Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/8129387

Pharmacokinetic/Pharmacodynamic relationship of antimicrobial drugs used


in veterinary medicine

Article  in  Journal of Veterinary Pharmacology and Therapeutics · January 2005


DOI: 10.1111/j.1365-2885.2004.00603.x · Source: PubMed

CITATIONS READS

275 884

3 authors, including:

Quintin A Mckellar Sergio Sánchez Bruni


University of Hertfordshire National University of the Center of the Buenos Aires Province
184 PUBLICATIONS   6,110 CITATIONS    96 PUBLICATIONS   2,111 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Increase of Albendazole efficacy by the formulation of self-dispersible nanocrystals View project

Immuno-pharmacological alternatives for the control of infectious diseases in animal model View project

All content following this page was uploaded by Sergio Sánchez Bruni on 22 July 2020.

The user has requested enhancement of the downloaded file.


J. vet. Pharmacol. Therap. 27, 503–514, 2004. REVIEW

Pharmacokinetic/pharmacodynamic relationships of antimicrobial drugs


used in veterinary medicine

Q. A. MCKELLAR* McKellar, Q. A., Sanchez Bruni, S. F. Jones, D. G. Pharmacokinetic/pharma-


S. F. SANCHEZ BRUNI &  codynamic relationships of antimicrobial drugs used in veterinary medicine.
J. vet. Pharmacol. Therap. 27, 503–514.
D. G. JONES 
The rise in incidence of antimicrobial resistance, consumer demands and
*Royal Veterinary College, Hawkshead improved understanding of antimicrobial action has encouraged international
Campus, Hatfield, Hertfordshire, UK
 agencies to review the use of antimicrobial drugs. More detailed understanding
Moredun Research Institute,
of relationships between the pharmacokinetics (PK) of antimicrobial drugs in
Pentlands Science Park, Penicuik, UK
target animal species and their action on target pathogens [pharmacodynamics
(PD)] has led to greater sophistication in design of dosage schedules which
improve the activity and reduce the selection pressure for resistance in
antimicrobial therapy. This, in turn, may be informative in the pharmaceutical
development of antimicrobial drugs and in their selection and clinical utility.
PK/PD relationships between area under the concentration time curve from
zero to 24 h (AUC0–24) and minimum inhibitory concentration (MIC),
maximum plasma concentration (Cmax) and MIC and time during which
plasma concentrations exceed the MIC have been particularly useful in
optimizing efficacy and minimizing resistance. Antimicrobial drugs have been
classified as concentration-dependent where increasing concentrations at the
locus of infection improve bacterial kill, or time-dependent where exceeding the
MIC for a prolonged percentage of the inter-dosing interval correlates with
improved efficacy. For the latter group increasing the absolute concentration
obtained above a threshold does not improve efficacy. The PK/PD relationship
for each group of antimicrobial drugs is ‘bug and drug’ specific, although ratios
of 125 for AUC0–24:MIC and 10 for Cmax:MIC have been recommended to
achieve high efficacy for concentration-dependent antimicrobial drugs, and
exceeding MIC by 1–5 multiples for between 40 and 100% of the inter-dosing
interval is appropriate for most time-dependent agents. Fluoroquinolones,
aminoglycosides and metronidazole are concentration-dependent and beta-
lactams, macrolides, lincosamides and glycopeptides are time-dependent. For
drugs of other classes there is limited and conflicting information on their
classification. Resistance selection may be reduced for concentration-dependent
antimicrobials by achieving an AUC0–24:MIC ratio of greater than 100 or a
Cmax:MIC ratio of greater than 8. The relationships between time greater than
MIC and resistance selection for time-dependent antimicrobials have not been
well characterized.

Q.A. McKellar, Royal Veterinary College, Hawkshead Campus, Hatfield, Hertford-


shire AL9 7TA, UK. E-mail: principal@rvc.ac.uk

INTRODUCTION case of fish, in confined water spaces. Antimicrobial drug


prophylaxis and therapy have permitted maintenance of animals
Antimicrobial drugs have revolutionized human and veterinary in these husbandry systems without the adverse impact on
medicine through the provision of effective and inexpensive animal health and welfare which bacterial disease would have
means of treating, and in some circumstances preventing, otherwise inflicted. Furthermore, it has long been recognized
bacterial infectious disease. In veterinary medicine, intensified that antimicrobial drugs confer growth-promoting effects, even
production methods have led to an increase in the spread of when administered to healthy animals at dosage rates lower
disease where animals are kept in confined air spaces, or in the than those effective in treating clinical disease (Stokestad &

 2004 Blackwell Publishing Ltd 503


504 Q. A. McKellar et al.

Jukes, 1949, 1950). However, three major factors have now bacteria resistant to the administered antimicrobial or to other
encouraged a review of antimicrobial drug use in animals. First, antimicrobials demonstrating common resistance mechanisms
the inexorable selection of bacteria resistant to available drugs in (Blaser et al., 1987; Baquero et al., 1997).
animals and ‘more particularly’ in man (House of Lords, 1998; The utility of PK/PD information is demonstrable in the
World Health Organisation, 2001). Added to this there is a development of new antimicrobials, the more specific selection
growing body of evidence that bacteria selected for resistance in of appropriate antimicrobials from formularies, the design of
animals can be transmitted to man where they may cause optimal dosage strategies and the reduction in selection of
disease or transmit the genetic material responsible for resistance antimicrobial resistance (Gunderson et al., 2001). The devel-
to human pathogenic bacteria (MAFF, 1998; Smith et al., 1999; opment of new drugs for use in veterinary medicine has
Sáenz et al., 2000). Moreover, the acquisition of bacterial generally been based on dosage titration against model
resistance has outstripped the ability of pharmaceutical com- infections of bacterial disease supported by subsequent field
panies to produce new products with mechanisms of action investigations using the selected dosage (The Rules Governing
which overcome resistance, as is evidenced by the growing Medicinal Products in the European Union, 1999). Models
number of virtually untreatable bacterial infections in man utilizing the dose titration approach provide very useful but
(World Health Organisation, 2001). Nevertheless, it is clear that limited information as the only variable generally tested is
the majority of human resistant infections which are difficult to absolute dosage and the pattern of drug disposition is
treat are derived from bacteria selected for resistance in man and essentially fixed. Furthermore, a selected bacterial population
not in animals (World Health Organisation, 2001). is tested, with a fixed susceptibility to the antimicrobial. Dose
Secondly, the sophistication of the consumer in developed titration has been shown to discriminate clinical outcome
markets has meant that production systems which are demon- (mortality) poorly, whereas bacterial shedding may correlate
strably favourable to animal health and welfare, and where audit well with dose (Yancey et al., 1990). Field studies based solely
can confirm minimal chemotherapeutic intervention, are gain- on monitoring clinical outcome are flawed by the ‘Pollyanna
ing popularity. While this conflicts somewhat with major market effect’, whereby the efficacy of very good drugs is underesti-
forces whereby consumers effectively ‘buy cheap’, it is mated and the efficacy of poor drugs is overestimated. Thus,
undoubtedly a trend which will continue and will be spurred Marchant et al. (1992) demonstrated a clinical success of 89%
by increased affluence, traceability and information availability. in children with otitis media treated with an antimicrobial
This socio-economic pressure has already caused a rapid move which conferred 100% bacterial eradication. A clinical success
away from the use of antimicrobial drugs as growth promoters in of 74% was achieved with an antimicrobial conferring only
Europe, where they have been labelled as a crutch for poor 27% bacteriological cure, thus suggesting apparent success on
production methods but where the hard evidence implicating a treatment which was essentially no better that placebo.
them in human health problems is scant (Bates et al., 1994; Integration of the subtle differences in plasma disposition and
Klare et al., 1995; Report from the Commission on Antimicrobial bacterial sensitivities of closely related antimicrobials will allow
Feed Additives, Stockholm, 1997). Thirdly, there is the realiza- more specific selection of antimicrobials from formularies and
tion over the last decade that the relationship between the thus more effective individualization of treatment strategies
concentration of an antimicrobial in an animal or man and its (Gunderson et al., 2001). In veterinary medicine such subtle-
effect on the target pathogen is not simple. Thus, dosage ties in dosage selection are in their infancy but they are
strategies have been developed which may increase the efficacy clearly appropriate and achievable targets. Perhaps the
or reduce the selection pressure for resistance associated with the greatest utility for PK/PD integration is in the design of
antimicrobial for which they are tailored (Hyatt et al., 1995). optimal dosage strategies. Thus not only can the absolute
Pharmacokinetic/pharmacodynamic (PK/PD) interactions for amount of drug required to treat an infection be determined
antimicrobial drugs result in gross observable readouts in but the way in which the drug is delivered to produce a
administered animals including clinical improvement, growth plasma (or tissue) profile optimum to effect removal of the
promotion and adverse reactions (toxicity). Clinical improvement causal bacteria can also be characterized (Sarasola et al.,
is associated with the antimicrobial causing bacterial cell death or 2002). Integration of kinetic and dynamic characteristics of
inhibiting bacterial cell growth and thus facilitating its removal antimicrobial drugs has been used widely virtually since their
by the host immune system (Prescott & Walker, 2000). Growth first introduction. However more sophisticated PK/PD model-
promotion is also effected by the interaction between drug and ling (see later for details) is now being utilized to predict
bacteria and thus a change in bacterial populations and in their optimum dosages and may provide a versatile tool to
metabolic profile (Lev & Forbes, 1959; Eyssen & De Somer, 1967). determine optimum administration strategies. Optimization of
Adverse reactions may be the result of the antimicrobial affecting dosage strategy may impact on the selection of antimicrobial
commensal bacterial populations and thus overgrowth by resistance in two ways. It is implicit that the optimal strategy
pathogenic organisms and for instance the development of to remove the offending pathogen delivers the minimal
enterocolitis (English & Roberts, 1983). Alternatively, the appropriate dosage (effective to eradicate the target pathogen),
antimicrobial drug may directly affect the host animal causing thus commensals are exposed to the minimal selection
specific organ or system pathology (Yeary, 1975; Appel & Neu, pressure for resistance. Furthermore PK/PD parameters can
1977). The PK/PD interaction may also result in the selection of be determined which minimize the selection window (see later)

