SlideShare a Scribd company logo
1 of 28
Download to read offline
Crit Care Clin 24 (2008) 393–420




      Vancomycin Revisited: A Reappraisal
               of Clinical Use
                   Burke A. Cunha, MD, MACPa,b,*
                  a
                  Infectious Disease Division, Winthrop-University Hospital,
                                  Mineola, NY 11501, USA
         b
          State University of New York, School of Medicine, Stony Brook, NY, USA



    Over the years, vancomycin has become the mainstay of methicillin-
resistant Staphylococcus aureus (MRSA) therapy. Although vancomycin is
active against a variety of other gram-positive organisms, other antibiotics
are preferred to treat infections due to these organisms (Table 1). In critical
care medicine, vancomycin has been mainly used to treat infections where
MRSA is the presumed cause of infection. These include central intravenous
line infections, soft tissue infections, bone infections, shunt infections in
hemodialysis patients, bacteremia, and acute bacterial endocarditis. The
widespread use of empiric vancomycin has resulted in adverse effects [1–5].
    The initial unpurified formulations of vancomycin were associated with
reports of potential nephrotoxicity [6,7]. Subsequently, the purified prepara-
tions of vancomycin have been used and have not been associated with neph-
rotoxicity. There is no evidence for vancomycin monotherapy–associated
nephrotoxicity [8,9]. The few reports of potential vancomycin nephrotoxicity
described patients receiving vancomycin and known nephrotoxic medica-
tions. Vancomycin plus an aminoglycoside does not appear to increase the
nephrotoxic potential of the aminoglycoside [10–15]. Endotoxin or cytokine
release from gram-negative bacilli treated with an aminoglycoside and van-
comycin may be responsible for an increase in creatinine in some cases, which
may have been mistakenly ascribed to vancomycin toxicity [16]. Because van-
comycin monotherapy is not nephrotoxic, the use of vancomycin serum
levels to avoid nephrotoxicity has no basis [3,17,18]. Because vancomycin
is renally eliminated by glomerular filtration, vancomycin dosing in renal in-
sufficiency can be accurately dosed based on the creatinine clearance (CrCl),
(ie, the daily dose of vancomycin should be reduced in proportion to the


  * Infectious Disease Division, Winthrop-University Hospital, 259 First Street, Mineola,
NY 11501.

0749-0704/08/$ - see front matter Ó 2008 Elsevier Inc. All rights reserved.
doi:10.1016/j.ccc.2007.12.012                                         criticalcare.theclinics.com
394                                        CUNHA


Table 1
Vancomycin: clinically useful microbiologic spectrum
Clinically effective                                         Clinically ineffective
Gram-positive aerobic cocci                                 Gram-negative aerobic cocci
  Staphylococci (MSSA/MRSA),                                 Neisseria meningitidis
  Nonenterococcal streptococci (Groups A, B, C, G)
  Staphylococcus epidermidis (coagulase negative
   staphylococci) (MSSE/MRSE)
  Penicillin-resistant Streptococcus pneumoniae
Gram-positive anaerobic cocci                               Gram-negative aerobic bacilli
  Peptococcus                                                Escherichia coli
  Peptostreptococcus                                         Klebsiella pneumoniae
                                                              Pseudomonas aeruginosa
Gram-positive aerobic bacilli                               Gram-positive aerobic bacilli
  Corynebacterium sp                                         Listeria monocytogenes
  Lactobacillus sp
Gram-positive anaerobic bacilli                             Gram-negative anaerobic bacilli
  Clostridium sp                                             Bacteroides fragilis
Group D enterococci                                         Group D enterococci
  Enterococcus faecalis (VSE)a                               Enterococcus faecium (VRE)
   Abbreviations: MRSE, methicillin-resistant Staphylococcus epidermidis; MSSA, methicillin-
sensitive Staphylococcus aureus; MSSE, methicillin-sensitive Staphylococcus epidermidis; VRE,
vancomycin-resistant enterococci; VSE, vancomycin-sensitive enterococci.
   a
      Only when combined with an aminoglycoside (eg, gentamicin).
   Data from Refs. [1,3,4].


decrease in renal function). In those responding to vancomycin therapy and
in those with a normal volume of distribution (Vd), vancomycin levels are
unhelpful, expensive, and unnecessary for vancomycin dosing [17–21].
    The pharmacokinetic and pharmacodynamic characteristics of vancomy-
cin have been well studied. Vancomycin obeys both ‘‘concentration depen-
dent’’ kinetics at concentrations O MIC and ‘‘concentration independent’’
kinetics at concentrations ! MIC [1,3,19,22]. Because nephrotoxicity is not
a consideration, high-dose vancomycin (eg, 2 g intravenously every 12 hours
[60 mg/kg/d]) has been used in special situations, eg, osteomyelitis without
nephrotoxicity [23,24]. After years of clinical experience, vancomycin side
effects have become more fully appreciated. In addition to ‘‘red neck’’ or
‘‘red man’’ syndrome, leukopenia, thrombocytopenia, and, rarely, sudden
death have been ascribed to vancomycin [1,3,25].
    Extensive vancomycin use over the years has resulted in two major prob-
lems. Firstly, excessive use of vancomycin has resulted in an increased
prevalence of vancomycin-resistant enterococci (VRE) worldwide. Vanco-
mycin-sensitive enterococci (VSE) represent the main group D enterococcal
component of feces. Vancomycin has sufficient anti-VSE activity to decrease
VSE in the fecal flora, resulting in a commensurate increase VRE in stools
[26–30]. Although the spectrum of infection caused by VSE and VRE are
the same, the number of antimicrobials available to treat VRE is limited,
making the therapy of VRE more difficult than the therapy of VSE [31,32].
VANCOMYCIN REVISITED                           395

   Another negative effect of vancomycin use over the years has been a rel-
ative increase S aureus resistance [33–37]. Vancomycin therapy results in
cell-wall thickening of S aureus strains, of both methicillin-sensitive S aureus
(MSSA) and methicillin-resistant S aureus (MRSA). Vancomycin-mediated
cell-wall thickening results in ‘‘permeability mediated’’ resistance to vanco-
mycin as well as to other anti-MSSA and anti-MRSA antibiotics [38–42].
Vancomycin-induced ‘‘permeability-mediated’’ resistance is manifested mi-
crobiologically by increased minimum inhibitory concentrations (MICs)
and clinically by delayed resolution or therapeutic failure in treating staph-
ylococcal bacteremias or acute bacterial endocarditis [43–48].
   To avoid selecting out heteroresistant vancomycin-intermediate S aureus
(hVISA), vancomycin use should be minimized and other MRSA antibiotics
should preferentially be used instead (eg, minocycline, daptomycin, line-
zolid, or tigecycline) [1,31,32].

Vancomycin pharmacokinetics and pharmacodynamics
   Although available orally and intravenously, vancomycin is primarily
administered via the intravenous route in the critical care setting. Oral van-
comycin is the preferred therapy for Clostridium difficile diarrhea because it
is not absorbed, is active against C difficile, and achieves high intraluminal
concentrations in the colon [1,3,32]. Because intravenous vancomycin does
not penetrate into the bowel lumen, intravenous vancomycin has no role
in the treatment of C difficile diarrhea or colitis [1,3,5,49].
   Vancomycin is usually administered as 1 g (intravenous) every 12 hours.
After a 1-g intravenous dose, vancomycin serum concentrations are predict-
ably R25 mg/mL [1,3,5,50]. High dose of vancomycin (eg, 2 g intravenously
every 12 hours) has been used in the treatment of central nervous system
(CNS) infections to overcome the relatively poor cerebrospinal fluid (CSF)
penetration of vancomycin (CSF penetration with noninflamed meninges is
0% and with inflamed meninges is only 15% of simultaneous serum concen-
trations) and osteomyelitis (achieves serum trough levels R 15 mg/mL
[1,32,51,52]. For cardiopulmonary bypass (CBP) prophylaxis, use 1 g intrave-
nously preoperatively because vancomycin is rapidly removed during CBP
[53,54]. Burn patients have increased Vd and may require higher doses of van-
comycin. Intravenous drug abusers (IVDAs) have higher vancomycin renal
clearances than do non-IVDAs, and may require higher doses (Box 1) [1].
   Vancomycin is not removed by hemodialysis [1,3]. In anuric patients
a 1 g (IV) dose results in peak serum concentrations of about 48 mg/mL.
Vancomycin’s mean elimination half-life is 7.5 days (vancomycin serum
concentration is 3.5 mg/mL after 18 days) [1,32,55,56]. In patients using
new ‘‘high flux’’ membranes for hemodialysis, about 40% of vancomycin
is removed. Therefore, a post–‘‘high flux’’ hemodialysis dose (eg, 500 mg
intravenously, which is w50% of the anuric dose) should be administered
post-hemodialysis [57].
396                                    CUNHA




  Box 1. Vancomycin: pharmacokinetic and pharmacodynamic
  characteristics
  Pharmacokinetic parameters
     Usual adult dose: 1 g intravenously every 12 hours (30 mg/kg/
        d); 2 g intravenously every 12 hours (60 mg/kg/d)
     Peak serum levels: 63 mg/mL (1-g dose); 120 mg/mL (2-g dose)
     Excreted unchanged (urine): 90%
     Serum half-life: 6 hours (normal); 180 hours (end-stage renal
        disease)
     Plasma protein binding: 55%
     Vd: 0.7 L/kg
  Therapeutic serum levels:
     Peak 1 g  25 mg/mL; 2g  50 mg/mL
     Trough: 1 g ‚ 7.5 m/mL; 2 g ‚ 15 m/mL
  Pharmacodynamic characteristics
     ‘‘Concentration dependent’’ kinetics
         at concentrations  MIC
     ‘‘Concentration independent’’ kinetics
         (time dependent) at concentrations  MIC
  Primary mode of elimination: renal
  Dosage adjustments:
     CrCl 50–80 mL/min: dose = 500 mg intravenously every
        12 hours
     CrCl 10–50 mL/min: dose = 500 mg intravenously every
        24 hours
     CrCl 10 mL/min: dose = 1 g intravenous every week
     Post-hemodialysis dose: none
     Post–‘‘high flux’’ hemodialysis dose: 500 mg intravenous
        every 12 hours
     Post–peritoneal dialysis dose: none
     CVVH dose: 1 gm intravenous every 24 hours

     Data from Cunha BA, editor. Antibiotic essentials, 7th edition. Royal Oak (MI):
  Physicians Press; 2008; with permission.



   Peritoneal dialysis (PD) removes no vancomycin. Therefore, after an ini-
tial 1-g intravenous dose, vancomycin should be dosed for a CrCl less than
10 mL/min and no post-PD dose is necessary [1,32]. For patients with peri-
tonitis on chronic ambulatory peritoneal dialysis (CAPD), vancomycin may
be added to peritoneal dialysis fluid. A 1-g intravenous loading dose of
vancomycin may be given to such patients after dialysis, followed by the
renally adjusted maintenance anuric dose [1,32]. In addition, to maintain
equilibrium between the dialysate fluid and the serum compartment,
VANCOMYCIN REVISITED                         397

vancomycin may also be given intraperitoneally (2–30 mg of vancomycin
per liter of dialysis fluid) with each CAPD [1,32]. Vancomycin has been
used to treat CNS infections, but CNS penetration of vancomycin is vari-
able. Vancomycin administered intravenously achieves only about 15% of
simultaneous serum levels in the CSF in patients with meningitis. If high
CSF concentrations are desired for treatment of gram-positive shunt infec-
tions, give 2 g intravenously every 12 hours until shunt removal. Intrave-
nous vancomycin may be supplemented by intrathecal doses (ie,
vancomycin 10–20 mg/d intrathecally), preferably administered via Om-
maya reservoir [1,3,32,58].
   With normal renal function, about 90% of intravenous vancomycin is
eliminated unchanged in the urine during the first 24 hours. After a single
1-g intravenous dose of vancomycin, urine concentrations are w500 mg/mL,
which are maintained for 24 hours [1,3]. Intravenously administered van-
comycin does not concentrate well into, bile (30%), synovial fluid, pleural
fluid, or lung parenchyma [1,58–62].
   Intravenous vancomycin diffuses well into most other body compart-
ments except feces, aqueous humor, and CSF [1,60]. While vancomycin pen-
etrates poorly into CSF, it penetrates well into brain tissue (Box 2)
[1,6,32,51,52,58].

Vancomycin levels
    Vancomycin is eliminated by glomerular filtration. The serum half-life of
IV vancomycin is 6 hours with normal renal function, and 7 days in anuria
[1,6,56,62]. Many approaches to vancomycin dosing for various degrees of
renal insufficiency have been devised and all are based on CrCl [1,6,21].
Formerly, vancomycin was administered as 500 mg intravenously every
6 hours. Today vancomycin is usually administered as 1 g intravenously
every 12 hours (for adults with a normal Vd and CrCl) [22,63,64]. For seri-
ous systemic infections due to susceptible organisms, is to administer vanco-
mycin 1 g intravenously every 12 hours (15 mg/kg/d) or 2 g intravenously
every 12 hours (30 mg/kg/d) [1,3,32]. Even when used in prolonged high
doses for special situations, such as for MRSA osteomyelitis (2 g intrave-
nously every 12 hours [30 mg/kg/d]), there has been no nephrotoxicity
[23,24].
    Since vancomycin was thought to be nephrotoxic, vancomycin dosing
was based on vancomycin levels [65,66]. However, vancomycin has no neph-
rotoxic potential. Thus, there is no rationale for using routine vancomycin
levels to dose patients to avoid nephrotoxicity. Accurate vancomycin dosing
is readily achieved more quickly, simply, less expensively, and without risk
of nephrotoxicity by dosing vancomycin based on CrCl rather then by van-
comycin levels [67–72]. It has been shown that vancomycin levels do not re-
duce nonexistent toxicity but vancomycin levels often result in unnecessary
vancomycin dosing changes [73–78].
398                                    CUNHA




  Box 2. Vancomycin tissue concentrations*
  Bone concentrations
     Cortical bone
         Uninfected: w5%
         Infected: w10%
     Cancellous bone
         Uninfected: w5%
         Infected: 5%
  Synovial fluid concentrations
     Uninflamed synovial fluid: w20%
  Urine Concentrations
     Uninfected urine: 100% (normal renal function)
  Stool concentrations
     Feces:
         Intravenous: 0%
         Orally: 1000%
  CSF concentrations
     Uninflamed meninges: 0% (prophylaxis of CSF seeding)
     Inflamed meninges: 15% (therapy of acute bacterial
        meningitis)
  Heart concentrations
     Cardiac valves: 20%
  Lung concentrations
     Lung parenchyma: 10%
  Bile concentrations
     Unobstructed bile: w50%
  Ascitic fluid concentrations
     Ascites: 10%

      * Relative to simultaneous serum levels




    There is often considerable delay between the time vancomycin peaks
and troughs are reported [70,74–76]. Two or more days have usually
elapsed when dosing adjustments are made, which means adjustments in
vancomycin dosing are based on previous, but no longer relevant estimates
of, renal function (CrCl) [70,76]. The other practical difficulty with vanco-
mycin serum peak and trough levels is in the timing of the samples which
influences interpretation of the levels. This, in addition, causes unnecessar-
ily frequent changes in vancomycin dosing, which serve no clinical purpose
[1,6,32,70–78].
    In anuria, on chronic hemodialysis, vancomycin peak serum concentra-
tions are predictable and vancomycin serum levels are unnecessary. In
VANCOMYCIN REVISITED                         399

patients receiving vancomycin on ‘‘high flux’’ hemodialysis, a posthemodial-
ysis dose should be given to replace the vancomycin removed. Since about
40% of vancomycin is removed by ‘‘high flux’’ hemodialysis, a 500-mg in-
travenous posthemodialysis dose should be given [57].
   Vancomycin serum concentrations may be used to assess adequacy of
therapy in patients with unusual Vd, such as burn patients, or in those
patients not responding to appropriately dosed vancomycin (ie, patients
showing vancomycin ‘‘tolerance’’ or resistance). For these purposes, vanco-
mycin serum levels should be obtained. Except to assess therapeutic efficacy
or ‘‘apparent therapeutic failure’’ there is no rationale for obtaining vanco-
mycin serum levels [69–78].


Vancomycin microbiologic activity and efficacy
Antistaphylococcal activity
Staphylococcus epidermidis
   The main use of parenteral vancomycin over the years has been in the
treatment of Staphylococci epidermidis, (ie, coagulase-negative staphylococci
(CoNS) infections, methicillin-sensitive S epidermidis (MSSE), and methicil-
lin-resistant S epidermidis (MRSE) infections). CoNS infections are usually
related to prosthetic implant materials (ie, infections involving prosthetic
joints, prosthetic heart valves, and CNS shunts). These infections are diffi-
cult to eradicate with antimicrobial therapy alone because antibiotics are
unable to eradicate these organisms in the glycocalyx on prosthetic
materials.

Methicillin-resistant Staphylococcus aureus
   Vancomycin has been used to treat serious staphylococcal infections,
especially MRSA. The prevalence of MRSA has increased during the past
several decades with three clinical variants recognized. Two of these are
hospital-acquired MRSA (HA-MRSA) and community-onset MRSA
(CO-MRSA). CO-MRSA strains originate in the hospital, circulate in the
community, and subsequently have their onset in the community before be-
ing readmitted to the hospital. A third is community-acquired (CA-MRSA),
a term often mistakenly applied to CO-MRSA because both come from the
community.
   However, CA-MRSA infections have two distinctive clinical presentations
readily differentiating them from CO-MRSA strains, which represent the
majority of MRSA admitted from the community (community onset) to the
hospital. CA-MRSA infections usually present either as severe pyoderma or
as necrotizing/hemorragic CAP. Although still rare, these two clinical CA-
MRSA syndromes are recognizable by their clinical presentations. Strains of
CA-MRSA, particularly those with the Panton-Valentine leukocidin gene
(PVL positive), are unusually virulent with a high degree of cytotoxic
400                                   CUNHA



activity, which accounts for their extensive tissue destruction and two
unique clinical presentations. While these strains are genetically distinctive
(ie, Staphylococcal chromosome cassette [SCC] mec IV), most hospital
laboratories cannot perform studies to identify PVL positive strains. Clini-
cians must rely on the clinical presentation to identify CA-MRSA strains
versus the great majority of MRSA strains from the community, which are
nearly all CO-MRSA. In spite of the increased virulence of CA-MRSA
PVL positive strains, these organisms are surprisingly sensitive to older
antibiotics, such as doxycycline, clindamycin, and trimethoprim-sulfame-
thoxazole (TMP-SMX), but these antibiotics are not effective against
HA-MRSA strains. Vancomycin, linezolid, daptomycin, minocycline, and
tigecycline are active against HA-MRSA, CO-MRSA, and CA-MRSA
strains. For CA-MRSA necrotizing pyodermas, treat with surgical de-
bridement and a HA-MRSA/CO-MRSA antibiotic, such as vancomycin,
linezolid, daptomycin, or tigecycline. For CA-MRSA CAP with influenza-
like illness, treat with influenza antivirals and linezolid [79,80].

Group D enterococci
   Since vancomycin was the initial antibiotic effective against MRSA, its
excessive use over time has resulted in two major clinical problems. Firstly,
extensive intravenous (not oral) vancomycin use has resulted in an increase
in the prevalence of E faecium (VRE), as well as increased vancomycin re-
sistance [81]. The increase in VRE isolates is explained by the activity of
vancomycin against the VSE component in the fecal flora, which is the pre-
dominant enterococcal species in the fecal flora. By decreasing VSE in the
fecal flora, there is a commensurate increase in VRE in feces. There has
been an increase worldwide in VRE prevalence due to vancomycin over
use. Instead, other anti-MRSA antibiotics that do not have this effect should
be used [26,81,82].


Vancomycin tolerance and resistance
Enterococci
    Enterococci and, to a lesser extent, staphylococci may develop ‘‘tolerance’’
to vancomycin. ‘‘Tolerance’’ may be defined as a minimum bactericidal
concentration (MBC) of R32 times the MIC of an antibiotic. Vancomycin
‘‘tolerance’’ may account for some cases of delayed or blunted therapeutic re-
sponse with enterococci or staphylococci [1,83–85].
    Vancomycin is a heptapeptide and is bactericidal for most gram-positive
organisms, including staphylococci but is bacteriostatic against enterococci.
Resistance to vancomycin is mediated by alterations in the permeability of
outer membrane porins in S aureus. Vancomycin binds rapidly and irrevers-
ibly to cell walls, inhibiting cell-wall synthesis. Interfering with peptidoglycan
VANCOMYCIN REVISITED                            401

cross-linkages causes defective cell-wall synthesis, resulting in bacterial cell-
wall lysis. Enterococci resistance to vancomycin may be of the high- or low-
grade variety. High-level (Van A) vancomycin resistance (MIC R64 mg/mL)
is mediated by plasmids and is inducible and transferable. Low-level
(Van B) resistance (MIC: 32–64 mg/mL) is nontransferable and chromoso-
mally encoded [1,85].

