of antimicrobial therapy are advised.
uncomplicated S aureus bloodstream infection;34 In summary, we found that an early oral switch was
upcoming platform trials might allow more efficient non-inferior to standard intravenous antimicrobial
recruitment.35 A study conducted in one of the therapy in patients with low-risk S aureus bloodstream
participating centres found that the proportion of infection. This study supports an early switch to oral
patients with multiple risk factors for complicated or antimicrobial therapy in patients with low-risk S aureus
difficult-to-treat S aureus bloodstream infection increased bloodstream infection provided a rigorous clinical
over 14 years,36 suggesting that low-risk S aureus assessment and close monitoring for complications are
bloodstream infection has decreased in prevalence. The done. Future studies should address other subgroups of
reduced recruitment rate could not be compensated for patients with S aureus bloodstream infection and confirm
by increasing the capacity of the clinical network and led the benefits of early oral switch in a detailed medico-
to early termination of the trial. To this end, the planned economic analysis.
interim analysis was converted into the final analysis, SABATO study group
which increased the attainable non-inferiority margin Adoración Valiente, Marina de Cueto (Hospital Universitario Virgen
to 10%. Macarena, Seville, Spain), Ángel Rodríguez, José Molina (Hospital
Universitario Virgen del Rocío, Seville, Spain), Julia Fischer (University
Generally, a combined endpoint is considered less of Münster, Germany), Gregor Paul (University of Cologne, Cologne,
robust than a single endpoint. However, this design Germany) Reinhild Prinz-Langenohl (Clinical Trials Centre Cologne,
decision was made consciously to reflect sequelae that Cologne, Germany), Gabriele Peyerl-Hoffmann, Daniel Hornuß
are specific to S aureus bloodstream infection and to (University Medical Centre Freiburg, Freiburg, Germany), Sébastien
Gallien, Vincent Fihman (Henri-Mondor University Hospital, AP-HP,
ensure that the required sample size could be handled by Creteil, France), Marion Lacasse, Francois Coustillères (Centre
the clinical network. Hospitalier Régional Universitaire de Tours, Tours, France),

10 www.thelancet.com/infection Published online January 17, 2024 https://doi.org/10.1016/S1473-3099(23)00756-9


Descargado para Anonymous User (n/a) en Austral University Faculty of Biomedical Sciences de ClinicalKey.es por Elsevier en enero 30, 2024. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
Articles

