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: Pharmacokinetics and pharmacodynamics

Guideline Calculated parenteral initial therapy

  • corresponding author Hartmut Derendorf - Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, USA
  • Tobias Heinrichs - Bayer AG, Klinische Pharmazie, Leverkusen, Germany
  • Tobias Reimers - Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, USA
  • Cordula Lebert - Apotheke, Klinikum Nürnberg, Nuremberg, Germany
  • Alexander Brinkmann - Klinik für Anästhesie, operative Intensivmedizin und spezielle Schmerztherapie, Klinikum Heidenheim, Germany

GMS Infect Dis 2020;8:Doc17

doi: 10.3205/id000061, urn:nbn:de:0183-id0000618

This is the English version of the article.
The German version can be found at: http://www.egms.de/de/journals/id/2020-8/id000061.shtml

Published: March 26, 2020

© 2020 Derendorf 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 third 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.

The chapter features the pharmacokinetic and pharmacodynamics properties of the most frequently used antiinfective agents.


Pharmacology

In addition to the antimicrobial properties (pharmacodynamics) of a substance, the pharmacokinetic properties, i.e. the behavior in an organism, play a decisive role. Ultimately the question is whether the concentrations at the site of action are sufficient to inhibit the pathogens, kill them and possibly prevent the development of resistant pathogens. Adverse drug reactions and interactions should be minimized.

For the purpose of predicting efficacy, one speaks of PK/PD (pharmacokinetics/pharmacodynamics) when pharmacokinetic parameters or, in the simplest case, plasma and tissue concentrations are associated with the antimicrobial properties in vitro or in vivo.


Pharmacokinetics

Pharmacokinetic properties of drugs are determined by their physicochemical characteristics. The acid or base strength of a substance, its lipophilicity or hydrophilicity determine how the substance behaves under the physiological conditions of an organism. For example beta-lactam antibiotics and aminoglycosides are poor at penetrating membranes and therefore are located mainly in the extracellular space. An overview of pharmacokinetic parameters of individual substance groups is shown in Table 1 [Tab. 1].

An important pharmacokinetic parameter that describes the distribution of the drug in the body is the volume of distribution. Lipophilic substances, which can easily pass through membranes, are passively taken up intracellularly. Their volume of distribution is therefore high; with fluoroquinolones and macrolides it can be a multiple of the body volume. Substances with large volumes of distribution have lower plasma and interstitial levels but high intracellular concentrations. Water-soluble substances, on the other hand, penetrate cell membranes with difficulty and therefore mainly remain in the plasma and interstitium. Most pathogens are located in the interstitium, so concentration in these cases is crucial.

An important aspect of drug distribution is protein binding in serum. Depending on their physicochemical properties, antibiotics mainly bind to albumin.

Concentration-dependent binding is reversible. There is a dynamic balance between the free and the bound portion. In general, only the free, non protein-bound portion of an antibiotic is responsible for its action. As demonstrated for some antibiotics, high protein binding need not adversely affect the efficacy of a substance as long as there are sufficiently high unbound concentrations at the site of action. Clinical studies that appear to demonstrate a negative influence of protein binding were often performed with low total doses [1], [2], [3]. Furthermore, protein binding plays a role in kidney replacement procedures. Only the free, non protein-bound active substance portion can be eliminated via the artificial membranes of a kidney replacement procedure.

Equally significant for predicting efficacy is the question of tissue concentration. Tissue concentrations, as determined from biopsy material or surgical resectates, represent average concentrations in tissue homogenate. They do not adequately represent the complex processes or the heterogeneous distribution in the tissue. The measurements of tissue concentrations are important, for example when comparing two substances or substance groups.

Big progress was made in this area with the development of microdialysis. The measurement of antibiotic concentrations in compartments such as cerebrospinal fluid, alveolar film, pleural fluid, peritoneal fluid, pancreatic and prostatic fluid is important. Disease-related microcirculatory disturbances with compromised tissue perfusion, cell membranes with special anatomic structures and the presence of specific tissue receptors can be obstacles to the even distribution of antibiotics and thus influence treatment success. Table 2 [Tab. 2] shows the accessibility of different compartments for antibiotics. Thus, not only the physicochemical properties of the anti-infective agents but also the perfusion of the deep compartments play a crucial role in the actual site concentration [4], [5], [6].


Interaction between pharmacokinetics and pharmacodynamics

Since insufficient data is available on the concentration profiles at the site of infection, the pharmacokinetic evaluation of the various substances is usually carried out today using the different plasma concentrations; in severely ill intensive care patients, site concentrations may differ from the measurements in the primary compartment (serum, plasma) (especially in infections in deep compartments: lungs, bones, soft tissues) [4], [6]. Depending on the mechanism of action, different indices are recommended for the different groups of active ingredients to manage treatment.

The differences in the pharmacodynamic profile of the antibiotic groups are also explained by their different modes of action – concentration-dependent effect of fluoroquinolones, aminoglycosides, tetracyclines and glycylcyclines (tigecycline) and the time-dependent (non concentration-dependent) effect of beta-lactam antibiotics, lincosamides and macrolides (Table 3 [Tab. 3]). In the case of aminoglycosides, fluoroquinolones and cyclic lipopeptides (daptomycin), it has been shown that the ratio of peak concentration (Cmax) to the minimal inhibitory concentration (MIC) of the pathogen correlates with treatment success. For beta-lactam antibiotics, on the other hand, it is the percentage of the dosing interval in which the plasma concentration is above the pathogen MIC (t>MIC or %t>MIC). For fluoroquinolones and cyclic lipopeptides (daptomycin), the quotient of AUC (area under the curve) and MIC are considered predictive (the area under the 24-hour concentration time curve relative to the MIC: AUC24/MIC). This also applies to the group of glycopeptides. Previous findings on oxazolidinones (linezolid, tedizolid) indicate that both concentration and duration of exposure are relevant. The validation of these models for humans has been shown for some antibiotic groups.

