SlideShare uma empresa Scribd logo
1 de 14
Baixar para ler offline
Optimizing Therapy for Vancomycin-
      Resistant Enterococci (VRE)
      Peter K. Linden, M.D.1




      ABSTRACT

                                                Enterococci are gram-positive, facultative bacteria with low intrinsic virulence but
                                      capable of causing a diverse variety of infections such as bacteremia with or without
                                      endocarditis, and intra-abdominal, wound, and genitourinary infection. During the past
                                      2 decades the incidence of hospital-acquired enterococcal infection has significantly risen
                                      and is increasingly due to multidrug-resistant strains, primarily to the coacquisition
                                      of genetic determinants that encode for the stable expression of high-level b-lactam,
                                      aminoglycoside, and glycopeptide resistance.
                                                Because enterococci constitute part of the normal colonizing flora, careful clinical
                                      interpretation of cultures that grow enterococci is paramount to avoid unnecessary and
                                      potentially deleterious antimicrobial therapy. Traditional antimicrobial treatment for
                                      ampicillin- and glycopeptide-susceptible enterococcal infection remains a penicillin-,
                                      ampicillin-, semisynthetic penicillin–based regimen, or vancomycin in a penicillin-intol-
                                      erant individual. The need for a bactericidal combination with a cell-wall active agent
                                      combined with an aminoglycoside is most supported for native- or prosthetic valve
                                      endocarditis but is unproven for the majority of infections due to enterococci. The
                                      emergence of vancomycin-resistant enterococci prompted the clinical development of
                                      several novel and modified antimicrobial compounds approved for VRE infection (quinu-
                                      pristin-dalfopristin, linezolid) and several approved for non-VRE indications (daptomycin,
                                      tigecycline).
                                                There is a paucity of comparative clinical trial data with these new agents,
                                      although linezolid, based upon its efficacy and tolerability, appears to be the cornerstone
                                      of current treatment approaches. Despite a relatively short period of clinical use, enter-
                                      ococcal resistance has now been described for quinupristin-dalfopristin and linezolid and
                                      more recently even for daptomycin and tigecycline. Moreover, the optimal treatment of
                                      endocarditis due to VRE strains is unknown because, with the exception of daptomycin,
                                      current treatment options only yield bacteriostasis. Nonantimicrobial measures to treat
                                      VRE infection, such as foreign body removal and percutaneous or surgical drainage of
                                      close-spaced infection, reduce both the need for and the duration of anti-enterococcal
                                      treatment and the emergence of resistance to the newer antimicrobials.

                                      KEYWORDS: Enterococcus, vancomycin, linezolid, antimicrobial resistance,
                                      nosocomial infection




      1
       Department of Critical Care Medicine, University of Pittsburgh        Optimizing Antimicrobial Therapy for Serious Infections in the
      Medical Center, Pittsburgh, Pennsylvania.                              Critically Ill; Guest Editor, David L. Paterson, M.D., Ph.D.
         Address for correspondence and reprint requests: Peter K. Linden,      Semin Respir Crit Care Med 2007;28:632–645. Copyright # 2007
      M.D., Department of Critical Care Medicine, University of Pittsburgh   by Thieme Medical Publishers, Inc., 333 Seventh Avenue, New York,
      Medical Center, 602 A Scaife Hall, 3550 Terrace St., Pittsburgh, PA    NY 10001, USA. Tel: +1(212) 584-4662.
      15261 (e-mail: lindenpk@ccm.upmc.edu).                                 DOI 10.1055/s-2007-996410. ISSN 1069-3424.
632
OPTIMIZING THERAPY FOR VANCOMYCIN-RESISTANT ENTEROCOCCI (VRE)/LINDEN           633

INCIDENCE OF ENTEROCOCCAL                                        sporins) is expressed in all enterococci due to the
INFECTION AND RESISTANCE TRENDS                                  expression of inner-cell-wall penicillin-binding proteins
Enterococci have become more common and problematic              (PBPs) with low affinity for these compounds.3 Expo-
pathogens over the past 2 decades, with a rise in both the       sure of such enterococcal strains to an effective b-lactam
overall incidence of enterococcal infection and multidrug        results in inhibitory but not bactericidal activity as
resistance. In a nationwide surveillance study [Surveil-         measured by time-killing kinetic curves.4
lance and Control of Pathogens of Epidemiological                       Low-level resistance to aminoglycosides is secon-
Importance (SCOPE)] between 1995 and 2002, enter-                dary to their low penetrability through the outer-perim-
ococci were the third most frequent cause of nosocomial          eter envelope of the organism, a property that can be
bloodstream infection, and high-level vancomycin-                overcome with the synergistic activity of an effective cell
resistance was present in 60% of Enterococcus faecium            wall active agent such as a penicillin or vancomycin.5
strains but only 2% of E. faecalis strains.1 A Centers for       Although the majority of enterococci exhibit in vitro
Disease Control and Prevention (CDC) surveillance                susceptibility to trimethoprim/sulfamethoxasole their
program during the same time period showed that VRE              ability to utilize exogenous folate in vivo precludes
accounted for 27.5% of intensive care unit (ICU) noso-           the clinical utility of trimethoprim-sulfamethoxazole
comial bacteremic and nonbacteremic infections.2 The             (TMP/SMX) and other agents that impair folate syn-
vast majority of E. faecium strains are multidrug resistant      thesis.6 A significant percentage of enterococci may also
with high-level resistance to penicillin and ampicillin          possess constitutive resistance to macrolides (erythromy-
(MIC > 128 mg/mL) and high-level resistance to                   cin, azithromycin) and lincosamides (clindamycin) pri-
gentamicin (MIC > 1000 mg/mL), which eliminates                  marily mediated by modification of the ribosomal
the potential for bactericidal ‘‘synergistic’’ treatment.        attachment site.7
The forces behind this important trend include the
increased prevalence and greater longevity of immuno-
compromised hosts due to native or iatrogenic immuno-            Acquired Resistance Mechanisms
suppression, the increased use of antimicrobials that            There are few other species of bacteria that have the
are devoid of enterococcal activity (cephalosporins,             proclivity and efficiency of the Enterococcus to acquire
quinolones) and thus selective for more resistant pheno-         new and multiple antimicrobial resistance mechanisms3,4
types, and, most importantly, the appearance of new              (Table 2). The genomic elements that encode for resist-
resistance mechanisms (i.e., high-level vancomycin               ance are carried on plasmid or larger transposon elements,
resistance), which confer resistance to previously effective     are stable, and often carry multiple resistance determi-
antimicrobial classes.                                           nants that culminate in multidrug-resistant strains. Enter-
                                                                 ococci acquire resistance to chloramphenicol (mediated by
                                                                 chloramphenicol acetyltransferase), quinolones (by gyrase
ANTIMICROBIAL RESISTANCE                                         mutations), rifampin (by mutation of the gene that enc-
MECHANISMS AMONG ENTEROCCI                                       odes for RNA polymerase), and tetracyclines (by a variety
                                                                 of mechanisms).8 However, the most clinically important
Intrinsic Resistance Mechanisms                                  antimicrobials to which enterococci have acquired resist-
Enterococci possess several constitutive, nontransferable        ance are discussed in more detail following here.
resistance mechanisms against a variety of antimicro-
bials, which limits therapeutic options even for vanco-
mycin-susceptible enterococci and magnifies the effect            High-Level b-Lactam Resistance
of superimposed intrinsic resistance traits (Table 1).           Overproduction and/or mutation of the penicillin-
Relative or absolute resistance to the b-lactams (pen-           binding protein 5 receptor leading to diminished affinity
icillin, ampicillin, antipseudomonal penicillins, cephalo-       for b-lactams has increased dramatically in E. faecium

       Table 1 Intrinsic Resistance Mechanisms among Enterococci
       Antimicrobial                       Mechanism(s)                        Comments

       Ampicillin, penicillin              Altered binding protein
       Aminoglycosides (LL)                Decreased permeability Altered      High-level gentamicin strains may be
                                             ribosomal binding                   susceptible to high-level streptomycin
       Clindamycin                         Altered ribosomal binding
       Erythromycin                        Altered ribosomal binding
       Tetracyclines                       Efflux pump
       Trimethoprim-sulfamethoxasole       Utilize exogenous folate
       LL, low level.
634   SEMINARS IN RESPIRATORY AND CRITICAL CARE MEDICINE/VOLUME 28, NUMBER 6          2007


             Table 2 Acquired Resistance Mechanisms among Enterococci
             Antimicrobial                  Mechanism(s)                               Comments

             Ampicillin, penicillin (HL)    Mutation of pbp-5                          95% of E. faecium < 5% E. faecalis
             Aminoglycoside (HL)            Enzyme modification                         Plasmid mediated Some HL-gentamicin
                                                                                         R strains may be streptomycin S
             Quinolones                     DNA gyrase mutation
             Chloramphenicol                Efflux pump
             Glycopeptide                   Altered cell wall binding                  Transposon 1546
             Quinupristin/dalfopristin      Ribosomal modification Efflux pump           ermB gene vatd, vate gene
             Linezolid                      Single point mutation                      G2476U mutation
             Daptomycin                     Unknown                                    Described in E. faecalis, E. faecium,
                                                                                         and E. durans
             HL, high level.


      but remains uncommon (< 5%) among E. faecalis                     England in 1986 was a major watershed mark in the
      strains.9,10 This property is expressed constitutively and        evolution of enterococcal antimicrobial resistance and
      carried by resistance genes located on chromosomal ele-           the final step toward the subsequent establishment of
      ments. E. faecium strains with acquired high-level ampi-          endemic multidrug-resistant enterococci.15,16 VRE
      cillin resistance have ampicillin MICs > 128 mg/mL and            strains did not first appear in the United States until
      are neither inhibited nor killed by ampicillin, penicillin,       1989, but thereafter their incidence rapidly increased
      or other b-lactams. The ubiquity of high-level ampicillin         from 0.3% of all enterococci in 1989 to 7.9% in 1993.17
      resistance has been a major step toward the eventual              During this early period the majority of reported VRE
      evolution of multidrug resistance among E. faecium as             isolates were almost exclusively E. faecium, were mono-
      the superimposition of other resistance traits have               or pauciclonal in origin, and predominantly originated
      appeared in such strains.                                         from ICU patients in tertiary care centers, particularly in
                                                                        the northeastern United States, where both vanA and
                                                                        vanB genotype outbreaks were observed; however, there
      High-Level Aminoglycoside Resistance                              was no discernible epidemiologic or clinical differentia-
      The first reports of high-level gentamicin resistant               tion between the two types. Local enhancement of
      (HLGR) strains in the United States were in 1979,                 contact precautions usually aborted or significantly
      appearing in both E. faecalis and E. faecium.11 More recent       modified such outbreaks.
      surveillance data from the SCOPE program between the                     A more contemporaneous surveillance study of
      years 1997 and 1999 showed 69 to 71% of all U.S.                  bloodstream isolates has shown a steady decrease in
      enterococcal strains were HLGR and 40% of all tested              vancomycin susceptibility among E. faecium strains
      vancomycin-resistant enterococci (VRE) strains.12 Enter-          from 60% in 1997 to only 39.1% in 2002, whereas the
      ococci acquire resistance to aminoglycosides via (1)              vast majority (96.1 to 99.4%) of E. faecalis strains
      changes in the ribosomal attachment sites; (2) diminished         continue to remain vancomycin susceptible over this
      aminoglycoside transport into the cell; and (3)                   5-year period.18 The incidence of VRE remains highest
      aminoglycoside-modifying enzymes (adenyltransferase,              in the intensive care unit setting. It has increased to a
      phosphotransferase, and bifunctional acetyl-phospho-              greater relative extent on hospital floors and parahospital
      transferase). Although the majority of HLGR strains               centers such as long-term acute care (LTAC) facilities
      also exhibit high-level streptomycin resistance, a minority       and skilled-care nursing facilities, which often receive
      retain sensitivity to streptomycin; thus susceptibility test-     patients from hospitals with endemic VRE epidemi-
      ing to high-level streptomycin is worthwhile in HLGR              ology.19
      strains.13 No reliable bactericidal activity can be achieved
      with any antimicrobial combination against strains with
      high-level aminoglycoside resistance.14                           Genetic Basis of Vancomycin Resistance
                                                                        Six distinct glycopeptide resistance phenotypes have
                                                                        been discovered: VanA, VanB, VanC, VanD, VanE,
      VANCOMYCIN AND OTHER                                              and VanG, distinguished based upon gene content,
      GLYCOPEPTIDE RESISTANCE                                           glycopeptide minimum inhibitory concentrations
                                                                        (MICs), and inducibility and transferability properties20
      Epidemiology                                                      (Table 3). The vanA and vanB phenotypes uniformly
      Without question, the appearance of E. faecium strains            confer high-level vancomycin resistance (MIC > 64 mg/
      with high-level vancomycin resistance in France and               mL) and have the highest prevalence and clinical
OPTIMIZING THERAPY FOR VANCOMYCIN-RESISTANT ENTEROCOCCI (VRE)/LINDEN                635

Table 3 Level and Type of Vancomycin Resistance in Enterococci
                                                  Acquired Resistance Level, Type                                  Intrinsic Resistance.
                                                                                                                   Low Level Type
Strain             High,               Variable,              Moderate,                         Low                VanC1/C2/C3
Characteristic     VanA                VanB                   VanD
                                                                                VanG               VanE

MIC, mg/L
  Vancomycin       64–1000             4–1000                 64–128            16                 8–32            2–32
Teicoplanin        16–512              0.5–1                  4–64              0.5                0.5             0.5-1
Conjugation        Positive            Positive               Negative          Positive           Negative        Negative
Mobile element     Tn1546              Tn1547 or Tn1549
Expression         Inducible           Inducible              Constitutive      Inducible          Inducible       Constititive
                                                                                                                     Inducible
Location           Plasmid             Plasmid                Choromosome       Choromosome        Choromosome     Choromosome
                     choromosome          choromosome
Modified target     D-Ala-D--Lac        D--Ala-D--Lac          D--Ala-D--Lac     D--Ala-D--Ser      D--Ala-D--Ser   D--Ala-D--Ser

D-Ala-D-Lac, D-alanine-D-lactate; D-Ala-D-ser, D-alanine-D-serine; MIC, minimum inhibitory concentration.
(With permission from Couvalin.20)

importance. Although vanB strains retain susceptibility                  antibiotic-induced mutation. (2) Amplification of the
to teicoplanin, this agent was never commercially avail-                 VRE inoculum within the gastrointestinal reservoir
able in the United States, and rapid resistance has been                 usually due to antimicrobial selective pressures. Prior
described when VanB strains undergo teicoplanin ex-                      or ongoing antimicrobials may also enhance the risk of
posure.21 Transposon 1546 (Tn1546) contains the vanA                     VRE colonization by reducing naturally competing gut
gene complex which encodes for an eight-peptide se-                      flora. (3) Natural- or iatrogenic anatomical or immune
quence culminating in ligase-mediated modification of                     defects that lead to bloodstream or nonbloodstream
the cell wall target for vancomycin from a high affinity                  (tissue) invasion. Perirectal, rectal, or, preferentially,
D-alanine-D-alanine linkage to a low affinity D-ala-
nine-D-lactate linkage on the cell wall peptidoglycan
terminus.22 The vanB gene cluster has partial DNA
homology with the vanA gene cluster and similarly
encodes for ligase modification of the vancomycin target.
The vanA gene has been shown to be transferable in
vitro to Staphylococcus aureus, and naturally occurring
vanA gene–mediated vancomycin resistance, probably
due to horizontal transposon transmission, has been
reported in four methicillin-resistant S. aureus (MRSA)
strains in three patients with protracted vancomycin
exposure for MRSA infection and a fourth patient
without prior vancomycin exposure.23–28


Dynamics and Risk Factors for VRE Colonization
and Infection
Colonization with VRE is a necessary prerequisite for
VRE superinfection, which will arise only when ana-
tomical or other predisposing factors become manifest.
Similar to the more susceptible enterococcal strains, the
natural colonizing reservoir for VRE is the intestinal
tract, with secondary contiguous reservoirs on the skin,
genitourinary tract, and oropharynx.29,30 There are three
sequential processes leading to detectable VRE coloni-
zation and potential subsequent infection with multiple
modifiers (Fig. 1): (1) Exposure to enterococci contain-
ing the vancomycin-resistant genome via contact with an
                                                                         Figure 1 Sequence of vancomycin-resistant enterococci (VRE)
animate or inanimate source. It should be emphasized                     exposure and antimicrobial amplification leading to VRE super-
that the vanA gene does not arise from a spontaneous or                  infection and increased VRE transmission.
636   SEMINARS IN RESPIRATORY AND CRITICAL CARE MEDICINE/VOLUME 28, NUMBER 6        2007