 2004 Blackwell Publishing Ltd, J. vet. Pharmacol. Therap. 27, 503–514


Pharmacokinetic/pharmacodynamic of antimicrobial drugs 505

for resistance associated with the target pathogen (Blondeau A clear relationship between Cmax:MIC, and favourable clinical
et al., 2001). response, has been demonstrated in human patients with Gram-
negative infections treated with aminoglycoside antibiotics. A
Cmax:MIC ratio of 2 resulted in favourable clinical response in
PK/PD RELATIONSHIPS about 50% of patients, whereas a ratio of 12 produced a positive
response in approximately 90% of patients (Moore et al., 1987).
In defining PK/PD relationships the most useful PK parameters A relationship has also been demonstrated between time above
are the area under the plasma concentration time curve (AUC) MIC and bacteriological cure in people with otitis media treated
from 0 time to 24 h, the maximum plasma concentration (Cmax) with beta-lactam antibacterial drugs. Bacteriological cure
achieved and time (T) during which concentrations exceed a increased from about 40 to 80% as the time above MIC
defined PD threshold. The most useful PD parameter is the increased from 10 to 100% of the interdosing interval (Craig &
minimum inhibitory concentration (MIC) (Hyatt et al., 1995). Andes, 1996). Each of the above studies was carried out in
The MIC is the lowest concentration of antimicrobial which models of human bacterial disease, or in human patients, and
inhibits the growth of the target bacteria and is generally each is complicated by the fact that there is covariance between
determined by growing the bacteria in doubling dilutions of the Cmax, AUC0–24 and T > MIC. In other words, as the maximum
antimicrobial for 24 h and assessing inhibition of growth by the concentration achieved in plasma increases so does the AUC and
resultant turbidity of the growth media (Walker, 2000a). The T > MIC.
sensitivity of this method can be improved by using overlapping In order to study the relationship between Cmax and
doubling dilutions (AliAbadi et al., 2003; AliAbadi & Lees, 2003) T > MIC in a representative animal model of respiratory
although this is impractical for routine screening of large disease, and to reduce the influence of covariance of PK/PD
numbers of organisms when determining MIC50 and MIC90 markers, a study utilizing bolus and infusion delivery of
(discussed later). danofloxacin was carried out. Danofloxacin was administered
However, as stated earlier the relationship between anti- to calves experimentally infected with Mannheimia (Pasteurella)
microbial concentration achieved in the target animal and the haemolytica by strategies conferring either a high Cmax:MIC
effect on the offending bacterial pathogen is not simple. Efficacy ratio (bolus injection) or a long T > MIC (i.v. infusion) but at
may depend upon achieving concentrations in plasma several the same absolute dose and producing similar AUC:MIC ratio
fold higher than the MIC of the pathogen (Blaser et al., 1987). in both treatments. The high Cmax:MIC ratio conferred
Alternatively it may be dependent on maintaining concentra- statistically better clinical and bacteriological cure than the
tions in plasma just above the MIC for a prolonged time (Craig, long T > MIC (Sarasola et al., 2002). The above studies and
1998a) and, for some drugs and pathogens, a combination of others (discussed later under individual classes) permit the
concentration and time of exposure may be important classification of antimicrobial drugs according to their opti-
(co-dependency). Furthermore, resistance may develop more mum activity. They may be concentration-dependent (time-
rapidly when insufficient concentration or time of exposure to independent) whereby the antimicrobial drugs kill bacteria to
the drug is achieved (Baquero & Negri, 1997; Blondeau et al., a greater extent at increasing exposure concentration. Alter-
2001). In quantitative terms the PK/PD parameters which have natively, they may be time-dependent (concentration-inde-
been most extensively investigated, and for which the most pendent) whereby bacteria are killed to the same extent once
robust information is currently available, are AUC0–24:MIC, a threshold concentration has been reached, but the antimi-
Cmax:MIC and T > MIC (Hyatt et al., 1995). crobial drugs are more effective when concentrations above
The relationship between AUC0–24:MIC and efficacy has been the MIC are maintained for a longer proportion of the
well demonstrated in a mouse model of a Gram-negative interdosing interval. These classifications are both antibacterial
bacterial infection in which treatment with fluoroquinolones and pathogen specific, and there may be drugs which are
reduced mortality from approximately 100% at a low co-dependent on concentration and time (Gunderson et al.,
AUC0–24:MIC ratio to almost zero at a ratio of >100 (Craig, 2001). A general classification of antimicrobial agents for
1998b). The AUC0–24:MIC ratio has also been shown to relate to which information is available is given in Table 1. It should be
bacteriological cure in seriously ill people treated with ciprofl- appreciated that many of the data utilized in classifying these
oxacin in which a ratio of >125 resulted in almost 80% agents are derived from human studies and that, for some
reduction in the number of patients from which pathogenic drugs, this information is conflicting. In this regard the fact
bacteria could be isolated (Schentag, 2000). In the same study, that some show apparent time and concentration dependence
when a ratio of <125 was achieved bacteriological cure was may be because of covariance of the PK parameters.
achieved in only about 50% of patients. Extrapolation of these
results to veterinary clinical situations should be made with
caution as models utilize neutropenic mice and seriously ill PHARMACOKINETIC ISSUES
human patients are also likely to be neutropenic. In infected but
immunocompetent domestic animals similar relationships al- Perhaps the most important PK consideration is where to
most certainly exist but the ratios are likely to differ as measure drug concentrations from which PK data are derived. It
antimicrobial drugs and immune mechanisms act in concert. is axiomatic that the antimicrobial should reach the locus of the

 2004 Blackwell Publishing Ltd, J. vet. Pharmacol. Therap. 27, 503–514


506 Q. A. McKellar et al.

Table 1. General classification of antimicrobial drugs for which infor- of the bacteria being tested (Walker, 2000a). Such factors should
mation is available on concentration or time-dependent killing activity be standardized according to internationally accepted method-
Concentration-dependent Time-dependent Co-dependent ologies (National Committee on Clinical Veterinary Standards,
NCCLS, 1999). Nevertheless, it is important to realize that MIC
Aminoglycosides Beta-lactams Beta-lactams* data may still be somewhat inaccurate as MICs are normally
Fluoroquinolones Macrolides Fluoroquinolones
determined using doubling dilutions raising the possibility of an
(except azithromycin)
almost 100% error between two sequential readouts. Running
Metronidazole Clindamycin Glycopeptides
(vs. anaerobes) Vancomycin parallel tests, with overlapping concentrations, can reduce the
error (Forrest et al., 1997; AliAbadi & Lees, 2002, 2003;
*In relation to reduction in resistance selection pressure. AliAbadi et al., 2003). Another important consideration is that