Staphylococci
   Widespread vancomycin use has increased staphylococcal resistance to
vancomycin manifested as increased MICs. Clinically, vancomycin resis-
tance is manifested as delayed resolution of infection or therapeutic failure.
It has long been appreciated that patients with MRSA bacteremia/endocar-
ditis often do not respond to vancomycin therapy [86–92]. This cannot be
ascribed to underdosing vancomycin. Even with optimal vancomycin
dosing, persistent MRSA bacteremias and therapeutic failures are not un-
common in clinical practice. Vancomycin therapy increases staphylococcal
cell-wall thickening, particularly in hVISA strains. Cell-wall thickening re-
sults in ‘‘permeability-mediated’’ resistance to vancomycin as well as to
other antistaphylococcal antibiotics. For these reasons, vancomycin may
no longer be the preferred antibiotic for treatment of serious systemic
MRSA infections. Other effective MRSA antibiotics are available and
may be used in place of vancomycin (eg, minocycline, linezolid, tigecycline,
and daptomycin). The empiric use of vancomycin to treat MRSA-colonized
patients should be minimized to avoid further increases in VRE prevalence
and MRSA resistance [1,6,32,93].


Vancomycin: clinical uses
Methicillin-sensitive Staphylococcus aureus and methicillin-resistant
Staphylococcus aureus bacteremias
   The finding of S aureus, either of the MSSA or MRSA variety, is common
in blood culture reports. Because MSSA and MRSA often colonize the skin,
these organisms are frequently introduced into blood cultures during veni-
puncture. S epidermidis, (CoNS), are also common blood culture contami-
nants. Approximately 20% of blood cultures are contaminated by MSSA,
MRSA, or CoNS. Clinicians should not equate-positive blood cultures with
bacteremia.
   Some organisms have clinical significance whenever present in blood cul-
tures (ie, Listeria monocytogenes, Brucella sp, Streptococcus pneumoniae).
The clinical significance of gram-positive cocci in blood cultures must be as-
sessed by the degree of blood culture positivity and considered in the appro-
priate clinical context. Ordinarily, four blood cultures should be obtained.
This usually allows the clinician to clearly differentiate between blood
402                                  CUNHA



culture positivity and bacteremia when dealing with blood culture reports
positive for MSSA, MRSA, or CoNS. One or two out of four positive blood
cultures usually represent blood culture contamination rather than infec-
tion. High-grade blood culture positivitydthree of four or four of fourd
is usually indicative of bacteremia. If it is established that the patient has
bacteremia, the clinician’s next task is to determine whether it is a transient
or sustained bacteremia. Staphylococci bacteremia is said to be persistent
when MSSA or MRSA is present repeatedly in consecutive blood cultures
demonstrating high blood culture positivity (ie, three of four or four of
four). One out of four or two of four blood cultures with CoNS almost al-
ways represents blood culture contamination. Conversely, persistent high-
grade blood culture positivity with CoNS, particularly in the presence of
implanted prosthetic materials, usually indicates a prosthetic material- or
device-associated infection [2,32,94,95].
   Much empiric vancomycin is used to ‘‘cover’’ ‘‘gram positive cocci in clus-
ters’’ in preliminary blood culture reports rather than bacteremia. The most
common cause of high-grade MSSA, MRSA, or CoNS bacteremia are central
venous catheter (CVC)–related infections. Empiric vancomycin used to
‘‘cover’’ the staphylococcal bacteremia or the ‘‘catheter’’ should be discour-
aged. If CVC line infection is suspected, the line should be removed and the
diagnosis confirmed by semiquantitative catheter tip cultures. Treatment for
CVC line–associated bacteremia is catheter removal, not just antibiotic ther-
apy. Overuse of empiric vancomycin for suspected or known CVC-line infec-
tions has increased the prevalence of VRE. If CVC access is necessary after
CVC removal for semiquantitative catheter tip culture, another line may be re-
placed over a guide wire without risk of pneumothorax, pending catheter tip
results. If the semiquantitative catheter tip cultures are later reported as neg-
ative, the new catheter placed over a guide wire may remain in place. If catheter
tip cultures are positive (ie, R15 colonies of the same organism as causing the
bacteremia), then the line replaced over the guide wire should be removed and
a new CVC placed in a different anatomic location (Boxes 3 and 4) [96,97].

Continuous Methicillin-sensitive Staphylococcus aureus and
methicillin-resistant Staphylococcus aureus bacteremias
   Patients presenting with a sustained or persistent MSSA or MRSA bac-
teremia should be treated empirically with an agent that offers a high degree
of activity against MSSA or MRSA, depending upon the hospital’s predom-
inant S aureus type (ie, MSSA versus MRSA) while a workup to determine
the cause of the persistent bacteremia is undertaken. The most common un-
derlying causes of persistent MSSA or MRSA bacteremia are intravascular
infections (ie, CVC-related infections) or acute bacterial endocarditis or par-
avalvular abscess, and less commonly are due to persistent bacteremias from
non-cardiac staphylococcal abscesses and, even less commonly, bone infec-
tions [47,97–101].
VANCOMYCIN REVISITED                       403



  Box 3. Vancomycin: appropriate prophylactic
  and therapeutic uses
  Prophylaxis
   Prosthetic (orthopaedic/cardiovascular) implant surgery
   MSSA/MRSA infections in hemodialysis patients
   Subacute bacterial endocarditis for:
    Oral and upper respiratory tract procedures
      (Streptococcus viridans) (in penicillin allergic patients)
    Gastrointestinal and genitourinary procedures
      (E faecalis, VSE) procedures in patients with previous
      subacute bacterial endocarditis or prosthetic valve
      endocarditis (plus gentamicin)
  Therapy
   MRSA, CoNS CNS shunt (ventriculo-atrial,
    ventriculo-peritoneal) infectionsa
   MRSA hemodialysis catheter infectionsb
   Endocarditisc
    Streptococus viridans subacute bacterial endocarditis (in
      penicillin-allergic patients)
    CoNS prosthetic valve endocarditis due to MSSE or MRSE
    VSE subacute bacterial endocarditisd
     a
       Preferred therapy (eg, linezolid).
     b
       Preferred therapy (eg, daptomycin or linezolid).
     c
       Alternative therapy preferred.
     d
       Combined with gentamicin.




Methicillin-sensitive Staphylococcus aureus and methicillin-resistant
Staphylococcus aureus central intravenous line infections
    The majority of intravenous line infections related to CVCs are due to
skin organisms (ie, MSSA, MRSA, MSSE, or MRSE). Because vancomycin
is active against these skin pathogens, it is frequently used as empiric mono-
therapy for presumed CVC line infections. There are two problems with this
approach. In institutions where the predominant pathogen causing CVC
line infections is MSSA, rather than MRSA, other agents should be used
in preference to vancomycin. After MSSA and MRSA, aerobic gram-nega-
tive bacilli are the next most important pathogens in CVC line infections.
The use of vancomycin empirically for CVC infections has two hospital-
wide consequences [96,97,101].
    Firstly, vancomycin increases institutional prevalence of VRE. Secondly,
exposure to vancomycin has the effect of increasing cell-wall thickness
among staphylococci. Vancomycin-induced thickened bacterial cell walls
404                                CUNHA




  Box 4. Clinical situations in which vancomycin
  should be avoided
  Prophylaxis
     General surgical procedures
     MSSA/MRSA neurosurgical shunts
     MSSA/MRSA chest tubes
     MSSA/MRSA intravenous lines
     MSSA/MRSA in febrile leukopenia
  Therapy
     MRSA colonization of respiratory secretions, wounds, or
      urine (avoid treating colonization; treat only infection)
     Most serious systemic MRSA infections
      Preferentially use other anti-MRSA antibiotics
        Intravenous MRSA antibiotics: linezolid, daptomycin,
          tigecycline, minocycline
        Oral MRSA antibiotics: linezolid, minocycline



are manifested as an increase in MICs, delayed resolution of infection, or
therapeutic failure. Staphylococci with thickened cell walls represent a per-
meability barrier to vancomycin as well as to other antibiotics. This is man-
ifested clinically in the microbiology laboratory as ‘‘MIC drift.’’ The
increase in MICs, often observed during vancomycin therapy, is reflective
of cell-wall thickening. A preferred approach to the empiric treatment to
CVC line infections pending CVC removal is to use meropenem in institu-
tions where CVC infections are more frequently due to MSSA/GNBs than
to MRSA. In institutions where the reverse is true, and MRSA is an
important CVC pathogen, empiric therapy with tigecycline is preferable
to combination therapy with vancomycin plus an anti–GNB antibiotic
(Box 5).

Methicillin-sensitive Staphylococcus aureus and methicillin-resistant
Staphylococcus aureus hemodialysis catheter infections
   The treatment of hemodialysis catheter infections in hemodialysis pa-
tients has been a major area of vancomycin usage because vancomycin is
eliminated by glomerular filtration. The increased half-life of vancomycin
in anuria may be used to a therapeutic advantage in patients with MSSA/
MRSA infections in end stage renal disease on hemodialysis. As with other
foreign body infections due to gram-positive organisms, shunt removal is
usually necessary for elimination the infection. Vancomycin has been used
prophylactically and therapeutically in hemodialysis patients to treat dialy-
sis catheter infections due to MSSA and MRSA [32,102–104].
VANCOMYCIN REVISITED                            405



  Box 5. Diagnostic clinical pathway: persistent S aureus
  (MSSA, MRSA) bacteremias
  Differentiate S aureus blood culture positivity (1/4 or 1/2) from
    bacteremia.
  With S aureus bacteremia, differentiate low-intensity intermittent
    bacteremia (1/2 or 2/4 positive blood cultures) from continuous
    high-intensity bacteremia (3/4–4/4 positive blood cultures).
  Determine the source of S aureus bacteremia.
    Abscesses
    Central intravenous lines
    Implanted prosthetic devices/materials
    Acute bacterial endocarditis
    Soft tissue and bone infections
  Review antibiotic-related factors.
    Determine drug dose/dosing interval.
    Determine MIC/MBCs.
    Evaluate in vivo versus in vitro effectiveness.
    Evaluate potential of permeability-related resistance due
       to vancomycin cell-wall thickening.
  With high-intensity continuous S aureus bacteremia, obtain
    a transthoracic or transesophageal echocardiogram to
    diagnose or rule out acute bacterial endocarditis.

     Data from Cunha BA. Persistent S aureus bacteremia: clinical pathway
  for diagnosis  treatment. Antibiotics for Clinicians 2006;10(S1):39–46; with
  permission.




Methicillin-sensitive Staphylococcus aureus and methicillin-resistant
Staphylococcus aureus acute bacterial endocarditis
   The diagnostic approach to persistent S aureus bacteremia is relatively
straightforward. First, in patients with CVCs, this source of infection should
be ruled out first because since CVC infections are readily diagnosable and
easily treated by CVC removal. If there is no CVC in place, or if the
removed CVC semiquantitative catheter tip cultures are negative, then
endocarditis should be the next diagnostic consideration. MSSA or
MRSA endocarditis presents as acute bacterial endocarditis (ABE). Except
for IVDAs, patients presenting with MSSA or MRSA acute bacterial endo-
carditis, usually present with fever of 102 F or higher. In addition, MSSA or
MRSA acute bacterial endocarditis usually presents without a heart mur-
mur early in the infectious process. Later, when there is valvular destruction,
a new or rapidly changing murmur becomes apparent.
406                                   CUNHA



    In patients with subacute bacterial endocarditis and positive blood cultures
due to subacute bacterial endocarditis pathogens, cardiac echocardiography
is indicated in patients who have a heart murmur. Cardiac echocardiography
may be the transthoracic (TTE) or the transesophageal (TEE) variety. For
screening purposes, TTE is preferred to TEE. For detection of prosthetic
valve endocarditis and myocardial abscess TEE is preferred. In patients
with subacute bacterial endocarditis, the indication for a TTE or a TEE is
the presence of continuous bacteremia due to a known subacute bacterial
endocarditis pathogen plus fever and a heart murmur [105–107].
    In contrast, in patients with possible acute bacterial endocarditis, the indi-
cations for a TTE or TEE are fever and a sustained high-grade MSSA or
MRSA bacteremia. A heart murmur is not usually present in acutely in pa-
tients presenting with acute bacterial endocarditis. If cardiac echocardiogra-
phy is negative for vegetations, acute bacterial endocarditis is effectively
eliminated from further consideration. If the high-grade continuous MSSA
or MRSA bacteremia continues, the echocardiography should be reviewed
for a potential paravalvular myocardial abscess. If a myocardial abscess is sus-
pected, TEE, is the preferred echocardiographic modality to demonstrate
a perivalvular septal, or myocardial abscess. In patients with potential staph-
ylococcal prosthetic valve endocarditis (PVE), a TEE is preferred to demon-
strate vegetations. Patients with prosthetic valve(s) should be suspected of
having prosthetic valve endocarditis if there is fever and a continuous high-
grade bacteremia with a known endocarditis pathogen. Fever is usually pres-
ent, but murmurs are difficult to appreciate because of the sounds generated by
the prosthetics valve. Subacute bacterial endocarditis, acute bacterial endo-
carditis, and prosthetic valve endocarditis may be accompanied by peripheral
findings associated with endocarditis (Box 6; Tables 2 and 3) [97,101,106,107].

Staphylococcal central nervous system infections
   Vancomycin has been used to treat CNS shunt infections, which are
usually due to CoNS of the MSSE or MRSE variety. CNS shunt infections
usually require shunt removal. If suppression is desired, vancomycin may
be given parenterally and, if necessary, CSF concentrations may be further
increased by intraventricular administration. Usually the best way to admin-
ister intrathecal vancomycin is via an Ommaya reservoir. Vancomycin CSF
levels may be obtained in patients to assure adequate CSF levels, but resolu-
tion of shunt infections almost always requires shunt removal. It is preferable
to use an antibiotic with good CSF penetration (eg, linezolid) [1,31,32].

Febrile neutropenia
   Recently, some clinicians have added vancomycin to regimens to treat
febrile neutropenia in compromised hosts receiving chemotherapy. The
increased incidence of gram-positive infections in febrile neutropenia are
not due to neutropenia per se, but are due to central line catheters or
VANCOMYCIN REVISITED                              407



  Box 6. Diagnostic clinical pathway: MSSA/MRSA
  acute bacterial endocarditis
  Differentiate S aureus blood culture positivity (1/2 or 1/4 positive
     blood cultures) from bacteremia (3/4–4/4 positive blood
     cultures).
  With S aureus bacteremia, differentiate low-intensity intermittent
     bacteremia (1/2 or 2/4 positive blood cultures) from continuous
     high-intensity bacteremia (3/4–4/4 positive blood cultures).
  Acute bacterial endocarditis is not a complication of low-
     intensity/intermittent S aureus bacteremia. TTE/TEE
     unnecessary, but will verify no vegetations.
  If continuous high-grade MSSA/MRSA bacteremia, obtain a TTE
     or TEE to document cardiac vegetations and/or paravalvular
     abscess and confirm diagnosis of acute bacterial endocarditis.
  Diagnostic criteria for MSSA/MRSA acute bacterial endocarditis
      Essential features
         Continuous high-grade MSSA/MRSA bacteremia
         Cardiac vegetations on TTE/TEE
         No other source
      Nonessential features
         Fever ‚102 F (non-IVDAs)
         Cardiac murmura
     a
       With early MSSA/MRSA, a murmur is not present. Later, a new murmur in
  acute bacterial endocarditis indicates a vegetation or valvular destruction.
     Data from Cunha BA: MSSA/MRSA acute bacterial endocarditis (ABE): clinical
  pathway for diagnosis  treatment. Antibiotics for Clinicians 2006;10(S1):29–34.



chemotherapy infusion devices in these patients. Depending upon the ge-
ography/epidemiology of the balance between MSSA versus MRSA strains
in patients in the community, empiric treatment with an anti-MSSA or an
anti-MRSA agent may be desired until the temporary or semipermanent
catheter is removed or replaced. For patients presenting with febrile neutro-
penia treated with an antibiotic effective against Pseudomonas aeruginosa
with good anti-MSSA activity, additional antistaphylococcal therapy is
usually unnecessary. Antibiotics commonly used empirically in febrile neu-
tropenia (eg, levofloxacin, meropenem) have anti-MSSA activity. In febrile
neutropencia, fungal infections usually present after 10 to 14 days of antibi-
otic therapy and are clinically manifested as an otherwise unexplained recru-
descence of fever in patients who have responded to empiric anti-P aeruginosa
therapy. If fungal infection can be ruled out, a temporary or semipermanent
central line infection should be suspected. If blood cultures are persistently
positive (high-grade positivity) with MRSA, then anti-MRSA therapy should
408                                           CUNHA


Table 2
Delayed resolution or failure of vancomycin therapy of MSSA or MRSA bacteremia and acute
bacterial endocarditis
                          Failure rates                 Duration of bacteremia        References
MSSA bacteremia           Nafcillin: 4%                 Nafcillin: 2 days             Sande [110]
                          Vancomycin: 20%               Vancomycin: 7 days
                                                          (20% O3 days)
                                                          (12% O7 days)
MSSA acute bacterial      Nafcillin: 1.4%–26%           Nafcillin: 2 days             Gentry [111]
 endocarditis             Vancomycin: 37%–50%           Vancomycin: 5 days            Geraci [112]
                                                                                      Esposito [4]
                                                                                      Chang [98]
MRSA acute bacterial                                    Nafcillin: Not applicable     Small [115]
 endocarditis                                           Vancomycin: O7 days


be given until the device is removed. The routine use of vancomycin in com-
bination therapy with an anti-P aeruginosa antibiotic for febrile neutropenia
should be discouraged to minimize further increases in VRE prevalence and
permeability-mediated resistance [1,32,108].

Penicillin-allergic patients
   Vancomycin has been used in penicillin-allergic patients to treat Strepto-
coccus viridans and enterococcal endocarditis. Vancomycin monotherapy
has been used successfully over the years to treat S viridans subacute bacte-
rial endocarditis. Because vancomycin is bacteristatic against E faecalis,
combination therapy with aminoglycoside (eg, gentamicin) has been used
to treat VSE endocarditis. Strains demonstrating high-level gentamicin resis-
tance may be treated with vancomycin plus ceftriaxone or, alternatively,
another antibiotic with bactericidal antienterococcal activity may be used
(eg, daptomycin) (see Box 4) [32,109].

Vancomycin: therapeutic alternatives
   The empiric therapeutic approach to central intravenous line–related
infections has been discussed. Empiric therapy during the workup for

Table 3
Combination therapy for MSSA and MRSA acute bacterial endocarditis
Antibiotic combinations                   Comments                                  References
MSSA acute bacterial endocarditis
 Nafcillin þ gentamicin                   Outcomes same without gentamicin          Cha [116]
 Vancomycin þ gentamicin                                                            Lee [117]
MRSA acute bacterial endocarditis
 Vancomycin                               Duration of bacteremia: 7 days            Levine [118]
 Vancomycin þ rifampin                    Duration of bacteremia: 9 days            Shelburne [119]
                                           (antagonistic; not synergistic)
VANCOMYCIN REVISITED                           409

ABE should consist of antistaphylococcal antibiotic that has a high degree
of activity against the presumed pathogen, does not cause resistance, does
not cause the emergence of other organism (eg, VRE), and has a good safety
profile. In the treatment of MSSA bacteremias, vancomycin has been shown
to be inferior to vancomycin in terms of delayed resolution of bacteremia. If
MSSA is the pathogen in bacteremia or endocarditis, vancomycin should
not be used as therapy in these patients and preferably another agent should
be used. In penicillin-allergic patients, vancomycin is often used empirically
to treat MSSA or MRSA bacteremias. However, there are many alternative
antibiotics available that do not cross-react with penicillins or cephalospo-
rins and that are effective to treat MSSA or MRSA bacteremias.
   The treatment of MSSA or MRSA bacteremia is optimal with a single
agent. Clinicians often assume there is benefit in adding a second drug for
enhanced antistaphylococcal activity. It has been shown that vancomycin
plus gentamicin offers no benefit in the treatment of MSSA bacteremias.
Another common misconception is that the addition of rifampin or other
drugs to vancomycin will enhance the antistaphylococcal activity of the pri-
mary anti-MSSA/MRSA agent. It has been shown the rifampin is likely to
be antagonistic, not synergistic, against S aureus [110]. While rifampin has
a high degree of in vitro activity against S aureus, rifampin monotherapy
should not be used because of resistance. The practice of adding an amino-
glycoside or rifampin to antistaphylococcal antibiotics has no benefit and
may have a negative effect on outcomes (see Tables 2 and 3) [1,3–
5,31,32,110].
   In penicillin-allergic patients, the treatment of serious systemic infections
due to MSSA (ie, CVC line infections, acute bacterial endocarditis, pros-
thetic valve endocarditis, and so on), may be treated with antibiotics other
than vancomycin (ie, meropenem, TMP-SMX, daptomycin). As with
MSSA, there are numerous therapeutic alternatives to vancomycin for the
treatment of serious MRSA infections. Useful antibiotics to treat all
MRSA types (HA-MRSA, CO-MRSA, CA-MRSA) include daptomycin
linezolid, tigecycline, and minocycline [1,32].
   If possible, vancomycin should be avoided in therapy of most serious sys-
temic infections due to MSSA/MRSA for two principal reasons. Prolonged
vancomycin exposure selects out heteroresistant strains (hVISA). These
strains are the ones that have thickened cell walls as the result of
vancomycin exposure. Thickened cell walls are manifested as an increase
in MICs and decreased cell-wall permeability to vancomycin as well as to
other antibiotics. The other untoward side effect of vancomycin use is in-
creased VRE prevalence [37–43].
   In conclusion, vancomycin should be used selectively for the parenteral
treatment of serious systemic infections due to MSSA, MRSA, or CoNS.
Available therapeutic alternatives do not increase the prevalence of VRE
and do not result in cell-wall thickening and permeability-mediated
resistance. Alternative agents more quickly resolve MSSA/MRSA
410                                  CUNHA



bacteremias than vancomycin. The addition of gentamicin or rifampin to
vancomycin does not enhance antistaphylococcal activity and may have
a negative effect (ie, antagonism) [47,98–100,111–117].


