gms | German Medical Science

GMS Infectious Diseases

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

ISSN 2195-8831

Calculated initial parenteral treatment of bacterial infections: Sepsis

Guideline Calculated parenteral initial therapy

  • corresponding author Klaus-Friedrich Bodmann - Klinik für Internistische Intensiv- und Notfallmedizin und Klinische Infektiologie, Klinikum Barnim GmbH, Werner Forßmann Krankenhaus, Eberswalde, Germany
  • Rainer Höhl - Institut für Klinikhygiene, Medizinische Mikrobiologie und Klinische Infektiologie, Klinikum Nürnberg, Germany
  • Wolfgang Krüger - Klinik für Anästhesiologie und Operative Intensivmedizin, Klinikum Konstanz, Germany
  • Beatrice Grabein - Stabsstelle Klinische Mikrobiologie und Krankenhaushygiene, Klinikum der Universität München, Munich, Germany
  • Wolfgang Graninger - Vienna, Austria

GMS Infect Dis 2020;8:Doc09

doi: 10.3205/id000053, urn:nbn:de:0183-id0000533

This is the English version of the article.
The German version can be found at: http://www.egms.de/de/journals/id/2020-8/id000053.shtml

Published: March 26, 2020

© 2020 Bodmann et al.
This is an Open Access article distributed under the terms of the Creative Commons Attribution 4.0 License. See license information at http://creativecommons.org/licenses/by/4.0/.


Abstract

This is the eleventh chapter of the guideline “Calculated initial parenteral treatment of bacterial infections in adults – update 2018” in the 2nd updated version. The German guideline by the Paul-Ehrlich-Gesellschaft für Chemotherapie e.V. (PEG) has been translated to address an international audience.

Sepsis, defined as a life threatening organ dysfunction caused by a misregulated host response to an infection, is the third leading cause of death in Germany with a lethality rate of 30% to over 50%. An early, effective antimicrobial therapy is, next to infectious source control, the most important causal treatment option. It should be complemented by the mainly supportive measures of general intensive care therapy. Prior antimicrobial therapy, the patient’s medical history (e.g. risk factors for multiresistant agents) and small-scale epidemiology are to be considered as part of the therapeutic and practical decisions. A modification of the often needed broad initial calculated combination therapy is desirable. In the future, prompt measurements of plasma concentrations of antiinfectives, especially for the sepsis patient with diverse and partly conflicting pathophysiological changes, will have great importance regarding efficacy, toxicity and resistance development. In order to apply those complex strategies in clinical routine, there is a requirement for a strong interdisciplinary collaboration between the intensive care unit, clinical infectiology, microbiology, and clinical pharmacology, ideally in the framework of a functional antimicrobial stewardship program.


Introduction

The treatment of sepsis, especially with increasingly multidrug-resistant or selective pathogens (acronym “ESCAPE” [1]), represents one of the greatest challenges for clinically active physicians [2], [3].

Particularly in the field of intensive care, sepsis and septic shock are of particular importance due to increasing incidence, a slight drop in mortality of 30–50% and high costs.

In 2013, according to a survey by Fleischmann et al. [4] 279,530 cases of sepsis were reported to the Institute for the Hospital Remuneration System (InEK), with 67,849 of these patients (24.3%) dying. The mortality rate appears to be particularly high in patients with severe sepsis (60.3%). Sepsis is the third leading cause of death in Germany.

These data are complemented by a prospective, multicenter point-prevalence study (INSEP Study), in which 11,883 patients from 133 German intensive care units were investigated. Of these, 1,503 patients (12.6%) had a diagnosis of severe sepsis or septic shock, of which 860 cases (57.2%) were of nosocomial origin. The mortality of patients with sepsis was 34.3% during intensive care stay, compared to 6% of patients without sepsis. Overall, this study confirmed the tendency towards slightly lower mortality compared to previous studies but increasing prevalence of sepsis [5].

International comparisons of the incidence and mortality of sepsis are difficult as disease patterns, age structures, epidemiologically available data, and criteria for hospitalization or intensive care units differ significantly between countries. With these limitations, the mortality of severe sepsis is estimated at an average of 28% [6].

Fungal infections must be a consideration in patients in non-neutropenic intensive care. In the German prevalence study [7], fungi were detected microbiologically as the cause of severe sepsis in 17.8% of cases. In the US Candida spp. are now the third most frequent pathogen in blood cultures from patients in intensive care units [8], in Germany the fourth most frequent with the highest pathogen-associated mortality [9].

The predisposing diseases of sepsis include all forms of immune deficiency, such as tumors, diabetes mellitus, kidney and liver diseases and hemoblastosis, in the field of surgical intensive care for example polytrauma, burns and major high-risk procedures such as organ transplants. Effective antimicrobial treatment is the most important causal treatment option in addition to early source control. It is complemented by general intensive care with its mainly supportive measures [10].

According to current knowledge, Schuster’s definition of microbial sepsis is still considered the best [11]: “Sepsis is the sum of the life-threatening clinical manifestations and pathophysiological changes in response to activity of pathogens and their products entering the bloodstream from a focal point of infection, activating large biological cascade systems and specialized cell systems and triggering the formation and release of humoral and cellular mediators.”

The current criteria for the diagnosis of sepsis consist of the detection of an infection and at least two of the following four criteria [12]:

1.
Fever above 38°C or in rare cases hypothermia below 36°C
2.
Tachypnea above 20/min or hypocapnia with a PaCO2 <32 mm Hg
3.
Tachycardia above 90/min
4.
Leukocytosis greater than 12,000/mm3 or leukopenia less than 4,000/mm3 or normal white blood cell count left shift in differential blood count (more than 10% immature forms)

The reduction to these four “SIRS criteria” or the necessity of ≥2 of the criteria was not uncontroversial because on the one hand up to a quarter of cases of sepsis are not covered by them and on the other hand SIRS criteria have been met even by simple, uncomplicated infections [13], [14], [15]. Septic conditions were classified into different degrees of clinical severity according to the old American consensus definition: SIRS, sepsis, severe sepsis, septic shock.

