Review
Class-dependent relevance of tissue distribution in the interpretation of anti-infective pharmacokinetic/pharmacodynamic indices

https://doi.org/10.1016/j.ijantimicag.2010.01.023Get rights and content

Abstract

The pharmacokinetic/pharmacodynamic (PK/PD) indices useful for predicting antimicrobial clinical efficacy are well established. The most common indices include the time free drug concentration in plasma is above the minimum inhibitory concentration (MIC) (fT>MIC) expressed as a percent of the dosing interval, the ratio of maximum concentration to MIC (Cmax/MIC), and the ratio of the area under the 24-h concentration–time curve to MIC (AUC0–24/MIC). A single PK/PD index may correlate well with an entire antimicrobial class. For example, the β-lactams correlate well with the fT>MIC. However, other classes may be more complex and a single index cannot be generalised to the class, e.g. the macrolides. The rationale behind which PK/PD index best correlates with efficacy depends on several factors, including the mechanism of action, the microbial kill kinetics, the degree of protein binding and the degree of tissue distribution. Studies have traditionally emphasised the first two factors, whilst the significance of protein binding and tissue distribution is increasingly appreciated. In fact, the latter two factors may partially elucidate why the magnitude of reported target indices are not always as expected. For example, tigecycline and telithromycin are clinically efficacious with average serum concentrations below their MICs over a 24-h period. Therefore, to understand more fully the PK/PD relationship of antibiotics and to better predict the clinical efficacy of antibiotic dosing regimens, assessment of free drug concentrations at the site of action is warranted.

Introduction

Pharmacokinetic/pharmacodynamic (PK/PD) characterisation of antimicrobial agents has allowed a better understanding of why a particular dosing regimen achieves clinical success or failure. Using techniques first pioneered by Eagle et al. [1], [2] and further developed by Craig and co-workers [3], [4], [5], dose selection has become a much more sophisticated process over previous empirical methods. In vitro and in vivo experiments are used to define a relationship between drug concentration (pharmacokinetics) and effect (pharmacodynamics) and allow for a clear target to be identified so that efficacy is achieved in the clinical setting [6]. The PK/PD indices typically used for antimicrobials include the time the free drug concentration remains above the minimum inhibitory concentration (MIC) (fT>MIC) expressed as a percent of the dosing interval, the ratio of the maximum concentration and MIC (Cmax/MIC), and the ratio of the 24-h area under the concentration–time curve and MIC (AUC0–24/MIC) [3]. In vivo PK parameters are usually determined from serum or plasma drug profiles, whilst in vitro PD parameters commonly use culture media drug concentrations.

The index that best correlates with a particular antibiotic depends on several factors. One of these factors is the pattern of microbial kill exhibited by the compound, which is frequently referred to as either time-dependent killing or concentration-dependent killing. Antibiotics that display time-dependent killing typically reach a maximum effect at a concentration ca. 4× MIC [7], [8]. Once this maximum effect is reached, increasing the concentration does not increase the rate of bacterial death, as shown for ticarcillin in Fig. 1[7]. Antibiotics that display concentration-dependent killing produce an increasing effect as the concentration increases, as shown for tobramycin and ciprofloxacin in Fig. 1. However, categorising an agent's antimicrobial activity as time- or concentration-dependent is not always obvious, as shown in Fig. 2 where profiles for concentrations >4× MIC appear similar for all compounds [9].

Concentration-dependent antibiotics usually correlate efficacy with exposure and the MIC, i.e. the Cmax/MIC or the AUC0–24/MIC. The efficacy of time-dependent antimicrobials usually depends on fT>MIC as a percentage of the dosing interval. However, time-dependent antibiotics that display a prolonged post-antibiotic effect (PAE), i.e. the persistence of activity after removal of the drug or after concentrations drop below the MIC [10], [11], [12], often correlate well with the AUC0–24/MIC. Although the mechanisms of the PAE have been speculated, the PAE appears to depend on the drug, the pathogen and the infection model [10]. The term PAE is used collectively to explain residual effects of complex PK and PD processes that are not well defined.

