You are on page 1of 11

burns 37 (2011) 16–26

available at www.sciencedirect.com

journal homepage: www.elsevier.com/locate/burns

Review

Antibiotics and the burn patient

François Ravat a,*, Ronan Le-Floch b, Christophe Vinsonneau c, Pierre Ainaud d,


Marc Bertin-Maghit e, Hervé Carsin f, Gérard Perro g
the Société Française d’Etude et de Traitement des Brûlures (SFETB)
a
Centre des brûlés, Centre hospitalier St Joseph et St Luc, 20 quai Claude Bernard, 69007 Lyon, France
b
Centre des brûlés, Centre hospitalo-universitaire, Nantes, France
c
Centre des brûlés, Groupe hospitalier Cochin, Paris, France
d
Centre des brûlés, Hôpital de la Conception, Marseille, France
e
Centre des brûlés, Hôpital Edouard Herriot, Lyon, France
f
Centre des brûlés, Hôpital d’Instruction des Armées Percy, Clamart, France
g
Centre des brûlés, Hôpital Pellegrin-Tripode, Bordeaux, France

article info abstract

Article history: Infection is a major problem in burn care and especially when it is due to bacteria with
Accepted 13 October 2009 hospital-acquired multi-resistance to antibiotics. Moreover, when these bacteria are Gram-
negative organisms, the most effective molecules are 20 years old and there is little hope of
Keywords: any new product available even in the distant future. Therefore, it is obvious that currently
Antibiotics available antibiotics should not be misused. With this aim in mind, the following review was
Antibiotic therapy conducted by a group of experts from the French Society for Burn Injuries (SFETB). It
Burn examined key points addressing the management of antibiotics for burn patients: when
Burn patient to use or not, time of onset, bactericidia, combination, adaptation, de-escalation, treatment
Bacterial resistance duration and regimen based on pharmacokinetic and pharmacodynamic characteristics of
Pharmacokinetics these compounds. The authors also considered antibioprophylaxis and some other key
Pharmacodynamics points such as: infection diagnosis criteria, bacterial inoculae and local treatment. French
Dosage guidelines for the use of antibiotics in burn patients have been designed up from this work.
# 2009 Elsevier Ltd and ISBI. All rights reserved.

Contents

1. General considerations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
1.1. Introduction. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
1.2. Infection criteria . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
1.3. Dealing with local infection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
1.4. Role of bacterial population (inoculum) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 17
2. Time for onset of antibiotic therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
3. Bactericidal or bacteriostatic molecules? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
4. Association or monotherapy? . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
5. Adaptation of antibiotherapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18

* Corresponding author. Tel.: +33 478 61 89 25; fax: +33 478 61 88 77.
E-mail address: fravat@ch-stjoseph-stluc-lyon.fr (F. Ravat).
0305-4179/$36.00 # 2009 Elsevier Ltd and ISBI. All rights reserved.
doi:10.1016/j.burns.2009.10.006
burns 37 (2011) 16–26 17

6. Duration of antibiotherapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
7. Administration methods (dosage and rhythm of injection) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
7.1. Notions of pharmacokinetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
7.2. Notions of pharmacodynamics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
7.3. Regimen . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
8. Monitoring antibiotic concentrations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
9. Perioperative antibiotic prophylaxis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
10. Guidelines from the French Society for Burn Injuries (SFETB) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
10.1. Guidelines for antibiotic therapy. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
10.2. Guidelines for antibiotics prophylaxis. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 20
10.3. Practical use. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 21
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 24

A study conducted in the French burns centres during the 1.2. Infection criteria
summer 2006 pointed out that 19% of inpatients developed
an infection. Although these facts are comparable to those Diagnosis of infection in burn patient is not easy because
observed in intensive care units, infection is a major problem clinical (hyperthermia) and biological (hyperleukocytosis,
in burn care and especially when it is due to bacteria increased CRP levels, etc.) infection criteria are poorly
with hospital-acquired multi-resistance to antibiotics, relevant, especially in heavier-burn patients. Actually, a
which has been proved to be directly related to the serious burn triggers a systemic inflammatory response
consumption of antibiotics. Moreover, when these bacteria syndrome (SIRS), which mimics usual clinical and biological
are Gram-negative organisms, the most effective molecules signs of infection [6–9]. Therefore, deciding an antibiotic
are 20 years old and there is no hope to see any new molecule therapy based on such usual infection criteria may lead to the
available even in the far future. Therefore, it is obvious that prescription of useless antibiotics. This has led experts to
current antibiotics – still active – should not be overused or define the criteria of infection in burn patient. The retained
misused. criteria are based on those validated for intensive care patients
Thus, the French Society for Burn Injuries (SFETB) has with two particular characteristics, namely the general criteria
published guidelines for the use of antibiotics in the burn of infection and criteria for burn wound infection. Differenti-
patient. These guidelines were drawn up by a group of ation between skin infection and colonisation should be
experts from the SFETB. Analysis of the literature, synthesis pointed out, as the presence of germs on the wound does not
of the relevant items and drafting the guidelines were led by necessarily mean that it is infected (see Appendix 1).
three members of the group. The changes, definitive draft
and validation were approved collectively by all the experts, 1.3. Dealing with local infection
according to the levels of evidence-based medicine [1,2]. The
following text is the review of literature conducted by Antibiotic therapy prescribed to prevent infection of burned
the French group of experts prior to the elaboration of skin does not actually prevent it and even facilitates the
guidelines. emergence of multi-resistant bacteria [10]. Diffusion of
antibiotic in the burned skin is questionable and cannot
achieve bactericidal concentrations so that bacteria can grow
1. General considerations and develop resistances. However, local topical agents are
known to be efficient in preventing and treating burned skin
1.1. Introduction infections [7]. For these two reasons, local infection – that is,
without general symptoms of sepsis – should require only
 Antibiotics should be used by keeping mind that they target local treatment.
not the patient but the bacteria: the clinician should select
the molecule or molecules with the greatest antibacterial 1.4. Role of bacterial population (inoculum)
efficacy and attempt to supply a concentration at the
infection site high enough to kill targeted pathogens The probability of antibiotic-resistant mutant strains appear-
(bactericidia). ing within a bacterial population is not void. For example, with
 Antibiotics should not allow bacterial resistance to arise by Pseudomonas aeruginosa, the probability of a fluoroquinolone-
lowering the selection pressure and thereby limiting the resistant strain appearing is estimated at 10 6. The risk of
consequences of antibiotics use in terms of bacterial appearance of resistant strains is therefore directly related to
ecology. Controlling the prescription of antibiotics, espe- the size of the bacterial population (inoculum). With P.
cially by avoiding useless antibiotic therapy, is an excellent aeruginosa, if the size of the bacterial population exceeds
means to decrease emergence of bacterial resistance [3–5]. 106, the probability of a fluoroquinolone-resistant strain
 Antibiotics are only one of the means to fight infection. appearing becomes higher. One of the means of limiting this
Antibiotic therapy is part of global hospital strategy as well probability is to reduce the inoculum [11]. For some infectious
as hygiene, and others. diseases, the reduction of inoculum alone ensures recovery
18 burns 37 (2011) 16–26