      stool cultures have been the traditional sites to detect      Table 4 Clinical and Epidemiological Clues That MDR
      VRE colonization.31,32 The duration of VRE intestinal         Enterococcal Infection Is Present
      colonization is variable, can last for months to years, and   Significant and recent antimicrobial exposure
      may be indefinite, in part subject to the inoculum-              Multiple agents
      detection threshold of the surveillance testing method          Third-generation cephalosporins
      employed.33–35 Spontaneous clearance of intestinal col-         Antianaerobic antimicrobials
      onization only occurs in the minority of patients in            Vancomycin
      several studies analyzing serial cultures in both antimi-     Positive rectal-fecal surveillance culture or vancomycin-resistant
      crobial- and nonantimicrobial-exposed patients.                 enterococci from a prior clinical site
              Multiple case control and cohort studies have         Intra-abdominal infection (abscess, peritonitis, cholangitis)
      analyzed risk factors for either VRE colonization,            Indwelling catheters in place (bladder, intravascular)
      VRE superinfection, or both.36–39 Two fundamental             High incidence of MDR enterococci in unit, hospital
      risk factor categories are demographic/illness severity       Prior liver transplant, neutropenia, chemotherapy-related
      variables and the type, intensity, and duration of recent       mucositis
      antimicrobial exposure. Demographic risks include du-         MDR, multidrug-resistant.
      ration of hospital- and ICU length of stay, physical
      proximity to VRE-colonized patients in the same unit,         gens, a careful clinical assessment of whether the re-
      and hospitalization in units with a high prevalence of        ported isolate is a likely cause of the patient’s clinical
      VRE, ‘‘colonization pressure.’’ Prior administration of       syndrome that merits specific treatment is always war-
      multiple antibiotics, third-generation cephalosporins,        ranted. Realistically, however, it may be difficult to make
      antimicrobials with anaerobic spectrums (metronidazole,       this distinction, particularly in patients who have major
      clindamycin), and parenteral vancomycin have been             comorbid conditions or critical illness that is naturally
      implicated in case-control analyses of colonization or        coupled with colonization or infection due to multidrug-
      superinfection. Such antimicrobials probably exert a          resistant enterococcal strains. Although enterococci are
      selective effect and amplify otherwise undetectable or        of low virulence, it bears emphasis that they are also
      smaller VRE inocula in the intestines and other secon-        quite capable of promulgating the systemic inflammatory
      dary reservoirs. Donskey and colleagues have demon-           response syndrome (SIRS), severe sepsis, and septic
      strated that the density of VRE as measured by serial         shock and have been a frequent inciting blood pathogen
      quantitative stool cultures increased significantly when       in recent prospective, randomized sepsis trials.42
      patients received ! 1 antianaerobic antimicrobial,                   Abundant and recent observational studies sup-
      whereas this effect was not seen in patients receiving        port the association between appropriate empirical
      antimicrobials with minimal antianaerobic activity.40         antimicrobial therapy and survival.43–45 Thus, for the
      Interestingly, parenteral vancomycin administration re-       severe end of the clinical spectrum, strong consideration
      sulted in no increase in the stool VRE density. More-         should be given to empirical antienterococcal therapy for
      over, patients with high VRE density coupled with fecal       patients whose demographic features and clinical pre-
      incontinence were also more likely to have positive           sentation place them at high risk for enterococcal in-
      environmental cultures for VRE. The ‘‘VRE-selective’’         fection or sepsis. Clearly a major part of this decision also
      effects of antimicrobials and other risk factors become       includes estimating the likelihood that the enterococcal
      relatively diminished when the proportion of patients         strain could be a multidrug-resistant strain. Such epi-
      already colonized with VRE is 50% or greater, which           demiological and clinical clues, which might prompt
      may explain some studies where newly introduced anti-         empirical enterococcal therapy, are summarized in
      biotic control measures may only yield modest reduc-          Table 4.
      tions in VRE colonization and infection rates in
      hyperendemic settings.41 Patients with comorbidities,
      including oncologic conditions, especially neutropenia,       Does the Enterococcus in the Culture Result
      and prior solid organ transplantation, especially liver       Require Antimicrobial Treatment?
      transplantation, appear to have the highest rates of          Microbiological culture data that report the presence of
      VRE bacteremia and poorest outcomes.                          enterococci always require some level of clinical discrim-
                                                                    ination to determine whether they merit treatment.
                                                                    Enterococcal isolates from a respiratory specimen (spu-
      GENERAL ISSUES IN THE TREATMENT OF                            tum, endotracheal aspirate, bronchoalveolar lavage), and
      ENTEROCOCCAL INFECTION                                        skin, wound, or mucosal surfaces almost always represent
      The treatment of serious enterococcal infection is chal-      colonization. Urine cultures obtained via indwelling
      lenging from several aspects. Because enterococci may         bladder (Foley) catheters often represent asymptomatic
      colonize skin, wound, and mucosal surfaces and their          bacteriuria. Wound and intra-abdominal drains often
      isolation is often accompanied by more virulent patho-        become colonized with skin flora, including enterococci.
OPTIMIZING THERAPY FOR VANCOMYCIN-RESISTANT ENTEROCOCCI (VRE)/LINDEN            637

However, such isolates may be significant when the             Table 5 Nonantimicrobial Interventions for
character of the drainage fluid reveals evidence of            Vancomycin-Resistant Enterococcal Infections
inflammatory response (i.e., pyuria or purulence).             Site of Infection         Nonantimicrobial Intervention
Although enterococci may be blood culture contami-
                                                              Bloodstream               Catheter removal
nants, particularly when specimens are obtained from
                                                                                        Consider endovascular infection if
indwelling intravascular catheters, the appropriate clin-
                                                                                          no primary source obvious or
ical bias should be that such cultures represent true
                                                                                          patient with risk factors for
pathogens in most instances. Finally, simple nonantimi-
                                                                                          endocarditis or other endovascular
crobial interventions may obviate the need for antienter-
                                                                                          material
ococcal therapy, such as removal of intravascular or
                                                              Closed-space              Consider percutaneous or surgical
bladder catheters or superficial wound debridement.46,47
                                                                infection                 drainage, e.g., cholangitis—
                                                                                          percutaneous transhepatic
                                                                                          drainage or endoscopic retrograde
Is Bactericidal Therapy Required?
                                                                                          cholangiopancreatography for
The majority of enterococcal infections are not proven to
                                                                                          stent placement
require bactericidal treatment and can be managed
                                                              Urinary tract infection   Removal of bladder catheter
successfully with a single effective agent.48,49 Either
                                                                or bacteriuria
native- or prosthetic-valve endocarditis is the prototyp-
                                                              Superficial wound          Incision and drainage or debridement
ical enterococcal infection for which bactericidal anti-
                                                                infection
microbial therapy is required, usually achieved with the
                                                              Suspected foreign         May require removal if refractory
combination of a cell wall–active agent such as ampicil-
                                                                body infection            to antimicrobial therapy
lin, penicillin, or vancomycin combined with an amino-
                                                                                          or adjacent to devitalized tissue
glycoside such as gentamicin or streptomycin. Other
sites of infection for which bactericidal treatment is
probably merited include enterococcal meningitis and          considerations is summarized in Table 5. It also bears
enterococcemia in a neutropenic host.                         emphasis that the inability to address the nonantimicro-
        However, a bactericidal combination is not pos-       bial considerations of such complex VRE infections has
sible to achieve with enterococci exhibiting high-level       been the principal cofactor leading to the development of
aminoglycoside resistance and almost all strains of           VRE strains that have evolved resistance to the recently
E. faecium strains with high-level vancomycin resistance.     approved VRE antimicrobials, quinupristin-dalfopristin
Uncommon exceptions are vancomycin-resistant E.               and linezolid.
faecalis strains, which retain ampicillin and high-level
gentamicin susceptibility. Successful treatment of such
cases has been reported with ampicillin and gentamicin,       Specific Antimicrobials for the Treatment of
ampicillin þ ofloxacin, penicillin þ streptomycin, and         Vancomycin-Resistant Enterococcal Infection
linezolid þ gentamicin.50 Limited clinical experience         Despite the established high prevalence of multidrug-
is available for the treatment of vancomycin-resistant        resistant enterococcal strains with high-level vancomy-
enterococcal endocarditis with the newer agents (see          cin-resistance there is a remarkable paucity of controlled,
later discussion).                                            comparative trial data on its antimicrobial treatment.
                                                              Major obstacles have been the slow development of
                                                              novel agents with VRE activity, high levels of comor-
Nonantimicrobial Treatment of VRE Infection                   bidity that confound outcome interpretation, complex
Many VRE infections may be partially or completely            surgical infection for which antimicrobial therapy alone
cured with conservative or aggressive nonantimicrobial        is not curative, and the polymicrobial nature of many
interventions. Less serious infections such as bladder        VRE infections, particularly those occurring in the
catheter–associated bacteriuria and urinary tract infec-      abdomen. Both approved and nonapproved treatment
tion may be adequately treated simply with catheter           options for VRE are summarized in Table 6. At present
removal. Postoperative superficial wound infections            there are only two U.S. Food and Drug Administration
may also respond to opening the incision and simple           (FDA)-approved treatments for VRE (E. faecium) in-
drainage or debridement. Closed-space infection such as       fection: quinupristin/dalfopristin (Q/D, Synercid, King
intra-abdominal abscesses, cholangitis due to biliary         Pharmaceuticals, Inc., Bristol, TN) and linezolid (Zyvox,
obstruction, devitalized tissue, or infected foreign bodies   Pfizer, New York, NY) and two other approved agents
(intravascular catheters, synthetic graft or mesh material,   that have in vitro activity against VRE but are not
prosthetics) are not infrequently the primary source of       approved for VRE infection; daptomycin (Cubicin,
VRE bacteremic or nonbacteremic infection. The treat-         Cubist Pharmaceuticals, Lexington, MA), which is ap-
ment implications for infections with such anatomical         proved for complicated skin–skin structure infection,
638   SEMINARS IN RESPIRATORY AND CRITICAL CARE MEDICINE/VOLUME 28, NUMBER 6              2007


      Table 6 Therapeutic Antimicrobial Options for VRE Infection
      Antimicrobial(s)                  Reported Evidence                             Comments

      High-dose ampicillin or           Case reports                                  May be effective with VRE strains
        ampicillin-sulbactam                                                             with ampicillin MIC 32–64 mg/mL
      Chloramphenicol                   Case series                                   Resistance reported
      Tetracycline, doxycycline         Case reports                                   Æ rifampin or ciprofloxacin
      Novobiocin                        Anecdotal                                     No longer manufactured
      Nitrofurantoin                    Small case series                             Only for urinary tract infections
      Teicoplanin                       Case reports                                  Not active against VanAResistance in VanB reported
      Quinupristin/dalfopristin         Large case series but noncomparative          Bacteriostatic
                                                                                      Not active against E. faecalis
                                                                                      Resistance reported
      Linezolid                         1. Dose comparative trial                     Bacteriostatic
                                        2. Large compassionate use series             Resistance reported
      Daptomycin                        1. Case report þ series                       Bactericidal
                                                                                      Resistance reported
      Tigecycline                       In vitro data only                            Bacteriostatic
      Dalbavancin                       In vitro data only                            VanA strains resistant
      Telavancin                        In vitro data only
      Oritavancin                       In vitro data only
      MIC, minimum inhibitory concentration.


      and S. aureus bacteremia and tigecycline (Tygacil, Wyeth              been reported with the use of tetracycline, doxycycline,
      Pharmaceuticals, Inc., Philadelphia, PA), which is ap-                and oral novobiocin combined with either ciprofloxacin or
      proved for complicated skin–skin structure and intra-                 doxycycline; however, such experience has never been rep-
      abdominal infection.                                                  roduced in larger clinical series of prospective trials.55–58
              Prior to the availability of Q/D and linezolid                       Teicoplanin, a glycopeptide not commercially
      approval several centers published their experience with              available in the United States, does have in vitro activity
      a variety of available agents or combinations that dem-               versus VanB phenotypic enterococci. In a European
      onstrated in vitro activity. Clinical success was described           study of 63 patients with vancomycin-susceptible enter-
      with high parenteral dosages of ampicillin or ampicillin/             ococcal infection, clinical and microbiological responses
      sulbactam (18 to 24 g/day), even including endocarditis.              were observed in 84% and 87% of cases, respectively.59
      Such a strategy appears limited to those uncommon VRE                 This agent remains unstudied for VanB enterococcal
      strains with ampicillin MICs of 32 to 64 mg/mL, a target              infection, perhaps in part due to the development of
      range for which plasma ampicillin levels can be achieved              teicoplanin resistance among VanB E. faecalis during
      with high dose therapy.50–52 Because no b-lactamase                   teicoplanin therapy.60,61
      elaboration occurs with VRE, the mechanism of sulbac-                        Nitrofurantoin has in vitro activity against both
      tam activity is not known, although a plausible explan-               VanA and VanB enterococci.62 Due to its ability to
      ation is its intrinsic penicillin-binding protein properties.         achieve high urinary concentrations nitrofurantoin has
              Chloramphenicol has bacteriostatic activity against           been shown to be effective in VRE urinary tract
      enterococci and VRE strains; however, its in vivo efficacy             infection.46,63 Nitrofurantoin cannot be employed for
      was never established. In a retrospective study of 80 cases           VRE outside the urinary tract and in patients with a
      of VRE bacteremia, 51 patients were treated with chlor-               creatinine clearance < 30 mL/min because elevated
      amphenicol from which 22/36 (61%) evaluable patients                  blood concentrations are associated with hepatic, pul-
      demonstrated a clinical response.53 A microbiological                 monary, hematologic, and other toxicities.
      response was also observed in 33/43 (77%) of the micro-
      biologically evaluable patients. No survival benefit was               QUINUPRISTIN/DALFOPRISTIN
      observed compared with VRE bacteremic patients in the                 Quinupristin/dalfopristin (Q/D) is a semisynthetic
      study cohort who did not receive chloramphenicol. Sub-                parenteral streptogramin compound, which is derived
      sequently at the same center, the prevalence of chloram-              from its parent natural compound pristinamycin, a
      phenicol resistance among VRE strains over a 10-year                  product of Streptomyces pristinaspiralis, an oral and top-
      period (1991 to 2000) were observed to increase from 0 to             ical antistaphylococcal agent that has been in clinical use
      11%, a trend that correlated significantly with prior                  in Europe since the 1980s. The major properties of this
      chloramphenicol or quinolone exposure.54 Isolated re-                 compound are summarized in Table 7. This antimicro-
      ports of favorable outcome for VRE infection have also                bial is a 30:70 mixture of quinupristin and dalfopristin,
OPTIMIZING THERAPY FOR VANCOMYCIN-RESISTANT ENTEROCOCCI (VRE)/LINDEN                     639

Table 7 Major Features of Quinupristin/Dalfopristin and Linezolid
Feature                                                     Quinupristin/ Dalfopristin                           Linezolid

Antimicrobial class                                         Streptogramn                                         Oxazolidinone
Peak serum concentrations (mg/L)                            10-12                                                15.1
Elimination half-life (h)                                   0.8 (Q) , 0.6 (D)                                    5.5
Major metabolic routes                                      Hepatobiliary                                        Peripheral nonoxidative
Major elimination routes                                    Faecal (70-75%)                                      Nonrenal (65%)
                                                            Urinary (19%)                                        Urinary (30%)
Protein binding (%)                                         30 (Q) 70 (D)                                        31
Mechanism of action                                         Protein synthesis inhibition                         Protein synthesis inhibition
Site of action                                              50S ribosome                                         70S initiation complex
Postantibiotic effect (h)                                   6–8                                                  1
Bactericidal (vs VRE)                                       No                                                   No
Cytochrome P-450 inhibition                                 Yes                                                  No
Formulations                                                Parenteral                                           Parenteral þ oral
Dose and administration                                     5–7.5 mg/kg q 8–12h                                  600 mg q12 h
Dosage adjustment                                           None                                                 None
Approved indications                                        VRE                                                  VRE
                                                            Complicated SSSI                                     Complicated SSSI
                                                            Nosocomial pneumonia                                 Nosocomial pneumonia
Major adverse effects                                       Phlebitis (peripheral)                               Myelosuppression
                                                            Myalgia/arthralgia
Cost ($US per day; 2000 values)                             $300-350                                             $115 (parenteral)
                                                                                                                 $80 (oral)
D, dalfopristin; Q, quinupristin; qXh, every X hours; SSSI, skin and skin structure infection; VRE, vancomycin-resistant enterocooci.
(With permission from Linden.42)

which are derivatives of streptogramin types B and A,                             Clinical interest in the utility of Q/D for serious
respectively. It is a unique antimicrobial because it acts                VRE infection began in the mid-1990s with a large-
through sequential ribosomal binding and is internally                    scale, noncomparative, open-label, emergency use
synergistic to produce a bactericidal effect. Dalfopristin                program for multiresistant gram-positive infection, prin-
initially binds to the 50S bacterial ribosome, which in-                  cipally vancomycin-resistant E. faecium and MRSA in-
duces a permanent conformational change that acceler-                     fection refractory or intolerant to vancomycin.66,67 The
ates quinupristin ribosomal binding.64 Protein synthesis                  patient populations in both series had a high prevalence
is impaired via both the interruption of peptide chain                    of acute and chronic comorbidities, including diabetes,
elongation and the inhibition of formed peptide extru-                    oncologic conditions, chronic liver disease, dialysis me-
sion. SSuch synergism results in bactericidal activity                    chanical ventilation, and prior organ transplantation.
against some important gram-positive species, including                   Q/D was administered at 7.5 mg/kg intravenously every
Streptococcus pneumoniae, Streptococcus agalacticae, and                  8 hours to patients with documented VRE bacteremia or
some strains of Staphylococcus aureus. However, only                      nonbacteremic VRE infection, with the duration of
bacteriostatic activity is present for the majority of E.                 treatment determined by the primary treating physi-
faecium strains by time–killing curve studies. This effect is             cians. The overall success rate defined as both clinical
primarily mediated by 23S ribosomal modification                           success and bacteriologic eradication was 65.8% in the
encoded for by the ermB gene (erythromycin methylase),                    initial study and 65.6% in the follow-up study. There
which reduces quinupristin affinity for its ribosomal                      have been several reports of clinical cure combining Q/D
binding site and thus limits activity to only the dalfopris-              with doxycycline or high-dose ampicillin in endocarditis;
tin moiety. Such strains are termed MLSb (macrolide-                      however, no larger-scale experience has been per-
lincosamide-streptogramin) phenotypes.65 Erythromycin                     formed.68–70
resistance serves as an excellent surrogate marker for the                        As Q/D usage increased both before and after its
presence of the MLSb phenotype among enterococci.                         regulatory approval in 1999, several important clinical
Q/D is also unique as an antienterococcal agent based                     limitations became apparent. Peripheral intravenous
upon its marked disparity in in vitro susceptibility be-                  administration was associated with a high rate of
tween E. faecium (MIC90 ¼ 1 to 2 mg/mL) and E. faecalis                   phlebitis necessitating central venous administration.
(MIC90 ¼ 8 to 16 mg/mL). This disparity is most likely                    Myalgia and arthralgia unassociated with objective
due to altered ribosomal binding or presence of an active                 inflammatory signs were observed in 7 to 10% of patients
efflux pump.                                                               in the emergency use program, with much higher rates in
640   SEMINARS IN RESPIRATORY AND CRITICAL CARE MEDICINE/VOLUME 28, NUMBER 6      2007