In particular, in immunosuppressed patients and in infections in hard to reach compartments (abscesses, osteomyelitis, meningitis, necrotizing infections, see also Table 2 [Tab. 2]), the consideration of PK/PD indices in the choice of dosage regimen is of crucial importance. Also, the pharmacokinetic characteristics in the critically ill – which are affected by their hyperdynamic circulatory situation, endothelial damage, increased capillary permeability, hypoalbuminemia, extracorporeal circuits, intravenous administration of large amounts of fluid or administration of vasopressors – can contribute to an increased volume of distribution and by increasing renal perfusion in the absence of relevant organ dysfunction to an increased clearance of hydrophilic antibiotics and reduction of their plasma concentration [4], [6]. For such seriously ill patients, other PD indices may be important besides MIC. For treatment success in certain groups of pathogens (non-fermenters, e.g. Pseudomonas aeruginosa) including the avoidance of resistance development, concentration profiles may be more favorable in which the site concentrations remain well above the MIC (corresponds to MPC, mutant prevention concentration) [4], [7].

The data on PK/PD correlations offer the possibility of adjusting the dosage individually using therapeutic drug monitoring (TDM), especially in high-risk populations (such as critically ill patients, geriatric patients, patients with organ failure, infections with multidrug-resistant pathogens [e.g. extended-spectrum beta-lactamase (ESBL) producers]) [6], [8], [9], [10].

Clearance and volume of distribution determine the half-life of a substance. These parameters also co-determine the time the plasma concentration is above the MIC and for the total exposure (AUC) and play an important role in the calculation of the dosing interval.

Decreased function of the drug-eliminating organs (especially the kidneys and the liver) results in reduced clearance of antibiotics and prolongs the half-life, which may be one reason for the increased rate of adverse effects. The relevance of impaired renal and hepatic function plays a lesser role for antibiotics with a wide therapeutic range (broad concentration range between the effective and the toxic levels, for instance for penicillins, cephalosporins, carbapenems, macrolides, lincosamides, fluoroquinolones, linezolid) than for antibiotics with a narrow therapeutic range (such as aminoglycosides or vancomycin). Nevertheless, in intensive care patients it has been shown that increased plasma levels of beta-lactam antibiotics are associated with a poorer neurological outcome [5]. In addition to the microbiological efficacy, the extent of renal and extrarenal elimination as well as any potential nephro- and/or hepatotoxic potential of the antibiotics themselves or their metabolites play an important role in the selection of suitable antibiotics. These antibiotics (potentially nephrotoxic: aminoglycosides, vancomycin, teicoplanin, telavancin; potentially hepatotoxic: amoxicillin/clavulanic acid, flucloxacillin, fluoroquinolones, tetracyclines, rifampicin) should only be administered in life-threatening situations if the corresponding organ has impaired function. Possible risks due to the accumulation of potential toxic metabolites in patients with pronounced renal and hepatic insufficiency should also be considered. In principle, antibiotics with high extrarenal elimination should be selected in cases of impaired renal function and, in hepatic insufficiency, antibiotics with a predominantly renal excretion mode.

Antibiotics which are predominantly eliminated via the kidneys to varying degrees are also secreted by glomerular or tubular filtration (e.g. penicillins) or reabsorbed. If renal function is impaired, the dosage should be adjusted to the degree of renal impairment according to creatinine clearance. The following are crucial in determining the need for dose adjustment:

  • the proportion of renal elimination of the drug in normal renal function,
  • the toxicity of the substance,
  • the degree of renal impairment; and
  • the increase of creatinine clearance beyond normal levels (for example with reduced muscle mass, pregnancy or early stage diabetes mellitus).

In general the dosage specifications of the manufacturers should be followed. If these are not available, the dosing regimen for renal insufficiency should be adjusted by calculating the individual elimination fraction (Q) according to Dettli [11], [12].

Helpful links for dose adjustment in renal insufficiency:

Critically ill intensive care patients have a special status in terms of substance-specific pharmacokinetics. Recommended dosages and sensitivities (tested as sensitive, intermediate or resistant) are based on the assumption that the pharmacokinetics of the drug are equivalent to those of a “standard patient”. In fact, however, the distribution and elimination capacity of drugs in the critically ill is very variable and difficult to predict. The renal function of patients with severe infections alone shows great inter- and intra-individual variability, so that the drug clearance and thus the optimal dosage of predominantly renally excreted anti-infective drugs can vary by a factor of 10 [13]. This problem is not only clinically apparent with beta-lactam antibiotics [13] but also with reserve substances such as linezolid [14]. A review article provides helpful guidance on individualized dosing of anti-infective agents (e.g. web-based calculation programs such as CADDy [Calculator to Approximate Drug Dosing in Dialysis] in seriously ill intensive care patients [6]. Patients with organ replacement procedures (such as renal replacement procedures [hemodialysis, hemofiltration] [6], [8], [15], ECMO [16], ECLS) present a particular challenge here.

Unlike creatinine clearance in renal insufficiency, clinical scores in hepatic insufficiency (Child-Pugh score, MELD score) are not good predictors of drug metabolization and elimination.

Liver diseases have a different, unpredictable influence on the individual cytochrome P450 isoenzymes. Existing tests allow only a rough assessment of the function of the individual isoenzymes. The reduction in hepatic clearance and the associated need for dose adjustment may be relevant to antibiotics that are almost exclusively metabolized by liver enzymes, predominantly those with high lipophilicity and low polarity that can be poorly eliminated via the kidney (antibiotics with high extrarenal clearance: clindamycin, tedizolid, chloramphenicol and minocycline). When dosing with other tetracyclines, clavulanic acid, flucloxacillin, macrolides or streptogramins, higher grade hepatic insufficiency with reduced metabolization performance must also be considered. For antibiotics with a high presystemic elimination rate (e.g. ciprofloxacin), hepatic impairment may significantly increase the bioavailability after oral administration and thus the plasma concentration.

For all stages of renal and hepatic insufficiency, the loading dose (initial dose), which depends on the volume of distribution, should be identical to that for a healthy kidney or liver. Otherwise, initially reduced doses of antibiotics may take several days to reach an effective level. Since the success of antibiotic treatment mainly depends on the initial selection and an adequate dosage, this would jeopardize treatment success.

The dosage of antibiotics in overweight patients is a particular pharmacotherapeutic problem. The kinetics of many antibiotics are sometimes unpredictable due to unusual distributional processes in these patients. There is no clear relationship between the lipophilicity of the substances and their distribution in obese patients. Altered volume of distribution, clearance and problems in assessing kidney function using creatinine clearance are just some of the reasons that often cause overweight patients to be inadequately treated with standard doses of antibiotics. Subtherapeutic concentrations may then lead to clinical treatment failure and development of resistance, while supratherapeutic/excessively high concentrations usually lead to undesirable side effects (with the exception of aminoglycosides). Since an increased volume of distribution and increased clearance is generally to be expected in these patients, a weight-adapted dose adjustment is necessary. Which weight (TBW – total body weight, IBW – ideal body weight, LBW – lean body weight or ABW – adjusted body weight) should be used as the basis for the dose calculation is dependent both on the antibiotic itself (e.g. in tigecycline with a distribution volume of 7 to 10 l/kg [17]) as well as the type and duration of administration [18], [19], [20], [21].

