Enterococcus species

Authors: Marcus J. Zervos, M.D.,  Joseph W. Chow, M.D.,  Anne Chen, M.D.,  Robert R. Muder, M.D. Updated October, 2010

MICROBIOLOGY

Enterococcus species are gram-positive, facultative anaerobic cocci that are morphologically similar to streptococci on Gram stain (181). The normal habitat of these microorganisms is the gastrointestinal tract of human and other mammals, although they can be isolated from the oropharynx, female genital tract, and skin. Most recently, 36 species of enterococci have been described, however 26 have been associated with human infection (65, 87, 225). Enterococcus faecalis is the most common human pathogen, but Enterococcus faecium has become increasingly prevalent in hospital-acquired infections. All the other enterococcal species together constitute less than 5% of enterococcal infections (87,225). These other species associated with human infections include Enterococcus gallinarum, Enterococcus casseliflavus, Enterococcus avium, Enterococcus cecorum, Enterococcus durans, Enterococcus hirae, Enterococcus malodoratus, Enterococcus mundtii, Enterococcus pseudoavium and Enterococcus raffinosus (49, 65, 87, 95,110,163, 203, 208,270).

back to top

EPIDEMIOLOGY

Enterococci are not as virulent as other gram-positive cocci and often occur as a component of a polymicrobial infection in debilitated hosts. Their increasing resistance to antimicrobial agents has led to their emergence as superinfecting nosocomial pathogens in patients receiving broad spectrum antibiotic therapy (79, 272, 286).

Vancomycin resistant enterococci (VRE) occur in both acute and long term care settings. When first introduced into a facility, an outbreak of VRE infection may occur. In acute outbreaks, the VRE isolates typically represent a single clone, or a limited number of clones, as identified by pulsed-field gel electrophoretic analysis of bacterial DNA (27, 66). After endemicity has become established, multiple clones of VRE are usually present (173, 176). A meta-analysis has reported that vancomycin resistance is an independent risk factor for mortality in patients with enterococcal bacteremia (51).

Patients with asymptomatic colonization of the gastrointestinal tract by VRE exceed those with clinically recognized infection by a ratio of 10:1 or greater (27). Thus, there is a potentially large reservoir of patients harboring VRE who may be the source of transmission to other patients. Unless these colonized patients are identified by appropriate surveillance procedures, they can continue to serve as reservoirs of VRE.

Risk factors for acquisition of or infection with VRE include admission to a critical care unit, severity of illness, exposure to other patients with VRE, duration of hospitalization, and exposure to antimicrobials (35, 58, 101,176, 263,272, 283). Antimicrobials exposures epidemiologically linked to VRE acquisition include vancomycin, cephalosporins, quinolones and agents with anti-anaerobic activity (29, 58, 76, 79,100,101,148,173, 201, 248). Receipt of the latter group of agents leads to a marked increase in stool density of VRE as well (53, 57,249). It should be noted that a meta-analysis concluded that many prior studies finding an association between receipt of vancomycin and VRE suffered from a number of flaws including selection of an inappropriate control group and failure to control for duration of hospitalization (30, 100). When these factors were taken into account, the association between VRE acquisition and antecedent vancomycin exposure was not statistically significant in this study.

Evidence is clear that with the consumption of antimicrobial agents that lack activity in vitro to enterococci administered to humans and food animals there is an association with an increase in antimicrobial resistance to that agent. Evidence suggests a relationship between streptogramin, aminoglycoside and glycopeptide resistance genes between humans and animals (56, 104, 105, 112). The degree of risk to humans related to the administration to animals of antimicrobial agents remains very controversial (112).

back to top

CLINICAL MANIFESTATION

Urinary tract infections and bacteremia are among the most common manifestations of enterococcal infection. Endocarditis is the most serious infection caused by enterococci, although it consists of only about 3% of enterococcal bacteremias (273). Enterococci are also commonly found in intraabdominal, pelvic, wound, and soft tissue infections, but in these cases they are often part of a mixed infection due to aerobic and anaerobic bacteria. Much less common are enterococcal meningitis, osteomyelitis, and septic arthritis.

LABORATORY DIAGNOSIS

Enterococci are readily and quickly identified in the clinical microbiology laboratory. On gram stainthey appear as gram-positive cocci in pairs and short chains. On blood agar plates they appear as grey colonies and are usually alpha-hemolytic. A rapid biochemical test can rapidly identify colonies of enterococci within minutes based on the ability of almost all enterococcal species to hydrolyze pyrrolidonyl-beta-naphthylamide (PYR). As all enterococci produce leucine aminopeptidase, this test is used on some rapid streptococcal identification panels. Other older tests that are used less frequently include the bile-esculin test, growth on broth containing 6.5% NaCl and ability to grow at both 10oC and 45oC. For identification of newer species of enterococci, a combination of conventional biochemical tests, and evaluation of DNA content is needed.

back to top

PATHOGENESIS

Enterococci are less virulent than many other bacteria that cause infections in humans. Some E. faecalis factors such as cytolysin (“hemolysin”), aggregation substance, gelatinase, serine protease, enterococcal surface protein, Ace protein, and enterococcal polysaccharide antigen, which have been associated with virulence in vitro and in animal models (85, 273). Some of these traits may play a pathogenic role in certain human infections, but this has not yet been proven conclusively.

SUSCEPTIBILITY IN VITRO AND IN VIVO

Enterococci are relatively resistant to many antibiotics that are active against gram positive cocci including cephalosporins, macrolides, and clindamycin. Antibiotics with varying degrees of in vitro activity against enterococci include the penicillins (especially penicillin, ampicillin, and piperacillin), glycopeptides (vancomycin and teicoplanin), carbapenems (imipenem and meropenem), aminoglycosides, tetracyclines (tetracycline and doxycycline), quinolones (including ciprofloxacin, moxifloxacin, gatifloxacin, and gemifloxacin), chloramphenicol, rifampin, the streptogramin combination quinupristin/dalfopristin and the oxazolidinone linezolid. The penicillins and the glycopeptides have the best activity, and ampicillin typically has greater in vitro killing ability than vancomycin (37). Enterococci have intrinsic low-level resistance to the aminoglycosides due to the decreased ability of these agents to penetrate the cell wall, but this can be overcome by the addition of cell wall-active agents (such as the penicillins and glycopeptides) that result in synergistic killing of the organisms (171). Antimicrobial agents undergoing development such as the oritavancin and dalbavancin, both of which are new glycopeptides, the lipoglycopeptide telavancin, iclaprim, which is a diaminopyrimidine, the glycylcycline tigecycline, also exhibit in vitro activity against enterococci (7, 11, 76, 82, 114, 116, 138, 180, 200, 213, 230, 234, 236).

In Vitro and In Vivo (Animal Data) Activity for Aminoglycoside-Resistant Enterococci

High-level gentamicin resistance (MICs > 500 - 2,000 mg/ml) in enterococci is usually due to the presence of the “bifunctional” aminoglycoside-modifying enzyme encoded by the aac(6’)-Ie-aph(2”)-Ia gene (9, 69). This enzyme modifies the aminoglycoside and eliminates synergism between a cell wall-active agent (such as ampicillin or vancomycin) and gentamicin, plus essentially all clinically available aminoglycosides except streptomycin. High-level streptomycin resistance can be caused by the streptomycin-modifying enzymes ANT(6’)-Ia or ANT(3”)-Ia or by a change in the ribosome binding site, and also eliminates synergism with the cell wall-active agents (43a, 62, 128, 199). Nosocomial isolates that possess genes for high-level resistance to both gentamicin and streptomycin are not uncommon (45, 87, 276).

In an in vitro study, both vancomycin and teicoplanin were bacteriostatic and not bactericidal against E. faecalis with high-level gentamicin resistance, and antagonism was often seen when either agent was combined with ciprofloxacin (266). In the same study, ampicillin alone was bactericidal against 8 of 13 E. faecalis, and ampicillin plus ciprofloxacin was bactericidal against all 13 E. faecalis strains (266). In a rabbit endocarditis model with a beta-lactamase-producing, high-level gentamicin resistant E. faecalis strain, ampicillin/sulbactam reduced bacterial titers in vegetations more effectively than vancomycin after 7 days, although neither regimen could sterilize the heart valve (134).

The combination of amoxicillin plus cefotaxime can exhibit an in vitro bacteriostatic synergistic effect against amoxicillin-susceptible E. faecalis isolates. This could be explained by the partial saturation of penicillin-binding proteins 2 and 3 by cefotaxime combined with the total saturation of penicillin-binding proteins 4 and 5 by amoxicillin (152). The combination of ampicillin plus ceftriaxone in time-kill studies produced synergistic killing against 10 of 10 ampicillin-susceptible E. faecalis isolates. Furthermore, ampicillin plus ceftriaxone in experimental E. faecalis rabbit endocarditis significantly lowered bacterial titers in aortic valve vegetations compared to ampicillin alone (83). However, no clinical data are available to confirm the superiority of this combination over ampicillin alone for infections caused by ampicillin-susceptible E. faecalis.

Although high-level aminoglycoside resistance is synonymous with resistance to synergism, there are rare enterococci with gentamicin MICs < 500 µg/ml that are also resistant to the synergistic effect of a cell wall-active agent plus gentamicin. Moellering and colleagues have detected several E. faecalis with gentamicin MICs of only 8 µg/ml that are resistant to ampicillin-gentamicin synergism but susceptible to ampicillin-tobramycin synergism (170). Hayden and colleagues have reported five E. faecium with gentamicin MICs between 4 and 32 µg/ml that are resistant to ampicillin-gentamicin synergism (103). The mechanisms of these resistance phenotypes have not yet been determined. A new gentamicin resistance gene, aph(2”)-Ic, has been found in E. faecalis, E. faecium, and E. gallinarum isolates (40). This gene compromises ampicillin-gentamicin synergism but confers a gentamicin MIC of typically only 256 µg/ml. If these three enterococcal phenotypes become more prevalent, synergy testing of a cell wall-active agent combined with gentamicin may be indicated in selected clinical situations, such as enterococcal endocarditis or meningitis, to confirm the efficacy of the antimicrobial combination utilized for therapy (43).

Heretofore, only gentamicin and streptomycin have been used for high-level aminoglycoside testing in enterococci, since the aac(6’)-Ie-aph(2”)-Ia gene confers high-level resistance to essentially all the clinically available aminoglycosides (including gentamicin, amikacin, tobramycin, netilmicin, and kanamycin) except for streptomycin. A second gene, aph(2”)-Id, that confers high-level gentamicin resistance has been detected (267). This gene also confers resistance to tobramycin, netilmicin, and kanamycin, but not to amikacin. Studies performed on the E. casseliflavus from which the gene was isolated showed synergistic killing with the combination of ampicillin and amikacin (267). However, ampicillin plus amikacin did not achieve as much killing against several E. faecium isolates that possess the aph(2”)-Id gene (267). Therefore, only a small percentage of enterococcal isolates with high-level gentamicin resistance might be susceptible to the combination of a cell wall-active agent plus amikacin.

The new aminoglycoside arbekacin is modified to a lesser degree than gentamicin is by the bifunctional AAC(6’)-Ie-APH(2”)-Ia enzyme (111). Ampicillin plus arbekacin produced in vitro synergistic killing against 40% of enterococci that possess the aac(6’)-Ie-aph(2”)-Ia gene (123). Compared to ampicillin alone, ampicillin plus arbekacin was significantly more effective in reducing bacteria in vegetations in a rabbit infective endocarditis model caused by an E. faecalis isolate that contained aac(6’)-Ie-aph(2”)-Ia (120). Ampicillin plus arbekacin has also produced in vitro synergistic killing against 8 of 13 vancomycin-, ampicillin-, and gentamicin-resistant E. faecium that possess the aph(2”)-Id gene (121).

In Vitro and In Vivo (Animal Data) for Enterococci Resistant to Penicillins and Glycopeptides, But Susceptible to Aminoglycosides

High-level penicillin resistance in enterococci is due predominantly to over-expression of penicillin-binding protein 5 (PBP 5), which has low affinity for the penicillins, but is able to substitute for the functions of the susceptible penicillin-binding proteins when they are inhibited by the beta-lactam agents (2, 71, 91, 141,142, 241). For an E. faecium strain resistant to penicillin (MIC = 200 µg/ml) but susceptible to gentamicin, no in vitro synergistic bactericidal activity was observed when penicillin was combined with gentamicin, and the combination did not significantly lower bacterial counts in vegetations in a rat endocarditis model when compared with no therapy (26). However, Torres and colleagues have found in vitro synergism when high levels of penicillin are combined with gentamicin against some E. faecium isolates with high-level penicillin resistance (MIC > 128 µg/ml), but low-level gentamicin resistance. Synergism was exhibited in nine of twelve strains (penicillin MIC = 200 µg/ml) when penicillin at 100 µg/ml was combined with gentamicin (5 µg/ml) (264). In addition, synergism was seen in all three strains for which the penicillin MIC was 400 µg/ml when penicillin at 200 µg/ml was combined with gentamicin at 5 µg/ml (264).

Glycopeptide resistance in enterococci is due the synthesis of modified peptidoglycan precursors that have decreased affinity for vancomycin and teicoplanin (10,136). Most glycopeptide-resistant clinical isolates are of the VanA or VanB phenotype, although VanA to VanG phenotypes have been described. Strains with the VanA phenotype have high-level resistance to both vancomycin (MICs = 64 - >1024 µg/ml) and teicoplanin (MICs = 16 - 512 µg/ml), while strains with the VanB phenotype have varying levels of resistance to vancomycin (MICs = 4 - 1024 µg/ml), but are susceptible to teicoplanin (10, 136). Heteroresistance to vancomycin has also been identified (3). E. gallinarum, E. casseliflavus, and E. flavescens strains are intrinsically resistant to vancomycin (MICs = 4 - 32 µg/ml), but remain susceptible to teicoplanin, and are of the VanC phenotype (136, 188, 223). Teicoplanin appears to be effective therapy against enterococcal strains lacking glycopeptide resistance determinants. In a rat endocarditis model, teicoplanin was actually more efficacious than vancomycin against a beta-lactamase-producing strain with high-level gentamicin resistance (281).

Teicoplanin’s efficacy for the treatment of VanB-type VRE is not as promising. Teicoplanin plus gentamicin produced in vitro bactericidal synergism against an E. faecalis resistant to vancomycin but susceptible to teicoplanin and gentamicin (12). However, teicoplanin resistance can be selected for in vitro with both E. faecalis and E. faecium strains (102), and teicoplanin-resistant E. faecalis mutants have arisen during teicoplanin therapy in a rabbit endocarditis model (12). The in vivo emergence of resistance to teicoplanin during vancomycin therapy for E. faecium (VanB phenotype) bacteremia has also been observed (102).

For strains truly resistant to ampicillin, vancomycin, and teicoplanin but still susceptible to aminoglycosides, several options have been suggested. The combination of ciprofloxacin plus rifampin plus gentamicin substantially reduced bacterial counts in vegetations in a rat endocarditis model (277). This triple antimicrobial combination has been used with success in two cases of enterococcal bacteremia (147). Ciprofloxacin plus gentamicin, ciprofloxacin plus rifampin, and ciprofloxacin plus rifampin plus gentamicin all showed bactericidal activity in time-kill studies (147). The activity of these combinations is related to the intrinsic activity of the fluoroquinolone. The newer quinolones with greater potency in vitro than ciprofloxacin against enterococci, such as moxifloxacin and gatifloxacin, may prove in the future to be better therapeutic choices than ciprofloxacin. Unfortunately, fluoroquinolone resistance in clinical enterococcal isolates is not uncommon (107, 117, 118, 156, 244).

Penicillin plus vancomycin plus gentamicin has produced in vitro synergistic killing against clinical enterococcal isolates resistant to vancomycin (240). In an E. faecium strain where a synergistic bacteriostatic effect between penicillin and vancomycin was demonstrated in vitro, the triple combination of high-dose penicillin plus vancomycin plus gentamicin proved effective in a rabbit endocarditis model. However, a bacterial subpopulation resistant to penicillin/vancomycin synergism was frequently isolated from vegetations at the end of therapy (31). In a subsequent study using the same model, the triple combination of ceftriaxone plus vancomycin plus gentamicin was even more effective in reducing bacterial counts than the combination of either penicillin plus vancomycin plus gentamicin or penicillin plus teicoplanin plus gentamicin. Still, a subpopulation resistant to ceftriaxone/vancomycin synergism emerged in 10% to 20% of the animals (32). Other investigators have not found the triple combination of ampicillin plus vancomycin plus gentamicin to be reliably bactericidal in vitro against highly ampicillin-resistant, vancomycin-resistant, gentamicin-susceptible E. faecium strains (77).

In Vitro and In Vivo (Animal Data) Activity for Enterococci Resistant to Penicillins, Glycopeptides, and Aminoglycosides

The combinations of ampicillin (or penicillin) plus vancomycin, imipenem plus vancomycin, and ceftriaxone plus vancomycin (or teicoplanin) have shown bacteriostatic synergism in vitro against some enterococci resistant to penicillin, glycopeptides, and aminoglycosides (96,135). However, bacteriostatic synergism may be limited to specific strains (92, 99, 103), and bactericidal synergism has not been shown with these combinations (36, 99,103, 131,135). The triple combination of ampicillin (MIC = 16 µg/ml) plus imipenem (MIC = 32 µg/ml) plus vancomycin (MIC = 512 µg/ml) exhibited bactericidal synergistic activity in vitro against an E. faecium isolate (22). Ampicillin plus imipenem was highly active against the same E. faecium isolate in a rabbit endocarditis model, but efficacy was not increased by the addition of vancomycin to the regimen (22). In the same model, imipenem plus vancomycin was significantly more effective than ampicillin alone or ampicillin plus vancomycin. The authors suggested that ampicillin plus imipenem synergism may be due to the saturation of different penicillin-binding proteins by the two agents (22, 72, 84).

