Skip to main content
Erschienen in: Infection 3/2016

Open Access 11.04.2016 | Guideline

Strategies to enhance rational use of antibiotics in hospital: a guideline by the German Society for Infectious Diseases

verfasst von: K. de With, F. Allerberger, S. Amann, P. Apfalter, H.-R. Brodt, T. Eckmanns, M. Fellhauer, H. K. Geiss, O. Janata, R. Krause, S. Lemmen, E. Meyer, H. Mittermayer, U. Porsche, E. Presterl, S. Reuter, B. Sinha, R. Strauß, A. Wechsler-Fördös, C. Wenisch, W. V. Kern

Erschienen in: Infection | Ausgabe 3/2016

insite
INHALT
download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN

Abstract

Introduction

In the time of increasing resistance and paucity of new drug development there is a growing need for strategies to enhance rational use of antibiotics in German and Austrian hospitals. An evidence-based guideline on recommendations for implementation of antibiotic stewardship (ABS) programmes was developed by the German Society for Infectious Diseases in association with the following societies, associations and institutions: German Society of Hospital Pharmacists, German Society for Hygiene and Microbiology, Paul Ehrlich Society for Chemotherapy, The Austrian Association of Hospital Pharmacists, Austrian Society for Infectious Diseases and Tropical Medicine, Austrian Society for Antimicrobial Chemotherapy, Robert Koch Institute.

Materials and methods

A structured literature research was performed in the databases EMBASE, BIOSIS, MEDLINE and The Cochrane Library from January 2006 to November 2010 with an update to April 2012 (MEDLINE and The Cochrane Library). The grading of recommendations in relation to their evidence is according to the AWMF Guidance Manual and Rules for Guideline Development.

Conclusion

The guideline provides the grounds for rational use of antibiotics in hospital to counteract antimicrobial resistance and to improve the quality of care of patients with infections by maximising clinical outcomes while minimising toxicity. Requirements for a successful implementation of ABS programmes as well as core and supplemental ABS strategies are outlined. The German version of the guideline was published by the German Association of the Scientific Medical Societies (AWMF) in December 2013.
Hinweise
AWMF (Association of the Scientific Medical Societies in Germany) Registry No. 092/001.
S3-Guideline of the German Society for Infectious diseases Reg.Soc. (Deutsche Gesellschaft für Infektiologie e.V., DGI) in association with the following professional societies/associations/institutions:
German Society of Hospital Pharmacists (ADKA).
German Society for Hygiene and Microbiology (DGHM).
Paul Ehrlich Society for Chemotherapy (PEG).
The Austrian Association of Hospital Pharmacists (AAHP).
Austrian Society for Infectious Diseases and Tropical Medicine (ÖGIT).
Austrian Society for Antimicrobial Chemotherapy (ÖGACH).
Robert Koch Institute (RKI), Berlin.
External reviewer: Prof. Dr. Stephan Harbarth, Genf.
Patient representative: Viktoria Mühlbauer, Düsseldorf.
Editor of the English version: Sina Helbig, Dresden.
H. Mittermayer: Deceased.

Introduction and aims of the guideline

The dramatic increase in antibiotic resistance seen in many areas and regions combined with the paucity of new drug development more than ever calls for prudent, controlled, and appropriate use of antiinfectives in all areas of medicine. This affects almost all disciplines and medical specialties. The density of antiinfective treatment—with all its implications for cost, toxicity, the emergence of resistance and recommendations on diagnosis and follow-up, as well as recommendations on further therapy in the outpatient setting—is so high, in particular in the hospital sector, that safety and quality assurance processes will no longer succeed without a panel of experts and strategic discussions. Following a first position paper of the European Commission in 2001, in a second report on “Prudent use of antimicrobial agents in human medicine” published in 2010, EU Member States were recommended to establish or enhance surveillance systems for antibiotic resistance and antibiotic consumption. Particular importance gains this recommendation in Germany in light of the amendment of the Infection Protection Act [Infektionsschutzgesetz (IfSG), especially §4 and §23] in July 2011. The Act not only stipulates collection of data on antibiotic consumption, pathogenic microorganisms and resistance, it also requires that data on antibiotic consumption be assessed taking into account the local resistance situation, and that appropriate conclusions be drawn regarding the use of antibiotics. Furthermore, that the necessary adjustments to antibiotic consumption be communicated to staff and implemented (IfSG § 23 paragraph 4). Antibiotic stewardship (ABS) programmes should and can assume this responsibility in combination with policies and programmes for infection prevention. The aim of ABS programmes in hospital is to continuously improve the quality of antiinfective prescribing with regard to agent selection, dosing, administration and duration of treatment in order to maximise clinical outcomes while minimising toxicity to the patient as well as the emergence of resistance and costs.
Many reviews published since 2005 [112] on antibiotic stewardship describe the requirements and elements needed to institutionalise this type of programme in hospitals. More recent publications also detail use of these programmes in intensive care units [1315], paediatrics [1618] or small community hospitals [1921]. A relatively new systematic review on ABS activities in critical care medicine, assesses 24 studies between 1996 and 2010, among them six methodologically ambitious investigations [15]. These were projects limiting cephalosporin use to minimise the emergence of resistance, studies on the implementation of computerised decision support systems and infectious diseases consultation services, as well as introduction of new guidelines for therapy and prophylaxis. The review provides good insight into the effects of various ABS strategies on consumption, costs and resistance, as demonstrated in recent years by a number of other original papers. Most studies show a 10–40 % reduction in antiinfective drug use, shorter treatment duration and cost reduction. Programmes that were active for longer than 6 months were also associated with an improvement in resistance rates depending on the drug–pathogen combination [15]. A recent Cochrane review of 2013 (89 studies up to 2007) reached a similar conclusion. The review shows that the effect of interventions (e.g. antimicrobial restriction) is usually delayed (6 months) in respect of microbiological endpoints (e.g. antibiotic resistance); however, a prompt effect (frequently as soon as 1 month) is noted with regard to prescribing endpoints. According to the meta-analysis of methodologically robust studies (including randomised and controlled before-after studies with interrupted time-series analyses), professional interventions to reduce excessive antiinfective prescribing are successful in minimising the emergence of resistance and reducing hospital-acquired infections, as well as in improving individual treatment outcomes [22].
The two most recent reviews mentioned demonstrate the importance of ABS programmes and rational prescribing strategies in terms of minimising resistance. Instituting ABS programmes to save costs is not any more the driving factor, although this aspect is still important. An analysis published in 2012 on the cost-effectiveness of an ABS programme initiated at a University Hospital in Maryland, USA, showed interesting results. Over a period of 3 years, gross savings of roughly USD 3 million were realised, i.e. approximately USD 1 million/year. This was opposed by expenses of roughly USD 200,000/year to finance the programme (personnel costs). Although yearly cost savings dropped to approximately USD 400,000 p.a. over the full 7-year term of the ABS programme, it nevertheless remained cost-effective and delivered net savings with only one full-time personnel per 500 beds (infectious diseases physician, pharmacist, IT specialist). When the programme was discontinued after 7 years, antiinfective costs rapidly rose by around USD 2 million during the subsequent 2 years [23]. Cost–benefit analyses performed in other more recent pharmacoeconomic investigations of ABS programmes are no longer limited to the potential “savings” achieved in drug and material costs; rather they show that adequate antiinfective therapy is associated with lower mortality, shorter length of hospital stay and duration of treatment, and that it can reduce the overall cost of treatment and improve patient safety [24].
This guideline recommends and outlines the requirements and main elements of ABS programmes with which the above objectives can be achieved. The recommendations are based on a systematic evaluation of many new observational and interventional studies with clinical and microbiological endpoints, as well as the endpoints antiinfective prescribing and costs, which were mainly conducted in adult patients in acute-care hospitals. The available literature was compiled based on the guideline published by two American societies (IDSA, SHEA) focusing on the development of facility-specific ABS programmes (“Guidelines for Developing an Institutional Program to Enhance Antimicrobial Stewardship”) as well as on a Cochrane Review by Davey et al. from 2005 on “Interventions to improve antibiotic prescribing practices for hospital inpatients (Review)”, taking into account its update (2013) [2, 6, 22, 25]. Further literature was systematically searched until 15 April 2012 and evaluated. For details see the methodology report published online (http://​www.​awmf.​org). Although some of the recommendations are not new in content, they altogether draw on much better study evidence and a greater number of examples for successful programmes. The recommendations were derived by consensus by the guideline development group based on review of the literature, taking into account relevance, evidence, applicability and practicability in German and Austrian acute-care hospitals. Key challenges are the current trends in multidrug-resistant pathogens (VRE, multidrug-resistant Gram-negative bacteria) and Clostridium difficile in Germany and Austria, the lack of skilled personnel—especially infectious diseases physicians—and limited experience with well-functioning infectious disease consultation services established elsewhere, increasing cost pressure in hospitals and outsourcing of microbiological diagnostics.
For the purpose of safety and quality assurance, it is recommended to use a selection of indicators from a catalogue developed and agreed upon by members of the guideline development group and users in Germany. Further experience with validation especially of process indicators as well as international experience gained in particularly France, England and Scotland on use of such indicators for internal and external quality assurance should be taken into account.

Summary of recommendations

Requirements

Availability of a team of ABS experts

For effective implementation of ABS programmes, it is essential that a multidisciplinary team should be instructed by the hospital administration and allocated with adequate resources to draw up guidelines derived by consensus with the users for the treatment of infectious diseases and to ensure their implementation through ABS strategies (A).
The team should consist of at least one infectious diseases physician (or clinician with infectious diseases training) and an experienced clinical pharmacist/hospital pharmacist, as well as a specialist in microbiology, virology and infection epidemiology being responsible for laboratory diagnostic and microbiological consultation; furthermore, the physician locally responsible for infection control. The team members should either have appropriate training in antibiotic stewardship or already be sufficiently experienced (A).
The team will receive the support and collaboration of the hospital administration, and activities within the ABS programme should be compensated with a minimum of one full-time equivalent (FTE) of 0.5 per 250 beds (A). There should be good collaboration between the Therapeutics and Drugs Committee, Hospital Infection Control Committee, pharmacy and representatives of clinical divisions/departments (ABS representatives), for which purpose the team should issue its own Rules of Procedure (A).
Significance in practice:
  • ABS programmes should be instituted facility-wide which necessitates a multidisciplinary team with the competence for interdisciplinary cooperation.
  • Infectious diseases specialists serving as consultants improve the clinical outcome of patients with infections, and ensure the quality of drug prescribing.
  • Clinical pharmacists improve the quality of drug prescribing (e.g. dosing and drug application, avoidance of adverse drug events).
  • Microbiologists facilitate high-grade infection medicine by ensuring the quality of microbiological diagnostics and preanalytics, and by expertly evaluating and conveying microbial culture results.
  • Various ABS programmes describe an FTE of 0.5 per 250 beds as being the minimum staff resources necessary to cost-effectively conduct an ABS programme.

Availability of surveillance data on pathogens, resistance, and antimicrobial consumption

Pathogens and resistance
Antimicrobial susceptibility data on major pathogens should be available and accessible at least yearly on a hospital-wide level and separately for general and intensive care units, or department-specific, as the case may be. Data on primary isolates should be shown by pathogen and type of specimen, e.g. blood, urine, miscellaneous samples. Culture results from screening tests should be shown separately. Susceptibility rates should indicate the number of isolates tested. Infection rates should relate consistently to a single denominator (e.g. patient-days/number of cases). Participation in an established surveillance system is recommended (A).
Significance in practice:
  • Conducting an additional material analysis (e.g. number of blood culture sets per patient or 1000 patient-days, number of urine cultures per patient, number of catheter-associated urine cultures, etc.) also with regard to material quality and positivity/contamination rates (e.g. blood cultures) can be useful.
  • Whether the susceptibility rates of pathogens should be limited to agents listed on the hospital formulary should be discussed within the team.
  • The amendment of the Infection Protection Act (Infektionsschutzgesetz, IfSG) (reporting, documentation) is mandatory.
Antimicrobial consumption
Data on antimicrobial consumption, expressed as use density (daily doses per 100 patient-days) should be collected at least annually or preferably quarterly and are generally reported by the pharmacist. Data are reported institution-wide, at the ward level as well as for individual (speciality) departments. On demand, data should be broken down to the agent level and should be provided to the ABS team. Participation in an established surveillance system is recommended (A).
Point prevalence surveys should be conducted for systematic quantitative and qualitative assessment of antiinfective use, and, if required should be reevaluated short-term (A). Antiinfective use data are collected at the patient level which allows to assess prescribing quality based on indication and type of infection, and to recognise the need for targeted ABS strategies. Access to patient-level data ought to be guaranteed.
Significance in practice:
  • Use density should be presented by antibiotic class and not only by individual agent.
  • Reporting consumption data and antiinfective costs ranked by individual agent or class (e.g. top 5 or 10) is also reasonable.
  • Point prevalence surveys are a simple tool to examine process quality.
  • The amendment of the Infection Protection Act (Infektionsschutzgesetz, IfSG) (reporting, documentation) must be observed.

ABS core strategies

Application of local treatment guidelines/pathways, hospital antiinfective formulary, formulary restrictions and approval requirements

Developing and updating local treatment guidelines, clinical pathways, and an antiinfective formulary is one of the ABS team’s chief responsibilities. The antiinfective formulary should be based on national and international guidelines as well as on the local/regional pathogen and resistance patterns, and possibly drug costs. Drugs on the antiinfective formulary should be categorised according to recommended versus reserve or special compounds. In addition, these should be tagged with special prescription status and be subject to approval and preauthorisation requirements. The antiinfective formulary is updated at least yearly based on therapy guidelines and whenever necessary and approved by the Therapeutics and Drugs Committee (A).
Adherence to guidelines regarding substance selection, dosing, route and duration of treatment may improve clinical outcome in terms of mortality, as well as treatment duration and length of hospital stay. To ensure adherence, users should be involved in developing the guidelines and be educated through audits of antiinfective use or antiinfective point-of-care chart reviews (A).
Individualising antiinfective prescriptions with or without special approval requirements improves targeted therapy and reduces inappropriate treatment. Various possibilities for implementation have been described and should be used, from simple antimicrobial order forms to highly differentiated antiinfective request forms that may be subject to specific time limits or limited to certain hospital areas (A). Guideline-based antiinfective drug use or use of individual defined substances can be controlled by this means, thus minimising consumption, costs and adverse drug events.
Restricting whole substance classes can—by shifting to an alternative substance—prove to be an effective strategy for controlling nosocomial infections and the development of critical resistance levels; accordingly, antiinfective restriction ought to be targeted (B). At the same time, routine surveillance of antibiotic consumption and locally prevalent pathogens and their susceptibility patterns should be performed to detect possible adverse effects of the strategy in time (A).
Significance in practice:
  • Local guidelines serve quality assurance and are a core strategy of every ABS programme.
  • The antiinfective formulary is a useful ABS tool especially in small and medium-sized hospitals.
  • Clinical pathways are rarely employed, can, however, be very helpful in the emergency room.
  • Special order forms are highly effective ABS tools. Efforts should be undertaken to foster acceptance by prescribers.
  • Restrictions on use to control resistance and nosocomial infections are frequently only temporarily effective. They should be time-restricted by the ABS team and reconsidered depending on the effects.

Design and implementation of education, training and information

Targeted education, training and information are essential elements of any ABS programme. They provide the foundation of knowledge needed to promote more rational use of antibiotics and reasonable microbiological diagnostic, and to improve acceptance of ABS programmes. They have the objective of optimising the therapeutic and diagnostic management of patients with infection through greater adherence to recommendations. They should preferably take place as an active training measure rather than in the form of passive communication of information (A).
Education, training and information in different formats and on various topics should be offered repeatedly as they are not sustainable as a one-off measure. They should be organised in agreement and integration with local ABS programmes (A).
Education, training and information should be independent of commercial interests, whereby the hospital administration is responsible for implementing and financing the measures (A).
Significance in practice:
  • The target group for local training and educational sessions should be clearly defined.
  • The handling of conflicts of interest should be laid down in writing (Rules of Procedure) by the ABS team.
  • Informative meetings and educational/training sessions should give special attention to a critical evaluation of published study results.

Conducting proactive audits of antiinfective use

Proactive on-site audits of antiinfective use in the context of antiinfective point-of-care chart reviews are important elements of ABS programmes and should be performed routinely by the ABS team (A). They enhance compliance with guidelines or clinical pathways, improve outcome in patients with infection and improve the quality of prescribing with regard to indication, choice of agent, dosing, dosing interval, administration route and treatment duration.
Depending on the problem and treatment target, besides point prevalence studies, agent-, indication- and/or diagnosis-related audits of antiinfective use should be conducted within the scope of regular antiinfective point-of-care chart reviews hospital-wide or at the unit level, whereby quality indicators should preferably be applied (A).
Results should be fed back in direct interaction with the prescribing physicians and discussed with them (A).
Significance in practice:
  • Performing proactive audit of antiinfective use with review and feedback is time-consuming; it does, however, promote interdisciplinary collaboration.
  • Antiinfective point-of-care chart reviews can increase the number of treatments complying with guidelines and thus substantially improve process quality.

Quality indicators

ABS programmes should be integrated within the hospital’s quality management. Content overlaps with the Therapeutics and Drugs Committee (drug safety) and Hospital Infection Control Committee (prevention of nosocomial infection) is useful and desired. Appropriate quality indicators to measure prescription practice (process measure), emergence of resistance or trend in consumption (outcome measure) and structure ought to be set and applied in every ABS programme (B). At least three indicators measuring structural quality and at least three indicators measuring process quality should be set regularly (A).
Significance in practice:
  • Quality indicators are used to evaluate the progress of an ABS programme.
  • Quality indicators help to recognise hospital areas which will benefit from the implementation of targeted and intensive ABS measures.

Supplemental ABS strategies

Special programmes for treatment optimisation

De-escalation
A key aspect of supplemental measures is to streamline treatment after initial empirical broad-spectrum therapy and conversion from empirical to targeted therapy. This ought to be done based on clinical criteria as well as microbiology results or other diagnostic findings. De-escalation measures ought to preferably be performed at the patient level in the context of antiinfective point-of-care chart reviews and proactive audits of antiinfective drug use (B). Programmes promoting antiinfective de-escalation are expected to, by reducing antibiotic load, impact beneficially on the emergence of resistance, the prevention of secondary infections, cost levels and adverse drug reactions (B).
Significance in practice:
  • De-escalation includes conversion from an empirical combination therapy to targeted monotherapy based on knowledge of the microorganism isolated, susceptibility and infectious disease.
  • De-escalation should be initiated early on (after 48–72 h), which also includes discontinuation of initial therapy if diagnosis is not secured. Observational studies show that this strategy is not adopted in 20–60 % of cases.
  • De-escalation programmes should point out that depending on the exact diagnosis in some cases instead of de-escalation, escalation may in fact be necessary.
Duration of treatment
It is possible to shorten the duration of antiinfective treatment for many indications (e.g. perioperative antibiotic prophylaxis) and this is recommended wherever backed by good studies and evidence. The ABS team should utilise local guidelines and antiinfective point-of-care chart reviews to draw attention to the excessive duration of treatment frequently encountered in practice. The ABS team should define the duration of treatment recommended as a rule, since this is expected to impact substantially on antiinfective drug use, side effects and costs (A). Use of biomarkers such as Procalcitonin may be useful for controlling the duration of treatment in cases where there is clinical uncertainty. As a result, the number of days of antibiotic therapy can be reduced and under certain circumstances costs can be cut (C).
Significance in practice:
  • Shortening the duration of treatment appropriately reduces the density of antiinfective use without compromising clinical outcome or costs, it also minimises the emergence of resistance by decreasing selection pressure.
  • The duration of treatment is well established for a number of indications, e.g. pneumonia, endocarditis, perioperative antibiotic prophylaxis. Therapy, thus, only needs to be individualised and extended in certain cases.
Parenteral-to-oral conversion
If sufficient bioavailability is assured, and if the patient’s condition allows, therapy should be switched from parenteral to oral antibiotic application (A). This measure reduces the length of hospital stay and the risk of line-related adverse events. Furthermore, it leads to a reduction in the total cost of treatment. Implementation of programmes allowing parenteral-to-oral conversion of antimicrobial agents at the institutional level ought to be facilitated by developing clinical criteria and through explicit designation in institutional guidelines or clinical pathways (B).
Significance in practice:
  • Switch to oral therapy should be assessed on day 3–4 of parenteral antiinfective therapy.
  • Switching to oral therapy not only results in direct cost savings (antiinfective agents, supplies, nursing time) and lowers risk of line infections, it also increases the patient’s mobility.
Dose optimisation
Adequate adjustment and optimisation of the dose and dosing interval is essential for effective, safe and responsible administration of antiinfective therapy, and an important part of ABS programmes. Besides individual patient factors, optimal dosing of antiinfectives should take into account the nature and severity of illness, the causative microorganism, concomitant medications, as well as the pharmacokinetics and pharmacodynamics of the agents prescribed. Strategies to optimise dosing in ABS programmes should include assessment of organ function for drug dose adjustment in order to avoid adverse drug events and unwanted drug interactions (A).
Furthermore, optimising the dosing interval and duration of infusion is recommended in particular in critically ill patients, best by employing a therapeutic drug monitoring (TDM) scheme; appropriate consented local institutional guidelines should be available and up to date (B).
Significance in practice:
  • Prolonged infusion of beta-lactams (taking into account physico-chemical stability) is reasonable and recommended particularly in critically ill patients.
  • TDM can avoid under-/over-dosing and minimise organ toxicity.
  • Programmes for doses optimisation are cost-effective.
Scheduled switch of antimicrobials
So-called “Cycling” programmes, which involve periodically removing a specific antimicrobial drug or an antimicrobial drug class as the standard recommended therapy and later reintroducing it (periodic scheduled rotation), are not suitable as a strategy to reverse critical emergence of resistance or to control nosocomial outbreaks with multiple resistant pathogens and, as such, should not be used as a strategy to do so (A).
Strategic rotation of specific antimicrobial drugs or antimicrobial drug classes ought to be undertaken to limit the selective pressures and to achieve a reduction of infectious microorganisms or microorganisms displaying specific resistance properties for a certain time (B). There is evidence to suggest that a balanced use of different antimicrobial drugs or antimicrobial drug classes (so-called “mixing”) can minimise the emergence of resistance. In both cases, routine surveillance of antimicrobial drug use and resistance should be performed (A).
Significance in practice:
  • Strategic rotation of specific antimicrobials or antimicrobial classes should be planned by the ABS team in consultation with the facility’s infection control team and the microbiology department. Continuous surveillance of pathogens, resistance patterns and consumption is imperative.
  • Guidelines and antiinfective formularies that recommend predominant use of fluoroquinolones or third-generation cephalosporins should be considered as critical.

Special rules for communication of microbiology results

The quality of microbiology diagnostics depends crucially on compliance with guidelines on procedures in the preanalytical phase. Expert consensus recommends that deviations from protocol ought to be reported and the reasons for rejecting the samples stated (B).
Technical progress and up-to-date molecular diagnostic methods for rapid pathogen detection should be used if they improve the quality of care and/or substantially improve identification and epidemiologic investigation of local outbreaks (A).
Positive blood culture findings, interim microscopic findings, results of rapid testing results and rapid susceptibility testing should be communicated promptly to the physician (A).
Antibiograms ought to adhere to local guidelines with respect to antimicrobial use and diagnostic findings, be presented selectively in agreement with the ABS team, and, if need be, include relevant interpretative comments. This procedure aids selection of a targeted, guideline-based antibiotic regimen (B).
The microbiology laboratory is responsible for the timely identification of critical trends in antimicrobial resistance and prompt communication of observations to the ABS team and the physicians responsible for infection control (A). This way, the clinical and epidemiological significance of the observations can be defined at an early stage.
Significance in practice:
  • Molecular diagnostic methods can expedite pathogen specification.
  • Selective reporting of susceptibility results with respect to choice and number of antimicrobial agents, and comments on daily treatment costs, route of administration, hospital formulary drug, resistance mechanisms supports adherence to local guidelines.

Special rules for management of patients with multidrug-resistant microorganisms and C. difficile

ABS strategies should be used to prevent infection with C. difficile (A). Restricting use of certain antimicrobial drugs or substitution of antimicrobial drug classes (e.g. penicillin for cephalosporins or fluoroquinolones) can considerably reduce the incidence of C. difficile infection. Infection prevention and control strategies are frequently also applied at the same time; however, they have less impact on the C. difficile incidence than in the epidemiology of MRSA or VRE.
Targeted ABS strategies are to varying degrees also effective in reducing multidrug resistant Gram-negative bacteria, particularly ESBL-producing microorganisms, MRSA and VRE, and ought to be specifically applied here too (B). In case of high prevalence of multidrug-resistant microorganisms, recommendations on diagnostic tests, evaluation of findings and treatment, as well as infection control management should be coordinated immediately and disseminated locally (A).
Routine surveillance of antimicrobial consumption and antimicrobial susceptibility data should be performed (A) to avoid indiscriminate compensatory use of other antimicrobial drug classes, since this can promote the unintentional and uncontrolled emergence of resistance.
Significance in practice:
  • Reducing consumption of cephalosporins and/or fluoroquinolones or substituting them for penicillin may reduce the frequency of C. difficile infection and possibly also have a beneficial effect on the incidence of infections caused by multidrug-resistant pathogens.

Computerised information technology

The ABS team should be supported by novel information and communication technology in the implementation of ABS programmes. Local treatment guidelines, the antiinfective formulary, and other ABS documents should be available electronically (A).
Electronic prescribing tools with and without linkage to electronic preauthorisation solutions, to ABS documents or to active communication of information using computerised reminders to the prescriber should be used to improve the use of antiinfectives in the interest of patient safety (A). They ought to be used to reduce consumption and/or costs (B).
Computerised decision support systems that are integrated into the hospital’s internal information system can, by utilising electronic medical records, help to evaluate and optimise the indication for antiinfective therapy, drug selection and dosing (C).
To implement computerised ABS measures, the ABS team must have hospital-wide access rights to electronic medical records (with due respect to data protection).
Significance in practice:
  • The local treatment guideline and the antiinfective formulary should be readily electronically accessible from every clinical computer workstation.
  • For ABS activities or for surveillance and analysis of antimicrobial usage, computer physician order entry (CPOE) systems should be designed in such a way as to allow automated generation of exact lists of the antiinfectives used.
  • Surgical software should be utilisable in such a manner as to ensure that antibiotic prophylaxis is compliant with guidelines.
  • Computer-based expert systems cannot replace a physician’s clinical judgement.

Recommendations of the guideline

Requirements

Availability of a team of ABS experts

The guideline development group recommends:
For effective implementation of ABS programmes, it is essential that a multidisciplinary team should be instructed by the hospital administration and allocated with adequate resources to draw up guidelines derived by consensus with the users for the treatment of infectious diseases and to ensure their implementation through ABS strategies (A).
The team should consist of at least one infectious diseases physician (or clinician with infectious diseases training) and an experienced clinical pharmacist/hospital pharmacist, as well as a specialist in microbiology, virology and infection epidemiology being responsible for laboratory diagnostic and microbiological consultation; furthermore, the physician locally responsible for infection control. The team members should either have appropriate training in antibiotic stewardship or already be sufficiently experienced (A).
The team will receive the support and collaboration of the hospital administration, and activities within the ABS programme should be compensated with a minimum of one full-time equivalent (FTE) of 0.5 per 250 beds (A). There should be good collaboration between the Therapeutics and Drugs Committee, Hospital Infection Control Committee, pharmacy and representatives of clinical divisions/departments (ABS representatives), for which purpose the team should issue its own Rules of Procedure (A).
In the community hospital setting, ABS programmes should be available hospital-wide, i.e. involving physicians across all operative and non-operative medical fields. A multidisciplinary team (so-called ABS team) of ABS-trained members (so-called ABS experts) is considered essential to the success of this type of programme. It should have the support of hospital administration, and collaboration of the infection control team and Therapeutics and Drugs committee, the pharmacy and of the responsible physicians (so-called ABS representatives) in the corresponding departments [2, 6]. The advantage of a multidisciplinary team is justified by the necessary diversity of ABS programmes which have different objectives of interventions depending on hospital, type of ward and speciality discipline [26]. At least one randomised controlled [27, 28] and several prospective before-and-after studies [2935] on the implementation of a trained ABS team, led to a decrease in mortality, a reduction in nosocomial infections and significantly shorter length of hospital stay. In addition, it resulted in an improved quality of prescribing, which in turn, led to fewer drug-related adverse events. The studies show that to achieve different objectives of interventions it is crucial to collect data on clinical, microbiological and prescribing endpoints, and that this can only be done by appropriately trained and sufficiently qualified professionals. Based on the IDSA/SHEA guideline and past experience [6], the ABS team should include at least one infectious diseases physician and a clinical pharmacist, ideally with infectious disease training. The importance of an infectious diseases-trained specialist and clinical pharmacist for effective ABS programmes was shown in several randomised, controlled as well as prospective, quasi-experimental studies. This was demonstrated particularly in regard to appropriate treatment of bacteremia [36], dosage adjustment and early conversion to oral therapy [3740].
The ABS team should issue Rules of Procedure defining the organisational structures and conditions for implementation of antibiotic stewardship programmes including their functions and objectives. The composition of the multidisciplinary ABS team should be described in detail, from mandate to staffing (qualification, status, objectives and functions, competences and cooperations) and amount of time compensated. Organisational charts can be useful to show internal and external communication structures. The Rules of Procedure should stipulate the frequency of meetings and the reporting obligations toward hospital administration. Potential conflicts of interest of members of the ABS team should be disclosed. Furthermore, it is necessary to lay down hospital-wide rules on how to deal with the pharmaceutical industry or third parties because commercial marketing strategies may possibly influence antimicrobial prescribing [4143].
Infectious diseases physicians are especially well-suited to planning and implementing ABS programmes and to developing guidelines because of their in-depth knowledge of the treatment of infectious diseases, their broad training in clinical internal or paediatric medicine, and not least their experience in conducting cross-departmental specialist consultations [43]. Infectious disease consultation services improved treatment quality in patients with bacteremia and in some studies also improved survival [36, 4549]. In the case of community, nosocomial or ventilator-associated pneumonia, introduction of a consultation service in an intensive care unit (incl. training) resulted in shorter length of stay (13.8 vs. 19.2 days), a decrease of ventilation time (7.4 vs. 11.8 days), reduction in duration of therapy (9.2 vs. 14.5 days) and a decrease in mortality by 6–13 % [27, 50] due to optimised empirical and targeted therapy strategies.
Several trails, including a randomised controlled trial, investigated the efficacy of a multidisciplinary ABS team which provides feedback to prescribing physicians. Particularly the feedback from the infectious disease consultation service resulted in a significantly more appropriate antibiogram-based therapy and in discontinuation of antimicrobial therapy [5154].
Clinical pharmacists/hospital pharmacists are involved in the activities of the Therapeutics and Drugs Committee and in developing local guidelines and formularies. They have special knowledge of pharmacology—such as the clinical relevance of adverse drug effects, dose optimisation or route of administration, and they have experience in conducting audits of antiinfective use, e.g. to ensure guideline adherence [37, 40, 5557]. Generally, the pharmacist is responsible for design, implementation, and compliance with formulary restrictions and preauthorisation requirements. He is also responsible for processing data on antimicrobial consumption and costs for the purpose of surveillance and benchmarking (pharmacoeconomics) [8, 5860]. Pharmacist-led parenteral-to-oral conversion programmes resulted in a significant reduction of parenteral therapy duration by 1–1.5 days without negatively impacting clinical outcomes [38, 39, 61]. This can, as shown in surgical departments of a German university hospital, lead to significant cost savings [6264]. Computerised physician order entry systems (CPOE) could aid the pharmacist in reviewing the appropriateness of antiinfective prescriptions as these systems allow to produce a daily report on the antiinfectives prescribed without review of individual patient charts on the ward.
Ideally, the team is complemented by a medical microbiologist (in Germany: specialist in microbiology, virology and infection epidemiology; in Austria: specialist in infection control and microbiology) and the physician locally responsible for infection control [65]. The expertise of the medical microbiologist is required to establish local guidelines for laboratory diagnostics of infection including preanalytical specimen management, and to report microbiological results in accordance with national and international quality standards. ABS interventions have to be in line with current microbiological diagnostics and reporting, as well as with easily accessible current surveillance data on pathogens. Medical microbiologists should provide support by using targeted diagnostic tests, rapid reporting and professional communication of results. Retrospective investigations indicate that introduction of point-of-care chart reviews focusing on diagnostics delivered by medical microbiologists with infectious diseases training leads to significant reduction in the use of broad-spectrum antibiotics [66, 67].
If infectious diseases specialists are not available in smaller hospitals, an experienced hospitalist can, in collaboration with an authorised pharmacist who has at least 2 years’ working experience in a hospital pharmacy, assume the leadership role in lieu of an infectious diseases physician. In this case, the team members must be ABS-trained, e.g. they must have completed training courses certifying them as ABS experts with knowledge in the following areas: design and implementation of ABS tools (treatment guidelines, antiinfective formulary, treatment pathways), application and implementation of point prevalence surveys of antiinfective prescribing practices, requirements for surveillance data (consumption, pathogens, resistance), the content of current guidelines and important ABS intervention strategies. The ABS experts should be capable, as a team, of developing and implementing a programme for continuous improvement of the quality of antiinfective prescribing that is tailored to the specific needs and situation of the respective hospital. Relevant continuous training in the field of ABS is recommended.
Based on the available evidence from the literature and repeated internal consultations, the guideline development group recommends that clinical infectious diseases physicians or clinical pharmacists with ABS training should principally assume core leadership function in the ABS team. Transferability of available experience (mainly American) to the German health care system is limited. The German Society for Hygiene and Microbiology (DGHM) and the Paul Ehrlich Society for Chemotherapy (PEG) point out that the conditions in Germany (several years further training in medical microbiology, medical microbiologists working in hospital and also providing infectious disease consultation, likewise the shortage of infectious diseases physicians and specialised pharmacists) and experience in some other European countries (e.g. England and the Netherlands) should allow to consider clinically oriented and experienced medical microbiologists (German: specialist in microbiology, virology and infection epidemiology), as being suitable for core leadership function, assumed they are for the most part present and available in the hospital and released from duties in the laboratory.
The team size depends primarily on the size of hospital. In the older and the current literature, between 0.5 and 1.5 full-time equivalent posts depending on the number of beds (~200 to ~900) or level of care provided, equating to one full-time equivalent of 0.5 per 250–300 beds, is well documented as being cost effective and associated with high net savings in the initial phase [23, 27, 29, 6871]. Ongoing activity by the teams is essential to preserve the effects, as the quality of use and cost usually deteriorate rapidly when ABS programmes are discontinued [23, 72]. In larger hospitals, it is therefore recommended to appoint department-specific ABS representatives to support the ABS team in its activities. The ABS team must be involved in decision-making in the Therapeutics and Drugs Committee and the Hospital Infection Control Committee as these committees may influence the design of ABS strategies, and jointly coordinated programmes (involving bundles of interventions) must be discussed to be effective particularly in the area of nosocomial infections. In the event of C. difficile outbreaks, or if the C. difficile incidence increase over time, infection control strategies alone are often not sufficiently effective. As demonstrated in multiple time-series analyses, restricting use of cephalosporins, fluoroquinolones or clindamycin is necessary to reduce C. difficile incidence effectively (see 2.1., 3.3.) [2, 25, 73].
The recommendations of the former ABS Group Austria on the further development of ABS programmes in Austrian Hospitals (“Antibiotika-Kultur in Krankenanstalten”) [74], the IDSA/SHEA Guideline on “Hospital Antibiotic Stewardship” [6] and the Australian recommendations [43] refer to the need for hospital administration to direct the ABS team to plan ABS activities, to support the implementation of these interventions and to provide necessary resources. Several quasi-experimental before-and-after studies emphasise the importance of support given by the hospital administration or departmental management, in particular in the development of guidelines, their establishment and successful implementation [75, 76].

Availability of surveillance data on pathogens, resistance, and antimicrobial consumption

Pathogens and resistance
The guideline development group recommends:
Antimicrobial susceptibility data on major pathogens should be available and accessible at least yearly on a hospital-wide level and separately for general and intensive care units, or department-specific, as the case may be. Data on primary isolates should be shown by pathogen and type of specimen, e.g. blood, urine, miscellaneous samples. Culture results from screening tests should be shown separately. Susceptibility rates should indicate the number of isolates tested. Infection rates should relate consistently to a single denominator (e.g. patient-days/number of cases). Participation in an established surveillance system is recommended (A).
A requirement for successful ABS programmes is the availability of current hospital-wide data on pathogens and antiinfective use. This will allow for weak-point analysis and optimisation potential [2, 8, 77]. In addition to provision of routine reporting on pathogen identification with antibiogram, the microbiology laboratory is, in coordination with the ABS team, responsible for surveillance of pathogen and resistance patterns. Expert consensus recommends that pathogen-specific susceptibility data should be updated at least annually. Data on primary isolates and subsequent isolates should be presented separately. In addition, data should include susceptibility and resistance rates according to generally recommended breakpoints as well as the number of isolates tested. Electronic data processing available in the microbiology laboratory can facilitate unit-specific (general ward vs ICU) or department-specific evaluation of resistance. This allows to recognise the distribution of individual pathogens and antibiotic susceptibility profiles in different departments in dependence on prescribing habits, which helps guide ABS interventions.
The available infrastructure and personnel resources must allow even hospitals without an on-site microbiology laboratory, to provide hospital-based or unit-based data on pathogens and antimicrobial susceptibility, if need be, at shorter intervals, whereby presentation of data on susceptibility rates on fewer than 10 tested isolates does not appear useful.
Expert consensus recommends reporting at least on S. aureus, E. coli other Enterobacteriaceae, P. aeruginosa and Candida spp. by specimen type (blood, urine and miscellaneous samples) as well as on C. difficile, whereby screening culture results should be reported separately. Standardised surveillance is a fundamental requirement for benchmarking with other institutions/departments. Interpretation of the data takes into account the size of hospital, the level of care, and the patient mix (e.g. hematologic-oncologic patients). Participation in established surveillance systems is recommended.
Antimicrobial consumption
The guideline development group recommends:
Data on antimicrobial consumption, expressed as use density (daily doses per 100 patient-days) should be collected at least annually or preferably quarterly and are generally reported by the pharmacist. Data are reported institution-wide, at the ward level as well as for individual (speciality) departments. On demand, data should be broken down to the agent level and should be provided to the ABS team. Participation in an established surveillance system is recommended (A).
Point prevalence surveys should be conducted for systematic quantitative and qualitative assessment of antiinfective use, and, if required should be reevaluated short-term (A). Antiinfective use data are collected at the patient level, allowing to assess prescribing quality based on indication and type of infection, and to recognise the need for targeted ABS strategies. Access to patient-level data ought to be guaranteed.
Continuous reporting of surveillance data on antiinfective consumption is useful in monitoring trends and identifying areas for evaluating appropriateness of prescribing. It therefore supports systematic audit of antimicrobial use with intervention and feedback to the prescriber [7880]. Consumption data are usually obtained from the pharmacy and are being presented as daily doses by the pharmacist. They are an essential prerequisite for medium and long-term assessment of the effectiveness of interventions [81]. Another goal of continuous surveillance is early identification of an increase in antibiotic consumption.
Expert consensus recommends that these data should be available institution-wide, for individual departments and at the ward level (e.g. general ward, intensive care unit) at least annually, preferably quarterly. Data should be collected by antimicrobial agents and reported in the form of daily doses per 100 patient-days (e.g. defined daily doses, DDD, according to the ATC Index of the WHO, and/or recommended daily doses, RDD) [82]. Upon the request of the ABS team, aggregate antibiotic usage data should be available for specific classes of antibiotics as well as stratified by different clinical units. Good examples for this form of data presentation such as so-called “antiinfective report” incl. graphical presentation are available for various German hospitals (Fig. 1).
Economic data (e.g. antibiotic costs) ought to be also documented; however, these data alone do not provide a suitable basis for analysis and intervention in terms of ABS. According to Article 23 (4) of the Infection Protection Act, usage data must be evaluated taking into account local resistance data and appropriate conclusions must be drawn regarding the use of antibiotics. Furthermore, the necessary adjustments of antibiotic consumption must be implemented and the staff must be informed. Participation in an established surveillance system provides a standardised method to calculate antimicrobial use density and is therefore recommended. Thus, depending on the patient mix, comparisons between different hospitals are also possible [81, 83]. However, IT-based patient-level consumption data, so-called prescribed daily doses (PDD) should be the ultimate goal.
Point prevalence surveys can be very helpful for temporary assessment of the quality of antimicrobial prescribing, e.g. before and after guideline amendment [8486]. A point prevalence survey provides information on the choice of substance, dose, dosing interval and route of administration. In addition, data on the indication for prescribing (nosocomial vs community acquired or prophylactic) and the type of infection can be collected at patient level, allowing to evaluate the consumption density in relation to the prescribing quality. The ABS team ought to have access to relevant patient data to conduct these surveys which are usually carried out as a 1-day point prevalence survey. Starting from the day of the survey, prescription data can also be collected retrospectively for a limited time interval (e.g. 6 days). On the day of the survey, patient-based data as mentioned above are documented. Additionally, it is recommended to document the number of patients per unit, to calculate the prevalence of antiinfective prescriptions per unit (e.g. ICU, department). This analysis allows to evaluate the relation of defined daily doses recommended by the WHO (DDD) to prescribed daily doses (PDD) derived from chart review. Additionally, other patient-relevant information can be investigated, e.g. on immunosuppression, organ insufficiencies or on presence of devices. European 1-day point prevalence surveys (http://​www.​esac.​be, http://​www.​ecdc.​europa.​eu/​en/​healthtopics/​Healthcare-associated_​infections/​database/​Pages/​database.​aspx) have shown that approximately one-third of all hospitalised patients received antiinfective treatment and that even on regular wards >50 % of total consumption was given intravenously. Furthermore, within hospitals fluoroquinolones and cephalosporins were prescribed frequently, more than 30 % of the patients received combination therapy, and >50 % of perioperative prophylaxis was administered longer than 1 day. Only 62 % of patients were treated in adherence with guidelines [8587]. Point prevalence surveys can also be used to verify the feasibility of quality indicators (see Sect. 2.2.4).

