Abstract

Vancomycin is one of only a few antibiotics available to treat patients infected with methicillin-resistant Staphylococcus aureus and methicillin-resistant, coagulase-negative Staphylococcus species. Therefore, understanding the clinical implications of the pharmacokinetic and pharmacodynamic properties of vancomycin is a necessity for clinicians. Vancomycin is a concentration-independent antibiotic (also referred to as a “time-dependent” antibiotic), and there are factors that affect its clinical activity, including variable tissue distribution, inoculum size, and emerging resistance. This article reviews the pharmacokinetic and pharmacodynamic data related to vancomycin and discusses such clinical issues as toxicities and serum concentration monitoring.

Vancomycin is a large glycopeptide compound with a molecular weight of ∼1450 Da [1]. It is not appreciably absorbed orally and is eliminated primarily via the renal route, with >80%–90% recovered unchanged in urine within 24 h after administration of a single dose [2]. The pharmacokinetic profile of vancomycin is complex and can be characterized by either a 2- or 3-compartment pharmacokinetic profile (figure 1) [3,4,5]. The drug is administered intravenously, with a standard infusion time of at least 1 h, to minimize infusion-related adverse effects. In patients with normal creatinine clearance, vancomycin has an α-distribution phase of ∼30 min to 1 h and a β-elimination half-life of 6–12 h. The volume of distribution is 0.4–1 L/kg [2, 4–7]. The binding of vancomycin to protein has been reported in the literature to range from 10% to 50% [8–11]. Factors that affect the overall activity of vancomycin include its tissue distribution, inoculum size, and protein-binding effects.

Figure 1

Schematic representation of a 2-compartment pharmacokinetic model, wherein C is the concentration, α and β are the respective elimination constants, e is the base of the natural logarithm, t is time, A and B are the respective zero time intercepts for α and β, Ko is the infusion rate constant, Vc is the volume of the central compartment, Vp is the volume of the peripheral compartment, K12 and K21 are intracompartmental rate constants, and KEL is the elimination rate constant from the central compartment.

Tissue Distribution

Vancomycin penetrates into most body spaces, although the concentrations obtained are variable and somewhat dependent on the degree of inflammation present. In studies examining the penetration of vancomycin into the CSF of patients with uninflamed meninges, fairly low concentrations have been demonstrated (range, 0–3.45 mg/L), with corresponding CSF-to-serum ratios of 0–0.18 [1, 12]. As expected, inflamed meninges improve penetration of vancomycin into the CNS, with reported concentrations of 6.4–11.1 mg/L and CSF-to-serum ratios of 0.36–0.48 [1, 12].

The penetration of vancomycin into the lung is highly variable. Cruciani et al. [13] investigated the penetration of vancomycin into the lung tissue of 36 patients undergoing a partial lobectomy. After intravenous administration of 1 g of vancomycin, concentrations ranged from 0 to 12.2 mg/L, with a mean concentration of 2.8 mg/L and a penetration of 41% [13]. In a recent study investigating the penetration of vancomycin into the epithelial lining fluid of healthy volunteers given 1 g of vancomycin every 12 h, the mean concentration at 12 h was 2.4 mg/L, which represented a 52% overall penetration rate [14]. However, in critically injured patients, penetration of vancomycin into epithelial lining fluid was more variable, ranging from 0 to 8.1 mg/L after several hours, with an overall blood-to—epithelial lining fluid penetration ratio of 6:1 [15]. Craig and Andes [16] recently compared the efficacy of vancomycin with that of oritavancin (a glycopeptide derivative similar to vancomycin) in a thigh and lung mouse infection model. Although the activity of oritavancin was comparable in both lung and thigh infection models, vancomycin activity was found to be 2–3-fold less potent in the lung infection model, compared with the thigh infection model.