Hydrophilic antibiotics (beta-lactams, aminoglycosides, glycopeptides) [22] are less well distributed in adipose tissue. When dosing these antibiotics IBW or ABW are usually used [23]. Using TBW can lead to overdoses. In contrast, lipophilic antibiotics (fluoroquinolones, macrolides, clindamycin, tetracyclines, tigecycline, cotrimoxazole, rifampicin, chloramphenicol) [22] have a higher volume of distribution. Consequently, increased adipose tissue in obese patients also leads to an increase in the volume of distribution compared to patients with normal weight. TBW tends to be used for dosing in this case [23]. It should be noted that the degree of hydrophilicity or lipophilicity within the two groups (hydrophilic and lipophilic antibiotics) differs from antibiotic to antibiotic.

For special patient populations (those with CF, sepsis, neutropenia, burns, or high body weight) Therapeutic Drug Monitoring (TDM) is recommended [6], [8], [9], [10] but only a few antibiotics have rapid tests available (for example, aminoglycosides, glycopeptides). Special dosage guidelines must be observed in the aforementioned patient groups. The different pharmacokinetic characteristics of the individual substances are summarized in Table 1 [Tab. 1].


Therapeutic drug monitoring

Many antibiotics are characterized by significant inter- and intra-individual differences in pharmacokinetic properties, especially in elimination behavior and volume of distribution. This is especially true in intensive care patients with severe sepsis, septic shock and consecutive multiple organ failure and profound changes in distribution spaces (e.g. capillary leak and infusion treatments) [6], [8]. As a result, even standard doses can result in a wide range of plasma concentrations [13], which, on the one hand, threatens the risk of under-dosing with insufficient therapeutic effect, on the other hand excessive plasma levels with the risk of undesirable toxic effects. The aim of therapeutic drug monitoring (TDM) is to find the optimal individual dosage for the patient taking into account pharmacokinetic principles and measurements of the drug concentration in the patient’s blood [6], [7], [8].

Prerequisites or indications for conducting TDM above all are:

  • Therapeutic and toxic effects are in a concentration related cause and effect relationship.
  • The substance has a narrow therapeutic range and exceeding the concentration range by even relatively degree can lead to toxic effects.
  • The pharmacokinetics of the drug are subject to significant intra- and inter-individual variability, especially in intensive care patients with severe sepsis and septic shock.
  • Pharmacokinetic target parameters (Cmax, Cmin, AUC) are known.
  • Sufficiently sensitive methods for determining concentration are available involving a reasonable amount of effort.

For many antibiotics, e.g. penicillins and cephalosporins, the risk of unwanted toxic effects is rather low, since they have a relatively large therapeutic range. For these antibiotics, treatment based on blood level is only recommended for certain patient groups (e.g. intensive care patients) [4], [6], [8], [10], [13]. In a mixed intensive care group, dosage adjustment is necessary in 20–30% of cases [4], [8]. Intensive care patients with elevated creatinine clearance are at particular risk of the associated under-dosing [4], [6], [8], [24], [25]. The measurement of beta-lactam concentrations is currently not widespread, as dedicated PK/PD targets and dose-adjustment strategies are currently being debated in scientific circles [6], [10]. The measurement is predominantly done by chromatography. Commercial measuring methods are not available in Germany [6], [8], [10]. Drugs where TDM is strongly recommended for their safe use include aminoglycosides and glycopeptides. Table 4 [Tab. 4] gives recommendations on the target ranges for the peak and trough levels of the most commonly used aminoglycosides and glycopeptides taking into account different patient populations.

In treatment with aminoglycosides, single administration of the total daily dose has become more widespread, with increased clinical effectiveness, lower toxicity and economic advantages [26], [27], [28], [29], [30], [31], [32], [33], [34], [35], [36]. Taking into account accepted PK/PD parameters, aminoglycoside peak levels well above the MIC of the pathogen (Cmax/MIC>10) are aimed for [37], [38]. The mean MIC of gentamicin is 2 mg/l for pathogens with reduced sensitivity (e.g. for Pseudomonas aeruginosa); thus, peak levels of at least 20 mg/l should be aimed for [39].

In the treatment of endocarditis and neutropenic patients, single dose administration is sufficient in most cases. For severe endocarditis (enterococci, heart valve prostheses), single dose administration is not recommended and multiple administration recommended, for example in combination with a synergistic antibiotic attacking the cell wall [40].

In treatment with the glycopeptide antibiotics vancomycin and teicoplanin the aims are permanent concentrations above the MIC of the relevant pathogens in accordance with their pharmacodynamic parameters. As mandated by TDM, trough levels are monitored [41]. For the treatment of life-threatening infections (meningitis and pneumonia) and reduced sensitivity agents, vancomycin trough levels of 15–20 mg/l should be the target [42], [43], [44]. However, the increased risk of nephrotoxicity above a vancomycin trough level >15 mg/l should be taken into account [45]. Evidence from recent literature suggests that continuous administration of vancomycin reduces the likelihood of nephrotoxic side effects [46], [47], [48], [49].

For the treatment of bone or prosthetic infections, teicoplanin trough levels of 20–25 mg/l are recommended [50]. When teicoplanin is used to treat bacterial endocarditis, trough levels should be at least 30–40 mg/l [51]. Trough levels above 60 mg/l are considered toxic [52].


Continuous or prolonged infusions of beta-lactam antibiotics

Beta-lactam antibiotics are effective if the MIC of the pathogens is exceeded as permanently as possible during the growth phase of the cell wall. Initially, the bactericidal activity increases with increasing concentrations of the antibiotic up to 4 to 5 times the MIC but higher levels will not improve the therapeutic outcome. This pharmacokinetic-pharmacodynamic relationship is described as a time-dependent (non-concentration-dependent) bactericide. For beta-lactam antibiotics, the concentration of unbound antibiotic should exceed the MIC of the pathogen at the site of infection for at least 40–60% of that time [53], with approximately 40% for carbapenems and higher for cephalosporins; penicillins are in between. These data are derived from animal studies. Clinical trial results in intensive care patients suggest that keeping above the MIC 100% of the time can improve outcome [54], [55], [56], [57], [58]. Since in intensive care patients with severe infections in deep compartments the plasma concentrations measured in the context of TDM do not correspond to active site concentrations, as a PK/PD target some experts recommend keeping the plasma level 4 to 5 times above the MIC for the full dosing interval [4], [6], [8], [10].