Christian Becker (Helios Klinikum Krefeld, Krefeld, Germany), publication. Data will be shared for any analysis with researchers who
André Fuchs (University Hospital of Augsburg, Augsburg, Germany), submit a methodologically sound proposal, which is approved by the
Laura Morata (Hospital Clínic, Barcelona, Spain), Sebastian Weis (Jena ethics committee at the institution of the corresponding author.
University Hospital—Friedrich Schiller University Jena, Jena, Germany), Requests should be directed to the corresponding author of the Article.
Diane Ponscarme (Saint-Louis Hospital, Paris, France), To gain access, a data sharing agreement will need to be signed. Data
Lydie Khatchatourian (Centre Hospitalier de Cornouaille, Quimper, will then be available from the University data warehouse.
France), Elisabeth Rouveix (Ambroise Paré University Hospital,
Acknowledgments
Boulogne-Billancourt, France), Dominique Merrien (Centre Hospitalier
AJK received funding for this trial from Deutsche
Départmentale Vendée, La Roche-sur-Yon, France), Raphaël Lecomte
Forschungsgemeinschaft (217917502). We thank the participants and staff
(Centre Hospitalier Universitaire de Nantes, Nantes, France),
from all sites who contributed to the trial (appendix 5 pp 3–4). We thank
Jacobien Veenemans (Amphia Hospital Breda, CK Breda, Netherlands),
Vance G Fowler, Stephan Harbarth, and Guy Thwaites for the expert
Helem H Vilchez (Fundació Institut d’Investigació Sanitària Illes
opinion in the trial steering committee; Vincent LeMoing,
Balears, Palma de Mallorca, Spain), Violaine Tolsma (Centre Hospitalier
Miquel Pujol Rojo, and Estée Török for participating in the clinical review
d’Annecy Genevois, France), Johanna Kessel (University Hospital
committee; and Walter E Haefeli, Alexandra Heininger, and
Frankfurt, Frankfurt, Germany), Marc J M Bonten (University Medical
Geraldine Rauch for participating in the data monitoring committee.
Center Utrecht, Utrecht, Netherlands), Jan Rupp (University Hospital of
This trial was supported by Clinical Trials Centre Cologne (Medical
Schleswig Holstein, Luebeck, Germany), Laurent Hocqueloux (Centre
Faculty, University of Cologne) with regard to project management,
Hospitalier Régional, Orléans, France), Frederic Lucht (Centre
database development, data management, monitoring, and safety
Hospitalier Universitaire de Saint-Étienne, France), Jean-Paul Stahl
management.
(Centre Hospitalier Universitaire Grenoble, Grenoble, France),
Anne L M Vlek (Diakonessenhuis, Utrecht, Netherlands). References
1 Kern WV, Rieg S. Burden of bacterial bloodstream
Contributors infection—a brief update on epidemiology and significance of
AJK was the coordinating principal investigator and sponsor’s multidrug-resistant pathogens. Clin Microbiol Infect 2020;
representative. GF served as the clinical trial lead until July, 2016. 26: 151–57.
AJK, HSe, WVK, SRi, and MH conceived and designed the study. 2 Bai AD, Lo CKL, Komorowski AS, et al. Staphylococcus aureus
AJK, AR, and MN managed the trial. AJK, LEL-C, JR-B, JMC, MDN, GF, bacteraemia mortality: a systematic review and meta-analysis.
NJ, WVK, SRi, RL, LC, LB, AL, KK, CRM, AS, SH, BF, ML, J-PT, AD, Clin Microbiol Infect 2022; 28: 1076–84.
TG, DB, TW, SRe, JK, MLM, EF, HSt, VV, and PT were site investigators 3 Kaasch AJ, Barlow G, Edgeworth JD, et al. Staphylococcus aureus
involved in participant recruitment and data collection. MH, AA, and bloodstream infection: a pooled analysis of five prospective,
AJK developed the statistical analysis plan. AJK, AR, AA, and observational studies. J Infect 2014; 68: 242–51.
MH curated the data. MH and AA had access to the data in the study 4 Kaasch AJ, Kern WV, Joost I, Hellmich M, Seifert H, Rieg S. Effect
and conducted the formal statistical analysis. AJK, MH, and AA verified of clinically uninfected orthopedic implants and pacemakers/
the data with the clinical review committee and the monitoring team. AICDs in low-risk Staphylococcus aureus bloodstream infection on
AJK, MH, AA, and HSe wrote the first draft of the manuscript. crude mortality rate: a post hoc analysis of a large cohort study.
All authors had full access to all the data in the study and had final Open Forum Infect Dis 2019; 6: ofz170.