Some with anaerobic activity. MICs are routinely determined in a culture broth, and not in the
environment in which the bacteria grow in vivo such as blood,
extracellular fluid, intracellular environment, urine and milk, or
offending bacteria to be effective and, therefore, it could be in the presence of pus, exudate or detritus. The pH, aerobic or
assumed that tissue or cellular concentrations would be the most anaerobic environment and growth phase are also important in
appropriate for determining the required kinetic parameters. vivo and, in particular, in specific anatomical or physiological
Nevertheless, plasma concentrations have been shown to be the situations such as the mammary gland and urinary tract and in
best predictors of clinical success even for most tissue infections pathological situations, such as severe inflammation or absces-
(Schentag, 1989; Cars, 1997; Toutain et al., 2002). This is sation (Ziv & Rasmussen, 1975; Verklin & Mandell, 1977;
because extravascular fluid drug penetration is generally Strausbaugh & Sande, 1978; Luscombe & Nicholls, 1988).
complete and plasma concentration of nonprotein bound drug, In vitro PD assessment of activity may underestimate the
therefore, provides an accurate measure of tissue concentrations. activity of an antimicrobial drug achieved in vivo because of
Where a specific anatomical or pathological barrier exists, for the postantibiotic effect (PAE) and postantibiotic leukocyte
example in the central nervous system or in an abscess, or where enhancement (PALE) (Prescott & Walker, 2000). The PAE
bacteria are intracellular (e.g mycoplasma, chlamydophila) it describes the persistent suppression of bacterial growth
may be more appropriate to use concentrations derived from that following removal of an antimicrobial from the locus of the
site (Toutain et al., 2002). However, this may also be true for bacteria (Fuursted, 1987). The occurrence and magnitude of
drugs which preferentially accumulate inside cells, such as the PAE are dependent on the micro-organism, the type and
macrolides, for which plasma concentrations do not appear to concentration of drug to which the micro-organism has been
correlate with observed efficacy (Gladue et al., 1989; Nightin- exposed, and the duration of exposure. The PAE is generally
gale, 1999). Of the available veterinary antimicrobials fluoro- longer in vivo than in vitro (Renneberg & Walder, 1989). The
quinolones, macrolides, lincosamides and trimethoprim display mechanisms of action of the PAE are varied and include, for
the greatest capacity for intracellular penetration. For these beta-lactams, the length of time that the organism takes to
agents intracellular disposition may also be important as synthesize new penicillin-binding proteins. For aminoglycosides
penetration of lysosomes and phagolysosomes, as well as the length of time taken for the drug to dissociate from the
cytosolic distribution, may be required to affect bacteria within ribosome and to diffuse from its site of activity, and then for
specific subcellular loci (Tulkens, 1990). As only free drug is protein synthesis to recommence confers a PAE. Beta-lactams
pharmacologically active, plasma PK parameters should be express PAE for Gram-positive bacteria only (except for
corrected to reflect the extent of protein binding. However, for carbapenems which may also have a Gram-negative PAE).
practical purposes, this appears to be of importance only for Antimicrobial drugs which inhibit DNA or protein synthesis,
drugs which are highly protein bound with a free fraction of tend to impart long PAE against Gram-negative bacteria
<20% in plasma (Toutain et al., 2002) and may, therefore, be of (Prescott & Walker, 2000). Macrolides, fluoroquinolones and
relevance for clindamycin, cloxacillin, doxycycline and some aminoglycosides consistently show PAE for Gram-negative
sulphonamides (Hardman et al., 1996). Protein binding of drugs bacteria (Dudley, 1991; Craig, 1998b; Lutsar et al., 1998).
varies between animal species and it is important to consider The efficacy of spaced administrations of concentration-
binding in the target animal. dependent antimicrobials may be associated with their PAE.
The PALE describes the increased susceptibility to phagocytosis
and intracellular killing demonstrated by bacteria following
PHARMACODYNAMIC ISSUES exposure to an antimicrobial agent. Drugs, which produce the
greatest PAE also, tend to produce the greatest PALE.
Many PD factors affect antimicrobial activity and these may be
important both in vitro, where the drug activity is being assessed,
and in vivo, where activity is the intent. In determining MIC, or RESISTANCE SELECTION
other in vitro PD parameters, pH and presence of oxygen in the
environment of the organism are critical to its growth; as are the Conventional wisdom suggests that underdosing with an
bacterial load applied to the test system and the phase of growth antimicrobial drug rapidly selects for resistance and this has

 2004 Blackwell Publishing Ltd, J. vet. Pharmacol. Therap. 27, 503–514


Pharmacokinetic/pharmacodynamic of antimicrobial drugs 507

been confirmed using PK/PD markers where an AUC0–24:MIC


ratio of 100 or greater for all antimicrobial drugs was shown to
reduce selection for resistance (Thomas et al., 1998). More
detailed examination for the fluoroquinolones indicates that an
independent risk of resistance selection is conferred when an
AUC0–24:MIC > 100 or a Cmax:MIC > 8 are not achieved
(Blaser et al., 1987; Forrest et al., 1993). Furthermore, under-
exposure to one fluoroquinolone can confer resistance to the
whole class and it must be anticipated that similar risks will
apply to other drug groups sharing resistance mechanisms.
Some caution is required in extrapolating this ratio from
severely ill and presumably immunocompromised humans to
immunocompetent (but bacterially infected) animals. Repeated
exposure to suboptimal drug concentrations is now recognized Fig. 1. The selection window for an antimicrobial lies between the
as the single most important factor for the emergence of concentration at which wildtype bacteria survive exposure and the
resistance (Burgess, 1999). Optimal dosing strategies confer mutant prevention concentration which confers bacterial eradication.
appropriate drug concentration and time of exposure for the The size of the selection window is affected by a drug’s pharmacokinetics
as demonstrated for the drug (or formulation) represented by the bold
target pathogen which is likely to have a specific or narrow
and dashed lines (Baquero & Negri, 1997; Drlica, 2001; Catry et al.,
range of susceptibility (MIC) on which the dosage is based. 2003).
However, commensal bacteria may express quite different
sensitivities, which could result in their underexposure to the
delivered drug, and thus the selection of resistant populations, per day but does not indicate how that dose should be divided for
which could then transfer resistance genes to pathogens optimal efficacy.
(Baquero et al., 1997). Thus, optimal dosing strategies for
AUIC  MIC  Cl
specific pathogens may not be those which minimize selection Dose per day ¼ ; ð1Þ
in the whole animal. Using the example of the fluoroquinolo- fu  F  24h
nes, it is possible to conceptualize a concentration window, where AUIC ¼ AUC/MIC ratio for optimal efficacy; MIC ¼ mini-
exposure to which is likely to select for resistance. Resistance in mum inhibitory concentration; Cl ¼ clearance per day; fu ¼ free
fluoroquinolones may be conferred by two successive muta- fraction of drug in plasma (ignore if minimal binding); F ¼ bio-
tions, on bacterial gyrase enzymes (Hooper & Wolfson, 1993). availability.
The first mutation reduces susceptibility to fluoroquinolones When both PK and PD data are available, a sigmoidal Emax
without conferring full resistance. In a treated animal the relationship for bacterial count vs. ex vivo AUC0–24:MIC has been
plasma concentration of fluoroquinolone declines from the Cmax utilized. In this PK/PD modelling approach the dose can be
to concentrations below which even the wildtype bacteria are adjusted to provide a specific desired effect, which may be
unaffected. However, as drug concentrations fall there is a bacteriostasis, bactericidal activity or bacterial eradication as
period during which the first-step (gyrase) mutants have a shown in Eqns 2 & 3 and in Fig. 2 (AliAbadi & Lees, 2001,
selective advantage over the wildtype, and during which the 2002, 2003; AliAbadi et al., 2003). In fact, Eqn 2 yields the
population of these mutants increases. As this population grows same result as Eqn 1.
so does the probability of the second mutation occurring and
AUC24h =MIC ex vivo MIC90
thus the selection of double mutants which are fully resistant. DO ¼ DE   ; ð2Þ
AUC24h =MIC in vivo MICE
The concentration between that where the first-step mutants
are killed [the mutant prevention concentration (Blondeau where DO ¼ optimal dose; DE ¼ dose used experimentally;
et al., 2001)] and that at which wildtype bacteria survive is MIC90 ¼ MIC for 90% of organisms; MICE ¼ MIC for organism
termed the selection window (Fig. 1) (Catry et al., 2003). The used experimentally; AUC24 h/MICin vivo ¼ ratio provided by dose
disposition PK of an antimicrobial drug can affect the size of the DE in vivo; AUC24 h/MICex vivo ¼ ratio provided by required effect
selection window as can the administration strategy for its ex vivo; required effect (see Eqn 3 and Fig. 2) ¼ bacteriostasis,
delivery. An optimum PK/PD ratio can be determined to reduce bactericidal activity, elimination of bacteria.
the selection window and thus resistance development.
E ¼ Emax CNc =ECN50 þ CNc ; ð3Þ

where E ¼ antibacterial effect measured as change in log10 CFU;


OPTIMAL DOSAGE Emax ¼ maximum antibacterial effect; EC50 ¼ AUIC24 h of drug
that results in 50% of maximum antibacterial effect;
For drugs whose optimal efficacy can be related to the Cc ¼ AUIC24 h of drug in effect compartment (e.g. serum);
AUC0–24:MIC ratio, optimum dosage can be determined if N ¼ Hill coefficient steepness of AUIC24 h vs. effect (e.g. bacte-
appropriate PK and PD data are known as shown in Eqn 1 riostasis); ex vivo antibacterial effect ¼ AUIC24 h in serum, exu-
(Toutain et al., 2002). This equation provides an absolute dose date, transudate required for bacteriostasis, bactericidal activity,

 2004 Blackwell Publishing Ltd, J. vet. Pharmacol. Therap. 27, 503–514


508 Q. A. McKellar et al.

1 physiological and developmental states apparent in veterinary


Bacteriostatic
0
AUIC24h = 18 Observed
patients make population kinetics very attractive for PK/PD
Log cfu/mL difference