The pharmacokinetic properties of beta-lactam-antibiotics do not show great variability one from another. Beta-lactam antibiotics are rapidly distributed in the extracellular space after parenteral administration. At steady state, similar concentrations are reached after intermittent administration and bolus administration followed by continuous infusion [59], [60], [61], [62], [63], [64].

The manufacturer’s dosage recommendations usually call for administering beta-lactam antibiotics 2 to 4 times (1 to 6 times) depending on the pharmacokinetic parameters. As a result, approved indications which are confirmed by clinical studies usually result in sufficient free efficacy levels which exceed the MIC of sensitive pathogens. However, intermittent application often fails to achieve the goal of exceeding the pathogen’s MIC permanently at the site of the infection, as shown in PK/PD simulations, experimental and clinical studies [4], [6], [8], [10]. This is especially true in patients with high extracellular distribution spaces and an increased clearance rate. In particular this includes patients with a hyperdynamic circulatory situation and a capillary leak, e.g. in sepsis, patients with cystic fibrosis, drainage, bleeding, large burns, ascites, severe pancreatitis, patients with a BMI >30 kg/m2, congestive heart failure, edema, haemofiltration (depending on balance), dialysis patients (pre-dialysis) and pregnant women [4], [6], [8]. In contrast, dehydrated patients, dialysis patients following dialysis and patients under volume restrictions have a lower volume of distribution than normal patients. For high-risk patients and in geriatrics, bespoke antibiotic treatment is therefore required [4], [6], [8], [65], [66], [67], [68], [69], [70], [71], [72], [73], [74], [75], [76], [77].

Recommendations for prolonged administration (over 3–4 hours) or continuous administration of beta-lactam antibiotics are based on theoretical considerations supported by experimental studies or simulations. Clinical examinations show advantages for prolonged or continuous administration with longer-lasting serum levels above the MIC even at lower daily doses [78], [79], [80], [81], [82], [83], [84], [85], [86],[87], [88], [89], [90], [91], [92], [93], [94], [95], [96] with comparable effectiveness and safety [71], [97], [98] in terms of clinical and microbiological efficacy. Advantages of prolonged antibiotic administration have been shown, above all, in severely ill intensive care patients (APACHE II score >17) [99]. There is currently no agreement regarding the superiority of continuous or intermittent administration [100], [101], [102], [103], [104]. A recent clinical study was not able to objectify differences in mortality [105]. However, another recent study underlined an improved healing rate following continuous administration [106]. This result was confirmed again by a recent meta-analysis [107]. Continuous application of beta-lactam antibiotics without TDM is not recommended without restriction because there is a risk of permanently falling below the pathogen’s MIC. Not only does falling below the MIC result in a lack of efficacy of the antibiotic but it can also favor the selection of resistant mutants. Securely reaching rational PK/PD goals can only be ensured with TDM and it is therefore of crucial importance in continuous application. Against this background prolonged application is much safer.

Beta-lactam antibiotics have limited stability after preparation. It is not only the degree of degradation that is crucial but also the type of decomposition products that have allergenic potential. This fact is insufficiently considered in numerous studies on the stability of the substances. According to these studies, solutions of beta-lactam antibiotics are considered to be stable over a period of time if their degradation level is below 10%. The extent of the degradation depends on the solvent, the effects of light, the concentration of the antibiotic, the type of application aids as well as their production and temperature. In the case of close-fitting pump application in outpatient parenteral antibiotic therapy (OPAT), significant stability losses must be taken into account due to increased ambient heat.

The use of the recommended solvents is of great practical importance to ensure optimum solubility and stability. Almost without exception, all penicillins (dry substances) must be dissolved in aqua ad injectabilia in order to accelerate the dissolution behavior and to ensure particle freedom. Further dilution is then usually possible in conventional infusion solutions. Many beta-lactam antibiotics show a number of incompatibility reactions with other medicines when administered in the same infusion system. The manufacturer’s information on compatibility must be observed.

The most common adverse drug reactions of penicillins are allergies and pseudoallergic reactions. The cause of these reactions is the presence of an unstable beta-lactam structure or specific side chains. Penicillins in solution vary in stability depending on their side chains and the pH. The degradation products of penicillins act as haptens and can form covalent bonds with the body’s own proteins. They form a hapten-protein complex that can induce an allergy-producing immune response. The degradation products of penicillins have a significant potential for allergic reactions. Further information on safety can be found in chapter 4 [108].

The nature and extent of the degradation of the beta-lactam antibiotics are substance-dependent. Acylaminopenicillins, isoxazolylpenicillins, cephalosporins and aztreonam are generally more stable than benzylpenicillin because of their structure. Ring-opening, however, is also possible with cephalosporins by nucleophilic or (more rarely) electrophilic attack, as the example of ceftazidime and other cephalosporins shows [109]. The chemical stability of carbapenems varies widely and above all depends on the concentration of the solution and the temperature [110], [111]. There are very contradictory data for the stability of various beta-lactam antibiotics in infusion solutions. Here the recommendation of the manufacturer of the product should be observed.

Like beta-lactam antibiotics in patients with severe infections, linezolid has a high variability in serum concentrations, with insufficient blood levels under standard dosage regimens. Current data indicate that a continuous application can also make a useful contribution to achieving PK/PD goals [112], [113].