responsibility for the decision to submit for publication. 5 Bupha-Intr O, Blackmore T, Bloomfield M. Efficacy of early oral
switch with β-lactams for low-risk Staphylococcus aureus bacteremia.
Declaration of interests Antimicrob Agents Chemother 2020; 64: e02345–19.
AJK received funding for this study from the Deutsche 6 Willekens R, Puig-Asensio M, Ruiz-Camps I, et al. Early oral switch
Forschungsgemeinschaft and is Chairperson of the German Sepsis to linezolid for low-risk patients with Staphylococcus aureus
Society. HSe received grants or research support from the bloodstream infections: a propensity-matched cohort study.
Bundesministerium für Bildung und Forschung (BMBF) Germany and Clin Infect Dis 2019; 69: 381–87.
the German Center for Infection Research, and has been a consultant 7 Liu C, Bayer A, Cosgrove SE, et al. Clinical practice guidelines by
for Debiopharm, Gilead, MSD, and Shionogi. AS has received grants the Infectious Diseases Society of America for the treatment of
from Gilead Sciences (IN-ES-540–6089) and Pfizer, consulting fees and methicillin-resistant Staphylococcus aureus infections in adults and
lecture honoraria from Pfizer, MSD, Angelini, Shionogi, Gilead, and children. Clin Infect Dis 2011; 52: e18–55.
Menarini, and travel support from Pfizer. JR-B has received grants or 8 Gudiol F, Aguado JM, Almirante B, et al. Executive summary of the
research support from the Spanish Network for Research in Infectious diagnosis and treatment of bacteremia and endocarditis due to
Diseases (REIPI RD16/0016/00001) and Centro de Investigación Staphylococcus aureus. A clinical guideline from the Spanish Society
of Clinical Microbiology and Infectious Diseases (SEIMC).
Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC,
Enferm Infecc Microbiol Clin 2015; 33: 626–32.
CB21/13/00012), Instituto de Salud Carlos III, Spanish Ministry of
9 Brown NM, Goodman AL, Horner C, Jenkins A, Brown EM.
Science and Innovation, which are co-financed by the European
Treatment of methicillin-resistant Staphylococcus aureus (MRSA):
Development Regional Fund. SH received honoraria and travel support updated guidelines from the UK. JAC Antimicrob Resist 2021;
from Shionogi, Pfizer, Infectopharm, and AdvanzPharma. NJ reports 3: dlaa114.
receiving honoraria for lectures from AbbVie, Bayer, Infectopharm, and 10 Dagher M, Fowler VG Jr, Wright PW, Staub MB. A narrative review
Medacta, travel support from Gilead, Pfizer, and Correvio, participation of early oral stepdown therapy for the treatment of uncomplicated
in paid advisory board meetings for MSD, and membership in the Staphylococcus aureus bacteremia: yay or nay? Open Forum Infect Dis
steering committee of the German Society of Internal Medicine. 2020; 7: ofaa151.
SRi received honoraria for lectures from Falk Foundation, Pfizer, 11 Iversen K, Ihlemann N, Gill SU, et al. Partial oral versus
bioMérieux, Akademie für Infektionsmedizin, Med Update, intravenous antibiotic treatment of endocarditis. N Engl J Med 2019;
streamedup!, and Deutscher Apotheker-Verlag. TW reports receiving 380: 415–24.
honoraria for presentations from AstraZeneca, Advanz, MSD, Pfizer, 12 Li H-K, Rombach I, Zambellas R, et al. Oral versus intravenous
and Shionogi, grants or research support from BMBF Germany, and antibiotics for bone and joint infection. N Engl J Med 2019;
travel support from Pfizer. All other authors declare no competing 380: 425–36.
interests. 13 Jorgensen SCJ, Lagnf AM, Bhatia S, Shamim M-D, Rybak MJ.
Sequential intravenous-to-oral outpatient antibiotic therapy for
Data sharing MRSA bacteraemia: one step closer. J Antimicrob Chemother 2019;
Individual participants’ data (including the data dictionary) that 74: 489–98.
underlie the results reported in this Article will be shared on request 14 Kouijzer IJE, van Leerdam EJ, Gompelman M, et al. Intravenous to
after de-identification. The study protocol, statistical analysis plan, and oral switch in complicated Staphylococcus aureus bacteremia without
informed consent form will also be shared. Data will be available endovascular infection: a retrospective single-center cohort study.
12 months after publication and ending 36 months following Clin Infect Dis 2021; 73: 895–98.