      oncologic patients and liver transplant recipients.71,72      infection.77 Among 549 cases of VRE infection, there
      Although the precise reason for this toxicity is unknown      was an 81.4% clinical cure rate at end-of-therapy.
      a neuropathic cause is suspected. Its higher incidence in     Because linezolid is a bacteriostatic agent that displays
      populations with diminished metabolism and excretion          no synergistic activity with other agents its efficacy in
      suggest it is due to either native drug or metabolite         VRE native- or prosthetic-valve endocarditis remains
      accumulation. Phenotypic resistance to Q/D among              questionable. Both clinical success and failure have been
      E. faecium (MIC ! 4 mg/mL) was observed in six                reported when linezolid has been used as a first-line
      (1.8%) and five (1.3%) of VRE cases, either during or          therapy or salvage treatment; however, no large-scale
      after treatment, from both published emergency use            randomized trial experience is yet available.78–80 In
      series.71,72 Clonal dissemination of Q/D-resistant            recent years, linezolid has become the dominant agent
      strains despite the absence of Q/D or other streptogra-       for the treatment of serious VRE infection. Multiple
      min exposure has been described among pediatric pa-           cases report of linezolid-resistance (MIC ! 8 mg/mL)
      tients.73 Three fundamental resistance mechanisms have        occurring in VRE (E. faecium) and vancomycin-suscep-
      been discovered: enzymatic modification (acetylation) of       tible E. faecalis strains that were susceptible (MIC 1 to
      dalfopristin encoded by the vatD or vatE genes, active        2 mg/mL) at baseline but developed a fourfold or greater
      efflux by an adenosine triphosphate (ATP)-binding              rise in MIC to 8 to 32 mg/mL.81–85 Common to most
      protein encoded by the msrC or lsa genes, and alteration      cases where linezolid-resistance appeared has been a
      of the ribosomal target site encoded for by the erm           protracted length of therapy (> 28 days) associated
      genes.70 Because phenotypic resistance requires the           with retained foci of VRE infection such as abscesses,
      presence of resistance mechanisms to both the quinu-          devitalized tissue, or foreign materials. The majority of
      pristin and the dalfopristin components, at least two or      linezolid-resistant isolates contain a single base-pair
      more resistance genes are present. Several surveillance       mutation in the genome encoding for domain V of the
      studies have uncovered large Q/D resistance reservoirs        23S ribosomal binding site (G2476U mutation). The
      among E. faecium isolated from both domestic poultry          phenotypic level of resistance as determined by elevation
      and livestock in the United States, which may relate to       in MIC level has been shown to correlate with the ‘‘gene
      the use of virginiamycin as a growth-promoting food           dose’’ or number of copies of 23S rDNA containing the
      additive in domestic poultry.74                               G2466U mutation.86 Notably this mutation was pre-
                                                                    dicted by earlier in vitro spiral plate serial passage
      LINEZOLID                                                     experiments with linezolid.87 Horizontal cross trans-
      Linezolid is an oxazolidinone compound, a novel syn-          mission of an identical clone of linezolid-resistant
      thetic class that inhibits bacterial protein synthesis in a   E. faecium among linezolid-naive patients within the
      unique fashion via inhibiting the formation of the 70S        same ICU or hospital center have also been de-
      initiation complex (50S and 30S ribosomes, mRNA,              scribed.88,89 A case- controlled analysis revealed that a
      initiation factors 2 and 3, and fMet-tRNA).75 The major       longer course of linezolid (38 days vs 11 days) and
      properties of linezolid are summarized in Table 6. Line-      linezolid exposure prior to hospitalization were risk
      zolid exhibits a broad gram-positive spectrum but has         factors for the emergence of linezolid-resistant VRE.90
      only bacteriostatic activity against vancomycin-resistant     Thus repeat linezolid susceptibility testing is advisable in
      or susceptible enterococci with an MIC90 of 2 mg/mL,          patients who have had prior linezolid exposure or per-
      which is right at the susceptibility breakpoint. FDA          sistent isolation of a VRE strain on therapy or in patients
      approval was granted in 2000 for vancomycin-resistant         treated in a nosocomial setting with prior linezolid
      E. faecium infection in addition for other indications,       resistance. Although gastrointestinal symptoms are the
      including community-acquired and nosocomial pneu-             most common reported toxicity, reversible myelosup-
      monia and complicated skin and skin structure infection.      pression (thrombocytopenia, leukopenia, and/or anemia)
      Due to the lack of an approved comparator agent,              has been the most important treatment-limiting side
      linezolid was evaluated for patients with clinical and        effect with higher rates observed than the original regis-
      microbiological evidence of serious VRE infection in a        tration studies. Bone marrow examination has shown
      blinded, parenteral, dose-comparative trial comparing         changes similar to those observed with reversible chlor-
      66 patients randomized to 200 mg q 12 hours to                amphenicol toxicity.91,92 Such toxicity is usually observed
      79 patients treated with 600 mg q 12 hours.76 Among           only with sustained linezolid treatment that exceeds
      evaluable patients at end-of-treatment, a modest dose         2 weeks. Other reported toxicities of note include
      response was observed, with 67% and 52% response rates        gastrointestinal upset, rare cases of serotonin syndrome,
      seen in the high-dose and low-dose groups, respectively.      optic- and peripheral neuropathy, and lactic acidosis.93–96
      In addition, efficacy and safety were also demonstrated
      in a large study (n ¼ 796 patients) emergency-use             DAPTOMYCIN
      program for resistant, or treatment-refractory, or treat-     Daptomycin is a novel cyclic lipopeptide compound with
      ment-intolerant patients with serious gram-positive           a broad gram-positive spectrum and rapid bactericidal
OPTIMIZING THERAPY FOR VANCOMYCIN-RESISTANT ENTEROCOCCI (VRE)/LINDEN               641

activity that is currently approved for complicated             tee for Clinical Laboratory Standards (NCCLS) break-
skin–skin structure infection and S. aureus bacteremia,         points for vancomycin-resistant E. faecium strains are not
including right-sided endocarditis.97 Its apparent              yet established.108,109 Although clinical experience with
mechanism of action includes attachment to the exterior         tigecycline for VRE infection is not yet available
of the bacterial cytoplasmic membrane with membrane             it appears to be a promising option, particularly for
penetration of a lipophilic tail with disruption of the         intra-abdominal sites, where it has shown comparable
transmembrane potential due to ion efflux, an effect that        efficacy to meropenem in non-VRE monomicrobial and
is both concentration- and calcium ion–dependent and            polymicrobial infection.
leads to nonlytic bacterial cell death. In vitro studies
have shown nearly uniform activity against vancomycin-          NOVEL GLYCOPEPTIDES (ORITAVANCIN, DALBAVANCIN,
resistant E. faecium and E. faecalis strains with an MIC        TELAVANCIN)
90 of 2–4 mg/mL.98,99 In one recent study examining             Several new glycopeptide derivatives have in vitro bac-
only VRE strains that were either linezolid- or Q/D-            tericidal activity against VRE. Dalbavancin is a long-
resistant, daptomycin demonstrated susceptibility using         acting (half-life 7 to 10 days) derivative of teicoplanin,
a 4 mg/mL provisional breakpoint.100 The MIC break-             which has received approval for the treatment of com-
point is 4 mg/mL for vancomycin-susceptible E. faecalis;        plicated skin–skin structure infection; however, similarly
however, there is no established breakpoint for vanco-          to teicoplanin, this agent lacks in vitro activity against
mycin-susceptible or -resistant E. faecium. Regardless          the more prevalent VanA enterococcal strains.110
of the testing method (E-test, disk diffusion, or broth         Oritavancin is a semisynthetic glycopeptide that blocks
dilution) the zone size or MIC result can be signifi-            peptidoglycan synthesis and exerts bactericidal activity
cantly elevated by a two- to eightfold magnitude with           across a broad gram-positive spectrum.111 It has superior
inadequate calcium supplementation. To the present              activity against vanA and vanB enterococci compared
time, clinical experience with daptomycin for serious           with dalbavancin and telavancin, with concentration-
VRE infection remains quite limited. The optimal                dependent bactericidal activity against both E. faecium
dosing for enterococcal infection is not yet established;       and E. faecalis strains (MIC90 ¼ 1 to 2 mg/mL) and is
however, daily dosing at 6 mg/kg in the absence of renal        synergistic with ampicillin against the majority of iso-
insufficiency has been the most common dosing scheme.            lates.112 This agent has completed phase 3 trials in cSSSI
A randomized phase 3 trial versus linezolid in VRE              (complicated skin-skin structure infection); however,
infection was aborted due to enrollment difficulties.            concerns pertaining to its long half-life, high protein
In a study of nine neutropenic patients with VRE                binding, and reports of spontaneous resistance may
bacteremia treated with daptomycin at 4 mg/kg/day               limit its development.
or 6 mg/kg/day, a clinical and/or microbiological re-                   Telavancin, a long-acting lipoglycopeptide with
sponse was observed in only 4/9 (44%).101 In a second           multiple sites of action at the cell membrane and cell
report a similar response rate of 5/11 (45%) was                wall has shown noninferiority versus standard therapy in
observed in patients with VRE bacteremia and endo-              gram-positive cSSSI, including MRSA; however, clin-
carditis treated with 6 mg/kg/day of daptomycin.102             ical data for VRE are not yet available.113,114
Unfortunately, daptomycin resistance has been re-
ported during treatment for vancomycin-resistant
E. faecalis, E. faecium, and E. durans infection with a         REFERENCES
rise in the MIC to ! 8 mg/mL.103–106                              1. Centers for Disease Control and Prevention. National
                                                                     Nosocomial Infection Surveillance (NNIS) system: report,
TIGECYCLINE                                                          data summary from January 1992 through June 2003, issued
Tigecycline is the first approved agent of the glycylcy-              August 2003. Am J Infect Control 2003;31:481–498
cline class, a group closely related to the tetracyclines but     2. Wisplinghoff H, Bischoff T, Tallent SM, Seiferd H,
                                                                     Wenzel RP, Edmond MB. Nosocomial bloodstream
synthetically modified to achieve an enhanced spectrum
                                                                     infections in US hospitals: analysis of 24,179 cases from a
of activity against MRSA, other multiresistant gram-                 prospective nationwide surveillance study. Clin Infect Dis
positive species, and many gram-negative bacilli.107                 2004;39:309–317
Tigecycline is currently approved for complicated                 3. Moellering RC Jr. The Garrod Lecture. The Enterococcus: a
skin–skin structure and intra-abdominal infection based              classic example of the impact of antimicrobial resistance on
upon phase 3 studies showing comparability to standard               therapeutic options. J Antimicrob Chemother 1991;28:1–12
comparator regimens. Vancomycin-resistant enterococci             4. Murray BE. The life and times of the Enterococcus. Clin
                                                                     Microbiol Rev 1990;3:46–65
were not included in these registration trials.
                                                                  5. Hodges TL, Zighelboim-Daum S, Eliopoulos GM,
However, daptomycin exhibits very low MICs for                       Wennersten C, Moellering RC Jr. Antimicrobial suscept-
both vancomycin-susceptible and vancomycin-resistant                 ibility changes in Enterococcus faecalis following various
versus clinical strains of E. faecium and E. faecalis                penicillin exposure regimens. Antimicrob Agents Chemo-
(MIC90 ¼ 0.012 mg/mL), although National Commit-                     ther 1992;36:121–125
642   SEMINARS IN RESPIRATORY AND CRITICAL CARE MEDICINE/VOLUME 28, NUMBER 6              2007


        6. Hamilton-Miller JM, Purves D. Enterococci and antifolate         25. Centers for Disease Control and Prevention (CDC).
           antibiotics. Eur J Clin Microbiol 1986;5:391–394                     Vancomycin-resistant Staphylococcus aureus—New York.
        7. LeClercq R, Couvalin P. Bacterial resistance to macrolide,           MMWR Morb Mortal Wkly Rep 2005;53:322–323
           lincosamide and streptogramin antibiotics by target mod-         26. Miller D, Urdaneta V, Weltman A. Vancomycin-resistant
           ification. Antimicrob Agents Chemother 1991;35:1267–                  Staphylococcusaureus—Pennsylvania 2002. MMWR Morb
           1272                                                                 Mortal Wkly Rep 2002;51:902
        8. Lefort A, Mainardi JL, Tod M, Lotholary O. Antienter-            27. Sievert DM, Boulton ML, Stolzman G, et al. Staphylococcus
           ococcal antibiotics. Med Clin North Am 2000;84:1471–                 aureus resistant to vancomycin—United States 2002.
           1495                                                                 MMWR Morb Mortal Wkly Rep 2002;51:565–567
        9. Rybkine T, Mainardi JL, Sougakoff W, Collatz E,                  28. Whitener CJ, Park SY, Browne FA, et al. Vancomycin-
           Gutmann L. Penicillin- binding protein 5 sequence                    resistant Staphylococcus aureus in the absence of vancomycin
           alterations in clinical isolates of Enterococcus faecium with        exposure. Clin Infect Dis 2004;38:1049–1055
           different levels of beta-lactam resistance. J Infect Dis 1998;   29. Donskey CJ. The role of the intestinal tract as a reservoir
           178:159–163                                                          and source for transmission of nosocomial pathogens. Clin
       10. Grayson ML, Eliopoulos GM, Wennersten CB, et al.                     Infect Dis 2004;39:219–226
           Increasing resistance to b-lactam antibiotics amongst            30. Beezhold DW, Slaughter S, Hayden MK, et al. Skin
           clinical isolates of Enterococcus faecium: a 22-year review at       colonization with vancomycin-resistant enterococci among
           one institution. Antimicrob Agents Chemother 1991;35:                hospitalized patients with bacteremia. Clin Infect Dis 1997;
           2180–2184                                                            24:704–706
       11. Horodniceanu T, Bougueleret L, El-Solh N, Bieth G,               31. D’Agata EM, Gautam S, Green WK, Tang YW. High rate
           Delbos F. High level resistance to gentamicin in Streptococcus       of false- negative results of the rectal swab culture method
           faecalis subsp. zymogenes. Antimicrob Agents Chemother               in detection of gastrointestinal colonization with vanco-
           1979;16:686–689                                                      mycin-resistant enterococci. Clin Infect Dis 2002;34:167–
       12. Mederski-Samoraj BD, Murray BE. High level resistance to             172
           gentamicin in clinical isolates of enterococci. J Infect Dis     32. Weinstein JW, Tallapragada S, Farrel P, Dembry LM.
           1983;147:751–757                                                     Comparison of rectal and peri-rectal swabs for detection of
       13. Spiegel CA, Hucyke M. Endocarditis due to streptomycin-              colonization with vancomycin-resistant enterococci. J Clin
           susceptible Enterococcus faecalis with high-level gentamicin         Microbiol 1996;34:210–212
           resistance. Arch Intern Med 1989;149:1873–1875                   33. Montecalvo MA, Lencastre H, Carraher M, et al. Natural
       14. Eliopoulos GM. Aminoglycoside resistant enterococcal                 history of colonization with vancomycin-resistant Enter-
           endocarditis. Infect Dis Clin North Am 1993;7:117–133                ococcus faecium. Infect Control Hosp Epidemiol 1995;16:
       15. Leclercq R, Dutka-Malen S, Brisson-Noel A, et al.                    680–685
           Resistance of enterococci to aminoglycosides and glycopep-       34. Baden LR, Thiemke W, Skolnik A, et al. Prolonged
           tides. Clin Infect Dis 1992;15:495–501                               colonization with vancomycin-resistant Enterococcus faecium
       16. Leclercq R, Derlot E, Duval J, Courvalin P. Plasmid                  in long-term care patients and the significance of ‘‘clear-
           mediated resistance to vancomycin and teicoplanin in                 ance.’’ Clin Infect Dis 2001;33:1654–1660
           Enterococcus faecium. N Engl J Med 1988;319:157–161              35. Roghmann MC, Aquiyumi S, Schwalbe R, Morris JG Jr.
       17. Centers for Disease Control and Prevention. Nosocomial               Natural history of colonization with vancomycin-resistant
           enterococci resistant to vancomycin—United States 1989–              Enterococcus faecium. Infect Control Hosp Epidemiol 1997;
           1993. MMWR Morb Mortal Wkly Rep. 1993;42:597–599                     18:679–680
       18. Biedenbach DJ, Moet GJ, Jones RN. Occurrence and                 36. Tornieporth NG, Roberts RB, Hafner JJ, Riley LW. Risk
           antimicrobial resistance pattern comparisons among blood-            factors associated with vancomycin-resistant Enterococcus
           stream infection isolates from the SENTRY Antimicrobial              faecium infection or colonization in 145 matched case and
           Surveillance Program (1997–2002). Diagn Microbiol Infect             control patients. Clin Infect Dis 1996;23:767–772
           Dis 2004;50:59–69                                                37. Bonten MJ, Hayden MK, Nathan C, et al. Epidemiology of
       19. Lai KK, Fontecchio SA, Kelly AL, Baker S, Melvin ZS.                 colonization of patients and environment with vancomycin-
           The changing epidemiology of vancomycin-resistant enter-             resistant enterococci. Lancet 1996;348:1615–1619
           ococci. Infect Control Hosp Epidemiol 2003;24:264–268            38. Morris JG, Shay DK, Hebden JN, et al. Enterococci
       20. Courvalin P. Vancomycin resistance in gram-positive cocci.           resistant to multiple antimicrobial agents including vanco-
           Clin Infect Dis 2006;42:S25–S34                                      mycin, establishment of endemicity in a university medical
       21. Handwerger S, Skoble J. Identification of chromosomal                 center. Ann Intern Med 1995;123:250–259
           mobile element conferring high-level vancomycin resistance       39. Warren DK, Kollef MH, Seiler SM, Fridkin SK, Fraser VJ.
           in Enterococcus faecium. Antimicrob Agents Chemother                 The epidemiology of vancomycin-resistant Enterococcus in a
           1995;39:2446–2453                                                    medical intensive care unit. Infect Control Hosp Epidemiol
       22. Gold HS. Vancomycin resistant enterococci: mechanisms                2003;24:257–263
           and clinical observations. Clin Infect Dis 2001;33:210–219       40. Donskey CJ, Chowdhry T, Hecker M, et al. Effect of
       23. Noble WC, Virani Z, Cree RG. Co-transfer of vancomycin               antibiotic therapy on the density of vancomycin-resistant
           and other resistance genes from Enterococcus faecalis NCTC           enterococci in the stool of colonized patients. N Engl J Med
           12201 to Staphylococcus aureus. FEMS Microbiol Lett 1992;            2000;343:1925–1932
           72:195–198                                                       41. Bonten MJ, Slaughter S, Ambergen AW, et al. The role of
       24. Chang S, Sievert DM, Hageman JC, et al. Infection with               ‘‘colonization pressure’’ in the spread of vancomycin-
           vancomycin- resistant Staphylococcus aureus containing the           resistant enterococci: an important infection control varia-
           vanA resistance gene. N Engl J Med 2003;348:1342–1347                ble. Arch Intern Med 1998;158:1127–1132
OPTIMIZING THERAPY FOR VANCOMYCIN-RESISTANT ENTEROCOCCI (VRE)/LINDEN                643