Inoculum Size

The impact of inoculum on the activity of vancomycin has recently been examined in an in vitro pharmacodynamic model incorporating simulated endocardial vegetations against methicillin-susceptible and methicillin-resistant Staphylococcus aureus [17]. The activity of vancomycin, nafcillin, daptomycin, and linezolid was examined at a moderate inoculum of 106 log10 cfu/g and a high inoculum of 109 log10 cfu/g over a 72-h period. At the lower inoculum of 106 log10 cfu/g, vancomycin, nafcillin, and daptomycin demonstrated similar bactericidal activity; however, at the higher inoculum, nafcillin and vancomycin had little to no impact over the 72-h period, whereas the effectiveness of daptomycin was minimally affected. Although linezolid is bacteriostatic, it was not affected by the size of the inoculum.

Protein-Binding Effects

Although most studies have shown that the binding of vancomycin to protein is moderate (⩽50%), there are a number of in vitro assessments that have demonstrated a 1–8-fold increase in the MIC as a result of the presence of albumin, whereas the presence of serum has had a more variable effect [17–19].

Vancomycin Concentration And Killing Activity

A number of in vitro and animal studies have been performed to determine the relationship between vancomycin concentration and killing activity. Most in vitro killing curve experiments evaluating fixed vancomycin exposure concentrations as small increments of the MIC demonstrate that killing activity does not change as a function of increasing concentration [20]. Using an in vitro pharmacodynamic model that could mimic the elimination half-life of vancomycin, Larson et al. [21] evaluated the effects of concentration and bacterial activity against a strain of methicillin-resistant S. aureus. The administration of stepwise increasing clinical concentrations (range, 5–40 mg/L) resulted in no appreciable difference in killing. The postantibiotic effect of vancomycin is dependent on the concentration. As drug concentrations exceed the MIC by 2–4-fold, the postantibiotic effect was been reported to increase from 0.2 to 2 h for S. aureus and from 4.3 to 6.5 h for Staphylococcus epidermidis [22]. In vitro and neutropenic mouse thigh infection models have demonstrated that the area under the concentration curve (AUC) divided by the MIC (AUC/MIC) is the best predictor of the activity of vancomycin against methicillin-susceptible S. aureus (figure 2), methicillin-resistant S. aureus, and glycopeptide-intermediate S. aureus (GISA). However, in a Streptococcus pneumoniae nonneutropenic mouse peritonitis model, Knudsen et al. [24] demonstrated that the peak serum concentration divided by the MIC (peak/MIC) was the pharmacodynamic parameter with the most predictive value. Why peak/MIC has predictive value is unknown, but the reason may be one of several factors, including the species of organism, very high susceptibility to vancomycin, and the contribution of neutrophils to elimination of the pathogen.

Figure 2

Relationship between pharmacokinetic/pharmacodynamic indices for vancomycin and bacteriologic efficacy against methicillin-susceptible Staphylococcus aureus. This plot, which delineates the change in colony-forming units (cfu) in an experimental mouse infection model 3 different ways, suggests that the area under the curve divided by the MIC (AUC/MIC) is the most valuable pharmacokinetic/pharmacodynamic parameter for predicting the activity of vancomycin against methicillin-susceptible S. aureus. Peak/MIC, peak serum concentration divided by the MIC. Data are from Ebert [23].

Human Pharmacodynamic Studies

There are very few human studies evaluating the pharmacodynamics of vancomycin, and the findings of most of those studies have not been conclusive in determining which parameter has the most value in predicting patient outcome. The majority of studies have involved relatively small patient populations and patients with a variety of infection types. One prospective evaluation randomized 106 patients with S. aureus infections, including bacteremia and endocarditis, to achieve 3 different trough concentration targets of 5–10 mg/L, 10–15 mg/L, and 15–25 mg/L. No relationships were found between peak concentrations, trough concentrations, or pharmacodynamic parameters (e.g., peak/MIC, time above the MIC, or AUC/MIC) and organism eradication or overall patient outcome [25]. Moise-Broder et al. [26] examined the relationship between the vancomycin AUC/MIC and the outcomes of 108 patients with methicillin-resistant S. aureus pneumonia. An AUC/MIC value of ⩾400 was associated with a successful outcome, whereas an AUC/MIC value of <400 was associated with a lower eradication rate and a higher mortality rate (P = .005) [26]. A recent study examined the relationship between the AUC/MIC value and a successful outcome in 168 patients with S. aureus bacteremia. The MIC50 was 0.5 mg/L (range, 0.25–1.0 mg/L), and the median AUC/MIC value was 1072. Overall, in this study, no relationship was found between successful outcome and a specific AUC/MIC value [27].

