gms | German Medical Science

GMS Infectious Diseases

Paul-Ehrlich-Gesellschaft für Infektionstherapie e.V. (PEG) (PEG)

ISSN 2195-8831

Recommendations for empiric parenteral initial antibiotic therapy of bacterial diseases in adults: Update 2010

Guideline

Suche in Medline nach

  • corresponding author Klaus-Friedrich Bodmann - Werner Forßmann Krankenhaus, Medizinische Klinik III – Abt. Intern. Intensivmedizin und Notaufnahme, Eberswalde, Germany
  • author Béatrice Grabein - Klinische Mikrobiologie und Krankenhaushygiene, Klinikum der Universität München, München, Germany
  • Expert Commission of the Paul-Ehrlich Society for Chemotherapy e.V.

GMS Infect Dis 2014;2:Doc05

doi: 10.3205/id000013, urn:nbn:de:0183-id0000136

Veröffentlicht: 25. August 2014

© 2014 Bodmann et al.
Dieser Artikel ist ein Open Access-Artikel und steht unter den Creative Commons Lizenzbedingungen (http://creativecommons.org/licenses/by-nc-nd/3.0/deed.de). Er darf vervielfältigt, verbreitet und öffentlich zugänglich gemacht werden, vorausgesetzt dass Autor und Quelle genannt werden.


Abstract

Under the auspices of the “Paul-Ehrlich Gesellschaft”, an expert panel of German infectious disease specialists and microbiologists compiled updated evidence-based treatment recommendations for parenteral antibiotic therapy in adult patients.

Subject-specific expert teams reviewed the available evidence from published data. Evidence levels and grades of recommendations were assigned using a standardized protocol.

The following indication areas were covered: respiratory, ENT, oral/maxillary, intra-abdominal, urinary tract, skin/soft tissue, bone/joint and eye infections, sepsis, endocarditis, meningitis, infections in the elderly and perioperative prophylaxis.

In addition, the recommendations cover relevant issues regarding the use of parenteral antibiotics: characteristics of drug classes, susceptibility testing, spread of resistance, pharmacokinetics, pharmacodynamics, drug monitoring, interactions, safety and pharmacoeconomics.


1 Introduction and antibiotics

Klaus-Friedrich Bodmann, Dieter Adam, Hans-Reinhard Brodt, Eva Susanne Dietrich, Béatrice Grabein, Gert Höffken, Michael Kresken, Pramod M. Shah, Egid Strehl, Christoph Wenisch, Thomas A. Wichelhaus

This document is an update of the recommendations published in 2004 [544]. The introduction of new agents and the results of recent studies necessitated a full revision. As in earlier updates, the current pathogen resistance situation and the results of new clinical studies and the information on the individual agents are summarized in tables.

The working groups updated the individual chapters, approved the contents within the group, and presented the results for plenum discussions at two consensus conferences. Proposals made at the two conferences were discussed and implemented as appropriate. Consensus was defined as agreement of 80% of the eligible conference participants.

The consensus panel ranked the items according to level of evidence and strength of recommendation (Table 1 [Tab. 1] and Table 2 [Tab. 2]).

Generally, a high level of evidence resulted in a high-grade recommendation. However, in some cases results from a therapeutic study with a high evidence level resulted in a low-grade recommendation and vice versa.

This process resulted in these recommendations for the empiric initial parenteral treatment for bacterial infections in adults. In cases in which several treatment options are named their microbiological activity spectrum may not be equivalent. Treatment alternatives allow the epidemiology of the pathogen to be taken into account, to avoid intolerance to antibiotics, and to escalate or de-escalate a treatment according to the situation. The treating physician can therefore better adapt his treatment decisions to the risk profiles of individual patients.

Evaluation of the licensed indications for individual antibiotics

As a result of less tight licensing requirements, many older antibiotics are approved for a much broader spectrum of diseases than substances licensed in the last 10 years by the German Federal Institute for Pharmaceuticals and Medicinal Products or the European Medicines Agency. Due to more stringent regulations in the latter period and the particular problems in Germany regarding studies in difficult disease areas (e.g. ventilator-associated pneumonia), drugs are being used to treat infections for which they are not explicitly licensed (off-label use).

In this context, we may point out the specific issues in Germany regarding clinical studies in legally incompetent patients that have been leading to discontinuation of trials in intensive care patients.

With respect to the legal aspects of off-label prescription of pharmaceuticals, the German Federal Social Court ruled on March 19, 2002 that the statutory health insurances are obliged to pay for pharmaceuticals prescribed for diseases for which they are not licensed if

  • the disease is severe,
  • there is no alternative treatment,
  • and the available data substantiate the expectation of successful therapy.

The issues and open questions regarding off-label use routine practice are covered in a short statement in the Federal Health Newsletter.

Each physician should make his treatment decision together with each individual patient. External evidence of Grades I to III is always based on studies and therefore on standardized patient groups. The physician will base his treatment decision on the best available evidence. However he should check whether the data on which he is basing his decision really apply to the patient for whom he is making the treatment decision (i.e. correlates with the internal evidence).

Due to resistance issues in intensive care units and in oncology, is it imperative to use different antibiotic groups to minimize selection pressure, therefore off-label use of microbiological active drugs is justified (e.g. for nosocomial pneumonia).

Characteristics of antibiotics

Penicillins

The penicillin group of antibiotics is subdivided according to the chemical structure of the agents into benzylpenicillins, aminopenicillins, acylaminopenicillins and isoxazolyl penicillins. Accordingly, penicillins show widely divergent behaviour towards pathogens and beta-lactamases. Penicillins are considered bactericidal with time-dependent killing kinetics. Post-antibiotic effects are limited to a short period of time. Refer to chapter 3 for information on the optimum mode of application.

Penicillins show large variations in terms of their pharmacokinetic characteristics. The distribution is predominantly extracellular, with a distribution volume of 0.2 to 0.4 mL/kg of body weight. Penetration of penicillins in cerebrospinal fluid (CSF) is satisfactory in case of inflamed meninges and adequate dosage. The plasma half-life in patients with normal renal function is in the range of 1 to 2 hours. The majority of the administered dose is eliminated mostly unchanged via the kidneys. The fraction bound to plasma proteins is highly variable reaching values >90% for isoxazolyl penicillins.

The antibacterial spectrum of penicillins varies from narrow to extensive according to subgroup and is the key determinant for decisions on clinical use.

Benzylpenicillin (Penicillin G)

The antimicrobial spectrum of penicillin G covers most strains of streptococci, pneumococci, meningococci, spirochetes, and some anaerobic pathogens such as clostridia and Actinomyces species. Penicillin G is rarely effective against staphylococci due to beta-lactamases or altered binding proteins. The licensing of penicillin G allows its use for almost any systemic and local infection independent of the localization if the infection is caused by a penicillin-susceptible pathogen.

As the antimicrobial spectrum is very narrow, severe infections should not be treated with single-drug regimens before identification of the pathogen. For erysipelas and single-species infections with streptococci and pneumococci, penicillin G is still the drug of choice due to its effective tissue penetration, highly favorable tolerability, and low resistance rates in Germany (refer to chapter 2 for data on the resistance situation in Germany). Much higher rates of resistance should be expected in patients from other countries (e.g. Spain, France and Hungary).

Benzylpenicillin is also available for intramuscular depot injection as a slowly dissolving salt (benzylpenicillin benzathine). The plasma concentrations achieved with these formulations are low with delayed appearance in the blood stream. Indications for depot penicillins are prevention of recurrence in rheumatic fever and erysipelas as well as the treatment of primary syphilis.

Isoxazolyl penicillins: flucloxacillin, oxacillin

This subgroup has a narrow antimicrobial spectrum covering gram-positive organisms, with good activity against staphylococci, including penicillinase-producing strains. These penicillin derivatives are ineffective against methicillin-resistant staphylococci. They are less active than benzylpenicillins against gram-positive pathogens other than staphylococci. Therefore they should be used exclusively in the targeted therapy of infections caused by methicillin-susceptible staphylococci.

Compared to other penicillins, isoxazolyl penicillins are bound to plasma proteins to an extent of >90% and less able to penetrate infected tissues effectively.

Aminopenicillins: ampicillin, amoxicillin/clavulanic acid, ampicillin/sulbactam

The antibacterial spectrum of the aminopenicillins covers gram-positive as well as some gram-negative pathogens. They show good efficacy against streptococci, including pneumococci. Aminopenicillins are more active than penicillin G against Enterococcus faecalis and Listeria spp.

However, aminopenicillins have very limited activity against staphylococci and gram-negative pathogens, particularly enterobacteriaceae, Moraxella catarrhalis and Bacteroides fragilis due to increasing resistance mediated by beta-lactamases. Up to 80% of these strains are not susceptible. However, combination of aminopenicillins with beta-lactamase inhibitors (BLI) extends the antibacterial spectrum to a range of beta-lactamase-producing gram-positive and gram-negative pathogens as well as anaerobes and enables an empiric therapy.

Ampicillin is licensed for the treatment of acute and chronic bacterial infections caused by pathogens proven to be susceptible, independent of localization and severity of illness. This includes endocarditis, meningitis, and sepsis. The drug is also licensed for upper and lower respiratory tract infections, urogenital tract infections, intraabdominal infections, skin and soft tissue infections, as well as perioperative antibacterial prophylaxis.

Fixed combinations of amoxicillin/clavulanic acid and ampicillin/sulbactam are commercially available. Sulbactam is also available as a monosubstance for free combination with other beta-lactams.

The most common adverse effects of aminopenicillins are pseudoallergic skin reactions, a measle-like skin eruption that usually appears 5 to 10 days after the initiation of treatment. The exanthema most often affects patients with simultaneous viral infections (e.g. EBV mononucleosis).

Ureidopenicillins: mezlocillin, piperacillin, piperacillin/tazobactam, combinations with sulbactam

The antibacterial spectrum of ureidopenicillins covers gram-positive and gram-negative pathogens, including Pseudomonas aeruginosa for piperacillin. Because of the increasing rate of beta-lactamase-producing staphylococci, enterobacteriaceae and important anaerobes, the efficacy of ureidopenicillins used on their own is often limited.

However, the antibacterial spectrum is extended to beta-lactamase-producing pathogens by combination with beta-lactamase inhibitors (BLI). Ureidopenicillin-BLI combinations are considered appropriate for empiric initial antibiotic therapy of severe nosocomial infections.

Fixed combinations of piperacillin with tazobactam or free combinations of mezlocillin or piperacillin with sulbactam are available. Tazobactam is the most effective BLI in vitro. Well-documented studies, practical advantages and pharmacokinetic aspects favor the use of the fixed piperacilin/tazobactam combination for evidence-based antibiotic therapy. Moreover, in patients with renal failure, piperacillin and tazobactam are absorbed, distributed and eliminated with very similar kinetics while piperacillin and sulbactam show more divergent pharmacokinetics.

Ureidopenicillins are licensed for a broad spectrum of indications including systemic and local infections by susceptible pathogens (gram-positive, gram-negative, aerobic, anaerobic and mixed infections), ENT (ear, nose and throat) infections (piperacillin only), severe systemic infections including sepsis, bacterial endocarditis, meningitis, respiratory tract infections, intra-abdominal infections, renal and urinary tract infections, gynaecological infections, skin and soft tissue infections (including burns), bone and joint infections (including osteomyelitis) and perioperative antibacterial prophylaxis.

Cephalosporins

According to the recommendations of the Paul Ehrlich Society (PEG), cephalosporins are categorized into 5 groups in Germany. The previous group 5 contained only cefoxitin which is no longer available in Germany. The free position was therefore filled by ceftobiprol, a new cephalosporin active against MRSA (see group 5 below).

The pharmacodynamic properties of cephalosporins are similar to those of penicillins. In terms of pharmacokinetics, the individual agents exhibit considerable variations. Most cephalosporins are renally eliminated as unchanged substance. The average plasma half-life in patients with normal renal function is approximately 2 hours. Ceftriaxon has an average half-life of about 8 hours and is eliminated mostly via biliary excretion. Like the structurally related penicillins, cephalosporines are distributed extracellularly with a relative distribution volume of 0.2 to 0.4 l/kg of body weight.

Generally, cephalosporins are very well tolerated. Allergic reactions are less frequently observed than with penicillins. Cross-allergies to penicillins are limited (<10%). Refer to chapter 2 for resistance data.

Group 1 cephalosporins: cefazoline

Cefazoline is predominantly effective against staphylococci and streptococci. Like all cephalosporins except for ceftobiprole (see group 5 cephalosporins), cefazolin is inactive against methicillin-resistant staphylococci. The percentage of susceptible enterobacteriaceae (Escherichia coli, Klebsiella spp., etc.) has declined in recent years. Cefazoline is primarily appropriate for the treatment of infections caused by methicillin-susceptible staphylococci, and for perioperative prophylaxis.

Group 2 cephalosporins: cefuroxim, cefotiam

Compared to cefazoline, these cephalosporins have an extended spectrum in the gram-negative range which also includes Haemophilus influenzae. In addition, they are effective against methicillin-susceptible staphylococci (cefotiam > cefuroxime). High resistance rates must be expected with AmpC-producing enterobacteriaceae, such as Enterobacter spp. and Citrobacter spp. as well as Morganella morganii and Proteus vulgaris. These antibiotics are licensed for use against a wide range infections caused by susceptible pathogens, e.g. skin and soft tissue infections, bone and joint infections, respiratory tract infections, as well as kidney and urinary tract infections.

Group 3 cephalosporins

3a: cefotaxime, ceftriaxone

3b: ceftazidime

Group 3 cephalosporins show a wide spectrum of activity with a pronounced antibacterial effect against gram-negative bacteria. However, their spectrum of antibacterial activity is being restricted by the spread of enterobacteriaceae producing extended-spectrum beta-lactamases (ESBL) which render group 3 cephalosporins ineffective. Compared with group 1 and 2 cephalosporins, the efficacy of cefotaxim and ceftriaxon is weaker against staphylococci, while ceftazidim is inadequate for this genus of pathogens.

Group 3 cephalosporins are inappropriate for use in suspected or proven staphylococcal infections. In contrast to cefotaxime and ceftriaxone, ceftazidime is clinically ineffective against streptococci and pneumococci. Cefotaxime and ceftriaxone (group 3a) are ineffective against Pseudomonas while ceftazidim (Group 3b) shows good activity against this particular pathogen. The licensed indications include infections of all organ systems caused by susceptible pathogens.

Group 4 cephalosporins: cefepime, cefpirome (Austria)

The activity of group 4 cephalosporins against staphylococci is comparable to group 3a; their efficacy against Pseudomonas is comparable to ceftazidim. Cefepime and cefpirome are effective against pathogens overexpressing AmpC beta-lactamases (predominantly Enterobacter spp., Citrobacter spp.), which differentiates them from group 3 cephalosporins. However, ESBL-producing pathogens are still resistant.

Group 5 cephalosporins: ceftobiprole

The activity of ceftobiprole against gram-negative pathogens is comparable to group 4 cephalosporins. However, it is also active against methicillin-resistant staphylococci and Enterococcus faecalis. The licensed indications are currently limited to severe skin and soft tissue infections. Adherence to the recommended infusion duration of 2 hours is warranted.

Addendum: ceftobiprole had been introduced in Canada and Switzerland. Currently (November 2013), the drug is no longer available worldwide.

Carbapenems

Carbapenems are well-tolerated beta-lactam antibiotics divided into 2 groups based on their spectrum of activity. They have a very broad overall efficacy spectrum in the gram-positive and gram-negative domains, including anaerobic and ESBL-producing pathogens. In the recent years nosocomial infections due to carbapenemase-producing strains have been increasingly reported. Carbapenems have no or very limited activity against these isolates.

Stenotrophomonas maltophilia is primarily resistant to carbapenems. Carbapenems are also inactive against methicillin-resistant staphylococci and Enterococcus faecium.

Doripenem, imipenem/cilastatin and meropenem belong to group 1. Ertapenem is categorized in group 2 as it has no efficacy against enterococci, Pseudomonas spp. and Acinetobacter spp.

Further differentiations can be made in terms of pharmacokinetics. Carbapenems are distributed extracellularly with a relative distribution volume of 0.1 L/kg (ertapenem) and 0.2 L/kg of body weight (doripemen, imipenem, and meropenem), respectively. The protein-bound fraction is >90% for ertapenem, 25% for imipemen/cilastatin, 8% for doripemen and 2% for meropenem. All carbapemens are partially metabolized and eliminated mostly via the kidneys. The half-life of group 1 carbapenems in patients with normal renal function is approximately 1 hour. Ertapenem has a longer half-life of approximately 4 hours and may be given once daily. The dosages of doripenem, imipenem/cilastatin and meropenem are equivalent. For doripenem a longer infusion time is recommended and licensed, especially for less susceptible pathogens and severe infections.

As for penicillins, a dose-dependent epileptogenic adverse drug reaction (ADR) is known for all carbapenems (imipenem > ertapenem > meropenem > doripenem). As this ADR is reported most frequently with imipenem, this drug is not appropriate for the treatment of CNS infections. Meropenem is the only carbapenem licensed for the treatment of meningitis.

Monobactams: aztreonam

The pharmacokinetics and pharmacodynamics of aztreonam are similar to those of the penicillins. It is active exclusively against gram-negative pathogens including P. aeruginosa. Acinetobacter spp., S. maltophilia and ESBL-producing enterobacteriaceae are resistant. However, strains which produce metallo-beta-lactamases (MBL) are susceptible to aztreonam.

Because of the structural differences, cross-allergies with beta-lactam antibiotics are unlikely. The clinical relevance of aztreonam is limited. It can be used in combination with antibiotics effective against gram-positive bacteria.

Fluoroquinolones

As recommended by the PEG, fluoroquinolones are categorized into 4 groups. Since only groups 2 through 4 include parenterally available drugs, only these three groups will be considered here.

Fluoroquinolones show concentration-dependent bactericidal activity with a generally broad spectrum of activity. The differences between the groups are discussed in the following section.

The growing resistance rates in Escherichia coli and other enterobacteriaceae clearly limit the use of fluoroquinolones for empiric initial monotherapy, particularly in nosocomial infections. Cross-resistance among all fluoroquinolones is the rule. The fluoroquinolones penetrate well into many tissues and show intracellular and extracellular distribution, with relatively high distribution volumes of 2–4 L/kg of body weight. Usually less than 40% of the drug is bound to plasma proteins.

Levofloxacin is eliminated almost exclusively via the kidneys while ciprofloxacin shows some biliary and transintestinal elimination as well. Moxifloxacin is metabolized extensively via conjugation. The half-life is 3–4 hours for ciprofloxacin, 7–8 hours for levofloxacin, and more than 10 hours for moxifloxacin, which explains the different dosing intervals.

Adverse drug reactions are reported in 4–10% of treated patients, most frequently as gastrointestinal disturbances, CNS disorders (sleeplessness and dizziness), or skin reactions.

Group 2 fluoroquinolones: ciprofloxacin, (ofloxacin)

Ciprofloxacin has excellent activity against gram-negative enterobacteria and H. influenzae, and good activity against P. aeruginosa. It is only weakly active against staphylococci and clinically inadequate against pneumococci and enterococci. Moreover, ciprofloxacin is less active against Chlamydia, Legionella and Mycoplasma spp. than group 3 and 4 fluoroquinolones.

Licensed indications include uncomplicated and complicated renal and urinary tract infections, ENT infections, respiratory tract infections (excluding pneumococcal infections), abdominal infections, genital organ infections, bones and joint infections, skin and soft tissue infections, bacterial sepsis, and infections in neutropenic patients. The use of ofloxacin is no longer recommended (see below).

Group 3 fluoroquinones: levofloxacin

Levofloxacin is the L enantiomer and therefore the active entity of the racemic substance ofloxacin: its antibacterial activity is twice as high. Compared with ciprofloxacin, it shows improved efficacy against gram-positive pathogens such as staphylococci, streptococci and pneumococci as well as against Legionella, Chlamydia and Mycoplasma spp. Against gram-negative pathogens it is similarly active as ciprofloxacin, although it is somewhat less effective against P. aeruginosa.

Levofloxacin is licensed for the treatment of community-acquired pneumonia, complicated urinary tract infections, and skin and soft tissue infections.

Group 4 fluoroquinolones: moxifloxacin

Due to its structural differences versus group 2 and 3 fluoroquinolones, moxifloxacin is considerably more active against gram-positive pathogens, i.e. staphylococci and streptococci, including S. pneumoniae. Its efficacy against Legionella, Chlamydia and Mycoplasma spp. is further improved. Moxifloxacin is the only fluoroquinolone with useful activity against gram-positive and gram-negative anaerobes. However, is not adequately effective against P. aeruginosa.

Moxifloxacin is licensed for the treatment of community-acquired pneumonia and complicated skin and soft tissue infections.

Macrolides and azalides: erythromycin, clarithromycin, azithromycin

Macrolides have good antibacterial activity against Mycoplasma, Legionella and Chlamydia spp. as well as streptococci, including S. pneumoniae and Bordetella pertussis. Macrolide resistance rates of pneumococci have reached values beyond 20% in Germany but are recently declining. Data on resistance rates are given in chapter 2.

Erythromycin has inadequate activity against H. influenzae. The microbiological activity of clarithromycin, its active metabolites and azithromycin is somewhat higher but the clinical efficacy of these drugs against infections with this pathogen is still considered insufficient.

Macrolides are basically bacteriostatic agents but develop bactericidal effects at higher concentrations. Their pharmacodynamics are time-dependent. In addition to the antibacterial activity, an immunomodulatory effect of macrolides is being discussed.

The macrolides show dose-proportional pharmacokinetics. The half-life is below 2.5 hours for erythromycin, between 2 and 5 hours for clarithromycin, and above 14 hours for azithromycin. The volumes of distribution also show significant differences: approximately 0.7 L/kg for erythromycin, 4 L/kg for clarithromycin and 25 L/kg of body weight for azithromycin.

The macrolides are extensively metabolized in the liver. They are eliminated primarily via biliary excretion.

The most frequent side effects of macrolides are gastrointestinal disorders and elevated liver enzyme levels. Extensive drug interactions are a problem with erythromycin and clarithromycin.

Licensed indications are respiratory tract infections (including those with Chlamydophila pneumoniae or Legionella), whooping cough, diphtheria, scarlet fever and erysipelas.

Glycopeptides: vancomycin, teicoplanin

Vancomycin and teicoplanin are exclusively active against gram-positive pathogens. Their spectrum of activity covers staphylococci including methicillin-resistant strains, streptococci, enterococci inclucing E. faecium, Corynebacterium spp., and Clostridium difficile. Resistance of Staphylococcus aureus against glycopeptides has been described only in individual cases to date. Teicoplanin resistance has been observed in coagulase-negative staphylococci. Elevated minimal inhibitory concentrations (MICs) of vancomycin in MRSA are considered to be asscociated with treatment failure and higher mortality rates (see chapter 2).

The glycopeptides should be used only if other agents cannot be used due to resistance or allergies, as they are clinically less effective against non-resistant pathogens and have tolerability issues (see below).

The antibacterial activity of glycopeptides is time-dependent, with a slow onset of the therapeutic effect. The volume of distribution is 0.4–0.9 L/kg for vancomycin and 1 L/kg for teicoplanin. Glycopeptide pharmacokinetics show extensive intraindividual and interindividual variability. The plasma half-life is usually 4–6 hours for vancomycin and 70–100 hours for teicoplanin.

In plasma, 55% of vancomycin is bound to proteins, compared to 90% of teicoplanin. Glycopeptides are eliminated predominantly unchanged via the kidneys. Glycopeptides are potentially nephrotoxic and ototoxic, depending on the drug used. Plasma level monitoring of vancomycin is therefore essential. Alternative antibiotics should be used in patients with renal dysfunction.

Intravenous infusion of vancomycin requires appropriate dilution and infusion times to avoid a “red man syndrome”. The licensed indications cover bacterial sepsis, endocarditis, and infections of bones and joints, respiratory tract, skin and soft tissue, kidneys and urinary tract.

Aminoglycosides: amikacin, gentamicin, tobramycin

Aminoglycosides are effective against gram-negative bacteria, primarily enterobacteriaceae. Against P. aeruginosa, tobramycin and amikacin are more active than gentamicin. Aminoglycosides are less effective against gram-positive pathogens. They are used in combination with beta-lactam antibiotics to enhance efficacy in Enterococcus infections.

Aminoglycosides exert a broad, rapid concentration-dependent bactericidal effect. Target serum and/or tissue concentrations should exceed 5 times the minimum inhibitory concentration (MIC) of the pathogen. The postantibiotic effect of aminoglycosides may last for several hours, depending on the serum concentration, the agents used in combination and the patient’s immune function. The efficacy of aminoglycosides is pH-dependent: they are inactive in acidic and anaerobic environments.

Aminoglycosides are distributed extracellularly and eliminated as unchanged drugs via the kidneys. The mean relative volume of distribution is approximately 0.25 L/kg of body weight (range 0.1 to 0.8 L/kg). The plasma half life is in the range of 2 hours in patients with normal renal function and considerably longer in patients with renal impairment. Therefore, the creatinin clearance must be considered when treating high-risk patients and therapeutic drug monitoring is mandatory. Particularly in combination treatment with beta-lactam antibiotics, the entire daily dose should be administered once daily rather than three times daily to reach the highest possible peak concentration. There is evidence for a lower toxicity rate and better clinical results with the once daily dosage. Within a 24-hour dosing interval, the target minimum concentration of <1 mg/L and (extrapolated) peak concentration between 15 and 20 mg/L for gentamicin and tobramycin and about 60 mg/L for amikacin should be achieved in patients with normal kidney function. There is insufficient data on the once-daily dosage in the treatment of endocarditis and these cases should be treated conventionally.

Aminoglycosides have pronounced ototoxic and nephrotoxic potential and should be used only if strictly required. If used appropriately (once daily dosing, short duration of treatment with plasma level monitoring), these antibiotics have an acceptable tolerance level (see chapter 4). The licensed indications include severe (nosocomial) infections caused by gram-negative bacilli, febrile neutropenia, and Pseudomonas infections in cystic fibrosis. Aminoglycosides should never be used as single agents. Rather, they are usually combined with beta-lactam antibiotics. They may be used in combination with aminopenicillins for enterococcal endocarditis and Listeria infections. Duration of therapy with aminoglycosides is usually limited to 3–5 days.

Oxazolidinones: linezolid

Linezolid is active only against gram-positive pathogens. It has a good efficacy against gram-positive cocci, primarily staphylococci including methicillin-resistant strains and enterococci including vancomycin-resistant strains (VRE). It is bactericidal against streptococci and bacteriostatic against staphylococci and enterococci.

The relative volume of distribution is about 0.6 L/kg of body weight. Approximately 30% of the drug is bound to plasma proteins. The half-life in plasma is 5 to 7 hours with primarily renal elimination.

Linezolid is licensed for community-acquired and nosocomial pneumonia as well as complicated skin and soft tissue infections.

Due to the potential side effect of thrombocytopenia, regular monitoring of the blood count during therapy is necessary. Treatment duration should not exceed 28 days.

Lincosamides: clindamycin

Clindamycin has a predominantly bacteriostatic, time-dependent effect on staphylococci, streptococci, Bacteroides spp., Corynebacterium spp. and Mycoplasma pneumoniae. Due to its mechanism of action, clindamycin inhibits the production of toxins by staphylococci and streptococci and is therefore an important combination partner in the treatment of infections with toxins as a major pathogenic factor.

The relative distribution volume is in the range of 0.6 l/kg of body weight and the half-life is 2 to 3 hours. More than 80% of clindamycin is converted to active metabolites. Licensed indications include infections of bones and joints including septic arthritis, maxillodental infections, ENT infections, respiratory tract infections, pelvic and intra-abdominal infections, skin and soft tissue infections as well as scarlet fever, sepsis and endocarditis caused by clindamycin-susceptible pathogens.

Tetracyclines: doxycycline

The antibacterial spectrum of doxycycline covers gram-positive and gram-negative pathogens as well as Chlamydia and Mycoplasma spp. Doxycycline has primarily bacteriostatic activity and acts both extra- and intracellularly. The relative distribution volume is 0.8 L/kg of body weight with a half-life of 10 to 22 hours. Doxycycline is metabolized to a small extent and eliminated primarily via biliary and renal routes. Doxycycline has a broad spectrum of indications including the treatment of infections caused by susceptible pathogens primarily in the ENT region, respiratory tract, urogenital tract, abdominal region and biliary tract as well as borreliosis. Intravenous doxycycline is currently the drug of choice for rickettsiosis, plague, brucellosis, and query fever among others.

Glycylcyclines: tigecycline

Tigecycline has a broad spectrum of activity that extends to multiresistant gram-positive pathogens such as MRSA and VRE and multiresistant gram-negative bacteria such as ESBL-producing enterobacteriaceae and multiresistant Acinetobacter baumannii. It includes anaerobes and Chlamydia, Mycoplasma and Legionella spp. as well. Tigecycline is inactive against P. aeruginosa, Proteus spp., and M. morganii. The mode of action is primarily bacteriostatic. However, bactericidal activity has been observed e.g. for S. pneumoniae and H. influenzae [574], [584]. The volume of distribution is the range of 7 to 9 L/kg of body weight. The average terminal half-life is 42 hours. 59% of the substance is eliminated via bile and faeces and 33% via urine. Tigecycline is licensed for the treatment of complicated skin and soft tissue infections as well as complicated intra-abdominal infections.

Ansamycins: rifampicin

In vitro, rifampicin is active against mycobacteria, staphylococci including methicillin-resistant strains, streptococci and E. faecalis, among others. It acts strongly bactericidal or bacteriostatic against proliferating cells, depending on the drug concentration and the activity of the pathogen.

Rifampicin should not be used as monotherapy due to a high risk of rapid resistance development. In plasma 70–90% of rifampicin is bound to protein. The agent has a high tissue penetration and accumulates intracellularly. Rifampicin has a relative distribution volume of >1 L/kg of body weight. The half-life depends on the duration of treatment. In long-term therapy, the half-life is as low as 2–3 hours due to autoinduction of metabolism. Rifampicin is eliminated via bile and urine.

The most frequent side effects are liver dysfunction and gastrointestinal disturbances. Changes of blood cell count may be observed. As a strong inducer of cytochrome P450 enzymes rifampicin has a high potential for pharmacokinetic interactions with other drugs.

Nitroimidazoles: metronidazole

Metronidazole is active against anaerobic gram-positive and gram-negative bacteria, except for propionibacteria and actinomycetes. Metronidazole exerts a concentration-dependent bactericidal effect. The relative distribution volume is in the range of 0.5 L/kg of body weight with a half-life of 6 to 8 hours. In plasma 10–20% of metronidazole is bound to protein. The agent is metabolized in the liver and eliminated primarily via urine.

Metronidazole is licensed for the treatment of proven or suspected infections caused by anaerobes in various localizations (including brain abscesses) and for perioperative prophylaxis. Metronidazole is usually used in combination with other antibiotics for mixed aerobic and anaerobic infections and in monotherapy for Clostridium difficile-associated disease.

Adverse drug reactions include peripheral and central neuropathies.

Fosfomycin

Fosfomycin has a broad spectrum of activity that covers gram-positive and gram-negative pathogens, with bactericidal activity against MRSA, ESBL-producing enterobacteriaceae and P. aeruginosa.

Fosfomycin is not bound to plasma proteins and is eliminated as unchanged drug via the kidneys. Its plasma half-life is 2 hours. Fosfomycin penetrates highly effectively into various tissues.

It is licensed for a wide spectrum of infections, including severe diseases such as sepsis, meningitis, brain abscess, endocarditis, bone and joint infections, respiratory tract infections, skin and soft tissue infections, kidney and urinary tract infections and ENT infections. Fosfomycin is not recommended for monotherapy of severe infections but can be used in combinations with many other antibiotics.

The most frequent side effects are associated with the high sodium content and increased potassium excretion.

Cotrimoxazole

Cotrimoxazole is a combination drug containing sulfamethoxazole and trimethoprim. It shows broad-spectrum activity against gram-positive and gram-negative pathogens as well as protozoa and Pneumocystis jirovecii.

Both agents are distributed intracellularly and extracellularly. The substances are metabolized in the liver. The average half-life of the active drugs is 6.4 hours for sulfamethoxazole and 7.8 hours for trimethoprim. Excretion occurs primarily via urine and to a lower extent via bile.

Like many older antibiotics, cotrimoxazole is licensed for a broad spectrum of indications. Rational indications include Pneumocystis pneumonia, S. maltophilia infections and nocardiosis. Reversible bone marrow suppression and allergic reactions (sometimes including Stevens-Johnson or Lyell syndrome) may occur, particularly in long-term usage.

Cyclic lipopeptides: daptomycin

Daptomycin is active exclusively against gram-positive bacteria including multi-resistant pathogens such as MRSA and VRE. It is bactericidal in both growth and stationary phases of the pathogen life cycle. The half-life is in the range of 8–9 hours, 92% of the drug is bound to proteins. Daptomycin has a low distribution volume of 0.1 l/kg of body weight. The agent is eliminated primarily via urine, 5% of the parent drug is excreted with faeces. Daptomycin is licensed for the treatment of bacteremia, endocarditis, and skin and soft tissue infections [298], [308], [470].

Polymyxins: colistin

Colistin shows bactericidal activity against gram-negative pathogens, including multi-resistant strains of P. aeruginosa and A. baumannii as well as ESBL- or carbapenemase-producing enterobacteriaceae. Proteus spp., M. morganii, Serratia marcescens, Burkholderia-cepacia complex, Neisseria spp. and M. catarrhalis are resistant against colistin. Recent data on the pharmacokinetics and pharmacodynamics are limitied. Nephrotoxicity and neurotoxicity, side effects that were frequently reported in the past, have more rarely been observed in newer case series and studies. Intravenous colistin is recommended exclusively for infections caused by multiresistant gram-negative pathogens [367].




2 Microbiology

Michael Kresken, Karsten Becker, Uwe Frank, Arne C. Rodloff, Matthias Trautmann, Cornelia Lass-Flörl, Thomas A. Wichelhaus, Pramod M. Shah, Martin Kaase

The effective and economical use of antibiotics is based on rational and reliable microbiological diagnostics. An essential prerequisite for the adequate treatment choice for empiric therapy is a sound knowledge of the usual spectrum of pathogens and the current local, regional, national and international resistance situation. This knowledge is also required for hospital hygiene and infection prevention management. A close cooperation of treating physician, microbiologist and hygiene specialists is required. Cooperation starts with the optimal choice, correct collection and adequate transportation of the specimen material, as mistakes made at this stage cannot be amended later. Pure cultures of the pathogen are a mandatory prerequisite for susceptibility testing and require the best possible specimen material (“tissue samples are superior to smears”). The collaboration continues with the joint evaluation of the detected microorganisms and their susceptibility, the clinical diagnosis, an agreement on rational antibiotic therapy and hospital hygiene measures if appropriate.

The close coordination between the clinical wards and the microbiology/hospital hygiene departments should enable joint compilation and implementation of local guidelines on antibiotic use, surveillance of resistance and nosocomial infections and anti-epidemic hygiene measures. The clinical microbiologist and/or the infection control specialist should be locally available for regular participation in patient visits and ad-hoc case discussions. This allows well-targeted diagnoses and ensures rational antibiotic use.

Susceptibility testing

The susceptibility of a pathogen to an antibiotic is established through the determination of in vitro activity. The reference method is the determination of the minimum inhibitory concentration (MIC in mg/L) according to ISO 20776-1 [130]. The minimum requirement is the use of methods complying to ISO 20776-2 in the laboratory routine. Agar diffusion tests are used as well.

The numerical value of the MIC and the inhibition zone diameter (in mm) provide information on the susceptibility of a pathogen in vitro. Clinical interpretation of the results is achieved with the help of threshold concentrations (cut-off values) in the categories susceptible, intermediate (if defined) or resistant. European harmonized cut-off values as developed by the European Committee of Antimicrobial Susceptibility Testing (EUCAST) (http://www.eucast.org/%20clinical_breakpoints/) are available for most antibiotics. In Germany, the Medical Standards Committee of the German Institute for Standards (DIN) is responsible for the provision of the cut-off values. To date the recommendations of the US Clinical Laboratory Standards Institute (CLSI) are used in parallel to European standards. The cut-off values of EUCAST and DIN are almost always identical. However, they often diverge from the CLSI values, potentially resulting in pronounced differences of resistance rates. Therefore resistant rates derived from different standards cannot be directly compared.

When interpreting a microbiological result, a species-specific consideration of the antibiogram is essential. In cases of doubt and in cases with critical therapeutic relevance of resistance results, additional methods including nucleic acid detection (PCR) or antigen detection can be used to confirm the evaluation of special susceptibility in selected pathogens.

In addition, even an optimal microbiological diagnosis cannot exclude discrepancies between antibiogram and treatment outcome. The most frequent causes are mistakes in the pre-analytical phase resulting in the examination of a bacterial isolate which is not the actual pathogen. Quality problems may also result from prolonged transportation times of the sample material, which can easily lead to the inactivation of sensitive pathogens, overgrowth of irrelevant pathogens and drying out of sample material.

Potential reasons for clinical failure with a sensitive pathogen or clinical success with a resistant pathogen are summarized in Table 3 [Tab. 3]. In conclusion, any susceptibility testing has its methodological limitations. While correlation to the clinical situation is imperfect it still can help to predict clinical efficacy!

Resistance situation

The resistance situation of important bacterial species in Germany and in Central Europe is investigated at regular intervals by the PEG Working Group for Susceptibility Testing and Resistance in selected laboratories in Germany, Austria and Switzerland using uniform and standardized methods (PEG Resistenzstudie, http://www.p-e-g.de/resistenz). Moreover, data are available from the PEG Working Group on Blood Culture Studies [41], the German Network for Antimicrobial Resistance Surveillance (GENARS, http://www.genars.de/), which is now being continued within the framework of the ARS (see below), and the SARI Project (sari.ipse-freiburg.de). The European Antimicrobial Resistance Surveillance Network (EARS-net) supplies country-specific resistance data for isolates from patients with systemic infections (EARS-net, http://www.rivm.nl/earss). Further data sources regarding the monitoring of important pathogens include the national and international resistance surveillance studies carried out by the pharmaceutical industry (G-TEST [268], MYSTIC [528], TEST [382], ZAAPS [244] etc.) as well as the national reference centres (NRZ, http://www.rki.de/DE/Content/Infekt/NRZ/nrz_uebersicht_gesamt_node.html).

Recently, the Robert Koch Institute created the Antibiotics Resistance Surveillance System (ARS, https://ars.rki.de/) operating within the framework of the German Antibiotics Resistance Strategy (DART). It provides data on the resistance situation in both hospital and community-based care. A summary of data on the use of antibiotics and the spread of resistances in human and veterinarian medicine is available in the GERMAP 2008 and 2010 report (http://www.p-e-g.org/econtext/germap) initiated by the Federal Ministry of Consumer Protection and Food Safety, the PEG and the Infection Diseases Department in Freiburg, and which will be regularly updated in the future.

Within the framework of the PEG Resistance Study of 2007, 5,908 bacteria strains from different sample sources (wounds 26%, respiratory tract 20%, urinary tract 18%, blood 12%) were investigated in 26 laboratories. About 56% of the samples were from patients treated in general wards, 24% from intensive care patients and 18% from outpatients. The results indicate that the resistance rates of many pathogens have increased considerably in the last decade. The PEG Blood Culture Study conducted in 2006/2007 included 7,652 isolates from 14 laboratories. The following section discusses important resistance trends in the time between 1998 and 2007 as identified from the PEG Resistance Study and some results from the 2006/2007 Blood Culture Study.

Beta-lactam antibiotics

According to the results of the resistance study for Escherichia coli, ampicillin resistance has increased from 41% in 1998 (n=783) to 55% in 2007 (n=648) while cefuroxim resistance increased from 6% to 15%. The percentage of strains with extended spectrum beta-lactamases (ESBL) which inactivate group 3, 4 and 5 cephalosporins (as identified by the new cephalosporin classification, see chapter 1) has increased from 1% to 10% for E. coli and from 4% in 1998 (n=275) to 10% in 2007 (n=273) for Klebsiella pneumoniae. In the 2006/2007 blood culture study, the percentage of isolates with resistance to cefotaxim was 8% for E. coli (n=1523) and 15% for K. pneumoniae (n=315). Between 1998 (n=859) and 2007 (n=761), the resistance of Pseudomonas aeruginosa to ceftazidim and piperacillin (± beta-lactamase inhibitor) increased from 5–6% to 12–14%.

The percentage of methicillin (i.e. oxacillin) resistant strains of Staphylococcus aureus (MRSA) increased from 12% in 1998 (n=873) to 20% in 2007 (n=782). In 1990 (n=1310), the MRSA rate had been as low as 2%. The frequency of methicillin (i.e. oxacillin) resistant strains of Staphylococcus epidermidis slightly increased from 68% to 74% between 1998 (n=555) and 2007 (n=423). In the 2006/2007 study, 24% and 81% of blood culture isolates of S. aureus (n=1108) and S. epidermidis (n=194), respectively, were resistant to oxacillin.

Penicillin-resistant pneumococci are very rare in Germany. The rate of isolates with reduced susceptibility to penicillin was 10% in the 2007 resistance study (n=406) and 5% in the 2006/2007 blood culture study (n=79). Carbapenem-resistant enterobacteriaceae as well are still rarely encountered in Germany. The percentage of P. aeruginosa strains with reduced susceptibility to meropenem and imipenem was 10% to 14% of isolates from 411 patients in general wards as opposed to 20% to 23% of isolates from 196 patients treated in intensive care units. 26% of P. aeruginosa isolates (n=224) in the 2006/2007 blood culture study showed reduced susceptibility to meropenem.

Fluoroquinolones

The percentage of ciprofloxacin-resistant strains increased from 8% in 1998 to 26% in 2007 for E. coli and from 14% to 18% for P. aeruginosa. In the 2007 resistance study, resistance to levofloxacin was 26% for E. coli and 20% for P. aeruginosa. Reflecting the multiresistance of MRSA isolates, the percentage of S. aureus strains resistant to fluoroquinolones increased from 17% to 28%. In the 2006/2007 blood culture study, 32% of the 1,523 E. coli isolates and 27% of the 224 P. aeruginosa isolates were resistant to ciprofloxacin and 31% of the 1,108 S. aureus isolates were resistant to moxifloxacin.

Macrolides

The rate of macrolide-resistant strains in pneumococci was 14% in the 2007 resistance study (n=406) and 25% in the 2006/2007 blood culture study (n=79). The data on invasive pneumococci from the National Reference Centre for Streptococci shows a decrease of the resistance rates from 16% to 13% for adults and 21% to 14% for children in 2008 (1,907 adults, 280 children) versus 2007 (1,676 adults, 284 children).

Glycopeptides

Glycopeptide resistance rates of staphylococci remained favourably low according to results from both the 2007 resistance study and the 2006/2007 blood culture study. While vancomycin-resistant MRSA strains (VRSA; MIC>8 mg/l) exhibiting vanA resistance mechanisms are extremely rare worldwide, the so-called VISA (vancomycin-intermediate S. aureus) with a MIC of 4–8 mg/L (according to CLSI criteria) have already been observed in many countries. Changes in the cell wall are believed to be responsible for the reduced susceptibility in these strains.

Heterogeneous VISA (hVISA) are potential VISA precursor strains which appear to be vancomycin-susceptible in MIC testing but contain subpopulations of organisms with increased MIC values (≥4 mg/l) [21], [31], [111]. In addition, a slow but steady increase in the average vancomycin MIC for MRSA and MSSA below the suspectibility break points has been reported (referred to as MIC creep or MIC shift in the literature) [447], [496], [564]. These changes are of particular importance as the antibacterial activity of vancomycin against MRSA is already reduced in strains with an MIC apporaching 2 mg/l and vancomycin treatment of bacteremic infections caused by these pathogens has been associated with high failure rates [340], [450]. Consequently, the EUCAST reduced the susceptibility cut-off values for vancomycin and teicoplanin (resistant: >2 mg/l) in 2009.

For Entercoccus faecium, the percentage of vancomycin-resistant strains detected in the resistance study increased from 5% in 1998 (n=110) to 11% in 2007 (n=250), whereas vancomycin-resistant isolates of Enterococcus faecalis were not found in 2007 (n=488). In the 2006/2007 blood culture study, vancomycin-resistance was observed in 4% and <1% of E. faecium and E. faecalis isolates, respectively.

Trimethoprim/sulfamethoxazol

For E. coli, an increase in the resistance rates from 27% in 1998 to 34% in 2007 was detected.

Daptomycin, linezolid, tigecycline

Resistance rates of staphylococci (including MRSA), enterococci (including VRE) and streptococci against daptomycin and linezolid are still very low worldwide. As in any antibiotic treatment, development of resistance during therapy remains a possibility, however [174], [221], [225], [506]. Recent reports described a plasmid-encoded mechanism of resistance against oxazolidinones in staphylococci, which may promote the spread of resistance [311].

Currently, tigecycline-resistant gram-positive pathogens remain rare as well. Practically all isolates of E. coli (including ESBL-producing strains) are susceptible to tigecycline, while 5% to 10% of Enterobacter cloacae and K. pneumoniae isolates are found to be resistant [268]. Development of resistance during treatment is possible in Acinetobacter baumannii and K. pneumoniae [20], [249], [426].

Imipenem-resistant strains of A. baumannii tend to be less susceptible to tigecyclin than imipenem-susceptible strains of this pathogen [268]. Evidence-based summaries of the resistance situation for important bacterial pathogens (Grade A evidence) are given in Table 4 [Tab. 4].

The results of the “PEG Resistenzstudie” (Resistance Study) are mainly provided by laboratories in tertiary care hospitals. Therefore, they cannot necessarily be extrapolated to other areas of medical care. The increase in resistance rates is largely due to the surge in multiresistant pathogens, which may cause pronounced difficulties in antibiotic treatment. In many cases, resistance rates and pathogen patterns in nosocomial infections correlate with the antibiotic usage pattern in the reporting hospital. Any empiric antibiotic therapy must take pathogen epidemiology and local resistance situation into account. Particularly on intensive care units, the regular acquisition of resistance data is a necessary prerequisite for successful treatment. However, the absolute usage figures are probably less important in clinical practice than the failure to comply with general rules of hygiene and infection control measures.

Mechanisms of antibiotic resistance

Resistance mechanisms of bacteria can be categorized in 3 groups:

  • deactivating enzymes
  • modification of target structures
  • modification of access to target structures (increased efflux, decreased influx).

The genetic determinants of resistance may be an intrinsic component of the bacterial chromosome but are more often found on chromosomal and/or extrachrom osomal mobile genetic elements (e.g. resistance plasmids, transposons, insertion sequences, genomic islands), which are responsible for a rapid horizontal spread of resistance among bacteria.

Collateral damage inflicted by antibiotics

Adverse ecological effects such as the selection of antibiotic resistance in the physiologically colonizing flora, the occurrence of Clostridium difficile-associated diarrhoea, and the colonization and infection with multiresistant pathogens, e.g. ESBL-producing enterobacteriaceae, MRSA or vancomycin-resistant enterococci (VRE), are summarized as collateral damage of antibiotic use. The risks of collateral damage associated with various antibiotics can be identified in epidemiological studies.

Patients with gram-negative infections treated with fluoroquinolones are at increased risk of being infected by fluoroquinolone-resistant pathogens [399]. This correlation was shown e.g. in a study of patients with urinary tract infections: those who had been treated with ciprofloxacin in the year before the onset of urinary tract infection were significantly more likely to be infected with ciprofloxacin-resistant E. coli [26]. In another study, there was a significant correlation between the detection of fluoroquinolone-resistant E. coli in community-acquired urinary tract infections and the level of fluoroquinolone use in the population [198]. Moreover, there is evidence that the use of fluoroquinolones increases the risk of acquiring MRSA and ESBL-producing pathogens [399]. This correlation may explain why the majority of MRSA and ESBL-producing pathogens are resistant to fluoroquinolones.

In numerous case-control studies, the use of group 3 cephalosporins has been identified as a risk factor for ESBL-producing pathogens. These drugs were also identified as a risk factor for infections with MRSA and VRE. These drugs probably also increase the risk of pathogens producing carbapenemases as these enzymes inactivate cephalosporins as well [399].

Carbapenems have a central role in the treatment of life-threatening infections. As a result of the increase in ESBL-producing pathogens which are resistant to cephalosporins and usually also to fluoroquinolones, the importance of carbapenems has increased significantly.

Because new antibiotics with new efficacy mechanisms against gram-negative bacteria will not be reliably available in the coming years, an increase in carbapenem resistance would have a dramatic impact on therapy. It has already been shown that the use of imipenem and meropenem is associated with a higher risk of colonization with MRSA, ciprofloxacin-resistant P. aeruginosa and VRE than therapy with cephalosporins, fluoroquinolones or piperacillin/tazobactam [509]. Carbapenems are also a risk factor for infections with S. maltophilia.

Medical measures against increasing resistance

The development of bacterial resistance during treatment is due to genetic variability and the selection of rare resistant variants through the use of antibiotics. Containment of resistance is primarily based on the reduction of selective pressure and the prevention of transmission of (multi)resistant pathogens. Resistance development and the spread of resistant bacteria can be influenced by the following measures:

  • rational, patient-specific, targeted use of antibiotics
  • adequate dosage and duration of treatment
  • combination therapy (using the same dosages as in monotherapy) in cases with higher risk of resistant variants appearing on monotherapy, e.g. empiric treatment of severe infections such as pneumonia or sepsis in which the involvement of P. aeruginosa is suspected
  • parallel use of different classes of antibiotics for the same indication
  • adjustment of therapy after microbiological results become available
  • restrictive prophylactic and topical use of antibiotics
  • strict adherence to hand disinfection rules and further infection control measures
  • continued compilation of statistics on pathogens and resistance (local, regional and supraregional) as a basis for infection control measures in hospitals and guidelines for antibiotic therapy (§ 23 section 1 infection protection act)
  • enhanced involvement of infectious disease specialists in hospitals
  • continued education in antibiotic therapy and increased interdisciplinary cooperation of all professional groups involved in infectious disease therapy
  • vaccination

3 Pharmacokinetics and pharmacodynamics

Bernd Drewelow, Hartmut Derendorf, Fritz Sörgel, Jolanta Majcher-Peszynska, Cordula Lebert

Pharmacology

Besides the antimicrobial properties of a substance (pharmacodynamics), its pharmacokinetics, i.e. the behaviour of a drug inside the human body, has a decisive role in determining its clinical utility. Ultimately it is a question of whether or not the concentration at the site of action is adequate to inhibit the growth of the pathogen. Adverse drug effects and interactions should be minimized.

Correlating pharmacokinetic parameters or simplistically plasma and tissue concentrations with antimicrobial characteristics in vitro or in vivo for the purpose of predicting efficacy, is the realm of PK/PD (pharmacokinetics/pharmacodynamics).

Pharmacokinetics

Pharmacokinetic properties of drugs are determined by their physicochemical characteristics. The acidity or basicity of a substance and its lipophilic or hydrophilic properties will determine how it behaves under physiological conditions inside the human organism. Beta-lactam antibiotics and aminoglycosides, for example, do not pass easily through membranes and therefore are found mostly in extracellular compartments. A summary of pharmacokinetic parameters of the individual groups of antibiotics is given in Table 5 [Tab. 5].

The distribution volume is an important pharmacokinetic parameter describing the distribution of the drug throughout the body. Lipophilic substances easily passing through membranes are taken up passively and intracellularly. Therefore, they show a large distribution volume: it may be as high as multiple body volumes for fluoroquinolones and macrolides. Drugs with large distribution volumes will have low plasma and interstitial levels but high intracellular concentrations. In contrast, water-soluble substances do not readily penetrate the cell membranes and therefore are found mainly in the plasma and interstitium, i.e. the concentration in these compartments is a most relevant measure.

An important determinant of drug distribution is the protein binding in serum. Antibiotics bind primarily to albumin, depending on their physicochemical properties. The concentration-dependent binding is reversible. A dynamic balance exists between free and bound drug. Generally speaking, only the free drug is responsible for efficacy. As shown for some antibiotics, high protein binding has no negative effects on the efficacy as long as there is a high enough unbound concentration at the site of action. Clinical studies which appear to substantiate a negative influence by the protein binding, were often conducted with inadequate total dosages [295], [363], [455].

Tissue concentrations are similarly important in predicting efficacy. A tissue concentration, as determined from biopsy material or surgical resected tissue, describes the average concentration in a tissue homogenate. These tissue concentrations fail to describe complex processes and heterogenous distribution inside the tissue. Measurements of tissue concentrations may be relevant, for example, in the comparison of two drugs or drug groups. The development of microdialysis could represent a big step forward in this field. The measurement of antibiotic concentrations are important in localizations such as cerebrospinal fluid, epithelial lining fluid, alveolar cells, pleural fluid, peritoneal fluid, and pancreatic or prostate secretion. Poor circulation, peculiarly structured cell membranes and the existence of specific tissue receptors may impede homogeneous distribution of antibiotics and therefore influence treatment success. Table 6 [Tab. 6] shows the access of antibiotics to various compartments.

Interaction of pharmacokinetics and pharmacodynamics

As data on the concentration profiles at the site of infection are limited, analysis of pharmacokinetics is currently based on plasma concentrations for many drugs. Different indices are recommended for drug groups according to their mechanism of action.

The differences in the pharmacodynamic profiles of antibiotic groups are explained by their divergent bactericidal activity – e.g. concentration-dependent bactericidal activity of fluoroquinolones and aminoglycosides versus time-dependent (i.e. not concentration-dependent) bactericidal activity of beta-lactam antibiotics and macrolides (see Table 7 [Tab. 7]). It was shown for aminoglycosides and fluoroquinolones that the ratio of maximum concentration (Cmax) and minimal inhibitor concentration (MIC) of the pathogen correlates with treatment success. In contrast, for beta-lactam antibiotics, the time (i.e. percentage of the dose interval) with plasma concentration above the MIC of the pathogen (t>MIC) is the relevant parameter. For fluoroquinolones, the ratio of AUC (area under the concentration-time curve) and MIC is also deemed important in predicting the outcome (area under the 24-hour concentration-time curve over the MIC: AUC24/MIC). Data available for oxazolidinones so far indicate that concentration and time are both relevant for the antimicrobial effect. This model was validated in humans for some groups of antibiotics.

Considering the PK/PD indices, when choosing a dose regimen, is vitally important particularly in immunosuppressed patients and for infections in poorly accessible localizations (abscesses, osteomyelitis, meningitis, necrotizing infections; see Table 6 [Tab. 6]). In addition, pathophysiological and therapeutic factors in the critically ill (capillary leakage due to endothelial damage, hypoalbuminemia, extracorporeal circulation, intravenous administration of high fluid volumes, vasopressor use) may result in enhanced distribution volumes and, due to increased renal perfusion in the absence of adequate organ function, increase the clearance of hydrophilic antibiotics and thus decrease their plasma concentrations.