ABS core strategies

Most previously published experience with ABS programmes in hospital shows that sustained efficacy can be achieved under the requirements mentioned above and on the basis of generally accepted strategies or bundles of strategies. Certain components of ABS programmes are considered and prioritised as core ABS strategies, while others are considered optional or supplemental [2, 6, 22, 25]. The following core strategies are recommended by the guideline development group.

Application of local treatment guidelines/pathways, hospital antiinfective formulary, formulary restriction and approval requirements

The guideline development group recommends:
Developing and updating local treatment guidelines, clinical pathways, and an antiinfective formulary is one of the ABS team’s chief responsibilities. The antiinfective formulary should be based on national and international guidelines as well as on the local/regional pathogen and resistance patterns, and possibly drug costs. Drugs on the antiinfective formulary should be categorised according to recommended versus reserve or special compounds. In addition, these should be tagged with special prescription status and be subject to approval and preauthorisation requirements. The antiinfective formulary is updated at least yearly based on therapy guidelines and whenever necessary and approved by the Therapeutics and Drugs Committee (A).
Adherence to guidelines regarding substance selection, dosing, route and duration of treatment may improve clinical outcome in terms of mortality, as well as treatment duration and length of hospital stay. To ensure adherence, users should be involved in developing the guidelines and be educated through audits of antiinfective use or antiinfective point-of-care chart reviews (A).
Individualising antiinfective prescriptions with or without special approval requirements improves targeted therapy and reduces inappropriate treatment. Various possibilities for implementation have been described and should be used, from simple antimicrobial order forms to highly differentiated antiinfective request forms that may be subject to specific time limits or limited to certain hospital areas (A). Guideline-based antiinfective drug use or use of individual defined substances can be controlled by this means, thus minimising consumption, costs and adverse drug events.
Restricting whole substance classes can—by shifting to an alternative substance—prove to be an effective strategy for controlling nosocomial infections and the development of critical resistance levels; accordingly, antiinfective restriction ought to be targeted (B). At the same time, routine surveillance of antibiotic consumption and locally prevalent pathogens and their susceptibility patterns should be performed to detect possible adverse effects of the strategy in time (A).
Local treatment guidelines and clinical pathways are established and regularly updated by the ABS team with the involvement of the ABS representatives delegated from other clinical departments. National and international guidelines, the patient mix and local microbiology and resistance patterns should be taken into account. The established or revised local guidelines should have institution-wide validity for which consensus must be obtained. The treatment guidelines are presented to the Therapeutics and Drugs Committee and the Hospital Infection Control Committee. It is recommended to provide local treatment guidelines in electronic or pocketbook format and to ensure acceptance among users through training and education [88]. Without these measures guideline adherence is rather poor, and effects in terms of improving clinical outcomes or other endpoints remain small [89].
Treatment guidelines or clinical pathways can improve outcomes related to mortality, length of hospital stay and duration of treatment [90, 91]. High adherence to guidelines or clinical pathways, e.g. for management of community-acquired or nosocomial pneumonia, can be achieved with training and education. Thus, mortality can be decreased and the medium duration of therapy and hospital stay can be shortened by 1.7–6.8 days, while antiinfective usage is reduced by up to 77 % [9298]. Various strategies of treatment optimisation have been studied for community-acquired or nosocomial pneumonia [99102] and have partly been addressed in international and national guidelines. Their implementation in local guidelines, guideline adherence assumed, can help to avoid that therapy is either too broad or too long. An American and a French observational study have shown that involving physicians in the development of local guidelines can improve acceptance. When local consensus guidelines were posted on the intranet and regularly distributed to physicians and presented in departmental staff meetings, guideline-conform management of nosocomial pneumonia increased from 46 to 81 %, and 14-day mortality dropped from 23 to 8 % [103]. In a study of endocarditis, compliance with antimicrobial therapy improved from 23 to 62 % and 1-year mortality significantly decreased from 19 to 8 % [104] (Table 1). Numerous new investigations on improving guideline compliance have shown that institutionalising guidelines can optimise the quality of therapy in different categories (e.g. dose adjustment to renal function, parenteral-to-oral conversion, timely administration) by about 10 or more percent [105110].
Table 1
Examples for use of treatment guidelines and clinical pathways
References
Study-type/evidence
Patients
Intervention
Endpoints
Results
Soo Hoo et al. [103]
Observational study (II)
Patients with community-acquired pneumonia (58 patients before intervention, 58 patients after intervention)
Establishment of guidelines for the diagnosis and management of nosocomial pneumonia
Mortality
Proportion of patients with guideline-conforming treatment
Lower mortality rate at 14 days (23 vs 8 %, p = 0.03)
Increase in the number of patients treated in conformity with guidelines (46 vs. 81 %, p < 0.01)
Botelho-Nevers et al. [104]
Observational study (II)
Patients with infectious endocarditis (173 patients before intervention, 160 patients after intervention)
Establishment of treatment guidelines for management of infectious endocarditis
Mortality
Guideline adherence (compound selection, duration of treatment)
Lower 1 year mortality (18.5–8.2 %, HR 0.41; 95 % CI, 0.21–0.79, p = 0.008)
Lower hospital mortality (12.7–4.4 %, p = 0.007)
Increase in guideline adherence: compound selection (31.6 % auf 95 %, p < 0.001)
Compound selection and duration of treatment (22.7 % auf 61.8 %, p < 0.001)
Marrie et al. [93]
Randomised, controlled study (i)
Patients with community-acquired pneumonia in the emergency room of a hospital (nine hospitals with clinical pathway, 10 hospitals without clinical pathway)
Establishment of a clinical pathway for treatment of community-acquired pneumonia in the emergency room of nine hospitals
Mortality
Length of hospital stay
Duration of treatment
Proportion of patients with monotherapy
No difference in mortality
Shorter length of hospital stay by 1.7 days (6.1–4.4 days, p = 0.04)
Shorter duration of treatment by 1.7 days (6.3–4.6 days, p = 0.01)
Increase in the proportion of patients with monotherapy (27–64 %, p < 0.001)
Singh et al. [102]
Randomised, controlled study (I)
Patients with ventilator-associated pneumonia (39 patients treated in accordance with a risk score-based clinical pathway, 42 patients received standard therapy)
Establishment of risk score-based clinical pathway
Mortality
Length of hospital stay (ICU)
Detection of MDR pathogens
Duration of treatment, costs
No difference in mortality
Shorter length of hospital stay (ICU) by 5.3 days (14.7–9.4 days; p = 0.04)
Reduced detection of MDR pathogens (38–14 %, p  = 0.017
Shorter duration of treatment (9.8–3 days, p = 0.0001)
Lower treatment costs (640$–259$, p = 0.0001)
Ibrahim et al. [97]
Observational study (II)
Patients with ventilator-associated pneumonia (50 patients before intervention, 52 patients after intervention)
Establishment of a treatment guideline for management of ventilator-associated pneumonia
Mortality
Length of hospital stay, antibiotic therapy complying with guidelines,
Duration of treatment
No difference in mortality
Increase in the proportion of antimicrobial therapy conforming to guidelines from 48 to 94.2 % (p < 0.001)
Shorter duration of treatment from 14.8 days ± 8.1 days to 8.6 ± 5.1 (p < 0.001)
Clinical pathways complement local treatment guidelines, often taking into account diagnostic algorithms and risk scores. They are designed as a flowchart to simplify and improve the management of patients with infectious diseases. In a controlled, multi-centre Canadian study a risk score (PSI, pneumonia severity index)-based clinical pathway was instituted, addressing criteria for inpatient admission, sequential therapy and discharge of patients with community-acquired pneumonia. Although patients in the “experimental” arm had more severe disease, hospital stay and duration of parenteral antibiotic therapy was significantly shortened in this patient group, and the patients received monotherapy significantly more often without negative impact on mortality. Within this framework, an Australian study showed an approximately 10 % reduction in the use of broad-spectrum antibiotics [93, 111, 112]. Similar results were achieved by a more recent observational study in the UK, where introduction of a risk score (CURB-65)-based clinical pathway for treatment of community-acquired pneumonia influenced prescribing behaviour. As expected, CURB65-guided therapy resulted in an overall reduction in the prescription of cephalosporins and macrolides by 19 and 14 %, respectively, without negatively affecting outcome (30-day mortality, clinical response, treatment outcome). There was a corresponding increase in use of aminopenicillin monotherapy, and guideline compliance increased from 25 % to over 60 % [113].
Acceptance and implementation of treatment guidelines not only improves by involving users in guideline development. Other supplemental ABS strategies such as repetitive education, training and audits of antibiotic prescribing with feedback to the prescriber improve acceptance and adherence [114]. This is shown by a controlled before-and-after study in which adherence was consistently improved by a combination of interventions involving distribution of information packs to staff, repeated compilation of prescription data and educational sessions followed by reminders in the form of posters [98]. Implementation of a uniform guideline for perioperative prophylaxis including recommendations for choice of agent, dosage and timing resulted in annual antimicrobial cost savings of approximately USD 112,000 in a 1400-bed hospital [115].
The institutional antiinfective formulary is established by the pharmacist in the ABS team based on therapeutic efficacy, toxicity and cost. Drugs of the formulary should be categorised into recommended versus reserve or special compounds depending on local treatment guidelines. Graphical overview with alerts (traffic light system), information on daily therapeutic costs or restrictions on use is advisable. Adding information on special prescription or approval requirements is desirable. Besides information on agent and trade names, these lists contain information on the recommended daily dose, including dose adjustments in regard to organ impairment (Table 2). The antiinfective formulary must be passed by the Therapeutics and Drugs Committee. The formulary has an immediate influence on prescribing behaviour [116].
Table 2
Example of a formulary
https://static-content.springer.com/image/art%3A10.1007%2Fs15010-016-0885-z/MediaObjects/15010_2016_885_Tab2_HTML.gif
Caution should be exercised in controlling antibiotic use via the formulary alone without an indication-based treatment concept and concomitant surveillance of antibiotic consumption and resistance. It was for instance observed that by adding levofloxacin to the antiinfective formulary fluoroquinolone use subsequently increased substantially, resulting in a higher rate of MRSA infection. When an alert was inserted next to the fluoroquinolone selections on the electronic order entry screen, indicating alternative antibiotic agents in accordance with local guidelines, levofloxacin use decreased again by 50 % from 12 to 6 DDD/100 patient-days and the MRSA infection rate decreased again from 1.37 to 0.63 cases per 1000 patient-days [117]. Similar effects have been observed for other substances and classes and pathogens [118, 119].
Individualised antiinfective orders with or without approval requirements extend from simple to highly differentiated, computer-assisted order forms with an automatic prescription stop after a defined time (so-called “automatic stop order”). These can be agent, patient or indication based, temporary or limited to certain hospital areas. Individualised antiinfective orders present an effective tool to quickly and effectively influence prescribing behaviour. Special order forms or approval requirements are usually implemented for broad-spectrum antibiotics, new/expensive substances or substances requiring extensive consultation. They require justification for prescription, which must be evaluated prior to approval, and can effectively control use and costs. These substances are separately marked in the antiinfective formulary.
Many older prospective before-and-after trials dating from the 1980s and 1990s documented that restricting use of new and expensive cephalosporins generated cost savings of between 19 and 46 %, and reduced consumption by up to 50 % [6, 120124]. Significant cost reductions being achieved through an antimicrobial-restriction policy are less commonly observed in recent years, because numerous antibiotics have lost patent protection. Nevertheless, more recent studies showed continuing effectiveness regarding reduction in antibiotic consumption of as much as 54 %. [23, 125127]. Newer research on restricting use of broad-spectrum antibiotics yielded a monthly reduction from 137 to 72 DDD/100 cases or from 181 to 102 DDD/1000 patient-days, respectively. Overall, after implementation, the ABS programme delivered effective cost savings of USD 300,000 p.a. (corresponding to net savings of USD 2350/100 cases or 2182/1000 patient-days, respectively). [59].
Use of special order forms limiting antibiotic duration has proved to be particularly effective within the field of perioperative antibiotic prophylaxis. In several prospective, quasi-experimental before-and-after studies the effect of automatic stop order forms on antibiotic consumption, costs and guideline adherence to avoid extended prophylaxis was evaluated. By educational training, an overall 20–30 % improvement in guideline adherence was observed with respect to choice of drug and duration of antimicrobial use, with one study also showing improvement in appropriate timing of perioperative antibiotic prophylaxis before incision. This resulted in a reduction of surgical site infections from 3.2 to 1.9 %, a reduction in cost of USD 3000/100 patient-days and a reduction in consumption of approximately 20 DDD/100 patient-days [128132]. Other equally effective automated stop orders limiting total duration of treatment (e.g. 14 days) or restricting duration of reserve drugs such as vancomycin or carbapenems (72 h for empiric therapy, 7 days for therapeutic indication) have been described. Treatment beyond was only possible following consultation with the infectious diseases specialist or pharmacist. As a result, consumption of these substances was reduced by 10–25 % [125, 133, 134].
Specific programmes to restrict antimicrobial use can minimise nosocomial infections (e.g. C. difficile) and the increase of resistant pathogens (ESBL, MRSA) by a rapid and marked alteration in consumption. However, such programmes are usually only temporary and lack sustainable efficacy [22]. Restriction strategies are adopted in coordination with the Therapeutics and Drugs Committee, Hospital Infection Control Committee, the pharmacy, and hospital management. Timely and continuous surveillance of consumption, infectious diseases and resistance data are to be assured, to monitor compliance, but also to be able to rapidly identify possible negative impacts. The importance of instituting a programme for the surveillance of antimicrobial use including unrestricted antibiotics, cost and the development of resistance demonstrated by a prospective quasi-experimental observational study at a 450-bed hospital in Greece. In the study, use of carbapenems, third-generation cephalosporins, and fluoroquinolones was restricted based on a national recommendation in context of growing resistance among Gram-negative microorganisms. As a result, ciprofloxacin and ceftazidime consumption decreased as desired by 28 and 42 %, respectively. Subsequently, susceptibility of P. aeruginosa (32–45 %) and E. coli (77–84 %) to ciprofloxacin increased. On the other hand, susceptibility of K. pneumoniae to ciprofloxacin (80–60 %) and ceftazidime (61–46 %) continued to decrease. Of note, piperacillin/tazobactam use increased by 271 % and overall costs and consumption were 12–13 % higher than before intervention [135, 136].
Programmes restricting use of cephalosporins and fluoroquinolones have been repeatedly examined for their “ecological” effects [137140]. Multicenter controlled investigations in France show a 90 % reduction in fluoroquinolone use after introduction of a time-limited restriction, resulting in a significant reduction in MRSA. Reintroduction of fluoroquinolones was associated with a significant increase in MRSA compared to the previous period [141, 142]. A new study from France shows that even less restrictive fluoroquinolone use (20 % reduction) combined with improved hand hygiene also reduces the rate of MRSA (moderate) and at the same time impacts positively on resistance of P. aeruginosa to fluoroquinolones [143]. Other new studies demonstrate effects of changes in fluoroquinolone prescribing practice on C. difficile-associated diarrhoea [144148]. The effects, however, are not always due to the fluoroquinolone reduction alone.

Design and implementation of education, training and information

The guideline development group recommends:
Targeted education, training and information are essential elements of any ABS programme. They provide the foundation of knowledge needed to promote more rational use of antibiotics and reasonable microbiological diagnostic, and to improve acceptance of ABS programmes. They have the objective of optimising the therapeutic and diagnostic management of patients with infection through greater adherence to recommendations. They should preferably take place as an active training measure rather than in the form of passive communication of information (A).
Education, training and information in different formats and on various topics should be offered repeatedly as they are not sustainable as a one-off measure. They should be organised in agreement and integration with local ABS programmes (A).
Education, training and information should be independent of commercial interests, whereby the hospital administration is responsible for implementing and financing the measures (A).
Education, training and information are essential elements of every ABS programme. Overall, in a systematic review active clinician education in the form of lectures, seminars, “bedside teaching” demonstrated greater effectiveness than passive education techniques like posters, pocket cards or written prescription recommendations [149]. Two multicenter, randomised, controlled, and some before-and-after studies demonstrated that an educational intervention improved compliance with guideline-recommended diagnostic, therapeutic and prophylactic measures and resulted in a reduction in the number of non-indicated treatments [150152]. By educational training of nurses and medical staff inappropriate submission of urine cultures decreased from 2.6 to 0.9 per 1000 patient-days; treatment of asymptomatic bacteriuria was reduced from 1.7 to 0.6 per 1000 patient-days, while in another study a significant reduction from 74 to 17 % was seen [153, 154]. In Canadian long-term health care facilities, a 1-year long educational intervention involving repeated mailing of antibiotic guidelines with feedback on individual antibiotic prescribing behaviour of urinary tract infection, pneumonia, skin and soft tissue infection and sepsis resulted in a significant 64 % reduction of nonadherent treatment compared to control facilities [155]. Nonadherent antibiotic prescriptions remained lower during follow-up, although after termination of the educational intervention, the effect was no longer significant compared to control facilities. A similar effect was achieved in a study on an educational programme for guideline-based treatment of respiratory tract infections in emergency departments, in which 1 year post-intervention a 10 %, albeit non-significant reduction in antibiotic consumption was still documented compared to sites without intervention [156].
With the aim of reducing extended use of perioperative antibiotic prophylaxis by means of information disseminated by e-mail, poster and lectures, 12 Australian hospitals succeeded in rapidly and effectively limiting the duration of antibiotic prophylaxis to maximally 48 h, thus achieving considerably lower costs, which more than outweighed the costs of the 1-year intervention [157]. However, the effect of the intervention rapidly declined with time, as seen in other studies [158]. Repeated guideline-based educational interventions are necessary. They were shown to be particularly effective in optimising perioperative antibiotic prophylaxis [6, 159]. In an Argentine multi-step ABS programme involving training and formulary restriction, antimicrobial consumption could be reduced from 43 to 28 DDD/100 patient-days, resulting in substantial savings (>900.000 USD) over 18 months. During the training period a significant increase in the rate of prescriptions based upon microbiology results (27–63 %) was found, and use of ceftriaxone and carbapenems subsequently more than halved [30]. The combination of one-on-one education (academic detailing) and special review of orders for either levofloxacin or ceftazidime was also seen as a highly effective method for reducing inadequate antibiotic use. Unnecessary antibiotic prescription was significantly reduced by 41 % with no change in clinical outcome [28, 160]. However, academic detailing is time-consuming and personnel-intensive [28, 160]. This can partly be compensated by less time-consuming feedback activities, e.g. in the form of written recommendations placed in the patient chart; however, these are not quite as effective and less sustainable [157].
Education, training and information should be independent and should not be guided by the commercial interests of the manufacturers of medical and diagnostics products, since this is the only way to ensure that prescribing and professional behaviour are not subject to direct or indirect influence (Table 3). A systematic review examined the impact of various strategies undertaken by the pharmaceutical industry such as visits, funding for travel or lodging, sponsoring educational events, free samples, etc., on prescribing practices. According to the study, industry-sponsored continuing medical education (CME) had the biggest impact on physician prescribing practices compared to other activities, leading to a 6–19 % increase in prescription rates of the sponsor’s medication [41, 42]. The responsibility of organising, holding and financing educational events should be assumed by hospital management.
Table 3
Examples of the influence of commercial interests on prescribing and formulary design [41, 42]
Meetings with pharmaceutical representatives
66 % less likelihood of prescribing generic products
Travel sponsoring (congresses, etc.)
Requests to add the sponsor’s drugs to the hospital formulary are associated with an odds ratio of 7.9 % (95 % CI, 1.1–55.6)
4.5- to 10-fold increase in hospital prescribing rate (sponsor’s products) as compared to before travel
Continuing medical education (CME funding)
Prescribing rate, (sponsor’s products) increases by 5.5–18.7 %
Research funding
Requests to add the sponsor’s drugs to the hospital formulary is associated with an odds ratio of 9.5 (95 % CI, 2.6–35.7)

Conducting proactive audits of antiinfective use

The guideline development group recommends:
Proactive on-site audits of antiinfective use in the context of antiinfective point-of-care chart reviews are important elements of ABS programmes and should be performed routinely by the ABS team (A). They enhance compliance with guidelines or clinical pathways, improve outcome in patients with infection and improve the quality of prescribing with regard to indication, choice of agent, dosing, dosing interval, administration route and treatment duration.
Depending on the problem and treatment target, besides point prevalence studies, agent-, indication- and/or diagnosis-related audits of antiinfective use should be conducted within the scope of regular antiinfective point-of-care chart reviews either hospital-wide or at the unit level, whereby quality indicators should preferably be applied (A).
Results should be fed back in direct interaction with prescribing physicians and discussed with them (A).
Proactive audit of antiinfective use with review and feedback include the collection and analysis of data on diagnosis, indication, choice of agent, dosing, administration route and treatment duration at patient level. The results are fed back to and discussed with the prescribing physicians (point-of-care interventions). In personal consultation with the prescribing physicians reasons for choice of drug could be asked for and therapy should be optimised based on clinical, laboratory, radiological and microbiological examination results. Concomitant disease, comedication, expected pathogens when microbiology is not yet known and local antimicrobial susceptibility patterns must be taken into account. The guideline of two North American medical societies has described this type of audit as a highly effective interventional tool that provides a core strategy for an antimicrobial stewardship programme—called “prospective audit with intervention and feedback” [6].
Proactive audit of antiinfective use with review and feedback by an ABS team has been described as effectively increasing the rate of adequate antiinfective use by 20 %; the strategy can reduce the rate of inadequate use by half [34, 53, 161164]. Methods of feedback can be modified, especially when computer-based assistance is available [165]. The quality of information is important; however, personal feedback is often more effective [165, 166]. In addition to a direct improvement in the quality of prescribing, audits of antiinfective use allow to recognise the need for education and training.
Audits of antiinfective use can be agent-, diagnosis- or indication-based and can be performed at the patient level, in individual departments, wards, or hospital-wide. Targeted (e.g. in relation to agent, speciality department or ward) as well as time-restricted antiinfective audits can be highly effective (examples are shown in Table 4). A programme in which an infectious diseases specialist or pharmacist conducted targeted point-of-care chart reviews (3×/week) of patients receiving multiple antibiotics, prolonged or high-cost therapy (120-bed hospital), achieved good acceptance: 69 % of the recommendations were accepted and implemented, of these 38 % were to discontinue therapy due to excessive duration, duplicate coverage or inappropriate use, and 33 % were to switch to oral application. Compared with the previous year, a cost reduction of 19 %, estimated savings of USD177,000 were achieved [167].
Table 4
Examples for performing targeted proactive audits of antiinfective use
• Perioperative antibiotic prophylaxis in selected surgical fields
 
• Targeted therapy of bacteremic patients hospital-wide
 
• Community-acquired pneumonia in the emergency department
 
• Sequential therapy on general wards with antibiotics of high bioavailability
 
Agent-related proactive audit of antiinfective use can address dose adjustment to organ dysfunction, switch to oral application, discontinuation of therapy or targeted therapy based on microbiology. In a multicenter, randomised, controlled study parenteral antibiotics could be significantly reduced by 1 day when patients who had received parenteral antibiotics for longer than 3 days were reviewed by an infectious diseases physician for possible sequential therapy based on defined clinical and laboratory criteria and a recommendation was made for switch to oral drug application [38]. The intervention showed lack of effect on length of hospital stay, but reduced mean antibiotic costs per patient significantly from USD 36 to USD 20. Following updated recommendations on aminoglycoside treatment, antiinfective visitations by infectious diseases physicians achieved a significant 11 % reduction in nephrotoxicity by shortening the treatment duration from 6 to 4 days and optimise dosing by monitoring drug levels [161]. In a study assessing the effects of intervention and feedback by the infectious diseases physician, empiric treatment with levofloxacin, vancomycin and carbapenems was switched to targeted antibiotic treatment in line with guidelines. Consumption subsequently decreased by 20 % and median duration of therapy was reduced from 6 to 4 days in comparison to a control group [168, 169]. In another intervention, a prospective audit and feedback programme was instituted in a teaching hospital by pharmacists and infectious diseases physicians to counteract a trend towards increasing use of expanded-spectrum antimicrobials. This resulted in a significant reduction in consumption of third-generation cephalosporins and aztreonam within a period of 6 years from 28 to 6 DDD/1000 patient-days. Furthermore, there was a significant decrease over time in infections caused by C. difficile from 2.2 to 1.4 cases/1000 patient-days [29]. According to another recent time-series analysis, a significant decrease in fluoroquinolone consumption from 118 to 78 DDD/1000 patient-days over 4 years was achieved by the ABS team following implementation of daily hospital-wide audits of fluoroquinolone use based on individual patient data. At the same time, the rate of fluoroquinolone-resistant P. aeruginosa continuously decreased from 42 to 26 % [143]. A similar intervention in intensive care units resulted in a sustained 22 % decrease in the number of days of therapy with extended spectrum antibiotics compared with the control group—without negative impact on mortality [170] (Table 5).
Table 5
Suggested evaluation categories in local audits of antimicrobial use
1.
Antimicrobial therapy adheres to established institutional guidelines with respect to:
 Choice of agent
 Dose
 Route of administration
 Duration of infusion
 Duration of treatment
2.
Antimicrobial prophylaxis adheres to established institutional guidelines with respect to:
 Choice of agent
 Dosing
 Route of administration
 Timing of preoperative dose
 Dosing interval
 Duration of administration
With proactive audit of antimicrobial use focussing on diagnosis and indication for antibiotic treatment by an infectious diseases physician-led ABS team with direct interaction and feedback as well as written documentation of recommendations, length of stay was shortened by 3.3 days and a 6 % decrease in mortality was achieved [27]. As a result of the intervention, median hospital costs were reduced by USD 2642/intervention. By optimising the process of perioperative antibiotic prophylaxis, appropriate dosing and timely administration significantly increased from 72 to 90 % and 36 to 79 %, respectively [171]. Within the frame of quality assurance, and for benchmarking purposes with other hospitals, targeted audits of selected process of care indicators for the management of important and frequent infections can also in small acute-care hospitals lead to a significant improvement in adherence to established guidelines. In a quasi-experimental before-and-after study of a total of 36 hospitals (<200 beds) the effect of proactive audit and feedback, by using quality indicators, on the management of pneumonia in the emergency department was investigated. The hospitals demonstrated a 30 % improvement in the performance of microbiological diagnostics (blood/sputum cultures) prior to therapy and antibiotic administration within 4 h of hospital admission. As a consequence, mortality was reduced significantly by 12–6 % [172]. Proactive audits of antiinfective use based on selected quality indicators should regularly take place (see Sect. 2.2.4).
The ABS team should determine the objective, type, contents and frequency of point-of-care chart reviews in agreement with the wards or departments involved and should give report on its effects. The ABS team should get project-specific to hospital-wide access to the laboratory, radiological and microbiological data needed. Computer-based information technology can facilitate audits of antiinfective use (see Sect. 3.3.4)

Quality indicators

The guideline development group recommends:
ABS programmes should be integrated within the hospital’s quality management. Content overlaps with the Therapeutics and Drugs Committee (drug safety) and Hospital Infection Control Committee (prevention of nosocomial infection) is useful and desired. Appropriate quality indicators to measure prescription practice (process measure), emergence of resistance or trend in consumption (outcome measure) and structure ought to be set and applied in every ABS programme (B). At least three indicators measuring structural quality and at least three indicators measuring process quality should be set regularly (A).
ABS programmes are to be regarded as a strategy to ensure quality and should preferably reside as a standard component within the hospitals’ existing quality management [25]. It is recommendable to utilise data captured pursuant to the new Infection Protection Act for surveillance of resistant microorganism or antiinfective drug use (IfSG §23 Abs. 4) as well as selected data on infection management provided by external quality assurance sources. Additional quality indicators for local use should be selected and applied regularly. This allows to evaluate and document whether ABS aims can be met [171]. Owing to the different structures and organisation of hospitals, ABS measures must be evaluated locally and if need be adjusted accordingly [2, 22].
Ideally, indicators ought to be evidence-based, i.e. guideline-derived, and ought to be supported by a formal consensus process in regard of their relevance and practicability; last but not least, they ought to be also put to the practical test. Indicators have been developed for community-acquired pneumonia and urinary tract infections. However, numerous suggestions have been put forward for indicators whose evidence base is rather small and whose relevance and practicability rests on consensus alone. In Germany, there are catalogues of quality indicators for instance for the Helios Hospital Group (“Initiative of Quality Medicine”) or for the Rhön, Sana und Asklepios Hospital Group (“Quality Hospitals”). They also exist for mandatory external health care quality assurance concepts, for whose development and implementation the German National Institute for Quality Measurement in Health Care (Bundesgeschäftsstelle Qualitätssicherung gGmbH; BQS) till 2009, and since then the AQUA Insitut (the Institute for Applied Quality Improvement and Research in Health Care GmBH) was commissioned by the Federal Joint Committee (Gemeinsamer Bundesausschuss; G-BA). However, only few quality indicators have been set for measurement of antibiotic prescribing of which some are already at goal (e.g. for community-acquired pneumonia, antibiotic prophylaxis for obstetric and gynaecological indications, femur fracture, as well as hip and knee endoprosthesis). Individual more or less plausible and consented catalogues of structural indicators are available outside Germany [65, 173, 174]. A lot of experience was especially gained in France. Process quality indicators, respective pneumonia and surgical prophylaxis, are available in multiple countries (e.g. http://​www.​qualitymeasures.​ahrq.​gov or http://​www.​jointcommission.​org or http://​www.​ic.​nhs.​uk).
The guideline committee in collaboration with the ABS Expert Network (http://​www.​antibiotic-stewardship.​de) and the University Hospital of Freiburg established a catalogue of consensus structural and process ABS quality indicators in a multistage procedure including Delphi survey. The catalogue should facilitate external and internal quality assurance. Clinical, ecological (resistance) and economical (cost, cost-effectiveness) relevance as well as the presumed practicability were assessed separately in several categories. In analogy with the so-called QUALIFY process [175], a provisional list of potentially suitable structural and process indicators was drafted. It was based on the draft of the Guideline itself, the current literature [1, 6, 25, 86, 105, 171, 176189], including documents and experience with the former ESAC Group (http://​www.​esac.​ua.​ac.​be) [190] and the former ABS International Group, an initiative of 9 EU member states for the improved use of antiinfectives (http://​www.​abs-international.​eu) [191]. Based on the results of a workshop (15 participants) held at the ABS Expert Network meeting in November 2011 in Freiburg, a later questionnaire survey (Delphi methods, n = 75 ABS experts, i.e. advanced members of the ABS training programme of varying professional background, incl. pharmacy and microbiology) and a further workshop, of the initial 99 potential indicators 67 were put forward for discussion and 21 structure and 21 process indicators subsequently selected as presumably most suitable (see Tables 7, 8 in the Appendix) [434]. Suitable indicators on the quality of antimicrobial prescribing (process indicator) and structure (structure indicator) taken from this list (Tables 7, 8 in the Appendix) ought to be set and used in every ABS programme. At least three indicators on structure and process quality each should be determined regularly.

Supplemental ABS strategies

There are various strategies or measures that can supplement and complement the core ABS activities described here. They can play a pivotal role in further improvement of outcomes of antibiotic stewardship programmes. Supplemental ABS strategies include special programmes and recommendations for therapy optimisation, special rules in reporting microbiology results, rules on the management of patients with multidrug-resistant pathogens (MDR) or C. difficile as well as computerised support systems. Evidence for their effectiveness varies. Their implementation partly depends on the hospital’s infrastructure, e.g. with computerised expert systems and information technology or the possibility for rapid measurement of antibiotic levels in serum.

Special programmes for treatment optimisation

As a rule, programmes for therapy optimisation such as de-escalation (streamlining) strategies, interventions to control duration of treatment, switch to oral administration and optimisation of antimicrobial dosing are carried out at the ward or patient level (also called “point-of-care interventions”). These are relatively focussed interventions that can be highly effective in improving the quality of antimicrobial prescribing. They are usually elements of proactive audit of antiinfective use or chart review, implementation can, however, also be computerised. [2, 22, 25].
De-escalation
The guideline development group recommends:
A key aspect of supplemental measures is to streamline treatment after initial empirical broad-spectrum therapy and conversion from empirical to targeted therapy. This ought to be done based on clinical criteria as well as microbiology results or other diagnostic findings. De-escalation measures ought to be preferably performed at the patient level in the context of antiinfective point-of-care chart reviews and proactive audits of antiinfective drug use (B). Programmes promoting antiinfective de-escalation are expected to, by reducing antibiotic load, impact beneficially on the emergence of resistance, the prevention of secondary infections, cost levels and adverse drug reactions (B).
De-escalation proposes to simplify treatment, i.e. monotherapy rather than combination therapy, targeted (narrow spectrum) rather than untargeted broad-spectrum therapy, discontinuation of empiric treatment if diagnosis is uncertain [192, 193]. There are insufficient data to favour combination therapy over monotherapy in the routine management of ventilator-associated pneumonia. In addition, combination therapy did not show a benefit with regard to decreasing superinfection rates or the emergence of resistant pathogens [6, 194197]. In a meta-analysis of eight randomised, controlled studies on patients with different infections β-lactam/aminoglycoside combination therapy did not impact favourably on the emergence of resistance. Fewer superinfections were observed with monotherapy (OR 0.62; 95 % CI, 0.42–0.93) than with combination therapy [198]. Furthermore, combination with aminoglycosides is associated with a higher incidence of nephro- and ototoxicity [199201]. Thus, de-escalation to monotherapy is recommended based on type of infection and microbial culture results [202]. Combination therapy is only recommended for selected indications.
Observational studies on general wards and intensive care units show that 20–60 % antibiotic treatments could be adjusted based on microbial findings alone [193, 203205]. Pharmacists and infectious diseases physicians assessed combination therapy as being unnecessary in 50 % of cases, measurable effects of de-escalation were reduced length of hospital stay and high-cost savings [206208]. In an intervention, a computer programme identified combination antibiotic therapy across the hospital, which was then evaluated by a pharmacist or infectious diseases physician for adequacy. 98 % of combination therapy was found to be redundant. The implementation led to considerable net savings [209].
The effect of de-escalation, or treatment adjustment based on microbial culture results and/or clinical criteria was well demonstrated in at least one multicenter clinical study of patients in intensive care. Patients suspected of having developed ventilator-associated pneumonia receiving treatment in 31 French intensive care units were switched to targeted treatment based on culture and sensitivity results of pathogens obtained by bronchoalveolar lavage or endotracheal aspiration. Patients who received treatment based on bronchoalveolar lavage culture results had significantly more antibiotic-free days (5 vs 2), significant 10 % lower mortality at day 14 and decreased sepsis-related organ failure at day 3 and 7 [100, 103]. The authors related this to the fact that invasive bronchoscopy allows to differentiate better between pulmonary infection and colonisation, and that antibiotic treatment can be discontinued earlier given negative cultures from bronchoalveolar lavage. In a randomised controlled trial conducted in an intensive care unit in North America, the course of antibiotic therapy of pneumonia was shortened by 2 days, based on clinical criteria, without negative impact on mortality [210]. Numerous other investigations confirm these observations and show that adjusting therapy is usually possible after 48–72 h [211214].
Duration of treatment
The guideline development group recommends:
It is possible to shorten the duration of antiinfective treatment for many indications (e.g. perioperative antibiotic prophylaxis) and this is recommended wherever backed by good studies and evidence. The ABS team should utilise local guidelines and antiinfective point-of-care chart reviews to draw attention to the excessive duration of treatment frequently encountered in practice. The ABS team should define the duration of treatment recommended as a rule, since this is expected to impact substantially on antiinfective drug use, side effects and costs (A). Use of biomarkers such as Procalcitonin may be useful for controlling the duration of treatment in cases where there is clinical uncertainty. As a result, the number of days of antibiotic therapy can be reduced and under certain circumstances costs can be cut (C).
A frequently encountered problem in regard to antibiotic therapy is the duration of antimicrobial treatment often being too long. Large-scale studies in the USA, in European countries and elsewhere have repeatedly demonstrated prolonged duration of perioperative antibiotic prophylaxis—in 50 % or more cases perioperative antibiotic prophylaxis was administered for longer than 24 h [85, 86, 215]. This unnecessarily increases selective pressure for resistance to emerge [102, 216219].
German recommendations with S3 Guideline level, e.g. on community-acquired and hospital-acquired pneumonia or uncomplicated community-acquired urinary tract infections, give explicit, evidence-based recommendations on duration of treatment (http://​www.​awmf.​org). The results of the studies on which the recommendations on pneumonia are based demonstrate convincingly that the mean duration of treatment can be reduced without negative impact on clinical outcome and mortality [102, 220, 221]. An important trial is a prospective randomised double-blind study of patients with ventilator-associated pneumonia undertaken in 51 French intensive care units. It was demonstrated that 8-day treatment had no disadvantage for these patients as compared to 15 days. Shorter treatment duration was associated with emergence of fewer multidrug-resistant pathogens (−20 %) [101, 102]. These findings and the results of other similar studies have had a decisive influence on the assessment and conclusions reached in recent meta-analyses [222, 223]. Other good scientific studies on urinary tract infection show similar results [224]. When implementing ABS programmes, treatment orders for patients with community-acquired pneumonia should for instance be linked to a note “no longer than 5–7 days” to remind users to undertake an individual clinical reassessment of further treatment beyond this point in time. This question can also be addressed in proactive audits of antiinfective drug use [225, 226].
Biomarkers can also be useful to guide duration of therapy. Corresponding studies are available on use of Procalcitonin particularly in the management of patients with respiratory tract infections, whereby treatment duration in the control arm does not correspond in some cases to today’s standards. Various systematic reviews and meta-analyses are available on Procalcitonin [227, 228]. Determination of Procalcitonin levels can influence the density of antibiotic treatment in intensive care units: in a prospective study antibiotic treatment was reduced by 23 %, in other studies this effect remains, whereby the cost-effectiveness is unclear and there seems to be no impact on mortality [229235].
Numerous studies have aimed to improve adherence to guidelines on perioperative antibiotic prophylaxis, especially with regard to duration. Training programmes, local guidelines and checklists in the operating room with and without use of computer-based information technology were most often applied. For various reasons the results are not always satisfactory and comparable, depending on intervention and speciality field [128, 129, 158, 188, 236242]. Some examples of successful outcome are the significant reduction of treatment duration from 2.4 to 1.6 days (Japan), the 15 % reduction in the amount of antibiotics prescribed for perioperative antibiotic prophylaxis (Germany), the increase in the proportion of guideline-adherent prophylaxis of no longer than 24 h duration from 3 to 66 % (Taiwan) and the reduction of prolonged prophylaxis >24 h from 21 to 8 % (Netherlands). A time-series analysis of 13 hospitals in the Netherlands demonstrated convincingly that targeted training programmes result in a decrease of perioperative antimicrobial drug use from 121 to 99 DDD (defined daily doses)/100 procedures, and in a cost reduction of 25 % per procedure [188]. If available, electronic prescribing systems can be used for automatic stop orders to reduce the proportion of patients with prolonged prophylaxis. In a US study, by computer-based order intervention, the proportion of patients who had prophylaxis discontinued in the appropriate time frame increased by 17 %, compared to no change without intervention [243].
Parenteral-to-oral conversion
The guideline development group recommends:
If sufficient bioavailability is assured, and if the patient’s condition allows, therapy should be switched from parenteral to oral antibiotic application (A). This measure reduces the length of hospital stay and the risk of line-related adverse events. Furthermore, it leads to a reduction in the total cost of treatment. Implementation of programmes allowing parenteral-to-oral conversion of antimicrobial agents at the institutional level ought to be facilitated by developing clinical criteria and through explicit designation in institutional guidelines or clinical pathways (B).
Critically ill patients suffering from an infection, initially receive parenteral antibiotics. Stabilised patients as well as patients with a less serious illness can be given oral agents with good bioavailability as long as there are no contraindications (e.g. disorders of gastrointestinal resorption or dysphagia) (Table 6). Conversion to oral administration has numerous advantages. Mobility is improved, patients can be discharged earlier, the risk of adverse line-related events is smaller, and the amount of nursing time required is usually reduced. Switch to oral antibiotics has been well investigated for certain indications, and recommendations are made in many guidelines, e.g. the German guideline for treatment of community-acquired pneumonia [6, 244246]. Safety of switch to oral administration was evaluated in at least one meta-analysis and several partly multi-centre randomised controlled clinical trials. It was demonstrated, that duration of parenteral treatment and length of stay can be reduced by approximately 2–3 days without increasing mortality [61, 247255]. In a prospective quasi-experimental observational study involving around 200 pneumonia patients, almost 70 % of the patients could be switched to oral antibiotics at day 3, and a further 20 % between day 4 and 7. Similar experience was made in a series of other observational studies [39, 76, 111, 256, 257]. Safety was also investigated with respect to study endpoint hospital readmission [258, 259].
Table 6
Substances with good-to-excellent bioavailability
• Fluoroquinolones (without norfloxacin)
 