Other factors that influence the significance of PK/PD indices for predicting clinical efficacy that are often overlooked include protein binding and tissue distribution. Recently, it has been stressed that only free concentrations should be taken into account in these indices as only free drug has the ability to exhibit a pharmacological effect [13], [14], [15]. Similarly, it follows that the most relevant concentration for antibiotic efficacy would be the concentration within the interstitial space fluid (ISF) of target tissues, as most infections are located outside the plasma or serum where drug concentrations are commonly monitored. The volume of distribution, and subsequently the degree of tissue distribution, varies greatly between antimicrobial classes, as shown in Table 1[16], [17], [18], [19], [20], [21], [22], [23], [24], [25], [26]. It is the goal of this paper to review the PK/PD relationship of various antimicrobial classes and to analyse this relationship by examining the mechanism of action, in vitro kill kinetics and free tissue concentrations. Additionally, the PAE will be discussed in some instances, as it may be used to account for additional uncharacterised PD effects.

Section snippets

β-Lactams

β-Lactam antibiotics, which include the penicillins, cephalosporins, carbapenems, penems and monobactams, work by inhibiting cell wall synthesis by binding to penicillin-binding proteins. The class displays time-dependent killing that is consistent with this mechanism of action. In general, the β-lactams correlate well with fT>MIC. A PAE is virtually absent with Gram-negative bacteria, except for the carbapenem subclass [11], [27], and only a modest PAE is reported with Gram-positive bacteria,

Aminoglycosides

The pharmacokinetics and pharmacodynamics of the aminoglycosides are fairly well characterised and appear to be consistent throughout the class. The mechanism of action of this class involves binding to the 30S ribosomal subunit and preventing protein synthesis [37]. Typically, antibiotics that inhibit protein synthesis or replication display a concentration-dependent killing pattern and a PAE, as is the case for the aminoglycosides [8], [38]. With these PD characteristics, the efficacy of

Fluoroquinolones

The fluoroquinolones prevent DNA replication by inhibiting type II topoisomerases, also called DNA gyrase, and topoisomerase IV [48], [49] and may also affect bacterial membranes [50]. Like the aminoglycosides, the fluoroquinolones display concentration-dependent killing [7] and both the AUC0–24/MIC and Cmax/MIC ratios are correlated with efficacy. However, several clinical studies have revealed that the AUC0–24/MIC ratio is the better index [51], [52], [53], [54]. The high target Cmax/MIC

Oxazolidinone

Linezolid is the only approved oxazolidinone on the market and has activity against several resistant pathogens, including meticillin- and vancomycin-resistant Staphylococcus aureus, vancomycin-resistant enterococci and penicillin-resistant S. pneumoniae[60], [61], [62]. The mechanism of action of this class is inhibition of protein synthesis at the initiation stage by binding to the 50S subunit of the bacterial ribosome [63] and preventing the first amino acid (methionine)–tRNA complex from

Tetracyclines and glycylcycline

The pharmacokinetics and pharmacodynamics of the tetracyclines and the glycylcycline, tigecycline, are not fully understood. The mechanism of action of this antibiotic class is inhibition of protein synthesis by binding to the 30S subunit of the prokaryotic ribosome [20]. The tetracyclines display a time-dependent killing pattern and exhibit a considerable PAE [3], [70], [71], [72], [73]. A prolonged PAE, as observed with tigecycline [71], suggests that the best PK/PD index is the AUC0–24/MIC.

Macrolides, ketolides and azalides

The macrolides are characterised by a 14–16-member macrocyclic lactone ring. The mechanism of action involves reversible binding to the 50S ribosomal subunit and prevention of protein synthesis [77]. Although this type of mechanism usually results in a bactericidal agent, the macrolides can display bacteriostatic or bactericidal killing depending on the pathogen [78], [79]. Overall, this class mainly displays time-dependent killing [80]. As with the tetracycline/glycylcycline class, a residual

Glycopeptides

Like the β-lactams, glycopeptides also inhibit cell wall synthesis. This antibiotic class works by forming hydrogen bonds with bacterial cell wall intermediate peptides, thereby inhibiting peptidoglycan synthesis [99]. As characteristic for antibiotics that inhibit cell wall synthesis, the glycopeptides mainly display time-dependent killing in vitro [100], [101], [102], [103], [104]. However, oritavancin and dalbavancin have been shown to display bactericidal concentration-dependent activity in

Conclusion

PK/PD characterisation of antimicrobial agents allows for a much more rational approach to antimicrobial dosing than traditional empirical methods. PK/PD indices have been well established for antimicrobial agents but are not fully understood among all antibiotics and/or antibiotic classes. The rationale behind which PK/PD index best predicts efficacy and the required magnitude is dependent on several factors. As a general rule, antimicrobial agents that have a mechanism of action of inhibition

References (115)

  • T.P. Lodise et al.

    Pharmacodynamics of an 800-mg dose of telithromycin in patients with community-acquired pneumonia caused by extracellular pathogens

    Diagn Microbiol Infect Dis

    (2005)
  • M. Nguyen et al.

