Nothing Special   »   [go: up one dir, main page]

Ni Hms 653800

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Internal Medicine Journal 2005; 35: S3-S16

Antibiotics currently used in the treatment of infections


caused by Staphylococcus aureus
C. RAYNER1* and W. J. MUNCKHOF 2

Facility for Anti-infective Drug Development and Innovation,Victorian College of Pharmacy, Monash University,
1

Parkville,Victoria and 2Infection Management Service, Princess Alexandra Hospital & District Health Service,
Brisbane, Queensland, Australia

Abstract typically multi-resistant (mrMRSA), and mrMRSA


Staphylococcal infections are a common and significant strains must always be treated with a combination of two
clinical problem in medical practice. Most strains of oral antimicrobials, typically rifampicin and fusidic acid,
Staphylococcus aureus are now resistant to penicillin, and because resistance develops rapidly if they are used as
methicillin-resistant strains of S. aureus (MRSA) are single agents. Most community-acquired strains of
common in hospitals and are emerging in the com- MRSA in Australia and New Zealand are non multi-
munity. Penicillinase-resistant penicillins (flucloxacillin, resistant (nmMRSA), and lincosamides (clindamycin,
dicloxacillin) remain the antibiotics of choice for the lincomycin) or cotrimoxazole are the antibiotics of
management of serious methicillin-susceptible S. aureus choice for less serious nmMRSA infections such as skin
(MSSA) infections, but first generation cephalosporins and soft tissue infections. New antibiotics such as
(cefazolin, cephalothin and cephalexin), clindamycin, linezolid and quinupristin / dalfopristin have good anti-
lincomycin and erythromycin have important therapeutic staphylococcal activity but are very expensive and should
roles in less serious MSSA infections such as skin and be reserved for patients who fail on or are intolerant of
soft tissue infections or in patients with penicillin hyper- conventional therapy or who have highly resistant strains
sensitivity, although cephalosporins are contra-indicated such as hVISA (heterogenous vancomycin-intermediate
in patients with immediate penicillin hypersensitivity S aureus). (Intern Med J 2005; 35: S3–S16)
(urticaria, angioedema, bronchospasm or anaphylaxis).
All serious MRSA infections should be treated with Key words: Staphylococcus aureus, antibiotics, drug
parenteral vancomycin or, if the patient is vancomycin selection, therapeutic drug monitoring, hospital-in-the-
allergic, teicoplanin. Nosocomial strains of MRSA are home.

INTRODUCTION This review discusses the efficacy and adverse effect


profiles of antibiotics that may be used to treat suspected
In Australia and New Zealand, antistaphylococcal peni- or confirmed infections caused by S. aureus, and provides a
cillins and glycopeptides are the antimicrobial agents background for subsequent reviews in this issue in which
most commonly selected to treat infections caused by more explicit indication-based recommendations for
methicillin-susceptible and methicillin-resistant Staphylo- managing staphylococcal infections are outlined. As well
coccus aureus, respectively. However, there are certain as systematically evaluating those antimicrobials used
clinical scenarios where these conventional therapies may primarily to treat infections caused by both methicillin-
not be optimal or feasible for patient care, and where susceptible and methicillin-resistant S. aureus, it also
other antibiotics may be more appropriate. The rationale examines the potential roles of more recently approved
for clinical use of certain antibiotics varies between indi- antibiotics, the controversies surrounding therapeutic drug
cations. Factors to consider when choosing an antibiotic monitoring of vancomycin and other antistaphylococcal
may include excellent efficacy, low incidence of toxicity, agents, and special considerations for ‘Hospital in the
low potential for resistance development, penetration of Home’ parenteral therapy of infections caused by S. aureus.
the antibiotic into certain tissues (e.g. bone, cerebrospinal
fluid), inexpensive cost, and stability and dosing con- DRUFUSED PRIMARILY TO TREAT
venience for home intravenous use.
METHICILLIN-SUSCEPTIBLE
STAPHYLOCOCCUS AUREUS (MSSA)
Correspondence to: Dr Wendy J. Munckhof, Infection Management INFECTIONS
Service, Princess Alexandra Hospital, and The University of
Queensland, Brisbane, Queensland, Australia. β-Lactam antibiotics
Email:Wendy_Munckhof@health.qld.gov.au Penicillins
*Current position: Clinical Pharmacologist, Roche Products Penicillins are the antimicrobial agents most commonly
Limited, 6 Falcon Way, Shire Park,Welwyn Garden City, UK. used to treat serious methicillin-susceptible staphylococcal
S4 Rayner & Munckhof

infections. β-Lactams exhibit time-dependent killing of particularly those requiring prolonged therapy, oral
S. aureus in vitro, with moderate postantibiotic effects.1 dicloxacillin may be preferable to oral flucloxacillin.
Maximal killing of S. aureus in animal models occurs The commonest toxicity of any of the penicillins is
when serum levels exceed the minimum inhibitory hypersensitivity, which occurs in between 1% and 10%
concentration (MIC) for 40% to 50% of the dosing of the population,12 and in these patients, an alternative
interval.2 High peaks are much less important than antimicrobial agent must be used. A careful history of
duration of exposure to drug, with maximal killing of S. the clinical manifestation of the penicillin allergy should
aureus occurring at concentrations only 3 to 4 times the be elicited. For a relatively mild and late manifestation
MIC.3 Hence, when considering pharmacodynamic such as rash, a cephalosporin antibiotic may be used,
principles, the goal of a dosing regimen for β-lactam although there is an increased risk of a reaction to these
antibiotics active against S. aureus would be to optimize drugs (see further below). In patients with a history of
the duration of exposure to the drug. For short half-life an immediate hypersensitivity reaction to penicillin
penicillins such as benzylpenicillin and flucloxacillin, this (urticaria, angioedema, bronchospasm, or anaphylaxis),
can best be done by continuous intravenous infusion, a other β-lactam agents are contraindicated,13 and a non-
therapeutic approach that can be utilized in ‘Hospital in β-lactam antibiotic should be chosen.
the Home’ programmes.
Several types of penicillins are used to treat staphylo- Penicillin/β-lactamase inhibitor combinations
coccal infections. As more than 80% of Australian strains The penicillins amoxicillin, ticarcillin and piperacillin are
of MSSA produce a penicillinase,4 a penicillinase- only active against the small percentage of strains of
resistant penicillin needs to be used in most patients. S. aureus that do not produce β-lactamase. However, the
However, for the minority of strains of S. aureus that do addition of a β-lactamase inhibitor such as clavulanate or
not produce penicillinase, benzylpenicillin remains an tazobactam will readily inhibit the β-lactamase of many
effective therapeutic agent. The most commonly used bacteria, including S. aureus and some Gram-negative
penicillinase-resistant penicillins in Australia are flu- and anaerobic bacteria.14,15 Hence, efficacy against
cloxacillin and dicloxacillin. Methicillin is no longer penicillin-resistant, methicillin-susceptible staphylococci
commercially available in Australia, due to the rela- will be restored with these combination agents, but the
tively high incidence of interstitial nephritis associated overall antibacterial spectrum is broader than with the
with its use, and nor are cloxacillin, oxacillin and penicillinase-resistant penicillins. For this reason,
nafcillin. penicillinase-resistant penicillins are recommended if
Flucloxacillin and dicloxacillin remain the most effec- MSSA is the sole pathogen. However, for mixed infec-
tive antibiotics for serious MSSA infections such as tions such as diabetic foot infections or infected bites
endocarditis or bone and joint infections. They have where Gram-negative organisms and anaerobes are often
better bactericidal activity than cephalosporins against present in addition to S. aureus, the combination prepa-
S. aureus in vitro and in the rabbit model of endocarditis.5 rations are often used.
However, first-generation cephalosporins are equally Penicillin/β-lactamase inhibitors currently available in
effective and potentially less toxic than flucloxacillin and Australia include amoxicillin/clavulanate (oral formula-
dicloxacillin for the treatment of minor staphylococcal tion), ticarcillin/clavulanate (parenteral) and piperacillin/
infections, such as skin and soft tissue infections. Both tazabactam (parenteral). As well as broadening the
flucloxacillin and dicloxacillin are available in oral and antibacterial spectrum, the addition of clavulanate
parenteral formulations, although due to precipitation to amoxicillin also increases the likelihood of adverse
problems and lack of evidence that it is stable in solution events such as diarrhoea and hepatitis.16,17 Ticarcillin/
over 24 h, parenteral dicloxacillin is not recommended clavulanate and piperacillin/tazabactam are more expen-
for use in continuous infusion regimens in ‘Hospital in sive than most other parenteral penicillins.
the Home’ programmes.
There are no significant differences in pharmacokinetic Cephalosporins
profiles or efficacy between flucloxacillin and di- First-generation (or moderate-spectrum) cephalosporins
cloxacillin,6,7 although there are some differences in their are stable to staphylococcal β-lactamase but are not as
toxicity profiles, leading to much recent debate about their bactericidal as penicillins against S. aureus in vitro or in
relative places in the therapy of staphylococcal infec- the rabbit model of endocarditis.5 Hence, penicillinase-
tions.8–10 Flucloxacillin is associated with a cholestatic resistant penicillins such as flucloxacillin or dicloxacillin
hepatitis in 1 per 15 000 treatment courses of the drug, are preferred to cephalosporins for serious staphylo-
and fatalities have been reported rarely. In case-control coccal infections such as endocarditis or bone and joint
studies, the main risk factors for flucloxacillin-associated infections, although first-generation cephalosporins are
cholestatic hepatitis were age over 55 years and duration useful alternatives in patients with penicillin hyper-
of therapy over 2 weeks.11 Cholestatic hepatitis has been sensitivity or intolerance. There is, however, a dearth of
reported less frequently for dicloxacillin than flucloxa- information from clinical trials that have directly
cillin, but dicloxacillin causes more infusion phlebitis and assessed the effectiveness of cephazolin or other first-
interstitial nephritis. If either dicloxacillin or flucloxacillin generation cephalosporins in endocarditis caused by
are to be used intravenously, consideration should be S. aureus.
given to prolonging the infusion time (to >1 h) or admin- Cephalosporins are also indicated in the treatment of
istration via a central line, if available. In older patients, less severe staphylococcal infections, such as skin and

Internal Medicine Journal 2005; 35: S3–S16


Antistaphylococcal anitbiotics S5

Table 1 Usual adult dosages of selected antistaphylococcal antibiotics†

Antibiotic Adult dosage/route of administration

Agents active against non-multiresistant MRSA strains (nmrMRSA)‡


Dicloxacillin 250–500 mg q6h (PO) or 0.5–2 g q6h (IV)
Flucloxacillin 250–1000 mg q6h (PO) or 0.5–2 g q6h (IV)
Amoxicillin/clavulanate 1 tablet (500 mg/125 mg or 875 mg/125 mg) PO bid
Ticarcillin/clavulanate 3.1 g IV q6-8h
Piperacillin/tazobactam 2 g/0.25 g–4 g/0.5 g IV q6h-8h
Cephazolin 0.5–2 g q8h IV
Cephalothin 0.5–1 g q6h IV
Cephalexin 250–1000 mg q6h PO
Clindamycin 150–450 mg q6h (PO) or 600–1200 mg/d in 3–4 equal doses (IV)
Co-trimoxazole (trimethoprim/sulphamethoxazole) 1 DS tablet (160 mg/800 mg) PO bid
Agents active against multiresistant MRSA strains (mrMRSA)§
Vancomycin 1000 mg q12h or 500 mg q6h (IV) [see section on therapeutic
drug monitoring]
Teicoplanin 12 mg/kg loading dose (2 doses in 12 h) followed by
3–6 mg/kg/day as a single daily maintenance dose (IV or IM)
[see section on therapeutic drug monitoring]
Rifampicin 300 mg q12h (IV or PO)
Fusidic acid 500 mg q8h-12h (PO or IV)
Ciprofloxacin 500 mg PO q12h or 400 mg IV q12h
Moxifloxacin 400 mg q24h (PO or IV)
Gatifloxacin 400 mg q24h (PO or IV)
Last-line agents
Linezolid 600 mg bid (IV or PO)
Quinupristin/dalfopristin 7.5 mg/kg q8h-q12h (IV) [Note: IV dosing; requires central venous
catheter due to occurrence of phlebitis]
†Range of doses listed may change depending on the nature of the specific infection, organ dysfunction, and other factors. Consult approved

product information for further details.