A task force of 19 experts has revised the definition of sepsis on behalf of the two world-leading societies ESICM (European Society of Intensive Care Medicine) and SCCM (Society of Critical Care Medicine), which is now called “Sepsis-3” [16], [17], [18]. According to this new definition, sepsis is defined as a “life threatening organ dysfunction caused by a misregulated host response to infection”, i.e. the new “sepsis” is the old “severe sepsis”. The focus is now on the SOFA score; the SIRS criteria on the systemic inflammatory response of the body have been dropped. A “qSOFA” (quick SOFA) is intended to facilitate screening without lab testing in non-intensive care patients (Figure 1 [Fig. 1]):

  • Respiratory rate ≥22/min
  • Altered consciousness (GCS <15)
  • Systolic blood pressure ≤100 mm Hg

qSOFA is considered positive if ≥2 criteria are met: follow by further search for organ dysfunction (SOFA score), start or escalate treatment, intensify monitoring.

Organ dysfunction is defined as an acute change of the SOFA score ≥2 points as a result of the infection (mortality ≥10%). The baseline SOFA score is assumed to be zero in patients with no known organ dysfunction. The SOFA score parameters are respiration, coagulation, liver, cardiovascular system, CNS, kidney.

Septic shock is defined as: Sepsis + vasopressor administration required to maintain a mean arterial blood pressure ≥65 mm Hg in persistent hypotension + serum lactate >2 mmol/l (>18 mg/dl) despite adequate volume replacement. For septic shock, hospital mortality exceeds 40%.

The pathophysiological findings of recent years show that septic conditions are caused by a complicated network of pro- and anti-inflammatory cytokines [19], [20]. A sepsis event is a dynamic process of transition from the stage of “simple sepsis” to “severe sepsis” or “septic shock” with organ dysfunction or organ failure but also the development of septic organ colonization. A detailed description of intensive care supportive and adjunctive treatment measures would go far beyond the objectives of these guidelines on calculated anti-infective initial treatment. For this we refer to the current guidelines of the Surviving Sepsis Campaign [21].

Rapid, adequate antimicrobial treatment and, whenever possible, early source control in the first few hours is key to patient survival. In a retrospective study from 2006 [22] Kumar was able to show that with every hour of treatment delay after onset of hypotension in septic shock mortality increases by 7.6%. To some extent conflicting studies were published on this topic in the following years. A recent meta-analysis [23] seems to suggest that there is no benefit regarding mortality if antibiotics are administered in sepsis within the first 3 hours after initial assessment in the emergency department or one hour after the onset of septic shock. In addition to methodological weaknesses (7 studies could not be included due to failure to communicate with the authors), no single randomized, controlled study was included because there were none. In addition, the studies were not limited to those with adequate, effective treatment and no statement was made on multiresistant pathogens or on source control.

In the current guidelines of the Surviving Sepsis Campaign, a strong recommendation is made for giving intravenous antibiotics no later than one hour after the diagnosis of sepsis or septic shock, although the evidence for this procedure is considered moderate [21]. This is supported by other current data [24], also from Germany [25].

Due to the increase in multidrug-resistant pathogens (especially MRSA, VRE but especially Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacteriaceae type 3MRGN and 4MRGN) [26] it is often necessary to start broad or even combined, antimicrobial treatment to adequately cover the pathogen spectrum [27]. Prior antimicrobial treatment and the patient’s history (for example risk factors for MRGN pathogens) should necessarily be included in the therapeutic and practical considerations (such as isolation).

The need for antimicrobial treatment should be reconsidered [28] and re-evaluated [29] daily. Combination treatment should be de-escalated once microbiological findings are available (less broad, discontinuation of a combination partner) [30].

In the physiologically and pharmacologically complex situation of sepsis, it is recommended to treat patients with high doses initially (i.e. in the first few days) in order to quickly reach a sufficiently effective level in sepsis patients with a high volume of distribution and creatinine clearance which is often elevated in hyperdynamic circulatory situations [31]. However, there are few data with good evidence on this topic. Attention should always be paid to signs of antimicrobial toxicity and possible interactions. Subsequently, the dosage should be adjusted to the organ deficiencies (kidney, liver). In the future, determination of plasma concentrations of antibiotics and antimycotics will be particularly important in sepsis patients in terms of efficacy, toxicity and development of resistance [31], [32], [33], [34], [35].

According to the current state of knowledge, in order to manage treatment, especially regarding the question of termination and effectiveness of antibiotic treatment, repeated determination of procalcitonin (PCT) in the serum should be carried out in addition to clinical assessment [22], [36], [37], [38], [39].

In order to be able to successfully implement the above-mentioned strategies in everyday clinical practice, it is necessary for intensive care physicians and clinicians to work closely with clinical infectiology, microbiology and clinical pharmacology. The modern term “antimicrobial stewardship” [40], [41] describes this approach. For example, infectiological advice for Staphylococcus aureus bacteremia significantly increased the quality of treatment and decreased mortality and length of hospital stay [42].

Overall, sepsis is a heterogeneous disease which is difficult to diagnose in the early stages and difficult to treat in the late stages. Early intervention improves the prognosis. Rapid and adequate antimicrobial therapy, microbiological diagnostics, source control and supportive treatment of internal organ dysfunction are the cornerstones of successful sepsis treatment. Nevertheless, excessive antimicrobial treatment must be avoided because of the expected collateral damage [43].


Microbiology and current resistance situation

The current recommendations on blood culture diagnostics were published within the scope of the “MiQ guidelines” (Quality standards in microbiological-infectiological diagnostics of the German Society for Hygiene and Microbiology, DGHM). This contains instructions on the collection of blood cultures, the place of collection, the procedure for venipuncture and for sample transport and processing, with and without an automatic detection system. When taking blood cultures, if possible before initiating antibiotic treatment, the following points should be observed in particular:

  • fresh puncture of a peripheral vein, take samples from existing catheters only in addition
  • hygienic hand disinfection
  • wipe or spray disinfect the skin on an area of at least 5x 5 cm with alcoholic disinfectant, exposure time 1 min.
  • second skin disinfection inside out with sterile swab
  • wear disposable gloves
  • no re-palpation of the puncture site
  • venipuncture and removal of 8–10 ml (5–10 ml) of blood per blood culture bottle, i.e. 16–20 ml per blood culture set
  • taking three blood culture sets
  • wipe septum tops of blood culture bottles with alcoholic disinfectant
  • wait for the disinfectant to dry
  • inoculate blood culture bottles with fresh cannula (not used!) or use closed sampling system (TRBA!)
  • do not aerate the aerobic bottle
  • immediately transport blood culture bottles to the lab

The spectrum of sepsis pathogens is broad. In the German SEPNET study, 55% of cases were caused by Gram-positive bacteria, 54% by Gram-negative microorganisms and almost 18% by Candida species. Their sum exceeding 100% is explained by polymicrobial infections [7].