    Telithromycin: the first ketolide antimicrobial

    Clin Ther

    (2005)
  • H. Eagle et al.

    ‘Continuous’ vs. ‘discontinuous’ therapy with penicillin; the effect of the interval between injections on therapeutic efficacy

    N Engl J Med

    (1953)
  • H. Eagle et al.

    The bactericidal action of penicillin in vivo: the participation of the host, and the slow recovery of the surviving organisms

    Ann Intern Med

    (1950)
  • W.A. Craig

    Pharmacokinetic/pharmacodynamic parameters: rationale for antibacterial dosing of mice and men

    Clin Infect Dis

    (1998)
  • D. Andes et al.

    In vivo pharmacodynamic activity of the glycopeptide dalbavancin

    Antimicrob Agents Chemother

    (2007)
  • D. Andes et al.

    In vivo pharmacodynamics of a new oxazolidinone (linezolid)

    Antimicrob Agents Chemother

    (2002)
  • P.G. Ambrose et al.

    Pharmacokinetics–pharmacodynamics of antimicrobial therapy: it's not just for mice anymore

    Clin Infect Dis

    (2007)
  • W.A. Craig et al.

    Killing and regrowth of bacteria in vitro: a review

    Scand J Infect Dis Suppl

    (1990)
  • M.K. Lacy et al.

    The pharmacodynamics of aminoglycosides

    Clin Infect Dis

    (1998)
  • E.I. Nielsen et al.

    Semimechanistic pharmacokinetic/pharmacodynamic model for assessment of activity of antibacterial agents from time–kill curve experiments

    Antimicrob Agents Chemother

    (2007)
  • R.W. Bundtzen et al.

    Postantibiotic suppression of bacterial growth

    Rev Infect Dis

    (1981)
  • W.A. Craig

    Post-antibiotic effects in experimental infection models: relationship to in-vitro phenomena and to treatment of infections in man

    J Antimicrob Chemother

    (1993)
  • D.E. Nix et al.

    Effect of ertapenem protein binding on killing of bacteria

    Antimicrob Agents Chemother

    (2004)
  • S. Schmidt et al.

    Effect of protein binding on the pharmacological activity of highly bound antibiotics

    Antimicrob Agents Chemother

    (2008)
  • M. Zeitlinger et al.

    Plasma protein binding of fluoroquinolones affects antimicrobial activity

    J Antimicrob Chemother

    (2008)
  • D.S. Burgess et al.

    Comparison of β-lactam regimens for the treatment of Gram-negative pulmonary infections in the intensive care unit based on pharmacokinetics/pharmacodynamics

    J Antimicrob Chemother

    (2005)
  • D.J. Stalker et al.

    Pharmacokinetics and tolerance of single- and multiple-dose oral or intravenous linezolid, an oxazolidinone antibiotic, in healthy volunteers

    J Antimicrob Chemother

    (2003)
  • J. Turnidge

    Pharmacokinetics and pharmacodynamics of fluoroquinolones

    Drugs

    (1999)
  • G.G. Zhanel et al.

    The glycylcyclines: a comparative review with the tetracyclines

    Drugs

    (2004)
  • G. Muralidharan et al.

    Pharmacokinetics of tigecycline after single and multiple doses in healthy subjects

    Antimicrob Agents Chemother

    (2005)
  • R. Jain et al.

    The macrolide antibiotics: a pharmacokinetic and pharmacodynamic overview

    Curr Pharm Des

    (2004)
  • J.P. Shaw et al.

    Pharmacokinetics, serum inhibitory and bactericidal activity, and safety of telavancin in healthy subjects

    Antimicrob Agents Chemother

    (2005)
  • A. Leighton et al.

    Tolerability, pharmacokinetics, and serum bactericidal activity of intravenous dalbavancin in healthy volunteers

    Antimicrob Agents Chemother

    (2004)
  • G.R. Matzke et al.

    Clinical pharmacokinetics of vancomycin

    Clin Pharmacokinet

    (1986)
  • C.I. Bustamante et al.

    Postantibiotic effect of imipenem on Pseudomonas aeruginosa

    Antimicrob Agents Chemother

    (1984)
  • R.F. Parker et al.

    The action of penicillin on Staphylococcus: further observations on the effect of a short exposure

    J Bacteriol

    (1948)
  • B. Vogelman et al.

    In vivo postantibiotic effect in a thigh infection in neutropenic mice

    J Infect Dis

    (1988)
  • W.A. Craig et al.