42. Linden P. Can enterococcal infections initiate sepsis            59. Taylor SE, Paterson DL, Yu VL. Treatment options for
    syndrome? Curr Infect Dis Rep 2003;5:372–378                         chronic prostatitis due to vancomycin-resistant Enterococcus
43. Kollef MH, Sherman G, Ward S, Fraser VJ. Inadequate                  faecium. Eur J Clin Microbiol Infect Dis 1998;17:798–800
    antimicrobial treatment of infections: a risk factor for         60. Schmit JL. Efficacy of teicoplanin for enterococcal infec-
    hospital mortality among critically ill patients. Chest 1999;        tions: 63 cases and review. Clin Infect Dis 1992;15:302–306
    115:462–472                                                      61. Aslangul E, Baptista M, Fantin B, et al. Selection of
44. Kumar A, Robert D, Wood KE, et al. Duration of                       glycopeptide-resistant mutants of VanB type Enterococcus
    hypotension before initiation of effective antimicrobial             faecalis BM4281 in vitro and in experimental endocarditis.
    therapy is the critical determinant of survival in human             J Infect Dis 1997;175:598–605
    septic shock. Crit Care Med 2006;34:1589–1596                    62. Hayden MK, Trenholme GM, Schultz JE, et al. In vivo
45. Ibrahim EH, Sherman G, Ward S, Fraser VJ, Kollef MH.                 development of teicoplanin resistance in a VanB Enterococcus
    The influence of inadequate antimicrobial treatment of                faecalis. J Infect Dis 1993;167:1224–1227
    bloodstream infections on patient outcomes in the ICU            63. Zhanel GG, Hoban DJ, Karlowsky JA. Nitrofurantoin is
    setting. Chest 2000;118:146–155                                      active against vancomycin-resistant enterococci. Antimicrob
46. Quale J, Landman D, Atwood E, et al. Experience with a               Agents Chemother 2001;45:324–326
    hospital-wide outbreak of vancomycin-resistant enterococci.      64. Linden P, Coley K, Kusne S. Bacteriologic efficacy of
    Am J Infect Control 1996;24:372–379                                  nitrofurantoin for the treatment of urinary tract infection
47. Lai KK. Treatment of vancomycin resistant Enterococcus               due to vancomycin-resistant Enterococcus faecium. Clin Infect
    faecium infections. Arch Intern Med 1996;156:2579–2584               Dis 1999;29:999
48. Pankey GA, Sabath LD. Clinical relevance of bacteriostatic       65. Cocito C, DiGiambattista M, Nyssen E, Vannuffel P.
    versus bactericidal mechanisms of action in the treatment of         Inhibition of protein synthesis by streptogramins and related
    gram-positive bacterial infections. Clin Infect Dis                  antibiotics. J Antimicrob Chemother 2007;39(Suppl A):
    2004;38:864–870                                                      7–13
49. Finberg RW, Moellering RC, Tally FP, et al. The                  66. Hershberger E, Donabedian S, Konstantinou K, Zervos MJ.
    importance of bactericidal drugs, future directions in               Quinupristin-dalfopristin resistance in gram-positive bac-
    infectious diseases. Clin Infect Dis 2004;39:1314–1320               teria: mechanism of resistance and epidemiology. Clin Infect
50. Tsigrelis C, Singh KV, Coutinho TD, Murray BE, Baddour               Dis 2004;38:92–98
    LM. Vancomycin-resistant Enterococcus faecalis endocarditis:     67. Moellering RC, Linden PK, Reinhardt J, et al. The efficacy
    linezolid failure and strain characterization of virulence           and safety of quinupristin/dalfopristin for the treatment of
    factors. J Clin Microbiol 2007;45:631–635                            vancomycin-resistant Enterococcus faecium. J Antimicrob
51. Mekonen ET, Noskin GA, Hacek DM, Peterson LR.                        Chemother 1999;44:251–271
    Successful treatment of persistent bacteremia due to vanco-      68. Linden PK, Moellering RC, Wood CA, et al. Treatment of
    mycin resistant ampicillin resistant Enterococcus faecium.           vancomycin-resistant Enterococcus faecium infections with
    Microb Drug Resist 1995;1:249–253                                    quinupristin/dalfopristin. Clin Infect Dis 2001;33:1816–
52. Dodge RA, Daly JS, Davaro R, Glew RH. High-dose                      1823
    ampicillin plus streptomycin for treatment of a patient with     69. Brown J, Freeman BB. Combining quinupristin/dalfopristin
    severe infection due to multi-drug resistant enterococci.            with other agents for resistant infections. Ann Pharmac-
    Clin Infect Dis 1997;25:1269–1270                                    other 2004;38:677–685
53. Murray BE. Vancomycin-resistant enterococcal infections.         70. Bethea JA, Walko CM, Targos PA. Treatment of vancomy-
    N Engl J Med 2000;342:710–721                                        cin-resistant Enterococcus with quinupristin/dalfopristin and
54. Lautenbach E, Schuster MG, Biler WB, et al. The role of              high dose-ampicillin. Ann Pharmacother 2004;38:989–91
    chloramphenicol in the treatment of bloodstream infection        71. Thompson RL, Lavin B, Talbot GH. Endocarditis due to
    due to vancomycin-resistant Enterococcus. Clin Infect Dis            vancomycin-resistant Enterococcus faecium in an immuno-
    1998;27:1259–1265                                                    compromised patient: cure by administering combination
55. Gould CV, Fishman NO, Nakamkin NI, Lautenbach E.                     therapy with quinupristin/dalfopristin and high-dose ampi-
    Chloramphenicol resistance in vancomycin resistant enter-            cillin. South Med J 2003;96:818–820
    ococcal bacteremia: impact of prior fluoroquinolone use.          72. Raad I, Hachem R, Hanna H, et al. Treatment of
    Infect Control Hosp Epidemiol 2004;25:138–145                        vancomycin-resistant enterococcal infections in the immu-
56. Howe RA, Robson M, Oakhill A, et al. Successful use of               nocompromised host: quinupristin-dalfopristin in combina-
    tetracycline as therapy of an immunocompromised patient              tion with minocycline. Antimicrob Agents Chemother
    with septicemia caused by a vancomycin-resistant Enterococcus.       2001;45:3202–3204
    J Antimicrob Chemother 1997;40:144–145                           73. Olsen KM, Rebuck JA, Rupp ME. Arthralgias and myalgias
57. Linden PK, Pasculle AW, Manez R, et al. Utilization of               related to quinupristin-dalfopristin administration. Clin
    novobiocin and ciprofloxacin for the treatment of serious             Infect Dis 2001;32:e83–e86
    infection due to vancomycin-resistant Enterococcus faecium.      74. Werner G, Klare I, Spencker FB, Witte W. Intra-hospital
    In: Program and Abstracts of the Thirty-Third Interscience           dissemination of quinupristin-dalfopristin- and vancomy-
    Conference on Antimicrobial Agents and Chemotherapy,                 cin-resistant Enterococcus faecium in a paediatric ward of a
    New Orleans, LA. Abstract 1027. Washington, DC:                      German hospital. J Antimicrob Chemother 2003;52:113–
    American Society for Microbiology; 1993:307                          115
58. Montecalvo MA, Horowitz H, Wormser GP, et al. Effect             75. Donabedian SM, Perri MB, Vager D, et al. Quinupristin-
    of novobiocin containing antimicrobial regimens on infec-            dalfopristin resistance in Enterococcus faecium isolates from
    tion and colonization with vancomycin resistant Enterococcus         humans, farm animals, and grocery store meat in the United
    faecium. Antimicrob Agents Chemother 1995;39:794                     States. J Clin Microbiol 2006;44:3361–3365
644   SEMINARS IN RESPIRATORY AND CRITICAL CARE MEDICINE/VOLUME 28, NUMBER 6                 2007


       76. Moellering RC. Linezolid: the first oxazolidinone anti-             92. Green SL, Maddox JC, Huttenbach ED. Linezolid and
           microbial. Ann Intern Med 2003;138:135–142                             reversible myelosuppression. JAMA 2001;285:1291
       77. Pharmacia and Upjohn. Linezolid for the Treatment of               93. Halpern M. Linezolid-induced pancytopenia. Clin Infect
           Vancomycin Resistant Enterococcal Infections: A Double-                Dis 2002;35:347–348
           Blind Trial Comparing 600 mg Linezolid Every 12 Hours              94. Wigen CL, Goetz MB. Serotonin syndrome and linezolid.
           with 200 mg Linezolid Every 12 Hours (study report M/                  Clin Infect Dis 2002;34:1651–1652
           1260/0054A). Peapack, NJ: Pharmacia Upjohn; 1999                   95. Saijo T, Hayashi K, Yamada H, Wakakura M. Linezolid-
       78. Birmingham MC, Raynes CR, Meagher AK, Flavin SM,                       induced optic neuropathy. Am J Ophthalmol 2005;139:
           Batts DH, Schentag JJ. Linezolid for the treatment of                  1114–1116
           multidrug-resistant gram-positive infections: experience           96. Bressler AM, Zimmer SM, Gilmore JL, Somani J. Peripheral
           from a compassionate use program. Clin Infect Dis 2003;                neuropathy associated with prolonged use of linezolid. Lancet
           36:159–168                                                             Infect Dis 2004;4:528–531
       79. Stevens MP, Edmond MB. Endocarditis due to vancomy-                97. Apodaca AA, Rakita RM. Linezolid-induced lactic acidosis.
           cin-resistant enterococci: case report and review of the               N Engl J Med 2003;348:86–87
           literature. Clin Infect Dis 2005;41:1134–1142                      98. Carpenter CF, Chambers HF. Daptomycin: another novel
       80. Archuleta S, Murphy B, Keller MJ. Successful treatment of              agent for treating infections due to drug-resistant gram-
           vancomycin-resistant Enterococcus faecium endocarditis with            positive pathogens. Clin Infect Dis 2004;38:994–1000
           linezolid in a renal transplant recipient with human immuno-       99. Pfaller MA, Sader HS, Jones RN. Evaluation of the in vitro
           deficiency virus infection. Transpl Infect Dis 2004;6:117–119           activity of daptomycin against 19615 clinical isolates of
       81. Babcock HM, Ritchie DJ, Christiansen E, Starlin R, Little              gram positive cocci collected in North American hospitals
           R, Stanley S. Successful treatment of vancomycin-resistant             (2002–2005). Diagn Microbiol Infect Dis 2007;57:459–
           Enterococcus endocarditis with oral linezolid. Clin Infect Dis         465
           2001;32:1373–1375                                                 100. Jorgensen JH, Crawford SA, Kelly CC, Patterson JE.
       82. Zimmer SM, Caliendo AM, Thigpen MC, Somani J.                          In vitro activity of daptomycin against vancomycin-resistant
           Failure of linezolid treatment for enterococcal endocarditis.          enterococci of various Van types and comparison of
           Clin Infect Dis 2003;37:e29–e30                                        susceptibility testing methods. Antimicrob Agents Chemo-
       83. Tsigrelis C, Singh KV, Coutinho TD, Murray BE, Baddour                 ther 2003;47:3760–3763
           LM. Vancomycin-resistant Enterococcus faecalis endocarditis:      101. Anastasiou DM, Thorne GM, Luperchio SA, Alder JD.
           linezolid failure and strain characterization of virulence             In vitro activity of daptomycin against clinical isolates with
           factors. J Clin Microbiol 2007;45:631–635                              reduced susceptibility to linezolid and quinupristin/dalfo-
       84. Gonzales RD, Schreckenberger PC, Graham MB, et al.                     pristin. Int J Antimicrob Agents 2006;28:385–388
           Infections due to vancomycin-resistant Enterococcus faecium       102. Poutsiaka DD, Skiffingeron S, Miller KB, Hadley S,
           resistant to linezolid. Lancet 2001;357:1179                           Snydman DR. Daptomycin in the treatment of vancomy-
       85. Seedat J, Zick G, Klare I, et al. Rapid emergence of                   cin-resistant Enterococcus faecium bacteremia in neutropenic
           resistance to linezolid during linezolid therapy of an                 patients. J Infect 2007;54:567–571
           Enterococcus faecium infection. Antimicrob Agents Chemo-          103. Segreti JA, Crank CW, Finney MS. Daptomycin for the
           ther 2006;50:4217–4219                                                 treatment of gram-positive bacteremia and infective endo-
       86. Marra AR, Major Y, Edmond MD. Central venous catheter                  carditis: a retrospective case series of 31 patients. Pharma-
           colonization by linezolid-resistant, vancomycin-susceptible            cotherapy 2006;26:347–352
           Enterococcus faecalis. J Clin Microbiol 2006;44:1915–1916         104. Long JK, Choueiri TK, Hall GS, Avery RK, Sekeres MA.
       87. Rahim S, Pillai SK, Gold HS, Venkataraman L, Inglima K,                Daptomycin-resistant Enterococcus faecium in a patient with
           Press RA. Linezolid-resistant, vancomycin-resistant Enter-             acute myeloid leukemia. Mayop Clin Proc 2005;80:
           ococcus faecium infection in patients without prior exposure to        1215–1216
           linezolid. Clin Infect Dis 2003;36:E146–148                       105. Munoz-Price LS, Lolans K, Quinn JP. Emergence of
       88. Swaney SM, Shinabarger DL, Schaadt RD, Bock JH,                        resistance to daptomycin during treatment of vancomycin-
           Slightom JL, Zurenko GE. Oxazolidinone resistance is                   resistant Enterococcus faecalis infection. Clin Infect Dis 2005;
           associated with a mutation in the peptidyl transferase region          41:565–566
           of 23S rRNA [abstract C-104]. In: Program and Abstracts           106. Lewis JS II, Owens A, Cadena J, et al. Emergence of
           of the 38th Interscience Conference on Antimicrobial                   daptomycin resistance in Enterococcus faecium during dapto-
           Agents and Chemotherapy; San Diego. Washington, DC:                    mycin therapy. Antimicrob Agents Chemother 2005;49:
           American Society for Microbiology; 1998:98–99                          1664–1665
       89. Herrero IA, Issa NC, Patel R. Nosocomial spread of                107. Green MR, Anasetti C, Sandin RL, Rolfe NE, Greene JN.
           linezolid-resistant vancomycin-resistant Enterococcus faecium.         Development of daptomycin resistance in a bone marrow
           N Engl J Med 2002;346:867–869                                          transplant patient with vancomycin-resistant Enterococcus
       90. Marshall SH, Donskey CJ, Hutton-Thomas R, Salata RA,                   durans. J Oncol Pharm Pract 2006;12:179–181
           Rice LB. Gene dosage and linezolid resistance in Enter-           108. Stein GE, Craig WA. Tigecycline: a critical analysis. Clin
           ococcus faecium and Enterococcus faecalis. Antimicrob Agents           Infect Dis 2006;43:518–524
           Chemother 2002;46:3334–3336                                       109. Sader HS, Jones RN, Stilwell MG, Dowzicky MJ, Fritsche
       91. Pai MP, Rodvold KA, Schreckenbefger PC, Gonezales RD,                  TR. Tigecycline activity tested against 26,474 bloodstream
           Petrolatti JM, Quinn JP. Risk factors associated with the              infection isolates: a collection from 6 continents. Diagn
           development of infection with linezolid- and vancomycin-               Microbiol Infect Dis 2005;52:181–186
           resistant Enterococcus faecium. Clin Infect Dis 2002;35:          110. Streit JM, Sader HS, Fritsche TR, Jones RN. Dalbabancin
           1269–1272                                                              activity against selected populations of antimicrobial resistant
OPTIMIZING THERAPY FOR VANCOMYCIN-RESISTANT ENTEROCOCCI (VRE)/LINDEN                645

     gram-positive pathogens. Diagn Microbiol Infect Dis           113. Stryjewski ME, Chu VH, O’Riordan WD, et al. Telavancin
     2005;53:307–310                                                    versus standard therapy for treatment of complicated skin
111. Barrett JF. Recent developments in glycopeptide antibacte-         and skin structure infections caused by gram-positive
     rials. Curr Opin Investig Drugs 2005;6:781–790                     bacteria: FAST 2 Study. Antimicrob Agents Chemother
112. Baltch AL, Smith RP, Ritz WJ, Bopp LH. Comparison of               2006;50:862–867
     inhibitory and bactericidal activities and postantibiotic     114. Poulakou G, Giamarellou H. Investigational treatments for
     effects of LY-333328 and ampicillin used singly and in             postoperative surgical site infections. Expert Opin Investig
     combination against Enterococcus faecium. Antimicrob Agents        Drugs 2007;16:137–155
     Chemother 1998;42:2564–2568

Mais conteúdo relacionado

Mais procurados

MDR Pathogens: The Nightmare of Intensivists
MDR Pathogens: The Nightmare of IntensivistsMDR Pathogens: The Nightmare of Intensivists
MDR Pathogens: The Nightmare of IntensivistsDr.Mahmoud Abbas
 
Multidrug resistance in Microbes
Multidrug resistance in MicrobesMultidrug resistance in Microbes
Multidrug resistance in MicrobesArjun Kumar
 
Role of pharmacists in combating drug resistatnce by neel ratnam.
Role of pharmacists in combating drug resistatnce by neel ratnam.Role of pharmacists in combating drug resistatnce by neel ratnam.
Role of pharmacists in combating drug resistatnce by neel ratnam.neel ratnam
 
Multiple drug resistance
Multiple drug resistanceMultiple drug resistance
Multiple drug resistanceAbdullah Memon
 
Bacterial resistance mechanisms and new trends for resistance overcoming
Bacterial resistance mechanisms and new trends for resistance overcoming Bacterial resistance mechanisms and new trends for resistance overcoming
Bacterial resistance mechanisms and new trends for resistance overcoming Mohammed Fawzy
 
Antbiotic resistance
Antbiotic resistanceAntbiotic resistance
Antbiotic resistancepodila shree
 
A trends of salmonella and antibiotic resistance
A trends of salmonella and antibiotic resistanceA trends of salmonella and antibiotic resistance
A trends of salmonella and antibiotic resistanceAlexander Decker
 
Plasmid and antibiotic resistance
Plasmid and antibiotic resistancePlasmid and antibiotic resistance
Plasmid and antibiotic resistanceyy4me
 
Multi drug resistance molecular pathogenesis
Multi drug resistance   molecular pathogenesisMulti drug resistance   molecular pathogenesis
Multi drug resistance molecular pathogenesisAlagar Suresh
 
Mechanism of development for drug resistance
Mechanism of development for drug resistanceMechanism of development for drug resistance
Mechanism of development for drug resistanceDr Nikita Ingale
 
Specific Multi-drug Resistance MRSA-VRSA-ESBL-KPC
 Specific Multi-drug Resistance MRSA-VRSA-ESBL-KPC Specific Multi-drug Resistance MRSA-VRSA-ESBL-KPC
Specific Multi-drug Resistance MRSA-VRSA-ESBL-KPCAIIMS, New Delhi, India
 
Engineering pathogen resistance in crop plants current trends and future pros...
Engineering pathogen resistance in crop plants current trends and future pros...Engineering pathogen resistance in crop plants current trends and future pros...
Engineering pathogen resistance in crop plants current trends and future pros...UdayenduL10
 
New critical care issues 2015 17
New critical care issues 2015 17New critical care issues 2015 17
New critical care issues 2015 17samirelansary
 
Molecular mechanism of multi drug resistance
Molecular mechanism of multi drug resistanceMolecular mechanism of multi drug resistance
Molecular mechanism of multi drug resistanceDr. Geetanjali Baruah
 

Mais procurados (19)

MDR Pathogens: The Nightmare of Intensivists
MDR Pathogens: The Nightmare of IntensivistsMDR Pathogens: The Nightmare of Intensivists
MDR Pathogens: The Nightmare of Intensivists
 
Multidrug resistance in Microbes
Multidrug resistance in MicrobesMultidrug resistance in Microbes
Multidrug resistance in Microbes
 
Role of pharmacists in combating drug resistatnce by neel ratnam.
Role of pharmacists in combating drug resistatnce by neel ratnam.Role of pharmacists in combating drug resistatnce by neel ratnam.
Role of pharmacists in combating drug resistatnce by neel ratnam.
 