Vancomycin: reassessment of use
   The main role of vancomycin has been in the therapy of serious systemic
MRSA infections [1,118]. The widespread use of vancomycin over the years
has resulted in increased prevalence of VRE in institutions worldwide. Re-
cently, it has been shown that vancomycin induces cell-wall thickening among
staphylococci, resulting in ‘‘permeability-mediated’’ resistance. To avoid
these problems associated with vancomycin, it is preferable to use other
anti-MRSA antibiotics in place of vancomycin. Fortunately, several other
anti-MRSA agents are available. Antibiotics with excellent anti-MRSA activ-
ity include minocycline, linezolid, daptomycin, and tigecycline. Although
vancomycin is available as an oral formulation for C difficile infections, it is
ineffective to treat systemic infections. For the parenteral treatment of serious
systemic MRSA infections, minocycline, daptomycin, and linezolid are effec-
tive alternative anti-MRSA antibiotics. For the oral therapy of MRSA, min-
ocycline or linezolid may be used (Boxes 7 and 8) [1,32,119,120].


  Box 7. Vancomycin: use in serious systemic infection
  Clinical advantages
     Extensive clinical experience
     Not nephrotoxic
  Clinical disadvantages
     Adverse side effects
         Red neck (red man) syndrome
         Thrombocytopenia
         Leukopenia
         Sudden death
         Increased VRE prevalence
         Related to volume of intravenous (not oral) use
        S. aureus infections: delayed resolution, therapeutic failures
           MSSA/MRSA bacteremias
           MSSA/MRSA acute bacterial endocarditis
        S. aureus infections: permeability-mediated resistance
           (increased MICs)
         Cell-wall thickening; decreased S. aureus penetration of
            antibiotics (vancomycin and other anti-S aureus
            antibiotics)
     Not inexpensive
VANCOMYCIN REVISITED                              411



  Box 8. Suboptimal uses of vancomycin
   Empiric treatment of central intravenous line infections
       In locations where MSSA/GNBs  MRSA CVC pathogens
       Remove intravenous line as soon as possible
   Therapy of MRSA bacteremia/acute bacterial endocarditis
       Delayed clinical responses
       Therapeutic failures
   Intraperitoneal therapy of S aureus/CoNS CAPD peritonitis
       Intravenous therapy achieves therapeutic concentration in
           ascitic fluid. Requires another antibiotic for
           anti–gram-negative bacillary coverage.
   Monotherapy of VSE
       Bacteriostatic if monotherapy used.
       Adequate anti-VSE activity only if given with an
           aminoglycoside (eg, gentamicin)
   S aureus chronic osteomyelitis
   Prevention of S pneumoniae CSF ‘‘seeding’’
    (2 to CAP pneumococcal bacteremia)
       Unnecessary when using antibiotics that achieve therapeutic
           CSF concentrations (eg, ceftriaxone)

    Data from Cunha BA, editor. Infections in critical care medicine. 2nd edition.
  New York: Informa Health Care USA, Inc.; 2007.



    For the therapy of gram-positive CNS infections, other agents are phar-
macokinetically preferable to vancomycin. Linezolid, available orally and
intravenously, achieves high CSF levels when given in the usual dose (ie,
600 mg intravenously or orally every 12 hours). Minocycline using the usual
dose (100 mg intravenously or orally every 12 hours) also achieves therapeu-
tic CSF levels. No data are available regarding treatment of CNS infections
with or tigecycline [1,32].
    In access catheter graft infections in chronic hemodialysis patients, dap-
tomycin, linezolid, or tigecycline may be used. Vancomycin can be used to
treat S viridans subacute bacterial endocarditis in penicillin-allergic patients,
but other agents are preferable to vancomycin for this use. Vancomycin
monotherapy is ‘‘bacteristatic’’ against VSE. Therefore, for VSE subacute
bacterial endocarditis, an aminoglycoside should be added to vancomycin
to achieve bactericidal anti-VSE activity alternate therapy for VSE subacute
bacterial endocarditis (eg, meropenem, linezolid, or daptomycin) is available
[3,32,119,120].
    Aside from increased prevalence of VRE and increasing resistance among
staphylococci, there are other reasons to avoid or minimize vancomycin use.
Because vancomycin is an older drug, it is often thought of as an inexpensive
412
Table 4
Vancomycin: clinical use and therapeutic alternatives
Clinical uses                             Disadvantages, problems              Therapeutic alternatives      Advantages of alternative antibiotics
Monotherapy
 Central IV catheter (CVC) infections     Increased VRE prevalence            Removal of CVC               High-degree MRSA activity
  ‘‘that may be due to MRSA’’              Increased permeability-mediated     Daptomycin (6 mg/kg IV       Also covers MSSA, VSE
                                            resistance                           every 24 hours)              No increased resistance/VRE
                                           MIC ‘‘drift’’                       Linezolid (600 mg IV
                                                                                 every 12 hours)
 MRSA bacteremias, acute bacterial        Increased permeability-mediated     Daptomycin (6–12 mg/kg       Most rapidly bactericidal MRSA
  endocarditis                              resistance                           IV every 24 hours)            antibiotic
                                           Delayed therapeutic response                                      Effective in vancomycin




                                                                                                                                                     CUNHA
                                            (persistent bacteremia)                                            ‘‘treatment failures’’
                                           MIC ‘‘drift’’                         Linezolid (600 mg          Also available orally
                                                                                   IV every 12 hours)          for prolonged therapy
                                                                                                              No increased resistance/VRE
 MRSA osteomyelitis                       Vancomycin: often delayed           Linezolid (600 mg            Also available orally
                                            response with 30 mg/kg/d; better     IV/orally every 12 hours)     for prolonged therapy
                                            results with 60 mg/kg/d             Minocycline (100 mgIV/       No increased resistance/VRE
                                           Increased permeability-mediated      orally every 12 hours)       Inexpensive and also available
                                            resistance                                                         orally for prolonged therapy.
                                           Increased MICs during therapy
                                           MIC ‘‘drift’’
 MRSA nosocomial and                      Essential to avoid ‘‘covering’’     Linezolid (600 mg IV         Effective in rare cases of bona
  ventilator associated pneumonia           MSSA/MRSA respiratory                every 12 hours)               fide MRSA nosocomial
  (rare)                                    secretion colonization                                             pneumonia and ventilator-
                                           Increases permeability-mediated                                    associated pneumonia
                                            resistance                                                        Therapeutic parenchymal lung
                                           MIC ‘‘drift’’                                                      concentrations
Combination Therapy
 Vancomycin and ceftriaxone              ‘‘Double drug’’ therapy          Ceftriaxone (1–2 g IV every  Good CSF penetration prevents
  to prevent ‘‘seeding’’ of CSF            unnecessary. Vancomycin:          12 hours)                     PRSP ‘‘seeding’’ of CSF
  from bacteremic S pneumoniae             relatively low CSF concentrations                              Ceftriaxone monotherapy provides
  (PRSP) CAP                                                                                               adequate CSF concentrations if PRSP
                                                                                                           seeding of CSF
 Vancomycin plus gentamicin therapy  Vancomycin alone ‘‘bacteriostatic’’  Daptomycin (6 mg/kg IV       MICs for VSE/VRE greater than
  of E faecalis (VSE) subacute bacterial   against VSE                       every 24 hours)               those for MSSA/MRSA
  endocarditis                            Vancomycin bactericidal only                                   Use 12 mg/kg IV every 24 hours
                                           when combined with an                                           for VSE endocarditis
                                           aminoglycoside (eg, gentamicin)  Linezolid (600 mg IV         Available orally for prolonged
                                                                             every 12 hours)               therapy of VSE subacute bacterial
                                                                                                           endocarditis




                                                                                                                                                 VANCOMYCIN REVISITED
                                                                                                          Bacteriostatic but effective
                                                                                                           in subacute bacterial endocarditis
   Abbreviations: IV, intravenous; PSRP, penicillin-resistant S pneumoniae.




                                                                                                                                                 413
414                                      CUNHA



antibiotic. However, because vancomycin must be administered intrave-
nously, the cost of intravenous administration and monitoring must be
added to the actual cost of vancomycin use. If serum vancomycin levels
are used to guide therapy, the cost of such levels plus the cost of serial serum
creatinine levels must also be included in the actual cost of administering
parenteral vancomycin. For patients requiring long-term parenteral therapy
with vancomycin, peripherally inserted central catheters (PICCs) are often
used. The use of a PICC line to administer vancomycin increases antibiotic
cost because other charges related to surgical insertion, radiological verifica-
tion of PICC line placement, and the cost of home intravenous agencies
must be added to vancomycin cost. While the cost of administering paren-
teral antibiotics via PICC lines for vancomycin is similar to that for other
antibiotics, most infections for which vancomycin is administered parenter-
ally may be treated equally effectively with oral linezolid which is much less
expensive (see Boxes 4 and 5, Table 4).


Summary
   The reassessment of vancomycin use today is based on an evaluation of
the advantages and disadvantages of vancomycin alternatives for the ther-
apy of MRSA. Other antibiotics exist as alternative therapy for VSE infec-
tions as well as for preventing the ‘‘seeding’’ of the CNS by penicillin-
resistant S pneumoniae from bacteremic pneumococcal CAP. Clinicians
should appreciate the limited role of vancomycin today in MRSA therapy.
Clinicians should also realize vancomycin has limited activity against MSSA
and VSE. Other antibiotics are preferable against MSSA and VSE. Pres-
ently, clinicians should consider alternatives to vancomycin because of the
negative effects of vancomycin therapy (ie, increased VRE prevalence and
increasing resistance among staphylococci), and also because better thera-
peutic alternatives are available with little or no resistance potential, no in-
crease in VRE prevalence, better pharmacokinetics and lower cost (eg,
linezolid) (see Box 6). For the reasons mentioned, clinicians should use van-
comycin sparingly and consider other antibiotic alternatives for MSSA,
MRSA, CoNS, and VSE infections [32,119,120].


References
 [1] Kucers A, Crowe SM, Grayson ML, et al. Vancomycin: the use of antibiotics: a clinical
     review of antibacterial, antifungal and antiviral drugs. 5th edition. Oxford (United
     Kingdom): Butterworth-Heinemann; 1997. p. 763–90.
 [2] Louria DB, Kaminski T, Buchman J. Vancomycin in severe staphylococcal infections. Arch
     Intern Med 1961;107:225–40.
 [3] Cunha BA. Vancomycin. Med Clin North Am 1995;79:817–31.
 [4] Esposito AL, Gleckman RA. Vancomcyin: a second look. JAMA 1977;238:1756–7.
 [5] Menzies D, Goel K, Cunha BA. Vancomycin. Antibiotics for Clinicians 1998;2:97–9.
VANCOMYCIN REVISITED                                    415

 [6] Farber BF, Moellering RC Jr. Retrospective study of the toxicity of preparations of
     vancomycin from 1974 to 1981. Antimicrob Agents Chemother 1983;23:138–41.
 [7] Elting LS, Rubenstein EB, Kurtin D, et al. Mississippi and mud in the 1990s: risks and
     outcomes of vancomycin-associated toxicity in general oncology practice. Cancer 1998;
     83:2597–607.
 [8] Appel GB, Given DB, Levine LR, et al. Vancomycin and the kidney. Am J Kidney Dis
     1986;8:75–80.
 [9] Mellor JA, Kingdom J, Cafferkey M, et al. Vancomycin toxicity: a prospective study.
     J Antimicrob Chemother 1985;15:773–80.
[10] Nahata MC. Lack of nephrotoxicity in pediatric patients receiving concurrent vancomycin
     and aminoglycoside therapy. Chemotherapy 1987;33:302–4.
[11] Goren MP, Baker DK Jr, Shenep JL. Vancomycin does not enhance amikacin-induced
     tubular nephrotoxicity in children. Pediatr Infect Dis J 1989;8:278–82.
[12] Goetz MB, Sayers J. Nephrotoxicity of vancomycin and aminoglycoside therapy separately
     and in combination. J Antimicrob Chemother 1993;32:325–34.
[13] Ergur T, Onarlioglu B, Gunay Y, et al. Does vancomycin increase aminoglycoside nephro-
     toxicity? Acta Paediatr Jpn 1997;39:422–7.
[14] Pauly DJ, Musa DM, Lestico MR, et al. Risk of nephrotoxicity with combination
     vancomycin-aminoglycoside antibiotic therapy. Pharmacotherapy 1990;10:378–82.
[15] Rybak MJ, Albrecht LM, Boike SC, et al. Nephrotoxicity of vancomycin alone and with an
     aminoglycoside. J Antimicrob Chemother 1990;25:679–87.
[16] Ngeleka M, Beauchamp D, Tardif D, et al. Endotoxin increases the nephrotoxic potential
     of gentamicin and vancomycin plus gentamicin. J Infect Dis 1990;161:721–7.
[17] Rybak MJ, Boike SC. Monitoring vancomycin therapy. Drug Intell Clin Pharm 1986;20:
     757–61.
[18] Edwards DJ, Pancorbo S. Routine monitoring of serum vancomycin concentrations:
     waiting for proof of its value. Clin Pharm 1987;6:652–4.
[19] Moellering RC Jr. Pharmacokinetics of vancomycin. J Antimicrob Chemother 1984;14:43–52.
[20] Matzke GR, Zhanel GG, Guay DR. Clinical pharmacokinetics of vancomycin. Clin
     Pharmacokinet 1986;11:257–82.
[21] Moellering RC Jr, Krogstad DJ, Greenblatt DJ. Vancomycin therapy in patients with
     impaired renal function: a nomogram for dosage. Ann Intern Med 1981;94:343–6.
[22] Leader WG, Chandler MH, Castiglia M. Pharmacokinetic optimization of vancomycin
     therapy. Clin Pharmacokinet 1995;28:327–42.
[23] Boffi el Amari E, Vuagnat A, Stern R, et al. High versus standard dose vancomycin for
     osteomyelitis. Scand J Infect Dis 2004;36:712–7.
[24] Hidayat LK, Hsu DI, Quist R, et al. High dose vancomycin therapy for methicillin-resistant
     Staphylococcus aureus infections. Arch Intern Med 2006;166:2138–44.
[25] Von Drygalski A, Curtis BR, Bougie DW, et al. Vancomycin-induced immune thrombocy-
     topenia. N Engl J Med 2007;356:904–10.
[26] Carmeli Y, Samore MH, Huskins C. The association between antecedent vancomycin
     treatment and hospital-acquired vancomycin-resistant enterococci: a meta-analysis. Arch
     Intern Med 1999;159:2461–8.
[27] Fishbane S, Cunha BA, Shea KW, et al. Vancomycin-resistant enterococcus (VRE) in
     hemodialysis patients. Am J Infect Control 1999;20:461–2.
[28] Fridkin SK, Lawton R, Edwards JR, et al. Monitoring antimicrobial use and resistance:
     comparison with a national benchmark on reducing vancomycin use and vancomycin-
     resistant enterococci. Emerg Infect Dis 2002;8:702–7.
[29] Fridkin SK, Edwards JR, Courval JM, et al. The effect of vancomycin and third-generation
     cephalosporins on prevalence of vancomycin-resistant enterococci in 126 U.S. adult
     intensive care units. Ann Intern Med 2001;135:175–83.
[30] Kolar M, Urbanek K, Vagnerova I, et al. The influence of antibiotic use on the occurrence
     of vancomycin-resistant enterococci. J Clin Pharm Ther 2006;31:67–72.
416                                         CUNHA


[31] Cunha BA. Antimicrobial therapy of multi-drug resistant S pneumoniae VRE  MRSA.
     Med Clin North Am 2006;90:1165–82.
[32] Cunha BA. Antibiotic essentials. 7th edition. Royal Oak (MI): Physicians Press; 2008.
[33] Tenover F, Biddle J, Lancaster M. Increasing resistance to vancomycin and other glycopep-
     tides in Staphylococcus aureus. Emerg Infect Dis 2001;7:327–32.
[34] Liu C, Chambers HF. Staphylococcus aureus with heterogeneous resistance to vancomycin:
     epidemiology, clinical significance, and critical assessment of diagnostic methods. Antimi-
     crob Agents Chemother 2003;47:3040–5.
[35] Cosgrove SE, Carroll KC, Perl TM. Staphylococcus aureus with reduced susceptibility to
     vancomycin. Clin Infect Dis 2004;39:539–45.
[36] Fridkin SK, Hageman J, McDougal LK, et al. Epidemiological and microbiological
     characterization of infections caused by Staphylococcus aureus with reduced susceptibility
     to vancomycin, United States, 1997–2001. Clin Infect Dis 2003;36:429–39.
[37] Wang G, Hindler JF, Ward KW, et al. Increased vancomycin MICs for Staphylococcus
     aureus. Clinical isolates from a university hospital during a 5-year period. J Clin Microbiol
     2006;44:3883–6.
[38] Cui L, Ma K, Sato K, et al. Cell wall thickening is a common feature of vancomycin
     resistance in Staphylococcus aureus. J Clin Microbiol 2003;41:5–14.
[39] Cui L, Tominaga E, Hiramatsu K. Correlation between reduced daptomycin susceptibility
     and vancomycin resistance in vancomycin-intermediate Staphylococcus aureus. Antimicrob
     Agents Chemother 2006;50:1079–82.
[40] Bell JM, Walters LJ, Turnidge JD, et al. Vancomycin hetero-resistance has a small but
     significant effect on the daptomycin minimum inhibitory concentration of Staphylococcus
     aureus [abstract D-814]. Paper presented at the 46th ICAAC. San Francisco (CA), Septem-
     ber 27–30, 2006.
[41] Sakoulas G, Alder J, Thauvin-Eliopoulos C, et al. Induction of daptomycin heterogeneous
     susceptibility in Staphylococcus aureus by exposure to vancomycin. Antimicrob Agents
     Chemother 2006;50:1581–5.
[42] Patel JB, Jevitt LA, Hageman J, et al. An association between a reduced susceptibility to
     daptomycin and reduced susceptibility to vancomycin in Staphylococcus aureus. Clin Infect
     Dis 2006;42(11):1652–3.
[43] Howden BP, Ward PB, Charles PGP, et al. Treatment outcomes for serious infections
     caused by methicillin-resistant Staphylococcus aureus with reduced vancomycin susceptibil-
     ity. Clin Infect Dis 2004;38:521–8.
[44] Charles PG, Ward PB, Johnson PD, et al. Clinical features associated with bacteremia due
     to heterogeneous vancomycin-intermediate Staphylococcus aureus. Clin Infect Dis 2004;38:
     448–51.
[45] 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;47:1262–6.
[46] Howden BP, Johnson PD, Ward PB, et al. Isolates with low-level vancomycin resistance
     associated with persistent methicillin-resistant Staphylococcus aureus bacteremia.
     Antimicrob Agents Chemother 2006;50:3049–57.
[47] Wong SS, Ng TK, Yam WC, et al. Bacteremia due to Staphylococcus aureus with reduced
     susceptibility to vancomycin. Diagn Microbiol Infect Dis 2000;36:261–8.
[48] Fridkin SK. Vancomycin-intermediate and -resistant Staphylococcus aureus: what the
     infectious disease specialist needs to know. Clin Infect Dis 2001;32:108–15.
[49] Bouza E, Burillo A, Munoz P. Antimicrobial therapy of Clostridium difficile–associated
     diarrhea. Med Clin North Am 2006;90:1141–63.
[50] Cunha BA, Klein NC. Vancomycin. In: Yoshikawa T, editor. Antibiotics in the elderly.
     New York: Marcel Dekker; 1994. p. 311–22.
VANCOMYCIN REVISITED                                     417