• Cotrimoxazole
 
• Doxycycline
 
• Metronidazole
 
• Linezolid
• Rifampicin
• Fluconazole
With the exception of endocarditis and meningitis, timely switch to oral antibiotic administration can also be reasonable for pyelonephritis, for skin and soft tissue infections, febrile neutropenia, infantile osteomyelitis/purulent arthritis [252, 260267]. Systematic review of good clinical studies partially performed in Europe shows that switch to oral antibiotic administration for another 7–11 days is already possible at day 3 of parenteral therapy for treatment of infantile pyelonephritis without a higher incidence of renal damage or other complications [268271]. Other studies have documented substantial cost savings from early shift to oral antibiotics and as a consequence earlier discharge from hospital [64, 272274]. Some of these investigations were conducted by hospital pharmacists themselves [39, 275]. Prospective observational studies investigating early switch to oral antibiotics have demonstrated that sustainability can be achieved by checklists, clinical pathways defining criteria for early conversion to oral therapy and its implementation supported by hospital pharmacists [258, 276280].
Dose optimisation
The guideline development group recommends:
Adequate adjustment and optimisation of the dose and dosing interval is essential for effective, safe and responsible administration of antiinfective therapy, and an important part of ABS programmes. Besides individual patient factors, optimal dosing of antiinfectives should take into account, the nature and severity of illness, the causative microorganism, concomitant medications, as well as the pharmacokinetics and pharmacodynamics of the agents prescribed. Strategies to optimise dosing in ABS programmes should include assessment of organ function for drug dose adjustment in order to avoid adverse drug events and unwanted drug interactions (A).
Furthermore, optimising the dosing interval and duration of infusion is recommended in particular in critically ill patients, best by employing a therapeutic drug monitoring (TDM) scheme; appropriate consented local institutional guidelines should be available and up to date (B).
Evaluation of ABS programmes showed that one-third of all interventions was in regard to dose optimisation of antimicrobial treatment. This was demonstrated in many retrospective studies which provided evidence of inappropriate choice of agent and inadequate dosing [281287]. As with all medication, antiinfective dosing requires individual review and adjustment. If need be, dose and dosing interval must be adjusted, whereby age, weight, gender, hepatic and renal function, underlying and concomitant disease as well as co-medication must be considered. Dosing is largely determined by pathogen susceptibility, the location and severity of infection [288]. Dose optimisation programmes have been implemented by pharmacists and infectious diseases physicians with similar success and can also be cost effective [37, 289297].
Pharmacokinetic and pharmacodynamic (PK/PD) properties are important to achieve optimal antiinfective dose levels [298, 299]. Emergence of resistance can be promoted by using incorrect, low dosages of antibiotics with low-resistance barrier [216]. Therefore, strategies to avoid incorrect drug dosage or suboptimal dispensing appear useful, at least in critical areas such as intensive care units, despite uncertain evidence for a clinical benefit [101, 300305]. Important strategies are optimisation of dosing intervals (e.g. higher concentrations of aminoglycoside with an extended dosing interval) [306] and prolonged infusions of beta-lactams especially in presence of critical illness or multidrug-resistant microorganisms [307]. In this setting, therapeutic drug monitoring (TDM) can improve antibiotic dosing [308310].
By using TDM, the proportion of patients with serum piperacillin concentrations within therapeutic range increased from 50 to 75 % in a 30-bed intensive care unit in a French teaching hospital [311]. A much cited, multi-centre study from the Netherlands conducted in medical and surgical wards of four hospitals was able to show that active TDM-guided dosing of aminoglycosides and dosing recommendations by clinical pharmacists can significantly shorten length of stay by approximately 6 days and reduce incidence of nephrotoxicity from 13 to 3 %. A highly detailed cost-effectiveness analysis showed overall cost savings of 30 % [312]. Similar results were reported from France [313, 314]. The benefits of single-dose aminoglycoside administration compared to multiple-dose and extended-interval aminoglycoside dosage regimens can be utilised to minimise nephrotoxicity in children [38, 315, 316].
PK/PD analyses show that with beta-lactam antibiotics the duration of time that drug levels exceed the MIC of the pathogen at the site of infection is important to achieve better treatment outcomes (time-dependent killing). Beta-lactams with short half lives (<2 h) should actually be given as extended or continuous infusion, which requires drug stability at room temperature [317, 318]. An older meta-analysis [319, 320] suggests that clinical outcome in continuous intravenous infusion of suitable beta-lactams seems to be superior to intermittent infusion of the same daily dose. More recent systematic reviews investigating continuous infusion have not confirmed superiority [310, 321], whereby different daily doses were compared, though in certain studies very good effects were observed. McKinnon et al. [322] for instance showed in a prospective randomised investigation in critically ill patients with bacteremia that continuous infusion ceftazidime or cefepime is superior to intermittent short-term regimen: clinical improvement and mortality differed significantly. In a randomised study of bacterial meningitis, where cefotaxime was administered in the first 24 h by continuous infusion versus intermittent bolus, a benefit was seen in favour of continuous infusion in terms of lower mortality [323]. Treatment success can be achieved by the drug concentration being 40–60 % of the time in the 2- to 4-fold range of the MIC. Drug concentrations remaining above the MIC 100 % of the time is not necessary. An intermittent dosing strategy with longer duration of infusion can thus suffice to guarantee optimal outcome [324]. A retrospective multicenter study showed that increasing length of infusion (4 h) gives better clinical results for intermittent dosing of piperacillin/tazobactam. Mortality was reduced from 18 to 10 % (p = 0.02) [325]. A similar (before and after) observational study (larger number of cases) with cefepime, piperacillin/tazobactam and meropenem could not reproduce these positive results [326]. In yet a further prospective investigation conducted across Australia and Hongkong with 2 × 30 patients (in addition to piperacillin/tazobactam, ticarcillin/clavulanate and meropenem were permitted), PK parameters were defined as primary endpoint. The clinical results are of interest nevertheless: clinical cure was observed in 70 vs 43 % (continuous vs. bolus) (p < 0.01), and survival in the two arms was 90 vs 80 % [327]. In a Czech study, 2 × 120 patients (intensive care, medium APACHE-II-Score >20, high incidence of Klebsiella infection) were randomised in continuous infusion group and bolus group. A high loading dose was given in both arms. Clinical cure (83 vs. 75 %, p = 0.18) and microbiological success rate (91 vs. 78 %, p = 0.02) were better with continuous infusion. However, a very high loading dose of meropenem was given in both arms (4 × 1 g over 6 h vs. 3 × 2 g over 30 min) [328]. Additional findings in the continuous infusion group were shorter ICU stay (10 vs. 12 days), shorter duration of therapy (7 vs. 8 days) and (as to be expected from the study design) lower total dose of meropenem. Concerning mortality, no significant statistical difference (hospital, ITT population) was detected (17 vs. 23 %).
Scheduled switch of antimicrobials
The guideline development group recommends:
So-called “Cycling” programmes, which involve periodically removing a specific antimicrobial drug or an antimicrobial drug class as the standard recommended therapy and later reintroducing it (periodic scheduled rotation), are not suitable as a strategy to reverse critical emergence of resistance or to control nosocomial outbreaks with multiple resistant pathogens and, as such, should not be used as a strategy to do so (A).
Strategic rotation of specific antimicrobial drugs or antimicrobial drug classes ought to be undertaken to limit the selective pressure and to achieve a reduction of infectious microorganisms or microorganisms displaying specific resistance properties for a certain time (B). There is evidence to suggest that a balanced use of different antimicrobial drugs or antimicrobial drug classes (so-called “mixing”) can minimise the emergence of resistance. In both cases, routine surveillance of antimicrobial drug use and resistance should be performed (A).
Repeated rotation of different antimicrobials or antimicrobial classes (e.g. cephalosporins, fluoroquinolones, penicillins, carbapenems) for empiric therapy of acute infection within an institution or specific unit—so-called “Cycling”—was designed to limit selective pressure and thus prevent development of resistance toward frequently prescribed antimicrobials/antimicrobial classes. Published experience with cycling strategies, mainly on aminoglycosides, is a few decades old, and, in view of the minor share of aminoglycosides used, of little interest in clinical routine today. Neither are the studies consistently successful in showing a detectable effect in minimising resistance [6, 329, 330]. In many cases, implementation was problematic, with up to 50 % of the patients in cycling programmes receiving “off cycle” antimicrobials, i.e. not receiving per protocol antimicrobial treatment [6].
More recent studies on cycling of broad-spectrum beta-lactams show little improvement in methodology or results [331337]. Neither do mathematical models of antimicrobial cycling demonstrate benefit in respect of avoiding resistance. Mathematical modelling suggests that antibiotic cycling strategies, prompting simultaneous diversity of antimicrobials/antimicrobial classes, perform better than temporary dominance of a single antimicrobial agent/antimicrobial class [338340]. Several clinical trials confirm this concept [341344]. Within the scope of a Spanish prospective intervention study on ventilator-associated pneumonia in an interdisciplinary intensive care unit, it could indeed be demonstrated that, compared with two strategies with heterogenous use of cephalosporins, penicillins, carbapenems and fluoroquinolones, cycling led to a significant increase in resistant nosocomial pneumonia pathogens [345]. Thus, with respect to antimicrobial classes, attention should be paid to use of balanced, guideline-adherent therapy. Above all, excessive use of cephalosporins and fluoroquinolones should be avoided. If surveillance data of cephalosporins and fluoroquinolones point to excessive consumption, a strategic class switch in preference of penicillins should be undertaken. There are several new publications on this topic. Following an educational campaign and subsequent introduction of restrictions, the policy “Reduction of routine use of ceftriaxone and ciprofloxacin in favour of aminopenicillins” was successfully implemented in a Scottish hospital. The endpoints observed were change in the incidence of hospital-acquired MRSA and ESBL-positive infections (without special screening) as well as C. difficile rates. As result of the new policy, consumption of ceftriaxone reduced by 95 % and that of ciprofloxacin by 73 % (comparison of the first and final 6 months of the study). At the same time, hospital-acquisition rates for C. difficile reduced by 77 %, MRSA by 25 % and ESBL cases by 17 %. The intervention had a sustained effect (up to 3 years later) [346]. Other observational studies on strategic antimicrobial substitution of cephalosporins in favour of penicillins conducted in China, Greece and India seem to confirm a decline in ESBL cases, however, cannot prove it [347350].

Special rules for communication of microbiology results

The guideline development group recommends:
The quality of microbiology diagnostics depends crucially on compliance with guidelines on procedures in the preanalytical phase. Expert consensus recommends that any deviations from protocol ought to be reported and the reasons for rejecting the samples stated (B).
Technical progress and up-to-date molecular diagnostic methods for rapid pathogen detection should be used if they improve the quality of care and/or substantially improve identification and epidemiologic investigation of local outbreaks (A).
Positive blood culture findings, interim microscopic findings, results of rapid testing and rapid susceptibility testing should be delivered promptly to the attending physician (A).
Antibiograms ought to adhere to local guidelines with respect to antimicrobial use and diagnostic findings, be presented selectively in agreement with the ABS team, and, if need be, include relevant interpretative comments. This procedure aids selection of a targeted, guideline-based antibiotic therapy (B).
The microbiology laboratory is responsible for the timely identification of trends in antimicrobial resistance and prompt communication of observations to the ABS team and the physicians responsible for infection control (A). This way, the clinical and epidemiological significance of the observations can be defined at an early stage.
The microbiology laboratory plays a crucial role in achieving the objectives of an antimicrobial stewardship programme by providing timely identification of relevant pathogens, selective antibiograms and active communication of diagnostic results and their interpretation, including interim reports. Microbiology diagnostics and susceptibility testing and reporting should be based on latest national [e.g. quality standards of microbiology and infectious diseases (MiQ) for Gemany] and international quality standards (http://​www.​eucast.​org), should address the specific requirements of the requesting physician and live up to the hospital’s obligation to provide medical care. A meaningful microbiological diagnostic requires optimal sample quality, storage, and timely transport of samples to the laboratory. The transport time of urine samples stored at room temperature should not exceed 2 h, since delays in transport of samples to the laboratory may increase pathogen growth and produce false-positive test results [351, 352]. Samples that deviate from guideline-adherent pre-analytics ought to be reported or ought to lead to the implementation of rejection criteria. Criteria ought to be defined for all common samples—e.g. sputum, urine, stool and swabs—to avoid unnecessary antimicrobial therapy. It is for instance recommended that purulent sputum with more than 25 squamous epithelial cells per field ought not to be further screened; rather, findings ought to be reported and the sputum sample discarded [353355]. Microbiological diagnostic of good-quality sputum samples can provide valuable information on the pathogen involved. Older studies investigating patients with pneumonia were able to show that high-quality sputum samples resulted in targeted antimicrobial monotherapy more often [356, 357] than inadequate sputum or no sputum culture. Reporting is also of importance on samples from non-implanted foreign bodies (drains, urinary catheters, venous catheters, tracheal cannula, wound sponge, etc.) or where low sample volume impairs sensitivity.
Automated MIC-based antibiograms and integration of molecular diagnostic assays such as PCR, PNA-FISH or MALDI-TOF into microbiology diagnostic can shorten time to pathogen identification and reporting of results [358]. Several prospective, randomised clinical studies on diagnostic of blood cultures and respiratory samples in pneumonia have shown that more rapid detection of pathogens by a few days resulted in earlier targeted antibiotic therapy. Thus, duration of empirical therapy, duration of ventilation and length of hospital stay were reduced by a few days and mortality decreased variably [36, 359366]. More recent prospective investigations on use of rapid Legionella urinary antigen test in pneumonia patients have shown that it is a useful tool with which targeted antimicrobial treatment can be provided more frequently. This was less conclusively demonstrated for pneumococcal antigen in urine [367, 368]. However, carefully developed and implemented algorithms are necessary to maintain the potential benefit. At least two controlled before-and-after studies have shown that antibiotic therapy is only modified and adjusted to microbiological findings, i.e. blood culture results, if these are communicated personally to the treating physician or the culture results are documented in the patient’s medical chart [369371].
The guideline development group recommends the use of selective reporting of susceptibility testing results with respect to choice and number of antimicrobial agents depending on pathogen, local susceptibility data and existing therapy guidelines, with the objective of supporting guideline-adherent antibiotic therapy [1, 6, 372]. Two methodologically different interrupted time-series analyses indicate that selective antibiotic susceptibility reporting can influence prescribing behaviour [373, 374]. Additional reporting, providing information on major resistance mechanisms, on contamination or colonisation according to pathogen and pathogen quantity, or information on diagnostic and therapeutic guidelines can support ABS measures. However, the effect of the mode of reporting microbiology results on prescribing behaviour is not well studied.
In case of unprecedented or critical levels of bacterial resistance, the microbiology laboratory should identify the cause by molecular biological methods as well as characterisation of clonal variants by typing, in particular during outbreaks and in this case especially with the assistance of reference laboratories. Depending on test results, targeted ABS strategies should be seized by the ABS team in accordance with appropriate hygiene interventions, implemented by the infection control team.

Special rules for management of patients with multidrug-resistant microorganisms and C. difficile

The guideline development group recommends:
ABS strategies should be used to prevent infection with C. difficile (A). Restricting use of certain antimicrobial drugs or substitution of antimicrobial drug classes (e.g. penicillin for cephalosporins or fluoroquinolones) can considerably reduce the incidence of C. difficile infection. Infection prevention and control strategies are frequently also applied at the same time; however, they have less impact on the C. difficile incidence than in the epidemiology of MRSA or VRE.
Targeted ABS strategies are to varying degrees also effective in reducing multidrug resistant Gram-negative bacteria, particularly ESBL-producing microorganisms, MRSA and VRE, and ought to be specifically applied here too (B). In case of high prevalence of multidrug resistant microorganisms, recommendations on diagnostic tests, evaluation of findings and treatment, as well as infection control management should be coordinated immediately and disseminated locally (A).
Routine surveillance of antimicrobial consumption and antimicrobial susceptibility data should be performed (A) to avoid indiscriminate compensatory use of other antimicrobial drug classes, since this can promote the unintentional and uncontrolled emergence of resistance.
The importance of appropriate ABS strategies for the management of patients with multidrug-resistant microorganisms and C. difficile is well documented in several systematic reviews and corresponding studies especially for C. difficile [2, 8, 22, 25, 375]. Pretreatment especially with third-generation cephalosporins and fluoroquinolones presents a risk factor for C. difficile infection, as well as for the increase in ESBL-producing Gram-negative microorganisms, MRSA and VRE [376, 377].
In before-and-after studies restriction of some types of antimicrobials, particularly third-generation cephalosporin and fluoroquinolones, but also macrolides and clindamycin, resulted in a 50 % or higher reduction in the incidence of C. difficile-associated disease. Frequently, interventions were accompanied by general infection control strategies. However, even in controlling outbreaks, infection control practices do not always appear to be sufficiently successful, as shown by the data collected retrospectively in a methodologically robust time-series analysis. It was not until restriction of cephalosporins, macrolides and clindamycin was imposed 6 months later that the incidence of C. difficile-associated disease decreased by 60 %. Utilisation of broad-spectrum penicillin subsequently increased compensatorily without repeated emergence of the epidemic C. difficile strain [73]. An investigation carried out over several years in a geriatric unit showed that the incidence of C. difficile diarrhoea was strongly associated with the density of cefotaxime use [378]. More recent time-series analyses over a period of 12–24 months confirm that substituting cephalosporins and fluoroquinolones for penicillins can lead to a decrease in the incidence of C. difficile-associated disease. Cephalosporin and fluoroquinolone use decreased considerably by more than 22–50 % [144, 146].
It is possible to influence the incidence of ESBL-positive isolates by implementing strategies to control antibiotic consumption. Various methodologically different but demanding studies on restriction of cephalosporin use showed a decrease in the rate of infection and colonisation with ESBL-producing microorganisms [349, 350, 379381]. However, in respect to sustained minimisation of resistance, the effects of a policy for targeted restriction of antimicrobials are less conclusive [124, 382] and sometimes even contradictory [383387]. Particularly, unplanned increased consumption of alternative agents, in setting of restriction, can rapidly impact negatively on the resistance situation [135, 388, 389]. The effect of restriction of third-generation cephalosporins, vancomycin and/or fluoroquinolones varied in regard to VRE and MRSA [117, 383385, 390, 391]. For instance, a short-term effect of restriction of vancomycin and cephalosporins on gastrointestinal colonisation with VRE (decrease from 47 to 15 %) was observed [392]. According to long-term observations, VRE eventually increased again [393]. Infection control practices seem to have a stronger sustained effect in this regard, and ABS strategies alone may not suffice.
In case of high incidence of multidrug-resistant microorganisms and cumulative outbreaks, appropriate recommendations on diagnostic tests, evaluation of findings and treatment, as well as infection control management must be coordinated immediately and disseminated locally. There is generally great uncertainty about optimal treatment [394]. In certain circumstances, use of unconventional antimicrobials or of conventional antimicrobials in unusual dose and combination may become necessary. In this situation, it is indispensible for the ABS team to draft appropriate guidance and recommendations in collaboration with the microbiology laboratory, to allow optimal treatment outcome and not to promote further spread of multidrug-resistant microorganisms through inadequate antibiotic use.

Computerised information technology

The guideline development group recommends:
The ABS team should be supported by novel information and communication technology in the implementation of ABS programmes. Local treatment guidelines, the antiinfective formulary, and other ABS documents should be available electronically (A).
Electronic prescribing tools with and without linkage to electronic preauthorisation solutions to ABS documents or to active communication of information using computerised reminders to the prescriber should be used to improve the use of antiinfectives in the interests of patient safety (A). They ought to be used to reduce consumption and/or costs (B).
Computerised decision support systems that are integrated into the hospital’s internal information system can, by utilising electronic medical records, help evaluate and optimise the indication for antiinfective therapy, drug selection and dosing (C).
To implement computerised ABS measures, the ABS team must have hospital-wide access rights to electronic medical records (with due respect to data protection).
The ABS team should be supported by novel computer-based information and communication technology through the provision of hospital-wide availability of ABS documents (antiinfective formularies, guidelines, treatment pathways). Development and application of electronic computerised decision support systems (CDSS) is to be encouraged. To utilise these systems optimally, it is useful to link them to an electronic patient record/chart or/and a patient-based computer physician order entry system (CPOE). In spite of the tremendous advances that have been made in the development of these systems in the hospital sector, availability varies in German and Austrian hospitals. Software designed specifically for ABS purposes hardly exists at all. Systematic reviews evaluating the impact of CDSSs specifically in the field of ABS (studies to 2007) found them to be of limited benefit [395, 396].
Electronic hospital information systems are used to varying extent in most hospitals in Germany. The system gives the treating physician access to patient-related and treatment-relevant data. In the interests of patient safety, and taking account of data protection, the ABS team should also have access to these data (antiinfectives, and microbiological laboratory results). Furthermore, electronic hospital-wide surveillance data on pathogens and antiinfective consumption should be available to the ABS team at all times. The ABS team should receive support on use and layout of computerised information systems from experts [397401].
Besides automated electronic alerts/short consultations and computerised decision support systems, in future, electronic prescribing systems, i.e. patient-based computer physician order entry systems (CPOE) with and without approval and reminder/alert functions will be particularly important for interventional ABS activities. As far as prescribing quality is concerned, these systems offer many advantages over paper-based medication orders with regard to legibility, completeness, fast delivery of information and possibly to approval or reminder/alert functions [402]; however, they are not as yet standard practice in German-speaking countries. By using CPOE systems, the rate of medication ordering errors can be reduced considerably (dosing, interactions, allergies) [403407]. On the other hand, the extent to which these systems influence resistance or clinical outcomes and mortality is inconclusive [408413]. In addition, CPOE systems could facilitate audits of antimicrobial prescribing through timely provision of patient-related, cross-unit and cross-departmental antimicrobial prescribing data [414, 415]. In a randomised controlled trial conducted in the USA, an ABS team [infectious diseases physician (50 % FTE), clinical pharmacist (80 % FTE)] reviewed antibiotic prescribing based on a list of alerts generated by an electronic decision support system. Among others, the following selection criteria were used: intravenous antimicrobial application in spite of good oral bioavailability, unnecessary combination therapy, and antibiogram discordant therapy. Compared with the control arm, USD 38 were saved per day per patient (around 16 % of total antibiotic costs) and 1 h of work a day with automated system alerts [416, 417]. A reduction in rates of antimicrobial use can be achieved by integrating an antimicrobial approval system into electronic prescribing [75, 243, 408, 409]. Web-based reviews of guideline-adherent prescribing of third-generation cephalosporins resulted in a significant 50 % reduction that was sustained over 15 months [75]. In another study, vancomycin use required an indication (by drop-down menu or free text) at initial electronic ordering and after 72 h. Treatment was stopped automatically if no indication was entered both times. As a result, initial vancomycin orders per physician were reduced significantly by 29 %, and 36 % fewer renewal orders were written after 72 h. The amount of days of vancomycin therapy decreased by 36 %. However, no information was provided on clinical outcome [418]. Electronic prescribing systems can contribute to reduce antibiotic expenses, if information on antibiotic cost/hospital day is provided while the order is placed [419]. This seems to be more effective than automated feedback to prescribers on antibiotic expenditure in comparison to others [420].
Electronic documentation of the timing of perioperative antibiotic prophylaxis with provider-specific feedback (confidentially to the anaesthetist) on the rate of prophylaxis given too early or too late appeared to be effective. At the time the intervention was initiated 69 % of the patients received antibiotics within 60 min of incision, and 92 % a year later [421]. Similar good-to-very good results were reported for the use of so-called electronic “Real-time Alerts” in improving timing of perioperative prophylaxis (>99 % of perioperative prophylaxis in one study). An electronic alert system was implemented after 4 h operating time to remind surgical teams to redose perioperative prophylactic antibiotics. The intervention was effective: 68 % of patients with cardiac surgery in the reminder group received intraoperative redosing versus only 40 % in the control group. The rate of surgical site infection was similar in both groups (4 und 6 %), but lower than in the pre-study period (10 %) [422, 423].
Several studies on electronic expert systems were conducted in a hospital in Salt Lake City (1986–2001). Implementation in many German hospitals, however, is difficult on account of inadequate technical prerequisites. These systems provide clinical decision support in the form of detailed therapy recommendations and alerts based on interlinked data such as allergies, laboratory results, microbiological findings, etc. Studies on this programme showed a significant reduction in the number of antibiotics prescribed, duration of treatment, costs, and adverse drug events [237, 424431]. A comprehensive computerised system consisting of an antimicrobial guideline and approval system in regard to reserve antibiotics also proved highly effective in an Australian hospital. It facilitated turning around a trend towards greater use of reserve antibiotics and resulted in increasing conformity with guideline-recommended therapies [432]. Effects were less strong in other studies. In a European multi-centre study (incl. Denmark, Italy, Germany) a computerised decision support system that looked at local susceptibility data, showed a rate of “appropriate” empirical antibiotic treatment of 73 %, which was only negligibly better than that of the control group of 64 % [433]. A reduction of just under 11 % in antibiotic consumption was achieved in an Australian intensive care unit, while the proportion of inappropriate antimicrobial use declined by only 10 % [412].
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.
insite
INHALT
download
DOWNLOAD
print
DRUCKEN