Data on PK/PD correlations provide guidance when adjusting the dosage to individual needs with the help of therapeutic drug monitoring (TDM), particularly in high-risk populations (e.g. critically ill patients, elderly patients, and those with organ dysfunction). Clearance and distribution volumes determine the half-life of a drug. Both parameters are used to determine the time with plasma concentrations above MIC and the total exposure (AUC). They thus have an important role in the calculation of dose intervals.

Functional impairment of drug-excreting organs (mainly kidneys and liver) results in a reduced clearance of antibiotics and prolongation of the half-life, which may result in an increased rate of side effects. Reduced kidney and liver function is less important for antibiotics with wide therapeutic range (broad concentration range between effective and toxic levels, e.g. for penicillins, cephalosporins, carbapemens, macrolides, lincosamides, fluoroquinolones and linezolid) than for antibiotics with narrow therapeutic margins (e.g. aminoglycosides and vancomycin). Therefore, besides microbiological efficacy, the degree of renal and extrarenal excretion plus any nephrotoxic and/or hepatotoxic potential of the antibiotic itself or its metabolites play an important role in the choice of suitable antibiotics.

These antibiotics (potentially nephrotoxic: aminoglycosides and vancomycin; potentially hepatotoxic: amoxicillin/clavulanic acid, flucloxacillin, fluoroquinolones, and intravenous tetracyclines) should be used only in vital indications if the function of the relevant organs is impaired. Potential risks by accumulation of toxic metabolites should be considered in patients with pronounced renal or hepatic impairment. As a rule, antibiotics with high extrarenal elimination should be chosen in patients with impaired kidney function and antibiotics primarily excreted via the kidneys should be used for patients with liver dysfunction.

Antibiotics primarily excreted via the kidneys are eliminated by glomerular filtration and tubular secretion (e.g. penicillins) and may be reabsorbed in different amounts. In patients with renal dysfunction, the dosage should be adapted to the degree of impairment, according to the creatinin clearance. Parameters of relevance for dose adjustments include

  • percentage of drug eliminated via the kidneys in individuals with normal kidney function,
  • drug toxicity
  • degree of renal impairment.

Generally, recommendations of the manufacturer should be followed when choosing the dosage for patients with organ dysfunction. In the absence of such recommendations, the adaptation of the dosage regimen for renal dysfunction should be carried out by calculating the individual excretion factor (Q) according to Dettli [129], [252].

Helpful links to websites on dosage adaptation for renal impairment include: http://www.zct-berlin.de/niereninsuff/; http://www.dosing.de/; http://doseadapt.unibas.ch/. Unlike creatinine clearance in renal insufficiency, clinical scores (Child Pugh score, MELD score) in liver insufficiency are inadequate predictors for drug metabolization and elimination.

Liver diseases have a varying and unpredictable influence on cytochrome P450 isoenzyme activities. Available tests provide only a rough estimate of the function of individual isoenzymes. The reduction in hepatic clearance and the associated need of dose adjustments may be relevant for antibiotics which are almost completely metabolized by liver enzymes, primarily those with high lipophilia and low polarity which are poorly excreted via the kidneys (antibiotics with high extrarenal clearance include clindamycin, chloramphenicol and minocyclin).

High-grade hepatic dysfunction with reduced metabolic capacity should also be considered when choosing the dosage of tetracyclins, clavulanic acid, flucloxacillin, macrolides and streptogramins. For antibiotics with high presystemic elimination rates (first-pass effect), the bioavailability and therefore the plasma concentration may increase significantly in patients with impaired hepatic function (e.g. ciprofloxacin). In patients with renal and liver dysfunction, the loading dose (initial dose), which is dependent on the distribution volume should be the same as for patients with normal kidney and liver function. If the initial dose of the antibiotic is reduced, the effective concentration may be reached after a delay of several days. This may jeopardize treatment success of antibiotic therapy, which primarily depends on the initial choice and adequate drug exposure.

A particular challenge in pharmacotherapy is the dosing of antibiotics in obese patients. The pharmacokinetics of many antibiotics are unpredictable in these patients due to alterations in drug distribution. There is no clear relationship between the lipophilic properties of the drug and its distribution in obese patients. Alterations in distribution volumes and clearance as well as problems in estimating kidney function based on creatinine clearance are among the factors commonly leading to underexposure in obese patients who receive standard dosages of antibiotics. Subtherapeutic concentrations may lead to therapeutic failure and development of resistance. As higher distribution volumes and higher clearance should generally be expected for these patients, weight-based dose adjustments are required. Which weight parameter (TBW – total body weight, IBW – ideal body weight, LBW – lean body weight or ABW – adjusted body weight) should be used as the basis of dosage calculation depends on the drug itself and on the mode and duration of administration [160], [211], [373], [394].

In difficult-to-treat patients, therapeutic drug monitoring (TDM) is advisable but rapid testing is available for only few antibiotics (e.g. for aminoglycosides and glycopeptides). Dosage recommendation should be followed particularly for pediatric patients with cystic fibrosis, sepsis, burns or high body weight. The pharmacokinetic characteristics of the individual drugs are summarized in Table 5 [Tab. 5].

Therapeutic drug monitoring

Many antibiotics are characterized by substantial interindividual and intraindividual variability of pharmacokinetic parameters, particularly regarding elimination and distribution volume. This is particularly true for intensive care patients with multiple organ failure and major alterations in fluid volumes (e.g. due to capillary leakage and infusion therapy). Plasma concentrations achieved with standard dosages may therefore show major variations resulting in underexposure and therapeutic failure or elevated plasma levels with the risk of toxic effects.

Therapeutic drug monitoring (TDM) is used to determine the optimum dose regimen for individual patients by measuring drug plasma levels and applying pharmacokinetic principles.

Prerequisites and/or indications for TDM primarily include the following:

  • Concentration-effect correlations are known for therapeutic and toxic effects.
  • The drug has a narrow therapeutic range. Exceeding this range by a relatively small margin results in a risk of toxic effects.
  • The pharmacokinetics of the drug is subject to substantial intraindividual and/or interindividual variability.
  • Pharmacokinetic targets (Cmax, Cmin, AUC) are known.
  • Analytical methods providing adequate sensitivity can be used with reasonable expenditure.

For many antibiotics, e.g. penicillins and cephalosporins, the risk of toxic effects is small as they have a relatively broad therapeutic range. Treatment with these antibiotics rarely requires plasma level based adjustments. In contrast, aminoglycosides and glycopeptides are examples of antibiotics where TDM is strongly recommended for safe use. Table 8 [Tab. 8] shows recommendations of target areas for maximum and minimum levels of commonly used aminoglycosides and glycopeptides with respect to patient group.

When using aminoglycosides, single-dose application of the entire daily dosage is associated with increased clinical efficacy, less toxicity and economic advantages [17], [45], [87], [92], [110], [128], [153], [223], [228], [344], [386]. Based on PK/PD parameters, peak levels clearly above the MIC of the pathogen (Cmax/MHK>10) are targeted for aminoglycosides [404], [436]. The mean MIC of gentamicine for pathogens with reduced susceptibility (e.g. Pseudomonas aeruginosa) is 2.0 mg/L. Therefore, the target peak levels are at least 20 mg/L [423]. There is insufficient data for once daily use in endocarditis and neutropenic patients.

For the glycopeptides vancomycin and teicoplanin, the pharmacodynamic parameters require continuous drug levels above the MIC of the pathogen. As a rule, trough levels are determined in TDM [315]. When treating life-threatening infections (meningitis or pneumonia) and infections by pathogens with reduced susceptibility, a vancomycin trough level of 15–20 mg/L should be achieved [259], [309], [403]. However, there is an increased risk of nephrotoxicity at vancomycin trough levels of 15 mg/L and above [227].

When treating infections of bone or prosthetic joint infections and endocarditis, teicoplanin trough levels of 20–25 mg/L are recommended [492].

Continuous infusion of beta-lactam antibiotics

Beta-lactam antibiotics are effectively active if the MIC of the pathogen is exceeded as continuously as possible during the growth phase of the cell wall. Initially, the bactericidal efficacy increases with ascending concentrations up to this value; however exceeding this level will not improve treatment results. This pharmacokinetic-pharmacodynamic relationship is described as time-dependent rather than concentration-dependent bactericidal. For beta-lactam antibiotics, the concentration of the unbound antibiotic should exceed the MIC of the pathogen at the site of infection for at least 40–60% of the dosing interval [375]: 40% appears adequate for carbapenems, the higher value applies for cephalosporines, with penicillins requiring around 50%.

There is not a large variation in the pharmacokinetic data for beta-lactam antibiotics. After parenteral administration, beta-lactam antibiotics disperse rapidly in the extracellular region. In dynamic equilibrium, similar concentrations are reached after intermittent administration and after bolus administration followed by continuous infusion [40], [57], [58], [292], [346], [347].

Manufacturers typically recommend the administration of beta-lactam antibiotics (1) 2 to 4 (6) times daily, depending on pharmacokinetic parameters. Thus, adequate levels of free active drug that exceed the MIC of susceptible pathogens are reached in licensed indications which are supported by clinical studies. However, when using intermittent administration concentrations often fail to exceed the MIC of the pathogen as long as possible in the infected region as has been shown in PK/PD simulations and in experimental and clinical investigations. This is particularly true for patients with large extracellular distribution volumes and increased clearance rates. This primarily applies to patients with capillary leakage e.g. due to sepsis, patients with cystic fibrosis, drainage, bleeding, large burns, ascites, severe pancreatitis, BMI >30 kg/m2, cardiac insufficiency, edema, hemofiltration (depending on net fluid balance), dialysis and pregnancy. In contrast, desiccated patients, dialysis patients after a dialysis session, and patients with volume restrictions have smaller distribution volumes than normal patients. Individualized antibiotic therapy is recommended for high-risk and elderly patients [91], [186], [270], [284], [291], [313], [376], [377], [434], [435], [437], [454], [473].

These newer considerations are already followed for doripenem. Doripemen can be administered by short infusion or prolonged infusion over 4 hours [162]. Recommendations for continuous administration of beta-lactam antibiotics are based on theoretical considerations supported by experimental investigations or simulations. Evidence from clinical investigations supports advantages for continuous administration with prolonged maintenance of serum levels above MIC even at low daily dosages [12], [13], [86], [88], [89], [90], [98], [120], [178], [282], [283], [288], [348], [349], [424], [438], [512], [515], [516] with comparable clinical and microbiological efficacy and safety [190], [200], [376]. However, a significant superiority of continuous administration was shown in only few cases to date [312] and reduction of mortality has not been shown yet.

The stability of beta-lactam antibiotics after reconstitution is limited. This implies inactivation by degradation and decomposition products which may potentially trigger allergies. This aspect is neglected in numerous investigations on the stability of drugs. Solutions of beta-lactam antibiotics are considered to be stable within a given interval if less than 10% of the drug is degraded. The rate of degradation depends on solvent, light exposure, drug concentration, type of administration device, manufacturing and temperature. In out-patient antibiotic therapy (APAT) with portable pumps carried close to the body stability may be significantly reduced due to increased ambient temperature.

The use of the recommended solvent is a point of high practical importance to achieve optimum solubility and stability. Practically all penicillins (dry substances) must be dissolved in water for injection in order to accelerate solubility and avoid particle formation. Further dilution in the usual infusion solvents is generally feasible. For many beta-lactam antibiotics, there is a list of incompatibilities with other drugs if administered via the same infusion system. To follow the manufacturer specifications on compatibility is imperative.

The most frequent side effects of penicillins include allergic and pseudoallergic reactions. These reactions are caused by the instable beta-lactam structure or specific side chains. Immediate and delayed-type reactions may occur. Immediate-type allergies to penicillin following prior sensitization (e.g. by food containing penicillins or contact with moulds) usually evolve within minutes as urticaria-type exanthema and/or angioedema, potentially with life-threatening respiratory or cardiovascular complications. About 10% of delayed-type reactions are polymorphic, e.g. maculopapular exanthema. Serum-sickness-like symptoms are observed in 2% to 4% of cases. Allergic reactions are generally more frequent after parenteral than after oral administration.

Penicillins have variable stability in solution, depending on molecular side chains and the pH of the solvent. Degradation products of penicillins act as haptens and may form covalent bonds with host proteins. The hapten-protein complex may induce an allergic immune reaction.

Degradation products of penicillins have a significant allergenic potential. The most common degradation product is penicilloic acid, a product generated by beta-lactam ring opening. The penicilloyl-protein complex is designated the major determinant (major epitope). It is the cause of most penicillin allergies. The penicilloate, penilloate, penicillenate, penicilloinic acid, penicillanyl, penamaldate, penaldate and D-penicillamine determinants are minor determinants (minor epitopes). These minor determinants appear to be more important for critical clinical events (shock), even though anaphylactic reactions due to penicilloylic acid sensitization were also described.

Improper storage and preparation may induce the formation of larger amounts of degradation products which greatly increase the risk of allergic reactions to penicillin solutions. Thus the incidence of proven (0.9%) or probable (1.7%) adverse drug reactions was significantly lower with short infusions of freshly prepared penicillin solutions compared to continuous administration or infusion of stored solutions (8.3%, 6.7%, respectively) [369], [370], [371]. However, these differences could not be confirmed in further clinical studies.

The type and extent of degradation observed with a beta-lactam antibiotic depends on the substance. As a rule, acylaminopenicillins, isoxazolylpenicillins, cephalosporins and aztreonam are more stable than benzylpenicillins due to their molecular structure. Ring opening induced by nucleophilic or electrophilic (less frequent) attacks is however also possible in cephalosporins as observed for example with ceftazidim and other cephalosporins [474]. The chemical stability of carbapenems varies greatly, only doripenem may be administered by prolonged 4-hour infusions (as described in the label) [162].

Summary
  • Due to pharmacokinetic/pharmacodynamic considerations, the continuous infusion of beta-lactam antibiotics is superior to intermittent administration in achieving the goal of exceeding the MIC of the pathogen as continuously as possible.
  • Few clinical data are available on any significant superiority of this treatment regimen.
  • Continuous and intermittent infusions of a beta-lactam antibiotic has comparable side-effect profiles.
  • Continuous administration is recommended for patients whose pharmacokinetic parameters (distribution volume, clearance) deviate significantly from normal populations (e.g. patients with cystic fibrosis or patients with severe septic infections caused by pathogens with reduced susceptibility).
  • Continuous administration of the antibiotic should be preceded by a single bolus administration.
  • Economic advantages may be associated with continuous administration, as in non-severely ill patients, steady-state serum concentrations are achieved with lower daily dosages compared to intermittent infusion.
  • Due of limited stability at room temperature, some beta-lactam antibiotics are not suitable for continuous administration. In these cases, only prolonged infusion (3 hours) is possible.
  • Strict adherence to the manufacturer’s recommendations on the type of solvent and concentrations of the antibiotic solution is mandatory. Deviations may cause considerably reduced stability.
  • It is essential to use a dedicated intravenous line or lumen specifically for the continuous administration of a beta-lactam antibiotic as numerous incompatibility reactions with other drugs can occur.
  • Continuous or prolonged infusions are not licensed except for doripemen. Use of these regimens in practice therefore must be considered “off-label use”.

Drug interactions

Interactions with other drugs are an important cause of adverse reactions. Particularly the inhibition of hepatic monooxygenases (cytochrome P450 isoenzymes), for example by some macrolides and fluoroquinolones as well as azole antifungals, may cause an increased risk of side effects.

In addition, some drugs, for example rifampicin, barbiturate and carbamazepine, induce enhanced expression of cytochrom P450 isoenzymes, resulting in lower plasma levels with reduced efficacy of the affected drug.

Further important drug-drug interactions of antibiotics are listed in Table 9 [Tab. 9].


4 Safety and tolerability

Ralf Stahlmann, Hartmut Lode

Adverse drug reactions must be expected in about 10% of patients treated with most parenterally administered antibiotics. In some drugs, the side-effect rates are even higher. Therefore the differences in the tolerability of the available medications are of major importance. However, any comparison of results generated in different clinical trials is inadequate to evaluate differences in the tolerability of antibiotics. Despite broad standardization of clinical trials, data generated in single head-to-head trials, preferably double-blind studies, are the only reliable basis for direct comparisons of different drugs. This applies for side effects as well as for clinical efficacy. The number of patients treated in clinical studies in insufficient to derive reliable conclusions on infrequent side effects. Therefore, evaluations of pooled data from multiple clinical studies or even the experience gained from postmarketing surveillance must be considered. However, the limitations associated with this type of data should be taken into account.

In general the adverse drug reactions of most antiinfectives prescribed for parenteral therapy predominantly affect three organ systems:

  • gastrointestinal tract (e.g. nausea, vomiting, diarrhea),
  • skin (e.g. rashes/eruptions, urticaria, phototoxicity),
  • CNS (e.g. headaches, dizziness, sleep disturbances).

There are significant differences in the severity and the frequency of a given side effect. Toxic, allergic and biological effects may be differentiated according to the pathogenesis of the adverse effects. In some cases it remains unclear if, for example, a disturbance of the gastrointestinal tract is due to direct effects on the affected organs or whether the changes are caused by the impact on the bacterial flora.

As a rule, any administration of an antimicrobial drug has an effect on the bacterial flora. The type and extent of the changes are determined primarily by the pharmacokinetic characteristics of the antibiotic. Therefore the biological side effects of a drug must be considered in the risk/benefit assessment of each antibacterial treatment.

Beta-lactam antibiotics

Parenterally administered beta-lactam antibiotics are generally well tolerated. The side effects are usually mild and temporary. Treatment discontinuation is rarely required. In about 1–2% of patients, hypersensitivity may occur as a morbilliform or scarlatiniform erythema. This may be associated with edema of face, tongue or glottis (e.g. Quincke’s edema) in rare cases (0.5–1%). Pneumonitis and/or interstitial pneumonia and interstitial nephritis are extremely rare. In isolated cases (<0.1%) severe acute allergic reactions (anaphylaxis with life-threatening shock) (independent of the administered dose) occur, usually within 30 minutes after administration. This type of reaction is more frequently reported for penicillins than for other beta-lactam antibiotics. Patients should be monitored for allergic reactions during the first 30 minutes after the administration of any beta-lactam antibiotic.

Cross-allergies of penicillins and cephalosporins are quite rare. Aztreonam may be used in patients who developed skin eruptions or other types of acute hypersensitivity reactions to penicillins or other beta-lactams, as cross-allergies are very rare based on the available experience. In some patients, allergies to aztreonam have been observed, These are more likey caused by the structure of the side chain than the beta-lactam ring itself. As the aztreonam side chain is identical with the corresponding structure in ceftazidim, aztreonam should not be used after an allergic reaction to ceftazidim and vice versa [18], [568].

Reduction on blood counts (thrombocytopenia and/or eosinophilia, rarely (<2%) as leukopenia) are mediated by allergic or toxic mechanisms. The effects are general reversible within a few days of treatment discontinuation.

Gastrointestinal intolerability manifested as loss of appetite, nausea, vomiting, adominal pain, meteorism or soft stools are often observed in patients receiving beta-lactam antibiotics. Diarrhea (more than 3 bowel movements with loose stool per day) occurs in 2–10% of patients.

In patients receiving intravenous ceftobiprol, the rate of gastrointestinal disturbances such as nausea and vomiting was dependent on duration of the infusion. Patients often reported dysgeusia during the clinical trials [381].

Reversible moderate changes in liver function parameters (e.g. transaminases, alkaline phosphatase) occur in up to 10% of patients. In some cases, transient cholestatic hepatitis was observed. The risk increases with increasing age and duration of therapy. Amoxicillin/clavulanic acid should only be used with liver function monitoring in elderly patients (>65 years).

During treatment with ceftriaxone, alterations of gallbladder ultrasound images have been observed in rare cases. These changes disappear after discontinuation or completion of therapy (transient biliary pseudolithiasis).

In patients with certain risk factors (severely impaired kidney function or epilepsy, breached blood-brain barrier, e.g. meningitis), seizures may occur after administration of beta-lactam antibiotics in very high doses. As observed in animal experiments, the seizure risk is lower with newer carbapenems versus imipenem/cilastatin. Therefore meropenem rather than imipenem/cilastatin is licensed for the treatment of meningitis [585].

If meropenem or other carbapemens are used in combination with valproic acid, the plasma concentrations of the antiepileptic are significantly reduced and may result in an increased risk of seizures. Valproic acid is predominantly metabolized by glucuronidation. However, the parent drug may subsequently be released from the metabolite by hydrolysis. Carbapenems apparently block this hydrolysis of the glucuronide resulting in a reduction of the plasma levels of free valproic acid. Therefore, the valproic acid serum level must be monitored and the dosage adjusted correspondingly if a carbapenem is used concomitantly [345].

Long-term and repeated use of beta-lactam antibiotics (particularly those with broad antibacterial spectrum) may lead to superinfection or colonization with resistant pathogens or yeasts (e.g. oral thrush, vulvovaginitis).

Fluoroquinolones

During treatment with fluoroquinolones (e.g. ciprofloxacin, levofloxacin, moxifloxacin) adverse effects occur in about 4–10% of patients [494], [531].

Adverse effects most often affect the gastrointestinal tract or the CNS (e.g. sleep disturbances, dizziness). Regarding skin reactions, the phototoxic potential of fluoroquinolones has attracted particular notice. Basically, direct exposure to sunlight (or UV light from other sources) should be avoided in all treatments with fluoroquinolones. Cardiotoxic effects were observed in animal experiments after administration of fluoroquinolones withdrawn from the market including sparfloxacin. Minor changes of QTc time may occur in humans as well. These resulted in the recommendation that fluoroquinolones should not be combined with other arrhythmogenic drugs. Patients with hypokalemia and hypomagnesemia have a higher risk of torsades de pointes [232]. In very rare cases, hepatic reactions from hepatitis to liver failure have been observed in conjunction with fluoroquinolones.

Fluoroquinolones are contraindicated for children, adolescents and pregnant women. The clinical relevance of typical fluoroquinolone effects on immature cartilage remains controversial, however. Ciprofloxacin is increasingly being used for example e.g. in teenage cystic fibrosis patients. No increase in the rates of clinically relevant joint disturbances was reported. Inflammation or ruptures of the achilles tendon may occur as rare adverse effects of all fluoroquinolones [472]. Among the fluoroquinolones available for parenteral therapy, ciprofloxacin inhibits the cytochrome P450 isoenzyme 1A2. Therefore, the metabolism of theophylline, caffeine and other pharmacological agents; e.g. clozapin, may be inhibited to a clinically relevant extent [83].

Macrolides, azalides

Besides the classical macrolide erythromycin, clarithromycin and azithromycin are currently available for parenteral therapy. Due to its divergent structure, azithromycin is also categorized as an azalide. Macrolides/azalides often cause local intolerability reactions at the infusion site.

Besides local intolerability, the most frequent side effects of these antibiotics – including their parenteral use – are gastrointestinal disorders. The gastrointestinal reactions are caused primarily by direct stimulation of smooth muscles as the macrolides/azalides act as motilin agonists. The newer derivates of the erythromycins (clarithromycin, azithromycin) are better tolerated gastrointestinally than the classical macrolide [183].

Macrolides may cause allergic reactions. These are clearly less frequent than with penicillin or other beta-lactam antibiotics.

Cases of reversible hearing loss have been reported in association with high doses of erythromycin (intravenous administration). Cardiotoxic effects may also occur after administration of macrolides due to QTc prolongation with a potential for serious arrhythmia (torsades de pointes). This risk appears to be relatively small with azithromycin [481].

Interactions between erythromycin/clarithromycin and numerous other pharmaceuticals metabolized by the cytochrome P450 monooxygenases (e.g. CYP3A) have long been known. The phase I metabolism of carbamazepine, glucocorticoids, terfenadine, theophylline, cyclosporins and many other drugs is inhibited by macrolides. Particularly drugs associated with QTc prolongation (e.g. terfenadine, pimozide) may cause torsades de pointes when administered in combination with these macrolides. Based on available experience to date, inhibition of metabolizing enzymes is not associated with the azalide azithromycin. Cases of interaction with digitalis glycosides have been described for all macrolides/azalides, these are apparently caused by other mechanisms [475].

Glycopeptides

Hypersensitivity reactions such as fever, urticaria and exanthema may be associated with the use of glycopeptides (vancomycin, teicoplanin) [350].

Rapid infusion of vancomycin may trigger the release of mediators, causing red skin discoloration (red man syndrome), pain and cramps in the chest and back muscles. These reactions generally disappear within 20 minutes to a few hours after the end of infusion. As these symptoms rarely occur after slow infusions, it is imperative that vancomycin is adequately diluted and infused over a sufficiently long period of time. Similar reactions almost never occur with teicoplanin [350].

Gastrointestinal disorders (e.g. nausea, vomiting) may sometimes occur during treatment with glycopeptides. Acute renal failure has been observed in some cases after the administration of vancomycin in high doses. The risk of nephrotoxic reactions increases with higher doses and if vancomycin is combined with other potentially nephrotoxic drugs [227], [310], [446]. Temporary or permanent hearing impairments have occasionally been reported [173].

Alterations of blood counts are rarely observed with glycopeptides (transient neutropenia, thrombocytopenia or eosinophilia). Glycopeptides may cause pain at the injection site (thrombophlebitis).

Aminoglycosides

Aminoglycosides have a narrow therapeutic range. All drugs of this group are potentially nephrotoxic and ototoxic. In addition they may disrupt neuromuscular signal transmission and are therefore contraindicated in patients with myasthenia gravis [192], [207].

Aminoglycosides accumulate in cochlear hair cells and in the renal cortex. Therefore the risk of toxic damage increases significantly if treatment is continued for more than 8 days or if the patient has been preexposed to an aminoglycoside within 6 weeks before the start of treatment [140].

Ototoxicity and nephrotoxicity tend to be less likely if the entire daily dose is given in a single short infusion versus three divided doses. A once-daily dosing also appears to be beneficial with respect to the antibacterial effect, this dosing concept has become increasingly accepted in recent years [192], [445].

Vestibular (dizziness, nystagmus) and cochleal damage occur particularly in patients with renal impairment or in those treated with high dosages. Initially hearing loss primarily affects high frequencies [207].

Allergic reactions to aminoglycosides are rare [192].

Oxazolidinones (linezolid)

Linezolid is the first oxazolidinone used in human medicine. During clinical trials it was as well-tolerated as the comparator drugs. Gastrointestinal disturbances, e.g. vomiting, and mild CNS reactions were the most common side effects. Hematologic events (thrombocytopenia, neutropenia, anemia) were observed in patients receiving prolonged linezolid treatment (>2 weeks). Therefore, the blood counts should be monitored weekly during treatment with linezolid.

Peripheral neuropathy and/or optical neuropathy, very rarely associated with progression to blindness, were reported in patients treated with linezolid. These affected patients who were treated for longer than the maximum recommended duration of 28 days. Cases of lactate acidosis also occurred in long-term treatment [42], [361], [488].

Linezolid is an inhibitor of monoaminoxidase. Relevant interactions with concomitantly used adrenergic or serotonergic drugs may therefore occur. This may be relevant in simultaneous treatment with selective serotonin reuptake inhibitors and other drugs such as tricyclic antidepressants, serotonin-5-HT1 receptor agonists (triptanes), directly or indirectly acting sympathicomimetics (including adrenergic bronchodilators, pseudoephedrine or phenylpropanolamine), vasopressors (e.g. epinephrine, norepinephrine), dopaminergic drugs (e.g. dopamine, dobutamine) and pethidine or buspirone. Linezolid should not be used in conjunction with these drugs [334], [513].

Lincosamides (clindamycin)

The most frequent side effect of clindamycin is diarrhea due to the deterioration of physiological gut flora (5–20%). Severe pseudomembranous enterocolitis may occur in patients treated with clindamycin [391]. Serum bilirubin and liver enzyme levels may sometimes be elevated on clindamycin. Hypersensitivity reactions are comparatively rare. Hematological disturbances including e.g. thromboyctopenia and leukopenia are mostly observed in patients receiving longer clindamycin treatment courses [481].

Metronidazole

The most frequent side effects of metronidazole are gastrointestinal disorders, i.e. bitter-tasting regurgitation, metallic taste alteration and nausea. Diarrhea is rare [452]. Potential neurological disturbances include headache, dizziness, ataxia, and paraesthesia. Reversible peripheral neuropathy may occur with high dosages and long-term treatment. Cases of aseptic meningitis in association with metronidazol have been described [254]. Allergic reactions and haematological disturbances are possible [261]. A disulfiram effect occurring on metronidazole with simultaneous alcohol consumption has been reported – available data on this effect are contradictory however [543].

Tetracyclines (doxycycline) and glycylcyclines (tigecycline)

Doxycycline

From the tetracycline group, only doxycycline can be administered intravenously. Gastrointestinal disturbances are the most frequent side effects of doxycycline. Nausea, vomiting or diarrhea (occasionally pseudomembranous enterocolitis) may occur. Tetracyclines are potentially phototoxic. CNS reactions may appear as headache, nausea and photophobia. Severe anaphylactic reactions are very rare. Rapid injection may cause dizziness, flushing, reddening of the face and collapse. Intravenous administration is associated with local irritation and may cause local inflammation (thrombophlebitis). In this case, treatment should be change to oral administration if possible [453].

Tigecycline

In clinical pivotal studies, tigecycline caused gastrointestinal side effects (e.g. nausea) more frequently than the comparator drugs [175]. In the phase III studies, tigecycline was associated with vomiting in 19%, imipenem in 14% and vancomycin/aztreonam in 3.6% of the patients. Increases in transaminase levels were observed more commonly in patients receiving vancomycin/aztreonam, skin reactions were significantly more frequent than in the tigecycline group (19% vs. 10.6%). In all groups, the rates of treatment discontinuations due to adverse events were similar. Gastrointestinal side effects are dose-dependent and occurred more commonly in female patients [398].

Concomitant therapy with tigecycline and warfarin resulted in increased plasma concentrations of R and S warfarin (AUC values) by 68% and 29%, respectively. Although no direct effect on coagulation was observed, monitoring of the INR during concomitant therapy is advised.

Daptomycin

Daptomycin was as well tolerated as the comparator drugs in clinical trials [169]. The most frequent side effects were constipation (6.2%), nausea (5.8%), injection site reactions (5.8%), and headache (5.4%). Daptomycin may cause adverse effects on skeletal muscle [289], [385]. In an early phase I study, many cases of reversible CPK (creatine phosphokinase) values were observed at a dosage of 3 mg/kg of body weight every 12 hours. These effects occur less frequently with once daily administration. In addition, increased transaminase levels associated with effects on the skeletal muscles may occur. Regular observation for clinical signs of myopathy and CPK monitoring (once weekly) are generally recommended for daptomycin use [22].

As daptomycin is primarily excreted renally, increased plasma levels of daptomycin are expected when administered concomitantly with drugs that decrease renal filtration (NSAIDs, COX-2 inhibitors). Treatment with drugs that may cause myopathy should be discontinued during treatment with daptomycin as, in some cases, a significant increase in CPK was observed and, in isolated cases, rhabdomyolysis occurred [395]. If a simultaneous administration cannot be avoided, the CPK values should be monitored more frequently than once per week and the patient should be carefully observed.

Colistin

Among the polymyxins, polymyxin E (colistin) in particular experienced a renaissance in the last few years [301]. Adverse effects of colistin include gastrointestinal disorders, CNS symptoms (dizziness, paraesthesia) and skin reactions [109]. The nephrotoxicity of polymyxins is dose-dependent. Colistin appears to be less nephrotoxic than polymyxin B. However, this advantage is at least partially abrogated by the required higher dosages. Therefore similar rates of nephrotoxic reactions are expected in clinical use. Only insufficient data on the nephrotoxicity of both antibiotics are available yet. In clinical studies, nephrotoxic reactions were observed in 7–45% of patients. Note, however, that different definitions of nephrotoxicity were used and that some of the data came from patients with severe underlying diseases, making the interpretation of these results rather difficult [255]. Using the RIFLE criteria in a group of relatively young male patients mostly without underlying diseases, almost every second patient had mild, reversible nephrotoxicity. Treatment was discontinued in 21% of patients due to renal dysfunction [218]. Significant nephrotoxic reactions were reported for the majority of patients who already had renal impairment before treatment was started [335]. For patients with existing kidney dysfunction, the dosage must be reduced to the level recommended by the manufacturer.

Fosfomycin

The most frequent adverse effects of fosfomycin include gastrointestinal symptoms (nausea, sickness, vomiting, diarrhea) and skin eruptions (exanthema). Fatigue, headache and taste disturbances were observed as additional side effects. Hematological changes such as eosinophilia or aplastic anemia are observed rarely or very rarely. Anaphylactic shock and liver dysfunctions occurred very rarely. However, phlebitis at the administration site is a frequent adverse event [144], [163], [165], [251].

Administration of 1 g of fosfomycin (equivalent to 1.32 g of fosfomycin sodium) carries 14.5 mmol Na+. Therefore, serum electrolytes should be monitored when using recommended dosages. This is particularly important in patients with, for example, congestive heart disease, edema, or secondary hyperaldosteronism. The amounts of sodium administered with fosfomycin may increase the elimination of potassium and cause a net loss of potassium. Therefore, replacement of potassium may be necessary to avoid hypokalemia.

Rifampicin

Gastrointestinal intolerance is often observed during treatment with rifampicin. Symptoms include loss of appetite, gastric pain, nausea, vomiting, meteorism and diarrhea. Cases of pancreatitis are occasionally reported [95].

Hypersensitivity reactions are frequently seen with rifampicin [321]. The most frequent manifestations are fever, multiform exsudative erythema, pruritus and urticaria. Occasionally, severe reactions such as dyspnea, pulmonary edema, other edemas and shock have been observed. A lupus-like syndrome with fever, asthenia, muscle and joint pain, and antinuclear antibodies has been reported very rarely.

Hepatic side effects of rifampicin are frequent to very frequent, primarily manifesting as elevated transaminases, alkaline phosphatase, gamma glutamyltranspeptidase and less frequently serum bilirubin. These values often normalize on treatment [203].

Visual disorders, loss of vision and neuromyelitis optica may occur as severe side effects. A reddish brown discoloration of tear fluid is a harmless effect caused by the color of the active drug.

In rare cases the use of rifampicin results in eosinophilia, leukopenia, granulocytopenia, thrombocytopenia, thrombocytopenic purpura, hypoprothrombinemia or haemolytic anaemia.

Rifampicin is a potent inducer of cytochrome enzymes, phase II enzymes and transporter proteins. It causes, for example, a significant induction of the CYP3A4, 1A2, 2C9, 2C8 and 2C18/19 isoenzymes in the intestinal epithelium and the liver, thereby accelerating the metabolism of other drugs. It inhibits N-acetyltransferases. Rifampicin also blocks transport proteins for organic anions (OATP2). In view of the complex and diverse effects on the pharmcokinetically relevant metabolization and transport systems, hospital physicians should expect pharmacokinetic interactions with other drugs in any concomitant use of rifampicin [491].


5 Respiratory infections

Klaus-Friedrich Bodmann, Béatrice Grabein, Mathias Pletz, Gerd Höffken, Hartmut Lode

Respiratory tract infections are the most common infectious diseases in adults treated in hospitals and practices. The successful treatment of bacterial infections is favoured by early diagnosis followed by adequate antimicrobial treatment which requires calculated treatment at least in the initial phase.

Viruses are the predominant pathogens of upper respiratory tract infections whereas bacteria are more prevalent in infections of the lower respiratory tract.

Pneumococci are the most frequent bacterial pathogens of community-acquired respiratory tract infections. Haemophilus influenzae, Moraxella catarrhalis, Mycoplasma pneumoniae, Legionella spp. and enterobacteriaceae are important organisms as well. Staphylococcus aureus and Chlamydophila pneumoniae may occasionally be isolated. The CAPNETZ analysis provided current epidemiological data for Germany.

The spectrum of pathogens causing nosocomial pneumonia is significantly broader and includes, besides the pathogens observed in community-acquired infections, potentially multiresistant nosocomial pathogens such as methicillin-resistant S. aureus (MRSA), extended spectrum beta-lactamase (ESBL) producing enterobacteriaceae, Pseudomonas aeruginosa, Acinetobacter spp. and Stenotrophomonas maltophilia.

The resistance situation in pneumococci is still favourable in Germany, with <10% of the isolates showing reduced susceptibility to penicillin and a maximum of 2% being resistant. A trend towards reduced resistance rates has been observed for macrolides (see chapter 2).

In Germany, H. influenzae and M. catarrhalis as well are still susceptible to the recommended antibiotics.

The resistance situation of pathogens involved in nosocomial pneumonias has not yet been investigated in a large epidemiological study in Germany. Large variability of resistance across hospitals and even across departments means that current information on the local epidemiology and susceptibility is particularly important for the local implementation of treatment recommen-dations and guidelines.

The PEG Resistance Study, which includes about 20% respiratory tract isolates, has shown an increasing prevalence of ESBL-producing enterobacteriaceae in Germany. The percentage of ESBL producers increased particularly in Escherichia coli (from 1% to 10%) and in Klebsiella pneumoniae (from 4% to 10%).

Data aquired by the Krankenhaus-Infektions-Surveillance-System (KISS) (Hospital Infections Surveillance System) during 2007 and 2008 show that the proportion of MRSA in ventilator-associated pneumonia is almost 37% in the participating intensive care units.

Previous antibiotics treatment within the last 3 months predisposes patients to infections caused by pathogens with resistance particularly to the antibiotic class used. This association has been shown for beta-lactam, macrolide and fluoroquinolone antibiotics.

In the following chapter on community-acquired lower respiratory tract infections, reference will be made exclusively to the S3 guidelines on the epidemiology, diagnostics, antimicrobial treatment and management of adult patients with community-acquired respiratory tract infections [230]. These guidelines were produced by the Paul-Ehrlich Society for Chemotherapy (PEG), the German Pneumology Society (DGP), the German Infectiology Society and the CAPNETZ foundation.

Acute exacerbations of COPD (AECOPD)

Definition of AECOPD

AECOPD is defined as acute deterioration of respiratory symptoms in known chronic obstructive pulmonary disease (COPD) which requires additional treatment beyond the chronic platform therapy.

Aetiology of AECOPD

Almost half of all AECOPD episodes are caused by infectious agents, primarily respiratory viruses including respiratory syncytial virus (RSV), rhinoviruses, coronaviruses, adenoviruses, human metapneumovirus (HMP), and influenza viruses.

The most frequent bacterial pathogens are H. influenzae, S. pneumoniae, M. catarrhalis, enterobacteriaceae, and P. aeruginosa.

Clinical symptoms of AECOPD

The cardinal symptoms of AECOPD include:

  • increasing dyspnea
  • more frequent coughing
  • increase in amount and/or viscosity of sputum
  • chest tightness
  • unspecific signs such as
    • early tiring
    • sleep disruption
    • depression
    • and/or impairment of consciousness up to coma (CO2 narcosis)

Indications for antimicrobial treatment of patients with moderate to severe AECOPD

Besides the severity of the AECOPD, the procalcitonin (PCT) level measured in serum is used in the treatment decisions regarding antibiotic use.

The S3 guidelines recommends the following medication (Table 10 [Tab. 10]):

Antibiotic therapy is recommended (level B) for:

  • moderately severe AECOPD (indication for hospitalization): antimicrobial therapy only if Stockley II (purulent sputum)
    *if PCT levels are available and the measured value is <0.1 ng/ml, antimicrobial treatment may be forgone.
  • severe AECOPD (indication for intensive care): antimicrobial therapy is always indicated.

Pneumonia

Pneumonia is diagnosed in patients with new or increasing infiltrates in chest x-ray and the following clinical signs:

  • body temperature >38°C (or occasionally <36°C) and/or
  • leukocytosis (>10/μl) and/or
  • leftward shift (>5%) and/or
  • CRP>5 mg/dl

and at least 2 of the following symptoms:

  • productive cough
  • purulent sputum
  • dyspnea, trachypnea
  • chills
  • fine crackles on auscultation
  • thoracic pain when breathing

Classification of pneumonias is made according to the recommendations of the American Thoracic Society (ATS) and the Infectious Diseases Society of America (IDSA), taking into consideration whether the disease was community-acquired or hospital-acquired.

For community-acquired pneumonias – depending on age, risk factors, severity, and course of illness – four patient groups could be identified whose disease may be attributed with high probability to the pathogens typical for the respective groups.

The classification of nosocomial pneumonias into three groups with different treatment strategies based on risk assessment using a clinical score follows the recommendations of the PEG and DGP from 2003. Time of onset of pneumonia after hospital admission, need for ventilator support, age of the patient, comorbidities, and previous antiinfective therapy are assessed.

Supportive measures in the management of pneumonia include adequate intake of fluids (>2 L per day), appropriate antipyretics, oxygen support for hypoxia, treatment of bronchial obstruction, and in some cases the administration of glucocorticoids. All patients should receive prophylaxis against thromboembolism.

Community-acquired pneumonia

CRB-65 Score

Test for the presence of the following criteria:

  • confusion
  • respiratory rate (≥30/min)
  • diastolic blood pressure ≤60 mmHg / systolic blood pressure <90 mmHg
  • age (≥65 years)

The score is calculated by adding one point for the presence of each of the listed items.

These recommendations apply to moderate to severe CAP since mild CAP should be treated orally.

  • Moderately severe community-acquired pneumonia: management in general wards (hospitalized CAP): clinical decision using the CRB-65 Score
  • Severe community-acquired pneumonia: management in a monitoring ward (intensive care, intermediate care, etc.) (severe CAP): clinical decision using the CRB-65 score
    • without indication for empirical treatment effective against P. aeruginosa (see risk factors for P. aeruginosa)
    • with indication for empirical treatment effective against P. aeruginosa (see risk factors for P. aeruginosa)
Pathogen spectrum of hospitalized CAP patients

The most frequent pathogens are S. pneumoniae, M. pneumoniae, H. influenzae, gram-negative enterobacteriaceae and respiratory viruses. The frequency of L. pneumophila varies across regions, it may be as high as 6%. Enterobacteriaceae were shown to be somewhat more frequent than in CAP patients treated as out-patients.

According to the latest data from CAPNETZ, P. aeruginosa has a minor role as a CAP pathogen in Germany. Therefore for CAP treated on general wards, Pseudomonas coverage in initial treatment is necessary only in patients with risk factors (see risk factors for P. aeruginosa).

Risk factors for P. aeruginosa
  • Severe chronic structural lung diseases such as severe COPD with previous antibiotic treatment or previous hospitalization, both within the last 3 months
  • known colonization by P. aeruginosa
  • bronchiectases
  • cystic fibrosis
Treatment of hospitalized CAP patients

Antimicrobial therapy should be started as early as possible (recommendation level B). A delay of treatment initiation by ≥8 hours after hospital admission is associated with higher mortality. Diagnostic measures must not delay the start of treatment.

After parenteral initial therapy, an early switch to an oral treatment is appropriate if the following requirements are fulfilled (recommendation level A):

  • heart rate ≤100/min
  • respiratory rate ≤24/min
  • systolic blood pressure ≥90 mm Hg
  • body temperature ≤37.8°C
  • ability to tolerate oral nutrition
  • normal level of consciousness
  • no hypoxemia (pO2≥60 mm Hg or SaO2≥90%) and
  • safe oral intake of medication
Duration of treatment

The S3 guidelines recommend (recommendation level A) (Table 11 [Tab. 11]): antibiotic treatment may be stopped 48 to 72 hours after clinical improvement with reduction in body temperature, but not before completion of 5 days of therapy. A duration of treatment of more than 7 days is not generally required. A treatment duration of 8 to 15 days is recommended for proven infections with P. aeruginosa.

Appropriate reasons for a shorter treatment (<8 days) include:

  • improvement of general health status
  • ability to tolerate oral nutrition
  • improvement of respiratory symptoms
  • body temperature <38°C

Management of severe community-acquired pneumonia (sCAP)

  • Indication for intensified monitoring (depending on facilities, intensive care unit, intermediate care unit or intensified monitoring on a general ward): patients with acute lower respiratory tract infection with or without positive local auscultatory findings, an infiltrate in chest x-ray and
    • 1 major criterion of the modified ATS score
    • ≥2 minor criteria of the modified ATS score or
    • CRB-65 index ≥2

In particular cases one minor criterion of the modified ATS score or a CRB-65 index of 1 can also be sufficient to warrant intensified monitoring. Thorough clinical evaluation of CAP severity is essential for decision on intensive care.

Modified ATS criteria for severe pneumonia (sCAP): major criteria, determined at admission or later (positive if 1 or 2 factors are present)

1.
Need for intubation and mechanical ventilation
2.
Need for vasopressor use >4 hours (septic shock)

Minor criteria, determined at admission (positive if 2 or 3 factors are present)

1.
Severe respiratory insufficiency (PaO2/FiO2<250)
2.
Multilobar infiltrates in chest x-ray
3.
Systolic blood pressure <90mm Hg
Pathogen spectrum in sCAP

The etiology of sCAP differs from less severe CAP forms by a wider pathogen spectrum. Approximately 10% of sCAPs are polymicrobial infections.

Determination of a potential involvement of P. aeruginosa is important for differential therapy.

Patients with sCAP are categorized in two risk groups:

  • Patients with sCAP without risk factors for P. aeruginosa infection
  • Patients with sCAP with risk factors for P. aeruginosa infection

CAP caused by P. aeruginosa occurs almost exclusively in patients with particular risk factors.

Treatment of severe community-acquired pneumonia (sCAP)
Calculated initial therapy

The risk of unfavourable outcomes due to inadequate therapy of infections with resistant pathogens is particularly high in sCAP. Considering the latest resistance data is crucial.

The treatment of choice for sCAP without risk factors for P. aeruginosa involvement, according to S3 guidelines (recommendation level B) is a combination of broad-spectrum beta-lactam antibiotic (cefotaxim, ceftriaxon, piperacillin/tazobactam, or ertapenem) and a macrolide (Table 12 [Tab. 12]). Monotherapy with a fluoroquinolone covering pneumococci (levofloxacin or moxifloxacin) is a potential alternative, but should be limited to patients without septic shock or invasive ventilation.

For patients with indication for empirical therapy covering P. aeruginosa, the S3 guidelines (Table 13 [Tab. 13]) recommend a combination of piperacillin/tazobactam, cefepim, imipenem or meropenem and a fluoroquinolone covering pseudomonas (levofloxacin or ciprofloxacin) or an aminoglycoside plus a macrolide. An important differential criterion is previous therapy with an antibiotic necessitating a switch of drug class. Ceftazidim is active against P. aeruginosa but – unlike cefepim – inadequately effective against S. pneumoniae and S. aureus. As a rule, the treatment should be changed to monotherapy after clinical improvement and/or pathogen detection with a susceptibility test.

Duration of therapy

The S3 guidelines recommend (level B) a treatment duration of 8 to 10 days for patients without complications or 5 days after defervescence. In patients with proven P. aeruginosa infections, treatment should be continued for 15 days. Prolonged treatment courses are necessary for sCAP caused by S. aureus as well.

Nosocomial pneumonia

Nosocomial pneumonia is a hospital-acquired lung infection with onset within three days after admission to seven days after discharge of the patient. In the USA and Europe, pneumonia is the second most frequent nosocomial infection. It is the most common infection in intensive care medicine. With mortality rates of 30–50% it is the most frequently lethal hospital-acquired infection. Particularly infections with multiresistant bacteria have a poor prognosis. Early and effective treatment of nosocomial pneumonia is essential in reducing morbidity and mortality [2], [81], [106].

Risk factors predisposing patients for nosocomial pneumonia include:

  • higher age (>65 years)
  • previous antibiotic treatment
  • immunosuppression
  • coma
  • prolonged intubation and ventilation
  • organ failure and septic shock
  • preexisting respiratory disease
  • thoracic or abdominal surgery
  • severe trauma
  • smoking
  • alcohol abuse
  • drug abuse

All patients are colonized with possible pathogens in the oropharynx, have an impaired immune function and potential aspirations caused by diminished protective larynx reflexes.

The choice of initial antimicrobial therapy is based on the classification of patients into defined groups with characteristic pathogen spectra and defined treatment recommendations. Each of these three groups has its particular risk profile resulting in an overall risk score. The individual factors are weighted with 1 to 4 points (Table 14 [Tab. 14]). Spontaneous breathing and artificial ventilation or severe respiratory insufficiency with early (until day 4) or late (day 5 or later) onset after the occurrence of pneumonia, age of the patient and other risk factors including previous antiinfective therapy, structural lung diseases or extrapulmonary organ failure are considered as well. The individual risk factors have divergent degrees of influence on the severity of illness and the expected pathogen spectrum.

The recommendations are based on evidence of variable quality and often reflect expert opinions. This procedure is presently investigated in a retrospective chart review in several large hospitals in Germany. The recommendation to treat patients with nosocomial pneumonia according to a risk-based point system compiled in 2003 at a joint PEG and DGP consensus conference on nosocomial pneumonia remains controversial. At the current consensus conference, 34% of the participants rejected this recommendation.

Some of the listed antibiotics are not licensed for the respective indication but are recommended on the basis of the available evidence. In group 3, the combination with a fluoroquinolone has evidence level IV.

The adequacy of monotherapy is independent of disease severity. Monotherapy is indicated in pneumonia with onset within the first four days of hospitalization in the absence of risk factors. Multiresistant pathogens are less frequently found in spontaneously breathing patients.

The superiority of combination therapies for late-onset pneumonia and/or patients with risk factors is not reliably established. Combination therapy is still recommended in cases with suspected Pseudomonas involvement [442], [530], [562] and in patients with ventilator-associated pneumonia [126], [262], [442], [522]. The rationales of combination therapy include the extension of the antibacterial spectrum and the exploitation of possible synergies. After 2 to 3 days of treatment, the initial combination therapy should be evaluated [428], [442], [522], [542]. A decisive factor for treatment success is the early start of adequate antimicrobial therapy [74], [276] at a sufficient dosage. The duration of therapy should be not more than 8 to 10 days [101], [222], [562]. Pneumonias caused by S. aureus or P. aeruginosa require longer treatment duration [14], [224], [523], [530].

Initial calculated therapy group I: spontaneously breathing patients with ≤2 risk points

The pathogen spectrum of this patient group largely corresponds to the endogenous flora of the upper respiratory tract, that patient brought in from his environment. This includes S. pneumoniae, methicillin-susceptible S. aureus, H. influenzae and other gram-negative pathogens. Multiresistant bacteria are rarely present. Pneumonias occurring later than the fifths day after admission are primarily caused by gram-negative enterobacteria.

Group 2 cephalosporins or aminopenicillins/BLI are sufficient to treat mild pneumonias detected before the fifth day after admission. In severe cases, group 3a cephalosporins are preferred. Group 3 or 4 fluoroquinolones or group 2 carbapenems could also be used; they are not yet licensed in Germany for the treatment of nosocomial pneumonias but are currently being tested in clinical studies.

The initial treatment should be administered parenterally. It may be changed to an oral therapy after clinical improvement. If gastrointestinal absorption is intact, fluoroquinolones may be initiated orally if patient compliance is ensured.

Initial calculated therapy group II: non-ventilated patients with risk factors or patients on mechanical ventilation without risk factors (patients with 3 to 5 risk points)

In this patient group, an increased incidence of Enterobacter spp., Serratia spp., Citrobacter spp., Pseudomonas spp., Acinetobacter spp., S. maltophilia and anaerobes must be anticipated besides the above mentioned pathogens. Therefore, antibiotics covering these pathogens in their spectra should be used for treatment. Available options include group 4 cephalosporins, acylaminopenicillins/BLI and group 1 carbapenems. Because of the resistance situation and the undesirable microbiological side effects (collateral damage) in terms of selection of antibiotic-resistant pathogens and colonization or infection with multiresistant pathogens, group 3 cephalosporins and fluoroquinolones should not be used for these patients [78], [372], [399], [566]. In the case of cephalosporins this means an increased occurrence of vancomycin-resistant enterococci (VRE), ESBL-producing enterobacteriaceae and beta-lactam antibiotic-resistant Acinetobacter spp. [78], [399]. When using fluoroquinolones, colonization by MRSA and the resistance situation of E. coli and P. aeruginosa must be taken into consideration [372], [566].

Initial calculated therapy group III: patients with high risk, usually on mechanical ventilation (patients with ≥6 risk points)

In these patients, pneumonias are often caused by multiresistant pathogens. For this reason a combination treatment should be initiated [126], [262], [442], [522], [530], [562]. The options include group 3b or 4 cephalosporins, acylaminopenicillins/BLI or group 1 carbapenems in combination with an aminoglycoside (once daily), high-dose group 2 or 3 fluoroquinolones, or fosfomycin. A group 2 or 3 fluoroquinolone or fosfomycin should be preferred as combination partner for beta-lactam antibiotics [442], [530], [571].

MRSA pneumonias

From the clinical point of view, there are no substances available for the treatment of MRSA pneumonias which have been tested in clinical studies except for linezolid and the glycopeptides. Linezolid showed statistically significant advantage in a post hoc analysis of two prospective studies [582] but was not superior to vancomycin in the primary endpoint in another clinical trial [581].

The critical disadvantage of vancomycin is its poor penetration into the lungs (11% of the plasma level), which could theoretically be partially compensated by combination with MRSA-effective substances (fosfomycin, rifampicin) with good tissue penetration. However, these combinations were not tested in randomized clinical trials.

Linezolid is therefore the preferred choice for pulmonary MRSA infections. Because linezolid, like vancomycin, is active against gram-positive pathogens only, it should be used in monotherapy only if the involvement of gram-negative or atypical pathogens has been ruled out.

Aspiration pneumonia and pulmonary abscess

Aspiration pneumonias are subdivided into insidious recurrent aspirations and acute aspirations of stomach contents.

  • Pathogen identification is difficult.
  • Polymicrobial aetiology is common (aerobic and anaerobic pathogens).
  • Gram-positive pathogens are more likely in cases with community-acquired aspiration.
  • Gram-negative or polymicrobial infections, some involving anaerobes, are common in patients with frequent hospitalization and antimicrobial therapies.

The pathogenesis of primary lung abscesses involves aspiration, virulence of the pathogen and immune impairment of the patient. Risk factors for aspiration include

  • previous CNS diseases,
  • intoxication,
  • difficulty of swallowing and/or
  • oesophageal pathologies.

Secondary lung abscesses are due to

  • bronchial obstruction by tumors,
  • bronchial obstruction by foreign bodies and poststenotic pneumonia,
  • colliquation,
  • super infections of infarct pneumonias,
  • rarely bacteremia.

Bacterial mixed infections predominate; obligate anaerobes were detected in 20% to 90%. In a German study, S. aureus was found to be the most frequent pathogen in aspiration pneumonias and lung abscesses [389].

Previous aspiration is a risk factor for the involvement of enterobacteriaceae. As an additional aetiological role of anaerobic bacteriae in aspiration pneumonia is possible and as a large number of anaerobes produce beta-lactamases, penicillins should be combined with a beta-lactamase inhibitor. Alternatively, a combination of a group 3a cephalosporin (cefotaxim, ceftriaxon) with clindamycin, a group 4 fluoroquinolone (moxifloxacin) or a group 2 carbapenem (ertapenem) may be used.

Pleural infections

There is little proven data on calculated treatment of pleural infections. The evidence is based primarily on retrospective investigations and expert opinions.