Multi drug resistanse
Multi drug resistanseMulti drug resistanse
Multi drug resistanse
 
Multiple drug resistance
Multiple drug resistanceMultiple drug resistance
Multiple drug resistance
 
Bacterial resistance mechanisms and new trends for resistance overcoming
Bacterial resistance mechanisms and new trends for resistance overcoming Bacterial resistance mechanisms and new trends for resistance overcoming
Bacterial resistance mechanisms and new trends for resistance overcoming
 
Antbiotic resistance
Antbiotic resistanceAntbiotic resistance
Antbiotic resistance
 
Antibiotic drug resistnc and susceptibility
Antibiotic drug resistnc and susceptibilityAntibiotic drug resistnc and susceptibility
Antibiotic drug resistnc and susceptibility
 
A trends of salmonella and antibiotic resistance
A trends of salmonella and antibiotic resistanceA trends of salmonella and antibiotic resistance
A trends of salmonella and antibiotic resistance
 
Plasmid and antibiotic resistance
Plasmid and antibiotic resistancePlasmid and antibiotic resistance
Plasmid and antibiotic resistance
 
Multi drug resistance molecular pathogenesis
Multi drug resistance   molecular pathogenesisMulti drug resistance   molecular pathogenesis
Multi drug resistance molecular pathogenesis
 
Mechanism of development for drug resistance
Mechanism of development for drug resistanceMechanism of development for drug resistance
Mechanism of development for drug resistance
 
Nci mdro
Nci mdroNci mdro
Nci mdro
 
Antibiotic resistance
Antibiotic resistanceAntibiotic resistance
Antibiotic resistance
 
Specific Multi-drug Resistance MRSA-VRSA-ESBL-KPC
 Specific Multi-drug Resistance MRSA-VRSA-ESBL-KPC Specific Multi-drug Resistance MRSA-VRSA-ESBL-KPC
Specific Multi-drug Resistance MRSA-VRSA-ESBL-KPC
 
Engineering pathogen resistance in crop plants current trends and future pros...
Engineering pathogen resistance in crop plants current trends and future pros...Engineering pathogen resistance in crop plants current trends and future pros...
Engineering pathogen resistance in crop plants current trends and future pros...
 
Antimicrobial Resistance (AMR)
Antimicrobial Resistance (AMR)Antimicrobial Resistance (AMR)
Antimicrobial Resistance (AMR)
 
New critical care issues 2015 17
New critical care issues 2015 17New critical care issues 2015 17
New critical care issues 2015 17
 
Molecular mechanism of multi drug resistance
Molecular mechanism of multi drug resistanceMolecular mechanism of multi drug resistance
Molecular mechanism of multi drug resistance
 

Semelhante a Optimizing Therapy for Drug-Resistant Bacterial Infections

QUINOLONES IN CARTIs
QUINOLONES IN CARTIsQUINOLONES IN CARTIs
QUINOLONES IN CARTIsJohnScreen
 
Resistance in enterococci
Resistance in enterococciResistance in enterococci
Resistance in enterococciNeelima Singh
 
13.Antimicrobial Resistance.pptx
13.Antimicrobial Resistance.pptx13.Antimicrobial Resistance.pptx
13.Antimicrobial Resistance.pptxRehmatRehmat2
 
Mona Ahmed.docxxx.pptx
Mona Ahmed.docxxx.pptxMona Ahmed.docxxx.pptx
Mona Ahmed.docxxx.pptxmominaishfaq1
 
Combating Antimicrobial Resistance:Aminoglocisides Back To The Future
Combating Antimicrobial Resistance:Aminoglocisides Back To The FutureCombating Antimicrobial Resistance:Aminoglocisides Back To The Future
Combating Antimicrobial Resistance:Aminoglocisides Back To The FutureDoctors Republic
 
En atbantibiotics
En atbantibioticsEn atbantibiotics
En atbantibioticsDr P Deepak
 
Antibiotic prescription and bacterial resistance
Antibiotic prescription and bacterial resistanceAntibiotic prescription and bacterial resistance
Antibiotic prescription and bacterial resistanceMamdouh Sabry
 
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
 
Antimicrobial agent
Antimicrobial agentAntimicrobial agent
Antimicrobial agentaiiinura
 
ANTIBACTERIAL DRUGS.pdfbbdbbdndbdnsjdbdjbdj
ANTIBACTERIAL DRUGS.pdfbbdbbdndbdnsjdbdjbdjANTIBACTERIAL DRUGS.pdfbbdbbdndbdnsjdbdjbdj
ANTIBACTERIAL DRUGS.pdfbbdbbdndbdnsjdbdjbdjHappychifunda
 
Managing MDR/XDR Gram Negative infections in ICU
Managing MDR/XDR Gram Negative infections in ICUManaging MDR/XDR Gram Negative infections in ICU
Managing MDR/XDR Gram Negative infections in ICUVitrag Shah
 
Basics of Antimicrobial Drugs
Basics of Antimicrobial DrugsBasics of Antimicrobial Drugs
Basics of Antimicrobial DrugsASSOCHAM
 
Management of antibiotic resistance upload
Management of antibiotic resistance uploadManagement of antibiotic resistance upload
Management of antibiotic resistance uploadAnimesh Gupta
 
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
 
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
 
Antibiotics resistance
Antibiotics resistanceAntibiotics resistance
Antibiotics resistanceAhmad Ali khan
 

Semelhante a Optimizing Therapy for Drug-Resistant Bacterial Infections (20)

QUINOLONES IN CARTIs
QUINOLONES IN CARTIsQUINOLONES IN CARTIs
QUINOLONES IN CARTIs
 
Resistance in enterococci
Resistance in enterococciResistance in enterococci
Resistance in enterococci
 
13.Antimicrobial Resistance.pptx
13.Antimicrobial Resistance.pptx13.Antimicrobial Resistance.pptx
13.Antimicrobial Resistance.pptx
 
Antibiotics
Antibiotics Antibiotics
Antibiotics
 
Mona Ahmed.docxxx.pptx
Mona Ahmed.docxxx.pptxMona Ahmed.docxxx.pptx
Mona Ahmed.docxxx.pptx
 
Combating Antimicrobial Resistance:Aminoglocisides Back To The Future
Combating Antimicrobial Resistance:Aminoglocisides Back To The FutureCombating Antimicrobial Resistance:Aminoglocisides Back To The Future
Combating Antimicrobial Resistance:Aminoglocisides Back To The Future
 
En atbantibiotics
En atbantibioticsEn atbantibiotics
En atbantibiotics
 
Antimicrobials
AntimicrobialsAntimicrobials
Antimicrobials
 
Antibiotic prescription and bacterial resistance
Antibiotic prescription and bacterial resistanceAntibiotic prescription and bacterial resistance
Antibiotic prescription and bacterial resistance
 
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
 
Antimicrobial agent
Antimicrobial agentAntimicrobial agent
Antimicrobial agent
 
ANTIBACTERIAL DRUGS.pdfbbdbbdndbdnsjdbdjbdj
ANTIBACTERIAL DRUGS.pdfbbdbbdndbdnsjdbdjbdjANTIBACTERIAL DRUGS.pdfbbdbbdndbdnsjdbdjbdj
ANTIBACTERIAL DRUGS.pdfbbdbbdndbdnsjdbdjbdj
 
Managing MDR/XDR Gram Negative infections in ICU
Managing MDR/XDR Gram Negative infections in ICUManaging MDR/XDR Gram Negative infections in ICU
Managing MDR/XDR Gram Negative infections in ICU
 
Basics of Antimicrobial Drugs
Basics of Antimicrobial DrugsBasics of Antimicrobial Drugs
Basics of Antimicrobial Drugs
 
Management of antibiotic resistance upload
Management of antibiotic resistance uploadManagement of antibiotic resistance upload
Management of antibiotic resistance upload
 
Prerna presentation
Prerna presentationPrerna presentation
Prerna presentation
 
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
 
ANTIMICROBIAL RESISTANCE
ANTIMICROBIAL RESISTANCEANTIMICROBIAL RESISTANCE
ANTIMICROBIAL RESISTANCE
 
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
 
Antibiotics resistance
Antibiotics resistanceAntibiotics resistance
Antibiotics resistance
 

Mais de 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
 
Vancomycin revisited ccc 2008
Vancomycin revisited ccc 2008Vancomycin revisited ccc 2008
Vancomycin revisited ccc 2008negrulo2013
 

Mais de negrulo2013 (17)

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
Vancomycin revisited ccc 2008Vancomycin revisited ccc 2008
Vancomycin revisited ccc 2008
 