Ampicillin plus ciprofloxacin has been demonstrated to produce in vitro synergistic killing against some of these multiresistant strains that were still susceptible to ciprofloxacin, but in a rabbit endocarditis model this combination did not produce a significant reduction of bacteria in vegetations (131, 132). Ciprofloxacin plus novobiocin was bactericidal in a time-kill study, and produced a significant decrease in bacterial counts in vegetations in a rabbit endocarditis model (131, 217). Linezolid was more active than vancomycin in decreasing vegetation size caused by vancomycin-resistant E. faecium in a rat endocarditis model (205). The combination of quinupristin-dalfopristin with doxycycline resulted in less resistance to either agent when administered in combination than with either agent alone, in an in vitro endocarditis model (1).

back to top

ANTIMICROBIAL THERAPY

Since the human reservoir is the gastrointestinal tract, polymicrobial infections involving the gastrointestinal tract, urinary tract, and female genital tract are the primary infections. Infections of the urinary tract, soft tissue, and intraabdominal infections, and perhaps some cases of septic arthritis, can be treated with a single antimicrobial agent, especially in patients with normal host defenses (167, 181, 222). For bacteremia (without endocarditis), combination antibiotic therapy is often used but studies are not definitive. Some studies suggest that enterococcal bacteremia without endocarditis can be treated with a single agent (88, 95, 153), although the sources of bacteremia were not considered, which may be an important factor in determining outcome (106). Other studies suggest that combination therapy may be more effective than monotherapy therapy for serious infections such as bacteremia (167, 181, 191). In one prospective, observational study of 110 serious enterococcal infections, 71% of patients who received appropriate combination therapy (cell wall-active agent + aminoglycoside) achieved clinical cure compared to 53% of patients who received an active single agent only (p = 0.08) (207). 

For monotherapy in patients without penicillin allergy, the drug of choice is ampicillin (MIC usually two to four-fold lower than penicillin). Beta-lactamase-producing enterococci had been a concern, since they are resistant to ampicillin but are not detected by routine ampicillin susceptibility testing (182, 184). However, it appears now that they are rare, and many hospitals that have screened for beta-lactamase production in enterococci for several years have not found any (87). If isolated, they can be treated with ampicillin/sulbactam. If the organism is resistant to ampicillin, a glycopeptide such as vancomycin may be used for monotherapy. Imipenem offers no advantage over ampicillin against (beta-lactamase negative) enterococci in vitro nor in animal endocarditis models (13, 61, 232). We have seen a few E. faecalis isolates that are susceptible to ampicillin (MIC = 0.5 -1.0 µg/ml) but relatively resistant to imipenem (MIC > 8.0 µg/ml). However, in situations such as empiric therapy for complex intraabdominal infections, where coverage that includes enterococci may be prudent, imipenem (or piperacillin/tazobactam or ampicillin/sulbactam) might be substituted for regimens that contain ampicillin. A summary of treatment recommendations for enterococcal infection due to strains susceptible in vitro to glycopeptides is shown in Table 1.

Urinary Tract Infections

Urinary tract infections are the most common infection caused by enterococci and are often associated with urinary catheters. Enterococcal urinary tract infections not accompanied by bacteremia generally require only single drug therapy. If the organism is susceptible, ampicillin is the drug of choice. Vancomycin can be used if the organism is ampicillin-resistant. Linezolid or quinupristin/dalfopristin are reasonable alternatives if the enterococcus is resistant to both ampicillin and vancomycin.

For simple urinary tract infections, a quinolones with a low MIC for a particular isolate might be considered as an alternative, but caution must be exerted since enterococcal superinfections have occurred in patients treated with ciprofloxacin for infections caused by other bacteria (67, 285).

Complicated urinary tract infections as prostatitis and pyelonephritis are less common. They can be treated with the same agents used for simple urinary tract infections, but the duration of therapy would be longer. A seriously ill patient with pyelonephritis or perinephric abscess may benefit from combination therapy with a beta-lactam agent plus an aminoglycoside. Nitrofurantoin and fosfomycin exhibit excellent activity in vitro versus urinary enterococcal isolates including VRE strains (209, 256). Oral rifampin plus nitrofurantoin has cured a case of chronic prostatitis due to vancomycin-resistant E. faecium (256).

Intraabdominal Infections

The therapeutic recommendations outlined in this chapter are based on studies of predominantly E. faecium and E. faecalis. Optimal antimicrobial therapy for E. gallinarum, E. casseliflavus, E. avium, E. cecorum, E. durans, E. hirae, E. malodoratus, E. mundtii, E. pseudoavium and E. raffinosus is not known. However, based on in vitro data and anecdotal reports, it would seem reasonable to suggest that therapy for these enterococcal species is the same as that for E. faecium and E. faecalis.

Intraabdominal infections are polymicrobial in origin in which coverage for enteric bacteria (E. coli, Klebsiella pneumonia, etc.), and anaerobes (Bacteroides fragilis), must be covered including Enterococcus species. Broad spectrum therapy for the gastrointestinal flora mentioned above include ampicillin/sulbactam, piperacillin/tazobactam, and imipenem.

A case control study has suggested that empiric therapy for enterococci be especially considered for hepatobiliary or pancreatic infection, where both the prevalence of enterococci and the rate of enterococcal bacteremia are high (44).

Endocarditis

The necessity for combination antibiotic therapy is well established for enterococcal endocarditis. Improved outcome has been demonstrated in patients who received a synergistic combination of a cell wall-active agent plus an aminoglycoside, compared to patients who received monotherapy (59, 78, 125, 157, 161, 167, 181, 278). Cell wall-active antimicrobial agents such as the penicillins and glycopeptides, when combined with aminoglycosides, produce a synergistic bactericidal effect against susceptible strains of enterococci due to the enhanced intracellular penetration and the use of the aminoglycoside in the presence of the cell wall-active agent (172).

Penicillin (or ampicillin) plus gentamicin is the most commonly used combination for enterococcal endocarditis, although penicillin plus streptomycin has been the most studied combination regimen for enterococcal endocarditis. No direct comparative studies of efficacy between penicillin (or ampicillin) plus streptomycin versus penicillin plus gentamicin have been done, although streptomycin may be more effective than gentamicin in treatment of enterococcal endocarditis (279). In a study of streptomycin-resistant enterococcal endocarditis, 16 of 20 (80%) patients treated with penicillin plus gentamicin were cured, compared with a cure in 33 of 36 (92%) patients treated with penicillin plus streptomycin for streptomycin-susceptible enterococci in the same study (279). This higher relapse rate in patients treated with penicillin plus gentamicin was also higher than the relapse rate among patients treated with penicillin plus streptomycin in previous studies (125, 157, 161). Streptomycin has traditionally been given intramuscularly, which involves greater pain for the patient, although intravenous streptomycin has been given to a few patients and seemed to be well-tolerated (175). Streptomycin is predominantly ototoxic, and gentamicin is primarily nephrotoxic; while nephrotoxicity is often reversible, ototoxicity is often not (278). Moreover, gentamicin serum levels are more readily available to aid in monitoring for potential toxicity, and gentamicin is less expensive than streptomycin. A sufficient number of patients have been treated successfully with penicillin (or ampicillin) plus gentamicin that this is a well-accepted regimen (161, 167, 224, 278). For these reasons, gentamicin has replaced streptomycin as the more commonly used aminoglycoside in combination regimens for enterococcal endocarditis.

The recommended streptomycin dosing in enterococcal endocarditis is 7.5 mg/kg IM every 12 hours, with target peak serum levels of 15-30 µg/ml (161, 278). Gentamicin is usually given at 1 mg/kg IV every eight hours with target peak serum levels of > 3 µg/ml (161, 278). Treatment should continue for at least 4 weeks. Patients with symptoms greater than 3 months duration before initiation of appropriate therapy and patients with prosthetic valve endocarditis should receive antimicrobial therapy for at least 6 weeks (224, 279). Animal data do not support the use of once-daily dosing of aminoglycosides in enterococcal endocarditis (68, 158). However, one patient with E. faecalis pulmonic and tricuspid endocarditis was treated successfully with ampicillin and once-daily gentamicin (253). The combination of vancomycin plus an aminoglycoside has not been as extensively studied, so vancomycin should be used only when the patient has a significant history of allergy to the penicillins (278). The addition of ciprofloxacin to a regimen of ampicillin plus gentamicin was associated with cure in a case of relapsing E. faecalis endocarditis (137, 227). In selected patients, a shortened course of aminoglycosides may be considered. A 5-year nationwide prospective study in Sweden during 1995-1999 identified 881 definite episodes of infective endocarditis (195). Definite enterococcal endocarditis was diagnosed in 93 episodes (11%), the largest series of enterococcal endocarditis so far presented. Mortality during treatment was 16%, the relapse rate was 3%, and clinical cure was achieved in the remaining 81% of the episodes. Clinical cure was achieved with a median duration of cell wall-active antimicrobial therapy of 42 days combined with an aminoglycoside (median treatment time, 15 days). International guidelines generally recommend a 4-6-week combined synergistic treatment course with a cell wall-active antibiotic and an aminoglycoside. Treatment regimens in Sweden often include a shortened aminoglycoside treatment course in order to minimize adverse effects in older patients. In this study, fatal outcome seemed not to be due to the shortened aminoglycoside therapy course. In some enterococcal endocarditis episodes, shortening the duration of aminoglycoside therapy to 2-3 weeks requires further study.

Endocarditis due to vancomycin resistant Enterococcus is rare. The literature consists of single case reports, with most patients having serious underlying disease including transplantation, dialysis and health care associated infection (4, 8, 14, 18,19,24, 28, 39, 81, 122, 126, 159, 206, 260, 262, 265, 271,274,275, 278, 282). Most patients survived despite the lack of bactericidal therapy and valve replacement. The optimal therapy for this infection remains undefined, as comparative data are not available, patients were treated with various and multiple agents in combination and sequentially over varying periods of time.

Meningitis

Most experts would concur that treatment of enterococcal meningitis requires combination therapy so that maximal bactericidal activity may be achieved (15, 167, 181). However, in contrast to endocarditis, there are scant clinical data available to state unequivocally that combination therapy is superior to monotherapy in meningitis. In a review of enterococcal meningitis, thirty of thirty-two cases had data on antibiotic therapy (247). The majority of patients (20 of 30) received penicillin or ampicillin plus an aminoglycoside (three patients also received intrathecal gentamicin); there was only one death in this group, compared with one death in the group of four patients who received penicillin or ampicillin alone. One patient failed his initial regimen of ampicillin plus chloramphenicol, but recovered when the chloramphenicol was replaced by gentamicin. One severely ill patient received chloramphenicol plus gentamicin, and subsequently died (247). Ciprofloxacin has been reported to cure one case of E. faecalis meningitis in an infant (127). The combination of quinupristin/dalfopristin plus chloramphenicol cured a case of neonatal E. faecium meningitis that had failed therapy with teicoplanin plus chloramphenicol (89). A child with E. faecium meningitis was treated successfully with chloramphenicol (208). Intrathecal teicoplanin has been used successfully to treat a case of E. faecium meningitis (149). Intraventricular plus intravenous quinupristin/dalfopristin plus intravenous chloramphenicol sterilized the CSF of a patient with vancomycin-resistant E. faecium shunt infection who had failed therapy with chloramphenicol alone (268). Linezolid used alone for 28 days cured a patient with vancomycin-resistant E. faecium meningitis who had failed therapy with chloramphenicol (284). Linezolid used for 3 weeks plus gentamicin for 5 days sterilized the CSF of a second patient with vancomycin-resistant enterococcal meningitis (97).

Therapy for Strains Resistant to Aminoglycosides

Treatment of patients with endocarditis caused by enterococci with high-level resistance to aminoglycosides is associated with a high incidence of failure or relapse (166). Therapy for strains resistant to all aminoglycosides is usually limited to the use of ampicillin alone. Vancomycin alone is potentially less efficacious. Optimal duration of therapy with ampicillin alone is not known, but the observation that valve cultures at surgery are positive after as much as one month of ampicillin therapy suggests that longer courses than the traditional 4 to 6 weeks, such as 8 to 12 weeks should be ly considered (59, 124, 278). Continuous, rather than intermittent, infusion of ampicillin may prove beneficial since there are data showing its greater efficacy in sterilizing cardiac vegetations and improving survival in a rat endocarditis model (259). Cardiac valve replacement may well be required for a cure in many of these patients (59, 166, 224, 278).

A patient with ampicillin- and vancomycin-susceptible but gentamicin- and streptomycin-resistant E. faecalis endocarditis was initially treated with ampicillin (4g IV q6h) plus vancomycin. Both peak and trough serum bactericidal titers were 1:2. After imipenem was added (500 mg IV q6h), the peak and trough serum bactericidal titers increased to 1:128 and 1:64, respectively. A total of 6 weeks of antibiotics was given and the patient was cured (6). The authors suggested that the increased serum bactericidal activity seen after the addition of imipenem may be due to the saturation of different penicillin-binding proteins (6). The same regimen of ampicillin plus vancomycin plus imipenem was used successfully in treating a case of gentamicin-resistant enterococcal bacteremia without endocarditis (5). Tripodi and colleagues reported microbiological and clinical cure of ampicillin-susceptible but gentamicin-resistant E. durans endocarditis with the combination of ampicillin (MIC = 0.5 µg/ml) plus ciprofloxacin (MIC = 8 µg/ml) after failure with ampicillin plus gentamicin (265). The efficacy of the treatment was predicted in vitro by time-kill studies.

Therapy for Strains Resistant to Penicillins and Glycopeptides But Susceptible to Aminoglycosides

If the cell wall-active agents (penicillins and glycopeptides) are inactive against some enterococci, the antibiotic combinations of cell wall-active agents plus aminoglycosides will no longer be synergistic. Given the in vitro resistance, for certain enterococcal strains discussed previously, synergism may be limited chiefly by the achievable penicillin serum concentration. Since 5 million units of penicillin G infused intravenously can achieve a peak serum concentration of 135 µg/ml (134), the authors conclude that some gentamicin-susceptible E. faecium strains with penicillin MICs between 50 and 200 µg/ml may be treated with high-dose penicillin plus gentamicin (264).

These findings for penicillin have been assumed to be true for ampicillin, also, because the two agents’ mechanism of action is the same. Since peak ampicillin serum levels of 109 to 150 µg/ml can be attained after a 2-gram intravenous dose (75, 214), some enterococci with an ampicillin MIC up to 64 or even 128 µg/ml may prove to be susceptible to ampicillin-gentamicin synergism if high-dose ampicillin is used. A hemodialysis patient with ampicillin-resistant (MIC = 32 µg/ml) and gentamicin-resistant (MIC = 1,000 µg/ml) E. faecium bacteremia was treated successfully with ampicillin 2 grams IV q6h and streptomycin (MIC = 32 µg/ml) 500 mg twice weekly after failing therapy with doxycycline + chloramphenicol (54). A patient with vancomycin-resistant E. faecium endocarditis had persistent bacteremia during therapy with 12 g of ampicillin (MIC = 64 µg/ml) per day plus gentamicin and subsequently with quinupristin/dalfopristin. Blood cultures finally became sterile when therapy was changed to ampicillin 20 g per day plus gentamicin (183).

A patient with ampicillin-resistant (MIC = 64 µg/ml) E. faecium bacteremia failed therapy with continuous infusion ampicillin (20 g/day) plus gentamicin (MIC < 500 µg/ml) q12h, but was cured with continuous infusion ampicillin/sulbactam (20 g ampicillin/10 g sulbactam) plus gentamicin q12h (162). No bactericidal effect was seen when the patient was on ampicillin plus gentamicin (serum ampicillin concentration of 103 mg/ml), but a serum bactericidal titer of 1:2 was detected when the patient was on ampicillin/sulbactam plus gentamicin (serum ampicillin concentration of 130 mg/ml). It is possible that, via unknown mechanism, sulbactam may slightly enhance the activity of ampicillin against certain strains of enterococci (162).

In an open trial of teicoplanin therapy for enterococcal endocarditis, 5 of 7 patients treated with teicoplanin alone and 6 of 7 patients treated with teicoplanin plus an aminoglycoside were cured (233). In a study of teicoplanin therapy for endocarditis caused by Gram-positive bacteria, five patients with E. faecalis endocarditis were treated with teicoplanin alone, and all five were cured (216).

Therapy for Strains Resistant to Penicillins, Glycopeptides, and Aminoglycosides

The terms “VRE” and “VREF” are used commonly to describe these multiply resistant strains, which to date have been predominantly E. faecium. Antibiotic selection is most problematic for these strains (Table 2). At the current time, linezolid is approved for the treatment of VRE and is the drug of choice. Based on in vitro susceptibility results and anecdotal reports, daptomycin and quinupristin-dalfopristin also warrant consideration.