‡nmrMRSA = resistant to 0, 1 or 2 non-β-lactam agents.

§mrMRSA = resistant to 3 or more non-β-lactam agents.

IV, intravenous; MRSA, methicillin-resistant S. aureus; PO, oral.

soft tissue infections, because there is potentially less efficacy in a twice-daily parenteral dose for the treat-
toxicity than penicillinase-resistant penicillins and ment of skin and soft tissue infections, and dosing may
dosing is more convenient (Table 1).The major toxicities be decreased to once-daily with the addition of
of the cephalosporins are hypersensitivity and rash, but probenecid to increase its half-life.23 This strategy makes
they are much less likely to cause serious adverse events it convenient for ‘Hospital in the Home’ use. Later
such as hepatitis, interstitial nephritis or phlebitis than generation cephalosporins (i.e. second-generation agents
the penicillinase-resistant penicillins.6 As between 3% with moderate antibacterial spectra and enhanced anti-
and 8% of patients who are hypersensitive to penicillins Haemophilus or and antianaerobic activity, and third- or
will exhibit cross-reactivity with cephalosporins, fourth-generation agents with broad antibacterial spectra
predominantly as a rash,12,18,19 if there is a history of an with or without anti-Pseudomonas activity) all have
immediate hypersensitivity reaction to penicillins broader antibacterial spectra than the first-generation
(urticaria, angioedema, bronchospasm or anaphylaxis), cephalosporins, with either the same (some second-
cephalosporins are contraindicated. generation, all fourth-generation agents) or less (some
Cephazolin is more susceptible than cephalothin to second-generation, all third-generation agents) anti-
hydrolysis by β-lactamases produced by some strains of staphylococcal activity, along with increased costs.6
S. aureus.20 Clinical failure of cephazolin in endocarditis Hence, first-generation cephalosporins are preferred in
due to S. aureus has been reported several times.21 One the specific treatment of staphylococcal infections,
group suggested only using cephazolin if high inoculum unless the infection is caused by mixed organisms, in
MIC testing or molecular characterization of the which case a broader spectrum, later-generation agent
isolate’s β-lactamase showed it was susceptible to may be required.
cephazolin.22
First-generation agents currently available in Carbapenems
Australia include cephalexin (oral formulation), cepha- Carbapenems such as imipenem and meropenem are
zolin (IV) and cephalothin (IV). Cephazolin has good extremely broad-spectrum, expensive, parenteral β-lactam

Internal Medicine Journal 2005; 35: S3–S16


S6 Rayner & Munckhof

agents with good activity against many bacterial species, and protein synthesis by binding to the 50s ribosomal
including MSSA.14,24 Carbapenems are occasionally unit. It has activity against S. pneumoniae, S. pyogenes and
used to treat MSSA in mixed infections but, in general, MSSA, as well as a broad range of Gram-positive and
should be regarded as agents to be held in reserve for the Gram-negative anaerobes.40 It is not active against
treatment of staphylococcal infections. multiresistant MRSA (mrMRSA), but MSSA and most
strains of non-multiresistant MRSA (nmrMRSA) are
Non-β-lactam antibiotics generally clindamycin susceptible.30–32 Clindamycin is
essentially bacteriostatic, displays time-dependent
Macrolides and azalides activity at concentrations >4 times the MIC,41 and has a
Macrolides such as erythromycin, clarithromycin and modest in vitro postantibiotic effect against S. aureus.42
roxithromycin, and the azalide agent azithromycin act by Like the macrolides, it is bacteriostatic and therefore
binding to the 50s ribosome and inhibiting bacterial should be avoided as the sole treatment for endocarditis.
protein synthesis.25 In general, they are inactive against Clindamycin is available in both intravenous and oral
multiresistant methicillin-resistant S. aureus (mrMRSA),26 formulations. It has an oral bioavailability of 90%,
but are active against approximately 85% of MSSA27 is extensively bound to plasma proteins (>90%),
and 65% to 75% of non-multiresistant MRSA distributed widely in most tissues and body fluids
(nmrMRSA).28–32 Against extracellular pathogens, (except for cerebrospinal fluid), and has a short plasma
including S. aureus, macrolides and azalides are generally half-life of approximately 2 h.40,43,44 It is extensively
considered bacteriostatic, time-dependent bacterial metabolized by the liver to active metabolites.40 Clinda-
killers, and they require concentrations to be at least 4 to mycin demonstrates good penetration into bone and
5 times the MIC, and to exceed the MIC for at least 50% abscesses,45 and hence is an effective therapy for MSSA
of the dosing interval, for an optimal outcome based on osteomyelitis and septic arthritis.46–48
general findings for time-dependent killing drugs from Clindamycin is now often used as a first-line agent to
in vivo infection models.33–35 treat skin and soft tissue infections caused by
In general, however, bacteriostatic agents should be nmrMRSA.22,31,36,49,50 Because of the relatively recent emer-
avoided in the treatment of serious infections such as gence of nmrMRSA, most publications are either small
endocarditis. Also, erythromycin is not recommended as case series of nmrMRSA infections31 or extrapolations
empirical therapy for staphylococcal skin and soft tissue from the treatment of MSSA with clindamycin;51 good
infections, as up to 15% of strains are erythromycin- clinical trials of the therapy of nmrMRSA have yet to be
resistant and there is a relatively high incidence of nausea conducted. Also, some erythromycin-resistant strains of
and vomiting (oral formulation) or phlebitis (IV formu- nmrMRSA may have inducible MLSB resistance to clin-
lation) with this drug.36 However, if alternative agents damycin,52 which can be tested for in the laboratory with
such as β-lactams or clindamycin are contraindicated, the erythromycin-clindamycin disc approximation test.
erythromycin is an appropriate therapeutic choice for If this is not possible, erythromycin-resistant strains
erythromycin-susceptible strains. should be reported as clindamycin resistant.
The macrolides and azalides are extensively
distributed into tissues – particularly azithromycin and Co-trimoxazole (trimethoprim/sulphamethoxazole)
roxithromycin which have long half-lives compared Co-trimoxazole is a combination of two antimicrobials,
with erythromycin (68 h and 8–15 h vs 2–4 h, respec- trimethoprim and sulphamethoxazole, which inhibit
tively).37,38 Resistance to erythromycin usually confers bacterial folate synthesis at two different enzymatic
resistance to the newer macrolides and azalides, as well stages. Most strains of MSSA and nmrMRSA in Australia
as reduced activity compared with other drug classes and New Zealand are currently susceptible to cotrimoxa-
including the lincosamides and streptogramins.39 zole,28–31 while most Australian strains of multiresistant
Prior to prescribing a macrolide, it is particularly MRSA (mrMRSA) are resistant.27,53 Co-trimoxazole is
important to consult the drug interaction profiles in rarely used to treat serious staphylococcal infections such
the prescribing information. This is especially important as endocarditis, as it is inferior to the β-lactams and
for erythromycin and clarithromycin, but less so for glycopeptides. In a study of the treatment of experimental
roxithromycin and azithromycin. Unlike the latter drugs, S. aureus endocarditis in rabbits, cotrimoxazole proved
erythromycin and clarithromycin are potent inhibitors of inferior to cloxacillin, vancomycin and teicoplanin,54 and
the cytochrome P450 isoenzymes (particularly 3A4 and in intravenous drug users with MSSA endocarditis, it was
1A2), which may lead to serious drug interactions with a inferior to vancomycin.55 A recent review of case reports,
large number of other drugs including warfarin, theo- case series and clinical studies that focused on the use of
phylline, ergot alkaloids, carbamazepine and cisapride. cotrimoxazole for treatment of severe S. aureus infections
suggested that it may have a limited role in treating severe
Lincosamides infections, but randomised comparator-controlled
The lincosamides, lincomycin and clindamycin are studies are required to establish its place in therapy.56
chemically unrelated to the macrolides, although there Hence, cotrimoxazole is mostly used orally in the treat-
are some structural similarities. Clindamycin is a ment of minor staphylococcal infections caused by
synthetic derivative of lincomycin, but has a broader MSSA, for which it is an effective therapy.57
spectrum of antibacterial activity and improved oral More recently, with the advent of nmrMRSA, cotri-
absorption. Clindamycin inhibits peptide chain initiation moxazole has experienced a therapeutic resurgence. In
Internal Medicine Journal 2005; 35: S3–S16
Antistaphylococcal anitbiotics S7