Unsere Produktempfehlungen

Neuer Inhalt

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

Weitere Produktempfehlungen anzeigen
Anhänge

Appendix

See Tables 7, 8 (Sect. 2.2.4).
Table 7
ABS structure indicators
https://static-content.springer.com/image/art%3A10.1007%2Fs15010-016-0885-z/MediaObjects/15010_2016_885_Tab7_HTML.gif
Table 8
ABS process indicators
https://static-content.springer.com/image/art%3A10.1007%2Fs15010-016-0885-z/MediaObjects/15010_2016_885_Tab8_HTML.gif
TEE transesophageal echocardiograph
aHACEK organisms are a group of Gram-negative pathogens that share an enhanced capacity to produce endocardial infections: Haemophilus species (H. parainfluenzae, H. aphrophilus, H. paraphrophilus), Actinobacillus actinomycetemcomitans, Cardiobacterium hominis, Eikenella corrodens, and Kingella species
Literatur
2.
Zurück zum Zitat Davey P, Brown E, Fenelon L, et al. Interventions to improve antibiotic prescribing practices for hospital inpatients. Cochrane Database Syst Rev. 2005;4:CD003543 (I).PubMed Davey P, Brown E, Fenelon L, et al. Interventions to improve antibiotic prescribing practices for hospital inpatients. Cochrane Database Syst Rev. 2005;4:CD003543 (I).PubMed
3.
Zurück zum Zitat Paterson DL. The role of antimicrobial management programs in optimizing antibiotic prescribing within hospitals. Clin Infect Dis. 2006;42:S90–5 (IV).PubMedCrossRef Paterson DL. The role of antimicrobial management programs in optimizing antibiotic prescribing within hospitals. Clin Infect Dis. 2006;42:S90–5 (IV).PubMedCrossRef
6.
Zurück zum Zitat Dellit TH, Owens RC, McGowan JE Jr, et al. Infectious Diseases Society of America and the Society for Healthcare Epidemiology of America guidelines for developing an institutional program to enhance antimicrobial stewardship. Clin Infect Dis. 2007;44:159–77 (I).PubMedCrossRef Dellit TH, Owens RC, McGowan JE Jr, et al. Infectious Diseases Society of America and the Society for Healthcare Epidemiology of America guidelines for developing an institutional program to enhance antimicrobial stewardship. Clin Infect Dis. 2007;44:159–77 (I).PubMedCrossRef
7.
Zurück zum Zitat Owens RC, Jr. Antimicrobial stewardship: concepts and strategies in the 21st century. Diagn Microbiol Infect Dis. 2008;61:110–28 (IV).PubMedCrossRef Owens RC, Jr. Antimicrobial stewardship: concepts and strategies in the 21st century. Diagn Microbiol Infect Dis. 2008;61:110–28 (IV).PubMedCrossRef
8.
Zurück zum Zitat Patel D, Lawson W, Guglielmo BJ. Antimicrobial stewardship programs: interventions and associated outcomes. Expert Rev Anti Infect Ther. 2008;6:209–22 (I).PubMedCrossRef Patel D, Lawson W, Guglielmo BJ. Antimicrobial stewardship programs: interventions and associated outcomes. Expert Rev Anti Infect Ther. 2008;6:209–22 (I).PubMedCrossRef
9.
Zurück zum Zitat Gould IM. Antibiotic policies to control hospital-acquired infection. J Antimicrob Chemother. 2008;61:763–5 (IV).PubMedCrossRef Gould IM. Antibiotic policies to control hospital-acquired infection. J Antimicrob Chemother. 2008;61:763–5 (IV).PubMedCrossRef
10.
Zurück zum Zitat Lesprit P, Brun-Buisson C. Hospital antibiotic stewardship. Curr Opin Infect Dis. 2008;21:344–9 (III).PubMedCrossRef Lesprit P, Brun-Buisson C. Hospital antibiotic stewardship. Curr Opin Infect Dis. 2008;21:344–9 (III).PubMedCrossRef
11.
Zurück zum Zitat Pagani L, Gyssens IC, Huttner B, Nathwani D, Harbarth S. Navigating the Web in search of resources on antimicrobial stewardship in health care institutions. Clin Infect Dis. 2009;48:626–32 (IV).PubMedCrossRef Pagani L, Gyssens IC, Huttner B, Nathwani D, Harbarth S. Navigating the Web in search of resources on antimicrobial stewardship in health care institutions. Clin Infect Dis. 2009;48:626–32 (IV).PubMedCrossRef
12.
Zurück zum Zitat Drew RH. Antimicrobial stewardship programs: how to start and steer a successful program. J Manag Care Pharm. 2009;15:S18–23 (IV).PubMed Drew RH. Antimicrobial stewardship programs: how to start and steer a successful program. J Manag Care Pharm. 2009;15:S18–23 (IV).PubMed
13.
Zurück zum Zitat Owens RC Jr. Antimicrobial stewardship: application in the intensive care unit. Infect Dis Clin North Am. 2009;23:683–702 (IV).PubMedCrossRef Owens RC Jr. Antimicrobial stewardship: application in the intensive care unit. Infect Dis Clin North Am. 2009;23:683–702 (IV).PubMedCrossRef
14.
Zurück zum Zitat Patel SJ, Saiman L. Principles and strategies of antimicrobial stewardship in the neonatal intensive care unit. Semin Perinatol. 2012;36:431–6 (IV).PubMedPubMedCentralCrossRef Patel SJ, Saiman L. Principles and strategies of antimicrobial stewardship in the neonatal intensive care unit. Semin Perinatol. 2012;36:431–6 (IV).PubMedPubMedCentralCrossRef
15.
Zurück zum Zitat Kaki R, Elligsen M, Walker S, Simor A, Palmay L, Daneman N. Impact of antimicrobial stewardship in critical care: a systematic review. J Antimicrob Chemother. 2011;66:1223–30 (I).PubMedCrossRef Kaki R, Elligsen M, Walker S, Simor A, Palmay L, Daneman N. Impact of antimicrobial stewardship in critical care: a systematic review. J Antimicrob Chemother. 2011;66:1223–30 (I).PubMedCrossRef
16.
Zurück zum Zitat Newland JG, Hersh AL. Purpose and design of antimicrobial stewardship programs in pediatrics. Pediatr Infect Dis J. 2010;29:862–3 (IV).PubMedCrossRef Newland JG, Hersh AL. Purpose and design of antimicrobial stewardship programs in pediatrics. Pediatr Infect Dis J. 2010;29:862–3 (IV).PubMedCrossRef
17.
Zurück zum Zitat Hersh AL, Beekmann SE, Polgreen PM, Zaoutis TE, Newland JG. Antimicrobial stewardship programs in pediatrics. Infect Control Hosp Epidemiol. 2009;30:1211–7 (IV).PubMedCrossRef Hersh AL, Beekmann SE, Polgreen PM, Zaoutis TE, Newland JG. Antimicrobial stewardship programs in pediatrics. Infect Control Hosp Epidemiol. 2009;30:1211–7 (IV).PubMedCrossRef
18.
Zurück zum Zitat Patel SJ, Larson EL, Kubin CJ, Saiman L. A review of antimicrobial control strategies in hospitalized and ambulatory pediatric populations. Pediatr Infect Dis J. 2007;26:531–7 (I).PubMedCrossRef Patel SJ, Larson EL, Kubin CJ, Saiman L. A review of antimicrobial control strategies in hospitalized and ambulatory pediatric populations. Pediatr Infect Dis J. 2007;26:531–7 (I).PubMedCrossRef
19.
Zurück zum Zitat Septimus EJ, Owens RC Jr. Need and potential of antimicrobial stewardship in community hospitals. Clin Infect Dis. 2011;53:S8–14 (IV).PubMedCrossRef Septimus EJ, Owens RC Jr. Need and potential of antimicrobial stewardship in community hospitals. Clin Infect Dis. 2011;53:S8–14 (IV).PubMedCrossRef
20.
Zurück zum Zitat Ohl CA, Dodds Ashley ES. Antimicrobial stewardship programs in community hospitals: the evidence base and case studies. Clin Infect Dis. 2011;53:S23–8 (IIa).PubMedCrossRef Ohl CA, Dodds Ashley ES. Antimicrobial stewardship programs in community hospitals: the evidence base and case studies. Clin Infect Dis. 2011;53:S23–8 (IIa).PubMedCrossRef
21.
Zurück zum Zitat Pate PG, Storey DF, Baum DL. Implementation of an antimicrobial stewardship program at a 60-bed long-term acute care hospital. Infect Control Hosp Epidemiol. 2012;33:405–8 (III).PubMedCrossRef Pate PG, Storey DF, Baum DL. Implementation of an antimicrobial stewardship program at a 60-bed long-term acute care hospital. Infect Control Hosp Epidemiol. 2012;33:405–8 (III).PubMedCrossRef
22.
Zurück zum Zitat Davey P, Brown E, Charani E, et al. Interventions to improve antibiotic prescribing practices for hospital inpatients. Cochrane Database Syst Rev. 2013;4:CD003543 (I).PubMed Davey P, Brown E, Charani E, et al. Interventions to improve antibiotic prescribing practices for hospital inpatients. Cochrane Database Syst Rev. 2013;4:CD003543 (I).PubMed
23.
Zurück zum Zitat Standiford HC, Chan S, Tripoli M, Weekes E, Forrest GN. Antimicrobial stewardship at a large tertiary care academic medical center: cost analysis before, during, and after a 7-year program. Infect Control Hosp Epidemiol. 2012;33:338–45 (II).PubMedCrossRef Standiford HC, Chan S, Tripoli M, Weekes E, Forrest GN. Antimicrobial stewardship at a large tertiary care academic medical center: cost analysis before, during, and after a 7-year program. Infect Control Hosp Epidemiol. 2012;33:338–45 (II).PubMedCrossRef
24.
Zurück zum Zitat Goldman MP, Nair R. Antibacterial treatment strategies in hospitalized patients: what role for pharmacoeconomics? Cleve Clin J Med. 2007;74:S38–47 (I).PubMedCrossRef Goldman MP, Nair R. Antibacterial treatment strategies in hospitalized patients: what role for pharmacoeconomics? Cleve Clin J Med. 2007;74:S38–47 (I).PubMedCrossRef
25.
26.
Zurück zum Zitat Knox K, Lawson W, Dean B, Holmes A. Multidisciplinary antimicrobial management and the role of the infectious diseases pharmacist—a UK perspective. J Hosp Infect. 2003;53:85–90 (I).PubMedCrossRef Knox K, Lawson W, Dean B, Holmes A. Multidisciplinary antimicrobial management and the role of the infectious diseases pharmacist—a UK perspective. J Hosp Infect. 2003;53:85–90 (I).PubMedCrossRef
27.
Zurück zum Zitat Gums JG, Yancey RW Jr, Hamilton CA, Kubilis PS. A randomized, prospective study measuring outcomes after antibiotic therapy intervention by a multidisciplinary consult team. Pharmacotherapy. 1999;19:1369–77 (I).PubMedCrossRef Gums JG, Yancey RW Jr, Hamilton CA, Kubilis PS. A randomized, prospective study measuring outcomes after antibiotic therapy intervention by a multidisciplinary consult team. Pharmacotherapy. 1999;19:1369–77 (I).PubMedCrossRef
28.
Zurück zum Zitat Solomon DH, Van HL, Glynn RJ, et al. Academic detailing to improve use of broad-spectrum antibiotics at an academic medical center. Arch Intern Med. 2001;161:1897–902 (I).PubMedCrossRef Solomon DH, Van HL, Glynn RJ, et al. Academic detailing to improve use of broad-spectrum antibiotics at an academic medical center. Arch Intern Med. 2001;161:1897–902 (I).PubMedCrossRef
29.
Zurück zum Zitat Carling P, Fung T, Killion A, Terrin N, Barza M. Favorable impact of a multidisciplinary antibiotic management program conducted during 7 years. Infect Control Hosp Epidemiol. 2003;24:699–706 (II).PubMedCrossRef Carling P, Fung T, Killion A, Terrin N, Barza M. Favorable impact of a multidisciplinary antibiotic management program conducted during 7 years. Infect Control Hosp Epidemiol. 2003;24:699–706 (II).PubMedCrossRef
30.
Zurück zum Zitat Bantar C, Franco D, Heft C, et al. Does a reduction in antibiotic consumption always represent a favorable outcome from an intervention program on prescribing practice? Int J Infect Dis. 2006;10:231–5 (II).PubMedCrossRef Bantar C, Franco D, Heft C, et al. Does a reduction in antibiotic consumption always represent a favorable outcome from an intervention program on prescribing practice? Int J Infect Dis. 2006;10:231–5 (II).PubMedCrossRef
31.
Zurück zum Zitat Bevilacqua S, Demore B, Erpelding ML, et al. Effects of an operational multidisciplinary team on hospital antibiotic use and cost in France: a cluster controlled trial. Int J Clin Pharm. 2011;33:521–8 (II).PubMedCrossRef Bevilacqua S, Demore B, Erpelding ML, et al. Effects of an operational multidisciplinary team on hospital antibiotic use and cost in France: a cluster controlled trial. Int J Clin Pharm. 2011;33:521–8 (II).PubMedCrossRef
32.
Zurück zum Zitat Danaher PJ, Milazzo NA, Kerr KJ, Lagasse CA, Lane JW. The antibiotic support team—a successful educational approach to antibiotic stewardship. Mil Med. 2009;174:201–5 (II).PubMedCrossRef Danaher PJ, Milazzo NA, Kerr KJ, Lagasse CA, Lane JW. The antibiotic support team—a successful educational approach to antibiotic stewardship. Mil Med. 2009;174:201–5 (II).PubMedCrossRef
33.
Zurück zum Zitat Diamantis S, Rioux C, Bonnal C, et al. Evaluation of initial antibiotic therapy for bacteremia and role of an antibiotic management team for antibiotic stewardship. Med Mal Infect. 2010;40:637–43 (II).PubMedCrossRef Diamantis S, Rioux C, Bonnal C, et al. Evaluation of initial antibiotic therapy for bacteremia and role of an antibiotic management team for antibiotic stewardship. Med Mal Infect. 2010;40:637–43 (II).PubMedCrossRef
34.
Zurück zum Zitat Gendrin V, Letranchant L, Henard S, et al. Impact of corrective measures on fluoroquinolones prescriptions for urinary tract infections during a 2-round relevance study. Presse Med. 2012;41:e10–4 (III).PubMedCrossRef Gendrin V, Letranchant L, Henard S, et al. Impact of corrective measures on fluoroquinolones prescriptions for urinary tract infections during a 2-round relevance study. Presse Med. 2012;41:e10–4 (III).PubMedCrossRef
35.
Zurück zum Zitat Bauer S, Bouldouyre MA, Oufella A, et al. Impact of a multidisciplinary staff meeting on the quality of antibiotherapy prescription for bone and joint infections in orthopedic surgery. Med Mal Infect. 2012;42:603–7 (III).PubMedCrossRef Bauer S, Bouldouyre MA, Oufella A, et al. Impact of a multidisciplinary staff meeting on the quality of antibiotherapy prescription for bone and joint infections in orthopedic surgery. Med Mal Infect. 2012;42:603–7 (III).PubMedCrossRef
36.
Zurück zum Zitat Byl B, Clevenbergh P, Jacobs F, et al. Impact of infectious diseases specialists and microbiological data on the appropriateness of antimicrobial therapy for bacteremia. Clin Infect Dis. 1999;29:60–6 (I).PubMedCrossRef Byl B, Clevenbergh P, Jacobs F, et al. Impact of infectious diseases specialists and microbiological data on the appropriateness of antimicrobial therapy for bacteremia. Clin Infect Dis. 1999;29:60–6 (I).PubMedCrossRef
37.
Zurück zum Zitat Dranitsaris G, Spizzirri D, Pitre M, McGeer A. A randomized trial to measure the optimal role of the pharmacist in promoting evidence-based antibiotic use in acute care hospitals. Int J Technol Assess Health Care. 2001;17:171–80 (I).PubMedCrossRef Dranitsaris G, Spizzirri D, Pitre M, McGeer A. A randomized trial to measure the optimal role of the pharmacist in promoting evidence-based antibiotic use in acute care hospitals. Int J Technol Assess Health Care. 2001;17:171–80 (I).PubMedCrossRef
38.
Zurück zum Zitat Bailey TC, Ritchie DJ, McMullin ST, et al. A randomized, prospective evaluation of an interventional program to discontinue intravenous antibiotics at two tertiary care teaching institutions. Pharmacotherapy. 1997;17:277–81 (I).PubMed Bailey TC, Ritchie DJ, McMullin ST, et al. A randomized, prospective evaluation of an interventional program to discontinue intravenous antibiotics at two tertiary care teaching institutions. Pharmacotherapy. 1997;17:277–81 (I).PubMed
39.
Zurück zum Zitat Dunn K, O’Reilly A, Silke B, Rogers T, Bergin C. Implementing a pharmacist-led sequential antimicrobial therapy strategy: a controlled before-and-after study. Int J Clin Pharm. 2011;33:208–14 (II).PubMedCrossRef Dunn K, O’Reilly A, Silke B, Rogers T, Bergin C. Implementing a pharmacist-led sequential antimicrobial therapy strategy: a controlled before-and-after study. Int J Clin Pharm. 2011;33:208–14 (II).PubMedCrossRef
40.
Zurück zum Zitat Walker SE. Physicians’ acceptance of a preformatted pharmacy intervention chart note in a community hospital antibiotic step-down program. J Pharm Technol. 1998;14:141–5 (I). Walker SE. Physicians’ acceptance of a preformatted pharmacy intervention chart note in a community hospital antibiotic step-down program. J Pharm Technol. 1998;14:141–5 (I).
41.
Zurück zum Zitat Wazana A. Physicians and the pharmaceutical industry: is a gift ever just a gift? JAMA. 2000;283:373–80 (I).PubMedCrossRef Wazana A. Physicians and the pharmaceutical industry: is a gift ever just a gift? JAMA. 2000;283:373–80 (I).PubMedCrossRef
42.
Zurück zum Zitat Zipkin DA, Steinman MA. Interactions between pharmaceutical representatives and doctors in training. A thematic review. J Gen Intern Med. 2005;20:777–86 (I).PubMedPubMedCentralCrossRef Zipkin DA, Steinman MA. Interactions between pharmaceutical representatives and doctors in training. A thematic review. J Gen Intern Med. 2005;20:777–86 (I).PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Cruickshank M, Duguid M. Antimicrobial stewardship in Australian hospitals 2011. Sydney: Australian Commission on Safety & Quality in Health Care; 2011(IV). Cruickshank M, Duguid M. Antimicrobial stewardship in Australian hospitals 2011. Sydney: Australian Commission on Safety & Quality in Health Care; 2011(IV).
44.
Zurück zum Zitat Petrak RM, Sexton DJ, Butera ML, et al. The value of an infectious diseases specialist. Clin Infect Dis. 2003;36:1013–7 IV.PubMedCrossRef Petrak RM, Sexton DJ, Butera ML, et al. The value of an infectious diseases specialist. Clin Infect Dis. 2003;36:1013–7 IV.PubMedCrossRef
45.
Zurück zum Zitat Fluckiger U, Zimmerli W, Sax H, Frei R, Widmer AF. Clinical impact of an infectious disease service on the management of bloodstream infection. Eur J Clin Microbiol Infect Dis. 2000;19:493–500 (II).PubMedCrossRef Fluckiger U, Zimmerli W, Sax H, Frei R, Widmer AF. Clinical impact of an infectious disease service on the management of bloodstream infection. Eur J Clin Microbiol Infect Dis. 2000;19:493–500 (II).PubMedCrossRef
46.
Zurück zum Zitat Kern WV. Management of Staphylococcus aureus bacteremia and endocarditis: progresses and challenges. Curr Opin Infect Dis. 2010;23:346–58 (II).PubMedCrossRef Kern WV. Management of Staphylococcus aureus bacteremia and endocarditis: progresses and challenges. Curr Opin Infect Dis. 2010;23:346–58 (II).PubMedCrossRef
47.
Zurück zum Zitat Honda H, Krauss MJ, Jones JC, Olsen MA, Warren DK. The value of infectious diseases consultation in Staphylococcus aureus bacteremia. Am J Med. 2010;123:631–7 (II).PubMedPubMedCentralCrossRef Honda H, Krauss MJ, Jones JC, Olsen MA, Warren DK. The value of infectious diseases consultation in Staphylococcus aureus bacteremia. Am J Med. 2010;123:631–7 (II).PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Gros H, Aslangul E, Lesprit P, Mainardi JL. Positive blood culture in hospital: notification methods and impact of recommendations by an infectious disease specialist. Med Mal Infect. 2012;42:76–9 (III).PubMedCrossRef Gros H, Aslangul E, Lesprit P, Mainardi JL. Positive blood culture in hospital: notification methods and impact of recommendations by an infectious disease specialist. Med Mal Infect. 2012;42:76–9 (III).PubMedCrossRef
49.
Zurück zum Zitat Schmitt S, McQuillen DP, Nahass R, et al. Infectious diseases specialty intervention is associated with decreased mortality and lower healthcare costs. Clin Infect Dis. 2014;58:22–28 (III). Schmitt S, McQuillen DP, Nahass R, et al. Infectious diseases specialty intervention is associated with decreased mortality and lower healthcare costs. Clin Infect Dis. 2014;58:22–28 (III).
50.
Zurück zum Zitat Raineri E, Pan A, Mondello P, Acquarolo A, Candiani A, Crema L. Role of the infectious diseases specialist consultant on the appropriateness of antimicrobial therapy prescription in an intensive care unit. Am J Infect Control. 2008;36:283–90 (II).PubMedCrossRef Raineri E, Pan A, Mondello P, Acquarolo A, Candiani A, Crema L. Role of the infectious diseases specialist consultant on the appropriateness of antimicrobial therapy prescription in an intensive care unit. Am J Infect Control. 2008;36:283–90 (II).PubMedCrossRef
51.
Zurück zum Zitat Borer A, Gilad J, Meydan N, Schlaeffer P, Riesenberg K, Schlaeffer F. Impact of regular attendance by infectious disease specialists on the management of hospitalised adults with community-acquired febrile syndromes. Clin Microbiol Infect. 2004;10:911–6 (II).PubMedCrossRef Borer A, Gilad J, Meydan N, Schlaeffer P, Riesenberg K, Schlaeffer F. Impact of regular attendance by infectious disease specialists on the management of hospitalised adults with community-acquired febrile syndromes. Clin Microbiol Infect. 2004;10:911–6 (II).PubMedCrossRef
52.
Zurück zum Zitat Della LP, Gherardi V, Pellegrino F, Cocchi I, Esposito R, Kiren V. Improving the appropriateness of antibiotic prescription in hospitals: a pilot study assessing the effectiveness of an infectious diseases specialist’s consultation programme. Int J Antimicrob Agents. 2008;31:488–9 (II).CrossRef Della LP, Gherardi V, Pellegrino F, Cocchi I, Esposito R, Kiren V. Improving the appropriateness of antibiotic prescription in hospitals: a pilot study assessing the effectiveness of an infectious diseases specialist’s consultation programme. Int J Antimicrob Agents. 2008;31:488–9 (II).CrossRef
53.
Zurück zum Zitat Camins BC, King MD, Wells JB, et al. Impact of an antimicrobial utilization program on antimicrobial use at a large teaching hospital: a randomized controlled trial. Infect Control Hosp Epidemiol. 2009;30:931–8 (I).PubMedPubMedCentralCrossRef Camins BC, King MD, Wells JB, et al. Impact of an antimicrobial utilization program on antimicrobial use at a large teaching hospital: a randomized controlled trial. Infect Control Hosp Epidemiol. 2009;30:931–8 (I).PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Kawanami GH, Fortaleza CM. Factors predictive of inappropriateness in requests for parenteral antimicrobials for therapeutic purposes: a study in a small teaching hospital in Brazil. Scand J Infect Dis. 2011;43:528–35 (III).PubMedCrossRef Kawanami GH, Fortaleza CM. Factors predictive of inappropriateness in requests for parenteral antimicrobials for therapeutic purposes: a study in a small teaching hospital in Brazil. Scand J Infect Dis. 2011;43:528–35 (III).PubMedCrossRef
55.
Zurück zum Zitat Pastel DA, Chang S, Nessim S, Shane R, Morgan MA. Department of pharmacy-initiated program for streamlining empirical antibiotic therapy. Hosp Pharm 1992;27:596–603, 614 (I). Pastel DA, Chang S, Nessim S, Shane R, Morgan MA. Department of pharmacy-initiated program for streamlining empirical antibiotic therapy. Hosp Pharm 1992;27:596–603, 614 (I).
56.
Zurück zum Zitat von Gunten V, Reymond JP, Beney J. Clinical and economic outcomes of pharmaceutical services related to antibiotic use: a literature review. Pharm World Sci. 2007;29:146–63 (I).CrossRef von Gunten V, Reymond JP, Beney J. Clinical and economic outcomes of pharmaceutical services related to antibiotic use: a literature review. Pharm World Sci. 2007;29:146–63 (I).CrossRef
57.
Zurück zum Zitat Toth NR, Chambers RM, Davis SL. Implementation of a care bundle for antimicrobial stewardship. Am J Health Syst Pharm. 2010;67:746–9 (II).PubMedCrossRef Toth NR, Chambers RM, Davis SL. Implementation of a care bundle for antimicrobial stewardship. Am J Health Syst Pharm. 2010;67:746–9 (II).PubMedCrossRef
58.
Zurück zum Zitat De RT, Willems L, Simoens S. Economic effects of clinical pharmacy interventions: a literature review. Am J Health Syst Pharm. 2008;65:1161–72 (I).CrossRef De RT, Willems L, Simoens S. Economic effects of clinical pharmacy interventions: a literature review. Am J Health Syst Pharm. 2008;65:1161–72 (I).CrossRef
59.
Zurück zum Zitat Ng CK, Wu TC, Chan WM, et al. Clinical and economic impact of an antibiotics stewardship programme in a regional hospital in Hong Kong. Qual Saf Health Care. 2008;17:387–92 (II).PubMedCrossRef Ng CK, Wu TC, Chan WM, et al. Clinical and economic impact of an antibiotics stewardship programme in a regional hospital in Hong Kong. Qual Saf Health Care. 2008;17:387–92 (II).PubMedCrossRef
60.
Zurück zum Zitat Tonna AP, Stewart D, West B, Gould I, McCaig D. Antimicrobial optimisation in secondary care: the pharmacist as part of a multidisciplinary antimicrobial programme—a literature review. Int J Antimicrob Agents. 2008;31:511–7 (I).PubMedCrossRef Tonna AP, Stewart D, West B, Gould I, McCaig D. Antimicrobial optimisation in secondary care: the pharmacist as part of a multidisciplinary antimicrobial programme—a literature review. Int J Antimicrob Agents. 2008;31:511–7 (I).PubMedCrossRef
61.
Zurück zum Zitat McLaughlin CM, Bodasing N, Boyter AC, Fenelon C, Fox JG, Seaton RA. Pharmacy-implemented guidelines on switching from intravenous to oral antibiotics: an intervention study. QJM. 2005;98:745–52 (II).PubMedCrossRef McLaughlin CM, Bodasing N, Boyter AC, Fenelon C, Fox JG, Seaton RA. Pharmacy-implemented guidelines on switching from intravenous to oral antibiotics: an intervention study. QJM. 2005;98:745–52 (II).PubMedCrossRef
62.
Zurück zum Zitat Weber A, Schneider C, Grill E, Strobl R, Vetter-Kerkhoff C, Jauch KW. Interventions by clinical pharmacists on surgical wards—impact on antibiotic therapy. Zentralbl Chir. 2011;136:66–73 (II).PubMedCrossRef Weber A, Schneider C, Grill E, Strobl R, Vetter-Kerkhoff C, Jauch KW. Interventions by clinical pharmacists on surgical wards—impact on antibiotic therapy. Zentralbl Chir. 2011;136:66–73 (II).PubMedCrossRef
63.
Zurück zum Zitat Grill E, Weber A, Lohmann S, Vetter-Kerkhoff C, Strobl R, Jauch KW. Effects of pharmaceutical counselling on antimicrobial use in surgical wards: intervention study with historical control group. Pharmacoepidemiol Drug Saf. 2011;20:739–46 III.PubMedCrossRef Grill E, Weber A, Lohmann S, Vetter-Kerkhoff C, Strobl R, Jauch KW. Effects of pharmaceutical counselling on antimicrobial use in surgical wards: intervention study with historical control group. Pharmacoepidemiol Drug Saf. 2011;20:739–46 III.PubMedCrossRef
64.
Zurück zum Zitat Yen YH, Chen HY, Wuan-Jin L, Lin YM, Shen WC, Cheng KJ. Clinical and economic impact of a pharmacist-managed i.v.-to-p.o. conversion service for levofloxacin in Taiwan. Int J Clin Pharmacol Ther. 2012;50:136–41 (III).PubMedCrossRef Yen YH, Chen HY, Wuan-Jin L, Lin YM, Shen WC, Cheng KJ. Clinical and economic impact of a pharmacist-managed i.v.-to-p.o. conversion service for levofloxacin in Taiwan. Int J Clin Pharmacol Ther. 2012;50:136–41 (III).PubMedCrossRef
65.
Zurück zum Zitat Van GE, Costers M, Peetermans WE, Struelens MJ. Nationwide implementation of antibiotic management teams in Belgian hospitals: a self-reporting survey. J Antimicrob Chemother. 2010;65:576–80 (IV).CrossRef Van GE, Costers M, Peetermans WE, Struelens MJ. Nationwide implementation of antibiotic management teams in Belgian hospitals: a self-reporting survey. J Antimicrob Chemother. 2010;65:576–80 (IV).CrossRef
66.
Zurück zum Zitat O’Neill E, Humphreys H, Smyth E. Impact of recommendations by clinical microbiologists on antimicrobial treatment in the intensive care units of a Dublin teaching hospital. Clin Infect Dis. 2005;40:636–7 (III).PubMedCrossRef O’Neill E, Humphreys H, Smyth E. Impact of recommendations by clinical microbiologists on antimicrobial treatment in the intensive care units of a Dublin teaching hospital. Clin Infect Dis. 2005;40:636–7 (III).PubMedCrossRef
67.
Zurück zum Zitat Kothari A, Sagar V, Panigrahi B, Selot N. Controlling costs in the intensive-care unit: role of daily microbiologist rounds in an Indian hospital. Clin Microbiol Infect. 2008;14:1187–8 (IV).PubMedCrossRef Kothari A, Sagar V, Panigrahi B, Selot N. Controlling costs in the intensive-care unit: role of daily microbiologist rounds in an Indian hospital. Clin Microbiol Infect. 2008;14:1187–8 (IV).PubMedCrossRef
68.
Zurück zum Zitat Fraser GL, Stogsdill P, Dickens JD Jr, Wennberg DE, Smith RP Jr, Prato BS. Antibiotic optimization. An evaluation of patient safety and economic outcomes. Arch Intern Med. 1997;157:1689–94 (I).PubMedCrossRef Fraser GL, Stogsdill P, Dickens JD Jr, Wennberg DE, Smith RP Jr, Prato BS. Antibiotic optimization. An evaluation of patient safety and economic outcomes. Arch Intern Med. 1997;157:1689–94 (I).PubMedCrossRef
69.
Zurück zum Zitat Philmon C, Smith T, Williamson S, Goodman E. Controlling use of antimicrobials in a community teaching hospital. Infect Control Hosp Epidemiol. 2006;27:239–44 (II).PubMedCrossRef Philmon C, Smith T, Williamson S, Goodman E. Controlling use of antimicrobials in a community teaching hospital. Infect Control Hosp Epidemiol. 2006;27:239–44 (II).PubMedCrossRef
70.
Zurück zum Zitat Uckay I, Vernaz-Hegi N, Harbarth S, et al. Activity and impact on antibiotic use and costs of a dedicated infectious diseases consultant on a septic orthopaedic unit. J Infect. 2009;58:205–12 (III).PubMedCrossRef Uckay I, Vernaz-Hegi N, Harbarth S, et al. Activity and impact on antibiotic use and costs of a dedicated infectious diseases consultant on a septic orthopaedic unit. J Infect. 2009;58:205–12 (III).PubMedCrossRef
71.
Zurück zum Zitat Beovic B, Kreft S, Seme K, Cizman M. The impact of total control of antibiotic prescribing by infectious disease specialist on antibiotic consumption and cost. J Chemother. 2009;21:46–51 (II).PubMedCrossRef Beovic B, Kreft S, Seme K, Cizman M. The impact of total control of antibiotic prescribing by infectious disease specialist on antibiotic consumption and cost. J Chemother. 2009;21:46–51 (II).PubMedCrossRef
72.
Zurück zum Zitat Larosa LA, Fishman NO, Lautenbach E, Koppel RJ, Morales KH, Linkin DR. Evaluation of antimicrobial therapy orders circumventing an antimicrobial stewardship program: investigating the strategy of “stealth dosing”. Infect Control Hosp Epidemiol. 2007;28:551–6 (III).PubMedPubMedCentralCrossRef Larosa LA, Fishman NO, Lautenbach E, Koppel RJ, Morales KH, Linkin DR. Evaluation of antimicrobial therapy orders circumventing an antimicrobial stewardship program: investigating the strategy of “stealth dosing”. Infect Control Hosp Epidemiol. 2007;28:551–6 (III).PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Valiquette L, Cossette B, Garant MP, Diab H, Pepin J. Impact of a reduction in the use of high-risk antibiotics on the course of an epidemic of Clostridium difficile-associated disease caused by the hypervirulent NAP1/027 strain. Clin Infect Dis. 2007;45:S112–21 (II).PubMedCrossRef Valiquette L, Cossette B, Garant MP, Diab H, Pepin J. Impact of a reduction in the use of high-risk antibiotics on the course of an epidemic of Clostridium difficile-associated disease caused by the hypervirulent NAP1/027 strain. Clin Infect Dis. 2007;45:S112–21 (II).PubMedCrossRef
74.
Zurück zum Zitat Allerberger F, Frank A, Gareis R. Antibiotic stewardship through the EU project “ABS International”. Wien Klin Wochenschr. 2008;120:256–63 (IV).PubMedCrossRef Allerberger F, Frank A, Gareis R. Antibiotic stewardship through the EU project “ABS International”. Wien Klin Wochenschr. 2008;120:256–63 (IV).PubMedCrossRef
75.
Zurück zum Zitat Richards MJ, Robertson MB, Dartnell JG, et al. Impact of a web-based antimicrobial approval system on broad-spectrum cephalosporin use at a teaching hospital. Med J Aust. 2003;178:386–90 (II).PubMed Richards MJ, Robertson MB, Dartnell JG, et al. Impact of a web-based antimicrobial approval system on broad-spectrum cephalosporin use at a teaching hospital. Med J Aust. 2003;178:386–90 (II).PubMed
76.
Zurück zum Zitat Schwartzberg E, Rubinovich S, Hassin D, et al. Developing and implementing a model for changing physicians’ prescribing habits—the role of clinical pharmacy in leading the change. J Clin Pharm Ther. 2006;31:179–85 (II).PubMedCrossRef Schwartzberg E, Rubinovich S, Hassin D, et al. Developing and implementing a model for changing physicians’ prescribing habits—the role of clinical pharmacy in leading the change. J Clin Pharm Ther. 2006;31:179–85 (II).PubMedCrossRef
77.
Zurück zum Zitat Wickens HJ, Jacklin A. Impact of the Hospital Pharmacy Initiative for promoting prudent use of antibiotics in hospitals in England. J Antimicrob Chemother. 2006;58:1230–7 (IV).PubMedCrossRef Wickens HJ, Jacklin A. Impact of the Hospital Pharmacy Initiative for promoting prudent use of antibiotics in hospitals in England. J Antimicrob Chemother. 2006;58:1230–7 (IV).PubMedCrossRef
78.
Zurück zum Zitat Biswal S, Mishra P, Malhotra S, Puri GD, Pandhi P. Drug utilization pattern in the intensive care unit of a tertiary care hospital. J Clin Pharmacol. 2006;46:945–51 (IV).PubMedCrossRef Biswal S, Mishra P, Malhotra S, Puri GD, Pandhi P. Drug utilization pattern in the intensive care unit of a tertiary care hospital. J Clin Pharmacol. 2006;46:945–51 (IV).PubMedCrossRef
79.
Zurück zum Zitat Burgmann H, Janata O, Allerberger F, Frank A. Hospital antibiotic management in Austria—results of the ABS maturity survey of the ABS International group. Wien Klin Wochenschr. 2008;120:280–3 (IV).PubMedCrossRef Burgmann H, Janata O, Allerberger F, Frank A. Hospital antibiotic management in Austria—results of the ABS maturity survey of the ABS International group. Wien Klin Wochenschr. 2008;120:280–3 (IV).PubMedCrossRef
80.
Zurück zum Zitat Dumpis U, Gulbinovic J, Struwe J, Lagergren A, Griskevicius L, Bergman U. Differences in antibiotic prescribing in three university hospitals in the Baltic region revealed by a simple protocol for quality assessment of therapeutic indications. Int J Clin Pharmacol Ther. 2007;45:568–76 (IV).PubMedCrossRef Dumpis U, Gulbinovic J, Struwe J, Lagergren A, Griskevicius L, Bergman U. Differences in antibiotic prescribing in three university hospitals in the Baltic region revealed by a simple protocol for quality assessment of therapeutic indications. Int J Clin Pharmacol Ther. 2007;45:568–76 (IV).PubMedCrossRef
81.
Zurück zum Zitat McNeil V, Cruickshank M, Duguid M. Safer use of antimicrobials in hospitals: the value of antimicrobial usage data. Med J Aust. 2010;193:S114–7 (IV).PubMed McNeil V, Cruickshank M, Duguid M. Safer use of antimicrobials in hospitals: the value of antimicrobial usage data. Med J Aust. 2010;193:S114–7 (IV).PubMed
82.
Zurück zum Zitat Schweickert B, Kern WV, de WK, et al. Surveillance of antibiotic consumption: clarification of the “definition of data on the nature and extent of antibiotic consumption in hospitals according to section sign 23 paragraph 4 sentence 2 of the IfSG”. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz. 2013;56:903–12 (IV).PubMedCrossRef Schweickert B, Kern WV, de WK, et al. Surveillance of antibiotic consumption: clarification of the “definition of data on the nature and extent of antibiotic consumption in hospitals according to section sign 23 paragraph 4 sentence 2 of the IfSG”. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz. 2013;56:903–12 (IV).PubMedCrossRef
83.
Zurück zum Zitat de WK, Maier L, Steib-Bauert M, Kern P, Kern WV. Trends in antibiotic use at a university hospital: defined or prescribed daily doses? Patient days or admissions as denominator? Infection. 2006;34:91–4 (IV).CrossRef de WK, Maier L, Steib-Bauert M, Kern P, Kern WV. Trends in antibiotic use at a university hospital: defined or prescribed daily doses? Patient days or admissions as denominator? Infection. 2006;34:91–4 (IV).CrossRef
84.
Zurück zum Zitat Cusini A, Rampini SK, Bansal V, et al. Different patterns of inappropriate antimicrobial use in surgical and medical units at a tertiary care hospital in Switzerland: a prevalence survey. PLoS One. 2010;5:e14011 (IV).PubMedPubMedCentralCrossRef Cusini A, Rampini SK, Bansal V, et al. Different patterns of inappropriate antimicrobial use in surgical and medical units at a tertiary care hospital in Switzerland: a prevalence survey. PLoS One. 2010;5:e14011 (IV).PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Zarb P, Goossens H. European Surveillance of Antimicrobial Consumption (ESAC): value of a point-prevalence survey of antimicrobial use across Europe. Drugs. 2011;71:745–55 (IV).PubMedCrossRef Zarb P, Goossens H. European Surveillance of Antimicrobial Consumption (ESAC): value of a point-prevalence survey of antimicrobial use across Europe. Drugs. 2011;71:745–55 (IV).PubMedCrossRef
86.
Zurück zum Zitat Zarb P, Amadeo B, Muller A, et al. Identification of targets for quality improvement in antimicrobial prescribing: the web-based ESAC Point Prevalence Survey 2009. J Antimicrob Chemother. 2011;66:443–9 (IV).PubMedCrossRef Zarb P, Amadeo B, Muller A, et al. Identification of targets for quality improvement in antimicrobial prescribing: the web-based ESAC Point Prevalence Survey 2009. J Antimicrob Chemother. 2011;66:443–9 (IV).PubMedCrossRef
87.
Zurück zum Zitat Ansari F, Erntell M, Goossens H, Davey P. The European surveillance of antimicrobial consumption (ESAC) point-prevalence survey of antibacterial use in 20 European hospitals in 2006. Clin Infect Dis. 2009;49:1496–504 (IV).PubMedCrossRef Ansari F, Erntell M, Goossens H, Davey P. The European surveillance of antimicrobial consumption (ESAC) point-prevalence survey of antibacterial use in 20 European hospitals in 2006. Clin Infect Dis. 2009;49:1496–504 (IV).PubMedCrossRef
88.
Zurück zum Zitat Pulver LK, Tett SE, Coombes J. The Queensland experience of participation in a national drug use evaluation project, community-acquired pneumonia towards improving outcomes nationally (CAPTION). BMC Pulm Med. 2009;9:38 (III).PubMedPubMedCentralCrossRef Pulver LK, Tett SE, Coombes J. The Queensland experience of participation in a national drug use evaluation project, community-acquired pneumonia towards improving outcomes nationally (CAPTION). BMC Pulm Med. 2009;9:38 (III).PubMedPubMedCentralCrossRef
89.
Zurück zum Zitat Lomas J, Anderson GM, Domnick-Pierre K, Vayda E, Enkin MW, Hannah WJ. Do practice guidelines guide practice? The effect of a consensus statement on the practice of physicians. N Engl J Med. 1989;321:1306–11 (II).PubMedCrossRef Lomas J, Anderson GM, Domnick-Pierre K, Vayda E, Enkin MW, Hannah WJ. Do practice guidelines guide practice? The effect of a consensus statement on the practice of physicians. N Engl J Med. 1989;321:1306–11 (II).PubMedCrossRef
90.
Zurück zum Zitat Martinez R, Reyes S, Lorenzo MJ, Menendez R. Impact of guidelines on outcome: the evidence. Semin Respir Crit Care Med. 2009;30:172–8 (I).PubMedCrossRef Martinez R, Reyes S, Lorenzo MJ, Menendez R. Impact of guidelines on outcome: the evidence. Semin Respir Crit Care Med. 2009;30:172–8 (I).PubMedCrossRef
91.
Zurück zum Zitat Bassi GL, Ferrer M, Saucedo LM, Torres A. Do guidelines change outcomes in ventilator-associated pneumonia? Curr Opin Infect Dis. 2010;23:171–7 (IV).PubMedCrossRef Bassi GL, Ferrer M, Saucedo LM, Torres A. Do guidelines change outcomes in ventilator-associated pneumonia? Curr Opin Infect Dis. 2010;23:171–7 (IV).PubMedCrossRef
92.
Zurück zum Zitat Dempsey CL. Nursing home-acquired pneumonia: outcomes from a clinical process improvement program. Pharmacotherapy. 1995;15:33S–8S (III).PubMed Dempsey CL. Nursing home-acquired pneumonia: outcomes from a clinical process improvement program. Pharmacotherapy. 1995;15:33S–8S (III).PubMed
93.
Zurück zum Zitat Marrie TJ, Lau CY, Wheeler SL, Wong CJ, Vandervoort MK, Feagan BG, CAPITAL Study Investigators. A controlled trial of a critical pathway for treatment of community-acquired pneumonia. Community-acquired pneumonia intervention trial assessing levofloxacin. JAMA. 2000;283:749–55 (I).PubMedCrossRef Marrie TJ, Lau CY, Wheeler SL, Wong CJ, Vandervoort MK, Feagan BG, CAPITAL Study Investigators. A controlled trial of a critical pathway for treatment of community-acquired pneumonia. Community-acquired pneumonia intervention trial assessing levofloxacin. JAMA. 2000;283:749–55 (I).PubMedCrossRef
94.
Zurück zum Zitat Dean NC, Silver MP, Bateman KA, James B, Hadlock CJ, Hale D. Decreased mortality after implementation of a treatment guideline for community-acquired pneumonia. Am J Med. 2001;110:451–7 (II).PubMedCrossRef Dean NC, Silver MP, Bateman KA, James B, Hadlock CJ, Hale D. Decreased mortality after implementation of a treatment guideline for community-acquired pneumonia. Am J Med. 2001;110:451–7 (II).PubMedCrossRef
95.
Zurück zum Zitat Dean NC, Bateman KA, Donnelly SM, Silver MP, Snow GL, Hale D. Improved clinical outcomes with utilization of a community-acquired pneumonia guideline. Chest. 2006;130:794–9 (II).PubMedCrossRef Dean NC, Bateman KA, Donnelly SM, Silver MP, Snow GL, Hale D. Improved clinical outcomes with utilization of a community-acquired pneumonia guideline. Chest. 2006;130:794–9 (II).PubMedCrossRef
96.
Zurück zum Zitat Price J, Ekleberry A, Grover A, et al. Evaluation of clinical practice guidelines on outcome of infection in patients in the surgical intensive care unit. Crit Care Med. 1999;27:2118–24 (II).PubMedCrossRef Price J, Ekleberry A, Grover A, et al. Evaluation of clinical practice guidelines on outcome of infection in patients in the surgical intensive care unit. Crit Care Med. 1999;27:2118–24 (II).PubMedCrossRef
97.
Zurück zum Zitat Ibrahim EH, Ward S, Sherman G, Schaiff R, Fraser VJ, Kollef MH. Experience with a clinical guideline for the treatment of ventilator-associated pneumonia. Crit Care Med. 2001;29:1109–15 (II).PubMedCrossRef Ibrahim EH, Ward S, Sherman G, Schaiff R, Fraser VJ, Kollef MH. Experience with a clinical guideline for the treatment of ventilator-associated pneumonia. Crit Care Med. 2001;29:1109–15 (II).PubMedCrossRef
98.
Zurück zum Zitat Barlow G, Nathwani D, Williams F, et al. Reducing door-to-antibiotic time in community-acquired pneumonia: controlled before-and-after evaluation and cost-effectiveness analysis. Thorax. 2007;62:67–74 (II).PubMedPubMedCentralCrossRef Barlow G, Nathwani D, Williams F, et al. Reducing door-to-antibiotic time in community-acquired pneumonia: controlled before-and-after evaluation and cost-effectiveness analysis. Thorax. 2007;62:67–74 (II).PubMedPubMedCentralCrossRef
99.
Zurück zum Zitat Kollef MH, Sherman G, Ward S, Fraser VJ. Inadequate antimicrobial treatment of infections: a risk factor for hospital mortality among critically ill patients. Chest. 1999;115:462–74 (IV).PubMedCrossRef Kollef MH, Sherman G, Ward S, Fraser VJ. Inadequate antimicrobial treatment of infections: a risk factor for hospital mortality among critically ill patients. Chest. 1999;115:462–74 (IV).PubMedCrossRef
100.
Zurück zum Zitat Fagon JY, Chastre J, Wolff M, et al. Invasive and noninvasive strategies for management of suspected ventilator-associated pneumonia. A randomized trial. Ann Intern Med. 2000;132:621–30 (I).PubMedCrossRef Fagon JY, Chastre J, Wolff M, et al. Invasive and noninvasive strategies for management of suspected ventilator-associated pneumonia. A randomized trial. Ann Intern Med. 2000;132:621–30 (I).PubMedCrossRef
101.
Zurück zum Zitat Chastre J, Wolff M, Fagon JY, et al. Comparison of 8 vs 15 days of antibiotic therapy for ventilator-associated pneumonia in adults: a randomized trial. JAMA. 2003;290:2588–98 (I).PubMedCrossRef Chastre J, Wolff M, Fagon JY, et al. Comparison of 8 vs 15 days of antibiotic therapy for ventilator-associated pneumonia in adults: a randomized trial. JAMA. 2003;290:2588–98 (I).PubMedCrossRef
102.
Zurück zum Zitat Singh N, Rogers P, Atwood CW, Wagener MM, Yu VL. Short-course empiric antibiotic therapy for patients with pulmonary infiltrates in the intensive care unit. A proposed solution for indiscriminate antibiotic prescription. Am J Respir Crit Care Med. 2000;162:505–11 (I).PubMedCrossRef Singh N, Rogers P, Atwood CW, Wagener MM, Yu VL. Short-course empiric antibiotic therapy for patients with pulmonary infiltrates in the intensive care unit. A proposed solution for indiscriminate antibiotic prescription. Am J Respir Crit Care Med. 2000;162:505–11 (I).PubMedCrossRef
103.
Zurück zum Zitat Soo Hoo GW, Wen YE, Nguyen TV, Goetz MB. Impact of clinical guidelines in the management of severe hospital-acquired pneumonia. Chest. 2005;128:2778–87 (II).PubMedCrossRef Soo Hoo GW, Wen YE, Nguyen TV, Goetz MB. Impact of clinical guidelines in the management of severe hospital-acquired pneumonia. Chest. 2005;128:2778–87 (II).PubMedCrossRef
104.
Zurück zum Zitat Botelho-Nevers E, Thuny F, Casalta JP, et al. Dramatic reduction in infective endocarditis-related mortality with a management-based approach. Arch Intern Med. 2009;169:1290–8 (II).PubMedCrossRef Botelho-Nevers E, Thuny F, Casalta JP, et al. Dramatic reduction in infective endocarditis-related mortality with a management-based approach. Arch Intern Med. 2009;169:1290–8 (II).PubMedCrossRef
105.
Zurück zum Zitat Schouten JA, Hulscher ME, Trap-Liefers J, et al. Tailored interventions to improve antibiotic use for lower respiratory tract infections in hospitals: a cluster-randomized, controlled trial. Clin Infect Dis. 2007;44:931–41 (I).PubMedCrossRef Schouten JA, Hulscher ME, Trap-Liefers J, et al. Tailored interventions to improve antibiotic use for lower respiratory tract infections in hospitals: a cluster-randomized, controlled trial. Clin Infect Dis. 2007;44:931–41 (I).PubMedCrossRef
106.
Zurück zum Zitat Jenkins TC, Knepper BC, Sabel AL, et al. Decreased antibiotic utilization after implementation of a guideline for inpatient cellulitis and cutaneous abscess. Arch Intern Med. 2011;171:1072–9 (III).PubMed Jenkins TC, Knepper BC, Sabel AL, et al. Decreased antibiotic utilization after implementation of a guideline for inpatient cellulitis and cutaneous abscess. Arch Intern Med. 2011;171:1072–9 (III).PubMed
107.
Zurück zum Zitat Grenier C, Pepin J, Nault V, et al. Impact of guideline-consistent therapy on outcome of patients with healthcare-associated and community-acquired pneumonia. J Antimicrob Chemother. 2011;66:1617–24 (III).PubMedCrossRef Grenier C, Pepin J, Nault V, et al. Impact of guideline-consistent therapy on outcome of patients with healthcare-associated and community-acquired pneumonia. J Antimicrob Chemother. 2011;66:1617–24 (III).PubMedCrossRef
108.
Zurück zum Zitat Thornhill MH, Dayer MJ, Forde JM, et al. Impact of the NICE guideline recommending cessation of antibiotic prophylaxis for prevention of infective endocarditis: before and after study. BMJ. 2011;342:d2392 (II).PubMedPubMedCentralCrossRef Thornhill MH, Dayer MJ, Forde JM, et al. Impact of the NICE guideline recommending cessation of antibiotic prophylaxis for prevention of infective endocarditis: before and after study. BMJ. 2011;342:d2392 (II).PubMedPubMedCentralCrossRef
109.
Zurück zum Zitat Westphal JF, Jehl F, Javelot H, Nonnenmacher C. Enhanced physician adherence to antibiotic use guidelines through increased availability of guidelines at the time of drug ordering in hospital setting. Pharmacoepidemiol Drug Saf. 2011;20:162–8 (III).PubMedCrossRef Westphal JF, Jehl F, Javelot H, Nonnenmacher C. Enhanced physician adherence to antibiotic use guidelines through increased availability of guidelines at the time of drug ordering in hospital setting. Pharmacoepidemiol Drug Saf. 2011;20:162–8 (III).PubMedCrossRef
110.
Zurück zum Zitat Schnoor M, Meyer T, Suttorp N, Raspe H, Welte T, Schafer T. Development and evaluation of an implementation strategy for the German guideline on community-acquired pneumonia. Qual Saf Health Care. 2010;19:498–502 (II).PubMed Schnoor M, Meyer T, Suttorp N, Raspe H, Welte T, Schafer T. Development and evaluation of an implementation strategy for the German guideline on community-acquired pneumonia. Qual Saf Health Care. 2010;19:498–502 (II).PubMed
111.
Zurück zum Zitat Lancaster JW, Lawrence KR, Fong JJ, et al. Impact of an institution-specific hospital-acquired pneumonia protocol on the appropriateness of antibiotic therapy and patient outcomes. Pharmacotherapy. 2008;28:852–62 (II).PubMedCrossRef Lancaster JW, Lawrence KR, Fong JJ, et al. Impact of an institution-specific hospital-acquired pneumonia protocol on the appropriateness of antibiotic therapy and patient outcomes. Pharmacotherapy. 2008;28:852–62 (II).PubMedCrossRef
112.
Zurück zum Zitat McIntosh KA, Maxwell DJ, Pulver LK, et al. A quality improvement initiative to improve adherence to national guidelines for empiric management of community-acquired pneumonia in emergency departments. Int J Qual Health Care. 2011;23:142–50 (III).PubMedCrossRef McIntosh KA, Maxwell DJ, Pulver LK, et al. A quality improvement initiative to improve adherence to national guidelines for empiric management of community-acquired pneumonia in emergency departments. Int J Qual Health Care. 2011;23:142–50 (III).PubMedCrossRef
113.
Zurück zum Zitat Chalmers JD, Singanayagam A, Akram AR, Choudhury G, Mandal P, Hill AT. Safety and efficacy of CURB65-guided antibiotic therapy in community-acquired pneumonia. J Antimicrob Chemother. 2011;66:416–23 (II).PubMedCrossRef Chalmers JD, Singanayagam A, Akram AR, Choudhury G, Mandal P, Hill AT. Safety and efficacy of CURB65-guided antibiotic therapy in community-acquired pneumonia. J Antimicrob Chemother. 2011;66:416–23 (II).PubMedCrossRef
114.
Zurück zum Zitat Mol PG, Wieringa JE, Nannanpanday PV, et al. Improving compliance with hospital antibiotic guidelines: a time-series intervention analysis. J Antimicrob Chemother. 2005;55:550–7 (II).PubMedCrossRef Mol PG, Wieringa JE, Nannanpanday PV, et al. Improving compliance with hospital antibiotic guidelines: a time-series intervention analysis. J Antimicrob Chemother. 2005;55:550–7 (II).PubMedCrossRef
115.
Zurück zum Zitat Willemsen I, van den BR, Bijsterveldt T, et al. A standardized protocol for perioperative antibiotic prophylaxis is associated with improvement of timing and reduction of costs. J Hosp Infect. 2007;67:156–60 (II).PubMedCrossRef Willemsen I, van den BR, Bijsterveldt T, et al. A standardized protocol for perioperative antibiotic prophylaxis is associated with improvement of timing and reduction of costs. J Hosp Infect. 2007;67:156–60 (II).PubMedCrossRef
