You are on page 1of 8

SUPPLEMENT ARTICLE

Vancomycin: A History
Donald P. Levine
Department of Medicine, Wayne State University, Detroit, Michigan

Vancomycin became available for clinical use 150 years ago but was soon discarded in favor of other antibiotics
that were deemed to be more efficacious and less toxic. The advent of pseudomembranous enterocolitis,
coupled with the spread of methicillin-resistant Staphylococcus aureus, led to a resurgence in the use of
vancomycin. Almost immediately, concerns arose with regard to its therapeutic utility. In addition, resistance
to vancomycin developed, first in enterococci and later in staphylococci. Several types of resistance have now
been identified, each with a unique effect on infections treated with vancomycin. Recent studies have rekindled
interest in the best way to administer the antibiotic. The findings of future studies may result in a return to
measuring levels of vancomycin in serum, to assure a successful therapeutic outcome.

There is greater interest in vancomycin now, 50 years in resistance to compound 05865 [4]. Isolates from
after its discovery, than at any time in its history. Re- other laboratories were also tested, and the results were
cently published articles about vancomycin reflect par- similar [5, 6]. Subsequent animal experiments sug-
allel increases in both the use of vancomycin and our gested that compound 05865 might be safe and effective
knowledge about this interesting antibiotic (figure 1). in humans. However, before clinical trials were begun,
A historical review puts this into perspective. In the the compound, dubbed “Mississippi mud” because of
1950s, with few options available to treat penicillin- its brown color, needed to be purified. A switch from
resistant staphylococcal infections, Eli Lilly and Com- picric acid precipitation to passage over an ion-ex-
pany initiated a program aimed at discovering antibi- change resin was an improvement, and the resulting
otics with activity against these pathogens. In 1952, a drug, which was named “vancomycin” (from the word
missionary in Borneo sent a sample of dirt to his friend “vanquish”), was made available for clinical trials.
Dr. E. C. Kornfield, an organic chemist at Eli Lilly. An In studies of volunteers, vancomycin reached ther-
organism isolated from that sample (Streptomyces or- apeutic concentrations in various tissue compartments
ientalis) produced a substance (“compound 05865”) and resulted in successful treatment of serious staph-
that was active against most gram-positive organisms, ylococcal infection in 8 of 9 patients, including 1 patient
including penicillin-resistant staphylococci [1]. Some with endocarditis. The sole therapeutic failure occurred
anaerobic organisms, including clostridia, were also in a patient with empyema, in whom an adequate con-
susceptible to compound 05865 [2], as was Neisseria centration could not be achieved in the empyema
pocket [2]. Six patients with endocarditis who had ex-
gonorrhoeae [3]. In vitro experiments were initiated to
perienced prior therapy failure with other antibiotics
determine whether the activity of compound 05865
were also treated with vancomycin; 5 patients were
would be preserved despite attempts to induce resis-
cured. The therapy failure occurred in a patient who
tance. After 20 serial passages of staphylococci from Eli
was admitted to the hospital with multiple complica-
Lilly laboratories, resistance to penicillin increased
tions, including shock and intractable heart failure, and
100,000-fold, compared with only a 4–8-fold increase
who died after receiving treatment for 5 days [7]. As
information about vancomycin spread, it was requested
Reprints or correspondence: Dr. Donald P. Levine, Wayne State University, Dept.
for cases for which other drugs had failed. Emergency
of Medicine, 4201 St. Antoine, Ste. 5C, Detroit, MI 48201 (dlevine@med shipments were made around the clock. Extra supplies
.wayne.edu).
at one hospital were shipped to another, if needed.
Clinical Infectious Diseases 2006; 42:S5–12
 2005 by the Infectious Diseases Society of America. All rights reserved.
Ultimately, reports of successful treatment were orga-
1058-4838/2006/4201S1-0003$15.00 nized and submitted to the US Food and Drug Ad-

History of Vancomycin • CID 2006:42 (Suppl 1) • S5

Downloaded from https://academic.oup.com/cid/article-abstract/42/Supplement_1/S5/275962


by guest
on 24 April 2018
Figure 1. Number of English-language articles with “vancomycin” in the title published since 1955, according to a 2004 search of Medline