Conclusion

  • Due to pharmacokinetic/pharmacodynamic considerations, prolonged or continuous infusion of beta-lactam antibiotics is superior to intermittent administration with respect to the therapeutic goal of exceeding the MIC of the pathogens as continually as possible.
  • There is some clinical data on the significant superiority of this treatment regimen.
  • Continuous and intermittent infusions of a beta-lactam antibiotic show a comparable side effect profile.
  • Prolonged/continuous administration is recommended in patients whose pharmacokinetic parameters (volume of distribution, clearance) are significantly different from normal population data (for example, patients with sepsis and septic shock, cystic fibrosis, or patients with severe infections due to pathogens with reduced sensitivity). Continuous application is only recommended under TDM. Prolonged administration of a beta-lactam antibiotic is safe even without TDM.
  • Prolonged/continuous administration of the antibiotic should always be preceded by a bolus dose.
  • For substances with a high volume of distribution (for example tigecycline), a higher dose should be given initially.
  • Possible economic advantages result from continuous administration, since similar serum concentrations in steady state compared to intermittent administration can be achieved with lower daily doses in patients who are not seriously ill.
  • Some beta-lactam antibiotics are not suitable for continuous administration due to their low stability at room temperature. In these cases, only a prolonged infusion period (3–4 hours) is possible.
  • The manufacturer’s recommendations regarding the type of solvents and the concentrations of the antibiotic solutions must be strictly adhered to. Deviations can result in considerably limited stability.
  • Continuous administration of beta-lactam antibiotics requires separate access or lumen, as there are numerous incompatibility reactions with other medicinal products.
  • Bioavailability data of the antibiotics for sequential therapy can be found in Table 5 [Tab. 5].

Drug interactions

An important cause of unwanted side effects may be interactions with other drugs. In particular, the inhibition of hepatic monooxygenases, the cytochrome P450 enzymes, usually cause a higher risk of side effects, for example through some macrolides and fluoroquinolones as well as azole antifungals.

Also, induction-enhanced expression of enzymes of the cytochrome P450 enzyme system is possible, for example through rifampicin, barbiturates and carbamazepine. The consequence is a reduced plasma level with reduced effectiveness of the particular drug concerned.

More important examples of interactions of antibiotics with other drugs are presented in Table 6 [Tab. 6].