Various other bacteriostatic antimicrobials with less in vitroactivity than the penicillins, glycopeptides, and aminoglycosides have been tried either alone or in combination against multiresistant enterococci. Oral rifampin plus nitrofurantoin has cured a case of chronic prostatitis due to vancomycin-resistant E. faecium (256). Chloramphenicol therapy for multiresistant enterococci in 16 severely ill patients in one study showed somewhat encouraging results (189). Eight of 14 patients in whom a clinical response could be determined showed improvement. Four patients were treated with rifampin in addition to chloramphenicol; most patients received other antibiotics that alone do not have activity against enterococci. Eleven of the 16 patients had a drainage procedure or debridement performed. In a study of E. faecium infections in liver transplant recipients, chloramphenicol was used as monotherapy in 16 cases, and 6 patients died (202). Single cases of successful therapy using chloramphenicol alone or in combination with vancomycin or rifampin have also been reported,(130,207, 208). In another study of vancomycin-resistant Enterococcus bacteremia treated with chloramphenicol, 22 of 36 patients demonstrated a clinical response, and 34 of 43 patients showed a microbiological response (133). However, no significant effect on mortality by chloramphenicol usage could be demonstrated (133). Lynch and colleagues studied 4 strains of E. faecalis and 1 strain of E. faecium and demonstrated efflux of chloramphenicol in all the strains (150). Their results suggest that many wild-type strains of E. faecalis, and presumably also of E. faecium, possess endogenous efflux pumps that excrete chloramphenicol (150). Therefore, chloramphenicol use probably should be limited to those cases where no other agents would likely be effective.

Intravenous doxycycline used alone for two weeks in a patient with catheter-related sepsis and probable endocarditis resulted in clearing of the bacteremia, but the patient subsequently died of congestive heart failure (130). Oral doxycycline for two weeks, plus removal of the Hickman catheter presumed to be the source of the bacteremia, was successful in treating a case of E. faecium bacteremia (174). An immunocompromised patient with vancomycin- and ampicillin-resistant E. faecium bacteremia who appeared to fail therapy with quinupristin/dalfopristin plus gentamicin was cured with five days of intravenous tetracycline followed by two months of oral doxycycline (with occasional periods of intravenous tetracycline when he could not tolerate oral therapy) (108).

The streptogramin combination quinupristin/dalfopristin has been used for treatment of E. faecium, but not E. faecalis, which are intrinsically resistant to the combination. Among 15 cases of multiresistant E. faecium infection treated with quinupristin/dalfopristin, three patients were cured of their infection, 5 had recurrence of the infection, and in 7 cases, the outcome was indeterminate (50). Blood cultures became negative for enterococcus during treatment in all patients with bacteremia. However, catheters were removed in the patients with catheter-related bacteremia prior to therapy, so it is not known whether removal of catheters alone would have been curative (50). Vancomycin-resistant E. faecium-associated mortality was significantly lower in a group of 20 patients with vancomycin-resistant E. faecium bacteremia treated with quinupristin/dalfopristin compared with an historical cohort of 42 patients with vancomycin-resistant E. faecium bacteremia treated with other agents (146). In another study, bacteriologic eradication occurred in 17 of 23 evaluable patients with serious E. faecium infections treated with quinupristin/dalfopristin (280). In a multicenter study of quinupristin/dalfopristin for vancomycin-resistant E. faecium infections, clinical success was achieved in 73.6% (142 of 193) of evaluable patients, and the bacteriological success rate was 70.5% (110 of 156), resulting in an overall success rate of 65.4% (102 of 156) (169). Successful treatment with quinupristin/dalfopristin for E. faecium vertebral osteomyelitis, peritonitis, ventriculitis, aortic graft infection, and endocarditis has also been reported (60, 81, 129, 150, 186, 229, 247, 254, 274). Quinupristin/dalfopristin has also been used successfully in treating pediatric patients (50). Since quinupristin/dalfopristin is not bactericidal against E. faecium, combining other agents with quinupristin/dalfopristin may be a reasonable approach in selected cases. A case of vancomycin-resistant E. faecium endocarditis with persistently positive blood cultures after two weeks of therapy with quinupristin/dalfopristin was cured after intravenous doxycycline and oral rifampin were added to the quinupristin/dalfopristin and therapy continued for 8 more weeks (159). Since E. faecium strains resistant to quinupristin/dalfopristin can be selected in vitro without difficulty (164), it is not surprising that emergence of increased resistance to quinupristin/dalfopristin has appeared during therapy for E. faecium infections (42, 145, 169, 280). Also, superinfection with E. faecalis has occurred during quinupristin/dalfopristin therapy for E. faecium infections (41, 169, 280). The agent is only available by intravenous infusion. Myalgias in 6.6% or patients, arthralgias in 9.1% and reports of antimicrobial resistance have been limiting features (60, 104).  

The oxazolidinones are a new class of antimicrobials that inhibit bacterial protein synthesis (143, 239, 252). Linezolid is a semisynthetic oxazolidinone agent that exhibits bacteriostatic in vitro and in vivo activity against enterococci (21, 63, 73, 192, 204, 226, 235). Although linezolid has in vitro activity against vancomycin-resistant E. faecalis, it has only received approval for vancomycin-resistant E. faecium infections because of the lack of patients with E. faecalis isolates in the phase III trials. In one case report, a neutropenic patient with ampicillin- and vancomycin-resistant E. faecium bacteremia was treated successfully with 14 days of linezolid (600 mg IV q12h) plus gentamicin. Time-kill studies with linezolid plus gentamicin did not show in vitro synergism (193). In another case report, a patient with acute myelogenous leukemia developed vancomycin-resistant E. faecium bacteremia after bone marrow transplantation. Blood cultures were persistently positive during 10 weeks of therapy with quinupristin/dalfopristin, and despite the addition of chloramphenicol and doxycycline at various times (160). The source of the E. faecium bacteremia was deemed to be an extensive central venous thrombus. The E. faecium remained susceptible in vitro to quinupristin/dalfopristin during therapy. Antimicrobial therapy was changed to intravenous linezolid alone and blood cultures became negative 8 days later, and all subsequent blood cultures were negative. The patient received 6 weeks of intravenous linezolid followed by 6 weeks of oral linezolid, and had no further complications (160). In an open-label, non-comparative, non-randomized compassionate-use program for linezolid, in which 66% of 826 treatment courses were prescribed for the treatment of VRE, the clinical and microbiological cure rates were 81% and 86% respectively (17). Sites of infection included bloodstream, intra-abdominal, complicated skin and soft tissue, urinary tract and bone.

In the largest case series published to date, 15 patients infected with vancomycin-resistant E. faecium were treated with linezolid after all other available therapeutic options were tried (39). The median duration of therapy was 20.5 (+ 3.5) days. Microbiologic cure was achieved in all 10 patients who survived and completed therapy, including a patient with infective endocarditis, who was treated for 42 days. All 7 patients alive at long-term follow-up were free of infection. No deaths were attributable to the index infection (39). Linezolid use has been reported in immunosuppressed patients. In a prospective, randomized study comparing quinupristin-dalfopristin with linezolid in forty cancer patients infected with VREF, comparable clinical efficacy was seen. Clinical response was seen in 58% of patients treated with linezolid vs. 43% treated with quinupristin-dalfopristin. (219). Five of six liver recipients with sepsis secondary to intra-abdominal VRE infection were cured at end of treatment, although a sixth patient relapsed with a linezolid-resistant E. faecium strain (144).

Although no comparative trials of linezolid have been done in patients with endocarditis, osteomyelitis, or meningitis, multiple case reports have been described. Oral linezolid was successful in treating a case of vancomycin-resistant E. faecium endocarditis that failed sequential monotherapy with chloramphenicol and quinupristin/dalfopristin (14). Oral linezolid was successful in treating a case of vancomycin–resistant E. faecium endocarditis in a renal transplant recipient with HIV infection. (8). Linezolid was also reported to be successful in treating cases of E. faecalis prosthetic valve endocarditis with (220) and without (275) high-level gentamicin-resistance. Failure of therapy for endocarditis with linezolid was noted in another report (287). Intravenous linezolid was successful in treating a patient with MRSA and VRE bacteremias secondary to vertebral osteomyelitis who failed therapy with intravenous vancomycin and oral amoxicillin and fusidic acid (165). Oral linezolid was reported to be successful in treating a case of E. faecalis (221) and E. faecium osteomyelitis (261) Although only bacteriostatic, there have been several case reports of successful treatment with linezolid of patients with VREF meningitis, including patients in the postoperative period, persons with infections associated with a device, and patients with infections in the setting of hyperinfection with yloides stercoralis (97, 246, 284).

The emergence of linezolid resistance during therapy is uncommon (288). Resistant mutants can be generated at a low frequency having mutations involving the domain V peptidyltransferase center of 23S rRNA, which is the same mutation in as in linezolid-resistant MRSA. How stable the resistance is in these isolates is uncertain. Other centers have isolated increased numbers of linezolid-resistant VRE and have noted a higher incidence of linezolid resistance emerging during therapy (1 of 45) (86). However, prior use with linezolid is not necessary before the development of resistance (185). A case of linezolid-resistant E. faecalis was isolated in a cord blood stem cell recipient following treatment with linezolid for VRE bacteremia (52). Three cases of linezolid resistant E. faecalis were reported after prolonged treatment with linezolid for VRE. One of these patients had been diagnosed with VRE aortic and mitral valve endocarditis that was sensitive to ampicillin. The patient was treated for six weeks with linezolid. Two weeks following treatment, the patient was readmitted with third-degree heart block and VRE bacteremia. The patient was desensitized to penicillin and treated successfully with ampicillin (25). Given the complexity in determining optimal antimicrobial therapy for multiply resistant enterococci (“VRE/VREF”), consultation with an infectious diseases specialist may be prudent in these cases.

Daptomycin is a novel cyclic lipopeptide that is a fermentation product of Streptomyces roseosporus with a unique mechanism of action. It acts specifically at bacterial cytoplasmic membrane wit multiple effects on cellular function including inhibition of lipoteichoic acid synthesis, disruption of membrane potential and inhibition of peptidoglycan synthesis. It displays rapid concentration-dependent killing and is bactericidal even in the stationary phase of growth. The agent is currently FDA approved in the United States for management of complicated skin and soft tissue infection. Current recommended dosage is 4 mg/kg once daily, however in clinical trials of serious staphylococcal and enterococcal infection, doses of 6 mg/kg/24 hours were used. In vitro studies of 289 isolates of E. faecalis demonstrated MICs < = 2 µg/ml in all isolates to daptomycin (109). A large multicenter European study with in vitro studies of 40 isolates of vancomycin-resistant E. faecalis and 114 isolates of vancomycin-resistant E. faecium demonstrated MICs<2 µg/ml in all isolates (47). In vitro activity of daptomycin against enterococci from the nosocomial and community environments in Portugal were studied. 1151 isolates were obtained, all of which demonstrated MICs <= 4 µg/ml (194). Similarly, 89 strains of VRE were tested in vitro. Daptomycin MICs were <=2 and <=4 µg/ml for E. faecalis and E. faecium, respectively (113).  In vitro studies have demonstrated additive or indifferent interactions with other antibiotics. Antagonism, as determined by kill curve studies, has not been observed. Daptomycin has been reported to show synergism in vitro with aminoglycosides, rifampin and beta lactams against some VRE (38, 243). In an in vitro study using 20 isolates of vancomycin-susceptible E. faecalis, daptomycin showed synergistic activity with ceftriaxone, cefepime and imipenem against 65%, 35% and 35% of the isolates, respectively (243). Unfortunately, very limited clinical data exists. A phase III trial comparing linezolid to daptomycin for the treatment of VRE was terminated early due to difficulty enrolling patients (33). Daptomycin-resistant strains are very difficult to generate in vitro (38, 238, 242). Resistance to daptomycin on therapy with the agent is rare, having been reported in 3 patients with enterococcal infection. The emergence of resistance to daptomycin occurred in 2 of more than 1000 (<0.2%) infected subjects across the entire set of Phase 2 and 3 clinical trials (one S. aureus and one E. faecalis). A case was reported where there was emergence of daptomycin resistance (MICs >32 µg/ml) in E. faecium during daptomycin therapy (140). Furthermore, a case of daptomycin resistance that developed during daptomycin therapy for E. faecalis has also been reported (178). At this time, no mechanism of resistance to daptomycin has been identified, there are no known transferable elements and cross-resistance has not been observed with any other class of antibiotic. Daptomycin appears to be a promising drug with good in vitro bactericidal activity against enterococci regardless of vancomycin susceptibility; however, data regarding clinical outcomes is lacking.

Tigecycline is a semisynthetic glycylcycline antimicrobial agent that is similar in structure to minocycline, but with excellent in vitro activity versus gram-positive bacteria including VRE. It is a bacteriostatic agent in vitro versus enterococci. The agent has been recently approved in the United States for therapy of complicated skin soft tissue and intraabdominal infection. It has linear pharmacokinetics, has a Cmax of 0.87 µg/ml a Cmin of 0.13 µg/ml, and AUC0-24hrs of 4.7 µg/h/ml, a t ½ of 42 hrs and significant tissue uptake (179). The drug is administered by intravenous infusion only, and in initial clinical trials had nausea that was more common than comparators (190, 245).

Antimicrobial agents undergoing development include dalbavancin and oritavancin which are both new glycopeptides, the lipoglycopeptide telavancin, and iclaprim, which is a diaminopyrimidine inhibitor of microbial dihydrofolate reductase. Dalbavancin is an investigational semisynthetic, glycopeptide antibiotic with excellent in vitro activity against gram-positive bacteria. It is active in vitro against vancomycin susceptible enterococci, and vancomycin resistant enterococci containing the VanB phenotype (250). It is structurally related to teicoplanin, and thereby will likely be of limited utility for therapy of vancomycin-resistant enterococci (154). Dalbavancin has a long half life 9-12 d; a single infusion of 1000mg results in mean plasma concentrations of >35mg/L for a 7 day period (94). It is being studied in skin soft tissue and catheter bloodstream infection with preliminary results showing clinical and bacteriologic success rates similar to comparators (94, 218) The oligosaccharide, evernimicin, and fluoroquinolone clinafloxacin exhibit good in vitro enterococcal activity, but did not complete clinical trials. (46, 80, 115, 119, 139, 187, 234, 237, 269). The new aminoglycoside arbekacin shows preliminary promise in treating some high-level gentamicin-resistant enterococci. Arbekacin is currently available only in Japan, where it is used to treat gentamicin- and methicillin-resistant Staphylococcus aureus infections (197).

back to top

ADJUNCTIVE THERAPY

Although infections by VRE and VREF appear to be increasing, such organisms are not necessarily highly virulent. There may be clinical settings where the patient will recover even when no appropriate antimicrobial therapy is available. For example, catheter-related bacteremias might sometimes be treatable with catheter removal alone (88, 129). Surgical site infections, skin and soft tissue infections, and intraabdominal abscesses may be manageable by surgical debridement and drainage and treated with antibiotics active against other gastrointestinal flora (44, 88, 129).

ENDPOINTS FOR MONITORING THERAPY

Duration of antimicrobial therapy for enterococcal infections depends predominantly on the site of infection and the patient’s clinical response to therapy. Treatment for simple urinary tract infections may require only a few days of oral or intravenous antibiotics. Bacteremia without endocarditis may require 10 to 14 days of antibiotics and typically depends on how quickly the patient responds to therapy. If the source of infection cannot be removed, such as central venous catheters that must remain or abscesses that cannot be drained, the duration of antimicrobial therapy might naturally be longer. As mentioned previously, endocarditis in certain clinical situations can require a minimum of 6 weeks of antibiotics.

VACCINES

There are no vaccines for this bacterium.

INFECTION CONTROL

Colonized patients are the primary reservoir of VRE. Patient to patient transmission occurs primarily via indirect transmission by health-care workers (34). Medical devices, such as electronic rectal thermometers, have also been implicated (147). VRE are relatively hardy and can persist for prolonged periods in the environment, and the immediate surroundings of a colonized patient may be heavily contaminated (20). The role of environmental contamination in transmission remains to be precisely defined. The epidemiology of vancomycin resistant E. faecalis is similar to that of vancomycin resistant E. faecium (196).

The Hospital Infection Control Practices Advisory Committee (HICPAC) has published “Recommendations for Preventing the Spread of Vancomycin Resistance” (92). They are summarized in Tables 3 and 4.

Key principals underlying an effective control strategy include:

1. The hospital laboratory must be able to accurately identify VRE. Automated microtitre systems may be unreliable.

2. The number of asymptomatically colonized patients exceeds the number of clinically infected patients by many fold. The former group of patients are a reservoir for spread of VRE; surveillance via stool or rectal swab cultures of patients at risk are needed to detect VRE carriage.

3. VRE infected or colonized patients must be identified and placed into appropriate isolation as rapidly as possible.

4. Since most transmission occurs via health care workers, hospital staff must be familiar with, and must follow, isolation and control procedures (Table 4).

5. Contamination of the environment and of medical equipment may play a role in transmission; effective disinfection procedures are necessary.

6. VRE carriers who are re-admitted or transferred to other facilities must be quickly identified and placed into isolation.

7. Antibiotic usage should be controlled. Although current CDC recommendations stress control of vancomycin usage, the strength of the association between VRE and vancomycin is controversial (see Section II-C). Reduction in the use of cephalosporins and agents with potent anaerobic activity may be beneficial.

Implementation of a multi-faceted control strategy such as that outlined in the HICPAC recommendations has reportedly led to a reduction in the rate of VRE transmission in several acute care facilities. In the setting of an acute outbreak in a facility or unit in which VRE was previously present at low levels, vigorous implementation of such a strategy may lead to the elimination of VRE (66) or return to baseline levels (27). Even after establishment of endemic VRE in a facility, transmission can be significantly reduced by a sustained and vigorous intervention that addresses key control issues (173). A coordinated, multi institutional control strategy has resulted in a sustained reduction in the prevalence of VRE among hospitals and long-term care facilities in a large geographic region (198).