without antibiotic therapy or becomes the main treatment probability of the combination is 10 12, which exceeds the
(this is especially the case of skin and soft-tissue infections usual inoculae sizes).
[12]). In burn patients, very significant inocula may be Some antibiotics should not be used in monotherapy due
observed during pneumonias and/or infection of burned skin. to their high selection risk (fosfomycin, fusidic acid, rifampi-
In these cases, reducing the inoculum has tremendous impact, cin and fluoroquinolones) [11]. Combination therapy is also
by clearing for pneumonia and debridement (or removal) of recommended against multi-resistant hospital bacteria to
infected burned tissues, respectively. avoid acquisition of new resistances, thereby maintaining
During bacterial colonisation of tissue, the shift from their sensitivity profile [11]. One should remember that the
colonisation to infection depends on three parameters, burn patient is immunocompromised and expresses a
namely the level of the bacterial inoculum, host defences number of factors altering the pharmacokinetics of anti-
and bacterial virulence [13–15]. Reducing the inoculum may biotics [25]; this means that the regimen of antibiotics should
therefore contribute to preventing infections. be altered when compared with that recommended in the
healthy volunteer [26]. Antibiotic therapy should be started
immediately and should be effective from the beginning
2. Time for onset of antibiotic therapy [11,12,16,17,19].
All these arguments are in favour of the use of antibiotic
In case of serious infection (i.e., poorly tolerated and/or life combinations for the management of serious bacterial
threatening), antibiotic therapy should be started within 6 h infections in burn patients.
following the diagnosis of infection [16,17] as delayed
antibiotic therapy increases mortality [17–19]. When the
infection is not very serious (i.e., well-tolerated and without 5. Adaptation of antibiotherapy
organ failure), initiation of antibiotic therapy can be delayed
until microbiological documentation. In serious undocument- Adaptation is a two-stage strategy [19,27,28]:
ed infection, bacteriological sampling should be performed
before starting antibiotic therapy [20], but without delaying it.  Initial clinical approach: start of empirical treatment when
As long as the infection is not documented, antibiotic infection is suspected.
therapy is empiric. Therefore, broad-spectrum molecules  Subsequent bacteriological approach: assessment of initial
should be chosen for maximum efficacy. Nevertheless, the treatment based on bacteriological documentation.
choice for empiric treatment depends on patient ecology,
ward ecology, length of stay, previous antibiotic therapy, Antibiotic therapy should be started immediately [16,17].
patient condition, and so on. Consequently, it is often started though bacteriological
documentation is lacking (empiric antibiotic therapy). This
empiric antibiotic therapy may be inappropriate, and is known
3. Bactericidal or bacteriostatic molecules? to increase mortality [17,29]. The antibiotic usually chosen has
a broad-spectrum activity (with multi-drug resistant strains
Burn patients exhibit immune deficiency, which is still selection risk) although bacteria involved are sensitive to
incompletely understood, and mainly affects cell-mediated narrower-spectrum antibiotics [30,31]. In these conditions,
immunity (lymphocytes, macrophages and neutrophils) [21]. any antibiotic therapy should be assessed after 48–72 h
In these conditions, antibiotic therapy will, by itself, probably [12,16,20], as soon as bacteriological results are available.
need to be effective, that is, without the help of host defences Antibiotic therapy will have to be adapted to the germ(s)
[22]. Moreover, the infections observed are often with heavy actually responsible for the infection.
inoculae (lung, wound). Bactericidal antibiotics will there help Shifting from broad-spectrum empiric antibiotic therapy to
to reduce inoculum. Lastly, in case of serious infection, a narrow-spectrum adapted strategy (guided by the antibio-
antibiotic therapy will need to be effective quickly. For all gram) is called de-escalation. It should be performed whenev-
these reasons, bactericidal molecules should be preferred. er possible [16,20,27,28,32]. De-escalation has two aims,
namely individual benefit (recovery of a patient) and collective
benefit (reducing selective pressure and source of bacterial
4. Association or monotherapy? resistance).
Three conditions are mandatory for de-escalation.
The literature does not provide powerful enough data to
recommend combination therapy rather than monotherapy  Bacteriological documentation available.
[23,24] except in particular cases. Nevertheless, combination  Antibiogram available (bacterial sensitivity to antibiotics
therapy has a number of theoretical advantages, namely established).
broader spectrum (useful in situations of empiric antibiotic  Improvement of clinical status after 72 h.
therapy), enhanced bactericidia (more important and with the
quickest bactericidal activity) and prevention of emergence of There are understandable limits:
resistant strains (especially when inoculum is heavy). The
probability of bacterial resistance to a combination of two  Reliability of bacteriological data (e.g., in the case of
molecules is the product of probability for each molecule (if ventilator-associated pneumonia, what type of sample
each molecule presents with a probability of 10 6, the should be chosen to confirm diagnosis?).
burns 37 (2011) 16–26 19

 How to assess the clinical evolution (e.g., which elements increase further (maximum bactericidal activity). With
should be used in pneumonia: hyperthermia? arterial blood these antibiotics, the determining factor is how long does
gases? imaging?)? the concentration surpass the MIC.