The development of staphylococcal resistance to vancomycin has been associated with prolonged exposure to low serum concentrations of the drug. GISA infection and subsequent failure of vancomycin therapy have been reported since the middle of the 1990s. By definition, these strains have a vancomycin MIC of 8–16 mg/L. The majority of cases of GISA infection have occurred among patients receiving peritoneal dialysis or hemodialysis who had received suboptimal, prolonged, and repeated courses of vancomycin [28]. Most cases of GISA infection have involved serum concentrations of vancomycin that were consistently ⩽10 mg/L. Although the number of cases of GISA infection has remained low, there appears to be some evidence that this type of resistance has occurred in the past but may have been underreported because of our inability to detect these strains in the clinical laboratory [29]. The Centers for Disease Control and Prevention recommends the use of vancomycin screening plates of 6 mg/L, which may increase our ability to detect these strains [30]. S. aureus strains that display heteroresistance to glycopeptides (i.e., heteroresistant GISA strains) have also been reported to be associated with vancomycin therapy failure [30, 31]. These strains typically have an MIC of 1–4 mg/L but contain a subpopulation of cells that exhibit higher MIC values when plated onto agar plates containing vancomycin or when tested with a heavy inocula by use of the Etest (AB BIODISK) methods for antimicrobial susceptibility. Similar to GISA strains, these organisms are difficult to detect in the clinical laboratory, and their prevalence may be underreported [32]. Recent in vitro evaluations have demonstrated a relationship between exposure to low vancomycin serum concentrations and the development of heteroresistant GISA [33]. However, because of the difficulty in detecting these strains clinically, the overall prevalence and clinical significance of heteroresistant GISA have not been established [32, 34].

Toxicity

In recent years, there appears to be less controversy with regard to the relationship between serum vancomycin concentrations and toxicity. Historically, vancomycin toxicities were related to impurities in the manufacturing process [1]. Although most animal studies have not found that vancomycin causes nephrotoxicity, there have been a number of studies involving humans that have attempted to link elevated serum vancomycin serum concentrations with renal damage [35–39]. Most of these studies are retrospective, and definitions for nephrotoxicity are highly variable. In many cases, serum vancomycin concentrations were measured after an elevation in serum creatinine levels, making it uncertain which came first. Despite these drawbacks, the nephrotoxic potential of vancomycin is reported to be ⩽5% [37, 39]. In one of the largest investigations to date, Pestotnik et al. [40] reported that the incidence of nephrotoxicity among 1750 patients was 1.4%. Of interest, vancomycin appears to potentiate the nephrotoxicity of aminoglycosides, when administered in combination with this class of drugs. Both animal and human studies have concluded that the combination may increase the nephrotoxicity of aminoglycosides by, on average, 3–4-fold [37, 39, 41].

With respect to ototoxicity, the overall incidence appears to be low [42]. Despite clinical case reports of a relationship between vancomycin serum concentrations and ototoxicity, there are no animal models that have demonstrated this relationship. The majority of experts believe that this drug is not ototoxic [42–45].

The so-called therapeutic range most often quoted for vancomycin monitoring is peak and trough serum concentrations of 30–40 mg/L and 5–10 mg/L, respectively [46, 47]. As stated earlier, there is little evidence to support a relationship between specific serum concentrations and efficacy and toxicity. Because vancomycin is a concentration-independent, or time-dependent, antibiotic and because there are practical issues associated with determining a precise peak serum concentration with this multicompartment antibiotic, most clinicians have abandoned the routine practice of determining peak serum concentrations. Therefore, most clinicians rely solely on monitoring trough serum concentrations for this antibiotic.