Data on the resistance situation in blood culture isolates in Germany are available from the ARS Antibiotic Resistance Surveillance System from 2015 [44] (see also chapter 2 [45]).

The proportion of methicillin-resistant strains of Staphylococcus aureus has fallen slightly in recent years, reaching 11.8% (n=7,740). However the proportion of methicillin-resistant isolates in coagulase-negative staphylococci remains high at 58.8% (n=27.804). The proportion of glycopeptide-resistant Enterococcus faecium strains in blood culture isolates stood at 12.2% in 2015 (n=1,729), falling slightly in 2011 from a previous high of 14.8% (n=573).

In the case of Escherichia coli, the proportion of fluoroquinolone-resistant strains has fallen slightly in recent years and for ciprofloxacin was 20.7% (n=11,611). The proportion of cefotaxime-resistant isolates expressing the presence of an ESBL is currently 11.5% (n=9,958). In Klebsiella pneumoniae, the ciprofloxacin resistance rate has remained more or less constant in recent years, reaching 12.1% in 2015 (n=2,051). The rate of ESBL-formers in Klebsiella pneumoniae, again measured by cefotaxime resistance, has also been virtually unchanged in recent years and last year was 13.0% (n=1,796). In the meantime, the first carbapenem-resistant Klebsiella isolates have been detected in blood cultures, even though the proportion of 0.2% (meropenem, n=2,032) is still very low.

In the case of Pseudomonas aeruginosa, the resistance rate to ceftazidime is 9.1% (n=10,769), to piperacillin/tazobactam 15.6% (n=1,073) and to meropenem 8.1% (n=1,081) but if intermediate strains are added to meropenem, this results in 16.7% intermediate and resistant isolates.


Pharmacokinetics and pharmacodynamics

The pharmacokinetics and pharmacodynamics of antibiotics in patients with severe sepsis and septic shock are sometimes significantly different from data collected in less severely ill patients. Pharmacokinetics are influenced by complex, sometimes counterproductive processes, so that antibiotic levels are difficult to predict. In the early stages of sepsis, hyperdynamic circulatory conditions dominate in many patients, leading to increased clearance of renally eliminated anti-infective agents (augmented renal clearance, ARC) compared to healthy people. The capillary leak also causes expansion of the extracellular space. These two factors lead to unexpectedly low plasma levels of hydrophilic and renally eliminated antibiotics, affecting most beta-lactams and also aminoglycosides and vancomycin [46], [47], [48]. As a result, therapeutic drug monitoring (TDM) should be carried out for these antibiotics, which is mandatory for aminoglycosides and vancomycin in any case because of their high toxic potential [49], [50]. For beta-lactams TDM would also be useful but it is rarely available for routine clinical work. Changes in pharmacokinetics are less pronounced for antibiotics with large volumes of distribution (for example fluoroquinolones), i.e. with predominantly intracellular accumulation [46].

As a progressing sepsis leads to more and more organ dysfunction, especially with renal insufficiency, reduced elimination leads to increased plasma levels and possibly the accumulation of mostly ineffective but potentially toxic metabolites of the drugs [47]. Added to this, for antibiotics with high protein binding, is displacement from binding through other drugs or due to pH shifts . If alternatives exist, consideration should therefore be given to antibiotics with lower protein binding and low toxic potential (for example in MSSA sepsis a cephalosporin instead of flucloxacillin, which is more than 90% protein-bound and has a high hepatotoxic risk).

There is however no answer to the question of whether the percentages given for protein binding can be transferred to the situation of treating critically ill patients. There is a discussion of whether it would not be better for the plasma levels of beta-lactam antibiotics in critically ill patients to be above the MIC during the entire dosing interval. In addition, it is stated that plasma levels should be up to 4 times above the MIC to ensure tissue penetration of the antibiotics. However in many cases this would mean a significantly higher dosage than previously used for beta-lactam antibiotics [32].

After initial administration of a loading dose to rapidly achieve the required effective level, continuous infusions of beta-lactam antibiotics, especially for intermediate-susceptibility pathogens, could improve treatment outcomes in critically ill patients. However in addition to practicability (shelf-life at room temperature, incompatibility with other drugs), the use of a continuous infusion without therapeutic drug monitoring (TDM) carries the risk that the plasma levels may be permanently below the MIC of the (often unknown) pathogen. Continuous antibiotic infusion should therefore only be used if TDM is available promptly, ideally complemented by determination of the MIC of the antibiotic for the pathogen (see chapter 3 [51]).

If TDM is not available, prolonged infusion of beta-lactams over 3–4 hours is a possible sensible compromise. In this way, the disadvantage of short infusion with (unnecessarily) high peak levels and rapid drop of the effective level below the MIC is avoided as well as the potential danger associated with continuous infusion of permanently staying below the MIC. For rapidly reaching a therapeutically effective level, the initial dose should be given in the form of a traditional short infusion.

In order to clinically translate the insights into the peculiarities of pharmacokinetics and pharmacodynamics, TDM should be established for the most important beta-lactams used at the hospital in question in patients with severe sepsis (for example ceftazidime or cefepime, piperacillin/tazobactam, meropenem or imipenem). Without TDM, the use of continuous infusions is discouraged.