Discontinuation of antibiotic therapy considered useless 7.3. Regimen


may be likened to de-escalation [33].
The regimen depends on the pharmacodynamic character-
istics of the molecules [47,49]:
6. Duration of antibiotherapy
 Concentration-related antibiotics (aminoglycosides, fluoroqui-
Excepted in few particular situations related to the microor- nolones and fosfomycin) [34,50–54]. The parameter to
ganism and/or severe immune failure, a well-conducted consider is the inhibitory quotient (IQ), defined as the ratio
antibiotic therapy enables a quick inoculum decrease [34]. between the maximum concentration (Cmax) and the MIC.
Prolonged administration of antibiotics is often unjustified Administration is intermittent; the interval between two
and leads to an increase in selective pressure. Following administrations depends on the elimination half-life of the
several prospective randomised studies conducted in ventila- molecule; it should not exceed 3 times this half-life [55].
tor-associated pneumonia [32,35,36], antibiotic therapy lasting Consequently, regimen of aminoglycosides is a single daily
7–8 days is recommended unless treatment provided initially dose (SDD) while, with fluoroquinolones, it consists of
was adequate. However, in P. aeruginosa-related infections, a several daily injections (ciprofloxacin 3–4 daily, ofloxacin 3
longer duration is probably necessary [35]. In that case, it is daily, pefloxacin 2 daily and levofloxacin 2–3 daily).
recommended not to exceed 15 days of antibiotic therapy [37]  Time-related antibiotics (beta-lactams, glycopeptides) [45,56–
or discontinue it after 48–72 h of apyrexia (or disappearance of 58]. Bactericidal activity is slow and poorly related to
signs that led to diagnosis of infection). concentration. The predictive parameter of therapeutic
success is the time during which the antibiotic serum
concentrations are above the MIC. The aim is therefore to
7. Administration methods (dosage and reach serum concentrations exceeding the MIC 100% of the
rhythm of injection) time. There are several methods to achieve this, namely
using molecules with a long half-life, shortening the time
7.1. Notions of pharmacokinetics period between the two injections or using continuous
intravenous infusion. Continuous infusion regimen seems
In intensive care and burn patients, all pharmacokinetics to be the optimal choice because it appears to provide more
parameters (absorption, distribution, metabolism and excre- stable serum levels in burn patients [59–61]. Continuous
tion) of many classes of drugs, including antimicrobials, are infusion needs the injection of a loading dose to achieve
altered, with significant intra-individual variations, and have levels above MIC within a reasonable time period. The
been documented over the past 30 years [26,38–44]. The main loading dose depends on the molecules used [12,16].
clinical consequence is a drop in tissue concentrations. It has
also been demonstrated that low serum concentrations of
antibiotics may lead to both therapeutic failures and emergence 8. Monitoring antibiotic concentrations
of resistant strains [45]. In addition, the resistance mechanisms
developed by bacteria lead to reduced efficacy of usual dosages Historically, antibiotic monitoring was suggested to prevent
of antibiotic (increase in minimum inhibitory concentration), toxicity. Nowadays, there is no doubt on its relevance in
and the combination of these factors brings about therapeutic guaranteeing, as soon as possible, the efficacy of the molecules
failures [46]. Consequently, the usually recommended regimen, used [38,56,62–64]. A new concept has been defined, based on
suitable for a healthy volunteer, is not recommended and both the interactions between the pharmacokinetics and pharma-
daily dosage and regimen should be altered in burn patients. codynamics of the drug, in which the key factors are serum
levels. The following two situations occur, depending on the
7.2. Notions of pharmacodynamics bactericidal activity of the antibiotic considered:

Bactericidal antibiotics may be divided into two groups,  Concentration-related activity: Achieving an IQ above 10 in
depending on their bactericidal activity profile [47,48]: most cases [34,50] and above 20 for P. aeruginosa (or similar
germ)-related infection is recommended. To determine the
 Concentration-related antibiotics. Bactericidal activity is IQ, both Cmax and MIC values are requested. MIC of targeted
proportional to the concentration obtained, that is, to the bacteria can be found by the bacteriology laboratory (E-test
administered dose: the higher the dose, the stronger the or other technique [65]). When MIC is unknown, the highest
bactericidal activity. The targeted pharmacokinetic objec- MIC in sensitive bacteria (low critical concentration (LCC))
tive is therefore the highest possible concentration, the only can be found by local scientific societies or CDC and should
limit being side effects. be the value used to calculate IQ.
 Time-related antibiotics. Bactericidal activity increases with To measure the maximum concentration (Cmax), sam-
the dose but reaches a plateau over which it does not pling should proceed 30 min after the end of the injection,
20 burns 37 (2011) 16–26

with the exception of ciprofloxacin, where a sample must be Brûlures in June 2008. They are available at www.sfetb.org or
taken as soon as infusion is completed, due to the fast www.brulure.org.
diffusion of the molecule. With aminoglycosides, concen-
tration measurement just before the second injection 10.1. Guidelines for antibiotic therapy
(through concentration) is recommended. It will help assess
the risk of toxicity.  No antibiotics without proven infections (level 1)
 Time-related antibiotics: When the continuous infusion regi-  A local infection requires a local treatment (level 1)
men is chosen, monitoring blood concentrations by steady However, when the local infection is associated with
state (Css) is mandatory. The experts recommend a Css general signs of infection, experts consider that the
between 4 and 5 times the MIC [59]. In some particular cases, infectious process is no longer purely local and that use
such as Pseudomonas infections, targeted Css could be 10 of an antibiotic may be indicated.
times the MIC [57]. The half-life of most (except for  Attempt to reduce the bacterial inoculum (level 5)
ceftriaxone, ertapénème, doripenem, céfépime and teico-  Antibiotics in serious infections is an emergency (level 1)
planin) is short. Provided that the steady state is reached  Use bactericidal antibiotics (level 5)
after 5 T1/2b, samples for Css monitoring can be obtained by  Know how to combine antibiotics (level 5)
the day following onset of the compound [59]. The steady Experts recommend the use of antibiotic combinations
state is more quickly achieved after the administration of a for the management of serious bacterial infections
loading dose. Furthermore, with time-related antimicro- during, at least, the first 72 h of the infection.
bials, initiating continuous infusion without loading dose  Adapt antibiotic therapy (level 1)
will probably lead to initial low concentrations, when Any antibiotic therapy should be assessed within 48–
inoculum and thereby risk of resistance selection is higher. 72 h, as soon as bacteriology is available. Antibiotic
In the continuous infusion regimen, the sample can be therapy should be adapted to the germ(s) responsible of
obtained for monitoring at any moment when the steady the infection.
state is thought to be achieved.  Practise de-escalation (level 5)
Anytime possible, shift broad-spectrum antibiotic for
narrow-spectrum one guided by the antibiogram.
 When to stop antibiotics therapy (level 5)
9. Perioperative antibiotic prophylaxis Antibiotic therapy lasting 7–8 days is recommended,
provided initial treatment was accurate.
In burn patients, antibiotic prophylaxis is only relevant during In P. aeruginosa infections antibiotherapy should not
the perioperative period as discussed above [10]. It aims to exceed 15 days.
fulfil three objectives [66,67]:  Respect the regimen (level 1)
With concentration-related antibiotics, administration is
 Reduce local inoculum and enhance grafts intake. intermittent and the interval between two injections
 Decrease wound-borne bacteraemia. should not exceed 3 times its half-life.
 Do not increase selection pressure. With time-related antibiotics, continuous infusion after
the loading dose should be used.
Antibiotic prophylaxis rules had been defined and spread In any burn patient, but those with kidney and/or liver
worldwide by several consensus conferences driven by other failure, higher dosage than usually recommended is
scientific societies [68]. These are: needed.
 Antibiotic monitoring is mandatory (level 2)
 Antibiotic prophylaxis should be started early enough before With concentration-related antibiotics, IQ over 10 should
surgery (approximately 1 h 30 min, usually just before be achieved (20 in P. aeuginosa infections).
anaesthesia induction). With time-related antibiotics, experts recommend to
 Half of the initial dose should be re-injected every 2 half- achieve a concentration at steady state between 4 and 5
lives of the molecule (for oxacillin, the re-injection should be times the MIC.
practised every 4 h).
 Antibiotic prophylaxis lasts at least 24 h and should never 10.2. Guidelines for antibiotics prophylaxis
exceed 48 h.
 If repeated injections are likely, continuous infusion after In burn patients, antibiotic prophylaxis could be used in
loading dose is possible, provided pharmacodynamics of the patients needing invasive surgery (excisions, flaps, etc.) but
molecule is suitable (time-related bactericidal molecules). not in dressing changes. The experts recommend [68] (level 5):