Pleuritis sicca is caused mainly by viruses, Chlamydia spp. or Mycoplasma spp. Therefore, macrolides or group 3 and 4 fluoroquinolones (evidence level IV) are used as calculated treatments.

The primary goals in treatment of parapneumonic effusions include control of the infection, drainage of infected effusion, (re-)expansion of the lungs and avoidance of pleural calluses.

Basic therapy includes sufficient, pathogen-specific antimicrobial treatment aiming at the control of the underlying infection. There are no controlled clinical trials on the optimum antibiotic treatment and its duration. The calculated antimicrobial treatment should cover gram-positive cocci, gram-negative pathogens (where applicable including P. aeruginosa) and anaerobes. Initially, parenteral administration is preferred to ensure sufficient plasma and intrapleural concentrations. Basically, treatment should be continued at least until the infected effusion has been completely drained. Long treatment durations of several weeks are often necessary.

A basic requirement in the treatment of complicated parapneumonic effusions is the effective and complete drainage of infected fluid. The following differentiated approach is suggested:

1.
No intervention
2.
Thoracocentesis
3.
Establishment of a thoracic suction drainage without local fibrinolysis
4.
Establishment of a thoracic suction drainage with local fibrinolysis
5.
Video-assisted thoracoscopy with post-intervenional thoracic suction drainage
6.
Surgical exploration (thoracotomy) with or without decortication and/or rib resection

If drainage is insufficient to rapidly remove the infected pleural fluid, video-assisted thoracoscopy (VATS) in a preferred option as well as a limited attempt of local fibrinolysis if appropriate.

Risk stratification
1.
Size of effusion
A 0: Minimal effusion (<5 cm in a lateral x-ray) or thorax sonography
A 1: Medium-large, freely discharging effusion (>5 cm and <½ hemithorax)
A 2: Large, freely discharging effusion (>½ hemithorax), chambered effusion or effusion with thickened pleura
2.
Bacteriology of exsudate
B x: Gram stain or culture not known
B 0: Negative culture and gram stain
B 1: Positive culture or gram stain
B 2: Pus
3.
Clinical chemistry of exsudate
C x: pH unknown
C 0: pH >7.2
C 1: pH <7.2

Additional patient risk due to pleural effusion is estimated using the following parameters: prolonged hospitalization, increased morbidity due to further interventions, prolonged physical disability, increased risk of a respiratory impairment, increased risk of local inflammation, mortality.

Treatment recommendation (recommendation B)

Category 1 (very low risk): presence of A 0, B x or C x: no drainage

Category 2 (low risk): presence of A 1, B 0 or C 0: no drainage

Category 3 (medium risk): presence of A 2, B 1 or C 1: drainage indicated

Category 4 (high risk): presence of B 2: drainage indicated

For patients of categories 3 and 4, a relief puncture appears to be insufficient in most cases. Therefore, thoracic drainage is indicated. In the case of chambered effusions, or category 4 patients, local fibrinolysis or video-assisted thoracoscopy is the most appropriate approach.


6 Infections in the ear, nose and throat or mouth, jaw and facial regions

Bilal Al-Nawas, Cordula Lebert, Pierre Federspil, Pramod M. Shah, Wolfgang Elies

In daily practice, bacterial infections in the head and throat region often require the use of antibiotics. The decision for an intravenous treatment depends on the severity of the infection and the individual prerequisites for administration. However mastoiditis, malignant otitis externa, sinusitis with orbital and other complications, epiglottitis, severe odontogenic abscesses and cervical phlegmons should generally be treated with intravenous antibiotics. Even if initial treatments are mostly calculated therapies, microbiological pathogen identification is mandatory for these severe diseases. Switching to an oral preparation after the initial intravenous treatment phase is possible in some cases. Recently introduced antibiotics have extended the range of indications for oral treatments. Specifically, fluoroquinolones may be administered orally for some severe infections. If possible, oral therapy should be preferred for economic reasons. As a matter of principle, any antibiotic therapy should be reassessed after 3 or 4 days. Interestingly, despite the high incidence of these infections, systematic investigations of the pathogen spectrum have been performed only rarely. Very few randomized comparative studies with large patients numbers that may support the present recommendations are available. Current expert recommendations of medical societies for antimicrobial treatment of head and throat infections were considered in the following recommendations (Table 15 [Tab. 15]) [8], [10], [167].

Malignant otitis externa

Malignant otitis externa is a very rare disease predominantly caused by Pseudomonas aeruginosa. In rare cases other pathogens might be involved [32], [100], [287], [485]. The infection usually affects older, male diabetes mellitus patients [214] and in rare cases patients with immunosuppression [16], [477]. The infection may spread to adjacent bone structures and requires surgical debridement of the affected area.

The disease may be lethal if left untreated. Therefore therapy must be initaited immediately upon admission to the hospital with a high-dosage intravenous antibiotic, e.g. from the group 2 fluoroquinolones (ciproflaxacin), ureidopenicillins (preferrably piperacillin), group 3b or group 4 cephalosporins (ceftazidime or cefepime, respectively), group 1 carbapenems (doripenem, imipenem/cilastatin, meropenem) or group 3 fluoroquinolones (levofloxacin) [56], [60]. In cases with insufficient response, treatment should be switched to a beta-lactam combined with an aminoglycoside.

Treatment duration should be approximately 6 weeks. If an aminoglycoside (e.g. tobramycin) is used due to a high-resistance situation, serum levels must be monitored due to the ototoxic and nephrotoxic potential of this group of drugs. A once-daily regimen resulting in high peak and low trough levels is preferred (see chapter 3).

Mastoiditis

Mastoiditis is a common complication of acute or chronic otitis media. The pathogen that caused the primary infection should be suspected as the cause of mastoiditis. The most commonly involved pathogens in order of frequency are Streptococcus pneumoniae, Streptococcus pyogenes, Haemophilus influenzae, Staphylococcus aureus, P. aeruginosa, Escherichia coli and Proteus mirabilis [167], [336].

Surgical debridement is strictly required. The initial calculated therapy should be started with an aminopenicillin/beta-lactamase inhibitor (BLI) combination (ampicillin/sulbactam, amoxicillin/clavulanic acid) or a cephalosporin from group 2 (cefuroxim, cefotiam), 3a (cefotaxim, ceftriaxon) or 4 (cefepim). Alternatively, group 3 or 4 fluoroquinolones (levofloxacin or moxifloxacin, respectively), ureidopenicillins/BLI (piperacillin/BLI, mezlocillin/BLI) or group 2 carbapenems (ertapenem) may be used.

Pathogens should be identified from samples taken intraoperatively if possible. Based on the microbiology results, the antimicrobial treatment may be adjusted to obtain better pathogen targeting. The treatment duration is about 1 week.

Frontal bone osteomyelitis

Frontal bone osteomyelitis is an infection of the frontal bone that spreads from acute or chronic sinusitis or dental infections of the upper jaw per continuitatem. It mostly affects adolescents. Surgical debridement of the affected frontal sinus and removal of the infected part of the bone is strictly indicated. The most common pathogens are S. aureus, S. pneumoniae, H. influenzae and P. aeruginosa.

Antimicrobial therapy initially consists of high-dose ureidopenicillin/BLI or aminopenicillin/BLI (piperacillin/tazobactam, piperacillin + sulbactam, mezlocillin + sulbactam, amoxicillin/clavulanic acid, ampicillin/sulbactam) or a carbapenem (doripenem, imipenem/cilastatin, meropenem, ertapenem). If cefotaxim or ceftriaxone is used, it should preferably be combined with clindamycin or metronidazol.

In severe cases, beta-lactam antibiotics are combined with an aminoglycoside or fluoroquinolone. Samples obtained by sinus puncture, surgical drainage fluid and blood cultures should be examined microbiologically. After pathogen indentification the treatment should be adjusted to obtain more specific targeting. Treatment duration should be approximately 6 weeks.

Orbital phlegmon

Orbital phlegmon is an acute infection of the soft tissue of the ocular orbit. It usually results from an infection spreading from the paranasal sinuses. Occasionally, the root cause are odontogenic or intracranial infections. In rare cases trauma, surgery or dacryocystitis may also be the causative event. Because of a high risk of complications in terms of loss of vision or intracranial spread of infection, surgical intervention is strictly required. Potential pathogens include S. aureus, S. pneumoniae, H. influenzae, M. catarrhalis, K. pneumoniae, P. aeruginosa and anaerobes. Fungi are detected less frequently. Polymicrobial infections are prevalent.

Antibiotic therapy should be initiated intravenously. High-dose ureidopenicillins/BLI or aminopenicillins/BLI (piperacillin/tazobactam, piperacillin + sulbactam, mezlocillin + sulbactam, amoxicillin/clavulanic acid, ampicillin/sulbactam) or carbapenems (doripenem, imipenem/cilastatin, meropenem, ertapenem) are recommended. If cefotaxim or ceftriaxone is used, it should be combined with clindamycin or metronidazol if possible.

In severe cases, an aminoglycoside or fluoroquinolone (ciprofloxacin, levofloxacin) is added to the basic treatment. A switch to oral moxifloxacin is a feasible option for sinusitis with orbital involvement. Samples obtained by sinus puncture, surgical drainage fluid and blood cultures should be examined microbiologically. After pathogen identification the treatment should be adjusted to obtain more specific targeting. The recommended treatment duration is 14 days.

Epiglottitis

Acute epiglottitis occurs mainly in children and is quite rare in adults. Usually it is an acute and severe rapidly progressing disease. Due to the imminent risk of an airway obstruction, it must immediately be treated in a hospital intensive care unit with the option of rapid intubation or tracheotomy.

In adults, potential pathogens are S. pyogenes, H. influenzae Type B, S. aureus, and S. pneumoniae, occasionally Haemophilus parainfluenzae or viruses. Group 3a cephalosporins (cefotaxim, ceftriaxon), aminopenicillins/BLI (amoxicillin/clavulanic acid, ampicillin/sulbactam) or – if S. aureus is detected – group 2 cephalsporins (cefuroxim, cefotiam) are recommended for calculated therapy. The incidence of epiglottitis has declined in children living in Western countries due to widespread vaccination against Haemophilus influenzae B [205], [285].

Perichondritis

This infection usually results from an earlier injury. In the ear region, common pathogens are P. aeruginosa and S. aureus but other pathogens may be involved as well, depending on the type of trauma. Samples should be obtained for microbiological diagnosis. The antibiotic treatment should be as targeted as possible based on the microbiological results. Hospitalization may be required in severe cases. Options for initial calculated therapy include intravenous piperacillin/BLI, cefepim, imipenem/cilastatin or meropenem. Alternatively ciprofloxacin, ceftazidime or levofloxacin may be used if the involvement of Pseudomonas is strongly suspected.

Nasal furuncles

Nasal furuncles are painful deep infections of the hair follicles in the nasal vestibulum with a phlegmonous spread towards the tip of the nose, nasal septum, upper lip or along the root of the nose. It is caused by S. aureus.

Out-patient therapy of mild forms is feasible. Hospitalization is necessary for complicated forms of disease which may require surgery (incision of the furuncle, drainage, ligation and transection of the angular vein) to avoid the risk of thrombosis with sepsis spreading to the cavernous sinus.

The antibiotic treatment should involve anti-staphylococcal, penicillinase-stable beta-lactams such as flucloxacillin, dicloxacillin, cefaclor, ampicillin/sulbactam or amoxicillin/clavulanic acid. Treatment should be administered intravenously if necessary (flucloxacillin, oxacillin, cefazoline).

Peritonsillar abscess

Peritonsillar abscesses occur predominantly in young adults [176], [216], [246] and rarely in children. The infection is almost exclusively unilateral. As a rule it stems from recurrent or acute exacerbated tonsillitis and blocked drainage of pus due to formation of scar tissue. Aerobic/anaerobic mixed infections are often present. Beta-haemolytic streptococci, staphylococci, Prevotella spp, Fusobacterium necrophorum [188], [209], [216], [444], other anaerobes and gram-negative aerobic pathogens [331] have been detected.

Abscesses require lancing due to the risk of spreading with formation of distant septic metastasis and a high risk of complications. In addition, antibiotic treatment is required. Out-patient treatment of mild cases is possible but hospitalization is required for severe cases or abscess tonsillectomy.

Antibiotic treatment is required in the perioperative and postoperative phase. It involves initial intravenous application of a group 1 or 2 cephalosporin (cefazolin, cefuroxim, cefotiam) in combination with clindamycin. Other options include aminopenicillins/BLI (ampicillin/sulbactam, amoxicillin/clavulanic acid), erythromycin, clarithromycin or azithromycin. A switch to a usual oral therapy used for tonsillitis (phenoxymethylpenicillin, cephalosporins, macrolides) may be feasible on the second or third day of therapy.

Chronic purulent sinusitis

Chronic sinusitis results from an incompletely cured acute sinusitis. It persists for more than 8 weeks or more than 4 episodes per year with residual symptoms. Potential causes include gradual obstruction of the sinus by increased tissue formation in the osseous parts, disruption of the mucociliary function and impaired drainage of secretions. In a substantial number of cases, odontogenic infections usually spreading from the root tips of the upper molars are responsible. Basically, it is necessary to remove the focal source of infection after the critical phase of disease has been overcome.

There are two forms of chronic sinusitis. The most frequent form is polypous-serous sinusitis. The mucous-purulent chronic sinusitis occurs less often and, as a rule, should be treated with oral antibiotics (and nasal decongestants). The most frequent pathogens are not those encountered in acute sinusitis but rather S. aureus, Streptococci, H. influenzae, various enterobacteriaceae, less often P. aeruginosa, anaerobes and in rare cases Aspergillus spp. [167]. Inhibitor-protected aminopenicillins (amoxicillin/clavulanic acid, sultamicillin), oral cephalosporins (cefuroxim axetil, loracarbef, cefpodoxim proxetil) or levofloxacin and moxifloxacin [546] are recommended for calculated oral antibiotic therapy. The spectrum of efficacy should include anaerobes if there is an odontogenic source of infection. Treatment may be required for up to 3 weeks. Intravenous antibiotic therapy is needed only in specific cases.

Odontogenic abscess/throat phlegmone

There are no systematic data on the resistance situation or the development of resistance in dental medicine, as most odontogenic infections can be treated orally on an outpatient basis. For uncomplicated odontogenic infections, identification of pathogens is not required in dental practices. However, it is essential to isolate the pathogens of parenterally treated severe odontogenic infections with the risk of spreading to adjacent tissues. Typical pathogens, also found in closed abscesses, include oral streptococci and less frequently staphylococci. Anaerobes or capnophilic pathogens often include Prevotella spp., Fusobacteria, Bacteroides spp., Veillonella and Peptostreptococci [7].

According to available date, pathogens from previously untreated abscesses have low resistance rates for penicillin and clindamycin [146], [147], [339]. On the other hand, beta-lactamase-producing pathogens were reported in about 15% to 35% of cases, particularly in complicated, previously treated abscesses which required parenteral treatment [152], [280], [412], [484], as well as sometimes critical rates of resistance against clindamycin in 25% to 45% of cases [11], [280], [412], [484]. It appears that particularly pretreatment with antibiotics results in an increase in penicillin-resistant pathogens [9], [281]. In severe odontogenic soft tissue infections, which typically have already been pretreated, higher resistance rates against penicillin and clindamycin are expected [148]. As indicated by the data discussed above, inhibitor-protected penicillins (e.g. amoxicillin/clavulanic acid) are almost universally effective against the relevant pathogens [411]. However, when judging resistance, it should be considered that the pathogenic role of the detected bacteria is not clear [390]. In life-threatening situations, carbapenems are the drugs of choice in empirical regimens [279], [412], [449]. Alternatively, metronidazole can be used in combination with a fluoroquinolone [9], [152], [280]. In the presence of allergies to beta-lacams, clindamycin as monotherapy with the above-mentioned limitations, is the established alternative. New data suggest that moxifloxacin is a potential treatment option as well [11], [565].

Osteomyelitis

The most important form is acute and secondary chronic osteomyelitis caused by bacteria (odontogenic infection, pulpal and parodontal infection, infected extraction wounds) with pus discharge, fistulation and sequestration. The pathogens are similar to those encountered in odontogenic polymicrobial abscesses [323], [413]. Colonization and infection with multiresistant gram-positive pathogens are reported, particularly after longer antibiotic pretreatment [19], [529]. Osteomyelitis is treated with a combination of surgery and antibiotics [323]. The less frequent primary chronic osteomyelitis is differentiated as a non-purulent chronic inflammation of unclear origin [323]. Special forms of inflammation in previously irradiated bones such as infected osteoradionecrosis or osteomyelitides induced by medications such as bisphosphonates [563], glucocorticoids and antineoplasic substances are not considered to be caused primarily by bacteria. Bacterial superinfections require targeted adjuvant antibiotic treatment.

The primary goal of osteomyelitis treatment is the eradication of the focus by resection of infected and necrotic bone combined with an initially empirical antibiotic treatment which is best followed by pathogen-specific antimicrobial therapy. Because of the protracted course of the disease parenteral treatment is usually necessary. For many years, locally implanted gentamicin-releasing polymethyl methacrylate (PMMA) chains have been successfully used, particularly for chronic disease [168], [202]. Any adjuvant antibiotic treatment should cover the anaerobic pathogen spectrum as well as staphylococci which are often isolated [265]. Clindamycin or penicillin are recommended for antimicrobial therapy in pretreated patients. However, the above-described limitations apply [478], as penicillin resistance is often found after previous therapy [413]. Due to the potentially long and critical course of the disease, pathogen identification should be attempted in all cases. Some authors recommend that antibiotic therapy should be administered for 4 to 6 weeks after surgery [34].

Acute necrotizing gingivitis and tonsillitis

The acute form of necrotizing gingivitis (ANUG) is often associated with other viral diseases and evolves into the disfiguring disease noma in malnourished patients [166]. Spirochetes typically appearing in ANUG can be controlled in most cases with local disinfection measures such as hydrogen peroxide or chlorhexidine mouthwash. In severe cases, additional adjuvant antibiotic treatment is recommended. Intravenous therapy is often required due to the severity of symptoms [257], [422]. The acute course of the disease and problems caused by culture evidence of spirochete involvement often means that classical pathogen identification is not reasonable. Mixed infections with fusobacteria are also reported [197], [415], [451]. Penicillin V in combination with metronidazole is the treatment of choice [478]. In allergy cases, clindamycin can also be used as an alternative. However, no comparative studies are available for these medications.

Cervicofacial actinomycosis

As a usually mixed infection with the major pathogen being Actinomyces israelii, this disease is treated successfully with antibiotics [483]. Depending on the severity of the findings, an additional surgical intervention may be necessary. Microbiological or at least histopathological examination of the Actinomyces plaques is important. Actinomycetes are typically susceptible to penicillin. In cases with allergy, doxycycline, clindamycin or a cephalosporin are recommended [478], [483]. The necessity of covering the accompanying obligatory anaerobes remains controversial [457], [483]. As with other chronic infections, high-dose long-term therapy is required due to the poor penetration into granulation tissue [478]. There is no exact data on the optimum treatment duration for the cervicofacial form. Treatment of up to 6 months is considered for complicated forms. In cases with less severe disease or adequate surgical treatment, recommended treatment durations of about 6 weeks are found in the literature [320], [483].

Sialadenitis

Sialadenitis is a bacterial or viral infection of the salivary glands. A sialadenitis often occurs as a superinfection following functional impairment of the salivary glands. The submandibular glands are most often affected. Secretory dysfunction of the salivary and mucous glands causes increased viscosity of the saliva, which promotes the precipitation of inorganic substances. The resulting sialoliths may promote bacterial colonization and infection. They should be removed during the chronic phase of disease. There are acute and chronic forms of the disease. The predominant pathogens are viruses (usually mumps virus, parainfluenza viruses or CMV), and in adults staphylococci and streptococci but also anaerobes. A recent publication reports evidence of an increased incidence of infections due to Fusobacterium necrophorum (14%), particularly in the presence of peritonsillar abscesses (91%) [209], [444]. F. necrophorum may cause the Lemierre syndrome as a severe complication. In the acute phase a conservative treatment is preferred in most cases. Because of the high prevalence of systemic symptoms, intravenous therapy or surgical release is often necessary, which generally requires hospitalization. Severe bacterial infections must be treated with intravenous antibiotics while oral treatment is also acceptable for less severe infections. In older studies, a causal role of streptococci and staphylococci is reported [286]. More recent work has drawn attention to the major importance of anaerobes in purulent sialadenitis [299]. Due to frequent penicillin resistance of the pathogens use of an aminopenicillin/BLI or clindamycin is recommended [167]. There are published recommendations to use cephalosporins [167]. This group of drugs, however, is ineffective against anaerobes.


7 Intraabdominal Infections

Christian Eckmann, Rainer Isenmann, Peter Kujath, Annette Pross, Arne C. Rodloff, Franz-Josef Schmitz, Matthias Trautmann, Hannes Wacha

Indications for Antimicrobial Treatment and Duration of Treatment

Intraabdominal infections (IAI) are rather common. In Germany about 150,000 patients per year are treated for an IAI [30]. National and international databases show that every fourth case of severe sepsis or septic shock can be attributed to IAI [159], [276]. Almost 90% of all intraabdominal infections require primarily a surgical intervention to control the source of infection (e.g. closure of gastric perforation). Nevertheless, the value of antibiotic therapy versus placebo is significant in this group of diseases [579]. Inadequate initial antibiotic treatment of IAI is associated with substantially worse outcomes and results in considerable increased cost [37], [118], [150], [364].

Recommendations on antibiotic therapy of intraabdominal infections are derived from numerous prospective randomized controlled studies. As the objective of most studies is the demonstration of therapeutic equivalence, the available evidence is not sufficient to prefer any specific regimen over another [579]. When choosing the appropriate antibiotic, individual patient characteristics (e.g. immunosuppression, previous therapy), anticipated pathogen spectrum, local pathogen and resistance situation, appropriate mode of administration, low toxicity and cost should be included in the decision.

No reliable data are available on the appropriate duration of treatment for intraabdominal infections [274]. Treatment durations given below are based on durations used in randomized studies, the peculiarities of special pathogens (e.g. Candida spp.), the local and systemic severity of infection and the experience of the authors. In general, discontinuation of the antibiotic should be considered if clinical condition and infection parameters are significantly improved. If treatment success is not achieved after 10 days, antimicrobial treatment should preferably be discontinued and new samples taken for susceptibility testing rather than continuing a regimen that may select resistant pathogens and/or unnecessarily expose the patient to potential toxicities.

Systematically, three forms of peritonitis with different pathogenesis, pathogen spectrum, and requirement of surgical and/or antimicrobial therapy can be differentiated [274].

Primary peritonitis

Primary (i.e. spontaneous bacterial) peritonitis (SBP) accounts for approximately 1% of all peritonitis cases. Juvenile peritonitis is a hematogeneous infection caused by streptococci, pneumococci, or rarely Haemophilus influenzae. In adults, primary peritonitis mostly affects patients with ascites caused by hepatic cirrhosis (approximately 70%) or immune impairment (approximately 30%) [171], [432]. Usually it is a monobacterial infection. In routine practice, the pathogen is identified in only about 35% of the cases, where Escherichia coli, Klebsiella spp., staphylococci, enterococci or streptococci, and sometimes pathogenic intestinal bacteria such as Aeromonas spp. or Salmonella spp. are isolated [171]. Primary peritonitis associated with tuberculosis is caused by hematogeneous dissemination.

Only few randomized trials have been investigating treatment of SBP. Most available studies are retrospective analyses. The investigated drugs included ceftriaxone, cefotaxim, ceftazidim as well as ureidopenicillins combined with a beta-lactamase inhibitor (BLI) [102] (Table 16 [Tab. 16]). Given in conjunction with albumin (to treat ascites) these therapies achieved clinically treatment success rates of about 80% [489].

Peritonitis associated with CAPD

Peritonitis associated with continuous ambulatory peritoneal dialysis (CAPD) is usually caused by a contamination of tubes or a catheter system. The most frequent pathogens are coagulase-negative staphylococci and Staphylococcus aureus. E. coli, enterococci, streptococci, Pseudomonas aeruginosa, anaerobes, Enterobacter spp. or Candida spp. have been less frequently identified [502]. Uncomplicated cases may be treated locally by adding antimicrobial substances to the dialysis fluid. Only in the less common severe forms, intravenous therapy is required in addition to the intraperitoneal treatment. Dosage should be adjusted for renal dysfunction as appropriate.

A group 2 cephalosporin optionally combined with ciprofloxacin is recommended for calculated therapy [302]. The treatment should be adjusted according to the results of microbiological diagnosis. The antibiotics listed in Tab. 19 may be used if there is evidence of MRSA, MRSE and enterococci (incl. VRE). If the infection remains uncontrolled after a week of therapy, the peritoneal dialysis catheter must be removed [545].

Secondary peritonitis

Comprising about 80% to 90% of all cases, secondary peritonitis due to a perforation of the gastrointestinal tract is by far the most frequent IAI. By definition surgical intervention is required to control the source of infection (e.g. appendectomy for perforated appendix). Increasingly, the preferred approach is primary surgical treatment of the infection source followed by definitive closure of the abdomen and subsequent clinical monitoring of the patient [220], [535]. Secondary peritonitis may be divided in community-acquired (approx. 70%) and postoperative (approx. 30%) forms.

Community-acquired secondary peritonitis

Community-acquired secondary peritonitis always involves mixed bacterial infection. The spectrum of pathogens originates from the gastrointestinal flora and depends on the pathogenesis and localization of the perforation and/or leakage. The predominant pathogens are E. coli, Bacteroides fragilis, enterococci and Candida spp. Besides surgical interventions, calculated initial antibiotic therapy should be started preferably before surgery.

The following recommendations are based on the duration of disease and the pathogen spectrum as determined by the cause of disease [545]. Aminopenicillins/BLI, ureidopenicillins/BLI, group 2 carbapenems, or alternatively group 2 or 3a cephalosporins or a group 2 fluoroquinolone in combination with metronidazole can be used for locally confined, acute peritonitis. In the absence of risk factors, therapy can be limited to 1 or 2 days (Table 17 [Tab. 17]). Group 2 fluoroquinolones and aminopenicillins/BLI should only be used if the local resistance epidemiology for major pathogens such as E. coli shows susceptibility rates ≥90% [486].

Drugs or combinations with a broad spectrum of efficacy should be used to treat diffuse peritonitis that already lasted more than 2 to 4 hours. Ureidopenicillins/BLI or group 1 and 2 carbapenems are used for calculated therapy. Alternatively, a combination of metronidazole with a group 2 and 3 fluoroquinolone as well as a group 4 fluoroquinolone or a glycylcycline (tigecycline) can be used. The involvement of enterococci should only be considered when choosing a drug regimen for postoperative peritonitis and severely ill patients [141], [217], [486], [550]. Aminoglycosides (even in combination with clindamycin or metronidazole) have been shown to be inferior to newer treatment regimens with beta-lactam antibiotics and fluoroquinolones and are no longer considered as drugs of choice [401]. However, aminoglycosides are useful as combination partners used together with broad-spectrum beta-lactam antibiotics (particularly carbapenems and ureidopenicillins/BLI) [194], [324]. Pharmacokinetic variability as well as ototoxicity and nephrotoxicity require regular plasma level monitoring (see chapter 3).

Postoperative peritonitis

Postoperative peritonitis is a nosocomial secondary peritonitis defined as an intraabdominal infectious complication following a surgical intervention (e.g. leaking anastomosis after anterior rectum resection). In contrast to tertiary peritonitis, postoperative peritonitis requires surgical intervention [141], [549]. Most patients have been treated with antibiotics before the disease becomes apparent. Therefore, postoperative peritonitis is characterized by a selected spectrum of pathogens including enterococci (including VRE), multiresistant gram-negative pathogens (ESBL producers) and fungi.

Antibiotics with broad antimicrobial spectra including group 1 and 2 carbapenems, tigecycline, piperacillin/tazobacam or moxifloxacin should be used. The use of antifungals should be considered if appropriate [29], [103], [236], [314], [316], [328].

Tertiary peritonitis

In tertiary peritonitis the intraabdominal infection persists without a surgically removable focus after surgical sanitation of the source of a secondary peritonitis [103], [274], [366]. Usually low-virulence pathogens cause persistent infection, due to sustained immunosuppression of the patient. This form of nosocomial peritonitis has a shifted pathogen spectrum similar to that found in secondary postoperative peritonitis. It commonly involves enterococci (including VRE), staphylococci (including MRSA), enterobacteriaceae, anaerobes and Candida spp. [141]. Group 1 and 2 carbapenems, tigecycline (in combination with a drug effective against Pseudomonas), ureidopenicillins combined with a beta-lactamase inhibitor or group 3a cephalosporins in combination with metronidazole can be used in antimicrobial therapy (Table 17 [Tab. 17]) [141], [236], [314], [328], [486]. Combination therapy is recommended if there is evidence of Enterobacter spp. and treatment with a carbapenem is not feasible.

Necrotizing pancreatitis with infected necrosis

Approximately 80% of all deaths due to acute pancreatitis are caused by septic complications. The translocation of pathogens from the colon into the peripancreatic tissue is the most common cause of secondary infected pancreatic necrosis [77], [572]. Infected pancreatic necrosis is assumed if fever, leukocytosis, an increase of CRP in serum and an unexpected clinical deterioration occur. Evidence of gas inclusions within the necrotic pancreas tissue in the abdominal CT is considered to demonstrate infected necrosis [35]. The conservative interventional treatment of infected pancreatic necrosis includes CT-guided endoscopic transgastric drainage. Open or minimally invasive surgical treatment is currently thought to be best performed after an interval of more than 3 weeks [61]. Recent publications and meta-analyses conclude that indiscriminate administration of antibiotics does not achieve significantly positive effects on the course of necrotizing pancreatitis but will rather select for resistant pathogens and Candida spp. [235], [327], [541]. International consensus conferences do not generally recommend antibiotic treatment [121], [365].

Absolute indications for antibiotic treatment of proven infected necrosis include infected pseudocysts, abscess formation, cholangitis and other extrapancreatic manifestations. The most important pathogens of infected pancreatic necrosis are enterobacteriaceae, enterococci, staphylococci, anaerobes and Candida spp. When choosing appropriate antimicrobials, the ability of the drug to penetrate the pancreatic tissue has to be taken into consideration (Table 18 [Tab. 18]). Investigations with reliable data showing adequate tissue penetration are available for quinolones (ciprofloxacin, moxifloxacin), carbapenems (doripenem, ertapenem, imipenem/cilastatin, meropenem), metronidazole, cephalosporins (cefotaxime, ceftazidime, cefepime) and penicillins (mezlocillin, piperacillin ± tazobactam). Inadequate tissue penetration has been demonstrated for aminoglycosides [400], [545].

Secondary cholangitis

Infection of the hepatic bile ducts is usually caused by an obstruction, most commonly caused by gall stones, benign structures and tumors. Isolation of the pathogen by endoscopic retrograde cholangiopancreaticography (ERCP), intraoperative sampling or blood cultures is successful in 75% to 100% of bile duct obstruction caused by stones. The spectrum of pathogens includes enterobacteriaceae, enterococci and anaerobes as well as Pseudomonas spp. The key intervention for choledocholithiasis consists of endoscopic clearing of the bile ducts and subsequent laparoscopic cholecystectomy. Calculated antibiotic treatment can be started with an aminopenicillin/BLI, ureidopenicillin/BLI or a group 3 or 4 cephalosporin in combination with metronidazole. Alternatively, group 1 or 2 carbapenems or group 2 or 3 fluoroquinolones are used. Treatment duration after successful bile duct clearing is less than 3 days, it should be prolonged if the bile flow remains impaired. If Pseudomonas infection is suspected (e.g. case of repeated intervention or prolonged hospitalization), a combination with another agent covering Pseudomonas is recommended [545].

Difficult to treat and multiresistant pathogens (MRP)

In the mid 1990s, 95% to 97% of all bacterial strains isolated from patients with intraabdominal infections were still susceptible to frequently used antibiotics (group 3a cephalosporins + metronidazole, group 2 quinolones) [266]. In recent years, however, the proportion of resistant strains (MRSA, VRE, ESBL producers, multiresistant Pseudomonas spp.) has markedly increased worldwide, particularly in postoperative peritonitis and tertiary peritonitis [103], [122], [149], [267], [441]. Particularly in cases with life-threatening disease that may be caused by resistant pathogens, it is of key importance to cover the expected pathogen spectrum with the initial antibiotic treatment. If microbiological results show no evidence of resistant pathogens, treatment should be deescalated.

The following sections discuss particular aspects of individual multiresistant pathogens (see Table 19 [Tab. 19]).

Methicillin-resistant Staphylococcus aureus (MRSA)

Intraabdominal infections caused by MRSA is rarely found in immunocompetent patients. In most cases MRSA involvement will be a colonization of open abdomen, for example after abdominal compartment syndrome and open irrigation. Antibiotic treatment is indicated for non-immunocompromized patients only in case of local and systemic signs of infections as well as persistent detection of MRSA. In immunocompromized patients (e.g. after transplants) any indication of MRSA involvement should prompt antimicrobial therapy. Tigecycline is the only novel MRSA-effective antibiotic licensed for the treatment of intraabdominal infections [29]. It also covers other expected gram-negative and anaerobic pathogens. Vancomycin does not penetrate well into the intraabdominal compartment. Clinical data are available for treatment of intraabdominal infections with linezolid [63]. Linezolid, daptomycin and vancomycin should be combined with antibiotics effective against gram-negative pathogens [22], [141], [567].

Enterococci (including VRE)

Some controversy prevails about the role of enterococci as a primary pathogen in polymicrobial IAI as multiple evidence indicates successful treatments of IAI by means of surgical source control and use of antibiotics that do not cover enterococci [217], [374], [441]. Therapy targeting enterococci is recommended for patients with postoperative or tertiary peritonitis, severe sepsis originating in the abdomen and a risk of endocarditis (peritonitis and cardiac valve replacement) [217]. In these situations, selective pressure favouring VRE is expected particularly after previous antimicrobial therapy (Table 19 [Tab. 19]). Case reports on linezolid-resistant enterococcal strains have been published [212].

ESBL producers

In the last few years the enterobacteriaceae (particularly E. coli and K. pneumoniae) have increasingly developed resistance against beta-lactam antibiotics, including group 3 and 4 cephalosporins, which are hydrolysed by extended-spectrum beta-lactamases (ESBL) [307], [576]. Surgical treatment areas focusing on the abdomen report a relevant prevalence of ESBL-producing pathogens [296]. Carbapenems are primarily indicated in infections with ESBL-producing pathogens. Depending on test results, fluoroquinolones or fosfomycin may be used as combination partners. Tigecycline and colistin are usually effective, but their activity must be tested as well [194], [324].

Pseudomonas spp., Acinetobacter spp.

Pseudomonas spp. is detected in about 8% of IAI. However, the percentage of causally relevant strains may be significantly lower [134], [490]. In general, group 3b and 4 cephalosporins, ciprofloxacin, group 1 carbapenems, piperacillin ± tazobactam and aminoglycosides may be used in antimicrobial therapy [139]. Combination therapy (e.g. a group 3b cephalosporin plus an aminoglycoside or plus ciprofloxacin) lowers the post-treatment resistance rate [526] but has no clinical advantage [400]. If three or four antibiotic classes are ineffective (multidrug resistance, MDR), usually only colistin can be used [194], [324]. The same applies for carbapenem-resistant Acinetobacter spp., where tigecycline may also be effective. In special cases, sulbactam used as monotherapy may be active as well (note test results).

Invasive intraabdominal fungal infections

Most invasive intraabdominal fungal infections (IAFI) are caused by Candida spp. Up to 18% of all cases of severe sepsis in Germany are caused by Candida spp. [159]. Antifungal therapy is not required if Candida is detected only once in intraoperatively acquired samples of a postoperatively stable and immunocompetent patient with community-acquired secondary peritonitis (e.g. after perforation of a gastric ulcer). From a surgical point of view, risk groups for IAFI include patients with severe postoperative peritonitis (e.g. caused by suture leakage after esophagojejunostomy) or tertiary peritonitis. In two studies with such risk groups, pre-emptive therapy with fluconazole achieved a significant reduction of the incidence of invasive fungal infections and a statistically non-significant reduction of mortality versus placebo [151], [407]. However, the necessity of pre-emptive treatment remains controversial [66].

In approximately 60% of cases, initial high-dose therapy with fluconazole is adequate (Table 17 [Tab. 17]). However, the rate of Candida strains with reduced susceptibility to fluconazole remains high at approximately 40% in Germany [410]. Therefore, based on new multicentre studies, the use of an echinochandin (anidulafungin, caspofungin) is preferred if the patient is instable or recently received treatment or prophylaxis with an azole. For toxicity reasons, initial therapy with amphotericin B is recommended only in patients with allergy to other antifungal substances [396]. Delaying the treatment of an IAFI is associated with poor outcomes [66], [271], [343]. Due to a lack of specific controlled study data for IAFI and because of the individual clinical situation (e.g. stable vs. unstable patient), no levels of recommendation have been assigned.


8 Infections of kidneys and genito-urinary tract

Florian Wagenlehner, Reinhard Fünfstück, Udo Hoyme, Martin Kaase, Eberhard Kniehl, Johannes Knobloch, Kurt Naber

Indications for initial intravenous antimicrobial therapy

Usually infections of the kidneys and urogenital tract in adults require initial (empirical) intravenous antibiotic therapy only for cases with severe clinical course and systemic manifestations, e.g. nausea and vomiting, or suspected sepsis (see chapter 10) [199]. This generally includes severe forms of uncomplicated or complicated and/or nosocomial pyelonephritis, acute prostatitis, and less frequently acute epididymitis with or without orchitis, acute salpingitis-pelvicoperitonitis or severe abscess-forming infections in kidneys and urogenital tract. Occasionally, intravenous therapy should be empirically initiated if, in certain clinical situations, multiresistant pathogens must be anticipated for which is no oral antibiotic available and the results of microbiological tests cannot be awaited as, for example, surgery must be immediately performed (e.g. acute urinary stone obstruction).

General criteria for choosing an antibiotic

The antimicrobial agent is chosen according to the expected pathogen spectrum, taking into consideration the pharmacokinetic and pharmacodynamic properties of the drug. When treating infections of the urinary tract, sufficient renal elimination of the active drug is required [405], [557]. In addition, the collateral damage of antibiotics, e.g. side effects and development of resistance, must be taken into account (see chapter 2). Other appropriate measures may be needed but are not the subject of this article [62], [72], [514].

Acute uncomplicated pyelonephritis

The most frequently detected pathogen is Escherichia coli, followed by Proteus mirabilis and Klebsiella pneumoniae. Other enterobacteriaciae have been detected less frequently in the urine. There are no large epidemiological studies on antimicrobial susceptibility. Studies on uncomplicated cystitis may be used as substitute, since they have a somewhat similar pathogen spectrum and resistance situation [357]. Early initiation of effective therapy may prevent damage to the renal parenchyma. An initial (empirical) intravenous therapy with a group 3a cephalosporin, a fluoroquinolone with high renal elimination (e.g. ciprofloxacin or levofloxacin), an aminopenicillin/beta-lactamase inhibitor (BLI) or an aminoglycoside is indicated in cases with severe systemic symptoms including nausea and vomiting [33], [199], [239], [354], [409]. After improvement of these symptoms, the intravenous therapy should be changed to an oral regimen as soon as possible. An oral fluoroquinolone such as ciprofloxacin or levofloxacin, an oral group 3 cephalosporin, an aminopenicillin/BLI, cotrimoxazol or trimethoprim would be appropriate, if the pathogen tested susceptible [199], [353], [431], [511]. Treatment duration depends on the clinical course of the disease. Seven to 10 days are usually sufficient. Sometimes an elevated initial dose of a fluoroquinolone, e.g. levofloxacin 750 mg per day, can shorten the duration of therapy to 5 days [409].

Complicated and/or nosocomial urinary tract infections

Definition

A complicated urinary tract infection (UTI) is defined as an infection of the urinary tract, which is associated with a morphological, functional or metabolic abnormality resulting in renal dysfunction, impairment of urine flow and/or local or systemic disruption of immune mechanisms [65], [240], [514], [553].

Indications for initial intravenous antimicrobial therapy

As mentioned above, the indications for any initial intravenous antimicrobial treatment depend on the general condition and risk profile of the patient. The successful antibiotic treatment of complicated UTI requires elimination of the complicating or causative factors [199].

Pathogen spectrum

In general, the expected pathogen spectrum is considerably broader than in uncomplicated UTI and depends on the circumstances under which the complicated UTI was acquired [65], [240], [556]. Thus the pathogen spectrum of a community-acquired, first-time, complicated UTI, e.g. acutely evolving as a result of calcium oxalate urinary stones, in a patient without recent antibiotic treatment and catheters, is relatively similar to the spectrum of an uncomplicated acute pyelonephritis [62]. In contrast, in a nosocomial complicated UTI, pathogens must be anticipated that do not belong to the usual spectrum of pathogens seen in primary urinary tract infections because these infections rather represent secondary events resulting from selection or colonization. These pathogens may include, Pseudomonas aeruginosa, Acinetobacter baumannii, and Providencia stuartii [240], [556]. If a complicated UTI is suspected, sampling for urine culture is mandatory before starting an antibiotic treatment. This enables to adjust the treatment to the results of the microbiological test despite the broad pathogen spectrum and potential resistance [199].

Choice of antibiotics

The empirical initial antibiotic treatment must take into account the regional resistance situation of the expected spectrum of pathogens [240]. The following previous clinical conditions may influence the expected pathogen spectrum and susceptibility [240]:

1.
Where was the UTI acquired, e.g. community, nursing home, hospital, after a diagnostic/therapeutic intervention?
2.
Was there a previous antibiotic therapy (how long ago, which antibiotic)?
3.
Was there a previous prolonged hospitalization?
4.
Did the patient have a catheter (which, how long ago, how treated)?
5.
If so, check the quality of the urine drainage and if necessary change the catheter (remove the infectious biofilm)
6.
Is this a recurrence or a treatment failure?

Group 3a cephalosporins, fluoroquinolones, aminopenicillins with a BLI or a group 2 carbapenem (ertapenem) can be used as parenteral initial treatment of a first-time, community-acquired, complicated UTI [33], [199], [239], [354], [409]. In patients with nosocomial or catheter-associated UTI, multiresistant pathogens may be involved [240], [551], [553], [556]. Therefore, in empirical therapy an antibiotic should be used that covers rare and multiresistant gram-negative pathogens (see above). Group 3b or 4 cephalosporins, group 2 or 3 fluoroquinolones (check for local E. coli resistance!) and group 1 carbapenems (doripenem, imipenem, meropenem) can be used [199], [355], [559]. If coverage of enterococci is desired, as mixed infections with enterococci are particularly frequent in catheter-associated urinary tract infections, ureidopenicillins in combination with a BLI (e.g. tazobactam) are appropriate for empirical therapy [199], [356]. If multiresistant pathogens are suspected (due to outbreaks or high endemic resistance rates), the appropriate drugs should be used in the empirical treatment. Since carbapenemases rarely occur in the German-speaking countries yet, a group 2 carbapenem (ertapenem) can be used for enterobacteriaceae expressing extended-spectrum beta lactamases (ESBL) or a group 1 carbapenem (doripenem, imipenem, meropenem) can be used if involvement of Pseudomonas is suspected [199], [355], [559]. For ESBL, fosfomycin could also be used as intravenous initial therapy. However, few data are available for complicated UTI [358]. For infections with methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant enterococci (VRE), several very effective drugs, e.g. daptomycin or linezolid are available [352], [552], [557]. However, there are no adequate studies on urinary tract infections, so the appropriate treatment must be chosen on an individual basis.

Patients with diabetes mellitus

Urinary tract infections in patients with diabetes mellitus are problematic as they may increase the pathogenetically significant insulin resistance and therefore deteriorate the instable metabolism situation. This is particularly true for patients with an HbA1c value >9%, with a tendency for hypo- or hyperglycaemia, a BMI >30 kg/m2 and in cases with diabetic nephropathy (stage 2: albumin excretion >200 mg/l, eGFR <60 ml/min). Glucosuria facilitates the colonization of the urinary tract with pathogens and facultative pathogenic microorganisms.

For asymptomatic bacteriuria in patients with stable diabetic metabolism situation and no obstruction or other anatomical abnormalities, an antimicrobial treatment is not necessary [179], [388]. For uncomplicated and complicated infections, the same recommendations for initial intravenous and subsequent oral treatment apply as for patients without diabetes mellitus. Note that cotrimoxazole may enhance the hypoglycaemic effect of oral antidiabetics. Other metabolic interactions of antibiotics and antidiabetics are rare.

Patients with impaired renal function or kidney transplant

No potentially nephrotoxic antibiotic, e.g. aminoglycosides or vancomycin, should be used for patients with impaired renal function, dialysis patients or kidney transplant recipients. The first dose should always be a full normal dose. The choice of subsequent dosage depends on the mode of drug elimination and renal function (Table 20 [Tab. 20]) [25], [73], [463], [495], [503], [587].

Urosepsis

Urosepsis occurs after haematogenic distribution from the infected urinary tract without or after previous urological interventions. Primarily E. coli and other enterobacteriaceae have been detected as pathogens. Following urological interventions and in patients with indwelling catheters, multiresistant Pseudomonas spp., Proteus spp., Serratia spp., Enterobacter spp., enterococci and staphylococci must be considered (see complicated UTI) [199], [240].

The initial parenteral antibiotic therapy must be started immediately at the first suspicion of urosepsis (within the first hour) and after taking appropriate samples of urine and blood cultures [123], [124], [154], [276], [525]. Treatment recommendations are given in chapter 10. In general, the maximum possible dosage should be chosen [405], [561] in addition to intensive care for sepsis.

Urosepsis usually involves obstructive uropathy, e.g. due to urolithiasis, tumors, benign prostatic hypertrophy or abscess-forming infection, specific urological diagnostics should detect and localize the obstructive uropathy and/or the abscess with the goal of controlling the source as early as possible or bypass the obstruction (e.g. by transurethral or suprapubic catheter, ureteral stent or nephrostomy) ensuring unimpaired urinary flow [123], [124], [558], [560].

Acute prostatitis, prostatic abscess

The empirical treatment for acute bacterial prostatitis (ABP) follows the same considerations as for complicated urinary tract infections [199], [555]. Predominantly E. coli and other enterobacteriaceae are responsible for spontaneously occurring ABP. In patients with acute prostatitis following urological surgery, other gram-negative pathogens, e.g. Pseudomonas spp. may be detected as well. In these patients the acute prostatitis is often accompanied by abscesses which frequently involve K. pneumoniae as well [337].

For the empirical treatment, the chosen drug should achieve high antibiotic concentration in the urine, and sufficient concentration in the prostate tissue, prostate secretion and ejaculate [199]. Initial intravenous antibiotic therapy is necessary only for severe forms of acute bacterial prostatitis with and without abscess. The drug of choice is a group 2 or 3 fluoroquinolone [554], [555]. History of previous antibiotic therapy is particularly important as many patients have already received fluoroquinolones and are thus at risk for fluoroquinolone-resistant pathogens. Alternatively, group 3 or 4 cephalosporins or an ureidopenicillin with a beta-lactamase inhibitor may be used for acute prostatitis. Since acute prostatitis is a less common infection and early initiation of antibiotic therapy is necessary, there are no prospective randomized studies available. Treatment recommendations are therefore based mainly on expert opinions [199], [458], [555].

After the pathogen has been isolated from a urine culture (prostate massage is contraindicated in cases of acute bacterial prostatitis) and the resistance situation has been clarified, the treatment should be switched to a targeted therapy. After improvement in the clinical situation it should be continued orally for at least two (to four) weeks to avoid complications such as acute urinary retention, epididymitis, prostate abscesses or chronic prostatitis [199], [458], [555].

Acute epididymitis, epididymal orchitis, including infections with abscesses

An epididymitis in sexually active men under 35 years of age is usually caused by Chlamydia trachomatis or Neisseria gonorrhoea, or enterobacteriaceae in men having sex with men. An (often asymptomatic) urethritis is usually present [199], [99].

Generally, intravenous therapy is only necessary in severe forms with a risk of complications, e.g. abscesses, or treatment failure. The intravenous treatment should be changed to an oral regimen as early as possible. In young men, ceftriaxon in combination with doxycycline is recommended. In older men or in the case of allergies against beta-lactam antibiotics, group 2 or 3 flurorquinolones can be considered. The increasing resistance to fluoroquinolones and the increasing occurrence of ESBL-producing enterobacteriaceae must also be taken into account in older men with urinary tract infections [65], [556].

Endometritis, salpingitis, tubo-ovarian abscess, pelvicoperitonitis

In infections of the female genital organs in sexually active, premenopausal women, a broad-spectrum of potential pathogens must be expected. Besides the sexually transmitted pathogens N. gonorrhoeae and C. trachomatis, potential pathogens involve the vaginal flora and pathogens of bacterial vaginosis – and in rare cases mycoplasma or ureaplasma [233], [306], [505]. With a few exceptions only laparoscopically acquired samples are diagnostically relevant for the aetiology of ascending infections [238]. Since none of the available antibiotics covers the complete spectrum of potential pathogens, there is no consensus on the treatment of choice. However, numerous investigations with drug combinations have shown positive results. Reliable investigations of intravenous and oral treatment regimes as well as comparisons of the out-patient and hospital treatment are still pending. Therefore the decision for one of the proposed regimes must be made on an individual basis, depending on the severity of disease, patient compliance and local resistance situation. After clinical improvement, an intravenous initial treatment can be changed to an oral regimen with one of the combination partners usually being doxycycline, clindamycin or a fluoroquinolone [99]. Cephalosporins should be combined primarily with metronidazole for anaerobic coverage. Alternatively, fluoroquinolones or aminopenicillins/BLI which are characterized by their broad spectrum of activity can also be used. Group 2 or 3 fluoroquinolones in combination with metronidazole, aminopenicillins/BLI plus doxycycline or a group 2 carbapenem can be used as well. Monitoring the course of the disease for 72 hours is essential, even in primarily uncomplicated infections [99]. In case of treatment failure, the antimicrobial therapy should be adapted to the meanwhile available microbiological results and, if necessary, surgical intervention should be performed [233], [306]. In pregnant women, the embryotoxic and teratogenic potential of antibiotics must be taken into consideration.


9 Skin, soft tissue, bone and joint infections

Peter Kujath, Christian Eckmann, Wolfgang Graninger, Gerd Gross, Cord Sunderkötte

There is a broad spectrum of skin and soft tissue infections, ranging from harmless, superficial pyodermia to life-threatening myonecrosis with high mortality. Skin and soft tissue infections caused by viruses, bacteria, parasites or fungi are among the most frequent infectious diseases [243], [272]. In order to organize this plethora of diseases, the British microbiologist Kingston devised three levels of severity in 1990 [258]. His classification is of clinical relevance as it considers the urgency of surgical intervention (Table 21 [Tab. 21]).

A further clinically useful approach is the classification of complicated versus uncomplicated skin and soft tissue infections. The term complicated used by the FDA is defined by major risk factors of skin and soft tissue infections (Table 22 [Tab. 22]). Complicated skin and soft tissue infections (cSSTI) or complicated skin and skin structure infections (cSSSI) formed the basis for the pivotal clinical licensing studies for new antibiotics in recent years [22], [156], [322].

Another consideration is the extent of infection in terms of local versus diffusely spread infection with sytemic symptoms [300]. Also the depth of infection, i.e. involvement of subcutaneous tissue, fascia or muscles must be considered (Table 23 [Tab. 23]).

Skin infections with predominantly conservative treatment

Examples of these primarily cutaneous, superficial bacterial infections include impetigo contagiosa, boils, carbuncles, erysipelas and erysipeloids.

Indication for the use of an antibiotic is an infection with general symptoms: fever (>38.5°C), leukocytosis (>10,000/µl) and markedly increased CRP.

Impetigo/ecthyma (deep, ulcerating form of impetigo) (Streptococcus pyogenes, beta-hemolytic group C and G streptococci, Staphylococcus aureus)

A widespread impetigo requires systemic but almost never intravenous antibiotic therapy. Intravenous administration is indicated only for infections of the face and failure to respond to oral therapy. Penicillin G is recommended for ecthyma caused by streptococci and an isoxazolyl penicillin (oxacillin or flucloxacillin) for ecthyma caused by S. aureus.

If the pathogen is unknown, a Group 2 cephalosporin or alternatively a macrolide is recommended [465].

Boils and carbuncles also require a systemic antibiotic treatment or a parenteral treatment only for localization on the face or failure to respond to the orally administered drug.

As S. aureus is the most frequent pathogen, treatment should be with Group 1 or 2 cephalosporins, clindamycin or an aminopenicillin in combination with a beta-lactamase inhibitor (BLI).

Erysipeloid (Erysipelothrix rhusiopathiae)

The treatment of choice for both local infections and rare systemic forms (with fever, endocarditis or arthritis) is penicillin G. Patients who are allergic to penicillin receive cephalosporins, clindamycin or fluoroquinolones. Erysipelothrix is resistant to glycopeptides.

Erysipelas (S. pyogenes)

Classic erysipelas is an infection caused by S. pyogenes with the characteristic symptoms of overheated erythema with shiny surface, sharply defined edges and tongue-shaped offshoots, usually at some centimeters distance from the focus of infection (e.g. interdigital mycosis). Depending on the enzyme and toxin expression of the bacteria, blisters and bleeding as well as already initially systemic infection reactions (fever, chills, increasing ESR, increasing CRP, and leukocytosis (>10,000/µl) occur. The drug of choice is penicillin, also in the case of a recurrence. To date, no penicillin-resistant S. pyogenes strains have been detected. Aminopenicillins or penicillinase-insensitive penicillins are not the first choice due to their low activity. In patients with allergy, clindamycin is the alternative treatment. Moxifloxacin may be used in case of clindamycin failure.

Parenteral antibiotic therapy is indicated for complicated erysipelas (e.g. haemorrhagic, necrotizing or blistered erysipelas) as well as localization in the face and impaired venous or arterial blood circulation. If symptoms improve after 5 to 7 days, treatment may be continued with oral penicillin V or an oral group 1 or 2 cephalosporin.

Soft tissue infections without need for immediate surgical intervention/cellulitis/plegmons (e.g. soft tissue infections in chronic wounds)

Other than in classical erysipelas which is caused only by Streptococci, the deep forms of soft tissue infections are clinically characterized by overheated, more edematous, painful reddening and doughy swelling, by livid, dull and less distinct borders, while initial systemic signs of infection may be absent. An appropriate differentiation is desirable for therapeutic reasons, since a broad spectrum of pathogens must be expected for soft tissue infections which are not classical erysipelas.

The term cellulitis in the broadest sense describes any skin infection spreading out from a point of primary infection. In a more narrow interpretation, cellulitis is an acute infection which spreads from a pre-existing skin lesion (wound, ulcer) into the dermis and subcutaneous tissue. Pathogens are usually S. aureus but also Escherichia coli, Klebsiella spp., etc. However, usage of this term is not restricted to this definition in the German and English-speaking countries. It is rather used as a generic term for all cutaneous soft tissue infections including erysipelas and phlegmons [504].