ministration, and, in 1958, in the absence of an effective al- [7]. In a retrospective analysis of patients receiving vancomycin
ternative, vancomycin was immediately approved [1]. Later between 1974 and 1981, auditory toxicity was not seen. Fever
published studies confirmed the efficacy of the drug [8, 9]. and rash were uncommon (1%–3% of patients). Phlebitis af-
However, methicillin, the first semisynthetic penicillin, was fected 13% of patients and necessitated discontinuation of ther-
also approved in 1958, followed shortly thereafter by cephal- apy for 2% of patients. Nephrotoxicity was infrequent (5% of
othin. Because of perceived toxicity, vancomycin became re- patients) and reversible, but it possibly was potentiated by con-
served for patients with serious b-lactam allergies or patients comitant aminoglycoside therapy. Neutropenia occurred in 2%
with infections caused by organisms that were resistant to the of patients but was rapidly reversible [13]. Thrombocytopenia
newer agents. associated with the use of vancomycin has been studied [14,
15], as has the nephrotoxic potential of vancomycin [16–18].
TOXICITY Vancomycin rarely causes interstitial nephritis [16], and the
In the initial trials, little toxicity was observed in association combination of vancomycin and aminoglycosides has been re-
with vancomycin use. Venous irritation, chills, and rash were ported to cause renal failure, especially in adults who are re-
reported but were considered to be infusion related. Ototoxicity ceiving prolonged therapy and whose trough serum vanco-
was observed, usually presenting as tinnitus, but was attributed mycin concentrations are 110 mg/mL [17, 18]. Notably, the
to elevated serum concentrations found in patients with renal combination does not appear to cause nephrotoxicity in chil-
failure. These problems occurred more frequently in association dren [19, 20].
with the use of early preparations of vancomycin and were Rapid infusion of vancomycin has been associated with the
considered to be the result of impurities in the compound. “red man,” or “red neck,” syndrome. This syndrome, which is
Reports of ototoxicity described patients who were also re- characterized by a combination of erythema, pruritis, hypo-
ceiving known ototoxic drugs. Traber and I [10] reported the tension, and angioedema, is a histamine-like response to rapid
acute onset of tinnitus and documented hearing loss 7 days infusion, but was not a feature of early reports. Rybak et al.
after beginning therapy with vancomycin for a patient who was [21] compared the incidence of red man syndrome in patients
receiving no other ototoxic drugs. His peak serum vancomycin taking either vancomycin or teicoplanin (a glycopeptide anti-
concentration was 49.2 mg/mL, and an audiogram revealed a biotic with an antimicrobial spectrum similar to that of van-
profound high-frequency sensorineural hearing loss that slowly comycin) with the incidence of red man syndrome in healthy
improved, despite continued treatment with vancomycin at a control subjects given vancomycin or placebo. For both patients
lower dose [10]. Nevertheless, subsequent studies indicated that and control subjects, vancomycin infusions lasted 60 min and
vancomycin-induced ototoxicity was an infrequent occurrence were administered via infusion pumps with identical tubing.
[11, 12]. Infusion-related chills seemed to be eliminated by use Nine of 10 control subjects who received vancomycin experi-
of the purified commercial product, and skin rash, which was enced symptoms, versus none of the patients who received
described in early reports, has not been a persistent problem either vancomycin or teicoplanin (P ! .001) [21]. Interestingly,

S6 • CID 2006:42 (Suppl 1) • Levine

Downloaded from https://academic.oup.com/cid/article-abstract/42/Supplement_1/S5/275962


by guest
on 24 April 2018
Figure 2. Use of vancomycin in the United States, France, Italy, Germany, the United Kingdom, and The Netherlands. Reprinted from Kirst et al.
[28] with permission from the American Society for Microbiology.

there was no difference in serum histamine levels between pa- according to nomogram data, whereas others measured serum
tients and control subjects, which could be explained only by concentrations in every case. Currently, the issue is being recon-
the hypothesis that infection blunts the effect of glycopeptides. sidered, as will be discussed later in this article.
No matter how much toxicity is truly related to vancomycin,
many researchers were convinced that problems could be CLINICAL USE
avoided by careful monitoring of serum concentrations.
As noted above, shortly after being introduced, vancomycin
was eclipsed by antibiotics that were considered to be less toxic
MONITORING
and equally or more efficacious. Beginning in the early 1980s,
Measuring vancomycin levels in serum might be helpful to a dramatic increase in vancomycin use occurred, with a 1100-
prevent toxicity and to assure an adequate therapeutic drug fold increase occurring over the next 2 decades [28] (figure 2).
concentration. However, there are no definitive studies relating Two events were primarily responsible for this dramatic resur-
serum concentrations of vancomycin to treatment outcome. gence. The first was the advent of pseudomembranous enter-
Indeed, the standard regimens of 500 mg of vancomycin every ocolitis. Prior to its release, vancomycin was used to treat an
6 h or 1 g of vancomycin every 12 h appear to have been entity called “postoperative micrococcal colitis” [2]. The re-
arbitrary. However, many clinicians preferred to know whether sponse to vancomycin was excellent, and the presence of this
the regimen they prescribed achieved an expected serum con- disease, also called “acute staphylococcal ileocolitis,” became
centration, especially if dosage adjustments were necessitated an indication for the use of vancomycin [29]. Although Clos-
by renal failure. Thus, to assure a desired concentration without tridium difficile is the primary agent of pseudomembranous
actually measuring serum levels, several investigators con- enterocolitis, Staphylococcus aureus is clearly an occasional cause
structed nomograms, several of which became popular [22, 23]. of the disorder [30]. Because vancomycin is active against both
Other investigators compared individualized dosing based on pathogens and is poorly absorbed from the intestinal tract, it
the actual serum concentration with dosing based on data from became the drug of choice for treating pseudomembranous
nomograms and found the individualized method to be su- enterocolitis [31]. However, widespread vancomycin use, es-
perior [24, 25]. However, by 1994, without data to support either pecially via the oral route, was responsible, at least in part, for
a therapeutic or a toxic range, Cantu et al. [26] questioned the the development of vancomycin-resistant enterococci (VRE)
value of monitoring serum concentrations of vancomycin. Moell- (discussed below) [32]. With the recent interest in intracolonic
ering [27] agreed but recommended monitoring of serum levels administration of vancomycin, VRE will likely remain a grow-
in certain situations—for example, when patients receive van- ing problem [33, 34].
comycin/aminoglycoside combinations, when anephric patients The second event leading to increased vancomycin use was
undergoing hemodialysis receive infrequent doses of vancomy- the widespread appearance of resistant pathogens: first, meth-
cin, and when patients receive higher-than-usual doses of van- icillin-resistant S. aureus (MRSA), and, then, penicillin-resistant
comycin. With no clear consensus, many clinicians chose to dose Streptococcus pneumoniae. Known for decades as a cause of