–1
studies as subpopulations requiring dosage alterations should be
Predicted
–2
Bactericidal identified.
–3
AUIC24h = 39
–4
Elimination
–5
AUIC24h = 90
–6 SPECIFIC AGENTS (FOR EXPERIMENTAL DATA SEE
–7 TABLE 2).
0 50 100 150 200 250 300
AUIC24h
Beta-lactams
Fig. 2. Sigmoidal Emax relationship for bacterial count vs. ex vivo
The available evidence indicates that beta-lactams are essentially
AUIC0–24 in a goat (AliAbadi & Lees, 2001).
time-dependent, whereby the time that the drug concentration
remains above the MIC is the greatest determinant of likely
and bacterial eradication; AUIC24 bacteriostasis E ¼ 0 (no efficacy (Eagle et al., 1950; Roosendaal et al., 1986). However,
change in bacterial count); AUIC24 bactericidal E ¼ )3(3 log or there is still considerable debate on how much above the MIC the
99.9% reduction in bacterial count); AUIC24 bacterial elimin- plasma concentration of the antimicrobial should be maintained,
ation, reduction to limit of detection 10 CFU/mL. and for what proportion of the interdosing interval the
A weighted AUC (WAUC) has also been applied in dosage concentration should be maintained at the desired level. There
optimization (Corvaisier et al., 1998). This incorporates the total is unlikely to be a single answer to each of these questions as
time for which the plasma drug concentrations exceed the MIC they are probably drug, pathogen and locus specific. As a rule of
(Eqn 4) and can be used for both concentration- and time- thumb, exceeding the MIC by 1–5 multiples for between 40 and
dependent drugs: 100% of the dosage interval is considered appropriate if not
AUCh  T > MICðhÞ highly specific (Gunderson et al., 2001). In deep-seated infections
WAUCðhÞ ¼ ; ð4Þ penetration of the bacterial locus may depend on high blood
MICðT > MICÞmaxðhÞ
concentrations as distribution is normally by simple diffusion
where WAUC(h) ¼ area under the concentration–time curve and thus affected by the drug concentration gradient. As a
weighted for total time at which plasma drug concentration consequence, distribution and activity of time-dependent drugs
exceeds the MIC (T > MIC)max ¼ 24 h. may therefore be associated with the AUC0–24 and Cmax (Lavoie
& Bergeron, 1985). Achieving concentrations several times
higher than the MIC may also be important to limit resistance
POPULATION PHARMACOKINETICS selection (see above).
In order to achieve the plasma concentration profile optimal for
Population PK methods were developed to study the kinetics of efficacy, several strategies may be adopted. It may be possible to
drugs in target human populations, generally during phase III select beta-lactams with longer terminal serum half-lives and this
clinical trials. As early drug development kinetic trials utilize, is a potential target for pharmaceutical companies producing
often normal, healthy, young, male volunteers they may not newer beta-lactams. Alternatively, they could be formulated in
identify inter- and intra-individual variation in the target repository dosage types such as the procaine or benzathine forms
population (Aarons, 1992; Sheiner & Ludden, 1992). Techni- Fig. 3. Although the benzathine formulations were conceived
ques were therefore developed which utilize sparse data sets before the time-dependent nature of beta-lactam activity was
typically with many patients but few observations per patient understood, by serendipity, they conferred the appropriate con-
(Samara & Granneman, 1997). In veterinary medicine popula- centration–time profiles of the active beta-lactams (Prescott,
tion PK have been used to study inter-individual variation and 2000). Concern has been expressed that concentrations of beta-
the influence of diverse pathophysiological factors in circum- lactams formulated with benzathine penicillin failed to exceed the
stances where sparse data per individual were available (Martin- MIC of relevant target pathogens and, although this is evidently
Jiminez et al., 1998; Whittem et al., 2000). It has also clear true, they were generally part of more complex formulations
utility for monitoring and predicting tissue residues (Martin- containing procaine also. It may be therefore that the release
Jiminez & Riviere, 1998; Whittem, 1999). The potential to profile associated with combined procaine and benzathine formu-
integrate population PK with dynamics for antimicrobial drugs lations did confer an appropriate plasma concentration–time
has recently been demonstrated in dogs administered marbofl- profile for target pathogens. Products containing benzathine
oxacin before cataract surgery. A limited number of aqueous penicillin are no longer authorized in the EU but are available
humor samples and blood samples were collected from 63 dogs elsewhere. It is also possible to extend the residence time of beta-
during surgery. Marbofloxacin concentrations were measured lactam antibacterials by co-administering them with agents such
and data analysed using population PK parameters. PD surro- as probenecid, which inhibit their active secretion in the kidney
gate markers were then used to predict in vivo antimicrobial tubules (Fig. 4; Sarasola & McKellar, 1992). The most obvious
activity (Regnier et al., 2003). The diversity of pathological, step, which the practitioner can take to improve the concentra-

 2004 Blackwell Publishing Ltd, J. vet. Pharmacol. Therap. 27, 503–514


Pharmacokinetic/pharmacodynamic of antimicrobial drugs 509

Table 2. Pharmacodynamic data from selected in vitro animal and human studies

Antibiotic Organism Parameter Source References

Beta lactams
Penicillin G Diplococcus pneumoniae T > MIC Mouse + rabbit Eagle et al., 1950
Ampicillin Escherichia coli AUC0–24:MIC IVPDM White et al., 1989
(to prevent resistance + regrowth)
Cephalosporins Sreptococcus pneumoniae T > MIC In vitro, murine Frimodt-Møller et al., 1986
Enterobacteriaceae T > MIC 60–70% Animal models Craig, 1995
Streptococcus spp. T > MIC 60–70% Animal models
Staphylococcus T > MIC 40–50% Animal models
Cephazolin E. coli T > MIC 100% Neutropenic murine Vogelman et al., 1988
Aminoglycosides
Gentamicin Pseudomonas aeruginosa, E. coli AUC0–24:MIC and T > MIC Neutropenic murine Vogelman et al., 1988
Gentamicin, Klebsiella pneumoniae AUC0–24:MIC and T > MIC Neutropenic murine Leggett et al., 1989
Netilmicin thigh and lung infection
Amikacin K. pneumoniae, E. coli, T > MIC – normal renal function Neutropenic murine Craig et al., 1991
P. aeruginosa
AUC0–24 – impaired renal function
Gentamicin E. coli, Klebsiella sp. Cmax:MIC > 10 Human retrospective Moore et al., 1987
Amikacin
Aminoglycosides Gram-negative bacteria Cmax:MIC > 8, T > 4 · MIC Human retrospective Deziel-Evans et al., 1986
Fluoroquinolones
Ciprofloxacin P. aeruginosa Cmax:MIC > 8 IVPDM Dudley et al., 1991
AUC0–24:MIC > 100 IVPDM Madaras-Kelly et al., 1996
S. pneumoniae AUC0–24:MIC > 35 IVPDM
S. aureus AUC0–24:MIC > 57 IVPDM Wright et al., 1998
S. pneumoniae, S. aureus Cmax:MIC 15–40 Serum ultrafiltrate Hoang et al., 1998
human Hyatt et al., 1994
P. aeruginosa Cmax:MIC20–50
Levofloxacin S. pneumoniae AUC0–24:MIC > 35 IVPDM Wright et al., 1998
Bacillus fragilis AUC0–24:MIC > 50 IVPDM Peterson et al., 1998
S. pneumoniae, S.aureus Cmax:MIC > 10 Human skin and Preston et al., 1998
UTI infections
Macrolides, glycopeptides, lincosamides and metronidazole
Erythromycin S. pneumoniae T > MIC 60% Neutropenic murine Vogelman et al., 1988
Clarithromycin S. pneumoniae T > MIC Neutropenic murine Ebert et al., 1991
Azithromycin S. pneumoniae AUC0–24:MIC Neutropenic murine Craig et al., 1992
Vancomycin S. aureus T > MIC  100% IVPDM Larsson et al., 1996
Vancomycin S. pneumoniae, S. aureus T > MIC, Cmax:MIC, AUC0–24:MIC Murine peritonitis Knudsen et al., 2000
Metronidazole Trichomonas vaginalis C:MLC > 10–25 IVPDM anaerobic Nix et al., 1995
Clindamycin S. pneumoniae T > MIC IVPDM Lewis et al., 1999

Table adapted from Gunderson et al. (2001).


IVPDM, in vitro pharmacodynamic model; MLC, minimum lethal concentrations.