The majority of patients colonized with VRE in long-term care facilities appear to have acquired the organism during an acute care hospitalization (184). Patient to patient transmission appears to occur at a much lower rate than in acute care facilities. This may be due to the lower level of medical care and infrequency of invasive procedures in long-term care. Thus, there is little justification for denying a patient colonized with VRE to a long-term care facility. Administration of antibiotics prolongs the duration of VRE carriage in long-term care patients; thus antibiotics should be administered only when clearly indicated. Recommendations for control of VRE in the long term care facility have been published (Table 5).

Testing of the molecular relatedness of isolates is a well established method for investigation of epidemics of hospital acquired infection. Pulse field gel electrophoresis is the method of choice for delineation of the relatedness of strains, however for evaluation of the possibility of gene dissemination, PCR methods are needed (55, 70). The use of the tests for endemic infection control requires further investigation. In one Chicago hospital system (210, 211) ,data on nosocomial infection was collected during a 24 month period before and 60 month after implementing a new program that included routine molecular testing of VRE, drug resistant gram-negative bacteria and methicillin-resistant Staphylococcus aureus. During the intervention period, infections per 1,000 patient days fell 13 percent, and the percentage of hospitalized patients with nosocomial infections decreased 23 percent. The rate of infection fell to 43 percent below the U.S. rate. Approximately 50 deaths were avoided in a 5 year period. The typing laboratory costs for the program were $ 400,000 per year, with a savings of $5.00 for each dollar spent in relation to nosocomial infection reduction. In this study, cost savings by use of the molecular tests for endemic nosocomial infection were accomplished by a combination of establishment of clonality of isolates so that early intervention could be accomplished and by determination of the unrelatedness of isolates, thereby avoiding unneeded and costly epidemic investigation. Cost reduction was also accomplished by earlier recognition of person to person spread of isolates as compared to traditional surveillance, and molecular testing was used to establish the presence of pseudoepidemics also avoiding further epidemic investigation.

back to top

SUMMARY AND CONCLUSION

Although part of the normal intestinal flora and once felt to be innocuous endogenous pathogens, enterococci have proven to have much more complex interactions with the human host, having emerged in recent years as important nosocomial pathogens. Strains with resistance to multiple antimicrobials are on the rise, posing significant therapeutic and epidemiological challenges. In order to achieve the goal of minimizing the impact of resistance, a more comprehensive, multidisciplinary effort is needed, including a better understanding of the epidemiology and pathogenicity of these micro-organisms, judicious use of antimicrobials, effective infection control measures in hospitals, and reduction of resistance in reservoirs including the environment and animal husbandry.

back to top

REFERENCES

1. Aeschlimann JR, Zervos MJ, Rybak MJ. Treatment of vancomycin-resistant Enterococcus faecium with RP 59500 (quinupristin-dalfopristin) administered by intermittent or continuous infusion, alone or in combination with doxycycline, in an in vitro pharmacodynamic infection model with simulated endocardial vegetations. Antimicrob Agents Chemother. 1998 Oct;42(10):2710-7. [PubMed]

2. Al-Obeid S, Gutmann L, Williamson R. Modification of penicillin-binding proteins of penicillin-resistant mutants of different species of enterococci. J Antimicrob Chemother 1990;26:613-8. [PubMed]

3. Alam MR, Donabedian S, Brown W, Gordon J, Chow JW, Zervos MJ, Hershberger E. Heteroresistance to vancomycin in Enterococcus faecium. J Clin Microbiol. 2001 ;39:3379-81.  [PubMed]

4. Ang JY, Lua JL, Turner DR, Asmar BI. Vancomycin-resistant Enterococcus faecium in a premature infant sucessfully treated with linezolid. Pediatr Infect Dis J 2003;22:1101-3.  [PubMed]

5. Antony SJ, Matheren P, Stratton CW. Increased bactericidal activity as documented by serum bactericidal titers for a triple combination of cell wall active agents against gentamicin-resistant enterococci. Scand J Infect Dis 1995;27:401-3. [PubMed]

6. Antony SJ, Ladner J, Stratton CW, Raudales F, Dummer SJ. High-level aminoglycoside-resistant enterococcus causing endocarditis successfully treated with a combination of ampicillin, imipenem and vancomycin. Scand J Infect Dis 1997;29:628-30. [PubMed]

7. Appelbaum PC, Jacobs MR. Recently approved and investigational antibiotics for treatment of severe infections caused by gram-positive bacteria. Curr Opin Microbiol. 2005.  [PubMed]

8. Archuleta S, Murphy B, Keller MJ. Successful treatment of vancomycin-resistant Enterococcus faecium endocarditis with linezolid in a renal transplant recipient with human immunodeficiency virus infection. Transpl Infect Dis 2004;6:117-9.  [PubMed]

9. Arduino RC, Murray BE. Enterococci: antimicrobial resistance. In: Mandell GL, Douglas RG, Bennett JE, eds. Update to Principles and Practice of Infectious Diseases. New York: Churchill Livingstone Inc; 1993:3-15. [PubMed]

10. Arthur M, Courvalin P. Genetics and mechanisms of glycopeptide resistance in enterococci. Antimicrob Agents Chemother 1993;37:1563-71. [PubMed]

11. Arthur M, Depardieu F, Reynolds P, Courvalin P. Moderate-level resistance to glycopeptide LY333328 mediated by genes of the vanA and vanB clusters in enterococci. Antimicrob Agents Chemother 1999;43:1875-80. [PubMed]

12. Aslangul E, Baptista M, Fantin B, Depardieu F, Arthur M, Courvalin P, Carbon C. Selection of glycopeptide-resistant mutants of VanB-type Enterococcus faecalis BM4281 in vitro and in experimental endocarditis. J Infect Dis 1997;175:598-605. [PubMed]

13. Auckenthaler R, Wilson WR, Wright AJ, Washington JA II, Durack DT, Geraci JE. Lack of an in vivo and in vitro bactericidal activity of N-formimidoyl thienamycin against enterococci. Antimicrob Agents Chemother 1982;22:448-52. [PubMed]

14. Babcock HM, Ritchie DJ, Christiansen E, Starlin R, Little R, Stanley S. Successful treatment of vancomycin-resistant Enterococcus endocarditis with oral linezolid. Clin Infect Dis 2001. 32:1373-1375. [PubMed]

15. Bayer AS, Seidel JS, Yoshikawa TT, Anthony BF, Guze LB. Group D enterococcal meningitis: clinical and therapeutic considerations with report of three cases and review of the literature. Arch Intern Med 1976;136:883-6. [PubMed]

16.Bethea JA, Walko CM, Targos PA.Treatment of vancomycin-resistant enterococcus with quinupristin/dalfopristin and high-dose ampicillin. Ann Pharmacother. 2004 ;38:989-91.  [PubMed]

17. Birmingham MC, Rayner CR, Meagher AK, Flavin SM, Batts DH, Schentag JJ.  Linezolid for the treatment of multidrug-resistant, gram-positive infections: experience from a compassionate-use program. Clin Infect Dis. 2003 Jan 15;36:159-68.  [PubMed]

18.Bishara J, Sagie A, Samra Z, Pitlik S. Polymicrobial endocarditis caused by methicillin resistant-Staphylococcus aureus and glycopeptide-resistant enterococci. Eur J Clin Microbiol Infect Dis; 1999;18:674-5. [PubMed]

19. Botha PL, Struwig MC, de Vries W, Hough J, Chalkley LJ. Enterococcal endocarditis-a case treated with teicoplanin and amoxycillin. S Afr Med J 1998;88:564-5 [PubMed]

20. Bonilla HF, Zervos MJ, Kauffman CA. Long term survival of vancomycin-resistant Enterococcus faecium on a contaminated surface. Infect Control Hosp Epidemiol. 1996 Dec;17:770-2. [PubMed]

21. Bostic GD, Perri MB, Thal LA, Zervos MJ. Comparative in vitro and bactericidal activity of oxazolidinone antibiotics against multidrug-resistant enterococci. Diagn Microbiol Infect Dis 1998;30:109-12. [PubMed]

22. Brandt CM, Rouse MS, Laue NW, Stratton CW, Wilson WR, Steckelberg JM. Effective treatment of multidrug-resistant enterococcal experimental endocarditis with combinations of cell wall-active agents. J Infect Dis 1996;173:909-13. [PubMed]

23. Brennen C, Wagener MM, Muder RR. Vancomycin-resistant Enterococcus faecium in a long-term care facility. J Am Geriatr Soc 1998; 46:157-160. [PubMed]

24. Brink AJ, van den Ende J, Routier RJ, Devenish L. A case of vancomycin-resistant enterococcal endocarditis. S Afr Med J 2000;90:1113-5.  [PubMed]

25. Burleson BS, Ritchie DJ, Micek ST, Dunne WM. Enterococcus faecalis resistant to linezolid: case series and review of the literature. Pharmacotherapy. 2004 Sep;24:1225-31.  [PubMed]

26. Bush LM, Calmon J, Cherney CL, Wendeler M, Pitsakis P, Poupard J, Levison ME, Johnson CC. High-level penicillin resistance among isolates of enterococci. Ann Intern Med 1989;110:515-20. [PubMed]

27. Byers KE, Anglim AM, Anneski CJ, Germanson TP, Gold HS, Durbin LJ, Simonton BM, Farr BM. A hospital epidemic of vancomycin-resistant Enterococcus: risk factors and control. Infect Control Hosp Epidemiol 2001; 22:140-147. [PubMed]

28. Carfagna P, Tarasi A, Cassone M, Del Grosso MF, Bianco G, Venditti M. Prosthetic biologic valve endocarditis caused by a vancomycin-resistant (vanA) Enterococcus faecalis: case report J Chemother 2000;12:416-20.  [PubMed]

29. Carmeli Y, Eliopoulos GM, Samore MH. Antecedent treatment with different antibiotic agents as a risk factor for vancomycin-resistant Enterococcus. Emerg Infect Dis 2002;8:02-807.  [PubMed]

30. Carmeli Y, Samore MH, Huskins WC. The association between antecedent vancomycin treatment and hospital-acquired vancomycin-resistant enterococci: a meta-analysis. Arch Intern Med 1999; 159:2461-2468. [PubMed]

31. Caron F, Lemeland JF, Humbert G, Klare I, Gutmann L. Triple combination penicillin-vancomycin-gentamicin for experimental endocarditis caused by a highly penicillin- and glycopeptide-resistant isolate of Enterococcus faecium. J Infect Dis 1993;168:681-6.  [PubMed]

32. Caron FM, Pestel M, Kitzis M-D, Lemeland J-F, Humbert G, Gutmann L. Comparison of different beta-lactam-glycopeptide-gentamicin combinations for an experimental endocarditis caused by a highly beta-lactam-resistant and highly glycopeptide-resistant isolate of Enterococcus faecium. J Infect Dis 1995;171:106-12. [PubMed]

33. Carpenter CF, Chambers HF. Daptomycin: another novel agent for treating infections due to drug-resistant gram-positive pathogens. Clin Infect Dis. 2004 Apr 1;38:994-1000.  [PubMed]

34. Centers for Disease Control and Prevention. Recommendations for preventing the spread of vancomycin resistance. Recommendations of the Hospital Infection Control Practices Advisory Committee (HICPAC). MMWR Recomm Rep. 1995;44(RR-12):1-13. [PubMed]

35. Cetinkaya Y, Falk P, Mayhall CG. Vancomycin-resistant enterococci. Clin Microbiol Rev 2000;13:686-707.  [PubMed]

36. Cercenado E, Eliopoulos GM, Wennersten CB, Moellering RC Jr. Absence of synergistic activity between ampicillin and vancomycin against highly vancomycin-resistant enterococci. Antimicrob Agents Chemother 1992; 36:2201-3. [PubMed]

37. Cercenado E, Eliopoulos GM, Wennersten CB, Moellering RC Jr. Influence of high-level gentamicin resistance and beta-hemolysis on susceptibility of enterococci to the bactericidal activities of ampicillin and vancomycin. Antimicrob Agents Chemother 1992; 36:2526-8. [PubMed]

38. Cha R, Rybak MJ. Daptomycin against multiple drug-resistant staphylococcus and enterococcus isolates in an in vitro pharmacodynamic model with simulated endocardial vegetations. Diagn Microbiol Infect Dis. 2003 ;47:539-46. [PubMed]

39. Chien JW, Kucia ML, Salata RA. Use of linezolid, an oxazolidinone, in the treatment of multidrug-resistant gram-positive bacterial infections. Clin Infect Dis 2000;30:146-51. [PubMed]

40. Chow JW, Zervos MJ, Lerner SA, Thal LA, Donabedian SM, Jaworski DD, Tsai S, Shaw KJ, Clewell DB. A novel gentamicin resistance gene in Enterococcus. Antimicrob Agents Chemother 1997;41:511-4. [PubMed]

41. Chow JW, Davidson A, Sanford E III, Zervos MJ. Superinfection with Enterococcus faecalis during quinupristin/dalfopristin therapy. Clin Infect Dis 1997;24:91-2. [PubMed]

42. Chow JW, Donabedian SM, Zervos MJ. Emergence of increased resistance to quinupristin/dalfopristin during therapy for Enterococcus faecium bacteremia. Clin Infect Dis 1997;24:90-1. [PubMed]

43. Chow JW, Donabedian SM, Clewell DB, Sahm DF, Zervos MJ. In vitro susceptibility and molecular analysis of gentamicin-resistant enterococci. Diagn Microbiol Infect Dis 1998;32:141-6. [PubMed]

43a. Clark NC, Olsvik Ø, Swenson JM, Spiegel CA, Tenover FC. Detection of a streptomycin/spectinomycin adenylyltransferase gene (aadA) in Enterococcus faecalis. Antimicrob Agents Chemother 1999;43:157-160.  [PubMed]

44. Cooper GS, Shlaes DM, Jacobs MR, Salata RA. The role of Enterococcus in intraabdominal infections: case control analysis. Infect Dis Clin Pract 1993;2:332-9.  [PubMed]

45. Coque TM, Arduino RC, Murray BE. High-level resistance to aminoglycosides: comparison of community and nosocomial fecal isolates of enterococci. Clin Infect Dis 1995;20:1048-51. [PubMed]

46. Cormican MG, Marshall SA, Jones RN. Preliminary interpretive criteria for disk diffusion susceptibility testing of SCH27899, a compound in the everninomicin class of antimicrobial agents. Diagn Microbiol Infect Dis 1995;23:157-60. [PubMed]

47. Critchley IA, Draghi DC, Sahm DF, Thornsberry C, Jones ME, Karlowsky JA. Activity of daptomycin against susceptible and multidrug-resistant Gram-positive pathogens collected in the SECURE study (Europe) during 2000-2001. J Antimicrob Chemother. 2003 Mar; 51:639-49.  [PubMed]

48. Dajani AS, Taubert KA, Wilson W, Bolger AF, Bayer A, Ferrieri P, Gewitz MH, Shulman ST, Nouri S, Newburger JW, Hutto C, Pallasch TJ, Gage TW, Levison ME, Peter G, Zuccaro G, Jr. Prevention of bacterial endocarditis. Recommendations by the American Heart Association. JAMA 1997;277:1794-801. [PubMed]

49. De Baere T, Claeys G, Verschraegen G, Devriese LA, Baele M, Van Vlem B, Vanholder R, Dequidt C, Vaneechoutte M. Continuous ambulatory peritoneal dialysis peritonitis due to Enterococcus cecorum . J Clin Microbiol 2000. 38:3511-3512. [PubMed]

50. Dever LL, Smith SM, DeJesus D, Masurekar M, Patel D, Kaminski ZC, Johanson WG Jr. Treatment of vancomycin-resistant Enterococcus faecium infections with an investigational streptogramin antibiotic (quinupristin/dalfopristin): a report of fifteen cases. Microbial Drug Resist 1996;2:407-13. [PubMed]

51. DiazGranados CA, Zimmer SM, Klein M, Jernigan JA. Comparison of mortality associated with vancomycin-resistant and vancomycin-susceptible enterococcal bloodstream infections: a meta-analysis. Clin Infect Dis 2005;41:327-333.  [PubMed]

52. Dibo I, Pillai SK, Gold HS, Baer MR, Wetzler M, Slack JL, Hazamy PA, Ball D, Hsiao CB, McCarthy PL Jr, Segal BH. Linezolid-resistant Enterococcus faecalis isolated from a cord blood transplant recipient. J Clin Microbiol. 2004 Apr;42:1843-5.  [PubMed]

53. DiNubile MJ, Chow JW, Satishchandran V, Polis A, Motyl MM, Abramson MA, Teppler H. Acquisition of resistant bowel flora during a double-blind randomized clinical trial of ertapenem versus piperacillin-tazobactam therapy for intraabdominal infections. Antimicrob Agents Chemother 2005;49:3217-3221.  [PubMed]

54. Dodge RA, Daly JS, Davaro R, Glew RH. High-dose ampicillin plus streptomycin for treatment of a patient with severe infection due to multiresistant enterococci. Clin Infect Dis 1997;25:1269-70. [PubMed]