History of Vancomycin • CID 2006:42 (Suppl 1) • S7

Downloaded from https://academic.oup.com/cid/article-abstract/42/Supplement_1/S5/275962


by guest
on 24 April 2018
nosocomial infection [35], MRSA appeared suddenly in com- Surprisingly, patients receiving vancomycin plus rifampin re-
munity isolates in Detroit and, later, throughout the world [36– mained bacteremic longer than did those receiving vancomycin
38]. With its predictable activity against MRSA, vancomycin alone, although the data were not statistically significant. In
was the drug of choice, thus beginning a new era in the history studies of coagulase-negative staphylococci, vancomycin and
of the antibiotic. rifampin are usually synergistic in vitro [45, 46]. The combi-
In 1982, Sorrell et al. [39] described 19 patients with MRSA nation is less predictable against S. aureus, and antagonism has
infection, including 10 with bacteremia. Vancomycin was been reported [47, 48]. Whether antagonism played a role in
equally as effective against MRSA as standard therapy was our study is unknown. In a search for better alternatives, Mar-
against methicillin-susceptible S. aureus, but the results may kowitz et al. [49] found vancomycin to be superior to tri-
have been influenced by adjunctive therapy with aminoglyco- methoprim-sulfamethoxazole in treating patients infected with
sides, rifampin, or trimethoprim-sulfamethoxazole. In addi- either MRSA or methicillin-susceptible S. aureus, although the
tion, although patients had peak and trough vancomycin levels, main difference was in patients infected with methicillin-sus-
which conformed to previously recommended ranges (18.0– ceptible S. aureus.
47.0 mg/mL and 4.8–12.9 mg/mL, respectively), tinnitus, rash, The emergence of penicillin-resistant S. pneumoniae presented
neutropenia, and possible nephrotoxicity were observed. At the new therapeutic challenges, especially for the treatment of men-
same time, my colleagues and I described 24 patients with ingitis. Vancomycin was well-established therapy for shunt-re-
infective endocarditis due to MRSA infection [40]. Most pa- lated meningitis, but there always was concern about its pene-
tients received other antibiotics in addition to vancomycin, so tration across the blood-brain barrier. Because penicillin-resistant
the mean duration of bacteremia (9.2 days) must be interpreted S. pneumoniae may also be cephalosporin resistant, vancomycin
in that light. One patient, the index case patient, remained plus a third-generation cephalosporin is the standard initial em-
bacteremic for 68 days without vancomycin therapy before un- pirical regimen for known or suspected cases of pneumococcal
dergoing tricuspid valve excision. Prior to surgery, she was given meningitis. Studies show variable penetration in adults, but re-
a variety of different regimens, and, each time, she appeared sults are favorable if high doses of vancomycin are used. However,
to respond initially. Therapy with vancomycin would have been concomitant steroid therapy, which is also standard for acute
started sooner, but the treating physician was reluctant to use meningitis in adults, may prevent adequate vancomycin levels in
it because of concerns about toxicity. CSF. In pediatric patients, the combination of vancomycin plus
With the increased use of vancomycin came data suggesting steroids does not appear to be harmful [50].
that vancomycin might not be equivalent to b-lactams. For
example, some patients with septic thrombophlebitis had per- RESISTANCE AND ITS IMPACT
sistent bacteremia, despite receiving treatment with heparin and With the accelerated use of vancomycin that began in the 1980s,
vancomycin. It was found that heparin causes vancomycin to it was inevitable that resistance would occur. VRE were reported
precipitate when administered through the same tubing, leading in Europe by 1986 and in the United States by 1987 [51]. Six
to subtherapeutic concentrations of vancomycin [41]. Small resistance patterns (designated “VanA” through “VanE,” and,
and Chambers [42] studied S. aureus isolates recovered from most recently, “VanG”) have been reported [52, 53]. VanA-type
patients who experienced failure of vancomycin therapy and strains are inducibly resistant to high levels of vancomycin (MIC,
found that, compared with nafcillin, vancomycin resulted in ⭓64 mg/mL) and teicoplanin (MIC, ⭓16 mg/mL). VanB-type
delayed bacterial eradication in vitro. strains are also inducibly resistant to vancomycin (MIC, 4 to
In an effort to address the question of the efficacy of van- 11024 mg/mL) but retain susceptibility to teicoplanin [54]. VanC,
comycin, my colleagues and I studied patients with endocarditis which is found in Enterococcus casseliflavus/Enterococcus flavescens
due to MRSA. We planned to compare vancomycin mono- and Enterococcus gallinarum, is constitutive and confers low-level
therapy with combination therapy involving vancomycin plus resistance to vancomycin (MIC, 4–32 mg/mL) but not to tei-
either rifampin or gentamicin. However, clinicians were reluc- coplanin [55]. VanD is also constitutive and has been found in
tant to participate, fearing that the vancomycin-gentamicin only a few isolates [55]. Like the Van A, VanB, and VanD types,
combination would prove to be too toxic. Thus, the final study the VanE type appears to be acquired and also confers low-level
consisted of only 2 treatment arms: vancomycin monotherapy resistance to vancomycin only (MIC, 16 mg/mL), as does VanG
or vancomycin plus rifampin [43]. Vancomycin monotherapy (MIC, 12–16 mg/mL). Multiple genes are responsible for these
resulted in prolonged bacteremia (9.0 days), which is consistent different phenotypes, but they produce resistance by only one or
with the results of our previous report but nearly 3 times as the other of 2 common pathways. The target for vancomycin is
long as the duration observed with nafcillin therapy [44]. Of the terminal d-alanyl-d-alanine of the growing peptidoglycan
equal importance, 6 (14%) of 43 patients experienced therapy chain. In vancomycin-resistant organisms, these peptidoglycan
failure; 3 died, and 3 required valve replacement or excision. intermediates are altered to either d-alanyl-d-lactate (VanA,