9
tion–time profile of the beta-lactams, is to administer dosages more
8
Sodium penicillin
frequently. Administration by continuous infusion would provide
7
an even more accurate method for achieving optimum plasma
Units per mL

6
concentrations but is unlikely to prove practical except perhaps in
5
4
intensive care or during anaesthesia (Sarasola & McKellar, 1993).
3 Procaine penicillin Benzathine penicillin
2 Aminoglycosides
1
0 The aminoglycosides are concentration-dependent drugs for the
0 4 8 12 16 20 24 2 3 4 5 Gram-negative bacteria against which they are generally used
Hours Days
(Moore et al., 1987), although they may demonstrate some
Fig. 3. Concentration of penicillin in blood after administration of concentration-independent activity when used as adjunctive
equivalent doses of sodium, procaine and benzathine penicillins (Bogan, therapy against Gram-positive bacteria. The Cmax:MIC ratio has
1983). been shown to be the most useful PK/PD parameter for

 2004 Blackwell Publishing Ltd, J. vet. Pharmacol. Therap. 27, 503–514


510 Q. A. McKellar et al.

100 2.5
1
2
Amplicillin (µg/mL)

3
10 4
2

Plasma concentration (µg/mL)


1 5
6
7
0.1 1.5
Ampicillin + 
Ampicillin alone  Probenecid
If the MIC is:
0.01
Cmax/MIC = 2.4; AUC/MIC = 14.3
0 2 4 6 8 10 12 1 1
Time (h)

Cmax/MIC = 4.7; AUC/MIC = 28.6


0.5 0.5
Cmax/MIC = 9.3; AUC/MIC = 57
0.25
Cmax/MIC = 19.4; AUC/MIC = 119
0.12
0
0 5 10 15 20
Hours

Fig. 5. Cmax:MIC and AUC0–24:MIC ratios for orbifloxacin where


Cmax ¼ 2.33 lg/mL and AUC0–¥ ¼ 14.3 lgÆh/mL (Walker, 2000b).

ded to prevent resistance selection (Dudley, 1991; Thomas


Fig. 4. Concentrations of ampicillin in horses after the administration of et al., 1998). The impact which MIC has on the attainment of
an intravenous bolus dose (10 mg/kg) of ampicillin sodium with or desirable PK/PD ratios (AUC0–24:MIC of 125) is clearly
without probenecid (75 mg/kg). The MIC of representative equine demonstrated for orbifloxacin (Fig. 5), and difloxacin in dogs
pathogens and T > MIC are given (Adamson et al., 1985; Hirsh & Jang,
where it is apparent that optimal AUC0–24:MIC is achievable
1987; Sarasola & McKellar, 1992).
only for organisms with low MIC (0.12 lg/mL or less) with
recommended dosage (Walker, 2000b). When PK and PD data
predicting efficacy of aminoglycosides, and increasing the are available the PK/PD ratios can be determined (Tables 3 &
Cmax:MIC ratio correlates with clinical response. A Cmax:MIC 4). These data demonstrate the substantial differences which
ratio of >10 has been recommended for single daily dosing, experimental methodology can make in determination of PK
although some care should be taken to avoid toxicity. As toxicity and the impact that this and the sensitivity of the pathogen
to aminoglycosides is related to the trough concentration of drug makes to the derived PK/PD ratios. Few of the determined
it is likely that single daily dosing will allow concentrations to fall results indicate optimal ratios, and this raises the question of
during the trough period below the threshold which would cause whether optimal ratios derived from in vitro studies, or by
toxicity (Marra et al., 1996). This may not be the case for extrapolation from man, can be directly applied to domestic
animals with impaired renal function, although aminoglycosides animals. Some of the newer fluoroquinolones which have been
should generally be contraindicated in these animals, or should developed in human medicine, with good activity against
be administered with extended interdosing interval (Riviere, Gram-positive bacteria and some against anaerobes, have been
2000). A relationship between the Cmax:MIC ratio and the shown to possess concentration-independent activity against
emergence of resistance exists for aminoglycosides as it has been these pathogens (Ibrahim et al., 1999). Furthermore, fluoro-
shown for netilmicin, used to treat either Escherichia coli or quinolones have been shown to retain activity against
Staphylococcus aureus infections, that regrowth is prevented if the Gram-positive bacteria at lower AUC0–24:MIC ratios than for
Cmax:MIC ratio exceeds 8 (Blaser et al., 1987). Gram-negative bacteria (Lacy et al., 1999; Lister & Sanders,
1999; Peterson et al., 1999), which is paradoxical given their
generally lower MICs for Gram-negative compared with Gram-
Fluoroquinolones
positive bacteria.
A great deal of information is now available on the PK/PD
relationships for fluoroquinolones used in human medicine.
Macrolides, glycopeptides, lincosamides and metronidazole
They conform to concentration dependency against Gram-
negative bacteria and the Cmax:MIC ratio has been shown to Relatively little information is available on the macrolides,
have particular utility in determining their optimal activity glycopeptides, lincosamides and metronidazole and most must
(Drusano et al., 1993). This has subsequently been confirmed be extrapolated from either mouse models or in vitro
in a M. (Pasteurella) haemolytica model of respiratory disease in simulations. Macrolides are thought to be time-dependent
cattle (Sarasola et al., 2002). A Cmax:MIC ratio of >8 and an drugs for which the time greater than MIC most closely relates
AUC0–24:MIC ratio of >100 have been shown to prevent to efficacy. Erythromycin has been demonstrated to have
bacterial regrowth during treatment and are thus recommen- greatest activity against Streptococcus pneumoniae when the

 2004 Blackwell Publishing Ltd, J. vet. Pharmacol. Therap. 27, 503–514


Pharmacokinetic/pharmacodynamic of antimicrobial drugs 511

Table 3. AUC0–¥:MIC ratios for fluoroquinolones in dogs (Walker, 2000b)

Staphylococcus intermedius Escherichia coli Klebsiella pneumoniae

Dose* AUC MICz90 MIC90 MIC90


(mg/kg) (lgÆh/mL) (lgÆh/mL) AUC/MIC (h) (lgÆh/mL) AUC/MIC (h) (lgÆh/mL) AUC/MIC (h)

Enrofloxacin 5.0 15.7 0.12–0.5 131–31 0.03–0.125 523–126 0.06–0.12 262–130


Orbifloxacin 2.5 14.3 0.5 29 0.5 29 0.25 57
Difloxacin 5.0 14.5 1.0 15 0.25 58 0.5 29
Marbofloxacin 2.0 22.0 0.25 88

*All doses given per os.



AUC, area under the curve from 0–¥ (not 0–24 h).

MIC90 more than 20 isolates, range is given where data generated from more than one study.

Table 4. AUC0–24:MIC ratios for fluoroquinolones in dogs (Pirro et al., some concentration-dependent activity in an anaerobic in vitro
1999) model of Trichomonas vaginalis infection (Nix et al., 1995). The
lincosamide clindamycin is time-dependent in an in vitro
AUC0–24/MIC90 (h)
Dose AUC0–24 S. pneumoniae model (Lewis et al., 1999) although there is little
(mg/kg) (lgÆh/mL) S. intermedius E. coli information on the in vivo activity of these antimicrobials.
Enrofloxacin 5.0 8.7 70 146
Orbifloxacin 2.5 12.7 51 102
Difloxacin 5.0 9.3 37 73 CONCLUSIONS
Marbofloxacin 2.0 13.1 52 105
The outstanding utility of PK/PD integration is readily apparent
MIC90, n ¼ 25.
from the human medical literature. Whilst there may be
Pirro et al. (1999) – pharmacodynamic (MIC) data.
Heinen (2002) – pharmacokinetic (AUC) data. quantitative differences between the optimal ratios determined
for man and domestic animals it is certain that there are
time greater than MIC exceeds 60% of the interdosing period qualitative similarities. The laudable progress towards optimizing
(Vogelman et al., 1988). The macrolide, azithromycin, used in (and minimizing) the use of chemotherapeutics in animals
humans only, has been reported to possess concentration- means that PK/PD integration for antimicrobial drugs for
dependent activity when the AUC0–24:MIC ratio is optimized veterinary research is a priority and provides an opportunity
(Craig et al., 1992). There is debate regarding the activity of which should be embraced as such with enthusiasm, urgency
macrolides for which the plasma concentrations appear and vigour.
insufficient to confer good efficacy against bacteria where
even wildtype rarely have MICs which would make them
susceptible. One suggestion is that activity is related to high REFERENCES
concentrations achieved in cells and at tissue sites of infection.
Aarons, L. (1992) Population pharmacokinetics. International Journal of
Clarithromycin has been shown to achieve concentrations of
Clinical Pharmacology, Therapeutics and Toxicology, 30, 520–522.
one- to 30-fold higher in lung epithelial lining and 200–1000- Adamson, P.J.W.M., Wilson, W.D., Hirsh, D.C., Baggot, J.D. & Martin,
fold higher in alveolar macrophages than in plasma (Patel L.D. (1985) Susceptibility of equine bacterial isolates to antimicrobial
et al., 1996; Rodvoed et al., 1997). Release from these agents. American Journal of Veterinary Research, 46, 447–450.
intracellular sites may subject bacteria to prolonged exposure AliAbadi, F.S. & Lees, P. (2001) Pharmacokinetics and pharmacody-
appropriate for a time-dependent drug (Gladue et al., 1989). namics of danofloxacin in serum and tissue fluids of goats following
Whether the reservoir capacity of the cellular deposits of intravenous and intramuscular administration. American Journal of
Veterinary Research, 62, 1979–1989.
macrolides is sufficient to endorse this hypothesis has been
AliAbadi, F.S. & Lees, P. (2002) Pharmacokinetics and pharmacokinetic/
contested (Toutain et al., 2002). The macrolides have been pharmacodynamic integration of marbofloxacin in calf serum, exu-
shown to express extended PAE, which may support their dates and transudate. Journal of Veterinary Pharmacology and Ther-
activity (Craig, 1998b). apeutics, 25, 161–174.
The glycopeptide vancomycin has greatest in vitro activity AliAbadi, F.S. & Lees, P. (2003) Pharmacokinetic-pharmacodynamic
when the time greater than MIC is achieved for the whole integration of danofloxacin in the calf. Research in Veterinary Science,
interdosing interval (Larsson et al., 1996). Nevertheless, in a 74, 247–259.
AliAbadi, F.S., Landoni, M.F. & Lees, P. (2003) Pharmacokinetics (PK),
murine peritonitis model of S. pneumoniae and Staphylococcus
pharmacodynamics (PD), and PK-PD integration of danofloxacin in
aureus time greater than MIC, Cmax:MIC and AUC0–24:MIC were sheep biological fluids. Antimicrobial Agents and Chemotherapy, 47,
all shown to correlate with efficacy suggesting that for glyco- 626–635.
peptides there may be co-dependency on time and concentration Appel, G.B. & Neu, H.C. (1977) Nephrotoxicity of antimicrobial agents.
(Knudsen et al., 2000). Metronidazole has been shown to have New England Journal of Medicine, 296, 722–728.