In some hospitals where TDM is not available, it is common practice to use higher doses for patients in the hyperdynamic phase of sepsis if the kidneys are still functioning, at least on the first day of treatment. Treatment may then be to administer the initial dose of the antibiotics mentioned above, when indicated, as a short infusion, followed by prolonged infusions at the usual times on the ward. It should be pointed out that only Doripenem – which was withdrawn from the market – was approved for prolonged infusion. For the other beta-lactams there are promising individual studies and meta-analyzes demonstrating better clinical efficacy of continuous or prolonged infusion. In addition to a few prospective studies, the meta-analyzes also include retrospective and cohort studies with limited significance [52], [53]. A recent meta-analysis [54] evaluated randomized, prospective studies of continuous versus intermittent beta-lactam infusion based on individual patient data [55], [56], [57]. There was a significant reduction in hospital mortality with continuous infusion (19.6% versus 26.3%) but without therapeutic drug monitoring [54].

Despite convincing data in vitro and in vivo, it remains difficult to demonstrate the superiority of continuous or prolonged infusions in clinical trials. The reasons for this are manifold, ranging from the difficulty of infection diagnosis to lack of pathogen identification to the fact that, in very sensitive pathogens, effective levels above the MIC can be achieved for a sufficient length of time even with traditional intermittent bolus administration [58], [59], [60].

In terms of pharmacodynamics, aminoglycosides are a mirror image to beta-lactams. This is because the bactericidal effect is improved by high peak levels, followed by pronounced post-antibiotic effects, which allow the plasma levels to drop below the MIC for many hours. TDM is established nationwide and is obligatory due to the high nephrotoxicity and ototoxicity. After bolus administration of the total daily dose, the next dose is given at the earliest after 24 hours, when the trough level for gentamicin or tobramycin is below 1 mg/l.

For fluoroquinolones, in order to optimize efficacy, it is recommended to generate the largest possible area of the time-plasma-mirror curve above the MIC (AUC>MIC). In clinical practice, this complicated mathematical term is of little use. Due to the mathematical link, fluoroquinolones can be better understood as analogous to aminoglycosides as peak-level-dependent antibiotics [47].


Treatment recommendations

In almost all patients, the initial, antimicrobial treatment is calculated according to the intervention treatment recommended by the Paul-Ehrlich-Gesellschaft. In some of the patients it is possible to modify initial intervention treatment through detecting the pathogen with an antibiogram. Initial anti-infective selection is influenced by the suspected source of infection, underlying diseases and risk factors (for example whether an infection is community-acquired or nosocomial, time of onset of infection and prior antimicrobial treatment).

Table 1 [Tab. 1] shows treatment recommendations for unknown pathogens in relation to the type and localization of the infection and Table 2 [Tab. 2] shows treatment recommendations for cases where the pathogens have been identified. Table 3 [Tab. 3] shows the recommendation grades for the use of antibiotics in the indication “nosocomially acquired sepsis with unknown pathogen and unknown site of infection”. The wide variety of treatment options listed in Table 1 [Tab. 1] and Table 2 [Tab. 2] is due to different degrees of severity of the disease and the risk factors of the patient. The duration of treatment should be 7–10 days. Exceptions are slow response to treatment, a non-restorable focus and immunosuppression [4]. In PCT-directed antibiotic treatment, the duration of antibiotic treatment may also be shorter than 7 days if by that time there has been a PCT decrease of more than 80% compared to the highest measured value or if the absolute PCT has a measured value ≤0.25 ng/l.

Although the data are insufficient, initial combination treatment should always be performed in patients suffering from life-threatening illness (Table 1 [Tab. 1]). This approach is supported, amongst other things, by the results of the Surviving Sepsis Campaign. Dellinger et al. recommend administration of one or more substances with a broad spectrum and good penetration into tissue for calculated initial treatment [28].

This strategy should be evaluated after 72 hours at the latest. Combination treatment is explicitly called for in cases of suspected or proven Pseudomonas infection [28], [61]. Traditionally, aminoglycosides have been the preferred combination partners for beta-lactam antibiotics. The option of using fluoroquinolones as a combination partner of beta-lactam antibiotics is backed up by the work of Paul et al. [62], [63]. Fluoroquinolones offer pharmacokinetic benefits, are associated with lower toxicity, and there is no need to measure levels regularly. However, resistance rates for fluoroquinolones are consistently higher than for aminoglycosides. In view of the occasionally high fluoroquinolones resistance rates, fosfomycin is another option as a combination partner with good tissue penetration.

In cases of sepsis, all anti-infective drugs must be administered intravenously and in high doses. Neither sequential therapy nor dose reduction are proven by studies in this indication.

In severe sepsis or septic shock and with unknown sepsis focus, it should be combined with a lipopeptide (daptomycin) [7], [40], [64], [65] or a glycopeptide in high-risk patients with a high rate of MRSA. Alternatively, ceftobiprole (group 5 cephalosporin) may also be used in combination with a fluoroquinolone or fosfomycin in these patients since ceftobiprole has good efficacy against MRSA. However it is doubtful whether the approved dosage of 3x 500 mg i.v., in the form of a 2-hour infusion in patients with normal renal function, is adequate. According to the data published in ECCMID 2015 by Torres et al. [66] one should aim for a dose of 3x 1,000 mg ceftobiprole in such patients. Ceftolozane/tazobactam could also be a useful treatment option in this indication. Here, however, the lack of efficacy of this combination of substances against staphylococci and most anaerobes has to be considered. The dose for the treatment of sepsis should be 3x 3 g i.v.

In addition, anaerobes can be expected in sepsis which originates from the respiratory tract, especially with Streptococcus pneumoniae, Staphylococcus aureus and various Enterobacteriaceae as well as in aspiration pneumonia. In case of serious risk situations or hospital stays of more than 5 days, Pseudomonas aeruginosa, Acinetobacter spp. and Stenotrophomonas maltophilia can be expected. The pathogen spectrum can vary greatly from hospital to hospital. A recent study indicates that Gram-negative pathogens in ventilated patients can increasingly also be expected in short hospital stays [67]. In high-risk patients with severe sepsis or septic shock and high MRSA hospital rates, it should be combined with oxazolidinone (linezolid) [40]. Ceftobiprole, in combination with a fluoroquinolone or fosfomycin, is a useful alternative. Ceftobiprole is a treatment option that can also be administered at a suitable dose (3x 1 g i.v.) in cases of pneumogenic sepsis [66].