S8 • CID 2006:42 (Suppl 1) • Levine

Downloaded from https://academic.oup.com/cid/article-abstract/42/Supplement_1/S5/275962


by guest
on 24 April 2018
VanB, and VanD) or d-alanyl-d-serine (VanC, VanE, and VanG). At present, there are no standardized methods to identify
These altered targets have reduced affinity for vancomycin, re- hVISA, which makes reports of treatment failures difficult to
sulting in much higher MICs. interpret [66]. Another problem is that routine clinical labo-
Depardieu et al. [52] contributed much to the understanding ratory techniques may be inadequate to detect staphylococci
of resistance to vancomycin and helped to characterize the with reduced susceptibility to vancomycin [67]. Nevertheless,
clinical significance of each glycopeptide-resistant phenotype. several published series indicate that vancomycin is ineffective
Using a strain of VanB-resistant Enterococcus faecalis, Aslangul for the treatment of hVISA strains. Wong et al. [68] attributed
et al. [54] were able to prevent the emergence of resistance by vancomycin therapy failure to heteroresistance in 4 patients
combining teicoplanin with gentamicin; monotherapy with ei- with bacteremia. Other investigators studied paired isolates re-
ther vancomycin or teicoplanin induced resistance. Later ex- covered from a patient who experienced relapse of infection
periments with a VanB isolate proved that a 2-step mechanism after receiving vancomycin therapy for infective endocarditis.
is required for the development of resistance to the combination The relapse isolate was identical to the initial isolate, as deter-
of teicoplanin and gentamicin [56]. VanE strains responded to mined by PFGE, but resistant populations were identified. De-
high doses of glycopeptides, whereas standard doses led to treat- spite MICs in the susceptible range (MIC, ⭐2 mg/mL), when
ment failure in the rabbit model of endocarditis. Notably, be- the relapse isolate was tested in the rabbit model of endocarditis,
cause such strains may be indistinguishable from other types vancomycin failed to reduce the organism count in vegetations
of resistant enterococci in clinical practice, Lafaurie et al. [57] [69]. Charles et al. [70] compared 48 episodes of bacteremia
recommended the routine use of combination therapy with due to vancomycin-susceptible MRSA with 5 cases of bacter-
gentamicin. Finally, Lefort et al. [58] found VanD strains to be emia due to hVISA during a 12-month period. Patients infected
affected by a significant inoculum effect. Detection of such with hVISA were more likely to experience persistent fever and
strains is important, because standard doses of vancomycin or bacteremia for 17 days (P ! .001). Although patients infected
teicoplanin might result in mutants with high-level glycopep- with hVISA tended to have low trough serum vancomycin levels
tide resistance in infections with high bacterial density. during the first week of therapy, vancomycin therapy failed
Perhaps the greatest concern about VRE is the potential for even after dosage adjustments. Fridkin et al. [71] reported a
resistance to spread from enterococci to other pathogens, in case-control analysis of infections due to S. aureus with reduced
particular to S. aureus. Enterococci possess several systems, susceptibility to vancomycin (SA-RVS). For that report, re-
including plasmids and transposons, that enable the transfer duced susceptibility was defined as an MIC of ⭓4 mg/mL and
of genetic material to other bacteria. Although the transfer of included isolates with MICs of 8–16 mg/mL, which are generally
resistance from enterococci to S. aureus was demonstrated in referred to as VISA strains. Patients infected with SA-RVS had
vitro in 1992 [59], some researchers questioned whether it a crude 6-month mortality rate of ∼80%, similar to patients
could occur in natural conditions. Intermediate resistance to infected with VISA [71]. Patients infected with SA-RVS also
vancomycin had already been reported among staphylococci, had a higher in-hospital mortality rate. Risk factors for SA-
but it was not caused by transfer of genetic material from RVS infection were antecedent vancomycin use and MRSA in-
enterococci. The first staphylococci with reduced susceptibility fection in the preceding 2 or 3 months. In another study of
to vancomycin were reported from Japan in 1997 [60, 61]. patients with serious infections due to SA-RVS, 76% experi-
Additional strains soon appeared in the United States. The enced glycopeptide therapy failure, and 40% died [72].
MICs of these isolates range from 4 to 8 mg/mL; hence, they Currently, there is growing interest in another aspect of
are called “vancomycin-intermediate S. aureus” (VISA). These staphylococcal function that affects therapeutic outcome. Ac-
strains produce an excess amount of non–cross-linked d-alanyl- cessory gene regulator group II polymorphism, which is dis-
d-alanine, which captures vancomycin molecules, thus pre- cussed in more detail by Sakoulas et al. [73] and Stevens [74]
venting them from reaching the target site [62]. Infection with elsewhere in this supplement and which may also play an im-
VISA has been associated with treatment failure [63], but these portant role in vancomycin resistance, has recently been rec-
organisms are rare and, thus far, have not had a major clinical ognized as a risk factor for vancomycin therapy failure in MRSA
impact [64]. More common are strains of MRSA that are “het- infection [75, 76].
eroresistant” to vancomycin (hVISA); such strains now account The question regarding the transfer of resistance from en-
for up to 26% of MRSA isolates in Japan [65]. The vancomycin terococcus to S. aureus in nature was answered when the first
MIC of hVISA is within the susceptible range (⭐4 mg/mL), but vancomycin-resistant S. aureus (VRSA; MIC, ⭓32 mg/mL) iso-
these strains contain subpopulations that exhibit reduced sus- late was recovered from a patient in Michigan. The patient had
ceptibility. These organisms have additional importance, be- diabetes, peripheral vascular disease, renal failure, and chronic
cause they may be precursors to VISA [63, 66]. If so, the fact infected foot ulcers that were treated intermittently with van-
that they are becoming widespread is of great concern. comycin. In addition to VRSA, a VRE isolate was also recovered