 2004 Blackwell Publishing Ltd, J. vet. Pharmacol. Therap. 27, 503–514


512 Q. A. McKellar et al.

Baquero, F. & Negri, M.C. (1997) Strategies to minimise the development Dudley, M.N., Blaser, J., Gilbert, D., Mayer, K.H. & Zinner, S.H. (1991)
of antibiotic resistance. Journal of Chemotherapy, 9, 29–37. Combination therapy with ciprofloxacin plus azlocillin against Pseu-
Baquero, F., Negri, M.C., Morosini, M.I. & Blázquez, J. (1997) The anti- domonas aeruginosa: effect of simultaneous versus staggered adminis-
biotic selective process: concentration-specific amplification of low-le- tration in an in vitro model of infection. Journal of Infectious Diseases,
vel resistant populations. CIBA Foundation Symposia, 207, 93–111. 164, 499–506.
Bates, J., Jordens, J.Z. & Griffiths, D.T. (1994) Farm animals as a putative Eagle, H., Fleischman, R. & Musselman, A.D. (1950) Effect of schedule of
reservoir for vancomycin-resistant enterococcal infection in man. administration on the therapeutic efficacy of penicillin. American
Journal of Antimicrobial Chemotherapy, 34, 507–516. Journal of Medicine, 9, 280–299.
Blaser, J., Stone, B.B., Groner, M.C. & Zinner, S.H. (1987) Comparative Ebert, S., Rikardsdottir, S. & Craig, W.A. (1991) Pharmacodynamic com-
study with enoxacin and netilmicin in a pharamacodynamic model to parison of clarithromycin vs erythromycin. In Program and Abstracts of
determine importance of ratio of antibiotic peak concentrations to MIC the 31st Interscience Conference on Antimicrobial Agents and Chemotherapy.
for bacterial activity and emergence of resistance. Antimicrobial Agents p. 509. American Society for Microbiology, Washington, DC.
and Chemotherapy, 31, 1054–1060. English, P.B. & Roberts, M.C. (1983) Adverse reaction to antimicrobial
Blondeau, J.M., Zhao, X., Hanson, G. & Drlica, K. (2001) Mutant pre- agents in the horse. Veterinary Research Communications, 7, 207–210.
vention concentrations of fluoroquinolones for clinical isolates of Eyssen, H. & De Somer, P. (1967) Effects of Streptococcus faecalis and a
Streptococcus pneumoniae. Antimicrobial Agents and Chemotherapy, 45, filterable agent in growth and nutrient absorption in gnotobiotic
433–438. chicks. Poultry Science, 46, 323–333.
Bogan, J.A. (1983) Absorption and distribution. In Pharmacological Basis Forrest, A., Nix, D.E., Ballow, C.H., Goss, T.F., Birmingham, M.C. &
of Large Animal Medicine. Eds Bogan, J.A., Lees, P. & Yoxall, A.T. pp. Schentag, J.J. (1993) Pharmacodynamics of intravenous ciprofloxacin
3–22. Blackwell Scientific Publications, Oxford. in seriously ill patients. Antimicrobial Agents and Chemotherapy, 37,
Burgess, D.S. (1999) Pharmacodynamic principles of antimicrobial 1073–1081.
therapy in the prevention of resistance. Chest, 115, 195–233. Forrest, A., Ghodosh, S., Amantea, M.A., Collins, D.A. & Schentag, J.J.
Cars, O. (1997) Efficacy of beta-lactam antibiotics: integration of phar- (1997) Pharmacokinetics and pharmacodynamics of oral grapa-
macokinetics and pharamcodynamics. Diagnostic Microbiology and floxacin in patients with acute bacterial exacerbations of chronic
Infectious Disease, 27, 29–33. bronchitis. Journal of Antimicrobial Chemotherapy, 40, 45–57.
Catry, B., Laevens, H., Devriese, L.A., Opsomer, G. & De Kruif, A. (2003) Frimodt-Møller, N., Bentzon, M.W. & Thomson, V.F. (1986) Experi-
Antimicrobial resistance in livestock. Journal of Veterinary Pharmacol- mental infection with Streptococcus pneumoniae in mice: correlation of
ogy and Therapeutics, 26, 81–93. in vitro and pharmacokinetic parameters with in vivo effect for 14
Corvaisier, S., Maire, P.H., Bouvier d’Yvoire, M.Y., Barbaut, X., Bleyzac, cephalosporins. Journal of Infectious Diseases, 154, 511–517.
N. & Jelliffe, R.W. (1998) Comparisons between antimicrobial phar- Fuursted, K. (1987) Post-antibiotic effect of ciprofloxacin on Pseudomonas
macodynamic indices and bacterial killing as described by using the aeruginosa. European Journal of Clinical Microbiology, 6, 271–274.
Zhi model. Antimicrobial Agents and Chemotherapy, 42, 1731–1737. Gladue, R.P., Bright, G.M., Isaacson, R.E. & Newborg, M.F. (1989) In
Craig, W.A. (1995) Interrelationship between pharmacokinetics and vitro and in vivo uptake of azithromycin (CP-62,995) by phagocytic
pharmacodynamics in determining dosage regimens for broad-spec- cells: possible mechanisms of delivery and release at sites of infection.
trum cephalosporins. Diagnostic Microbiology and Infectious Disease, 22, Antimicrobial Agents and Chemotherapy, 33, 277–282.
89–96. Gunderson, B.W., Ross, G.H., Ibrahim, K.H. & Rotschafer, J.C. (2001)
Craig, W.A. (1998a) Choosing an antibiotic on the basis of pharmaco- What do we really know about antibiotic pharmacodynamics. Phar-
dynamics. Ear Nose and Throat Journal, 77 (Suppl. 6), 7–12. macotherapy, 21, 302s–318s.
Craig, W.A. (1998b) Pharmacokinetic/pharmacodynamic parameters: Hardman, J.G., Limbird, L.E., Molinoff, P.B., Ruddon, R.W. & Gilman,
rationale for antibacterial dosing of mice and men. Clinical Infectious A.G. (1996) Design and optimization of dosage regimens; Pharmaco-
Diseases, 26, 1–12. kinetic data. In: Goodman and Gilman’s The Pharmacological Basis of
Craig, W.A. & Andes, W. (1996) Pharmacokinetics and pharmacody- Therapeutics, 9th edn. Eds Hardman, J.G.G., Gilman, A. & Limbird, L.L.
namic of antibiotics in otitis media. Pediatric Infectious Diseases Journal, pp. 1712–1792. McGraw-Hill, New York.
15, 255–259. Heinen, E. (2002) Comparative serum pharmacokinetics of the fluoro-
Craig, W.A., Redington, J. & Ebert, G.C. (1991) Pharmacodynamics of quinolones enrofloxacin, difloxacin, marbofloxacin and orbifloxacin in
amikacin in vitro and in mouse thigh and lung infections. Journal of dogs after single oral administration. Journal of Veterinary Pharmacology
Antimicrobial Chemotherapy, 27 (Suppl. C), 29–40. and Therapeutics, 25, 1–5.
Craig, W.A., Rikardsdottir, S. & Wantanabe, Y. (1992) In vivo and in vitro Hirsh, D.C. & Jang, S.S. (1987) Antimicrobial susceptibility of bacterial
post-antibiotic effects (PAEs) of azithromycin. In Program and Abstracts pathogens from horses. Veterinary Clinics of North America: Equine
of the 32nd Interscience Conference on Antimicrobial Agents and Chemo- Practice, 3, 181–190.
therapy. p. 45.American Society for Microbiology, Washington, DC. Hoang, A.D., Peterson, M., Hovde, L.B., Wright, D. & Rotschafer, J.C.
Deziel-Evans, L.M., Murphy, J.E. & Job, M.L. (1986) Correlation of (1998) Investigation of AUC/MIC ratio as a generic predictor of
pharmacokinetic indices with therapeutic outcome in patients receiv- fluoroquinolone activity against Staphylococcus aureus using trovafl-
ing aminoglycosides. Clinical Pharmacy, 5, 319–324. oxacin, ciprofloxacin, sparfloxacin and levofloxacin in an in vitro
Drlica, K. (2001) A strategy for fighting antibiotic resistance. ASM News, pharmacodynamic model. In Program and Abstracts of the 98th General
67, 27–33. Meeting of the American Society for Microbiology, Washington, DC. p. 50.
Drusano, G.L., Johnson, D.E., Rosen, M. & Standiford, H.C. (1993) Hooper, D.C. & Wolfson, J.S. (1993) Mechanisms of bacterial resistance
Pharmacodynamics of a fluoroquinolone antimicrobial agent in a to quinolones. In Quinolone Antimicrobial Agents, 2nd edn. Eds Hooper,
neutropenic rat model of pseudomonas sepsis. Antimicrobial Agents and D.C. & Wolfson, J.S. pp. 97–118. American Society for Microbiology,
Chemotherapy, 37, 483–490. Washington, DC.
Dudley, M.N. (1991) Pharmacodynamics and pharmacokinetics of House of Lords (1998) Resistance to Antibiotics and Other Antimicrobial
antibiotics with special reference to the fluoroquinolones. American Agents. Select Committee on Science and Technology 7th Report,
Journal of Medicine, 91, 545–550. London. The Stationery Office, London.