In addition, ceftolozane/tazobactam should be considered as a further option for calculated initial treatment in patients with severe sepsis or septic shock and unknown septic focus as well as in pneumogenic sepsis because of its excellent efficacy against pseudomonads (including MDR) and ESBL-producers. Its ineffectiveness against staphylococci and anaerobics must, however, be compensated by an appropriate combination partner.

If sepsis originates in the urinary tract without previous instrumental intervention, primarily Escherichia coli and Proteus mirabilis are to be expected as sepsis pathogens. After urological interventions other Enterobacteriaceae, Pseudomonas aeruginosa, enterococci and staphylococci must also be considered.

If the starting point is the intestine or a gynecological organ, the following pathogens must be expected: Enterobacteriaceae, anaerobes, enterococci, Pseudomonas spp., Staphylococcus aureus.

In biliary sepsis, pathogen colonization in the bile ducts and thus the risk of bacteremia increases with the degree of outflow obstruction. In occlusive ictus, more than 75% of patients have pathogens in their blood. The spectrum includes Enterobacteriaceae, enterococci and anaerobes. In post-operative bacteremia, cholangiotic sepsis and sub-hepatic abscesses as well as in interventional procedures (ERCP or endoscopic papillotomy), other Gram-negative pathogens, including Pseudomonas aeruginosa, have been identified. If sepsis originates from the gut/gynecological organs and biliary tract, in severe sepsis or septic shock, it can be combined with a glycylcycline (tigecycline) [40], [68], [69].

If the source is the skin or soft tissue, infections by Streptococcus pyogenes, Staphylococcus aureus (also MRSA) and mixed infections with the additional involvement of non-A streptococci, anaerobes, Enterobacteriaceae or Pseudomonas aeruginosa are possible.

The pathogen spectrum of catheter-associated sepsis includes coagulase-negative staphylococci, Staphylococcus aureus, Gram-negative rod bacteria, Candida spp., Corynebacterium jeikeium and propionibacteria. Another therapeutic option is the lipopeptide daptomycin [7], [65] as an alternative to the glycopeptide.

The monotherapy recommendations presented in Table 1 [Tab. 1] are based on the results of well-documented, randomized clinical studies. In contrast, there is a general lack of clinical studies on combination treatment recommendations. Accordingly, these recommendations are based on expert opinions, this is especially true for combination therapy with a fluoroquinolone.


Note

This is the eleventh chapter of the guideline “Calculated initial parenteral treatment of bacterial infections in adults – update 2018” in the 2nd updated version. The German guideline by the Paul-Ehrlich-Gesellschaft für Chemotherapie e.V. (PEG) has been translated to address an international audience.


Competing interests

The authors declare that they have no competing interests.