The overall AUC/MIC value may be the pharmacodynamic parameter that best correlates with a successful outcome associated with the use of vancomycin; however, further studies seem to be warranted. The nephrotoxic and ototoxic effects of this drug are minimal and are not related to specific serum concentrations. Prolonged exposure to serum concentrations close to the MIC are associated with the emergence of resistance; therefore, it is important to maintain adequate serum concentrations in patients with fast or rapidly changing creatinine clearance. There are several body compartments in which penetration is poor, such as the lung and the CNS. It would also seem prudent to keep concentrations from being suboptimal in patients with pneumonia or meningitis, as well as in patients receiving vancomycin who are receiving dialysis for renal failure. The American Thoracic Society recently published guidelines for hospital-acquired, ventilator-associated, and health care—associated pneumonia. These guidelines recommend vancomycin trough concentrations of 15–20 mg/L for the treatment of methicillin-resistant S. aureus pneumonia. The recommendations are based on vancomycin susceptibility and pharmacokinetic/pharmacodynamic properties, as well as reported clinical experience, since no definitive clinical studies have evaluated the recommended serum concentrations with respect to patient outcome [48]. Although precise and aggressive pharmacokinetic adjustments via the successive measurement of serum concentrations may not be necessary for most infections, empirical adjustments made using standard pharmacokinetic equations or a validated nomogram that maintains trough serum concentrations at 4–5 times the MIC would seem to be reasonable. Higher concentrations may be needed for sequestered infections or in situations where vancomycin penetration has been documented to be poor. Further research examining the outcome of patients as it relates to vancomycin serum concentrations is warranted.

Acknowledgments

Potential conflicts of interest. M.J.R. receives grant and/or research support from Advancis, Bayer, Cubist, and Oscient, is a consultant for Advancis, Bayer, Cubist, and Oscient, and is on speakers' bureaus of Aventis, Bayer, and Cubist.

References

1
Cooper
GL
Given
DB
Pharmacokinetics of vancomycin
Vancomycin: a comprehensive review of 30 years clinical experience
1986
San Diego
Park Row Publishers
(pg. 
23
-
38
)
2
Matzke
GR
Zhanel
GG
Guay
DR
Clinical pharmacokinetics of vancomycin
Clin Pharmacokinet
1986
, vol. 
11
 (pg. 
257
-
82
)
3
Matzke
GR
McGory
RW
Halstenson
CE
Keane
WF
Pharmacokinetics of vancomycin in patients with various degrees of renal function
Antimicrob Agents Chemother
1984
, vol. 
25
 (pg. 
433
-
7
)
4
Rodvold
KA
Blum
RA
Fischer
JH
, et al. 
Vancomycin pharmacokinetics in patients with various degrees of renal function
Antimicrob Agents Chemother
1988
, vol. 
32
 (pg. 
848
-
52
)
5
Rotschafer
JC
Crossley
K
Zaske
DE
Mead
K
Sawchuk
RJ
Solem
LD
Pharmacokinetics of vancomycin: observations in 28 patients and dosage recommendations
Antimicrob Agents Chemother
1982
, vol. 
22
 (pg. 
391
-
4
)
6
Blouin
RA
Bauer
LA
Miller
DD
Record
KE
Griffen
WO
Jr
Vancomycin pharmacokinetics in normal and morbidly obese subjects
Antimicrob Agents Chemother
1982
, vol. 
21
 (pg. 
575
-
80
)
7
Golper
TA
Noonan
HM
Elzinga
L
, et al. 
Vancomycin pharmacokinetics, renal handling, and nonrenal clearances in normal human subjects
Clin Pharmacol Ther
1988
, vol. 
43
 (pg. 
565
-
70
)
8
Ackerman
BH
Taylor
EH
Olsen
KM
Abdel-Malak
W
Pappas
AA
Vancomycin serum protein binding determination by ultrafiltration
Drug Intell Clin Pharm
1988
, vol. 
22
 (pg. 
300
-
3
)
9
Bailey
EM
Rybak
MJ
Kaatz
GW
Comparative effect of protein binding on the killing activities of teicoplanin and vancomycin
Antimicrob Agents Chemother
1991
, vol. 
35
 (pg. 
1089
-
92
)
10
Zokufa
HZ
Solem
LD
Rodvold
KA
Crossley
KB
Fischer
JH
Rotschafer
JC
The influence of serum albumin and alpha 1-acid glycoprotein on vancomycin protein binding in patients with burn injuries
J Burn Care Rehabil
1989
, vol. 
10
 (pg. 
425
-
8
)
11
Albrecht
LM
Rybak
MJ
Warbasse
LH
Edwards
DJ
Vancomycin protein binding in patients with infections caused by Staphylococcus aureus
DICP
1991
, vol. 
25
 (pg. 
713
-
5
)
12
Albanese
J
Leone
M
Bruguerolle
B
Ayem
ML
Lacarelle
B
Martin
C
Cerebrospinal fluid penetration and pharmacokinetics of vancomycin administered by continuous infusion to mechanically ventilated patients in an intensive care unit
Antimicrob Agents Chemother
2000
, vol. 
44
 (pg. 
1356
-
8
)
13
Cruciani
M
Gatti
G
Lazzarini
L
, et al. 
Penetration of vancomycin into human lung tissue
J Antimicrob Chemother
1996
, vol. 
38
 (pg. 
865
-
9
)
14
Rodvold
K
Gotfried
MH
Loutit
JS
Porter
SB
Plasma and intrapulmonary concentrations of oritavancin and vancomycin in normal healthy adults [abstract O254]
Program and abstracts of the 14th European Congress of Clinical Microbiology and Infectious Diseases (Prague)
2004
Basel
European Society of Clinical Microbiology and Infectious Diseases
pg. 
44
 