Optimizing Therapy for Drug-Resistant Bacterial Infections

  • 1. Optimizing Therapy for Vancomycin- Resistant Enterococci (VRE) Peter K. Linden, M.D.1 ABSTRACT Enterococci are gram-positive, facultative bacteria with low intrinsic virulence but capable of causing a diverse variety of infections such as bacteremia with or without endocarditis, and intra-abdominal, wound, and genitourinary infection. During the past 2 decades the incidence of hospital-acquired enterococcal infection has significantly risen and is increasingly due to multidrug-resistant strains, primarily to the coacquisition of genetic determinants that encode for the stable expression of high-level b-lactam, aminoglycoside, and glycopeptide resistance. Because enterococci constitute part of the normal colonizing flora, careful clinical interpretation of cultures that grow enterococci is paramount to avoid unnecessary and potentially deleterious antimicrobial therapy. Traditional antimicrobial treatment for ampicillin- and glycopeptide-susceptible enterococcal infection remains a penicillin-, ampicillin-, semisynthetic penicillin–based regimen, or vancomycin in a penicillin-intol- erant individual. The need for a bactericidal combination with a cell-wall active agent combined with an aminoglycoside is most supported for native- or prosthetic valve endocarditis but is unproven for the majority of infections due to enterococci. The emergence of vancomycin-resistant enterococci prompted the clinical development of several novel and modified antimicrobial compounds approved for VRE infection (quinu- pristin-dalfopristin, linezolid) and several approved for non-VRE indications (daptomycin, tigecycline). There is a paucity of comparative clinical trial data with these new agents, although linezolid, based upon its efficacy and tolerability, appears to be the cornerstone of current treatment approaches. Despite a relatively short period of clinical use, enter- ococcal resistance has now been described for quinupristin-dalfopristin and linezolid and more recently even for daptomycin and tigecycline. Moreover, the optimal treatment of endocarditis due to VRE strains is unknown because, with the exception of daptomycin, current treatment options only yield bacteriostasis. Nonantimicrobial measures to treat VRE infection, such as foreign body removal and percutaneous or surgical drainage of close-spaced infection, reduce both the need for and the duration of anti-enterococcal treatment and the emergence of resistance to the newer antimicrobials. KEYWORDS: Enterococcus, vancomycin, linezolid, antimicrobial resistance, nosocomial infection 1 Department of Critical Care Medicine, University of Pittsburgh Optimizing Antimicrobial Therapy for Serious Infections in the Medical Center, Pittsburgh, Pennsylvania. Critically Ill; Guest Editor, David L. Paterson, M.D., Ph.D. Address for correspondence and reprint requests: Peter K. Linden, Semin Respir Crit Care Med 2007;28:632–645. Copyright # 2007 M.D., Department of Critical Care Medicine, University of Pittsburgh by Thieme Medical Publishers, Inc., 333 Seventh Avenue, New York, Medical Center, 602 A Scaife Hall, 3550 Terrace St., Pittsburgh, PA NY 10001, USA. Tel: +1(212) 584-4662. 15261 (e-mail: lindenpk@ccm.upmc.edu). DOI 10.1055/s-2007-996410. ISSN 1069-3424. 632
  • 2. OPTIMIZING THERAPY FOR VANCOMYCIN-RESISTANT ENTEROCOCCI (VRE)/LINDEN 633 INCIDENCE OF ENTEROCOCCAL sporins) is expressed in all enterococci due to the INFECTION AND RESISTANCE TRENDS expression of inner-cell-wall penicillin-binding proteins Enterococci have become more common and problematic (PBPs) with low affinity for these compounds.3 Expo- pathogens over the past 2 decades, with a rise in both the sure of such enterococcal strains to an effective b-lactam overall incidence of enterococcal infection and multidrug results in inhibitory but not bactericidal activity as resistance. In a nationwide surveillance study [Surveil- measured by time-killing kinetic curves.4 lance and Control of Pathogens of Epidemiological Low-level resistance to aminoglycosides is secon- Importance (SCOPE)] between 1995 and 2002, enter- dary to their low penetrability through the outer-perim- ococci were the third most frequent cause of nosocomial eter envelope of the organism, a property that can be bloodstream infection, and high-level vancomycin- overcome with the synergistic activity of an effective cell resistance was present in 60% of Enterococcus faecium wall active agent such as a penicillin or vancomycin.5 strains but only 2% of E. faecalis strains.1 A Centers for Although the majority of enterococci exhibit in vitro Disease Control and Prevention (CDC) surveillance susceptibility to trimethoprim/sulfamethoxasole their program during the same time period showed that VRE ability to utilize exogenous folate in vivo precludes accounted for 27.5% of intensive care unit (ICU) noso- the clinical utility of trimethoprim-sulfamethoxazole comial bacteremic and nonbacteremic infections.2 The (TMP/SMX) and other agents that impair folate syn- vast majority of E. faecium strains are multidrug resistant thesis.6 A significant percentage of enterococci may also with high-level resistance to penicillin and ampicillin possess constitutive resistance to macrolides (erythromy- (MIC > 128 mg/mL) and high-level resistance to cin, azithromycin) and lincosamides (clindamycin) pri- gentamicin (MIC > 1000 mg/mL), which eliminates marily mediated by modification of the ribosomal the potential for bactericidal ‘‘synergistic’’ treatment. attachment site.7 The forces behind this important trend include the increased prevalence and greater longevity of immuno- compromised hosts due to native or iatrogenic immuno- Acquired Resistance Mechanisms suppression, the increased use of antimicrobials that There are few other species of bacteria that have the are devoid of enterococcal activity (cephalosporins, proclivity and efficiency of the Enterococcus to acquire quinolones) and thus selective for more resistant pheno- new and multiple antimicrobial resistance mechanisms3,4 types, and, most importantly, the appearance of new (Table 2). The genomic elements that encode for resist- resistance mechanisms (i.e., high-level vancomycin ance are carried on plasmid or larger transposon elements, resistance), which confer resistance to previously effective are stable, and often carry multiple resistance determi- antimicrobial classes. nants that culminate in multidrug-resistant strains. Enter- ococci acquire resistance to chloramphenicol (mediated by chloramphenicol acetyltransferase), quinolones (by gyrase ANTIMICROBIAL RESISTANCE mutations), rifampin (by mutation of the gene that enc- MECHANISMS AMONG ENTEROCCI odes for RNA polymerase), and tetracyclines (by a variety of mechanisms).8 However, the most clinically important Intrinsic Resistance Mechanisms antimicrobials to which enterococci have acquired resist- Enterococci possess several constitutive, nontransferable ance are discussed in more detail following here. resistance mechanisms against a variety of antimicro- bials, which limits therapeutic options even for vanco- mycin-susceptible enterococci and magnifies the effect High-Level b-Lactam Resistance of superimposed intrinsic resistance traits (Table 1). Overproduction and/or mutation of the penicillin- Relative or absolute resistance to the b-lactams (pen- binding protein 5 receptor leading to diminished affinity icillin, ampicillin, antipseudomonal penicillins, cephalo- for b-lactams has increased dramatically in E. faecium Table 1 Intrinsic Resistance Mechanisms among Enterococci Antimicrobial Mechanism(s) Comments Ampicillin, penicillin Altered binding protein Aminoglycosides (LL) Decreased permeability Altered High-level gentamicin strains may be ribosomal binding susceptible to high-level streptomycin Clindamycin Altered ribosomal binding Erythromycin Altered ribosomal binding Tetracyclines Efflux pump Trimethoprim-sulfamethoxasole Utilize exogenous folate LL, low level.
  • 3. 634 SEMINARS IN RESPIRATORY AND CRITICAL CARE MEDICINE/VOLUME 28, NUMBER 6 2007 Table 2 Acquired Resistance Mechanisms among Enterococci Antimicrobial Mechanism(s) Comments Ampicillin, penicillin (HL) Mutation of pbp-5 95% of E. faecium < 5% E. faecalis Aminoglycoside (HL) Enzyme modification Plasmid mediated Some HL-gentamicin R strains may be streptomycin S Quinolones DNA gyrase mutation Chloramphenicol Efflux pump Glycopeptide Altered cell wall binding Transposon 1546 Quinupristin/dalfopristin Ribosomal modification Efflux pump ermB gene vatd, vate gene Linezolid Single point mutation G2476U mutation Daptomycin Unknown Described in E. faecalis, E. faecium, and E. durans HL, high level. but remains uncommon (< 5%) among E. faecalis England in 1986 was a major watershed mark in the strains.9,10 This property is expressed constitutively and evolution of enterococcal antimicrobial resistance and carried by resistance genes located on chromosomal ele- the final step toward the subsequent establishment of ments. E. faecium strains with acquired high-level ampi- endemic multidrug-resistant enterococci.15,16 VRE cillin resistance have ampicillin MICs > 128 mg/mL and strains did not first appear in the United States until are neither inhibited nor killed by ampicillin, penicillin, 1989, but thereafter their incidence rapidly increased or other b-lactams. The ubiquity of high-level ampicillin from 0.3% of all enterococci in 1989 to 7.9% in 1993.17 resistance has been a major step toward the eventual During this early period the majority of reported VRE evolution of multidrug resistance among E. faecium as isolates were almost exclusively E. faecium, were mono- the superimposition of other resistance traits have or pauciclonal in origin, and predominantly originated appeared in such strains. from ICU patients in tertiary care centers, particularly in the northeastern United States, where both vanA and vanB genotype outbreaks were observed; however, there High-Level Aminoglycoside Resistance was no discernible epidemiologic or clinical differentia- The first reports of high-level gentamicin resistant tion between the two types. Local enhancement of (HLGR) strains in the United States were in 1979, contact precautions usually aborted or significantly appearing in both E. faecalis and E. faecium.11 More recent modified such outbreaks. surveillance data from the SCOPE program between the A more contemporaneous surveillance study of years 1997 and 1999 showed 69 to 71% of all U.S. bloodstream isolates has shown a steady decrease in enterococcal strains were HLGR and 40% of all tested vancomycin susceptibility among E. faecium strains vancomycin-resistant enterococci (VRE) strains.12 Enter- from 60% in 1997 to only 39.1% in 2002, whereas the ococci acquire resistance to aminoglycosides via (1) vast majority (96.1 to 99.4%) of E. faecalis strains changes in the ribosomal attachment sites; (2) diminished continue to remain vancomycin susceptible over this aminoglycoside transport into the cell; and (3) 5-year period.18 The incidence of VRE remains highest aminoglycoside-modifying enzymes (adenyltransferase, in the intensive care unit setting. It has increased to a phosphotransferase, and bifunctional acetyl-phospho- greater relative extent on hospital floors and parahospital transferase). Although the majority of HLGR strains centers such as long-term acute care (LTAC) facilities also exhibit high-level streptomycin resistance, a minority and skilled-care nursing facilities, which often receive retain sensitivity to streptomycin; thus susceptibility test- patients from hospitals with endemic VRE epidemi- ing to high-level streptomycin is worthwhile in HLGR ology.19 strains.13 No reliable bactericidal activity can be achieved with any antimicrobial combination against strains with high-level aminoglycoside resistance.14 Genetic Basis of Vancomycin Resistance Six distinct glycopeptide resistance phenotypes have been discovered: VanA, VanB, VanC, VanD, VanE, VANCOMYCIN AND OTHER and VanG, distinguished based upon gene content, GLYCOPEPTIDE RESISTANCE glycopeptide minimum inhibitory concentrations (MICs), and inducibility and transferability properties20 Epidemiology (Table 3). The vanA and vanB phenotypes uniformly Without question, the appearance of E. faecium strains confer high-level vancomycin resistance (MIC > 64 mg/ with high-level vancomycin resistance in France and mL) and have the highest prevalence and clinical
  • 4. OPTIMIZING THERAPY FOR VANCOMYCIN-RESISTANT ENTEROCOCCI (VRE)/LINDEN 635 Table 3 Level and Type of Vancomycin Resistance in Enterococci Acquired Resistance Level, Type Intrinsic Resistance. Low Level Type Strain High, Variable, Moderate, Low VanC1/C2/C3 Characteristic VanA VanB VanD VanG VanE MIC, mg/L Vancomycin 64–1000 4–1000 64–128 16 8–32 2–32 Teicoplanin 16–512 0.5–1 4–64 0.5 0.5 0.5-1 Conjugation Positive Positive Negative Positive Negative Negative Mobile element Tn1546 Tn1547 or Tn1549 Expression Inducible Inducible Constitutive Inducible Inducible Constititive Inducible Location Plasmid Plasmid Choromosome Choromosome Choromosome Choromosome choromosome choromosome Modified target D-Ala-D--Lac D--Ala-D--Lac D--Ala-D--Lac D--Ala-D--Ser D--Ala-D--Ser D--Ala-D--Ser D-Ala-D-Lac, D-alanine-D-lactate; D-Ala-D-ser, D-alanine-D-serine; MIC, minimum inhibitory concentration. (With permission from Couvalin.20) importance. Although vanB strains retain susceptibility antibiotic-induced mutation. (2) Amplification of the to teicoplanin, this agent was never commercially avail- VRE inoculum within the gastrointestinal reservoir able in the United States, and rapid resistance has been usually due to antimicrobial selective pressures. Prior described when VanB strains undergo teicoplanin ex- or ongoing antimicrobials may also enhance the risk of posure.21 Transposon 1546 (Tn1546) contains the vanA VRE colonization by reducing naturally competing gut gene complex which encodes for an eight-peptide se- flora. (3) Natural- or iatrogenic anatomical or immune quence culminating in ligase-mediated modification of defects that lead to bloodstream or nonbloodstream the cell wall target for vancomycin from a high affinity (tissue) invasion. Perirectal, rectal, or, preferentially, D-alanine-D-alanine linkage to a low affinity D-ala- nine-D-lactate linkage on the cell wall peptidoglycan terminus.22 The vanB gene cluster has partial DNA homology with the vanA gene cluster and similarly encodes for ligase modification of the vancomycin target. The vanA gene has been shown to be transferable in vitro to Staphylococcus aureus, and naturally occurring vanA gene–mediated vancomycin resistance, probably due to horizontal transposon transmission, has been reported in four methicillin-resistant S. aureus (MRSA) strains in three patients with protracted vancomycin exposure for MRSA infection and a fourth patient without prior vancomycin exposure.23–28 Dynamics and Risk Factors for VRE Colonization and Infection Colonization with VRE is a necessary prerequisite for VRE superinfection, which will arise only when ana- tomical or other predisposing factors become manifest. Similar to the more susceptible enterococcal strains, the natural colonizing reservoir for VRE is the intestinal tract, with secondary contiguous reservoirs on the skin, genitourinary tract, and oropharynx.29,30 There are three sequential processes leading to detectable VRE coloni- zation and potential subsequent infection with multiple modifiers (Fig. 1): (1) Exposure to enterococci contain- ing the vancomycin-resistant genome via contact with an Figure 1 Sequence of vancomycin-resistant enterococci (VRE) animate or inanimate source. It should be emphasized exposure and antimicrobial amplification leading to VRE super- that the vanA gene does not arise from a spontaneous or infection and increased VRE transmission.
  • 5. 636 SEMINARS IN RESPIRATORY AND CRITICAL CARE MEDICINE/VOLUME 28, NUMBER 6 2007 stool cultures have been the traditional sites to detect Table 4 Clinical and Epidemiological Clues That MDR VRE colonization.31,32 The duration of VRE intestinal Enterococcal Infection Is Present colonization is variable, can last for months to years, and Significant and recent antimicrobial exposure may be indefinite, in part subject to the inoculum- Multiple agents detection threshold of the surveillance testing method Third-generation cephalosporins employed.33–35 Spontaneous clearance of intestinal col- Antianaerobic antimicrobials onization only occurs in the minority of patients in Vancomycin several studies analyzing serial cultures in both antimi- Positive rectal-fecal surveillance culture or vancomycin-resistant crobial- and nonantimicrobial-exposed patients. enterococci from a prior clinical site Multiple case control and cohort studies have Intra-abdominal infection (abscess, peritonitis, cholangitis) analyzed risk factors for either VRE colonization, Indwelling catheters in place (bladder, intravascular) VRE superinfection, or both.36–39 Two fundamental High incidence of MDR enterococci in unit, hospital risk factor categories are demographic/illness severity Prior liver transplant, neutropenia, chemotherapy-related variables and the type, intensity, and duration of recent mucositis antimicrobial exposure. Demographic risks include du- MDR, multidrug-resistant. ration of hospital- and ICU length of stay, physical proximity to VRE-colonized patients in the same unit, gens, a careful clinical assessment of whether the re- and hospitalization in units with a high prevalence of ported isolate is a likely cause of the patient’s clinical VRE, ‘‘colonization pressure.’’ Prior administration of syndrome that merits specific treatment is always war- multiple antibiotics, third-generation cephalosporins, ranted. Realistically, however, it may be difficult to make antimicrobials with anaerobic spectrums (metronidazole, this distinction, particularly in patients who have major clindamycin), and parenteral vancomycin have been comorbid conditions or critical illness that is naturally implicated in case-control analyses of colonization or coupled with colonization or infection due to multidrug- superinfection. Such antimicrobials probably exert a resistant enterococcal strains. Although enterococci are selective effect and amplify otherwise undetectable or of low virulence, it bears emphasis that they are also smaller VRE inocula in the intestines and other secon- quite capable of promulgating the systemic inflammatory dary reservoirs. Donskey and colleagues have demon- response syndrome (SIRS), severe sepsis, and septic strated that the density of VRE as measured by serial shock and have been a frequent inciting blood pathogen quantitative stool cultures increased significantly when in recent prospective, randomized sepsis trials.42 patients received ! 1 antianaerobic antimicrobial, Abundant and recent observational studies sup- whereas this effect was not seen in patients receiving port the association between appropriate empirical antimicrobials with minimal antianaerobic activity.40 antimicrobial therapy and survival.43–45 Thus, for the Interestingly, parenteral vancomycin administration re- severe end of the clinical spectrum, strong consideration sulted in no increase in the stool VRE density. More- should be given to empirical antienterococcal therapy for over, patients with high VRE density coupled with fecal patients whose demographic features and clinical pre- incontinence were also more likely to have positive sentation place them at high risk for enterococcal in- environmental cultures for VRE. The ‘‘VRE-selective’’ fection or sepsis. Clearly a major part of this decision also effects of antimicrobials and other risk factors become includes estimating the likelihood that the enterococcal relatively diminished when the proportion of patients strain could be a multidrug-resistant strain. Such epi- already colonized with VRE is 50% or greater, which demiological and clinical clues, which might prompt may explain some studies where newly introduced anti- empirical enterococcal therapy, are summarized in biotic control measures may only yield modest reduc- Table 4. tions in VRE colonization and infection rates in hyperendemic settings.41 Patients with comorbidities, including oncologic conditions, especially neutropenia, Does the Enterococcus in the Culture Result and prior solid organ transplantation, especially liver Require Antimicrobial Treatment? transplantation, appear to have the highest rates of Microbiological culture data that report the presence of VRE bacteremia and poorest outcomes. enterococci always require some level of clinical discrim- ination to determine whether they merit treatment. Enterococcal isolates from a respiratory specimen (spu- GENERAL ISSUES IN THE TREATMENT OF tum, endotracheal aspirate, bronchoalveolar lavage), and ENTEROCOCCAL INFECTION skin, wound, or mucosal surfaces almost always represent The treatment of serious enterococcal infection is chal- colonization. Urine cultures obtained via indwelling lenging from several aspects. Because enterococci may bladder (Foley) catheters often represent asymptomatic colonize skin, wound, and mucosal surfaces and their bacteriuria. Wound and intra-abdominal drains often isolation is often accompanied by more virulent patho- become colonized with skin flora, including enterococci.