 2004 Blackwell Publishing Ltd, J. vet. Pharmacol. Therap. 27, 503–514


Pharmacokinetic/pharmacodynamic of antimicrobial drugs 513

Hyatt, J.M., Nix, D.E. & Schentag, J.J. (1994) Pharmacokinetic and pharmacodynamic model. Antimicrobial Agents and Chemotherapy, 40,
pharmacodynamic activities against strains of Streptococcus pneumo- 627–632.
niae, Staphylococcus aureus and Pseudomonas aeruginosa for which MICs MAFF (1998) A Review of Antimicrobial Resistance in the Food Chain. A
are similar. Antimicrobial Agents and Chemotherapy, 38, 2730–2737. technical report for MAFF. MAFF Publications, London.
Hyatt, J.M., McKinnon, P.S., Zimmer, G.S. & Schentag, J.J. (1995) The Marchant, C.D., Carlin, S.A., Johnson, C.E. & Shurin, P.A. (1992) Mea-
importance of pharmacokinetic/pharmacodynamic surrogate markers suring the comparative efficacy of antibacterial agents for acute otitis
to outcome, focus on antibacterial agents. Clinical Pharmacokinetics, media: the ‘Pollyanna phenomenon’. Journal of Pediatrics, 120, 72–77.
28, 143–160. Marra, F., Partoni, N. & Jewesson, P. (1996) Aminoglycoside adminis-
Ibrahim, K.H., Hovde, L.B., Brown, G.H. & Rotschafer, J.C. (1999) Lev- trations as a single daily dose: an improvement to current practice or a
ofloxacin pharmacodynamics vs Streptococcus pneumoniae In Program repeat of previous errors? Drugs, 52, 344–376.
and Abstracts of the 39th Interscience Conference on Antimicrobial Agents Martin-Jiminez, T. & Riviere, J.E. (1998) Population pharmacokinetics in
and Chemotherapy. p. 50. American Society for Microbiology, veterinary medicine: potential use for therapeutic drug monitoring and
Washington, DC. prediction of tissue residues. Journal of Veterinary Pharmacology and
Klare, I., Heir, H., Claus, H., Böhma, G., Marin, S., Selfman, G., Haken- Therapeutics, 21, 167–189.
beck, R., Antanassova, V. & Wilte, W. (1995) Enterococcus faecium Martin-Jiminez, T., Papich, M.G. & Riviere, J.E. (1998) Population
strains with vanA-mediated high level glycopeptide resistance from pharmacokinetics of gentamicin in horses. American Journal of Veter-
animal food stuffs and faecal samples of humans in the community. inary Research, 59, 1589–1598.
Microbial Drug Resistance, 1, 265–272. Moore, R.D., Lietman, P.S. & Smith, C.R. (1987) Clinical response to
Knudsen, J.D., Fuursted, K., Raber, S., Espersen, F. & Frimodt-Moller, N. aminoglycoside therapy: importance of the ratio of peak concentration
(2000) Pharmacodynamics of glycopeptides in the mouse peritonitis to minimal inhibitory concentration. Journal of Infectious Diseases, 155,
model of Streptococcus pneumoniae and Staphylococcus aureus infection. 93–99.
Antimicrobial Agents and Chemotherapy, 44, 1247–1254. NCCLS (1999) Performance Standards for Antimicrobial Disk and Dilution
Lacy, M.K., Lu, W. Xu, X., Tessier, P.R., Nicolan, D.P., Quintiliani, R. & Susceptibility Tests for Bacteria Isolated from Animals; Approved Standard.
Nightingale, C.H. (1999) Pharmacodynamic comparisons of levo- National Committee on Clinical Veterinary Standards Document M31-
floxacin, ciprofloxacin and ampicillin against Streptococcus pneumoniae A. NCCLS, Wayne, PA.
in an in vitro model of infection. Antimicrobial Agents and Chemother- Nightingale, C.H. (1999) Determining outcomes in respiratory tract
apy, 43, 672–677. infections: pathogen, patient and antibiotic properties. Infections in
Larsson, A.J., Walker, K.J., Raddatz, J.K. & Rotschafer, J.C. (1996) The Medicine, 16, 37–41.
concentration-independent effect of monoexponential and biexponen- Nix, D.F., Tyrrell, R. & Muller, M. (1995) Pharmacodynamics of
tial decay in vancomycin concentrations on the killing of Staphylo- metronidazole determined by a time-kill assay for Trichomonas vagina-
coccus aureus under aerobic and anaerobic conditions. Journal of lis. Antimicrobial Agents and Chemotherapy, 39, 1848–1852.
Antimicrobial Chemotherapy, 38, 589–597. Patel, K.B., Xuan, D.W., Tessier, P.R., Russomanno, J.H., Quintilani, R. &
Lavoie, G.Y. & Bergeron, M.G. (1985) Influence of four modes of ad- Nightingale, C.H. (1996) Comparison of bronchopulmonary pharma-
ministration on penetration of aztreonam, cefuroxine and ampicillin cokinetics of clarithromycin and azithromycin. Antimicrobial Agents
into interstitial fluid and fibrin clots and on in vivo efficacy against and Chemotherapy, 40, 2375–2379.
Haemophilus influenzae. Antimicrobial Agents and Chemotherapy, 28, Peterson, M.L., Hovde, L.B., Wright, D.H., Hoang, A.D. & Rotschafer, J.C.
404–412. (1998) Pharmacodynamic outcome parameters as predictors for
Leggett, J.E., Fantin, B., Ebert, S., Totsuka, K., Vogelman, B., Calame, W., fluoroquinolones in the treatment of anaerobic infections. In Program
Mattie, H. & Craig, W.A. (1989) Comparative antibiotic dose-effect and Abstracts of the 98th General Meeting of the American Society for
relations at several dosing intervals in murine pneumonitis and thigh Microbiology, Washington, DC. p. 52.
infection models. Journal of Infectious Disease, 159, 281–292. Peterson, M.L., Hovde, L.B., Wright, D.H., Hoang, A.D., Raddatz, J.K.,
Lev, M. & Forbes, M. (1959) Growth response to dietary penicillin of Boysen, P.J. & Rotschafer, J.C. (1999) Fluoroquinolone resistance in
germ-free chicks and of chicks with a defined intestinal flora. British Bacteroides fragilis following sparfloxacin exposure. Antimicrobial Agents
Journal of Nutrition, 13, 78–84. and Chemotherapy, 43, 2251–2255.
Lewis, R.E., Klepsier, M.E., Ernst, E.J., Lund, B.C., Biedenbach, D.J. & Pirro, F., Edingloh, M. & Schmeer, N. (1999) Bactericidal and inhibitory
Jones, R.N. (1999) Evalutaion of low dose extended interval clin- activity of enrofloxacin and other fluoroquinolones in small animal
damycin regimes against Staphylococcus aureus and Streptococcus pathogens. Compendium on Continuing Education for the Practising
pneumoniae using a dynamic in vitro model of infection. Antimicrobial Veterinarian, 21 (Suppl), 19–25.
Agents and Chemotherapy, 43, 2005–2009. Prescott, J.F. (2000) Beta-lactam antibiotics: penam penicillins. In Anti-
Lister, P.D. & Sanders, C.C. (1999) Pharmacodynamics of levafloxacin microbial Therapy in Veterinary Medicine, 3rd edn. Eds Prescott, J.F.,
and ciprofloxacin against Streptococcus pneumoniae. Journal of Anti- Baggot, J.D. & Walker, R.D. pp. 105–133. Iowa State University Press,
microbial Chemotherapy, 43, 79–86. Ames, IA.
Luscombe, D.K. & Nicholls, P.J. (1988) Processes of drug handling by the Prescott, J.F. & Walker, R.D. (2000) Principles of antimicrobial drug
body. In Introduction to the Principles of Drug Design, 2nd edn. Ed. selection and use. In Antimicrobial Therapy in Veterinary Medicine, 3rd
Smith, H.J. pp. 24–313. Butterworth (Publishers Ltd), London. edn. Eds Prescott, J.F., Baggot, J.D. & Walker, R.D. pp. 88–104. Iowa
Lutsar, I., McCracken, G.H., Jr & Friedland, I.R. (1998) Antibiotic State University Press, Ames, IA.
pharmacodynamics in cerebrospinal fluid. Clinical Infectious Diseases, Preston, S.L., Drusano, G.L., Berman, A.L., Fowler, C.L., Chow, A.T.,
27, 1117–1127. Dornseif, B., Reichl, V., Natarajan, J. & Corrado, M. (1998) Pharma-
Madaras-Kelly, K.J., Ostergaard, B.F., Hovde, L.B. & Rotschafer, J.C. codynamics of levofloxacin: in a new paradigm for early clinical trials.
(1996) Twenty-four hour area under the concentration-time curve/ Journal of the American Medical Association, 279, 125–129.
MIC ratio as a generic predictor of fluoroquinolone antimicrobial effect Regnier, A., Concordet, D., Schneider, M., Boisramé, B. & Toutain, P.L.
by using three strains of Pseudomonus aeruginosa and an in vitro (2003) Population pharmacokinetics of marbofloxacin in aqueous