116.
Zurück zum Zitat Winters BD, Thiemann DR, Brotman DJ. Impact of a restrictive antimicrobial policy on the process and timing of antimicrobial administration. J Hosp Med. 2010;5:E41–5 (III).PubMedCrossRef Winters BD, Thiemann DR, Brotman DJ. Impact of a restrictive antimicrobial policy on the process and timing of antimicrobial administration. J Hosp Med. 2010;5:E41–5 (III).PubMedCrossRef
117.
Zurück zum Zitat Madaras-Kelly KJ, Remington RE, Lewis PG, Stevens DL. Evaluation of an intervention designed to decrease the rate of nosocomial methicillin-resistant Staphylococcus aureus infection by encouraging decreased fluoroquinolone use. Infect Control Hosp Epidemiol. 2006;27:155–69 (II).PubMedCrossRef Madaras-Kelly KJ, Remington RE, Lewis PG, Stevens DL. Evaluation of an intervention designed to decrease the rate of nosocomial methicillin-resistant Staphylococcus aureus infection by encouraging decreased fluoroquinolone use. Infect Control Hosp Epidemiol. 2006;27:155–69 (II).PubMedCrossRef
118.
Zurück zum Zitat Church EC, Mauldin PD, Bosso JA. Antibiotic resistance in Pseudomonas aeruginosa related to quinolone formulary changes: an interrupted time series analysis. Infect Control Hosp Epidemiol. 2011;32:400–2 (II).PubMedCrossRef Church EC, Mauldin PD, Bosso JA. Antibiotic resistance in Pseudomonas aeruginosa related to quinolone formulary changes: an interrupted time series analysis. Infect Control Hosp Epidemiol. 2011;32:400–2 (II).PubMedCrossRef
119.
Zurück zum Zitat Bassetti M, Righi E, Ansaldi F, et al. Impact of limited cephalosporin use on prevalence of methicillin-resistant Staphylococcus aureus in the intensive care unit. J Chemother. 2009;21:633–8 (II).PubMedCrossRef Bassetti M, Righi E, Ansaldi F, et al. Impact of limited cephalosporin use on prevalence of methicillin-resistant Staphylococcus aureus in the intensive care unit. J Chemother. 2009;21:633–8 (II).PubMedCrossRef
120.
Zurück zum Zitat Britton HL, Schwinghammer TL, Romano MJ. Cost containment through restriction of cephalosporins. Am J Hosp Pharm. 1981;38:1897–900 (II).PubMed Britton HL, Schwinghammer TL, Romano MJ. Cost containment through restriction of cephalosporins. Am J Hosp Pharm. 1981;38:1897–900 (II).PubMed
121.
Zurück zum Zitat Hayman JN, Sbravati EC. Controlling cephalosporin and aminoglycoside costs through pharmacy and therapeutics committee restrictions. Am J Hosp Pharm. 1985;42:1343–7 (II).PubMed Hayman JN, Sbravati EC. Controlling cephalosporin and aminoglycoside costs through pharmacy and therapeutics committee restrictions. Am J Hosp Pharm. 1985;42:1343–7 (II).PubMed
122.
Zurück zum Zitat Woodward RS, Medoff G, Smith MD, Gray JL III. Antibiotic cost savings from formulary restrictions and physician monitoring in a medical-school-affiliated hospital. Am J Med. 1987;83:817–23 (III).PubMedCrossRef Woodward RS, Medoff G, Smith MD, Gray JL III. Antibiotic cost savings from formulary restrictions and physician monitoring in a medical-school-affiliated hospital. Am J Med. 1987;83:817–23 (III).PubMedCrossRef
123.
Zurück zum Zitat Maswoswe JJ, Okpara AU. Enforcing a policy for restricting antimicrobial drug use. Am J Health Syst Pharm. 1995;52:1433–5 (III).PubMed Maswoswe JJ, Okpara AU. Enforcing a policy for restricting antimicrobial drug use. Am J Health Syst Pharm. 1995;52:1433–5 (III).PubMed
124.
Zurück zum Zitat White AC, Atmar RL, Wilson J, Cate TR, Stager CE, Greenberg SB. Effects of requiring prior authorization for selected antimicrobials: expenditures, susceptibilities, and clinical outcomes. Clin Infect Dis. 1997;25:230–9 (II).PubMedCrossRef White AC, Atmar RL, Wilson J, Cate TR, Stager CE, Greenberg SB. Effects of requiring prior authorization for selected antimicrobials: expenditures, susceptibilities, and clinical outcomes. Clin Infect Dis. 1997;25:230–9 (II).PubMedCrossRef
125.
Zurück zum Zitat Siddiqui S, Hussein K, Manasia R, et al. Impact of antibiotic restriction on broad spectrum antibiotic usage in the ICU of a developing country. J Pak Med Assoc. 2007;57:484–7 (III).PubMed Siddiqui S, Hussein K, Manasia R, et al. Impact of antibiotic restriction on broad spectrum antibiotic usage in the ICU of a developing country. J Pak Med Assoc. 2007;57:484–7 (III).PubMed
126.
Zurück zum Zitat Tunger O, Karakaya Y, Cetin CB, Dinc G, Borand H. Rational antibiotic use. J Infect Dev Ctries. 2009;3:88–93 (III).PubMedCrossRef Tunger O, Karakaya Y, Cetin CB, Dinc G, Borand H. Rational antibiotic use. J Infect Dev Ctries. 2009;3:88–93 (III).PubMedCrossRef
127.
Zurück zum Zitat Altunsoy A, Aypak C, Azap A, Ergonul O, Balik I. The impact of a nationwide antibiotic restriction program on antibiotic usage and resistance against nosocomial pathogens in Turkey. Int J Med Sci. 2011;8:339–44 (III).PubMedPubMedCentralCrossRef Altunsoy A, Aypak C, Azap A, Ergonul O, Balik I. The impact of a nationwide antibiotic restriction program on antibiotic usage and resistance against nosocomial pathogens in Turkey. Int J Med Sci. 2011;8:339–44 (III).PubMedPubMedCentralCrossRef
128.
Zurück zum Zitat Gomez MI, Acosta-Gnass SI, Mosqueda-Barboza L, Basualdo JA. Reduction in surgical antibiotic prophylaxis expenditure and the rate of surgical site infection by means of a protocol that controls the use of prophylaxis. Infect Control Hosp Epidemiol. 2006;27:1358–65 (II).PubMedCrossRef Gomez MI, Acosta-Gnass SI, Mosqueda-Barboza L, Basualdo JA. Reduction in surgical antibiotic prophylaxis expenditure and the rate of surgical site infection by means of a protocol that controls the use of prophylaxis. Infect Control Hosp Epidemiol. 2006;27:1358–65 (II).PubMedCrossRef
129.
Zurück zum Zitat Hermsen ED, Smith SS, Puumala SE, Rupp ME. Improvement in prescribing habits and economic outcomes associated with the introduction of a standardized approach for surgical antimicrobial prophylaxis. Infect Control Hosp Epidemiol. 2008;29:457–61 (II).PubMedCrossRef Hermsen ED, Smith SS, Puumala SE, Rupp ME. Improvement in prescribing habits and economic outcomes associated with the introduction of a standardized approach for surgical antimicrobial prophylaxis. Infect Control Hosp Epidemiol. 2008;29:457–61 (II).PubMedCrossRef
130.
Zurück zum Zitat Parker BM, Henderson JM, Vitagliano S, et al. Six sigma methodology can be used to improve adherence for antibiotic prophylaxis in patients undergoing noncardiac surgery. Anesth Analg. 2007;104:140–6 (II).PubMedCrossRef Parker BM, Henderson JM, Vitagliano S, et al. Six sigma methodology can be used to improve adherence for antibiotic prophylaxis in patients undergoing noncardiac surgery. Anesth Analg. 2007;104:140–6 (II).PubMedCrossRef
131.
Zurück zum Zitat Perez A, Dennis RJ, Rodriguez B, et al. An interrupted time series analysis of parenteral antibiotic use in Colombia. J Clin Epidemiol. 2003;56:1013–20 (II).PubMedCrossRef Perez A, Dennis RJ, Rodriguez B, et al. An interrupted time series analysis of parenteral antibiotic use in Colombia. J Clin Epidemiol. 2003;56:1013–20 (II).PubMedCrossRef
132.
Zurück zum Zitat Salama S, Rotstein C, Mandell L. A multidisciplinary hospital-based antimicrobial use program: impact on hospital pharmacy expenditures and drug use. Can J Infect Dis. 1996;7:104–9 (II).PubMedPubMedCentralCrossRef Salama S, Rotstein C, Mandell L. A multidisciplinary hospital-based antimicrobial use program: impact on hospital pharmacy expenditures and drug use. Can J Infect Dis. 1996;7:104–9 (II).PubMedPubMedCentralCrossRef
133.
Zurück zum Zitat Belliveau PP, Rothman AL, Maday CE. Limiting vancomycin use to combat vancomycin-resistant Enterococcus faecium. Am J Health Syst Pharm. 1996;53:1570–5 (II).PubMed Belliveau PP, Rothman AL, Maday CE. Limiting vancomycin use to combat vancomycin-resistant Enterococcus faecium. Am J Health Syst Pharm. 1996;53:1570–5 (II).PubMed
134.
Zurück zum Zitat Marra AR, de Almeida SM, Correa L, et al. The effect of limiting antimicrobial therapy duration on antimicrobial resistance in the critical care setting. Am J Infect Control. 2009;37:204–9 (II). Marra AR, de Almeida SM, Correa L, et al. The effect of limiting antimicrobial therapy duration on antimicrobial resistance in the critical care setting. Am J Infect Control. 2009;37:204–9 (II).
135.
Zurück zum Zitat Falagas ME, Bliziotis IA, Michalopoulos A, et al. Effect of a policy for restriction of selected classes of antibiotics on antimicrobial drug cost and resistance. J Chemother. 2007;19:178–84 (II).PubMedCrossRef Falagas ME, Bliziotis IA, Michalopoulos A, et al. Effect of a policy for restriction of selected classes of antibiotics on antimicrobial drug cost and resistance. J Chemother. 2007;19:178–84 (II).PubMedCrossRef
136.
Zurück zum Zitat Lewis GJ, Fang X, Gooch M, Cook PP. Decreased resistance of Pseudomonas aeruginosa with restriction of ciprofloxacin in a large teaching hospital’s intensive care and intermediate care units. Infect Control Hosp Epidemiol. 2012;33:368–73 (II).PubMedCrossRef Lewis GJ, Fang X, Gooch M, Cook PP. Decreased resistance of Pseudomonas aeruginosa with restriction of ciprofloxacin in a large teaching hospital’s intensive care and intermediate care units. Infect Control Hosp Epidemiol. 2012;33:368–73 (II).PubMedCrossRef
137.
Zurück zum Zitat Pepin J, Saheb N, Coulombe MA, et al. Emergence of fluoroquinolones as the predominant risk factor for Clostridium difficile-associated diarrhea: a cohort study during an epidemic in Quebec. Clin Infect Dis. 2005;41:1254–60 (III).PubMedCrossRef Pepin J, Saheb N, Coulombe MA, et al. Emergence of fluoroquinolones as the predominant risk factor for Clostridium difficile-associated diarrhea: a cohort study during an epidemic in Quebec. Clin Infect Dis. 2005;41:1254–60 (III).PubMedCrossRef
138.
Zurück zum Zitat LeBlanc L, Pepin J, Toulouse K, et al. Fluoroquinolones and risk for methicillin-resistant Staphylococcus aureus, Canada. Emerg Infect Dis. 2006;12:1398–405 (III).PubMedPubMedCentralCrossRef LeBlanc L, Pepin J, Toulouse K, et al. Fluoroquinolones and risk for methicillin-resistant Staphylococcus aureus, Canada. Emerg Infect Dis. 2006;12:1398–405 (III).PubMedPubMedCentralCrossRef
139.
Zurück zum Zitat Weber SG, Gold HS, Hooper DC, Karchmer AW, Carmeli Y. Fluoroquinolones and the risk for methicillin-resistant Staphylococcus aureus in hospitalized patients. Emerg Infect Dis. 2003;9:1415–22 (III).PubMedPubMedCentralCrossRef Weber SG, Gold HS, Hooper DC, Karchmer AW, Carmeli Y. Fluoroquinolones and the risk for methicillin-resistant Staphylococcus aureus in hospitalized patients. Emerg Infect Dis. 2003;9:1415–22 (III).PubMedPubMedCentralCrossRef
140.
Zurück zum Zitat MacDougall C, Powell JP, Johnson CK, Edmond MB, Polk RE. Hospital and community fluoroquinolone use and resistance in Staphylococcus aureus and Escherichia coli in 17 US hospitals. Clin Infect Dis. 2005;41:435–40 (III).PubMedCrossRef MacDougall C, Powell JP, Johnson CK, Edmond MB, Polk RE. Hospital and community fluoroquinolone use and resistance in Staphylococcus aureus and Escherichia coli in 17 US hospitals. Clin Infect Dis. 2005;41:435–40 (III).PubMedCrossRef
141.
Zurück zum Zitat Charbonneau P, Parienti JJ, Thibon P, et al. Fluoroquinolone use and methicillin-resistant Staphylococcus aureus isolation rates in hospitalized patients: a quasi experimental study. Clin Infect Dis. 2006;42:778–84 (II).PubMedCrossRef Charbonneau P, Parienti JJ, Thibon P, et al. Fluoroquinolone use and methicillin-resistant Staphylococcus aureus isolation rates in hospitalized patients: a quasi experimental study. Clin Infect Dis. 2006;42:778–84 (II).PubMedCrossRef
142.
Zurück zum Zitat Parienti JJ, Cattoir V, Thibon P, et al. Hospital-wide modification of fluoroquinolone policy and methicillin-resistant Staphylococcus aureus rates: a 10-year interrupted time-series analysis. J Hosp Infect. 2011;78:118–22 (II).PubMedCrossRef Parienti JJ, Cattoir V, Thibon P, et al. Hospital-wide modification of fluoroquinolone policy and methicillin-resistant Staphylococcus aureus rates: a 10-year interrupted time-series analysis. J Hosp Infect. 2011;78:118–22 (II).PubMedCrossRef
143.
Zurück zum Zitat Lafaurie M, Porcher R, Donay JL, Touratier S, Molina JM. Reduction of fluoroquinolone use is associated with a decrease in methicillin-resistant Staphylococcus aureus and fluoroquinolone-resistant Pseudomonas aeruginosa isolation rates: a 10 year study. J Antimicrob Chemother. 2012;67:1010–5 (II).PubMedCrossRef Lafaurie M, Porcher R, Donay JL, Touratier S, Molina JM. Reduction of fluoroquinolone use is associated with a decrease in methicillin-resistant Staphylococcus aureus and fluoroquinolone-resistant Pseudomonas aeruginosa isolation rates: a 10 year study. J Antimicrob Chemother. 2012;67:1010–5 (II).PubMedCrossRef
144.
Zurück zum Zitat Talpaert MJ, Gopal RG, Cooper BS, Wade P. Impact of guidelines and enhanced antibiotic stewardship on reducing broad-spectrum antibiotic usage and its effect on incidence of Clostridium difficile infection. J Antimicrob Chemother. 2011;66:2168–74 (III).PubMedCrossRef Talpaert MJ, Gopal RG, Cooper BS, Wade P. Impact of guidelines and enhanced antibiotic stewardship on reducing broad-spectrum antibiotic usage and its effect on incidence of Clostridium difficile infection. J Antimicrob Chemother. 2011;66:2168–74 (III).PubMedCrossRef
145.
Zurück zum Zitat Kallen AJ, Thompson A, Ristaino P, et al. Complete restriction of fluoroquinolone use to control an outbreak of Clostridium difficile infection at a community hospital. Infect Control Hosp Epidemiol. 2009;30:264–72 (III).PubMedCrossRef Kallen AJ, Thompson A, Ristaino P, et al. Complete restriction of fluoroquinolone use to control an outbreak of Clostridium difficile infection at a community hospital. Infect Control Hosp Epidemiol. 2009;30:264–72 (III).PubMedCrossRef
146.
Zurück zum Zitat Price J, Cheek E, Lippett S, et al. Impact of an intervention to control Clostridium difficile infection on hospital- and community-onset disease; an interrupted time series analysis. Clin Microbiol Infect. 2010;16:1297–302 (III).PubMedCrossRef Price J, Cheek E, Lippett S, et al. Impact of an intervention to control Clostridium difficile infection on hospital- and community-onset disease; an interrupted time series analysis. Clin Microbiol Infect. 2010;16:1297–302 (III).PubMedCrossRef
147.
Zurück zum Zitat Willemsen I, Cooper B, van BC, Winters M, Andriesse G, Kluytmans J. Improving quinolone use in hospitals by using a bundle of interventions in an interrupted time series analysis. Antimicrob Agents Chemother. 2010;54:3763–9 (II).PubMedPubMedCentralCrossRef Willemsen I, Cooper B, van BC, Winters M, Andriesse G, Kluytmans J. Improving quinolone use in hospitals by using a bundle of interventions in an interrupted time series analysis. Antimicrob Agents Chemother. 2010;54:3763–9 (II).PubMedPubMedCentralCrossRef
148.
Zurück zum Zitat Aldeyab MA, Devine MJ, Flanagan P, et al. Multihospital outbreak of Clostridium difficile ribotype 027 infection: epidemiology and analysis of control measures. Infect Control Hosp Epidemiol. 2011;32:210–9 (III).PubMedCrossRef Aldeyab MA, Devine MJ, Flanagan P, et al. Multihospital outbreak of Clostridium difficile ribotype 027 infection: epidemiology and analysis of control measures. Infect Control Hosp Epidemiol. 2011;32:210–9 (III).PubMedCrossRef
149.
Zurück zum Zitat Ranji SR, Steinman MA, Shojania KG, Gonzales R. Interventions to reduce unnecessary antibiotic prescribing: a systematic review and quantitative analysis. Med Care. 2008;46:847–62 (I).PubMedCrossRef Ranji SR, Steinman MA, Shojania KG, Gonzales R. Interventions to reduce unnecessary antibiotic prescribing: a systematic review and quantitative analysis. Med Care. 2008;46:847–62 (I).PubMedCrossRef
150.
Zurück zum Zitat Serisier DJ, Bowler SD. Effect of a simple educational intervention on the hospital management of community-acquired pneumonia. Respirology. 2007;12:389–93 (III).PubMedCrossRef Serisier DJ, Bowler SD. Effect of a simple educational intervention on the hospital management of community-acquired pneumonia. Respirology. 2007;12:389–93 (III).PubMedCrossRef
151.
Zurück zum Zitat Akter SF, Heller RD, Smith AJ, Milly AF. Impact of a training intervention on use of antimicrobials in teaching hospitals. J Infect Dev Ctries. 2009;3:447–51 (II).PubMedCrossRef Akter SF, Heller RD, Smith AJ, Milly AF. Impact of a training intervention on use of antimicrobials in teaching hospitals. J Infect Dev Ctries. 2009;3:447–51 (II).PubMedCrossRef
152.
Zurück zum Zitat Kao LS, Lew DF, Doyle PD, et al. A tale of 2 hospitals: a staggered cohort study of targeted interventions to improve compliance with antibiotic prophylaxis guidelines. Surgery. 2010;148:255–62 (II).PubMedCrossRef Kao LS, Lew DF, Doyle PD, et al. A tale of 2 hospitals: a staggered cohort study of targeted interventions to improve compliance with antibiotic prophylaxis guidelines. Surgery. 2010;148:255–62 (II).PubMedCrossRef
153.
Zurück zum Zitat Zabarsky TF, Sethi AK, Donskey CJ. Sustained reduction in inappropriate treatment of asymptomatic bacteriuria in a long-term care facility through an educational intervention. Am J Infect Control. 2008;36:476–80 (III).PubMedCrossRef Zabarsky TF, Sethi AK, Donskey CJ. Sustained reduction in inappropriate treatment of asymptomatic bacteriuria in a long-term care facility through an educational intervention. Am J Infect Control. 2008;36:476–80 (III).PubMedCrossRef
154.
Zurück zum Zitat Pavese P, Saurel N, Labarere J, et al. Does an educational session with an infectious diseases physician reduce the use of inappropriate antibiotic therapy for inpatients with positive urine culture results? A controlled before-and-after study. Infect Control Hosp Epidemiol. 2009;30:596–9 (II).PubMedCrossRef Pavese P, Saurel N, Labarere J, et al. Does an educational session with an infectious diseases physician reduce the use of inappropriate antibiotic therapy for inpatients with positive urine culture results? A controlled before-and-after study. Infect Control Hosp Epidemiol. 2009;30:596–9 (II).PubMedCrossRef
155.
Zurück zum Zitat Monette J, Miller MA, Monette M, et al. Effect of an educational intervention on optimizing antibiotic prescribing in long-term care facilities. J Am Geriatr Soc. 2007;55:1231–5 (I).PubMedCrossRef Monette J, Miller MA, Monette M, et al. Effect of an educational intervention on optimizing antibiotic prescribing in long-term care facilities. J Am Geriatr Soc. 2007;55:1231–5 (I).PubMedCrossRef
156.
Zurück zum Zitat Metlay JP, Camargo CA Jr, MacKenzie T, et al. Cluster-randomized trial to improve antibiotic use for adults with acute respiratory infections treated in emergency departments. Ann Emerg Med. 2007;50:221–30 (I).PubMedCrossRef Metlay JP, Camargo CA Jr, MacKenzie T, et al. Cluster-randomized trial to improve antibiotic use for adults with acute respiratory infections treated in emergency departments. Ann Emerg Med. 2007;50:221–30 (I).PubMedCrossRef
157.
Zurück zum Zitat Landgren FT, Harvey KJ, Mashford ML, Moulds RF, Guthrie B, Hemming M. Changing antibiotic prescribing by educational marketing. Med J Aust. 1988;149:595–9 (II).PubMed Landgren FT, Harvey KJ, Mashford ML, Moulds RF, Guthrie B, Hemming M. Changing antibiotic prescribing by educational marketing. Med J Aust. 1988;149:595–9 (II).PubMed
158.
Zurück zum Zitat Ozgun H, Ertugrul BM, Soyder A, Ozturk B, Aydemir M. Peri-operative antibiotic prophylaxis: adherence to guidelines and effects of educational intervention. Int J Surg. 2010;8:159–63 (II).PubMedCrossRef Ozgun H, Ertugrul BM, Soyder A, Ozturk B, Aydemir M. Peri-operative antibiotic prophylaxis: adherence to guidelines and effects of educational intervention. Int J Surg. 2010;8:159–63 (II).PubMedCrossRef
159.
Zurück zum Zitat Kanter G, Connelly NR, Fitzgerald J. A system and process redesign to improve perioperative antibiotic administration. Anesth Analg. 2006;103:1517–21 (II).PubMedCrossRef Kanter G, Connelly NR, Fitzgerald J. A system and process redesign to improve perioperative antibiotic administration. Anesth Analg. 2006;103:1517–21 (II).PubMedCrossRef
160.
Zurück zum Zitat Pettersson E, Vernby A, Molstad S, Lundborg CS. Can a multifaceted educational intervention targeting both nurses and physicians change the prescribing of antibiotics to nursing home residents? A cluster randomized controlled trial. J Antimicrob Chemother. 2011;66:2659–66 (I).PubMedCrossRef Pettersson E, Vernby A, Molstad S, Lundborg CS. Can a multifaceted educational intervention targeting both nurses and physicians change the prescribing of antibiotics to nursing home residents? A cluster randomized controlled trial. J Antimicrob Chemother. 2011;66:2659–66 (I).PubMedCrossRef
161.
Zurück zum Zitat Zahar JR, Rioux C, Girou E, et al. Inappropriate prescribing of aminoglycosides: risk factors and impact of an antibiotic control team. J Antimicrob Chemother. 2006;58:651–6 (II).PubMedCrossRef Zahar JR, Rioux C, Girou E, et al. Inappropriate prescribing of aminoglycosides: risk factors and impact of an antibiotic control team. J Antimicrob Chemother. 2006;58:651–6 (II).PubMedCrossRef
162.
Zurück zum Zitat Kisuule F, Wright S, Barreto J, Zenilman J. Improving antibiotic utilization among hospitalists: a pilot academic detailing project with a public health approach. J Hosp Med. 2008;3:64–70 (II).PubMedCrossRef Kisuule F, Wright S, Barreto J, Zenilman J. Improving antibiotic utilization among hospitalists: a pilot academic detailing project with a public health approach. J Hosp Med. 2008;3:64–70 (II).PubMedCrossRef
163.
Zurück zum Zitat Kiyatkin DE, Wright S, Zenilman J, Kisuule F. Improving utilization of antimicrobial drugs among physician assistants in the ED. JAAPA 2011;24:44, 47-1 (II). Kiyatkin DE, Wright S, Zenilman J, Kisuule F. Improving utilization of antimicrobial drugs among physician assistants in the ED. JAAPA 2011;24:44, 47-1 (II).
164.
Zurück zum Zitat Linnebur SA, Fish DN, Ruscin JM, et al. Impact of a multidisciplinary intervention on antibiotic use for nursing home-acquired pneumonia. Am J Geriatr Pharmacother. 2011;9:442–50 (II).PubMedCrossRef Linnebur SA, Fish DN, Ruscin JM, et al. Impact of a multidisciplinary intervention on antibiotic use for nursing home-acquired pneumonia. Am J Geriatr Pharmacother. 2011;9:442–50 (II).PubMedCrossRef
165.
Zurück zum Zitat Cosgrove SE, Patel A, Song X, et al. Impact of different methods of feedback to clinicians after postprescription antimicrobial review based on the Centers for Disease Control and Prevention’s 12 steps to prevent antimicrobial resistance among hospitalized adults. Infect Control Hosp Epidemiol. 2007;28:641–6 (II).PubMedCrossRef Cosgrove SE, Patel A, Song X, et al. Impact of different methods of feedback to clinicians after postprescription antimicrobial review based on the Centers for Disease Control and Prevention’s 12 steps to prevent antimicrobial resistance among hospitalized adults. Infect Control Hosp Epidemiol. 2007;28:641–6 (II).PubMedCrossRef
166.
Zurück zum Zitat Ivers N, Jamtvedt G, Flottorp S, et al. Audit and feedback: effects on professional practice and healthcare outcomes. Cochrane Database Syst Rev. 2012;6:CD000259 (I).PubMed Ivers N, Jamtvedt G, Flottorp S, et al. Audit and feedback: effects on professional practice and healthcare outcomes. Cochrane Database Syst Rev. 2012;6:CD000259 (I).PubMed
167.
Zurück zum Zitat LaRocco A Jr. Concurrent antibiotic review programs—a role for infectious diseases specialists at small community hospitals. Clin Infect Dis. 2003;37:742–3 (II).PubMedCrossRef LaRocco A Jr. Concurrent antibiotic review programs—a role for infectious diseases specialists at small community hospitals. Clin Infect Dis. 2003;37:742–3 (II).PubMedCrossRef
168.
Zurück zum Zitat Masia M, Matoses C, Padilla S, et al. Limited efficacy of a nonrestricted intervention on antimicrobial prescription of commonly used antibiotics in the hospital setting: results of a randomized controlled trial. Eur J Clin Microbiol Infect Dis. 2008;27:597–605 (I).PubMedCrossRef Masia M, Matoses C, Padilla S, et al. Limited efficacy of a nonrestricted intervention on antimicrobial prescription of commonly used antibiotics in the hospital setting: results of a randomized controlled trial. Eur J Clin Microbiol Infect Dis. 2008;27:597–605 (I).PubMedCrossRef
169.
Zurück zum Zitat Rattanaumpawan P, Sutha P, Thamlikitkul V. Effectiveness of drug use evaluation and antibiotic authorization on patients’ clinical outcomes, antibiotic consumption, and antibiotic expenditures. Am J Infect Control. 2010;38:38–43 (II).PubMedCrossRef Rattanaumpawan P, Sutha P, Thamlikitkul V. Effectiveness of drug use evaluation and antibiotic authorization on patients’ clinical outcomes, antibiotic consumption, and antibiotic expenditures. Am J Infect Control. 2010;38:38–43 (II).PubMedCrossRef
170.
Zurück zum Zitat Elligsen M, Walker SA, Pinto R, et al. Audit and feedback to reduce broad-spectrum antibiotic use among intensive care unit patients: a controlled interrupted time series analysis. Infect Control Hosp Epidemiol. 2012;33:354–61 (II).PubMedCrossRef Elligsen M, Walker SA, Pinto R, et al. Audit and feedback to reduce broad-spectrum antibiotic use among intensive care unit patients: a controlled interrupted time series analysis. Infect Control Hosp Epidemiol. 2012;33:354–61 (II).PubMedCrossRef
171.
Zurück zum Zitat Zvonar RK, Bush P, Roth V. Practice changes to improve delivery of surgical antibiotic prophylaxis. Healthc Q. 2008;11:141–4 (II).PubMedCrossRef Zvonar RK, Bush P, Roth V. Practice changes to improve delivery of surgical antibiotic prophylaxis. Healthc Q. 2008;11:141–4 (II).PubMedCrossRef
172.
Zurück zum Zitat Chu LA, Bratzler DW, Lewis RJ, et al. Improving the quality of care for patients with pneumonia in very small hospitals. Arch Intern Med. 2003;163:326–32 (II).PubMedCrossRef Chu LA, Bratzler DW, Lewis RJ, et al. Improving the quality of care for patients with pneumonia in very small hospitals. Arch Intern Med. 2003;163:326–32 (II).PubMedCrossRef
173.
Zurück zum Zitat Cooke J, Alexander K, Charani E, et al. Antimicrobial stewardship: an evidence-based, antimicrobial self-assessment toolkit (ASAT) for acute hospitals. J Antimicrob Chemother. 2010;65:2669–73 (IV).PubMedCrossRef Cooke J, Alexander K, Charani E, et al. Antimicrobial stewardship: an evidence-based, antimicrobial self-assessment toolkit (ASAT) for acute hospitals. J Antimicrob Chemother. 2010;65:2669–73 (IV).PubMedCrossRef
174.
Zurück zum Zitat Amadeo B, Dumartin C, Parneix P, Fourrier-Reglat A, Rogues AM. Relationship between antibiotic consumption and antibiotic policy: an adjusted analysis in the French healthcare system. J Antimicrob Chemother. 2011;66:434–42 (III).PubMedCrossRef Amadeo B, Dumartin C, Parneix P, Fourrier-Reglat A, Rogues AM. Relationship between antibiotic consumption and antibiotic policy: an adjusted analysis in the French healthcare system. J Antimicrob Chemother. 2011;66:434–42 (III).PubMedCrossRef
175.
Zurück zum Zitat Nothacker M, Reiter A. Qualitätsindikatoren für Nationale VersorgungsLeitlinien. In: ÄZQ (Hrsg.) Programm für Nationale VersorgungsLeitlinien von BÄK, KBV und AWMF—Qualitätsindikatoren, Manual für Autoren. 18–31. 2009. Nothacker M, Reiter A. Qualitätsindikatoren für Nationale VersorgungsLeitlinien. In: ÄZQ (Hrsg.) Programm für Nationale VersorgungsLeitlinien von BÄK, KBV und AWMF—Qualitätsindikatoren, Manual für Autoren. 18–31. 2009.
176.
Zurück zum Zitat Afshar N, Tabas J, Afshar K, Silbergleit R. Blood cultures for community-acquired pneumonia: are they worthy of two quality measures? A systematic review. J Hosp Med. 2009;4:112–23.PubMedCrossRef Afshar N, Tabas J, Afshar K, Silbergleit R. Blood cultures for community-acquired pneumonia: are they worthy of two quality measures? A systematic review. J Hosp Med. 2009;4:112–23.PubMedCrossRef
177.
Zurück zum Zitat Dumartin C, Rogues AM, Amadeo B, et al. Antibiotic stewardship programmes: legal framework and structure and process indicator in Southwestern French hospitals, 2005–2008. J Hosp Infect. 2011;77:123–8 (III).PubMedCrossRef Dumartin C, Rogues AM, Amadeo B, et al. Antibiotic stewardship programmes: legal framework and structure and process indicator in Southwestern French hospitals, 2005–2008. J Hosp Infect. 2011;77:123–8 (III).PubMedCrossRef
178.
Zurück zum Zitat Hermanides HS, Hulscher ME, Schouten JA, Prins JM, Geerlings SE. Development of quality indicators for the antibiotic treatment of complicated urinary tract infections: a first step to measure and improve care. Clin Infect Dis. 2008;46:703–11 (IV).PubMedCrossRef Hermanides HS, Hulscher ME, Schouten JA, Prins JM, Geerlings SE. Development of quality indicators for the antibiotic treatment of complicated urinary tract infections: a first step to measure and improve care. Clin Infect Dis. 2008;46:703–11 (IV).PubMedCrossRef
179.
Zurück zum Zitat Kanwar M, Brar N, Khatib R, Fakih MG. Misdiagnosis of community-acquired pneumonia and inappropriate utilization of antibiotics: side effects of the 4-h antibiotic administration rule. Chest. 2007;131:1865–9 (III).PubMedCrossRef Kanwar M, Brar N, Khatib R, Fakih MG. Misdiagnosis of community-acquired pneumonia and inappropriate utilization of antibiotics: side effects of the 4-h antibiotic administration rule. Chest. 2007;131:1865–9 (III).PubMedCrossRef
180.
Zurück zum Zitat Morris AM, Brener S, Dresser L, et al. Use of a structured panel process to define quality metrics for antimicrobial stewardship programs. Infect Control Hosp Epidemiol. 2012;33:500–6 (IV).PubMedCrossRef Morris AM, Brener S, Dresser L, et al. Use of a structured panel process to define quality metrics for antimicrobial stewardship programs. Infect Control Hosp Epidemiol. 2012;33:500–6 (IV).PubMedCrossRef
181.
Zurück zum Zitat Nathwani D, Sneddon J, Patton A, Malcolm W. Antimicrobial stewardship in Scotland: impact of a national programme. Antimicrob Resist Infect Control. 2012;1:7 (III).PubMedPubMedCentralCrossRef Nathwani D, Sneddon J, Patton A, Malcolm W. Antimicrobial stewardship in Scotland: impact of a national programme. Antimicrob Resist Infect Control. 2012;1:7 (III).PubMedPubMedCentralCrossRef
182.
Zurück zum Zitat Nguyen HB, Corbett SW, Steele R, et al. Implementation of a bundle of quality indicators for the early management of severe sepsis and septic shock is associated with decreased mortality. Crit Care Med. 2007;35:1105–12 (II).PubMedCrossRef Nguyen HB, Corbett SW, Steele R, et al. Implementation of a bundle of quality indicators for the early management of severe sepsis and septic shock is associated with decreased mortality. Crit Care Med. 2007;35:1105–12 (II).PubMedCrossRef
183.
Zurück zum Zitat Pines JM, Isserman JA, Hinfey PB. The measurement of time to first antibiotic dose for pneumonia in the emergency department: a white paper and position statement prepared for the American Academy of Emergency Medicine. J Emerg Med. 2009;37:335–40 (IV).PubMedCrossRef Pines JM, Isserman JA, Hinfey PB. The measurement of time to first antibiotic dose for pneumonia in the emergency department: a white paper and position statement prepared for the American Academy of Emergency Medicine. J Emerg Med. 2009;37:335–40 (IV).PubMedCrossRef
184.
Zurück zum Zitat Pulcini C, Defres S, Aggarwal I, Nathwani D, Davey P. Design of a ‘day 3 bundle’ to improve the reassessment of inpatient empirical antibiotic prescriptions. J Antimicrob Chemother. 2008;61:1384–8 (III).PubMedCrossRef Pulcini C, Defres S, Aggarwal I, Nathwani D, Davey P. Design of a ‘day 3 bundle’ to improve the reassessment of inpatient empirical antibiotic prescriptions. J Antimicrob Chemother. 2008;61:1384–8 (III).PubMedCrossRef
185.
Zurück zum Zitat Quattromani E, Powell ES, Khare RK, et al. Hospital-reported data on the pneumonia quality measure “Time to First Antibiotic Dose” are not associated with inpatient mortality: results of a nationwide cross-sectional analysis. Acad Emerg Med. 2011;18:496–503 (III).PubMedPubMedCentralCrossRef Quattromani E, Powell ES, Khare RK, et al. Hospital-reported data on the pneumonia quality measure “Time to First Antibiotic Dose” are not associated with inpatient mortality: results of a nationwide cross-sectional analysis. Acad Emerg Med. 2011;18:496–503 (III).PubMedPubMedCentralCrossRef
186.
Zurück zum Zitat Saizy-Callaert S, Causse R, Furhman C, Le Paih MF, Thebault A, Chouaid C. Impact of a multidisciplinary approach to the control of antibiotic prescription in a general hospital. J Hosp Infect. 2003;53:177–82 (III).PubMedCrossRef Saizy-Callaert S, Causse R, Furhman C, Le Paih MF, Thebault A, Chouaid C. Impact of a multidisciplinary approach to the control of antibiotic prescription in a general hospital. J Hosp Infect. 2003;53:177–82 (III).PubMedCrossRef
187.
Zurück zum Zitat Shorr AF, Owens RC Jr. Guidelines and quality for community-acquired pneumonia: measures from the Joint Commission and the Centers for Medicare and Medicaid Services. Am J Health Syst Pharm. 2009;66:S2–7 (IV).PubMedCrossRef Shorr AF, Owens RC Jr. Guidelines and quality for community-acquired pneumonia: measures from the Joint Commission and the Centers for Medicare and Medicaid Services. Am J Health Syst Pharm. 2009;66:S2–7 (IV).PubMedCrossRef
188.
Zurück zum Zitat Van Kasteren ME, Mannien J, Kullberg BJ, et al. Quality improvement of surgical prophylaxis in Dutch hospitals: evaluation of a multi-site intervention by time series analysis. J Antimicrob Chemother. 2005;56:1094–102 (II).PubMedCrossRef Van Kasteren ME, Mannien J, Kullberg BJ, et al. Quality improvement of surgical prophylaxis in Dutch hospitals: evaluation of a multi-site intervention by time series analysis. J Antimicrob Chemother. 2005;56:1094–102 (II).PubMedCrossRef
189.
Zurück zum Zitat von Gunten V, Troillet N, Beney J, et al. Impact of an interdisciplinary strategy on antibiotic use: a prospective controlled study in three hospitals. J Antimicrob Chemother. 2005;55:362–6 (II).CrossRef von Gunten V, Troillet N, Beney J, et al. Impact of an interdisciplinary strategy on antibiotic use: a prospective controlled study in three hospitals. J Antimicrob Chemother. 2005;55:362–6 (II).CrossRef
190.
Zurück zum Zitat Buyle FM, Metz-Gercek S, Mechtler R, et al. Prospective multicentre feasibility study of a quality of care indicator for intravenous to oral switch therapy with highly bioavailable antibiotics. J Antimicrob Chemother. 2012;67:2043–6 (II).PubMedCrossRef Buyle FM, Metz-Gercek S, Mechtler R, et al. Prospective multicentre feasibility study of a quality of care indicator for intravenous to oral switch therapy with highly bioavailable antibiotics. J Antimicrob Chemother. 2012;67:2043–6 (II).PubMedCrossRef
191.
Zurück zum Zitat Kern WV, Metz-Gercek S, Mechtler R, et al. Staphylococcus aureus blood-stream infection management indicators as quality indicators for hospital antibiotic stewardship: feasibility study by the ABS International Quality Indicators (ABS QI) team. Clin Microbiol Infect 2009;15 Suppl 4:S188 (IV). Kern WV, Metz-Gercek S, Mechtler R, et al. Staphylococcus aureus blood-stream infection management indicators as quality indicators for hospital antibiotic stewardship: feasibility study by the ABS International Quality Indicators (ABS QI) team. Clin Microbiol Infect 2009;15 Suppl 4:S188 (IV).
192.
Zurück zum Zitat Drew RH, White R, MacDougall C, Hermsen ED, Owens RC Jr. Insights from the Society of Infectious Diseases Pharmacists on antimicrobial stewardship guidelines from the Infectious Diseases Society of America and the Society for Healthcare Epidemiology of America. Pharmacotherapy. 2009;29:593–607 (IV).PubMedCrossRef Drew RH, White R, MacDougall C, Hermsen ED, Owens RC Jr. Insights from the Society of Infectious Diseases Pharmacists on antimicrobial stewardship guidelines from the Infectious Diseases Society of America and the Society for Healthcare Epidemiology of America. Pharmacotherapy. 2009;29:593–607 (IV).PubMedCrossRef
193.
Zurück zum Zitat Huttner B, Jones M, Rubin MA, et al. Double trouble: how big a problem is redundant anaerobic antibiotic coverage in Veterans Affairs medical centres? J Antimicrob Chemother. 2012;67:1537–9 (III).PubMedCrossRef Huttner B, Jones M, Rubin MA, et al. Double trouble: how big a problem is redundant anaerobic antibiotic coverage in Veterans Affairs medical centres? J Antimicrob Chemother. 2012;67:1537–9 (III).PubMedCrossRef
194.
Zurück zum Zitat Aarts MA, Hancock JN, Heyland D, McLeod RS, Marshall JC. Empiric antibiotic therapy for suspected ventilator-associated pneumonia: a systematic review and meta-analysis of randomized trials. Crit Care Med. 2008;36:108–17 (I).PubMedCrossRef Aarts MA, Hancock JN, Heyland D, McLeod RS, Marshall JC. Empiric antibiotic therapy for suspected ventilator-associated pneumonia: a systematic review and meta-analysis of randomized trials. Crit Care Med. 2008;36:108–17 (I).PubMedCrossRef
195.
Zurück zum Zitat De Waele JJ, Ravyts M, Depuydt P, Blot SI, Decruyenaere J, Vogelaers D. De-escalation after empirical meropenem treatment in the intensive care unit: fiction or reality? J Crit Care. 2010;25:641–6.PubMedCrossRef De Waele JJ, Ravyts M, Depuydt P, Blot SI, Decruyenaere J, Vogelaers D. De-escalation after empirical meropenem treatment in the intensive care unit: fiction or reality? J Crit Care. 2010;25:641–6.PubMedCrossRef
196.
Zurück zum Zitat Johnson SJ, Ernst EJ, Moores KG. Is double coverage of gram-negative organisms necessary? Am J Health Syst Pharm. 2011;68:119–24 (IV).PubMedCrossRef Johnson SJ, Ernst EJ, Moores KG. Is double coverage of gram-negative organisms necessary? Am J Health Syst Pharm. 2011;68:119–24 (IV).PubMedCrossRef
197.
Zurück zum Zitat Niederman MS, Soulountsi V. De-escalation therapy: is it valuable for the management of ventilator-associated pneumonia? Clin Chest Med. 2011;32:517–34 (I).PubMedCrossRef Niederman MS, Soulountsi V. De-escalation therapy: is it valuable for the management of ventilator-associated pneumonia? Clin Chest Med. 2011;32:517–34 (I).PubMedCrossRef
198.
Zurück zum Zitat Bliziotis IA, Samonis G, Vardakas KZ, Chrysanthopoulou S, Falagas ME. Effect of aminoglycoside and beta-lactam combination therapy versus beta-lactam monotherapy on the emergence of antimicrobial resistance: a meta-analysis of randomized, controlled trials. Clin Infect Dis. 2005;41:149–58 (I).PubMedCrossRef Bliziotis IA, Samonis G, Vardakas KZ, Chrysanthopoulou S, Falagas ME. Effect of aminoglycoside and beta-lactam combination therapy versus beta-lactam monotherapy on the emergence of antimicrobial resistance: a meta-analysis of randomized, controlled trials. Clin Infect Dis. 2005;41:149–58 (I).PubMedCrossRef
199.
Zurück zum Zitat Paul M, Benuri-Silbiger I, Soares-Weiser K, Leibovici L. Beta lactam monotherapy versus beta lactam-aminoglycoside combination therapy for sepsis in immunocompetent patients: systematic review and meta-analysis of randomised trials. BMJ. 2004;328:668 (I).PubMedPubMedCentralCrossRef Paul M, Benuri-Silbiger I, Soares-Weiser K, Leibovici L. Beta lactam monotherapy versus beta lactam-aminoglycoside combination therapy for sepsis in immunocompetent patients: systematic review and meta-analysis of randomised trials. BMJ. 2004;328:668 (I).PubMedPubMedCentralCrossRef
200.
Zurück zum Zitat Marcus R, Paul M, Elphick H, Leibovici L. Clinical implications of beta-lactam-aminoglycoside synergism: systematic review of randomised trials. Int J Antimicrob Agents. 2011;37:491–503 (I).PubMedCrossRef Marcus R, Paul M, Elphick H, Leibovici L. Clinical implications of beta-lactam-aminoglycoside synergism: systematic review of randomised trials. Int J Antimicrob Agents. 2011;37:491–503 (I).PubMedCrossRef
201.
Zurück zum Zitat Abad CL, Kumar A, Safdar N. Antimicrobial therapy of sepsis and septic shock—when are two drugs better than one? Crit Care Clin. 2011;27:e1–27 (I).PubMedCrossRef Abad CL, Kumar A, Safdar N. Antimicrobial therapy of sepsis and septic shock—when are two drugs better than one? Crit Care Clin. 2011;27:e1–27 (I).PubMedCrossRef
202.
Zurück zum Zitat Boyd N, Nailor MD. Combination antibiotic therapy for empiric and definitive treatment of gram-negative infections: insights from the Society of Infectious Diseases Pharmacists. Pharmacotherapy. 2011;31:1073–84 (IV).PubMedCrossRef Boyd N, Nailor MD. Combination antibiotic therapy for empiric and definitive treatment of gram-negative infections: insights from the Society of Infectious Diseases Pharmacists. Pharmacotherapy. 2011;31:1073–84 (IV).PubMedCrossRef
203.
Zurück zum Zitat Alvarez-Lerma F, Alvarez B, Luque P, et al. Empiric broad-spectrum antibiotic therapy of nosocomial pneumonia in the intensive care unit: a prospective observational study. Crit Care. 2006;10:R78 (II).PubMedPubMedCentralCrossRef Alvarez-Lerma F, Alvarez B, Luque P, et al. Empiric broad-spectrum antibiotic therapy of nosocomial pneumonia in the intensive care unit: a prospective observational study. Crit Care. 2006;10:R78 (II).PubMedPubMedCentralCrossRef
204.
Zurück zum Zitat Mettler J, Simcock M, Sendi P, et al. Empirical use of antibiotics and adjustment of empirical antibiotic therapies in a university hospital: a prospective observational study. BMC Infect Dis. 2007;7:21 (III).PubMedPubMedCentralCrossRef Mettler J, Simcock M, Sendi P, et al. Empirical use of antibiotics and adjustment of empirical antibiotic therapies in a university hospital: a prospective observational study. BMC Infect Dis. 2007;7:21 (III).PubMedPubMedCentralCrossRef
205.
Zurück zum Zitat Montravers P, Dupont H, Gauzit R, Veber B, Bedos JP, Lepape A. Strategies of initiation and streamlining of antibiotic therapy in 41 French intensive care units. Crit Care. 2011;15:R17 (II).PubMedPubMedCentralCrossRef Montravers P, Dupont H, Gauzit R, Veber B, Bedos JP, Lepape A. Strategies of initiation and streamlining of antibiotic therapy in 41 French intensive care units. Crit Care. 2011;15:R17 (II).PubMedPubMedCentralCrossRef
206.
Zurück zum Zitat Briceland LL, Nightingale CH, Quintiliani R, Cooper BW, Smith KS. Antibiotic streamlining from combination therapy to monotherapy utilizing an interdisciplinary approach. Arch Intern Med. 1988;148:2019–22 (III).PubMedCrossRef Briceland LL, Nightingale CH, Quintiliani R, Cooper BW, Smith KS. Antibiotic streamlining from combination therapy to monotherapy utilizing an interdisciplinary approach. Arch Intern Med. 1988;148:2019–22 (III).PubMedCrossRef
207.
Zurück zum Zitat Schlueter M, James C, Dominguez A, Tsu L, Seymann G. Practice patterns for antibiotic de-escalation in culture-negative healthcare-associated pneumonia. Infection. 2010;38:357–62 (IV).PubMedPubMedCentralCrossRef Schlueter M, James C, Dominguez A, Tsu L, Seymann G. Practice patterns for antibiotic de-escalation in culture-negative healthcare-associated pneumonia. Infection. 2010;38:357–62 (IV).PubMedPubMedCentralCrossRef
208.
Zurück zum Zitat Shime N, Satake S, Fujita N. De-escalation of antimicrobials in the treatment of bacteraemia due to antibiotic-sensitive pathogens in immunocompetent patients. Infection. 2011;39:319–25 (IV).PubMedCrossRef Shime N, Satake S, Fujita N. De-escalation of antimicrobials in the treatment of bacteraemia due to antibiotic-sensitive pathogens in immunocompetent patients. Infection. 2011;39:319–25 (IV).PubMedCrossRef
209.
Zurück zum Zitat Glowacki RC, Schwartz DN, Itokazu GS, Wisniewski MF, Kieszkowski P, Weinstein RA. Antibiotic combinations with redundant antimicrobial spectra: clinical epidemiology and pilot intervention of computer-assisted surveillance. Clin Infect Dis. 2003;37:59–64 (II).PubMedCrossRef Glowacki RC, Schwartz DN, Itokazu GS, Wisniewski MF, Kieszkowski P, Weinstein RA. Antibiotic combinations with redundant antimicrobial spectra: clinical epidemiology and pilot intervention of computer-assisted surveillance. Clin Infect Dis. 2003;37:59–64 (II).PubMedCrossRef
210.
Zurück zum Zitat Micek ST, Ward S, Fraser VJ, Kollef MH. A randomized controlled trial of an antibiotic discontinuation policy for clinically suspected ventilator-associated pneumonia. Chest. 2004;125:1791–9 (I).PubMedCrossRef Micek ST, Ward S, Fraser VJ, Kollef MH. A randomized controlled trial of an antibiotic discontinuation policy for clinically suspected ventilator-associated pneumonia. Chest. 2004;125:1791–9 (I).PubMedCrossRef
211.
Zurück zum Zitat Kollef MH, Kollef KE. Antibiotic utilization and outcomes for patients with clinically suspected ventilator-associated pneumonia and negative quantitative BAL culture results. Chest. 2005;128:2706–13 (II).PubMedCrossRef Kollef MH, Kollef KE. Antibiotic utilization and outcomes for patients with clinically suspected ventilator-associated pneumonia and negative quantitative BAL culture results. Chest. 2005;128:2706–13 (II).PubMedCrossRef
212.
Zurück zum Zitat Giantsou E, Liratzopoulos N, Efraimidou E, et al. De-escalation therapy rates are significantly higher by bronchoalveolar lavage than by tracheal aspirate. Intensive Care Med. 2007;33:1533–40 (II).PubMedCrossRef Giantsou E, Liratzopoulos N, Efraimidou E, et al. De-escalation therapy rates are significantly higher by bronchoalveolar lavage than by tracheal aspirate. Intensive Care Med. 2007;33:1533–40 (II).PubMedCrossRef
213.
Zurück zum Zitat Heyland DK, Dodek P, Muscedere J, Day A, Cook D. Randomized trial of combination versus monotherapy for the empiric treatment of suspected ventilator-associated pneumonia. Crit Care Med. 2008;36:737–44 (II).PubMedCrossRef Heyland DK, Dodek P, Muscedere J, Day A, Cook D. Randomized trial of combination versus monotherapy for the empiric treatment of suspected ventilator-associated pneumonia. Crit Care Med. 2008;36:737–44 (II).PubMedCrossRef
214.
Zurück zum Zitat Eachempati SR, Hydo LJ, Shou J, Barie PS. Does de-escalation of antibiotic therapy for ventilator-associated pneumonia affect the likelihood of recurrent pneumonia or mortality in critically ill surgical patients? J Trauma. 2009;66:1343–8 (III).PubMedCrossRef Eachempati SR, Hydo LJ, Shou J, Barie PS. Does de-escalation of antibiotic therapy for ventilator-associated pneumonia affect the likelihood of recurrent pneumonia or mortality in critically ill surgical patients? J Trauma. 2009;66:1343–8 (III).PubMedCrossRef
215.
Zurück zum Zitat Amadeo B, Zarb P, Muller A, et al. European surveillance of antibiotic consumption (ESAC) point prevalence survey 2008: paediatric antimicrobial prescribing in 32 hospitals of 21 European countries. J Antimicrob Chemother. 2010;65:2247–52 (III).PubMedCrossRef Amadeo B, Zarb P, Muller A, et al. European surveillance of antibiotic consumption (ESAC) point prevalence survey 2008: paediatric antimicrobial prescribing in 32 hospitals of 21 European countries. J Antimicrob Chemother. 2010;65:2247–52 (III).PubMedCrossRef
216.
Zurück zum Zitat Guillemot D, Carbon C, Balkau B, et al. Low dosage and long treatment duration of beta-lactam: risk factors for carriage of penicillin-resistant Streptococcus pneumoniae. JAMA. 1998;279:365–70 (II).PubMedCrossRef Guillemot D, Carbon C, Balkau B, et al. Low dosage and long treatment duration of beta-lactam: risk factors for carriage of penicillin-resistant Streptococcus pneumoniae. JAMA. 1998;279:365–70 (II).PubMedCrossRef
217.
Zurück zum Zitat Harbarth S, Harris AD, Carmeli Y, Samore MH. Parallel analysis of individual and aggregated data on antibiotic exposure and resistance in gram-negative bacilli. Clin Infect Dis. 2001;33:1462–8 (II).PubMedCrossRef Harbarth S, Harris AD, Carmeli Y, Samore MH. Parallel analysis of individual and aggregated data on antibiotic exposure and resistance in gram-negative bacilli. Clin Infect Dis. 2001;33:1462–8 (II).PubMedCrossRef
218.
Zurück zum Zitat Albrich WC, Monnet DL, Harbarth S. Antibiotic selection pressure and resistance in Streptococcus pneumoniae and Streptococcus pyogenes. Emerg Infect Dis. 2004;10:514–7 (III).PubMedPubMedCentralCrossRef Albrich WC, Monnet DL, Harbarth S. Antibiotic selection pressure and resistance in Streptococcus pneumoniae and Streptococcus pyogenes. Emerg Infect Dis. 2004;10:514–7 (III).PubMedPubMedCentralCrossRef
219.
Zurück zum Zitat Zillich AJ, Sutherland JM, Wilson SJ, et al. Antimicrobial use control measures to prevent and control antimicrobial resistance in US hospitals. Infect Control Hosp Epidemiol. 2006;27:1088–95 (III).PubMedCrossRef Zillich AJ, Sutherland JM, Wilson SJ, et al. Antimicrobial use control measures to prevent and control antimicrobial resistance in US hospitals. Infect Control Hosp Epidemiol. 2006;27:1088–95 (III).PubMedCrossRef
220.
Zurück zum Zitat Dunbar LM, Wunderink RG, Habib MP, et al. High-dose, short-course levofloxacin for community-acquired pneumonia: a new treatment paradigm. Clin Infect Dis. 2003;37:752–60 (I).PubMedCrossRef Dunbar LM, Wunderink RG, Habib MP, et al. High-dose, short-course levofloxacin for community-acquired pneumonia: a new treatment paradigm. Clin Infect Dis. 2003;37:752–60 (I).PubMedCrossRef
221.