[51] Gump DW. Vancomycin for treatment of bacterial meningitis. Rev Infect Dis 1981;3:
     289–92.
[52] Nagl M, Neher C, Hager J, et al. Bactericidal activity of vancomycin in cerebrospinal fluid.
     Antimicrob Agents Chemother 1999;43:1932–4.
[53] Skhirtladze K, Hutschala H, Fleck T, et al. Impaired target site penetration of vancomycin
     in diabetic patients following cardiac surgery. Antimicrob Agents Chemother 2006;50:
     1372–5.
[54] Massias L, Dubois C, de Lentdecker P, et al. Penetration of vancomycin in uninfected
     sternal bone. Antimicrob Agents Chemother 1992;36:2539–41.
[55] Ackerman BH, Vannier AM. Necessity of a loading dose when using vancomycin in
     critically ill patients. J Antimicrob Chemother 1992;29:460–1.
[56] Cunha BA, Deglin J, Chow M, et al. Pharmacokinetics of vancomycin in patients undergo-
     ing chronic hemodialysis. Rev Infect Dis 1981;3:269–72.
[57] DelDot ME, Lipman J, Tett SE. Vancomycin pharmacokinetics in critically ill patients
     receiving continuous venovenous haemodiafiltration. Br J Clin Pharmacol 2004;58:
     259–68.
[58] Lutsar I, McCracken GH Jr, Friedland IR. Antibiotic pharmacodynamics in cerebrospinal
     fluid. Clin Infect Dis 1998;27:1117–27.
[59] Byl B, Jacobs F, Wallemacq P, et al. Vancomycin penetration of uninfected pleural fluid
     exudate after continuous or intermittent infusion. Antimicrob Agents Chemother 2003;
     47:2015–7.
[60] Cruciani M, Gatti G, Lazzarini L, et al. Penetration of vancomycin into human lung tissue.
     J Antimicrob Chemother 1996;38:865–9.
[61] Cruciani M, Gatti G, Lazzarini L, et al. Penetration of vancomycin in severe staphylococcal
     infections: prospective multicenter randomized study. Antimicrob Agents Chemother
     1996;38:865–9.
[62] Rybak MJ. The pharmacokinetic and pharmacodynamic properties of vancomycin. Clin
     Infect Dis 1994;18:544–6.
[63] Zimmermann AE, Katona BG, Plaisance KI. Association of vancomycin serum concentra-
     tions with outcomes in patients with gram-positive bacteremia. Pharmacotherapy 1995;15:
     85–91.
[64] James JK, Palmer SM, Levine DR, et al. Comparison of conventional dosing versus
     continuous-infusion vancomycin therapy for patients with suspected or documented
     gram-positive infections. Antimicrob Agents Chemother 1996;40:696–700.
[65] Kralovicov K, Spanik S, Halko J, et al. Do vancomycin serum levels predict failures of
     vancomycin therapy or nephrotoxicity in cancer patients? J Chemother 1997;9:420–6.
[66] Masur H, Francioli P, Ruddy M, et al. Vancomycin serum levels and toxicity in chronic
     haemodialysis patients with Staphylococcus aureus bacteremia. Clin Nephrol 1983;20:85–8.
[67] Andres I, Lopez R, Pou L, et al. Vancomycin monitoring: one or two serum levels? Ther
     Drug Monit 1997;19:614–9.
[68] Sayers JF, Shimasaki R. Routine monitoring of serum vancomycin concentrations: The
     answer lies in the middle. Clin Pharm 1988;7:18.
[69] Moellering RC Jr. Monitoring serum vancomycin levels: climbing the mountain because it
     is there? Clin Infect Dis 1994;18:544–6.
[70] Rodvold KA, Zokufa H, Rotschafer JC. Routine monitoring of serum vancomycin concen-
     trations: Can waiting be justified? Clin Pharm 1987;6:655–8.
[71] Freeman CD, Quintiliani R, Nightingale CH. Vancomycin therapeutic drug monitoring: Is
     it necessary. Ann Pharmacother 1993;27:594–8.
[72] Saunders NJ. Vancomycin administration and monitoring reappraisal. J Antimicrob
     Chemother 1995;36:279–82.
[73] Karam CM, McKinnon PS, Neuhauser MM, et al. Outcome assessment of minimizing
     vancomycin monitoring and dosing adjustments. Pharmacotherapy 1999;19:257–66.
418                                          CUNHA


[74] Darko W, Medicis JJ, Smith A, et al. Mississippi mud no more: cost-effectiveness of
     pharmacokinetic dosage adjustment of vancomycin to prevent nephrotoxicity. Pharmaco-
     therapy 2003;23:643–50.
[75] Cantu TG, Yamanaka-Yuen NA, Lietman PS. Serum vancomycin concentrations:
     reappraisal of their clinical value. Clin Infect Dis 1994;18:533–43.
[76] Cunha BA. Vancomycin serum levels: unnecessary, unhelpful, and costly. Antibiotics for
     Clinicians 2004;8:273–7.
[77] Cunha BA, Mohan SS, Hamid N, et al. Cost ineffectiveness of serum vancomycin levels.
     Eur J Clin Microbiol Infect Dis 2007;13:509–11.
[78] Allegaert K, van den Anker JN. Predictability of vancomycin pharmacokinetics in
     neonates. Eur J Clin Microbiol Infect Dis 2007;26:847–8.
[79] Cunha BA. Clinical manifestations and antimicrobial therapy of methicillin resistant
     Staphylococcus aureus (MRSA). Clin Microbiol Infect 2005;11:33–42.
[80] Alder JD. Staphylococcus aureus: antibiotic resistance and antibiotic selection. Antibiotics
     for Clinicians 2006;10(S1):19–24.
[81] Krol V, Cunha BA, Schoch PE, et al. Empiric gentamicin and vancomycin therapy for
     bacteremias in chronic dialysis outpatient units in the era of antibiotic resistance. J Chemo-
     ther 2006;18:490–3.
[82] Quale J, Landman D, Saurina G, et al. Manipulation of hospital antimicrobial formulary to
     control an outbreak of vancomycin-resistant enterococci. Clin Infect Dis 1996;23:1020–5.
[83] Saribas S, Bagdatli Y. Vancomycin tolerance in enterococci. Chemotherapy 2004;50:
     250–4.
[84] Watanakunakorn C. Antibiotic-tolerant Staphylococcus aureus. J Antimicrob Chemother
     1978;4:561–8.
[85] Cunha BA, Mickail N, Eisenstein L. E faecalis vancomycin sensitive enterococci (VSE)
     bacteremia unresponsive to vancomycin successfully treated with high dose daptomycin.
     Heart Lung 2007;36:456–61.
[86] Hussain FM, Boyle-Vavra S, Shete PB, et al. Evidence for a continuum of decreased
     vancomycin susceptibility in unselected Staphylococcus aureus clinical isolates. J Infect
     Dis 2002;186:661–7.
[87] Sakoulas G, Moise-Broder PA, Schentag JJ, et al. Relationship of MIC and bactericidal
     activity to efficacy of vancomycin for treatment of methicillin-resistant Staphylococcus
     aureus bacteremia. J Clin Microbiol 2004;42:2398–402.
[88] Schwaber MJ, Wright SB, Carmeli Y, et al. Clinical implications of varying degrees of
     vancomycin susceptibility in methicillin-resistant Staphylococcus aureus bacteremia. Emerg
     Infect Dis 2003;9:657–64.
[89] Srinivasan A, Dick JD, Perl TM. Vancomcyin resistance in staphylococci. Clin Microbiol
     Rev 2002;15:430–8.
[90] Walsh TR, Howe RA. The prevalence and mechanisms of vancomycin resistance in
     Staphylococcus aureus. Annu Rev Microbiol 2002;56:657–75.
[91] Wooton M, Walsh TR, Macgowan AP. Evidence for reduction in breakpoints used to
     determine vancomycin susceptibility in Staphylococcus aureus. Antimicrob Agents
     Cheomther 2005;49:3982–3.
[92] Ward PB, Johnson PD, Grabsch EA, et al. Treatment failure due to methicillin-resistant
     Staphylococcus aureus (MRSA) with reduced susceptibility to vancomycin. Med J Aust
     2001;175:480–3.
[93] Wooten M, MacGowan AP, Walsh TR. Comparative bactericidal activities of daptomycin
     and vancomycin against glycopeptide-intermediate Staphylococcus aureus (GISA) and
     Heterogenous GISA isolates. Antimicrob Agents Chemother 2006;50:4195–7.
[94] Burillo A, Bouza E. Epidemiology of MSSA and MRSA: colonization and infection.
     Antibiotics for Clinicians 2006;10(S1):3–10.
[95] Kim SH, Park WB, Lee KD, et al. Outcome of Staphylococcus aureus bacteremia in patients
     with eradicable foci versus noneradicable foci. Clin Infect Dis 2003;37:794–9.
VANCOMYCIN REVISITED                                      419

 [96] Evans M, Finch R. Approach to staphylococcal central IV line infections and bacteremias.
      Antibiotics for Clinicians 2006;10(S1):25–8.
 [97] Cunha BA. Persistent S aureus bacteremia: clinical pathway for diagnosis  treatment.
      Antibiotics for Clinicians 2006;10(S1):39–46.
 [98] Chang FY, Peacock JE Jr, Musher DM, et al. Staphylococcus aureus bacteremia: recurrence
      and the impact of antibiotic treatment in a prospective multicenter study. Medicine
      (Baltimore) 2003;82:333–9.
 [99] Lodise TP, McKinnon PS, Swiderski L, et al. Outcomes analysis of delayed antibiotic treatment
      for hospital-acquired Staphylococcus aureus bacteremia. Clin Infect Dis 2003;36:1418–23.
[100] Rotun SS, Mcmath V, Schoonmaker DJ, et al. Staphylococcus aureus with reduced suscep-
      tibility to vancomycin isolated from a patient with fatal bacteremia. Emerg Infect Dis 1999;
      5:147–9.
[101] Cunha BA. IV line infections. In: Cunha BA, editor. Infectious diseases in critical care
      medicine. 2nd edition. New York: Informa Healthcare; 2007.
[102] Morris AJ, Bilinsky RT. Prevention of staphylococcal shunt infections by continuous
      vancomycin prophylaxis. Am J Med Sci 1971;262:87–92.
[103] Eykyn S, Phillips I, Evans J. Vancomycin for staphylococcal shunt site infections in patients
      on regular haemodialysis. Br Med J 1970;3:8–82.
[104] Tofte RW, Solliday J, Rotschafer J, et al. Staphylococcus aureus infection of dialysis shunt:
      absence of synergy with vancomycin and rifampin. South Med J 1981;74:612–6.
[105] Cunha BA, Gill MV, Lazar J. Acute infective endocarditis. Infect Dis Clin North Am 1996;
      10:811–34.
[106] Cunha BA. MSSA/MRSA acute bacterial endocarditis (ABE): clinical pathway for
      diagnosis  treatment. Antibiotics for Clinicians 2006;10(S1):29–34.
[107] Brusch JL. Infective endocarditis. New York: Informa Healthcare; 2007. p. 143–272.
[108] Picazo JJ. Association for Health Research and Development (ACINDES). Management
      of the febrile neutropenic patient. Int J Antimicrob Agents 2005;2(S1):S120–2.
[109] Cunha BA. Clinical approach to antibiotic therapy in the penicillin allergic patient. Med
      Clin North Am 2006;90:1257–64.
[110] Bliziotis IA, Ntziora F, Lawrence KR, et al. Rifampin as adjuvant treatment of gram-
      positive bacterial infections: a systemic review of comparative clinical trials. Eur J Clin
      Microbiol Infect Dis 2007;26:849–56.
[111] Gentry CA, Rodvold KA, Novack RM, et al. Retrospective evaluation of therapies for
      Staphylococcus aureus endocarditis. Pharmacotherapy 1977;17:990–7.
[112] Geraci JE, Wilson WR. Vancomycin therapy for infective endocarditis. Rev Infect Dis
      1981;3(Suppl):S520–58.
[113] Small PM, Chambers HF. Vancomycin for Staphylococcal aureus endocarditis in intrave-
      nous drug users. Antimicrob Agents Chemother 1990;34:1227–31.
[114] Lee DG, Chun HS, Yim DS, et al. Efficacies of vancomycin, arbekacin, and gentamicin
      alone or in combination against methicillin-resistant Staphylococcus aureus in an in vitro
      infective endocarditis model. Antimicrob Agents Chemother 2003;47:3768–73.
[115] Levine DP, Fromm BS, Reddy BR. Slow response to vancomycin or vancomycin plus
      rifampin in methicillin-resistant Staphylococcus aureus endocarditis. Ann Intern Med
      1991;115:674–80.
[116] Shelburne SA, Musher DM, Hulten K, et al. In vitro killing of community-associated
      methicillin-resistant Staphylococcus aureus with drug combinations. Antimicrob Agents
      Chemother 2004;48:4016–9.
[117] Cha R, Brown WJ, Ryback MJ. Bactericidal activities of daptomycin, quinupristin-
      dalfopristin, and linezolid against vancomycin-resistant Staphylococcus aureus in an in
      vitro pharmacodynamic model with simulated endocardial vegetations. Antimicrob Agents
      Chemother 2003;47:3960–3.
[118] Kirst HA, Thompson DG, Nicas TI. Historical yearly usage of vancomycin. Antimicrob
      Agents Chemother 1998;42:1303–4.
420                                       CUNHA


[119] Stevens DL. The role of vancomycin in the treatment paradigm. Clin Infect Dis 2006;42:
      S51–7.
[120] Pope SD, Roecker A. Vancomycin for treatment of invasive, multi-drug resistant
      Staphylococcus aureus infections. Expert Opin Pharmacother 2007;8:1245–61.

More Related Content

What's hot

Micafungin vs. caspofungin in hsct
Micafungin vs. caspofungin in hsctMicafungin vs. caspofungin in hsct
Micafungin vs. caspofungin in hsctHarsh shaH
 
NORFLOXACIN AND ADVANCED CIRRHOSIS
NORFLOXACIN AND ADVANCED CIRRHOSISNORFLOXACIN AND ADVANCED CIRRHOSIS
NORFLOXACIN AND ADVANCED CIRRHOSISAjay Kandpal
 
Antibiotics guide
Antibiotics guide Antibiotics guide
Antibiotics guide Badheeb
 
Newer antibiotics for resistant gram +ve infections in icu
Newer antibiotics for resistant gram +ve infections in icuNewer antibiotics for resistant gram +ve infections in icu
Newer antibiotics for resistant gram +ve infections in icuDR.pankaj omar
 
Antibiotics in icu
Antibiotics in icuAntibiotics in icu
Antibiotics in icuimran80
 
Antibiotics 2015 in icu
Antibiotics 2015 in icuAntibiotics 2015 in icu
Antibiotics 2015 in icusamirelansary
 
Cefiderocol - Dr. ANCY GEORGE
Cefiderocol - Dr. ANCY GEORGECefiderocol - Dr. ANCY GEORGE
Cefiderocol - Dr. ANCY GEORGEAncy George
 
Antibiotics surgery
Antibiotics surgeryAntibiotics surgery
Antibiotics surgerysauvik2014
 
glycopeptides and lipopeptides
glycopeptides and lipopeptidesglycopeptides and lipopeptides
glycopeptides and lipopeptidesKGMU, Lucknow
 
Role of echinocandins in invasive fungal infection
Role of echinocandins in invasive fungal infectionRole of echinocandins in invasive fungal infection
Role of echinocandins in invasive fungal infectionHarsh shaH
 
DNA Gyrase Inhibitors -Quinolones,Fluoroquinolones
DNA Gyrase Inhibitors -Quinolones,Fluoroquinolones DNA Gyrase Inhibitors -Quinolones,Fluoroquinolones
DNA Gyrase Inhibitors -Quinolones,Fluoroquinolones Vijay Salvekar
 
Optimizing antimicrobial therapy for hospitalized pneumonia: Focus on PK/PD p...
Optimizing antimicrobial therapy for hospitalized pneumonia: Focus on PK/PD p...Optimizing antimicrobial therapy for hospitalized pneumonia: Focus on PK/PD p...
Optimizing antimicrobial therapy for hospitalized pneumonia: Focus on PK/PD p...WAidid
 
Drugs against bugs - antibiotics in the ICU
Drugs against bugs - antibiotics in the ICUDrugs against bugs - antibiotics in the ICU
Drugs against bugs - antibiotics in the ICUAndrew Ferguson
 
Newer antibiotics and uses
Newer antibiotics and usesNewer antibiotics and uses
Newer antibiotics and usesSujit Shrestha
 

What's hot (20)

New antibiotics 3 2014
New antibiotics   3   2014New antibiotics   3   2014
New antibiotics 3 2014
 
Micafungin vs. caspofungin in hsct
Micafungin vs. caspofungin in hsctMicafungin vs. caspofungin in hsct
Micafungin vs. caspofungin in hsct
 
streptogramin final
streptogramin finalstreptogramin final
streptogramin final
 
Antibiotics in icu
Antibiotics in icuAntibiotics in icu
Antibiotics in icu
 
NORFLOXACIN AND ADVANCED CIRRHOSIS
NORFLOXACIN AND ADVANCED CIRRHOSISNORFLOXACIN AND ADVANCED CIRRHOSIS
NORFLOXACIN AND ADVANCED CIRRHOSIS
 
Quinolones
QuinolonesQuinolones
Quinolones
 
Antibiotics guide
Antibiotics guide Antibiotics guide
Antibiotics guide
 
Newer antibiotics for resistant gram +ve infections in icu
Newer antibiotics for resistant gram +ve infections in icuNewer antibiotics for resistant gram +ve infections in icu
Newer antibiotics for resistant gram +ve infections in icu
 
Antibiotics in icu
Antibiotics in icuAntibiotics in icu
Antibiotics in icu
 
Antibiotics 2015 in icu
Antibiotics 2015 in icuAntibiotics 2015 in icu
Antibiotics 2015 in icu
 
Cefiderocol - Dr. ANCY GEORGE
Cefiderocol - Dr. ANCY GEORGECefiderocol - Dr. ANCY GEORGE
Cefiderocol - Dr. ANCY GEORGE
 
Antibiotics surgery
Antibiotics surgeryAntibiotics surgery
Antibiotics surgery
 
Rational use of Antibiotics
Rational use of AntibioticsRational use of Antibiotics
Rational use of Antibiotics
 
glycopeptides and lipopeptides
glycopeptides and lipopeptidesglycopeptides and lipopeptides
glycopeptides and lipopeptides
 
Role of echinocandins in invasive fungal infection
Role of echinocandins in invasive fungal infectionRole of echinocandins in invasive fungal infection
Role of echinocandins in invasive fungal infection
 
DNA Gyrase Inhibitors -Quinolones,Fluoroquinolones
DNA Gyrase Inhibitors -Quinolones,Fluoroquinolones DNA Gyrase Inhibitors -Quinolones,Fluoroquinolones
DNA Gyrase Inhibitors -Quinolones,Fluoroquinolones
 
Optimizing antimicrobial therapy for hospitalized pneumonia: Focus on PK/PD p...
Optimizing antimicrobial therapy for hospitalized pneumonia: Focus on PK/PD p...Optimizing antimicrobial therapy for hospitalized pneumonia: Focus on PK/PD p...
Optimizing antimicrobial therapy for hospitalized pneumonia: Focus on PK/PD p...
 