 2004 Blackwell Publishing Ltd, J. vet. Pharmacol. Therap. 27, 503–514


514 Q. A. McKellar et al.

humor after intravenous administration in dogs. American Journal of The Rules Governing Medicinal Products in the European Union (1999)
Veterinary Research, 64, 889–893. Guidelines, Veterinary Medicinal Products, General Efficacy, Environmental
Renneberg, J. & Walder, M. (1989) Post-antibiotic effect of imipenem, Risk Assessment, Vol. 7A. Office for Official Publications of the Euro-
norfloxacin and amikacin in vitro and in vivo. Antimicrobial Agents and pean Union, Luxembourg.
Chemotherapy, 33, 1714–1720. Thomas, J.K., Forrest, A., Bhaveni, S.M., Hyatt, J.M., Cheng, A., Ballow,
Report from the Commission on Antimicrobial Feed Additives, Stockholm L.H. & Schentag, J.J. (1998) Pharmacodynamic evaluation of factors
(1997) Antimicrobial Feed Additives, Vol. 106. Government Official associated with the development of bacterial resistance in acutely ill
Reports 1997: 132. p. 47. Ministry of Agriculture, Stockholm. patients during therapy. Antimicrobial Agents and Chemotherapy, 42,
Riviere, J.E. (2000) Renal impairment. In Antimicrobial Therapy in Vet- 521–527.
erinary Medicine, 3rd edn. Eds Prescott, J.F., Baggot, J.D. & Walker, R.D. Toutain, P.L., del Castillo, J.R.E. & Bousquet-Mélou, A. (2002) The
pp. 453–458. Iowa State University Press, Ames, IA. pharmacokinetic–pharmacodynamic approach to a rational dosage
Rodvoed, K., Gotfried, M., Danziger, L. & Servi, R. (1997) regimen for antibiotics. Research in Veterinary Science, 73, 105–114.
Intrapulmonary steady-state concentrations of clarithromycin and Tulkens, P.M. (1990) Accumulation and subcellular distribution of
azithromycin in healthy adult volunteers. Antimicrobial Agents and antibiotics in macrophages in relation to activity against intracellular
Chemotherapy, 41, 1399–1402. bacteria. In Ciprofloxacin in Pulmonology. Ed. Fass, R.J. pp. 12–20. W.
Roosendaal, R., Bakker-Wondenberg, I.A., van den Berghe-van Raffe, M. Zuckschwerdt Verlag, Munich.
& Michel, M.F. (1986) Continuous versus intermittent administration Verklin, R.M., Jr & Mandell, G.L. (1977) Alteration of effectiveness of
of ceflazidime in experimental Klebsiella pneumoniae pneumonia in antibiotics by anaerobiosis. Journal of Laboratory and Clinical Medicine,
normal and leukopenic rats. Antimicrobial Agents and Chemotherapy, 89, 65–71.
30, 403–408. Vogelman, B., Gudmundsson, S., Leggett, J., Turnridge, J., Fibert, S. &
Sáenz, Y., Zarazaga, M., Lantero, M., Gastaňares, M.J., Baquero, F. & Craig, W.A. (1988) Correlation of antimicrobial pharmacokinetic
Torres, C. (2000) Antibiotic resistance in Campylobacter strains isolated parameters with therapeutic efficacy in an animal model. Journal of
from animals, foods and humans in Spain in 1997–1998. Antimicro- Infectious Diseases, 158, 831–847.
bial Agents and Chemotherapy, 44, 267–271. Walker, R.D. (2000a) Antimicrobial susceptibility testing and interpret-
Samara, E. & Granneman, R. (1997) Role of population pharmacoki- ation of results. In Antimicrobial Therapy in Veterinary Medicine, 3rd
netics in drug development. A pharmaceutical industry perspective. edn. Eds Prescott, J.F., Baggot, J.D. & Walker, R.D. pp. 12–26. Iowa
Clinical Pharmacokinetics, 32, 294–312. State University Press, Ames, IA.
Sarasola, P. & McKellar, Q.A. (1992) Effect of probenecid on disposi- Walker, R.D. (2000b) Fluoroquinolones in antimicrobial therapy. In
tion kinetics of ampicillin in horses. Veterinary Record, 131, 173– Antimicrobial Therapy in Veterinary Medicine, 3rd edn. Eds Prescott, J.F.,
175. Baggot, J.D. & Walker, R.D. pp. 315–338. Iowa State University Press,
Sarasola, P. & McKellar, Q.A. (1993) Pharmacokinetics and applications Ames, IA.
of ampicillin sodium as an intravenous infusion in the horse. Journal of White, C.A., Toothaker, R.D., Smith, A.L. & Slattery, J.J. (1989) In vitro
Veterinary Pharmacology and Therapeutics, 16, 63–69. evaluation of the detriments of bactericidal activity of ampicillin dosing
Sarasola, P., Lees, P., AliAbadi, F.S., McKellar, Q.A., Donachie, W., Marr, regimens against Escherichia coli. Antimicrobial Agents and Chemother-
K.A. Sunderland, S.J. & Rowan, T.G. (2002) Pharmacokinetic and apy, 33, 1046–1051.
pharmacodynamic profiles of danofloxacin administered by two dosing Whittem, T. (1999) Pharmacokinetics and milk discard times of pirli-
regimens in calves infected with Mannheimia (Pasteurella) haemolytica. mycin after intramammary infusion: a population approach. Journal of
Antimicrobial Agents and Chemotherapy, 46, 3013–3019. Veterinary Pharmacology and Therapeutics, 22, 41–51.
Schentag, J.J. (1989) Clinical significance of antibiotic tissue penetration. Whittem, T., Hogan, D., Sisson, D. & Cooper, T. (2000) The population
Clinical Pharmacokinetics, 16, 25–31. pharmacokinetics of digoxin in dogs with heart disease. Journal of
Schentag, J.J. (2000) Clinical pharmacology of the fluoroquinolones Veterinary Pharmacology and Therapeutics, 23, 261–263.
studies in human dynamic/kinetic models. Clinical Infectious Diseases, World Health Organisation (2001) WHO Global Strategy for Containment
2000 (Suppl. 2), S40–S44. of Antimicrobial Resistance, Vol. 1211. WHO, Geneva.
Sheiner, L.B. & Ludden, T.M. (1992) Population pharmacokinetics/ Wright, D.H., Hovde, L.B., Peterson, M.L., Hoang, A.D. & Rotschafer, J.C.
dynamics. Annual Review of Pharmacology and Toxicology, 32, 185–209. (1998) In vitro evaluation of pharmacodynamic parameters for three
Smith, K.F., Besser, J.M., Hedberg, C.W., Leano, F.T., Bender, J.B., fluoroquinolones against Streptococcus pneumoniae. In Program and
Wickland, J.H., Johnson, B.P., Moore, K.A. & Osterholm, M.T. (1999) Abstracts of the 98th General Meeting of the American Society for
Quinolone-resistant Campylobacter jejuni infections in Minnesota, Microbiology, Washington, DC, p. 55.
1992–1998. New England Journal of Medicine, 340, 1525–1532. Yancey, R.J., Evans, R.A., Kratzer, D.D., Paulissen, J.B. & Carmer, S.G.
Stokestad, E.L.R. & Jukes, T.H. (1949) Further observations on the ani- (1990) Efficacy of ceftriofur hydrochloride for treatment of experi-
mal protein factor. Proceedings of the Society for Biological and Experi- mentally induced colibacillosis in neonatal swine. American Journal of
mental Medicine, 73, 523–528. Veterinary Research, 51, 349–353.
Stokestad, E.L.R. & Jukes, T.H. (1950) The multiple nature of the animal Yeary, R.A. (1975) Systemic toxic effects of chemotherapeutics agents in
protein factor. Journal of Biological Chemistry, 180, 647–654. domestic animals. Veterinary Clinics of North America, 5, 51–69.
Strausbaugh, L.J. & Sande, M.A. (1978) Factors influencing the therapy Ziv, G. & Rasmussen, F. (1975) Distribution of labelled antibiotics in dif-
of experimental Proteus mirabilis meningitis in rabbits. Journal of ferent components of milk following intramammary and intramuscular
Infectious Diseases, 137, 251–260. administration. Journal of Dairy Science, 58, 1637–1644.

 2004 Blackwell Publishing Ltd, J. vet. Pharmacol. Therap. 27, 503–514

View publication stats

You might also like