Zurück zum Zitat van NC, van’t Wout JW, Assendelft WJ, et al. Treatment duration of febrile urinary tract infection (FUTIRST trial): a randomized placebo-controlled multicenter trial comparing short (7 days) antibiotic treatment with conventional treatment (14 days). BMC Infect Dis. 2009;9:131 (I).CrossRef van NC, van’t Wout JW, Assendelft WJ, et al. Treatment duration of febrile urinary tract infection (FUTIRST trial): a randomized placebo-controlled multicenter trial comparing short (7 days) antibiotic treatment with conventional treatment (14 days). BMC Infect Dis. 2009;9:131 (I).CrossRef
222.
Zurück zum Zitat Pugh R, Grant C, Cooke RP, Dempsey G. Short-course versus prolonged-course antibiotic therapy for hospital-acquired pneumonia in critically ill adults. Cochrane Database Syst Rev. 2011;10:CD007577 (I).PubMed Pugh R, Grant C, Cooke RP, Dempsey G. Short-course versus prolonged-course antibiotic therapy for hospital-acquired pneumonia in critically ill adults. Cochrane Database Syst Rev. 2011;10:CD007577 (I).PubMed
223.
Zurück zum Zitat Havey TC, Fowler RA, Daneman N. Duration of antibiotic therapy for bacteremia: a systematic review and meta-analysis. Crit Care. 2011;15:R267 (I).PubMedPubMedCentralCrossRef Havey TC, Fowler RA, Daneman N. Duration of antibiotic therapy for bacteremia: a systematic review and meta-analysis. Crit Care. 2011;15:R267 (I).PubMedPubMedCentralCrossRef
224.
Zurück zum Zitat Eliakim-Raz N, Yahav D, Paul M, Leibovici L. Duration of antibiotic treatment for acute pyelonephritis and septic urinary tract infection—7 days or less versus longer treatment: systematic review and meta-analysis of randomized controlled trials. J Antimicrob Chemother. 2013;68:2183–91 (I). Eliakim-Raz N, Yahav D, Paul M, Leibovici L. Duration of antibiotic treatment for acute pyelonephritis and septic urinary tract infection—7 days or less versus longer treatment: systematic review and meta-analysis of randomized controlled trials. J Antimicrob Chemother. 2013;68:2183–91 (I).
225.
Zurück zum Zitat Hayashi Y, Paterson DL. Strategies for reduction in duration of antibiotic use in hospitalized patients. Clin Infect Dis. 2011;52:1232–40 (III).PubMedCrossRef Hayashi Y, Paterson DL. Strategies for reduction in duration of antibiotic use in hospitalized patients. Clin Infect Dis. 2011;52:1232–40 (III).PubMedCrossRef
226.
Zurück zum Zitat Liew YX, Chlebicki MP, Lee W, Hsu LY, Kwa AL. Use of procalcitonin (PCT) to guide discontinuation of antibiotic use in an unspecified sepsis is an antimicrobial stewardship program (ASP). Eur J Clin Microbiol Infect Dis. 2011;30:853–5 (III).PubMedCrossRef Liew YX, Chlebicki MP, Lee W, Hsu LY, Kwa AL. Use of procalcitonin (PCT) to guide discontinuation of antibiotic use in an unspecified sepsis is an antimicrobial stewardship program (ASP). Eur J Clin Microbiol Infect Dis. 2011;30:853–5 (III).PubMedCrossRef
227.
Zurück zum Zitat Schuetz P, Muller B, Christ-Crain M, et al. Procalcitonin to initiate or discontinue antibiotics in acute respiratory tract infections. Cochrane Database Syst Rev. 2012;9:CD007498 (I).PubMed Schuetz P, Muller B, Christ-Crain M, et al. Procalcitonin to initiate or discontinue antibiotics in acute respiratory tract infections. Cochrane Database Syst Rev. 2012;9:CD007498 (I).PubMed
228.
Zurück zum Zitat Agarwal R, Schwartz DN. Procalcitonin to guide duration of antimicrobial therapy in intensive care units: a systematic review. Clin Infect Dis. 2011;53:379–87 (I).PubMedCrossRef Agarwal R, Schwartz DN. Procalcitonin to guide duration of antimicrobial therapy in intensive care units: a systematic review. Clin Infect Dis. 2011;53:379–87 (I).PubMedCrossRef
229.
Zurück zum Zitat Bouadma L, Luyt CE, Tubach F, et al. Use of procalcitonin to reduce patients’ exposure to antibiotics in intensive care units (PRORATA trial): a multicentre randomised controlled trial. Lancet. 2010;375:463–74 (I).PubMedCrossRef Bouadma L, Luyt CE, Tubach F, et al. Use of procalcitonin to reduce patients’ exposure to antibiotics in intensive care units (PRORATA trial): a multicentre randomised controlled trial. Lancet. 2010;375:463–74 (I).PubMedCrossRef
230.
Zurück zum Zitat Schroeder S, Hochreiter M, Koehler T, et al. Procalcitonin (PCT)-guided algorithm reduces length of antibiotic treatment in surgical intensive care patients with severe sepsis: results of a prospective randomized study. Langenbecks Arch Surg. 2009;394:221–6 (I).PubMedCrossRef Schroeder S, Hochreiter M, Koehler T, et al. Procalcitonin (PCT)-guided algorithm reduces length of antibiotic treatment in surgical intensive care patients with severe sepsis: results of a prospective randomized study. Langenbecks Arch Surg. 2009;394:221–6 (I).PubMedCrossRef
231.
Zurück zum Zitat Kopterides P, Siempos II, Tsangaris I, Tsantes A, Armaganidis A. Procalcitonin-guided algorithms of antibiotic therapy in the intensive care unit: a systematic review and meta-analysis of randomized controlled trials. Crit Care Med. 2010;38:2229–41 (I).PubMedCrossRef Kopterides P, Siempos II, Tsangaris I, Tsantes A, Armaganidis A. Procalcitonin-guided algorithms of antibiotic therapy in the intensive care unit: a systematic review and meta-analysis of randomized controlled trials. Crit Care Med. 2010;38:2229–41 (I).PubMedCrossRef
232.
Zurück zum Zitat Heyland DK, Johnson AP, Reynolds SC, Muscedere J. Procalcitonin for reduced antibiotic exposure in the critical care setting: a systematic review and an economic evaluation. Crit Care Med. 2011;39:1792–9 (I).PubMedCrossRef Heyland DK, Johnson AP, Reynolds SC, Muscedere J. Procalcitonin for reduced antibiotic exposure in the critical care setting: a systematic review and an economic evaluation. Crit Care Med. 2011;39:1792–9 (I).PubMedCrossRef
233.
Zurück zum Zitat Nobre V, Harbarth S, Graf JD, Rohner P, Pugin J. Use of procalcitonin to shorten antibiotic treatment duration in septic patients: a randomized trial. Am J Respir Crit Care Med. 2008;177:498–505 (I).PubMedCrossRef Nobre V, Harbarth S, Graf JD, Rohner P, Pugin J. Use of procalcitonin to shorten antibiotic treatment duration in septic patients: a randomized trial. Am J Respir Crit Care Med. 2008;177:498–505 (I).PubMedCrossRef
234.
Zurück zum Zitat Tang H, Huang T, Jing J, Shen H, Cui W. Effect of procalcitonin-guided treatment in patients with infections: a systematic review and meta-analysis. Infection. 2009;37:497–507 (I).PubMedCrossRef Tang H, Huang T, Jing J, Shen H, Cui W. Effect of procalcitonin-guided treatment in patients with infections: a systematic review and meta-analysis. Infection. 2009;37:497–507 (I).PubMedCrossRef
235.
Zurück zum Zitat Hohn A, Schroeder S, Gehrt A, et al. Procalcitonin-guided algorithm to reduce length of antibiotic therapy in patients with severe sepsis and septic shock. BMC Infect Dis. 2013;13:158 (III).PubMedPubMedCentralCrossRef Hohn A, Schroeder S, Gehrt A, et al. Procalcitonin-guided algorithm to reduce length of antibiotic therapy in patients with severe sepsis and septic shock. BMC Infect Dis. 2013;13:158 (III).PubMedPubMedCentralCrossRef
236.
Zurück zum Zitat Gyssens IC, Geerligs IE, Dony JM, et al. Optimising antimicrobial drug use in surgery: an intervention study in a Dutch university hospital. J Antimicrob Chemother. 1996;38:1001–12 (II).PubMedCrossRef Gyssens IC, Geerligs IE, Dony JM, et al. Optimising antimicrobial drug use in surgery: an intervention study in a Dutch university hospital. J Antimicrob Chemother. 1996;38:1001–12 (II).PubMedCrossRef
237.
Zurück zum Zitat Burke JP. Maximizing appropriate antibiotic prophylaxis for surgical patients: an update from LDS Hospital, Salt Lake City. Clin Infect Dis. 2001;33:S78–83 (II).PubMedCrossRef Burke JP. Maximizing appropriate antibiotic prophylaxis for surgical patients: an update from LDS Hospital, Salt Lake City. Clin Infect Dis. 2001;33:S78–83 (II).PubMedCrossRef
238.
Zurück zum Zitat Hosoglu S, Aslan S, Akalin S, Bosnak V. Audit of quality of perioperative antimicrobial prophylaxis. Pharm World Sci. 2009;31:14–7 (II).PubMedCrossRef Hosoglu S, Aslan S, Akalin S, Bosnak V. Audit of quality of perioperative antimicrobial prophylaxis. Pharm World Sci. 2009;31:14–7 (II).PubMedCrossRef
239.
Zurück zum Zitat Burkitt KH, Mor MK, Jain R, et al. Toyota production system quality improvement initiative improves perioperative antibiotic therapy. Am J Manag Care. 2009;15:633–42 (III).PubMed Burkitt KH, Mor MK, Jain R, et al. Toyota production system quality improvement initiative improves perioperative antibiotic therapy. Am J Manag Care. 2009;15:633–42 (III).PubMed
240.
Zurück zum Zitat Takahashi Y, Takesue Y, Nakajima K, et al. Implementation of a hospital-wide project for appropriate antimicrobial prophylaxis. J Infect Chemother. 2010;16:418–23 (II).PubMedCrossRef Takahashi Y, Takesue Y, Nakajima K, et al. Implementation of a hospital-wide project for appropriate antimicrobial prophylaxis. J Infect Chemother. 2010;16:418–23 (II).PubMedCrossRef
241.
Zurück zum Zitat Meyer E, Schwab F, Pollitt A, Bettolo W, Schroeren-Boersch B, Trautmann M. Impact of a change in antibiotic prophylaxis on total antibiotic use in a surgical intensive care unit. Infection. 2010;38:19–24 (III).PubMedCrossRef Meyer E, Schwab F, Pollitt A, Bettolo W, Schroeren-Boersch B, Trautmann M. Impact of a change in antibiotic prophylaxis on total antibiotic use in a surgical intensive care unit. Infection. 2010;38:19–24 (III).PubMedCrossRef
242.
Zurück zum Zitat Sun TB, Chao SF, Chang BS, Chen TY, Gao PY, Shyr MH. Quality improvements of antimicrobial prophylaxis in coronary artery bypass grafting. J Surg Res. 2011;167:329–35 (III).PubMedCrossRef Sun TB, Chao SF, Chang BS, Chen TY, Gao PY, Shyr MH. Quality improvements of antimicrobial prophylaxis in coronary artery bypass grafting. J Surg Res. 2011;167:329–35 (III).PubMedCrossRef
243.
Zurück zum Zitat Haynes K, Linkin DR, Fishman NO, et al. Effectiveness of an information technology intervention to improve prophylactic antibacterial use in the postoperative period. J Am Med Inform Assoc. 2011;18:164–8 (II).PubMedPubMedCentralCrossRef Haynes K, Linkin DR, Fishman NO, et al. Effectiveness of an information technology intervention to improve prophylactic antibacterial use in the postoperative period. J Am Med Inform Assoc. 2011;18:164–8 (II).PubMedPubMedCentralCrossRef
244.
Zurück zum Zitat Lim WS, Baudouin SV, George RC, et al. BTS guidelines for the management of community acquired pneumonia in adults: update 2009. Thorax. 2009;64:iii1–55 (I).PubMedCrossRef Lim WS, Baudouin SV, George RC, et al. BTS guidelines for the management of community acquired pneumonia in adults: update 2009. Thorax. 2009;64:iii1–55 (I).PubMedCrossRef
245.
Zurück zum Zitat Wiersinga WJ, Bonten MJ, Boersma WG, et al. SWAB/NVALT (Dutch Working Party on Antibiotic Policy and Dutch Association of Chest Physicians) guidelines on the management of community-acquired pneumonia in adults. Neth J Med. 2012;70:90–101 (I).PubMed Wiersinga WJ, Bonten MJ, Boersma WG, et al. SWAB/NVALT (Dutch Working Party on Antibiotic Policy and Dutch Association of Chest Physicians) guidelines on the management of community-acquired pneumonia in adults. Neth J Med. 2012;70:90–101 (I).PubMed
246.
Zurück zum Zitat Hoffken G, Lorenz J, Kern W, et al. S3-guideline on ambulant acquired pneumonia and deep airway infections. Pneumologie. 2005;59:612–64 (I).PubMedCrossRef Hoffken G, Lorenz J, Kern W, et al. S3-guideline on ambulant acquired pneumonia and deep airway infections. Pneumologie. 2005;59:612–64 (I).PubMedCrossRef
247.
Zurück zum Zitat Siegel RE, Halpern NA, Almenoff PL, Lee A, Cashin R, Greene JG. A prospective randomized study of inpatient IV antibiotics for community-acquired pneumonia—the optimal duration of therapy. Chest. 1996;110:965–71 (I).PubMedCrossRef Siegel RE, Halpern NA, Almenoff PL, Lee A, Cashin R, Greene JG. A prospective randomized study of inpatient IV antibiotics for community-acquired pneumonia—the optimal duration of therapy. Chest. 1996;110:965–71 (I).PubMedCrossRef
248.
Zurück zum Zitat Athanassa Z, Makris G, Dimopoulos G, Falagas ME. Early switch to oral treatment in patients with moderate to severe community-acquired pneumonia: a meta-analysis. Drugs. 2008;68:2469–81 (I).PubMedCrossRef Athanassa Z, Makris G, Dimopoulos G, Falagas ME. Early switch to oral treatment in patients with moderate to severe community-acquired pneumonia: a meta-analysis. Drugs. 2008;68:2469–81 (I).PubMedCrossRef
249.
Zurück zum Zitat Oosterheert JJ, Bonten MJ, Schneider MM, et al. Effectiveness of early switch from intravenous to oral antibiotics in severe community acquired pneumonia: multicentre randomised trial. BMJ. 2006;333:1193 (I).PubMedPubMedCentralCrossRef Oosterheert JJ, Bonten MJ, Schneider MM, et al. Effectiveness of early switch from intravenous to oral antibiotics in severe community acquired pneumonia: multicentre randomised trial. BMJ. 2006;333:1193 (I).PubMedPubMedCentralCrossRef
250.
Zurück zum Zitat Rhew DC, Tu GS, Ofman J, Henning JM, Richards MS, Weingarten SR. Early switch and early discharge strategies in patients with community-acquired pneumonia: a meta-analysis. Arch Intern Med. 2001;161:722–7 (I).PubMedCrossRef Rhew DC, Tu GS, Ofman J, Henning JM, Richards MS, Weingarten SR. Early switch and early discharge strategies in patients with community-acquired pneumonia: a meta-analysis. Arch Intern Med. 2001;161:722–7 (I).PubMedCrossRef
251.
Zurück zum Zitat Castro-Guardiola A, Viejo-Rodriguez AL, Soler-Simon S, et al. Efficacy and safety of oral and early-switch therapy for community-acquired pneumonia: a randomized controlled trial. Am J Med. 2001;111:367–74 (I).PubMedCrossRef Castro-Guardiola A, Viejo-Rodriguez AL, Soler-Simon S, et al. Efficacy and safety of oral and early-switch therapy for community-acquired pneumonia: a randomized controlled trial. Am J Med. 2001;111:367–74 (I).PubMedCrossRef
252.
Zurück zum Zitat Li JZ, Willke RJ, Rittenhouse BE, Rybak MJ. Effect of linezolid versus vancomycin on length of hospital stay in patients with complicated skin and soft tissue infections caused by known or suspected methicillin-resistant Staphylococci: results from a randomized clinical trial. Surg Infect (Larchmt). 2003;4:57–70 (I).CrossRef Li JZ, Willke RJ, Rittenhouse BE, Rybak MJ. Effect of linezolid versus vancomycin on length of hospital stay in patients with complicated skin and soft tissue infections caused by known or suspected methicillin-resistant Staphylococci: results from a randomized clinical trial. Surg Infect (Larchmt). 2003;4:57–70 (I).CrossRef
253.
Zurück zum Zitat Chan R, Hemeryck L, O’Regan M, Clancy L, Feely J. Oral versus intravenous antibiotics for community acquired lower respiratory tract infection in a general hospital: open, randomised controlled trial. BMJ. 1995;310:1360–2 (I).PubMedPubMedCentralCrossRef Chan R, Hemeryck L, O’Regan M, Clancy L, Feely J. Oral versus intravenous antibiotics for community acquired lower respiratory tract infection in a general hospital: open, randomised controlled trial. BMJ. 1995;310:1360–2 (I).PubMedPubMedCentralCrossRef
254.
Zurück zum Zitat Omidvari K, de Boisblanc BP, Karam G, Nelson S, Haponik E, Summer W. Early transition to oral antibiotic therapy for community-acquired pneumonia: duration of therapy, clinical outcomes, and cost analysis. Respir Med. 1998;92:1032–9 (I).PubMedCrossRef Omidvari K, de Boisblanc BP, Karam G, Nelson S, Haponik E, Summer W. Early transition to oral antibiotic therapy for community-acquired pneumonia: duration of therapy, clinical outcomes, and cost analysis. Respir Med. 1998;92:1032–9 (I).PubMedCrossRef
255.
Zurück zum Zitat Manuel O, Burnand B, Bady P, et al. Impact of standardised review of intravenous antibiotic therapy 72 hours after prescription in two internal medicine wards. J Hosp Infect. 2010;74:326–31 (II).PubMedCrossRef Manuel O, Burnand B, Bady P, et al. Impact of standardised review of intravenous antibiotic therapy 72 hours after prescription in two internal medicine wards. J Hosp Infect. 2010;74:326–31 (II).PubMedCrossRef
256.
Zurück zum Zitat Ramirez JA, Vargas S, Ritter GW, et al. Early switch from intravenous to oral antibiotics and early hospital discharge: a prospective observational study of 200 consecutive patients with community-acquired pneumonia. Arch Intern Med. 1999;159:2449–54 (II).PubMedCrossRef Ramirez JA, Vargas S, Ritter GW, et al. Early switch from intravenous to oral antibiotics and early hospital discharge: a prospective observational study of 200 consecutive patients with community-acquired pneumonia. Arch Intern Med. 1999;159:2449–54 (II).PubMedCrossRef
257.
Zurück zum Zitat Lee RW, Lindstrom ST. Early switch to oral antibiotics and early discharge guidelines in the management of community-acquired pneumonia. Respirology. 2007;12:111–6 (II).PubMedCrossRef Lee RW, Lindstrom ST. Early switch to oral antibiotics and early discharge guidelines in the management of community-acquired pneumonia. Respirology. 2007;12:111–6 (II).PubMedCrossRef
258.
Zurück zum Zitat Mertz D, Koller M, Haller P, et al. Outcomes of early switching from intravenous to oral antibiotics on medical wards. J Antimicrob Chemother. 2009;64:188–99 (II).PubMedPubMedCentralCrossRef Mertz D, Koller M, Haller P, et al. Outcomes of early switching from intravenous to oral antibiotics on medical wards. J Antimicrob Chemother. 2009;64:188–99 (II).PubMedPubMedCentralCrossRef
259.
Zurück zum Zitat Nathan RV, Rhew DC, Murray C, Bratzler DW, Houck PM, Weingarten SR. In-hospital observation after antibiotic switch in pneumonia: a national evaluation. Am J Med. 2006;119:512–7 (IV).PubMedCrossRef Nathan RV, Rhew DC, Murray C, Bratzler DW, Houck PM, Weingarten SR. In-hospital observation after antibiotic switch in pneumonia: a national evaluation. Am J Med. 2006;119:512–7 (IV).PubMedCrossRef
260.
Zurück zum Zitat Bocquet N, Sergent AA, Jais JP, et al. Randomized trial of oral versus sequential IV/oral antibiotic for acute pyelonephritis in children. Pediatrics. 2012;129:e269–75 (I).PubMedCrossRef Bocquet N, Sergent AA, Jais JP, et al. Randomized trial of oral versus sequential IV/oral antibiotic for acute pyelonephritis in children. Pediatrics. 2012;129:e269–75 (I).PubMedCrossRef
261.
Zurück zum Zitat Klastersky J, Paesmans M. Risk-adapted strategy for the management of febrile neutropenia in cancer patients. Support Care Cancer. 2007;15:477–82 (IV).PubMedCrossRef Klastersky J, Paesmans M. Risk-adapted strategy for the management of febrile neutropenia in cancer patients. Support Care Cancer. 2007;15:477–82 (IV).PubMedCrossRef
262.
Zurück zum Zitat Bachur R, Pagon Z. Success of short-course parenteral antibiotic therapy for acute osteomyelitis of childhood. Clin Pediatr (Phila). 2007;46:30–5 (II).CrossRef Bachur R, Pagon Z. Success of short-course parenteral antibiotic therapy for acute osteomyelitis of childhood. Clin Pediatr (Phila). 2007;46:30–5 (II).CrossRef
263.
Zurück zum Zitat Kern WV. Risk assessment and treatment of low-risk patients with febrile neutropenia. Clin Infect Dis. 2006;42:533–40 (I).PubMedCrossRef Kern WV. Risk assessment and treatment of low-risk patients with febrile neutropenia. Clin Infect Dis. 2006;42:533–40 (I).PubMedCrossRef
264.
Zurück zum Zitat van NC, van’t Wout JW, Spelt IC, et al. Prospective cohort study of acute pyelonephritis in adults: safety of triage towards home based oral antimicrobial treatment. J Infect. 2010;60:114–21 (II).CrossRef van NC, van’t Wout JW, Spelt IC, et al. Prospective cohort study of acute pyelonephritis in adults: safety of triage towards home based oral antimicrobial treatment. J Infect. 2010;60:114–21 (II).CrossRef
265.
Zurück zum Zitat Jagodzinski NA, Kanwar R, Graham K, Bache CE. Prospective evaluation of a shortened regimen of treatment for acute osteomyelitis and septic arthritis in children. J Pediatr Orthop. 2009;29:518–25 (III).PubMedCrossRef Jagodzinski NA, Kanwar R, Graham K, Bache CE. Prospective evaluation of a shortened regimen of treatment for acute osteomyelitis and septic arthritis in children. J Pediatr Orthop. 2009;29:518–25 (III).PubMedCrossRef
266.
Zurück zum Zitat Stengel D, Bauwens K, Sehouli J, Ekkernkamp A, Porzsolt F. Systematic review and meta-analysis of antibiotic therapy for bone and joint infections. Lancet Infect Dis. 2001;1:175–88 (I).PubMedCrossRef Stengel D, Bauwens K, Sehouli J, Ekkernkamp A, Porzsolt F. Systematic review and meta-analysis of antibiotic therapy for bone and joint infections. Lancet Infect Dis. 2001;1:175–88 (I).PubMedCrossRef
267.
Zurück zum Zitat Cenizal MJ, Skiest D, Luber S, et al. Prospective randomized trial of empiric therapy with trimethoprim-sulfamethoxazole or doxycycline for outpatient skin and soft tissue infections in an area of high prevalence of methicillin-resistant Staphylococcus aureus. Antimicrob Agents Chemother. 2007;51:2628–30 (I).PubMedPubMedCentralCrossRef Cenizal MJ, Skiest D, Luber S, et al. Prospective randomized trial of empiric therapy with trimethoprim-sulfamethoxazole or doxycycline for outpatient skin and soft tissue infections in an area of high prevalence of methicillin-resistant Staphylococcus aureus. Antimicrob Agents Chemother. 2007;51:2628–30 (I).PubMedPubMedCentralCrossRef
268.
Zurück zum Zitat Hodson EM, Willis NS, Craig JC. Antibiotics for acute pyelonephritis in children. Cochrane Database Syst Rev. 2007;4:CD003772 (I).PubMed Hodson EM, Willis NS, Craig JC. Antibiotics for acute pyelonephritis in children. Cochrane Database Syst Rev. 2007;4:CD003772 (I).PubMed
269.
Zurück zum Zitat Neuhaus TJ, Berger C, Buechner K, et al. Randomised trial of oral versus sequential intravenous/oral cephalosporins in children with pyelonephritis. Eur J Pediatr. 2008;167:1037–47 (I).PubMedCrossRef Neuhaus TJ, Berger C, Buechner K, et al. Randomised trial of oral versus sequential intravenous/oral cephalosporins in children with pyelonephritis. Eur J Pediatr. 2008;167:1037–47 (I).PubMedCrossRef
270.
Zurück zum Zitat Vouloumanou EK, Rafailidis PI, Kazantzi MS, Athanasiou S, Falagas ME. Early switch to oral versus intravenous antimicrobial treatment for hospitalized patients with acute pyelonephritis: a systematic review of randomized controlled trials. Curr Med Res Opin. 2008;24:3423–34 (I).PubMedCrossRef Vouloumanou EK, Rafailidis PI, Kazantzi MS, Athanasiou S, Falagas ME. Early switch to oral versus intravenous antimicrobial treatment for hospitalized patients with acute pyelonephritis: a systematic review of randomized controlled trials. Curr Med Res Opin. 2008;24:3423–34 (I).PubMedCrossRef
271.
Zurück zum Zitat Hom J. Are oral antibiotics equivalent to intravenous antibiotics for the initial management of pyelonephritis in children? Paediatr Child Health. 2010;15:150–2 (I).PubMedPubMedCentral Hom J. Are oral antibiotics equivalent to intravenous antibiotics for the initial management of pyelonephritis in children? Paediatr Child Health. 2010;15:150–2 (I).PubMedPubMedCentral
272.
Zurück zum Zitat Kuti JL, Le TN, Nightingale CH, Nicolau DP, Quintiliani R. Pharmacoeconomics of a pharmacist-managed program for automatically converting levofloxacin route from i.v. to oral. Am J Health Syst Pharm. 2002;59:2209–15 (II).PubMed Kuti JL, Le TN, Nightingale CH, Nicolau DP, Quintiliani R. Pharmacoeconomics of a pharmacist-managed program for automatically converting levofloxacin route from i.v. to oral. Am J Health Syst Pharm. 2002;59:2209–15 (II).PubMed
273.
Zurück zum Zitat Rigaud B, Malbranche C, Pioud V, et al. Good clinical practices and inpatient antibiotics: optimization of fluoroquinolone switch therapy. Presse Med. 2007;36:1159–66 (II).PubMedCrossRef Rigaud B, Malbranche C, Pioud V, et al. Good clinical practices and inpatient antibiotics: optimization of fluoroquinolone switch therapy. Presse Med. 2007;36:1159–66 (II).PubMedCrossRef
274.
Zurück zum Zitat Lorgelly PK, Atkinson M, Lakhanpaul M, et al. Oral versus i.v. antibiotics for community-acquired pneumonia in children: a cost-minimisation analysis. Eur Respir J. 2010;35:858–64 (I).PubMedCrossRef Lorgelly PK, Atkinson M, Lakhanpaul M, et al. Oral versus i.v. antibiotics for community-acquired pneumonia in children: a cost-minimisation analysis. Eur Respir J. 2010;35:858–64 (I).PubMedCrossRef
275.
Zurück zum Zitat Przybylski KG, Rybak MJ, Martin PR, et al. A pharmacist-initiated program of intravenous to oral antibiotic conversion. Pharmacotherapy. 1997;17:271–6 (II).PubMed Przybylski KG, Rybak MJ, Martin PR, et al. A pharmacist-initiated program of intravenous to oral antibiotic conversion. Pharmacotherapy. 1997;17:271–6 (II).PubMed
276.
Zurück zum Zitat Vogtlander NP, Van Kasteren ME, Natsch S, Kullberg BJ, Hekster YA, Van Der Meer JW. Improving the process of antibiotic therapy in daily practice: interventions to optimize timing, dosage adjustment to renal function, and switch therapy. Arch Intern Med. 2004;164:1206–12 (II).PubMedCrossRef Vogtlander NP, Van Kasteren ME, Natsch S, Kullberg BJ, Hekster YA, Van Der Meer JW. Improving the process of antibiotic therapy in daily practice: interventions to optimize timing, dosage adjustment to renal function, and switch therapy. Arch Intern Med. 2004;164:1206–12 (II).PubMedCrossRef
277.
Zurück zum Zitat Waagsbo B, Sundoy A, Paulsen EQ. Reduction of unnecessary i.v. antibiotic days using general criteria for antibiotic switch. Scand J Infect Dis. 2008;40:468–73 (II).PubMedCrossRef Waagsbo B, Sundoy A, Paulsen EQ. Reduction of unnecessary i.v. antibiotic days using general criteria for antibiotic switch. Scand J Infect Dis. 2008;40:468–73 (II).PubMedCrossRef
278.
Zurück zum Zitat Shindo Y, Sato S, Maruyama E, et al. Implication of clinical pathway care for community-acquired pneumonia in a community hospital: early switch from an intravenous beta-lactam plus a macrolide to an oral respiratory fluoroquinolone. Intern Med. 2008;47:1865–74 (IV).PubMedCrossRef Shindo Y, Sato S, Maruyama E, et al. Implication of clinical pathway care for community-acquired pneumonia in a community hospital: early switch from an intravenous beta-lactam plus a macrolide to an oral respiratory fluoroquinolone. Intern Med. 2008;47:1865–74 (IV).PubMedCrossRef
279.
Zurück zum Zitat Buyle F, Vogelaers D, Peleman R, Van MG, Robays H. Implementation of guidelines for sequential therapy with fluoroquinolones in a Belgian hospital. Pharm World Sci. 2010;32:404–10 (II).PubMedCrossRef Buyle F, Vogelaers D, Peleman R, Van MG, Robays H. Implementation of guidelines for sequential therapy with fluoroquinolones in a Belgian hospital. Pharm World Sci. 2010;32:404–10 (II).PubMedCrossRef
280.
Zurück zum Zitat Dryden M, Saeed K, Townsend R, et al. Antibiotic stewardship and early discharge from hospital: impact of a structured approach to antimicrobial management. J Antimicrob Chemother. 2012;67:2289–96 (II).PubMedCrossRef Dryden M, Saeed K, Townsend R, et al. Antibiotic stewardship and early discharge from hospital: impact of a structured approach to antimicrobial management. J Antimicrob Chemother. 2012;67:2289–96 (II).PubMedCrossRef
281.
Zurück zum Zitat Metjian TA, Prasad PA, Kogon A, Coffin SE, Zaoutis TE. Evaluation of an antimicrobial stewardship program at a pediatric teaching hospital. Pediatr Infect Dis J. 2008;27:106–11 (II).PubMed Metjian TA, Prasad PA, Kogon A, Coffin SE, Zaoutis TE. Evaluation of an antimicrobial stewardship program at a pediatric teaching hospital. Pediatr Infect Dis J. 2008;27:106–11 (II).PubMed
282.
Zurück zum Zitat Hoefel HH, Lautert L, Schmitt C, Soares T, Jordan S. Vancomycin administration: mistakes made by nursing staff. Nurs Stand. 2008;22:35–42 (IV).PubMedCrossRef Hoefel HH, Lautert L, Schmitt C, Soares T, Jordan S. Vancomycin administration: mistakes made by nursing staff. Nurs Stand. 2008;22:35–42 (IV).PubMedCrossRef
283.
Zurück zum Zitat Magnotti LJ, Schroeppel TJ, Fabian TC, et al. Reduction in inadequate empiric antibiotic therapy for ventilator-associated pneumonia: impact of a unit-specific treatment pathway. Am Surg. 2008;74:516–22 (III).PubMed Magnotti LJ, Schroeppel TJ, Fabian TC, et al. Reduction in inadequate empiric antibiotic therapy for ventilator-associated pneumonia: impact of a unit-specific treatment pathway. Am Surg. 2008;74:516–22 (III).PubMed
284.
Zurück zum Zitat Dalley AJ, Lipman J, Venkatesh B, Rudd M, Roberts MS, Cross SE. Inadequate antimicrobial prophylaxis during surgery: a study of beta-lactam levels during burn debridement. J Antimicrob Chemother. 2007;60:166–9 (III).PubMedCrossRef Dalley AJ, Lipman J, Venkatesh B, Rudd M, Roberts MS, Cross SE. Inadequate antimicrobial prophylaxis during surgery: a study of beta-lactam levels during burn debridement. J Antimicrob Chemother. 2007;60:166–9 (III).PubMedCrossRef
285.
Zurück zum Zitat Luna CM, Aruj P, Niederman MS, et al. Appropriateness and delay to initiate therapy in ventilator-associated pneumonia. Eur Respir J. 2006;27:158–64 (III).PubMedCrossRef Luna CM, Aruj P, Niederman MS, et al. Appropriateness and delay to initiate therapy in ventilator-associated pneumonia. Eur Respir J. 2006;27:158–64 (III).PubMedCrossRef
286.
Zurück zum Zitat Ramsay C, Brown E, Hartman G, Davey P. Room for improvement: a systematic review of the quality of evaluations of interventions to improve hospital antibiotic prescribing. J Antimicrob Chemother. 2003;52:764–71 (I).PubMedCrossRef Ramsay C, Brown E, Hartman G, Davey P. Room for improvement: a systematic review of the quality of evaluations of interventions to improve hospital antibiotic prescribing. J Antimicrob Chemother. 2003;52:764–71 (I).PubMedCrossRef
287.
Zurück zum Zitat Zaragoza R, Artero A, Camarena JJ, Sancho S, Gonzalez R, Nogueira JM. The influence of inadequate empirical antimicrobial treatment on patients with bloodstream infections in an intensive care unit. Clin Microbiol Infect. 2003;9:412–8 (III).PubMedCrossRef Zaragoza R, Artero A, Camarena JJ, Sancho S, Gonzalez R, Nogueira JM. The influence of inadequate empirical antimicrobial treatment on patients with bloodstream infections in an intensive care unit. Clin Microbiol Infect. 2003;9:412–8 (III).PubMedCrossRef
288.
Zurück zum Zitat McKenzie C. Antibiotic dosing in critical illness. J Antimicrob Chemother. 2011;66:ii25–31 (IV).PubMedCrossRef McKenzie C. Antibiotic dosing in critical illness. J Antimicrob Chemother. 2011;66:ii25–31 (IV).PubMedCrossRef
289.
Zurück zum Zitat Cordova MA, Generali JA, White SJ, Godwin HN. The effects of two pharmacy intervention methods on cefazolin dosing intervel for surgical prophylaxis. Hospital pharmacy. 1986;21:405–14 (II). Cordova MA, Generali JA, White SJ, Godwin HN. The effects of two pharmacy intervention methods on cefazolin dosing intervel for surgical prophylaxis. Hospital pharmacy. 1986;21:405–14 (II).
290.
Zurück zum Zitat Avorn J, Soumerai SB, Taylor W, Wessels MR, Janousek J, Weiner M. Reduction of incorrect antibiotic dosing through a structured educational order form. Arch Intern Med. 1988;148:1720–4 (II).PubMedCrossRef Avorn J, Soumerai SB, Taylor W, Wessels MR, Janousek J, Weiner M. Reduction of incorrect antibiotic dosing through a structured educational order form. Arch Intern Med. 1988;148:1720–4 (II).PubMedCrossRef
291.
Zurück zum Zitat Burton ME, Ash CL, Hill DP Jr, Handy T, Shepherd MD, Vasko MR. A controlled trial of the cost benefit of computerized bayesian aminoglycoside administration. Clin Pharmacol Ther. 1991;49:685–94 (I).PubMedCrossRef Burton ME, Ash CL, Hill DP Jr, Handy T, Shepherd MD, Vasko MR. A controlled trial of the cost benefit of computerized bayesian aminoglycoside administration. Clin Pharmacol Ther. 1991;49:685–94 (I).PubMedCrossRef
292.
Zurück zum Zitat Destache CJ, Meyer SK, Bittner MJ, Hermann KG. Impact of a clinical pharmacokinetic service on patients treated with aminoglycosides: a cost-benefit analysis. Ther Drug Monit. 1990;12:419–26 (I).PubMedCrossRef Destache CJ, Meyer SK, Bittner MJ, Hermann KG. Impact of a clinical pharmacokinetic service on patients treated with aminoglycosides: a cost-benefit analysis. Ther Drug Monit. 1990;12:419–26 (I).PubMedCrossRef
293.
Zurück zum Zitat McNabb JJ, Nightingale CH, Quintiliani R, Nicolau DP. Cost-effectiveness of ceftazidime by continuous infusion versus intermittent infusion for nosocomial pneumonia. Pharmacotherapy. 2001;21:549–55 (II).PubMedCrossRef McNabb JJ, Nightingale CH, Quintiliani R, Nicolau DP. Cost-effectiveness of ceftazidime by continuous infusion versus intermittent infusion for nosocomial pneumonia. Pharmacotherapy. 2001;21:549–55 (II).PubMedCrossRef
294.
Zurück zum Zitat Grant EM, Kuti JL, Nicolau DP, Nightingale C, Quintiliani R. Clinical efficacy and pharmacoeconomics of a continuous-infusion piperacillin-tazobactam program in a large community teaching hospital. Pharmacotherapy. 2002;22:471–83 (II).PubMedCrossRef Grant EM, Kuti JL, Nicolau DP, Nightingale C, Quintiliani R. Clinical efficacy and pharmacoeconomics of a continuous-infusion piperacillin-tazobactam program in a large community teaching hospital. Pharmacotherapy. 2002;22:471–83 (II).PubMedCrossRef
295.
Zurück zum Zitat Scaglione F. Can PK/PD be used in everyday clinical practice. Int J Antimicrob Agents. 2002;19:349–53 (III).PubMedCrossRef Scaglione F. Can PK/PD be used in everyday clinical practice. Int J Antimicrob Agents. 2002;19:349–53 (III).PubMedCrossRef
296.
Zurück zum Zitat Kaufman SE, Donnell RW, Hickey WS. Rationale and evidence for extended infusion of piperacillin-tazobactam. Am J Health Syst Pharm. 2011;68:1521–6 (I).PubMedCrossRef Kaufman SE, Donnell RW, Hickey WS. Rationale and evidence for extended infusion of piperacillin-tazobactam. Am J Health Syst Pharm. 2011;68:1521–6 (I).PubMedCrossRef
297.
Zurück zum Zitat Duszynska W, Taccone FS, Switala M, Hurkacz M, Kowalska-Krochmal B, Kubler A. Continuous infusion of piperacillin/tazobactam in ventilator-associated pneumonia: a pilot study on efficacy and costs. Int J Antimicrob Agents. 2012;39:153–8 (III).PubMedCrossRef Duszynska W, Taccone FS, Switala M, Hurkacz M, Kowalska-Krochmal B, Kubler A. Continuous infusion of piperacillin/tazobactam in ventilator-associated pneumonia: a pilot study on efficacy and costs. Int J Antimicrob Agents. 2012;39:153–8 (III).PubMedCrossRef
298.
Zurück zum Zitat DeRyke CA, Lee SY, Kuti JL, Nicolau DP. Optimising dosing strategies of antibacterials utilising pharmacodynamic principles: impact on the development of resistance. Drugs. 2006;66:1–14 (IV).PubMedCrossRef DeRyke CA, Lee SY, Kuti JL, Nicolau DP. Optimising dosing strategies of antibacterials utilising pharmacodynamic principles: impact on the development of resistance. Drugs. 2006;66:1–14 (IV).PubMedCrossRef
299.
Zurück zum Zitat Owens RC Jr, Shorr AF. Rational dosing of antimicrobial agents: pharmacokinetic and pharmacodynamic strategies. Am J Health Syst Pharm. 2009;66:S23–30 (IV).PubMedCrossRef Owens RC Jr, Shorr AF. Rational dosing of antimicrobial agents: pharmacokinetic and pharmacodynamic strategies. Am J Health Syst Pharm. 2009;66:S23–30 (IV).PubMedCrossRef
300.
Zurück zum Zitat Markou N, Markantonis SL, Dimitrakis E, et al. Colistin serum concentrations after intravenous administration in critically ill patients with serious multidrug-resistant, gram-negative bacilli infections: a prospective, open-label, uncontrolled study. Clin Ther. 2008;30:143–51 (II).PubMedCrossRef Markou N, Markantonis SL, Dimitrakis E, et al. Colistin serum concentrations after intravenous administration in critically ill patients with serious multidrug-resistant, gram-negative bacilli infections: a prospective, open-label, uncontrolled study. Clin Ther. 2008;30:143–51 (II).PubMedCrossRef
301.
Zurück zum Zitat Daikos GL, Lolans VT, Jackson GG. First-exposure adaptive resistance to aminoglycoside antibiotics in vivo with meaning for optimal clinical use. Antimicrob Agents Chemother. 1991;35:117–23 (III).PubMedPubMedCentralCrossRef Daikos GL, Lolans VT, Jackson GG. First-exposure adaptive resistance to aminoglycoside antibiotics in vivo with meaning for optimal clinical use. Antimicrob Agents Chemother. 1991;35:117–23 (III).PubMedPubMedCentralCrossRef
302.
Zurück zum Zitat Gumbo T, Louie A, Deziel MR, Parsons LM, Salfinger M, Drusano GL. Selection of a moxifloxacin dose that suppresses drug resistance in Mycobacterium tuberculosis, by use of an in vitro pharmacodynamic infection model and mathematical modeling. J Infect Dis. 2004;190:1642–51 (III).PubMedCrossRef Gumbo T, Louie A, Deziel MR, Parsons LM, Salfinger M, Drusano GL. Selection of a moxifloxacin dose that suppresses drug resistance in Mycobacterium tuberculosis, by use of an in vitro pharmacodynamic infection model and mathematical modeling. J Infect Dis. 2004;190:1642–51 (III).PubMedCrossRef
303.
Zurück zum Zitat Henderson-Begg SK, Livermore DM, Hall LM. Effect of subinhibitory concentrations of antibiotics on mutation frequency in Streptococcus pneumoniae. J Antimicrob Chemother. 2006;57:849–54 (III).PubMedCrossRef Henderson-Begg SK, Livermore DM, Hall LM. Effect of subinhibitory concentrations of antibiotics on mutation frequency in Streptococcus pneumoniae. J Antimicrob Chemother. 2006;57:849–54 (III).PubMedCrossRef
304.
Zurück zum Zitat Olofsson SK, Geli P, Andersson DI, Cars O. Pharmacodynamic model to describe the concentration-dependent selection of cefotaxime-resistant Escherichia coli. Antimicrob Agents Chemother. 2005;49:5081–91 (III).PubMedPubMedCentralCrossRef Olofsson SK, Geli P, Andersson DI, Cars O. Pharmacodynamic model to describe the concentration-dependent selection of cefotaxime-resistant Escherichia coli. Antimicrob Agents Chemother. 2005;49:5081–91 (III).PubMedPubMedCentralCrossRef
305.
Zurück zum Zitat Tam VH, Schilling AN, Neshat S, Poole K, Melnick DA, Coyle EA. Optimization of meropenem minimum concentration/MIC ratio to suppress in vitro resistance of Pseudomonas aeruginosa. Antimicrob Agents Chemother. 2005;49:4920–7 (III).PubMedPubMedCentralCrossRef Tam VH, Schilling AN, Neshat S, Poole K, Melnick DA, Coyle EA. Optimization of meropenem minimum concentration/MIC ratio to suppress in vitro resistance of Pseudomonas aeruginosa. Antimicrob Agents Chemother. 2005;49:4920–7 (III).PubMedPubMedCentralCrossRef
306.
Zurück zum Zitat Smyth AR, Bhatt J. Once-daily versus multiple-daily dosing with intravenous aminoglycosides for cystic fibrosis. Cochrane Database Syst Rev. 2012;2:CD002009 (I).PubMed Smyth AR, Bhatt J. Once-daily versus multiple-daily dosing with intravenous aminoglycosides for cystic fibrosis. Cochrane Database Syst Rev. 2012;2:CD002009 (I).PubMed
307.
Zurück zum Zitat Sime FB, Roberts MS, Peake SL, Lipman J, Roberts JA. Does beta-lactam pharmacokinetic variability in critically ill patients justify therapeutic drug monitoring? A systematic review. Ann Intensive Care. 2012;2:35 (I).PubMedPubMedCentralCrossRef Sime FB, Roberts MS, Peake SL, Lipman J, Roberts JA. Does beta-lactam pharmacokinetic variability in critically ill patients justify therapeutic drug monitoring? A systematic review. Ann Intensive Care. 2012;2:35 (I).PubMedPubMedCentralCrossRef
308.
Zurück zum Zitat Roberts JA, Joynt GM, Choi GY, Gomersall CD, Lipman J. How to optimise antimicrobial prescriptions in the intensive care unit: principles of individualised dosing using pharmacokinetics and pharmacodynamics. Int J Antimicrob Agents. 2012;39:187–92 (IV).PubMedCrossRef Roberts JA, Joynt GM, Choi GY, Gomersall CD, Lipman J. How to optimise antimicrobial prescriptions in the intensive care unit: principles of individualised dosing using pharmacokinetics and pharmacodynamics. Int J Antimicrob Agents. 2012;39:187–92 (IV).PubMedCrossRef
309.
310.
Zurück zum Zitat Roberts JA, Webb S, Paterson D, Ho KM, Lipman J. A systematic review on clinical benefits of continuous administration of beta-lactam antibiotics. Crit Care Med. 2009;37:2071–8 (I).PubMedCrossRef Roberts JA, Webb S, Paterson D, Ho KM, Lipman J. A systematic review on clinical benefits of continuous administration of beta-lactam antibiotics. Crit Care Med. 2009;37:2071–8 (I).PubMedCrossRef
311.
Zurück zum Zitat Blondiaux N, Wallet F, Favory R, et al. Daily serum piperacillin monitoring is advisable in critically ill patients. Int J Antimicrob Agents. 2010;35:500–3 (II).PubMedCrossRef Blondiaux N, Wallet F, Favory R, et al. Daily serum piperacillin monitoring is advisable in critically ill patients. Int J Antimicrob Agents. 2010;35:500–3 (II).PubMedCrossRef
312.
Zurück zum Zitat van Lent-Evers NA, Mathot RA, Geus WP, van Hout BA, Vinks AA. Impact of goal-oriented and model-based clinical pharmacokinetic dosing of aminoglycosides on clinical outcome: a cost-effectiveness analysis. Ther Drug Monit. 1999;21:63–73 (I).PubMedCrossRef van Lent-Evers NA, Mathot RA, Geus WP, van Hout BA, Vinks AA. Impact of goal-oriented and model-based clinical pharmacokinetic dosing of aminoglycosides on clinical outcome: a cost-effectiveness analysis. Ther Drug Monit. 1999;21:63–73 (I).PubMedCrossRef
313.
Zurück zum Zitat Leon-Djian CB, Bourguignon L, Spath HM, Maire P. Cost-effectiveness analysis of active TDM in elderly patients treated with aminoglycosides. Therapie. 2011;66:445–52 (IV).PubMedCrossRef Leon-Djian CB, Bourguignon L, Spath HM, Maire P. Cost-effectiveness analysis of active TDM in elderly patients treated with aminoglycosides. Therapie. 2011;66:445–52 (IV).PubMedCrossRef
314.
Zurück zum Zitat Duszynska W. Pharmacokinetic-pharmacodynamic modelling of antibiotic therapy in severe sepsis. Anaesthesiol Intensive Ther. 2012;44:158–64 (IV).PubMed Duszynska W. Pharmacokinetic-pharmacodynamic modelling of antibiotic therapy in severe sepsis. Anaesthesiol Intensive Ther. 2012;44:158–64 (IV).PubMed
315.
Zurück zum Zitat Hennessy S, Leonard CE, Localio AR, et al. Prescriber adherence to pharmacokinetic monitoring service recommendations for aminoglycoside dosing and the risk of acute kidney injury. Int J Clin Pharmacol Ther. 2011;49:536–44 (III).PubMedCrossRef Hennessy S, Leonard CE, Localio AR, et al. Prescriber adherence to pharmacokinetic monitoring service recommendations for aminoglycoside dosing and the risk of acute kidney injury. Int J Clin Pharmacol Ther. 2011;49:536–44 (III).PubMedCrossRef
316.
Zurück zum Zitat Mueller EW, Boucher BA. The use of extended-interval aminoglycoside dosing strategies for the treatment of moderate-to-severe infections encountered in critically ill surgical patients. Surg Infect (Larchmt). 2009;10:563–70 (I).CrossRef Mueller EW, Boucher BA. The use of extended-interval aminoglycoside dosing strategies for the treatment of moderate-to-severe infections encountered in critically ill surgical patients. Surg Infect (Larchmt). 2009;10:563–70 (I).CrossRef
317.
Zurück zum Zitat Craig WA. Pharmacokinetic/pharmacodynamic parameters: rationale for antibacterial dosing of mice and men. Clin Infect Dis. 1998;26:1–10 (IV).PubMedCrossRef Craig WA. Pharmacokinetic/pharmacodynamic parameters: rationale for antibacterial dosing of mice and men. Clin Infect Dis. 1998;26:1–10 (IV).PubMedCrossRef
318.
Zurück zum Zitat Drusano GL. Antimicrobial pharmacodynamics: critical interactions of ‘bug and drug’. Nat Rev Microbiol. 2004;2:289–300 (IV).PubMedCrossRef Drusano GL. Antimicrobial pharmacodynamics: critical interactions of ‘bug and drug’. Nat Rev Microbiol. 2004;2:289–300 (IV).PubMedCrossRef
319.
Zurück zum Zitat Kasiakou SK, Lawrence KR, Choulis N, Falagas ME. Continuous versus intermittent intravenous administration of antibacterials with time-dependent action: a systematic review of pharmacokinetic and pharmacodynamic parameters. Drugs. 2005;65:2499–511 (I).PubMedCrossRef Kasiakou SK, Lawrence KR, Choulis N, Falagas ME. Continuous versus intermittent intravenous administration of antibacterials with time-dependent action: a systematic review of pharmacokinetic and pharmacodynamic parameters. Drugs. 2005;65:2499–511 (I).PubMedCrossRef
320.
Zurück zum Zitat Kasiakou SK, Sermaides GJ, Michalopoulos A, Soteriades ES, Falagas ME. Continuous versus intermittent intravenous administration of antibiotics: a meta-analysis of randomised controlled trials. Lancet Infect Dis. 2005;5:581–9 (I).PubMedCrossRef Kasiakou SK, Sermaides GJ, Michalopoulos A, Soteriades ES, Falagas ME. Continuous versus intermittent intravenous administration of antibiotics: a meta-analysis of randomised controlled trials. Lancet Infect Dis. 2005;5:581–9 (I).PubMedCrossRef
321.
Zurück zum Zitat Tamma PD, Putcha N, Suh YD, Van Arendonk KJ, Rinke ML. Does prolonged beta-lactam infusions improve clinical outcomes compared to intermittent infusions? A meta-analysis and systematic review of randomized, controlled trials. BMC Infect Dis. 2011;11:181 (I).PubMedPubMedCentralCrossRef Tamma PD, Putcha N, Suh YD, Van Arendonk KJ, Rinke ML. Does prolonged beta-lactam infusions improve clinical outcomes compared to intermittent infusions? A meta-analysis and systematic review of randomized, controlled trials. BMC Infect Dis. 2011;11:181 (I).PubMedPubMedCentralCrossRef
322.
Zurück zum Zitat McKinnon PS, Paladino JA, Schentag JJ. Evaluation of area under the inhibitory curve (AUIC) and time above the minimum inhibitory concentration (T>MIC) as predictors of outcome for cefepime and ceftazidime in serious bacterial infections. Int J Antimicrob Agents. 2008;31:345–51 (III).PubMedCrossRef McKinnon PS, Paladino JA, Schentag JJ. Evaluation of area under the inhibitory curve (AUIC) and time above the minimum inhibitory concentration (T>MIC) as predictors of outcome for cefepime and ceftazidime in serious bacterial infections. Int J Antimicrob Agents. 2008;31:345–51 (III).PubMedCrossRef
323.
Zurück zum Zitat Pelkonen T, Roine I, Cruzeiro ML, Pitkaranta A, Kataja M, Peltola H. Slow initial beta-lactam infusion and oral paracetamol to treat childhood bacterial meningitis: a randomised, controlled trial. Lancet Infect Dis. 2011;11:613–21 (I).PubMedCrossRef Pelkonen T, Roine I, Cruzeiro ML, Pitkaranta A, Kataja M, Peltola H. Slow initial beta-lactam infusion and oral paracetamol to treat childhood bacterial meningitis: a randomised, controlled trial. Lancet Infect Dis. 2011;11:613–21 (I).PubMedCrossRef
324.
Zurück zum Zitat Dandekar PK, Maglio D, Sutherland CA, Nightingale CH, Nicolau DP. Pharmacokinetics of meropenem 0.5 and 2 g every 8 hours as a 3-hour infusion. Pharmacotherapy. 2003;23:988–91 (III).PubMedCrossRef Dandekar PK, Maglio D, Sutherland CA, Nightingale CH, Nicolau DP. Pharmacokinetics of meropenem 0.5 and 2 g every 8 hours as a 3-hour infusion. Pharmacotherapy. 2003;23:988–91 (III).PubMedCrossRef
325.
Zurück zum Zitat Yost RJ, Cappelletty DM. The retrospective cohort of extended-infusion piperacillin-tazobactam (RECEIPT) study: a multicenter study. Pharmacotherapy. 2011;31:767–75 (III).PubMedCrossRef Yost RJ, Cappelletty DM. The retrospective cohort of extended-infusion piperacillin-tazobactam (RECEIPT) study: a multicenter study. Pharmacotherapy. 2011;31:767–75 (III).PubMedCrossRef
326.
Zurück zum Zitat Arnold HM, Hollands JM, Skrupky LP, et al. Prolonged infusion antibiotics for suspected gram-negative infections in the ICU: a before-after study. Ann Pharmacother. 2013;47:170–80 (II).PubMedCrossRef Arnold HM, Hollands JM, Skrupky LP, et al. Prolonged infusion antibiotics for suspected gram-negative infections in the ICU: a before-after study. Ann Pharmacother. 2013;47:170–80 (II).PubMedCrossRef
327.
Zurück zum Zitat Dulhunty JM, Roberts JA, Davis JS, et al. Continuous infusion of beta-lactam antibiotics in severe sepsis: a multicenter double-blind, randomized controlled trial. Clin Infect Dis. 2013;56:236–44 (I).PubMedCrossRef Dulhunty JM, Roberts JA, Davis JS, et al. Continuous infusion of beta-lactam antibiotics in severe sepsis: a multicenter double-blind, randomized controlled trial. Clin Infect Dis. 2013;56:236–44 (I).PubMedCrossRef
328.
Zurück zum Zitat Chytra I, Stepan M, Benes J, et al. Clinical and microbiological efficacy of continuous versus intermittent application of meropenem in critically ill patients: a randomized open-label controlled trial. Crit Care. 2012;16:R113 (I).PubMedPubMedCentralCrossRef Chytra I, Stepan M, Benes J, et al. Clinical and microbiological efficacy of continuous versus intermittent application of meropenem in critically ill patients: a randomized open-label controlled trial. Crit Care. 2012;16:R113 (I).PubMedPubMedCentralCrossRef
329.
Zurück zum Zitat Toltzis P, Dul MJ, Hoyen C, et al. The effect of antibiotic rotation on colonization with antibiotic-resistant bacilli in a neonatal intensive care unit. Pediatrics. 2002;110:707–11 (II).PubMedCrossRef Toltzis P, Dul MJ, Hoyen C, et al. The effect of antibiotic rotation on colonization with antibiotic-resistant bacilli in a neonatal intensive care unit. Pediatrics. 2002;110:707–11 (II).PubMedCrossRef
330.
Zurück zum Zitat de CC, Franchineau P, Gourgand JM, Loriette Y, Gaulme J, Sirot J. Clinical and bacteriological survey after change in aminoglycoside treatment to control an epidemic of Enterobacter cloacae. J Hosp Infect. 1994;28:219–29 (II).CrossRef de CC, Franchineau P, Gourgand JM, Loriette Y, Gaulme J, Sirot J. Clinical and bacteriological survey after change in aminoglycoside treatment to control an epidemic of Enterobacter cloacae. J Hosp Infect. 1994;28:219–29 (II).CrossRef