References

1.
Peterson LR. Bad bugs, no drugs: no ESCAPE revisited. Clin Infect Dis. 2009 Sep;49(6):992-3. DOI: 10.1086/605539 External link
2.
Angus DC, van der Poll T. Severe sepsis and septic shock. N Engl J Med. 2013 Aug;369(9):840-51. DOI: 10.1056/NEJMra1208623 External link
3.
Marik PE. Don't miss the diagnosis of sepsis!. Crit Care. 2014 Sep;18(5):529. DOI: 10.1186/s13054-014-0529-6 External link
4.
Fleischmann C, Thomas-Rueddel DO, Hartmann M, Hartog CS, Welte T, Heublein S, Dennler U, Reinhart K. Hospital Incidence and Mortality Rates of Sepsis. Dtsch Arztebl Int. 2016 Mar;113(10):159-66. DOI: 10.3238/arztebl.2016.0159 External link
5.
SepNet Critical Care Trials Group. Incidence of severe sepsis and septic shock in German intensive care units: the prospective, multicentre INSEP study. Intensive Care Med. 2016 Dec;42(12):1980-1989. DOI: 10.1007/s00134-016-4504-3 External link
6.
Fleischmann C, Scherag A, Adhikari NK, Hartog CS, Tsaganos T, Schlattmann P, Angus DC, Reinhart K; International Forum of Acute Care Trialists. Assessment of Global Incidence and Mortality of Hospital-treated Sepsis. Current Estimates and Limitations. Am J Respir Crit Care Med. 2016 Feb;193(3):259-72. DOI: 10.1164/rccm.201504-0781OC External link
7.
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 External link
8.
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 External link
9.
Meyer E, Geffers C, Gastmeier P, Schwab F. No increase in primary nosocomial candidemia in 682 German intensive care units during 2006 to 2011. Euro Surveill. 2013 Jun 13;18(24). pii: 20505. DOI: 10.2807/ese.18.24.20505-en External link
10.
Seymour CW, Rosengart MR. Septic Shock: Advances in Diagnosis and Treatment. JAMA. 2015 Aug;314(7):708-17. DOI: 10.1001/jama.2015.7885 External link
11.
Schuster HP, Werdan K, editors. Intensivtherapie bei Sepsis und Multiorganversagen. Berlin, Heidelberg: Springer; 2000. DOI: 10.1007/978-3-662-07962-1 External link
12.
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; 101(6):1644-55. DOI: 10.1378/chest.101.6.1644 External link
13.
Liu V, Escobar GJ, Greene JD, Soule J, Whippy A, Angus DC, Iwashyna TJ. Hospital deaths in patients with sepsis from 2 independent cohorts. JAMA. 2014 Jul;312(1):90-2. DOI: 10.1001/jama.2014.5804 External link
14.
Kaukonen KM, Bailey M, Pilcher D, Cooper DJ, Bellomo R. Systemic inflammatory response syndrome criteria in defining severe sepsis. N Engl J Med. 2015 Apr;372(17):1629-38. DOI: 10.1056/NEJMoa1415236 External link
15.
Shankar-Hari M, Deutschman CS, Singer M. Do we need a new definition of sepsis? Intensive Care Med. 2015 May;41(5):909-11. DOI: 10.1007/s00134-015-3680-x External link
16.
Singer M, Deutschman CS, Seymour CW, Shankar-Hari M, Annane D, Bauer M, Bellomo R, Bernard GR, Chiche JD, Coopersmith CM, Hotchkiss RS, Levy MM, Marshall JC, Martin GS, Opal SM, Rubenfeld GD, van der Poll T, Vincent JL, Angus DC. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA. 2016 Feb;315(8):801-10. DOI: 10.1001/jama.2016.0287 External link
17.
Seymour CW, Liu VX, Iwashyna TJ, Brunkhorst FM, Rea TD, Scherag A, Rubenfeld G, Kahn JM, Shankar-Hari M, Singer M, Deutschman CS, Escobar GJ, Angus DC. Assessment of Clinical Criteria for Sepsis: For the Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA. 2016 Feb;315(8):762-74. DOI: 10.1001/jama.2016.0288 External link
18.
Shankar-Hari M, Phillips GS, Levy ML, Seymour CW, Liu VX, Deutschman CS, Angus DC, Rubenfeld GD, Singer M; Sepsis Definitions Task Force. Developing a New Definition and Assessing New Clinical Criteria for Septic Shock: For the Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA. 2016 Feb 23;315(8):775-87. DOI: 10.1001/jama.2016.0289 External link
19.
Hotchkiss RS, Monneret G, Payen D. Sepsis-induced immunosuppression: from cellular dysfunctions to immunotherapy. Nat Rev Immunol. 2013 Dec;13(12):862-74. DOI: 10.1038/nri3552 External link
20.
Uhle F, Lichtenstern C, Brenner T, Weigand MA. Sepsis und Multiorganversagen – Pathophysiologie der Sepsis [Pathophysiology of sepsis]. Anasthesiol Intensivmed Notfallmed Schmerzther. 2015 Feb;50(2):114-22. DOI: 10.1055/s-0041-100391 External link
21.
Rhodes A, Evans LE, Alhazzani W, Levy MM, Antonelli M, Ferrer R, Kumar A, Sevransky JE, Sprung CL, Nunnally ME, Rochwerg B, Rubenfeld GD, Angus DC, Annane D, Beale RJ, Bellinghan GJ, Bernard GR, Chiche JD, Coopersmith C, De Backer DP, French CJ, Fujishima S, Gerlach H, Hidalgo JL, Hollenberg SM, Jones AE, Karnad DR, Kleinpell RM, Koh Y, Lisboa TC, Machado FR, Marini JJ, Marshall JC, Mazuski JE, McIntyre LA, McLean AS, Mehta S, Moreno RP, Myburgh J, Navalesi P, Nishida O, Osborn TM, Perner A, Plunkett CM, Ranieri M, Schorr CA, Seckel MA, Seymour CW, Shieh L, Shukri KA, Simpson SQ, Singer M, Thompson BT, Townsend SR, Van der Poll T, Vincent JL, Wiersinga WJ, Zimmerman JL, Dellinger RP. Surviving Sepsis Campaign: International Guidelines for Management of Sepsis and Septic Shock: 2016. Crit Care Med. 2017 Mar;45(3):486-552. DOI: 10.1097/CCM.0000000000002255 External link
22.
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; 34(6):1589-96. DOI: 10.1097/01.CCM.0000217961.75225.E9 External link
23.
Sterling SA, Miller WR, Pryor J, Puskarich MA, Jones AE. The Impact of Timing of Antibiotics on Outcomes in Severe Sepsis and Septic Shock: A Systematic Review and Meta-Analysis. Crit Care Med. 2015 Sep;43(9):1907-15. DOI: 10.1097/CCM.0000000000001142 External link
24.
Seymour CW, Gesten F, Prescott HC, Friedrich ME, Iwashyna TJ, Phillips GS, Lemeshow S, Osborn T, Terry KM, Levy MM. Time to Treatment and Mortality during Mandated Emergency Care for Sepsis. N Engl J Med. 2017 06;376(23):2235-2244. DOI: 10.1056/NEJMoa1703058 External link
25.
Bloos F, Rüddel H, Thomas-Rüddel D, Schwarzkopf D, Pausch C, Harbarth S, Schreiber T, Gründling M, Marshall J, Simon P, Levy MM, Weiss M, Weyland A, Gerlach H, Schürholz T, Engel C, Matthäus-Krämer C, Scheer C, Bach F, Riessen R, Poidinger B, Dey K, Weiler N, Meier-Hellmann A, Häberle HH, Wöbker G, Kaisers UX, Reinhart K; MEDUSA study group. Effect of a multifaceted educational intervention for anti-infectious measures on sepsis mortality: a cluster randomized trial. Intensive Care Med. 2017 Nov;43(11):1602-1612. DOI: 10.1007/s00134-017-4782-4 External link