  • 6. OPTIMIZING THERAPY FOR VANCOMYCIN-RESISTANT ENTEROCOCCI (VRE)/LINDEN 637 However, such isolates may be significant when the Table 5 Nonantimicrobial Interventions for character of the drainage fluid reveals evidence of Vancomycin-Resistant Enterococcal Infections inflammatory response (i.e., pyuria or purulence). Site of Infection Nonantimicrobial Intervention Although enterococci may be blood culture contami- Bloodstream Catheter removal nants, particularly when specimens are obtained from Consider endovascular infection if indwelling intravascular catheters, the appropriate clin- no primary source obvious or ical bias should be that such cultures represent true patient with risk factors for pathogens in most instances. Finally, simple nonantimi- endocarditis or other endovascular crobial interventions may obviate the need for antienter- material ococcal therapy, such as removal of intravascular or Closed-space Consider percutaneous or surgical bladder catheters or superficial wound debridement.46,47 infection drainage, e.g., cholangitis— percutaneous transhepatic drainage or endoscopic retrograde Is Bactericidal Therapy Required? cholangiopancreatography for The majority of enterococcal infections are not proven to stent placement require bactericidal treatment and can be managed Urinary tract infection Removal of bladder catheter successfully with a single effective agent.48,49 Either or bacteriuria native- or prosthetic-valve endocarditis is the prototyp- Superficial wound Incision and drainage or debridement ical enterococcal infection for which bactericidal anti- infection microbial therapy is required, usually achieved with the Suspected foreign May require removal if refractory combination of a cell wall–active agent such as ampicil- body infection to antimicrobial therapy lin, penicillin, or vancomycin combined with an amino- or adjacent to devitalized tissue glycoside such as gentamicin or streptomycin. Other sites of infection for which bactericidal treatment is probably merited include enterococcal meningitis and considerations is summarized in Table 5. It also bears enterococcemia in a neutropenic host. emphasis that the inability to address the nonantimicro- However, a bactericidal combination is not pos- bial considerations of such complex VRE infections has sible to achieve with enterococci exhibiting high-level been the principal cofactor leading to the development of aminoglycoside resistance and almost all strains of VRE strains that have evolved resistance to the recently E. faecium strains with high-level vancomycin resistance. approved VRE antimicrobials, quinupristin-dalfopristin Uncommon exceptions are vancomycin-resistant E. and linezolid. faecalis strains, which retain ampicillin and high-level gentamicin susceptibility. Successful treatment of such cases has been reported with ampicillin and gentamicin, Specific Antimicrobials for the Treatment of ampicillin þ ofloxacin, penicillin þ streptomycin, and Vancomycin-Resistant Enterococcal Infection linezolid þ gentamicin.50 Limited clinical experience Despite the established high prevalence of multidrug- is available for the treatment of vancomycin-resistant resistant enterococcal strains with high-level vancomy- enterococcal endocarditis with the newer agents (see cin-resistance there is a remarkable paucity of controlled, later discussion). comparative trial data on its antimicrobial treatment. Major obstacles have been the slow development of novel agents with VRE activity, high levels of comor- Nonantimicrobial Treatment of VRE Infection bidity that confound outcome interpretation, complex Many VRE infections may be partially or completely surgical infection for which antimicrobial therapy alone cured with conservative or aggressive nonantimicrobial is not curative, and the polymicrobial nature of many interventions. Less serious infections such as bladder VRE infections, particularly those occurring in the catheter–associated bacteriuria and urinary tract infec- abdomen. Both approved and nonapproved treatment tion may be adequately treated simply with catheter options for VRE are summarized in Table 6. At present removal. Postoperative superficial wound infections there are only two U.S. Food and Drug Administration may also respond to opening the incision and simple (FDA)-approved treatments for VRE (E. faecium) in- drainage or debridement. Closed-space infection such as fection: quinupristin/dalfopristin (Q/D, Synercid, King intra-abdominal abscesses, cholangitis due to biliary Pharmaceuticals, Inc., Bristol, TN) and linezolid (Zyvox, obstruction, devitalized tissue, or infected foreign bodies Pfizer, New York, NY) and two other approved agents (intravascular catheters, synthetic graft or mesh material, that have in vitro activity against VRE but are not prosthetics) are not infrequently the primary source of approved for VRE infection; daptomycin (Cubicin, VRE bacteremic or nonbacteremic infection. The treat- Cubist Pharmaceuticals, Lexington, MA), which is ap- ment implications for infections with such anatomical proved for complicated skin–skin structure infection,
  • 7. 638 SEMINARS IN RESPIRATORY AND CRITICAL CARE MEDICINE/VOLUME 28, NUMBER 6 2007 Table 6 Therapeutic Antimicrobial Options for VRE Infection Antimicrobial(s) Reported Evidence Comments High-dose ampicillin or Case reports May be effective with VRE strains ampicillin-sulbactam with ampicillin MIC 32–64 mg/mL Chloramphenicol Case series Resistance reported Tetracycline, doxycycline Case reports Æ rifampin or ciprofloxacin Novobiocin Anecdotal No longer manufactured Nitrofurantoin Small case series Only for urinary tract infections Teicoplanin Case reports Not active against VanAResistance in VanB reported Quinupristin/dalfopristin Large case series but noncomparative Bacteriostatic Not active against E. faecalis Resistance reported Linezolid 1. Dose comparative trial Bacteriostatic 2. Large compassionate use series Resistance reported Daptomycin 1. Case report þ series Bactericidal Resistance reported Tigecycline In vitro data only Bacteriostatic Dalbavancin In vitro data only VanA strains resistant Telavancin In vitro data only Oritavancin In vitro data only MIC, minimum inhibitory concentration. and S. aureus bacteremia and tigecycline (Tygacil, Wyeth been reported with the use of tetracycline, doxycycline, Pharmaceuticals, Inc., Philadelphia, PA), which is ap- and oral novobiocin combined with either ciprofloxacin or proved for complicated skin–skin structure and intra- doxycycline; however, such experience has never been rep- abdominal infection. roduced in larger clinical series of prospective trials.55–58 Prior to the availability of Q/D and linezolid Teicoplanin, a glycopeptide not commercially approval several centers published their experience with available in the United States, does have in vitro activity a variety of available agents or combinations that dem- versus VanB phenotypic enterococci. In a European onstrated in vitro activity. Clinical success was described study of 63 patients with vancomycin-susceptible enter- with high parenteral dosages of ampicillin or ampicillin/ ococcal infection, clinical and microbiological responses sulbactam (18 to 24 g/day), even including endocarditis. were observed in 84% and 87% of cases, respectively.59 Such a strategy appears limited to those uncommon VRE This agent remains unstudied for VanB enterococcal strains with ampicillin MICs of 32 to 64 mg/mL, a target infection, perhaps in part due to the development of range for which plasma ampicillin levels can be achieved teicoplanin resistance among VanB E. faecalis during with high dose therapy.50–52 Because no b-lactamase teicoplanin therapy.60,61 elaboration occurs with VRE, the mechanism of sulbac- Nitrofurantoin has in vitro activity against both tam activity is not known, although a plausible explan- VanA and VanB enterococci.62 Due to its ability to ation is its intrinsic penicillin-binding protein properties. achieve high urinary concentrations nitrofurantoin has Chloramphenicol has bacteriostatic activity against been shown to be effective in VRE urinary tract enterococci and VRE strains; however, its in vivo efficacy infection.46,63 Nitrofurantoin cannot be employed for was never established. In a retrospective study of 80 cases VRE outside the urinary tract and in patients with a of VRE bacteremia, 51 patients were treated with chlor- creatinine clearance < 30 mL/min because elevated amphenicol from which 22/36 (61%) evaluable patients blood concentrations are associated with hepatic, pul- demonstrated a clinical response.53 A microbiological monary, hematologic, and other toxicities. response was also observed in 33/43 (77%) of the micro- biologically evaluable patients. No survival benefit was QUINUPRISTIN/DALFOPRISTIN observed compared with VRE bacteremic patients in the Quinupristin/dalfopristin (Q/D) is a semisynthetic study cohort who did not receive chloramphenicol. Sub- parenteral streptogramin compound, which is derived sequently at the same center, the prevalence of chloram- from its parent natural compound pristinamycin, a phenicol resistance among VRE strains over a 10-year product of Streptomyces pristinaspiralis, an oral and top- period (1991 to 2000) were observed to increase from 0 to ical antistaphylococcal agent that has been in clinical use 11%, a trend that correlated significantly with prior in Europe since the 1980s. The major properties of this chloramphenicol or quinolone exposure.54 Isolated re- compound are summarized in Table 7. This antimicro- ports of favorable outcome for VRE infection have also bial is a 30:70 mixture of quinupristin and dalfopristin,
  • 8. OPTIMIZING THERAPY FOR VANCOMYCIN-RESISTANT ENTEROCOCCI (VRE)/LINDEN 639 Table 7 Major Features of Quinupristin/Dalfopristin and Linezolid Feature Quinupristin/ Dalfopristin Linezolid Antimicrobial class Streptogramn Oxazolidinone Peak serum concentrations (mg/L) 10-12 15.1 Elimination half-life (h) 0.8 (Q) , 0.6 (D) 5.5 Major metabolic routes Hepatobiliary Peripheral nonoxidative Major elimination routes Faecal (70-75%) Nonrenal (65%) Urinary (19%) Urinary (30%) Protein binding (%) 30 (Q) 70 (D) 31 Mechanism of action Protein synthesis inhibition Protein synthesis inhibition Site of action 50S ribosome 70S initiation complex Postantibiotic effect (h) 6–8 1 Bactericidal (vs VRE) No No Cytochrome P-450 inhibition Yes No Formulations Parenteral Parenteral þ oral Dose and administration 5–7.5 mg/kg q 8–12h 600 mg q12 h Dosage adjustment None None Approved indications VRE VRE Complicated SSSI Complicated SSSI Nosocomial pneumonia Nosocomial pneumonia Major adverse effects Phlebitis (peripheral) Myelosuppression Myalgia/arthralgia Cost ($US per day; 2000 values) $300-350 $115 (parenteral) $80 (oral) D, dalfopristin; Q, quinupristin; qXh, every X hours; SSSI, skin and skin structure infection; VRE, vancomycin-resistant enterocooci. (With permission from Linden.42) which are derivatives of streptogramin types B and A, Clinical interest in the utility of Q/D for serious respectively. It is a unique antimicrobial because it acts VRE infection began in the mid-1990s with a large- through sequential ribosomal binding and is internally scale, noncomparative, open-label, emergency use synergistic to produce a bactericidal effect. Dalfopristin program for multiresistant gram-positive infection, prin- initially binds to the 50S bacterial ribosome, which in- cipally vancomycin-resistant E. faecium and MRSA in- duces a permanent conformational change that acceler- fection refractory or intolerant to vancomycin.66,67 The ates quinupristin ribosomal binding.64 Protein synthesis patient populations in both series had a high prevalence is impaired via both the interruption of peptide chain of acute and chronic comorbidities, including diabetes, elongation and the inhibition of formed peptide extru- oncologic conditions, chronic liver disease, dialysis me- sion. SSuch synergism results in bactericidal activity chanical ventilation, and prior organ transplantation. against some important gram-positive species, including Q/D was administered at 7.5 mg/kg intravenously every Streptococcus pneumoniae, Streptococcus agalacticae, and 8 hours to patients with documented VRE bacteremia or some strains of Staphylococcus aureus. However, only nonbacteremic VRE infection, with the duration of bacteriostatic activity is present for the majority of E. treatment determined by the primary treating physi- faecium strains by time–killing curve studies. This effect is cians. The overall success rate defined as both clinical primarily mediated by 23S ribosomal modification success and bacteriologic eradication was 65.8% in the encoded for by the ermB gene (erythromycin methylase), initial study and 65.6% in the follow-up study. There which reduces quinupristin affinity for its ribosomal have been several reports of clinical cure combining Q/D binding site and thus limits activity to only the dalfopris- with doxycycline or high-dose ampicillin in endocarditis; tin moiety. Such strains are termed MLSb (macrolide- however, no larger-scale experience has been per- lincosamide-streptogramin) phenotypes.65 Erythromycin formed.68–70 resistance serves as an excellent surrogate marker for the As Q/D usage increased both before and after its presence of the MLSb phenotype among enterococci. regulatory approval in 1999, several important clinical Q/D is also unique as an antienterococcal agent based limitations became apparent. Peripheral intravenous upon its marked disparity in in vitro susceptibility be- administration was associated with a high rate of tween E. faecium (MIC90 ¼ 1 to 2 mg/mL) and E. faecalis phlebitis necessitating central venous administration. (MIC90 ¼ 8 to 16 mg/mL). This disparity is most likely Myalgia and arthralgia unassociated with objective due to altered ribosomal binding or presence of an active inflammatory signs were observed in 7 to 10% of patients efflux pump. in the emergency use program, with much higher rates in
  • 9. 640 SEMINARS IN RESPIRATORY AND CRITICAL CARE MEDICINE/VOLUME 28, NUMBER 6 2007 oncologic patients and liver transplant recipients.71,72 infection.77 Among 549 cases of VRE infection, there Although the precise reason for this toxicity is unknown was an 81.4% clinical cure rate at end-of-therapy. a neuropathic cause is suspected. Its higher incidence in Because linezolid is a bacteriostatic agent that displays populations with diminished metabolism and excretion no synergistic activity with other agents its efficacy in suggest it is due to either native drug or metabolite VRE native- or prosthetic-valve endocarditis remains accumulation. Phenotypic resistance to Q/D among questionable. Both clinical success and failure have been E. faecium (MIC ! 4 mg/mL) was observed in six reported when linezolid has been used as a first-line (1.8%) and five (1.3%) of VRE cases, either during or therapy or salvage treatment; however, no large-scale after treatment, from both published emergency use randomized trial experience is yet available.78–80 In series.71,72 Clonal dissemination of Q/D-resistant recent years, linezolid has become the dominant agent strains despite the absence of Q/D or other streptogra- for the treatment of serious VRE infection. Multiple min exposure has been described among pediatric pa- cases report of linezolid-resistance (MIC ! 8 mg/mL) tients.73 Three fundamental resistance mechanisms have occurring in VRE (E. faecium) and vancomycin-suscep- been discovered: enzymatic modification (acetylation) of tible E. faecalis strains that were susceptible (MIC 1 to dalfopristin encoded by the vatD or vatE genes, active 2 mg/mL) at baseline but developed a fourfold or greater efflux by an adenosine triphosphate (ATP)-binding rise in MIC to 8 to 32 mg/mL.81–85 Common to most protein encoded by the msrC or lsa genes, and alteration cases where linezolid-resistance appeared has been a of the ribosomal target site encoded for by the erm protracted length of therapy (> 28 days) associated genes.70 Because phenotypic resistance requires the with retained foci of VRE infection such as abscesses, presence of resistance mechanisms to both the quinu- devitalized tissue, or foreign materials. The majority of pristin and the dalfopristin components, at least two or linezolid-resistant isolates contain a single base-pair more resistance genes are present. Several surveillance mutation in the genome encoding for domain V of the studies have uncovered large Q/D resistance reservoirs 23S ribosomal binding site (G2476U mutation). The among E. faecium isolated from both domestic poultry phenotypic level of resistance as determined by elevation and livestock in the United States, which may relate to in MIC level has been shown to correlate with the ‘‘gene the use of virginiamycin as a growth-promoting food dose’’ or number of copies of 23S rDNA containing the additive in domestic poultry.74 G2466U mutation.86 Notably this mutation was pre- dicted by earlier in vitro spiral plate serial passage LINEZOLID experiments with linezolid.87 Horizontal cross trans- Linezolid is an oxazolidinone compound, a novel syn- mission of an identical clone of linezolid-resistant thetic class that inhibits bacterial protein synthesis in a E. faecium among linezolid-naive patients within the unique fashion via inhibiting the formation of the 70S same ICU or hospital center have also been de- initiation complex (50S and 30S ribosomes, mRNA, scribed.88,89 A case- controlled analysis revealed that a initiation factors 2 and 3, and fMet-tRNA).75 The major longer course of linezolid (38 days vs 11 days) and properties of linezolid are summarized in Table 6. Line- linezolid exposure prior to hospitalization were risk zolid exhibits a broad gram-positive spectrum but has factors for the emergence of linezolid-resistant VRE.90 only bacteriostatic activity against vancomycin-resistant Thus repeat linezolid susceptibility testing is advisable in or susceptible enterococci with an MIC90 of 2 mg/mL, patients who have had prior linezolid exposure or per- which is right at the susceptibility breakpoint. FDA sistent isolation of a VRE strain on therapy or in patients approval was granted in 2000 for vancomycin-resistant treated in a nosocomial setting with prior linezolid E. faecium infection in addition for other indications, resistance. Although gastrointestinal symptoms are the including community-acquired and nosocomial pneu- most common reported toxicity, reversible myelosup- monia and complicated skin and skin structure infection. pression (thrombocytopenia, leukopenia, and/or anemia) Due to the lack of an approved comparator agent, has been the most important treatment-limiting side linezolid was evaluated for patients with clinical and effect with higher rates observed than the original regis- microbiological evidence of serious VRE infection in a tration studies. Bone marrow examination has shown blinded, parenteral, dose-comparative trial comparing changes similar to those observed with reversible chlor- 66 patients randomized to 200 mg q 12 hours to amphenicol toxicity.91,92 Such toxicity is usually observed 79 patients treated with 600 mg q 12 hours.76 Among only with sustained linezolid treatment that exceeds evaluable patients at end-of-treatment, a modest dose 2 weeks. Other reported toxicities of note include response was observed, with 67% and 52% response rates gastrointestinal upset, rare cases of serotonin syndrome, seen in the high-dose and low-dose groups, respectively. optic- and peripheral neuropathy, and lactic acidosis.93–96 In addition, efficacy and safety were also demonstrated in a large study (n ¼ 796 patients) emergency-use DAPTOMYCIN program for resistant, or treatment-refractory, or treat- Daptomycin is a novel cyclic lipopeptide compound with ment-intolerant patients with serious gram-positive a broad gram-positive spectrum and rapid bactericidal
  • 10. OPTIMIZING THERAPY FOR VANCOMYCIN-RESISTANT ENTEROCOCCI (VRE)/LINDEN 641 activity that is currently approved for complicated tee for Clinical Laboratory Standards (NCCLS) break- skin–skin structure infection and S. aureus bacteremia, points for vancomycin-resistant E. faecium strains are not including right-sided endocarditis.97 Its apparent yet established.108,109 Although clinical experience with mechanism of action includes attachment to the exterior tigecycline for VRE infection is not yet available of the bacterial cytoplasmic membrane with membrane it appears to be a promising option, particularly for penetration of a lipophilic tail with disruption of the intra-abdominal sites, where it has shown comparable transmembrane potential due to ion efflux, an effect that efficacy to meropenem in non-VRE monomicrobial and is both concentration- and calcium ion–dependent and polymicrobial infection. leads to nonlytic bacterial cell death. In vitro studies have shown nearly uniform activity against vancomycin- NOVEL GLYCOPEPTIDES (ORITAVANCIN, DALBAVANCIN, resistant E. faecium and E. faecalis strains with an MIC TELAVANCIN) 90 of 2–4 mg/mL.98,99 In one recent study examining Several new glycopeptide derivatives have in vitro bac- only VRE strains that were either linezolid- or Q/D- tericidal activity against VRE. Dalbavancin is a long- resistant, daptomycin demonstrated susceptibility using acting (half-life 7 to 10 days) derivative of teicoplanin, a 4 mg/mL provisional breakpoint.100 The MIC break- which has received approval for the treatment of com- point is 4 mg/mL for vancomycin-susceptible E. faecalis; plicated skin–skin structure infection; however, similarly however, there is no established breakpoint for vanco- to teicoplanin, this agent lacks in vitro activity against mycin-susceptible or -resistant E. faecium. Regardless the more prevalent VanA enterococcal strains.110 of the testing method (E-test, disk diffusion, or broth Oritavancin is a semisynthetic glycopeptide that blocks dilution) the zone size or MIC result can be signifi- peptidoglycan synthesis and exerts bactericidal activity cantly elevated by a two- to eightfold magnitude with across a broad gram-positive spectrum.111 It has superior inadequate calcium supplementation. To the present activity against vanA and vanB enterococci compared time, clinical experience with daptomycin for serious with dalbavancin and telavancin, with concentration- VRE infection remains quite limited. The optimal dependent bactericidal activity against both E. faecium dosing for enterococcal infection is not yet established; and E. faecalis strains (MIC90 ¼ 1 to 2 mg/mL) and is however, daily dosing at 6 mg/kg in the absence of renal synergistic with ampicillin against the majority of iso- insufficiency has been the most common dosing scheme. lates.112 This agent has completed phase 3 trials in cSSSI A randomized phase 3 trial versus linezolid in VRE (complicated skin-skin structure infection); however, infection was aborted due to enrollment difficulties. concerns pertaining to its long half-life, high protein In a study of nine neutropenic patients with VRE binding, and reports of spontaneous resistance may bacteremia treated with daptomycin at 4 mg/kg/day limit its development. or 6 mg/kg/day, a clinical and/or microbiological re- Telavancin, a long-acting lipoglycopeptide with sponse was observed in only 4/9 (44%).101 In a second multiple sites of action at the cell membrane and cell report a similar response rate of 5/11 (45%) was wall has shown noninferiority versus standard therapy in observed in patients with VRE bacteremia and endo- gram-positive cSSSI, including MRSA; however, clin- carditis treated with 6 mg/kg/day of daptomycin.102 ical data for VRE are not yet available.113,114 Unfortunately, daptomycin resistance has been re- ported during treatment for vancomycin-resistant E. faecalis, E. faecium, and E. durans infection with a REFERENCES rise in the MIC to ! 8 mg/mL.103–106 1. Centers for Disease Control and Prevention. National Nosocomial Infection Surveillance (NNIS) system: report, TIGECYCLINE data summary from January 1992 through June 2003, issued Tigecycline is the first approved agent of the glycylcy- August 2003. Am J Infect Control 2003;31:481–498 cline class, a group closely related to the tetracyclines but 2. Wisplinghoff H, Bischoff T, Tallent SM, Seiferd H, Wenzel RP, Edmond MB. Nosocomial bloodstream synthetically modified to achieve an enhanced spectrum infections in US hospitals: analysis of 24,179 cases from a of activity against MRSA, other multiresistant gram- prospective nationwide surveillance study. Clin Infect Dis positive species, and many gram-negative bacilli.107 2004;39:309–317 Tigecycline is currently approved for complicated 3. Moellering RC Jr. The Garrod Lecture. The Enterococcus: a skin–skin structure and intra-abdominal infection based classic example of the impact of antimicrobial resistance on upon phase 3 studies showing comparability to standard therapeutic options. J Antimicrob Chemother 1991;28:1–12 comparator regimens. Vancomycin-resistant enterococci 4. Murray BE. The life and times of the Enterococcus. Clin Microbiol Rev 1990;3:46–65 were not included in these registration trials. 5. Hodges TL, Zighelboim-Daum S, Eliopoulos GM, However, daptomycin exhibits very low MICs for Wennersten C, Moellering RC Jr. Antimicrobial suscept- both vancomycin-susceptible and vancomycin-resistant ibility changes in Enterococcus faecalis following various versus clinical strains of E. faecium and E. faecalis penicillin exposure regimens. Antimicrob Agents Chemo- (MIC90 ¼ 0.012 mg/mL), although National Commit- ther 1992;36:121–125