Note

This is the third 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.
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. DOI: 10.1128/AAC.35.12.2505 External link
2.
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 External link
3.
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. DOI: 10.1093/jac/26.suppl_A.1 External link
4.
Abdul-Aziz MH, Lipman J, Mouton JW, Hope WW, Roberts JA. Applying pharmacokinetic/pharmacodynamic principles in critically ill patients: optimizing efficacy and reducing resistance development. Semin Respir Crit Care Med. 2015 Feb;36(1):136-53. DOI: 10.1055/s-0034-1398490 External link
5.
Beumier M, Casu GS, Hites M, Wolff F, Cotton F, Vincent JL, Jacobs F, Taccone FS. Elevated β-lactam concentrations associated with neurological deterioration in ICU septic patients. Minerva Anestesiol. 2015 May;81(5):497-506.
6.
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; 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 Jun;14(6):498-509. DOI: 10.1016/S1473-3099(14)70036-2 External link
7.
Henrichfreise B, Wiegand I, Luhmer-Becker I, Wiedemann B. Development of resistance in wild-type and hypermutable Pseudomonas aeruginosa strains exposed to clinical pharmacokinetic profiles of meropenem and ceftazidime simulated in vitro. Antimicrob Agents Chemother. 2007 Oct;51(10):3642-9. DOI: 10.1128/AAC.00160-07 External link
8.
Frey OR, Helbig S, Brinkmann A, Fuchs T, Köberer A, König C, Röhr AC, Preisenberger J. Fragen und Antworten zur individuellen Dosierung von ß-Lactam-Antibiotika bei kritisch Kranken. Intensiv-News. 2015;19(4):30-3.
9.
Sime FB, Roberts MS, Tiong IS, Gardner JH, Lehman S, Peake SL, Hahn U, Warner MS, Roberts JA. Can therapeutic drug monitoring optimize exposure to piperacillin in febrile neutropenic patients with haematological malignancies? A randomized controlled trial. J Antimicrob Chemother. 2015 Aug;70(8):2369-75. DOI: 10.1093/jac/dkv123 External link
10.
Wong G, Brinkman A, Benefield RJ, Carlier M, De Waele JJ, El Helali N, Frey O, Harbarth S, Huttner A, McWhinney B, Misset B, Pea F, Preisenberger J, Roberts MS, Robertson TA, Roehr A, Sime FB, Taccone FS, Ungerer JP, Lipman J, Roberts JA. An international, multicentre survey of β-lactam antibiotic therapeutic drug monitoring practice in intensive care units. J Antimicrob Chemother. 2014 May;69(5):1416-23. DOI: 10.1093/jac/dkt523 External link
11.
Dettli L. The kidney in pre-clinical and clinical pharmacokinetics. J Clin Pharmacol Ther. 1984;15:241-54. DOI: 10.3999/jscpt.15.241 External link
12.
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.
13.
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
14.
Zoller M, Maier B, Hornuss C, Neugebauer C, Döbbeler G, Nagel D, Holdt LM, Bruegel M, Weig T, Grabein B, Frey L, Teupser D, Vogeser M, Zander J. Variability of linezolid concentrations after standard dosing in critically ill patients: a prospective observational study. Crit Care. 2014 Jul 10;18(4):R148. DOI: 10.1186/cc13984 External link
15.
Carlier M, Carrette S, Roberts JA, Stove V, Verstraete A, Hoste E, Depuydt P, Decruyenaere J, Lipman J, Wallis SC, De Waele JJ. Meropenem and piperacillin/tazobactam prescribing in critically ill patients: does augmented renal clearance affect pharmacokinetic/pharmacodynamic target attainment when extended infusions are used? Crit Care. 2013 May 3;17(3):R84. DOI: 10.1186/cc12705 External link
16.
Andes D, Craig WA. Pharmacokinetics and pharmacodynamics of outpatient intravenous antimicrobial therapy. Infect Dis Clin North Am. 1998 Dec;12(4):849-60, vi.
17.
Begg EJ, Barclay ML, Kirkpatrick CM. The therapeutic monitoring of antimicrobial agents. Br J Clin Pharmacol. 2001;52 Suppl 1:35S-43S.
18.
Jamal JA, Mueller BA, Choi GY, Lipman J, Roberts JA. How can we ensure effective antibiotic dosing in critically ill patients receiving different types of renal replacement therapy? Diagn Microbiol Infect Dis. 2015 May;82(1):92-103. DOI: 10.1016/j.diagmicrobio.2015.01.013 External link
19.
Donadello K, Antonucci E, Cristallini S, Roberts JA, Beumier M, Scolletta S, Jacobs F, Rondelet B, de Backer D, Vincent JL, Taccone FS. β-Lactam pharmacokinetics during extracorporeal membrane oxygenation therapy: A case-control study. Int J Antimicrob Agents. 2015 Mar;45(3):278-82. DOI: 10.1016/j.ijantimicag.2014.11.005 External link
20.
Muralidharan G, Micalizzi M, Speth J, Raible D, Troy S. Pharmacokinetics of tigecycline after single and multiple doses in healthy subjects. Antimicrob Agents Chemother. 2005 Jan;49(1):220-9. DOI: 10.1128/AAC.49.1.220-229.2005 External link
21.
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 External link
22.
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 External link
23.
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 External link
24.
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 External link
25.
Falagas ME, Karageorgopoulos DE. Adjustment of dosing of antimicrobial agents for bodyweight in adults. Lancet. 2010 Jan 16;375(9710):248-51. DOI: 10.1016/S0140-6736(09)60743-1 External link
26.
Al-Dorzi HM, Al Harbi SA, Arabi YM. Antibiotic therapy of pneumonia in the obese patient: dosing and delivery. Curr Opin Infect Dis. 2014 Apr;27(2):165-73. DOI: 10.1097/QCO.0000000000000045 External link
27.
Sime FB, Udy AA, Roberts JA. Augmented renal clearance in critically ill patients: etiology, definition and implications for beta-lactam dose optimization. Curr Opin Pharmacol. 2015 Oct;24:1-6. DOI: 10.1016/j.coph.2015.06.002 External link
28.
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 External link
29.
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 External link
30.
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 External link
31.
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 External link
32.
El Desoky E, Klotz U. Value, limitations and clinical impact of therapeutic drug-monitoring in adults. Drug Invest. 1993;6(3):127-36. DOI: 10.1007/BF03259732 External link
33.
Hehl EM, Drewelow B. Therapeutisches Drug Monitoring von Aminoglykosiden. In: Hitzenberger G, editor. Therapeutisches Drug Monitoring. Wien: Blackwell-MZV; 1994. p. 117-28.
34.
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.
35.
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 External link
36.
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 External link
37.
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 External link
38.
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 External link
39.
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 External link
40.
Hanberger H, Edlund C, Furebring M, G Giske C, Melhus A, Nilsson LE, Petersson J, Sjölin J, Ternhag A, Werner M, Eliasson E; Swedish Reference Group for Antibiotics. Rational use of aminoglycosides – review and recommendations by the Swedish Reference Group for Antibiotics (SRGA). Scand J Infect Dis. 2013 Mar;45(3):161-75. DOI: 10.3109/00365548.2012.747694 External link
41.
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 External link
42.
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 External link
43.
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 External link
44.
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 External link
45.
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 External link
46.
Hanrahan T, Whitehouse T, Lipman J, Roberts JA. Vancomycin-associated nephrotoxicity: A meta-analysis of administration by continuous versus intermittent infusion. Int J Antimicrob Agents. 2015 Sep;46(3):249-53. DOI: 10.1016/j.ijantimicag.2015.04.013 External link
47.