15
Lamer
C
de Beco
V
Soler
P
, et al. 
Analysis of vancomycin entry into pulmonary lining fluid by bronchoalveolar lavage in critically ill patients
Antimicrob Agents Chemother
1993
, vol. 
37
 (pg. 
281
-
6
)
16
Craig
WA
Andes
DR
Activity of oritavancin versus vancomycin in the neutropenic murine thigh-and lung-infection models [abstract A-1863]
Program and abstracts of the 44th Interscience Conference on Antimicrobial Agents and Chemotherapy (Washington, DC)
2004
Washington, DC
American Society for Microbiology
pg. 
37
 
17
LaPlante
KL
Rybak
MJ
Impact of high-inoculum Staphylococcus aureus on the activities of nafcillin, vancomycin, linezolid, and daptomycin, alone and in combination with gentamicin, in an in vitro pharmacodynamic model
Antimicrob Agents Chemother
2004
, vol. 
48
 (pg. 
4665
-
72
)
18
Stanley
D
McGrath
BJ
Lamp
KC
Rybak
MJ
Effect of human serum on killing activity of vancomycin and teicoplanin against Staphylococcus aureus
Pharmacotherapy
1994
, vol. 
14
 (pg. 
35
-
9
)
19
Lamp
KC
Rybak
MJ
Bailey
EM
Kaatz
GW
In vitro pharmacodynamic effects of concentration, pH, and growth phase on serum bactericidal activities of daptomycin and vancomycin
Antimicrob Agents Chemother
1992
, vol. 
36
 (pg. 
2709
-
14
)
20
Ross
GH
Wright
DH
Rotschafer
JC
Ibrahim
KH
Glycopeptide pharmacodynamics. In: Nightingale CH, Murakawa T, Ambrose PG, eds. Antimicrobial pharmacodynamics in theory and clinical practice. 1st ed
New York: Marcel Dekker
2002
(pg. 
177
-
204
)
21
Larsson
AJ
Walker
KJ
Raddatz
JK
Rotschafer
JC
The concentration-independent effect of monoexponential and biexponential decay in vancomycin concentrations on the killing of Staphylococcus aureus under aerobic and anaerobic conditions
J Antimicrob Chemother
1996
, vol. 
38
 (pg. 
589
-
97
)
22
Lowdin
E
Odenholt
I
Cars
O
In vitro studies of pharmacodynamic properties of vancomycin against Staphylococcus aureus and Staphylococcus epidermidis
Antimicrob Agents Chemother
1998
, vol. 
42
 (pg. 
2739
-
44
)
23
Ebert
S
In vivo cidal activity and pharmacokinetic parameters for vancomycin against methicillin-susceptible and -resistant S. aureus [abstract 439]. In: Program and abstracts of the 27th Interscience Conference on Antimicrobial Agents and Chemotherapy (New York)
Washington, DC
American Society for Microbiology
(pg. 
1987
-
173
)
24
Knudsen
JD
Fuursted
K
Espersen
F
Frimodt-Moller
N
Activities of vancomycin and teicoplanin against penicillin-resistant pneumococci in vitro and in vivo and correlation to pharmacokinetic parameters in the mouse peritonitis model
Antimicrob Agents Chemother
1997
, vol. 
41
 (pg. 
1910
-
5
)
25
Rybak
MJ
Cappelletty
DM
Ruffing
MJ
, et al. 
Influence of vancomycin serum concentrations on the outcome of patients being treated for gram-positive infections [abstract A-46]
Program and abstracts of the 37th Interscience Conference on Antimicrobial Agents and Chemotherapy (Toronto)
1997
Washington, DC
American Society for Microbiology
pg. 
9
 