 No identified local infection and undefined bacterial target.


10. Guidelines from the French Society for Target methicillin-sensitive Staphylococcus, that is, oxa-
Burn Injuries (SFETB) cillin or cloxacillin (30 mg kg 1) or first-generation ceph-
alosporin (30 mg kg 1). In case of allergy, clindamycin
The following guidelines have been established according to should be used (10 mg kg 1).
the levels of evidence-based medicine [1,2]. They have been  No identified local infection but isolation of a pathogen on
validated by Société Française d’Etude et de Traitement des skin samples.
burns 37 (2011) 16–26 21

Target this one.  To the burns


 Documented or non-documented local infection. - Presence of pus
This is no longer prophylaxis, as the infection is ongoing. - Rapid cleansing and detachment
Administration should follow the usual rules about - Appearance of blackish marks (necrosis or haemor-
curative treatment and consider the identified or rhage)
presumed pathogen. - Unexplained conversion of a lesion from superficial to
 Application of inert skin substitute such as artificial dermis. deep (>48th hour)
In the absence of guidelines in the literature, the experts  To the donor graft sites
suggest if bacteria are isolated from skin samples, target - Presence of pus
them. Otherwise, target Staphylococcus. - Unexplained delayed healing
- Scab
10.3. Practical use  To the recipient grafts
- Presence of pus
For examples of regimen for some antibiotics in burn patients, - Lysis of grafts
see Appendix 2. - Necrosis of fat located under the graft
 To the healed areas
- Impetigo
Conflict of interest statement - Lysis of healed areas
(2) Bacteriological skin samples:
The authors have no financial interests to declare. They are used to find out the germ(s) involved.
More often, a simple swab is enough.
The biopsy is never systematic. It might be performed in
Appendix A difficult cases, followed by

General definitions  Microbiology examination


- Direct microscope examination with staining and
No predictive value of infection semi-quantitative measurement of germs
 In adults, presence of SIRS: two or more criteria of the four - Quantification of germs present per gram of tissue
below: after homogenate status: threshold of 105 CFU g 1 is
T (8C)>38.5 8C or <36 8C, retained as significant of the risk of haematogenous
Heart rate >90 bpm, dissemination
RR >20 per minute or capnia <25 mmHg,  An extemporaneous pathology examination after
Leucocytes >12 G or <4 G or >10% of immature forms. freezing enabling one to appreciate the level of
 Any burn patient >20% BSA and/or with smoke inhalation invasivity
injury is likely to present with SIRS criteria in any infectious - Colonisation: germs in the non-vascularised tissue
process. - Infection: germs in the living tissue and in contact with
vessels
(3) Summary:
Predictive values of infection Skin infection with general signs is considered as a
 Appearance of SIRS criteria in an adult whose lesions are systemic infection originated from skin.
<15% or 20% BSA and without any smoke inhalation injury.
 Two or more of the four criteria below in an adult with a burn
General signs + + + +
>BSA and/or with smoke inhalation injury:
Local signs + + + +
- T (8C) >39.5 8C or <35.5 8C,
Skin culture + + + +
- 50% basal HR, Skin infection + S + S + ? +
- 50% basal RR,
- or 100% of number of leucocytes,
- Haemodynamic failure with onset or enhancement of Fungal skin infection
catecholamine treatment. The diagnosis may be confirmed with a biopsy.

Herpes skin infection


Definitions of infection criteria for burned skin The diagnosis is clinical and may be confirmed with the
onset of a serology conversion and the presence of the virus in
The diagnosis of a skin infection is clinical. local samples.

Bacterial infection
(1) Positive local signs: Definitions of infection criteria for the other sites
 Presence of a local or loco-regional inflammatory
reaction Definitions per site (below) originate from those main-
 Unfavourable and unexpected local evolution tained by the CCLIN (French Central Comity for Struggle
22 burns 37 (2011) 16–26

Against Nosocomial Infections), the survey of the REA-  In case of central venous catheter (CVC)-associated bacter-
REACAT/RAISIN 2006 monitoring network. These definitions aemia, the following will be necessary:
are taken up in the ‘Guide de définition des infections - Positive blood culture in presence of a CVC (or withdrawn
nosocomiales’ – of the CCLIN Paris-Nord (1995), itself adapted within 48 h) in the absence of any other infection to the
from 1988 CDC definitions (CDC definitions for nosocomial same germ
infections, Gardner JS, Jarvis WR, Emori TG, et al., Am J Infect - AND one of the following criteria:
Contl 1988;16:128–40.) and the 1992 CSHPF (100 recommanda- - 103 CFU ml 1 of the same germ in quantitative culture of
tions pour la surveillance et la prévention des infections, BEH the catheter
June 1992) (100 guidelines for the monitoring and prevention of - Differential blood cultures with CVC/periph 5 or positiv-
infections). ity time period CVC/periph 2 h to the same germ.