Australia and New Zealand, most nmrMRSA strains are endocarditis, the doses of gentamicin used were 1 mg/kg
currently susceptible to cotrimoxazole,28–31 and either IV three times daily.64
clindamycin or cotrimoxazole are the agents most
commonly used to treat skin and soft tissue infections DRUGS USED PRIMARILY TO TREAT
caused by nmrMRSA,36 although published experience METHICILLIN-RESISTANT
is limited.58 STAPHYLOCOCCUS AUREUS (MRSA)
The most common adverse events of cotrimoxazole INFECTIONS
are nausea, diarrhoea and rash. Stevens–Johnson
syndrome, neutropenia, thrombocytopenia and nephro- Vancomycin
toxicity may also occur but are rare.6 Age is a risk factor Vancomycin is a Gram-positive, cell-wall active
for developing serious adverse reactions, and special care glycopeptide antibiotic that demonstrates antibacterial
must be exercised when prescribing cotrimoxazole to behaviour largely independent of its concentration. In
elderly patients. vitro, maximal antibacterial effects occur with concentra-
tions 4–5 times the MIC. Vancomycin also displays
Tetracyclines persistent effects which minimize the occurrence of
The tetracyclines are currently not widely used for the bacterial regrowth at times when the concentration of
treatment of staphylococcal infections. Most strains of antibiotic has declined to a level less than the MIC.67–69
mrMRSA, and many strains of nmrMRSA and MSSA Vancomycin is extensively used to treat staphylococcal
are resistant to this group of drugs.27 However, some infections, especially those caused by MRSA. In vitro,
strains of mrMRSA that are tetracycline-resistant are animal model, and human clinical studies (including
minocycline-susceptible, and minocycline is sometimes endocarditis)70 have demonstrated that vancomycin
used in the treatment of MRSA infections in developing evokes slower and incomplete bactericidal effects in
countries where vancomycin is not available, although MSSA compared with β-lactams.71–74 Hence, when
clinical data are relatively limited.59,60 sensitivity allows, antistaphylococcal penicillins are
The major toxicities of the tetracyclines are gastroin- preferred to glycopeptides against S. aureus. Also, in the
testinal upset, candidal superinfection, photosensitivity, last few years, vancomycin-intermediate Staphylococcus
and, in children, discoloration of teeth. aureus (VISA), heterogenous VISA (hVISA), and
vancomycin-resistant Staphylococcus aureus (VRSA) have
Aminoglycosides evolved.75,76 A clinical pointer to their possible presence
Most strains of MSSA and nmrMRSA are susceptible to may be failure of vancomycin therapy for MRSA infec-
gentamicin, but most strains of mrMRSA are gentamicin- tion despite therapeutic vancomycin levels. VISA and
resistant.61,62 Hence, many microbiology laboratories hVISA can be difficult to detect with routine suscept-
currently use gentamicin susceptibility as a rough guide ibility testing, and the microbiology laboratory should
to likely community acquisition of MRSA. The main be alerted so that additional methodology is used
exception is a British hospital-acquired strain of (see review on S. aureus with reduced susceptibility to
nmrMRSA, UK EMRSA-15, which is gentamicin- vancomycin in this issue).
susceptible and has caused outbreaks in Australian Vancomycin is a high molecular weight compound that
hospitals.63 is not absorbed orally and has to be administered by intra-
As the antibacterial activity of gentamicin is primarily venous infusion. In comparison with other antibiotics
against Gram-negative bacteria, its role in the treatment such as the β-lactams, vancomycin has a long plasma half-
of staphylococcal infections is limited. It is primarily life of approximately 6 h in patients with normal renal
used for synergy in the treatment of staphylococcal function.77 This is considerably extended in patients with
endocarditis. In a study in which gentamicin was added renal dysfunction (half-life is up to 7.5 days in anephric
to nafcillin in the treatment of staphylococcal endo- patients, indicating minimal non-renal clearance), and
carditis, there was a more rapid clinical defervescence dosage adjustment is therefore required in these patients.77
and sterilization of blood cultures during the first week, Vancomycin is generally considered to have a high degree
but the ultimate outcome was unaffected. Hence, the of tissue penetration,77–79 although some investigators
authors recommended that gentamicin be added in contend that suboptimal tissue concentrations may
the first 5–7 days only.64 Gentamicin is also some- contribute to poor outcomes in patients with MRSA
times used in the treatment of prosthetic valve pneumonia80 and other deep-seated MRSA infections.
endocarditis, although this is mostly for infection caused Penetration of the drug into cerebrospinal fluid (CSF) is
by methicillin-resistant, gentamicin-susceptible strains of variable and dependent on whether or not meningitis
Staphylococcus epidermidis65 (see also review on endo- is also present.81 One study demonstrated average
carditis in this issue). CSF/serum concentration ratios of 0.18 in patients
Gentamicin must be given parenterally, and it without meningitis and 0.48 in patients with meningitis.82
commonly causes nephrotoxicity and ototoxicity. As it Vancomycin can cause an infusion rate-dependent
has a narrow therapeutic window, gentamicin concentra- and histamine-mediated flushing of the skin, sometimes
tions and renal function need to be monitored carefully. accompanied by angioedema, known as ‘red man’
Once-daily dosing is now used for most therapeutic indi- syndrome.83,84 It has also been linked to ototoxicity and
cations, but this has not been studied in staphylococcal nephrotoxicity, although it is now thought to be much
endocarditis.66 In studies in patients with staphylococcal less frequently associated with these adverse effects than
Internal Medicine Journal 2005; 35: S3–S16
S8 Rayner & Munckhof

initially believed.83 The majority of reports relating to Rifampicin is administered intravenously or orally,
vancomycin-associated ototoxicity and nephrotoxicity and it extensively penetrates into tissues (including
originated from data obtained nearly four decades pyogenic bone and pulmonary lesions), body fluids and
ago.83,85 Much of the data implicating vancomycin with abscesses.97 As approximately 30% of a dose of
toxicity is plagued by concomitant administration of rifampicin is excreted unchanged in the urine, doses
other ototoxic or nephrotoxic drugs, associated disease, should be reduced in the presence of hepatic dysfunc-
poor documentation of the timing of serum concentra- tion.92 Patients taking rifampicin should be warned
tion measurements, varied definitions of toxicity, and the about reddish-orange staining of urine and contact
absence of appropriate audiometric or renal function lenses, and about the potential for drug interactions, as
data.85 The role of therapeutic drug monitoring for rifampicin may cause numerous interactions including
vancomycin toxicity has also been questioned widely in enhanced metabolism of the oral contraceptive pill and
the literature.85 Dosing considerations and therapeutic decreased concentrations (and effects) of cytochrome
drug monitoring of vancomycin are discussed below. P450 3A4 (CYP3A4) substrates such as cyclosporin,
nifedipine and prednisolone.92 Rifampicin may cause
Teicoplanin gastrointestinal upsets in up to 17% of patients, while
Like vancomycin, teicoplanin is a glycopeptide that less than 5% may develop a mild maculopapular rash
inhibits bacterial growth by inhibiting cell wall biosynthesis with long-term therapy or elevations in liver function
and, in general, it has similar activity against Gram- tests.98 Rarely, rifampicin has caused immune-mediated
positive aerobic and anaerobic cocci (including MRSA influenza symptoms, haematological abnormalities and
and C. difficile, respectively).86,87 Teicoplanin also exhibits interstitial nephritis.98
time-dependent bacterial killing and possesses a signifi- The major therapeutic indication for rifampicin in
cant postantibiotic effect in vitro.86 In vivo, it appears to staphylococcal infections is in oral combination therapy
be at least as effective as vancomycin,88,89 although the of mrMRSA infections. In Australia, it is usually
number of clinical trials comparing it with vancomycin is combined with fusidic acid or, if the isolate is suscep-
limited. Hence, it cannot be stated with certainty that the tible, ciprofloxacin. However, rifampicin is a relatively
two drugs are equally therapeutically effective. In one difficult and expensive drug for patients to obtain
non-comparative retrospective study in patients treated outside the public hospital system in Australia, as it is
with teicoplanin (mostly in combination with rifampicin currently not approved under the Pharmaceutical
and/or gentamicin) for endocarditis, cure without Benefits Scheme for the treatment of staphylococcal
surgery was achieved in 48% of cases.90 infections. Hence, for the treatment of MSSA and
Unlike vancomycin, ‘red man’ syndrome is extremely nmrMRSA infections, there are usually cheaper, less
rare with teicoplanin, and it can be administered as toxic and more readily available therapeutic alternatives.
either an intramuscular or intravenous injection.83 Sometimes rifampicin has been considered for
Teicoplanin also has a lesser propensity to cause nephro- addition to a standard regimen in the treatment of
toxicity than vancomycin.83 It is 90% bound to plasma serious staphylococcal infection. An unpublished multi-
albumin, and as a consequence of its prolonged terminal centre Australian study of 97 patients given either
half life of 155–168 h, is administered once daily.86,87 rifampicin/flucloxacillin or flucloxacillin for methicillin-
Since its pharmacokinetics are variable and affected by a susceptible S. aureus found no difference in outcome,
number of patient factors (e.g. renal function, burns, with a tendency for patients given rifampicin to have
intravenous drug abuse), therapeutic drug monitoring of prolonged fever duration (J. Turnidge, personal commu-
teicoplanin is useful to ensure efficacy (see further below). nication). Of more concern is the anecdotal evidence
that adding rifampicin to vancomycin selects for
Rifampicin rifampicin resistant S. aureus,99 despite US recommen-
Rifampicin is a complex macrocyclic antibiotic that specif- dations to use this combination.100 The addition of
ically inhibits chain initiation of bacterial DNA-dependent rifampicin to failing regimens remains controversial at
RNA polymerase by binding to the B-subunit of the this time.
enzyme.91,92 It is bactericidal and has a concentration-
dependent, prolonged postantibiotic effect. Rifampicin is Fusidic acid
active against many pathogens including S. aureus, Fusidic acid arrests bacterial protein synthesis by inter-
coagulase-negative staphylococci, and mycobacteria. In fering with an elongation factor that promotes
Australia, about 90% of hospital strains of mrMRSA are translocation on the bacterial ribosome.101,102 At higher
rifampicin-susceptible.4 Unfortunately, resistance occurs concentrations it is bactericidal, but otherwise is bacte-
rapidly via 1-step target-site mutations in the B-subunit of riostatic.103 There are no studies, to our knowledge, of
RNA polymerase. Therefore, rifampicin must always be pharmacodynamic predictors of efficacy of fusidic acid,
administered in combination with another antibiotic,93,94 and the drug has a relatively short postantibiotic
especially when the minimum bactericidal concentration effect.103,104 Unlike other agents with Gram-positive
of the original antibiotic is high, and in patients with antibacterial activity, fusidic acid has only modest
deep-seated infections such as osteomyelitis or refractory activity against streptococci, but retains activity against
endocarditis.92 The development of rifampicin resistance MSSA and MRSA.105 Currently, about 95% of strains of
in MRSA is rare if the drug is combined with fusidic acid95 mrMRSA are fusidic acid-susceptible, but there has
or therapeutic concentrations of linezolid.96 been a relatively recent increase in resistance, possibly
Internal Medicine Journal 2005; 35: S3–S16
Antistaphylococcal anitbiotics S9