History of Vancomycin • CID 2006:42 (Suppl 1) • S9

Downloaded from https://academic.oup.com/cid/article-abstract/42/Supplement_1/S5/275962


by guest
on 24 April 2018
from a foot lesion; this is a noteworthy fact because, by this ago, might be required, but it might also extend the useful life
time, Enterococcus faecium had largely supplanted E. faecalis as of this antibiotic. After 50 years, we are learning more about
the predominant species found in hospital settings. It appears vancomycin than ever before, and this knowledge may lead us
that the staphylococcus acquired the VanA gene from the VRE to a much more successful approach to patient care. Even after
[77, 78]. A second VRSA strain was soon isolated from a patient 50 years, there is still much to add to the history of vancomycin.
in Pennsylvania, who also had received intermittent courses of
vancomycin, but none during the 5 years prior to developing Acknowledgment
VRSA infection [79]. She was colonized with VRE and MRSA,
Potential conflict of interest. D.P.L. receives grant/research support
both of which were isolated on multiple occasions. As in the from Cubist, Nabi, and Wyeth and is a consultant for Cubist.
Michigan case, it appears that the MRSA acquired VanA from
the VRE, even though the patient was not being treated with References
vancomycin at the time [80, 81]. In April 2004, a third VRSA
1. Anderson RCGR, Higgins HM Jr, Pettinga CD. Symposium: how a
strain (vancomycin MIC, 64 mg/mL) was isolated from a pa- drug is born. Cincinnati J Med 1961; 42:49–60.
tient in New York [82]. This strain was found to be vancomycin 2. Geraci JE, Heilman FR, Nichols DR, Ross GT, Wellman WE. Some
susceptible, as determined by standard automated antimi- laboratory and clinical experiences with a new antibiotic, vancomycin.
Mayo Clin Proc 1956; 31:564–82.
crobial susceptibility testing. The observation that automated
3. Geraci JE, Wilson WR. Vancomycin therapy for infective endocarditis.
techniques might misinterpret antibiotic susceptibility among Rev Infect Dis 1981; 3(Suppl):S250–8.
staphylococci prompted the Centers for Disease Control and 4. McGuire JM, Wolfe RN, Ziegler DW. Vancomycin, a new antibiotic.
Prevention to publish new guidelines for laboratory testing of II. In vitro antibacterial studies. Antibiot Annu 1955; 3:612–8.
5. Griffith RS. Vancomycin: continued clinical studies. Antibiot Annu
S. aureus isolates [83]. 1956–1957:118–22.
As we advance further into the era of vancomycin resistance, 6. Griffith RS, Peck FB Jr. Vancomycin, a new antibiotic. III. Preliminary
it is important to recall that failure of vancomycin therapy may clinical and laboratory studies. Antibiot Annu 1955; 3:619–22.
7. Geraci JE, Heilman FR, Nichols DR, Wellman WE. Antibiotic therapy
still occur in infections due to susceptible organisms. Fowler, of bacterial endocarditis. VII. Vancomycin for acute micrococcal en-
Jr. et al. [84] found vancomycin therapy to be an independent docarditis: preliminary report. Mayo Clin Proc 1958; 33:172–81.
risk factor for recurrent S. aureus bacteremia, and, in another 8. Geraci JE, Heilman FR. Vancomycin in the treatment of staphylococcal
endocarditis. Prog Neurobiol 1960; 35:316–26.
study, vancomycin treatment was cited as a potential reason
9. Kirby WM, Perry DM, Bauer AW. Treatment of staphylococcal sep-
for increased mortality among patients with MRSA infection ticemia with vancomycin: report of thirty-three cases. N Engl J Med
[85]. Recent evidence suggests that outcomes might be deter- 1960; 262:49–55.
mined by even slight differences in the MIC. In a recent study 10. Traber PG, Levine DP. Vancomycin ototoxicity in patient with normal
renal function. Ann Intern Med 1981; 95:458–60.
by Fowler, Jr. et al. [76], patients with bacteremia due to MRSA 11. Brummett RE, Fox KE. Vancomycin- and erythromycin-induced hear-
with an MIC of ⭐0.05 mg/mL had a 55.6% success rate, com- ing loss in humans. Antimicrob Agents Chemother 1989; 33:791–6.
pared with a 9.5% success rate among patients whose bacter- 12. Brummett RE. Ototoxicity of vancomycin and analogues. Otolaryngol
Clin North Am 1993; 26:821–8.
emia was due to MRSA with an MIC of 1–2 mg/mL (P p .03). 13. Farber BF, Moellering RC Jr. Retrospective study of the toxicity of
Such a result might be avoided if there were reliable data in- preparations of vancomycin from 1974 to 1981. Antimicrob Agents
dicating what serum vancomycin level predicts therapeutic suc- Chemother 1983; 23:138–41.
14. Marraffa J, Guharoy R, Duggan D, Rose F, Nazeer S. Vancomycin-
cess (i.e., the therapeutic concentration). Knowledge of the
induced thrombocytopenia: a case proven with rechallenge. Pharma-
therapeutic concentration and the MIC of the pathogen might cotherapy 2003; 23:1195–8.
overcome the intrinsic deficiencies of the drug and improve its 15. Zenon GJ, Cadle RM, Hamill RJ. Vancomycin-induced thrombocy-
topenia. Arch Intern Med 1991; 151:995–6.
utility. As an example, the serum vancomycin level was noted
16. Bergman MM, Glew RH, Ebert TH. Acute interstitial nephritis asso-
to play a role in the treatment of pneumonia. Penetration of ciated with vancomycin therapy. Arch Intern Med 1988; 148:2139–40.
vancomycin into lung tissue is poor [86], resulting in thera- 17. Cimino MA, Rotstein C, Slaughter RL, Emrich LJ. Relationship of
peutic failure in patients with pneumonia [87]. However, when serum antibiotic concentrations to nephrotoxicity in cancer patients
receiving concurrent aminoglycoside and vancomycin therapy. Am J
the serum concentration was maintained at a level sufficiently Med 1987; 83:1091–7.
above the MIC of the infecting organism, the outcome was 18. Rybak MJ, Albrecht LM, Boike SC, Chandrasekar PH. Nephrotoxicity
significantly improved. Moise et al. [88] showed that 78% of of vancomycin, alone and with an aminoglycoside. J Antimicrob Che-
mother 1990; 25:679–87.
the patients being treated for pneumonia who achieved an area
19. Goren MP, Baker DK Jr, Shenep JL. Vancomycin does not enhance
under the inhibition curve of 1345 survived, versus 24% of the amikacin-induced tubular nephrotoxicity in children. Pediatr Infect
patients with an area under the inhibition curve of ⭐345. Fu- Dis J 1989; 8:278–82.
20. Nahata MC. Lack of nephrotoxicity in pediatric patients receiving con-
ture studies must address whether specific therapeutic ranges
current vancomycin and aminoglycoside therapy. Chemotherapy
can be determined for different types of infections. A return 1987; 33:302–4.
to monitoring serum levels, a practice almost abandoned long 21. Rybak MJ, Bailey EM, Warbasse LH. Absence of “red man syndrome”