Lung infection
Pneumonia Infection of central catheter
General signs + specific organ signs  microbiological cri- Local or general infection signs with all the following
teria: criteria:

 At least two chest X-rays, with new image of pneumonia or a  No blood culture to the same germ
change of a previous image  CVC culture 103 CFU ml 1
 At least one of the following signs (two in the absence of  Regression of the infectious syndrome within 48 h following
microbiological criteria): catheter withdrawal
- Appearance of purulent secretions or change in their
characteristics (colour, smell, consistency and Urinary tract infection
quantity)  Positive urine cytology (104 leucocytes ml 1)
- Dyspnoea, tachypnoea or cough (if not ventilated)  Asymptomatic (without general signs):
- Recent onset or worsened hypoxemia - Bladder catheter within past 7 days: urine culture
 Microbiological diagnosis (one of the following criteria): 105 CFU ml 1 if the patient has had urinary catheter
- BAL with a threshold of 104 CFU ml 1 or 5% of cells with insertion within the previous 7 days.
direct bacterial inclusion - In the absence of urinary catheterisation, no bladder
- Wimberley brush technique with a threshold of catheter: two consecutive urine cultures 105 CFU ml 1 to
103 CFU ml 1 the same germ(s) without the presence of more than two
- PDP with threshold of 103 CFU ml 1 species (no more than two different species)
- Quantitative bronchial aspiration with a threshold of  Symptomatic (general signs):
106 CFU ml 1 - Urine culture 105 CFU ml 1 (at maximum of two species)
- Blood culture or positive sample of bronchial tissue or
(histology) or pleural fluid in the absence of any other - or 103 CFU ml 1 with 104 leukocytes ml 1 and general
source of infection signs
- Specific examinations for viral pneumonia or pneumonia
due to particular microorganisms (Ag or Ac in bronchial
secretions, direct examinations or positive cultures of
bronchial secretions, urinary antigens or serology con-
versions) Appendix B

Bronchitis Methods of administration of some antibiotics in burn


General signs, cough, recent change in expectorations or patients.
bronchial aspirations, bronchial crepitations + isolation of
germ(s) in bronchial aspirations + no radiological sign of Beta-lactams
infection.
Beta-lactams are time-related bactericidal antibiotics.
Bacteraemia They should be administered by continuous infusion
General signs + positive blood culture(s): whenever possible [55,69–73].
 At least one blood culture (sample taken during a tempera- Continuous perfusion is immediately preceded by a
ture peak) positive to a germ known to be a pathogen loading dose, depending on the molecule [12,16], and is
 Two blood cultures in a maximum interval of 48 h usually equal to a single dose in repeated administration
(sample taken during a temperature peak) positive to one regimen.
of the following germs: coagulase-negative Staphylococcus, Continuous administration is sometimes rendered difficult
Bacillus sp., Corynebacterium sp., Propionibacterium sp., Micro- by poor stability of the molecule along time (clavulanic acid
coccus sp. and Acinetobacter sp. and imipenem), physical and chemical incompatibility of
 If bacteraemia is the consequence of another infection or is molecules and differences in the pharmacokinetics of a
responsible for secondary localisations, local signs of molecule and its co-factor (amoxicillin–clavulanic acid/imi-
infection will be associated. penem–cilastatin).
burns 37 (2011) 16–26 23

Penicillins Iimipenem
Continuous infusion
Cloxacillin and oxacillin 50–100 mg kg 1 daily
Continuous infusion Loading dose 10 mg kg 1
150–200 mg kg 1 daily Stability: 3 h 30 min at 25 8C
Loading dose 50 mg 1 kg 1 Targeted steady-state concentration is at 16–20 mg l 1 or 4–5 times
1
Targeted steady-state concentration is at 8–10 mg l or 4–5 times MIC. For germs at risk (specifically Acinetobacter baumanii) it is 32–
the MIC. 40 mg l 1 or 8–10 times MIC

Amoxicillin, amoxicillin + clavulanate Fluoroquinolones


Continuous infusion
150–200 mg kg 1 daily
Concentration-related bactericidal antibiotics active on
Loading dose 50 mg 1 kg 1
Targeted steady-state concentration is at 64–80 mg l 1 or 4–5 times
Gram-negative and Gram-positive bacteria [74]. These pro-
the MIC. ducts should be administered repeatedly. The frequency of
Amoxicillin: stable 6 h at 25 8C in NaCl solvent. In case of electric injections is determined by the half-life of the molecules [74].
syringe administration, replace the syringe every 6 h. Only few data about pharmacokinetics of fluoroquinolones
Amoxicillin–clavulanate: clavulanate tends to accumulate because such as ciprofloxacin being available, the experts can only
its half-life is longer than amoxicillin’s. In continuous infusion
provide guidelines for ciprofloxacin.
regimen, mix 50% amoxicillin and 50% as amoxicillin–clavulanate
and change syringe every 6 h. In repeated injections regimen, the
Ciprofloxacin [26,75]
recommended targeted concentration (16–20 mg l 1) is a trough
Repeated administration
concentration.
3–4 injections daily
10–20 mg kg 1 per injection (total dose 30–80 mg kg 1 daily)
Carboxy- and Ureidopenicillins Infuse for 30 min. Sample for Cmax immediately at the end of the
injection (due to the very quick diffusion time of the molecule)
Ticarcillin, ticarcillin + clavulanic acid Target (Cmax): >30 mg l 1 (optimal = 40 mg l 1) or >10 times the
Continuous infusion MIC
150–200 mg kg 1 daily Beware of occult water administration (0.5 ml per mg ciproflox-
Loading dose 50 mg 1 kg 1 acin)
Targeted steady-state concentration 64–80 mg l 1 or 4–5 times the
MIC
Ticarcillin: stable for 24 h at 25 8C Aminoglycosides
Ticarcillin–clavulanic acid: stable for 6 h at 25 8C. Clavulanate tends to
accumulate because its half-life is longer than ticarcillin’s. In Concentration-related bactericidal antibiotics.
continuous infusion regimen, mix 50% ticarcillin and 50% ticarcil-
Single daily dose (SDD).
lin–clavulanate and change syringe every 6 h. In repeated injections
The dosage should be increased in burn patients [42].
regimen, the recommended target concentration (16–20 mg l 1) is a
trough concentration
Amikacin
30 mg kg 1 once a day
Piperacillin, piperacillin + tazobactam
Infuse for 60 min
Continuous infusion
Sample for peak concentration 30 min after the end of infusion
At least 200 mg kg 1 daily
Targeted peak value: >80 mg l 1 or >10 times the MIC
Loading dose 50 mg kg 1
Sample for trough concentration immediately before the second
There are no stability or accumulation problems. Targeted steady-
infusion
state concentration is reached at 64–80 mg l 1 or 4–5 times the MIC
Trough concentration < 5 mg l 1