The most frequent pathogen is S. aureus. However, infections originating from chronic wounds such as pressure sores, ulcers associated with peripheral arterial vascular disease (pAVD) or venous insufficiency are often polymicrobial (S. aureus, haemolytic streptococci, enterobacteriaceae).

Mild to moderately severe infections can be treated with oral clindamycin (3–4x300–600 mg/day orally). For moderately severe to severe S. aureus infections or critical localization (e.g. hands or face), treatment with an intravenous isoxazolyl penicillin (flucloxacillin or oxacillin 4x1–2 g/day IV) or alternatively a Group 2 cephalosporin (e.g. cefuroxim 3x0.75–1.5 g/day IV) is recommended.

Complicated chronic, usually polymicrobial infections (e.g. pressure sores, ulcers) are often caused by mixed infections of gram-positive and gram-negative pathogens and typically involve anaerobes as well. Initial therapy is preferably IV aminopenicillin/BLI (e.g. amoxicillin/clavulanic acid 3x2.2 g/day or ampicillin/sulbactam 3–4x1.5–3 g IV) or moxifloxacin. In case of treatment failure, a therapy targeted to the detected pathogen and susceptibility results should be started. he pathogen should preferably isolated from a tissue sample.

  • For infections complicated by diabetes mellitus or pAVK see below.
  • For infections with evidence of MRSA see below.

For septic diseases which aetiologically result from polymicrobially infected ulcers, the recommended initial therapy is an intravenous carbapenem, piperacillin/BLI or Group 2 or 3 fluoroquinolone combined with metronidazole, or moxifloxacin given as monotherapy.

The following pathogens require specific antibiotics or antifungals, respectively:

  • Aeromonas hydrophila after exposure to fresh water and Vibrio spp. (Vibrio vulnificus, Vibrio alginolyticus, Vibrio parahaemolyticus) after exposure to salt water (see above)
  • Mycobacteria
  • Haemophilus influenzae in children (periorbital cellulitis)
  • Pseudomonas aeruginosa with neutropenia
  • Cryptococcus neoformans with disturbances of the cell-mediated immune response
  • Pasteurella spp. and Capnocytophaga spp. after animal bites (see below) [155], [161], [294], [504]

Infections needing immediate surgical intervention For these infections as for abscesses, panaritium, phlegmons or suppurative bursitis, surgical intervention with adequate debridement is essential. If risk factors are present such as a complicated skin and soft tissue infection and general symptoms, oral or parenteral antibiotic treatment is indicated. Local antibiotic therapy is inappropriate. The calculated initial treatment should involve an aminopenicillin/BLI, an ureidopenicillin/BLI, a Group 1 or 2 cephalosporin, an isoxazolyl penicillin or clindamycin. Group 4 fluoroquinolones may be used for these infections because of their broad spectrum of activity [67], [193], [305]. After identification of the pathogen and testing its susceptibility, the treatment can be optimized by further targeting. Antibiotic therapy can be stopped after an improvement of fever, general symptoms, CRP and leukocytosis (to values below 8,000/µl). A 5-day antibiotic regime is equivalent to a 10-day treatment [499].

Severe, life-threatening soft tissue infections

A hallmark of these rare necrotizing soft tissue infections is the acute course of disease with early organ failure. These diseases are grouped under the designation “necrotizing skin and soft tissue infections” (nSSTI) in English-speaking countries [305]. They are often associated with agonizing pain and toxin-induced microthromboses causing pronounced tissue necrosis with decreased perfusion and hypoxia in the affected area [85].

The initial clinical presentation is often unspecific making timely and life-saving diagnosis more difficult. The typical early tissue necrosis is often detected intraoperatively or by means of biopsies. Necrosis of the skin is generally absent at the time of diagnosis and appears later in the course of disease. Immediate measures involve radical surgical debridement with antibiotic therapy started at latest during surgery and intensive medical care. The disease patterns include myonecrosis (gas gangrene), necrotizing fasciitis including Fournier’s gangrene, secondary infected injection abscess and myositis. Streptococcal toxic shock syndrome (STSS) can also be grouped into this type of diseases [82].

Aside from hematogenic dissemination, potential pathogen entry points include trivial traumatic wounds, infected surgical wounds and injection sites, less frequently infected periurethral glands or perianal infections (Fournier’s gangrene). Usually these are mixed infections with grampositive pathogens (beta-haemolytic streptococci, S. aureus), anaerobes (Bacteroides fragilis, Prevotella melaninogenica) and enterobacteriaceae often with a resulting synergistic increase of virulence.

Antimicrobial therapy with an ureidopenicillin/BLI or a Group 1 or 2 carbapenem is recommended. Alternatively a combination of a Group 3a cephalosporin with metronidazole or clindamycin is appropriate. Because of its broad spectrum, monotherapy with moxifloxacin is an alternative. The additional administration of clindamycin or linezolid inhibits toxic protein biosynthesis in gram-positive bacteria. This alleviates septic complications of exotoxin production (e.g. superantigens). There is evidence that the combination of a penicillin with clindamycin is effective in streptococcal toxic shock syndrome [64], [499].

Certain skin and soft tissue infections require special treatment.

Bite wounds

The jaws of mammals generate bite pressures of up to 1 t/cm2. Therefore, their teeth may inflict severe tissue distruction with accompanying contamination. For deep wounds surgical treatment and primary antibiotic therapy is indicated. An aminopenicillin/BLI or, in case of allergy, a Group 1 or 2 cephalosporin or a Group 4 fluoroquinolone is recommended [278], [510].

The pathogen spectrum generally originates from the physiological oral flora of the animal which inflicted the wound or from the bitten individual. Cat and dog bites are usually aerobic/anaerobic mixed infections of various pathogens. Pasteurella, staphylococci and streptococci are regularly found. Particularly after cat bites, the transmitted pathogens may reach deeper tissue layers due to the puncture type of the bite.

If bones or tendons are injured, chronic osteomyelitis or tendomyositis and/or tendosynovitis can result.

The most frequently isolated anaerobes after a dog or cat bite are Bacteroides spp., Fusobacterium spp., Porphyromonas spp., Prevotella spp., Propionibacterium spp. and Peptostreptococcus spp.

After rat bites, Streptobacillus moniliformis, the pathogen of rat-bite fever, must be expected.

Bite wounds inflicted by humans may cause acute and smoldering chronic infections. Usually there are Gram-positive (mostly Streptococcus spp.) and Gram-negative pathogens (e.g. Haemophilus spp., Eikenella corrodens) as well as anaerobes (Fusobacterium, Prevotella and Porphyromonas species).

In all cases of bite wounds, the patient’s tetanus vaccination status must be checked.

Diabetic foot syndrome

Foot lesions in diabetics are the result of complex long-term neuropathic and angiopathic damage. Reduced immune reactions after trivial trauma and permanent mechanical stress often lead to painless soft tissue infections which may extend to adjacent tendons, joint capsules and bone or the entire foot. Antibiotics which are sufficiently active in soft and osseous tissue should be used.

Diabatic foot infections usually involve mutiple pathogens including staphylococci, streptococci, enterobacteriaceae, pseudomonads and/or anaerobes.

For mild infections, oral aminopenicillins/BLI or Group 2 and 3 fluoroquinolones are recommended. Moderate to severe illnesses must be treated surgically and initially parenteral antimicrobials must be administered. Here ureidopenicillins/BLI, a Group 1 or 2 carbapenem, or a combination of clindamycin with a Group 2, 3a or 4 cephalosporin or a Group 4 fluoroquinolone (moxifloxacin) is recommended. A long-term treatment over several weeks is often necessary [55], [273], [304], [538].

Decubital ulcers (bedsores)

Pressure sores may evolve even with appropriate prophylactic measures [96].

The surgical treatment depends on the sore stage of, according to the scale suggested by Campbell. Treatment with an antibiotic is usually indicated for infected sores. The responsible pathogens are Gram-positive and Gram-negative aerobic bacteria and anaerobes. Ureidopenicillins/BLI, a Group 2, 3 or 4 fluoroquinolone, or a Group 3 or 4 cephalosporin are recommended. Clindamycin or metronidazole may be added.

MRSA in skin and soft tissue infections

Skin and soft tissue infections (mostly in hospitals and long-term care facilities) may be caused by methicillin-resistant S. aureus (MRSA). These are most frequently postoperative or chronic wounds. As for other skin and soft tissue infections, local treatment is indicated. The decolonization is carried out locally according to the corresponding hygiene guidelines [248].

The indication for systemic use of antibiotics should be handled restrictlively (e.g. for infections with systemic reactions such as fever [>38.5°C], leukocytosis [>10,000/µl] and marked increase in CRP). Detection of pathogens is often due to bacterial colonization rather than wound infection.

According to available data, linezolid achieves significantly higher eradication rates in MRSA monoinfections than vancomycin [567]. Daptomycin may also be used in cases of systemic infection or MRSA bacteremia [22], [450]. Tigecycline is a therapeutic alternative for polymicrobial infections involving MRSA. It is recommended to use glycopeptides (vancomycin or teicoplanin) in combination with rifampicin or fosfomycin.

In recent years, primarily in the USA, outbreaks of MRSA strains susceptible to clindamycin, cotrimoxazol and partially fluoroquinolones have occurred. These community-associated MRSA (caMRSA) occur in groups of healthy patients (military, sport clubs). Because of their high virulence mediated by toxin production (particularly panton-valentine-leukocidin), they may cause skin and soft tissue infections of all severity levels. Early diagnosis and consistent treatment currently appear to be the most effective approach to prevent severe disease. Strict hygiene measures are important to avoid outbreaks within hospitals. Wound closure should be achieved.

Mediastinitis

The most frequent forms of mediastinitis are caused by perforations of oesophagus or trachea, by descending infections from the mouth/throat, and postoperative sternum infections. The source of the infection must be surgically removed. In addition, a high-dose antibiotic therapy is indicated. The pathogen spectrum of hematogenous mediastinitis mainly includes gram-positive cocci. Primarily Gram-positive cocci, anaerobes and Candida spp. may be involved after perforation of the esophagus or postoperative complications following esophageal surgery. A group 1 or 2 carbapenem, an ureidopenicillin/BLI or a group 3 or 4 cephalosporin, optionally in combination with metronidazole is recommended. Moxifloxacin may be used as a monotherapy. Initially, fluconazole may be added in patients at high-risk of fungal involvement. The use of daptomycin, linezolid and tigecycline is also possible.

Postoperative wound infections

The incidence of postoperative wound infections has significantly decreased in the last two decades after the introduction of perioperative antibiotic prophylaxis. The management of postoperative wound infections depends on the stage of infection according to Cruse’s wound classification and the additional risk factors (see Chapter 16) [115].

Postoperative wound infections are divided into superficial and deep wound infections defined by the Centers for Disease Control (CDC) in Atlanta, Georgia, USA [38].

Microbiological investigation to determine the pathogen should be carried out for all postoperative wound infections. The treatment of choice is to open the infected wound and treatment continuation. Treatment with antibiotics is indicated only in exceptional cases (immunosuppression, sepsis, multiple risk factors). The calculated initial treatment should complement the perioperatively administered antibiotic. Either an ureidopenicillin/BLI, a group 3 cephalosporin or a group 2, 3, or 4 fluoroquinolone is recommended. In any perioperative wound infection, the possibility of a surgical complication (suture insufficiency, infected prosthesis, foreign body left in situ) must be considered and excluded during diagnosis. In severe disease and in patients at risk of MRSA infection, the initial additional use of linezolid or daptomycin should be considered. If there is no evidence of a resistant pathogen, treatment can be de-escalated early.

A summary of the antibiotic treatments for the skin and soft tissue infections discussed here is given in Table 24 [Tab. 24].

Bone and joint infections

The course of and prognosis of bone and joint infections critically depend on early diagnosis and adequate treatment. The latter consists of extensive surgical debridement and/or synovectomy, stabilization of fracture, and treatment of a skin/soft tissue defect. Antibiotic therapy is indicated (Table 25 [Tab. 25]), initially given at high-dose and via the intravenous route. A switch to oral therapy is feasible if adequate drug exposure is reached with an oral medication.

Osteomyelitis

Osteomyelitis is an infection of the bone marrow canal, which is usually caused by the hematogenous spread of endogenous pathogens. The pathogen spectrum varies according to the age of the patient. In adults, monoinfections by S. aureus, streptococci, Serratia marcescens or Proteus spp. are dominant pathogens. Calculated therapy is started with a Group 2 cephalosporin in combination with clindamycin or an aminopenicillin/BLI. Alternatively, a Group 2 or 3 fluoroquinolone in combination with clindamycin or moxifloxacin as monotherapy can be used. In complicated cases (e.g. severe spondylodiscitis), the combination of fosfomycin with a cephalosporin can be considered [459]. As soon as evidence of the pathogen and the results of a susceptibility test are available, the treatment should be changed to a more targeted regimen.

Posttraumatic or postoperative osteitis

Osteitis is defined as an infection of any bony skeletal structure. This generally occurs posttraumatically or postoperatively due to direct contamination during a trauma or surgery. Mixed infection with staphylococci, streptococci, enterobacteriaceae and anaerobes are common. Staphylococci dominate in postoperative osteitis. Treatment must start as soon as possible with surgical debridement, stabilization of the bone and initial calculated antibiotic therapy. An aminopenicillin/BLI, a Group 2 cephalosporin and clindamycin are recommended. In cases with high risk of multiresistant staphylococci, daptomycin, linezolid or teicoplanin or a combination with fosfomycin can be used. Rifampicin penetrates well into biofilms. In cases of chronic osteitis, the infected bone must be removed. This should be followed by a targeted antibiotic therapy [182], [247].

Sternum osteitis

Sternum osteitis is essentially caused by S. aureus and coagulase-negative staphylococci which are frequently multiresistant. The initial high-dose antibiotic therapy should involve an isoxazolyl penicillin or a group 2 cephalosporin combined with clindamycin or fosfomycin. In cases of MRSA or methicillin-resistant coagulase-negative staphylococci such as, for example, Staphylococcus epidermidis, the use of linezolid or daptomycin is recommended [461].

Bacterial arthritis

The fundamental cause of bacterial arthritis is an iatrogenic infection. With regard to the prognosis, the early infection is differentiated from the late infection. The pathogen spectrum includes mainly staphylococci and beta-hemolytic group A, B, C or G streptococci. Other pathogens such as enterobacteriaceae and gonococci are rare. Besides surgical treatment, the calculated antibiotic treatment is the same as recommended for osteitis. In rare cases of infections caused by Salmonella spp. or gonococci, antibiotic treatment alone is sufficient.

Prosthetic infections

Infected prostheses should preferably be removed or changed after extensive surgical debridement and antibiotic treatment at the maximum feasible dosage [325]. Only in cases of life-threatening contraindications surgical exchange of the prothesis may be omitted. The infection rarely resolves if purely conservative treatments are used.


10 Sepsis

Klaus-Friedrich Bodmann, Rainer Höhl, Wolfgang Krüger, Béatrice Grabein

The treatment of sepsis is one of the greatest challenges for hospital physicians [174], [184], [204], [226], [378], [427], [448], [507], [508], [570]. Particularly in intensive care medicine, sepsis and septic shock are very important due to the high mortality of 40% to 60%. A large study on the epidemiology of sepsis in German intensive care units [159] found prevalence rates of 12.4% for sepsis and 11% for severe sepsis (including septic shock), with an overall mortality of 48.4% and a hospital mortality of 55.2%. The incidence of newly diagnosed severe sepsis in Germany was 76 to 110 per 100,000 population. Fungal sepsis in non-neutropenic patients must also be taken into consideration [396]. In the German prevalence study, 17.8% of severe sepsis cases with microbiological documentation of pathogens were caused by fungi. In the USA, Candida spp. is the third most frequent pathogen detected in blood cultures of patients in intensive care units [575]. All immunocompromized patients, including those with tumors, diabetes mellitus, kidney and liver diseases, hematological malignant diseases, surgical intensive care (e.g. after polytrauma and burns) and high-risk surgery (e.g. organ transplants) carry an increased risk of developing sepsis. Aside from source control, effective antimicrobial therapy is the most important causal intervention which is supplemented by supportive and adjunctive intensive care measures [123], [124].

According to current knowledge, microbial sepsis appears to be best described by the definition of Schuster [469]: “Sepsis is the entirety of symptoms of life-threatening clinical diseases and pathophysiological changes occurring as reactions to pathogens and their products which spread from the point of infection into the blood stream, activating biological cascades and special cell systems thus triggering the production and release of humoral and cellular mediators.”

The generally accepted criteria for the diagnosis of sepsis include evidence of infection and at least two of the four criteria listed below [71]

1.
fever >38°C or, in rare cases, hypothermia <36°C
2.
tachypnea >20/min or hypocapnia with a PaCO2<32 mm Hg
3.
tachycardia >90/min.
4.
leukocytosis >12,000/mm3 or leukopenia <4,000/mm3.

Sepsis as a sydrome is divided into different levels of clinical severity according to the American consensus definition:

  • SIRS (Systemic Inflammatory Response Syndrome) is a general inflammatory defense reaction to various events (e.g. trauma, hypoxia, pancreatitis), which exhibits at least two of the above-listed criteria, although an infection need not be present.
  • Sepsis is the systemic reaction to an infection. As with SIRS, at least two of the above-listed criteria must be present. The international consensus definition of sepsis does not include the presence of organ dysfunction.
  • Severe sepsis is defined by organ dysfunction in addition to sepsis symptoms. Dysfunctions may affect: lungs (hypoxia, respiratory acidosis), kidneys (oliguria/anuria, metabolic acidosis), liver (e.g. jaundice, progressive sclerosing cholangitis), heart (heart failure, septic cardiomyopathy) but also CNS, gastrointestinal tract, bone marrow, coagulation and immune system. From the intensive care point of view and in contrast to the international consensus recommendation, the term sepsis is used if organ failure is present. Recommendations for antimicrobial therapy also refer to severely ill patients with organ dysfunction.
  • Septic shock is accompanied by a persistent reduction of blood pressure despite adequate fluid supply. Refractory septic shock is present if blood pressure falls for longer than one hour and cannot be corrected by infusion of fluids and use of vasopressors.

Pathophysiological evidence indicates that sepsis is caused by a complicated network of pro and anti-inflammatory cytokines. Following the pro-inflammatory phase of SIRS, an excess of anti-inflammatory cytokines is produced. This phase is called CARS (compensatory anti-inflammatory response syndrome). The SIRS and CARS phases often overlap in patients, making for a mixed antagonistic response syndrome (MARS). Sepsis is a dynamic process with transitions from the stage of “mild” sepsis to “severe” sepsis to “septic shock” with organ dysfunction or failure but also with the development of septic organ metastases.

Sepsis research in the last 20 years has concentrated mainly on supportive (volume and circulatory treatments) and adjunctive measures (anti-inflammatory agents). Initial positive study results with newly tested interventions often could not be confirmed in large, multicentre trials. Low-dose hydrocortisone can no longer be recommended as routine treatment for patients with septic shock (increased incidence of superinfections and no reduction of mortality) [493]. Likewise an intensified intravenous insulin treatment (glucose target level ≤110 mg/dl) is generally no longer recommended (increased incidence of severe hypoglycemic events), nor are antithrombin (AT III), low-dosed dopamin, vasopressin and hydroxyethyl starch in the investigated formulations [84].

Lung-protective ventilation [23], treatment guided by haemodynamic target values (early goal-directed therapy) [433] and – for patients at high risk of lethal outcome (failures of at least two organs) and without contraindications – early administration (<48 hours) of recombinant activated protein C [59] have become established treatments.

Addendum: the use of activated protein C is no longer recommended. The failure of new therapies for the treatment of severe sepsis and septic shock is related to the lack of an early and differentiated diagnosis. Whenever sepsis is suspected, an early “aggressive” diagnostic workup (e.g. CT, BAL) is required [521].

Early, adequate antimicrobial therapy is an essential determenant of patient survival.

For septic shock patients, Kumar et al. [276] showed in a retrospective study published in 2006 that the mortality rate increases by 7.6% with every hour of delay of therapy after the onset of hypotension. In a follow-up article published in 2009 [275], these data were impressively confirmed.

The severity of illness may be determined rather simple according to clinical criteria: need for ventilation, need for catecholamine therapy (particularly vasopressors), and organ dysfunction, primarily renal impairment. However, no clinical study was performed to validate this procedure.

Due to the increased prevalence of multiresistant pathogens (MRSA, VRE, Pseudomonas aeruginosa, ESBL-producing pathogens, etc.), broadly active, optionally combined antimicrobial therapy must often be started in order to adequately cover the spectrum of potential pathogens. Any previous antimicrobial treatment must be considered in therapeutic decisions.

The need for antimicrobial therapy should be established at latest by day 3 [116] and re-evaluated at least every 48 hours [479]. If feasible, combination therapy should be de-escalated to a therapy with a more narrow spectrum after 3 to 5 days and after microbiological data become available.

Given the physiologically and pharmacologically complex situation and the high volume of distribution in sepsis patients, high-dose therapy on the first day is needed to rapidly establish sufficiently high plasma levels. However, there is little study data supporting this plausible notion.

In the following days, dosage should be adjusted for organ (kidney and liver) function with consideration of potential drug-drug interactions.

Therapy can be guided by monitoring the procalcitonin (PCT) in the serum. In a study in severe sepsis patients [379], the duration of antibiotic therapy could be reduced by 3.5 days and the duration of stay in the intensive care unit was shortened by 2 days without any clinical disadvantages. The antibiotic was stopped when the PCT value had fallen by more than 90% of the initial value.

In order to be able to implement this strategy in the clinical routine setting, a close cooperation between the intensive care physicians and clinical infectiology and microbiology departments is required. The term “antimicrobial stewardship” [125] describes this interdisciplinary approach.

Microbiology and current resistance situation

The current recommendations for blood culture diagnostics are published within the framework of the “MIQ Guidelines“ (quality standards in the microbiological-infectiological diagnostics of the German Society for Hygiene and Microbiology, DGHM). The guidelines describe procedures for blood culture sampling, choice of access site, venipuncture, as well as sample transport and processing with and without automatic detection systems. Blood cultures samples should be taken before starting antibiotic therapy. The following procedures are recommended:

  • make a fresh puncture in a peripheral vein; use an indwelling catheter only for additional samples
  • disinfect hands of staff
  • wipe or spray an area of at least 5x5 cm with alcolhol-based disinfectant, wait for 1 min
  • disinfect the skin a second time from centre outwards with a sterile swab
  • put on disposable gloves
  • do not palpate the puncture site again
  • puncture the vein and take samples of (5–) 8 to 10 ml of blood per blood culture bottle, i.e. 16 to 20 ml per blood culture set
  • use at least two, better 3 blood culture sets
  • wipe the stoppers of blood culture bottles with alcohol-based disinfectant
  • wait until the disinfectant has dried
  • inoculate the blood culture bottles with fresh syringes (not inserted!) or a closed removal system (TRBA!)
  • do not ventilate the anaerobic bottles
  • transport the blood culture bottles to the laboratory immediately

Sepsis may involve a broad spectrum of potential pathogens. In the SEPNET study in Germany, 55% of the cases were caused by gram-positive bacteria, 54% by gram-negative microorganisms and almost 18% by Candida strains; these add up to more than 100% due to polymicrobial infections [159].

In the fourth PEG Blood Culture Study, with 14 laboratories all over Germany plus 1 centre in Austria participating, the distribution of the 7,652 pathogens detected from all clinically relevant blood culture isolates collected between July 1, 2006 and June 30, 2007 was as follows: 46.1% gram-positive pathogens, 46.7% gram-negative pathogens, 1.6% anaerobes and 5.6% fungi. The most frequently detected pathogen was Escherichia coli, followed by S. aureus. Enterococci, particularly Enterococcus faecium, as well as fungi showed the largest increase of prevalence.

The current resistance situation in blood culture isolates in Germany was also analyzed in the fourth PEG Blood Culture Study. According to these data, the proportion of methicillin-resistant S. aureus strains (22.8%) has increased significantly compared to the years 2000/2001. However, there was a large cross-center variation of 8.7% to 41%.

The proportion of glycopeptide-resistant E. faecium strains was 5.4%, which is in the same range as the rate of 6.3% found for Germany in the European Antimicrobial Resistance Surveillance Study (EARSS) of 2008.

The proportion of fluoroquinolone-resistant E. coli strains has increased to 31.2%. The proportion of cefotaxime-resistance as a marker of ESBL production was 7% in the current study. The fluoroquinolone resistance rate in Klebsiella pneumoniae and K. oxytoca increased to 17% and 15%, respectively. The rate of ESBL producers in K. pneumoniae increased to 14%, as measured by cefotaxime resistance.

For P. aeruginosa, the rates of resistance to ceftazidim and meropenem have remained stable at 14.4% and 11.8%, respectively. Ciprofloxacin resistance actually decreased slightly to 23.9%.

Pharmacokinetics and Pharmacodynamics

Investigations on pharmacokinetics and pharmacodynamics of antibiotics used in patients with sepsis remain limited to date. Pharmacokinetics are influenced by complex, partially antagonistic processes making antibiotic levels difficult to predict. In many patients, the early phase of sepsis is dominated by a hyperdynamic circulatory situation which may lead to enhanced clearance of renally eliminated antibiotics as compared to in healthy individuals. Capillary leakage may cause significant expansion of the extracellular space. These two factors result in unexpectedly low plasma level of hydrophilic and renally eliminated antibiotics which applies to most beta lactams and aminoglycosides. This effect is less pronounced for antibiotics with larger volumes of distribution, i.e. mostly intracellular accumulation. If organ function – primarily renal clearance – declines in the further course of sepsis, plasma levels will increase due to decreased elimination. This may lead to accumulation of usually ineffective but potentially toxic drug metabolites [303]. In addition, antibiotics with high protein binding are dissociated from plasma proteins by other drugs or changes in pH. For these reasons antibiotics with low protein binding and low potential toxicity should be chosen (e.g. for MSSA sepsis, cephalosporins should be used rather than flucloxacillin, with its high affinity to proteins (>90%) and a high risk of hepatotoxicity). Pharmacodynamics describes antimicrobial efficacy as a function of drug exposure parameters. For example beta lactams (penicillins, cephalosporins, carbapenems) are classified as time-dependent antibiotics, which means that the plasma level should remain above the MIC of the infecting pathogen as long as possible, while high plasma level spikes do not confer an advantage in terms of pathogen killing. Continuous infusions may improve the treatment results, particularly in severly ill patients with moderately susceptible pathogens [269], [312].

Group 1 carbapenems, with their postantibiotic effect against gram-negative pathogens and limited half-life at room temperature, are also suitable for prolonged infusions (3–4 hours), thus optimally exploiting the pharmacokinetic-pharmacodynamic (PK/PD) profile. In contrast, peak level dependent antibiotics such as aminoglycosides should be administered as a bolus dose once daily. This similarly applies for fluoroquinolones whose PK/PD profile is determined by the parameter AUC (area under the plasma concentration-time curve) >MIC. To allow for dose adjustments, an early measurement of plasma levels (i.e. therapeutic drug monitoring) is urgently recommended but is available only in exceptional cases.

Treatment recommendations

Almost all patients receive calculated initial antimicrobial treatment as recommended by the PEG (Paul-Ehrlich Society). For some patients, where information of pathogen susceptibility is available, the initial therapy may be modified accordingly.

The initial choice of antibiotic is influenced by the suspected source of infection, the underlying disease, the risk factors, whether an infection is community-acquired or nosocomial, the onset of the infection and whether the patient was previously treated with antimicrobials.

Table 26 [Tab. 26] shows treatment recommendations for an unknown pathogen, based on the type and origin of the infection. Table 27 [Tab. 27] lists treatment recommendations for known pathogens. The diversity of the treatment options in Table 26 [Tab. 26] and Table 27 [Tab. 27] stems from the different degrees of disease severity and various risk factors. Treatment duration should be in the range of 7–10 days. Exceptions are a slow response to treatment, failure of source control and immunosuppression [79].

Although the database is not sufficient, patients with life-threatening disease should always receive therapy with a combination regimen (Table 26 [Tab. 26]). This approach is supported by the results of the Surviving Sepsis Campaign [79]. Dellinger et al. recommended the administration of one or more drugs with broad spectrum of antimicrobial activity and good tissue penetration for calculated initial therapy [123].

A combination treatment is explicitly required if a Pseudomonas infection is suspected or detected [79], [84], [112]. Traditionally, aminoglycosides are the preferred combination partner of beta-lactam antibiotics. The option to use fluoroquinolones as combination partners for a beta-lactam is substantiated by the work of Paul [400], [401], pharmacokinetic advantages, lower toxicity and the fact that plasma levels need not be determined regularly, while direct treatment costs are higher. In view of the increasing rates of resistance to fluoroquinolones, fosfomycin is another option as a combination partner. It achieves good tissue penetration (Table 28 [Tab. 28]).

In sepsis patients, all antibiotics must be administered intravenously and at high doses. Neither intravenous to oral sequential treatment nor dose reductions have been documented in a study on this indication.

A lipopeptid (daptomycin) [116], [125], [159] or a glycopeptide should be added to the combination in high-risk patients with severe sepsis, septic shock or unknown focus of sepsis, particularly if local MRSA prevalence is high.

In sepsis with a focus in the respiratory tract, predominantly Streptococcus pneumoniae, various enterobacteriaceae and anaerobes from aspiration pneumonia must be anticipated. In high risk patients and those who have been hospitalized for more than 5 days, P. aeruginosa, Acinetobacter spp. and Stenotrophomonas maltophilia must be considered as potential pathogens. The pathogen spectrum can vary greatly from hopsital to hospital. A recent study has shown that gram-negative pathogens should be expected in patients on mechanical ventilation, even in those with shorter periods of hospitalization [187]. An oxazolidinone (linezolid) should be added to the combination for severe sepsis or septic shock in high-risk patients and in settings with high local rates of MRSA [125].

In cases of sepsis originating in the urinary tract without previous instrumental intervention, E. coli and Proteus mirabilis are the most likely pathogens. After urological surgery, other enterobacteriaceae, P. aeruginosa, enterococci and staphylococci must also be considered.

If the source of sepsis is the intestinal tract or a gynaecological organ, the following pathogens must be expected: enterobacteriaceae, anaerobes, enterococci, Pseudomonas spp., and S. aureus.

In a biliary sepsis, pathogen colonization of bile ducts and bacteraemia increase with the degree of bile duct obstruction. In cases with obstructive jaundice, pathogens are detected in the blood in more than 75% of patients. The spectrum of pathogen includes enterobacteriaceae, enterococci and anaerobes. Additional gram-negative pathogens including P. aeruginosa have been detected in postoperative bacteraemia, cholangitic sepsis, subhepatic abscesses and interventional surgery (ERCP or endoscopic papillotomy). For severe sepsis or septic shock originating in the intestines, gynaecological organs and bile ducts, a glycylcycline (tigecycline) can be added to the combination [123], [124], [125], [185].

If the focus of sepsis is located in skin or soft tissue, infections are mostly due to Streptococcus pyogenes, S. aureus (including MRSA) as well as mixed pathogens with the possilbe additional involvement of non-A streptococci, anaerobes, enterobacteriaceae and/or P. aeruginosa.

The pathogen spectrum of catheter-related sepsis includes coagulase-negative staphylococci, S. aureus, gram-negative rods, Candida spp., Corynebacterium jeikeium and propionibacteria. The lipopeptide daptomycin is an alternative to the use of a glycopeptide [116], [159].

The recommendation to use a single-agent regimen is based on the results of well-documented randomized clinical studies corresponding to evidence level I.

In general, there is insufficient evidence from clinical studies to support recommendations for combination therapies. These recommendations are based on expert opinion and are predominantly classified as evidence level IV. This is particularly true for combination treatments involving fluoroquinolones.


11 Bacterial endocarditis

Christoph Naber, Peter Kern, Eberhard Straube, Mathias Herrmann, Wolfgang Graninger

Endocarditis is an endovascular infection, mostly caused by bacteria. It affects the native heart valve or intravascularly implanted devices such as prosthetic valves or pacemaker electrodes. Despite substantial diagnostic and therapeutic advances, bacterial endocarditis is still a severe disease with a mortality rate of 20% to 30%. The exact incidence in Germany is not known. In France, it is in the range of 30 cases per million population [229]. The disease is categorized clinically according to severity as subacute or acute endocarditis. The former is more frequently caused by streptococci and enterococci, the latter by staphylococci.

Clinical diagnosis

Many cases of endocarditis are definitively diganosed and adequetely treated only after long delay of often over a month after the onset of symptoms [54]. The classical key symptoms are often difficult to assess (e.g. aggravated heart murmur), or rather unspecific (e.g. fever, loss of body weight, night sweats, exhaustion or myalgia). In many cases, the first symptoms already indicate complications, including progressive dyspnea as as sign of valvular destruction with significant regurgitation and volume overload. Septic embolisms from a heart vegetation may cause neurological conplications and are often the first symptoms.

The possibility of endocarditis must be considered in the differential diagnosis of even unspecific symptoms particularly in patients with risk factors, e.g. a prosthetic valve or intravenous drug abuse.

Echocardiography

After the clinical diagnosis endocardial involvement must be confirmed by further diagnostic workup which should include immediate transthoracic echocardiography to detect serious valve destruction or septic cardiomyopathy. Transoesophageal echocardiography (TEE) will be necessary in most cases. One single negative TEE does not rule out an infective endocarditis. If clinical suspicion remains, the TEE should be repeated after 6 to 10 days. After the diagnosis has been made, weekly echocardiography will be used to monitor the course of disease. Even in the absence of fever, local progression may result in a growth of vegetations or formation of abscesses or fistulas. In cases with clinical worsening, immediate examination is necessary to identify complications as soon as possible.

Pathogen identification

Besides echocardiographic imaging, the identification of the causative pathogen is vital for targeted therapy. It is of key importance to take blood cultures correctly before starting the antimicrobial treatment. At least three blood culture sets must be taken (aerobic and anaerobic) at different times from a peripheral freshly punctured vein after adequate disinfection. This should be carried out without regard to body temperature, as continuous bacteremia must be suspected. The most common cause for negative blood cultures is ongoing treatment with antibiotics. Therefore, in clinically stable patients who have already received an antibiotic before the diagnosis or who have had negative blood cultures, the interruption of an ongoing antibiotic therapy (>48 hours) before taking a blood culture should be considered. Another reason for a negative blood culture might be the presence of a microorganism which is difficult to culture. Therefore, it is important to notify the laboratory of the suspected diagnosis of endocarditis to ensure adequate analysis (e.g. sufficiently long incubation period). For instance, a serological analysis must be carried out for Bartonella, Brucella and Coxiella spp. Molecular biological methods to detect pathogens in EDTA blood samples using polymerase chain reaction (PCR) are available for clinical testing and could offer a workaround for the described diagnostic problems. However, their clinical validity in endocarditis has not been sufficiently evaluated to date.

The microbiological testing of surgically excised heart valve material is obligatory. Other than in the analysis of blood, the PCR can deliver directive results. It should be noted that the presence of bacterial DNA does not yield any information about the activity of the infection.

Diagnostic criteria such as the Duke criteria help in the categorization of findings. Particularly in cases with negative blood cultures, their sensitivity is often sufficient for infections of prosthetic valves or pacemaker electrodes or if the right side of the heart is affected. However, they can never replace a rational clinical judgement.

Essentials of endocarditis treatment

The antibiotic therapy, surgical treatment and management of complications are the three essential treatment approaches in bacterial endocarditis. For this reason, endocarditis is always treated by a team of physicians, consisting of cardiologists, infectiologists, microbiologists and heart surgeons. The prognosis for bacterial endocarditis depends on many factors which are, among others,

  • the source of the infection (nosocomial or community-acquired),
  • the underlying pathogen,
  • the local resistance situation, and
  • the presence of foreign material.

In general, cerebral complications are less frequent in right-sided endocarditis and the success rate of conservative management is usually higher than in left-sided endocarditis. An endocarditis involving a prosthetic valve usually needs surgical intervention more urgently and earlier than a native valve endocarditis. An infection with Staphylococcus aureus is usually more severe than a streptococcal infection.

Antibiotic therapy

If the general condition of the patient is critical, empirical antimicrobial treatment is started immediately but always after taking blood culture samples. In native heart valve endocarditis and late endocarditis after cardiac valve replacement (>1 year after surgery), methicillin-sensitive S. aureus strains (MSSA), various streptococci and Enterococcus faecalis are the most frequent pathogens (Table 29 [Tab. 29]). The microbiological results from the first infection may help in choosing the calculated treatment.

In early endocarditis after a valve replacement (<1 year after surgery), methicillin-resistant S. aureus strains (MRSA), coagulase-negative staphylococci and gram-negative bacteria are more frequently found as causative pathogens (Table 29 [Tab. 29]).

Any empirical treatment should be modified as necessary when the results of pathogen identification and antibiotic susceptibility are available. Treatment regimens for endocarditis are listed in Table 30 [Tab. 30] for the most frequent pathogens. Further detailed treatment recommendations as well as advice on the management of complications, indications for surgical intervention and antibiotic therapy after surgery are given in the guidelines of the European Society for Cardiology [208].

Evaluation of recommendations for treatment of endocarditis with antibiotics given by the European Society for Cardiology (ESC) in 2009

Considerable changes appear in the newly revised European guidelines, particularly regarding the treatment of staphylococcal endocarditis. The additional treatment of native valve endocarditis with gentamicin is recommended only as an option of new data [113], [164], [400] show significantly increased nephrotoxicity and no survival advantage. In addition, the expert committee of the PEG sees insufficient evidence for recommending a combination therapy with gentamicin for native valve endocarditis caused by streptococci with penicillin MICs<0.125 mg/l.

Gentamicin is usually administered once daily because kidney toxicity is reduced and efficacy increased by application of high doses in long intervals.

As increased efficacy is assumed, enterococcal endocarditis should be treated with a multiple-drug combination including an aminopenicillin. Increased efficacy is also assumed against frequently encountered strains with low-level resistance to gentamicin but not against those with high-level resistance [580].

When treating documented infections with methicillin-susceptible staphylococci, vancomycin should strictly be avoided in accordance with the ESC guidelines. Several studies showed that patients were less successfully treated with vancomycin than with a beta-lactam antibiotic [256], [501]. The beta-lactam antibiotics primarily recommended are still flucloxacillin, alternatively cefazolin or cefuroxim; a combination of aminopenicillin or ureidopenicillin plus beta-lactamase inhibitor (e.g. ampicillin/sulbactam) are not the drugs of choice for this indication.

When using potentially toxic agents such as vancomycin or gentamicin, monitoring of plasma levels and kidney function should be initiated. The revised ESC guidelines recommend a much higher therapeutic level for vancomycin than earlier guidelines (particularly for S. aureus endocarditis). The rationale is the prevalence of S. aureus strains with selectable subpopulations of bacteria showing reduced susceptibility to vancomycin, so-called hVISA (heterogenous vancomycin-intermediate S. aureus) [21], [31], [351], [487]. However, it should be stressed that the recommendation of a higher trough plasma level is based on expert opinion. The vancomycin trough plasma level should be in the range of 15–20 mg/L for infectious endocarditis according to the PEG expert committee.

In the revised guidelines, daptomycin [174] at 6 mg/kg/day IV is also recommended as an alternative to vancomycin in the treatment of staphylococcal endocarditis if methicillin resistant pathogens are involved. Higher dosages, e.g. 9 mg/kg/day, may be more effective but are not licensed.

In the current revision of the European guidelines, amoxicillin and ampicillin are often recommended almost as synonyms. Amoxicillin in contrast to ampicillin is not available as a monosubstance for IV administration in Germany. When given in combination with clavulanic acid, the high dosage level of amoxicillin recommended for enterococci endocarditis cannot be reached due to possible hepatotoxicity of clavulanic acid [4]. Thus, ampicillin at the appropriate high dosage level remains the drug of choice. The revised European guidelines also open the possibility of an IM (intramuscular) administration of an antibiotic. This is not recommended in Germany. The treatment should be administered strictly intravenously and patients should be hospitalized during initial treatment.

An out-patient treatment may be considered after 2 weeks of treatment in hospital and only for restricively chosen patients with clearly defined indication. The reason for this recommendation is that complications frequently occur in the first two weeks after starting antibiotic treatment and close monitoring must therefore be recommended even in an apparently favourable course of disease [15].

Surgical treatment and postoperative care

In general a surgical treatment should be an integral part of the therapeutic concept and not as a fall-back approach after failure of the antibiotic therapy. Conversely, eradicating bacteria in vegetations is often unfeasible before surgery and attempts to do so should not lead to a delay of surgical intervention in cases with a clear indication for an operation (e.g. abscess formation, progressive heart failure). Oral follow-up therapy after intravenous treatment according to guideline recommendations does not appear appropriate. Blood cultures are not only useful for diagnosis but also for monitoring therapeutic success. Blood culture samples should again be collected 2–4 weeks after after the end of antimicrobial therapy, with high vigilance for the occurrence of fever in the meantime.


12 Bacterial meningitis

Pramod M. Shah, Hans-Reinhard Brodt, Thomas A. Wichelhaus, Matthias Trautmann, Roland Nau

Acute bacterial meningitis is characterized by the clinical key symptoms of fever, headache and meningeal irritation (meningism). In addition confusion, reduced vigilance or coma may define the clinical picture [69], [143], [418], [421]. Acute bacterial meningitis is differentiated from viral meningitis. Overall, the incidence of acute bacterial meningitis is 5 to 10 cases per 100,000 population per year [467]. The pathogen spectrum depends on age, the most common pathogens in adults being pneumococci and meningococci. Since the introduction of active vaccination against Haemophilus influenzae type b, the formerly dominant pathogen of bacterial meningitis in children is declining [143], [467]. Other less frequent pathogens of acute bacterial meningitis include Listeria monocytogenes and, in meningitis associated with CSF-drainage and prior surgery, staphylococci, enterobacteriaceae and Pseudomonas spp. Meningitis due to a spread from craniofacial infections is caused primarily by pneumococci and other streptococci. Other infectious diseases with septic manifestations may also lead to CNS manifestation, e.g. Leptospira or Borrelia burgdorferi infections. Subacute or chronic meningitis syndrome is caused primarily by mycobacteria, Candida, Cryptococcus neoformans, Coccidioides immitis and Treponema pallidum. In patients with severe immunosuppression, atypical and rather subacute forms of meningitis may occur.

Diagnostics

Blood culture sample must be collected from all patients. Depending on the localization of a coexisting infection, it is essential to take throat swabs, samples of bronchial secretions, urine samples or wound smears.

The diagnosis of bacterial meningitis is confirmed by lumbar puncture and examination of cerebrospinal fluid (CSF). A granulocytic pleocytosis above 1,000 cells/µl, CSF protein levels above 100 mg/dl, lactate above 3.5 mmol/l and a liquor-serum-glucose quotient below 0.3 are potential indicators of meningitis. Methylene blue test and gram stains of the CSF sediment may provide evidence of the type of pathogen (gram-negative rods or cocci, gram-positive rods or cocci).

Helpful supplementary diagnostics include antigen detection from the spinal fluid, serum and urine (e.g. [319]), PCR from CSF (particularly tuberculous meningitis and/or evidence or exclusion of viral CNS infection), CRP/PCT determination in serum and differential blood analysis. Evidence of pathogen-specific antibody synthesis by determination of the CSF/serum antibody index is very important in subacute meningitis and encephalitis, particularly in neuroborreliosis [425].

In patients without impairment of consciousness and without focal neurological deficits, CSF and blood samples should be collected before the immediate start of antibiotic therapy [219], [264]. Subsequent imaging (cranial CT or MRI) and examination in collaboration with an ENT specialist help to localize the source of infection in the CNS region (e.g. sinuses, mastoid) and to recognize early intracranial complications. In patients with impairment of consciousness or focal neurological deficits, CSF samples should not be taken before cerebral imaging. However, blood cultures, urine samples, throat swabs and/or bronchial secretions (for evidence of pneumococci and meningococci) should be collected before an immediate start of antibiotic therapy [264].

Treatment

A delay in antibiotic treatment initiation is strongly associated with poor prognosis [24], [27]. Due to the pathogen spectrum of community-acquired bacterial meningitis, calculated initial therapy should be started (Table 31 [Tab. 31]) with a group 3a cephalosporin [406], [466] in combination with ampicillin (which is effective against Listeria monocytogenes). For nosocomial bacterial meningitis and infected CSF drainage devices, the empirical initial treatment should consist of vancomycin plus meropenem or vancomycin plus ceftazidim. If the pathogen can be identified, the treatment should be readjusted according to the results of the microbiological examination (Table 32 [Tab. 32]). As a rule, any infected drainage device must be removed and replaced by an external device. The minimum duration of treatment for unknown pathogens, H. influenzae or Streptococcus pneumoniae should be at least 10 days, and 7 days for meningococci. The antibiotic treatment of patients with meningitis due to Listeria, Staphylococcus aureus, Pseudomonas aeruginosa or enterobacteriaceae should last for 3 weeks.

The success of the antimicrobial treatment, except in meningitis caused by meningococci, should preferably be aided by a control puncture at latest after 48 hours [293]. Fever or an increase in pleocytosis in sterile CSF should not prompt changes in treatment. A final lumbar puncture at or after the end of treatment is not necessary if no complications occurred.

For pathogens with reduced susceptibility to antibiotics, an intraventricular antibiotic treatment might be necessary to eliminate the pathogens from the CNS. In Germany, no antibiotics are currently licensed for intraventricular administration and there are no randomized clinical studies showing an improvement in treatment results by intraventricular administration. The intraventricular administration of antibiotics is an experimental treatment. Table 33 [Tab. 33] shows antibiotics with published experience where intraventricular administration would be rational because of low CSF penetration and high systemic toxicity. Therapeutic monitoring via determination of CSF drug concentrations is recommended [586].

Adjuvant therapy with dexamethasone [421], [460], [466] improves the prognosis in community-acquired bacterial meningitis in adults, primarily in pneumococcal meningitis. In countries with high standards of medical care (diagnostics and therapy), dexamethasone is recommended to reduce mortality and to avoid long-term damage, particularly hearing loss [119], [532]. A 10 mg dose of dexamethasone (adults) is given 20 minutes or immediately before or with the first administration of an antibiotic and then every 6 hours for 4 days. In countries with low standards of medical care, the adjuvant treatment with dexamethasone is not recommended [341], [456]. As there are no data available for patients with nosocomial meningitis or for immunosuppressed patients with bacterial meningitis, the adjuvant treatment with dexamethasone is not recommended in these groups. There is insufficient experience for adults with other adjuvant strategies which were effective in animal experiments.

It has been shown that adjuvant use of corticosteroids improves the treatment results, particularly in severe tuberculous menigitis [419], [519]. Depending on the neurological deficits, adults and adolescents receive dexamethasone for 4 or 8 weeks (stage 2 or 3) as follows: 0.4, 0.3, 0.2 and 0.1 mg/kg IV per day in weeks 1, 2, 3 and 4, respectively, followed by 4, 3, 2, and 1 mg per day orally in weeks 5, 6, 7, and 8, respectively. Alternatively, prednisone may be started at 60 to 80 mg/day and tapered out over 4 to 6 weeks. To date, experience and studies also indicate a trend towards an improvement of results in children and immunosuppressed patients (HIV). However, the available data do not yet justify a general recommendation. Low-dose heparin is recommended for prophylaxis against thromboembolism and proton-pump inhibitors are used for gastric protection.

Prophylaxis

The most frequent pathogen causing meningitis after splenectomy is S. pneumoniae, followed by other encapsulated bacteria. Therefore, active vaccination against pneumococci, H. influenzae type b and meningococci should be provided before splenectomy (in patients with emergency surgery after the operation). With regard to other indications for vaccination against Haemophilus, pneumococci and meningococci, refer to the homepage of the Permanent Commission on Vaccination of the Robert Koch Institute (http://www.rki.de/nn_199596/DE/Content/Infekt/Impfen/impfen.html). Based on current resistance data, people in close contact to patients with meningococcal meningitis acquired in Germany should receive an antimicrobial prophylactic treatment. The prophylaxis can be administered up to ten days after the last contact with the patient. Recommended antibiotics are ciprofloxacin, rifampicin or ceftriaxone (http://www.meningococcus.uni-wuerzburg.de/startseite/berichte/daten_2008) [177]. Adults (except when pregnant) receive ciprofloxacin (a single dose of 500–750 mg orally) or alternatively rifampicin 600 mg every 12 hours for 2 days. Pregnant women receive ceftriaxone (a single dose of 250 mg IM). Children receive rifampicin (10 mg/kg body weight every 12 hours for 2 days orally). Rapid development of resistance to rifampicin (even in prophylactic treatment) has been reported [177]. Resistance to ciprofloxacin appears to increase in N. meningitidis isolated from patients from Southern and Western Europe and Southeast Asia [5], [480], [482]. If there is at least one unvaccinated child or a person with a relevant immunodeficiency among a group of people around a close contact to a patient with H. influenzae meningitis, they should receive antimicrobial prophylaxis up to 7 days after the last contact to the patient (https://www.rki.de/DE/Content/Infekt/EpidBull/Archiv/2002/Ausgabenlinks/30_02.pdf?__blob=publicationFile): adults (unless pregnant) receive rifampicin 600 g every 24 hours for 4 days orally; infants and children receive rifampicin 10 and 20 mg/kg body weight, respectively, every 24 hours for 4 days orally.

Addendum

In a recently published meta-analysis, the efficacy of adjuvant dexamethasone in bacterial meningitis was not confirmed [533].


13 Eye infections

Wolfgang Behrens-Baumann, Hermann O. C. Gümbel, Michael Kresken

Infections in ophthalmology affect the eyelids, orbita, conjunctiva, cornea, and the interior of the eyeball, as endophthalmitis, or more specifically retinitis or chorioiditis. Infections of the surface of the eye are best treated with topical antiinfectives for pharmacokinetic reasons. However, in cases of conjunctivitis caused by Chlamydia spp. and Haemophilus spp. it is often preferable to use a systemic antibiotic in addition to the topical therapy as these pathogens also colonize the urogenital tract and the nose and throat region [48].

Endophthalmitis is an inflammatory reaction to an intraocular infection by bacteria, fungi or (rarely) parasites. Inoculation occurs exogenously (postoperative, posttraumatic, spread from neighbouring tissues) or endogenously (bacteremia). The PEG recommendations follow the guidelines of the German-speaking Society for Intraocular Lens Implantation and Refractive Surgery [51], which formed the basis of the guidelines on prevention, investigation and management of postoperative endophthalmitis of the European Society of Cataract and Refractive Surgeons (ESCRS) [39].

The spectrum of pathogens encountered in eye infections is broad (Table 34 [Tab. 34]). Therefore, a diagnostic pars plana vitrectomy (ppV) is recommended [39]. This can be performed simultaneously to the injection of an intravitreal antibiotic. This mode of application yields the highest drug concentration directly at the site of infection and is therefore indispensable. However, sufficient concentrations of the active substance are maintained for a limited period of time only. While injection of antibiotics may be successful per se [402], it is often combined with a ppV. A dose of 1 mg/0.1 ml of vancomycin is recommended for infections caused by gram-positive pathogens [317]. This dosage results in concentrations above the MIC90 of Staphylococcus epidermidis for >48 hours [210]. Using 0.2 mg/0.1 ml vancomycin, therapeutically useful levels are maintained for about 3 to 4 days [181]. With regard to Gram-negative pathogens, it is increasingly recommended not to use aminoglycosides (due to their comparatively narrow antibacterial spectrum and the risk of retinotoxicity) [180], [237] but instead prefer 2 mg/0.1 ml of ceftazidime [97], [180], [237].

Although the Early Vitrectomy Study (EVS) [158] apparently indicates that systemic antibiotic treatment is unnecessary, it should nevertheless be performed. EVS tested the efficacy of intravenous ceftazidime plus amikacin for 5 to 10 days but no antibiotic with higher activity against gram-positive pathogens, e.g. vancomycin, was included. However, 38% of eyes with a final visual acuity of <5/200 were infected with Gram-positive bacteria. It is widely known that ceftazidime is less effective against Gram-positive bacteria than vancomycin [104], and amikacin does not penetrate into infected rabbit eyes [157]. All in all, based on the study design, the EVS cannot answer the question whether intravenous antibiotic treatment is useful [142], [498]. In fact, answering this question was not intended in the study, as its name already indicates (see [158]). Therefore Sternberg and Martin state: “intravenous antibiotic therapy is considered the standard of care” [498].

The main rationale of using additional systemic antibiotics is that sufficiently high levels of active drug are maintained for only a limited period of time after intravitreal injection. Since the clearance of antibiotics in the human vitreous has not been systematically investigated, systemic administration makes sense. Alternatively, intravitreal injection may be given every one to three days, according to animal studies [136].

In parallel to the intravitreal treatment, the same antibiotic could be administered intravenously, corresponding to level III of the Magdeburg Three Level Plan [49], [50] (Table 35 [Tab. 35]). This treatment scheme for infections with unknown pathogens, has become an established standard in the German-speaking countries and is regularly updated [50]. If there are contraindications to the drugs listed in level III, antibiotics from level II may be used. The antibiotics of levels II and III should always be given intravenously because of better pharmacokinetics and tolerability. Using high dosages is also recommended to overcome potential low-level resistance mediated by efflux pumps [471]. Due to the resistance situation in nosocomial infections, the fluoroquinolones are not used in the primary therapy of unknown pathogens (see chapter 2). If there is evidence of vancomycin-resistant enterococci (VRE), methicillin-resistant staphylococcus aureus (MRSA) or methicillin-resistant staphylococcus epidermidis (MRSE), daptomycin is available as a back-up alternative antibiotic [22], [395]. Ceftobiprole is a new treatment option, which albeit is available in Switzerland only [242], [380]. (Note added in the translated version: ceftobiprole had been introduced in Canada and Switzerland. Currently (November 2013), the drug is no longer available worldwide.) If the anterior eye chamber is involved in endophthalmitis, the use of group 3 and 4 fluoroquinolones (levofloxacin or moxifloxacin) eye drops every 30–60 minutes appear useful [52].

Glucocorticoids. Endophthalmitis is often characterized by invasion of leukocytes and monocytes from the peripheral blood mainly into the vitreous. These cells secrete proteases, cations and cytokines stimulating strong exsudation and infiltration, leading to “immune-associated damage” [245], [417]. This damage continues even after the pathogen has been eradicated. Glucocorticoids have cytoprotective effects including the inhibition of phospholipase activity, cytokine expression and monocyte adhesion. Therefore, steroids should be used in endopthalmitis (e.g. prednisone 200 mg/70 kg body weight). Dexamethasone (0.4 mg in 0.1 ml) is commonly injected intravitreally during the initial treatment.

Late endophthalmitis (after >2 weeks to several years) after a cataract operation is often caused by Propionibacterium acnes, less frequently by a fungus. The pathogens are usually localized within the intraocular lens (IOL) inside the cicatrized capsular sack. Thus, diagnostic and therapeutic access is difficult. A treatment attempt with systemic clarithromycin can be successful [384] and appears rational as the initial, less invasive measure. A dose of 2x500 mg per day is recommended. If the condition recurs, a ppV with opening of the posterior lens capsule is recommended [127]. Although flushing the capsule with vancomycin solution can be successful [6], [241], [462], many cases require surgical removal of the IOL and the capsular sack [6], [105].