    In vivo pharmacodynamics of ceftobiprole against multiple bacterial pathogens in murine thigh and lung infection models

    Antimicrob Agents Chemother

    (2008)
  • A.K. Majumdar et al.

    Pharmacokinetics of ertapenem in healthy young volunteers

    Antimicrob Agents Chemother

    (2002)
  • AstraZeneca. Merrem® I.V. prescribing information. AstraZeneca; 2007. http://www1.astrazeneca-us.com/pi/MerremIV.pdf...
  • Bhavnani S. Which PK/PD index for which drug? In: International Society of Antimicrobial Pharmacology William A. Craig...
  • J. Karjagin et al.

    Pharmacokinetics of meropenem determined by microdialysis in the peritoneal fluid of patients with severe peritonitis associated with septic shock

    Clin Pharmacol Ther

    (2008)
  • F.J. Legat et al.

    Penetration of piperacillin and tazobactam into inflamed soft tissue of patients with diabetic foot infection

    Antimicrob Agents Chemother

    (2005)
  • F. Tomaselli et al.

    Penetration of meropenem into pneumonic human lung tissue as measured by in vivo microdialysis

    Antimicrob Agents Chemother

    (2004)
  • E.J. Begg et al.

    Comparison of gentamicin dosing regimens using an in-vitro model

    J Antimicrob Chemother

    (1992)
  • A.D. Kashuba et al.

    Optimizing aminoglycoside therapy for nosocomial pneumonia caused by Gram-negative bacteria

    Antimicrob Agents Chemother

    (1999)
  • B. Vogelman et al.

    Correlation of antimicrobial pharmacokinetic parameters with therapeutic efficacy in an animal model

    J Infect Dis

    (1988)
  • J. Blaser et al.

    Comparative study with enoxacin and netilmicin in a pharmacodynamic model to determine importance of ratio of antibiotic peak concentration to MIC for bactericidal activity and emergence of resistance

    Antimicrob Agents Chemother

    (1987)
  • L.E. Bryan et al.

    Effects of membrane-energy mutations and cations on streptomycin and gentamicin accumulation by bacteria: a model for entry of streptomycin and gentamicin in susceptible and resistant bacteria

    Antimicrob Agents Chemother

    (1977)
  • Cited by (59)

    • Comparison of In Vivo Transportability of Anti-Methicillin-Resistant Staphylococcus aureus (MRSA) Agents Into Intracellular and Extracellular Tissue Spaces in Rats

      2021, Journal of Pharmaceutical Sciences
      Citation Excerpt :

      There is a possibility of alteration in the pharmacokinetic profiles of drugs in MRSA infection and/or inflammation. Under inflammatory condition, phagocyte extravasation increased and the pH decreased in ISF, thus altering the distribution of antimicrobial agents into the infected and inflammatory sites.48–50 In clinical patients with sepsis, the physiological changes lead to pharmacokinetic alterations of antimicrobial agents.51

    • Excessive lysosomal ion-trapping of hydroxychloroquine and azithromycin

      2020, International Journal of Antimicrobial Agents
      Citation Excerpt :

      The volume of distribution of azithromycin was reported to be 31 L/kg and the half-life was 68 h, despite a relatively rapid clearance of 630 mL/min [18]. Furthermore, the pharmacokinetics of azithromycin have been shown to be affected by the disease state [19]. In a preclinical investigation, azithromycin was injected into rats and interstitial concentrations were measured in both thighs using microdialysis.

    • Concentration-dependent plasma protein binding: Expect the unexpected

      2018, European Journal of Pharmaceutical Sciences
      Citation Excerpt :

      Plasma protein binding can influence the pharmacokinetics and/or pharmacodynamics of a drug, thereby modifying its pharmacological or toxicological effects, or both. The scientific basis for this modifying influence of the extent of protein binding, or more correctly the proportion of drug unbound in plasma, on the overall biological effects of a drug have been extensively elaborated and reviewed by many investigators including Wilkinson, Shand, Benet and Derendorf (Barbour et al., 2010; Benet and Hoener, 2002; Heuberger et al., 2013; Sansom and Evans, 1995; Schmidt et al., 2010; Schmidt et al., 2008; van Steeg et al., 2009; Wilkinson, 1987; Wilkinson and Shand, 1975; Zeitlinger et al., 2011). For many drugs, the unbound fraction in plasma is relatively constant across a range of clinically relevant concentrations because the molar concentration of unbound drug is low in comparison to the molar concentration of binding sites on the protein(s) to which the drug binds.

    View all citing articles on Scopus
    View full text