  • 11. 642 SEMINARS IN RESPIRATORY AND CRITICAL CARE MEDICINE/VOLUME 28, NUMBER 6 2007 6. Hamilton-Miller JM, Purves D. Enterococci and antifolate 25. Centers for Disease Control and Prevention (CDC). antibiotics. Eur J Clin Microbiol 1986;5:391–394 Vancomycin-resistant Staphylococcus aureus—New York. 7. LeClercq R, Couvalin P. Bacterial resistance to macrolide, MMWR Morb Mortal Wkly Rep 2005;53:322–323 lincosamide and streptogramin antibiotics by target mod- 26. Miller D, Urdaneta V, Weltman A. Vancomycin-resistant ification. Antimicrob Agents Chemother 1991;35:1267– Staphylococcusaureus—Pennsylvania 2002. MMWR Morb 1272 Mortal Wkly Rep 2002;51:902 8. Lefort A, Mainardi JL, Tod M, Lotholary O. Antienter- 27. Sievert DM, Boulton ML, Stolzman G, et al. Staphylococcus ococcal antibiotics. Med Clin North Am 2000;84:1471– aureus resistant to vancomycin—United States 2002. 1495 MMWR Morb Mortal Wkly Rep 2002;51:565–567 9. Rybkine T, Mainardi JL, Sougakoff W, Collatz E, 28. Whitener CJ, Park SY, Browne FA, et al. Vancomycin- Gutmann L. Penicillin- binding protein 5 sequence resistant Staphylococcus aureus in the absence of vancomycin alterations in clinical isolates of Enterococcus faecium with exposure. Clin Infect Dis 2004;38:1049–1055 different levels of beta-lactam resistance. J Infect Dis 1998; 29. Donskey CJ. The role of the intestinal tract as a reservoir 178:159–163 and source for transmission of nosocomial pathogens. Clin 10. Grayson ML, Eliopoulos GM, Wennersten CB, et al. Infect Dis 2004;39:219–226 Increasing resistance to b-lactam antibiotics amongst 30. Beezhold DW, Slaughter S, Hayden MK, et al. Skin clinical isolates of Enterococcus faecium: a 22-year review at colonization with vancomycin-resistant enterococci among one institution. Antimicrob Agents Chemother 1991;35: hospitalized patients with bacteremia. Clin Infect Dis 1997; 2180–2184 24:704–706 11. Horodniceanu T, Bougueleret L, El-Solh N, Bieth G, 31. D’Agata EM, Gautam S, Green WK, Tang YW. High rate Delbos F. High level resistance to gentamicin in Streptococcus of false- negative results of the rectal swab culture method faecalis subsp. zymogenes. Antimicrob Agents Chemother in detection of gastrointestinal colonization with vanco- 1979;16:686–689 mycin-resistant enterococci. Clin Infect Dis 2002;34:167– 12. Mederski-Samoraj BD, Murray BE. High level resistance to 172 gentamicin in clinical isolates of enterococci. J Infect Dis 32. Weinstein JW, Tallapragada S, Farrel P, Dembry LM. 1983;147:751–757 Comparison of rectal and peri-rectal swabs for detection of 13. Spiegel CA, Hucyke M. Endocarditis due to streptomycin- colonization with vancomycin-resistant enterococci. J Clin susceptible Enterococcus faecalis with high-level gentamicin Microbiol 1996;34:210–212 resistance. Arch Intern Med 1989;149:1873–1875 33. Montecalvo MA, Lencastre H, Carraher M, et al. Natural 14. Eliopoulos GM. Aminoglycoside resistant enterococcal history of colonization with vancomycin-resistant Enter- endocarditis. Infect Dis Clin North Am 1993;7:117–133 ococcus faecium. Infect Control Hosp Epidemiol 1995;16: 15. Leclercq R, Dutka-Malen S, Brisson-Noel A, et al. 680–685 Resistance of enterococci to aminoglycosides and glycopep- 34. Baden LR, Thiemke W, Skolnik A, et al. Prolonged tides. Clin Infect Dis 1992;15:495–501 colonization with vancomycin-resistant Enterococcus faecium 16. Leclercq R, Derlot E, Duval J, Courvalin P. Plasmid in long-term care patients and the significance of ‘‘clear- mediated resistance to vancomycin and teicoplanin in ance.’’ Clin Infect Dis 2001;33:1654–1660 Enterococcus faecium. N Engl J Med 1988;319:157–161 35. Roghmann MC, Aquiyumi S, Schwalbe R, Morris JG Jr. 17. Centers for Disease Control and Prevention. Nosocomial Natural history of colonization with vancomycin-resistant enterococci resistant to vancomycin—United States 1989– Enterococcus faecium. Infect Control Hosp Epidemiol 1997; 1993. MMWR Morb Mortal Wkly Rep. 1993;42:597–599 18:679–680 18. Biedenbach DJ, Moet GJ, Jones RN. Occurrence and 36. Tornieporth NG, Roberts RB, Hafner JJ, Riley LW. Risk antimicrobial resistance pattern comparisons among blood- factors associated with vancomycin-resistant Enterococcus stream infection isolates from the SENTRY Antimicrobial faecium infection or colonization in 145 matched case and Surveillance Program (1997–2002). Diagn Microbiol Infect control patients. Clin Infect Dis 1996;23:767–772 Dis 2004;50:59–69 37. Bonten MJ, Hayden MK, Nathan C, et al. Epidemiology of 19. Lai KK, Fontecchio SA, Kelly AL, Baker S, Melvin ZS. colonization of patients and environment with vancomycin- The changing epidemiology of vancomycin-resistant enter- resistant enterococci. Lancet 1996;348:1615–1619 ococci. Infect Control Hosp Epidemiol 2003;24:264–268 38. Morris JG, Shay DK, Hebden JN, et al. Enterococci 20. Courvalin P. Vancomycin resistance in gram-positive cocci. resistant to multiple antimicrobial agents including vanco- Clin Infect Dis 2006;42:S25–S34 mycin, establishment of endemicity in a university medical 21. Handwerger S, Skoble J. Identification of chromosomal center. Ann Intern Med 1995;123:250–259 mobile element conferring high-level vancomycin resistance 39. Warren DK, Kollef MH, Seiler SM, Fridkin SK, Fraser VJ. in Enterococcus faecium. Antimicrob Agents Chemother The epidemiology of vancomycin-resistant Enterococcus in a 1995;39:2446–2453 medical intensive care unit. Infect Control Hosp Epidemiol 22. Gold HS. Vancomycin resistant enterococci: mechanisms 2003;24:257–263 and clinical observations. Clin Infect Dis 2001;33:210–219 40. Donskey CJ, Chowdhry T, Hecker M, et al. Effect of 23. Noble WC, Virani Z, Cree RG. Co-transfer of vancomycin antibiotic therapy on the density of vancomycin-resistant and other resistance genes from Enterococcus faecalis NCTC enterococci in the stool of colonized patients. N Engl J Med 12201 to Staphylococcus aureus. FEMS Microbiol Lett 1992; 2000;343:1925–1932 72:195–198 41. Bonten MJ, Slaughter S, Ambergen AW, et al. The role of 24. Chang S, Sievert DM, Hageman JC, et al. Infection with ‘‘colonization pressure’’ in the spread of vancomycin- vancomycin- resistant Staphylococcus aureus containing the resistant enterococci: an important infection control varia- vanA resistance gene. N Engl J Med 2003;348:1342–1347 ble. Arch Intern Med 1998;158:1127–1132
  • 12. OPTIMIZING THERAPY FOR VANCOMYCIN-RESISTANT ENTEROCOCCI (VRE)/LINDEN 643 42. Linden P. Can enterococcal infections initiate sepsis 59. Taylor SE, Paterson DL, Yu VL. Treatment options for syndrome? Curr Infect Dis Rep 2003;5:372–378 chronic prostatitis due to vancomycin-resistant Enterococcus 43. Kollef MH, Sherman G, Ward S, Fraser VJ. Inadequate faecium. Eur J Clin Microbiol Infect Dis 1998;17:798–800 antimicrobial treatment of infections: a risk factor for 60. Schmit JL. Efficacy of teicoplanin for enterococcal infec- hospital mortality among critically ill patients. Chest 1999; tions: 63 cases and review. Clin Infect Dis 1992;15:302–306 115:462–472 61. Aslangul E, Baptista M, Fantin B, et al. Selection of 44. Kumar A, Robert D, Wood KE, et al. Duration of glycopeptide-resistant mutants of VanB type Enterococcus hypotension before initiation of effective antimicrobial faecalis BM4281 in vitro and in experimental endocarditis. therapy is the critical determinant of survival in human J Infect Dis 1997;175:598–605 septic shock. Crit Care Med 2006;34:1589–1596 62. Hayden MK, Trenholme GM, Schultz JE, et al. In vivo 45. Ibrahim EH, Sherman G, Ward S, Fraser VJ, Kollef MH. development of teicoplanin resistance in a VanB Enterococcus The influence of inadequate antimicrobial treatment of faecalis. J Infect Dis 1993;167:1224–1227 bloodstream infections on patient outcomes in the ICU 63. Zhanel GG, Hoban DJ, Karlowsky JA. Nitrofurantoin is setting. Chest 2000;118:146–155 active against vancomycin-resistant enterococci. Antimicrob 46. Quale J, Landman D, Atwood E, et al. Experience with a Agents Chemother 2001;45:324–326 hospital-wide outbreak of vancomycin-resistant enterococci. 64. Linden P, Coley K, Kusne S. Bacteriologic efficacy of Am J Infect Control 1996;24:372–379 nitrofurantoin for the treatment of urinary tract infection 47. Lai KK. Treatment of vancomycin resistant Enterococcus due to vancomycin-resistant Enterococcus faecium. Clin Infect faecium infections. Arch Intern Med 1996;156:2579–2584 Dis 1999;29:999 48. Pankey GA, Sabath LD. Clinical relevance of bacteriostatic 65. Cocito C, DiGiambattista M, Nyssen E, Vannuffel P. versus bactericidal mechanisms of action in the treatment of Inhibition of protein synthesis by streptogramins and related gram-positive bacterial infections. Clin Infect Dis antibiotics. J Antimicrob Chemother 2007;39(Suppl A): 2004;38:864–870 7–13 49. Finberg RW, Moellering RC, Tally FP, et al. The 66. Hershberger E, Donabedian S, Konstantinou K, Zervos MJ. importance of bactericidal drugs, future directions in Quinupristin-dalfopristin resistance in gram-positive bac- infectious diseases. Clin Infect Dis 2004;39:1314–1320 teria: mechanism of resistance and epidemiology. Clin Infect 50. Tsigrelis C, Singh KV, Coutinho TD, Murray BE, Baddour Dis 2004;38:92–98 LM. Vancomycin-resistant Enterococcus faecalis endocarditis: 67. Moellering RC, Linden PK, Reinhardt J, et al. The efficacy linezolid failure and strain characterization of virulence and safety of quinupristin/dalfopristin for the treatment of factors. J Clin Microbiol 2007;45:631–635 vancomycin-resistant Enterococcus faecium. J Antimicrob 51. Mekonen ET, Noskin GA, Hacek DM, Peterson LR. Chemother 1999;44:251–271 Successful treatment of persistent bacteremia due to vanco- 68. Linden PK, Moellering RC, Wood CA, et al. Treatment of mycin resistant ampicillin resistant Enterococcus faecium. vancomycin-resistant Enterococcus faecium infections with Microb Drug Resist 1995;1:249–253 quinupristin/dalfopristin. Clin Infect Dis 2001;33:1816– 52. Dodge RA, Daly JS, Davaro R, Glew RH. High-dose 1823 ampicillin plus streptomycin for treatment of a patient with 69. Brown J, Freeman BB. Combining quinupristin/dalfopristin severe infection due to multi-drug resistant enterococci. with other agents for resistant infections. Ann Pharmac- Clin Infect Dis 1997;25:1269–1270 other 2004;38:677–685 53. Murray BE. Vancomycin-resistant enterococcal infections. 70. Bethea JA, Walko CM, Targos PA. Treatment of vancomy- N Engl J Med 2000;342:710–721 cin-resistant Enterococcus with quinupristin/dalfopristin and 54. Lautenbach E, Schuster MG, Biler WB, et al. The role of high dose-ampicillin. Ann Pharmacother 2004;38:989–91 chloramphenicol in the treatment of bloodstream infection 71. Thompson RL, Lavin B, Talbot GH. Endocarditis due to due to vancomycin-resistant Enterococcus. Clin Infect Dis vancomycin-resistant Enterococcus faecium in an immuno- 1998;27:1259–1265 compromised patient: cure by administering combination 55. Gould CV, Fishman NO, Nakamkin NI, Lautenbach E. therapy with quinupristin/dalfopristin and high-dose ampi- Chloramphenicol resistance in vancomycin resistant enter- cillin. South Med J 2003;96:818–820 ococcal bacteremia: impact of prior fluoroquinolone use. 72. Raad I, Hachem R, Hanna H, et al. Treatment of Infect Control Hosp Epidemiol 2004;25:138–145 vancomycin-resistant enterococcal infections in the immu- 56. Howe RA, Robson M, Oakhill A, et al. Successful use of nocompromised host: quinupristin-dalfopristin in combina- tetracycline as therapy of an immunocompromised patient tion with minocycline. Antimicrob Agents Chemother with septicemia caused by a vancomycin-resistant Enterococcus. 2001;45:3202–3204 J Antimicrob Chemother 1997;40:144–145 73. Olsen KM, Rebuck JA, Rupp ME. Arthralgias and myalgias 57. Linden PK, Pasculle AW, Manez R, et al. Utilization of related to quinupristin-dalfopristin administration. Clin novobiocin and ciprofloxacin for the treatment of serious Infect Dis 2001;32:e83–e86 infection due to vancomycin-resistant Enterococcus faecium. 74. Werner G, Klare I, Spencker FB, Witte W. Intra-hospital In: Program and Abstracts of the Thirty-Third Interscience dissemination of quinupristin-dalfopristin- and vancomy- Conference on Antimicrobial Agents and Chemotherapy, cin-resistant Enterococcus faecium in a paediatric ward of a New Orleans, LA. Abstract 1027. Washington, DC: German hospital. J Antimicrob Chemother 2003;52:113– American Society for Microbiology; 1993:307 115 58. Montecalvo MA, Horowitz H, Wormser GP, et al. Effect 75. Donabedian SM, Perri MB, Vager D, et al. Quinupristin- of novobiocin containing antimicrobial regimens on infec- dalfopristin resistance in Enterococcus faecium isolates from tion and colonization with vancomycin resistant Enterococcus humans, farm animals, and grocery store meat in the United faecium. Antimicrob Agents Chemother 1995;39:794 States. J Clin Microbiol 2006;44:3361–3365
  • 13. 644 SEMINARS IN RESPIRATORY AND CRITICAL CARE MEDICINE/VOLUME 28, NUMBER 6 2007 76. Moellering RC. Linezolid: the first oxazolidinone anti- 92. Green SL, Maddox JC, Huttenbach ED. Linezolid and microbial. Ann Intern Med 2003;138:135–142 reversible myelosuppression. JAMA 2001;285:1291 77. Pharmacia and Upjohn. Linezolid for the Treatment of 93. Halpern M. Linezolid-induced pancytopenia. Clin Infect Vancomycin Resistant Enterococcal Infections: A Double- Dis 2002;35:347–348 Blind Trial Comparing 600 mg Linezolid Every 12 Hours 94. Wigen CL, Goetz MB. Serotonin syndrome and linezolid. with 200 mg Linezolid Every 12 Hours (study report M/ Clin Infect Dis 2002;34:1651–1652 1260/0054A). Peapack, NJ: Pharmacia Upjohn; 1999 95. Saijo T, Hayashi K, Yamada H, Wakakura M. Linezolid- 78. Birmingham MC, Raynes CR, Meagher AK, Flavin SM, induced optic neuropathy. Am J Ophthalmol 2005;139: Batts DH, Schentag JJ. Linezolid for the treatment of 1114–1116 multidrug-resistant gram-positive infections: experience 96. Bressler AM, Zimmer SM, Gilmore JL, Somani J. Peripheral from a compassionate use program. Clin Infect Dis 2003; neuropathy associated with prolonged use of linezolid. Lancet 36:159–168 Infect Dis 2004;4:528–531 79. Stevens MP, Edmond MB. Endocarditis due to vancomy- 97. Apodaca AA, Rakita RM. Linezolid-induced lactic acidosis. cin-resistant enterococci: case report and review of the N Engl J Med 2003;348:86–87 literature. Clin Infect Dis 2005;41:1134–1142 98. Carpenter CF, Chambers HF. Daptomycin: another novel 80. Archuleta S, Murphy B, Keller MJ. Successful treatment of agent for treating infections due to drug-resistant gram- vancomycin-resistant Enterococcus faecium endocarditis with positive pathogens. Clin Infect Dis 2004;38:994–1000 linezolid in a renal transplant recipient with human immuno- 99. Pfaller MA, Sader HS, Jones RN. Evaluation of the in vitro deficiency virus infection. Transpl Infect Dis 2004;6:117–119 activity of daptomycin against 19615 clinical isolates of 81. Babcock HM, Ritchie DJ, Christiansen E, Starlin R, Little gram positive cocci collected in North American hospitals R, Stanley S. Successful treatment of vancomycin-resistant (2002–2005). Diagn Microbiol Infect Dis 2007;57:459– Enterococcus endocarditis with oral linezolid. Clin Infect Dis 465 2001;32:1373–1375 100. Jorgensen JH, Crawford SA, Kelly CC, Patterson JE. 82. Zimmer SM, Caliendo AM, Thigpen MC, Somani J. In vitro activity of daptomycin against vancomycin-resistant Failure of linezolid treatment for enterococcal endocarditis. enterococci of various Van types and comparison of Clin Infect Dis 2003;37:e29–e30 susceptibility testing methods. Antimicrob Agents Chemo- 83. Tsigrelis C, Singh KV, Coutinho TD, Murray BE, Baddour ther 2003;47:3760–3763 LM. Vancomycin-resistant Enterococcus faecalis endocarditis: 101. Anastasiou DM, Thorne GM, Luperchio SA, Alder JD. linezolid failure and strain characterization of virulence In vitro activity of daptomycin against clinical isolates with factors. J Clin Microbiol 2007;45:631–635 reduced susceptibility to linezolid and quinupristin/dalfo- 84. Gonzales RD, Schreckenberger PC, Graham MB, et al. pristin. Int J Antimicrob Agents 2006;28:385–388 Infections due to vancomycin-resistant Enterococcus faecium 102. Poutsiaka DD, Skiffingeron S, Miller KB, Hadley S, resistant to linezolid. Lancet 2001;357:1179 Snydman DR. Daptomycin in the treatment of vancomy- 85. Seedat J, Zick G, Klare I, et al. Rapid emergence of cin-resistant Enterococcus faecium bacteremia in neutropenic resistance to linezolid during linezolid therapy of an patients. J Infect 2007;54:567–571 Enterococcus faecium infection. Antimicrob Agents Chemo- 103. Segreti JA, Crank CW, Finney MS. Daptomycin for the ther 2006;50:4217–4219 treatment of gram-positive bacteremia and infective endo- 86. Marra AR, Major Y, Edmond MD. Central venous catheter carditis: a retrospective case series of 31 patients. Pharma- colonization by linezolid-resistant, vancomycin-susceptible cotherapy 2006;26:347–352 Enterococcus faecalis. J Clin Microbiol 2006;44:1915–1916 104. Long JK, Choueiri TK, Hall GS, Avery RK, Sekeres MA. 87. Rahim S, Pillai SK, Gold HS, Venkataraman L, Inglima K, Daptomycin-resistant Enterococcus faecium in a patient with Press RA. Linezolid-resistant, vancomycin-resistant Enter- acute myeloid leukemia. Mayop Clin Proc 2005;80: ococcus faecium infection in patients without prior exposure to 1215–1216 linezolid. Clin Infect Dis 2003;36:E146–148 105. Munoz-Price LS, Lolans K, Quinn JP. Emergence of 88. Swaney SM, Shinabarger DL, Schaadt RD, Bock JH, resistance to daptomycin during treatment of vancomycin- Slightom JL, Zurenko GE. Oxazolidinone resistance is resistant Enterococcus faecalis infection. Clin Infect Dis 2005; associated with a mutation in the peptidyl transferase region 41:565–566 of 23S rRNA [abstract C-104]. In: Program and Abstracts 106. Lewis JS II, Owens A, Cadena J, et al. Emergence of of the 38th Interscience Conference on Antimicrobial daptomycin resistance in Enterococcus faecium during dapto- Agents and Chemotherapy; San Diego. Washington, DC: mycin therapy. Antimicrob Agents Chemother 2005;49: American Society for Microbiology; 1998:98–99 1664–1665 89. Herrero IA, Issa NC, Patel R. Nosocomial spread of 107. Green MR, Anasetti C, Sandin RL, Rolfe NE, Greene JN. linezolid-resistant vancomycin-resistant Enterococcus faecium. Development of daptomycin resistance in a bone marrow N Engl J Med 2002;346:867–869 transplant patient with vancomycin-resistant Enterococcus 90. Marshall SH, Donskey CJ, Hutton-Thomas R, Salata RA, durans. J Oncol Pharm Pract 2006;12:179–181 Rice LB. Gene dosage and linezolid resistance in Enter- 108. Stein GE, Craig WA. Tigecycline: a critical analysis. Clin ococcus faecium and Enterococcus faecalis. Antimicrob Agents Infect Dis 2006;43:518–524 Chemother 2002;46:3334–3336 109. Sader HS, Jones RN, Stilwell MG, Dowzicky MJ, Fritsche 91. Pai MP, Rodvold KA, Schreckenbefger PC, Gonezales RD, TR. Tigecycline activity tested against 26,474 bloodstream Petrolatti JM, Quinn JP. Risk factors associated with the infection isolates: a collection from 6 continents. Diagn development of infection with linezolid- and vancomycin- Microbiol Infect Dis 2005;52:181–186 resistant Enterococcus faecium. Clin Infect Dis 2002;35: 110. Streit JM, Sader HS, Fritsche TR, Jones RN. Dalbabancin 1269–1272 activity against selected populations of antimicrobial resistant
  • 14. OPTIMIZING THERAPY FOR VANCOMYCIN-RESISTANT ENTEROCOCCI (VRE)/LINDEN 645 gram-positive pathogens. Diagn Microbiol Infect Dis 113. Stryjewski ME, Chu VH, O’Riordan WD, et al. Telavancin 2005;53:307–310 versus standard therapy for treatment of complicated skin 111. Barrett JF. Recent developments in glycopeptide antibacte- and skin structure infections caused by gram-positive rials. Curr Opin Investig Drugs 2005;6:781–790 bacteria: FAST 2 Study. Antimicrob Agents Chemother 112. Baltch AL, Smith RP, Ritz WJ, Bopp LH. Comparison of 2006;50:862–867 inhibitory and bactericidal activities and postantibiotic 114. Poulakou G, Giamarellou H. Investigational treatments for effects of LY-333328 and ampicillin used singly and in postoperative surgical site infections. Expert Opin Investig combination against Enterococcus faecium. Antimicrob Agents Drugs 2007;16:137–155 Chemother 1998;42:2564–2568