The incidence of traumatic endophthalmitis (TE) is in the range of 5–14% [3], [429]. The symptoms of TE are the same as those observed in postoperative endophthalmitis. Immediate tissue sampling (vitreous) is required and samples sent for microbiological examination in the case of an open eyeball injury. In doing so, the microbiological-infectiological quality standards on microbiological diagnostics in eye infections must be observed [569].

Open eyeball traumas are associated with a particularly high risk of traumatic endophthalmitis. Therefore, antiseptic treatment and antibiotic prophylaxis – both topically and systemically – is clearly required, as periorbital structures (tear ducts, sinuses, etc.) which are not reached by antibiotic drops and creams are often affected by the trauma [46].

Based on a study carried out by Narang et al. in 70 patients with open eye injuries, the intravitreal administration of 1 mg/0.1 ml of vancomycin plus 2.25 mg of ceftazidime appears to be reasonable [360]. An international randomized study is currently investigating the role of these intravitreal injections as adjuvant treatment added to the systemic administration of intravenous moxifloxacin in low-risk (e.g. metal foreign objects) and high-risk (e.g. injuries in the agricultural industry, organic foreign material) patients [539].

The antibiotic treatment of acute traumatic endophthalmitis after primary wound closure follows level III of the Magdeburg three-level plan,

Endogenous endophthalmitis should be treated with targeted systemic antibiotic therapy (according to the result of blood culture diagnostics) in order to control the source of infection and bacteremia.


14 Antibiotic therapy in elderly patients

Peter Walger

Even if chronological age is of limited value to describe the probability of health problems, multimorbidity, the need for care and rates of dementia increase significantly beyond the age of 80 to 85 years. In general, the elderly or seniors are defined as people in the age of at least 65 years, while the aged or very old people are those of 85 years and beyond. While aging and illness are two separate phenomena, age-related health deterioration and age-correlated diseases are mutually interacting processes. Diseases with long latency, illnesses manifesting after decades of exposure to various risk factors, or chronic diseases aging with their carriers shape the medical implications of age as much as acute illnesses which are more frequent, have divergent symptoms and are more hazardous in old age.

Biological-physiological, medical and psychosocial parameters change with increasing age. Impaired immune function, reduced mobility, malnutrition, exsiccosis, and multiple chronic comorbidities commonly result in polypharmacotherapy with a high risk of drug interactions and increasing non-adherence.

Both the morbidity and the mortality of numerous infectious diseases increase with advancing age. Community-acquired pneumonia (CAP) is the most frequent infection-related cause of death in patients beyond 65 years of age. The incidences of many other infections such as urinary tract infections, sepsis, skin and soft tissue infections, bacterial endocarditis, cholecystitis and diverticulitis are elevated as well. Atypical clinical manifestations, e.g. reduced fever reaction, unspecific general symptoms or early impairment of brain functions impede the diagnosis leading to delays of adequate therapy [68], [196], [326].

Frequent side effects of antibiotics in the elderly

More than 30% of people >70 years of age have at least five chronic diseases [326], [342]. Extensive use of prescription and over-the-counter drugs as well as plant-based medicines is common in elderly people. In the USA, 25% of women in the age group of >65 years take 5 prescription drugs, 12% take at least 10.

Data for Germany are similar. People in the age group of >70 years take on average 3 different medicines per day. The 80–85-year-old individuals receive the highest number of drugs on a daily basis; 35% of the >70-year-old people take 5–8 and 15% take >13 different medications [195]. Polypharmacotherapy increases with age. Additionally, herbal medications or nutritional supplements are taken by 14% (1998) [250] or 26–27% (2002) [253], [359], respectively. According to treatment guidelines (USA 2005), a ficticious 79-year-old patient with five of the most common comorbidities (COPD, Type 2 diabetes mellitus, hypertension, osteoporosis, and osteoarthritis) receives 12 medications daily in a complicated regimen, with unpredictable interactions of diseases and/or medications, and with numerous adverse drug effects [76].

In general, side effects of medications are up to three times more prevalent in the elderly than in younger adulty around the age of 30 years [537]. Taking up to 5 medications entails a risk of adverse drug reactions (ADRs) in the range of 4%, 6–10 medications are associated with an ADR risk of 10%, and taking 11–15 medications increases the ADR risk to 28% [362]. Overall, ADRs occur in 15–35% of elderly patients. Adverse drug reactions cause 20–25% of geriatric hospital admissions. Anticoagulants, non-steroidal anti-inflammatory drugs (NSAIDs), antidiabetics, diuretics and digitalis glycosides are the drugs most commonly associated with ADRs leading to hospitalization [518]. Interactions play a role in approximately 40% of the ADRs. Low body weight in particular is frequently associated with ADRs. More than 80% of hospitalizations associated with ADRs are avoidable [53], [416].

Prevalence of inappropriate drug prescriptions

Data from the USA, Canada and Europe indicate a high rate of inappropriate use of medications in elderly patients; e.g. in the US the rate was 23.5% in 1994, 20% in 1996 (3% of these drugs were on the Beers list of 11 “always avoid” medications with increased risk of hospitalization and death), an 19% in 2002; in Europe approximately 20% in 2005. The typical patient receiving polypharmacotherapy with a high rate of inappropriate medications is a >85-year-old female, who lives alone, has a low health and social status [439], [583].

The Beers Criteria [44] provide a list of inappropriate medications categorized in three groups: “always avoid” (11 drugs), “rarely appropriate” (8 drugs) and “some indication but often misused” (14 drugs). Based on the criteria, a number of revised PIM lists (PIM= potencially inappropriate medications) were published in the USA, France, the Netherlands and Canada [43], [170], [290], [330]. The risk of adverse drug reactions leading to the hospitalization of elderly patients is strongly increased if several drugs are taken concomitantly (high risk of interactions). Polypharmacotherapy and prescription of neuroleptics or anti-dementia drugs are significant risk factors for ADRs in people living in retirement homes [215]. A list adapted to the situation in Germany „Potentially inadequate medications for elderly people“ is being prepared in cooperation with the Drug Commission of the German Medical Association [231].

The treatment of elderly patients with bacterial infections therefore typically means adding an antibiotic – with its own side effects and interaction potential – to a long list of various medications with partially unclear interaction potential and diverse side effects.

Prescribing antibiotics for the elderly

The available classes of antibiotics (or individual drugs) have been assessed according to specific aspects and risks:

1.
Risk of selection for Clostridium difficile
Hazardous antibiotics: cephalosporins, fluoroquinolones, ampicillin, amoxicillin, clindamycin.
2.
Neuropsychiatric side effects (encephalopathy, seizures, ototoxicity) and interactions with psychopharmacons
Hazardous antibiotics: fluoroquinolones, metronidazole, carbapenems (particularly imipenem), high-dose penicillins (particularly penicillin G), cephalosporins (particularly cefazolin), linezolid (serotonin syndrome), sulfamethoxazole, clarithromycin (psychoses), gentamycin, tobramycin, streptomycin, isoniazide. The neurotoxicity of aminoglycosides is actually a cochlear and vestibular toxicity, more rarely a neuromuscular blockade. The ototoxicity is enhanced by comedication with loop diuretics and vancomycin but also by loud ambient noise. Age is an important risk factor per se. For QT prolongation, see point 4).
3.
Interactions with other important drugs
Potentially hazardous antibiotics
– Anticoagulant therapy with phenprocoumon: cephalosporins, amoxicillin/clavulanic acid, cotrimoxazole, clarithromycin; ADR: increased INR
– Lipid reduction with CSE inhibitors (statins): macrolides (CYP-3A4 inhibition); ADR: rhabdomyolysis
– Antihypertensive therapy with calcium antagonists (dihydropyridines): macrolides (CYP-3A4 inhibitors); ADRs: hypotension, reflex tachycardia
– Bronchodilator therapy with theophyllin or caffeine: fluoroquinolones (CYP-1A2 inhibition); ADR: increased CNS stimulation
– Diuretic therapy with loop diuretics, anti-inflammatory treatment with NSAIDs, therapy with cisplatin or amphotericin B: enhaced nephrotoxicity of aminoglycosides, glycopeptides and beta-lactam antibiotics, enhanced ototoxicity of aminoglycosides and erythromycin
– Enzyme induction of hepatic CYP450 isoenzymes by rifampicin leads to decreased efficacy of calcium channel blockers, theophyllin, phenoprocoumon, contraceptives and phenytoin
– Erythromycin and clarithromycin inhibit the hepatic CYP3A system, potentially causing hazardous enhancement of toxicity of various drugs by inhibition of hepatic drug metabolism. This particularly affects many psychotropic drugs such as risperidon, clozapin and clomipramin; antiepileptics such as phenytoin, carpamazepin and valproic acid, as well as simvastatin and other statins, theophyllin, warfarin and some anti-HIV drugs. Azithromycin has the smallest interaction potential among the macrolides.
4.
QT prolongation
Potentially hazardous antibiotics: fluoroquinolones, macrolides and metronidazole, particularly in polypharmacotherapy in combination with beta-blockers, antiarrhythmic drugs, psychotropic drugs and phenytoin
5.
Nephrotoxicity
Potentially hazardous antibiotics: aminoglycosides, glycopeptides, cotrimoxazole, and nitrofurantoin. All beta-lactam antibiotics, particularly penicillins and cephalosporins, may cause interstitial nephritis in <1% of patients. Dosage levels of most antibiotics must be adjusted to impaired renal function; among the fluoroquinolones, ofloxacin/levofloxacin need dose-adjustements at a creatinine clearance of <50 ml/min, and ciprofloxacin at <30 ml/min
6.
Ototoxicity
Potentially hazardous antibiotics: aminoglycosides, erythromycin, less frequently clarithromycin (transient hearing loss due to high dosage or use in patients with renal impairment)
7.
Hepatotoxicity
Potentially hazardous antibiotics: rifampicin, combinations with clavulanic acid, isoxazolyl penicillins, erythromycin. In 1% to 10% of patients, fluoroquinolones cause a small increase in transaminases, which in most cases does not require treatment discontinuation or dose reduction. Due of a few severe, sometimes lethal hepatic complications, oral moxifloxacin was downgraded in 2008 to a reserve status after failure of other antibiotics in non-life-threatening infections such as bronchitis, sinusitis or community-acquired pneumonia. Therefore, monitoring liver enzyme levels is essential in patiens receiving fluoroquinolones. For some antibiotics, dose adjustments are only required in patients with severe liver dysfunction, these include macrolides, clindamycin, ciprofloxacin, minocyclin, metronidazole, linezolid, and tigecycline
8.
Drug rash, allergic reactions, hypersensitivity
50% of all allergic skin reactions to medications in hospitalized patients are caused by beta-lactam antibiotics. Depending on the time of appearance, these are divided into immediate (0–1 hour), delayed (1–72 hours) and late (>72 hours) [297]. The classification of Gell and Coombs [189] differentiates four immunopathological reactions: type I is mediated by IgE (anaphylaxis), type II is mediated by IgG or IgM (cytotoxic, cytolytic), type III is mediated by soluble immune complexes (immune-complex disease) and type IV is mediated by T-lymphocytes (delayed or T-cell-dependent hypersensitivity). In addition, there are other “idiopathic” types with unknown pathogenesis (e.g. maculopapular rash, Stevens-Johnson syndrome, and exfoliative dermatitis) [414].
9.
Malnutrition, nutritional deficiency and exsiccosis
Dose adustments according to renal function and physiological parameters
10.
Multiresistance induction or selection (collateral damage)
Hazardous antibotics if overused: cephalosporins, fluoroquinolones, carbapenems
11.
Rare side effects associated including those with high risk of morbidity
Hazardous antibiotics: fluoroquinolones (Achilles tendon rupture), linezolid (serotonin syndrome) if concomitant use of selective serotinin reuptake inhibitors (SSRI), monoamine oxidase inhibitors (MAOI), specific antidepressants, valproic acid, tryptophan, lithium salts, ritonavir [497].

Evaluation of kidney function in the elderly

The average renal blood flow decreases by about 10% per decade of age from 600 ml/min per 1.73 m2 in the fourth decade to about 300 ml/min per 1.73 m2 in the ninth decade. Accordingly, the glomerular filtration rate as well decreases by about 10% per decade. However, as the production of creatinine decreases with progressive loss of muscle mass with age, the serum creatinin level remains constant. Therefore creatinine levels in the upper normal range already indicate a reduced renal function. Increase in creatinin should be a critical factor when choosing the dosage of antibiotics. Many laboratories report the glomerular filtration rate based on the MDRD formula (MDRD= Modification of Diet in Renal Disease Study). However, this formula was not validated for people over 70 years of age in the MDRD study. The calculation of GFR by the Cockcroft-Gault formula also has significant limitations regarding higher age and large variations in body weight. In comparative studies, the most reliable GFR estimates were obtained by 24-hour collection of urine, even though there was a trend towards false high values. However, in practice, collection errors severely limit the utility of the method. Another alternative is the determination of cystatin C, which is independent of age and muscle mass. This method appears to be most reliable for incipient renal dysfunction without a detectable increase of creatinin levels. As all available methods of renal monitoring have significant limitations in elderly people, a potential overestimation of glomerular filtration rate should be compensated for by a restrictive use of potentially nephrotoxic substances [573].

Antibiotic resistance in the elderly

All risk factors promoting the colonization or infection by multiresistant pathogens become more prominent with age. Multimorbidity and specific comorbidities such as diabetes mellitus or COPD, prior antibiotic therapy, previous hospitalizations, care in nursing homes, rehabilitation facilities and other tertiary healthcare structures, presence of invasive devices, e.g. gastric feeding tubes, central intravenous catheters, tracheal cannula and urinary catheters. Other nosocomial risks such as dialysis, treatment of chronic ulcers and other long-term care requirements and pre-existing colonizations also accumulate with increasing age. The risk of multiresistance represents a major challenge in the selection of an adequate antimicrobial regimen, i.e. the choice of a broad-spectrum antibiotic or appropriate combinations. Any inappropriate treatment carries the risk of prolonged hospitalization, increased costs and, ultimately, increased hospital mortality [70], [263].

Summary

In principle, antibiotics can be used in the elderly following the same principles as in younger adults. No antibiotic must be generally regarded as inappropriate for older people.

However, the choice and the dosing of antibiotics must be adapted to the general medical problems and physiological changes of old age. At the same time the increased risk of resistant and multiresistant pathogens in the context of multiple hospitalizations and previous antibiotic therapies has become increasingly important during the last few years.

Because of the higher frequency and potentially serious consequences of antibiotic side effects in the elderly versus younger adults, the following measures are essential for treatment optimization: restrictive indication; choice of the optimal antibiotic, with respect to rare but harzardous side effects, choice of dosage, dosing interval and duration of treatment appropriate for the physiological status of the patient; and monitoring of efficacy and toxicity aiming at the early detection of expected and unexpected side effects. Any recommendations must reflect the special requirements of antibiotic therapy in elderly patients.


15 Pharmacoeconomics

Eva Susanne Dietrich, Egid Strehl, Katja de With, Wolfgang Kämmerer, Michael Wilke

The total expenditures of the German statutory health insurances in 2008 were €160 billion. Of that amount, €29 billion (18%) was paid for drugs used in out-patients and about €3 billion was for drugs used in hospitalized patients.

The price disparity between generics, established products and newly licensed medications is becoming ever larger while the evidence for patient-relevant use is heterogeneous.

In order to allocate the available financial ressources with the maximum possible effect on quality of care, avoid hidden rationing, and keep the available ressources and expeditures in balance, the decision-makers on the level of healthcare suppliers, insurance companies and administrations are increasingly looking for rationalization possibilites.

Important instruments are benefit and cost-benefit analyses of treatments and workflows, budget and health-impact analysis, as well as process optimization.

While treatment guidelines are based mainly on these aspects, doctors, lawyers and scientists are still debating whether or not economic evaluations of individual medications should be included in guidelines. Therefore the following recommendations do not focus on individual medications but rather on general measures to optimize treatments and processes.

Implementation of recommendations

Besides improving care and the compliance, guideline-based prescription may help to reduce the total cost of therapy and increase cost-effectiveness. This is particularly true in hospitals where numerous studies have produced evidence in favor of this approach (e.g. [133], [332]). In the office-based sector, there is considerable evidence of additional costs [131]. In many cases, extra costs are induced through increased treatment quotas (by lowering the intervention threshold), higher dosages, and better compliance of patients with chronic diseases. Cost savings may stem from avoidance of polypharmacotherapy and from adequate treatment duration in acute diseases.

Calculated antibiotic therapy

Microbiological diagnostics

Compared to the total cost of treatment in a hospital, the cost of microbiological diagnostics is small. Therefore a reduction in microbiological investigations should not be a primary target for cost-saving efforts, particularly in hospitals. For example, a rapid antibiotic susceptibility test may help to significantly reduce the costs of antibiotics as well as entire treatment costs while reducing mortality through early adjustment of antimicrobial therapy according to the test results [135]. In studies, 8–20% of patients with sepsis did not receive appropriate antibiotic therapy, even though the culture results including an antibiogram were fully available [75], [107]. Active communication of blood culture results with meaningful interpretation of the antibiogram by microbiologists or infectious disease specialists may improve the prescribing quality of antimicrobial treatment and thereby reduce costs and length of hospitalization [75], [94], [172].

Step-down therapy

Effective oral antibiotics enhance the opportunity to switch patients from parenteral to oral treatment (step-down therapy) even in hospitalized patients and enable continuation of treatment as an oral follow-up out-patient treatment after discharge. In principle the oral follow-up antibiotic is not necessarily identical to the parenteral drug or even from the same substance group. The choice rather depends on the indication and the expected pathogen spectrum. High bioavailability of the oral drug is particularly advantageous.

Compared to the oral only treatment, the step-down therapy has the advantage of rapidly reaching high drug levels by initial IV administration. Depending on the clinical course of infection, the treatment may already be changed to a less expensive oral follow-up therapy after 1 to 3 days.

Numerous clinical and economic studies have shown that a step-down therapy and fully intraveneous therapy are equivalent in many indications, while the step-down approach is significantly more economical (e.g. [206], [333], [355], [392], [547]).

An early switch to oral therapy effectively shortens the length of hospitalization [234], [329], [430]. This is very important given the current paradigm of hospital financing of lump payments according to diagnosis-related groups (DRG) in Germany and other countries.

Other reasons for the economical advantage of step-down therapy versus fully parenteral administration include:

  • lower antibiotic costs
  • lower staff costs for preparation and administration
  • earlier mobilization of patients and shorter hospitalization
  • fewer complications by intravenous injections application.

In cases with pharmacokinetically equivalent formulations and adequate absorption, the oral application should be preferred over parenteral use. If an oral therapy is prescribed, equivalent drugs with longer half-life are preferred because of the better compliance.

Switching to an oral administration requires the following:

  • reduction of infection parameters, particularly fever for 24–48 hours, CRP, leukocytosis with leftward shift
  • reduction in clinical signs, e.g. reduction in coughing, sputum production and thoracic pain in respiratory infections
  • significant improvement of general health condition
  • appropriate oral treatment available
  • abiity to swallow and tolerate oral medication
  • no impairment of intestinal absorption.
De-escalation

Besides step-down therapy, de-escalation may contribute to the optimization of the clinical-economical balance. The principle of this treatment strategy is the initial calculated use of a highly effective broad-spectrum antibiotic followed by an equally effective but more targeted therapy with a narrower spectrum after availability of microbiological findings. De-escalation should minimize the selection pressure with respect to multiresistant bacteria (e.g. Klebsiella spp. with extended-spectrum beta-lactamases (ESBL), Stenotrophomonas maltophilia, Bacteroides spp. producing metallo-beta-lactamases) or fungi [440].

Reducing the spectrum of therapy is usually associated with a reduction of cost. Prerequisites for de-escalation include:

  • availability of specific microbiological results,
  • clinical improvement (see above).

There is good evidence for the equivalence of a de-escalation treatment and other treatment concepts (e.g. [145], [476]).

Monotherapy

As studies on various indications have shown, monotherapies with modern substances are as effective as combination therapies which often result from polypragmatism (e.g. [138], [408]). Monotherapies have the following advantages:

  • fewer side effects
  • exclusion of potential antagonism between antiinfectives
  • fewer pharmacokinetic interactions
  • lower drug and subsequent costs.

Once-daily dosage

Antibiotics with a long biological half-life (4 to 6 hours) and a long-lasting postantibiotic effect enable once-daily dosage, potentially saving costs for staff and materials. In addition, side effect rates could be lower.

Antibiotic cycling and diversity

Increased use of antibiotics may cause increased selection pressure resulting in the development of resistance which in turn leads to increased costs. Cycling strategies try to minimize the selection pressure thereby reducing or preventing the development of resistance by transient removal and re-introduction of a given antimicrobial drug or class. Studies testing cycling strategies are difficult to implement. Due to the complexity of study protocols and the related difficulty of consistant implementation, only one substance or antibiotic class is switched by schedule. The mathematical results of published studies on the effects of cycling strategies on costs or use of drug as well as mortality, duration of hospitalization and resistance rates are scantly meaningful or poorly reliable for the above reasons. Consequently, it remains unclear whether this treatment concept is able to favorably influence resistance development and subsequent costs. In addition, models on the minimization of resistance development show that cycling strategies are probably less effective than antibiotic diversity, i.e. the simultaneous use of different antimicrobials in terms of heterogeneous prescribing on the same ward.

Therapeutic drug monitoring (TDM)

Therapeutic drug monitoring may have various goals:

  • establish a rational basis for a formerly empirical dosage [527]
  • identify reasons for failure of a rational therapy in a specific case
  • avoid severe and/or persistent side effects (with respect to morbidity/mortality) of drugs with a narrow therapeutic range
  • avoid unnecessary drug expenses and other follow-up and incidental costs within the DRG reimbursement system by determining the optimal dose and treatment duration for expensive drugs (e.g. parenteral antifungals) thus saving additional cost of underdosing, overdosing (in this case mostly harmless) or unnecessarily long duration of treatment [397].

Drug monitoring should absolutely be cost-effective. That means that any savings derived from the reduction of drug consumption and the reduction of side effects and their treatment must be greater than the total cost of equipment, substances and staff time used in drug monitoring [524].

The following aspects should be kept in mind when considering TDM:

  • What percentage of patients remain free of side effects with a suitable TDM?
  • What percentage of samples for a TDM are taken at exactly the optimal time?
  • In what percentage of patients can the serum concentration level be adjusted and maintained at a pre-defined therapeutic target range by means of TDM?

There is high-level evidence that TDM is cost-effective even in treatments with the relatively low-priced aminoglycoside antibiotics [534].

With a lower level of evidence, it has also been shown that a TDM of vancomycin is cost-effective by reducing the nephrotoxicity e.g. for patients in intensive care or with malignant diseases or those simultaneously taking potentially nephrotoxic medications [117].

Prolonged infusion instead of bolus administration or short infusion of meropenem and other carbapenems may be more costeffective and advantageous in achieving adequate penetration in the respiratory tract as necessary, for example, in cystic fibrosis patients. Here a TDM will result in both economic and therapeutic benefits [527].

Optimization of procedures and costs

An important instrument in the optimization of procedures and costs is the establishment of clinical treatment protocols and/or of standard operating procedures (SOP). As part of the treatment protocol, standards of medication use must be developed, established and evaluated. Because of their impact on cost but also on the quality and success of patient care, antiinfectives are a very important drug group to be considered in treatment standards and SOPs.

An important criterion for the choice of appropriate antiinfectives for the treatment protocol/procedure is an economic-pharmacoeconomic analysis of treatment alternatives from the hospital perspective. This analysis should include an examination of the procedure costs of an antiinfective treatment from purchase to application in the patient. Another important question is to what degree the administered antiinfective fulfills the aspects of quality management and assurance, procedure management, patient orientation and staff orientation. Besides the purchase price of relevant alternatives, the following parameters must be included in the procedure analysis:

  • Staff time and expenditure per administration, as a reduction in dosing frequency is favorable under DRG conditions (higher performance, less staff time). The key parameter is the staff cost per administration which in the literature ranges between 2 to 4 Euros or US Dollars per dose [476], [536].
  • The cost of drug application material such as needles, catheters, infusion equipment, etc. These costs range from as €1 to €4 by type of administration in the literature [536].
  • A lower error rate. Investigations and the resulting recommendations for Anglo-Saxon countries showed that a lower dosing frequency and simpler preparation reduce the probability of errors in the use of a drug [114]. The number of preparation steps should also be taken into account. Therefore, whenever possible, ready-to-use preparations should be employed.
  • The risk of confusion/mix-ups.
  • The cost of necessary monitoring.

Table 36 [Tab. 36] shows key properties of antibiotic therapies affecting the total costs of treatment.

Useful literature

Cost-effectiveness, budget-impact and health-impact analyses are valuable decision aids to identify the best treatments among the numerous options from both a patient and a budget perspective.

Both physicans and pharmacists are increasingly confronted with pharmacoeconomic studies reporting positive results for new products. It is therfore necessary to evaluate the quality of these investigations, e.g. based on the suggestions for planning, conduct and publication of pharmacoeconomic evaluations of antiinfectives given by the working group “Pharmacoeconomics of Antiinfectives” [132].

Alternatively, information on the evaluation of drugs (e.g. health-technology assessments) may be obtained from recognized institutions such as the National Institute for Health and Clinical Excellence (http://www.evidence.nhs.uk/default.aspx), the Institute for Quality and Cost-effectiveness in Health Care (http://www.iqwig.de/publikationen-des-iqwig.114.html) or the Canadian Agency for Drugs and Technologies in Health (http://www.cadth.ca/en/products/health-technology-assessment). Besides detailed presentation of available evidence and assessment of its quality, these institutions usually provide evaluations of the cost-effectiveness of medications under consideration, in part based on their own economic studies. Because of the increased networking among international HTA agencies and an advanced standardization of methods, it is increasingly rare that the conclusions of the HTAs differ.

Information on pharmacoeconomics as well as individual clinical trials and HTAs can be obtained from the databank of the NHS Centre for Review and Dissemination (http://www.crd.york.ac.uk/crdweb/). Here, the studies are described in much more detail than on Medline. Based on a comprehensive table of approximately 30 criteria, study goals, design of clinical and economic parts of the studies as well as their clinical and economic results are presented clearly and in detail. In addition, a short evaluation of the quality of the study is provided.

Future prospects

Pharmacoeconomic analyses are an important basis for benefit-dependent reimbursement in many countries and thereby set the legal framework for physician therapeutic decisions. Future clinical practice guidelines will increasingly consider the economic and legal framework and critically deal with the cost-effectiveness of drugs. This will differentiate them from systematic reviews or textbooks that evaluate the evidence with respect to quality and efficacy.

Furthermore, pharmacoeconomic analyses also offer an opportunity, as they combine a critical, evidence-based evaluation of the patient-relevant benefit with economic considerations thus rendering these aspects inseparable. If these analyses are more strongly involved in decision making, the outcome will be an enhanced selection of high-quality medications which are better adapted to patient needs.


16 Perioperative antibiotic prophylaxis

Hannes Wacha, Udo Hoyme, Rainer Isenmann, Peter Kujath, Cordula Lebert, Kurt Naber, Bernd Salzberger

Perioperative prophylaxis in surgical interventions is usually a short-term, single-shot administration of an antibiotic shortly before, at the start of or at latest during the intervention to avoid postoperative wound complications. The prevention of other types of postoperative infections such as sepsis, pneumonia, urinary tract infections (except for urological surgery) or meningitis has been targeted and differentially examined in only a few studies to date. Data on these infections have been mostly derived from studies conducted with the goal of investigating the effects on wound infection rates. Antibiotic prophylaxis is complementing rather than replacing evidence-based hygiene measures for the prevention of postoperative infections.

The indication for perioperative prophylaxis is based on the wound classification and additional risk factors of the patient. It is carried out for all patients with “contaminated” or “dirty” wounds, independent of other factors. The administration of antibiotics is also established in aseptic surgery with implantation of foreign material. In “clean” or “clean-contaminated” surgery or wounds, the indication should be validated by the presence of risk factors. Antibiotic prophylaxis is recommended in surgical interventions where infectious complications have particularly severe consequences (e.g. intracranial surgery) independent of risk factors and scientific evidence from clinical studies.

The essential features of perioperative prophylaxis are summarized as follows:

  • Antibiotic prophylaxis and its indications are established along with measures of asepsis.
  • The primary goal of perioperative prophylaxis is the reduction of wound infection rates. The secondary goal is avoidance of local and systemic postoperative complications due to infection.
  • Prophylaxis should be customized to the individual patient and the involved risks.
  • The risk begins with the operation. Effective levels of the antibiotic agent should be ensured for the duration of the risk period. In cases with persistent risk, the drug level must therefore be maintained according to the pharmacokinetic properties.
  • Premature application of an antibiotic is useless. The continuation of prophylaxis beyond the operation as a preventative treatment requires special indications.
  • The risk profile and regional epidemiology must be taken into consideration when choosing the antibiotic. Special attention must be given to possible secondary infections, particularly those caused by gram-negative pathogens.
  • Only substances which have been proven effective in the relevant indications should be used. The broadest experience is available for beta-lactam antibiotics.
  • The choice of the prophylactic drug should be based primarily on the pathogen spectrum, the pharmacokinetics and the licensing status.
  • For the individual patient, the risk of resistance development is small. However, this is not true for the entire patient population of a hospital.
  • The economic aspects are important, even if the cost of antibiotic prophylaxis is less than the cost of postoperative infectious complications.

Table 37 [Tab. 37] gives an overview of recommendations for perioperative prophylaxis in selected surgical indications. A detailed account is given in the full version of this chapter in the Chemotherapy Journal [548].

The listed substance groups and substances have been shown to be effective in clinical studies. Because of the large number of choices in some substance groups, individual evidence grades were not assigned for each single option and in individual cases analogous conclusions were allowed.

Available meta-analyses and prospective randomized studies on antibiotic prophylaxis in surgery often vary in quality and sometimes group different surgical interventions under the general term of abdominal surgery and allow for different substances, with the effect of sometimes less definitive recommendations. Therefore apparently equivalent studies were given recommendation levels ranging from A to C by the experts.


Notes

Origin

This guideline is the translated version of: Bodmann KF, Grabein B; Expertenkommission der Paul-Ehrlich-Gesellschaft für Chemotherapie e.V. Empfehlungen zur kalkulierten parenteralen Initialtherapie bakterieller Erkrankungen bei Erwachsenen: Update 2010. Chemother J. 2010;19(6):179-255. Available from: http://www.chemotherapie-journal.de/archiv/artikel/2010/06/186.html

Competing interests

Potential conflicts of interest were recorded with the following questions:

1.
Have you received fees for consulting or participation in an advisory board?
2.
Do you have financial ties to companies (e.g. pharmaceutical companies)?
3.
Have you received fees for lectures or support for conference attendance (e.g. travel costs), etc.?
4.
Have you received research aid (“grants”) of nonprofit organizations or public institutions?

Specifically, the authors reported the following potential conflicts of interest (both alphabetical listing of companies/institutions):

  • D. Adam: 1. Pfizer/Wyeth (Prevenar Advisory Board); 3. Pfizer
  • B. Al-Nawas: 1. Sanofi-Aventis; 3. Astratech, Camlog, Roche, Stravmann; 4. ITI Foundation, Krebshilfe, Osteology Foundation
  • K. Becker: 3. Pfizer (lecture fees, travel cost support); 4. BMWi KF 2279801AJ9
  • W. Behrens-Baumann: 3. Alcon, AMO (Abbott Medical Optics), Bausch & Lomb, Hoya, Polytech; 4. Alcon, AMO, Bausch & Lomb, Hoya, Polytech
  • K.F. Bodmann: 1. Bayer, Brahms, Novartis, Pfizer, Wyeth; 3. Bayer, Brahms, Fresenius, Janssen-Cilag, Lilly, MSD, Novartis, Pfizer, Wyeth
  • H.R. Brodt: 1. Bristol-Myers Squibb, Novartis; 3. Bayer Vital, Essex Pharma, Gilead Sciences, Janssen-Cilag, Pfizer Pharma, Wyeth Pharma; 4. Bundesministerium für Bildung und Forschung (BMBF), DG SANCO (Generaldirektion Gesundheit und Verbraucher) of EU-Commission
  • H. Derendorf: 1. Johnson & Johnson; 3. Pfizer, Wyeth
  • K. de With: No conflicts of interest
  • E. S. Dietrich: 1.-4. AOK Bundesverband Deutschland, Aventis Deutschland, Bayer AG Deutschland/Schweiz, Bundesamt für Gesundheit (BAG) Schweiz, GlaxoSmithKline Deutschland, IKK-Landesverband Westfalen-Lippe Deutschland, Pfizer GmbH Deutschland, Wyeth Pharma GmbH (all in the context of the activities at Techniker Krankenkasse and Healthecon AG)
  • B. Drewelow: 1. Janssen-Cilag, Novartis, Pfizer; 3. Astellas, AstraZeneca, Bayer, Boehringer-Ingelheim, Janssen-Cilag, Novartis, Pfizer
  • Ch. Eckmann: 1. Novartis, Pfizer; 3. Bayer Vital, MSD, Novartis, Pfizer
  • W. Elies: No conflicts of interest
  • P. Federspil: No conflicts of interest
  • U. Frank: 2. Occasionally third party funded projects: Astellas, Astra, Cilag, Novartis, Pfizer, Schwabe; 3. AOK Freiburg, AstraZeneca, Bayer Vital, Diakoniestation Trossingen, Forum Rottweil, GlaxoSmithKline, Landesapothekerkammer Baden-Württemberg, Herzzentrum Bad Krozingen, Pfizer, Repha, Spirig Pharma, Vivantes; 4. EU-Commission (DG Sanco), EU-Project Burden, Implement
  • R. Fünfstück: 1. MSD, Novartis; 3. Kirchheim-Verlag, Lilly Pharma Deutschland GmbH, MSD, Novartis Pharma, Rosen-Pharma.
  • B. Grabein: 1. Astellas, Gilead, Novartis, Pfizer; 3. AstraZeneca, Bayer, BectonDickinson, Biomérieux, Essex, Gilead, Janssen-Cilag, MSD, Novartis, Pfizer, Wyeth
  • W. Graninger: 3. Abbott, Actavis, Aesca, Astellas, AstraZeneca, Bayer, Leo, Novartis, Orphan, Pfizer, Sandoz, Sanofi, Wyeth
  • G. Gross: 1. GlaxoSmithKline, Sanofi Pasteur MSD; 3. Abbott, Sanofi Pasteur MSD
  • H. Gümbel: No conflicts of interest
  • M. Herrmann: 1. Astellas, Novartis; 2. Bayer Vital, MSD, Novartis, Pfizer, Wyeth; 3. Bayer Vital, Novartis, Pfizer; 4. BMBF, Deutsche Forschungsgemeinschaft, Saarland
  • G. Höffken: 1. Brahms, Lilly, Novartis, Roche; 3. Bayer Schering, Boehringer Ingelheim, Brahms, Janssen-Cilag, Lilly, MSD, Novartis, Nycomed, Pfizer, Roche, Sanofi-Aventis, Wyeth; 4. Brahms, Lilly, Novartis, Roche
  • R. Höhl: 1. MSD; Novartis, Pfizer; 3. Astellas, Bayer Vital, Brahms, Essex, Gilead, Lilly, MSD, Novartis, Pfizer, Pulsion, Wyeth
  • U. B. Hoyme: 2. 2 patents (Merete Medical GmbH); 3. through HELIOS-contract (Abbott, AstraZeneca Onkologie, Bayer Schering, Chugai, Gramanns Veranstaltungs- und Tagungsorganisation, Inverness Medical, Jenapharm, Dr. Kade, Novartis, Strathmann, Dr. August Wolff)
  • R. Isenmann: No conflicts of interest
  • M. Kaase: No conflicts of interest
  • W. Kämmerer: 1. Boehringer Ingelheim, Fresenius-Kabi, Pfizer, Pharmallergan; 4. Amgen, Bayer, Fresenius-Kabi, Gilead, Johnson & Johnson, MSD, Novartis, Pfizer, Sanofi-Aventis
  • P. Kern: 1. Johnson & Johnson; 3. Novartis, Tibotec; 4. Bundesministerium für Forschung (BMFT), Deutsche Forschungsgemeinschaft (DFG)
  • E. Kniehl: 3. Instrumentation Laboratory
  • J. Knobloch: 3. Bayer, Janssen, Novartis, Pfizer
  • W. Krüger: 1. Novartis, Wyeth; 3. AstraZeneca, Janssen-Cilag, Novartis, Pfizer, Wyeth
  • P. Kujath: 1. Astellas, Pfizer; 3. Bayer, Novartis, Pfizer
  • C. Lass-Flörl: 1. Gilead Sciences, MSD, Pfizer, Schering Pflough; 2. Astellas Pharma, Gilead Sciences, MSD, Pfizer, Schering Plough; 3. Astellas, Gilead Sciences, MSD, Schering Plough; 4. Austrian Science Fund, MFF Tirol
  • C. Lebert: 3. Essex, MSD; Pfizer
  • H. Lode: 1. Bayer, GSK, Janssen, Oti, Novartis, Pfizer, Protez; 3. Basilea, Bayer, Pfizer
  • J. Majcher-Peszynska: 3. Bayer, Pfizer; 4. BMBF
  • Ch. Naber: 1. Astellas, Novartis; 3. Astellas, Bayer, Novartis; 4. Bayer
  • K. G. Naber: 1. Bionorica, Merlion, OM Pharma, Pierre Fabre, RosenPharma, Zambon; 3. Daiichi-Sankyo, Janssen-Cilag, Johnson & Johnson, OM Pharma, Pierre Fabre, Sanofi-Aventis
  • R. Nau: 2. Bayer, Pfizer; 3. to a small extent (ASC Dienstleistungs GmbH, Bildungszentren Klinikum Braunschweig, Ärzte-Seminare Karlsruhe, Polizei-Akademie Niedersachsen); 4. Else Kröner-Fresenius-Stiftung, European Commission (project CARE-PNEUMO)
  • M. W. Pletz: 1. Sandoz, Wyeth; 3. AstraZeneca, Bayer, Brahms, Gilead, MSD, Novartis, Pfizer; 4. ARGUS, BMBF, CAPNETZ, DFG, Fraunhofer
  • A. Pross: 1. Bayer, Pfizer; 3. Novartis, Pfizer, Wyeth
  • A. C. Rodloff: 1. MSD; Wyeth; 3. Essex, Janssen-Cilag, MSD, Novartis, Pfizer, Sanofi-Aventis, Wyeth; 4. RKI
  • B. Salzberger: 1. Novartis, Wyeth; 3. AstraZeneca, MSD, Novartis, Pfizer, Wyeth; 4. Universität Regensburg
  • F.-J. Schmitz: 3.lectures: AstraZeneca, Bayer, Johnson & Johnson, MSD, Pfizer, Wyeth
  • P. M. Shah: 1. Janssen-Cilag, Novartis, Sanofi-Aventis
  • F. Sörgel: 3. Pfizer (lecture), Sandoz (lectures and clinical studies), Siemens (lecture), Wyeth (lectures and analytical works)
  • R. Stahlmann: 2. Third party funding of a master’s degree program in toxicology (Bayer-Schering, Berlin-Chemie, Merz, Nycomed)
  • E. Straube: 3. Reimbursement of travel costs for the consensus conferences by PEG; 4. research funds of BMBF (for projects not connected to the above topic/article)
  • E. Strehl: 3. Pfizer, Sanofi-Aventis; 4. Pfizer (financing a graduate pharmacist for a project in clinical pharmacy)
  • C. Sunderkötter: 1. Actelion, Infectopharm, Pfizer, Wyeth; 3. Actelion, Essex, MSD, Novartis, Pfizer, Wyeth; 4. Actelion
  • M. Trautmann: 3. Bayer Healthcare, 3M, Pall, Pfizer, Wyeth
  • H. Wacha: 1. Novartis
  • F. Wagenlehner: 1. Astellas, Janssen-Cilag, Pierre Fabre, Rosen-Pharma; 3. Bayer, OM Pharma, Pfleger, Sanofi-Aventis, Serag Wiessner; 4. Land Hessen – Landes-Offensive zur Entwicklung Wissenschaftlich-Ökonomischer Exzellenz (LOEWE)
  • P. Walger: 3. Lecture fees from Astra-Zeneca, Novartis, Pfizer, Sanofi-Aventis, Wyeth, support for conference attendance from Pfizer, Wyeth
  • Ch. Wenisch: 1. Wyeth; 2. Baxter, Bayer, GSK, Novartis, Pfizer, Wyeth; 3. Bayer, GSK, Pfizer; 4. AGES
  • T. A. Wichelhaus: 1. Almirall, Novartis; 3. Biomérieux, Novartis, Pfizer
  • M. Wilke: 1. Novartis, Pfizer, Wyeth; 3. MSD, Novartis, Wyeth