Cephalosporins
Gentamicin, tobramycin and netilmicin
10 mg kg 1 once a day
Cefotaxime
Infuse for 60 min
Continuous infusion
Sample for peak concentration 30 min after the end of infusion
100–150 mg kg 1 daily
Targeted peak value: >20 mg l 1 or >10 times the MIC
Loading dose 25 mg kg 1
Sample for trough concentration immediately before the second
Stability: 3 h 30 min at 25 8C
1 infusion
Targeted steady-state concentration is at 16–20 mg l or 4–5 times
Trough concentration < 2 mg l 1
MIC

Ceftazidime
Continuous infusion Glycopeptides
100–150 mg kg 1 daily
Loading dose 25 mg kg 1
Administration in soft bags with volumetric pump is preferred due Vancomycin
to the risk of gas release, but continuous administration with electric Continuous infusion
syringe is possible 30 mg kg 1 per day
Targeted steady-state concentration is at 16–20 mg l 1 or 4–5 times Loading dose = 5 mg kg 1
MIC. For germs at risk (specifically ticarcillin-R P. aeruginosa), the Concentration at steady state: 20–30 mg l 1. Sample can be taken
targeted steady-state concentration is 32–40 mg l 1 or 8–10 times MIC any time, more than 12 h after infusion onset
24 burns 37 (2011) 16–26

Oxazolidinones management of severe sepsis and septic shock. Intensive


Care Med 2004;30:536–55.
[17] Kumar A, Roberts D, Wood KE, Light B, Parrillo JE, Sharma S,
a et al. Duration of hypotension before initiation of effective
Linezolide
antimicrobial therapy is the critical determinant of survival
Spectrum limited to Gram-positive cocci. Bactericidal activity
in human septic shock. Crit Care Med 2006;34:1589–96.
limited to streptococci [76].
[18] Luna CM, Vujacich P, Niederman MS, Vay C, Gherardi C,
Continuous infusion (time-related bactericidal activity)
Matera J, et al. Impact of BAL data on the therapy and
1200 mg daily in adults
outcome of ventilator-associated pneumonia. Chest
Loading dose = 5 mg kg 1
1997;111:676–85.
Target concentration is 10 mg l 1 or 5 times the MIC
a [19] Iregui M, Ward S, Sherman G, Fraser VJ, Kollef MH. Clinical
Few data available by this day [77–79].
importance of delays in the initiation of appropriate
treatment for ventilator-associated pneumonia. Chest
references
2002;122:262–8.
[20] Kollef MH, Micek ST. Strategies to prevent antimicrobial
resistance in the intensive care unit. Crit Care Med
[1] Sackett DL, Straus SE, Richardson WS, Rosenberg W, 2005;33:1845–53.
Haynes RB. Evidence-based medicine: how to practice and [21] Munster AM. The immunological response and strategies
teach EBM, second ed., London: Churchill Livingstone; for intervention. In: Herndon DH, et al., editors. Total burn
2000. care. second ed., London: WB Saunders; 2002. p. 316–30.
[2] Delvenne C, Pasleau F. Comment résoudre en pratique un [22] Zhao X, Drlica K. Restricting the selection of antibiotic-
problème diagnostique ou thérapeutique en suivant une resistant mutants: a general strategy derived from
démarche EBM? Rev Med Liege 2000;55:226–32. fluoroquinolones studies. Clin Infect Dis 2001;33(Suppl.
[3] Kollef MH. Is there a role for antibiotic cycling in the 3):S147–56.
intensive care unit? Crit Care Med 2001;28(Suppl. 4): [23] Brun-Buisson C. Associations d’antibiotiques ou
N135–42. monothérapie?In: Actualités en Réanimation et Urgences.
[4] Goldmann DA, Weinstein RA, Wenzel RP, Tablan OC, Paris: Elsevier Masson; 2007. pp. 463–473.
Dumas RJ, Gaynes RP, et al. Strategies to prevent and [24] Safdar N, Handelsman J, Maki DG. Does combination
control the emergence and spread of antimicrobial antimicrobial therapy reduce mortality in Gram-negative
resistant microorganisms in hospitals: a challenge to bacteraemia? A meta-analysis. Lancet Infect Dis
hospital leadership. JAMA 1996;275:234–40. 2004;4:519–27.
[5] Haute Autorité de Santé. Recommandations [25] Blanchet B, Julien V, Vinsonneau C, Tod M. Influence of
professionnelles: stratégies d’antibiothérapie et prévention burns on pharmacokinetics and pharmacodynamics of
des résistances bactériennes en établissement de santé; drugs used in the care of burn patients. Clin Pharmacokinet
Avril 2008. www.has.fr. 2008;47:635–54.
[6] Latarjet J, Echinard C. Les brûlures. Paris: Masson; 1993. [26] Lesne-Hulin A, Bourget P, Ravat F, Goudin C, Latarjet J.
[7] Heggers JP, Hawkins H, Edgar P, Villareal C, Herndon D. Clinical pharmacokinetics of ciprofloxacin in patients with
Treatment of infection in burns. In: Herndon DH, et al., major burns. Eur J Clin Pharmacol 1999;55:515–9.
editors. Total burn care. second ed., London: WB Saunders; [27] Rello J, Vidaur L, Sandiumenge A, Rodriguez A, Gualis B,
2002. p. 120–69. Boque C, et al. De-escalation therapy in ventilator-
[8] Sherwood ER, Traber DL. The systemic inflammatory associated pneumonia. Crit Care Med 2004;32:2183–90.
response syndrome. In: Herndon DH, editor. Total burn [28] Hoffken G, Niederman MS. Nosocomial pneumonia: the
care. 3rd ed, Saunders-Elsevier; 2007. p. 293–309. importance of a de-escalating strategy for antibiotic
[9] Jeschke MG, Mlcak RP, Finnerty CC, Norbury WB, Gauglitz treatment of pneumonia in the ICU. Chest 2002;122:
GG, Kulp GA, et al. Burn size determines the inflammatory 21483–2196.
and hypermetabolic response. Crit Care Med [29] Dupont H, Mentec H, Sollet JP, Bleichner G. Impact of
2007;35(Suppl.):S519–23. appropriateness of initial antibiotic therapy on the
[10] Ugburo AO, Atoyebi OA, Oyeneyin JO, Sowemimo GOA. An outcome of ventilator-associated pneumonia. Intensive
evaluation of the role of systemic antibiotic prophylaxis in Care Med 2001;27:355–62.
the control of burn wound infection at the Lagos University [30] Kollef MH. Optimizing antibiotic therapy in the intensive
Teaching Hospital. Burns 2004;30:43–8. care unit setting. Crit Care 2001;5:189–95.
[11] Conférence d’experts de la SFAR (Société Française [31] Kollef MH. Gram negative bacterial resistance: evolving
d’Anesthésie et de Réanimation). Associations patterns and treatment paradigms. Clin Inf Dis
d’antibiotiques ou monothérapie en réanimation 2005;40(Suppl. 2):S85–88.
chirurgicale et en chirurgie; 1999. www.sfar.org. [32] Micek ST, Heuring TJ, Hollands JM, Shah RA, Kollef MH.
[12] Antibiothérapie probabiliste des états septiques, graves. Optimizing antibiotic treatment for ventilator-associated
Ann Fr Anesth Reanim 2004;23:1020–6. pneumonia. Pharmacotherapy 2006;26(2):204–13.
[13] Lepape A. Epidémiologie et écologie bactérienne des [33] Kollef MH, Kollef KE. Antibiotic utilization and outcomes
infections nosocomiales en réanimation. In: Martin C, Gouin for patients with clinically suspected ventilator-associated
F, editors. Infections et antibiothérapie en réanimation aux pneumonia and negative quantitative BAL culture results.
urgences et en chirurgie. Paris: Arnette; 2000. p. 427–39. Chest 2005;128:2706–13.
[14] Greene JN. The microbiology of colonization, including [34] Forrest A, Nix DE, Ballow CH, Goss TF, Birmingham MC,
techniques for assessing and measuring colonization. Schentag JJ. Pharmacodynamics of intravenous
Infect Contl Hosp Epidemiol 1996;17:114–8. ciprofloxacin in seriously ill patients. Antimicrob Agents
[15] Bonten MJ, Weinstein RA. The role of colonization in the Chemother 1993;37:1073–81.
pathogenesis of nosocomial infections. Infect Contl Hosp [35] Chastre J, Wolff M, Fagon JY, Chevret S, Thomas F, Wermert
Epidemiol 1996;17:193–200. D, et al. Comparison of 8 vs 15 days of antibiotic therapy for
[16] Dellinger RP, Carlet JM, Masur H, Gerlach H, Calandra T, ventilator associated pneumonia in adults: a randomized
Cohen J, et al. Surviving. Sepsis Campaign guidelines for trial. JAMA 2003;290:2588–98.
burns 37 (2011) 16–26 25