26.
Raman G, Avendano E, Berger S, Menon V. Appropriate initial antibiotic therapy in hospitalized patients with gram-negative infections: systematic review and meta-analysis. BMC Infect Dis. 2015 Sep;15:395. DOI: 10.1186/s12879-015-1123-5 External link
27.
Kumar A, Safdar N, Kethireddy S, Chateau D. A survival benefit of combination antibiotic therapy for serious infections associated with sepsis and septic shock is contingent only on the risk of death: a meta-analytic/meta-regression study. Crit Care Med. 2010 Aug;38(8):1651-64. DOI: 10.1097/CCM.0b013e3181e96b91 External link
28.
Dellinger RP, Levy MM, Rhodes A, Annane D, Gerlach H, Opal SM, Sevransky JE, Sprung CL, Douglas IS, Jaeschke R, Osborn TM, Nunnally ME, Townsend SR, Reinhart K, Kleinpell RM, Angus DC, Deutschman CS, Machado FR, Rubenfeld GD, Webb SA, Beale RJ, Vincent JL, Moreno R; Surviving Sepsis Campaign Guidelines Committee including the Pediatric Subgroup. Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock: 2012. Crit Care Med. 2013 Feb;41(2):580-637. DOI: 10.1097/CCM.0b013e31827e83af External link
29.
Dellinger RP. The Surviving Sepsis Campaign: Where have we been and where are we going? Cleve Clin J Med. 2015 Apr;82(4):237-44. DOI: 10.3949/ccjm.82gr.15001 External link
30.
Garnacho-Montero J, Gutiérrez-Pizarraya A, Escoresca-Ortega A, Corcia-Palomo Y, Fernández-Delgado E, Herrera-Melero I, Ortiz-Leyba C, Márquez-Vácaro JA. De-escalation of empirical therapy is associated with lower mortality in patients with severe sepsis and septic shock. Intensive Care Med. 2014 Jan;40(1):32-40. DOI: 10.1007/s00134-013-3077-7 External link
31.
Roberts JA, Taccone FS, Lipman J. Understanding PK/PD. Intensive Care Med. 2016 Nov;42(11):1797-1800. DOI: 10.1007/s00134-015-4032-6 External link
32.
Taccone FS, Laterre PF, Dugernier T, Spapen H, Delattre I, Wittebole X, De Backer D, Layeux B, Wallemacq P, Vincent JL, Jacobs F. Insufficient β-lactam concentrations in the early phase of severe sepsis and septic shock. Crit Care. 2010;14(4):R126. DOI: 10.1186/cc9091 External link
33.
Mouton JW, Ambrose PG, Canton R, Drusano GL, Harbarth S, MacGowan A, Theuretzbacher U, Turnidge J. Conserving antibiotics for the future: new ways to use old and new drugs from a pharmacokinetic and pharmacodynamic perspective. Drug Resist Updat. 2011 Apr;14(2):107-17. DOI: 10.1016/j.drup.2011.02.005 External link
34.
Drusano GL, Lodise TP. Saving lives with optimal antimicrobial chemotherapy. Clin Infect Dis. 2013 Jan;56(2):245-7. DOI: 10.1093/cid/cis863 External link
35.
Lipman J, Roberts J. Does Appropriate Antibiotic Therapy Mean Only Adequate Spectrum and Timing? Crit Care Med. 2015 Aug;43(8):1773-4. DOI: 10.1097/CCM.0000000000001060 External link
36.
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 External link
37.
Bouadma L, Luyt CE, Tubach F, Cracco C, Alvarez A, Schwebel C, Schortgen F, Lasocki S, Veber B, Dehoux M, Bernard M, Pasquet B, Régnier B, Brun-Buisson C, Chastre J, Wolff M; PRORATA trial group. Use of procalcitonin to reduce patients’ exposure to antibiotics in intensive care units (PRORATA trial): a multicentre randomised controlled trial. Lancet. 2010 Feb 6;375(9713):463-74. DOI: 10.1016/S0140-6736(09) 61879-1 External link
38.
Wacker C, Prkno A, Brunkhorst FM, Schlattmann P. Procalcitonin as a diagnostic marker for sepsis: a systematic review and meta-analysis. Lancet Infect Dis. 2013 May;13(5):426-35. DOI: 10.1016/S1473-3099(12) 70323-7 External link
39.
Bodmann KF. Diagnostische Marker und Effektivitätsbewertung in der antibakteriellen Therapie [Diagnostic markers and assessment of efficacy of antibacterial therapy]. Med Klin Intensivmed Notfmed. 2014 Apr;109(3):187-90. DOI: 10.1007/s00063-013-0312-5 External link
40.
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 External link
41.
Deutsche Gesellschaft für Infektiologie (DGI), et al. Strategien zur Sicherung rationaler Antibiotika-Anwendung im Krankenhaus. S3 - Guideline. AWMF-Registernummer 092-001. AWMF; 2013. Available from: http://www.awmf.org/uploads/tx_szleitlinien/092-001l_S3_Antibiotika_Anwendung_im_Krankenhaus_2013-verlaengert.pdf External link
42.
Bai AD, Showler A, Burry L, Steinberg M, Ricciuto DR, Fernandes T, Chiu A, Raybardhan S, Science M, Fernando E, Tomlinson G, Bell CM, Morris AM. Impact of Infectious Disease Consultation on Quality of Care, Mortality, and Length of Stay in Staphylococcus aureus Bacteremia: Results From a Large Multicenter Cohort Study. Clin Infect Dis. 2015 May;60(10):1451-61. DOI: 10.1093/cid/civ120 External link
43.
Bretonnière C, Leone M, Milési C, Allaouchiche B, Armand-Lefevre L, Baldesi O, Bouadma L, Decré D, Figueiredo S, Gauzit R, Guery B, Joram N, Jung B, Lasocki S, Lepape A, Lesage F, Pajot O, Philippart F, Souweine B, Tattevin P, Timsit JF, Vialet R, Zahar JR, Misset B, Bedos JP; Société de Réanimation de Langue Française (SRLF); Société Française d’Anesthésie et de Réanimation (SFAR). Strategies to reduce curative antibiotic therapy in intensive care units (adult and paediatric). Intensive Care Med. 2015 Jul;41(7):1181-96. DOI: 10.1007/s00134-015-3853-7 External link
44.
Robert-Koch-Institute. ARS - Antibiotic Resistance Surveillance. [Timestamp: 22.8.2016]. [cited 2017 Feb 13]. Available from: https://ars.rki.de External link
45.
Kresken M, Grabein B, Becker K, Straube E, Wichelhaus TA, Willinger B. Kalkulierte parenterale Initialtherapie bakterieller Infektionen: Mikrobiologie [Calculated parenteral initial treatment of bacterial infections: Microbiology]. GMS Infect Dis. 2020;8:Doc18. DOI: 10.3205/id000062 External link
46.
Roberts JA, Paul SK, Akova M, Bassetti M, De Waele JJ, Dimopoulos G, Kaukonen KM, Koulenti D, Martin C, Montravers P, Rello J, Rhodes A, Starr T, Wallis SC, Lipman J; DALI Study. DALI: defining antibiotic levels in intensive care unit patients: are current β-lactam antibiotic doses sufficient for critically ill patients? Clin Infect Dis. 2014 Apr;58(8):1072-83. DOI: 10.1093/cid/ciu027 External link