26
Moise-Broder
PA
Forrest
A
Birmingham
MC
Schentag
JJ
Pharmacodynamics of vancomycin and other antimicrobials in patients with Staphylococcus aureus lower respiratory tract infections
Clin Pharmacokinet
2004
, vol. 
43
 (pg. 
925
-
42
)
27
Drew
RH
Lu
I
Joyce
M
Benjamin
DK
Jr
Fowler
VG
Jr
Lack of relationship between predicted area under the time-concentration curve/minimum inhibitory concentration and outcome in vancomycin-treated patients with Staphylococcus aureus bacteremia [abstract A-1493]
Program and abstracts of the 44th Interscience Conference on Antimicrobial Agents and Chemotherapy (Washington, DC)
2004
Washington, DC
American Society for Microbiology
pg. 
36
 
28
Rybak
MJ
Akins
RL
Emergence of methicillin-resistant Staphylococcus aureus with intermediate glycopeptide resistance: clinical significance and treatment options
Drugs
2001
, vol. 
61
 (pg. 
1
-
7
)
29
Fridkin
SK
Vancomycin-intermediate and -resistant Staphylococcus aureus: what the infectious disease specialist needs to know
Clin Infect Dis
2001
, vol. 
32
 (pg. 
108
-
15
)
30
Carey
RB
Patel
JB
McAllister
S
, et al. 
Failure of automated systems to detect vancomycin-resistant Staphylococcus aureus [abstract D-66]
Program and abstracts of the 44th Interscience Conference on Antimicrobial Agents and Chemotherapy (Washington, DC)
2004
Washington, DC
American Society for Microbiology
pg. 
142
 
31
Moore
MR
Perdreau-Remington
F
Chambers
HF
Vancomycin treatment failure associated with heterogeneous vancomycin-intermediate Staphylococcus aureus in a patient with endocarditis and in the rabbit model of endocarditis
Antimicrob Agents Chemother
2003
, vol. 
47
 (pg. 
1262
-
6
)
32
Howden
BP
Ward
PB
Charles
PG
, et al. 
Treatment outcomes for serious infections caused by methicillin-resistant Staphylococcus aureus with reduced vancomycin susceptibility
Clin Infect Dis
2004
, vol. 
38
 (pg. 
521
-
8
)
33
Liu
C
Chambers
HF
Staphylococcus aureus with heterogeneous resistance to vancomycin: epidemiology, clinical significance, and critical assessment of diagnostic methods
Antimicrob Agents Chemother
2003
, vol. 
47
 (pg. 
3040
-
5
)
34
Charles
PG
Ward
PB
Johnson
PD
Howden
BP
Grayson
ML
Clinical features associated with bacteremia due to heterogeneous vancomycin-intermediate Staphylococcus aureus
Clin Infect Dis
2004
, vol. 
38
 (pg. 
448
-
51
)
35
Wood
CA
Kohlhepp
SJ
Kohnen
PW
Houghton
DC
Gilbert
DN
Vancomycin enhancement of experimental tobramycin nephrotoxicity
Antimicrob Agents Chemother
1986
, vol. 
30
 (pg. 
20
-
4
)
36
Wold
JS
Turnipseed
SA
Toxicology of vancomycin in laboratory animals
Rev Infect Dis
1981
, vol. 
3
 