331.
Zurück zum Zitat Martinez JA, Nicolas JM, Marco F, et al. Comparison of antimicrobial cycling and mixing strategies in two medical intensive care units. Critical Care Medicine. 2006;34:329–36 (III).PubMedCrossRef Martinez JA, Nicolas JM, Marco F, et al. Comparison of antimicrobial cycling and mixing strategies in two medical intensive care units. Critical Care Medicine. 2006;34:329–36 (III).PubMedCrossRef
332.
Zurück zum Zitat Martinez JA, Delgado E, Marti S, et al. Influence of antipseudomonal agents on Pseudomonas aeruginosa colonization and acquisition of resistance in critically ill medical patients. Intensive Care Med. 2009;35:439–47 (III). Martinez JA, Delgado E, Marti S, et al. Influence of antipseudomonal agents on Pseudomonas aeruginosa colonization and acquisition of resistance in critically ill medical patients. Intensive Care Med. 2009;35:439–47 (III).
333.
Zurück zum Zitat Cadena J, Taboada CA, Burgess DS, et al. Antibiotic cycling to decrease bacterial antibiotic resistance: a 5-year experience on a bone marrow transplant unit. Bone Marrow Transpl. 2007;40:151–5 (IV).CrossRef Cadena J, Taboada CA, Burgess DS, et al. Antibiotic cycling to decrease bacterial antibiotic resistance: a 5-year experience on a bone marrow transplant unit. Bone Marrow Transpl. 2007;40:151–5 (IV).CrossRef
334.
Zurück zum Zitat Craig M, Cumpston AD, Hobbs GR, Devetten MP, Sarwari AR, Ericson SG. The clinical impact of antibacterial prophylaxis and cycling antibiotics for febrile neutropenia in a hematological malignancy and transplantation unit. Bone Marrow Transpl. 2007;39:477–82 (III).CrossRef Craig M, Cumpston AD, Hobbs GR, Devetten MP, Sarwari AR, Ericson SG. The clinical impact of antibacterial prophylaxis and cycling antibiotics for febrile neutropenia in a hematological malignancy and transplantation unit. Bone Marrow Transpl. 2007;39:477–82 (III).CrossRef
335.
Zurück zum Zitat de Araujo OR, da Silva DC, Diegues AR, et al. Cefepime restriction improves gram-negative overall resistance patterns in neonatal intensive care unit. Braz J Infect Dis. 2007;11:277–80 (II).PubMedCrossRef de Araujo OR, da Silva DC, Diegues AR, et al. Cefepime restriction improves gram-negative overall resistance patterns in neonatal intensive care unit. Braz J Infect Dis. 2007;11:277–80 (II).PubMedCrossRef
336.
Zurück zum Zitat Francetic I, Kalenic S, Huic M, et al. Impact of aminoglycoside cycling in six tertiary intensive care units: prospective longitudinal interventional study. Croat Med J. 2008;49:207–14 (IV).PubMedPubMedCentralCrossRef Francetic I, Kalenic S, Huic M, et al. Impact of aminoglycoside cycling in six tertiary intensive care units: prospective longitudinal interventional study. Croat Med J. 2008;49:207–14 (IV).PubMedPubMedCentralCrossRef
337.
Zurück zum Zitat Hedrick TL, Schulman AS, McElearney ST, et al. Outbreak of resistant Pseudomonas aeruginosa infections during a quarterly cycling antibiotic regimen. Surg Infect (Larchmt). 2008;9:139–52 (IV).CrossRef Hedrick TL, Schulman AS, McElearney ST, et al. Outbreak of resistant Pseudomonas aeruginosa infections during a quarterly cycling antibiotic regimen. Surg Infect (Larchmt). 2008;9:139–52 (IV).CrossRef
338.
Zurück zum Zitat Bonhoeffer S, Lipsitch M, Levin BR. Evaluating treatment protocols to prevent antibiotic resistance. Proc Natl Acad Sci USA. 1997;94:12106–11 (III).PubMedPubMedCentralCrossRef Bonhoeffer S, Lipsitch M, Levin BR. Evaluating treatment protocols to prevent antibiotic resistance. Proc Natl Acad Sci USA. 1997;94:12106–11 (III).PubMedPubMedCentralCrossRef
339.
Zurück zum Zitat Bergstrom CT, Lo M, Lipsitch M. Ecological theory suggests that antimicrobial cycling will not reduce antimicrobial resistance in hospitals. Proc Natl Acad Sci USA. 2004;101:13285–90 (III).PubMedPubMedCentralCrossRef Bergstrom CT, Lo M, Lipsitch M. Ecological theory suggests that antimicrobial cycling will not reduce antimicrobial resistance in hospitals. Proc Natl Acad Sci USA. 2004;101:13285–90 (III).PubMedPubMedCentralCrossRef
340.
Zurück zum Zitat Bal AM, Kumar A, Gould IM. Antibiotic heterogeneity: from concept to practice. Ann N Y Acad Sci. 2010;1213:81–91 (IV).PubMedCrossRef Bal AM, Kumar A, Gould IM. Antibiotic heterogeneity: from concept to practice. Ann N Y Acad Sci. 2010;1213:81–91 (IV).PubMedCrossRef
341.
Zurück zum Zitat Erdeljic V, Francetic I, Bosnjak Z, et al. Distributed lags time series analysis versus linear correlation analysis (Pearson’s r) in identifying the relationship between antipseudomonal antibiotic consumption and the susceptibility of Pseudomonas aeruginosa isolates in a single Intensive Care Unit of a tertiary hospital. Int J Antimicrob Agents. 2011;37:467–71 (III).PubMedCrossRef Erdeljic V, Francetic I, Bosnjak Z, et al. Distributed lags time series analysis versus linear correlation analysis (Pearson’s r) in identifying the relationship between antipseudomonal antibiotic consumption and the susceptibility of Pseudomonas aeruginosa isolates in a single Intensive Care Unit of a tertiary hospital. Int J Antimicrob Agents. 2011;37:467–71 (III).PubMedCrossRef
342.
Zurück zum Zitat Miliani K, L’Heriteau F, Lacave L, Carbonne A, Astagneau P. Imipenem and ciprofloxacin consumption as factors associated with high incidence rates of resistant Pseudomonas aeruginosa in hospitals in northern France. J Hosp Infect. 2011;77:343–7 (III).PubMedCrossRef Miliani K, L’Heriteau F, Lacave L, Carbonne A, Astagneau P. Imipenem and ciprofloxacin consumption as factors associated with high incidence rates of resistant Pseudomonas aeruginosa in hospitals in northern France. J Hosp Infect. 2011;77:343–7 (III).PubMedCrossRef
343.
Zurück zum Zitat Takesue Y, Nakajima K, Ichiki K, et al. Impact of a hospital-wide programme of heterogeneous antibiotic use on the development of antibiotic-resistant Gram-negative bacteria. J Hosp Infect. 2010;75:28–32 (II).PubMedCrossRef Takesue Y, Nakajima K, Ichiki K, et al. Impact of a hospital-wide programme of heterogeneous antibiotic use on the development of antibiotic-resistant Gram-negative bacteria. J Hosp Infect. 2010;75:28–32 (II).PubMedCrossRef
344.
Zurück zum Zitat Pluss-Suard C, Pannatier A, Kronenberg A, Muhlemann K, Zanetti G. Impact of antibiotic use on carbapenem resistance in Pseudomonas aeruginosa: is there a role for antibiotic diversity? Antimicrob Agents Chemother. 2013;57:1709–13 (III).PubMedPubMedCentralCrossRef Pluss-Suard C, Pannatier A, Kronenberg A, Muhlemann K, Zanetti G. Impact of antibiotic use on carbapenem resistance in Pseudomonas aeruginosa: is there a role for antibiotic diversity? Antimicrob Agents Chemother. 2013;57:1709–13 (III).PubMedPubMedCentralCrossRef
345.
Zurück zum Zitat Sandiumenge A, Lisboa T, Gomez F, Hernandez P, Canadell L, Rello J. Effect of antibiotic diversity on ventilator-associated pneumonia caused by ESKAPE organisms. Chest. 2011;140:643–51 (II).PubMedCrossRef Sandiumenge A, Lisboa T, Gomez F, Hernandez P, Canadell L, Rello J. Effect of antibiotic diversity on ventilator-associated pneumonia caused by ESKAPE organisms. Chest. 2011;140:643–51 (II).PubMedCrossRef
346.
Zurück zum Zitat Dancer SJ, Kirkpatrick P, Corcoran DS, Christison F, Farmer D, Robertson C. Approaching zero: temporal effects of a restrictive antibiotic policy on hospital-acquired Clostridium difficile, extended-spectrum beta-lactamase-producing coliforms and meticillin-resistant Staphylococcus aureus. Int J Antimicrob Agents. 2013;41:137–42 (II).PubMedCrossRef Dancer SJ, Kirkpatrick P, Corcoran DS, Christison F, Farmer D, Robertson C. Approaching zero: temporal effects of a restrictive antibiotic policy on hospital-acquired Clostridium difficile, extended-spectrum beta-lactamase-producing coliforms and meticillin-resistant Staphylococcus aureus. Int J Antimicrob Agents. 2013;41:137–42 (II).PubMedCrossRef
347.
Zurück zum Zitat Lee J, Pai H, Kim YK, et al. Control of extended-spectrum beta-lactamase-producing Escherichia coli and Klebsiella pneumoniae in a children’s hospital by changing antimicrobial agent usage policy. J Antimicrob Chemother. 2007;60:629–37 (II).PubMedCrossRef Lee J, Pai H, Kim YK, et al. Control of extended-spectrum beta-lactamase-producing Escherichia coli and Klebsiella pneumoniae in a children’s hospital by changing antimicrobial agent usage policy. J Antimicrob Chemother. 2007;60:629–37 (II).PubMedCrossRef
348.
Zurück zum Zitat Petrikkos G, Markogiannakis A, Papaparaskevas J, et al. Differences in the changes in resistance patterns to third- and fourth-generation cephalosporins and piperacillin/tazobactam among Klebsiella pneumoniae and Escherichia coli clinical isolates following a restriction policy in a Greek tertiary care hospital. Int J Antimicrob Agents. 2007;29:34–8 (II).PubMedCrossRef Petrikkos G, Markogiannakis A, Papaparaskevas J, et al. Differences in the changes in resistance patterns to third- and fourth-generation cephalosporins and piperacillin/tazobactam among Klebsiella pneumoniae and Escherichia coli clinical isolates following a restriction policy in a Greek tertiary care hospital. Int J Antimicrob Agents. 2007;29:34–8 (II).PubMedCrossRef
349.
Zurück zum Zitat Wen Z, Wei X, Xiao Y, et al. Intervention study of the association of antibiotic utilization measures with control of extended-spectrum beta-lactamase (ESBL)-producing bacteria. Microbes Infect. 2010;12:710–5 (II).PubMedCrossRef Wen Z, Wei X, Xiao Y, et al. Intervention study of the association of antibiotic utilization measures with control of extended-spectrum beta-lactamase (ESBL)-producing bacteria. Microbes Infect. 2010;12:710–5 (II).PubMedCrossRef
350.
Zurück zum Zitat Murki S, Jonnala S, Mohammed F, Reddy A. Restriction of cephalosporins and control of extended spectrum beta-lactamase producing gram negative bacteria in a neonatal intensive care unit. Indian Pediatr. 2010;47:785–8 (III).PubMedCrossRef Murki S, Jonnala S, Mohammed F, Reddy A. Restriction of cephalosporins and control of extended spectrum beta-lactamase producing gram negative bacteria in a neonatal intensive care unit. Indian Pediatr. 2010;47:785–8 (III).PubMedCrossRef
351.
Zurück zum Zitat Wilson ML, Gaido L. Laboratory diagnosis of urinary tract infections in adult patients. Clin Infect Dis. 2004;38:1150–8 (II).PubMedCrossRef Wilson ML, Gaido L. Laboratory diagnosis of urinary tract infections in adult patients. Clin Infect Dis. 2004;38:1150–8 (II).PubMedCrossRef
352.
Zurück zum Zitat Gatermann SG, Fünfstück R, Handrick W, et al. MiQ 2 Harnwegsinfektionen. 2. Auflage. 2005. München, Urban & Fischer. Mikrobiologisch-infektiologische Qualitätsstandards (MiQ). Gatermann SG, Fünfstück R, Handrick W, et al. MiQ 2 Harnwegsinfektionen. 2. Auflage. 2005. München, Urban & Fischer. Mikrobiologisch-infektiologische Qualitätsstandards (MiQ).
353.
Zurück zum Zitat Anevlavis S, Petroglou N, Tzavaras A, et al. A prospective study of the diagnostic utility of sputum Gram stain in pneumonia. J Infect. 2009;59:83–9 (II).PubMedCrossRef Anevlavis S, Petroglou N, Tzavaras A, et al. A prospective study of the diagnostic utility of sputum Gram stain in pneumonia. J Infect. 2009;59:83–9 (II).PubMedCrossRef
354.
Zurück zum Zitat Barenfanger J, Drake C, Leon N, Mueller T, Troutt T. Clinical and financial benefits of rapid detection of respiratory viruses: an outcomes study. J Clin Microbiol. 2000;38:2824–8 (III).PubMedPubMedCentral Barenfanger J, Drake C, Leon N, Mueller T, Troutt T. Clinical and financial benefits of rapid detection of respiratory viruses: an outcomes study. J Clin Microbiol. 2000;38:2824–8 (III).PubMedPubMedCentral
355.
Zurück zum Zitat Podbielski A, Herrmann M, Kniehl E, Mauch H, Rüssmann H. MiQ 7 Infektionen der tiefen Atemwege—Teil I. [2. Auflage]. 2010. München, Urban & Fischer. Mikrobiologisch-infektiologische Qualitätsstandards (MiQ). Podbielski A, Herrmann M, Kniehl E, Mauch H, Rüssmann H. MiQ 7 Infektionen der tiefen Atemwege—Teil I. [2. Auflage]. 2010. München, Urban & Fischer. Mikrobiologisch-infektiologische Qualitätsstandards (MiQ).
356.
Zurück zum Zitat Roson B, Carratala J, Verdaguer R, Dorca J, Manresa F, Gudiol F. Prospective study of the usefulness of sputum Gram stain in the initial approach to community-acquired pneumonia requiring hospitalization. Clin Infect Dis. 2000;31:869–74 (II).PubMedCrossRef Roson B, Carratala J, Verdaguer R, Dorca J, Manresa F, Gudiol F. Prospective study of the usefulness of sputum Gram stain in the initial approach to community-acquired pneumonia requiring hospitalization. Clin Infect Dis. 2000;31:869–74 (II).PubMedCrossRef
357.
Zurück zum Zitat Gleckman R, DeVita J, Hibert D, Pelletier C, Martin R. Sputum gram stain assessment in community-acquired bacteremic pneumonia. J Clin Microbiol. 1988;26:846–9 (II).PubMedPubMedCentral Gleckman R, DeVita J, Hibert D, Pelletier C, Martin R. Sputum gram stain assessment in community-acquired bacteremic pneumonia. J Clin Microbiol. 1988;26:846–9 (II).PubMedPubMedCentral
358.
Zurück zum Zitat Tenover FC. Potential impact of rapid diagnostic tests on improving antimicrobial use. Ann N Y Acad Sci. 2010;1213:70–80 (I).PubMedCrossRef Tenover FC. Potential impact of rapid diagnostic tests on improving antimicrobial use. Ann N Y Acad Sci. 2010;1213:70–80 (I).PubMedCrossRef
359.
Zurück zum Zitat Bauer KA, West JE, Balada-Llasat JM, Pancholi P, Stevenson KB, Goff DA. An antimicrobial stewardship program’s impact with rapid polymerase chain reaction methicillin-resistant Staphylococcus aureus/S. aureus blood culture test in patients with S. aureus bacteremia. Clin Infect Dis. 2010;51:1074–80 (II).PubMedCrossRef Bauer KA, West JE, Balada-Llasat JM, Pancholi P, Stevenson KB, Goff DA. An antimicrobial stewardship program’s impact with rapid polymerase chain reaction methicillin-resistant Staphylococcus aureus/S. aureus blood culture test in patients with S. aureus bacteremia. Clin Infect Dis. 2010;51:1074–80 (II).PubMedCrossRef
360.
Zurück zum Zitat Kerremans JJ, Verboom P, Stijnen T, et al. Rapid identification and antimicrobial susceptibility testing reduce antibiotic use and accelerate pathogen-directed antibiotic use. J Antimicrob Chemother. 2008;61:428–35 (I).PubMedCrossRef Kerremans JJ, Verboom P, Stijnen T, et al. Rapid identification and antimicrobial susceptibility testing reduce antibiotic use and accelerate pathogen-directed antibiotic use. J Antimicrob Chemother. 2008;61:428–35 (I).PubMedCrossRef
361.
Zurück zum Zitat Bouza E, Torres MV, Radice C, et al. Direct E-test (AB Biodisk) of respiratory samples improves antimicrobial use in ventilator-associated pneumonia. Clin Infect Dis. 2007;44:382–7 (I).PubMedCrossRef Bouza E, Torres MV, Radice C, et al. Direct E-test (AB Biodisk) of respiratory samples improves antimicrobial use in ventilator-associated pneumonia. Clin Infect Dis. 2007;44:382–7 (I).PubMedCrossRef
362.
Zurück zum Zitat Doern GV, Vautour R, Gaudet M, Levy B. Clinical impact of rapid in vitro susceptibility testing and bacterial identification. J Clin Microbiol. 1994;32:1757–62 (I).PubMedPubMedCentral Doern GV, Vautour R, Gaudet M, Levy B. Clinical impact of rapid in vitro susceptibility testing and bacterial identification. J Clin Microbiol. 1994;32:1757–62 (I).PubMedPubMedCentral
363.
Zurück zum Zitat Forrest GN, Roghmann MC, Toombs LS, et al. Peptide nucleic acid fluorescent in situ hybridization for hospital-acquired enterococcal bacteremia: delivering earlier effective antimicrobial therapy. Antimicrob Agents Chemother. 2008;52:3558–63 (II).PubMedPubMedCentralCrossRef Forrest GN, Roghmann MC, Toombs LS, et al. Peptide nucleic acid fluorescent in situ hybridization for hospital-acquired enterococcal bacteremia: delivering earlier effective antimicrobial therapy. Antimicrob Agents Chemother. 2008;52:3558–63 (II).PubMedPubMedCentralCrossRef
364.
Zurück zum Zitat Lucignano B, Ranno S, Liesenfeld O, et al. Multiplex PCR allows rapid and accurate diagnosis of bloodstream infections in newborns and children with suspected sepsis. J Clin Microbiol. 2011;49:2252–8 (III).PubMedPubMedCentralCrossRef Lucignano B, Ranno S, Liesenfeld O, et al. Multiplex PCR allows rapid and accurate diagnosis of bloodstream infections in newborns and children with suspected sepsis. J Clin Microbiol. 2011;49:2252–8 (III).PubMedPubMedCentralCrossRef
365.
Zurück zum Zitat Frye AM, Baker CA, Rustvold DL, et al. Clinical impact of a real-time PCR assay for rapid identification of staphylococcal bacteremia. J Clin Microbiol. 2012;50:127–33 (III).PubMedPubMedCentralCrossRef Frye AM, Baker CA, Rustvold DL, et al. Clinical impact of a real-time PCR assay for rapid identification of staphylococcal bacteremia. J Clin Microbiol. 2012;50:127–33 (III).PubMedPubMedCentralCrossRef
366.
Zurück zum Zitat Buchan BW, Riebe KM, Ledeboer NA. Comparison of the MALDI Biotyper system using Sepsityper specimen processing to routine microbiological methods for identification of bacteria from positive blood culture bottles. J Clin Microbiol. 2012;50:346–52 (III).PubMedPubMedCentralCrossRef Buchan BW, Riebe KM, Ledeboer NA. Comparison of the MALDI Biotyper system using Sepsityper specimen processing to routine microbiological methods for identification of bacteria from positive blood culture bottles. J Clin Microbiol. 2012;50:346–52 (III).PubMedPubMedCentralCrossRef
367.
Zurück zum Zitat Falguera M, Ruiz-Gonzalez A, Schoenenberger JA, et al. Prospective, randomised study to compare empirical treatment versus targeted treatment on the basis of the urine antigen results in hospitalised patients with community-acquired pneumonia. Thorax. 2010;65:101–6 (I).PubMedCrossRef Falguera M, Ruiz-Gonzalez A, Schoenenberger JA, et al. Prospective, randomised study to compare empirical treatment versus targeted treatment on the basis of the urine antigen results in hospitalised patients with community-acquired pneumonia. Thorax. 2010;65:101–6 (I).PubMedCrossRef
368.
Zurück zum Zitat Sorde R, Falco V, Lowak M, et al. Current and potential usefulness of pneumococcal urinary antigen detection in hospitalized patients with community-acquired pneumonia to guide antimicrobial therapy. Arch Intern Med. 2011;171:166–72 (II).PubMedCrossRef Sorde R, Falco V, Lowak M, et al. Current and potential usefulness of pneumococcal urinary antigen detection in hospitalized patients with community-acquired pneumonia to guide antimicrobial therapy. Arch Intern Med. 2011;171:166–72 (II).PubMedCrossRef
369.
Zurück zum Zitat Arbo MD, Snydman DR. Influence of blood culture results on antibiotic choice in the treatment of bacteremia. Arch Intern Med. 1994;154:2641–5 (II).PubMedCrossRef Arbo MD, Snydman DR. Influence of blood culture results on antibiotic choice in the treatment of bacteremia. Arch Intern Med. 1994;154:2641–5 (II).PubMedCrossRef
370.
Zurück zum Zitat Bouza E, Sousa D, Munoz P, Rodriguez-Creixems M, Fron C, Lechuz JG. Bloodstream infections: a trial of the impact of different methods of reporting positive blood culture results. Clin Infect Dis. 2004;39:1161–9 (I).PubMedCrossRef Bouza E, Sousa D, Munoz P, Rodriguez-Creixems M, Fron C, Lechuz JG. Bloodstream infections: a trial of the impact of different methods of reporting positive blood culture results. Clin Infect Dis. 2004;39:1161–9 (I).PubMedCrossRef
371.
Zurück zum Zitat Holtzman C, Whitney D, Barlam T, Miller NS. Assessment of impact of peptide nucleic acid fluorescence in situ hybridization for rapid identification of coagulase-negative staphylococci in the absence of antimicrobial stewardship intervention. J Clin Microbiol. 2011;49:1581–2 (II).PubMedPubMedCentralCrossRef Holtzman C, Whitney D, Barlam T, Miller NS. Assessment of impact of peptide nucleic acid fluorescence in situ hybridization for rapid identification of coagulase-negative staphylococci in the absence of antimicrobial stewardship intervention. J Clin Microbiol. 2011;49:1581–2 (II).PubMedPubMedCentralCrossRef
372.
Zurück zum Zitat Nathwani D. Antimicrobial prescribing policy and practice in Scotland: recommendations for good antimicrobial practice in acute hospitals. J Antimicrob Chemother. 2006;57:1189–96 (IV).PubMedCrossRef Nathwani D. Antimicrobial prescribing policy and practice in Scotland: recommendations for good antimicrobial practice in acute hospitals. J Antimicrob Chemother. 2006;57:1189–96 (IV).PubMedCrossRef
373.
Zurück zum Zitat Steffee CH, Morrell RM, Wasilauskas BL. Clinical use of rifampicin during routine reporting of rifampicin susceptibilities: a lesson in selective reporting of antimicrobial susceptibility data. J Antimicrob Chemother. 1997;40:595–8 (III).PubMedCrossRef Steffee CH, Morrell RM, Wasilauskas BL. Clinical use of rifampicin during routine reporting of rifampicin susceptibilities: a lesson in selective reporting of antimicrobial susceptibility data. J Antimicrob Chemother. 1997;40:595–8 (III).PubMedCrossRef
374.
Zurück zum Zitat McNulty CA, Lasseter GM, Charlett A, et al. Does laboratory antibiotic susceptibility reporting influence primary care prescribing in urinary tract infection and other infections? J Antimicrob Chemother. 2011;66:1396–404 (II).PubMedCrossRef McNulty CA, Lasseter GM, Charlett A, et al. Does laboratory antibiotic susceptibility reporting influence primary care prescribing in urinary tract infection and other infections? J Antimicrob Chemother. 2011;66:1396–404 (II).PubMedCrossRef
375.
Zurück zum Zitat Hsu J, Abad C, Dinh M, Safdar N. Prevention of endemic healthcare-associated Clostridium difficile infection: reviewing the evidence. Am J Gastroenterol. 2010;105:2327–39 (I).PubMedCrossRef Hsu J, Abad C, Dinh M, Safdar N. Prevention of endemic healthcare-associated Clostridium difficile infection: reviewing the evidence. Am J Gastroenterol. 2010;105:2327–39 (I).PubMedCrossRef
376.
Zurück zum Zitat Wilcox MH, Planche T. Clostridium difficile infection. BMJ. 2009;338:b2528 (IV).PubMed Wilcox MH, Planche T. Clostridium difficile infection. BMJ. 2009;338:b2528 (IV).PubMed
377.
Zurück zum Zitat Debast SB, Vaessen N, Choudry A, Wiegers-Ligtvoet EA, van den Berg RJ, Kuijper EJ. Successful combat of an outbreak due to Clostridium difficile PCR ribotype 027 and recognition of specific risk factors. Clin Microbiol Infect. 2009;15:427–34 (III).PubMedCrossRef Debast SB, Vaessen N, Choudry A, Wiegers-Ligtvoet EA, van den Berg RJ, Kuijper EJ. Successful combat of an outbreak due to Clostridium difficile PCR ribotype 027 and recognition of specific risk factors. Clin Microbiol Infect. 2009;15:427–34 (III).PubMedCrossRef
378.
Zurück zum Zitat Wilcox MH, Freeman J, Fawley W, et al. Long-term surveillance of cefotaxime and piperacillin-tazobactam prescribing and incidence of Clostridium difficile diarrhoea. J Antimicrob Chemother. 2004;54:168–72 (II).PubMedCrossRef Wilcox MH, Freeman J, Fawley W, et al. Long-term surveillance of cefotaxime and piperacillin-tazobactam prescribing and incidence of Clostridium difficile diarrhoea. J Antimicrob Chemother. 2004;54:168–72 (II).PubMedCrossRef
379.
Zurück zum Zitat Brahmi N, Blel Y, Kouraichi N, et al. Impact of ceftazidime restriction on gram-negative bacterial resistance in an intensive care unit. J Infect Chemother. 2006;12:190–4 (II).PubMedCrossRef Brahmi N, Blel Y, Kouraichi N, et al. Impact of ceftazidime restriction on gram-negative bacterial resistance in an intensive care unit. J Infect Chemother. 2006;12:190–4 (II).PubMedCrossRef
380.
Zurück zum Zitat Lipworth AD, Hyle EP, Fishman NO, et al. Limiting the emergence of extended-spectrum beta-lactamase-producing enterobacteriaceae: influence of patient population characteristics on the response to antimicrobial formulary interventions. Infect Control Hosp Epidemiol. 2006;27:279–86 (II).PubMedCrossRef Lipworth AD, Hyle EP, Fishman NO, et al. Limiting the emergence of extended-spectrum beta-lactamase-producing enterobacteriaceae: influence of patient population characteristics on the response to antimicrobial formulary interventions. Infect Control Hosp Epidemiol. 2006;27:279–86 (II).PubMedCrossRef
381.
Zurück zum Zitat Tangden T, Eriksson BM, Melhus A, Svennblad B, Cars O. Radical reduction of cephalosporin use at a tertiary hospital after educational antibiotic intervention during an outbreak of extended-spectrum beta-lactamase-producing Klebsiella pneumoniae. J Antimicrob Chemother. 2011;66:1161–7 (II-III).PubMedCrossRef Tangden T, Eriksson BM, Melhus A, Svennblad B, Cars O. Radical reduction of cephalosporin use at a tertiary hospital after educational antibiotic intervention during an outbreak of extended-spectrum beta-lactamase-producing Klebsiella pneumoniae. J Antimicrob Chemother. 2011;66:1161–7 (II-III).PubMedCrossRef
382.
Zurück zum Zitat Bamberger DM, Dahl SL. Impact of voluntary vs enforced compliance of 3Rd-generation cephalosporin use in a teaching hospital. Arch Intern Med. 1992;152:554–7 (II).PubMedCrossRef Bamberger DM, Dahl SL. Impact of voluntary vs enforced compliance of 3Rd-generation cephalosporin use in a teaching hospital. Arch Intern Med. 1992;152:554–7 (II).PubMedCrossRef
383.
Zurück zum Zitat Anglim AM, Klym B, Byers KE, Scheld WM, Farr BM. Effect of a vancomycin restriction policy on ordering practices during an outbreak of vancomycin-resistant Enterococcus faecium. Arch Intern Med. 1997;157:1132–6 (II).PubMedCrossRef Anglim AM, Klym B, Byers KE, Scheld WM, Farr BM. Effect of a vancomycin restriction policy on ordering practices during an outbreak of vancomycin-resistant Enterococcus faecium. Arch Intern Med. 1997;157:1132–6 (II).PubMedCrossRef
384.
Zurück zum Zitat May AK, Melton SM, McGwin G, Cross JM, Moser SA, Rue LW. Reduction of vancomycin-resistant enterococcal infections by limitation of broad-spectrum cephalosporin use in a trauma and burn intensive care unit. Shock. 2000;14:259–64 (II).PubMedCrossRef May AK, Melton SM, McGwin G, Cross JM, Moser SA, Rue LW. Reduction of vancomycin-resistant enterococcal infections by limitation of broad-spectrum cephalosporin use in a trauma and burn intensive care unit. Shock. 2000;14:259–64 (II).PubMedCrossRef
385.
Zurück zum Zitat Stiefel U, Paterson DL, Pultz NJ, Gordon SM, Aron DC, Donskey CJ. Effect of the increasing use of piperacillin/tazobactam on the incidence of vancomycin-resistant enterococci in four academic medical centers. Infect Contr Hosp Epidemiol. 2004;25:380–3 (IV).CrossRef Stiefel U, Paterson DL, Pultz NJ, Gordon SM, Aron DC, Donskey CJ. Effect of the increasing use of piperacillin/tazobactam on the incidence of vancomycin-resistant enterococci in four academic medical centers. Infect Contr Hosp Epidemiol. 2004;25:380–3 (IV).CrossRef
386.
Zurück zum Zitat de Man P, Verhoeven BAN, Verbrugh HA, Vos MC, van den Anker JN. An antibiotic policy to prevent emergence of resistant bacilli. Lancet. 2000;355:973–8 (II).PubMedCrossRef de Man P, Verhoeven BAN, Verbrugh HA, Vos MC, van den Anker JN. An antibiotic policy to prevent emergence of resistant bacilli. Lancet. 2000;355:973–8 (II).PubMedCrossRef
387.
Zurück zum Zitat Toltzis P, Yamashita T, Vilt L, et al. Antibiotic restriction does not alter endemic colonization with resistant Gram-negative rods in a pediatric intensive care unit. Crit Care Med. 1998;26:1893–9 (II).PubMedCrossRef Toltzis P, Yamashita T, Vilt L, et al. Antibiotic restriction does not alter endemic colonization with resistant Gram-negative rods in a pediatric intensive care unit. Crit Care Med. 1998;26:1893–9 (II).PubMedCrossRef
388.
Zurück zum Zitat Rahal JJ, Urban C, Horn D, et al. Glass restriction of cephalosporin use to control total cephalosporin resistance in nosocomial Klebsiella. JAMA J Am Med Assoc. 1998;280:1233–7 (II).CrossRef Rahal JJ, Urban C, Horn D, et al. Glass restriction of cephalosporin use to control total cephalosporin resistance in nosocomial Klebsiella. JAMA J Am Med Assoc. 1998;280:1233–7 (II).CrossRef
389.
Zurück zum Zitat Arda B, Sipahi OR, Yamazhan T, et al. Short-term effect of antibiotic control policy on the usage patterns and cost of antimicrobials, mortality, nosocomial infection rates and antibacterial resistance. J Infect. 2007;55:41–8 (III).PubMedCrossRef Arda B, Sipahi OR, Yamazhan T, et al. Short-term effect of antibiotic control policy on the usage patterns and cost of antimicrobials, mortality, nosocomial infection rates and antibacterial resistance. J Infect. 2007;55:41–8 (III).PubMedCrossRef
390.
Zurück zum Zitat Landman D, Chockalingam M, Quale JM. Reduction in the incidence of methicillin-resistant Staphylococcus aureus and ceftazidime-resistant Klebsiella pneumoniae following changes in a hospital antibiotic formulary. Clin Infect Dis. 1999;28:1062–6 (II).PubMedCrossRef Landman D, Chockalingam M, Quale JM. Reduction in the incidence of methicillin-resistant Staphylococcus aureus and ceftazidime-resistant Klebsiella pneumoniae following changes in a hospital antibiotic formulary. Clin Infect Dis. 1999;28:1062–6 (II).PubMedCrossRef
391.
Zurück zum Zitat Leverstein-van Hall MA, Fluit AC, Blok HE, et al. Control of nosocomial multiresistant Enterobacteriaceae using a temporary restrictive antibiotic agent policy. Eur J Clin Microbiol Infect Dis. 2001;20:785–91 (III).PubMedCrossRef Leverstein-van Hall MA, Fluit AC, Blok HE, et al. Control of nosocomial multiresistant Enterobacteriaceae using a temporary restrictive antibiotic agent policy. Eur J Clin Microbiol Infect Dis. 2001;20:785–91 (III).PubMedCrossRef
392.
Zurück zum Zitat Quale J, Landman D, Saurina G, Atwood E, DiTore V, Patel K. Manipulation of a hospital antimicrobial formulary to control an outbreak of vancomycin-resistant enterococci. Clin Infect Dis. 1996;23:1020–5 (II).PubMedCrossRef Quale J, Landman D, Saurina G, Atwood E, DiTore V, Patel K. Manipulation of a hospital antimicrobial formulary to control an outbreak of vancomycin-resistant enterococci. Clin Infect Dis. 1996;23:1020–5 (II).PubMedCrossRef
393.
Zurück zum Zitat Lautenbach E, Larosa LA, Marr AM, Nachamkin I, Bilker WB, Fishman NO. Changes in the prevalence of vancomycin-resistant enterococci in response to antimicrobial formulary interventions: impact of progressive restrictions on use of vancomycin and third-generation cephalosporins. Clin Infect Dis. 2003;36:440–6 (II).PubMedCrossRef Lautenbach E, Larosa LA, Marr AM, Nachamkin I, Bilker WB, Fishman NO. Changes in the prevalence of vancomycin-resistant enterococci in response to antimicrobial formulary interventions: impact of progressive restrictions on use of vancomycin and third-generation cephalosporins. Clin Infect Dis. 2003;36:440–6 (II).PubMedCrossRef
394.
Zurück zum Zitat Zhou JJ, Patel SJ, Jia H, et al. Clinicians’ knowledge, attitudes, and practices regarding infections with multidrug-resistant gram-negative bacilli in intensive care units. Infect Control Hosp Epidemiol. 2013;34:274–83 (III).PubMedPubMedCentralCrossRef Zhou JJ, Patel SJ, Jia H, et al. Clinicians’ knowledge, attitudes, and practices regarding infections with multidrug-resistant gram-negative bacilli in intensive care units. Infect Control Hosp Epidemiol. 2013;34:274–83 (III).PubMedPubMedCentralCrossRef
395.
Zurück zum Zitat Pearson SA, Moxey A, Robertson J, et al. Do computerised clinical decision support systems for prescribing change practice? A systematic review of the literature (1990–2007). BMC Health Serv Res. 2009;9:154 (I).PubMedPubMedCentralCrossRef Pearson SA, Moxey A, Robertson J, et al. Do computerised clinical decision support systems for prescribing change practice? A systematic review of the literature (1990–2007). BMC Health Serv Res. 2009;9:154 (I).PubMedPubMedCentralCrossRef
396.
Zurück zum Zitat Linder JA, Schnipper JL, Tsurikova R, et al. Documentation-based clinical decision support to improve antibiotic prescribing for acute respiratory infections in primary care: a cluster randomised controlled trial. Inform Prim Care. 2009;17:231–40 (I).PubMed Linder JA, Schnipper JL, Tsurikova R, et al. Documentation-based clinical decision support to improve antibiotic prescribing for acute respiratory infections in primary care: a cluster randomised controlled trial. Inform Prim Care. 2009;17:231–40 (I).PubMed
397.
Zurück zum Zitat Haux R, Swinkels W, Ball M, Knaup P, Lun KC. Transformation of health care through innovative use of information technology: challenges for health and medical informatics education. Int J Med Inform. 1998;50:1–6 (IV).PubMedCrossRef Haux R, Swinkels W, Ball M, Knaup P, Lun KC. Transformation of health care through innovative use of information technology: challenges for health and medical informatics education. Int J Med Inform. 1998;50:1–6 (IV).PubMedCrossRef
398.
Zurück zum Zitat Haux R, Knaup P, Bauer AW, et al. Information processing in healthcare at the start of the third millennium: potential and limitations. Methods Inf Med. 2001;40:156–62 (IV).PubMed Haux R, Knaup P, Bauer AW, et al. Information processing in healthcare at the start of the third millennium: potential and limitations. Methods Inf Med. 2001;40:156–62 (IV).PubMed
399.
Zurück zum Zitat Haux R. Health care in the information society: what should be the role of medical informatics? Methods Inf Med. 2002;41:31–5 (IV).PubMed Haux R. Health care in the information society: what should be the role of medical informatics? Methods Inf Med. 2002;41:31–5 (IV).PubMed
400.
Zurück zum Zitat Haux R, Ammenwerth E, Herzog W, Knaup P. Health care in the information society. A prognosis for the year 2013. Int J Med Inform. 2002;66:3–21 (IV).PubMedCrossRef Haux R, Ammenwerth E, Herzog W, Knaup P. Health care in the information society. A prognosis for the year 2013. Int J Med Inform. 2002;66:3–21 (IV).PubMedCrossRef
401.
Zurück zum Zitat Haux R. Health information systems—past, present, future. Int J Med Inform. 2006;75:268–81 (IV).PubMedCrossRef Haux R. Health information systems—past, present, future. Int J Med Inform. 2006;75:268–81 (IV).PubMedCrossRef
402.
Zurück zum Zitat Cunningham TR, Geller ES, Clarke SW. Impact of electronic prescribing in a hospital setting: a process-focused evaluation. Int J Med Inform. 2008;77:546–54 (III).PubMedCrossRef Cunningham TR, Geller ES, Clarke SW. Impact of electronic prescribing in a hospital setting: a process-focused evaluation. Int J Med Inform. 2008;77:546–54 (III).PubMedCrossRef
403.
Zurück zum Zitat Sturzlinger H, Hiebinger C, Pertl D, Traurig P. Computerized physician order entry—effectiveness and efficiency of electronic medication ordering with decision support systems. GMS Health Technol Assess. 2009;5:Doc07 (I).PubMedPubMedCentral Sturzlinger H, Hiebinger C, Pertl D, Traurig P. Computerized physician order entry—effectiveness and efficiency of electronic medication ordering with decision support systems. GMS Health Technol Assess. 2009;5:Doc07 (I).PubMedPubMedCentral
404.
Zurück zum Zitat Di Pentima MC, Chan S, Eppes SC, Klein JD. Antimicrobial prescription errors in hospitalized children: role of antimicrobial stewardship program in detection and intervention. Clin Pediatr (Phila). 2009;48:505–12 (II).CrossRef Di Pentima MC, Chan S, Eppes SC, Klein JD. Antimicrobial prescription errors in hospitalized children: role of antimicrobial stewardship program in detection and intervention. Clin Pediatr (Phila). 2009;48:505–12 (II).CrossRef
405.
Zurück zum Zitat Kazemi A, Ellenius J, Pourasghar F, et al. The effect of computerized physician order entry and decision support system on medication errors in the neonatal ward: experiences from an Iranian teaching hospital. J Med Syst. 2011;35:25–37 (II).PubMedCrossRef Kazemi A, Ellenius J, Pourasghar F, et al. The effect of computerized physician order entry and decision support system on medication errors in the neonatal ward: experiences from an Iranian teaching hospital. J Med Syst. 2011;35:25–37 (II).PubMedCrossRef
406.
Zurück zum Zitat Cox ZL, Nelsen CL, Waitman LR, McCoy JA, Peterson JF. Effects of clinical decision support on initial dosing and monitoring of tobramycin and amikacin. Am J Health Syst Pharm. 2011;68:624–32 (II).PubMedPubMedCentralCrossRef Cox ZL, Nelsen CL, Waitman LR, McCoy JA, Peterson JF. Effects of clinical decision support on initial dosing and monitoring of tobramycin and amikacin. Am J Health Syst Pharm. 2011;68:624–32 (II).PubMedPubMedCentralCrossRef
407.
Zurück zum Zitat Traugott KA, Maxwell PR, Green K, Frei C, Lewis JS. Effects of therapeutic drug monitoring criteria in a computerized prescriber-order-entry system on the appropriateness of vancomycin level orders. Am J Health Syst Pharm. 2011;68:347–52 (II).PubMedCrossRef Traugott KA, Maxwell PR, Green K, Frei C, Lewis JS. Effects of therapeutic drug monitoring criteria in a computerized prescriber-order-entry system on the appropriateness of vancomycin level orders. Am J Health Syst Pharm. 2011;68:347–52 (II).PubMedCrossRef
408.
Zurück zum Zitat Kim JY, Sohn JW, Park DW, Yoon YK, Kim YM, Kim MJ. Control of extended-spectrum {beta}-lactamase-producing Klebsiella pneumoniae using a computer-assisted management program to restrict third-generation cephalosporin use. J Antimicrob Chemother. 2008;62:416–21 (II).PubMedCrossRef Kim JY, Sohn JW, Park DW, Yoon YK, Kim YM, Kim MJ. Control of extended-spectrum {beta}-lactamase-producing Klebsiella pneumoniae using a computer-assisted management program to restrict third-generation cephalosporin use. J Antimicrob Chemother. 2008;62:416–21 (II).PubMedCrossRef
409.
Zurück zum Zitat Yong MK, Buising KL, Cheng AC, Thursky KA. Improved susceptibility of Gram-negative bacteria in an intensive care unit following implementation of a computerized antibiotic decision support system. J Antimicrob Chemother. 2010;65:1062–9 (II).PubMedCrossRef Yong MK, Buising KL, Cheng AC, Thursky KA. Improved susceptibility of Gram-negative bacteria in an intensive care unit following implementation of a computerized antibiotic decision support system. J Antimicrob Chemother. 2010;65:1062–9 (II).PubMedCrossRef
410.
Zurück zum Zitat Garg AX, Adhikari NK, McDonald H, et al. Effects of computerized clinical decision support systems on practitioner performance and patient outcomes: a systematic review. JAMA. 2005;293:1223–38 (I).PubMedCrossRef Garg AX, Adhikari NK, McDonald H, et al. Effects of computerized clinical decision support systems on practitioner performance and patient outcomes: a systematic review. JAMA. 2005;293:1223–38 (I).PubMedCrossRef
411.
Zurück zum Zitat McKinley BA, Moore LJ, Sucher JF, et al. Computer protocol facilitates evidence-based care of sepsis in the surgical intensive care unit. J Trauma. 2011;70:1153–66 (III).PubMedCrossRef McKinley BA, Moore LJ, Sucher JF, et al. Computer protocol facilitates evidence-based care of sepsis in the surgical intensive care unit. J Trauma. 2011;70:1153–66 (III).PubMedCrossRef
412.
Zurück zum Zitat Thursky K. Use of computerized decision support systems to improve antibiotic prescribing. Expert Rev Anti Infect Ther. 2006;4:491–507 (I).PubMedCrossRef Thursky K. Use of computerized decision support systems to improve antibiotic prescribing. Expert Rev Anti Infect Ther. 2006;4:491–507 (I).PubMedCrossRef
413.
Zurück zum Zitat Sintchenko V, Coiera E, Gilbert GL. Decision support systems for antibiotic prescribing. Curr Opin Infect Dis. 2008;21:573–9 (I).PubMedCrossRef Sintchenko V, Coiera E, Gilbert GL. Decision support systems for antibiotic prescribing. Curr Opin Infect Dis. 2008;21:573–9 (I).PubMedCrossRef
414.
Zurück zum Zitat Di Pentima MC, Chan S. Impact of antimicrobial stewardship program on vancomycin use in a pediatric teaching hospital. Pediatr Infect Dis J. 2010;29:707–11 (II).PubMedCrossRef Di Pentima MC, Chan S. Impact of antimicrobial stewardship program on vancomycin use in a pediatric teaching hospital. Pediatr Infect Dis J. 2010;29:707–11 (II).PubMedCrossRef
415.
Zurück zum Zitat Lesprit P, Duong T, Girou E, Hemery F, Brun-Buisson C. Impact of a computer-generated alert system prompting review of antibiotic use in hospitals. J Antimicrob Chemother. 2009;63:1058–63 (II).PubMedCrossRef Lesprit P, Duong T, Girou E, Hemery F, Brun-Buisson C. Impact of a computer-generated alert system prompting review of antibiotic use in hospitals. J Antimicrob Chemother. 2009;63:1058–63 (II).PubMedCrossRef
416.
Zurück zum Zitat Hulgan T, Rosenbloom ST, Hargrove F, et al. Oral quinolones in hospitalized patients: an evaluation of a computerized decision support intervention. J Intern Med. 2004;256:349–57 (II).PubMedCrossRef Hulgan T, Rosenbloom ST, Hargrove F, et al. Oral quinolones in hospitalized patients: an evaluation of a computerized decision support intervention. J Intern Med. 2004;256:349–57 (II).PubMedCrossRef
417.
Zurück zum Zitat McGregor JC, Weekes E, Forrest GN, et al. Impact of a computerized clinical decision support system on reducing inappropriate antimicrobial use: a randomized controlled trial. J Am Med Inform Assoc. 2006;13:378–84 (I).PubMedPubMedCentralCrossRef McGregor JC, Weekes E, Forrest GN, et al. Impact of a computerized clinical decision support system on reducing inappropriate antimicrobial use: a randomized controlled trial. J Am Med Inform Assoc. 2006;13:378–84 (I).PubMedPubMedCentralCrossRef
418.
Zurück zum Zitat Shojania KG, Yokoe D, Platt R, Fiskio J, Ma’luf N, Bates DW. Reducing vancomycin use utilizing a computer guideline: results of a randomized controlled trial. J Am Med Inform Assoc. 1998;5:554–62 (I).PubMedPubMedCentralCrossRef Shojania KG, Yokoe D, Platt R, Fiskio J, Ma’luf N, Bates DW. Reducing vancomycin use utilizing a computer guideline: results of a randomized controlled trial. J Am Med Inform Assoc. 1998;5:554–62 (I).PubMedPubMedCentralCrossRef
419.
Zurück zum Zitat Rubinstein E, Barzilai A, Segev S, et al. Antibiotic cost reduction by providing cost information. Eur J Clin Pharmacol. 1988;35:269–72 (II).PubMedCrossRef Rubinstein E, Barzilai A, Segev S, et al. Antibiotic cost reduction by providing cost information. Eur J Clin Pharmacol. 1988;35:269–72 (II).PubMedCrossRef
420.
Zurück zum Zitat Parrino TA. The nonvalue of retrospective peer comparison feedback in containing hospital antibiotic costs. Am J Med. 1989;86:442–8 (II).PubMedCrossRef Parrino TA. The nonvalue of retrospective peer comparison feedback in containing hospital antibiotic costs. Am J Med. 1989;86:442–8 (II).PubMedCrossRef
421.
Zurück zum Zitat O’Reilly M, Talsma A, VanRiper S, Kheterpal S, Burney R. An anesthesia information system designed to provide physician-specific feedback improves timely administration of prophylactic antibiotics. Anesth Analg. 2006;103:908–12 (II).PubMedCrossRef O’Reilly M, Talsma A, VanRiper S, Kheterpal S, Burney R. An anesthesia information system designed to provide physician-specific feedback improves timely administration of prophylactic antibiotics. Anesth Analg. 2006;103:908–12 (II).PubMedCrossRef
422.
Zurück zum Zitat Nair BG, Newman SF, Peterson GN, Wu WY, Schwid HA. Feedback mechanisms including real-time electronic alerts to achieve near 100% timely prophylactic antibiotic administration in surgical cases. Anesth Analg. 2010;111:1293–300 (II).PubMedCrossRef Nair BG, Newman SF, Peterson GN, Wu WY, Schwid HA. Feedback mechanisms including real-time electronic alerts to achieve near 100% timely prophylactic antibiotic administration in surgical cases. Anesth Analg. 2010;111:1293–300 (II).PubMedCrossRef
423.
Zurück zum Zitat Zanetti G, Flanagan HL Jr, Cohn LH, Giardina R, Platt R. Improvement of intraoperative antibiotic prophylaxis in prolonged cardiac surgery by automated alerts in the operating room. Infect Control Hosp Epidemiol. 2003;24:13–6 (II).PubMedCrossRef Zanetti G, Flanagan HL Jr, Cohn LH, Giardina R, Platt R. Improvement of intraoperative antibiotic prophylaxis in prolonged cardiac surgery by automated alerts in the operating room. Infect Control Hosp Epidemiol. 2003;24:13–6 (II).PubMedCrossRef
424.
Zurück zum Zitat Burke JP, Classen DC, Pestotnik SL, Evans RS, Stevens LE. The HELP system and its application to infection control. J Hosp Infect. 1991;18:424–31 (IV).PubMedCrossRef Burke JP, Classen DC, Pestotnik SL, Evans RS, Stevens LE. The HELP system and its application to infection control. J Hosp Infect. 1991;18:424–31 (IV).PubMedCrossRef
425.
Zurück zum Zitat Burke JP. Surveillance, reporting, automation, and interventional epidemiology. Infect Control Hosp Epidemiol. 2003;24:10–2 (IV).PubMedCrossRef Burke JP. Surveillance, reporting, automation, and interventional epidemiology. Infect Control Hosp Epidemiol. 2003;24:10–2 (IV).PubMedCrossRef
426.
Zurück zum Zitat Evans RS, Larsen RA, Burke JP, et al. Computer surveillance of hospital-acquired infections and antibiotic use. JAMA J Am Med Assoc. 1986;256:1007–11 (II).CrossRef Evans RS, Larsen RA, Burke JP, et al. Computer surveillance of hospital-acquired infections and antibiotic use. JAMA J Am Med Assoc. 1986;256:1007–11 (II).CrossRef
427.
Zurück zum Zitat Evans RS, Pestotnik SL, Classen DC, et al. A computer-assisted management program for antibiotics and other antiinfective agents. N Engl J Med. 1998;338:232–8 (II).PubMedCrossRef Evans RS, Pestotnik SL, Classen DC, et al. A computer-assisted management program for antibiotics and other antiinfective agents. N Engl J Med. 1998;338:232–8 (II).PubMedCrossRef
428.
Zurück zum Zitat Evans RS, Pestotnik SL, Classen DC, Burke JP. Evaluation of a computer-assisted antibiotic-dose monitor. Ann Pharmacother. 1999;33:1026–31 (II).PubMedCrossRef Evans RS, Pestotnik SL, Classen DC, Burke JP. Evaluation of a computer-assisted antibiotic-dose monitor. Ann Pharmacother. 1999;33:1026–31 (II).PubMedCrossRef
429.
Zurück zum Zitat Pestotnik SL, Classen DC, Evans RS, Burke JP. Implementing antibiotic practice guidelines through computer-assisted decision support: clinical and financial outcomes. Ann Intern Med. 1996;124:884 (II).PubMedCrossRef Pestotnik SL, Classen DC, Evans RS, Burke JP. Implementing antibiotic practice guidelines through computer-assisted decision support: clinical and financial outcomes. Ann Intern Med. 1996;124:884 (II).PubMedCrossRef
430.
Zurück zum Zitat Mullett CJ, Evans RS, Christenson JC, Dean JM. Development and impact of a computerized pediatric antiinfective decision support program. Pediatrics. 2001;108:art-e75 (II).CrossRef Mullett CJ, Evans RS, Christenson JC, Dean JM. Development and impact of a computerized pediatric antiinfective decision support program. Pediatrics. 2001;108:art-e75 (II).CrossRef
431.
Zurück zum Zitat Mullett CJ, Thomas JG, Smith CL, Sarwari AR, Khakoo RA. Computerized antimicrobial decision support: an offline evaluation of a database-driven empiric antimicrobial guidance program in hospitalized patients with a bloodstream infection. Int J Med Inform. 2004;73:455–60 (IV).PubMedCrossRef Mullett CJ, Thomas JG, Smith CL, Sarwari AR, Khakoo RA. Computerized antimicrobial decision support: an offline evaluation of a database-driven empiric antimicrobial guidance program in hospitalized patients with a bloodstream infection. Int J Med Inform. 2004;73:455–60 (IV).PubMedCrossRef
432.
Zurück zum Zitat Buising KL, Thursky KA, Robertson MB, et al. Electronic antibiotic stewardship—reduced consumption of broad-spectrum antibiotics using a computerized antimicrobial approval system in a hospital setting. J Antimicrob Chemother. 2008;62:608–16 (II).PubMedCrossRef Buising KL, Thursky KA, Robertson MB, et al. Electronic antibiotic stewardship—reduced consumption of broad-spectrum antibiotics using a computerized antimicrobial approval system in a hospital setting. J Antimicrob Chemother. 2008;62:608–16 (II).PubMedCrossRef
433.
Zurück zum Zitat Paul M, Andreassen S, Tacconelli E, et al. Improving empirical antibiotic treatment using TREAT, a computerized decision support system: cluster randomized trial. J Antimicrob Chemother. 2006;58:1238–45 (I).PubMedCrossRef Paul M, Andreassen S, Tacconelli E, et al. Improving empirical antibiotic treatment using TREAT, a computerized decision support system: cluster randomized trial. J Antimicrob Chemother. 2006;58:1238–45 (I).PubMedCrossRef
434.
Zurück zum Zitat Thern J, de With K, Strauss R, Steib-Bauert M, Weber N, Kern WV. Selection of hospital antimicrobial prescribing quality indicators: a consensus among German antibiotic stewardship (ABS) networkers. Infection. 2014;42:351–62 (III). Thern J, de With K, Strauss R, Steib-Bauert M, Weber N, Kern WV. Selection of hospital antimicrobial prescribing quality indicators: a consensus among German antibiotic stewardship (ABS) networkers. Infection. 2014;42:351–62 (III).
Metadaten
Titel
Strategies to enhance rational use of antibiotics in hospital: a guideline by the German Society for Infectious Diseases
verfasst von
K. de With
F. Allerberger
S. Amann
P. Apfalter
H.-R. Brodt
T. Eckmanns
M. Fellhauer
H. K. Geiss
O. Janata
R. Krause
S. Lemmen
E. Meyer
H. Mittermayer
U. Porsche
E. Presterl
S. Reuter
B. Sinha
R. Strauß
A. Wechsler-Fördös
C. Wenisch
W. V. Kern
Publikationsdatum
11.04.2016
Verlag
Springer Berlin Heidelberg
Erschienen in
Infection / Ausgabe 3/2016
Print ISSN: 0300-8126
Elektronische ISSN: 1439-0973
DOI
https://doi.org/10.1007/s15010-016-0885-z