55. Donabedian S, Hershberger E, Thal LA, Chow JW, Clewell DB, Robinson-Dunn B, Zervos MJ.  PCR fragment length polymorphism analysis of vancomycin-resistant Enterococcus faecium. J Clin Microbiol. 2000 ;38:2885-8.  [PubMed]

56. Donabedian SM, Thal LA, Hershberger E, Perri MB, Chow JW, Bartlett P, Jones R, Joyce K, Rossiter S, Gay K, Johnson J, Mackinson C, Debess E, Madden J, Angulo F, Zervos MJ. Molecular characterization of gentamicin-resistant Enterococci in the United States: evidence of spread from animals to humans through food. J Clin Microbiol. 2003 ;41:1109-13. [PubMed]

57. Donskey CJ, Chowdhry TK, Hecker MT, et al. Effect of antibiotic therapy on the density of vancomycin-resistant enterococci in the stool of colonized patients. N Engl J Med 2000; 343:1925-1932. [PubMed]

58. Edmond MB, Ober JF, Weinbaum DL, Pfaller MA, Hwang T, Sanford MD, Wenzel RP. Vancomycin-resistant Enterococcus faecium bacteremia: risk factors for infection. Clin Infect Dis 1995; 20:1126-1133. [PubMed]

59. Eliopoulos GM. Aminoglycoside resistant enterococcal endocarditis. Infect Dis Clinics N Am 1993;7:117-33. [PubMed]

60. Eliopoulos GM. Quinupristin-Dalfopristin and linezolid: evidence and opinion. Clin Infect Dis 2003;36:473-81. [PubMed]

61. Eliopoulos GM, Eliopoulos CT. Therapy of enterococcal infections. Eur J Clin Microbiol Infect Dis 1990;9:118-26. [PubMed]

62. Eliopoulos GM, Farber BF, Murray BE, Wennersten C, Moellering RC Jr. Ribosomal resistance of clinical enterococcal isolates to streptomycin. Antimicrob Agents Chemother 1984;25:398-9. [PubMed]

63. Eliopoulos GM, Wennersten CB, Gold HS, Moellering RC Jr. In vitro activities of new oxazolidinone antimicrobial agents against enterococci. Antimicrob Agents Chemother 1996;40:1745-7. [PubMed]

64. El-Khoury J, Fishman JA. Linezolid in the treatment of vancomycin-resistant Enterococcus faecium in solid organ transplant recipients: report of a multicenter compassionate-use trial.Transpl Infect Dis. 2003 Sep;5:121-5. [PubMed]

65. Facklam RR, Collins MD. Identification of Enterococcus species isolated from human infections by a conventional test scheme. J Clin Microbiol 1989;27:731-4. [PubMed]

66. Falk PS, Winnike J, Woodmansee C, Desai M, Mayhall CG. Outbreak of vancomycin-resistant enterococci in a burn unit. Infect Control Hosp Epidemiol 2000; 21:575-582.  [PubMed]

67. Fang G, Brennen C, Wagener M, Swanson D, Hilf M, Zadecky L, DeVine J, Yu VL. Use of ciprofloxacin versus use of aminoglycoside for therapy of complicated urinary tract infection: prospective, randomized clinical and pharmacokinetic study. Antimicrob Agents Chemother 1991;35:1849-55. [PubMed]

68. Fantin B, Carbon C. Importance of the aminoglycoside dosing regimen in the penicillin-netilmicin combination for treatment of Enterococcus faecalis-induced experimental endocarditis. Antimicrob Agents Chemother 1990;34:2387-91. [PubMed]

69. Ferretti JJ, Gilmore KS, Courvalin P. Nucleotide sequence analysis of the gene specifying the bifunctional 6’-aminoglycoside acetyltransferase 2”-aminoglycoside phosphotransferase enzyme in Streptococcus faecalis and identification and cloning of gene regions specifying the two activities. J Bacteriol 1986;167:631-8. [PubMed]

70. Flannagan SE, Chow JW, Donabedian SM, Brown WJ, Perri MB, Zervos MJ, Ozawa Y, Clewell DB. Plasmid content of a vancomycin-resistant Enterococcus faecalis isolate from a patient also colonized by Staphylococcus aureus with a VanA phenotype. Antimicrob Agents Chemother. 2003 ;47:3954-9. [PubMed]

71. Fontana R, Aldegheri M, Ligozzi M, Lopez H, Sucari A, Satta G. Overproduction of a low-affinity penicillin-binding protein and high-level ampicillin resistance in Enterococcus faecium. Antimicrob Agents Chemother 1994;38:1980-3. [PubMed]

72. Fontana R, Cerini R, Longoni P, Grossato A, Canepari P. Identification of a streptococcal penicillin-binding protein that reacts very slowly with penicillin. J Bacteriol 1983;155:1343-50. [PubMed]

73. Ford CW, Hamel JC, Wilson DM, Moerman JK, Stapert D, Yancey RJ Jr, Hutchinson DK, Barbachyn MR, Brickner SJ. In vivo activities of U-100592 and U-100766, novel oxazolidinone antimicrobial agents, against experimental bacterial infections. Antimicrob Agent Chemother 1996;40:1508-13. [PubMed]

74. Fortun J, Coque TM, Martin-Davila P, Moreno L, Canton R, Loza E, Baquero F, Moreno S. Risk factors associated with ampicillin resistance in patients with bacteraemia caused by Enterococcus faecium .J Antimicrob Chemother. 2002 Dec;50:1003-9.  [PubMed]

75. Foulds G. Pharmacokinetics of sulbactam/ampicillin in humans: a review. Rev Infect Dis 1986;8(suppl 5):S503-11. [PubMed]

76. Fraise AP, Andrews J, Wise R. Activity of a new glycopeptide antibiotic (LY333328) against enterococci and other resistant Gram-positive organisms. J Antimicrob Chemother 1997;40:423-5. [PubMed]

77. Fraimow HS, Venuti E. Inconsistent bactericidal activity of triple-combination therapy with vancomycin, ampicillin, and gentamicin against vancomycin-resistant, highly ampicillin-resistant Enterococcus faecium. Antimicrob Agents Chemother 1992;36:1563-6.  [PubMed]

78. Francioli P. Antibiotic treatment of streptococcal and enterococcal endocarditis: an overview. Eur Heart J 1995;16(Suppl B):75-9. [PubMed]

79. Fridkin SK, Edwards JR, Courval JM, Hill H, Tenover FC, Lawton R, Gaynes RP, McGowan JE Jr. The effect of vancomycin and third-generation cephalosporins on prevalence of vancomycin-resistant enterococci in 126 U.S. adult intensive care units. Ann Int Med 2001;135:175-183.  [PubMed]

80. Fuchs PC, Barry AL, Brown SD. In vitro activities of SCH27899 alone and in combination with 17 other antimicrobial agents. Antimicrob Agents Chemother 1999;43:2996-7. [PubMed]

81. Furlong WB, Rakowski TA. Therapy with RP 59500 (quinupristin/dalfopristin) for prosthetic valve endocarditis due to enterococci with vanA/vanB resistance patterns. Clin Infect Dis 1997;25:163-4. [PubMed]

82. Gales AC, Jones RN. Antimicrobial activity and spectrum of the new glycylcycline, GAR-936 tested against 1,203 recent clinical bacterial isolates. Diagn Microbiol Infect Dis 2000;36:19-36. [PubMed]

83. Gavalda J, Torres C, Tenorio C, López P, Zaragoza M, Capdevila JA, Almirante B, Ruiz F, Borrell N, Gomis X, Pigrau C, Baquero F, Pahissa A. Efficacy of ampicillin plus ceftriaxone in treatment of experimental endocarditis due to Enterococcus faecalis strains highly resistant to aminoglycosides. Antimicrob Agents Chemother 1999;43:639-46.  [PubMed]

84. Georgopapadakou NH. Penicillin-binding proteins and bacterial resistance to beta-lactams. Antimicrob Agents Chemother 1993;37:2045-53. [PubMed]

85. Gilmore MS, Coburn PS, Nallapareddy SR, Murray BE. Enterococcal virulence. In: Gilmore MS, ed. The Enterococci. Pathogenesis, Molecular Biology, and Antibiotic Resistance. Washington, DC: ASM Press; 2002:301-354  [PubMed].

86. Gonzales RD, Schreckenberger PC, Graham MB, Kelkar S, DenBesten K, Quinn JP. Infections due to vancomycin-resistant Enterococcus faecium resistant to linezolid. Lancet 2001. 357:1179. [PubMed]

87. Gordon S, Swenson JM, Hill BC, Pigott NE, Facklam RR, Cooksey RC, Thornsberry C, Enterococcal Study Group, Jarvis WR, Tenover FC. Antimicrobial susceptibility patterns of common and unusual species of enterococci causing infections in the United States. J Clin Microbiol 1992;30:2373-8. [PubMed]

88. Graninger W, Ragette R. Nosocomial bacteremia due to Enterococcus faecalis without endocarditis. Clin Infect Dis 1992;15:49-57. [PubMed]

89. Gransden WR, King A, Marossy D, Rosenthal E. Quinupristin/dalfopristin in neonatal Enterococcus faecium meningitis. Arch Dis Child Fetal Neonatal Ed 1998;78:F235-6.  [PubMed]

90. Gray JW, Darbyshire PJ, Beath SV, Kelly D, Mann JR. Experience with quinupristin/dalfopristin in treating infections with vancomycin-resistant Enterococcus faecium in children. Pediatr Infect Dis J 2000;19:234-8. [PubMed]

91. Grayson ML, Eliopoulos GM, Wennersten CB, Ruoff KL, De Girolami PC, Ferraro MJ, Moellering RC Jr. Increasing resistance to beta-lactam antibiotics among clinical isolates of Enterococcus faecium: a 22-year review at one institution. Antimicrob Agents Chemother 1991;35:2180-4. [PubMed]

92. Green M, Binczewski B, Pasculle AW, Edmund M, Barbadora K, Kusne S, Shlaes DM. Constitutively vancomycin-resistant Enterococcus faecium resistant to synergistic beta-lactam combinations. Antimicrob Agents Chemother 1993;37:1238-42. [PubMed]

93. Greub G, Devriese LA, Pot B, Dominguez J, Bille J. Enterococcus cecorum septicemia in a malnourished adult patient. Eur J Clin Microbiol Infect Dis 1997;16:594-8. [PubMed]

94. Guay DRP. Dalbavancin: an investigational glycopeptide. Expert Rev Anti Infect Ther 2004;2:845-52. [PubMed]

95. Gullberg RM, Homann SR, Phair JP. Enterococcal bacteremia: analysis of 75 episodes. Rev Infect Dis 1989;11:74-85. [PubMed]

96. Gutmann L, Al-Obeid S, Billot-Klein D, Guerrier M-L, Collatz E. Synergy and resistance to synergy between beta-lactam antibiotics and glycopeptides against glycopeptide-resistant strains of Enterococcus faecium. Antimicrob Agents Chemother 1994;38:824-9. [PubMed]

97. Hachem R, Afif C, Gokasian, Raad I. Successful treatment of vancomycin-resistant Enterococcus meningitis with linezolid. Eur J Clin Microbiol Infect Dis 2001;20:432-434.  [PubMed]

98. Hachem R, Raad I. Failure of oral antimicrobial agents in eradicating gastrointestinal colonization with vancomycin-resistant enterococci. Infect Control Hosp Epidemiol. 2002;23:43-4. [PubMed]

99. Handwerger SD, Perlman C, Altarac D, McAuliffe V. Concomitant high-level vancomycin and penicillin resistance in clinical isolates of enterococci. Clin Infect Dis 1992;14:655-61. [PubMed]

100. Harbarth S, Cosgrove S, Carmeli Y. Effects of antibiotics on nosocomial epidemiology of vancomycin-resistant enterococci. Antimicrob Agents Chemother 2002;46:1619-1628.  [PubMed]

101. Hayden MK. Insights into the epidemiology and control of infection with vancomycin-resistant enterococci. Clin Infect Dis 2000; 31:1058-1065. [PubMed]     

102. Hayden MK, Trenholme GM, Schultz JE, Sahm DF. In vivo development of teicoplanin resistance in a VanB Enterococcus faecium isolate. J Infect Dis 1993;167:1224-7.[PubMed]

103. Hayden M, Koenig GI, Trenholme GM. Bactericidal activities of antibiotics against vancomycin-resistant Enterococcus faecium blood isolates and synergistic activities of combinations. Antimicrob Agents Chemother 1994;38:1225-9.[PubMed]

104. Hershberger E, Donabedian S, Konstantinou K, Zervos MJ. Quinupristin-dalfopristin resistance in gram-positive bacteria: mechanism of resistance and epidemiology. Clin Infect Dis. 2004;38:92-8. [PubMed]

105. Hershberger E, Oprea SF, Donabedian SM, Perri M, Bozigar P, Bartlett P, Zervos MJ.  Epidemiology of antimicrobial resistance in enterococci of animal origin. J Antimicrob Chemother. 2005 ;55:127-30. [PubMed]

106. Hoge CW, Adams J, Buchanan B, Sears SD. Enterococcal bacteremia: to treat or not to treat, a reappraisal. Rev Infect Dis 1991;13:600-5.[PubMed]

107. Hoogkamp-Korstanje JAA, Roelofs-Willemse J. Comparative in vitro activity of moxifloxacin against Gram-positive clinical isolates. J Antimicrob Chemother 2000;45:31-9. [PubMed]

108. Howe RA, Robson M, Oakhill A, Cornish JM, Millar MR. Successful use of tetracycline as therapy of an immunocompromised patient with septicaemia caused by a vancomycin-resistant enterococcus. J Antimicrob Chemother 1997;40:144-5.[PubMed]

109. Hsueh PR, Chen WH, Teng LJ, Luh KT. Nosocomial infections due to methicillin-resistant Staphylococcus aureus and vancomycin-resistant enterococci at a university hospital in Taiwan from 1991 to 2003: resistance trends, antibiotic usage and in vitro activities of newer antimicrobial agents. Int J Antimicrob Agents. 2005 Jul;26:43-9. [PubMed]

110. Hsueh PR, Teng LJ, Chen YC, Yang PC, Ho SW, Luh KT. Recurrent bacteremic peritonitis caused by Enterococcus cecorum in a patient with liver cirrhosis. J Clin Microbiol 2000;38:2450-2452.[PubMed]

111. Inoue M, Nonoyama M, Okamoto R, Ida T. Antimicrobial activity of arbekacin, a new aminoglycoside antibiotic, against methicillin-resistant Staphylococcus aureus. Drugs Exp Clin Res 1994;22:233-40.[PubMed]

112. Jensen VF, Neimann J, Hammerum AM, Molbak K, Wegener HC. Does the use of antibiotics in food animals pose a risk to human health? An unbiased review? J Antimicrob Chemother. 2004;54:274-5.[PubMed]

113. Johnson AP, Mushtaq S, Warner M, Livermore DM. Activity of daptomycin against multi-resistant Gram-positive bacteria including enterococci and Staphylococcus aureus resistant to linezolid. Int J Antimicrob Agents. 2004 Oct;24:315-9. 69. [PubMed]

114. Jones RN. Disk diffusion susceptibility test development for the new glycylcycline, GAR-936. Diagn Microbiol Infect Dis 1999;35:249-52.[PubMed]

115. Jones RN, Barrett MS. Antimicrobial Activity of SCH 27899, Oligosaccharide Member of the Everninomycin Class with a Wide Gram-Positive Spectrum. Clin Microbiol Infect 1995;1:35-43.[PubMed]

116. Jones RN, Barrett MS, Erwin ME. In vitro activity and spectrum of LY333328, a novel glycopeptide derivative. Antimicrob Agents Chemother 1997;41:488-93.[PubMed]

117. Jones RN, Beach ML, Pfaller MA, Doern GV. Antimicrobial activity of gatifloxacin tested against 1676 strains of ciprofloxacin-resistant Gram-positive cocci isolated from patient infections in North and South America. Diagn Microbiol Infect Dis 1998;32:247-52.[PubMed]

118. Jones RN, Kugler KC, Erwin ME, Biedenbach DJ, Beach ML, Pfaller MA, The Quality Control Study Group. Gatifloxacin (AM-1155, CG 5501) susceptibility testing interpretive criteria and quality control guidelines for dilution and disk (5-mg) diffusion methods. Diagn Microbiol Infect Dis 1999;33:247-53.[PubMed]

119. Jones RN, Marshall SA, Erwin ME, The Quality control Study Group. Antimicrobial activity and spectrum of SCH27899 (Ziracinâ) tested against gram-positive species including recommendations for routine susceptibility testing methods and quality control. Diagn Microbiol Infect Dis 1999;34:103-10.[PubMed]

120. Kak V, Donabedian SM, Zervos MJ, Kariyama R, Kumon H, Chow JW. Efficacy of ampicillin plus arbekacin in experimental rabbit endocarditis caused by an Enterococcus faecalis strain with high-level gentamicin resistance Antimicrob Agents Chemother 2000; 44:2545-2546.[PubMed]

121. Kak V, You I, Zervos MJ, Kariyama R, Kumon H, Chow JW. In-vitro synergistic activity of the combination of ampicillin and arbekacin against vancomycin- and high-level gentamicin-resistant Enterococcus faecium with the aph(2”)-Id gene. Diagn Microbiol Infect Dis 2000; 37:297-299.[PubMed]

122. Kapur D, Dorsky D, Feingold JM, Bona RD, Edwards RL, Aslanzadeh J, Tutschka PJ, Bilgrami S. Incidence and outcome of vancomycin-resistant Enterococcal bacteremia following autologous peripheral blood stem cell transplantation. Bone Marrow Transplant 2000;25:147-52. [PubMed]