due to selection by the community use of a topical The newer fluoroquinolones gatifloxacin and moxi-
preparation of fusidic acid.4 floxacin are more potent antistaphylococcal agents than
Like rifampicin, fusidic acid is available for intra- ciprofloxacin, and are 5- to 10-fold more active against
venous or oral administration, and it extensively S. aureus.108 However, they do not have useful activity
penetrates into tissues.106 However, it is more highly against ciprofloxacin-resistant strains.116 The majority of
bound to plasma proteins (95% to 97%) and has a mrMRSA strains are resistant117 and hence, in Australia,
longer plasma half life (14 h vs 4–5 h for rifampicin).106 these agents are usually not indicated for the treatment
Gastrointestinal upset may be dose-limiting, and hyper- of mrMRSA infections. If strains are susceptible, care
bilirubinaemia may occur in up to 50% of patients.107 should be taken when they are used to treat serious
Use of fusidic acid is generally restricted to the con- staphylococcal infections such as osteomyelitis or septic
tinuing oral and outpatient treatment of mrMRSA arthritis. As with all fluoroquinolones, gatifloxacin and
infections in combination with rifampicin or, if the moxifloxacin are best combined with another active
isolate is susceptible, ciprofloxacin.107 Like rifampicin, agent in the treatment of mrMRSA infections due to
there are usually cheaper, less toxic and more readily potential resistance development. In vitro studies have
available therapeutic alternatives for the treatment of shown that resistant mutants of S. aureus occur readily
MSSA and nmrMRSA infections. after serial passage on media containing either gati-
floxacin or moxifloxacin,118,119 although more serial
Fluoroquinolones passages were required for S. aureus to acquire resistance
Broad-spectrum fluoroquinolones such as ciprofloxacin, with moxifloxacin than the older fluoroquinolones
gatifloxacin and moxifloxacin are active against S. aureus, ofloxacin or levofloxacin.120 It has been postulated that
and exhibit concentration-dependent killing and a the newer fluoroquinolones do not select for resistance
moderate postantibiotic effect in vitro and in vivo.3,108 as rapidly as the older compounds due to their dual
Studies in animals models and humans have shown that activity against DNA gyrase and topoisomerase IV.121,122
the principal predictor of bacterial killing by fluoro- Moxifloxacin and gatifloxacin both have excellent
quinolones is the 24-h AUC (area under the plasma bioavailability and rarely need to be given parenterally.
concentration-time curve)/MIC ratio (AUC24/MIC Unlike ciprofloxacin, cytochrome P450 is not involved in
ratio). A peak concentration/MIC ratio of >10 is also the metabolism of either moxifloxacin or gatifloxacin,
important in the prevention of selection of resistant and there is no interaction with theophylline. However,
mutants during treatment.109–111 gatifloxacin may interact with digoxin, probenecid and
Fluoroquinolones are not first-line agents in the treat- oral antidiabetic agents, and moxifloxacin may rarely
ment of staphylococcal infections for several reasons. interact with warfarin, so care should be taken if these
Ciprofloxacin, which is the fluoroquinolone most drugs are used concomitantly.123 As with all fluoro-
commonly used to treat staphylococcal infections in quinolones, absorption may be decreased if drugs
Australia, is a relatively expensive antimicrobial agent, containing multivalent cations (antacids, sucralfate,
and its extensive use has been associated with relatively calcium, zinc, magnesium and iron) are given at the
rapid emergence of resistance in Gram-positive same time. Also, patients on drugs that prolong the
bacteria.61,112,113 Whereas 95% of mrMRSA strains were cardiac QTc interval (e.g. quinidine, procainamide,
susceptible to ciprofloxacin in 1989, only 25% are now amiodarone and sotalol) should not receive either gati-
susceptible, although most strains of MSSA remain floxacin or moxifloxacin, as the QTc interval may be
ciprofloxacin susceptible. Ciprofloxacin is much more further lengthened, resulting in ventricular arrhythmias,
active against Gram-negative bacteria than against including Torsades de pointes.124,125
staphylococci, and staphylococcal MICs are usually close The adverse effects of both moxifloxacin and gati-
to the susceptibility breakpoint.114 Also, staphylococci floxacin are mostly mild and the most common are
may develop ciprofloxacin resistance during treatment, nausea, diarrhoea and dizziness. Rare adverse events
especially when used to treat mrMRSA and when include tendonitis, hepatotoxicity126–128 and, with gati-
monotherapy is used.61 As with rifampicin and fusidic floxacin, hypoglycaemia129 which may be related to age
acid, ciprofloxacin should never be used alone to treat and AUC.130
mrMRSA infections, but it is occasionally used in combi-
nation with rifampicin or fusidic acid to treat the minority NEW DRUGS USED PRIMARILY TO
of mrMRSA infections that are ciprofloxacin-susceptible. TREAT STAPHYLOCOCCUS AUREUS
Ciprofloxacin is well absorbed orally and rarely needs INFECTIONS WHERE
to be given parenterally. Adverse events are relatively CONVENTIONAL THERAPY IS
uncommon but include gastrointestinal effects, INAPPROPRIATE
headaches and dizziness, and rash. Ciprofloxacin does
inhibit cytochrome P450 1A2, and can result in Streptogramins
increased serum concentrations of drugs such as theo-
phyllline, caffeine, clopidogrel, fluvoxamine, propranolol Quinupristin/dalfopristin
and warfarin. Also, like all fluoroquinolones, it binds to Quinupristin/dalfopristin is a new parenteral anti-
drugs containing multivalent cations (antacids, sucral- microbial preparation comprised of two streptogramins.
fate, calcium, zinc, magnesium and iron), resulting in a Each drug binds to a different site on the bacterial ribo-
marked decrease in oral absorption of the drug.115 somal subunit and acts synergistically in disrupting both
Internal Medicine Journal 2005; 35: S3–S16
S10 Rayner & Munckhof

early and late phases of bacterial protein synthesis.25 was one-tenth that of linezolid. Subsequent to publi-
Quinupristin/dalfopristin exerts a time-dependent, cation of the above, pristinamycin use at the study
bacteriostatic (or slow bactericidal) activity and a hospital increased substantially, being used in situations
prolonged in vitro postantibiotic effect (4.6–7 h) against where rifampicin/fusidate is problematic (resistance,
S. aureus.35 Based on in vivo models, concentrations intolerance, or interactions, e.g. rifampcin with warfarin)
should exceed the MIC for ~50% of the dosing interval as a cheaper and better tolerated alternative to oral line-
for bactericidal activity, and the AUC of quinupristin/ zolid (I. Gosbell, personal communication).
dalfopristin divided by the MIC of quinupristin has been
found to be associated with efficacy.35,131 The postanti- Oxazolidinones
biotic effect may be reduced significantly if the MRSA Linezolid is a synthetic antimicrobial and the first of the
strain is constitutively resistant to erythromycin.132 oxazolidinone class. Virtually all Gram-positive bacteria
In preregistration clinical studies, quinupristin/ are susceptible (including MRSA), regardless of their
dalfopristin has been shown to have similar success rates susceptibility to other agents.135,139 Linezolid acts on
to comparators in complicated skin and skin structure bacteria via the inhibition of a novel site in ribosomal
infections, catheter-related bacteraemia caused by protein synthesis, interfering with functional initiation
staphylococci (including MRSA), and hospital-acquired complex formation. Linezolid is available for both IV
pneumonia caused by staphylococci and Streptococcus and oral administration, has essentially 100% oral
pneumoniae.133–135 As quinupristin/dalfopristin inhibits bioavailability, and penetrates well into extravascular
CYP 3A4 and the metabolism of compounds such as sites including bone.140–142 In vitro pharmacodynamic
cyclosporin and midazolam, it increases plasma concen- studies have demonstrated that it has bacteriostatic
trations of these CYP3A4 substrates.135 Peripheral activity against enterococci, minimal bactericidal activity
infusion site reactions occur in about 75% of patients, against staphylococci, and is highly bactericidal against
requiring IV administration via central venous access. streptococci. Clinical and bacteriologic outcomes in
Arthralgia and myalgia may also occur in 13% to 50% patients are optimized by maintaining concentrations
of patient courses.135 above the MIC at all times and an AUC/MIC ratio of
Quinupristin/dalfopristin is indicated for the treatment 80–120 for bacteraemia, lower respiratory tract infec-
of suspected or proven MRSA or vancomycin-resistant tions, and skin and soft tissue infections.143
E. faecium infections requiring intravenous therapy where Linezolid has been shown to have equal or superior
other antibiotics are inappropriate. It is, however, very success rates to comparators in treating infections
expensive in comparison with older agents. caused by Gram-positive organisms, including MRSA,
in studies of community-acquired pneumonia, noso-
Pristinamycin comial pneumonia, and skin and soft tissue
Pristinamycin is an older oral streptogramin antibiotic infections.144–146 In non-comparative compassionate use
that is currently licensed in some European countries, programmes, linezolid at a dose of 600 mg twice daily
including France and Spain, but is not commercially has been shown to achieve clinical cure rates of 91.5% in
available in Australia. Its therapeutic role is therefore evaluable patients when treating vancomycin-resistant
likely to be small, but it has been used as an oral alter- enterococci (E. faecium and E. faecalis), MRSA, and
native in patients with mrMRSA infections where other infections in adults.147 Linezolid may also be
rifampicin, fusidic acid or ciprofloxacin are contraindi- effective in treating deep-seated infections such as
cated by resistance or allergy. It is substantially cheaper osteomyelitis.148
than either quinupristin/dalfopristin or linezolid. In an Resistance to linezolid has been reported in entero-
open-label study of the treatment of mrMRSA chest or cocci and rarely in MRSA.149,150 Reversible suppression
skin and soft tissue infections, pristinamycin proved an of indices of haematologic function, including anaemia,
effective therapy.136 In this study, it was combined with thrombocytopenia and leukopenia, may occur in some
oral doxycycline (if the isolate was doxycycline- patients and may be related to patient factors, linezolid
susceptible) due to in vitro synergy between tetracyclines concentrations, and long durations of therapy.151 Line-
and streptogramins and a lessening of the risk of zolid is currently reserved for the treatment of patients
selecting resistance.137 who either fail on or are intolerant of conventional
In a recent Australian study, 27 patients with staphyl- therapy. Like quinupristin/dalfopristin, linezolid is
ococcal bone and joint infections were treated with substantially more expensive than conventional therapy.
pristinamycin, because of patient intolerance or isolate
resistance to rifampicin and/or fusidic acid.138 Twenty- THERAPEUTIC DRUG MONITORING
four had MRSA, three coagulase-negative staphylococci, CONSIDERATIONS
and four multiple organisms. Pristinamycin monotherapy
was given in 19 cases. No haematological or biochemical Vancomycin
toxicity was observed; seven had gastrointestinal side- In the mid-1990s, a number of excellent reviews and
effects and one developed a rash, but only four required editorials called into question the age-old practice of
cessation of the drug due to side-effects. Treatment therapeutic drug monitoring (TDM) of vancomycin,
outcome was evaluated in 23 cases: 16 were cured and citing a lack of evidence for linking concentrations with
five were suppressed. Development of resistance was not toxicity or for improving the effectiveness or safety of
seen.The acquisition cost of pristinamycin to the authors vancomycin, and suggesting that empirical vancomycin
Internal Medicine Journal 2005; 35: S3–S16
Antistaphylococcal anitbiotics S11