47.
Roberts JA, Abdul-Aziz MH, Lipman J, Mouton JW, Vinks AA, Felton TW, Hope WW, Farkas A, Neely MN, Schentag JJ, Drusano G, Frey OR, Theuretzbacher U, Kuti JL; International Society of Anti-Infective Pharmacology and the Pharmacokinetics and Pharmacodynamics Study Group of the European Society of Clinical Microbiology and Infectious Diseases. Individualised antibiotic dosing for patients who are critically ill: challenges and potential solutions. Lancet Infect Dis. 2014; 14(6):498-509. DOI: 10.1016/S1473-3099(14) 70036-2 External link
48.
Udy AA, Lipman J, Jarrett P, Klein K, Wallis SC, Patel K, Kirkpatrick CM, Kruger PS, Paterson DL, Roberts MS, Roberts JA. Are standard doses of piperacillin sufficient for critically ill patients with augmented creatinine clearance? Crit Care. 2015 Jan;19:28. DOI: 10.1186/s13054-015-0750-y External link
49.
Hao JJ, Chen H, Zhou JX. Continuous versus intermittent infusion of vancomycin in adult patients: A systematic review and meta-analysis. Int J Antimicrob Agents. 2016 Jan;47(1):28-35. DOI: 10.1016/j.ijantimicag.2015.10.019 External link
50.
Kumpf O, Braun JP, Brinkmann A, Bause H, Bellgardt M, Bloos F, Dubb R, Greim C, Kaltwasser A, Marx G, Riessen R, Spies C, Weimann J, Wöbker G, Muhl E, Waydhas C. Quality indicators in intensive care medicine for Germany – third edition 2017. Ger Med Sci. 2017 Aug 1;15:Doc10. DOI: 10.3205/000251 External link
51.
Derendorf H, Heinrichs T, Reimers T, Lebert C, Brinkmann A. Kalkulierte parenterale Initialtherapie bakterieller Infektionen: Pharmakokinetik und Pharmakodynamik [Calculated parenteral initial treatment of bacterial infections: Pharmacokinetics and pharmacodynamics]. GMS Infect Dis. 2020;8:Doc17. DOI: 10.3205/id000061 External link
52.
Chant C, Leung A, Friedrich JO. Optimal dosing of antibiotics in critically ill patients by using continuous/extended infusions: a systematic review and meta-analysis. Crit Care. 2013 Nov 29;17(6):R279. DOI: 10.1186/cc13134 External link
53.
Falagas ME, Tansarli GS, Ikawa K, Vardakas KZ. Clinical outcomes with extended or continuous versus short-term intravenous infusion of carbapenems and piperacillin/tazobactam: a systematic review and meta-analysis. Clin Infect Dis. 2013 Jan;56(2):272-82. DOI: 10.1093/cid/cis857 External link
54.
Roberts JA, Abdul-Aziz MH, Davis JS, Dulhunty JM, Cotta MO, Myburgh J, Bellomo R, Lipman J. Continuous versus Intermittent β-Lactam Infusion in Severe Sepsis. A Meta-analysis of Individual Patient Data from Randomized Trials. Am J Respir Crit Care Med. 2016 Sep;194(6):681-91. DOI: 10.1164/rccm.201601-0024OC External link
55.
Dulhunty JM, Roberts JA, Davis JS, Webb SA, Bellomo R, Gomersall C, Shirwadkar C, Eastwood GM, Myburgh J, Paterson DL, Lipman J. Continuous infusion of beta-lactam antibiotics in severe sepsis: a multicenter double-blind, randomized controlled trial. Clin Infect Dis. 2013 Jan;56(2):236-44. DOI: 10.1093/cid/cis856 External link
56.
Dulhunty JM, Roberts JA, Davis JS, Webb SA, Bellomo R, Gomersall C, Shirwadkar C, Eastwood GM, Myburgh J, Paterson DL, Starr T, Paul SK, Lipman J; BLING II Investigators for the ANZICS Clinical Trials Group. A Multicenter Randomized Trial of Continuous versus Intermittent β-Lactam Infusion in Severe Sepsis. Am J Respir Crit Care Med. 2015 Dec 1;192(11):1298-305. DOI: 10.1164/rccm.201505-0857OC External link
57.
Abdul-Aziz MH, Sulaiman H, Mat-Nor MB, Rai V, Wong KK, Hasan MS, Abd Rahman AN, Jamal JA, Wallis SC, Lipman J, Staatz CE, Roberts JA. Beta-Lactam Infusion in Severe Sepsis (BLISS): a prospective, two-centre, open-labelled randomised controlled trial of continuous versus intermittent beta-lactam infusion in critically ill patients with severe sepsis. Intensive Care Med. 2016 Oct;42(10):1535-1545. DOI: 10.1007/s00134-015-4188-0 External link
58.
Lipman J, Boots R. A new paradigm for treating infections: “go hard and go home”. Crit Care Resusc. 2009 Dec;11(4):276-81.
59.
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 External link
60.
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 External link
61.
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 External link
62.
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 External link
63.
Paul M, Lador A, Grozinsky-Glasberg S, Leibovici L. Beta lactam antibiotic monotherapy versus beta lactam-aminoglycoside antibiotic combination therapy for sepsis. Cochrane Database Syst Rev. 2014 Jan 7;(1):CD003344. DOI: 10.1002/14651858.CD003344.pub3 External link
64.
Cosgrove SE, Fowler VG Jr. Management of methicillin-resistant Staphylococcus aureus bacteremia. Clin Infect Dis. 2008 Jun 1;46 Suppl 5:S386-93. DOI: 10.1086/533595 External link
65.
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 External link
66.
Torres A, Sanches-Garcia M, Demeyer I, Saulay M, Schmitt-Hoffmann AH, Engelhardt M, et al. Pharmacokinetics, safety and tolerability of high-dose ceftobiprole medocaril administrered as prolonged infusion in intensive-car-unit (ICU) patients [Abstract]. In: 25th European Congress of Clinical Microbiology and Infectious Diseases; 2015 Apr 25-28; Copenhagen. O199.
67.
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 External link
68.
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 External link
69.
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 External link
70.
Grabein B, Ebenhoch M, Kühnen E, Thalhammer F. Kalkulierte parenterale Initialtherapie bakterieller Infektionen: Infektionen durch multiresistente gramnegative Stäbchen – ESBL-Bildner, Carbapenemase-bildende Enterobacteriaceae, Carbapenem-resistente Acinetobacter baumannii [Calculated parenteral initial treatment of bacterial infections: Infections with multi-resistant Gram-negative rods – ESBL producers, carbapenemase-producing Enterobacteriaceae, carbapenem-resistant Acinetobacter baumannii]. GMS Infect Dis. 2020;8:Doc04. DOI: 10.3205/id000048 External link