123. Kariyama R, Kumon H, Chow L, Zervos MJ, Takata T, Tabata M, Chow JW. In-vitro activity of the combination of ampicillin and arbekacin against high-level gentamicin-resistant enterococci. J Antimicrob Chemother 1998;42:836-8.[PubMed]

124. Kaye D. Treatment of infective endocarditis. Ann Intern Med 1996;124:606-8. [PubMed]

125. Koenig MG, Kaye D. Enterococcal endocarditis: report of nineteen cases with long-term follow-up data. N Engl J Med 1961;264:257-64.[PubMed]

126. Konstantinov IE, Zehr KJ. Aortic root replacement in a patient with vancomycin-resistant Enterococcus faecium endocarditis and leukemia. Chest 2001;120:1744-6.[PubMed]

127. Krcméry V Jr, Filka J, Uher J, Kurak H, Šagát T, Tuharský J, Novák I, Urbanová T, Kralinský K, Mateicka F, Krcméryová T, Jurga L, Šulcová M, Štencl J, Krúpová I. Ciprofloxacin in treatment of nosocomial meningitis in neonates and in infants: report of 12 cases and review. Diagn Microb Infect Dis 1999;35:75-80.[PubMed]

128. Krogstad DJ, Korfhagen TR, Moellering RC Jr, Wennersten C, Swartz MN, Perzynski S, Davies J. Aminoglycoside-inactivating enzymes in clinical isolates of Streptococcus faecalis: an explanation for antibiotic synergism. J Clin Invest 1978;62:480-6.[PubMed]

129. Lai KK. Treatment of vancomycin-resistant Enterococcus faecium infections. Arch Intern Med 1996;156:2579-84.[PubMed]

130. Lam S, Singer C, Tucci V, Morthland VH, Pfaller MA, Isenberg HD. The challenge of vancomycin-resistant enterococci: a clinical and epidemiologic study. Am J Infect Control 1995;23:170-80.[PubMed]

131. Landman D, Mobarakai NK, Quale JM. Novel antibiotic regimens against Enterococcus faecium resistant to ampicillin, vancomycin, and gentamicin. Antimicrob Agents Chemother 1993;37:1904-8.[PubMed]

132. Landman D, Quale JM, Mobarakai N, Zaman MM. Ampicillin plus ciprofloxacin therapy of experimental endocarditis caused by multidrug-resistant Enterococcus faecium. J Antimicrob Chemother 1995;36:253-8.[PubMed]

133. Lautenbach E, Schuster MG, Bilker WB, Brennan PJ. The role of chloramphenicol in the treatment of bloodstream infection due to vancomycin-resistant Enterococcus. Clin Infect Dis 1998;27:1259-65. [PubMed]

134. Lavoie SR, Wong ES, Coudron PE, Williams DS, Markowitz SM. Comparison of ampicillin-sulbactam with vancomycin for treatment of experimental endocarditis due to a beta-lactamase-producing, highly gentamicin-resistant isolate of Enterococcus faecalis. Antimicrob Agents Chemother 1993;37:1447-51.[PubMed]

135. Leclercq R, Bingen E, Su QH, Lambert-Zechovski N, Courvalin P, Duval J. Effects of combinations of beta-lactams, daptomycin, gentamicin, and glycopeptides against glycopeptide-resistant enterococci. Antimicrob Agents Chemother 1991;35:92-8.[PubMed]

136. Leclercq R, Courvalin P. Resistance to glycopeptides in enterococci. Clin Infect Dis 1997;24:545-56.[PubMed]

137. Lee PY, Das SS, Stevens PJ. Achieving bactericidal therapy and high-level aminoglycoside resistance. J Antimicrob Chemother 1993;31:608-9.[PubMed]

138. Lefort A, Saleh-Mghir A, Garry L, Carbon C, Fantin B. Activity of LY333328 combined with gentamicin in vitro and in rabbit experimental endocarditis due to vancomycin-susceptible or -resistant Enterococcus faecalis . Antimicrob Agents Chemother 2000. 44:3017-3021.[PubMed]

139. Levine DP, Holley HP, Eiseman I, Willcox P, Tack K. Clinafloxacin for the treatment of bacterial endocarditis. Clin Infect Dis. 2004 ;38:620-31. [PubMed]

140. Lewis JS 2nd, Owens A, Cadena J, Sabol K, Patterson JE, Jorgensen JH. Emergence of daptomycin resistance in Enterococcus faecium during daptomycin therapy. Antimicrob Agents Chemother. 2005 Apr;49:1664-5.[PubMed]

141. Ligozzi M, Pittaluga F, Fontana R. Identification of a genetic element (psr) which negatively controls expression of Enterococcus hirae penicillin-binding protein 5. J Bacteriol 1993;175:2046-51.[PubMed]

142. Ligozzi M, Pittaluga F, Fontana R. Modification of penicillin-binding protein 5 associated with high-level ampicillin resistance in Enterococcus faecium. Antimicrob Agents Chemother 1996;40:354-7.[PubMed]

143. Lin AH, Murray RW, Vidmar TJ, Marotti KR. The oxazolidinone eperezolid binds to the 50S ribosomal subunit and competes with binding of chloramphenicol and lincomycin. Antimicrob Agents Chemother 1997;41:2127-31.[PubMed]

144. Linden PK. Treatment options for vancomycin-resistant enterococcal infections. Drugs. 2002;62(3):425-41. [PubMed]

145. Linden PK, Pasculle AW, Manez R, Kramer DJ, Fung JJ,Pinna AD, Kusne S. Differences in outcomes for patients with bacteremia due to vancomycin-resistant Enterococcus faecium or vancomycin-susceptible E. faecium. Clin Infect Dis 1996;22:663-70.[PubMed]

146. Linden PK, Pasculle AW, McDevitt D, Kramer DJ. Effect of quinupristin/dalfopristin on the outcome of vancomycin-resistant Enterococcus faecium bacteraemia: comparison with a control cohort. J Antimicrob Chemother 1997;39 (suppl A):145-51.[PubMed]

147. Livornese LL Jr, Dias S, Samel C, Romanowski B, Taylor S, May P, Pitsakis P, Woods G, Kaye D, Levison ME, Johnson CC. Hospital-acquired infection with vancomycin-resistant Enterococcus faecium transmitted by electronic thermometers. Ann Intern Med 1992;117:112-6.[PubMed]

148. Loeb M, Salama S, Arm-evans M, Capretta G, Olde J. A case-control study to detect modifiable risk factors for colonization with vancomycin-resistant enterococci. Infect Control Hosp Epidemiol 1999; 20:760-763.[PubMed]

149. Losonsky GA, Wolf A, Schwalbe RS, Nataro J, Gibson CB, Lewis EW. Successful treatment of meningitis due to multiply resistant Enterococcus faecium with a combination of intrathecal teicoplanin and intravenous antimicrobial agents. Clin Infect Dis 1994;19:163-5.[PubMed]

150. Lynch C, Courvalin P, Nikaido H. Active efflux of antimicrobial agents in wild-type strains of enterococci. Antimicrob Agents Chemother 1997;41:869-871.[PubMed]

151. Lynn WA, Clutterbuck E, Want S, Markides V, Lacey S, Rogers TR, Cohen J. Treatment of CAPD-peritonitis due to glycopeptide-resistant Enterococcus faecium with quinupristin/dalfopristin. Lancet 1994;344:1025-6.[PubMed]

152. Mainardi JL, Gutmann L, Acar JF, Goldstein FW. Synergistic effect of amoxicillin and cefotaxime against Enterococcus faecalis. Antimicrob Agents Chemother 1995;39:1984-7.[PubMed]

153. Maki DG, Agger WA. Enterococcal bacteremia: clinical features, the risk of endocarditis, and management. Medicine (Baltimore) 1988;67:248-69.[PubMed]

154. Malabarba A, Goldstein BP. Origin, structure, and activity in vitro and in-vivo of dalbavancin. J Antimicrob Chemo 2005;55:ii15-20. [PubMed]

155. Malani PN, Thal L, Donabedian SM, Robinson-Dunn B, Kauffman CA, Chow JW, Hershberger E, Zervos MJ. Molecular analysis of vancomycin-resistant Enterococcus faecalis from Michigan hospitals during a 10 year period. J Antimicrob Chemother. 2002 May;49:841-3. [PubMed]

156. Malathum K, Singh KV, Murray BE. In vitro activity of moxifloxacin, a new 8-methoxyquinolone, against Gram-positive bacteria. Diagn Microb Infect Dis 1999;35:127-33.[PubMed]

157. Mandell GL, Kaye D, Levison ME, Hook EW. Enterococcal endocarditis: an analysis of 38 patients observed at the New York Hospital-Cornell Medical Center. Arch Intern Med 1970;125:258-64.[PubMed]

158. Marangos MN, Nicolau DP, Quintiliani R, Nightingale CH. Influence of gentamicin dosing interval on the efficacy of penicillin-containing regimens in experimental Enterococcus faecalis endocarditis. J Antimicrob Chemother 1997;39:519-22. [PubMed]

159. Matsumura S, Simor AE. Treatment of endocarditis due of vancomycin-resistant Enterococcus faecium with quinupristin/dalfopristin, doxycycline, and rifampin: a synergistic drug combination. Clin Infect Dis 1998;27:1554-6.[PubMed]

160. McNeil SA, Clark NM, Chandrasekar PH, Kauffman CA. Successful treatment of vancomycin-resistant Enterococcus faecium bacteremia with linezolid after failure of treatment with Synercid (quinupristin/dalfopristin). Clin Infect Dis 2000;30:403-4.[PubMed]

161. Megran DW. Enterococcal endocarditis. Clin Infect Dis 1992;15:63-71.[PubMed]

162. Mekonen ET, Noskin GA, Hacek DM, Peterson LR. Successful treatment of persistent bacteremia due to vancomycin-resistant, ampicillin-resistant Enterococcus faecium. Microb Drug Resist 1995;1:249-53.[PubMed]

163. Mellman RL, Spisak GM, Burakoff R. Enterococcus avium bacteremia in association with ulcerative colitis. Am J Gast 1992;87:337-8.[PubMed]

164. Millichap J, Ristow RA, Noskin GA, Peterson LR. Selection of Enterococcus faecium strains with stable and unstable resistance to the streptogramin RP 59500 using stepwise in vitro exposure. Diagn Microbiol Infect Dis 1996;25:15-20.[PubMed]

165. Melzer M, Goldsmith D, Gransden W. Successful treatment of vertebral osteomyelitis with linezolid in a patient receiving hemodialysis and with persistent methicillin-resistant Staphylococcus aureus and vancomycin-resistant Enterococcus bacteremias. Clin Infect Dis 2000. 31:208-209.[PubMed]

166. Moellering RC Jr. The enterococcus: a classic example of the impact of antimicrobial resistance on therapeutic options. J Antimicrob Chemother 1991;28:1-12.[PubMed]

167. Moellering RC Jr. Emergence of enterococcus as a significant pathogen. Clin Infect Dis 1992;14:1173-8.[PubMed]

168. Moellering RC. Linezolid: the first oxazolidinone antimicrobial. Ann Intern Med. 2003 Jan 21;138(2):135-42. [PubMed]

169. Moellering RC, Linden PK, Reinhardt J, Blumberg EA, Bompart F, Talbot GH. The efficacy and safety of quinupristin/dalfopristin for the treatment of infections caused by vancomycin-resistant Enterococcus faecium. J Antimicrob Chemother 1999;44:251-61.[PubMed]

170. Moellering RC Jr, Murray BE, Schoenbaum SC, Adler J, Wennersten CB. A novel mechanism of resistance to penicillin-gentamicin synergism in Streptococcus faecalis. J Infect Dis 1980;141:81-6.[PubMed]

171. Moellering RC Jr, Weinberg AN. Studies on antibiotic synergism against enterococci. II. Effect of various antibiotics on the uptake of 14C-labelled streptomycin by enterococci. J Clin Invest 1971;50:2580-4.[PubMed]  

172. Moellering RC Jr, Wennersten C, Weinberg AN. Studies on antibiotic synergism against enterococci. I. Bacteriologic studies. J Lab Clin Med 1971;77:821-8.[PubMed]

173. Montecalvo MA, Jarvis WR, Uman J, Shay DK, Petrullo C, Rodney K, Gedris C, Horowitz HW, Wormser GP. Infection-control measures reduce transmission of vancomycin-resistant enterococci in an endemic setting. Ann Intern Med 1999; 131:269-272.[PubMed]

174. Moreno F, Jorgensen JH, Weiner MH. An old antibiotic for a new multiple-resistant Enterococcus faecium? Diagn Microbiol Infect Dis 1994;20:41-3.[PubMed]

175. Morris JT, Cooper RH. Intravenous streptomycin: a useful route of administration. Clin Infect Dis 1994;19:1150-1.[PubMed]

176. Morris JG Jr, Shay DK, Hebden JN, McCarter RJ Jr, Perdue BE, Jarvis W, Johnson JA, Dowling TC, Polish LB, Schwalbe RS. Enterococci resistant to multiple antimicrobial agents, including vancomycin. Ann Intern Med 1995; 123:250-259.[PubMed]

177. Mouton Y, Deboscker Y. Les beta-lactamines. Encycl Med Chir Paris Ther 1983;25007:B10-20.[PubMed]

178. Munoz-Price LS, Lolans K, Quinn JP. Emergence of resistance to daptomycin during treatment of vancomycin-resistant Enterococcus faecalis infection. Clin Infect Dis. 2005;41:565-6. [PubMed]

179. Muralidharan G, Micalizzi M, Speth J, Raible D, Troy S. Pharmacokinetics of tigecycline after a single and multiple doses in healthy subjects. Antimicrob Agents Chemother 2005;49:220-9. [PubMed]

180. Murphy TM, Deitz JM, Petersen PJ, Mikels SM, Weiss WJ. Therapeutic efficacy of GAR-936, a novel glycylcycline, in a rat model of experimental endocarditis. Antimicrob Agents Chemother 2000. 44:3022-3027.[PubMed]

181. Murray BE. The life and times of the Enterococcus. Clin Microbiol Rev 1990;3:46-65.[PubMed]

182. Murray BE. beta-lactamase-producing enterococci. Antimicrob Agents Chemother 1992;36:2355-9.[PubMed]

183. Murray BE. Vancomycin-resistant enterococcal infections. N Engl J Med 2000;342:710-21.[PubMed]

184. Murray BE, Singh KV, Markowitz SM, Lopardo HA, Patterson JE, Zervos MJ, Rubeglio E, Eliopoulos GM, Rice LB, Goldstein FW, Jenkins SG, Caputo GM, Nasnas R, Moore LS, Wong ES, Weinstock G. Evidence for clonal spread of a single strain of beta-lactamase-producing Enterococcus (Streptococcus) faecalis to six hospitals in five states. J Infect Dis 1991;163:780-5.[PubMed]

185. Mutnick AH, Enne V, Jones RN. Linezolid resistance since 2001: SENTRY Antimicrobial Surveillance Program.Ann Pharmacother. 2003 ;37:769-74. [PubMed]

186. Nachtman A, Verma R, Egnor M. Vancomycin-resistant Enterococcus faecium shunt infection in an infant: an antibiotic cure. Microbial Drug Resist 1995;1:95-6.[PubMed]

187. Nakashio S, Iwasawa H, Don FY, Kanemitso K, Shimada I. Everninomicin, a new oligosaccharide antibiotic: its antimicrobial activity, post-antibiotic effect and synergistic bactericidal activity. Drugs Exptl Clin Res 1995;1:7-16.[PubMed]

188. Navarro F, Courvalin P. Analysis of genes encoding D-alanine-D-alanine ligase-related enzymes in Enterococcus casseliflavus and Enterococcus flavescens. Antimicrob Agents Chemother 1994;38:1788-93.[PubMed]

189. Norris AH, Reilly JP, Edelstein PH, Brennan PJ, Schuster MG. Chloramphenicol for the treatment of vancomycin-resistant enterococcal infections. Clin Infect Dis 1995;20:1137-44. [PubMed]

190. Noskin GA. Tigecycline: a new glycylcycline for treatment of serious infection. Clin Infect Dis 2005;41:S303-14. [PubMed]

191. Noskin GA, Peterson LR, Warren JR. Enterococcus faecium and Enterococcus faecalis bacteremia: acquisition and outcome. Clin Infect Dis 1995;20:296-301. [PubMed]

192. Noskin GA, Siddiqui F, Stosor V, Hacek D, Peterson LR. In vitro activities of linezolid against important Gram-positive bacterial pathogens including vancomycin-resistant enterococci. Antimicrob Agents Chemother 1999;43:2059-62.[PubMed]

193. Noskin GA, Siddiqui F, Stosor V, Kruzynski J, Peterson LR. Successful treatment of persistent vancomycin-resistant Enterococcus faecium bacteremia with linezolid and gentamicin. Clin Infect Dis 1999;28:689-90.[PubMed]

194. Novais C, Sousa JC, Coque TM, Peixe LV; Portuguese Resistance Study Group. In vitro activity of daptomycin against enterococci from nosocomial and community environments in Portugal. J Antimicrob Chemother. 2004 Nov;54:964-6. [PubMed]

195. Olaison L, Schadewitz K; Swedish Society of Infectious Diseases Quality Assurance Study Group for Endocarditis. Enterococcal endocarditis in Sweden, 1995-1999: can shorter therapy with aminoglycosides be used? Clin Infect Dis. 2002;34:159-66. [PubMed]