dosing regimens (e.g. nomograms that take account of occasionally for teicoplanin to ensure efficacy, especially
renal function and the patient’s age and weight) do not in patients who have high clearances, such as burns
compromise efficacy or safety compared with TDM- patients, children and injecting drug users.86 Trough
based dosing and are more cost-effective.85,152,153 concentrations should be >10 mg/L for most infections,
Currently, there is considerable variability in the utili- except endocarditis where they should be >20 mg/L.158
zation of TDM for vancomycin therapy at different Therapeutic drug monitoring of other antistaphylo-
hospitals, and its use is probably of most importance for coccal antibiotics such as β-lactams is not routine,
making sure that concentrations are adequate for although it is occasionally done for research purposes.
treating an infection.
The vancomycin MIC90 for staphylococci is 2 mg/L,
and maximal antibacterial effects occur with concen-
ANTIBIOTICS USED IN ‘HOSPITAL IN
trations 4–5 times the MIC in vitro. A number of THE HOME’ PROGRAMMES
investigators have shown that serum bactericidal titres Choice of therapy
(serum MBCs) of 1:8 (requiring serum drug concen-
trations of >12 mg/L in one cohort of 20 paediatric Selection of antimicrobials for use in a parenteral
patients)154 are necessary for clinical success.155 Thus, the ‘Hospital in the Home’ programme presents unique
targeting of trough plasma concentrations of 5 mg/L are challenges in addition to inpatient therapeutic consider-
of concern, especially for deep-seated staphylococcal ations. Of particular importance are the issues of
infections. Trough plasma concentrations >10–12 mg/L practicality and economics. For the management of
(which achieve concentrations >4–5 times the MIC and staphylococcal infections in particular, the short plasma
serum MBC titres of >1:8) would appear to be prudent, half-lives of most of the appropriate intravenous
but on the basis of tissue penetration may provide inad- β-lactams means that traditional dosing frequencies
equate local concentrations for some deep-seated are too cumbersome and impractical in the ‘Hospital in
infections. The most recent version (No. 12) of the the Home’ setting. In general, antibiotics given once or
Antibiotics Therapeutic Guidelines has adopted a target twice daily are most convenient and cost-effective for
trough vancomycin concentration of 10–20 mg/L, which home intravenous use, unless programmable pumps
is higher than in previous editions.36 are used, although this technology is relatively expen-
Recently, a 12-month retrospective review of MRSA sive. Hence, two approaches have been used to recruit
bacteraemias in a Melbourne teaching hospital revealed patients into such programs: the consideration of novel
that 9% were identified as hVISA, and that hVISA dosing strategies for short half-life β-lactams, and the use
episodes were more likely than MRSA episodes to occur of alternative antibiotics with longer half-lives (e.g.
in patients who had low trough serum vancomycin levels glycopeptides, ceftriaxone).
(<10 mg/L) during the first week of therapy.156 However,
it is yet to be determined whether dosing to trough Novel therapeutic approaches
concentrations >10 mg/L might improve the response to In recent years, consideration of pharmacokinetic and
hVISA infections or influence the emergence of pharmacodynamic relationships has led to the develop-
glycopeptide resistance. ment of a number of novel dosing strategies, making
Although there are reports of ototoxicity occurring outpatient parenteral treatment feasible for drugs that
with vancomycin, there is a paucity of data implicating would traditionally be considered inappropriate for such
the role of measuring serum vancomycin concentrations. a setting.
In a prospective study, the role of vancomycin alone in
causing nephrotoxicity was found to be very low, but Continuous infusion
nephrotoxicity increased to 22% to 35% with coadmin- Continuous infusions at concentrations above the MIC
istration of aminoglycosides.157 It is likely that high have been suggested to maximize the effectiveness of
vancomycin concentrations in many patients with renal those antibiotics that cause minimal suppression of
impairment may be more a consequence than a cause bacterial growth once concentrations fall below the MIC
of renal dysfunction. Overall, it is considered that (minimal persistent, sub-MIC or postantibiotic effects)
vancomycin monotherapy is unlikely to cause either and display time-dependent bacterial killing. The anti-
nephrotoxicity or ototoxicity, and the use of serum biotics most suited to this approach in dosing are the
concentrations to help identify actual or potential stable, short half-life β-lactams.159
toxicity has not been established.85 Despite the limita- A number of studies have shown that continuous
tions of the evidence linking concentration to toxicity, infusion of a short half-life β-lactam demonstrated
one should be cognizant of the possibility that use of comparable efficacy versus intermittent dosing, with a
larger doses to achieve higher trough concentrations reduced total daily dose and potential cost savings. Such
might increase toxicity. studies have included ceftazidime for pseudomonal
infections in cystic fibrosis patients160 and nosocomial
Other drugs pneumonia,161,162 and cefuroxime for a variety of infec-
Assays are available for the quantification of a variety of tions, including pneumonia.159 Improved efficacy has yet
antibiotics used to treat staphylococcal infections in to be noted with continuous infusions, since most
plasma and other biological fluids in some specialist studies have involved small numbers of patients and have
institutions. Therapeutic drug monitoring is performed mostly used intermittent regimens that are predicted to
Internal Medicine Journal 2005; 35: S3–S16
S12 Rayner & Munckhof

produce adequate concentrations, and no differences in broad-spectrum cephalosporins. Diagn Microbiol Infect Dis
toxicity have been demonstrated. Continuous infusion of 1995; 22: 89–96.
flucloxacillin may be an alternative for the outpatient 3 Craig WA, Ebert SC. Killing and regrowth of bacteria in
vitro: a review. Scand J Infect Dis Suppl 1990; 74: 63–70.
management of staphylococcal infections,163,164 and it
4 Nimmo GR, Bell JM, Mitchell D, Gosbell IB, Pearman JW,
is usually given by a disposable infusion device or Turnidge JD. Antimicrobial resistance in Staphylococcus
programmable pump. However, the cost of these devices aureus in Australian teaching hospitals, 1989–1999. Microb
means they are usually reserved for the management of Drug Resist 2003; 9: 155–60.
serious staphylococcal infections such as endocarditis 5 Kaye D. Treatment of staphylococcal endocarditis. In:
and bone and joint infections. Sande MA, Kaye D, Root RK, eds. Endocarditis:
There is no likely advantage for administering Contemporary Issues in Infectious Diseases, Vol. 2. New
glycopeptides as a continuous infusion, given their York: Churchill Livingstone; 1985: 135–9.
moderate to long postantibiotic effects and long plasma 6 Kucers A, Crowe SM, Grayson ML, Hoy JF. The Use of
half-lives. Antibiotics, 5th edn. Oxford, UK: Butterworth Heinemann;
1997.
Pharmacokinetic enhancement 7 Sutherland R, Croydon EA, Rolinson GN. Flucloxacillin, a
new isoxazolyl penicillin, compared with oxacillin, cloxacillin,
Probenecid has also been used to prolong the plasma and dicloxacillin. Br Med J 1970; 4: 455–60.
half-lives of β-lactam antibiotics that have a significant 8 Aders H. Case study: flucloxacillin – worth the risk? United
tubular secretion component to their elimination165 by Med Protect J 2000; 1: 5–6.
competitively inhibiting their active tubular secretion. 9 Adverse Drug Reactions Advisory Committee Australian
For example, the half-life of benzylpenicillin is increased Drug Evaluation Committee (ADRAC) Commonwealth of
by 2.6-fold in the presence of probenecid.166 The clinical Australia. A comparison of flucloxacillin with dicloxacillin.
value of probenecid ‘pharmacokinetically enhanced’ Aust Adv Drug React Bull 1999; 18: 7.
β-lactam therapy is well established, particularly in the 10 Gosbell IB, Turnidge JD, Tapsall JW, Benn RA. Toxicities of
treatment of gonorrhoea, where current therapeutic flucloxacillin and dicloxacillin – is there really a difference?
Med J Aust 2000; 173: 500–1.
recommendations still advocate use of this combination.
11 McNeil JJ, Grabsch EA, McDonald MM. Postmarketing
The decrease in total clearance (and consequent increase surveillance: strengths and limitations. The flucloxacillin-
in half-life) of the β-lactam is advantageous since it will dicloxacillin story. Med J Aust 1999; 170: 270–3.
increase the total time the β-lactam concentration 12 Lin RY. A perspective on penicillin allergy. Arch Intern Med
exceeds the MIC, allowing for the use of some agents 1992; 152: 930–7.
(e.g. cephazolin) in the home IV setting that otherwise 13 Pumphrey RS, Davis S. Under-reporting of antibiotic ana-
would not have been feasible.23 It should be noted that phylaxis may put patients at risk. Lancet 1999; 353: 1157–8.
probenecid does not significantly alter the pharmaco- 14 Acar JF, Goldstein FW, Kitzis MD. Susceptibility survey of
kinetics of all β-lactams, since a number of these agents piperacillin alone and in the presence of tazobactam.
rely on elimination mechanisms other than tubular J Antimicrob Chemother 1993; 31: 23–8.
15 Bush K. Beta-lactamase inhibitors from laboratory to clinic.
secretion or have a higher affinity than probenecid for
Clin Microbiol Rev 1988; 1: 109–23.
the tubular transport mechanism.167 Thus, it would be 16 Conner CS. Beta-Lactamase inhibitors. Drug Intell Clin
prudent to consult the primary literature when consid- Pharm 1985; 19: 475–6.
ering probenecid-enhanced dosing strategies. 17 Thomson JA, Fairley CK, Ugoni AM, Forbes AB, Purcell
PM, Desmond PV et al. Risk factors for the development of
ACKNOWLEDGEMENTS amoxycillin-clavulanic acid associated jaundice. Med J Aust
1995; 162: 638–40.
The authors would like to thank: the Australian Society for Antimicro-
bials and Australasian Society for Infectious Diseases, who provided all 18 Dash CH. Penicillin allergy and the cephalosporins.
the financial support for the costs of production and publication of this J Antimicrob Chemother 1975; 1: 107–18.
work; Professor John Turnidge and Associate Professor Keryn Chris- 19 Boguniewicz M, Leung DY. Hypersensitivity reactions to
tiansen of the Australian Society for Antimicrobials, for devising the antibiotics commonly used in children. Pediatr Infect Dis J
concept of writing guidelines for the treatment of deep-seated staphy- 1995; 14: 221–31.
lococcal infections and for assistance with various aspects of 20 Fong IW, Engelking ER, Kirby VMM. Relative inactivation
publication; Associate Professor Iain Gosbell, Department of Micro- by Staphylococcus aureus of eight cephalosporin antibiotics.
biology and Infectious Diseases, South-western Area Pathology Antimicrob Agents Chemother 1976; 9: 939–44.
Service, Liverpool, NSW, for convening the ASA Staphylococcus aureus
21 Quinn EL, Pohlod D, Madhavan T, Burch K, Fisher E, Cox F.
Working Party and co-ordinating the writing of the body of
manuscripts and editing this manuscript; Mr Trevor Speight, Medical Clinical experiences with cephazolin and other
Editor, Medicines Information Co. Ltd, Auckland, NZ, for final editing cephalosporins in bacterial endocarditis. J Infect Dis 1973;
and formatting of this manuscript; and the members of the 1328 Suppl: S386–9.
Australasian Society for Infectious Diseases who provided peer review 22 Nannini EC, Singh KV, Murray BE. Relapse of type A beta-
of this work. lactamase-producing Staphylococcus aureus native valve
endocarditis during cefazolin therapy: revisiting the issue.
REFERENCES Clin Infect Dis 2003; 37: 1194–8.
23 Grayson ML, McDonald M, Gibson K, Athan E,
1 Bundtzen RW, Gerber AU, Cohn DL, Craig WA. Munckhof WJ, Paull P et al. Once-daily intravenous cefazolin
Postantibiotic suppression of bacterial growth. Rev Infect Dis plus oral probenecid is equivalent to once-daily intravenous
1981; 3: 28–37. ceftriaxone plus oral placebo for the treatment of moderate-
2 Craig WA. Interrelationship between pharmacokinetics and to-severe cellulitis in adults. Clin Infect Dis 2002; 34:
pharmacodynamics in determining dosage regimens for 1440–8.