Suppl
(pg. 
224
-
9
)
37
Rybak
MJ
Albrecht
LM
Boike
SC
Chandrasekar
PH
Nephrotoxicity of vancomycin, alone and with an aminoglycoside
J Antimicrob Chemother
1990
, vol. 
25
 (pg. 
679
-
87
)
38
Cimino
MA
Rotstein
C
Slaughter
RL
Emrich
LJ
Relationship of serum antibiotic concentrations to nephrotoxicity in cancer patients receiving concurrent aminoglycoside and vancomycin therapy
Am J Med
1987
, vol. 
83
 (pg. 
1091
-
7
)
39
Farber
BF
Moellering
RC
Jr
Retrospective study of the toxicity of preparations of vancomycin from 1974 to 1981
Antimicrob Agents Chemother
1983
, vol. 
23
 (pg. 
138
-
41
)
40
Pestotnik
SL
Adverse effects of intravenous vancomycin in hospitalized patients: attributable costs and excess length of stay [abstract 376]
Program and abstracts of the 35th Annual Meeting of the Infectious Diseases Society of America (San Francisco)
1997
Alexandria, VA
Infectious Diseases Society of America
pg. 
424
 
41
Sorrell
TC
Collignon
PJ
A prospective study of adverse reactions associated with vancomycin therapy
J Antimicrob Chemother
1985
, vol. 
16
 (pg. 
235
-
41
)
42
Elting
LS
Rubenstein
EB
Kurtin
D
, et al. 
Mississippi mud in the 1990s: risks and outcomes of vancomycin-associated toxicity in general oncology practice
Cancer
1998
, vol. 
83
 (pg. 
2597
-
607
)
43
Tange
RA
Kieviet
HL
von Marle
J
Bagger-Sjoback
D
Ring
W
An experimental study of vancomycin-induced cochlear damage
Arch Otorhinolaryngol
1989
, vol. 
246
 (pg. 
67
-
70
)
44
Brummett
RE
Fox
KE
Vancomycin- and erythromycin-induced hearing loss in humans
Antimicrob Agents Chemother
1989
, vol. 
33
 (pg. 
791
-
6
)
45
Cantu
TG
Dick
JD
Elliott
DE
Humphrey
RL
Kornhauser
DM
Protein binding of vancomycin in a patient with immunoglobulin A myeloma
Antimicrob Agents Chemother
1990
, vol. 
34
 (pg. 
1459
-
61
)
46
Lundstrom
TS
Sobel
JD
Vancomycin, trimethoprim-sulfamethoxazole, and rifampin
Infect Dis Clin North Am
1995
, vol. 
9
 (pg. 
747
-
67
)
47
Lundstrom
TS
Sobel
JD
Antibiotics for gram-positive bacterial infections: vancomycin, quinupristin-dalfopristin, linezolid, and daptomycin
Infect Dis Clin North Am
2004
, vol. 
18
 (pg. 
651
-
68
x
48
American Thoracic Society
Infectious Diseases Society of America. Guidelines for the management of adults with hospital-acquired, ventilator-associated, and healthcare-associated pneumonia
Am J Respir Crit Care Med
2005
, vol. 
171
 (pg. 
388
-
416
)

Comments

0 Comments
Submit a comment
You have entered an invalid code
Thank you for submitting a comment on this article. Your comment will be reviewed and published at the journal's discretion. Please check for further notifications by email.