www.thelancet.com/infection Published online January 17, 2024 https://doi.org/10.1016/S1473-3099(23)00756-9 11


Descargado para Anonymous User (n/a) en Austral University Faculty of Biomedical Sciences de ClinicalKey.es por Elsevier en enero 30, 2024. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.
Articles

15 Mun SJ, Kim S-H, Huh K, et al. Oral step-down therapy in patients 28 European Medicines Agency. Points to consider on switching
with uncomplicated Staphylococcus aureus primary bacteremia and between superiority and non-inferiority. July 27, 2000. https://www.
catheter-related bloodstream infections. J Chemother 2022; ema.europa.eu/en/documents/scientific-guideline/points-consider-
34: 319–25. switching-between-superiority-and-non-inferiority_en.pdf (accessed
16 Yeager SD, Oliver JE, Shorman MA, Wright LR, Veve MP. Dec 21, 2023).
Comparison of linezolid step-down therapy to standard parenteral 29 European Medicines Agency. Guideline on the evaluation of
therapy in methicillin-resistant Staphylococcus aureus bloodstream medicinal products indicated for treatment of bacterial infections—
infections. Int J Antimicrob Agents 2021; 57: 106329. revision 3. May 19, 2022. https://www.ema.europa.eu/en/
17 Pérez-Rodríguez MT, Sousa A, Moreno-Flores A, et al. The benefits documents/scientific-guideline/guideline-evaluation-medicinal-
and safety of oral sequential antibiotic therapy in non-complicated products-indicated-treatment-bacterial-infections-revision-3_en.pdf
and complicated Staphylococcus aureus bacteremia. Int J Infect Dis (accessed Dec 21, 2023).
2021; 102: 554–60. 30 Fine JP, Gray RJ. A proportional hazards model for the
18 Itoh N, Hadano Y, Saito S, Myokai M, Nakamura Y, Kurai H. subdistribution of a competing risk. J Am Stat Assoc 1999;
Intravenous to oral switch therapy in cancer patients with catheter- 94: 496–509.
related bloodstream infection due to methicillin-sensitive 31 Schrenzel J, Harbarth S, Schockmel G, et al. A randomized clinical
Staphylococcus aureus: a single-center retrospective observational trial to compare fleroxacin–rifampicin with flucloxacillin or
study. PLoS One 2018; 13: e0207413. vancomycin for the treatment of staphylococcal infection.
19 Diego-Yagüe I, Mora-Vargas A, Vázquez-Comendador JM, et al. Clin Infect Dis 2004; 39: 1285–92.
Sequential oral antibiotic in uncomplicated Staphylococcus aureus 32 Thwaites GE, Scarborough M, Szubert A, et al. Adjunctive
bacteraemia: a propensity-matched cohort analysis. rifampicin for Staphylococcus aureus bacteraemia (ARREST):
Clin Microbiol Infect 2023; 29: 744–50. a multicentre, randomised, double-blind, placebo-controlled trial.
20 Liu C, Strnad L, Beekmann SE, Polgreen PM, Chambers HF. Lancet 2018; 391: 668–78.
Clinical practice variation among adult infectious disease 33 Holland TL, Raad I, Boucher HW, et al. Effect of algorithm-based
physicians in the management of Staphylococcus aureus bacteremia. therapy vs usual care on clinical success and serious adverse events
Clin Infect Dis 2019; 69: 530–33. in patients with staphylococcal bacteremia: a randomized clinical
21 Hospenthal DR, Waters CD, Beekmann SE, Polgreen PM. Practice trial. JAMA 2018; 320: 1249–58.
patterns of infectious diseases physicians in transitioning from 34 Holland TL, Chambers HF, Boucher HW, et al. Considerations for
intravenous to oral therapy in patients with bacteremia. clinical trials of Staphylococcus aureus bloodstream infection in
Open Forum Infect Dis 2019; 7: ofz386. adults. Clin Infect Dis 2019; 68: 865–72.
22 Kaasch AJ, Fätkenheuer G, Prinz-Langenohl R, et al. Early oral 35 Tong SYC, Mora J, Bowen AC, et al. The Staphylococcus aureus
switch therapy in low-risk Staphylococcus aureus bloodstream Network Adaptive Platform trial protocol: new tools for an old foe.
infection (SABATO): study protocol for a randomized controlled Clin Infect Dis 2022; 75: 2027–34.
trial. Trials 2015; 16: 450. 36 Mathé P, Göpel S, Hornuss D, et al. Increasing numbers and
23 Kaasch AJ, Rommerskirchen A, Hellmich M, et al. Protocol update complexity of Staphylococcus aureus bloodstream infection—14 years
for the SABATO trial: a randomized controlled trial to assess early of prospective evaluation at a German tertiary care centre with
oral switch therapy in low-risk Staphylococcus aureus bloodstream multi-centre validation of findings. Clin Microbiol Infect 2023;
infection. Trials 2020; 21: 175. 29: 1197.e9–15.
24 Bone RC, Balk RA, Cerra FB, et al. Definitions for sepsis and organ 37 Nambiar K, Seifert H, Rieg S, et al. Survival following
failure and guidelines for the use of innovative therapies in sepsis. Staphylococcus aureus bloodstream infection: a prospective
Chest 1992; 101: 1644–55. multinational cohort study assessing the impact of place of care.
25 US Department of Health and Human Services, National Institutes J Infect 2018; 77: 516–25.
of Health, National Cancer Institute. Common terminology criteria 38 Ghanem-Zoubi N, Kagna O, Abu-Elhija J, et al. Integration of
for adverse events Version 4.0. Bethesda, MD: National Cancer FDG-PET/CT in the diagnostic workup for Staphylococcus aureus
Institute, 2009. bacteremia: a prospective interventional matched-cohort study.
26 Zhao G. Tests of non-null hypothesis on proportions for stratified Clin Infect Dis 2021; 73: e3859–66.
data. Stat Med 2008; 27: 1429–46. 39 Tubiana S, Duval X, Alla F, et al. The VIRSTA score, a prediction
27 European Medicines Agency. Guideline on the evaluation of score to estimate risk of infective endocarditis and determine
medicinal products indicated for treatment of bacterial infections— priority for echocardiography in patients with Staphylococcus aureus
revision 2. Dec 15, 2011. https://www.ema.europa.eu/en/ bacteremia. J Infect 2016; 72: 544–53.
documents/scientific-guideline/guideline-evaluation-medicinal- 40 Cao Y, Guimaraes AO, Peck MC, et al. Risk stratification biomarkers
products-indicated-treatment-bacterial-infections-revision-2_en.pdf for Staphylococcus aureus bacteraemia. Clin Transl Immunology 2020;
(accessed Dec 21, 2023). 9: e1110.

12 www.thelancet.com/infection Published online January 17, 2024 https://doi.org/10.1016/S1473-3099(23)00756-9


Descargado para Anonymous User (n/a) en Austral University Faculty of Biomedical Sciences de ClinicalKey.es por Elsevier en enero 30, 2024. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2024. Elsevier Inc. Todos los derechos reservados.

You might also like