References

1.
Aaberg TM Jr, Flynn HW Jr, Schiffman J, Newton J. Nosocomial acute-onset postoperative endophthalmitis survey. A 10-year review of incidence and outcomes. Ophthalmology. 1998 Jun;105(6):1004-10. DOI: 10.1016/S0161-6420(98)96000-6 Externer Link
2.
Abrahamian FM, Deblieux PM, Emerman CL, Kollef MH, Kupersmith E, Leeper KV Jr, Paterson DL, Shorr AF. Health care-associated pneumonia: identification and initial management in the ED. Am J Emerg Med. 2008 Jul;26(6 Suppl):1-11. DOI: 10.1016/j.ajem.2008.03.015 Externer Link
3.
Affeldt JC, Flynn HW Jr, Forster RK, Mandelbaum S, Clarkson JG, Jarus GD. Microbial endophthalmitis resulting from ocular trauma. Ophthalmology. 1987 Apr;94(4):407-13. DOI: 10.1016/S0161-6420(87)33447-5 Externer Link
4.
Agarwal VK, McHutchison JG, Hoofnagle JH; Drug-Induced Liver Injury Network. Important elements for the diagnosis of drug-induced liver injury. Clin Gastroenterol Hepatol. 2010 May;8(5):463-70. DOI: 10.1016/j.cgh.2010.02.008 Externer Link
5.
Alcalá B, Salcedo C, de la Fuente L, Arreaza L, Uría MJ, Abad R, Enríquez R, Vázquez JA, Motgé M, de Batlle J. Neisseria meningitidis showing decreased susceptibility to ciprofloxacin: first report in Spain. J Antimicrob Chemother. 2004 Feb;53(2):409. DOI: 10.1093/jac/dkh075 Externer Link
6.
Aldave AJ, Stein JD, Deramo VA, Shah GK, Fischer DH, Maguire JI. Treatment strategies for postoperative Propionibacterium acnes endophthalmitis. Ophthalmology. 1999 Dec;106(12):2395-401. DOI: 10.1016/S0161-6420(99)90546-8 Externer Link
7.
Al-Nawas B, Grötz KA, Brahm R, Maeurer M, Wagner W. Infektionen im Mund-, Kiefer- und Gesichtsbereich: Was hat sich in den letzten 25 Jahren geändert? Dtsch Zahnarztl Z. 2000;55:765-9.
8.
Al-Nawas B, Grötz KA, Piesold J. Leitlinien der Deutschen Gesellschaft für Mund-, Kiefer- und Gesichtschirurgie – Osteomyelitis S2. AWMF; 2008. Available from: http://www.awmf.org/leitlinien/detail/ll/007-045.html Externer Link
9.
Al-Nawas B, Maeurer M. Severe versus local odontogenic bacterial infections: comparison of microbial isolates. Eur Surg Res. 2008;40(2):220-4. DOI: 10.1159/000110864 Externer Link
10.
Al-Nawas B, Piesold J, Otten JE. Leitlinien der Deutschen Gesellschaft für Mund-, Kiefer- und Gesichtschirurgie – Speicheldrüseninfektionen S2. AWMF; 2008. Available from: http://www.awmf.org/leitlinien/detail/ll/007-056.html Externer Link
11.
Al-Nawas B, Walter C, Morbach T, Seitner N, Siegel E, Maeurer M, Krummenauer F. Clinical and microbiological efficacy of moxifloxacin versus amoxicillin/clavulanic acid in severe odontogenic abscesses: a pilot study. Eur J Clin Microbiol Infect Dis. 2009 Jan;28(1):75-82. DOI: 10.1007/s10096-008-0587-2 Externer Link
12.
Alou L, Aguilar L, Sevillano D, Giménez MJ, Echeverría O, Gómez-Lus ML, Prieto J. Is there a pharmacodynamic need for the use of continuous versus intermittent infusion with ceftazidime against Pseudomonas aeruginosa? An in vitro pharmacodynamic model. J Antimicrob Chemother. 2005 Feb;55(2):209-13. DOI: 10.1093/jac/dkh536 Externer Link
13.
Ambrose PG, Quintiliani R, Nightingale CH, Nicolau DP. Continuous vs. intermittent infusion of cefuroxime for the therapy of community-acquired pneumonia. Infect Dis Clin Pract. 1998;7:463-70. DOI: 10.1097/00019048-199812000-00007 Externer Link
14.
American Thoracic Society. Guidelines for the management of adults with hospital-acquired, ventilator-associated, and healthcare-associated pneumonia. Am J Respir Crit Care Med. 2005 Feb;171(4):388-416. DOI: 10.1164/rccm.200405-644ST Externer Link
15.
Amodeo MR, Clulow T, Lainchbury J, Murdoch DR, Gallagher K, Dyer A, Metcalf SL, Pithie AD, Chambers ST. Outpatient intravenous treatment for infective endocarditis: safety, effectiveness and one-year outcomes. J Infect. 2009 Dec;59(6):387-93. DOI: 10.1016/j.jinf.2009.09.009 Externer Link
16.
Anderhuber W, Walch C, Köle W. Der seltene Fall einer Otitis externa maligna beim Nichtdiabetiker [A rare case of malignant otitis external in a non-diabetic patient]. Laryngorhinootologie. 1995 Jul;74(7):456-9. DOI: 10.1055/s-2007-997779 Externer Link
17.
Andes D, Craig WA. Pharmacokinetics and pharmacodynamics of outpatient intravenous antimicrobial therapy. Infect Dis Clin North Am. 1998 Dec;12(4):849-60, vi. DOI: 10.1016/S0891-5520(05)70024-6 Externer Link
18.
Andes DR, Craig WA. Cephalosporins. In: Mandell GL, Bennett JE, Dolin R, editors. Mandell, Douglas, and Bennett’s Principles and Practice of Infectious Diseases. 7th ed. Churchill Livingstone: Elsevier; 2010. p. 323-39.
19.
Ang JY, Asmar BI. Multidrug-resistant viridans streptococcus (MDRVS) osteomyelitis of the mandible successfully treated with moxifloxacin. South Med J. 2008 May;101(5):539-40. DOI: 10.1097/SMJ.0b013e3181684ee3 Externer Link
20.
Anthony KB, Fishman NO, Linkin DR, Gasink LB, Edelstein PH, Lautenbach E. Clinical and microbiological outcomes of serious infections with multidrug-resistant gram-negative organisms treated with tigecycline. Clin Infect Dis. 2008 Feb;46(4):567-70. DOI: 10.1086/526775 Externer Link
21.
Appelbaum PC. Reduced glycopeptide susceptibility in methicillin-resistant Staphylococcus aureus (MRSA). Int J Antimicrob Agents. 2007 Nov;30(5):398-408. DOI: 10.1016/j.ijantimicag.2007.07.011 Externer Link
22.
Arbeit RD, Maki D, Tally FP, Campanaro E, Eisenstein BI; Daptomycin 98-01 and 99-01 Investigators. The safety and efficacy of daptomycin for the treatment of complicated skin and skin-structure infections. Clin Infect Dis. 2004 Jun;38(12):1673-81. DOI: 10.1086/420818 Externer Link
23.
ARDS-Network. Ventilation with lower tidal volumes as compared with traditional tidal volumes for acute lung injury and the acute respiratory distress syndrome. The Acute Respiratory Distress Syndrome Network. N Engl J Med. 2000 May;342(18):1301-8. DOI: 10.1056/NEJM200005043421801 Externer Link
24.
Aronin SI, Peduzzi P, Quagliarello VJ. Community-acquired bacterial meningitis: risk stratification for adverse clinical outcome and effect of antibiotic timing. Ann Intern Med. 1998 Dec;129(11):862-9. DOI: 10.7326/0003-4819-129-11_Part_1-199812010-00004 Externer Link
25.
Aronoff G, Brater DC, Schrier R, Bennett WM. Use of drugs in patients with renal insufficiency. Workshop report. Blood Purif. 1994;12(1):14-9. DOI: 10.1159/000170140 Externer Link
26.
Arslan H, Azap OK, Ergönül O, Timurkaynak F; Urinary Tract Infection Study Group. Risk factors for ciprofloxacin resistance among Escherichia coli strains isolated from community-acquired urinary tract infections in Turkey. J Antimicrob Chemother. 2005 Nov;56(5):914-8. DOI: 10.1093/jac/dki344 Externer Link
27.
Auburtin M, Wolff M, Charpentier J, Varon E, Le Tulzo Y, Girault C, Mohammedi I, Renard B, Mourvillier B, Bruneel F, Ricard JD, Timsit JF. Detrimental role of delayed antibiotic administration and penicillin-nonsusceptible strains in adult intensive care unit patients with pneumococcal meningitis: the PNEUMOREA prospective multicenter study. Crit Care Med. 2006 Nov;34(11):2758-65. DOI: 10.1097/01.CCM.0000239434.26669.65 Externer Link
28.
Augsten R, Königsdörffer E, Strobel J. Endogene Endophthalmitis bei schweren Allgemeinerkrankungen [Endogenous endophthalmitis in severe generalized diseases]. Ophthalmologe. 1997 Jun;94(6):397-400. DOI: 10.1007/s003470050132 Externer Link
29.
Babinchak T, Ellis-Grosse E, Dartois N, Rose GM, Loh E; Tigecycline 301; 306 Study Groups. The efficacy and safety of tigecycline for the treatment of complicated intra-abdominal infections: analysis of pooled clinical trial data. Clin Infect Dis. 2005 Sep;41 Suppl 5:S354-67. DOI: 10.1086/431676 Externer Link
30.
Bader FG, Schröder M, Kujath P, Muhl E, Bruch HP, Eckmann C. Diffuse postoperative peritonitis - value of diagnostic parameters and impact of early indication for relaparotomy. Eur J Med Res. 2009 Nov;14(11):491-6. DOI: 10.1186/2047-783X-14-11-491 Externer Link
31.
Bae IG, Federspiel JJ, Miró JM, Woods CW, Park L, Rybak MJ, Rude TH, Bradley S, Bukovski S, de la Maria CG, Kanj SS, Korman TM, Marco F, Murdoch DR, Plesiat P, Rodriguez-Creixems M, Reinbott P, Steed L, Tattevin P, Tripodi MF, Newton KL, Corey GR, Fowler VG Jr; International Collaboration on Endocarditis–Microbiology Investigator. Heterogeneous vancomycin-intermediate susceptibility phenotype in bloodstream methicillin-resistant Staphylococcus aureus isolates from an international cohort of patients with infective endocarditis: prevalence, genotype, and clinical significance. J Infect Dis. 2009 Nov;200(9):1355-66. DOI: 10.1086/606027 Externer Link
32.
Bae WK, Lee KS, Park JW, Bae EH, Ma SK, Kim NH, Choi KC, Shin JH, Cho HH, Cho YB, Kim SW. A case of malignant otitis externa caused by Candida glabrata in a patient receiving haemodialysis. Scand J Infect Dis. 2007;39(4):370-2. DOI: 10.1080/00365540600978971 Externer Link
33.
Bailey RR, Begg EJ, Smith AH, Robson RA, Lynn KL, Chambers ST, Barclay ML, Hornibrook J. Prospective, randomized, controlled study comparing two dosing regimens of gentamicin/oral ciprofloxacin switch therapy for acute pyelonephritis. Clin Nephrol. 1996 Sep;46(3):183-6.
34.
Baltensperger MM, Eyrich GK. Osteomyelitis of the jaws: Definition and classification. In: Baltensperger MM, Eyrich GK, editors. Osteomyelitis of the jaws. Berlin, Heidelberg: Springer; 2009. DOI: 10.1007/978-3-540-28766-7 Externer Link
35.
Banks PA, Gerzof SG, Langevin RE, Silverman SG, Sica GT, Hughes MD.CT-guided aspiration of suspected pancreatic infection: bacteriology and clinical outcome. Int J Pancreatol. 1995 Dec;18(3):265-70. DOI: 10.1007/BF02784951 Externer Link
36.
Barbour A, Schmidt S, Rand KH, Derendorf H. Ceftobiprole: a novel cephalosporin with activity against Gram-positive and Gram-negative pathogens, including methicillin-resistant Staphylococcus aureus (MRSA). Int J Antimicrob Agents. 2009 Jul;34(1):1-7. DOI: 10.1016/j.ijantimicag.2008.12.012 Externer Link
37.
Barie PS. The cost of failure. Surg Infect (Larchmt). 2008 Jun;9(3):313-5. DOI: 10.1089/sur.2007.9964 Externer Link
38.
Barie PS, Eachempati SR. Surgical site infections. Surg Clin North Am. 2005 Dec;85(6):1115-35, viii-ix. DOI: 10.1016/j.suc.2005.09.006 Externer Link
39.
Barry P, Behrens-Baumann W, Pleyer U, Seal D. ESCRS Guidelines on prevention, investigation and management of post-operative endophthalmitis, Version 2. J Cataract Refract Surg. 2007;33:1-36.
40.
Barza M, Brusch J, Bergeron MG, Weinstein L. Penetration of antibiotics into fibrin loci in vivo. 3. Intermittent vs. continuous infusion and the effect of probenecid. J Infect Dis. 1974 Jan;129(1):73-8. DOI: 10.1093/infdis/129.1.73 Externer Link
41.
Becker A, Rosenthal EJK; Studiengruppe. Antibiotika-Empfindlichkeit von Sepsis-Erregern 2006–2007. Chemother J. 2010;19(2):28-39.
42.
Beekmann SE, Gilbert DN, Polgreen PM; IDSA Emerging Infections Network. Toxicity of extended courses of linezolid: results of an Infectious Diseases Society of America Emerging Infections Network survey. Diagn Microbiol Infect Dis. 2008 Dec;62(4):407-10. DOI: 10.1016/j.diagmicrobio.2008.08.009 Externer Link
43.
Beers MH. Explicit criteria for determining potentially inappropriate medication use by the elderly. An update. Arch Intern Med. 1997 Jul;157(14):1531-6. DOI: 10.1001/archinte.1997.00440350031003 Externer Link
44.
Beers MH, Ouslander JG, Rollingher I, Reuben DB, Brooks J, Beck JC. Explicit criteria for determining inappropriate medication use in nursing home residents. UCLA Division of Geriatric Medicine. Arch Intern Med. 1991 Sep;151(9):1825-32. DOI: 10.1001/archinte.1991.00400090107019 Externer Link
45.
Begg EJ, Barclay ML, Kirkpatrick CM. The therapeutic monitoring of antimicrobial agents. Br J Clin Pharmacol. 2001;52 Suppl 1:35S-43S.
46.
Behrens-Baumann W. Antiinfektiva bei Augenverletzungen [Anti-infectives in eye injuries]. Klin Monbl Augenheilkd. 2004 Aug;221(8):674-6. DOI: 10.1055/s-2004-813188 Externer Link
47.
Behrens-Baumann W. Antiinfektive medikamentöse Therapie in der Augenheilkunde - Teil 1: Bakterielle Infektionen [Anti-infective drug therapy in ophthalmology -- Part 1: bacterial infections]. Klin Monbl Augenheilkd. 2004 Jul;221(7):539-45. DOI: 10.1055/s-2004-813317 Externer Link
48.
Behrens-Baumann W. Chlamydienerkrankungen des Auges. Eine kurze Ubersicht [Chlamydial diseases of the eye. A short overview]. Ophthalmologe. 2007 Jan;104(1):28-34. DOI: 10.1007/s00347-006-1463-z Externer Link
49.
Behrens-Baumann W. Magdeburger Dreistufenplan für systemische Antibiotikatherapie. 1991–2009. Available from: http://www.med.uni-magdeburg.de/augenklinik Externer Link
50.
Behrens-Baumann W. Zur aktuellen Therapie der postoperativen Endophthalmitis [Current therapy for postoperative endophthalmitis]. Klin Monbl Augenheilkd. 2008 Nov;225(11):919-23. DOI: 10.1055/s-2008-1027512 Externer Link
51.
Behrens-Baumann W, Augustin AJ, Dick B, Fabian E, Huber-Spitzy V, Klauß V, Kramer A, Pitten FA, Pleyer U, Zeitz J. Leitlinie zur Prophylaxe und Therapie von Endophthalmitiden (Stand Juli 2005). Hyg Med. 2005;30:342-62.
52.
Behrens-Baumann W, Pleyer U. Therapie und Prognose der bakteriellen Keratitis [Therapy and prognosis of bacterial keratitis]. Ophthalmologe. 2007 Jan;104(1):15-20. DOI: 10.1007/s00347-006-1464-y Externer Link
53.
Beijer HJ, de Blaey CJ. Hospitalisations caused by adverse drug reactions (ADR): a meta-analysis of observational studies. Pharm World Sci. 2002 Apr;24(2):46-54. DOI: 10.1023/A:1015570104121 Externer Link
54.
Benetka O, Block M, Sangha O, Praetorius F, Gottwik M, Uebis R, Neuhaus KL, von Essen R; Arbeitsgemeinschaft Leitender Kardiologischer Krankenhausärzte. Clinical course of infective endocarditis in the late nineties: preliminary results of the ALKK endocarditis registry. Eur Heart J. 1999;20:362.
55.
Berendt AR, Peters EJ, Bakker K, Embil JM, Eneroth M, Hinchliffe RJ, Jeffcoate WJ, Lipsky BA, Senneville E, Teh J, Valk GD. Specific guidelines for treatment of diabetic foot osteomyelitis. Diabetes Metab Res Rev. 2008 May-Jun;24 Suppl 1:S190-1. DOI: 10.1002/dmrr.853 Externer Link
56.
Berenholz L, Katzenell U, Harell M. Evolving resistant pseudomonas to ciprofloxacin in malignant otitis externa. Laryngoscope. 2002 Sep;112(9):1619-22. DOI: 10.1097/00005537-200209000-00017 Externer Link
57.
Bergeron MG, Nguyen M, Gauvreau L. Influence of constant infusion versus bolus injections of antibiotics on in vivo synergy. Infection. 1978;6:38-46. DOI: 10.1007/BF01646064 Externer Link
58.
Bergeron MG, Simard P. Influence of three modes of administration on the penetration of latamoxef into interstitial fluid and fibrin clots and its in-vivo activity against Haemophilus influenzae. J Antimicrob Chemother. 1986 Jun;17(6):775-84. DOI: 10.1093/jac/17.6.775 Externer Link
59.
Bernard GR, Vincent JL, Laterre PF, LaRosa SP, Dhainaut JF, Lopez-Rodriguez A, Steingrub JS, Garber GE, Helterbrand JD, Ely EW, Fisher CJ Jr; Recombinant human protein C Worldwide Evaluation in Severe Sepsis (PROWESS) study group. Efficacy and safety of recombinant human activated protein C for severe sepsis. N Engl J Med. 2001 Mar;344(10):699-709. DOI: 10.1056/NEJM200103083441001 Externer Link
60.
Bernstein JM, Holland NJ, Porter GC, Maw AR. Resistance of Pseudomonas to ciprofloxacin: implications for the treatment of malignant otitis externa. J Laryngol Otol. 2007 Feb;121(2):118-23. DOI: 10.1017/S0022215106002775 Externer Link
61.
Besselink MG, Verwer TJ, Schoenmaeckers EJ, Buskens E, Ridwan BU, Visser MR, Nieuwenhuijs VB, Gooszen HG. Timing of surgical intervention in necrotizing pancreatitis. Arch Surg. 2007 Dec;142(12):1194-201. DOI: 10.1001/archsurg.142.12.1194 Externer Link
62.
Bichler KH, Eipper E, Naber K, Braun V, Zimmermann R, Lahme S. Urinary infection stones. Int J Antimicrob Agents. 2002 Jun;19(6):488-98. DOI: 10.1016/S0924-8579(02)00088-2 Externer Link
63.
Birmingham MC, Rayner CR, Meagher AK, Flavin SM, Batts DH, Schentag JJ. Linezolid for the treatment of multidrug-resistant, gram-positive infections: experience from a compassionate-use program. Clin Infect Dis. 2003 Jan;36(2):159-68. DOI: 10.1086/345744 Externer Link
64.
Bisno AL, Stevens DL. Streptococcal infections of skin and soft tissues. N Engl J Med. 1996 Jan;334(4):240-5. DOI: 10.1056/NEJM199601253340407 Externer Link
65.
Bjerklund Johansen TE, Cek M, Naber K, Stratchounski L, Svendsen MV, Tenke P; PEP and PEAP study investigators; European Society of Infections in Urology. Prevalence of hospital-acquired urinary tract infections in urology departments. Eur Urol. 2007 Apr;51(4):1100-11; discussion 1112. DOI: 10.1016/j.eururo.2006.08.012 Externer Link
66.
Blot SI, Vandewoude KH, De Waele JJ. Candida peritonitis. Curr Opin Crit Care. 2007 Apr;13(2):195-9. DOI: 10.1097/MCC.0b013e328028fd92 Externer Link
67.
Bogner J. 25 Jahre Fortschritt in der Infektiologie: neue Ansätze im Kampf gegen resistente Erreger [25 years of progress in infectiology: new approaches in the fight against resistant pathogens]. MMW Fortschr Med. 2008 Nov;150(48):123-4.
68.
Böhm K, Tesch-Römer C, Ziese T, editors. Beiträge zur Gesundheitsberichterstattung des Bundes: Gesundheit und Krankheit im Alter. Berlin: Robert-Koch Institut; 2009.
69.
Bohr V, Hansen B, Jessen O, Johnsen N, Kjersem H, Kristensen HS, Nyboe J, Rasmussen N. Eight hundred and seventy-five cases of bacterial meningitis. Part I of a three-part series: clinical data, prognosis, and the role of specialised hospital departments. J Infect. 1983 Jul;7(1):21-30. DOI: 10.1016/S0163-4453(83)90894-0 Externer Link
70.
Bolon M, Weber SG. General Principles of antimicrobial selection. In: Halter JB, Ouslander JG, Tinetti ME, Studenski S, High KP, Asthana S, editors. Hazzard’s geriatric medicine and gerontology. 6th ed. New York, Chicago: Mc Craw Hill Medical; 2009. p. 1517-30.
71.
Bone RC, Balk RA, Cerra FB, Dellinger RP, Fein AM, Knaus WA, Schein RM, Sibbald WJ. Definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. The ACCP/SCCM Consensus Conference Committee. American College of Chest Physicians/Society of Critical Care Medicine. Chest. 1992 Jun;101(6):1644-55. DOI: 10.1378/chest.101.6.1644 Externer Link
72.
Botto H, Naber KG, Bishop MC, Jarlier V. Antibiotic policy in prophylaxis and treatment of nosocomial urinary tract infection. In: Naber KG, Pechere JC, Kumazawa J, Khoury S, editors. Nosocomial and health care associated infections in urology. Plymouth, UK: Plymbridge Distributors; 2001. p. 179-91.
73.
Bouvier d’Yvoir MJ, Maire PH. Dosage regimens of antibacterials. Clin Drug Invest. 1996;11(4):229-39. DOI: 10.2165/00044011-199611040-00006 Externer Link
74.
Bouza E, Burillo A. Advances in the prevention and management of ventilator-associated pneumonia. Curr Opin Infect Dis. 2009 Aug;22(4):345-51. DOI: 10.1097/QCO.0b013e32832d8910 Externer Link
75.
Bouza E, Sousa D, Muñoz P, Rodríguez-Créixems 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 Oct;39(8):1161-9. DOI: 10.1086/424520 Externer Link
76.
Boyd CM, Darer J, Boult C, Fried LP, Boult L, Wu AW. Clinical practice guidelines and quality of care for older patients with multiple comorbid diseases: implications for pay for performance. JAMA. 2005 Aug;294(6):716-24. DOI: 10.1001/jama.294.6.716 Externer Link
77.
Bradley EL 3rd. A clinically based classification system for acute pancreatitis. Summary of the International Symposium on Acute Pancreatitis, Atlanta, Ga, September 11 through 13, 1992. Arch Surg. 1993 May;128(5):586-90. DOI: 10.1001/archsurg.1993.01420170122019 Externer Link
78.
Bradley SJ, Wilson AL, Allen MC, Sher HA, Goldstone AH, Scott GM. The control of hyperendemic glycopeptide-resistant Enterococcus spp. on a haematology unit by changing antibiotic usage. J Antimicrob Chemother. 1999 Feb;43(2):261-6. DOI: 10.1093/jac/43.2.261 Externer Link
79.
Briegel J. Update der Surviving Sepsis Campaign Guidelines 2008 [Update of the Surviving Sepsis Campaign guidelines 2008]. Anaesthesist. 2008 Mar;57(3):284-6. DOI: 10.1007/s00101-008-1335-9 Externer Link
80.
Brinton GS, Topping TM, Hyndiuk RA, Aaberg TM, Reeser FH, Abrams GW. Posttraumatic endophthalmitis. Arch Ophthalmol. 1984 Apr;102(4):547-50. DOI: 10.1001/archopht.1984.01040030425016 Externer Link
81.
Brito V, Niederman MS. Healthcare-associated pneumonia is a heterogeneous disease, and all patients do not need the same broad-spectrum antibiotic therapy as complex nosocomial pneumonia. Curr Opin Infect Dis. 2009 Jun;22(3):316-25. DOI: 10.1097/QCO.0b013e328329fa4e Externer Link
82.
Broll R, Eckmann C, Kujath P, Bruch HP. “Streptococcal toxic shock-like syndrome”. Chirurg. 1998 Aug;69(8):806-12. DOI: 10.1007/s001040050494 Externer Link
83.
Brouwers EE, Söhne M, Kuipers S, van Gorp EC, Schellens JH, Koks CH, Beijnen JH, Huitema AD. Ciprofloxacin strongly inhibits clozapine metabolism: two case reports. Clin Drug Investig. 2009;29(1):59-63. DOI: 10.2165/0044011-200929010-00006 Externer Link
84.
Brunkhorst FM, Engel C, Bloos F, Meier-Hellmann A, Ragaller M, Weiler N, Moerer O, Gruendling M, Oppert M, Grond S, Olthoff D, Jaschinski U, John S, Rossaint R, Welte T, Schaefer M, Kern P, Kuhnt E, Kiehntopf M, Hartog C, Natanson C, Loeffler M, Reinhart K; German Competence Network Sepsis (SepNet). Intensive insulin therapy and pentastarch resuscitation in severe sepsis. N Engl J Med. 2008 Jan;358(2):125-39. DOI: 10.1056/NEJMoa070716 Externer Link
85.
Bryant AE. Biology and pathogenesis of thrombosis and procoagulant activity in invasive infections caused by group A streptococci and Clostridium perfringens. Clin Microbiol Rev. 2003 Jul;16(3):451-62. DOI: 10.1128/CMR.16.3.451-462.2003 Externer Link
86.
Buck C, Bertram N, Ackermann T, Sauerbruch T, Derendorf H, Paar WD. Pharmacokinetics of piperacillin-tazobactam: intermittent dosing versus continuous infusion. Int J Antimicrob Agents. 2005 Jan;25(1):62-7. DOI: 10.1016/j.ijantimicag.2004.08.012 Externer Link
87.
Buijk SE, Mouton JW, Gyssens IC, Verbrugh HA, Bruining HA. Experience with a once-daily dosing program of aminoglycosides in critically ill patients. Intensive Care Med. 2002 Jul;28(7):936-42. DOI: 10.1007/s00134-002-1313-7 Externer Link
88.
Burgess DS, Hastings RW, Hardin TC. Pharmacokinetics and pharmacodynamics of cefepime administered by intermittent and continuous infusion. Clin Ther. 2000 Jan;22(1):66-75. DOI: 10.1016/S0149-2918(00)87978-3 Externer Link
89.
Burgess DS, Summers KK, Hardin TC. Pharmacokinetics and pharmacodynamics of aztreonam administered by continuous intravenous infusion. Clin Ther. 1999 Nov;21(11):1882-9. DOI: 10.1016/S0149-2918(00)86736-3 Externer Link
90.
Burgess DS, Waldrep T. Pharmacokinetics and pharmacodynamics of piperacillin/tazobactam when administered by continuous infusion and intermittent dosing. Clin Ther. 2002 Jul;24(7):1090-104. DOI: 10.1016/S0149-2918(02)80021-2 Externer Link
91.
Burkhardt O, Derendorf H, Welte T. Ertapenem: the new carbapenem 5 years after first FDA licensing for clinical practice. Expert Opin Pharmacother. 2007 Feb;8(2):237-56. DOI: 10.1517/14656566.8.2.237 Externer Link
92.
Burkhardt O, Lehmann C, Madabushi R, Kumar V, Derendorf H, Welte T. Once-daily tobramycin in cystic fibrosis: better for clinical outcome than thrice-daily tobramycin but more resistance development? J Antimicrob Chemother. 2006 Oct;58(4):822-9. DOI: 10.1093/jac/dkl328 Externer Link
93.
Burton ME, Shaw LM, Schentag JJ, Evans, WE. Applied Pharmacokinetics and Pharmacodynamics, Principles of Therapeutic Drug Monitoring. 4th ed. Baltimore: Lippincott Williams & Wilkins; 2006.
94.
Byl B, Clevenbergh P, Jacobs F, Struelens MJ, Zech F, Kentos A, Thys JP. Impact of infectious diseases specialists and microbiological data on the appropriateness of antimicrobial therapy for bacteremia. Clin Infect Dis. 1999 Jul;29(1):60-6. DOI: 10.1086/520182 Externer Link
95.
Calfee DP. Rifamycins. In: Mandell GL, Bennett JE, Dolin R, editors. Mandell, Douglas, and Bennett’s Principles and Practice of Infectious Diseases. 7th ed. Churchill Livingstone: Elsevier; 2010. p. 403-17.
96.
Campbell RM. The surgical management of pressure sores. Surg Clin North Am. 1959 Apr;39(2):509-30.
97.
Campochiaro PA, Lim JI. Aminoglycoside toxicity in the treatment of endophthalmitis. The Aminoglycoside Toxicity Study Group. Arch Ophthalmol. 1994 Jan;112(1):48-53. DOI: 10.1001/archopht.1994.01090130058017 Externer Link
98.
Cappelletty DM, Kang SL, Palmer SM, Rybak MJ. Pharmacodynamics of ceftazidime administered as continuous infusion or intermittent bolus alone and in combination with single daily-dose amikacin against Pseudomonas aeruginosa in an in vitro infection model. Antimicrob Agents Chemother. 1995 Aug;39(8):1797-801. DOI: 10.1128/AAC.39.8.1797 Externer Link
99.
Centers for Disease Control and Prevention. Sexually transmitted diseases treatment guidelines, 2006. MMWR Recomm Rep. 2006 Aug 4;55(RR-11):1-94.
100.
Chai FC, Auret K, Christiansen K, Yuen PW, Gardam D. Malignant otitis externa caused by Malassezia sympodialis. Head Neck. 2000 Jan;22(1):87-9.
101.
Chastre J, Wolff M, Fagon JY, Chevret S, Thomas F, Wermert D, Clementi E, Gonzalez J, Jusserand D, Asfar P, Perrin D, Fieux F, Aubas S; PneumA Trial Group. Comparison of 8 vs 15 days of antibiotic therapy for ventilator-associated pneumonia in adults: a randomized trial. JAMA. 2003 Nov;290(19):2588-98. DOI: 10.1001/jama.290.19.2588 Externer Link
102.
Chavez-Tapia NC, Soares-Weiser K, Brezis M, Leibovici L. Antibiotics for spontaneous bacterial peritonitis in cirrhotic patients. Cochrane Database Syst Rev. 2009;(1):CD002232. DOI: 10.1002/14651858.CD002232.pub2 Externer Link
103.
Chromik AM, Meiser A, Hölling J, Sülberg D, Daigeler A, Meurer K, Vogelsang H, Seelig MH, Uhl W. Identification of patients at risk for development of tertiary peritonitis on a surgical intensive care unit. J Gastrointest Surg. 2009 Jul;13(7):1358-67. DOI: 10.1007/s11605-009-0882-y Externer Link
104.
Ciulla TA. Update on acute and chronic endophthalmitis. Ophthalmology. 1999 Dec;106(12):2237-8. DOI: 10.1016/S0161-6420(99)90521-3 Externer Link
105.
Clark WL, Kaiser PK, Flynn HW Jr, Belfort A, Miller D, Meisler DM. Treatment strategies and visual acuity outcomes in chronic postoperative Propionibacterium acnes endophthalmitis. Ophthalmology. 1999 Sep;106(9):1665-70. DOI: 10.1016/S0161-6420(99)90348-2 Externer Link
106.
Clec’h C, Timsit JF, De Lassence A, Azoulay E, Alberti C, Garrouste-Orgeas M, Mourvilier B, Troche G, Tafflet M, Tuil O, Cohen Y. Efficacy of adequate early antibiotic therapy in ventilator-associated pneumonia: influence of disease severity. Intensive Care Med. 2004 Jul;30(7):1327-33. DOI: 10.1007/s00134-004-2292-7 Externer Link
107.
Cobo J, Oliva J, Sanz J, Aguado JM, Del Pozo JL, del Pozo J, Moreno S. Influence of microbiological reports on physician's choice of antimicrobial treatment for susceptible pathogens. Eur J Clin Microbiol Infect Dis. 2003 Sep;22(9):569-72. DOI: 10.1007/s10096-003-0978-3 Externer Link
108.
Colleaux KM, Hamilton WK. Effect of prophylactic antibiotics and incision type on the incidence of endophthalmitis after cataract surgery. Can J Ophthalmol. 2000 Dec;35(7):373-8.
109.
Coly-Mycin M. Parenteral, Full Prescribing Information. Monarch Pharmaceuticals; 2006 [cited 2009 Sept 27]. Available from: http://www.jhppharma.com/products/colymfp.pdf
110.
Conil JM, Georges B, Breden A, Segonds C, Lavit M, Seguin T, Coley N, Samii K, Chabanon G, Houin G, Saivin S. Increased amikacin dosage requirements in burn patients receiving a once-daily regimen. Int J Antimicrob Agents. 2006 Sep;28(3):226-30. DOI: 10.1016/j.ijantimicag.2006.04.015 Externer Link
111.
Conly JM, Johnston BL. VISA, hetero-VISA and VRSA: the end of the vancomycin era? Can J Infect Dis. 2002 Sep;13(5):282-4.
112.
Cosgrove SE, Fowler VG Jr. Management of Methicillin-Resistant Staphylococcus aureus Bacteremia. Clin Infect Dis. 2008;46 Suppl 5:S386-93. DOI: 10.1086/533595 Externer Link
113.
Cosgrove SE, Vigliani GA, Fowler VG Jr, Abrutyn E, Corey GR, Levine DP, Rupp ME, Chambers HF, Karchmer AW, Boucher HW. Initial low-dose gentamicin for Staphylococcus aureus bacteremia and endocarditis is nephrotoxic. Clin Infect Dis. 2009 Mar;48(6):713-21. DOI: 10.1086/597031 Externer Link
114.
Cousins D. Safe medication initiatives – sustaining good practice. Hospital Pharmacist. 2006;13:215-7.
115.
Cruse PJ, Foord R. The epidemiology of wound infection. A 10-year prospective study of 62,939 wounds. Surg Clin North Am. 1980 Feb;60(1):27-40.
116.
Cunha BA. Sepsis and septic shock: selection of empiric antimicrobial therapy. Crit Care Clin. 2008 Apr;24(2):313-34, ix. DOI: 10.1016/j.ccc.2007.12.015 Externer Link
117.
Darko W, Medicis JJ, Smith A, Guharoy R, Lehmann DE. Mississippi mud no more: cost-effectiveness of pharmacokinetic dosage adjustment of vancomycin to prevent nephrotoxicity. Pharmacotherapy. 2003 May;23(5):643-50. DOI: 10.1592/phco.23.5.643.32199 Externer Link
118.
Davey PG, Marwick C. Appropriate vs. inappropriate antimicrobial therapy. Clin Microbiol Infect. 2008 Apr;14 Suppl 3:15-21. DOI: 10.1111/j.1469-0691.2008.01959.x Externer Link
119.
De Gans J, van de Beek D; European Dexamethasone in Adulthood Bacterial Meningitis Study Investigators. Dexamethasone in adults with bacterial meningitis. N Engl J Med. 2002 Nov;347(20):1549-56. DOI: 10.1056/NEJMoa021334 Externer Link
120.
De Jongh R, Hens R, Basma V, Mouton JW, Tulkens PM, Carryn S. Continuous versus intermittent infusion of temocillin, a directed spectrum penicillin for intensive care patients with nosocomial pneumonia: stability, compatibility, population pharmacokinetic studies and breakpoint selection. J Antimicrob Chemother. 2008 Feb;61(2):382-8. DOI: 10.1093/jac/dkm467 Externer Link
121.
De Waele JJ. A role for prophylactic antibiotics in necrotizing pancreatitis? Why we may never know the answer... Crit Care. 2008;12(6):195. DOI: 10.1186/cc7122 Externer Link
122.
De Waele JJ, Hoste EA, Blot SI. Blood stream infections of abdominal origin in the intensive care unit: characteristics and determinants of death. Surg Infect (Larchmt). 2008 Apr;9(2):171-7. DOI: 10.1089/sur.2006.063 Externer Link
123.
Dellinger RP, Levy MM, Carlet JM, Bion J, Parker MM, Jaeschke R, Reinhart K, Angus DC, Brun-Buisson C, Beale R, Calandra T, Dhainaut JF, Gerlach H, Harvey M, Marini JJ, Marshall J, Ranieri M, Ramsay G, Sevransky J, Thompson BT, Townsend S, Vender JS, Zimmerman JL, Vincent JL. Surviving Sepsis Campaign: international guidelines for management of severe sepsis and septic shock: 2008. Intensive Care Med. 2008 Jan;34(1):17-60. DOI: 10.1007/s00134-007-0934-2 Externer Link
124.
Dellinger RP, Levy MM, Carlet JM, Bion J, Parker MM, Jaeschke R, Reinhart K, Angus DC, Brun-Buisson C, Beale R, Calandra T, Dhainaut JF, Gerlach H, Harvey M, Marini JJ, Marshall J, Ranieri M, Ramsay G, Sevransky J, Thompson BT, Townsend S, Vender JS, Zimmerman JL, Vincent JL; International Surviving Sepsis Campaign Guidelines Committee; American Association of Critical-Care Nurses; American College of Chest Physicians; American College of Emergency Physicians; Canadian Critical Care Society; European Society of Clinical Microbiology and Infectious Diseases; European Society of Intensive Care Medicine; European Respiratory Society; International Sepsis Forum; Japanese Association for Acute Medicine; Japanese Society of Intensive Care Medicine; Society of Critical Care Medicine; Society of Hospital Medicine; Surgical Infection Society; World Federation of Societies of Intensive and Critical Care Medicine. Surviving Sepsis Campaign: international guidelines for management of severe sepsis and septic shock: 2008. Crit Care Med. 2008 Jan;36(1):296-327. DOI: 10.1097/01.CCM.0000298158.12101.41 Externer Link
125.
Dellit TH, Owens RC, McGowan JE Jr, Gerding DN, Weinstein RA, Burke JP, Huskins WC, Paterson DL, Fishman NO, Carpenter CF, Brennan PJ, Billeter M, Hooton TM; Infectious Diseases Society of America; Society for Healthcare Epidemiology of America. 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 Jan;44(2):159-77. DOI: 10.1086/510393 Externer Link
126.
Dembinski R, Rossaint R. Ventilatorassoziierte Pneumonie [Ventilator-associated pneumonia]. Anaesthesist. 2008 Aug;57(8):825-42. DOI: 10.1007/s00101-008-1415-x Externer Link
127.
Deramo VA, Ting TD. Treatment of Propionibacterium acnes endophthalmitis. Curr Opin Ophthalmol. 2001 Jun;12(3):225-9. DOI: 10.1097/00055735-200106000-00015 Externer Link
128.
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 Sep;12(5):419-26. DOI: 10.1097/00007691-199009000-00003 Externer Link
129.
Dettli L. The kidney in pre-clinical and clinical pharmacokinetics. Jpn J Clin Pharmacol Ther. 1984;15:241-54. DOI: 10.3999/jscpt.15.241 Externer Link
130.
Deutsches Institut für Normung (DIN). Labormedizinische Untersuchungen und In-vitro-Diagnostika-Systeme – Empfindlichkeitsprüfung von Infektionserregern und Evaluation von Geräten zur antimikrobiellen Empfindlichkeitsprüfung – Teil 1: Referenzmethode zur Testung der In-vitro-Aktivität von antimikrobiellen Substanzen gegen schnell wachsende aerobe Bakterien, die Infektionskrankheiten verursachen (ISO/FDIS 20776-1:2006). 2006.
131.
Dietrich ES, Jopp R, Schreier U, Gilge R, Bartmann P, Berthold H. Kosten einer leitliniengerechten Arzneimitteltherapie in Deutschland. Gesundh Ökon Qual Manag. 2005;10(1):35-43. DOI: 10.1055/s-2004-813943 Externer Link
132.
Dietrich ES, Mast O, Fischer H, Hoppe-Tichy T, Kullmann KH, Stodt HJ, Strehl E, Tassler H, Wittmann D. Vorschläge zur Planung, Durchführung und Publikation pharmakoökonomischer Evaluationen zu Antiinfektiva. Chemother J. 2001;10(3):110-5.
133.
Dietrich ES, Schmid M, Bucher A, Lemmen S, Frank U, Daschner FD. Kostenreduktion in der Antibiotika-Therapie. Krankenhauspharmazie. 1998;19(6):287-92.
134.
Dinubile MJ, Friedland I, Chan CY, Motyl MR, Giezek H, Shivaprakash M, Weinstein RA, Quinn JP. Bowel colonization with resistant gram-negative bacilli after antimicrobial therapy of intra-abdominal infections: observations from two randomized comparative clinical trials of ertapenem therapy. Eur J Clin Microbiol Infect Dis. 2005 Jul;24(7):443-9. DOI: 10.1007/s10096-005-1356-0 Externer Link
135.
Doern GV, Vautour R, Gaudet M, Levy B. Clinical impact of rapid in vitro susceptibility testing and bacterial identification. J Clin Microbiol. 1994 Jul;32(7):1757-62.
136.
Doft BH, Weiskopf J, Nilsson-Ehle I, Wingard LB Jr. Amphotericin clearance in vitrectomized versus nonvitrectomized eyes. Ophthalmology. 1985 Nov;92(11):1601-5. DOI: 10.1016/S0161-6420(85)33838-1 Externer Link
137.
Driebe WT Jr, Mandelbaum S, Forster RK, Schwartz LK, Culbertson WW. Pseudophakic endophthalmitis. Diagnosis and management. Ophthalmology. 1986 Apr;93(4):442-8. DOI: 10.1016/S0161-6420(86)33722-9 Externer Link
138.
Drinkovic D, Morris AJ, Pottumarthy S, MacCulloch D, West T. Bacteriological outcome of combination versus single-agent treatment for staphylococcal endocarditis. J Antimicrob Chemother. 2003 Nov;52(5):820-5. DOI: 10.1093/jac/dkg440 Externer Link
139.
Driscoll JA, Brody SL, Kollef MH. The epidemiology, pathogenesis and treatment of Pseudomonas aeruginosa infections. Drugs. 2007;67(3):351-68. DOI: 10.2165/00003495-200767030-00003 Externer Link
140.
Drusano GL, Ambrose PG, Bhavnani SM, Bertino JS, Nafziger AN, Louie A. Back to the future: using aminoglycosides again and how to dose them optimally. Clin Infect Dis. 2007 Sep;45(6):753-60. DOI: 10.1086/520991 Externer Link
141.
Dupont H. The empiric treatment of nosocomial intra-abdominal infections. Int J Infect Dis. 2007 May;11 Suppl 1:S1-6. DOI: 10.1016/S1201-9712(07)60001-0 Externer Link
142.
Durand ML. The post-endophthalmitis vitrectomy study era. Arch Ophthalmol. 2002 Feb;120(2):233-4.
143.
Durand ML, Calderwood SB, Weber DJ, Miller SI, Southwick FS, Caviness VS Jr, Swartz MN. Acute bacterial meningitis in adults. A review of 493 episodes. N Engl J Med. 1993 Jan;328(1):21-8. DOI: 10.1056/NEJM199301073280104 Externer Link
144.
Durupt S, Josserand RN, Sibille M, Durieu I. Acute, recurrent fosfomycin-induced liver toxicity in an adult patient with cystic fibrosis. Scand J Infect Dis. 2001;33(5):391-2. DOI: 10.1080/003655401750174200 Externer Link
145.
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 May;66(5):1343-8. DOI: 10.1097/TA.0b013e31819dca4e Externer Link
146.
Eckert AW, Höhne C, Schubert J. Erregerspektrum und Resistenzsituation bei rein anaeroben odontogenen Infektionen [Pathogen spectrum and resistance status of exclusively anaerobic odontogenic infections]. Mund Kiefer Gesichtschir. 2000 May;4(3):153-8. DOI: 10.1007/s100060050188 Externer Link
147.
Eckert AW, Maurer P, Wilhelms D, Schubert J. Keimspektren und Antibiotika bei odontogenen Infektionen. Renaissance der Penicilline? [Bacterial spectra and antibiotics in odontogenic infections. Renaissance of the penicillins?]. Mund Kiefer Gesichtschir. 2005 Nov;9(6):377-83. DOI: 10.1007/s10006-005-0646-0 Externer Link
148.
Eckert AW, Maurer P, Wilhelms D, Schubert J. Weichteilinfektionen in der Mund-, Kiefer- und Plastischen Gesichtschirurgie. Keimspektren und Antibiotika [Soft tissue infections in oral, maxillofacial, and plastic surgery. Bacterial spectra and antibiotics]. Mund Kiefer Gesichtschir. 2005 Nov;9(6):389-95. DOI: 10.1007/s10006-005-0645-1 Externer Link
149.
Eckmann C, Leitner E, Heizmann W, Bodmann KF. Wirksamkeit von Tigecyclin bei komplizierten Infektionen unter Routinebdingungen in Deutschland [Posterpräsentation]. In: 9. Kongress der Deutschen Interdisziplinären Vereinigung für Intensiv- und Notfallmedizin (DIVI); 03.–06.12.2008; Hamburg.
150.
Edelsberg J, Berger A, Schell S, Mallick R, Kuznik A, Oster G. Economic consequences of failure of initial antibiotic therapy in hospitalized adults with complicated intra-abdominal infections. Surg Infect (Larchmt). 2008 Jun;9(3):335-47. DOI: 10.1089/sur.2006.100 Externer Link
151.
Eggimann P, Francioli P, Bille J, Schneider R, Wu MM, Chapuis G, Chiolero R, Pannatier A, Schilling J, Geroulanos S, Glauser MP, Calandra T. Fluconazole prophylaxis prevents intra-abdominal candidiasis in high-risk surgical patients. Crit Care Med. 1999 Jun;27(6):1066-72. DOI: 10.1097/00003246-199906000-00019 Externer Link
152.
Eick S, Pfister W, Straube E. Antimicrobial susceptibility of anaerobic and capnophilic bacteria isolated from odontogenic abscesses and rapidly progressive periodontitis. Int J Antimicrob Agents. 1999 Jun;12(1):41-6. DOI: 10.1016/S0924-8579(99)00056-4 Externer Link
153.
El Desoky E, Klotz U. Value, limitations and clinical impact of therapeutic drug monitoring in adults. Drug Invest. 1993;6:127-36. DOI: 10.1007/BF03259732 Externer Link
154.
Elhanan G, Sarhat M, Raz R. Empiric antibiotic treatment and the misuse of culture results and antibiotic sensitivities in patients with community-acquired bacteraemia due to urinary tract infection. J Infect. 1997 Nov;35(3):283-8. DOI: 10.1016/S0163-4453(97)93194-7 Externer Link
155.
Elliott DJ, Zaoutis TE, Troxel AB, Loh A, Keren R. Empiric antimicrobial therapy for pediatric skin and soft-tissue infections in the era of methicillin-resistant Staphylococcus aureus. Pediatrics. 2009 Jun;123(6):e959-66. DOI: 10.1542/peds.2008-2428 Externer Link
156.
Ellis-Grosse EJ, Babinchak T, Dartois N, Rose G, Loh E; Tigecycline 300 cSSSI Study Group; Tigecycline 305 cSSSI Study Group. The efficacy and safety of tigecycline in the treatment of skin and skin-structure infections: results of 2 double-blind phase 3 comparison studies with vancomycin-aztreonam. Clin Infect Dis. 2005 Sep;41 Suppl 5:S341-53. DOI: 10.1086/431675 Externer Link
157.
El-Massry A, Meredith TA, Aguilar HE, Shaarawy A, Kincaid M, Dick J, Mahmoud MI. Minoglycoside levels in the rabbit vitreous cavity after intravenous administration. Am J Ophthalmol. 1996 Nov;122(5):684-9.
158.
Endophthalmitis Vitrectomy Study Group. Results of the Endophthalmitis Vitrectomy Study. A randomized trial of immediate vitrectomy and of intravenous antibiotics for the treatment of postoperative bacterial endophthalmitis. Endophthalmitis Vitrectomy Study Group. Arch Ophthalmol. 1995 Dec;113(12):1479-96. DOI: 10.1001/archopht.1995.01100120009001 Externer Link
159.
Engel C, Brunkhorst FM, Bone HG, Brunkhorst R, Gerlach H, Grond S, Gruendling M, Huhle G, Jaschinski U, John S, Mayer K, Oppert M, Olthoff D, Quintel M, Ragaller M, Rossaint R, Stuber F, Weiler N, Welte T, Bogatsch H, Hartog C, Loeffler M, Reinhart K. Epidemiology of sepsis in Germany: results from a national prospective multicenter study. Intensive Care Med. 2007 Apr;33(4):606-18. DOI: 10.1007/s00134-006-0517-7 Externer Link
160.
Erstad BL. Dosing of medications in morbidly obese patients in the intensive care unit setting. Intensive Care Med. 2004 Jan;30(1):18-32. DOI: 10.1007/s00134-003-2059-6 Externer Link
161.
Esposito S, Leone S, Petta E, Noviello S, Ianniello F. Treatment options for skin and soft tissue infections caused by meticillin-resistant Staphylococcus aureus:oralvs.parenteral; home vs. hospital. Int J Antimicrob Agents. 2009 Jul;34 Suppl 1:S30-5. DOI: 10.1016/S0924-8579(09)70547-3 Externer Link
162.
Fachinformation Doribax® 9/2009.
163.
Falagas ME, Giannopoulou KP, Kokolakis GN, Rafailidis PI. Fosfomycin: use beyond urinary tract and gastrointestinal infections. Clin Infect Dis. 2008 Apr;46(7):1069-77. DOI: 10.1086/527442 Externer Link
164.
Falagas ME, Matthaiou DK, Bliziotis IA. The role of aminoglycosides in combination with a beta-lactam for the treatment of bacterial endocarditis: a meta-analysis of comparative trials. J Antimicrob Chemother. 2006 Apr;57(4):639-47. DOI: 10.1093/jac/dkl044 Externer Link
165.
Falagas ME, Roussos N, Gkegkes ID, Rafailidis PI, Karageorgopoulos DE. Fosfomycin for the treatment of infections caused by Gram-positive cocci with advanced antimicrobial drug resistance: a review of microbiological, animal and clinical studies. Expert Opin Investig Drugs. 2009 Jul;18(7):921-44. DOI: 10.1517/13543780902967624 Externer Link
166.
Falkler WA Jr, Enwonwu CO, Idigbe EO. Microbiological understandings and mysteries of noma (cancrum oris). Oral Dis. 1999 Apr;5(2):150-5. DOI: 10.1111/j.1601-0825.1999.tb00081.x Externer Link
167.
Federspil PH. Antibiotika-Therapie der Infektionen an Kopf und Hals – Leitlinien, im Auftrag des Präsidiums der Deutschen Gesellschaft für Hals-Nasen-Ohren-Heilkunde, Kopf- und Hals-Chirurgie. Arzneimitteltherapie. 2000;18:350-61.
168.
Feifel H, Friebel S, Riediger D. Verlauf und Therapie der Osteomyelitis des Gesichtsschädels. Dtsch Zahnarztl Z. 1997:691-4.
169.
Fenton C, Keating GM, Curran MP. Daptomycin. Drugs. 2004;64(4):445-55. DOI: 10.2165/00003495-200464040-00009 Externer Link
170.
Fick DM, Cooper JW, Wade WE, Waller JL, Maclean JR, Beers MH. Updating the Beers criteria for potentially inappropriate medication use in older adults: results of a US consensus panel of experts. Arch Intern Med. 2003 Dec 8-22;163(22):2716-24. DOI: 10.1001/archinte.163.22.2716 Externer Link
171.
Fleig WE, Grothe W, Lotterer E, Behl S. Spontane bakterielle Peritonitis (SBP). Retrospektive und prospektive Daten einer multizentrischen Studie zu Prävalenz, Diagnostik und Therapie in Deutschland [Spontaneous bacterial peritonitis (SBP). Retrospective and prospective data from a multicenter study on prevalence, diagnosis and therapy in Germany]. Dtsch Med Wochenschr. 2004 Aug;129(34-35):1792-7. DOI: 10.1055/s-2004-829030 Externer Link
172.
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 Jul;19(7):493-500. DOI: 10.1007/s100960000306 Externer Link
173.
Forouzesh A, Moise PA, Sakoulas G. Vancomycin ototoxicity: a reevaluation in an era of increasing doses. Antimicrob Agents Chemother. 2009 Feb;53(2):483-6. DOI: 10.1128/AAC.01088-08 Externer Link
174.
Fowler VG Jr, Boucher HW, Corey GR, Abrutyn E, Karchmer AW, Rupp ME, Levine DP, Chambers HF, Tally FP, Vigliani GA, Cabell CH, Link AS, DeMeyer I, Filler SG, Zervos M, Cook P, Parsonnet J, Bernstein JM, Price CS, Forrest GN, Fätkenheuer G, Gareca M, Rehm SJ, Brodt HR, Tice A, Cosgrove SE; S. aureus Endocarditis and Bacteremia Study Group. Daptomycin versus standard therapy for bacteremia and endocarditis caused by Staphylococcus aureus. N Engl J Med. 2006 Aug;355(7):653-65. DOI: 10.1056/NEJMoa053783 Externer Link
175.
Frampton JE, Curran MP. Tigecycline. Drugs. 2005;65(18):2623-35. DOI: 10.2165/00003495-200565180-00008 Externer Link
176.
Franzese CB, Isaacson JE. Peritonsillar and parapharyngeal space abscess in the older adult. Am J Otolaryngol. 2003 May-Jun;24(3):169-73. DOI: 10.1016/S0196-0709(02)32428-1 Externer Link
177.
Fraser A, Gafter-Gvili A, Paul M, Leibovici L. Antibiotics for preventing meningococcal infections. Cochrane Database Syst Rev. 2006;(4):CD004785. DOI: 10.1002/14651858.CD004785.pub3 Externer Link
178.
Frei CR, Burgess DS. Continuous infusion beta-lactams for intensive care unit pulmonary infections. Clin Microbiol Infect. 2005 May;11(5):418-21. DOI: 10.1111/j.1469-0691.2005.01106.x Externer Link
179.
Fünfstück R, Stein G. Asymptomatische Bakteriurie. Nieren- und Hochdruckkrankheiten. 2007;26:269-78. DOI: 10.5414/NHP36269 Externer Link
180.
Galloway G, Ramsay A, Jordan K, Vivian A. Macular infarction after intravitreal amikacin: mounting evidence against amikacin. Br J Ophthalmol. 2002 Mar;86(3):359-60. DOI: 10.1136/bjo.86.3.359 Externer Link
181.
Gan IM, van Dissel JT, Beekhuis WH, Swart W, van Meurs JC. Intravitreal vancomycin and gentamicin concentrations in patients with postoperative endophthalmitis. Br J Ophthalmol. 2001 Nov;85(11):1289-93. DOI: 10.1136/bjo.85.11.1289 Externer Link
182.
García-Lechuz J, Bouza E. Treatment recommendations and strategies for the management of bone and joint infections. Expert Opin Pharmacother. 2009 Jan;10(1):35-55. DOI: 10.1517/14656560802611766 Externer Link
183.
Garey KW, Amsden GW. Intravenous azithromycin. Ann Pharmacother 1999;33(2):218-28. DOI: 10.1345/aph.18046 Externer Link
184.
Garnacho-Montero J, Ortiz-Leyba C, Herrera-Melero I, Aldabó-Pallás T, Cayuela-Dominguez A, Marquez-Vacaro JA, Carbajal-Guerrero J, Garcia-Garmendia JL. Mortality and morbidity attributable to inadequate empirical antimicrobial therapy in patients admitted to the ICU with sepsis: a matched cohort study. J Antimicrob Chemother. 2008 Feb;61(2):436-41. DOI: 10.1093/jac/dkm460 Externer Link
185.
Garnacho-Montero J, Sa-Borges M, Sole-Violan J, Barcenilla F, Escoresca-Ortega A, Ochoa M, Cayuela A, Rello J. Optimal management therapy for Pseudomonas aeruginosa ventilator-associated pneumonia: an observational, multicenter study comparing monotherapy with combination antibiotic therapy. Crit Care Med. 2007 Aug;35(8):1888-95. DOI: 10.1097/01.CCM.0000275389.31974.22 Externer Link
186.
Garraffo R. Pharmacodynamic bases for continuous infusion of beta-lactams: optimisation of antibacterial activities against gram-negative bacilli. Antibiotiques. 2002;4:22–8.
187.
Gastmeier P, Sohr D, Geffers C, Rüden H, Vonberg RP, Welte T. Early- and late-onset pneumonia: is this still a useful classification? Antimicrob Agents Chemother. 2009 Jul;53(7):2714-8. DOI: 10.1128/AAC.01070-08 Externer Link
188.
Gavriel H, Lazarovitch T, Pomortsev A, Eviatar E. Variations in the microbiology of peritonsillar abscess. Eur J Clin Microbiol Infect Dis. 2009 Jan;28(1):27-31. DOI: 10.1007/s10096-008-0583-6 Externer Link
189.
Gell PGH, Coombs RRA. Classification of allergic reactions responsible for clinical hypersensitivity and disease. In: Gell PGH, Coombs RRA, Hachmann PJ, editors. Clinical Aspects of Immunology. 3rd ed. Oxford: Blackwell Scientific Publications; 1975. p. 761-81.
190.
Georges B, Conil JM, Cougot P, Decun JF, Archambaud M, Seguin T, Chabanon G, Virenque C, Houin G, Saivin S. Cefepime in critically ill patients: continuous infusion vs. an intermittent dosing regimen. Int J Clin Pharmacol Ther. 2005 Aug;43(8):360-9. DOI: 10.5414/CPP43360 Externer Link
191.
Giamarellou H. Clinical experience with the fourth generation cephalosporins. J Chemother. 1996 Feb;8 Suppl 2:91-104.
192.
Gilbert DN, Leggett JE. Aminoglycosides. In: Mandell GL, Bennett JE, Dolin R, editors. Mandell, Douglas, and Bennett’s Principles and Practice of Infectious Diseases. 7th ed. Churchill Livingstone: Elsevier; 2010. p. 359-84.
193.
Giordano P, Song J, Pertel P, Herrington J, Kowalsky S. Sequential intravenous/oral moxifloxacin versus intravenous piperacillin-tazobactam followed by oral amoxicillin-clavulanate for the treatment of complicated skin and skin structure infection. Int J Antimicrob Agents. 2005 Nov;26(5):357-65. DOI: 10.1016/j.ijantimicag.2005.07.017 Externer Link
194.