S10 • CID 2006:42 (Suppl 1) • Levine

Downloaded from https://academic.oup.com/cid/article-abstract/42/Supplement_1/S5/275962


by guest
on 24 April 2018
in patients being treated with vancomycin or high-dose teicoplanin. 45. Ein ME, Smith NJ, Aruffo JF, Heerema MS, Bradshaw MW, Williams
Antimicrob Agents Chemother 1992; 36:1204–7. TW Jr. Susceptibility and synergy studies of methicillin-resistant Staph-
22. Matzke GR, McGory RW, Halstenson CE, Keane WF. Pharmacokinetics ylococcus epidermidis. Antimicrob Agents Chemother 1979; 16:655–9.
of vancomycin in patients with various degrees of renal function. An- 46. Lowy FD, Wexler MA, Steigbigel NH. Therapy of methicillin-resistant
timicrob Agents Chemother 1984; 25:433–7. Staphylococcus epidermidis experimental endocarditis. J Lab Clin Med
23. Moellering RC Jr, Krogstad DJ, Greenblatt DJ. Vancomycin therapy in 1982; 100:94–104.
patients with impaired renal function: a nomogram for dosage. Ann 47. Hackbarth CJ, Chambers HF, Sande MA. Serum bactericidal activity
Intern Med 1981; 94:343–6. of rifampin in combination with other antimicrobial agents against
24. Rybak MJ, Boike SC. Monitoring vancomycin therapy. Drug Intell Clin Staphylococcus aureus. Antimicrob Agents Chemother 1986; 29:611–3.
Pharm 1986; 20:757–61. 48. Watanakunakorn C, Guerriero JC. Interaction between vancomycin
25. Rybak MJ, Boike SC. Individualized adjustment of vancomycin dosage: and rifampin against Staphylococcus aureus. Antimicrob Agents Che-
comparison with two dosage nomograms. Drug Intell Clin Pharm mother 1981; 19:1089–91.
1986; 20:64–8. 49. Markowitz N, Quinn EL, Saravolatz LD. Trimethoprim-sulfamethox-
26. Cantu TG, Yamanaka-Yuen NA, Lietman PS. Serum vancomycin con- azole compared with vancomycin for the treatment of Staphylococcus
centrations: reappraisal of their clinical value. Clin Infect Dis 1994; 18: aureus infection. Ann Intern Med 1992; 117:390–8.
533–43. 50. Ahmed A. A critical evaluation of vancomycin for treatment of bacterial
27. Moellering RC Jr. Monitoring serum vancomycin levels: climbing the meningitis. Pediatr Infect Dis J 1997; 16:895–903.
mountain because it is there? Clin Infect Dis 1994; 18:544–6. 51. Murray BE. Vancomycin-resistant enterococcal infections. N Engl J
28. Kirst HA, Thompson DG, Nicas TI. Historical yearly usage of van- Med 2000; 342:710–21.
comycin. Antimicrob Agents Chemother 1998; 42:1303–4. 52. Depardieu F, Bonora MG, Reynolds PE, Courvalin P. The vanG gly-
29. Geraci JE. Vancomycin. Mayo Clin Proc 1977; 52:631–4. copeptide resistance operon from Enterococcus faecalis revisited. Mol
30. Gravet A, Rondeau M, Harf-Monteil C, et al. Predominant Staphy- Microbiol 2003; 50:931–48.
lococcus aureus isolated from antibiotic-associated diarrhea is clinically 53. McKessar SJ, Berry AM, Bell JM, Turnidge JD, Paton JC. Genetic
relevant and produces enterotoxin A and the bicomponent toxin LukE- characterization of vanG, a novel vancomycin resistance locus of En-
LukD. J Clin Microbiol 1999; 37:4012–9. terococcus faecalis. Antimicrob Agents Chemother 2000; 44:3224–8.
31. Fekety R, Shah AB. Diagnosis and treatment of Clostridium difficile 54. Aslangul E, Baptista M, Fantin B, et al. Selection of glycopeptide-
colitis. JAMA 1993; 269:71–5. resistant mutants of VanB-type Enterococcus faecalis BM4281 in vitro
32. Gerding DN. Is there a relationship between vancomycin-resistant en- and in experimental endocarditis. J Infect Dis 1997; 175:598–605.
terococcal infection and Clostridium difficile infection? Clin Infect Dis 55. Cetinkaya Y, Falk P, Mayhall CG. Vancomycin-resistant enterococci.
1997; 25(Suppl 2):S206–10. Clin Microbiol Rev 2000; 13:686–707.
33. Apisarnthanarak A, Khoury H, Reinus WR, Crippin JS, Mundy LM. 56. Lefort A, Baptista M, Fantin B, et al. Two-step acquisition of resistance
Severe Clostridium difficile colitis: the role of intracolonic vancomycin? to the teicoplanin-gentamicin combination by VanB-type Enterococcus
Am J Med 2002; 112:328–9. faecalis in vitro and in experimental endocarditis. Antimicrob Agents