Internal Medicine Journal 2005; 35: S3–S16


Antistaphylococcal anitbiotics S13

24 Jones RN, Barry AL, Thornsberry C. In-vitro studies of 43 Flaherty JF, Rodondi LC, Guglielmo BJ, Fleishaker JC,
meropenem. J Antimicrob Chemother 1989; 24: 9–29. Townsend RJ, Gambertoglio JG. Comparative
25 Vannuffel P, Cocito C. Mechanism of action of strepto- pharmacokinetics and serum inhibitory activity of
gramins and macrolides. Drugs 1996; 51 Suppl 1: 20–30. clindamycin in different dosing regimens. Antimicrob Agents
26 Hardy DJ, Hensey DM, Beyer JM, Vojtko C, McDonald EJ, Chemother 1988; 32: 1825–9.
Fernandes PB. Comparative in vitro activities of new 14-, 44 Flaherty JF Jr, Gatti G, White J, Bubp J, Borin M,
15-, and 16-membered macrolides. Antimicrob Agents Gambertoglio JG. Protein binding of clindamycin in sera of
Chemother 1988; 32: 1710–19. patients with AIDS. Antimicrob Agents Chemother 1996; 40:
27 Turnidge J, Bell J. Report for National Antimicrobial 1134–8.
Resistance Surveillance Program. Atlanta, GA: National 45 Norden CW, Shinners E, Niederriter K. Clindamycin
Centre for Diseases Control; 1998. treatment of experimental chronic osteomyelitis due to
28 Collignon P, Gosbell I, Vickery A, Nimmo G, Stylianopoulos T, Staphylococcus aureus. J Infect Dis 1986; 153: 956–9.
Gottlieb T. Community-acquired meticillin-resistant 46 Peltola H, Unkila-Kallio L, Kallio MJ. Simplified treatment
Staphylococcus aureus in Australia. Australian Group on of acute staphylococcal osteomyelitis of childhood. The
Antimicrobial Resistance. Lancet 1998; 352: 145–6. Finnish Study Group. Pediatrics 1997; 99: 846–50.
29 Gosbell IB, Mercer JL, Neville SA, Crone SA, Chant KG, 47 Rodriguez W, Ross S, Khan W, McKay D, Moskowitz P.
Jalaludin BB et al. Non-multiresistant and multiresistant Clindamycin in the treatment of osteomyelitis in children: a
methicillin-resistant Staphylococcus aureus in community- report of 29 cases. Am J Dis Child 1977; 131: 1088–93.
acquired infections. Med J Aust 2001; 174: 627–30. 48 Kaplan SL, Mason EO Jr, Feigin RD. Clindamycin versus
30 Adhikari RP, Cook GM, Lamont I, Lang S, Heffernan H, nafcillin or methicillin in the treatment of Staphylococcus aureus
Smith JM. Phenotypic and molecular characterization of osteomyelitis in children. South Med J 1982; 75: 138–42.
community occurring, Western Samoan phage pattern 49 Frank AL, Marcinak JF, Mangat PD, Schreckenberger PC.
methicillin-resistant Staphylococcus aureus. J Antimicrob Community-acquired and clindamycin-susceptible
Chemother 2002; 50: 825–31. methicillin-resistant Staphylococcus aureus in children. Pediatr
31 Munckhof WJ, Schooneveldt J, Coombs GW, Hoare J, Infect Dis J 1999; 18: 993–1000.
Nimmo GR. Emergence of community-acquired 50 Nimmo GR, Looke DF. Non-multiresistant methicillin-
methicillin-resistant Staphylococcus aureus (MRSA) resistant Staphylococcus aureus in the community. Next
infection in Queensland, Australia. Int J Infect Dis 2003; chapter in the story of staphylococcal sepsis? Med J Aust
7: 259–64. 2001; 174: 617–18.
32 Riley TV, Pearman JW, Rouse IL. Changing epidemiology of 51 Pusponegoro EH, Wiryadi BE. Clindamycin and cloxacillin
methicillin-resistant Staphylococcus aureus in Western compared in the treatment of skin and soft-tissue infections.
Australia. Med J Aust 1995; 163: 412–14. Clin Ther 1990; 12: 236–41.
33 Bauernfeind A, Jungwirth R, Eberlein E. Comparative 52 Munckhof WJ, Harper J, Schooneveldt J, Nimmo GR. Recent
pharmacodynamics of clarithromycin and azithromycin appearance of clindamycin resistance in community-acquired
against respiratory pathogens. Infection 1995; 23: 316–21. methicillin-resistant Staphylococcus aureus (MRSA) in south-
34 den Hollander JG, Knudsen JD, Mouton JW, Fuursted K, east Queensland. Med J Aust 2002; 176: 243–4.
Frimodt-Moller N, Verbrugh HA et al. Comparison of 53 Turnidge J, Lawson P, Munro R, Benn R. A national survey of
pharmacodynamics of azithromycin and erythromycin antimicrobial resistance in Staphylococcus aureus in Australian
in vitro and in vivo. Antimicrob Agents Chemother 1998; teaching hospitals. Med J Aust 1989; 150: 65, 9–72.
42: 377–82. 54 de Gorgolas M, Aviles P, Verdejo C, Fernandez Guerrero ML.
35 Carbon C. Pharmacodynamics of macrolides, azalides, and Treatment of experimental endocarditis due to methicillin-
streptogramins: effect on extracellular pathogens. Clin Infect susceptible or methicillin-resistant Staphylococcus aureus with
Dis 1998; 27: 28–32. trimethoprim-sulfamethoxazole and antibiotics that inhibit cell
36 Antibiotic Writing Group. Therapeutic Guidelines: Antibiotic, wall synthesis. Antimicrob Agents Chemother 1995; 39: 953–7.
Version 12. Melbourne: Therapeutic Guidelines Ltd; 2003. 55 Markowitz N, Quinn EL, Saravolatz LD. Trimethoprim-
37 Nilsen OG. Pharmacokinetics of macrolides. Comparison of sulfamethoxazole compared with vancomycin for the
plasma, tissue and free concentrations with special reference treatment of Staphylococcus aureus infection. Ann Intern Med
to roxithromycin. Infection 1995; 23: S5–9. 1992; 117: 390–8.
38 Stevens RC, Reed MD, Shenep JL, Baker DK, Foulds G, 56 Adra M, Lawrence KR. Trimethoprim/sulfamethoxazole for
Luke DR et al. Pharmacokinetics of azithromycin after single- treatment of severe Staphylococcus aureus infections. Ann
and multiple-doses in children. Pharmacotherapy 1997; 17: Pharmacother 2004; 38: 338–41.
874–80. 57 Ardati KO, Thirumoorthi MC, Dajani AS. Intravenous
39 Lewis RE, Klepser ME, Ernst EJ, Lund BC, Biedenbach DJ, trimethoprim-sulfamethoxazole in the treatment of serious
Jones RN. Evaluation of low-dose, extended-interval infections in children. J Pediatr 1979; 95(5 Pt 1): 801–6.
clindamycin regimens against Staphylococcus aureus and 58 Mitchell JM, MacCulloch D, Morris AJ. MRSA in the
Streptococcus pneumoniae using a dynamic in vitro model of community. N Z Med J 1996; 109: 411.
infection. Antimicrob Agents Chemother 1999; 43: 2005–9. 59 Yuk JH, Dignani MC, Harris RL, Bradshaw MW,
40 Falagas ME, Gorbach SL. Clindamycin and metronidazole. Williams TW Jr. Minocycline as an alternative
Med Clin North Am 1995; 79: 845–67. antistaphylococcal agent. Rev Infect Dis 1991; 13: 1023–4.
41 Klepser ME, Banevicius MA, Quintiliani R, Nightingale CH. 60 Nicolau DP, Freeman CD, Nightingale CH, Coe CJ,
Characterization of bactericidal activity of clindamycin Quintiliani R. Minocycline versus vancomycin for treatment
against Bacteroides fragilis via kill curve methods. Antimicrob of experimental endocarditis caused by oxacillin-resistant
Agents Chemother 1996; 40: 1941–4. Staphylococcus aureus. Antimicrob Agents Chemother 1994;
42 Xue IB, Davey PG, Phillips G. Variation in postantibiotic 38: 1515–18.
effect of clindamycin against clinical isolates of Staphylococcus 61 Turnidge JD, Bell JM. Methicillin-resistant Staphylococcus
aureus and implications for dosing of patients with osteo- aureus evolution in Australia over 35 years. Microb Drug
myelitis. Antimicrob Agents Chemother 1996; 40: 1403–7. Resist 2000; 6: 223–9.