[36] Christ-Crain M, Stolz D, Bingisser R, Müller C, Miedinger D, [55] Schentag J. Pharmacokinetic and pharmacodynamic
Huber PR, et al. Procalcitonin guidance of antibiotic therapy surrogate markers: studies with fluoroquinolones in
in community-acquired pneumonia: a randomized trial. patients. Am J Health-Syst Pharm 1999;56(Suppl. 3):S21–4.
Am J Respir Crit Care Med 2006;174(1):84–93. [56] Craig WA. Pharmacokinetics/pharmacodynamics
[37] Guidelines for the management of adults with hospital- parameters: rationale for antimicrobial dosing of men and
acquired ventilator-associated and healthcare-associated, mice. Clin Infect Dis 1998;26:1–12.
pneumonia. Am J Resp Crit Care Med 2005;171:388–416. [57] Manduru M, Mihm LB, White RL, Friedrich LW, Flume RA,
[38] Potel G, Caillon J, Jacqueline C, Navas D, Kergueris MF, Bosso JA. In vitro pharmacodynamics of ceftazidime
Batard E. Dosage des antibiotiques en réanimation: quand against P. aeruginosa isolates from cystic fibrosis patients.
et comment demander et interpréter les tests. Réanimation Antimicrob Agents Chemother 1997;41:2053–6.
2006;15:187–92. [58] Wysocki M, Delatour F, Faurisson F, Rauss A, Pean Y, Misset
[39] Weinbren MJ. Pharmacokinetics of antibiotics in burn B, et al. Continuous versus intermittent infusion of
patients. J Antimicrob Chemother 1999;44:319–27. vancomycin in severe Staphylococcal infections:
[40] Zaske DE, Sawchuk RJ, Gerding DR, Strate RG. Increased prospective multicenter randomised study. Antimicrob
dosage requirements of gentamycin in burn patients. J Agents Chemother 2001;45:2460–7.
Trauma 1976;6:824–8. [59] Garaffo R. Bases pharmacodynamiques de l’administration
[41] Bourget P, Lesne-Hulin A, Le Reveillé R, Le Bever H, Carsin IV des beta lactamines par perfusion continue:
H. Clinical pharmacokinetics of piperacillin–tazobactam optimisation de l’activité antibactérienne sur les bacilles à
combination in patients with major burns and signs gram négatif. Antibiotics 2002;4:22–7.
of infection. Antimicrob Agents Chemother 1996;40: [60] Viaene E, Chanteux H, Servais H, Mingeot – Leclerc MP,
139–45. Tulkens P. Comparative stability of antipseudomonal
[42] Conil JM, Georges B, Breden A, Segonds C, Lavit M, Seguin T, agents administration through portable elastomeric pumps
et al. Increased amikacin dosage requirements in burn (home therapy for cystic fibrosis patients) and motor
patients receiving a once-daily regimen. Int J Antimicrob operated syringes (intensive care units). Antimicrob Agents
Agents 2006;28:226–30. Chemother 2002;8:2327–32.
[43] Kiser TH, Hoody DW, Obritsch MD, Wegzyn CO, Bauling PC, [61] Swanson D, De Angelis C, Smith I, Schentag J. Degradation
Fish DN. Levofloxacin pharmacokinetics and kinetics of imipenem in normal saline and in human
pharmacodynamics in patients with severe burn injury. serum. Antimicrob Agents Chemother 1986;5:936–7.
Antimicrob Agents Chemother 2006;50:1937–45. [62] Scaglione F. Can PK/PD be used in everyday clinical
[44] Mohr 3rd JF, Ostrosky-Zeichner L, Wainright DJ, Parks DH, practice. Int J Antimicrob Agents 2002;19:349–53.
Hollenbeck TC, Ericsson CD. Pharmacokinetic evaluation of [63] Frimodt-Moller N. How predictive is PK/PD for antibacterial
single-dose intravenous daptomycin in patients with agents? Int J Antimicrob Agents 2002;19:333–9.
thermal burn injury. Antimicrob Agents Chemother [64] Carlet J, Tabah A. Antibiotherapie des etats infectieux
2008;52:1891–3. graves. Med Mal Infect 2006;36:299–303.
[45] Craig W. Does the dose matter? Clin Infect Dis [65] Bolmstrom A. Determination of minimum bactericidal
2001;33(Suppl. 3):S233–7. concentrations, kill curves, and postantibiotic effects with
[46] Roberts JA, Kruger P, Paterson DL, Lipman J. Antibiotic the E-test technology. Diagn Microbiol Infect Dis
resistance—what’s dosing got to do with it? Crit Care Med 1994;19:187–95.