34. Apisarnthanarak A, Razavi B, Mundy LM. Adjunctive intracolonic van- Chemother 1999; 43:476–82.
comycin for severe Clostridium difficile colitis: case series and review 57. Lafaurie M, Perichon B, Lefort A, Carbon C, Courvalin P, Fantin B.
of the literature. Clin Infect Dis 2002; 35:690–6. Consequences of VanE-type resistance on efficacy of glycopeptides in
35. Peacock JE Jr, Marsik FJ, Wenzel RP. Methicillin-resistant Staphylo- vitro and in experimental endocarditis due to Enterococcus faecalis.
coccus aureus: introduction and spread within a hospital. Ann Intern Antimicrob Agents Chemother 2001; 45:2826–30.
Med 1980; 93:526–32. 58. Lefort A, Garry L, Depardieu F, Courvalin P, Fantin B. Influence of
36. Centers for Disease Control (CDC). Community-acquired methicillin- VanD type resistance on activities of glycopeptides in vitro and in
resistant Staphylococcus aureus infections—Michigan. MMWR Morb experimental endocarditis due to Enterococcus faecium. Antimicrob
Mortal Wkly Rep 1981; 30:185–7. Agents Chemother 2003; 47:3515–8.
37. Saravolatz LD, Pohlod DJ, Arking LM. Community-acquired methi- 59. Noble WC, Virani Z, Cree RG. Co-transfer of vancomycin and other
cillin-resistant Staphylococcus aureus infections: a new source for nos- resistance genes from Enterococcus faecalis NCTC 12201 to Staphylo-
ocomial outbreaks. Ann Intern Med 1982; 97:325–9. coccus aureus. FEMS Microbiol Lett 1992; 72:195–8.
38. Saravolatz LD, Markowitz N, Arking L, Pohlod D, Fisher E. Methicillin- 60. Centers for Disease Control and Prevention (CDC). Reduced suscep-
resistant Staphylococcus aureus: epidemiologic observations during a tibility of Staphylococcus aureus to vancomycin—Japan, 1996. MMWR
community-acquired outbreak. Ann Intern Med 1982; 96:11–6. Morb Mortal Wkly Rep 1997; 46:624–6.
39. Sorrell TC, Packham DR, Shanker S, Foldes M, Munro R. Vancomycin 61. Hiramatsu K, Hanaki H, Ino T, Yabuta K, Oguri T, Tenover FC. Meth-
therapy for methicillin-resistant Staphylococcus aureus. Ann Intern Med icillin-resistant Staphylococcus aureus clinical strain with reduced van-
1982; 97:344–50. comycin susceptibility. J Antimicrob Chemother 1997; 40:135–6.
40. Levine DP, Cushing RD, Jui J, Brown WJ. Community-acquired meth- 62. Sieradzki K, Roberts RB, Haber SW, Tomasz A. The development of
icillin-resistant Staphylococcus aureus endocarditis in the Detroit Med- vancomycin resistance in a patient with methicillin-resistant Staphy-
ical Center. Ann Intern Med 1982; 97:330–8. lococcus aureus infection. N Engl J Med 1999; 340:517–23.
41. Barg NL, Supena RB, Fekety R. Persistent staphylococcal bacteremia 63. Fridkin SK. Vancomycin-intermediate and -resistant Staphylococcus au-
in an intravenous drug abuser. Antimicrob Agents Chemother 1986; reus: what the infectious disease specialist needs to know. Clin Infect
29:209–11. Dis 2001; 32:108–15.
42. Small PM, Chambers HF. Vancomycin for Staphylococcus aureus en- 64. Tenover FC, Biddle JW, Lancaster MV. Increasing resistance to van-
docarditis in intravenous drug users. Antimicrob Agents Chemother comycin and other glycopeptides in Staphylococcus aureus. Emerg Infect
1990; 34:1227–31. Dis 2001; 7:327–32.
43. Levine DP, Fromm BS, Reddy BR. Slow response to vancomycin or 65. Schwaber MJ, Wright SB, Carmeli Y, et al. Clinical implications of
vancomycin plus rifampin in methicillin-resistant Staphylococcus aureus varying degrees of vancomycin susceptibility in methicillin-resistant
endocarditis. Ann Intern Med 1991; 115:674–80. Staphylococcus aureus bacteremia. Emerg Infect Dis 2003; 9:657–64.
44. Korzeniowski O, Sande MA. Combination antimicrobial therapy for 66. Liu C, Chambers HF. Staphylococcus aureus with heterogeneous resis-
Staphylococcus aureus endocarditis in patients addicted to parenteral tance to vancomycin: epidemiology, clinical significance, and critical
drugs and in nonaddicts: a prospective study. Ann Intern Med 1982; assessment of diagnostic methods. Antimicrob Agents Chemother
97:496–503. 2003; 47:3040–5.