Drugs against bugs - antibiotics in the ICU
Drugs against bugs - antibiotics in the ICUDrugs against bugs - antibiotics in the ICU
Drugs against bugs - antibiotics in the ICU
 
Antibiotic choices
Antibiotic choicesAntibiotic choices
Antibiotic choices
 
Newer antibiotics and uses
Newer antibiotics and usesNewer antibiotics and uses
Newer antibiotics and uses
 

Similar to Vancomycin revisited ccc 2008

Vancomycin Dosing in Patients on Intermittent Hemodialysis
Vancomycin Dosing in Patients on Intermittent HemodialysisVancomycin Dosing in Patients on Intermittent Hemodialysis
Vancomycin Dosing in Patients on Intermittent HemodialysisMahmoud Eid
 
Invitro Potency Analysis of Vancomyc in Capsules by Microbiological Assay
Invitro Potency Analysis of Vancomyc in Capsules by Microbiological AssayInvitro Potency Analysis of Vancomyc in Capsules by Microbiological Assay
Invitro Potency Analysis of Vancomyc in Capsules by Microbiological Assayiosrjce
 
7 Years of Experience in Osteomyelitis Management in Caracas, Venezuela_Crims...
7 Years of Experience in Osteomyelitis Management in Caracas, Venezuela_Crims...7 Years of Experience in Osteomyelitis Management in Caracas, Venezuela_Crims...
7 Years of Experience in Osteomyelitis Management in Caracas, Venezuela_Crims...CrimsonpublishersCJMI
 
Optimizing vancomycin enterococci resistant vre therapy 2008
Optimizing  vancomycin enterococci resistant vre therapy 2008Optimizing  vancomycin enterococci resistant vre therapy 2008
Optimizing vancomycin enterococci resistant vre therapy 2008negrulo2013
 
Multidrug resistance in Microbes
Multidrug resistance in MicrobesMultidrug resistance in Microbes
Multidrug resistance in MicrobesArjun Kumar
 
Antibiotic in ED
Antibiotic in EDAntibiotic in ED
Antibiotic in EDEM OMSB
 
Resistance in enterococci
Resistance in enterococciResistance in enterococci
Resistance in enterococciNeelima Singh
 
Main antimicrobial resistance pattern in bacteria.pptx
Main antimicrobial resistance pattern in bacteria.pptxMain antimicrobial resistance pattern in bacteria.pptx
Main antimicrobial resistance pattern in bacteria.pptxShinee13
 
SUPERBUGS , NEW HOPE NEW ANTIBIOTICS
SUPERBUGS , NEW HOPE NEW ANTIBIOTICSSUPERBUGS , NEW HOPE NEW ANTIBIOTICS
SUPERBUGS , NEW HOPE NEW ANTIBIOTICSAhmed Sarhan
 
Management of Skin and Soft Tissue Infections: IDSA Guideline 2014
Management of Skin and Soft Tissue Infections: IDSA Guideline 2014Management of Skin and Soft Tissue Infections: IDSA Guideline 2014
Management of Skin and Soft Tissue Infections: IDSA Guideline 2014Sun Yai-Cheng
 
18. treatment of infectious diseases
18. treatment of infectious diseases18. treatment of infectious diseases
18. treatment of infectious diseasesAhmad Hamadi
 
Antibiotic resistance a global concern part ii
Antibiotic resistance  a global concern part iiAntibiotic resistance  a global concern part ii
Antibiotic resistance a global concern part iiRohan Jagdale
 
Vancomycin resistant bacteria
Vancomycin resistant bacteriaVancomycin resistant bacteria
Vancomycin resistant bacteriaZablon Faith
 
Antibiotics and Neonatal Sepsis
Antibiotics and Neonatal SepsisAntibiotics and Neonatal Sepsis
Antibiotics and Neonatal SepsisKing_maged
 
Antibiotic use in an intensive care setting iacm, medicine update 2012
Antibiotic use in an intensive care setting   iacm, medicine update 2012Antibiotic use in an intensive care setting   iacm, medicine update 2012
Antibiotic use in an intensive care setting iacm, medicine update 2012Sachin Adukia
 

Similar to Vancomycin revisited ccc 2008 (20)

Vancomycin Dosing in Patients on Intermittent Hemodialysis
Vancomycin Dosing in Patients on Intermittent HemodialysisVancomycin Dosing in Patients on Intermittent Hemodialysis
Vancomycin Dosing in Patients on Intermittent Hemodialysis
 
Invitro Potency Analysis of Vancomyc in Capsules by Microbiological Assay
Invitro Potency Analysis of Vancomyc in Capsules by Microbiological AssayInvitro Potency Analysis of Vancomyc in Capsules by Microbiological Assay
Invitro Potency Analysis of Vancomyc in Capsules by Microbiological Assay
 
7 Years of Experience in Osteomyelitis Management in Caracas, Venezuela_Crims...
7 Years of Experience in Osteomyelitis Management in Caracas, Venezuela_Crims...7 Years of Experience in Osteomyelitis Management in Caracas, Venezuela_Crims...
7 Years of Experience in Osteomyelitis Management in Caracas, Venezuela_Crims...
 
Antibiotic update in icu
Antibiotic update in icuAntibiotic update in icu
Antibiotic update in icu
 
Optimizing vancomycin enterococci resistant vre therapy 2008
Optimizing  vancomycin enterococci resistant vre therapy 2008Optimizing  vancomycin enterococci resistant vre therapy 2008
Optimizing vancomycin enterococci resistant vre therapy 2008
 
Antibiotics in PICU.pptx
Antibiotics in PICU.pptxAntibiotics in PICU.pptx
Antibiotics in PICU.pptx
 
Multidrug resistance in Microbes
Multidrug resistance in MicrobesMultidrug resistance in Microbes
Multidrug resistance in Microbes
 
AUPDATE
AUPDATEAUPDATE
AUPDATE
 
Antibiotic in ED
Antibiotic in EDAntibiotic in ED
Antibiotic in ED
 
Resistance in enterococci
Resistance in enterococciResistance in enterococci
Resistance in enterococci
 
antibiotics in ICU
antibiotics in ICUantibiotics in ICU
antibiotics in ICU
 
Main antimicrobial resistance pattern in bacteria.pptx
Main antimicrobial resistance pattern in bacteria.pptxMain antimicrobial resistance pattern in bacteria.pptx
Main antimicrobial resistance pattern in bacteria.pptx
 
SUPERBUGS , NEW HOPE NEW ANTIBIOTICS
SUPERBUGS , NEW HOPE NEW ANTIBIOTICSSUPERBUGS , NEW HOPE NEW ANTIBIOTICS
SUPERBUGS , NEW HOPE NEW ANTIBIOTICS
 
Management of Skin and Soft Tissue Infections: IDSA Guideline 2014
Management of Skin and Soft Tissue Infections: IDSA Guideline 2014Management of Skin and Soft Tissue Infections: IDSA Guideline 2014
Management of Skin and Soft Tissue Infections: IDSA Guideline 2014
 
18. treatment of infectious diseases
18. treatment of infectious diseases18. treatment of infectious diseases
18. treatment of infectious diseases
 
Antibiotic resistance a global concern part ii
Antibiotic resistance  a global concern part iiAntibiotic resistance  a global concern part ii
Antibiotic resistance a global concern part ii
 
Vancomycin resistant bacteria
Vancomycin resistant bacteriaVancomycin resistant bacteria
Vancomycin resistant bacteria
 
Mrsa
MrsaMrsa
Mrsa
 
Antibiotics and Neonatal Sepsis
Antibiotics and Neonatal SepsisAntibiotics and Neonatal Sepsis
Antibiotics and Neonatal Sepsis
 
Antibiotic use in an intensive care setting iacm, medicine update 2012
Antibiotic use in an intensive care setting   iacm, medicine update 2012Antibiotic use in an intensive care setting   iacm, medicine update 2012
Antibiotic use in an intensive care setting iacm, medicine update 2012
 

More from negrulo2013

Recomendaciones sobre el diagnóstico de la enfermedad fúngica invasora seimc
Recomendaciones sobre el diagnóstico de la enfermedad fúngica invasora seimcRecomendaciones sobre el diagnóstico de la enfermedad fúngica invasora seimc
Recomendaciones sobre el diagnóstico de la enfermedad fúngica invasora seimcnegrulo2013
 
Nuevos agentes para el tratamiento de la hepatitis c
Nuevos agentes para el tratamiento de la hepatitis cNuevos agentes para el tratamiento de la hepatitis c
Nuevos agentes para el tratamiento de la hepatitis cnegrulo2013
 
Normas relacsalud
Normas relacsaludNormas relacsalud
Normas relacsaludnegrulo2013
 
Manualde organizacion
Manualde organizacionManualde organizacion
Manualde organizacionnegrulo2013
 
Man orginfectologia
Man orginfectologiaMan orginfectologia
Man orginfectologianegrulo2013
 
International clinical practice guidelines for the treatment of acute uncompl...
International clinical practice guidelines for the treatment of acute uncompl...International clinical practice guidelines for the treatment of acute uncompl...
International clinical practice guidelines for the treatment of acute uncompl...negrulo2013
 
Guideline useofantimicrobialagentsinneutropenicpatients
Guideline useofantimicrobialagentsinneutropenicpatientsGuideline useofantimicrobialagentsinneutropenicpatients
Guideline useofantimicrobialagentsinneutropenicpatientsnegrulo2013
 
Dosificación de antibióticos en el tratamiento de las infecciones por sarm
Dosificación de antibióticos en el tratamiento de las infecciones por sarmDosificación de antibióticos en el tratamiento de las infecciones por sarm
Dosificación de antibióticos en el tratamiento de las infecciones por sarmnegrulo2013
 
Diagnóstico de laboratorio de la enfermedad fúngica invasora
Diagnóstico de laboratorio de la enfermedad fúngica invasoraDiagnóstico de laboratorio de la enfermedad fúngica invasora
Diagnóstico de laboratorio de la enfermedad fúngica invasoranegrulo2013
 
Codigoeticaprofesional
CodigoeticaprofesionalCodigoeticaprofesional
Codigoeticaprofesionalnegrulo2013
 
Clinical practice guidelines by the infectious diseases society of america fo...
Clinical practice guidelines by the infectious diseases society of america fo...Clinical practice guidelines by the infectious diseases society of america fo...
Clinical practice guidelines by the infectious diseases society of america fo...negrulo2013
 
Boletin del programa provincial de inmunizaciones2012
Boletin del programa provincial de inmunizaciones2012Boletin del programa provincial de inmunizaciones2012
Boletin del programa provincial de inmunizaciones2012negrulo2013
 
Afiche ii jornadas cuyanas hiv 2012
Afiche ii jornadas cuyanas hiv 2012Afiche ii jornadas cuyanas hiv 2012
Afiche ii jornadas cuyanas hiv 2012negrulo2013
 
Abordaje integral de las infecciones respiratorias agudas
Abordaje integral de las infecciones respiratorias agudasAbordaje integral de las infecciones respiratorias agudas
Abordaje integral de las infecciones respiratorias agudasnegrulo2013
 

More from negrulo2013 (16)

Recomendaciones sobre el diagnóstico de la enfermedad fúngica invasora seimc
Recomendaciones sobre el diagnóstico de la enfermedad fúngica invasora seimcRecomendaciones sobre el diagnóstico de la enfermedad fúngica invasora seimc
Recomendaciones sobre el diagnóstico de la enfermedad fúngica invasora seimc
 
Nuevos agentes para el tratamiento de la hepatitis c
Nuevos agentes para el tratamiento de la hepatitis cNuevos agentes para el tratamiento de la hepatitis c
Nuevos agentes para el tratamiento de la hepatitis c
 
Normas relacsalud
Normas relacsaludNormas relacsalud
Normas relacsalud
 
Manualde organizacion
Manualde organizacionManualde organizacion
Manualde organizacion
 
Mgi07102
Mgi07102Mgi07102
Mgi07102
 
Man orginfectologia
Man orginfectologiaMan orginfectologia
Man orginfectologia
 
International clinical practice guidelines for the treatment of acute uncompl...
International clinical practice guidelines for the treatment of acute uncompl...International clinical practice guidelines for the treatment of acute uncompl...
International clinical practice guidelines for the treatment of acute uncompl...
 
Guideline useofantimicrobialagentsinneutropenicpatients
Guideline useofantimicrobialagentsinneutropenicpatientsGuideline useofantimicrobialagentsinneutropenicpatients
Guideline useofantimicrobialagentsinneutropenicpatients
 
Dosificación de antibióticos en el tratamiento de las infecciones por sarm
Dosificación de antibióticos en el tratamiento de las infecciones por sarmDosificación de antibióticos en el tratamiento de las infecciones por sarm
Dosificación de antibióticos en el tratamiento de las infecciones por sarm
 
Diagnóstico de laboratorio de la enfermedad fúngica invasora
Diagnóstico de laboratorio de la enfermedad fúngica invasoraDiagnóstico de laboratorio de la enfermedad fúngica invasora
Diagnóstico de laboratorio de la enfermedad fúngica invasora
 
Codigoeticaprofesional
CodigoeticaprofesionalCodigoeticaprofesional
Codigoeticaprofesional
 
Clinical practice guidelines by the infectious diseases society of america fo...
Clinical practice guidelines by the infectious diseases society of america fo...Clinical practice guidelines by the infectious diseases society of america fo...
Clinical practice guidelines by the infectious diseases society of america fo...
 
Botulismo uti
Botulismo utiBotulismo uti
Botulismo uti
 
Boletin del programa provincial de inmunizaciones2012
Boletin del programa provincial de inmunizaciones2012Boletin del programa provincial de inmunizaciones2012
Boletin del programa provincial de inmunizaciones2012
 
Afiche ii jornadas cuyanas hiv 2012
Afiche ii jornadas cuyanas hiv 2012Afiche ii jornadas cuyanas hiv 2012
Afiche ii jornadas cuyanas hiv 2012
 
Abordaje integral de las infecciones respiratorias agudas
Abordaje integral de las infecciones respiratorias agudasAbordaje integral de las infecciones respiratorias agudas
Abordaje integral de las infecciones respiratorias agudas
 