196. Oprea SF, Zaidi N, Donabedian SM, Balasubramaniam M, Hershberger E, Zervos MJ.  Molecular and clinical epidemiology of vancomycin-resistant Enterococcus faecalis. J Antimicrob Chemother. 2004;53:626-30. [PubMed]

197. Osakabe Y, Takahashi Y, Narihara K. The utility and dosage and administration of arbekacin in patients with MRSA infection. Antibiotics Chemother 1996;12:120-7. [PubMed]

198. Ostrowsky BE, Trick WE, Sohn AH, et al. Control of vancomycin-resistant enterococcus in health care facilities in a region. N Engl J Med 2001; 344:1427-1433.[PubMed]

199. Ounissi H, Courvalin P. Appendix B. Nucleotide sequences of streptococcal genes. In: Ferretti JJ, Curtis R III, eds. Streptococcal genetics. Washington, DC: American Society for Microbiology; 1987:275.[PubMed]

200. Pace JL, Judice JK. Telavancin. Curr Opin Investig Drugs. 2005 Feb;6:216-25. [PubMed]

201. Padiglione AA, Wolfe R, Grabsch EA, Olden D, Pearson S, Franklin C, Spelman D, Mayall B, Johnson PDR, Grayson ML. Risk factors for new detection of vancomycin-resistant enterococci in acute-care hospitals that employ strict infection control procedures. Antimicrob Agents Chemother 2003;47:2492-2498.[PubMed]

202. Papanicolaou GA, Meyers RR, Meyers J, Mendelson MH, Lou W, Emre S, Sheiner P, Miller C. Nosocomial infections with vancomycin-resistant Enterococcus faecium in liver transplant recipients: risk factors for acquisition and mortality. Clin Infect Dis 1996;23:760-6.[PubMed]

203. Patel R, Keating MR, Cockerill FR III, Steckelberg JM. Bacteremia due to Enterococcus avium. Clin Infect Dis 1993;17:1006-11.[PubMed]

204. Patel R, Rouse MS, Piper KE, Steckelberg JM. In vitro activity of linezolid against vancomycin-resistant enterococci, methicillin-resistant Staphylococcus aureus and penicillin-resistant Streptococcus pneumoniae. Diagn Microbiol Infect Dis 1999;34:119-22.[PubMed]

205. Patel RM, Rouse S, Piper KE, Steckelberg JM. Linezolid therapy of vancomycin-resistant Enterococcus faecium experimental endocarditis. Antimicrob Agents Chemother 2001. 45:621-623.[PubMed]

206. Paterson DL, Dominguez EA, Chang F-Y, Snydman DR, Singh N. Infective endocarditis in solid organ transplant patients, Clin Infect Dis 1998;26:689-94. [PubMed]

207. Patterson JE, Sweeney AH, Simms M, Carley N, Mangi R, Sabetta J, Lyons RW. An analysis of 110 serious enterococcal infections: epidemiology, antibiotic susceptibility, and outcome. Medicine (Baltimore) 1995;74:191-200.[PubMed]

208. Pérez Mato S, Robinson S, Bégué RE. Vancomycin-resistant Enterococcus faecium meningitis successfully treated with chloramphenicol. Ped Infect Dis J 1999;18:483-4. [PubMed]

209. Perri MB, Hershberger E, Ionescu M, Lauter C, Zervos MJ.   In vitro susceptibility of vancomycin-resistant enterococci (VRE) to fosfomycin. Diagn Microbiol Infect Dis. 2002 Apr;42:269-71. [PubMed]

210. Peterson LR, Noskin GA. New technology for detecting multidrug-resistant pathogens in the clinical microbiology laboratory. Emerg Infect Dis 2001;7: 1-12. [PubMed]

211. Peterson LR, Petzel RA, Clabots CR, Fasching CE, Gerding DN. Medical technologists using molecular epidemiology as part of the infection control team. Diagn. Microbiol. Infect. Dis. 1993;16:303-311. [PubMed]

212. Peterson PJ, Bradford PA, Weiss WJ, Murphy TM, Sum PE, Projan SJ. In vitro and in vivo activities of tigecycline (GAR-936), daptomycin, and comparative antimicrobial agents against glycopeptide-intermediate Staphylococcus aureus and other resistant gram-positive pathogens. Antimicrob Agents Chemother 2002;46:2595-601.[PubMed]

213. Petersen PJ, Jacobus NV, Weiss WJ, Sum PE, Testa RT. In vitro and in vivo antibacterial activities of a novel glycylcycline, the 9-t-bytylglycylamido derivative of minocycline (GAR-936). Antimicrob Agents Chemother 1999;43:738-44.[PubMed]

214. Physicians’ Desk Reference. 51st ed. Montvale, NJ: Medical Economics Company, Inc, 1997:2035.[PubMed]

215. Pintado V, Cabellos C, Moreno S, Meseguer MA, Ayats J, Viladrich PF. Enterococcal meningitis: a clinical study of 39 cases and review of the literature. Medicine (Baltimore). 2003 Sep;82:346-64. [PubMed]

216. Presterl E, Graninger W, Georgopoulos A. The efficacy of teicoplanin in the treatment of endocarditis caused by Gram-positive bacteria. J Antimicrob Chemother 1993;31:755-66.[PubMed]

217. Quale JM, Landman D, Mobarakai N. Treatment of experimental endocarditis due to multidrug resistant Enterococcus faecium with ciprofloxacin and novobiocin. J Antimicrob Chemother 1994;34:797-802.[PubMed]

218. Raad I, Darouiche R, Vazquez J, Lentnek, A, Hachem R, Hanna H, Goldstein B, Henkel T, Seltzer E. Efficacy and safety of weekly dalbavancin therapy for catheter-related bloodstream infection caused by gram-positive pathogens. Clin Infect Dis 2005;40:374-80. [PubMed]

219. Raad I, Hachem R, Hanna H, Afif C, Escalante C, Kantarjian H, Rolston K. Prospective, randomized study comparing quinupristin-dalfopristin with linezolid in the treatment of vancomycin-resistant Enterococcus faecium infections. J Antimicrob Chemother. 2004 Apr;53:646-9. [PubMed]

220. Rao N, White GJ. Successful treatment of Enterococcus faecalis prosthetic valve endocarditis with linezolid. Clin Infect Dis. 2002 Oct 1;35:902-4. [PubMed]

221. Rao N, Ziran BH, Hall RA, Santa ER. Successful treatment of chronic bone and joint infections with oral linezolid. Clin Orthop Relat Res. 2004;427:67-71. [PubMed]

222. Raymond N J, Henry J, Workowski KA. Enterococcal arthritis: case report and review. Clin Infect Dis 1995;21:516-22.[PubMed]

223. Reynolds PE, Snaith HA, Maguire AJ, Dutka-Malen S, Courvalin P. Analysis of peptidoglycan precursors in vancomycin-resistant Enterococcus gallinarum BM4174. Biochem J 1994;301:5-8.[PubMed]

224. Rice LB, Calderwood SB, Eliopoulos GM, Farber BF, Karchmer AW. Enterococcal endocarditis: a comparison of prosthetic and native valve disease. Rev Infect Dis 1991;13:1-7.[PubMed]

225. Ruoff KL, de la Maza L, Murtagh MJ, Spargo JD, Ferraro MJ. Species identities of enterococci isolated from clinical specimens. J Clin Microbiol 1990;28:435-7.[PubMed]

226. Rybak MJ, Cappelletty DM, Moldovan T, Aeschlimann JR, Kaatz GW. Comparative in vitro activities and postantibiotic effects of the oxazolidinone compounds eperezolid (PNU-100592) and linezolid (PNU-100766) versus vancomycin against Staphylococcus aureus, coagulase-negative staphylococci, Enterococcus faecalis, and Enterococcus faecium. Antimicrob Agents Chemother 1998;42:721-4.[PubMed]

227. Sacher HL, Miller WC, Landau SW, Sacher ML, Dixon WA, Dietrich KA. Relapsing native-valve enterococcal endocarditis: a unique cure with oral ciprofloxacin combination drug therapy. J Clin Pharmacol 1991;31:719-21.[PubMed]

228. Sadfar AJ, Bryan CS, Stinson S, Saunders DE. Prosthetic valve endocarditis due to vancomycin resistant Enterococcus faecium: treatment with chloramphenicol and minocycline. Clin Infect Dis 2002;34:61-3 .[PubMed]

229. Sahgal VS, Urban C, Mariano N, Weinbaum F, Turner J, Rahal JJ. Quinupristin/dalfopristin (RP 59500) therapy for vancomycin-resistant Enterococcus faecium aortic graft infection: case report. Microbial Drug Resist 1995;1:245-7.[PubMed]

230. Saleh-Mghir A, Lefort A, Petegnief Y, Dautrey S, Vallois JM, Le Guludec D, Carbon C, Fantin B. Activity and diffusion of LY333328 in experimental endocarditis due to vancomycin-resistant Enterococcus faecalis. Antimicrob Agents Chemother 1999;43:115-20.[PubMed]

231. Sandoe JA, Witherden IR, Au-Yeung HK, Kite P, Kerr KG, Wilcox MH.  Enterococcal intravascular catheter-related bloodstream infection: management and outcome of 61 consecutive cases.J Antimicrob Chemother. 2002 Oct;50:577-82.[PubMed]

232. Scheld WE, Keeley JM. Imipenem therapy of experimental Staphylococcus aureus and Streptococcus faecalis endocarditis. J Antimicrob Chemother 1983;12 (Suppl D):65-78.[PubMed]

233. Schmit JL. Efficacy of teicoplanin for enterococcal infections: 63 cases and review. Clin Infect Dis 1992;15:302-6.[PubMed]

234. Schouten MA, Voss A, Hoogkamp-Korstanje JAA, The European VRE Study Group. Antimicrobial susceptibility patterns of enterococci causing infections in Europe. Antimicrob Agents Chemother 1999;43:2542-6.[PubMed]

235. Schülin T, Thauvin-Eliopoulos C, Moellering RC Jr, Eliopoulos GM. Activities of the oxazolidinones linezolid and eperezolid in experimental intra-abdominal abscess due to Enterococcus faecalis or vancomycin-resistant Enterococcus faecium. Antimicrob Agents Chemother 1999;43:2873-6.[PubMed]

236. Schwalbe RS, McIntosh AC, Qaiyumi S, Johnson JA, Johnson RJ, Furness KM, Holloway WJ, Steele-Moore L. In vitro activity of LY333328, an investigational glycopeptide antibiotic against enterococci and staphylococci 1996;40:2416-9.[PubMed]

237. Shah P, Trostmann U, Tack K. Open-label, multicentre, emergency-use study of clinafloxacin (CI-960) in the treatment of patients with serious life-threatening infections. Int J Antimicrob Agents. 2002 ;19:245-8. [PubMed]

238. Shah PM. The need for new therapeutic agents: what is the pipeline? Clin Microbiol Infect. 2005 May;11 Suppl 3:36-42. [PubMed]

239. Shinabarger DL, Marotti KR, Murray RW, Lin AH, Melchior EP, Swaney SM, Dunyak DS, Demyan WF, Buysse JM. Mechanism of action of oxazolidinones: effects of linezolid and eperezolid on translation reactions. Antimicrob Agents Chemother 1997;41:2132-6.[PubMed]

240. Shlaes DM, Etter L, Gutmann L. Synergistic killing of vancomycin-resistant enterococci of classes A, B, and C by combinations of vancomycin, penicillin, and gentamicin. Antimicrob Agents Chemother 1991;35:776-9.[PubMed]

241. Signoretto C, Boaretti M, Canepari P. Cloning, sequencing and expression in Escherichia coli of the low-affinity penicillin-binding protein of Enterococcus faecalis. FEMS Microbiol Lett 1994;123:99-106.[PubMed]

242. Silverman JA, Oliver N, Andrew T, Li T. Resistance studies with daptomycin. Antimicrob Agents Chemother. 2001 Jun;45:1799-802. [PubMed]

243. Snydman DR, McDermott LA, Jacobus NV. Determination of synergistic effects of daptomycin with gentamicin or in vitro effect of beta-lactam antibiotics against Staphylococcus aureus and enterococci by FIC index and time kill kinetics (abstract no. E-533). 41st Interscience Conference on Antimicrobial Agents and Chemotherapy; 2001 Dec 16-19; Chicago, 175. [PubMed]

244. Speciale A, Aleo G, La Ferla K, Musumeci R, Caccamo F, Nicoletti G. Moxifloxacin - comparative inhibitory bactericidal activity against.  Int J Antimicrob Agents. 2002 Feb;19:111-8.[PubMed]

245. Stein GE. Safety of the newer parenteral antibiotics. Clin Infect Dis 2005;41:S293-302. [PubMed]

246. Steinmetz MP, Vogelbaum MA, De Georgia MA, Andrefsky JC, Isada C. Successful treatment of vancomycin-resistant enterococcus meningitis with linezolid: case report and review of the literature. Crit Care Med. 2001 Dec;29:2383-5. [PubMed]

247. Stevenson KB, Murray EW, Sarubbi FA. Enterococcal meningitis: report of four cases and review. Clin Infect Dis 1994;18:233-9.[PubMed]

248. Stiefel U, Paterson DL, Pultz NJ, Gordon SM, Aron DC, Donskey CJ. Effect of the increasing use of piperacillin/tazobactam on the incidence of vancomycin-resistant enterococci in four academic medical centers. Infect Control Hosp Epidemiol 2004;25:380-383.[PubMed]

249. Stiefel U, Pultz NJ, Helfand MS, Donskey CJ. Increased susceptibility to vancomycin-resistant Enterococcus intestinal colonization persists after completion of anti-anaerobic antibiotic treatment in mice. Infect Control Hosp Epidemiol 2004;25:373-379. [PubMed]

250. Streit JM, Fritsche TR, Sader HS, Jones RN. Worldwide assessment of dalbavancin activity and spectrum against over 6,000 clinical isolates. Diagn Microbiol Infect Dis. 2004 ;48:137-43. [PubMed]

251. Summers M, Misenhimer GR, Antony SJ. Vancomycin-resistant Enterococcus faecium osteomyelitis: successful treatment with quinupristin-dalfopristin. South Med J. 2001. 94:353-5.[PubMed]

252. Swaney SM, Aoki H, Ganoza MC, Shinabarger DL. The oxazolidinone linezolid inhibits initiation of protein synthesis in bacteria. Antimicrob Agents Chemother 1998. 42:3251-3255.[PubMed]

253. Tam VH, Mckinnon PS, Levine DP, Brandel SM, Rybak MJ. Once-daily aminoglycoside in the treatment of Enterococcus faecalis endocarditis: case report and review. Pharmacotherapy. 2000 20:1116-9.[PubMed]

254. Tan TY, Pitman I, Penrose-Stevens A, Simpson BA, Flanagan PG. Treatment of a vancomycin-resistant Enterococcus faecium ventricular drain infection with quinupristin/dalfopristin and review of the literature. J Infect 2000. 41:85-97.[PubMed]

255. Tarr PE, Sakoulas G, Ganesan A, Smith MA, Lucey DR. Hematogenous enterococcal vertebral osteomyelitis: report of 2 cases and review of the literature. J Infect. 2004;48:354-62.[PubMed]

256. Taylor SE, Paterson DL, Yu VL. Treatment options for chronic prostatitis due to vancomycin-resistant Enterococcus faecium. Eur J clin Microbiol Infect Dis 1998;17:798-800.[PubMed]  

257. Tedla FM, Salifu M, Friedman EA. Successful treatment of vancomycin-resistant enterococcal peritonitis with linezolid in a kidney transplant patient: a case report and review of the literature.Perit Dial Int. 2004 ;24:70-2.  [PubMed]

258. Thal L, Donabedian S, Robinson-Dunn B, Chow JW, Dembry L, Clewell DB, Alshab D, Zervos MJ. Molecular analysis of glycopeptide-resistant Enterococcus faecium isolates collected from Michigan hospitals over a 6-year period J Clin Microbiol. 1998 Nov;36:3303-8.  [PubMed]

259. Thauvin C, Eliopoulos GM, Willey S, Wennersten C, Moellering RC Jr. Continuous-infusion ampicillin therapy of enterococcal endocarditis in rats. Antimicrob Agents Chemother 1989;31:139-43. [PubMed]

260. Thompson RL, Lavin B, Talbot GH. Endocarditis due to vancomycin-resistant Enterococcus faecium in an immunocompromised patient: cure by administering combination therapy with quinupristin-dalfopristin and high-dose ampicillin. South Med J 2003;96:818-20.  [PubMed]

261. Till M, Wixson RL, Pertel PE. Linezolid treatment for osteomyelitis due to vancomycin-resistant Enterococcus faecium. Clin Infect Dis. 2002 May 15;34:1412-4.  [PubMed]

262. Tiong IY, Novaro GM, Jefferson B, Monson M, Smedira N, Penn MS. Bacterial endocarditis and functional mitral stenosis: a report of two cases and literature review. Chest 2002;122:2259-62.  [PubMed]

263. Tornieporth NG, Roberts RB, John J, Hafner A, Riley LW. Risk factors associated with vancomycin-resistant Enterococcus faecium infection or colonization in 145 matched case patients and control patients. Clin Infect Dis 1996; 23:767-772. [PubMed]