Hanrahan TP, Kotapati C, Roberts MJ, Rowland J, Lipman J, Roberts JA, Udy A. Factors associated with vancomycin nephrotoxicity in the critically ill. Anaesth Intensive Care. 2015 Sep;43(5):594-9.
48.
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
49.
Cristallini S, Hites M, Kabtouri H, Roberts JA, Beumier M, Cotton F, Lipman J, Jacobs F, Vincent JL, Creteur J, Taccone FS. New Regimen for Continuous Infusion of Vancomycin in Critically Ill Patients. Antimicrob Agents Chemother. 2016 Aug;60(8):4750-6. DOI: 10.1128/AAC.00330-16 External link
50.
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 External link
51.
Fachinformation Targocid®. Mai 2016.
52.
Matthews PC, Chue AL, Wyllie D, Barnett A, Isinkaye T, Jefferies L, Lovering A, Scarborough M. Increased teicoplanin doses are associated with improved serum levels but not drug toxicity. J Infect. 2014 Jan;68(1):43-9. DOI: 10.1016/j.jinf.2013.08.018 External link
53.
Nicolau DP. Pharmacodynamic optimization of beta-lactams in the patient care setting. Crit Care. 2008;12 Suppl 4:S2. DOI: 10.1186/cc6818 External link
54.
Li C, Du X, Kuti JL, Nicolau DP. Clinical pharmacodynamics of meropenem in patients with lower respiratory tract infections. Antimicrob Agents Chemother. 2007 May;51(5):1725-30. DOI: 10.1128/AAC.00294-06 External link
55.
Li C, Kuti JL, Nightingale CH, Nicolau DP. Population pharmacokinetic analysis and dosing regimen optimization of meropenem in adult patients. J Clin Pharmacol. 2006 Oct;46(10):1171-8. DOI: 10.1177/0091270006291035 External link
56.
McKinnon PS, Paladino JA, Schentag JJ. Evaluation of area under the inhibitory curve (AUIC) and time above the minimum inhibitory concentration (T>MIC) as predictors of outcome for cefepime and ceftazidime in serious bacterial infections. Int J Antimicrob Agents. 2008 Apr;31(4):345-51. DOI: 10.1016/j.ijantimicag.2007.12.009 External link
57.
Tam VH, Nikolaou M. A novel approach to pharmacodynamic assessment of antimicrobial agents: new insights to dosing regimen design. PLoS Comput Biol. 2011 Jan 6;7(1):e1001043. DOI: 10.1371/journal.pcbi.1001043 External link
58.
Tam VH, Schilling AN, Neshat S, Poole K, Melnick DA, Coyle EA. Optimization of meropenem minimum concentration/MIC ratio to suppress in vitro resistance of Pseudomonas aeruginosa. Antimicrob Agents Chemother. 2005 Dec;49(12):4920-7. DOI: 10.1128/AAC.49.12.4920-4927.2005 External link
59.
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 External link
60.
Bergeron MG, Nguyen BM, Gauvreau L. Influence of constant infusion versus bolus injections of antibiotics on invivo synergy. Infection. 1978;6(Suppl 1):S38-S46. DOI: 10.1007/BF01646064 External link
61.
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 External link
62.
Lavoie GY, Bergeron MG. Influence of four modes of administration on penetration of aztreonam, cefuroxime, and ampicillin into interstitial fluid and fibrin clots and on in vivo efficacy against Haemophilus influenzae. Antimicrob Agents Chemother. 1985 Sep;28(3):404-12. DOI: 10.1128/AAC.28.3.404 External link
63.
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 External link
64.
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 External link
65.
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 External link
66.
Garraffo R. Pharmacodynamic bases for continuous infusion of beta-lactams: optimisation of antibacterial activities against gram-negative bacilli. Antibiotiques. 2002;4:22-8.
67.
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 External link
68.
Kuzemko J, Crawford C. Continuous infusion of ceftazidime in cystic fibrosis. Lancet. 1989 Aug 12;2(8659):385. DOI: 10.1016/S0140-6736(89)90561-8 External link
69.
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 External link
70.
Lortholary O, Lefort A, Tod M, Chomat AM, Darras-Joly C, Cordonnier C; Club de Reflexion sur les Infections en Onco-Hématologie. 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 External link
71.
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 External link
72.
Nicolau DP, Lacy MK, McNabb J, Quintiliani R, Nightingale, CH. Pharmacokinetics of continuous and intermittent ceftazidime in intensive care unit patients with nosocomial pneumonia. Infect Dis Clin Pract (Baltim Md). 1999;81:45-9.
73.
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 External link
74.
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 External link
75.
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 External link
76.
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 External link
77.
Sermet-Gaudelus I, Hulin A, Ferroni A, Silly C, Gaillard JL, Berche P, Lenoir G. L’antibiotherapie dans la mucoviscidose. I. Particularites 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 External link
78.
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 External link
79.
Ambrose PG, Quintiliani R, Nightingale CH, Nicolau DP. Continuous vs. intermittent infusion of cefuroxime for the treatment of community-acquired pneumonia. Infect Dis Clin Pract (Baltim Md). 1998;7(9):463-70. DOI: 10.1097/00019048-199812000-00007 External link
80.
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 External link
81.
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 External link
82.
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 External link
83.
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 External link
84.
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 External link
85.
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 External link
86.
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 External link
87.
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 External link
88.
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 External link
89.
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 External link
90.
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.
91.
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 External link
92.
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 External link
93.
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 External link
94.
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 External link
95.
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 External link
96.
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 External link
97.
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 External link
98.
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 External link
99.
Lodise TP Jr, Lomaestro B, Drusano GL. Piperacillin-tazobactam for Pseudomonas aeruginosa infection: clinical implications of an extended-infusion dosing strategy. Clin Infect Dis. 2007 Feb;44(3):357-63. DOI: 10.1086/510590 External link
100.
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
101.
Shiu J, Wang E, Tejani AM, Wasdell M. Continuous versus intermittent infusions of antibiotics for the treatment of severe acute infections. Cochrane Database Syst Rev. 2013 Mar 28;(3):CD008481. DOI: 10.1002/14651858.CD008481.pub2 External link
102.
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
103.
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;17(6):R279. DOI: 10.1186/cc13134 External link
104.
Teo J, Liew Y, Lee W, Kwa AL. Prolonged infusion versus intermittent boluses of β-lactam antibiotics for treatment of acute infections: a meta-analysis. Int J Antimicrob Agents. 2014 May;43(5):403-11. DOI: 10.1016/j.ijantimicag.2014.01.027 External link
105.
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 12;192(11):1298-305. DOI: 10.1164/rccm.201505-0857OC External link
106.
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
107.
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