Vancomycin revisited ccc 2008

  • 1. Crit Care Clin 24 (2008) 393–420 Vancomycin Revisited: A Reappraisal of Clinical Use Burke A. Cunha, MD, MACPa,b,* a Infectious Disease Division, Winthrop-University Hospital, Mineola, NY 11501, USA b State University of New York, School of Medicine, Stony Brook, NY, USA Over the years, vancomycin has become the mainstay of methicillin- resistant Staphylococcus aureus (MRSA) therapy. Although vancomycin is active against a variety of other gram-positive organisms, other antibiotics are preferred to treat infections due to these organisms (Table 1). In critical care medicine, vancomycin has been mainly used to treat infections where MRSA is the presumed cause of infection. These include central intravenous line infections, soft tissue infections, bone infections, shunt infections in hemodialysis patients, bacteremia, and acute bacterial endocarditis. The widespread use of empiric vancomycin has resulted in adverse effects [1–5]. The initial unpurified formulations of vancomycin were associated with reports of potential nephrotoxicity [6,7]. Subsequently, the purified prepara- tions of vancomycin have been used and have not been associated with neph- rotoxicity. There is no evidence for vancomycin monotherapy–associated nephrotoxicity [8,9]. The few reports of potential vancomycin nephrotoxicity described patients receiving vancomycin and known nephrotoxic medica- tions. Vancomycin plus an aminoglycoside does not appear to increase the nephrotoxic potential of the aminoglycoside [10–15]. Endotoxin or cytokine release from gram-negative bacilli treated with an aminoglycoside and van- comycin may be responsible for an increase in creatinine in some cases, which may have been mistakenly ascribed to vancomycin toxicity [16]. Because van- comycin monotherapy is not nephrotoxic, the use of vancomycin serum levels to avoid nephrotoxicity has no basis [3,17,18]. Because vancomycin is renally eliminated by glomerular filtration, vancomycin dosing in renal in- sufficiency can be accurately dosed based on the creatinine clearance (CrCl), (ie, the daily dose of vancomycin should be reduced in proportion to the * Infectious Disease Division, Winthrop-University Hospital, 259 First Street, Mineola, NY 11501. 0749-0704/08/$ - see front matter Ó 2008 Elsevier Inc. All rights reserved. doi:10.1016/j.ccc.2007.12.012 criticalcare.theclinics.com
  • 2. 394 CUNHA Table 1 Vancomycin: clinically useful microbiologic spectrum Clinically effective Clinically ineffective Gram-positive aerobic cocci Gram-negative aerobic cocci Staphylococci (MSSA/MRSA), Neisseria meningitidis Nonenterococcal streptococci (Groups A, B, C, G) Staphylococcus epidermidis (coagulase negative staphylococci) (MSSE/MRSE) Penicillin-resistant Streptococcus pneumoniae Gram-positive anaerobic cocci Gram-negative aerobic bacilli Peptococcus Escherichia coli Peptostreptococcus Klebsiella pneumoniae Pseudomonas aeruginosa Gram-positive aerobic bacilli Gram-positive aerobic bacilli Corynebacterium sp Listeria monocytogenes Lactobacillus sp Gram-positive anaerobic bacilli Gram-negative anaerobic bacilli Clostridium sp Bacteroides fragilis Group D enterococci Group D enterococci Enterococcus faecalis (VSE)a Enterococcus faecium (VRE) Abbreviations: MRSE, methicillin-resistant Staphylococcus epidermidis; MSSA, methicillin- sensitive Staphylococcus aureus; MSSE, methicillin-sensitive Staphylococcus epidermidis; VRE, vancomycin-resistant enterococci; VSE, vancomycin-sensitive enterococci. a Only when combined with an aminoglycoside (eg, gentamicin). Data from Refs. [1,3,4]. decrease in renal function). In those responding to vancomycin therapy and in those with a normal volume of distribution (Vd), vancomycin levels are unhelpful, expensive, and unnecessary for vancomycin dosing [17–21]. The pharmacokinetic and pharmacodynamic characteristics of vancomy- cin have been well studied. Vancomycin obeys both ‘‘concentration depen- dent’’ kinetics at concentrations O MIC and ‘‘concentration independent’’ kinetics at concentrations ! MIC [1,3,19,22]. Because nephrotoxicity is not a consideration, high-dose vancomycin (eg, 2 g intravenously every 12 hours [60 mg/kg/d]) has been used in special situations, eg, osteomyelitis without nephrotoxicity [23,24]. After years of clinical experience, vancomycin side effects have become more fully appreciated. In addition to ‘‘red neck’’ or ‘‘red man’’ syndrome, leukopenia, thrombocytopenia, and, rarely, sudden death have been ascribed to vancomycin [1,3,25]. Extensive vancomycin use over the years has resulted in two major prob- lems. Firstly, excessive use of vancomycin has resulted in an increased prevalence of vancomycin-resistant enterococci (VRE) worldwide. Vanco- mycin-sensitive enterococci (VSE) represent the main group D enterococcal component of feces. Vancomycin has sufficient anti-VSE activity to decrease VSE in the fecal flora, resulting in a commensurate increase VRE in stools [26–30]. Although the spectrum of infection caused by VSE and VRE are the same, the number of antimicrobials available to treat VRE is limited, making the therapy of VRE more difficult than the therapy of VSE [31,32].
  • 3. VANCOMYCIN REVISITED 395 Another negative effect of vancomycin use over the years has been a rel- ative increase S aureus resistance [33–37]. Vancomycin therapy results in cell-wall thickening of S aureus strains, of both methicillin-sensitive S aureus (MSSA) and methicillin-resistant S aureus (MRSA). Vancomycin-mediated cell-wall thickening results in ‘‘permeability mediated’’ resistance to vanco- mycin as well as to other anti-MSSA and anti-MRSA antibiotics [38–42]. Vancomycin-induced ‘‘permeability-mediated’’ resistance is manifested mi- crobiologically by increased minimum inhibitory concentrations (MICs) and clinically by delayed resolution or therapeutic failure in treating staph- ylococcal bacteremias or acute bacterial endocarditis [43–48]. To avoid selecting out heteroresistant vancomycin-intermediate S aureus (hVISA), vancomycin use should be minimized and other MRSA antibiotics should preferentially be used instead (eg, minocycline, daptomycin, line- zolid, or tigecycline) [1,31,32]. Vancomycin pharmacokinetics and pharmacodynamics Although available orally and intravenously, vancomycin is primarily administered via the intravenous route in the critical care setting. Oral van- comycin is the preferred therapy for Clostridium difficile diarrhea because it is not absorbed, is active against C difficile, and achieves high intraluminal concentrations in the colon [1,3,32]. Because intravenous vancomycin does not penetrate into the bowel lumen, intravenous vancomycin has no role in the treatment of C difficile diarrhea or colitis [1,3,5,49]. Vancomycin is usually administered as 1 g (intravenous) every 12 hours. After a 1-g intravenous dose, vancomycin serum concentrations are predict- ably R25 mg/mL [1,3,5,50]. High dose of vancomycin (eg, 2 g intravenously every 12 hours) has been used in the treatment of central nervous system (CNS) infections to overcome the relatively poor cerebrospinal fluid (CSF) penetration of vancomycin (CSF penetration with noninflamed meninges is 0% and with inflamed meninges is only 15% of simultaneous serum concen- trations) and osteomyelitis (achieves serum trough levels R 15 mg/mL [1,32,51,52]. For cardiopulmonary bypass (CBP) prophylaxis, use 1 g intrave- nously preoperatively because vancomycin is rapidly removed during CBP [53,54]. Burn patients have increased Vd and may require higher doses of van- comycin. Intravenous drug abusers (IVDAs) have higher vancomycin renal clearances than do non-IVDAs, and may require higher doses (Box 1) [1]. Vancomycin is not removed by hemodialysis [1,3]. In anuric patients a 1 g (IV) dose results in peak serum concentrations of about 48 mg/mL. Vancomycin’s mean elimination half-life is 7.5 days (vancomycin serum concentration is 3.5 mg/mL after 18 days) [1,32,55,56]. In patients using new ‘‘high flux’’ membranes for hemodialysis, about 40% of vancomycin is removed. Therefore, a post–‘‘high flux’’ hemodialysis dose (eg, 500 mg intravenously, which is w50% of the anuric dose) should be administered post-hemodialysis [57].
  • 4. 396 CUNHA Box 1. Vancomycin: pharmacokinetic and pharmacodynamic characteristics Pharmacokinetic parameters Usual adult dose: 1 g intravenously every 12 hours (30 mg/kg/ d); 2 g intravenously every 12 hours (60 mg/kg/d) Peak serum levels: 63 mg/mL (1-g dose); 120 mg/mL (2-g dose) Excreted unchanged (urine): 90% Serum half-life: 6 hours (normal); 180 hours (end-stage renal disease) Plasma protein binding: 55% Vd: 0.7 L/kg Therapeutic serum levels: Peak 1 g 25 mg/mL; 2g 50 mg/mL Trough: 1 g ‚ 7.5 m/mL; 2 g ‚ 15 m/mL Pharmacodynamic characteristics ‘‘Concentration dependent’’ kinetics at concentrations MIC ‘‘Concentration independent’’ kinetics (time dependent) at concentrations MIC Primary mode of elimination: renal Dosage adjustments: CrCl 50–80 mL/min: dose = 500 mg intravenously every 12 hours CrCl 10–50 mL/min: dose = 500 mg intravenously every 24 hours CrCl 10 mL/min: dose = 1 g intravenous every week Post-hemodialysis dose: none Post–‘‘high flux’’ hemodialysis dose: 500 mg intravenous every 12 hours Post–peritoneal dialysis dose: none CVVH dose: 1 gm intravenous every 24 hours Data from Cunha BA, editor. Antibiotic essentials, 7th edition. Royal Oak (MI): Physicians Press; 2008; with permission. Peritoneal dialysis (PD) removes no vancomycin. Therefore, after an ini- tial 1-g intravenous dose, vancomycin should be dosed for a CrCl less than 10 mL/min and no post-PD dose is necessary [1,32]. For patients with peri- tonitis on chronic ambulatory peritoneal dialysis (CAPD), vancomycin may be added to peritoneal dialysis fluid. A 1-g intravenous loading dose of vancomycin may be given to such patients after dialysis, followed by the renally adjusted maintenance anuric dose [1,32]. In addition, to maintain equilibrium between the dialysate fluid and the serum compartment,
  • 5. VANCOMYCIN REVISITED 397 vancomycin may also be given intraperitoneally (2–30 mg of vancomycin per liter of dialysis fluid) with each CAPD [1,32]. Vancomycin has been used to treat CNS infections, but CNS penetration of vancomycin is vari- able. Vancomycin administered intravenously achieves only about 15% of simultaneous serum levels in the CSF in patients with meningitis. If high CSF concentrations are desired for treatment of gram-positive shunt infec- tions, give 2 g intravenously every 12 hours until shunt removal. Intrave- nous vancomycin may be supplemented by intrathecal doses (ie, vancomycin 10–20 mg/d intrathecally), preferably administered via Om- maya reservoir [1,3,32,58]. With normal renal function, about 90% of intravenous vancomycin is eliminated unchanged in the urine during the first 24 hours. After a single 1-g intravenous dose of vancomycin, urine concentrations are w500 mg/mL, which are maintained for 24 hours [1,3]. Intravenously administered van- comycin does not concentrate well into, bile (30%), synovial fluid, pleural fluid, or lung parenchyma [1,58–62]. Intravenous vancomycin diffuses well into most other body compart- ments except feces, aqueous humor, and CSF [1,60]. While vancomycin pen- etrates poorly into CSF, it penetrates well into brain tissue (Box 2) [1,6,32,51,52,58]. Vancomycin levels Vancomycin is eliminated by glomerular filtration. The serum half-life of IV vancomycin is 6 hours with normal renal function, and 7 days in anuria [1,6,56,62]. Many approaches to vancomycin dosing for various degrees of renal insufficiency have been devised and all are based on CrCl [1,6,21]. Formerly, vancomycin was administered as 500 mg intravenously every 6 hours. Today vancomycin is usually administered as 1 g intravenously every 12 hours (for adults with a normal Vd and CrCl) [22,63,64]. For seri- ous systemic infections due to susceptible organisms, is to administer vanco- mycin 1 g intravenously every 12 hours (15 mg/kg/d) or 2 g intravenously every 12 hours (30 mg/kg/d) [1,3,32]. Even when used in prolonged high doses for special situations, such as for MRSA osteomyelitis (2 g intrave- nously every 12 hours [30 mg/kg/d]), there has been no nephrotoxicity [23,24]. Since vancomycin was thought to be nephrotoxic, vancomycin dosing was based on vancomycin levels [65,66]. However, vancomycin has no neph- rotoxic potential. Thus, there is no rationale for using routine vancomycin levels to dose patients to avoid nephrotoxicity. Accurate vancomycin dosing is readily achieved more quickly, simply, less expensively, and without risk of nephrotoxicity by dosing vancomycin based on CrCl rather then by van- comycin levels [67–72]. It has been shown that vancomycin levels do not re- duce nonexistent toxicity but vancomycin levels often result in unnecessary vancomycin dosing changes [73–78].
  • 6. 398 CUNHA Box 2. Vancomycin tissue concentrations* Bone concentrations Cortical bone Uninfected: w5% Infected: w10% Cancellous bone Uninfected: w5% Infected: 5% Synovial fluid concentrations Uninflamed synovial fluid: w20% Urine Concentrations Uninfected urine: 100% (normal renal function) Stool concentrations Feces: Intravenous: 0% Orally: 1000% CSF concentrations Uninflamed meninges: 0% (prophylaxis of CSF seeding) Inflamed meninges: 15% (therapy of acute bacterial meningitis) Heart concentrations Cardiac valves: 20% Lung concentrations Lung parenchyma: 10% Bile concentrations Unobstructed bile: w50% Ascitic fluid concentrations Ascites: 10% * Relative to simultaneous serum levels There is often considerable delay between the time vancomycin peaks and troughs are reported [70,74–76]. Two or more days have usually elapsed when dosing adjustments are made, which means adjustments in vancomycin dosing are based on previous, but no longer relevant estimates of, renal function (CrCl) [70,76]. The other practical difficulty with vanco- mycin serum peak and trough levels is in the timing of the samples which influences interpretation of the levels. This, in addition, causes unnecessar- ily frequent changes in vancomycin dosing, which serve no clinical purpose [1,6,32,70–78]. In anuria, on chronic hemodialysis, vancomycin peak serum concentra- tions are predictable and vancomycin serum levels are unnecessary. In
  • 7. VANCOMYCIN REVISITED 399 patients receiving vancomycin on ‘‘high flux’’ hemodialysis, a posthemodial- ysis dose should be given to replace the vancomycin removed. Since about 40% of vancomycin is removed by ‘‘high flux’’ hemodialysis, a 500-mg in- travenous posthemodialysis dose should be given [57]. Vancomycin serum concentrations may be used to assess adequacy of therapy in patients with unusual Vd, such as burn patients, or in those patients not responding to appropriately dosed vancomycin (ie, patients showing vancomycin ‘‘tolerance’’ or resistance). For these purposes, vanco- mycin serum levels should be obtained. Except to assess therapeutic efficacy or ‘‘apparent therapeutic failure’’ there is no rationale for obtaining vanco- mycin serum levels [69–78]. Vancomycin microbiologic activity and efficacy Antistaphylococcal activity Staphylococcus epidermidis The main use of parenteral vancomycin over the years has been in the treatment of Staphylococci epidermidis, (ie, coagulase-negative staphylococci (CoNS) infections, methicillin-sensitive S epidermidis (MSSE), and methicil- lin-resistant S epidermidis (MRSE) infections). CoNS infections are usually related to prosthetic implant materials (ie, infections involving prosthetic joints, prosthetic heart valves, and CNS shunts). These infections are diffi- cult to eradicate with antimicrobial therapy alone because antibiotics are unable to eradicate these organisms in the glycocalyx on prosthetic materials. Methicillin-resistant Staphylococcus aureus Vancomycin has been used to treat serious staphylococcal infections, especially MRSA. The prevalence of MRSA has increased during the past several decades with three clinical variants recognized. Two of these are hospital-acquired MRSA (HA-MRSA) and community-onset MRSA (CO-MRSA). CO-MRSA strains originate in the hospital, circulate in the community, and subsequently have their onset in the community before be- ing readmitted to the hospital. A third is community-acquired (CA-MRSA), a term often mistakenly applied to CO-MRSA because both come from the community. However, CA-MRSA infections have two distinctive clinical presentations readily differentiating them from CO-MRSA strains, which represent the majority of MRSA admitted from the community (community onset) to the hospital. CA-MRSA infections usually present either as severe pyoderma or as necrotizing/hemorragic CAP. Although still rare, these two clinical CA- MRSA syndromes are recognizable by their clinical presentations. Strains of CA-MRSA, particularly those with the Panton-Valentine leukocidin gene (PVL positive), are unusually virulent with a high degree of cytotoxic
  • 8. 400 CUNHA activity, which accounts for their extensive tissue destruction and two unique clinical presentations. While these strains are genetically distinctive (ie, Staphylococcal chromosome cassette [SCC] mec IV), most hospital laboratories cannot perform studies to identify PVL positive strains. Clini- cians must rely on the clinical presentation to identify CA-MRSA strains versus the great majority of MRSA strains from the community, which are nearly all CO-MRSA. In spite of the increased virulence of CA-MRSA PVL positive strains, these organisms are surprisingly sensitive to older antibiotics, such as doxycycline, clindamycin, and trimethoprim-sulfame- thoxazole (TMP-SMX), but these antibiotics are not effective against HA-MRSA strains. Vancomycin, linezolid, daptomycin, minocycline, and tigecycline are active against HA-MRSA, CO-MRSA, and CA-MRSA strains. For CA-MRSA necrotizing pyodermas, treat with surgical de- bridement and a HA-MRSA/CO-MRSA antibiotic, such as vancomycin, linezolid, daptomycin, or tigecycline. For CA-MRSA CAP with influenza- like illness, treat with influenza antivirals and linezolid [79,80]. Group D enterococci Since vancomycin was the initial antibiotic effective against MRSA, its excessive use over time has resulted in two major clinical problems. Firstly, extensive intravenous (not oral) vancomycin use has resulted in an increase in the prevalence of E faecium (VRE), as well as increased vancomycin re- sistance [81]. The increase in VRE isolates is explained by the activity of vancomycin against the VSE component in the fecal flora, which is the pre- dominant enterococcal species in the fecal flora. By decreasing VSE in the fecal flora, there is a commensurate increase in VRE in feces. There has been an increase worldwide in VRE prevalence due to vancomycin over use. Instead, other anti-MRSA antibiotics that do not have this effect should be used [26,81,82]. Vancomycin tolerance and resistance Enterococci Enterococci and, to a lesser extent, staphylococci may develop ‘‘tolerance’’ to vancomycin. ‘‘Tolerance’’ may be defined as a minimum bactericidal concentration (MBC) of R32 times the MIC of an antibiotic. Vancomycin ‘‘tolerance’’ may account for some cases of delayed or blunted therapeutic re- sponse with enterococci or staphylococci [1,83–85]. Vancomycin is a heptapeptide and is bactericidal for most gram-positive organisms, including staphylococci but is bacteriostatic against enterococci. Resistance to vancomycin is mediated by alterations in the permeability of outer membrane porins in S aureus. Vancomycin binds rapidly and irrevers- ibly to cell walls, inhibiting cell-wall synthesis. Interfering with peptidoglycan
  • 9. VANCOMYCIN REVISITED 401 cross-linkages causes defective cell-wall synthesis, resulting in bacterial cell- wall lysis. Enterococci resistance to vancomycin may be of the high- or low- grade variety. High-level (Van A) vancomycin resistance (MIC R64 mg/mL) is mediated by plasmids and is inducible and transferable. Low-level (Van B) resistance (MIC: 32–64 mg/mL) is nontransferable and chromoso- mally encoded [1,85]. Staphylococci Widespread vancomycin use has increased staphylococcal resistance to vancomycin manifested as increased MICs. Clinically, vancomycin resis- tance is manifested as delayed resolution of infection or therapeutic failure. It has long been appreciated that patients with MRSA bacteremia/endocar- ditis often do not respond to vancomycin therapy [86–92]. This cannot be ascribed to underdosing vancomycin. Even with optimal vancomycin dosing, persistent MRSA bacteremias and therapeutic failures are not un- common in clinical practice. Vancomycin therapy increases staphylococcal cell-wall thickening, particularly in hVISA strains. Cell-wall thickening re- sults in ‘‘permeability-mediated’’ resistance to vancomycin as well as to other antistaphylococcal antibiotics. For these reasons, vancomycin may no longer be the preferred antibiotic for treatment of serious systemic MRSA infections. Other effective MRSA antibiotics are available and may be used in place of vancomycin (eg, minocycline, linezolid, tigecycline, and daptomycin). The empiric use of vancomycin to treat MRSA-colonized patients should be minimized to avoid further increases in VRE prevalence and MRSA resistance [1,6,32,93]. Vancomycin: clinical uses Methicillin-sensitive Staphylococcus aureus and methicillin-resistant Staphylococcus aureus bacteremias The finding of S aureus, either of the MSSA or MRSA variety, is common in blood culture reports. Because MSSA and MRSA often colonize the skin, these organisms are frequently introduced into blood cultures during veni- puncture. S epidermidis, (CoNS), are also common blood culture contami- nants. Approximately 20% of blood cultures are contaminated by MSSA, MRSA, or CoNS. Clinicians should not equate-positive blood cultures with bacteremia. Some organisms have clinical significance whenever present in blood cul- tures (ie, Listeria monocytogenes, Brucella sp, Streptococcus pneumoniae). The clinical significance of gram-positive cocci in blood cultures must be as- sessed by the degree of blood culture positivity and considered in the appro- priate clinical context. Ordinarily, four blood cultures should be obtained. This usually allows the clinician to clearly differentiate between blood