2008;36:2433–40. [66] Griswold JA, Grube BJ, Engrav LH, Marvin JA, Heimbach DM.
[47] Petitjean O, Nicolas P, Tod M. Pharmacodynamie des Determinants of donor sites infections in small burn grafts.
antibiotiques. In: Martin C, Gouin F, editors. Infections et J Burn Care Rehabil 1989;10:531–5.
antibiothérapie en réanimation aux urgences et en [67] Piel P, Scarnati S, Goldfarb W, Slater H. Antibiotics
chirurgie. Paris: Arnette; 2000. p. 13–82. prophylaxis in patients undergoing burn wound excision. J
[48] Vogelman B, Craig WA. Kinetics of antimicrobial activity. J Burn Care Rehabil 1985;6:422–4.
Pediatr 1986;108:835–40. [68] Recommandations pour la pratique de l’antibioprophylaxie
[49] Hyatt JM, Mckinnon PS, Zimmer GS, Schentag JJ. The en chirurgie. Ann Fr Anesth Reanim 1999;18:75–85.
importance of pharmacokinetic/pharmacodynamic [69] Eagle H, Fleishman R, Levy M. «Continuous» vs
surrogate markers to outcome. Focus on antibacterial «discontinuous» therapy with penicillin: the effect of the
agents. Clin Pharmacokinet 1995;28:143–60. interval between injections on therapeutic efficacy. N Engl J
[50] Moore RD, Lietman PS, Smith CR. Clinical response to Med 1953;12:481–8.
aminoglycoside therapy: importance of the ratio of peak [70] Craig W, Ebert S. Continuous infusion of beta-lactam
concentration to minimal inhibitory concentration. J infect antibiotics. Antimicrob Agents Chemother 1992;36:2577–83.
Dis 1987;155:93–9. [71] Mouton J, Vinks A. Is continuous infusion of beta lactam
[51] Marik PE. Aminoglycoside volumer of distribution and antibiotics worthwhile? Efficacy and pharmacokinectic
illness severity in clinically ill patients. Anesth intensive considerations. J Antimicrob Chemother 1996;38:5–15.
Care 1993;21:172–3. [72] Leder K, Turnidge J, Korman T, Grayson L. The clinical
[52] Barza M, Ioannidis JP, Cappelleri JC, Lau J. Single or multiple efficacy of continuous infusion flucloxacillin in serious
daily doses of aminoglycosides: a meta-analysis. BMJ staphylococcal sepsis. J Antimicrob Chemother
1996;312:338–45. 1999;43:113–8.
[53] Lipman J, Scribante J, Gous AG, Hon H, Tshukutsoane S, The [73] Roberts JA, Paratz J, Paratz E, Krueger WA, Lipman J.
Baragwanath Ciprofloxacin Study Group. Pharmacokinetics Continuous infusion of beta-lactam antibiotics in severe
profiles of high dose intravenous ciprofloxacin in severe infections: a review of its role. Int J Antimicrob Agents
sepsis. Antimicrob Agents Chemother 1998;42:2235–9. 2007;30:11–8.
[54] Thomas JK, Forrest A, Bhavnani SM, Hyatt JM, Cheng A, [74] Lode H, Borner K, Koeppe P. Pharmacodynamics of
Ballow CH, et al. Pharmacodynamics evaluation of factor fluoroquinolones. CID 1998;27:33–9.
associated with the development of bacterial resistance in [75] Garrelts JC, Jost G, Kowalsky SF, Krol GJ, Lettieri JT.
acutely ill patients during therapy. Antimicrob Agents Ciprofloxacin pharmacokinetics in burn patients.
Chemother 1998;42:521–7. Antimicrob Agents Chemother 1996;40:1153–6.
26 burns 37 (2011) 16–26

[76] Johnson AP, Warren M, Livermore DM. Activity of linezolid [78] Adembri C, Fallani S, Cassetta MI, Arrigucci S, Ottaviano A,
against multi resistant Gram-positive bacteria from diverse Pecile P, et al. Linezolid pharmacokinetic–pharmacodynamic
hospitals in the UK. J Antimicrob Chemother 2000;45: profile in critically ill septic patients: intermittent versus
225–30. continuous infusion. Int J Antimicrob Agents 2008;31:122–9.
[77] Whitehouse T, Cepeda JA, Shulman R, Aarons L, Nalda- [79] Lovering AM, Le Floch R, Hovsepian L, Stephanazzi J, Bret P,
Molina R, Tobin C, et al. Pharmacokinetic studies of Birraux G, et al. Pharmacokinetic evaluation of linezolid in
linezolid and teicoplanin in the critically ill. J Antimicrob patients with major thermal injuries. J Antimicrob
Chemother 2005;55:333–40. Chemother 2009 [Epub ahead of print].

You might also like