Internal Medicine Journal 2005; 35: S3–S16


S14 Rayner & Munckhof

62 Nimmo GR, Schooneveldt J, O’Kane G, McCall B, Vickery A. 79 Ziglam HM, Finch RG. Limitations of presently available
Community acquisition of gentamicin-sensitive methicillin- glycopeptides in the treatment of Gram-positive infection.
resistant Staphylococcus aureus in southeast Queensland, Clin Microbiol Infect 2001; 7: 53–65.
Australia. J Clin Microbiol 2000; 38: 3926–31. 80 Lamer C, de Beco V, Soler P, Calvat S, Fagon JY,
63 Pearman JW, Coombs GW, Grubb WB, O’Brien F. A British Dombret MC et al. Analysis of vancomycin entry into
epidemic strain of methicillin-resistant Staphylococcus aureus pulmonary lining fluid by bronchoalveolar lavage in
(UK EMRSA-15) has become established in Western critically ill patients. Antimicrob Agents Chemother 1993;
Australia. Med J Aust 2001; 174: 662. 37: 281–6.
64 Korzeniowski O, Sande MA. Combination antimicrobial 81 Matzke GR, Zhanel GG, Guay DR. Clinical
therapy for Staphylococcus aureus endocarditis in patients pharmacokinetics of vancomycin. Clin Pharmacokinet 1986;
addicted to parenteral drugs and in nonaddicts: a prospective 11: 257–82.
study. Ann Intern Med 1982; 97: 496–503. 82 Albanese J, Leone M, Bruguerolle B, Ayem ML, Lacarelle B,
65 Karchmer AW, Archer GL, Dismukes WE. Staphylococcus Martin C. Cerebrospinal fluid penetration and
epidermidis causing prosthetic valve endocarditis: pharmacokinetics of vancomycin administered by continuous
microbiologic and clinical observations as guides to therapy. infusion to mechanically ventilated patients in an intensive
Ann Intern Med 1983; 98: 447–55. care unit. Antimicrob Agents Chemother 2000; 44: 1356–8.
66 Munckhof WJ, Grayson ML, Turnidge JD. A meta-analysis of 83 Wilson AP. Comparative safety of teicoplanin and
studies on the safety and efficacy of aminoglycosides given vancomycin. Int J Antimicrob Agents 1998; 10: 143–52.
either once daily or as divided doses. J Antimicrob 84 Healy DP, Sahai JV, Fuller SH, Polk RE. Vancomycin-
Chemother 1996; 37: 645–63. induced histamine release and ‘red man syndrome’:
67 Duffull SB, Begg EJ, Chambers ST, Barclay ML. Efficacies comparison of 1- and 2-hour infusions. Antimicrob Agents
of different vancomycin dosing regimens against Chemother 1990; 34: 550–4.
Staphylococcus aureus determined with a dynamic in vitro 85 Freeman CD, Quintiliani R, Nightingale CH. Vancomycin
model. Antimicrob Agents Chemother 1994; 38: 2480–2. therapeutic drug monitoring: is it necessary? Ann
68 Larsson AJ, Walker KJ, Raddatz JK, Rotschafer JC. The Pharmacother 1993; 27: 594–8.
concentration-independent effect of monoexponential and 86 Brogden RN, Peters DH, Teicoplanin. A reappraisal of its
biexponential decay in vancomycin concentrations on the antimicrobial activity, pharmacokinetic properties and
killing of Staphylococcus aureus under aerobic and anaerobic therapeutic efficacy. Drugs 1994; 47: 823–54.
conditions. J Antimicrob Chemother 1996; 38: 589–97. 87 Campoli-Richards DM, Brogden RN, Faulds D. Teicoplanin.
69 Lowdin E, Odenholt I, Cars O. In vitro studies of A review of its antibacterial activity, pharmacokinetic
pharmacodynamic properties of vancomycin against properties and therapeutic potential. Drugs 1990; 40:
Staphylococcus aureus and Staphylococcus epidermidis. 449–86.
Antimicrob Agents Chemother 1998; 42: 2739–44. 88 Greenberg RN, Benes CA. Time-kill studies with oxacillin,
70 Chang FY, Peacock JE Jr, Musher DM, Triplett P, vancomycin, and teicoplanin versus Staphylococcus aureus.
MacDonald BB, Mylotte JM et al. Staphylococcus aureus J Infect Dis 1990; 161: 1036–7.
bacteremia: recurrence and the impact of antibiotic 89 Maple PA, Hamilton-Miller JM, Brumfitt W. Comparative
treatment in a prospective multicenter study. Medicine in-vitro activity of vancomycin, teicoplanin, ramoplanin
(Baltimore) 2003; 82: 333–9. (formerly A16686), paldimycin, DuP 721 and DuP 105
71 Apellaniz G, Valdes M, Perez R, Martin F, Soria F, Garcia A against methicillin and gentamicin resistant Staphylococcus
et al. Comparison of the effectiveness of various antibiotics in aureus. J Antimicrob Chemother 1989; 23: 517–25.
the treatment of methicillin-susceptible Staphylococcus aureus 90 Wilson AP, Gaya H. Treatment of endocarditis with
experimental infective endocarditis. J Chemother 1991; 3: teicoplanin: a retrospective analysis of 104 cases. J
91–7. Antimicrob Chemother 1996; 38: 507–21.
72 Cantoni L, Wenger A, Glauser MP, Bille J. Comparative 91 Wehrli W, Knusel F, Schmid K, Staehelin M. Interaction of
efficacy of amoxicillin-clavulanate, cloxacillin, and rifamycin with bacterial RNA polymerase. Proc Natl Acad
vancomycin against methicillin-sensitive and methicillin- Sci USA 1968; 61: 667–73.
resistant Staphylococcus aureus endocarditis in rats. J Infect 92 Vesely JJ, Pien FD, Pien BC. Rifampin, a useful drug for
Dis 1989; 159: 989–93. nonmycobacterial infections. Pharmacotherapy 1998; 18:
73 Chambers HF, Miller RT, Newman MD. Right-sided 345–57.
Staphylococcus aureus endocarditis in intravenous drug 93 Jin DJ, Gross CA. Mapping and sequencing of mutations in
abusers: two-week combination therapy. Ann Intern Med the Escherichia coli rpoB gene that lead to rifampicin
1988; 109: 619–24. resistance. J Mol Biol 1988; 202: 45–58.
74 Small PM, Chambers HF. Vancomycin for Staphylococcus 94 Aubry-Damon H, Soussy CJ, Courvalin P. Characterization
aureus endocarditis in intravenous drug users. Antimicrob of mutations in the rpoB gene that confer rifampin resistance
Agents Chemother 1990; 34: 1227–31. in Staphylococcus aureus. Antimicrob Agents Chemother
75 Hiramatsu K, Hanaki H, Ino T,Yabuta K, Oguri T, 1998; 42: 2590–4.
Tenover FC. Methicillin-resistant Staphylococcus aureus 95 Melchior NH. Combined in vitro activity of fusidic acid and
clinical strain with reduced vancomycin susceptibility. rifampicin against methicillin-resistant Staphylococcus aureus
J Antimicrob Chemother 1997; 40: 135–6. strains. In: Ishigami J, ed. Recent Advances in
76 Anonymous. Staphylococcus aureus resistance to vancomycin – Chemotherapy, Antimicrobial Section I. Proceedings of the
United Sates 2002. MMWR Morb Mortal Wkly Rep 2002; 14th International Congress of Chemotherapy, Kyoto. Tokyo:
51: 565–6. University of Tokyo Press; 1985: 582–3.
77 Moellering RC Jr. Pharmacokinetics of vancomycin. 96 Dailey CF, Pagano PJ, Buchanan LV, Paquette JA, Haas JV,
J Antimicrob Chemother 1984; 14 Suppl D: 43–52. Gibson JK. Efficacy of linezolid plus rifampin in an
78 Ross GH, Wright DH, Rotschafer JC, Ibrahim KH. experimental model of methicillin-susceptible Staphylococcus
Glycopeptide Pharmacodynamics. New York: Marcel Dekker; aureus endocarditis. Antimicrob Agents Chemother 2003; 47:
2002. 2655–8.

Internal Medicine Journal 2005; 35: S3–S16


Antistaphylococcal anitbiotics S15

97 Kenny MT, Strates B. Metabolism and pharmacokinetics of five other fluoroquinolones. J Antimicrob Chemother 2003;
the antibiotic rifampin. Drug Metab Rev 1981; 12: 159–218. 51: 1403–7.
98 Grosset J, Leventis S. Adverse effects of rifampin. Rev Infect 119 Roychoudhury S, Twinem TL, Makin KM, Nienaber MA,
Dis 1983; 5 Suppl 3: S440–50. Li C, Morris TW et al. Staphylococcus aureus mutants isolated
99 Simon GL, Smith RH, Sande MA. Emergence of rifampin- via exposure to nonfluorinated quinolones: detection of
resistant strains of Staphylococcus aureus during combination known and unique mutations. Antimicrob Agents Chemother
therapy with vancomycin and rifampin: a report of two cases. 2001; 45: 3422–6.
Rev Infect Dis 1983; 5: S507–8. 120 Dalhoff A. Comparative in vitro and in vivo activity of the
100 Watanakunakorn C, Guerriero JC. Interaction between C-8 methoxy quinolone moxifloxacin and the C-8 chlorine
vancomycin and rifampin against Staphylococcus aureus. quinolone BAY y 3118. Clin Infect Dis 2001; 32 Suppl 1:
Antimicrob Agents Chemother 1981; 19: 1089–91. S16–22.
101 Collignon P, Turnidge J. Fusidic acid in vitro activity. Int J 121 Drlica K, Zhao X. DNA gyrase, topoisomerase IV, and the
Antimicrob Agents 1999; 12: S45–58. 4-quinolones. Microbiol Mol Biol Rev 1997; 61: 377–92.
102 Tanaka S, Kaji A. Does translocase (G-factor) require the 122 Appelbaum PC, Hunter PA. The fluoroquinolone
presence of unesterified +RNA on the donor site for its antibacterials: past, present and future perspectives. Int J
action? – the effect of fusidic acid. Biochem Biophys Res Antimicrob Agents 2000; 16: 5–15.
Commun 1972; 46: 136–42. 123 Melvani S. A review of fluoroquinolones available in
103 Turnidge J. Fusidic acid pharmacology, pharmacokinetics Australia. Australian Society of Antimicrobials Newsletter
and pharmacodynamics. Int J Antimicrob Agents 1999; 12: 2003; 13: 4–5.
S23–34. 124 Bertino JS Jr, Owens RC Jr, Carnes TD, Iannini PB.
104 Munckhof WJ, Turnidge JD. The postantibiotic effect of Gatifloxacin-associated corrected QT interval prolongation,
fusidic acid against gram-positive bacteria. J Antimicrob torsades de pointes, and ventricular fibrillation in patients
Chemother 1997; 40: 433–6. with known risk factors. Clin Infect Dis 2002; 34: 861–3.
105 Verbist L. The antimicrobial activity of fusidic acid. 125 Ball P. Quinolone-induced QT interval prolongation: a
J Antimicrob Chemother 1990; 25 Suppl B: 1–5. not-so-unexpected class effect. J Antimicrob Chemother
106 Reeves DS. The pharmacokinetics of fusidic acid. 2000; 45: 557–9.
J Antimicrob Chemother 1987; 20: 467–76. 126 Coleman CI, Spencer JV, Chung JO, Reddy P. Possible
107 Turnidge J, Grayson ML. Optimum treatment of gatifloxacin-induced fulminant hepatic failure. Ann
staphylococcal infections. Drugs 1993; 45: 353–66. Pharmacother 2002; 36: 1162–7.
108 Turnidge J. Pharmacokinetics and pharmacodynamics of 127 Henann NE, Zambie MF. Gatifloxacin-associated acute
fluoroquinolones. Drugs 1999; 58 Suppl 2: 29–36. hepatitis. Pharmacotherapy 2001; 21: 1579–82.
109 Preston SL, Drusano GL, Berman AL, Fowler CL, Chow AT, 128 Soto S, Lopez-Roses L, Avila S, Lancho A, Gonzalez A,
Dornseif B et al. Pharmacodynamics of levofloxacin: a new Santos E et al. Moxifloxacin-induced acute liver injury. Am J
paradigm for early clinical trials. JAMA 1998; 279: 125–9. Gastroenterol 2002; 97: 1853–4.
110 Forrest A, Nix DE, Ballow CH, Goss TF, Birmingham MC, 129 Baker SE, Hangii MC. Possible gatifloxacin-induced
Schentag JJ. Pharmacodynamics of intravenous ciprofloxacin hypoglycemia. Ann Pharmacother 2002; 36: 1722–6.
in seriously ill patients. Antimicrob Agents Chemother 1993; 130 Ambrose PG, Bhavnani SM, Cirincione BB, Piedmonte M,
37: 1073–81. Grasela TH. Gatifloxacin and the elderly: pharmacokinetic-
111 Craig WA. Pharmacokinetic/pharmacodynamic parameters: pharmacodynamic rationale for a potential age-related dose
rationale for antibacterial dosing of mice and men. Clin reduction. J Antimicrob Chemother 2003; 52: 435–40.
Infect Dis 1998; 26: 1–10; quiz 1–2. 131 Chambers HF. Studies of RP 59500 in vitro and in a rabbit
112 Blumberg HM, Rimland D, Carroll DJ, Terry P, Wachsmuth model of aortic valve endocarditis caused by methicillin-
IK. Rapid development of ciprofloxacin resistance in resistant Staphylococcus aureus. J Antimicrob Chemother
methicillin-susceptible and -resistant Staphylococcus aureus. 1992; 30: 117–22.
J Infect Dis 1991; 163: 1279–85. 132 Fantin B, Leclercq R, Merle Y, Saint-Julien L, Veyrat C,
113 Wadsworth SJ, Kim KH, Satishchandran V, Axelrod P, Duval J et al. Critical influence of resistance to streptogramin
Truant AL, Suh B. Development of new antibiotic resistance B-type antibiotics on activity of RP 59500 (quinupristin-
in methicillin-resistant but not methicillin-susceptible Staphy- dalfopristin) in experimental endocarditis due to Staphylococcus
lococcus aureus. J Antimicrob Chemother 1992; 30: 821–6. aureus. Antimicrob Agents Chemother 1995; 39: 400–5.
114 Eliopoulos CT, Eliopoulos GM. Activity in vitro of the 133 Nichols RL, Graham DR, Barriere SL, Rodgers A, Wilson SE,
quinolones. In: Hooper DC, Rubenstein E, eds. Quinolone Zervos M et al. Treatment of hospitalized patients with
Antimicrobial Agents, 3rd ed. Washington, DC: American complicated gram-positive skin and skin structure infections:
Society for Microbiology; 2003; 91–111. two randomized, multicentre studies of
115 Munckhof WJ. Concurrent prescribing of antibiotics: beware quinupristin/dalfopristin versus cefazolin, oxacillin or
of drug interactions. Aust Fam Physician 1998; 27: 895–901. vancomycin. Synercid Skin and Skin Structure Infection
116 Andersson MI, MacGowan AP. Development of the Group. J Antimicrob Chemother 1999; 44: 263–73.
quinolones. J Antimicrob Chemother 2003; 51 Suppl 1: 1–11. 134 Fagon J, Patrick H, Haas DW, Torres A, Gibert C, Cheadle WG
117 Pfaller MA, Jones RN, Doern GV, Sader HS, Kugler KC, et al. Treatment of gram-positive nosocomial pneumonia.
Beach ML. Survey of blood stream infections attributable to Prospective randomized comparison of quinupristin/
gram-positive cocci: frequency of occurrence and dalfopristin versus vancomycin. Nosocomial Pneumonia
antimicrobial susceptibility of isolates collected in 1997 in Group. Am J Respir Crit Care Med 2000; 161: 753–62.
the United States, Canada, and Latin America from the 135 Eliopoulos GM. Quinupristin-dalfopristin and linezolid:
SENTRY Antimicrobial Surveillance Program. SENTRY evidence and opinion. Clin Infect Dis 2003; 36: 473–81.
Participants Group. Diagn Microbiol Infect Dis 1999; 33: 136 Dancer SJ, Robb A, Crawford A, Morrison D. Oral
283–97. streptogramins in the management of patients with
118 Griggs DJ, Marona H, Piddock LJ. Selection of methicillin-resistant Staphylococcus aureus (MRSA) infections.
moxifloxacin-resistant Staphylococcus aureus compared with J Antimicrob Chemother 2003; 51: 731–5.