Giske CG, Monnet DL, Cars O, Carmeli Y; ReAct-Action on Antibiotic Resistance. Clinical and economic impact of common multidrug-resistant gram-negative bacilli. Antimicrob Agents Chemother. 2008 Mar;52(3):813-21. DOI: 10.1128/AAC.01169-07 Externer Link
195.
Glaeske G, Janhsen K, editors. GEK Arzneimittel-Report 2005. Auswertungsergebnisse der GEK-Arzneimitteldaten aus den Jahren 2003–2004. St. Augustin: Asgard; 2005. (Schriftenreihe zur Gesundheitsanalyse; 36).
196.
Glaeske G, Trittin C, editors. Weichenstellung: Altern in unserer Gesellschaft. Neue Herausforderungen für das Gesundheitswesen. St. Augustin: Asgard; 2007. (Schriftenreihe zur Gesundheitsanalyse; 62).
197.
Gmür R, Wyss C, Xue Y, Thurnheer T, Guggenheim B. Gingival crevice microbiota from Chinese patients with gingivitis or necrotizing ulcerative gingivitis. Eur J Oral Sci. 2004 Feb;112(1):33-41. DOI: 10.1111/j.0909-8836.2004.00103.x Externer Link
198.
Gottesman BS, Carmeli Y, Shitrit P, Chowers M. Impact of quinolone restriction on resistance patterns of Escherichia coli isolated from urine by culture in a community setting. Clin Infect Dis. 2009 Sep;49(6):869-75. DOI: 10.1086/605530 Externer Link
199.
Grabe M, Bishop MC, Bjerklund-Johansen TE, Botto H. Guidelines on urological infections. In: European Association of Urology Guidelines. Arnhem, Netherlands: European Association of Urology; 2009. p. 1-110.
200.
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 Apr;22(4):471-83. DOI: 10.1592/phco.22.7.471.33665 Externer Link
201.
Greenfield DS, Suñer IJ, Miller MP, Kangas TA, Palmberg PF, Flynn HW Jr. Endophthalmitis after filtering surgery with mitomycin. Arch Ophthalmol. 1996 Aug;114(8):943-9. DOI: 10.1001/archopht.1996.01100140151007 Externer Link
202.
Grime PD, Bowerman JE, Weller PJ. Gentamicin impregnated polymethylmethacrylate (PMMA) beads in the treatment of primary chronic osteomyelitis of the mandible. Br J Oral Maxillofac Surg. 1990 Dec;28(6):367-74. DOI: 10.1016/0266-4356(90)90033-H Externer Link
203.
Grosset J, Leventis S. Adverse effects of rifampin. Rev Infect Dis. 1983 Jul-Aug;5 Suppl 3:S440-50. DOI: 10.1093/clinids/5.Supplement_3.S440 Externer Link
204.
Grossi P, Gasperina DD. Antimicrobial treatment of sepsis. Surg Infect (Larchmt). 2006;7 Suppl 2:S87-91. DOI: 10.1089/sur.2006.7.s2-87 Externer Link
205.
Guldfred LA, Lyhne D, Becker BC. Acute epiglottitis: epidemiology, clinical presentation, management and outcome. J Laryngol Otol. 2008 Aug;122(8):818-23. DOI: 10.1017/S0022215107000473 Externer Link
206.
Gupta A, Swaroop C, Agarwala S, Pandey RM, Bakhshi S. Randomized controlled trial comparing oral amoxicillin-clavulanate and ofloxacin with intravenous ceftriaxone and amikacin as outpatient therapy in pediatric low-risk febrile neutropenia. J Pediatr Hematol Oncol. 2009 Sep;31(9):635-41. DOI: 10.1097/MPH.0b013e3181acd8cd Externer Link
207.
Guthrie OW. Aminoglycoside induced ototoxicity. Toxicology. 2008 Jul 30;249(2-3):91-6. DOI: 10.1016/j.tox.2008.04.015 Externer Link
208.
Habib G, Hoen B, Tornos P, Thuny F, Prendergast B, Vilacosta I, Moreillon P, de Jesus Antunes M, Thilen U, Lekakis J, Lengyel M, Müller L, Naber CK, Nihoyannopoulos P, Moritz A, Zamorano JL; ESC Committee for Practice Guidelines. Guidelines on the prevention, diagnosis, and treatment of infective endocarditis (new version 2009): the Task Force on the Prevention, Diagnosis, and Treatment of Infective Endocarditis of the European Society of Cardiology (ESC). Endorsed by the European Society of Clinical Microbiology and Infectious Diseases (ESCMID) and the International Society of Chemotherapy (ISC) for Infection and Cancer. Eur Heart J. 2009 Oct;30(19):2369-413. DOI: 10.1093/eurheartj/ehp285 Externer Link
209.
Hagelskjaer Kristensen L, Prag J. Localised Fusobacterium necrophorum infections: a prospective laboratory-based Danish study. Eur J Clin Microbiol Infect Dis. 2008 Aug;27(8):733-9. DOI: 10.1007/s10096-008-0497-3 Externer Link
210.
Haider SA, Hassett P, Bron AJ. Intraocular vancomycin levels after intravitreal injection in post cataract extraction endophthalmitis. Retina (Philadelphia, Pa). 2001;21(3):210-3. DOI: 10.1097/00006982-200106000-00002 Externer Link
211.
Hall RG 2nd, Payne KD, Bain AM, Rahman AP, Nguyen ST, Eaton SA, Busti AJ, Vu SL, Bedimo R. Multicenter evaluation of vancomycin dosing: emphasis on obesity. Am J Med. 2008 Jun;121(6):515-8. DOI: 10.1016/j.amjmed.2008.01.046 Externer Link
212.
Halle E, Padberg J, Rosseau S, Klare I, Werner G, Witte W. Linezolid-resistant Enterococcus faecium and Enterococcus faecalis isolated from a septic patient: report of first isolates in Germany. Infection. 2004 Jun;32(3):182-3. DOI: 10.1007/s15010-004-3009-0 Externer Link
213.
Han DP, Wisniewski SR, Wilson LA, Barza M, Vine AK, Doft BH, Kelsey SF. Spectrum and susceptibilities of microbiologic isolates in the Endophthalmitis Vitrectomy Study. Am J Ophthalmol. 1996 Jul;122(1):1-17.
214.
Handzel O, Halperin D. Necrotizing (malignant) external otitis. Am Fam Physician. 2003 Jul;68(2):309-12.
215.
Hanke F, Szymanski J, Jaehde U, Thürmann PA. Drug-related problems in nursing homes – a prospective study. Int J Clin Pharmacol Ther. 2006;44:500.
216.
Hanna BC, McMullan R, Gallagher G, Hedderwick S. The epidemiology of peritonsillar abscess disease in Northern Ireland. J Infect. 2006 Apr;52(4):247-53. DOI: 10.1016/j.jinf.2005.07.002 Externer Link
217.
Harbarth S, Uckay I. Are there patients with peritonitis who require empiric therapy for enterococcus? Eur J Clin Microbiol Infect Dis. 2004 Feb;23(2):73-7. DOI: 10.1007/s10096-003-1078-0 Externer Link
218.
Hartzell JD, Neff R, Ake J, Howard R, Olson S, Paolino K, Vishnepolsky M, Weintrob A, Wortmann G. Nephrotoxicity associated with intravenous colistin (colistimethate sodium) treatment at a tertiary care medical center. Clin Infect Dis. 2009 Jun;48(12):1724-8. DOI: 10.1086/599225 Externer Link
219.
Hasbun R, Abrahams J, Jekel J, Quagliarello VJ. Computed tomography of the head before lumbar puncture in adults with suspected meningitis. N Engl J Med. 2001 Dec;345(24):1727-33. DOI: 10.1056/NEJMoa010399 Externer Link
220.
Hau T, Ohmann C, Wolmershäuser A, Wacha H, Yang Q. Planned relaparotomy vs relaparotomy on demand in the treatment of intra-abdominal infections. The Peritonitis Study Group of the Surgical Infection Society-Europe. Arch Surg. 1995 Nov;130(11):1193-6; discussion 1196-7. DOI: 10.1001/archsurg.1995.01430110051009 Externer Link
221.
Hayden MK, Rezai K, Hayes RA, Lolans K, Quinn JP, Weinstein RA. Development of Daptomycin resistance in vivo in methicillin-resistant Staphylococcus aureus. J Clin Microbiol. 2005 Oct;43(10):5285-7. DOI: 10.1128/JCM.43.10.5285-5287.2005 Externer Link
222.
Hedrick TL, McElearney ST, Smith RL, Evans HL, Pruett TL, Sawyer RG. Duration of antibiotic therapy for ventilator-associated pneumonia caused by non-fermentative gram-negative bacilli. Surg Infect (Larchmt). 2007 Dec;8(6):589-97. DOI: 10.1089/sur.2006.021 Externer Link
223.
Hehl EM, Drewelow B. Therapeutisches Drug Monitoring von Aminoglykosiden. In: Hitzenberger G, editors. Therapeutisches Drug Monitoring. Wien: Blackwell-MZV; 1994. p. 117-28.
224.
Heininger A, Unertl K. Beatmungsassoziierte Pneumonie und multiresistente bakterielle Erreger [Ventilator-associated pneumonia and multiresistant bacteria]. Anasthesiol Intensivmed Notfallmed Schmerzther. 2007 Feb;42(2):122-9. DOI: 10.1055/s-2007-971164 Externer Link
225.
Hentschke M, Saager B, Horstkotte MA, Scherpe S, Wolters M, Kabisch H, Grosse R, Heisig P, Aepfelbacher M, Rohde H. Emergence of linezolid resistance in a methicillin resistant Staphylococcus aureus strain. Infection. 2008 Feb;36(1):85-7. DOI: 10.1007/s15010-007-7220-7 Externer Link
226.
Heyland DK, Dodek P, Muscedere J, Day A, Cook D; Canadian Critical Care Trials Group. Randomized trial of combination versus monotherapy for the empiric treatment of suspected ventilator-associated pneumonia. Crit Care Med. 2008 Mar;36(3):737-44. DOI: 10.1097/01.CCM.0B013E31816203D6 Externer Link
227.
Hidayat LK, Hsu DI, Quist R, Shriner KA, Wong-Beringer A. High-dose vancomycin therapy for methicillin-resistant Staphylococcus aureus infections: efficacy and toxicity. Arch Intern Med. 2006 Oct;166(19):2138-44. DOI: 10.1001/archinte.166.19.2138 Externer Link
228.
Hitt CM, Klepser ME, Nightingale CH, Quintiliani R, Nicolau DP. Pharmacoeconomic impact of once-daily aminoglycoside administration. Pharmacotherapy. 1997 Jul-Aug;17(4):810-4.
229.
Hoen B, Alla F, Selton-Suty C, Béguinot I, Bouvet A, Briançon S, Casalta JP, Danchin N, Delahaye F, Etienne J, Le Moing V, Leport C, Mainardi JL, Ruimy R, Vandenesch F; Association pour l'Etude et la Prévention de l'Endocardite Infectieuse (AEPEI) Study Group. Changing profile of infective endocarditis: results of a 1-year survey in France. JAMA. 2002 Jul;288(1):75-81. DOI: 10.1001/jama.288.1.75 Externer Link
230.
Höffken G, Lorenz J, Kern W, Welte T, Bauer T, Dalhoff K, Dietrich E, Ewig S, Gastmeier P, Grabein B, Halle E, Kolditz M, Marre R, Sitter H. Epidemiologie, Diagnostik, antimikrobielle Therapie und Management von erwachsenen Patienten mit ambulant erworbenen unteren Atemwegsinfektionen sowie ambulant erworbener Pneumonie. Pneumologie. 2009;63(10):e1-e68. DOI: 10.1055/s-0029-1215037 Externer Link
231.
Holt S, Thürmann PA. Erstellung einer Liste potenziell inadäquater Medikation für ältere, multimorbide Patienten. In: 7. Deutscher Kongress für Versorgungsforschung des Deutschen Netzwerks für Versorgungsforschung; 16.–18.10.2008; Köln. Düsseldorf: German Medical Science GMS Publishing House; 2008. DocP6.9. Available from: http://www.egms.de/en/meetings/dkvf2008/08dkvf182.shtml Externer Link
232.
Hooper D, Strahilevitz J. Quinolones. In: Mandell GL, Bennett JE, Dolin R, editors. Mandell, Douglas, and Bennett’s Principles and Practice of Infectious Diseases. 7th ed. Churchill Livingstone: Elsevier; 2010. p. 487-510.
233.
Hoyme UB. Tuboovarialabszess, Pelveoperitonitis und septischer Schock. Gynäkologe. 2002;35(4):353-62. DOI: 10.1007/s00129-002-1172-x Externer Link
234.
Innes HE, Smith DB, O'Reilly SM, Clark PI, Kelly V, Marshall E. Oral antibiotics with early hospital discharge compared with in-patient intravenous antibiotics for low-risk febrile neutropenia in patients with cancer: a prospective randomised controlled single centre study. Br J Cancer. 2003 Jul;89(1):43-9. DOI: 10.1038/sj.bjc.6600993 Externer Link
235.
Isenmann R, Rünzi M, Kron M, Kahl S, Kraus D, Jung N, Maier L, Malfertheiner P, Goebell H, Beger HG; German Antibiotics in Severe Acute Pancreatitis Study Group. Prophylactic antibiotic treatment in patients with predicted severe acute pancreatitis: a placebo-controlled, double-blind trial. Gastroenterology. 2004 Apr;126(4):997-1004. DOI: 10.1053/j.gastro.2003.12.050 Externer Link
236.
Jaccard C, Troillet N, Harbarth S, Zanetti G, Aymon D, Schneider R, Chiolero R, Ricou B, Romand J, Huber O, Ambrosetti P, Praz G, Lew D, Bille J, Glauser MP, Cometta A. Prospective randomized comparison of imipenem-cilastatin and piperacillin-tazobactam in nosocomial pneumonia or peritonitis. Antimicrob Agents Chemother. 1998 Nov;42(11):2966-72.
237.
Jackson TL, Williamson TH. Amikacin retinal toxicity. Br J Ophthalmol. 1999 Oct;83(10):1199-200. DOI: 10.1136/bjo.83.10.1194f Externer Link
238.
Jacobson L, Weström L. Objectivized diagnosis of acute pelvic inflammatory disease. Diagnostic and prognostic value of routine laparoscopy. Am J Obstet Gynecol. 1969 Dec;105(7):1088-98.
239.
Jimenez-Cruz F, Jasovich A, Cajigas J, Jiang Q, Imbeault D, Woods GL, Gesser RM; Protocol 021 Study Group. A prospective, multicenter, randomized, double-blind study comparing ertapenem and ceftriaxone followed by appropriate oral therapy for complicated urinary tract infections in adults. Urology. 2002 Jul;60(1):16-22. DOI: 10.1016/S0090-4295(02)01664-3 Externer Link
240.
Johansen TE, Cek M, Naber KG, Stratchounski L, Svendsen MV, Tenke P; PEP and PEAP-study investigators; Board of the European Society of Infections in Urology. Hospital acquired urinary tract infections in urology departments: pathogens, susceptibility and use of antibiotics. Data from the PEP and PEAP-studies. Int J Antimicrob Agents. 2006 Aug;28 Suppl 1:S91-107. DOI: 10.1016/j.ijantimicag.2006.05.005 Externer Link
241.
Jonas JB. Postoperative proprionibacterium acnes endophthalmitis. Ophthalmology. 2001 Apr;108(4):633.
242.
Jones ME. In-vitro profile of a new beta-lactam, ceftobiprole, with activity against methicillin-resistant Staphylococcus aureus. Clin Microbiol Infect. 2007 Jun;13 Suppl 2:17-24. DOI: 10.1111/j.1469-0691.2007.01722.x Externer Link
243.
Jones ME, Karlowsky JA, Draghi DC, Thornsberry C, Sahm DF, Nathwani D. Epidemiology and antibiotic susceptibility of bacteria causing skin and soft tissue infections in the USA and Europe: a guide to appropriate antimicrobial therapy. Int J Antimicrob Agents. 2003 Oct;22(4):406-19. DOI: 10.1016/S0924-8579(03)00154-7 Externer Link
244.
Jones RN, Kohno S, Ono Y, Ross JE, Yanagihara K. ZAAPS International Surveillance Program (2007) for linezolid resistance: results from 5591 Gram-positive clinical isolates in 23 countries. Diagn Microbiol Infect Dis. 2009 Jun;64(2):191-201. DOI: 10.1016/j.diagmicrobio.2009.03.001 Externer Link
245.
Kain HL. Prinzipien bei der Behandlung der Endophthalmitis [Basic principles in treatment of endophthalmitis]. Klin Monbl Augenheilkd. 1997 May;210(5):274-88. DOI: 10.1055/s-2008-1035053 Externer Link
246.
Kanebayashi H, Ogawa Y, Yamanishi T, Fujita H, et al. Clinical investigation of the peritonsillar abscess. Oto-Rhino-Laryngol Tokyo. 2003;46:24-8.
247.
Kaplan SL. Challenges in the evaluation and management of bone and joint infections and the role of new antibiotics for gram positive infections. Adv Exp Med Biol. 2009;634:111-20. DOI: 10.1007/978-0-387-79838-7_10 Externer Link
248.
Kappstein I, van der Mühlen K, Meschzan D, Vatou V, Bieg-Habermann S. Prävention von MRSA-Ubertragungen: Standardhygiene statt Isolierung: 6 Jahre Uberwachung in einem Universitätsklinikum [Prevention of transmission of methicillin-resistant Staphylococcus aureus (MRSA) infection: standard precautions instead of isolation: a 6-year surveillance in a university hospital]. Chirurg. 2009 Jan;80(1):49-61. DOI: 10.1007/s00104-008-1565-z Externer Link
249.
Karageorgopoulos DE, Kelesidis T, Kelesidis I, Falagas ME. Tigecycline for the treatment of multidrug-resistant (including carbapenem-resistant) Acinetobacter infections: a review of the scientific evidence. J Antimicrob Chemother. 2008 Jul;62(1):45-55. DOI: 10.1093/jac/dkn165 Externer Link
250.
Kaufman DW, Kelly JP, Rosenberg L, Anderson TE, Mitchell AA. Recent patterns of medication use in the ambulatory adult population of the United States: the Slone survey. JAMA. 2002 Jan;287(3):337-44. DOI: 10.1001/jama.287.3.337 Externer Link
251.
Kawabata T, Yokota M, Mayama T, Shimizu K. A post-marketing surveillance study of fosfomycin sodium for injection. A nationwide prospective study of safety in 100,000 cases. Jap J Chemother. 2000;48:851-74.
252.
Keller F, Frankewitsch T, Zellner D, Simon S. Unifying concept of pharmacokinetics derived from drug distribution and elimination in renal failure. Int J Clin Pharmacol Ther. 1995 Oct;33(10):546-9.
253.
Kelly JP, Kaufman DW, Kelley K, Rosenberg L, Anderson TE, Mitchell AA. Recent trends in use of herbal and other natural products. Arch Intern Med. 2005 Feb;165(3):281-6. DOI: 10.1001/archinte.165.3.281 Externer Link
254.
Khan S, Sharrack B, Sewell WA. Metronidazole-induced aseptic meningitis during Helicobacter pylori eradication therapy. Ann Intern Med. 2007 Mar 6;146(5):395-6. DOI: 10.7326/0003-4819-146-5-200703060-00016 Externer Link
255.
Kim J, Lee KH, Yoo S, Pai H. Clinical characteristics and risk factors of colistin-induced nephrotoxicity. Int J Antimicrob Agents. 2009 Nov;34(5):434-8. DOI: 10.1016/j.ijantimicag.2009.06.028 Externer Link
256.
Kim SH, Kim KH, Kim HB, Kim NJ, Kim EC, Oh MD, Choe KW. Outcome of vancomycin treatment in patients with methicillin-susceptible Staphylococcus aureus bacteremia. Antimicrob Agents Chemother. 2008 Jan;52(1):192-7. DOI: 10.1128/AAC.00700-07 Externer Link
257.
Kinane DF, Marshall GJ. Periodontal manifestations of systemic disease. Aust Dent J. 2001 Mar;46(1):2-12. DOI: 10.1111/j.1834-7819.2001.tb00267.x Externer Link
258.
Kingston D, Seal DV. Current hypotheses on synergistic microbial gangrene. Br J Surg. 1990 Mar;77(3):260-4. DOI: 10.1002/bjs.1800770309 Externer Link
259.
Kitzis MD, Goldstein FW. Monitoring of vancomycin serum levels for the treatment of staphylococcal infections. Clin Microbiol Infect. 2006 Jan;12(1):92-5. DOI: 10.1111/j.1469-0691.2005.01306.x Externer Link
260.
Klugman KP, Friedland IR, Bradley JS. Bactericidal activity against cephalosporin-resistant Streptococcus pneumoniae in cerebrospinal fluid of children with acute bacterial meningitis. Antimicrob Agents Chemother. 1995 Sep;39(9):1988-92. DOI: 10.1128/AAC.39.9.1988 Externer Link
261.
Knowles S, Choudhury T, Shear NH. Metronidazole hypersensitivity. Ann Pharmacother. 1994 Mar;28(3):325-6.
262.
Koenig SM, Truwit JD. Ventilator-associated pneumonia: diagnosis, treatment, and prevention. Clin Microbiol Rev. 2006 Oct;19(4):637-57. DOI: 10.1128/CMR.00051-05 Externer Link
263.
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 Feb;115(2):462-74. DOI: 10.1378/chest.115.2.462 Externer Link
264.
Kommission „Leitlinien der Deutschen Gesellschaft für Neurologie“. Leitlinien für Diagnostik und Therapie in der Neurologie. 4. überarb. Aufl. Stuttgart, New York: Thieme; 2008.
265.
Koorbusch GF, Fotos P, Goll KT. Retrospective assessment of osteomyelitis. Etiology, demographics, risk factors, and management in 35 cases. Oral Surg Oral Med Oral Pathol. 1992 Aug;74(2):149-54. DOI: 10.1016/0030-4220(92)90373-X Externer Link
266.
Krepel CJ, Gohr CM, Edmiston CE, Condon RE. Surgical sepsis: constancy of antibiotic susceptibility of causative organisms. Surgery. 1995 May;117(5):505-9. DOI: 10.1016/S0039-6060(05)80249-1 Externer Link
267.
Kresken M, Hafner D, Schmitz FJ, Wichelhaus TA. Resistenz bei häufig isolierten Enterobacteriaceae gegenüber Breitspektrumantibiotika. Chemother J. 2006;15(6):179-90.
268.
Kresken M, Leitner E, Seifert H, Peters G, von Eiff C. Susceptibility of clinical isolates of frequently encountered bacterial species to tigecycline one year after the introduction of this new class of antibiotics: results of the second multicentre surveillance trial in Germany (G-TEST II, 2007). Eur J Clin Microbiol Infect Dis. 2009 Aug;28(8):1007-11. DOI: 10.1007/s10096-009-0725-5 Externer Link
269.
Krueger WA, Bulitta J, Kinzig-Schippers M, Landersdorfer C, Holzgrabe U, Naber KG, Drusano GL, Sörgel F. Evaluation by monte carlo simulation of the pharmacokinetics of two doses of meropenem administered intermittently or as a continuous infusion in healthy volunteers. Antimicrob Agents Chemother. 2005 May;49(5):1881-9. DOI: 10.1128/AAC.49.5.1881-1889.2005 Externer Link
270.
Kuang D, Verbine A, Ronco C. Pharmacokinetics and antimicrobial dosing adjustment in critically ill patients during continuous renal replacement therapy. Clin Nephrol. 2007 May;67(5):267-84. DOI: 10.5414/CNP67267 Externer Link
271.
Kujath P, editor. Mykosen in der operativen Medizin. Grundlagen der Diagnostik und Therapie. 1st ed. Stuttgart, New York: Thieme; 1998. p. 21-4.
272.
Kujath P. Haut- und Weichgewebsinfektionen. 2nd ed. Bremen: Uni-Med; 2004.
273.
Kujath P, Michelsen A. Wounds – from physiology to wound dressing. Dtsch Arztebl Int. 2008 Mar;105(13):239-48. DOI: 10.3238/arztebl.2008.0239 Externer Link
274.
Kujath P, Rodloff AC, editors. Peritonitis. 2nd ed. Heidelberg: Uni-Med Verlag; 2005. p. 13-5.
275.
Kumar A, Ellis P, Arabi Y, Roberts D, Light B, Parrillo JE, Dodek P, Wood G, Kumar A, Simon D, Peters C, Ahsan M, Chateau D; Cooperative Antimicrobial Therapy of Septic Shock Database Research Group. Initiation of inappropriate antimicrobial therapy results in a fivefold reduction of survival in human septic shock. Chest. 2009 Nov;136(5):1237-48. DOI: 10.1378/chest.09-0087 Externer Link
276.
Kumar A, Roberts D, Wood KE, Light B, Parrillo JE, Sharma S, Suppes R, Feinstein D, Zanotti S, Taiberg L, Gurka D, Kumar A, Cheang M. Duration of hypotension before initiation of effective antimicrobial therapy is the critical determinant of survival in human septic shock. Crit Care Med. 2006 Jun;34(6):1589-96. DOI: 10.1097/01.CCM.0000217961.75225.E9 Externer Link
277.
Kunimoto DY, Das T, Sharma S, Jalali S, Majji AB, Gopinathan U, Athmanathan S, Rao TN. Microbiologic spectrum and susceptibility of isolates: part II. Posttraumatic endophthalmitis. Endophthalmitis Research Group. Am J Ophthalmol. 1999 Aug;128(2):242-4. DOI: 10.1016/S0002-9394(99)00113-0 Externer Link
278.
Kuntz P, Pieringer-Mueller E, Hof H. Infektionsgefährdung durch Bissverletzungen. Dtsch Arztebl. 1996;93:A969-72.
279.
Kuriyama T, Karasawa T, Nakagawa K, Nakamura S, Yamamoto E. Antimicrobial susceptibility of major pathogens of orofacial odontogenic infections to 11 beta-lactam antibiotics. Oral Microbiol Immunol. 2002 Oct;17(5):285-9. DOI: 10.1034/j.1399-302X.2002.170504.x Externer Link
280.
Kuriyama T, Karasawa T, Nakagawa K, Yamamoto E, Nakamura S. Incidence of beta-lactamase production and antimicrobial susceptibility of anaerobic gram-negative rods isolated from pus specimens of orofacial odontogenic infections. Oral Microbiol Immunol. 2001 Feb;16(1):10-5. DOI: 10.1034/j.1399-302x.2001.160102.x Externer Link
281.
Kuriyama T, Nakagawa K, Karasawa T, Saiki Y, Yamamoto E, Nakamura S. Past administration of beta-lactam antibiotics and increase in the emergence of beta-lactamase-producing bacteria in patients with orofacial odontogenic infections. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2000 Feb;89(2):186-92. DOI: 10.1067/moe.2000.102040 Externer Link
282.
Kuti JL, Dandekar PK, Nightingale CH, Nicolau DP. Use of Monte Carlo simulation to design an optimized pharmacodynamic dosing strategy for meropenem. J Clin Pharmacol. 2003 Oct;43(10):1116-23. DOI: 10.1177/0091270003257225 Externer Link
283.
Kuti JL, Nightingale CH, Knauft RF, Nicolau DP. Pharmacokinetic properties and stability of continuous-infusion meropenem in adults with cystic fibrosis. Clin Ther. 2004 Apr;26(4):493-501. DOI: 10.1016/S0149-2918(04)90051-3 Externer Link
284.
Kuzemko J, Crawford C. Continuous infusion of ceftazidime in cystic fibrosis. Lancet. 1989 Aug;2(8659):385. DOI: 10.1016/S0140-6736(89)90561-8 Externer Link
285.
Ladhani S, Neely F, Heath PT, Nazareth B, Roberts R, Slack MP, McVernon J, Ramsay ME. Recommendations for the prevention of secondary Haemophilus influenzae type b (Hib) disease. J Infect. 2009 Jan;58(1):3-14. DOI: 10.1016/j.jinf.2008.10.007 Externer Link
286.
Lamey PJ, Boyle MA, MacFarlane TW, Samaranayake LP. Acute suppurative parotitis in outpatients: microbiologic and posttreatment sialographic findings. Oral Surg Oral Med Oral Pathol. 1987 Jan;63(1):37-41. DOI: 10.1016/0030-4220(87)90337-9 Externer Link
287.
Lancaster J, Alderson DJ, McCormick M. Non-pseudomonal malignant otitis externa and jugular foramen syndrome secondary to cyclosporin-induced hypertrichosis in a diabetic renal transplant patient. J Laryngol Otol. 2000 May;114(5):366-9. DOI: 10.1258/0022215001905580 Externer Link
288.
Landersdorfer CB, Kirkpatrick CM, Kinzig-Schippers M, Bulitta JB, Holzgrabe U, Drusano GL, Sörgel F. Population pharmacokinetics at two dose levels and pharmacodynamic profiling of flucloxacillin. Antimicrob Agents Chemother. 2007 Sep;51(9):3290-7. DOI: 10.1128/AAC.01410-06 Externer Link
289.
LaPlante KL, Rybak MJ. Daptomycin – a novel antibiotic against Gram-positive pathogens. Expert Opin Pharmacother. 2004 Nov;5(11):2321-31. DOI: 10.1517/14656566.5.11.2321 Externer Link
290.
Laroche ML, Charmes JP, Merle L. Potentially inappropriate medications in the elderly: a French consensus panel list. Eur J Clin Pharmacol. 2007 Aug;63(8):725-31. DOI: 10.1007/s00228-007-0324-2 Externer Link
291.
Lau WK, Mercer D, Itani KM, Nicolau DP, Kuti JL, Mansfield D, Dana A. Randomized, open-label, comparative study of piperacillin-tazobactam administered by continuous infusion versus intermittent infusion for treatment of hospitalized patients with complicated intra-abdominal infection. Antimicrob Agents Chemother. 2006 Nov;50(11):3556-61. DOI: 10.1128/AAC.00329-06 Externer Link
292.
Lavole GY, Bergeron MG. Influence of four modes of administration on penetration of aztreonam, cefuroxime, and ampicillin into interstitial fluid and fibrin clots and on invivo efficacy against Haemophilus influenzae. Antimicrob Agents Chemother. 1985;28:404-12. DOI: 10.1128/AAC.28.3.404 Externer Link
293.
Lebel MH, McCracken GH Jr. Delayed cerebrospinal fluid sterilization and adverse outcome of bacterial meningitis in infants and children. Pediatrics. 1989 Feb;83(2):161-7.
294.
Leclerc S, Teixeira A, Mahé E, Descamps V, Crickx B, Chosidow O. Recurrent erysipelas: 47 cases. Dermatology (Basel). 2007;214(1):52-7. DOI: 10.1159/000096913 Externer Link
295.
Lee BL, Sachdeva M, Chambers HF. Effect of protein binding of daptomycin on MIC and antibacterial activity. Antimicrob Agents Chemother. 1991 Dec;35(12):2505-8.
296.
Lehner S, Grabein B, Pfaller P, Kopp R. Bedeutung ESBL-produzierender Keime für die klinische Chirurgie : Diagnostik, Prävention und Therapie [Relevance of ESBL-producing pathogens for clinical surgery: diagnostics, therapy, and prevention]. Chirurg. 2009 Jun;80(6):527-36. DOI: 10.1007/s00104-008-1664-x Externer Link
297.
Levine BB. Immunologic mechanisms of penicillin allergy. A haptenic model system for the study of allergic diseases of man. N Engl J Med. 1966 Nov;275(20):1115-25. DOI: 10.1056/NEJM196611172752009 Externer Link
298.
Levine DP. Clinical experience with daptomycin: bacteraemia and endocarditis. J Antimicrob Chemother. 2008 Nov;62 Suppl 3:iii35-39. DOI: 10.1093/jac/dkn369 Externer Link
299.
Lewis MA, Lamey PJ, Gibson J. Quantitative bacteriology of a case of acute parotitis. Oral Surg Oral Med Oral Pathol. 1989 Nov;68(5):571-5. DOI: 10.1016/0030-4220(89)90242-9 Externer Link
300.
Lewis RT. Necrotizing soft-tissue infections. Infect Dis Clin North Am. 1992 Sep;6(3):693-703.
301.
Li J, Nation RL, Turnidge JD, Milne RW, Coulthard K, Rayner CR, Paterson DL. Colistin: the re-emerging antibiotic for multidrug-resistant Gram-negative bacterial infections. Lancet Infect Dis. 2006 Sep;6(9):589-601. DOI: 10.1016/S1473-3099(06)70580-1 Externer Link
302.
Lima RC, Barreira A, Cardoso FL, Lima MH, Leite M Jr. Ciprofloxacin and cefazolin as a combination for empirical initial therapy of peritoneal dialysis-related peritonitis: five-year follow-up. Perit Dial Int. 2007 Jan-Feb;27(1):56-60.
303.
Lipman J, Boots R. A new paradigm for treating infections: “go hard and go home”. Crit Care Resusc. 2009 Dec;11(4):276-81.
304.
Lipsky BA, Berendt AR, Deery HG, Embil JM, Joseph WS, Karchmer AW, LeFrock JL, Lew DP, Mader JT, Norden C, Tan JS; Infectious Diseases Society of America. Diagnosis and treatment of diabetic foot infections. Clin Infect Dis. 2004 Oct;39(7):885-910. DOI: 10.1086/424846 Externer Link
305.
Lipsky BA, Weigelt JA, Gupta V, Killian A, Peng MM. Skin, soft tissue, bone, and joint infections in hospitalized patients: epidemiology and microbiological, clinical, and economic outcomes. Infect Control Hosp Epidemiol. 2007 Nov;28(11):1290-8. DOI: 10.1086/520743 Externer Link
306.
Livengood III CH. Tubo-ovarian abscess. In: Mead PB, Hager WD, Faro S, editors. Protocols for infectious diseases in obstetrics and gynecology. 2nd ed. Malden: Blackwell Science Inc; 2000. p. 412-8.
307.
Livermore DM. Beta-Lactamases in laboratory and clinical resistance. Clin Microbiol Rev. 1995 Oct;8(4):557-84.
308.
Livermore DM. Future directions with daptomycin. J Antimicrob Chemother. 2008 Nov;62 Suppl 3:iii41-iii49. DOI: 10.1093/jac/dkn371 Externer Link
309.
Llopis-Salvia P, Jiménez-Torres NV. Population pharmacokinetic parameters of vancomycin in critically ill patients. J Clin Pharm Ther. 2006 Oct;31(5):447-54. DOI: 10.1111/j.1365-2710.2006.00762.x Externer Link
310.
Lodise TP, Lomaestro B, Graves J, Drusano GL. Larger vancomycin doses (at least four grams per day) are associated with an increased incidence of nephrotoxicity. Antimicrob Agents Chemother. 2008 Apr;52(4):1330-6. DOI: 10.1128/AAC.01602-07 Externer Link
311.
Long KS, Poehlsgaard J, Kehrenberg C, Schwarz S, Vester B. The Cfr rRNA methyltransferase confers resistance to Phenicols, Lincosamides, Oxazolidinones, Pleuromutilins, and Streptogramin A antibiotics. Antimicrob Agents Chemother. 2006 Jul;50(7):2500-5. DOI: 10.1128/AAC.00131-06 Externer Link
312.
Lorente L, Lorenzo L, Martín MM, Jiménez A, Mora ML. Meropenem by continuous versus intermittent infusion in ventilator-associated pneumonia due to gram-negative bacilli. Ann Pharmacother. 2006 Feb;40(2):219-23. DOI: 10.1345/aph.1G467 Externer Link
313.
Lortholary O, Lefort A, Tod M, Chomat AM, Darras-Joly C, Cordonnier C. Pharmacodynamics and pharmacokinetics of antibacterial drugs in the management of febrile neutropenia. Lancet Infect Dis. 2008 Oct;8(10):612-20. DOI: 10.1016/S1473-3099(08)70228-7 Externer Link
314.
Lucasti C, Jasovich A, Umeh O, Jiang J, Kaniga K, Friedland I. Efficacy and tolerability of IV doripenem versus meropenem in adults with complicated intra-abdominal infection: a phase III, prospective, multicenter, randomized, double-blind, noninferiority study. Clin Ther. 2008 May;30(5):868-83. DOI: 10.1016/j.clinthera.2008.04.019 Externer Link
315.
MacGowan AP. Pharmacodynamics, pharmacokinetics, and therapeutic drug monitoring of glycopeptides. Ther Drug Monit. 1998 Oct;20(5):473-7. DOI: 10.1097/00007691-199810000-00005 Externer Link
316.
Malangoni MA, Song J, Herrington J, Choudhri S, Pertel P. Randomized controlled trial of moxifloxacin compared with piperacillin-tazobactam and amoxicillin-clavulanate for the treatment of complicated intra-abdominal infections. Ann Surg. 2006 Aug;244(2):204-11. DOI: 10.1097/01.sla.0000230024.84190.a8 Externer Link
317.
Mamalis N, Kearsley L, Brinton E. Postoperative endophthalmitis. Curr Opin Ophthalmol. 2002 Feb;13(1):14-8. DOI: 10.1097/00055735-200202000-00004 Externer Link
318.
Mandell LA, Bergeron MG, Ronald AR, Vega C, Harding G, Saginur R, Feld R, Duperval R, Landis SJ, Miedzinski LJ. Once-daily therapy with ceftriaxone compared with daily multiple-dose therapy with cefotaxime for serious bacterial infections: a randomized, double-blind study. J Infect Dis. 1989 Sep;160(3):433-41.
319.
Marcos MA, Martínez E, Almela M, Mensa J, Jiménez de Anta MT. New rapid antigen test for diagnosis of pneumococcal meningitis. Lancet. 2001 May;357(9267):1499-500. DOI: 10.1016/S0140-6736(00)04658-4 Externer Link
320.
Martin MV. The use of oral amoxycillin for the treatment of actinomycosis. A clinical and in vitro study. Br Dent J. 1984 Apr;156(7):252-4. DOI: 10.1038/sj.bdj.4805331 Externer Link
321.
Martínez E, Collazos J, Mayo J. Hypersensitivity reactions to rifampin. Pathogenetic mechanisms, clinical manifestations, management strategies, and review of the anaphylactic-like reactions. Medicine (Baltimore). 1999 Nov;78(6):361-9. DOI: 10.1097/00005792-199911000-00001 Externer Link
322.
Martone WJ, Lamp KC. Efficacy of daptomycin in complicated skin and skin-structure infections due to methicillin-sensitive and -resistant Staphylococcus aureus: results from the CORE Registry. Curr Med Res Opin. 2006 Dec;22(12):2337-43. DOI: 10.1185/030079906X148427 Externer Link
323.
Marx RE. Chronic Osteomyelitis of the jaws. Oral Maxillofac Surg Clin North Am. 1991;3:367-81.
324.
Masterton RG, Galloway A, French G, Street M, Armstrong J, Brown E, Cleverley J, Dilworth P, Fry C, Gascoigne AD, Knox A, Nathwani D, Spencer R, Wilcox M. Guidelines for the management of hospital-acquired pneumonia in the UK: report of the working party on hospital-acquired pneumonia of the British Society for Antimicrobial Chemotherapy. J Antimicrob Chemother. 2008 Jul;62(1):5-34. DOI: 10.1093/jac/dkn162 Externer Link
325.
Matthews PC, Berendt AR, McNally MA, Byren I. Diagnosis and management of prosthetic joint infection. BMJ. 2009;338:b1773. DOI: 10.1185/030079906X148427 Externer Link
326.
Mayer KU, Baltes PB, editors. Die Berliner Altersstudie. Berlin: Akademie Verlag; 1996.
327.
Mazaki T, Ishii Y, Takayama T. Meta-analysis of prophylactic antibiotic use in acute necrotizing pancreatitis. Br J Surg. 2006 Jun;93(6):674-84. DOI: 10.1002/bjs.5389 Externer Link
328.
Mazuski JE. Antimicrobial treatment for intra-abdominal infections. Expert Opin Pharmacother. 2007 Dec;8(17):2933-45. DOI: 10.1517/14656566.8.17.2933 Externer Link
329.
McCollum M, Rhew DC, Parodi S. Cost analysis of switching from i.v. vancomycin to p.o. linezolid for the management of methicillin-resistant Staphylococcus species. Clin Ther. 2003 Dec;25(12):3173-89. DOI: 10.1016/S0149-2918(03)90101-9 Externer Link
330.
McLeod PJ, Huang AR, Tamblyn RM, Gayton DC. Defining inappropriate practices in prescribing for elderly people: a national consensus panel. CMAJ. 1997 Feb;156(3):385-91.
331.
Megalamani SB, Suria G, Manickam U, Balasubramanian D, Jothimahalingam S. Changing trends in bacteriology of peritonsillar abscess. J Laryngol Otol. 2008 Sep;122(9):928-30. DOI: 10.1017/S0022215107001144 Externer Link
332.
Menéndez R, Reyes S, Martínez R, de la Cuadra P, Manuel Vallés J, Vallterra J. Economic evaluation of adherence to treatment guidelines in nonintensive care pneumonia. Eur Respir J. 2007 Apr;29(4):751-6. DOI: 10.1183/09031936.00052506 Externer Link
333.
Mertz D, Koller M, Haller P, Lampert ML, Plagge H, Hug B, Koch G, Battegay M, Flückiger U, Bassetti S. Outcomes of early switching from intravenous to oral antibiotics on medical wards. J Antimicrob Chemother. 2009 Jul;64(1):188-99. DOI: 10.1093/jac/dkp131 Externer Link
334.
Metaxas EI, Falagas ME. Update on the safety of linezolid. Expert Opin Drug Saf. 2009 Jul;8(4):485-91. DOI: 10.1517/14740330903049706 Externer Link
335.
Michalopoulos AS, Tsiodras S, Rellos K, Mentzelopoulos S, Falagas ME. Colistin treatment in patients with ICU-acquired infections caused by multiresistant Gram-negative bacteria: the renaissance of an old antibiotic. Clin Microbiol Infect. 2005 Feb;11(2):115-21. DOI: 10.1111/j.1469-0691.2004.01043.x Externer Link
336.
Michalski G, Hocke T, Hoffmann K, Esser D. Die Mastoiditis als Folge einer akuten Otitis media – Eine retrospektive Studie [Therapy of acute mastoiditis]. Laryngorhinootologie. 2002 Dec;81(12):857-60. DOI: 10.1055/s-2002-36098 Externer Link
337.
Millán-Rodríguez F, Palou J, Bujons-Tur A, Musquera-Felip M, Sevilla-Cecilia C, Serrallach-Orejas M, Baez-Angles C, Villavicencio-Mavrich H. Acute bacterial prostatitis: two different sub-categories according to a previous manipulation of the lower urinary tract. World J Urol. 2006 Feb;24(1):45-50. DOI: 10.1007/s00345-005-0040-4 Externer Link
338.
Miño de Kaspar H, Kollmann M, Klauss V. Endophthalmitis. Bedeutung mikrobiologischer Untersuchungen für Therapie und Prognose [Endophthalmitis. Importance of microbiologic studies for therapy and prognosis]. Ophthalmologe. 1993 Dec;90(6):726-36.
339.
Mischkowski RA, Hidding J, Gruber G, Klesper B, Fangmann R. Risikofaktoren und Management von Abszessen im MKG-Bereich – Eine retrospektive Studie von über 1000 Fällen. Dtsch Zahnarztl Z. 1997;52:697-700.
340.
Moise PA, Sakoulas G, Forrest A, Schentag JJ. Vancomycin in vitro bactericidal activity and its relationship to efficacy in clearance of methicillin-resistant Staphylococcus aureus bacteremia. Antimicrob Agents Chemother. 2007 Jul;51(7):2582-6. DOI: 10.1128/AAC.00939-06 Externer Link
341.
Molyneux EM, Walsh AL, Forsyth H, Tembo M, Mwenechanya J, Kayira K, Bwanaisa L, Njobvu A, Rogerson S, Malenga G. Dexamethasone treatment in childhood bacterial meningitis in Malawi: a randomised controlled trial. Lancet. 2002 Jul;360(9328):211-8. DOI: 10.1016/S0140-6736(02)09458-8 Externer Link
342.
Monane M, Monane S, Semla T. Optimal medication use in elders. Key to successful aging. West J Med. 1997 Oct;167(4):233-7.
343.
Montravers P, Dupont H, Gauzit R, Veber B, Auboyer C, Blin P, Hennequin C, Martin C. Candida as a risk factor for mortality in peritonitis. Crit Care Med. 2006 Mar;34(3):646-52. DOI: 10.1097/01.CCM.0000201889.39443.D2 Externer Link
344.
Moore RD, Lietman PS, Smith CR. Clinical response to aminoglycoside therapy: importance of the ratio of peak concentration to minimal inhibitory concentration. J Infect Dis. 1987 Jan;155(1):93-9. DOI: 10.1093/infdis/155.1.93 Externer Link
345.
Mori H, Takahashi K, Mizutani T. Interaction between valproic acid and carbapenem antibiotics. Drug Metab Rev. 2007;39(4):647-57. DOI: 10.1080/03602530701690341 Externer Link
346.
Mouton JW, Horrevorts AM, Mulder PG, Prens EP, Michel MF. Pharmacokinetics of ceftazidime in serum and suction blister fluid during continuous and intermittent infusions in healthy volunteers. Antimicrob Agents Chemother. 1990 Dec;34(12):2307-11. DOI: 10.1128/AAC.34.12.2307 Externer Link
347.
Mouton JW, Michel MF. Pharmacokinetics of meropenem in serum and suction blister fluid during continuous and intermittent infusion. J Antimicrob Chemother. 1991 Dec;28(6):911-8. DOI: 10.1093/jac/28.6.911 Externer Link
348.
Mouton JW, Vinks AA, Punt NC. Pharmacokinetic-pharmacodynamic modeling of activity of ceftazidime during continuous and intermittent infusion. Antimicrob Agents Chemother. 1997 Apr;41(4):733-8.
349.
Munckhof WJ, Carney J, Neilson G, Neilson J, Carroll J, McWhinney B, Whitby M. Continuous infusion of ticarcillin-clavulanate for home treatment of serious infections: clinical efficacy, safety, pharmacokinetics and pharmacodynamics. Int J Antimicrob Agents. 2005 Jun;25(6):514-22. DOI: 10.1016/j.ijantimicag.2005.02.008 Externer Link
350.
Murray B, Nannini EC. Glycopeptides (Vancomycin and Teicoplanin), Streptogramins (Quinupristin-Dalfopristin), and Lipopeptides (Daptomycin). In: Mandell GL, Bennett JE, Dolin R, editors. Mandell, Douglas, and Bennett’s Principles and Practice of Infectious Diseases. 7th ed. Churchill Livingstone: Elsevier; 2010. p. 449-67.
351.
Naber CK, Baddour LM, Giamarellos-Bourboulis EJ, Gould IM, Herrmann M, Hoen B, Karchmer AW, Kobayashi Y, Kozlov RS, Lew D, Miró JM, Moellering RC Jr, Moreillon P, Peters G, Rubinstein E, Seifert H, Corey GR. Clinical consensus conference: survey on Gram-positive bloodstream infections with a focus on Staphylococcus aureus. Clin Infect Dis. 2009 May;48 Suppl 4:S260-70. DOI: 10.1086/598185 Externer Link
352.
Naber KG, Eisenstein BI, Tally FP. Daptomycin versus ciprofloxacin in the treatment of complicated urinary tract infection due to grampositive bacteria. Infect Dis Clin Pract (Baltim Md). 2004;12(6):322-7. DOI: 10.1097/01.idc.0000144898.12957.eb Externer Link
353.
Naber KG, Hauke W. Cefpodoxime proxetil in patients with acute uncomplicated pyelonephritis. International, prospective, randomized comparative study versus ciprofloxacin in general practice. Chemother J. 2001;10:29-34.
354.
Naber KG, Landen H. Rapid resolution of symptoms with ciprofloxacin therapy in 3859 hospitalised patients with urinary tract infection. Int J Antimicrob Agents. 2004 Mar;23 Suppl 1:S35-40. DOI: 10.1016/j.ijantimicag.2003.09.010 Externer Link
355.
Naber KG, Llorens L, Kaniga K, Kotey P, Hedrich D, Redman R. Intravenous doripenem at 500 milligrams versus levofloxacin at 250 milligrams, with an option to switch to oral therapy, for treatment of complicated lower urinary tract infection and pyelonephritis. Antimicrob Agents Chemother. 2009 Sep;53(9):3782-92. DOI: 10.1128/AAC.00837-08 Externer Link
356.
Naber KG, Savov O, Salmen HC. Piperacillin 2 g/tazobactam 0.5 g is as effective as imipenem 0.5 g/cilastatin 0.5 g for the treatment of acute uncomplicated pyelonephritis and complicated urinary tract infections. Int J Antimicrob Agents. 2002 Feb;19(2):95-103. DOI: 10.1016/S0924-8579(01)00481-2 Externer Link
357.
Naber KG, Schito G, Botto H, Palou J, Mazzei T. Surveillance study in Europe and Brazil on clinical aspects and Antimicrobial Resistance Epidemiology in Females with Cystitis (ARESC): implications for empiric therapy. Eur Urol. 2008 Nov;54(5):1164-75. DOI: 10.1016/j.eururo.2008.05.010 Externer Link
358.
Naber KG, Timmler R. Keimelimination durch Fosfomycin bei komplizierten Harnwegsinfektionen. Therapiewoche. 1983;33:3300-6.
359.
Nahin RL, Fitzpatrick AL, Williamson JD, Burke GL, Dekosky ST, Furberg C; GEM Study Investigators. Use of herbal medicine and other dietary supplements in community-dwelling older people: Baseline data from the ginkgo evaluation of memory study. J Am Geriatr Soc. 2006 Nov;54(11):1725-35. DOI: 10.1111/j.1532-5415.2006.00942.x Externer Link
360.
Narang S, Gupta V, Gupta A, Dogra MR, Pandav SS, Das S. Role of prophylactic intravitreal antibiotics in open globe injuries. Indian J Ophthalmol. 2003 Mar;51(1):39-44.
361.
Narita M, Tsuji BT, Yu VL. Linezolid-associated peripheral and optic neuropathy, lactic acidosis, and serotonin syndrome. Pharmacotherapy. 2007 Aug;27(8):1189-97. DOI: 10.1592/phco.27.8.1189 Externer Link
362.
Nash DB, Koenig JB, Chatterton ML. Why the elderly need individualized pharmaceutical care. Philadelphia: Office of Health Policy and Clinical Outcomes, Jefferson University; 2000. Available from: http://www.npcnow.org/system/files/research/download/Why-the-Elderly-Need-Individualized-Pharmaceutical-Care.pdf Externer Link
363.
Nath SK, Foster GA, Mandell LA, Rotstein C. Antimicrobial activity of ceftriaxone versus cefotaxime: negative effect of serum albumin binding of ceftriaxone. J Antimicrob Chemother. 1994 Jun;33(6):1239-43. DOI: 10.1093/jac/33.6.1239 Externer Link
364.
Nathens AB, Cook CH, Machiedo G, Moore EE, Namias N, Nwariaku F. Defining the research agenda for surgical infection: a consensus of experts using the Delphi approach. Surg Infect (Larchmt). 2006 Apr;7(2):101-10. DOI: 10.1089/sur.2006.7.101 Externer Link
365.
Nathens AB, Curtis JR, Beale RJ, Cook DJ, Moreno RP, Romand JA, Skerrett SJ, Stapleton RD, Ware LB, Waldmann CS. Management of the critically ill patient with severe acute pancreatitis. Crit Care Med. 2004 Dec;32(12):2524-36. DOI: 10.1097/01.CCM.0000148222.09869.92 Externer Link
366.
Nathens AB, Rotstein OD, Marshall JC. Tertiary peritonitis: clinical features of a complex nosocomial infection. World J Surg. 1998 Feb;22(2):158-63. DOI: 10.1007/s002689900364 Externer Link
367.
Nation RL, Li J. Colistin in the 21st century. Curr Opin Infect Dis. 2009 Dec;22(6):535-43. DOI: 10.1097/QCO.0b013e328332e672 Externer Link
368.
Nau R, Brück W. Neuronal injury in bacterial meningitis: mechanisms and implications for therapy. Trends Neurosci. 2002 Jan;25(1):38-45. DOI: 10.1016/S0166-2236(00)02024-5 Externer Link
369.
Neftel KA, Wälti M, Schulthess HK, Gubler J. Adverse reactions following intravenous penicillin-G relate to degradation of the drug in vitro. Klin Wochenschr. 1984 Jan;62(1):25-9. DOI: 10.1093/jac/33.6.1239 Externer Link
370.
Neftel KA, Wälti M, Spengler H, de Weck AL. Effect of storage of penicillin-G solutions on sensitisation to penicillin-G after intravenous administration. Lancet. 1982 May;1(8279):986-8. DOI: 10.1016/S0140-6736(82)91991-2 Externer Link
371.
Neftel KA, Wälti M, Spengler H, von Felten A, Weitzman SA, Bürgi H, de Weck AL. Neutropenia after penicillins: toxic or immune-mediated? Klin Wochenschr. 1981 Aug;59(16):877-88. DOI: 10.1007/BF01721921 Externer Link
372.
Neuhauser MM, Weinstein RA, Rydman R, Danziger LH, Karam G, Quinn JP. Antibiotic resistance among gram-negative bacilli in US intensive care units: implications for fluoroquinolone use. JAMA. 2003 Feb;289(7):885-8. DOI: 10.1001/jama.289.7.885 Externer Link
373.
Newman D, Scheetz MH, Adeyemi OA, Montevecchi M, Nicolau DP, Noskin GA, Postelnick MJ. Serum piperacillin/tazobactam pharmacokinetics in a morbidly obese individual. Ann Pharmacother. 2007 Oct;41(10):1734-9. DOI: 10.1345/aph.1K256 Externer Link
374.
Nichols RL, Muzik AC. Enterococcal infections in surgical patients: the mystery continues. Clin Infect Dis. 1992 Jul;15(1):72-6. DOI: 10.1093/clinids/15.1.72 Externer Link
375.
Nicolau DP. Pharmacodynamic optimization of beta-lactams in the patient care setting. Crit Care. 2008;12 Suppl 4:S2. DOI: 10.1186/cc6818 Externer Link
376.
Nicolau DP, McNabb J, Lacy MK, Quintiliani R, Nightingale CH. Continuous versus intermittent administration of ceftazidime in intensive care unit patients with nosocomial pneumonia. Int J Antimicrob Agents. 2001 Jun;17(6):497-504. DOI: 10.1016/S0924-8579(01)00329-6 Externer Link
377.
Nicolau DP, McNabb J, Lacy MK, Quintiliani R, Nightingale CH. Pharmacokinetics of continuous and intermittent ceftazidime in intensive care unit patients with nosocomial pneumonia. Infect Dis Clin Pract. 1999;8:45-9. DOI: 10.1097/00019048-199901000-00009 Externer Link
378.
Niederman MS. Use of broad-spectrum antimicrobials for the treatment of pneumonia in seriously ill patients: maximizing clinical outcomes and minimizing selection of resistant organisms. Clin Infect Dis. 2006 Jan;42 Suppl 2:S72-81. DOI: 10.1086/499405 Externer Link
379.
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 Mar;177(5):498-505. DOI: 10.1164/rccm.200708-1238OC Externer Link
380.
Noel GJ. Clinical profile of ceftobiprole, a novel beta-lactam antibiotic. Clin Microbiol Infect. 2007 Jun;13 Suppl 2:25-9. DOI: 10.1111/j.1469-0691.2007.01725.x Externer Link
381.
Noel GJ, Bush K, Bagchi P, Ianus J, Strauss RS. A randomized, double-blind trial comparing ceftobiprole medocaril with vancomycin plus ceftazidime for the treatment of patients with complicated skin and skin-structure infections. Clin Infect Dis. 2008 Mar;46(5):647-55. DOI: 10.1086/526527 Externer Link
382.
Nørskov-Lauritsen N, Marchandin H, Dowzicky MJ. Antimicrobial susceptibility of tigecycline and comparators against bacterial isolates collected as part of the TEST study in Europe (2004-2007). Int J Antimicrob Agents. 2009 Aug;34(2):121-30. DOI: 10.1016/j.ijantimicag.2009.02.003 Externer Link
383.
Okada AA, Johnson RP, Liles WC, D'Amico DJ, Baker AS. Endogenous bacterial endophthalmitis. Report of a ten-year retrospective study. Ophthalmology. 1994 May;101(5):832-8. DOI: 10.1016/S0161-6420(13)31255-X Externer Link
384.
Okhravi N, Guest S, Matheson MM, Kees F, Ficker LA, Tuft SJ, Lightman S. Assessment of the effect of oral clarithromycin on visual outcome following presumed bacterial endophthalmitis. Curr Eye Res. 2000 Sep;21(3):691-702. DOI: 10.1076/0271-3683(200009)2131-RFT691 Externer Link
385.
Oleson FB Jr, Berman CL, Kirkpatrick JB, Regan KS, Lai JJ, Tally FP. Once-daily dosing in dogs optimizes daptomycin safety. Antimicrob Agents Chemother. 2000 Nov;44(11):2948-53. DOI: 10.1128/AAC.44.11.2948-2953.2000 Externer Link
386.
Olsen KM, Rudis MI, Rebuck JA, Hara J, Gelmont D, Mehdian R, Nelson C, Rupp ME. Effect of once-daily dosing vs. multiple daily dosing of tobramycin on enzyme markers of nephrotoxicity. Crit Care Med. 2004 Aug;32(8):1678-82. DOI: 10.1097/01.CCM.0000134832.11144.CB Externer Link
387.
Olson RJ. Reducing the risk of postoperative endophthalmitis. Surv Ophthalmol. 2004 Mar;49 Suppl 2:S55-61. DOI: 10.1016/j.survophthal.2004.01.002 Externer Link
388.
Ooi ST, Frazee LA, Gardner WG. Management of asymptomatic bacteriuria in patients with diabetes mellitus. Ann Pharmacother. 2004 Mar;38(3):490-3. DOI: 10.1345/aph.1D355 Externer Link
389.
Ott SR, Allewelt M, Lorenz J, Reimnitz P, Lode H; German Lung Abscess Study Group. Moxifloxacin vs ampicillin/sulbactam in aspiration pneumonia and primary lung abscess. Infection. 2008 Feb;36(1):23-30. DOI: 10.1007/s15010-007-7043-6 Externer Link
390.
Otten JE, Weingart D, Pelz K, Poukens J. Bakterien in Sekreten extra- und intraoraler Operationswunden Kontamination oder Infektion? Dtsch Zahnarztl Z. 1994;49:1031-3.
391.
Owens RC Jr, Donskey CJ, Gaynes RP, Loo VG, Muto CA. Antimicrobial-associated risk factors for Clostridium difficile infection. Clin Infect Dis. 2008 Jan;46 Suppl 1:S19-31. DOI: 10.1086/521859 Externer Link
392.
Pablos AI, Escobar I, Albiñana S, Serrano O, Ferrari JM, Herreros de Tejada A. Evaluation of an antibiotic intravenous to oral sequential therapy program. Pharmacoepidemiol Drug Saf. 2005 Jan;14(1):53-9. DOI: 10.1002/pds.1042 Externer Link
393.
Paffrath U, Schwabe D, editors. Arzneiverordnungsreport (AVR). Berlin: Springer Verlag; 2005.
394.
Pai MP, Bearden DT. Antimicrobial dosing considerations in obese adult patients. Pharmacotherapy. 2007 Aug;27(8):1081-91. DOI: 10.1592/phco.27.8.1081 Externer Link