Weitere Artikel der Ausgabe 3/2016

Infection 3/2016 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Notfall-TEP der Hüfte ist auch bei 90-Jährigen machbar

26.04.2024 Hüft-TEP Nachrichten

Ob bei einer Notfalloperation nach Schenkelhalsfraktur eine Hemiarthroplastik oder eine totale Endoprothese (TEP) eingebaut wird, sollte nicht allein vom Alter der Patientinnen und Patienten abhängen. Auch über 90-Jährige können von der TEP profitieren.

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Bei schweren Reaktionen auf Insektenstiche empfiehlt sich eine spezifische Immuntherapie

Insektenstiche sind bei Erwachsenen die häufigsten Auslöser einer Anaphylaxie. Einen wirksamen Schutz vor schweren anaphylaktischen Reaktionen bietet die allergenspezifische Immuntherapie. Jedoch kommt sie noch viel zu selten zum Einsatz.

Therapiestart mit Blutdrucksenkern erhöht Frakturrisiko

25.04.2024 Hypertonie Nachrichten

Beginnen ältere Männer im Pflegeheim eine Antihypertensiva-Therapie, dann ist die Frakturrate in den folgenden 30 Tagen mehr als verdoppelt. Besonders häufig stürzen Demenzkranke und Männer, die erstmals Blutdrucksenker nehmen. Dafür spricht eine Analyse unter US-Veteranen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.