Internal Medicine Journal 2005; 35: S3–S16


S16 Rayner & Munckhof

137 Bonhoeffer S, Lipsitch M, Levin BR. Evaluating treatment clinical experience. Antimicrob Agents Chemother 2002; 46:
protocols to prevent antibiotic resistance. Proc Natl Acad Sci 2723–6.
USA 1997; 94: 12106–11. 152 Moellering RC Jr. Monitoring serum vancomycin levels:
138 Ng J, Gosbell IB. Successful oral pristinamycin therapy for climbing the mountain because it is there? Clin Infect Dis
osteoarticular infections due to methicillin-resistant 1994; 18: 544–6.
Staphylococcus aureus (MRSA) and other Staphylococcus spp. 153 Cantu TG,Yamanaka-Yuen NA, Lietman PS. Serum
J Antimicrob Chemother 2005; 55: 1008–12. vancomycin concentrations: reappraisal of their clinical value.
139 Jones RN, Johnson DM, Erwin ME. In vitro antimicrobial Clin Infect Dis 1994; 18: 533–43.
activities and spectra of U-100592 and U-100766, two novel 154 Schaad UB, McCracken GH Jr, Nelson JD. Clinical
fluorinated oxazolidinones. Antimicrob Agents Chemother pharmacology and efficacy of vancomycin in pediatric
1996; 40: 720–6. patients. J Pediatr 1980; 96: 119–26.
140 Meagher AK, Forrest A, Rayner CR, Birmingham MC, 155 Lisby-Sutch SM, Nahata MC. Dosage guidelines for the use
Schentag JJ. Population pharmacokinetics of linezolid in of vancomycin based on its pharmacokinetics in infants. Eur
patients treated in a compassionate-use program. Antimicrob J Clin Pharmacol 1988; 35: 637–42.
Agents Chemother 2003; 47: 548–53. 156 Charles PG, Ward PB, Johnson PD, Howden BP, Grayson ML.
141 Lovering AM, Zhang J, Bannister GC, Lankester BJ, Brown Clinical features associated with bacteremia due to
JH, Narendra G et al. Penetration of linezolid into bone, fat, heterogeneous vancomycin-intermediate Staphylococcus
muscle and haematoma of patients undergoing routine hip aureus. Clin Infect Dis 2004; 38: 448–51.
replacement. J Antimicrob Chemother 2002; 50: 73–7. 157 Rybak MJ, Albrecht LM, Boike SC, Chandrasekar PH.
142 Rana B, Butcher I, Grigoris P, Murnaghan C, Seaton R. Nephrotoxicity of vancomycin, alone and with an
Linezolid penetration into osteo-articular tissues. aminoglycoside. J Antimicrob Chemother 1990; 25: 679–87.
J Antimicrob Chemother 2002; 50: 747–50. 158 Reeves DS, MacGowan AP, Holt HA, Lovering AM,
143 Rayner CR, Forrest A, Meagher AK, Birmingham MC, Warnock DW, White LO. Therapeutic monitoring of
Schentag JJ. Clinical pharmacodynamics of linezolid in antimicrobials: a summary of the information presented at
seriously ill patients treated in a compassionate use the UK NEQAS for antibiotic Assays meeting for
programme. Clin Pharmacokinet 2003; 42: 1411–23. participants, October 1993. J Antimicrob Chemother 1995;
144 Stevens DL, Smith LG, Bruss JB, McConnell-Martin MA, 35: 213–26.
Duvall SE, Todd WM et al. Randomized comparison of 159 MacGowan AP, Bowker KE. Continuous infusion of beta-
linezolid (PNU-100766) versus oxacillin-dicloxacillin for lactam antibiotics. Clin Pharmacokinet 1998; 35: 391–402.
treatment of complicated skin and soft tissue infections. 160 Vinks AA, Brimicombe RW, Heijerman HG, Bakker W.
Antimicrob Agents Chemother 2000; 44: 3408–13. Continuous infusion of ceftazidime in cystic fibrosis patients
145 Stevens DL, Herr D, Lampiris H, Hunt JL, Batts DH, during home treatment: clinical outcome, microbiology and
Hafkin B. Linezolid versus vancomycin for the treatment of pharmacokinetics. J Antimicrob Chemother 1997; 40: 125–33.
methicillin-resistant Staphylococcus aureus infections. Clin 161 Nicolau DP, McNabb J, Lacy MK, Quintiliani R,
Infect Dis 2002; 34: 1481–90. Nightingale CH. Continuous versus intermittent
146 Rubinstein E, Cammarata S, Oliphant T, Wunderink R. administration of ceftazidime in intensive care unit patients
Linezolid (PNU-100766) versus vancomycin in the with nosocomial pneumonia. Int J Antimicrob Agents 2001;
treatment of hospitalized patients with nosocomial 17: 497–504.
pneumonia: a randomized, double-blind, multicenter study. 162 McNabb JJ, Nightingale CH, Quintiliani R, Nicolau DP.
Clin Infect Dis 2001; 32: 402–12. Cost-effectiveness of ceftazidime by continuous infusion
147 Birmingham MC, Rayner CR, Meagher AK, Flavin SM, versus intermittent infusion for nosocomial pneumonia.
Batts DH, Schentag JJ. Linezolid for the treatment of Pharmacotherapy 2001; 21: 549–55.
multidrug-resistant, gram-positive infections: experience from 163 Leder K, Turnidge JD, Korman TM, Grayson ML. The
a compassionate-use program. Clin Infect Dis 2003; 36: clinical efficacy of continuous-infusion flucloxacillin in
159–68. serious staphylococcal sepsis. J Antimicrob Chemother 1999;
148 Rayner CR, Baddour LM, Birmingham MC, Norden C, 43: 113–18.
Meagher AK, Schentag JJ. Linezolid in the treatment of 164 Howden BP, Richards MJ. The efficacy of continuous
osteomyelitis: results of compassionate use experience. infusion flucloxacillin in home therapy for serious
Infection 2004; 32: 8–14. staphylococcal infections and cellulitis. J Antimicrob
149 Lomaestro BM. Resistance to linezolid. Are we surprised? Chemother 2001; 48: 311–14.
How hard should we look? Ann Pharmacother 2003; 37: 165 Gibaldi M, Schwartz MA. Apparent effect of probenecid on
909–11. the distribution of penicillins in man. Clin Pharmacol Ther
150 Wilson P, Andrews JA, Charlesworth R, Walesby R, Singer M, 1968; 9: 345–9.
Farrell DJ et al. Linezolid resistance in clinical isolates of 166 Kampmann J, Lindahl F, Hansen JM, Siersbaek-Nielsen K.
Staphylococcus aureus. J Antimicrob Chemother 2003; 51: Effect of probenecid on the excretion of ampicillin in human
186–8. bile. Br J Pharmacol 1973; 47: 782–6.
151 Gerson SL, Kaplan SL, Bruss JB, Le V, Arellano FM, 167 Brown GR. Cephalosporin–probenecid drug interactions.
Hafkin B et al. Hematologic effects of linezolid: summary of Clin Pharmacokinet 1993; 24: 289–300.

Internal Medicine Journal 2005; 35: S3–S16

You might also like