264. Torres C, Tenorio C, Lantero M, Gastañares M-J, Baquero F. High-level penicillin resistance and penicillin-gentamicin synergy in Enterococcus faecium. Antimicrob Agents Chemother 1993;37:2427-31.[PubMed]

265. Tripodi MF, Locatelli A, Adinolfi LE, Andreana A, Utili R. Successful treatment with ampicillin and fluoroquinolones of human endocarditis due to high-level gentamicin-resistant enterococci. Eur J Clin Microbiol Infect Dis 1998;17:734-6.[PubMed]

266. Tripodi MF, Utili R, Rambaldi A, Locatelli A, Rosario P, Florio A, Ruggiero G. Unorthodox antibiotic combinations including ciprofloxacin against high-level gentamicin resistant enterococci. J Antimicrob Chemother 1996;37:727-36. [PubMed]

267. Tsai SF, Zervos MJ, Clewell DB, Donabedian SM, Sahm DF, Chow JW. A new high-level gentamicin resistance gene, aph(2”)-Id, in Enterococcus spp. Antimicrob Agents Chemother 1998;42:1229-1232. [PubMed]

268. Tush GM, Huneycutt S, Phillips A, Ward JD. Intraventricular quinupristin/dalfopristin for the treatment of vancomycin-resistant Enterococcus faecium shunt infection. Clin Infect Dis 1998;26:1460-1. [PubMed]

269. Urban C, Mariano N, Mosinka-Snipas K, Wadee C, Chahrour T, Rahal JJ. Comparative in vitro activity of SCH-27899, a novel everninomicin, and vancomycin. J Antimicrob Chemother 1996;37:361-4. [PubMed]

270. Van Goethem GF, Louwagie BM, Simoens MJ, Vandeven JM, Verhaegen JL, Boogaerts MA. Enterococcus casseliflavus septicaemia in a patient with acute myeloid leukaemia. Eur J Clin Microbiol Infect Dis 1994;13:519-20. [PubMed]

271. Venditti M, Biavasco F, Varaldo PE, Macchiarelli A, De Biase L, Marino B, Serra P. Catheter related endocarditis due to glycopeptide-resistant Enterococcus faecalis in a transplanted heart. Clin Infect Dis 1993;17:524-5164a.  [PubMed]

272. Vergis EN, Hayden MK, Chow JW, Snydman Dr, Zervos MJ, Linden PK, Wagener MM, Schmitt B, Muder RR. Determinants of vancomycin resistance and mortality rates in enterococcal bacteremia: a prospective multicenter study. Ann Intern Med 2001;135:484-92. [PubMed]

273.Vergis EN, Shankar N, Chow JW, Hayden MK, Snydman DR, Zervos MJ, Linden PK, Wagener MM, Muder RR Association between the presence of enterococcal virulence factors gelatinase, hemolysin, and enterococcal surface protein and mortality among patients with bacteremia due to Enterococcus faecalis. Clin Infect Dis. 2002;35:570-5.  [PubMed]

274. Vijayvargiya R, Veis JH. Antibiotic-resistant endocarditis in a hemodialysis patient. J Am Soc Nephrol 1996;7:536-42. [PubMed]

275. Wareham DW, Abbas H, Karcher AM, Das SS. Treatment of prosthetic valve infective endocarditis due to multi-resistant Gram-positive bacteria with linezolid. J Infect. 2005 Aug 11.  [PubMed]

276. Watanakunakorn C. Rapid increase in the prevalence of high-level aminoglycoside resistance among enterococci isolated from blood cultures during 1989-1991. J Antimicrob Chemother 1992;30:289-93. [PubMed]

277. Whitman MS, Pitsakis PG, Zausner A, Livornese LL, Osborne AJ, Johnson CC, Levison ME. Antibiotic treatment of experimental endocarditis due to vancomycin- and ampicillin-resistant Enterococcus faecium. Antimicrob Agents Chemother 1993;37:2069-73. [PubMed]

278. Wilson WR, Karchmer AW, Dajani AS, Taubert KA, Bayer A, Kaye D, Bisno AL, Ferrieri P, Shulman ST, Durack DT. Antibiotic treatment of adults with infective endocarditis due to streptococci, enterococci staphylococci, and HACEK microorganisms. JAMA 1995;274:1706-13. [PubMed]

279. Wilson WR, Wilkowske CJ, Wright AJ, Sande MA, Geraci JE. Treatment of streptomycin-susceptible and streptomycin-resistant enterococcal endocarditis. Ann Intern Med 1984;100:816-23. [PubMed]

280. Winston DJ, Emmanouilides C, Kroeber A, Hindler J, Buruckner DA, Territo MC, Busuttil RW. Quinupristin/dalfopristin therapy for infections due to vancomycin-resistant Enterococcus faecium. Clin Infect Dis 2000;30:790-797. [PubMed]

281. Yao JDC, Thauvin-Eliopoulos C, Eliopoulos GM, Moellering RC Jr. Efficacy of teicoplanin in two dosage regimens for experimental endocarditis caused by a beta-lactamase-producing strain of Enterococcus faecalis with high-level resistance to gentamicin. Antimicrob Agents Chemother 1990;34:827-30. [PubMed]

282. Yelamanchili S, Cunliffe NA, Miles RS. Prosthetic valve endocarditis caused by a vancomycin-resistant Enterococcus faecalis. J Infect Dis 1998;36:348-9  [PubMed]

283. Zaas AK, Song X, Tucker P, Perl TM. Risk factors for development of vancomycin-resistant enterococcal bloodstream infection in patients with cancer who are colonized with vancomycin-resistant enterococci. Clin Infect Dis 2002;35:1139-1146.  [PubMed]

284. Zeana C, Kubin CJ, Della-Latta P, Hammer SM. Vancomycin-resistant Enterococcus faecium meningitis successfully managed with linezolid: case report and review of the literature. Clin Infect Dis 2001;33:477-82. [PubMed]

285. Zervos MJ, Bacon AE III, Patterson JE, Schaberg DR, Kauffman CA. Enterococcal superinfection in patients treated with ciprofloxacin. J Antimicrob Chemother 1988;21:113-5. [PubMed]

286. Zervos MJ, Kauffman CA, Therasse P, Bergman A, Mikesell TS, Schaberg DR. Nosocomial infection caused by gentamicin resistant Streptococcus faecalis: an epidemiologic study. Ann Intern Med. 1987;106:687-691.  [PubMed]

287. Zimmer SM, Caliendo AM, Thigpen MC, Somani J. Failure of linezolid treatment for enterococcal endocarditis. Clin Infect Dis. 2003 ;37:e29-30. [PubMed]

288. Zurenko GE, Todd WM, Hafkin B, Meyers B, Kauffman C, Bock J, Slightom J, Shinabarger D. Development of linezolid-resistant Enterococcus faecium in two compassionate use program patients treated with linezolid [abstract]. Abstracts of the 39th Interscience Conference on Antimicrobial Agents and Chemotherapy, San Francisco, CA, Sept 24-27, 1999. Washington, DC: American Society for Microbiology, 1999:118. [PubMed]

Table 1. Antimicrobial Therapy for Enterococcus faecalis Susceptible to Glycopeptides [Download PDF]

Infection Antibiotic(s) primary: alternative Comments

Urinary tract infection

 Intraadominal infection

Ampicillin: Vancomycin, Nitrofurantoin Fosfomycin

Ampicillin: Beta lactamase inhibitors, Vancomycin

Nitrofurantoin only for (cystitis) with isolates susceptible, not in sepsis or renal failure, usual duration 7-10d Not essential to treat for Enterococcus in all intraabdominal infection, unless organisms cultured, patient severely ill, duration 10-14d

Endocarditis

Ampicillin or Penicillin plus Gentamicin: Streptomycin, vancomycin (Penicillin allergy) To be used in combination  for the treatment of enterococcal endocarditis caused by organisms susceptible in vitro to either agent; streptomycin is used when gentamicin cannot be used because of high level resistance. Ampicillin plus gentamicin for 4-6 weeks is treatment of choice for endocarditis

Intravenous Catheter Bacteremia

Ampicillin: Vancomycin Duration 10-14d, some patients (single positive blood culture will respond to removal of the line alone

 See table 2 for drug dosages

Table 2. Antimicrobial Therapy for Vancomycin Resistant Enterococci (VRE)[Download PDF]

Antibiotic(s) primary Dose, Duration Comments
Ampicillin 12g/d IV For rare ampicillin-susceptible isolates of Enterococcus faecium;vancomycin resistant E. faecalis are usually susceptible
Gentamicin or streptomycin 1 mg/kg q 8 hrs to achieve serum peaks of 3-4 mg/ml and trough <1 mg/ml for endocarditis, treat for at least 4-6 weeks To be used in combination with ampicillin for the treatment of enterococcal endocarditis caused by organisms susceptible in vitro to either agent; streptomycin is used when gentamicin cannot be used because of resistance
Linezolid 600 mg PO or IV q 12 hr For linezolid-susceptible isolates of E faecium and E faecalis.  An agent of choice for serious enterococcal VREF infections
Daptomycin Use dose of 6 mg/kg/24 hrs for serious enterococcal infection; 6-8 weeks for endocarditis. Not approved for treatment of VRE infection. Not approved for treatment of endocarditis. Limited clinical information for VREF, but bactericidal activity makes therapy with this is agent a consideration for serious infections
Antibiotic(s) alternative Dose, Duration Comments
Doxycycline 100 mg PO or IV q 12 hr Not a first line therapy. For susceptible isolates, not bacteremia or endocarditis
Nitrofurantoin 100 mg PO Q 6 hr For urinary tract infections (cystitis) with isolates susceptible to nitrofurantoin, not indicated in renal failure
Fosfomycin 3 g X1 For urinary tract infections (cystitis) with isolates susceptible to fosfomycin
Chloramphenicol 50 mg/kg/d IV (in q 6hr divided doses) For chloramphenicol-susceptible isolates of E faecium and E. faecalis. Not a first-line therapy
Tigecycline 100 mg IV then 50 mg IV q 12 hrs Not indicated for VRE, approved in US for skin soft tissue infection, excellent in-vitro activity vs VRE
Quinupristin/dalfopristin 7.5 mg/kg Q8hr IV For-susceptible isolates of E faecium only

Table 3. Summary of Recommendations for Preventing the Spread of Vancomycin Resistance (adapted from CDC-HICPAC). [Download PDF]

1. Appropriate use of vancomycin
 

a. Treatment of infection due to B-lactam resistant gram-positive organisms. 

b. Treatment of infection due to gram-positive organisms in patients with serious beta-lactam allergy.

c. Treatment of antibiotic associated colitis in cases of metronidazole failure or potentially life threatening illness. 

d. Endocarditis prophylaxis, as recommended by the American Heart Association (Dajani). 

e. Prophylaxis for surgical procedures involving implantation of a prosthesis in institutions with a high rate of infection due to MRSA or methicillin-resistant S. epidermidis.

2. Education Program
 

a. Include physicians, nurses, pharmacy and laboratory personnel, students, and all other direct patient care providers. 

b. Program should include information on epidemiology of VRE and impact of VRE on cost and outcome of patient care.

3. Role of the Microbiology Laboratory
 

a. Laboratory should be able to identify and speciate enterococci.

b. Fully automated methods of testing enterococci for susceptibility testing are unreliable; disk diffusion, gradient disk diffusion, agar dilation, or manual broth dilution are acceptable. 

c. Vancomycin resistance should be confirmed by repeating one of the above tests, or by streaking onto brain heart infusion containing 6 ug/ml of vancomycin. Preliminary and confirmatory identification of VRE should be immediately reported to patient care personnel and infection control. 

d. Screening for VRE should be conducted periodically in hospitals where VRE has not been previously detected.

4. Prevention and control of nosocomial transmission of VRE
  a. For all hospitals, including those with no or infrequent isolation of VRE: 
 

1. Notify appropriate staff immediately when VRE are detected. 

2. Educate clinical staff about hospital policies regarding VRE colonized or infected patients so that appropriate procedures can be implemented immediately. 

3. Establish systems for monitoring process and outcome measures. 

4. Isolation precautions to prevent patient to patient transmission of VRE: REFER TO TABLE 2.

  b. In Hospitals with endemic VRE of continued VRE transmission despite implementation of above measures: 
 

a. Focus initial control efforts on critical care units and other areas where VRE transmission rates are highest. 

b. Where feasible cohort staff caring for VRE-positive and VRE-negative patients. 

c. Carriage of enterococci by hospital staff are rarely implicated in transmission. Investigation and culturing of hospital staff should be at the direction of infection control staff. 

d. Verify that environmental disinfection procedures are adequate, and that procedures are correctly performed. 

e. Consider sending representative VRE isolates to reference laboratories for strain typing as an aid in identifying reservoirs and patterns of transmission.

Table 4.  Isolation precautions to prevent patient to patient transmission of VRE (Adapted from CDC-HICPAC) [Download PDF]

1. Place VRE colonized or infected patients in single rooms, or cohort with other patients with VRE.

2. Wear gloves when entering the room of a VRE-infected or colonized patient.

3. Wear a gown when entering the room of a VRE-infected or colonized patient if:

 

a. Substantial contact with the patient or environmental surfaces in the room is anticipate. 

b. The patient is incontinent 

c. The patient has an ileostomy, colostomy or wound drainage not contained by dressing.

4. Remove gloves and gown before leaving the patient’s room and wash hands immediately with an antiseptic soap or waterless antiseptic agent. 

5. Dedicate the use of non-critical items, such as stethoscope, sphygmomanometer or rectal thermometer to a single patient or cohort of isolated patients. Devices must be disinfected before used on other patients.

6. Obtain stool or rectal swab cultures of roommates of patients newly found to be infected of colonized with VRE. Perform additional patient screening at the discretion of the infection control staff.

7. Adopt a policy for determining when patients infected or colonized with VRE can be removed from isolation precautions. As VRE colonization may be prolonged, negative cultures from multiple sites on 3 separate occasions at least one week apart is recommended.

8. The hospital should adopt a system by which infected and colonized patients can be recognized and placed into isolation promptly on transfer or re-admission.

9. Develop a plan, in consultation with public health authorities, for discharge or transfer of colonized or infection patients to other health facilities, including nursing homes and home health care.

Table 5. Recommendations for control of VRE in long-term care facilities (adapted from Brennen et al.) [Download PDF]

1. Patients colonized with VRE should not be denied admission to long-term care.

2. Patients colonized with VRE should be placed in private rooms or cohorting them with other colonized patients.

3. Staff should wear gowns and gloves during patient activities likely to result in VRE transmission. This includes dressing changes, bathing, changing bed linens, toileting, and care of indwelling catheters.

4. Hands must be washed after patient contact.

5. Patients who are colonized with VRE who are continent of stool and who do not have wounds that cannot be contained by dressings may attend activities such as physical therapy and recreation.

6. Antibiotic therapy should be given only when clearly indicated.

7. If a patient colonized with VRE is transferred to another health care facility, the receiving institution should be notified of the patient’s status.

What's New

Heintz BH, et al. Vancomycin-resistant enterococcal urinary tract infections. Pharmacotherapy 2010;30(11):1136-49.

Ergaz Z, et al. Elimination of Vancomycin-Resistant Enterococci from a Neonatal Intensive Care Unit Following an Outbreak. J Hosp Infect. 2010 Apr;74:370-6.

Kosowska-Shick K, Clark C, et al. Activity of Telavancin Against Staphylococci and Enterococci Determined by MIC and Resistance Selection Studies. Antimicrob Agents Chemother. 2009 Oct; 53:4217-24.

Theilacker C, Jonas D, et al. Outcomes of Invasive Infection due to Vancomycin-Resistant Enterococcus faecium during a Recent Outbreak. Infection. 2009 Aug 7. [Epub ahead of print]

Climo MW et al. The Effect of Daily Bathing with Chlorhexidine on the Acquisition of Methicillin-Resistant Staphylococcus aureus, Vancomycin-resistant Enterococci, and Healthcare-associated Bloodstream Infections: Results of a Quasi-experimental Multicenter Trial. Crit Care Med. 2009 Apr 20. [Epub ahead of print]

Hall RG 2nd, et al. Multicenter evaluation of vancomycin dosing: emphasis on obesity. Am J Med. 2008 Jun; 121(6):515-8.

Arias CA, et al.  Failure of daptomycin monotherapy for endocarditis caused by an Enterococcus faecium strain with vancomycin-resistant and vancomycin-susceptible subpopulations and evidence of in vivo loss of the vanA gene cluster. Clin Infect Dis 2007; 45:1343-1346.

Cunha BA, et al.  E. faecalis vancomycin-sensitive enterococcal bacteremia unresponsive to a vancomycin tolerant strain successfully treated with high-dose daptomycin.  Heart Lung 2007;36(6):456-61.

Gavalda J, et al.  Brief Communication: Treatment of Enterococcus faecalis Endocarditis with Ampicillin plus Ceftriaxone.  Ann Intern Med 2007;146:574-579.

Milstone AM, et al.  Cerebrospinal Fluid Penetration and Bacteriostatic Activity of Linezolid against Enterococcus faecalis in a Child with a Ventriculoperitoneal Shunt Infection.  Pediatr Neurosurg 2007;43:406-409.

back to top

GUIDED MEDLINE SEARCH FOR:

Epidemiology

Clinical Manifestations

Pathogenesis

Laboratory Diagnosis

Therapy

Prevention

History

[Moellering RC Jr: Synergistic antibiotic combinations for Enterococcal Endocarditis]

GUIDED MEDLINE SEARCH FOR HISTORICAL ASPECTS

Enterococcus