108.
Stahlmann R, Lode H. Kalkulierte parenterale Initialtherapie bakterieller Infektionen: Sicherheit und Verträglichkeit [Calculated parenteral initial treatment of bacterial infections: Safety and tolerabilty]. GMS Infect Dis. 2020;8:Doc16. DOI: 10.3205/id000060 External link
109.
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 External link
110.
Berthoin K, Le Duff CS, Marchand-Brynaert J, Carryn S, Tulkens PM. Stability of meropenem and doripenem solutions for administration by continuous infusion. J Antimicrob Chemother. 2010 May;65(5):1073-5. DOI: 10.1093/jac/dkq044 External link
111.
Carlier M, Stove V, Verstraete AG, De Waele JJ. Stability of generic brands of meropenem reconstituted in isotonic saline. Minerva Anestesiol. 2015 Mar;81(3):283-7.
112.
Adembri C, Fallani S, Cassetta MI, Arrigucci S, Ottaviano A, Pecile P, Mazzei T, De Gaudio R, Novelli A. Linezolid pharmacokinetic/pharmacodynamic profile in critically ill septic patients: intermittent versus continuous infusion. Int J Antimicrob Agents. 2008 Feb;31(2):122-9. DOI: 10.1016/j.ijantimicag.2007.09.009 External link
113.
Taubert M, Zander J, Frechen S, Scharf C, Frey L, Vogeser M, Fuhr U, Zoller M. Optimization of linezolid therapy in the critically ill: the effect of adjusted infusion regimens. J Antimicrob Chemother. 2017 Aug 1;72(8):2304-2310. DOI: 10.1093/jac/dkx149 External link
114.
Nierenberg DW. Drug inhibition of penicillin tubular secretion: concordance between in vitro and clinical findings. J Pharmacol Exp Ther. 1987 Mar;240(3):712-6.
115.
Appel GB. Aminoglycoside nephrotoxicity. Am J Med. 1990 Mar 23;88(3C):16S-20S; discussion 38S-42S. DOI: 10.1016/0002-9343(90)90082-O External link
116.
Fachinformation Zevtera. Mai 2015.
117.
Kim J, Ohtani H, Tsujimoto M, Sawada Y. Quantitative comparison of the convulsive activity of combinations of twelve fluoroquinolones with five nonsteroidal antiinflammatory agents. Drug Metab Pharmacokinet. 2009;24(2):167-74. DOI: 10.2133/dmpk.24.167 External link
118.
Marchbanks CR. Drug-drug interactions with fluoroquinolones. Pharmacotherapy. 1993 Mar-Apr;13(2 Pt 2):23S-28S.
119.
Schelleman H, Bilker WB, Brensinger CM, Han X, Kimmel SE, Hennessy S. Warfarin with fluoroquinolones, sulfonamides, or azole antifungals: interactions and the risk of hospitalization for gastrointestinal bleeding. Clin Pharmacol Ther. 2008 Nov;84(5):581-8. DOI: 10.1038/clpt.2008.150 External link
120.
Simkó J, Csilek A, Karászi J, Lorincz I. Proarrhythmic potential of antimicrobial agents. Infection. 2008 Jun;36(3):194-206. DOI: 10.1007/s15010-007-7211-8 External link
121.
Spriet I, Goyens J, Meersseman W, Wilmer A, Willems L, Van Paesschen W. Interaction between valproate and meropenem: a retrospective study. Ann Pharmacother. 2007 Jul;41(7):1130-6. DOI: 10.1345/aph.1K079 External link
122.
Nahata M. Drug interactions with azithromycin and the macrolides: an overview. J Antimicrob Chemother. 1996 Jun;37 Suppl C:133-42.
123.
Ludden TM. Pharmacokinetic interactions of the macrolide antibiotics. Clin Pharmacokinet. 1985 Jan-Feb;10(1):63-79. DOI: 10.2165/00003088-198510010-00003 External link
124.
Westphal JF. Macrolide-induced clinically relevant drug interactions with cytochrome P-450A (CYP) 3A4: an update focused on clarithromycin, azithromycin and dirithromycin. Br J Clin Pharmacol. 2000 Oct;50(4):285-95. DOI: 10.1046/j.1365-2125.2000.00261.x External link
125.
Schreiber DH, Anderson TR. Statin-induced rhabdomyolysis. J Emerg Med. 2006 Aug;31(2):177-80. DOI: 10.1016/j.jemermed.2005.08.020 External link
126.
Snaith A, Pugh L, Simpson CR, McLay JS. The potential for interaction between warfarin and coprescribed medication: a retrospective study in primary care. Am J Cardiovasc Drugs. 2008;8(3):207-12. DOI: 10.2165/00129784-200808030-00007 External link
127.
Robertson SM, Penzak SR, Pau AK. Drug interactions in the management of HIV infection. Expert Opin Pharmacother. 2005 Feb;6(2):233-53. DOI: 10.1517/14656566.6.2.233 External link
128.
Neuvonen PJ, Penttilä O. Interaction between doxycycline and barbiturates. Br Med J. 1974 Mar;1(5907):535-6. DOI: 10.1136/bmj.1.5907.535 External link
129.
Hasan SA. Interaction of doxycycline and warfarin: an enhanced anticoagulant effect. Cornea. 2007 Jul;26(6):742-3. DOI: 10.1097/ICO.0b013e318053387f External link
130.
Pichard L, Fabre I, Fabre G, Domergue J, Saint Aubert B, Mourad G, Maurel P. Cyclosporin A drug interactions. Screening for inducers and inhibitors of cytochrome P-450 (cyclosporin A oxidase) in primary cultures of human hepatocytes and in liver microsomes. Drug Metab Dispos. 1990 Sep-Oct;18(5):595-606.
131.
Fachinformation Ciclosporin Pro. April 2016.
132.
Agwuh KN, MacGowan A. Pharmacokinetics and pharmacodynamics of the tetracyclines including glycylcyclines. J Antimicrob Chemother. 2006 Aug;58(2):256-65. DOI: 10.1093/jac/dkl224 External link
133.
Berg ML, Estes LL, Dierkhising RA, Curran B, Enzler MJ. Evaluation of impact of statin use on development of CPK elevation during daptomycin therapy. Ann Pharmacother. 2014 Mar;48(3):320-7. DOI: 10.1177/1060028013514377 External link
134.
Lee JH, Lee SI, Chung CJ, Lee JH, Lee SC, Choi SR, Oh JN, Bae JY. The synergistic effect of gentamicin and clindamycin on rocuronium-induced neuromuscular blockade. Korean J Anesthesiol. 2013 Feb;64(2):143-51. DOI: 10.4097/kjae.2013.64.2.143 External link
135.
Rybak MJ, Albrecht LM, Boike SC, Chandrasekar PH. Nephrotoxicity of vancomycin, alone and with an aminoglycoside. J Antimicrob Chemother. 1990 Apr;25(4):679-87. DOI: 10.1093/jac/25.4.679 External link
136.
Paradelis AG, Triantaphyllidis C, Giala MM. Neuromuscular blocking activity of aminoglycoside antibiotics. Methods Find Exp Clin Pharmacol. 1980 Feb;2(1):45-51.
137.
Fanos V, Cataldi L. Amphotericin B-induced nephrotoxicity: a review. J Chemother. 2000 Dec;12(6):463-70. DOI: 10.1179/joc.2000.12.6.463 External link
138.
Mahatthanatrakul W, Nontaput T, Ridtitid W, Wongnawa M, Sunbhanich M. Rifampin, a cytochrome P450 3A inducer, decreases plasma concentrations of antipsychotic risperidone in healthy volunteers. J Clin Pharm Ther. 2007 Apr;32(2):161-7. DOI: 10.1111/j.1365-2710.2007.00811.x External link
139.
Blassmann U, Roehr AC, Frey OR, Koeberer A, Briegel J, Huge V, Vetter-Kerkhoff C. Decreased Linezolid Serum Concentrations in Three Critically Ill Patients: Clinical Case Studies of a Potential Drug Interaction between Linezolid and Rifampicin. Pharmacology. 2016;98(1-2):51-5. DOI: 10.1159/000445194 External link
140.
Antal EJ, Hendershot PE, Batts DH, Sheu WP, Hopkins NK, Donaldson KM. Linezolid, a novel oxazolidinone antibiotic: assessment of monoamine oxidase inhibition using pressor response to oral tyramine. J Clin Pharmacol. 2001 May;41(5):552-62. DOI: 10.1177/00912700122010294 External link
141.
Ramsey TD, Lau TT, Ensom MH. Serotonergic and adrenergic drug interactions associated with linezolid: a critical review and practical management approach. Ann Pharmacother. 2013 Apr;47(4):543-60. DOI: 10.1345/aph.1R604 External link
142.
Sakai Y, Naito T, Arima C, Miura M, Qin L, Hidaka H, Masunaga K, Kakuma T, Watanabe H. Potential drug interaction between warfarin and linezolid. Intern Med. 2015;54(5):459-64. DOI: 10.2169/internalmedicine.54.3146 External link
143.
Bolhuis MS, Panday PN, Pranger AD, Kosterink JG, Alffenaar JC. Pharmacokinetic Drug Interactions of Antimicrobial Drugs: A Systematic Review on Oxazolidinones, Rifamycines, Macrolides, Fluoroquinolones, and Beta-Lactams. Pharmaceutics. 2011 Nov 18;3(4):865-913. DOI: 10.3390/pharmaceutics3040865 External link
144.
Fachinformation Sivextro®. November 2016.