395.
Papadopoulos S, Ball AM, Liewer SE, Martin CA, Winstead PS, Murphy BS. Rhabdomyolysis during therapy with daptomycin. Clin Infect Dis. 2006 Jun;42(12):e108-10. DOI: 10.1086/504379 Externer Link
396.
Pappas PG, Kauffman CA, Andes D, Benjamin DK Jr, Calandra TF, Edwards JE Jr, Filler SG, Fisher JF, Kullberg BJ, Ostrosky-Zeichner L, Reboli AC, Rex JH, Walsh TJ, Sobel JD; Infectious Diseases Society of America. Clinical practice guidelines for the management of candidiasis: 2009 update by the Infectious Diseases Society of America. Clin Infect Dis. 2009 Mar;48(5):503-35. DOI: 10.1086/596757 Externer Link
397.
Pascual A, Calandra T, Bolay S, Buclin T, Bille J, Marchetti O. Voriconazole therapeutic drug monitoring in patients with invasive mycoses improves efficacy and safety outcomes. Clin Infect Dis. 2008 Jan;46(2):201-11. DOI: 10.1086/524669 Externer Link
398.
Passarell J, Ludwig E, Liolios K, Meagher AK, Grasela TH, Babinchak T, Ellis-Grosse EJ. Exposure-response analyses of tigecycline tolerability in healthy subjects. Diagn Microbiol Infect Dis. 2009 Oct;65(2):123-9. DOI: 10.1016/j.diagmicrobio.2009.06.019 Externer Link
399.
Paterson DL. “Collateral damage” from cephalosporin or quinolone antibiotic therapy. Clin Infect Dis. 2004 May 15;38 Suppl 4:S341-5. DOI: 10.1086/382690 Externer Link
400.
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 Mar;328(7441):668. DOI: 10.1136/bmj.38028.520995.63 Externer Link
401.
Paul M, Silbiger I, Grozinsky S, Soares-Weiser K, Leibovici L. Beta lactam antibiotic monotherapy versus beta lactam-aminoglycoside antibiotic combination therapy for sepsis. Cochrane Database Syst Rev. 2006;(1):CD003344. DOI: 10.1002/14651858.CD003344.pub2 Externer Link
402.
Pavan PR, Oteiza EE, Hughes BA, Avni A. Exogenous endophthalmitis initially treated without systemic antibiotics. Ophthalmology. 1994 Jul;101(7):1289-96; discussion 1296-7. DOI: 10.1016/S0161-6420(94)31177-8 Externer Link
403.
Pea F, Furlanut M, Negri C, Pavan F, Crapis M, Cristini F, Viale P. Prospectively validated dosing nomograms for maximizing the pharmacodynamics of vancomycin administered by continuous infusion in critically ill patients. Antimicrob Agents Chemother. 2009 May;53(5):1863-7. DOI: 10.1128/AAC.01149-08 Externer Link
404.
Pea F, Viale P. Bench-to-bedside review: Appropriate antibiotic therapy in severe sepsis and septic shock--does the dose matter? Crit Care. 2009;13(3):214. DOI: 10.1186/cc7774 Externer Link
405.
Pea F, Viale P, Furlanut M. Antimicrobial therapy in critically ill patients: a review of pathophysiological conditions responsible for altered disposition and pharmacokinetic variability. Clin Pharmacokinet. 2005;44(10):1009-34. DOI: 10.2165/00003088-200544100-00002 Externer Link
406.
Peltola H, Anttila M, Renkonen OV; Finnish Study Group. Randomised comparison of chloramphenicol, ampicillin, cefotaxime, and ceftriaxone for childhood bacterial meningitis. Lancet. 1989;1:1281-7. DOI: 10.1016/S0140-6736(89)92685-8 Externer Link
407.
Pelz RK, Hendrix CW, Swoboda SM, Diener-West M, Merz WG, Hammond J, Lipsett PA. Double-blind placebo-controlled trial of fluconazole to prevent candidal infections in critically ill surgical patients. Ann Surg. 2001 Apr;233(4):542-8. DOI: 10.1097/00000658-200104000-00010 Externer Link
408.
Pereira CA, Petrilli AS, Carlesse FA, Luisi FA, da Silva KV, de Martino Lee ML. Cefepime monotherapy is as effective as ceftriaxone plus amikacin in pediatric patients with cancer and high-risk febrile neutropenia in a randomized comparison. J Microbiol Immunol Infect. 2009 Apr;42(2):141-7.
409.
Peterson J, Kaul S, Khashab M, Fisher AC, Kahn JB. A double-blind, randomized comparison of levofloxacin 750 mg once-daily for five days with ciprofloxacin 400/500 mg twice-daily for 10 days for the treatment of complicated urinary tract infections and acute pyelonephritis. Urology. 2008 Jan;71(1):17-22. DOI: 10.1016/j.urology.2007.09.002 Externer Link
410.
Pfaller MA, Diekema DJ. Epidemiology of invasive candidiasis: a persistent public health problem. Clin Microbiol Rev. 2007 Jan;20(1):133-63. DOI: 10.1128/CMR.00029-06 Externer Link
411.
Piesold J, Al-Nawas B, Otten JE, Grötz KA. Leitlinien der Deutschen Gesellschaft für Mund-Kiefer-Gesichtschirurgie – Odontogene Infektionen und Abszesse S2. AWMF; 2008. Available from: http://www.awmf.org/leitlinien/detail/ll/007-006.html Externer Link
412.
Piesold J, Vent S, Schönfeldt S. Odontogene pyogene Infektionen. 10-Jahres-Analyse [Odontogenic pyogenic infections. 10-year analysis]. Mund Kiefer Gesichtschir. 1999 Mar;3(2):82-91.
413.
Pigrau C, Almirante B, Rodriguez D, Larrosa N, Bescos S, Raspall G, Pahissa A. Osteomyelitis of the jaw: resistance to clindamycin in patients with prior antibiotics exposure. Eur J Clin Microbiol Infect Dis. 2009 Apr;28(4):317-23. DOI: 10.1007/s10096-008-0626-z Externer Link
414.
Pillai SK, Eliopoulos GM, Moellering, RC Jr. Section E: anti-infective therapy: principles of anti-infective therapy. In: Mandell GL, Bennett JE, Dolin R, editors. Mandell, Douglas, and Bennett’s Principles and Practice of Infectious Diseases. 7th ed. Philadelphia: Churchill Livingstone/Elsevier; 2010.
415.
Pino Rivero V, González Palomino A, Pantoja Hernández CG, Trinidad Ramos G, Marcos García M, Rejas Ugena E, Blasco Huelva A. Amigdalitis aguda ulceronecrótica fusoespirilar de Plaut-Vincent asociada a mononucleosis infecciosa [Plaut-Vincent's acute necrotizing ulcerative fusospirillum tonsillitis associated to infectious mononucleosis]. An Otorrinolaringol Ibero Am. 2006;33(3):219-24.
416.
Pirmohamed M, James S, Meakin S, Green C, Scott AK, Walley TJ, Farrar K, Park BK, Breckenridge AM. Adverse drug reactions as cause of admission to hospital: prospective analysis of 18 820 patients. BMJ. 2004 Jul;329(7456):15-9. DOI: 10.1136/bmj.329.7456.15 Externer Link
417.
Pleyer U, Mondino BJ, Adamu SA, Pitchekian-Halabi H, Engstrom RE, Glasgow BJ. Immune response to Staphylococcus epidermidis-induced endophthalmitis in a rabbit model. Invest Ophthalmol Vis Sci. 1992 Aug;33(9):2650-63.
418.
Prange H, Bitsch A, editors. Infektionserkrankungen des Zentralnervensystems. 2nd ed. Stuttgart: Wissenschaftliche Verlagsgesellschaft; 2001.
419.
Prasad K, Singh MB. Corticosteroids for managing tuberculous meningitis. Cochrane Database Syst Rev. 2008;(1):CD002244. DOI: 10.1002/14651858.CD002244.pub3 Externer Link
420.
Puliafito CA, Baker AS, Haaf J, Foster CS. Infectious endophthalmitis. Review of 36 cases. Ophthalmology. 1982 Aug;89(8):921-9. DOI: 10.1016/S0161-6420(82)34704-1 Externer Link
421.
Quagliarello VJ, Scheld WM. Treatment of bacterial meningitis. N Engl J Med. 1997 Mar;336(10):708-16. DOI: 10.1056/NEJM199703063361007 Externer Link
422.
Rams TE, Andriolo M Jr, Feik D, Abel SN, McGivern TM, Slots J. Microbiological study of HIV-related periodontitis. J Periodontol. 1991 Jan;62(1):74-81. DOI: 10.1902/jop.1991.62.1.74 Externer Link
423.
Rea RS, Capitano B, Bies R, Bigos KL, Smith R, Lee H. Suboptimal aminoglycoside dosing in critically ill patients. Ther Drug Monit. 2008 Dec;30(6):674-81. DOI: 10.1097/FTD.0b013e31818b6b2f Externer Link
424.
Reese AM, Frei CR, Burgess DS. Pharmacodynamics of intermittent and continuous infusion piperacillin/tazobactam and cefepime against extended-spectrum beta-lactamase-producing organisms. Int J Antimicrob Agents. 2005 Aug;26(2):114-9. DOI: 10.1016/j.ijantimicag.2005.06.004 Externer Link
425.
Reiber H, Lange P. Quantification of virus-specific antibodies in cerebrospinal fluid and serum: sensitive and specific detection of antibody synthesis in brain. Clin Chem. 1991 Jul;37(7):1153-60.
426.
Reid GE, Grim SA, Aldeza CA, Janda WM, Clark NM. Rapid development of Acinetobacter baumannii resistance to tigecycline. Pharmacotherapy. 2007 Aug;27(8):1198-201. DOI: 10.1592/phco.27.8.1198 Externer Link
427.
Reinhart K, Brunkhorst F, Bone H, Gerlach H, Gründling M, Kreymann G, Kujath P, Marggraf G, Mayer K, Meier-Hellmann A, Peckelsen C, Putensen C, Quintel M, Ragaller M, Rossaint R, Stüber F, Weiler N, Welte T, Werdan K; Deutsche Sepsis-Gesellschaft eV. Diagnose und Therapie der Sepsis: S2-Leitlinien der Deutschen Sepsis-Gesellschaft e.V. (DSG) und der Deutschen Interdisziplinären Vereinigung für Intensiv- und Notfallmedizin (DIVI) [Diagnosis and therapy of sepsis: guidelines of the German Sepsis Society Inc. and the German Interdisciplinary Society for Intensive and Emergency Medicine]. Anaesthesist. 2006 Jun;55 Suppl 1:43-56. DOI: 10.1007/s00101-006-1020-9 Externer Link
428.
Rello J, Vidaur L, Sandiumenge A, Rodríguez A, Gualis B, Boque C, Diaz E. De-escalation therapy in ventilator-associated pneumonia. Crit Care Med. 2004 Nov;32(11):2183-90.
429.
Reynolds DS, Flynn HW Jr. Endophthalmitis after penetrating ocular trauma. Curr Opin Ophthalmol. 1997 Jun;8(3):32-8. DOI: 10.1097/00055735-199706000-00006 Externer Link
430.
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 Mar;161(5):722-7. DOI: 10.1001/archinte.161.5.722 Externer Link
431.
Richard GA, Klimberg IN, Fowler CL, Callery-D'Amico S, Kim SS. Levofloxacin versus ciprofloxacin versus lomefloxacin in acute pyelonephritis. Urology. 1998 Jul;52(1):51-5. DOI: 10.1016/S0090-4295(98)00160-5 Externer Link
432.
Rimola A, García-Tsao G, Navasa M, Piddock LJ, Planas R, Bernard B, Inadomi JM. Diagnosis, treatment and prophylaxis of spontaneous bacterial peritonitis: a consensus document. International Ascites Club. J Hepatol. 2000 Jan;32(1):142-53. DOI: 10.1016/S0168-8278(00)80201-9 Externer Link
433.
Rivers E, Nguyen B, Havstad S, Ressler J, Muzzin A, Knoblich B, Peterson E, Tomlanovich M; Early Goal-Directed Therapy Collaborative Group. Early goal-directed therapy in the treatment of severe sepsis and septic shock. N Engl J Med. 2001 Nov;345(19):1368-77. DOI: 10.1056/NEJMoa010307 Externer Link
434.
Roberts JA, Lipman J. Antibacterial dosing in intensive care: pharmacokinetics, degree of disease and pharmacodynamics of sepsis. Clin Pharmacokinet. 2006;45(8):755-73. DOI: 10.2165/00003088-200645080-00001 Externer Link
435.
Roberts JA, Lipman J. Optimizing use of beta-lactam antibiotics in the critically ill. Semin Respir Crit Care Med. 2007 Dec;28(6):579-85. DOI: 10.1055/s-2007-996404 Externer Link
436.
Roberts JA, Lipman J. Pharmacokinetic issues for antibiotics in the critically ill patient. Crit Care Med. 2009 Mar;37(3):840-51. DOI: 10.1097/CCM.0b013e3181961bff Externer Link
437.
Roberts JA, Paratz J, Paratz E, Krueger WA, Lipman J. Continuous infusion of beta-lactam antibiotics in severe infections: a review of its role. Int J Antimicrob Agents. 2007 Jul;30(1):11-8. DOI: 10.1016/j.ijantimicag.2007.02.002 Externer Link
438.
Roberts JA, Roberts MS, Robertson TA, Dalley AJ, Lipman J. Piperacillin penetration into tissue of critically ill patients with sepsis--bolus versus continuous administration? Crit Care Med. 2009 Mar;37(3):926-33. DOI: 10.1097/CCM.0b013e3181968e44 Externer Link
439.
Rochon PA, Lane CJ, Bronskill SE, Sykora K, Anderson GM, Mamdani MM, Gurwitz JH, Dhalla IA. Potentially inappropriate prescribing in Canada relative to the US. Drugs Aging. 2004;21(14):939-47. DOI: 10.2165/00002512-200421140-00004 Externer Link
440.
Rodloff AC, Christiansen B, Deutrich C, Engelmann L, Schreiter D, Gaus W. Entwicklung der Antibiotika-Resistenz bei bakteriellen Krankheitserregern von Patienten eines Universitätsklinikums. Chemother J. 2002;11(3):105-12.
441.
Roehrborn A, Thomas L, Potreck O, Ebener C, Ohmann C, Goretzki PE, Röher HD. The microbiology of postoperative peritonitis. Clin Infect Dis. 2001 Nov;33(9):1513-9. DOI: 10.1086/323333 Externer Link
442.
Rotstein C, Evans G, Born A, Grossman R, Light RB, Magder S, McTaggart B, Weiss K, Zhanel GG. Clinical practice guidelines for hospital-acquired pneumonia and ventilator-associated pneumonia in adults. Can J Infect Dis Med Microbiol. 2008 Jan;19(1):19-53.
443.
Rupprecht TA, Pfister HW. Clinical experience with linezolid for the treatment of central nervous system infections. Eur J Neurol. 2005 Jul;12(7):536-42. DOI: 10.1111/j.1468-1331.2005.01001.x Externer Link
444.
Rusan M, Klug TE, Ovesen T. An overview of the microbiology of acute ear, nose and throat infections requiring hospitalisation. Eur J Clin Microbiol Infect Dis. 2009 Mar;28(3):243-51. DOI: 10.1007/s10096-008-0619-y Externer Link
445.
Rybak MJ, Abate BJ, Kang SL, Ruffing MJ, Lerner SA, Drusano GL. Prospective evaluation of the effect of an aminoglycoside dosing regimen on rates of observed nephrotoxicity and ototoxicity. Antimicrob Agents Chemother. 1999 Jul;43(7):1549-55.
446.
Rybak MJ, Lomaestro BM, Rotschafer JC, Moellering RC Jr, Craig WA, Billeter M, Dalovisio JR, Levine DP. Therapeutic monitoring of vancomycin in adults summary of consensus recommendations from the American Society of Health-System Pharmacists, the Infectious Diseases Society of America, and the Society of Infectious Diseases Pharmacists. Pharmacotherapy. 2009 Nov;29(11):1275-9. DOI: 10.1592/phco.29.11.1275 Externer Link
447.
Sader HS, Fey PD, Limaye AP, Madinger N, Fish DN, Pankey G, Rahal J, Rybak MJ, Snydman DR, Steed LL, Waites K, Jones RN. Evaluation of vancomycin and daptomycin potency trends (MIC creep) against methicillin-resistant Staphylococcus aureus isolates collected in nine U.S. medical centers from 2002 to 2006. Antimicrob Agents Chemother. 2009 Oct;53(10):4127-32. DOI: 10.1128/AAC.00616-09 Externer Link
448.
Safdar N, Handelsman J, Maki DG. Does combination antimicrobial therapy reduce mortality in Gram-negative bacteraemia? A meta-analysis. Lancet Infect Dis. 2004 Aug;4(8):519-27. DOI: 10.1016/S1473-3099(04)01108-9 Externer Link
449.
Sakamoto H, Aoki T, Kise Y, Watanabe D, Sasaki J. Descending necrotizing mediastinitis due to odontogenic infections. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2000 Apr;89(4):412-9. DOI: 10.1016/S1079-2104(00)70121-1 Externer Link
450.
Sakoulas G, Moise-Broder PA, Schentag J, Forrest A, Moellering RC Jr, Eliopoulos GM. Relationship of MIC and bactericidal activity to efficacy of vancomycin for treatment of methicillin-resistant Staphylococcus aureus bacteremia. J Clin Microbiol. 2004 Jun;42(6):2398-402. DOI: 10.1128/JCM.42.6.2398-2402.2004 Externer Link
451.
Salama C, Finch D, Bottone EJ. Fusospirochetosis causing necrotic oral ulcers in patients with HIV infection. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2004 Sep;98(3):321-3. DOI: 10.1016/j.tripleo.2004.03.002 Externer Link
452.
Salvatore M, Meyers BR. Metronidazole. In: Mandell GL, Bennett JE, Dolin R, editors. Mandell, Douglas, and Bennett’s Principles and Practice of Infectious Diseases. 7th ed. Churchill Livingstone: Elsevier; 2010. p. 419-26.
453.
Salvatore M, Meyers BR. Tetracyclines and chloramphenicol. In: Mandell GL, Bennett JE, Dolin R, editors. Mandell, Douglas, and Bennett’s Principles and Practice of Infectious Diseases. 7th ed. Churchill Livingstone: Elsevier; 2010. p. 385-401.
454.
Scaglione F, Paraboni L. Pharmacokinetics/pharmacodynamics of antibacterials in the Intensive Care Unit: setting appropriate dosing regimens. Int J Antimicrob Agents. 2008 Oct;32(4):294-301. DOI: 10.1016/j.ijantimicag.2008.03.015 Externer Link
455.
Scaglione F, Raichi M, Fraschini F. Serum protein binding and extravascular diffusion of methoxyimino cephalosporins. Time courses of free and total concentrations of cefotaxime and ceftriaxone in serum and pleural exudate. J Antimicrob Chemother. 1990 Sep;26 Suppl A:1-10.
456.
Scarborough M, Gordon SB, Whitty CJ, French N, Njalale Y, Chitani A, Peto TE, Lalloo DG, Zijlstra EE. Corticosteroids for bacterial meningitis in adults in sub-Saharan Africa. N Engl J Med. 2007 Dec;357(24):2441-50. DOI: 10.1056/NEJMoa065711 Externer Link
457.
Schaal KP, Herzog M, Pape HD, Pulverer G, Herzog S. Kölner Therapiekonzept zur Behandlung der menschlichen Aktinomykose von 1952–1982. In: Schuchard K, Pfeifer G, Schwenzer N, editors. Fortschritte der Kiefer- und Gesichtschirurgie. Stuttgart: Thieme; 1984. p. 151-6.
458.
Schaeffer AJ, Anderson RU, Krieger JN, Lobel B, et al. Consensus statement on prostatitis. The assessment and management of male pelvic pain syndrome, including prostatitis. In: 6th International Conference on New Developments in Prostate Cancer and Prostate Diseases. M. L. U. T. D. Edition. Paris: Health Publications; 2006. p. 343-75.
459.
Scheffer D, Hofmann S, Pietsch M, Wenisch C. Infektionen in der Orthopädie und Traumatologie. Pathogenese und Therapie [Infections in orthopedics and traumatology. Pathogenesis and therapy]. Orthopade. 2008 Jul;37(7):709-18; quiz 719. DOI: 10.1007/s00132-008-1301-x Externer Link
460.
Scheld WM, Dacey RG, Winn HR, Welsh JE, Jane JA, Sande MA. Cerebrospinal fluid outflow resistance in rabbits with experimental meningitis. Alterations with penicillin and methylprednisolone. J Clin Invest. 1980 Aug;66(2):243-53. DOI: 10.1172/JCI109850 Externer Link
461.
Schimmer C, Sommer SP, Bensch M, Elert O, Leyh R. Management of poststernotomy mediastinitis: experience and results of different therapy modalities. Thorac Cardiovasc Surg. 2008 Jun;56(4):200-4. DOI: 10.1055/s-2008-1038386 Externer Link
462.
Schlichtenbrede FC. Management of low grade endophthalmitis by capsular bag irrigation. 22. Kongress der Deutschsprachigen Gesellschaft für Intraokularlinsen-Implantation, interventionelle und refraktive Chirurgie 15.02. und 16.02.08 in Heidelberg. Klin Monbl Augenheilkd. 2008;225(Suppl 1):19. DOI: 10.1055/s-2008-1058022 Externer Link
463.
Schmechel H, Fünfstück R, Folger U, Robiller F, Schmechel R. Kritische Betrachtung der rechnerischen Bestimmung der glomerulären Filtrationsrate nach Cockroft und Gault – Versuch einer Korrektur. Nieren- und Hochdruckkrankheiten. 2005;34(10):433-44. DOI: 10.5414/NHP34433 Externer Link
464.
Schmidseder E, Miño de Kaspar H, Klauss V, Kampik A. Posttraumatische Endophthalmitis nach penetrierenden Augenverletzungen. Risikofaktoren, mikrobiologische Diagnostik und funktionelle Ergebnisse [Post-traumatic endophthalmitis after penetrating eye injuries. Risk factors, microbiological diagnosis and functional outcome]. Ophthalmologe. 1998 Mar;95(3):153-7. DOI: 10.1007/s003470050254 Externer Link
465.
Schöfer H, Brockmeyer N, Dissemond J, Effendy I, Esser S, Geiss HK, Harder S, Hartmann M, Jappe U, Plettenberg A, Reimann H, Shah P, Tschachler E, Wichelhaus TA; Deutschen Dermatologischen Gesellschaft (DDG); Arbeitsgemeinschaft für Dermatologische Infektiologie (ADI). Staphylokokken-Infektionen der Haut und Schleimhäute. Leitlinie der Deutschen Dermatologischen Gesellschaft (DDG), Arbeitsgemeinschaft für Dermatologische Infektiologie (ADI) [Staphylococcal infections of the skin and mucous membranes. Guideline of the German Dermatologic Society, Study Group of Dermatologic Infectiology]. J Dtsch Dermatol Ges. 2005 Sep;3(9):726-34.
466.
Scholz H, Hofmann T, Noack R, Edwards DJ, Stoeckel K. Prospective comparison of ceftriaxone and cefotaxime for the short-term treatment of bacterial meningitis in children. Chemotherapy. 1998 Mar-Apr;44(2):142-7. DOI: 10.1159/000007106 Externer Link
467.
Schuchat A, Robinson K, Wenger JD, Harrison LH, Farley M, Reingold AL, Lefkowitz L, Perkins BA. Bacterial meningitis in the United States in 1995. Active Surveillance Team. N Engl J Med. 1997 Oct;337(14):970-6. DOI: 10.1056/NEJM199710023371404 Externer Link
468.
Schultz BR, Allen CH, Kaplan SL. Corticosteroids for acute bacterial meningitis. Ann Emerg Med. 2009 Jul;54(1):136-7; author reply 137-8. DOI: 10.1016/j.annemergmed.2008.11.030 Externer Link
469.
Schuster HP, Werdan K, editors. Intensivtherapie bei Sepsis und Multiorganversagen. Berlin, Heidelberg: Springer; 2000. DOI: 10.1007/978-3-662-07962-1 Externer Link
470.
Seaton RA. Daptomycin: rationale and role in the management of skin and soft tissue infections. J Antimicrob Chemother. 2008 Nov;62 Suppl 3:iii15-23. DOI: 10.1093/jac/dkn368 Externer Link
471.
Seeger MA, Schiefner A, Eicher T, Verrey F, Diederichs K, Pos KM. Structural asymmetry of AcrB trimer suggests a peristaltic pump mechanism. Science. 2006 Sep;313(5791):1295-8. DOI: 10.1126/science.1131542 Externer Link
472.
Sendzik J, Lode H, Stahlmann R. Quinolone-induced arthropathy: an update focusing on new mechanistic and clinical data. Int J Antimicrob Agents. 2009 Mar;33(3):194-200. DOI: 10.1016/j.ijantimicag.2008.08.004 Externer Link
473.
Sermet-Gaudelus I, Hulin A, Ferroni A, Silly C, Gaillard JL, Berche P, Lenoir G. L'antibiothérapie dans la mucoviscidose. I. Particularités pharmacologiques des antibiotiques [Antibiotic therapy in cystic fibrosis. I. Pharmacologic specifics of antibiotics]. Arch Pediatr. 2000 May;7(5):519-28. DOI: 10.1016/S0929-693X(00)89009-0 Externer Link
474.
Servais H, Tulkens PM. Stability and compatibility of ceftazidime administered by continuous infusion to intensive care patients. Antimicrob Agents Chemother. 2001 Sep;45(9):2643-7. DOI: 10.1128/AAC.45.9.2643-2647.2001 Externer Link
475.
Shakeri-Nejad K, Stahlmann R. Drug interactions during therapy with three major groups of antimicrobial agents. Expert Opin Pharmacother. 2006 Apr;7(6):639-51. DOI: 10.1517/14656566.7.6.639 Externer Link
476.
Sharma S, Kumar A. Antimicrobial management of sepsis and septic shock. Clin Chest Med. 2008 Dec;29(4):677-87, ix. DOI: 10.1016/j.ccm.2008.06.004 Externer Link
477.
Shpitzer T, Stern Y, Cohen O, Levy R, Segal K, Feinmesser R. Malignant external otitis in nondiabetic patients. Ann Otol Rhinol Laryngol. 1993 Nov;102(11):870-2.
478.
Simon C, Stille W. Antibiotika Therapie in Klinik und Praxis. 10th ed. Stuttgart, New York: Schattauer; 2000.
479.
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 Aug;162(2 Pt 1):505-11. DOI: 10.1164/ajrccm.162.2.9909095 Externer Link
480.
Singhal S, Purnapatre KP, Kalia V, Dube S, Nair D, Deb M, Aggarwal P, Gupta S, Upadhyay DJ, Rattan A, Raj VS. Ciprofloxacin-resistant Neisseria meningitidis, Delhi, India. Emerging Infect Dis. 2007 Oct;13(10):1614-6. DOI: 10.3201/eid1310.060820 Externer Link
481.
Sivapalasingam S, Steigbigl NH. Macrolides, clindamycin, and ketolides. In: Mandell GL, Bennett JE, Dolin R, editors. Mandell, Douglas, and Bennett’s Principles and Practice of Infectious Diseases. 7th ed. Churchill Livingstone: Elsevier; 2010. p. 427-48.
482.
Skoczynska A, Alonso JM, Taha MK. Ciprofloxacin resistance in Neisseria meningitidis, France. Emerging Infect Dis. 2008 Aug;14(8):1322-3. DOI: 10.3201/eid1408.080040 Externer Link
483.
Smego RA Jr, Foglia G. Actinomycosis. Clin Infect Dis. 1998 Jun;26(6):1255-61; quiz 1262-3. DOI: 10.1086/516337 Externer Link
484.
Sobottka I, Cachovan G, Stürenburg E, Ahlers MO, Laufs R, Platzer U, Mack D. In vitro activity of moxifloxacin against bacteria isolated from odontogenic abscesses. Antimicrob Agents Chemother. 2002 Dec;46(12):4019-21. DOI: 10.1128/AAC.46.12.4019-4021.2002 Externer Link
485.
Soldati D, Mudry A, Monnier P. Necrotizing otitis externa caused by Staphylococcus epidermidis. Eur Arch Otorhinolaryngol. 1999;256(9):439-41. DOI: 10.1007/s004050050184 Externer Link
486.
Solomkin JS, Mazuski JE, Bradley JS, Rodvold KA, Goldstein EJ, Baron EJ, O'Neill PJ, Chow AW, Dellinger EP, Eachempati SR, Gorbach S, Hilfiker M, May AK, Nathens AB, Sawyer RG, Bartlett JG. Diagnosis and management of complicated intra-abdominal infection in adults and children: guidelines by the Surgical Infection Society and the Infectious Diseases Society of America. Clin Infect Dis. 2010 Jan;50(2):133-64. DOI: 10.1086/649554 Externer Link
487.
Soriano A, Marco F, Martínez JA, Pisos E, Almela M, Dimova VP, Alamo D, Ortega M, Lopez J, Mensa J. Influence of vancomycin minimum inhibitory concentration on the treatment of methicillin-resistant Staphylococcus aureus bacteremia. Clin Infect Dis. 2008 Jan;46(2):193-200. DOI: 10.1086/524667 Externer Link
488.
Soriano A, Miró O, Mensa J. Mitochondrial toxicity associated with linezolid. N Engl J Med. 2005 Nov;353(21):2305-6. DOI: 10.1056/NEJM200511243532123 Externer Link
489.
Sort P, Navasa M, Arroyo V, Aldeguer X, Planas R, Ruiz-del-Arbol L, Castells L, Vargas V, Soriano G, Guevara M, Ginès P, Rodés J. Effect of intravenous albumin on renal impairment and mortality in patients with cirrhosis and spontaneous bacterial peritonitis. N Engl J Med. 1999 Aug;341(6):403-9. DOI: 10.1056/NEJM199908053410603 Externer Link
490.
Sotto A, Lefrant JY, Fabbro-Peray P, Muller L, Tafuri J, Navarro F, Prudhomme M, De La Coussaye JE. Evaluation of antimicrobial therapy management of 120 consecutive patients with secondary peritonitis. J Antimicrob Chemother. 2002 Oct;50(4):569-76. DOI: 10.1093/jac/dkf167 Externer Link
491.
Sousa M, Pozniak A, Boffito M. Pharmacokinetics and pharmacodynamics of drug interactions involving rifampicin, rifabutin and antimalarial drugs. J Antimicrob Chemother. 2008 Nov;62(5):872-8. DOI: 10.1093/jac/dkn330 Externer Link
492.
Soy D, López E, Ribas J. Teicoplanin population pharmacokinetic analysis in hospitalized patients. Ther Drug Monit. 2006 Dec;28(6):737-43. DOI: 10.1097/01.ftd.0000249942.14145.ff Externer Link
493.
Sprung CL, Annane D, Keh D, Moreno R, Singer M, Freivogel K, Weiss YG, Benbenishty J, Kalenka A, Forst H, Laterre PF, Reinhart K, Cuthbertson BH, Payen D, Briegel J; CORTICUS Study Group. Hydrocortisone therapy for patients with septic shock. N Engl J Med. 2008 Jan;358(2):111-24. DOI: 10.1056/NEJMoa071366 Externer Link
494.
Stahlmann R, Lode H. Safety Overview. Toxicity, adverse effects, and drug interactions. In: Andriole VT, editor. The Quinolones. 3rd ed. London: Academic Press; 2000. p. 397-453. DOI: 10.1016/B978-012059517-4/50015-5 Externer Link
495.
Stein G, Eichhorn T, Fünfstück R. Harnwegsinfektionen bei Patienten mit eingeschränkter Nierenfunktion. Nieren- und Hochdruckkrankheiten. 2007;36(7):288-291. DOI: 10.5414/NHP36288 Externer Link
496.
Steinkraus G, White R, Friedrich L. Vancomycin MIC creep in non-vancomycin-intermediate Staphylococcus aureus (VISA), vancomycin-susceptible clinical methicillin-resistant S. aureus (MRSA) blood isolates from 2001-05. J Antimicrob Chemother. 2007 Oct;60(4):788-94. DOI: 10.1093/jac/dkm258 Externer Link
497.
Sternbach H. The serotonin syndrome. Am J Psychiatry. 1991 Jun;148(6):705-13.
498.
Sternberg P Jr, Martin DF. Management of endophthalmitis in the post-endophthalmitis vitrectomy study era. Arch Ophthalmol. 2001 May;119(5):754-5. DOI: 10.1001/archopht.119.5.754 Externer Link
499.
Stevens DL, Bisno AL, Chambers HF, Everett ED, Dellinger P, Goldstein EJ, Gorbach SL, Hirschmann JV, Kaplan EL, Montoya JG, Wade JC; Infectious Diseases Society of America. Practice guidelines for the diagnosis and management of skin and soft-tissue infections. Clin Infect Dis. 2005 Nov;41(10):1373-406. DOI: 10.1086/497143 Externer Link
500.
Stille W, Brodt HR, Groll A, Just-Nübling G, editors. Antibiotikatherapie. 11th ed. Stuttgart: Schattauer; 2005.
501.
Stryjewski ME, Szczech LA, Benjamin DK Jr, Inrig JK, Kanafani ZA, Engemann JJ, Chu VH, Joyce MJ, Reller LB, Corey GR, Fowler VG Jr. Use of vancomycin or first-generation cephalosporins for the treatment of hemodialysis-dependent patients with methicillin-susceptible Staphylococcus aureus bacteremia. Clin Infect Dis. 2007 Jan;44(2):190-6. DOI: 10.1086/510386 Externer Link
502.
Stuart S, Booth TC, Cash CJ, Hameeduddin A, Goode JA, Harvey C, Malhotra A. Complications of continuous ambulatory peritoneal dialysis. Radiographics. 2009 Mar-Apr;29(2):441-60. DOI: 10.1148/rg.292085136 Externer Link
503.
Swan SK, Bennett WM. Drug dosing guidelines in patients with renal failure. West J Med. 1992 Jun;156(6):633-8.
504.
Swartz MN. Clinical practice. Cellulitis. N Engl J Med. 2004 Feb;350(9):904-12. DOI: 10.1056/NEJMcp031807 Externer Link
505.
Sweet RL. Pelvic Inflammatory Disease: Treatment. In: Mead PB, Hager WD, Faro S, editors. Protocols for infectious diseases in obstetrics and gynecology. 2nd ed. Malden: Blackwell Science Inc; 2000. p. 400-5.
506.
Swoboda S, Fritz S, Martignoni ME, Feldhues RA, Hoppe-Tichy T, Buchler MW, Geiss HK. Varying linezolid susceptibility of vancomycin-resistant Enterococcus faecium isolates during therapy: a case report. J Antimicrob Chemother. 2005 Oct;56(4):787-9. DOI: 10.1093/jac/dki318 Externer Link
507.
Swoboda S, Hoppe-Tichy T, Geiss HK, Hainer C, Nguyen TH, Knaebel HP, Weigand MA. Septischer Schock durch Vancomycin-resistente Enterokokken. Tigecyclin-Monotherapie [Septic shock due to vancomycin-resistant enterococci infection. Tigecycline monotherapy]. Anaesthesist. 2007 Feb;56(2):169-74. DOI: 10.1007/s00101-006-1088-2 Externer Link
508.
Swoboda S, Ober M, Hainer C, Lichtenstern C, Seiler C, Wendt C, Hoppe-Tichy T, Büchler M, Weigand MA. Tigecycline for the treatment of patients with severe sepsis or septic shock: a drug use evaluation in a surgical intensive care unit. J Antimicrob Chemother. 2008 Mar;61(3):729-33. DOI: 10.1093/jac/dkm541 Externer Link
509.
Tacconelli E, De Angelis G, Cataldo MA, Mantengoli E, Spanu T, Pan A, Corti G, Radice A, Stolzuoli L, Antinori S, Paradisi F, Carosi G, Bernabei R, Antonelli M, Fadda G, Rossolini GM, Cauda R. Antibiotic usage and risk of colonization and infection with antibiotic-resistant bacteria: a hospital population-based study. Antimicrob Agents Chemother. 2009 Oct;53(10):4264-9. DOI: 10.1128/AAC.00431-09 Externer Link
510.
Talan DA, Citron DM, Abrahamian FM, Moran GJ, Goldstein EJ. Bacteriologic analysis of infected dog and cat bites. Emergency Medicine Animal Bite Infection Study Group. N Engl J Med. 1999 Jan;340(2):85-92. DOI: 10.1056/NEJM199901143400202 Externer Link
511.
Talan DA, Stamm WE, Hooton TM, Moran GJ, Burke T, Iravani A, Reuning-Scherer J, Church DA. Comparison of ciprofloxacin (7 days) and trimethoprim-sulfamethoxazole (14 days) for acute uncomplicated pyelonephritis pyelonephritis in women: a randomized trial. JAMA. 2000 Mar 22-29;283(12):1583-90. DOI: 10.1001/jama.283.12.1583 Externer Link
512.
Tam VH, Louie A, Lomaestro BM, Drusano GL. Integration of population pharmacokinetics, a pharmacodynamic target, and microbiologic surveillance data to generate a rational empiric dosing strategy for cefepime against Pseudomonas aeruginosa. Pharmacotherapy. 2003 Mar;23(3):291-5. DOI: 10.1592/phco.23.3.291.32110 Externer Link
513.
Taylor JJ, Wilson JW, Estes LL. Linezolid and serotonergic drug interactions: a retrospective survey. Clin Infect Dis. 2006 Jul;43(2):180-7. DOI: 10.1086/504809 Externer Link
514.
Tenke P, Kovacs B, Bjerklund Johansen TE, Matsumoto T, Tambyah PA, Naber KG. European and Asian guidelines on management and prevention of catheter-associated urinary tract infections. Int J Antimicrob Agents. 2008 Feb;31 Suppl 1:S68-78. DOI: 10.1016/j.ijantimicag.2007.07.033 Externer Link
515.
Tessier PR, Nicolau DP, Onyeji CO, Nightingale CH. Pharmacodynamics of intermittent- and continuous-infusion cefepime alone and in combination with once-daily tobramycin against Pseudomonas aeruginosa in an in vitro infection model. Chemotherapy. 1999 Jul-Aug;45(4):284-95. DOI: 10.1159/000007198 Externer Link
516.
Thalhammer F, Traunmüller F, El Menyawi I, Frass M, Hollenstein UM, Locker GJ, Stoiser B, Staudinger T, Thalhammer-Scherrer R, Burgmann H. Continuous infusion versus intermittent administration of meropenem in critically ill patients. J Antimicrob Chemother. 1999 Apr;43(4):523-7. DOI: 10.1093/jac/43.4.523 Externer Link
517.
Thompson JT, Parver LM, Enger CL, Mieler WF, Liggett PE. Infectious endophthalmitis after penetrating injuries with retained intraocular foreign bodies. National Eye Trauma System. Ophthalmology. 1993 Oct;100(10):1468-74. DOI: 10.1016/S0161-6420(93)31454-5 Externer Link
518.
Thürmann PA, Werner U, Hanke F, et al. Arzneimittelrisiken bei hochbetagten Patienten: Ergebnisse deutscher Studien. In: Bundesärztekammer, editor. Fortschritt und Fortbildung in der Medizin. Referate und Diskussionen des 31. interdisziplinären Forums der Bundesärztekammer. Köln: Deutscher Ärzte-Verlag; 2007. p. 216-24. (Fortschritt und Fortbildung in der Medizin; 31)
519.
Thwaites GE, Nguyen DB, Nguyen HD, Hoang TQ, Do TT, Nguyen TC, Nguyen QH, Nguyen TT, Nguyen NH, Nguyen TN, Nguyen NL, Nguyen HD, Vu NT, Cao HH, Tran TH, Pham PM, Nguyen TD, Stepniewska K, White NJ, Tran TH, Farrar JJ. Dexamethasone for the treatment of tuberculous meningitis in adolescents and adults. N Engl J Med. 2004 Oct;351(17):1741-51. DOI: 10.1056/NEJMoa040573 Externer Link
520.
Tice AD, Turpin RS, Hoey CT, Lipsky BA, Wu J, Abramson MA. Comparative costs of ertapenem and piperacillin-tazobactam in the treatment of diabetic foot infections. Am J Health Syst Pharm. 2007 May;64(10):1080-6.
521.
Torgersen C, Moser P, Luckner G, Mayr V, Jochberger S, Hasibeder WR, Dünser MW. Macroscopic postmortem findings in 235 surgical intensive care patients with sepsis. Anesth Analg. 2009 Jun;108(6):1841-7. DOI: 10.1213/ane.0b013e318195e11d Externer Link
522.
Torres A. The new American Thoracic Society/Infectious Disease Society of North America guidelines for the management of hospital-acquired, ventilator-associated and healthcare-associated pneumonia: a current view and new complementary information. Curr Opin Crit Care. 2006 Oct;12(5):444-5. DOI: 10.1097/01.ccx.0000244124.46871.0d Externer Link
523.
Torres A, Ewig S, Lode H, Carlet J; European HAP working group. Defining, treating and preventing hospital acquired pneumonia: European perspective. Intensive Care Med. 2009 Jan;35(1):9-29. DOI: 10.1007/s00134-008-1336-9 Externer Link
524.
Touw DJ, Neef C, Thomson AH, Vinks AA. Cost-effectivensess of therapeutic drug monitoring: an update. Eur J Hosp Pharm Sci. 2007;13(4):83-91.
525.
Townsend SR, Schorr C, Levy MM, Dellinger RP. Reducing mortality in severe sepsis: the Surviving Sepsis Campaign. Clin Chest Med. 2008 Dec;29(4):721-33, x. DOI: 10.1016/j.ccm.2008.06.011 Externer Link
526.
Trautmann M, Lepper PM, Bodmann KF. Resistenzentwicklung von Pseudomonas aeruginosa unter antibiotischer Therapie. Arzneimitteltherapie. 2006;24:50-6
527.
Trittler R, Ehrlich M, Egle H, Kümmerer K. Antiinfektives Drug-Monitoring – nicht nur Nebenwirkungen vermindern! Krankenhauspharmazie. 2004;25:53-8.
528.
Turner PJ. MYSTIC Europe 2007: activity of meropenem and other broad-spectrum agents against nosocomial isolates. Diagn Microbiol Infect Dis. 2009 Feb;63(2):217-22. DOI: 10.1016/j.diagmicrobio.2008.11.004 Externer Link
529.
Tuzuner-Oncul AM, Ungor C, Dede U, Kisnisci RS. Methicillin-resistant Staphylococcus aureus (MRSA) osteomyelitis of the mandible. Oral Surg Oral Med Oral Pathol Oral Radiol Endod. 2009 Jun;107(6):e1-4. DOI: 10.1016/j.tripleo.2009.03.002 Externer Link
530.
Valencia M, Torres A. Ventilator-associated pneumonia. Curr Opin Crit Care. 2009 Feb;15(1):30-5.
531.
Van Bambeke F, Tulkens PM. Safety profile of the respiratory fluoroquinolone moxifloxacin: comparison with other fluoroquinolones and other antibacterial classes. Drug Saf. 2009;32(5):359-78. DOI: 10.2165/00002018-200932050-00001 Externer Link
532.
Van de Beek D, de Gans J, McIntyre P, Prasad K. Corticosteroids for acute bacterial meningitis. Cochrane Database Syst Rev. 2007;(1):CD004405. DOI: 10.1002/14651858.CD004405.pub2 Externer Link
533.
Van de Beek D, Farrar JJ, de Gans J, Mai NT, Molyneux EM, Peltola H, Peto TE, Roine I, Scarborough M, Schultsz C, Thwaites GE, Tuan PQ, Zwinderman AH. Adjunctive dexamethasone in bacterial meningitis: a meta-analysis of individual patient data. Lancet Neurol. 2010 Mar;9(3):254-63. DOI: 10.1016/S1474-4422(10)70023-5 Externer Link
534.
Van Lent-Evers NA, Mathôt 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 Feb;21(1):63-73. DOI: 10.1097/00007691-199902000-00010 Externer Link
535.
Van Ruler O, Mahler CW, Boer KR, Reuland EA, Gooszen HG, Opmeer BC, de Graaf PW, Lamme B, Gerhards MF, Steller EP, van Till JW, de Borgie CJ, Gouma DJ, Reitsma JB, Boermeester MA; Dutch Peritonitis Study Group. Comparison of on-demand vs planned relaparotomy strategy in patients with severe peritonitis: a randomized trial. JAMA. 2007 Aug;298(8):865-72. DOI: 10.1001/jama.298.8.865 Externer Link
536.
Van Zanten AR, Engelfriet PM, van Dillen K, van Veen M, Nuijten MJ, Polderman KH. Importance of nondrug costs of intravenous antibiotic therapy. Crit Care. 2003 Dec;7(6):R184-90. DOI: 10.1186/cc2388 Externer Link
537.
Veehof LJ, Stewart RE, Meyboom-de Jong B, Haaijer-Ruskamp FM. Adverse drug reactions and polypharmacy in the elderly in general practice. Eur J Clin Pharmacol. 1999 Sep;55(7):533-6. DOI: 10.1007/s002280050669 Externer Link
538.
Vick-Fragoso R, Hernández-Oliva G, Cruz-Alcázar J, Amábile-Cuevas CF, Arvis P, Reimnitz P, Bogner JR; STIC Study Group. Efficacy and safety of sequential intravenous/oral moxifloxacin vs intravenous/oral amoxicillin/clavulanate for complicated skin and skin structure infections. Infection. 2009 Oct;37(5):407-17. DOI: 10.1007/s15010-009-8468-x Externer Link
539.
Viestenz A, Schrader W, Behrens-Baumann W. Traumatic Endophthalmitis Prevention Trial (TEPT) [Traumatic Endophthalmitis Prevention Trial (TEPT)]. Klin Monbl Augenheilkd. 2008 Nov;225(11):941-6. DOI: 10.1055/s-2008-1027840 Externer Link
540.
Viladrich PF, Gudiol F, Liñares J, Pallarés R, Sabaté I, Rufí G, Ariza J. Evaluation of vancomycin for therapy of adult pneumococcal meningitis. Antimicrob Agents Chemother. 1991 Dec;35(12):2467-72. DOI: 10.1128/AAC.35.12.2467 Externer Link
541.
Villatoro E, Bassi C, Larvin M. Antibiotic therapy for prophylaxis against infection of pancreatic necrosis in acute pancreatitis. Cochrane Database Syst Rev. 2006;(4):CD002941.
542.
Vincent JL. Ventilator-associated pneumonia. J Hosp Infect. 2004 Aug;57(4):272-80. DOI: 10.1016/j.jhin.2003.06.001 Externer Link
543.
Visapää JP, Tillonen JS, Kaihovaara PS, Salaspuro MP. Lack of disulfiram-like reaction with metronidazole and ethanol. Ann Pharmacother. 2002 Jun;36(6):971-4. DOI: 10.1345/aph.1A066 Externer Link
544.
Vogel F, Bodmann KF; Expertenkommission der Paul-Ehrlich-Gesellschaft. Empfehlungen zur kalkulierten parenteralen Initialtherapie bakterieller Erkrankungen bei Erwachsenen. Chemother J. 2004;13(2):46-105. Available from: http://www.chemotherapie-journal.de/uploads/media/CTJ_2004_02_Parenterale_Initialtherapie.pdf Externer Link
545.
Vogel F, Bodmann KF; Expertenkommission der Paul-Ehrlich-Gesellschaft. Empfehlungen zur kalkulierten parenteralen Initialtherapie bakterieller Erkrankungen bei Erwachsenen. Intraabdominale Infektionen. Chemother J. 2004;13(2):74-8. Available from: http://www.chemotherapie-journal.de/uploads/media/CTJ_2004_02_Parenterale_Initialtherapie.pdf
546.
Vogel F, Scholz H, al Nawas B, Elies W, Kresken M, Lode H. Rationaler Einsatz oraler Antibiotika bei Erwachsenen und Schulkindern. Chemother J. 2002;11(2):47-58.
547.
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 Dec;24(12):3423-34. DOI: 10.1185/03007990802550679 Externer Link
548.
Wacha H, Hoyme U, Isenmann R, Kujath P, Lebert C, Naber K, Salzbergeret B. Perioperative Antibiotika-Prophylaxe – Empfehlungen einer Expertenkommission der Paul-Ehrlich-Gesellschaft für Chemotherapie e.V. Chemother J. 2010;19(3):70-84.
549.
Wacha H, Schäfer V, Schöffel U, Holzheimer RG. Peritonitis und andere intraabdominelle Infektionen. In: Adam D, Doerr E, Linke S, Lode H, editors. Die Infektiologie. Heidelberg: Springer; 2004. p. 332-52.
550.
Wacha H, Warren B, Bassaris H, Nikolaidis P; Intra-Abdominal Infections Study Group. Comparison of sequential intravenous/oral ciprofloxacin plus metronidazole with intravenous ceftriaxone plus metronidazole for treatment of complicated intra-abdominal infections. Surg Infect (Larchmt). 2006 Aug;7(4):341-54. DOI: 10.1089/sur.2006.7.341 Externer Link
551.
Wagenlehner FM, Krcmery S, Held C, Klare I, Witte W, Bauernfeind A, Schneider I, Naber KG. Epidemiological analysis of the spread of pathogens from a urological ward using genotypic, phenotypic and clinical parameters. Int J Antimicrob Agents. 2002 Jun;19(6):583-91. DOI: 10.1016/S0924-8579(02)00093-6 Externer Link
552.
Wagenlehner FM, Lehn N, Witte W, Naber KG. In vitro activity of daptomycin versus linezolid and vancomycin against gram-positive uropathogens and ampicillin against enterococci, causing complicated urinary tract infections. Chemotherapy. 2005 May;51(2-3):64-9. DOI: 10.1159/000085611 Externer Link
553.
Wagenlehner FM, Loibl E, Vogel H, Naber KG. Incidence of nosocomial urinary tract infections on a surgical intensive care unit and implications for management. Int J Antimicrob Agents. 2006 Aug;28 Suppl 1:S86-90. DOI: 10.1016/j.ijantimicag.2006.05.011 Externer Link
554.
Wagenlehner FM, Naber KG. Fluoroquinolone Antimicrobial Agents in the Treatment of Prostatitis and Recurrent Urinary Tract Infections in Men. Curr Infect Dis Rep. 2005 Jan;7(1):9-16. DOI: 10.1007/s11908-005-0018-9 Externer Link
555.
Wagenlehner FM, Naber KG, Bschleipfer T, Brähler E, Weidner W. Prostatitis and male pelvic pain syndrome: diagnosis and treatment. Dtsch Arztebl Int. 2009 Mar;106(11):175-83. DOI: 10.3238/arztebl.2009.0175 Externer Link
556.
Wagenlehner FM, Niemetz AH, Weidner W, Naber KG. Spectrum and antibiotic resistance of uropathogens from hospitalised patients with urinary tract infections: 1994-2005. Int J Antimicrob Agents. 2008 Feb;31 Suppl 1:S25-34. DOI: 10.1016/j.ijantimicag.2007.07.029 Externer Link
557.
Wagenlehner FM, Pilatz A, Naber KG, Perletti G, Wagenlehner CM, Weidner W. Anti-infective treatment of bacterial urinary tract infections. Curr Med Chem. 2008;15(14):1412-27. DOI: 10.2174/092986708784567699 Externer Link
558.
Wagenlehner FM, Pilatz A, Naber KG, Weidner W. Therapeutic challenges of urosepsis. Eur J Clin Invest. 2008 Oct;38 Suppl 2:45-9. DOI: 10.1111/j.1365-2362.2008.02008.x Externer Link
559.
Wagenlehner FM, Wagenlehner C, Redman R, Weidner W, Naber KG. Urinary bactericidal activity of Doripenem versus that of levofloxacin in patients with complicated urinary tract infections or pyelonephritis. Antimicrob Agents Chemother. 2009 Apr;53(4):1567-73. DOI: 10.1128/AAC.01133-08 Externer Link
560.
Wagenlehner FM, Weidner W, Naber KG. Optimal management of urosepsis from the urological perspective. Int J Antimicrob Agents. 2007 Nov;30(5):390-7. DOI: 10.1016/j.ijantimicag.2007.06.027 Externer Link
561.
Wagenlehner FM, Weidner W, Naber KG. Pharmacokinetic characteristics of antimicrobials and optimal treatment of urosepsis. Clin Pharmacokinet. 2007;46(4):291-305. DOI: 10.2165/00003088-200746040-00003 Externer Link
562.
Wall RJ, Ely EW, Talbot TR, Weinger MB, Williams MV, Reischel J, Burgess LH, Englebright J, Dittus RS, Speroff T, Deshpande JK. Evidence-based algorithms for diagnosing and treating ventilator-associated pneumonia. J Hosp Med. 2008 Sep;3(5):409-22. DOI: 10.1002/jhm.317 Externer Link
563.
Walter C, Grötz KA, Kunkel M, Al-Nawas B. Prevalence of bisphosphonate associated osteonecrosis of the jaw within the field of osteonecrosis. Support Care Cancer. 2007 Feb;15(2):197-202. DOI: 10.1007/s00520-006-0120-z Externer Link
564.
Wang G, Hindler JF, Ward KW, Bruckner DA. Increased vancomycin MICs for Staphylococcus aureus clinical isolates from a university hospital during a 5-year period. J Clin Microbiol. 2006 Nov;44(11):3883-6. DOI: 10.1128/JCM.01388-06 Externer Link
565.
Warnke PH, Becker ST, Springer IN, Haerle F, Ullmann U, Russo PA, Wiltfang J, Fickenscher H, Schubert S. Penicillin compared with other advanced broad spectrum antibiotics regarding antibacterial activity against oral pathogens isolated from odontogenic abscesses. J Craniomaxillofac Surg. 2008 Dec;36(8):462-7. DOI: 10.1016/j.jcms.2008.07.001 Externer Link
566.
Weber SG, Gold HS, Hooper DC, Karchmer AW, Carmeli Y. Fluoroquinolones and the risk for methicillin-resistant Staphylococcus aureus in hospitalized patients. Emerging Infect Dis. 2003 Nov;9(11):1415-22. DOI: 10.3201/eid0911.030284 Externer Link
567.
Weigelt J, Itani K, Stevens D, Lau W, Dryden M, Knirsch C; Linezolid CSSTI Study Group. Linezolid versus vancomycin in treatment of complicated skin and soft tissue infections. Antimicrob Agents Chemother. 2005 Jun;49(6):2260-6. DOI: 10.1128/AAC.49.6.2260-2266.2005 Externer Link
568.
Weiss ME, Adkinson NF. ß-Lactam Allergy. In: Mandell GL, Bennett JE, Dolin R, editors. Mandell, Douglas, and Bennett’s Principles and Practice of Infectious Diseases. 7th ed. Churchill Livingstone: Elsevier; 2010. p. 347-54.
569.
Wellinghausen N, Bialasiewicz A, Mino de Kaspar H, Korn K, Schaller U, Zimmermann S. Mikrobiologisch-infektiologische Qualitätstandards: MiQ 31 – Infektionen des Auges. In: Podbielski A, Herrmann M, Kniehl E, Mauch H, Rüssmann H, editors. Qualitätsstandards in der mikrobiologisch-infektiologischen Diagnostik. München: Elsevier; 2011.
570.
Welte T, Brunkhorst FM. Antibiotika-Therapie der Sepsis. Med Welt. 2007;58:315-21.
571.
Wenisch C, Laferl H, Szell M, Krause R. Cefpirom plus Fosfomycin in der Behandlung von späten Beatmungsassoziierten Pneumonien. Chemother J. 2007;16(6):182-5.
572.
Whitcomb DC. Clinical practice. Acute pancreatitis. N Engl J Med. 2006 May;354(20):2142-50. DOI: 10.1056/NEJMcp054958 Externer Link
573.
Wiggins J, Patel SR. Changes in Kidney Function. In: Halter JB, Ouslander JG, Tinetti ME, Studenski S, High KP, Asthana S, editors. Hazzard’s geriatric medicine and gerontology. 6th ed. New York, Chicago: Mc Craw Hill Medical; 2009. p. 1009-15.
574.
Wilcox MH. Tigecycline and the need for a new broad-spectrum antibiotic class. Surg Infect (Larchmt). 2006 Feb;7(1):69-80. DOI: 10.1089/sur.2006.7.69 Externer Link
575.
Wisplinghoff H, Bischoff T, Tallent SM, Seifert H, Wenzel RP, Edmond MB. Nosocomial bloodstream infections in US hospitals: analysis of 24,179 cases from a prospective nationwide surveillance study. Clin Infect Dis. 2004 Aug;39(3):309-17. DOI: 10.1086/421946 Externer Link
576.
Witte W, Mielke M. Beta-Laktamasen mit breitem Wirkungsspektrum: Grundlagen, Epidemiologie, Schlussfolgerungen für die Prävention. Bundesgesundheitsblatt Gesundheitsforschung Gesundheitsschutz. 2004;46:881-90. DOI: 10.1007/s00103-003-0693-3 Externer Link
577.
Wolner B, Liebmann JM, Sassani JW, Ritch R, Speaker M, Marmor M. Late bleb-related endophthalmitis after trabeculectomy with adjunctive 5-fluorouracil. Ophthalmology. 1991 Jul;98(7):1053-60. DOI: 10.1016/S0161-6420(91)32177-8 Externer Link
578.
Wong JS, Chan TK, Lee HM, Chee SP. Endogenous bacterial endophthalmitis: an east Asian experience and a reappraisal of a severe ocular affliction. Ophthalmology. 2000 Aug;107(8):1483-91. DOI: 10.1016/S0161-6420(00)00216-5 Externer Link
579.
Wong PF, Gilliam AD, Kumar S, Shenfine J, O'Dair GN, Leaper DJ. Antibiotic regimens for secondary peritonitis of gastrointestinal origin in adults. Cochrane Database Syst Rev. 2005;(2):CD004539. DOI: 10.1002/14651858.CD004539.pub2 Externer Link
580.
Woodford N, Johnson AP, Morrison D, Speller DC. Current perspectives on glycopeptide resistance. Clin Microbiol Rev. 1995 Oct;8(4):585-615.
581.
Wunderink RG, Mendelson MH, Somero MS, Fabian TC, May AK, Bhattacharyya H, Leeper KV Jr, Solomkin JS. Early microbiological response to linezolid vs vancomycin in ventilator-associated pneumonia due to methicillin-resistant Staphylococcus aureus. Chest. 2008 Dec;134(6):1200-7. DOI: 10.1378/chest.08-0011 Externer Link
582.
Wunderink RG, Rello J, Cammarata SK, Croos-Dabrera RV, Kollef MH. Linezolid vs vancomycin: analysis of two double-blind studies of patients with methicillin-resistant Staphylococcus aureus nosocomial pneumonia. Chest. 2003 Nov;124(5):1789-97.
583.
Zahn C, Sangl J, Bierman AS, Miller MR, Friedman B, Wickizer SW, Meyer GS. Potentially inappropriate medication use in the community-dwelling elderly: findings from the 1996 Medical Expenditure Panel Survey. JAMA. 2001 Dec;286(22):2823-9. DOI: 10.1001/jama.286.22.2823 Externer Link
584.
Zhanel GG, Karlowsky JA, Rubinstein E, Hoban DJ. Tigecycline: a novel glycylcycline antibiotic. Expert Rev Anti Infect Ther. 2006 Feb;4(1):9-25. DOI: 10.1586/14787210.4.1.9 Externer Link
585.
Zhanel GG, Wiebe R, Dilay L, Thomson K, Rubinstein E, Hoban DJ, Noreddin AM, Karlowsky JA. Comparative review of the carbapenems. Drugs. 2007;67(7):1027-52. DOI: 10.2165/00003495-200767070-00006 Externer Link
586.
Ziai WC, Lewin JJ 3rd. Improving the role of intraventricular antimicrobial agents in the management of meningitis. Curr Opin Neurol. 2009 Jun;22(3):277-82. DOI: 10.1097/WCO.0b013e32832c1396 Externer Link
587.
Zolk O, Eschenhagen T. Infektionserkrankungen – Medikamentinteraktion in der Antibiotikatherapie. Notfallmedizin. 2003;29(9):358-63. DOI: 10.1055/s-2003-42565 Externer Link