  • 10. 402 CUNHA culture positivity and bacteremia when dealing with blood culture reports positive for MSSA, MRSA, or CoNS. One or two out of four positive blood cultures usually represent blood culture contamination rather than infec- tion. High-grade blood culture positivitydthree of four or four of fourd is usually indicative of bacteremia. If it is established that the patient has bacteremia, the clinician’s next task is to determine whether it is a transient or sustained bacteremia. Staphylococci bacteremia is said to be persistent when MSSA or MRSA is present repeatedly in consecutive blood cultures demonstrating high blood culture positivity (ie, three of four or four of four). One out of four or two of four blood cultures with CoNS almost al- ways represents blood culture contamination. Conversely, persistent high- grade blood culture positivity with CoNS, particularly in the presence of implanted prosthetic materials, usually indicates a prosthetic material- or device-associated infection [2,32,94,95]. Much empiric vancomycin is used to ‘‘cover’’ ‘‘gram positive cocci in clus- ters’’ in preliminary blood culture reports rather than bacteremia. The most common cause of high-grade MSSA, MRSA, or CoNS bacteremia are central venous catheter (CVC)–related infections. Empiric vancomycin used to ‘‘cover’’ the staphylococcal bacteremia or the ‘‘catheter’’ should be discour- aged. If CVC line infection is suspected, the line should be removed and the diagnosis confirmed by semiquantitative catheter tip cultures. Treatment for CVC line–associated bacteremia is catheter removal, not just antibiotic ther- apy. Overuse of empiric vancomycin for suspected or known CVC-line infec- tions has increased the prevalence of VRE. If CVC access is necessary after CVC removal for semiquantitative catheter tip culture, another line may be re- placed over a guide wire without risk of pneumothorax, pending catheter tip results. If the semiquantitative catheter tip cultures are later reported as neg- ative, the new catheter placed over a guide wire may remain in place. If catheter tip cultures are positive (ie, R15 colonies of the same organism as causing the bacteremia), then the line replaced over the guide wire should be removed and a new CVC placed in a different anatomic location (Boxes 3 and 4) [96,97]. Continuous Methicillin-sensitive Staphylococcus aureus and methicillin-resistant Staphylococcus aureus bacteremias Patients presenting with a sustained or persistent MSSA or MRSA bac- teremia should be treated empirically with an agent that offers a high degree of activity against MSSA or MRSA, depending upon the hospital’s predom- inant S aureus type (ie, MSSA versus MRSA) while a workup to determine the cause of the persistent bacteremia is undertaken. The most common un- derlying causes of persistent MSSA or MRSA bacteremia are intravascular infections (ie, CVC-related infections) or acute bacterial endocarditis or par- avalvular abscess, and less commonly are due to persistent bacteremias from non-cardiac staphylococcal abscesses and, even less commonly, bone infec- tions [47,97–101].
  • 11. VANCOMYCIN REVISITED 403 Box 3. Vancomycin: appropriate prophylactic and therapeutic uses Prophylaxis Prosthetic (orthopaedic/cardiovascular) implant surgery MSSA/MRSA infections in hemodialysis patients Subacute bacterial endocarditis for: Oral and upper respiratory tract procedures (Streptococcus viridans) (in penicillin allergic patients) Gastrointestinal and genitourinary procedures (E faecalis, VSE) procedures in patients with previous subacute bacterial endocarditis or prosthetic valve endocarditis (plus gentamicin) Therapy MRSA, CoNS CNS shunt (ventriculo-atrial, ventriculo-peritoneal) infectionsa MRSA hemodialysis catheter infectionsb Endocarditisc Streptococus viridans subacute bacterial endocarditis (in penicillin-allergic patients) CoNS prosthetic valve endocarditis due to MSSE or MRSE VSE subacute bacterial endocarditisd a Preferred therapy (eg, linezolid). b Preferred therapy (eg, daptomycin or linezolid). c Alternative therapy preferred. d Combined with gentamicin. Methicillin-sensitive Staphylococcus aureus and methicillin-resistant Staphylococcus aureus central intravenous line infections The majority of intravenous line infections related to CVCs are due to skin organisms (ie, MSSA, MRSA, MSSE, or MRSE). Because vancomycin is active against these skin pathogens, it is frequently used as empiric mono- therapy for presumed CVC line infections. There are two problems with this approach. In institutions where the predominant pathogen causing CVC line infections is MSSA, rather than MRSA, other agents should be used in preference to vancomycin. After MSSA and MRSA, aerobic gram-nega- tive bacilli are the next most important pathogens in CVC line infections. The use of vancomycin empirically for CVC infections has two hospital- wide consequences [96,97,101]. Firstly, vancomycin increases institutional prevalence of VRE. Secondly, exposure to vancomycin has the effect of increasing cell-wall thickness among staphylococci. Vancomycin-induced thickened bacterial cell walls
  • 12. 404 CUNHA Box 4. Clinical situations in which vancomycin should be avoided Prophylaxis General surgical procedures MSSA/MRSA neurosurgical shunts MSSA/MRSA chest tubes MSSA/MRSA intravenous lines MSSA/MRSA in febrile leukopenia Therapy MRSA colonization of respiratory secretions, wounds, or urine (avoid treating colonization; treat only infection) Most serious systemic MRSA infections Preferentially use other anti-MRSA antibiotics Intravenous MRSA antibiotics: linezolid, daptomycin, tigecycline, minocycline Oral MRSA antibiotics: linezolid, minocycline are manifested as an increase in MICs, delayed resolution of infection, or therapeutic failure. Staphylococci with thickened cell walls represent a per- meability barrier to vancomycin as well as to other antibiotics. This is man- ifested clinically in the microbiology laboratory as ‘‘MIC drift.’’ The increase in MICs, often observed during vancomycin therapy, is reflective of cell-wall thickening. A preferred approach to the empiric treatment to CVC line infections pending CVC removal is to use meropenem in institu- tions where CVC infections are more frequently due to MSSA/GNBs than to MRSA. In institutions where the reverse is true, and MRSA is an important CVC pathogen, empiric therapy with tigecycline is preferable to combination therapy with vancomycin plus an anti–GNB antibiotic (Box 5). Methicillin-sensitive Staphylococcus aureus and methicillin-resistant Staphylococcus aureus hemodialysis catheter infections The treatment of hemodialysis catheter infections in hemodialysis pa- tients has been a major area of vancomycin usage because vancomycin is eliminated by glomerular filtration. The increased half-life of vancomycin in anuria may be used to a therapeutic advantage in patients with MSSA/ MRSA infections in end stage renal disease on hemodialysis. As with other foreign body infections due to gram-positive organisms, shunt removal is usually necessary for elimination the infection. Vancomycin has been used prophylactically and therapeutically in hemodialysis patients to treat dialy- sis catheter infections due to MSSA and MRSA [32,102–104].
  • 13. VANCOMYCIN REVISITED 405 Box 5. Diagnostic clinical pathway: persistent S aureus (MSSA, MRSA) bacteremias Differentiate S aureus blood culture positivity (1/4 or 1/2) from bacteremia. With S aureus bacteremia, differentiate low-intensity intermittent bacteremia (1/2 or 2/4 positive blood cultures) from continuous high-intensity bacteremia (3/4–4/4 positive blood cultures). Determine the source of S aureus bacteremia. Abscesses Central intravenous lines Implanted prosthetic devices/materials Acute bacterial endocarditis Soft tissue and bone infections Review antibiotic-related factors. Determine drug dose/dosing interval. Determine MIC/MBCs. Evaluate in vivo versus in vitro effectiveness. Evaluate potential of permeability-related resistance due to vancomycin cell-wall thickening. With high-intensity continuous S aureus bacteremia, obtain a transthoracic or transesophageal echocardiogram to diagnose or rule out acute bacterial endocarditis. Data from Cunha BA. Persistent S aureus bacteremia: clinical pathway for diagnosis treatment. Antibiotics for Clinicians 2006;10(S1):39–46; with permission. Methicillin-sensitive Staphylococcus aureus and methicillin-resistant Staphylococcus aureus acute bacterial endocarditis The diagnostic approach to persistent S aureus bacteremia is relatively straightforward. First, in patients with CVCs, this source of infection should be ruled out first because since CVC infections are readily diagnosable and easily treated by CVC removal. If there is no CVC in place, or if the removed CVC semiquantitative catheter tip cultures are negative, then endocarditis should be the next diagnostic consideration. MSSA or MRSA endocarditis presents as acute bacterial endocarditis (ABE). Except for IVDAs, patients presenting with MSSA or MRSA acute bacterial endo- carditis, usually present with fever of 102 F or higher. In addition, MSSA or MRSA acute bacterial endocarditis usually presents without a heart mur- mur early in the infectious process. Later, when there is valvular destruction, a new or rapidly changing murmur becomes apparent.
  • 14. 406 CUNHA In patients with subacute bacterial endocarditis and positive blood cultures due to subacute bacterial endocarditis pathogens, cardiac echocardiography is indicated in patients who have a heart murmur. Cardiac echocardiography may be the transthoracic (TTE) or the transesophageal (TEE) variety. For screening purposes, TTE is preferred to TEE. For detection of prosthetic valve endocarditis and myocardial abscess TEE is preferred. In patients with subacute bacterial endocarditis, the indication for a TTE or a TEE is the presence of continuous bacteremia due to a known subacute bacterial endocarditis pathogen plus fever and a heart murmur [105–107]. In contrast, in patients with possible acute bacterial endocarditis, the indi- cations for a TTE or TEE are fever and a sustained high-grade MSSA or MRSA bacteremia. A heart murmur is not usually present in acutely in pa- tients presenting with acute bacterial endocarditis. If cardiac echocardiogra- phy is negative for vegetations, acute bacterial endocarditis is effectively eliminated from further consideration. If the high-grade continuous MSSA or MRSA bacteremia continues, the echocardiography should be reviewed for a potential paravalvular myocardial abscess. If a myocardial abscess is sus- pected, TEE, is the preferred echocardiographic modality to demonstrate a perivalvular septal, or myocardial abscess. In patients with potential staph- ylococcal prosthetic valve endocarditis (PVE), a TEE is preferred to demon- strate vegetations. Patients with prosthetic valve(s) should be suspected of having prosthetic valve endocarditis if there is fever and a continuous high- grade bacteremia with a known endocarditis pathogen. Fever is usually pres- ent, but murmurs are difficult to appreciate because of the sounds generated by the prosthetics valve. Subacute bacterial endocarditis, acute bacterial endo- carditis, and prosthetic valve endocarditis may be accompanied by peripheral findings associated with endocarditis (Box 6; Tables 2 and 3) [97,101,106,107]. Staphylococcal central nervous system infections Vancomycin has been used to treat CNS shunt infections, which are usually due to CoNS of the MSSE or MRSE variety. CNS shunt infections usually require shunt removal. If suppression is desired, vancomycin may be given parenterally and, if necessary, CSF concentrations may be further increased by intraventricular administration. Usually the best way to admin- ister intrathecal vancomycin is via an Ommaya reservoir. Vancomycin CSF levels may be obtained in patients to assure adequate CSF levels, but resolu- tion of shunt infections almost always requires shunt removal. It is preferable to use an antibiotic with good CSF penetration (eg, linezolid) [1,31,32]. Febrile neutropenia Recently, some clinicians have added vancomycin to regimens to treat febrile neutropenia in compromised hosts receiving chemotherapy. The increased incidence of gram-positive infections in febrile neutropenia are not due to neutropenia per se, but are due to central line catheters or
  • 15. VANCOMYCIN REVISITED 407 Box 6. Diagnostic clinical pathway: MSSA/MRSA acute bacterial endocarditis Differentiate S aureus blood culture positivity (1/2 or 1/4 positive blood cultures) from bacteremia (3/4–4/4 positive blood cultures). With S aureus bacteremia, differentiate low-intensity intermittent bacteremia (1/2 or 2/4 positive blood cultures) from continuous high-intensity bacteremia (3/4–4/4 positive blood cultures). Acute bacterial endocarditis is not a complication of low- intensity/intermittent S aureus bacteremia. TTE/TEE unnecessary, but will verify no vegetations. If continuous high-grade MSSA/MRSA bacteremia, obtain a TTE or TEE to document cardiac vegetations and/or paravalvular abscess and confirm diagnosis of acute bacterial endocarditis. Diagnostic criteria for MSSA/MRSA acute bacterial endocarditis Essential features Continuous high-grade MSSA/MRSA bacteremia Cardiac vegetations on TTE/TEE No other source Nonessential features Fever ‚102 F (non-IVDAs) Cardiac murmura a With early MSSA/MRSA, a murmur is not present. Later, a new murmur in acute bacterial endocarditis indicates a vegetation or valvular destruction. Data from Cunha BA: MSSA/MRSA acute bacterial endocarditis (ABE): clinical pathway for diagnosis treatment. Antibiotics for Clinicians 2006;10(S1):29–34. chemotherapy infusion devices in these patients. Depending upon the ge- ography/epidemiology of the balance between MSSA versus MRSA strains in patients in the community, empiric treatment with an anti-MSSA or an anti-MRSA agent may be desired until the temporary or semipermanent catheter is removed or replaced. For patients presenting with febrile neutro- penia treated with an antibiotic effective against Pseudomonas aeruginosa with good anti-MSSA activity, additional antistaphylococcal therapy is usually unnecessary. Antibiotics commonly used empirically in febrile neu- tropenia (eg, levofloxacin, meropenem) have anti-MSSA activity. In febrile neutropencia, fungal infections usually present after 10 to 14 days of antibi- otic therapy and are clinically manifested as an otherwise unexplained recru- descence of fever in patients who have responded to empiric anti-P aeruginosa therapy. If fungal infection can be ruled out, a temporary or semipermanent central line infection should be suspected. If blood cultures are persistently positive (high-grade positivity) with MRSA, then anti-MRSA therapy should
  • 16. 408 CUNHA Table 2 Delayed resolution or failure of vancomycin therapy of MSSA or MRSA bacteremia and acute bacterial endocarditis Failure rates Duration of bacteremia References MSSA bacteremia Nafcillin: 4% Nafcillin: 2 days Sande [110] Vancomycin: 20% Vancomycin: 7 days (20% O3 days) (12% O7 days) MSSA acute bacterial Nafcillin: 1.4%–26% Nafcillin: 2 days Gentry [111] endocarditis Vancomycin: 37%–50% Vancomycin: 5 days Geraci [112] Esposito [4] Chang [98] MRSA acute bacterial Nafcillin: Not applicable Small [115] endocarditis Vancomycin: O7 days be given until the device is removed. The routine use of vancomycin in com- bination therapy with an anti-P aeruginosa antibiotic for febrile neutropenia should be discouraged to minimize further increases in VRE prevalence and permeability-mediated resistance [1,32,108]. Penicillin-allergic patients Vancomycin has been used in penicillin-allergic patients to treat Strepto- coccus viridans and enterococcal endocarditis. Vancomycin monotherapy has been used successfully over the years to treat S viridans subacute bacte- rial endocarditis. Because vancomycin is bacteristatic against E faecalis, combination therapy with aminoglycoside (eg, gentamicin) has been used to treat VSE endocarditis. Strains demonstrating high-level gentamicin resis- tance may be treated with vancomycin plus ceftriaxone or, alternatively, another antibiotic with bactericidal antienterococcal activity may be used (eg, daptomycin) (see Box 4) [32,109]. Vancomycin: therapeutic alternatives The empiric therapeutic approach to central intravenous line–related infections has been discussed. Empiric therapy during the workup for Table 3 Combination therapy for MSSA and MRSA acute bacterial endocarditis Antibiotic combinations Comments References MSSA acute bacterial endocarditis Nafcillin þ gentamicin Outcomes same without gentamicin Cha [116] Vancomycin þ gentamicin Lee [117] MRSA acute bacterial endocarditis Vancomycin Duration of bacteremia: 7 days Levine [118] Vancomycin þ rifampin Duration of bacteremia: 9 days Shelburne [119] (antagonistic; not synergistic)
  • 17. VANCOMYCIN REVISITED 409 ABE should consist of antistaphylococcal antibiotic that has a high degree of activity against the presumed pathogen, does not cause resistance, does not cause the emergence of other organism (eg, VRE), and has a good safety profile. In the treatment of MSSA bacteremias, vancomycin has been shown to be inferior to vancomycin in terms of delayed resolution of bacteremia. If MSSA is the pathogen in bacteremia or endocarditis, vancomycin should not be used as therapy in these patients and preferably another agent should be used. In penicillin-allergic patients, vancomycin is often used empirically to treat MSSA or MRSA bacteremias. However, there are many alternative antibiotics available that do not cross-react with penicillins or cephalospo- rins and that are effective to treat MSSA or MRSA bacteremias. The treatment of MSSA or MRSA bacteremia is optimal with a single agent. Clinicians often assume there is benefit in adding a second drug for enhanced antistaphylococcal activity. It has been shown that vancomycin plus gentamicin offers no benefit in the treatment of MSSA bacteremias. Another common misconception is that the addition of rifampin or other drugs to vancomycin will enhance the antistaphylococcal activity of the pri- mary anti-MSSA/MRSA agent. It has been shown the rifampin is likely to be antagonistic, not synergistic, against S aureus [110]. While rifampin has a high degree of in vitro activity against S aureus, rifampin monotherapy should not be used because of resistance. The practice of adding an amino- glycoside or rifampin to antistaphylococcal antibiotics has no benefit and may have a negative effect on outcomes (see Tables 2 and 3) [1,3– 5,31,32,110]. In penicillin-allergic patients, the treatment of serious systemic infections due to MSSA (ie, CVC line infections, acute bacterial endocarditis, pros- thetic valve endocarditis, and so on), may be treated with antibiotics other than vancomycin (ie, meropenem, TMP-SMX, daptomycin). As with MSSA, there are numerous therapeutic alternatives to vancomycin for the treatment of serious MRSA infections. Useful antibiotics to treat all MRSA types (HA-MRSA, CO-MRSA, CA-MRSA) include daptomycin linezolid, tigecycline, and minocycline [1,32]. If possible, vancomycin should be avoided in therapy of most serious sys- temic infections due to MSSA/MRSA for two principal reasons. Prolonged vancomycin exposure selects out heteroresistant strains (hVISA). These strains are the ones that have thickened cell walls as the result of vancomycin exposure. Thickened cell walls are manifested as an increase in MICs and decreased cell-wall permeability to vancomycin as well as to other antibiotics. The other untoward side effect of vancomycin use is in- creased VRE prevalence [37–43]. In conclusion, vancomycin should be used selectively for the parenteral treatment of serious systemic infections due to MSSA, MRSA, or CoNS. Available therapeutic alternatives do not increase the prevalence of VRE and do not result in cell-wall thickening and permeability-mediated resistance. Alternative agents more quickly resolve MSSA/MRSA
  • 18. 410 CUNHA bacteremias than vancomycin. The addition of gentamicin or rifampin to vancomycin does not enhance antistaphylococcal activity and may have a negative effect (ie, antagonism) [47,98–100,111–117]. Vancomycin: reassessment of use The main role of vancomycin has been in the therapy of serious systemic MRSA infections [1,118]. The widespread use of vancomycin over the years has resulted in increased prevalence of VRE in institutions worldwide. Re- cently, it has been shown that vancomycin induces cell-wall thickening among staphylococci, resulting in ‘‘permeability-mediated’’ resistance. To avoid these problems associated with vancomycin, it is preferable to use other anti-MRSA antibiotics in place of vancomycin. Fortunately, several other anti-MRSA agents are available. Antibiotics with excellent anti-MRSA activ- ity include minocycline, linezolid, daptomycin, and tigecycline. Although vancomycin is available as an oral formulation for C difficile infections, it is ineffective to treat systemic infections. For the parenteral treatment of serious systemic MRSA infections, minocycline, daptomycin, and linezolid are effec- tive alternative anti-MRSA antibiotics. For the oral therapy of MRSA, min- ocycline or linezolid may be used (Boxes 7 and 8) [1,32,119,120]. Box 7. Vancomycin: use in serious systemic infection Clinical advantages Extensive clinical experience Not nephrotoxic Clinical disadvantages Adverse side effects Red neck (red man) syndrome Thrombocytopenia Leukopenia Sudden death Increased VRE prevalence Related to volume of intravenous (not oral) use S. aureus infections: delayed resolution, therapeutic failures MSSA/MRSA bacteremias MSSA/MRSA acute bacterial endocarditis S. aureus infections: permeability-mediated resistance (increased MICs) Cell-wall thickening; decreased S. aureus penetration of antibiotics (vancomycin and other anti-S aureus antibiotics) Not inexpensive
  • 19. VANCOMYCIN REVISITED 411 Box 8. Suboptimal uses of vancomycin Empiric treatment of central intravenous line infections In locations where MSSA/GNBs MRSA CVC pathogens Remove intravenous line as soon as possible Therapy of MRSA bacteremia/acute bacterial endocarditis Delayed clinical responses Therapeutic failures Intraperitoneal therapy of S aureus/CoNS CAPD peritonitis Intravenous therapy achieves therapeutic concentration in ascitic fluid. Requires another antibiotic for anti–gram-negative bacillary coverage. Monotherapy of VSE Bacteriostatic if monotherapy used. Adequate anti-VSE activity only if given with an aminoglycoside (eg, gentamicin) S aureus chronic osteomyelitis Prevention of S pneumoniae CSF ‘‘seeding’’ (2 to CAP pneumococcal bacteremia) Unnecessary when using antibiotics that achieve therapeutic CSF concentrations (eg, ceftriaxone) Data from Cunha BA, editor. Infections in critical care medicine. 2nd edition. New York: Informa Health Care USA, Inc.; 2007. For the therapy of gram-positive CNS infections, other agents are phar- macokinetically preferable to vancomycin. Linezolid, available orally and intravenously, achieves high CSF levels when given in the usual dose (ie, 600 mg intravenously or orally every 12 hours). Minocycline using the usual dose (100 mg intravenously or orally every 12 hours) also achieves therapeu- tic CSF levels. No data are available regarding treatment of CNS infections with or tigecycline [1,32]. In access catheter graft infections in chronic hemodialysis patients, dap- tomycin, linezolid, or tigecycline may be used. Vancomycin can be used to treat S viridans subacute bacterial endocarditis in penicillin-allergic patients, but other agents are preferable to vancomycin for this use. Vancomycin monotherapy is ‘‘bacteristatic’’ against VSE. Therefore, for VSE subacute bacterial endocarditis, an aminoglycoside should be added to vancomycin to achieve bactericidal anti-VSE activity alternate therapy for VSE subacute bacterial endocarditis (eg, meropenem, linezolid, or daptomycin) is available [3,32,119,120]. Aside from increased prevalence of VRE and increasing resistance among staphylococci, there are other reasons to avoid or minimize vancomycin use. Because vancomycin is an older drug, it is often thought of as an inexpensive