History of Vancomycin • CID 2006:42 (Suppl 1) • S11

Downloaded from https://academic.oup.com/cid/article-abstract/42/Supplement_1/S5/275962


by guest
on 24 April 2018
67. Tenover FC, Lancaster MV, Hill BC, et al. Characterization of staph- resistant Staphylococcus aureus containing the vanA resistance gene. N
ylococci with reduced susceptibilities to vancomycin and other gly- Engl J Med 2003; 348:1342–7.
copeptides. J Clin Microbiol 1998; 36:1020–7. 79. Miller D, Urdaneta V, Weltman A. Vancomycin-resistant Staphylococcus
68. Wong SS, Ng TK, Yam WC, et al. Bacteremia due to Staphylococcus aureus—Pennsylvania, 2002. MMWR Morb Mortal Wkly Rep 2002;
aureus with reduced susceptibility to vancomycin. Diagn Microbiol 51:902.
Infect Dis 2000; 36:261–8. 80. Whitener CJ, Park SY, Browne FA, et al. Vancomycin-resistant Staph-
69. Moore MR, Perdreau-Remington F, Chambers HF. Vancomycin treat- ylococcus aureus in the absence of vancomycin exposure. Clin Infect
ment failure associated with heterogeneous vancomycin-intermediate Dis 2004; 38:1049–55.
Staphylococcus aureus in a patient with endocarditis and in the rabbit 81. Tenover FC, Weigel LM, Appelbaum PC, et al. Vancomycin-resistant
model of endocarditis. Antimicrob Agents Chemother 2003; 47:1262–6. Staphylococcus aureus isolate from a patient in Pennsylvania. Antimi-
70. Charles PG, Ward PB, Johnson PD, Howden BP, Grayson ML. Clinical crob Agents Chemother 2004; 48:275–80.
features associated with bacteremia due to heterogeneous vancomycin- 82. Kacica M, McDonald LC. Vancomycin-resistant Staphylococcus au-
intermediate Staphylococcus aureus. Clin Infect Dis 2004; 38:448–51. reus—New York, 2004. MMWR Morb Mortal Wkly Rep 2004; 53:
71. Fridkin SK, Hageman J, McDougal LK, et al. Epidemiological and 322–3.
microbiological characterization of infections caused by Staphylococcus 83. Centers for Disease Control and Prevention. Investigation and control
aureus with reduced susceptibility to vancomycin, United States,
of vancomycin-intermediate and -resistant Staphylococcus aureus: a
1997–2001. Clin Infect Dis 2003; 36:429–39.
guide for health departments and infection control personnel. Atlanta:
72. Howden BP, Ward PB, Charles PG, et al. Treatment outcomes for
Centers for Disease Control and Prevention, 2004.
serious infections caused by methicillin-resistant Staphylococcus aureus
84. Fowler VG Jr, Kong LK, Corey GR, et al. Recurrent Staphylococcus
with reduced vancomycin susceptibility. Clin Infect Dis 2004; 38:521–8.
aureus bacteremia: pulsed-field gel electrophoresis findings in 29 pa-
73. Sakoulas G, Moellering RC Jr, Eliopoulos GM. Adaptation of methi-
tients. J Infect Dis 1999; 179:1157–61.
cillin-resistant Staphylococcus aureus in the face of vancomycin therapy.
85. Cosgrove SE, Sakoulas G, Perencevich EN, Schwaber MJ, Karchmer
Clin Infect Dis 2006; 42(Suppl 1):S40–50 (in this supplement).
AW, Carmeli Y. Comparison of mortality associated with methicillin-
74. Stevens DL. The role of vancomycin in the treatment paradigm. Clin
Infect Dis 2006; 42(Suppl 1):S51–7 (in this supplement). resistant and methicillin-susceptible Staphylococcus aureus bacteremia:
75. Moise-Broder PA, Sakoulas G, Eliopoulos GM, Schentag JJ, Forrest A, a meta-analysis. Clin Infect Dis 2003; 36:53–9.
Moellering RC Jr. Accessory gene regulator group II polymorphism in 86. Cruciani M, Gatti G, Lazzarini L, et al. Penetration of vancomycin into
methicillin-resistant Staphylococcus aureus is predictive of failure of human lung tissue. J Antimicrob Chemother 1996; 38:865–9.
vancomycin therapy. Clin Infect Dis 2004; 38:1700–5. 87. Gonzalez C, Rubio M, Romero-Vivas J, Gonzalez M, Picazo JJ. Bac-
76. Fowler VG Jr, Sakoulas G, McIntyre LM, et al. Persistent bacteremia teremic pneumonia due to Staphylococcus aureus: a comparison of
due to methicillin-resistant Staphylococcus aureus infection is associated disease caused by methicillin-resistant and methicillin-susceptible or-
with agr dysfunction and low-level in vitro resistance to thrombin- ganisms. Clin Infect Dis 1999; 29:1171–7.
induced platelet microbicidal protein. J Infect Dis 2004; 190:1140–9. 88. Moise PA, Forrest A, Bhavnani SM, Birmingham MC, Schentag JJ.
77. Sievert DM, Boulton ML, Stolzman G, et al. Staphylococcus aureus Area under the inhibitory curve and a pneumonia scoring system for
resistant to vancomycin—United States, 2002. MMWR Morb Mortal predicting outcomes of vancomycin therapy for respiratory infections
Wkly Rep 2002; 51:565–7. by Staphylococcus aureus. Am J Health Syst Pharm 2000; 57(Suppl 2):
78. Chang S, Sievert DM, Hageman JC, et al. Infection with vancomycin- S4–9.

S12 • CID 2006:42 (Suppl 1) • Levine

Downloaded from https://academic.oup.com/cid/article-abstract/42/Supplement_1/S5/275962


by guest
on 24 April 2018

You might also like