  • 20. 412 Table 4 Vancomycin: clinical use and therapeutic alternatives Clinical uses Disadvantages, problems Therapeutic alternatives Advantages of alternative antibiotics Monotherapy Central IV catheter (CVC) infections Increased VRE prevalence Removal of CVC High-degree MRSA activity ‘‘that may be due to MRSA’’ Increased permeability-mediated Daptomycin (6 mg/kg IV Also covers MSSA, VSE resistance every 24 hours) No increased resistance/VRE MIC ‘‘drift’’ Linezolid (600 mg IV every 12 hours) MRSA bacteremias, acute bacterial Increased permeability-mediated Daptomycin (6–12 mg/kg Most rapidly bactericidal MRSA endocarditis resistance IV every 24 hours) antibiotic Delayed therapeutic response Effective in vancomycin CUNHA (persistent bacteremia) ‘‘treatment failures’’ MIC ‘‘drift’’ Linezolid (600 mg Also available orally IV every 12 hours) for prolonged therapy No increased resistance/VRE MRSA osteomyelitis Vancomycin: often delayed Linezolid (600 mg Also available orally response with 30 mg/kg/d; better IV/orally every 12 hours) for prolonged therapy results with 60 mg/kg/d Minocycline (100 mgIV/ No increased resistance/VRE Increased permeability-mediated orally every 12 hours) Inexpensive and also available resistance orally for prolonged therapy. Increased MICs during therapy MIC ‘‘drift’’ MRSA nosocomial and Essential to avoid ‘‘covering’’ Linezolid (600 mg IV Effective in rare cases of bona ventilator associated pneumonia MSSA/MRSA respiratory every 12 hours) fide MRSA nosocomial (rare) secretion colonization pneumonia and ventilator- Increases permeability-mediated associated pneumonia resistance Therapeutic parenchymal lung MIC ‘‘drift’’ concentrations
  • 21. Combination Therapy Vancomycin and ceftriaxone ‘‘Double drug’’ therapy Ceftriaxone (1–2 g IV every Good CSF penetration prevents to prevent ‘‘seeding’’ of CSF unnecessary. Vancomycin: 12 hours) PRSP ‘‘seeding’’ of CSF from bacteremic S pneumoniae relatively low CSF concentrations Ceftriaxone monotherapy provides (PRSP) CAP adequate CSF concentrations if PRSP seeding of CSF Vancomycin plus gentamicin therapy Vancomycin alone ‘‘bacteriostatic’’ Daptomycin (6 mg/kg IV MICs for VSE/VRE greater than of E faecalis (VSE) subacute bacterial against VSE every 24 hours) those for MSSA/MRSA endocarditis Vancomycin bactericidal only Use 12 mg/kg IV every 24 hours when combined with an for VSE endocarditis aminoglycoside (eg, gentamicin) Linezolid (600 mg IV Available orally for prolonged every 12 hours) therapy of VSE subacute bacterial endocarditis VANCOMYCIN REVISITED Bacteriostatic but effective in subacute bacterial endocarditis Abbreviations: IV, intravenous; PSRP, penicillin-resistant S pneumoniae. 413
  • 22. 414 CUNHA antibiotic. However, because vancomycin must be administered intrave- nously, the cost of intravenous administration and monitoring must be added to the actual cost of vancomycin use. If serum vancomycin levels are used to guide therapy, the cost of such levels plus the cost of serial serum creatinine levels must also be included in the actual cost of administering parenteral vancomycin. For patients requiring long-term parenteral therapy with vancomycin, peripherally inserted central catheters (PICCs) are often used. The use of a PICC line to administer vancomycin increases antibiotic cost because other charges related to surgical insertion, radiological verifica- tion of PICC line placement, and the cost of home intravenous agencies must be added to vancomycin cost. While the cost of administering paren- teral antibiotics via PICC lines for vancomycin is similar to that for other antibiotics, most infections for which vancomycin is administered parenter- ally may be treated equally effectively with oral linezolid which is much less expensive (see Boxes 4 and 5, Table 4). Summary The reassessment of vancomycin use today is based on an evaluation of the advantages and disadvantages of vancomycin alternatives for the ther- apy of MRSA. Other antibiotics exist as alternative therapy for VSE infec- tions as well as for preventing the ‘‘seeding’’ of the CNS by penicillin- resistant S pneumoniae from bacteremic pneumococcal CAP. Clinicians should appreciate the limited role of vancomycin today in MRSA therapy. Clinicians should also realize vancomycin has limited activity against MSSA and VSE. Other antibiotics are preferable against MSSA and VSE. Pres- ently, clinicians should consider alternatives to vancomycin because of the negative effects of vancomycin therapy (ie, increased VRE prevalence and increasing resistance among staphylococci), and also because better thera- peutic alternatives are available with little or no resistance potential, no in- crease in VRE prevalence, better pharmacokinetics and lower cost (eg, linezolid) (see Box 6). For the reasons mentioned, clinicians should use van- comycin sparingly and consider other antibiotic alternatives for MSSA, MRSA, CoNS, and VSE infections [32,119,120]. References [1] Kucers A, Crowe SM, Grayson ML, et al. Vancomycin: the use of antibiotics: a clinical review of antibacterial, antifungal and antiviral drugs. 5th edition. Oxford (United Kingdom): Butterworth-Heinemann; 1997. p. 763–90. [2] Louria DB, Kaminski T, Buchman J. Vancomycin in severe staphylococcal infections. Arch Intern Med 1961;107:225–40. [3] Cunha BA. Vancomycin. Med Clin North Am 1995;79:817–31. [4] Esposito AL, Gleckman RA. Vancomcyin: a second look. JAMA 1977;238:1756–7. [5] Menzies D, Goel K, Cunha BA. Vancomycin. Antibiotics for Clinicians 1998;2:97–9.
  • 23. VANCOMYCIN REVISITED 415 [6] Farber BF, Moellering RC Jr. Retrospective study of the toxicity of preparations of vancomycin from 1974 to 1981. Antimicrob Agents Chemother 1983;23:138–41. [7] Elting LS, Rubenstein EB, Kurtin D, et al. Mississippi and mud in the 1990s: risks and outcomes of vancomycin-associated toxicity in general oncology practice. Cancer 1998; 83:2597–607. [8] Appel GB, Given DB, Levine LR, et al. Vancomycin and the kidney. Am J Kidney Dis 1986;8:75–80. [9] Mellor JA, Kingdom J, Cafferkey M, et al. Vancomycin toxicity: a prospective study. J Antimicrob Chemother 1985;15:773–80. [10] Nahata MC. Lack of nephrotoxicity in pediatric patients receiving concurrent vancomycin and aminoglycoside therapy. Chemotherapy 1987;33:302–4. [11] Goren MP, Baker DK Jr, Shenep JL. Vancomycin does not enhance amikacin-induced tubular nephrotoxicity in children. Pediatr Infect Dis J 1989;8:278–82. [12] Goetz MB, Sayers J. Nephrotoxicity of vancomycin and aminoglycoside therapy separately and in combination. J Antimicrob Chemother 1993;32:325–34. [13] Ergur T, Onarlioglu B, Gunay Y, et al. Does vancomycin increase aminoglycoside nephro- toxicity? Acta Paediatr Jpn 1997;39:422–7. [14] Pauly DJ, Musa DM, Lestico MR, et al. Risk of nephrotoxicity with combination vancomycin-aminoglycoside antibiotic therapy. Pharmacotherapy 1990;10:378–82. [15] Rybak MJ, Albrecht LM, Boike SC, et al. Nephrotoxicity of vancomycin alone and with an aminoglycoside. J Antimicrob Chemother 1990;25:679–87. [16] Ngeleka M, Beauchamp D, Tardif D, et al. Endotoxin increases the nephrotoxic potential of gentamicin and vancomycin plus gentamicin. J Infect Dis 1990;161:721–7. [17] Rybak MJ, Boike SC. Monitoring vancomycin therapy. Drug Intell Clin Pharm 1986;20: 757–61. [18] Edwards DJ, Pancorbo S. Routine monitoring of serum vancomycin concentrations: waiting for proof of its value. Clin Pharm 1987;6:652–4. [19] Moellering RC Jr. Pharmacokinetics of vancomycin. J Antimicrob Chemother 1984;14:43–52. [20] Matzke GR, Zhanel GG, Guay DR. Clinical pharmacokinetics of vancomycin. Clin Pharmacokinet 1986;11:257–82. [21] Moellering RC Jr, Krogstad DJ, Greenblatt DJ. Vancomycin therapy in patients with impaired renal function: a nomogram for dosage. Ann Intern Med 1981;94:343–6. [22] Leader WG, Chandler MH, Castiglia M. Pharmacokinetic optimization of vancomycin therapy. Clin Pharmacokinet 1995;28:327–42. [23] Boffi el Amari E, Vuagnat A, Stern R, et al. High versus standard dose vancomycin for osteomyelitis. Scand J Infect Dis 2004;36:712–7. [24] Hidayat LK, Hsu DI, Quist R, et al. High dose vancomycin therapy for methicillin-resistant Staphylococcus aureus infections. Arch Intern Med 2006;166:2138–44. [25] Von Drygalski A, Curtis BR, Bougie DW, et al. Vancomycin-induced immune thrombocy- topenia. N Engl J Med 2007;356:904–10. [26] Carmeli Y, Samore MH, Huskins C. The association between antecedent vancomycin treatment and hospital-acquired vancomycin-resistant enterococci: a meta-analysis. Arch Intern Med 1999;159:2461–8. [27] Fishbane S, Cunha BA, Shea KW, et al. Vancomycin-resistant enterococcus (VRE) in hemodialysis patients. Am J Infect Control 1999;20:461–2. [28] Fridkin SK, Lawton R, Edwards JR, et al. Monitoring antimicrobial use and resistance: comparison with a national benchmark on reducing vancomycin use and vancomycin- resistant enterococci. Emerg Infect Dis 2002;8:702–7. [29] Fridkin SK, Edwards JR, Courval JM, et al. The effect of vancomycin and third-generation cephalosporins on prevalence of vancomycin-resistant enterococci in 126 U.S. adult intensive care units. Ann Intern Med 2001;135:175–83. [30] Kolar M, Urbanek K, Vagnerova I, et al. The influence of antibiotic use on the occurrence of vancomycin-resistant enterococci. J Clin Pharm Ther 2006;31:67–72.
  • 24. 416 CUNHA [31] Cunha BA. Antimicrobial therapy of multi-drug resistant S pneumoniae VRE MRSA. Med Clin North Am 2006;90:1165–82. [32] Cunha BA. Antibiotic essentials. 7th edition. Royal Oak (MI): Physicians Press; 2008. [33] Tenover F, Biddle J, Lancaster M. Increasing resistance to vancomycin and other glycopep- tides in Staphylococcus aureus. Emerg Infect Dis 2001;7:327–32. [34] Liu C, Chambers HF. Staphylococcus aureus with heterogeneous resistance to vancomycin: epidemiology, clinical significance, and critical assessment of diagnostic methods. Antimi- crob Agents Chemother 2003;47:3040–5. [35] Cosgrove SE, Carroll KC, Perl TM. Staphylococcus aureus with reduced susceptibility to vancomycin. Clin Infect Dis 2004;39:539–45. [36] Fridkin SK, Hageman J, McDougal LK, et al. Epidemiological and microbiological characterization of infections caused by Staphylococcus aureus with reduced susceptibility to vancomycin, United States, 1997–2001. Clin Infect Dis 2003;36:429–39. [37] Wang G, Hindler JF, Ward KW, et al. Increased vancomycin MICs for Staphylococcus aureus. Clinical isolates from a university hospital during a 5-year period. J Clin Microbiol 2006;44:3883–6. [38] Cui L, Ma K, Sato K, et al. Cell wall thickening is a common feature of vancomycin resistance in Staphylococcus aureus. J Clin Microbiol 2003;41:5–14. [39] Cui L, Tominaga E, Hiramatsu K. Correlation between reduced daptomycin susceptibility and vancomycin resistance in vancomycin-intermediate Staphylococcus aureus. Antimicrob Agents Chemother 2006;50:1079–82. [40] Bell JM, Walters LJ, Turnidge JD, et al. Vancomycin hetero-resistance has a small but significant effect on the daptomycin minimum inhibitory concentration of Staphylococcus aureus [abstract D-814]. Paper presented at the 46th ICAAC. San Francisco (CA), Septem- ber 27–30, 2006. [41] Sakoulas G, Alder J, Thauvin-Eliopoulos C, et al. Induction of daptomycin heterogeneous susceptibility in Staphylococcus aureus by exposure to vancomycin. Antimicrob Agents Chemother 2006;50:1581–5. [42] Patel JB, Jevitt LA, Hageman J, et al. An association between a reduced susceptibility to daptomycin and reduced susceptibility to vancomycin in Staphylococcus aureus. Clin Infect Dis 2006;42(11):1652–3. [43] Howden BP, Ward PB, Charles PGP, et al. Treatment outcomes for serious infections caused by methicillin-resistant Staphylococcus aureus with reduced vancomycin susceptibil- ity. Clin Infect Dis 2004;38:521–8. [44] Charles PG, Ward PB, Johnson PD, et al. Clinical features associated with bacteremia due to heterogeneous vancomycin-intermediate Staphylococcus aureus. Clin Infect Dis 2004;38: 448–51. [45] 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;47:1262–6. [46] Howden BP, Johnson PD, Ward PB, et al. Isolates with low-level vancomycin resistance associated with persistent methicillin-resistant Staphylococcus aureus bacteremia. Antimicrob Agents Chemother 2006;50:3049–57. [47] Wong SS, Ng TK, Yam WC, et al. Bacteremia due to Staphylococcus aureus with reduced susceptibility to vancomycin. Diagn Microbiol Infect Dis 2000;36:261–8. [48] Fridkin SK. Vancomycin-intermediate and -resistant Staphylococcus aureus: what the infectious disease specialist needs to know. Clin Infect Dis 2001;32:108–15. [49] Bouza E, Burillo A, Munoz P. Antimicrobial therapy of Clostridium difficile–associated diarrhea. Med Clin North Am 2006;90:1141–63. [50] Cunha BA, Klein NC. Vancomycin. In: Yoshikawa T, editor. Antibiotics in the elderly. New York: Marcel Dekker; 1994. p. 311–22.
  • 25. VANCOMYCIN REVISITED 417 [51] Gump DW. Vancomycin for treatment of bacterial meningitis. Rev Infect Dis 1981;3: 289–92. [52] Nagl M, Neher C, Hager J, et al. Bactericidal activity of vancomycin in cerebrospinal fluid. Antimicrob Agents Chemother 1999;43:1932–4. [53] Skhirtladze K, Hutschala H, Fleck T, et al. Impaired target site penetration of vancomycin in diabetic patients following cardiac surgery. Antimicrob Agents Chemother 2006;50: 1372–5. [54] Massias L, Dubois C, de Lentdecker P, et al. Penetration of vancomycin in uninfected sternal bone. Antimicrob Agents Chemother 1992;36:2539–41. [55] Ackerman BH, Vannier AM. Necessity of a loading dose when using vancomycin in critically ill patients. J Antimicrob Chemother 1992;29:460–1. [56] Cunha BA, Deglin J, Chow M, et al. Pharmacokinetics of vancomycin in patients undergo- ing chronic hemodialysis. Rev Infect Dis 1981;3:269–72. [57] DelDot ME, Lipman J, Tett SE. Vancomycin pharmacokinetics in critically ill patients receiving continuous venovenous haemodiafiltration. Br J Clin Pharmacol 2004;58: 259–68. [58] Lutsar I, McCracken GH Jr, Friedland IR. Antibiotic pharmacodynamics in cerebrospinal fluid. Clin Infect Dis 1998;27:1117–27. [59] Byl B, Jacobs F, Wallemacq P, et al. Vancomycin penetration of uninfected pleural fluid exudate after continuous or intermittent infusion. Antimicrob Agents Chemother 2003; 47:2015–7. [60] Cruciani M, Gatti G, Lazzarini L, et al. Penetration of vancomycin into human lung tissue. J Antimicrob Chemother 1996;38:865–9. [61] Cruciani M, Gatti G, Lazzarini L, et al. Penetration of vancomycin in severe staphylococcal infections: prospective multicenter randomized study. Antimicrob Agents Chemother 1996;38:865–9. [62] Rybak MJ. The pharmacokinetic and pharmacodynamic properties of vancomycin. Clin Infect Dis 1994;18:544–6. [63] Zimmermann AE, Katona BG, Plaisance KI. Association of vancomycin serum concentra- tions with outcomes in patients with gram-positive bacteremia. Pharmacotherapy 1995;15: 85–91. [64] James JK, Palmer SM, Levine DR, et al. Comparison of conventional dosing versus continuous-infusion vancomycin therapy for patients with suspected or documented gram-positive infections. Antimicrob Agents Chemother 1996;40:696–700. [65] Kralovicov K, Spanik S, Halko J, et al. Do vancomycin serum levels predict failures of vancomycin therapy or nephrotoxicity in cancer patients? J Chemother 1997;9:420–6. [66] Masur H, Francioli P, Ruddy M, et al. Vancomycin serum levels and toxicity in chronic haemodialysis patients with Staphylococcus aureus bacteremia. Clin Nephrol 1983;20:85–8. [67] Andres I, Lopez R, Pou L, et al. Vancomycin monitoring: one or two serum levels? Ther Drug Monit 1997;19:614–9. [68] Sayers JF, Shimasaki R. Routine monitoring of serum vancomycin concentrations: The answer lies in the middle. Clin Pharm 1988;7:18. [69] Moellering RC Jr. Monitoring serum vancomycin levels: climbing the mountain because it is there? Clin Infect Dis 1994;18:544–6. [70] Rodvold KA, Zokufa H, Rotschafer JC. Routine monitoring of serum vancomycin concen- trations: Can waiting be justified? Clin Pharm 1987;6:655–8. [71] Freeman CD, Quintiliani R, Nightingale CH. Vancomycin therapeutic drug monitoring: Is it necessary. Ann Pharmacother 1993;27:594–8. [72] Saunders NJ. Vancomycin administration and monitoring reappraisal. J Antimicrob Chemother 1995;36:279–82. [73] Karam CM, McKinnon PS, Neuhauser MM, et al. Outcome assessment of minimizing vancomycin monitoring and dosing adjustments. Pharmacotherapy 1999;19:257–66.
  • 26. 418 CUNHA [74] Darko W, Medicis JJ, Smith A, et al. Mississippi mud no more: cost-effectiveness of pharmacokinetic dosage adjustment of vancomycin to prevent nephrotoxicity. Pharmaco- therapy 2003;23:643–50. [75] Cantu TG, Yamanaka-Yuen NA, Lietman PS. Serum vancomycin concentrations: reappraisal of their clinical value. Clin Infect Dis 1994;18:533–43. [76] Cunha BA. Vancomycin serum levels: unnecessary, unhelpful, and costly. Antibiotics for Clinicians 2004;8:273–7. [77] Cunha BA, Mohan SS, Hamid N, et al. Cost ineffectiveness of serum vancomycin levels. Eur J Clin Microbiol Infect Dis 2007;13:509–11. [78] Allegaert K, van den Anker JN. Predictability of vancomycin pharmacokinetics in neonates. Eur J Clin Microbiol Infect Dis 2007;26:847–8. [79] Cunha BA. Clinical manifestations and antimicrobial therapy of methicillin resistant Staphylococcus aureus (MRSA). Clin Microbiol Infect 2005;11:33–42. [80] Alder JD. Staphylococcus aureus: antibiotic resistance and antibiotic selection. Antibiotics for Clinicians 2006;10(S1):19–24. [81] Krol V, Cunha BA, Schoch PE, et al. Empiric gentamicin and vancomycin therapy for bacteremias in chronic dialysis outpatient units in the era of antibiotic resistance. J Chemo- ther 2006;18:490–3. [82] Quale J, Landman D, Saurina G, et al. Manipulation of hospital antimicrobial formulary to control an outbreak of vancomycin-resistant enterococci. Clin Infect Dis 1996;23:1020–5. [83] Saribas S, Bagdatli Y. Vancomycin tolerance in enterococci. Chemotherapy 2004;50: 250–4. [84] Watanakunakorn C. Antibiotic-tolerant Staphylococcus aureus. J Antimicrob Chemother 1978;4:561–8. [85] Cunha BA, Mickail N, Eisenstein L. E faecalis vancomycin sensitive enterococci (VSE) bacteremia unresponsive to vancomycin successfully treated with high dose daptomycin. Heart Lung 2007;36:456–61. [86] Hussain FM, Boyle-Vavra S, Shete PB, et al. Evidence for a continuum of decreased vancomycin susceptibility in unselected Staphylococcus aureus clinical isolates. J Infect Dis 2002;186:661–7. [87] Sakoulas G, Moise-Broder PA, Schentag JJ, et al. Relationship of MIC and bactericidal activity to efficacy of vancomycin for treatment of methicillin-resistant Staphylococcus aureus bacteremia. J Clin Microbiol 2004;42:2398–402. [88] Schwaber MJ, Wright SB, Carmeli Y, et al. Clinical implications of varying degrees of vancomycin susceptibility in methicillin-resistant Staphylococcus aureus bacteremia. Emerg Infect Dis 2003;9:657–64. [89] Srinivasan A, Dick JD, Perl TM. Vancomcyin resistance in staphylococci. Clin Microbiol Rev 2002;15:430–8. [90] Walsh TR, Howe RA. The prevalence and mechanisms of vancomycin resistance in Staphylococcus aureus. Annu Rev Microbiol 2002;56:657–75. [91] Wooton M, Walsh TR, Macgowan AP. Evidence for reduction in breakpoints used to determine vancomycin susceptibility in Staphylococcus aureus. Antimicrob Agents Cheomther 2005;49:3982–3. [92] Ward PB, Johnson PD, Grabsch EA, et al. Treatment failure due to methicillin-resistant Staphylococcus aureus (MRSA) with reduced susceptibility to vancomycin. Med J Aust 2001;175:480–3. [93] Wooten M, MacGowan AP, Walsh TR. Comparative bactericidal activities of daptomycin and vancomycin against glycopeptide-intermediate Staphylococcus aureus (GISA) and Heterogenous GISA isolates. Antimicrob Agents Chemother 2006;50:4195–7. [94] Burillo A, Bouza E. Epidemiology of MSSA and MRSA: colonization and infection. Antibiotics for Clinicians 2006;10(S1):3–10. [95] Kim SH, Park WB, Lee KD, et al. Outcome of Staphylococcus aureus bacteremia in patients with eradicable foci versus noneradicable foci. Clin Infect Dis 2003;37:794–9.
  • 27. VANCOMYCIN REVISITED 419 [96] Evans M, Finch R. Approach to staphylococcal central IV line infections and bacteremias. Antibiotics for Clinicians 2006;10(S1):25–8. [97] Cunha BA. Persistent S aureus bacteremia: clinical pathway for diagnosis treatment. Antibiotics for Clinicians 2006;10(S1):39–46. [98] Chang FY, Peacock JE Jr, Musher DM, et al. Staphylococcus aureus bacteremia: recurrence and the impact of antibiotic treatment in a prospective multicenter study. Medicine (Baltimore) 2003;82:333–9. [99] Lodise TP, McKinnon PS, Swiderski L, et al. Outcomes analysis of delayed antibiotic treatment for hospital-acquired Staphylococcus aureus bacteremia. Clin Infect Dis 2003;36:1418–23. [100] Rotun SS, Mcmath V, Schoonmaker DJ, et al. Staphylococcus aureus with reduced suscep- tibility to vancomycin isolated from a patient with fatal bacteremia. Emerg Infect Dis 1999; 5:147–9. [101] Cunha BA. IV line infections. In: Cunha BA, editor. Infectious diseases in critical care medicine. 2nd edition. New York: Informa Healthcare; 2007. [102] Morris AJ, Bilinsky RT. Prevention of staphylococcal shunt infections by continuous vancomycin prophylaxis. Am J Med Sci 1971;262:87–92. [103] Eykyn S, Phillips I, Evans J. Vancomycin for staphylococcal shunt site infections in patients on regular haemodialysis. Br Med J 1970;3:8–82. [104] Tofte RW, Solliday J, Rotschafer J, et al. Staphylococcus aureus infection of dialysis shunt: absence of synergy with vancomycin and rifampin. South Med J 1981;74:612–6. [105] Cunha BA, Gill MV, Lazar J. Acute infective endocarditis. Infect Dis Clin North Am 1996; 10:811–34. [106] Cunha BA. MSSA/MRSA acute bacterial endocarditis (ABE): clinical pathway for diagnosis treatment. Antibiotics for Clinicians 2006;10(S1):29–34. [107] Brusch JL. Infective endocarditis. New York: Informa Healthcare; 2007. p. 143–272. [108] Picazo JJ. Association for Health Research and Development (ACINDES). Management of the febrile neutropenic patient. Int J Antimicrob Agents 2005;2(S1):S120–2. [109] Cunha BA. Clinical approach to antibiotic therapy in the penicillin allergic patient. Med Clin North Am 2006;90:1257–64. [110] Bliziotis IA, Ntziora F, Lawrence KR, et al. Rifampin as adjuvant treatment of gram- positive bacterial infections: a systemic review of comparative clinical trials. Eur J Clin Microbiol Infect Dis 2007;26:849–56. [111] Gentry CA, Rodvold KA, Novack RM, et al. Retrospective evaluation of therapies for Staphylococcus aureus endocarditis. Pharmacotherapy 1977;17:990–7. [112] Geraci JE, Wilson WR. Vancomycin therapy for infective endocarditis. Rev Infect Dis 1981;3(Suppl):S520–58. [113] Small PM, Chambers HF. Vancomycin for Staphylococcal aureus endocarditis in intrave- nous drug users. Antimicrob Agents Chemother 1990;34:1227–31. [114] Lee DG, Chun HS, Yim DS, et al. Efficacies of vancomycin, arbekacin, and gentamicin alone or in combination against methicillin-resistant Staphylococcus aureus in an in vitro infective endocarditis model. Antimicrob Agents Chemother 2003;47:3768–73. [115] Levine DP, Fromm BS, Reddy BR. Slow response to vancomycin or vancomycin plus rifampin in methicillin-resistant Staphylococcus aureus endocarditis. Ann Intern Med 1991;115:674–80. [116] Shelburne SA, Musher DM, Hulten K, et al. In vitro killing of community-associated methicillin-resistant Staphylococcus aureus with drug combinations. Antimicrob Agents Chemother 2004;48:4016–9. [117] Cha R, Brown WJ, Ryback MJ. Bactericidal activities of daptomycin, quinupristin- dalfopristin, and linezolid against vancomycin-resistant Staphylococcus aureus in an in vitro pharmacodynamic model with simulated endocardial vegetations. Antimicrob Agents Chemother 2003;47:3960–3. [118] Kirst HA, Thompson DG, Nicas TI. Historical yearly usage of vancomycin. Antimicrob Agents Chemother 1998;42:1303–4.
  • 28. 420 CUNHA [119] Stevens DL. The role of vancomycin in the treatment paradigm. Clin Infect Dis 2006;42: S51–7. [120] Pope SD, Roecker A. Vancomycin for treatment of invasive, multi-drug resistant Staphylococcus aureus infections. Expert Opin Pharmacother 2007;8:1245–61.