You are on page 1of 26

REVIEW

crossm

Methicillin-Resistant Staphylococcus aureus Prosthetic Valve


Endocarditis: Pathophysiology, Epidemiology, Clinical
Presentation, Diagnosis, and Management
Alicia Galar,a,b Ana A. Weil,c,d David M. Dudzinski,d,e Patricia Muñoz,a,b,f,g Mark J. Siednerc,d

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest


a Clinical Microbiology and Infectious Diseases Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
b Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain
c Division of Infectious Diseases, Medicine Department, Massachusetts General Hospital, Boston, Massachusetts, USA
d Harvard Medical School, Boston, Massachusetts, USA
e Cardiology Division, Medicine Department, Massachusetts General Hospital, Boston, Massachusetts, USA
f
Department of Medicine, School of Medicine, Universidad Complutense de Madrid, Madrid, Spain
g CIBER de Enfermedades Respiratorias-CIBERES (CB06/06/0058), Madrid, Spain

SUMMARY . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1
INTRODUCTION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
TIMING, PATHOPHYSIOLOGY, PATHOGENESIS, AND HISTOPATHOLOGY . . . . . . . . . . . . . . . . 2
Timing of Infection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
Pathophysiology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
Pathogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
Histopathology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
EPIDEMIOLOGY . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
CLINICAL PRESENTATION, ASSESSMENT, AND DIAGNOSIS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
TREATMENT . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
Vancomycin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
Rifampin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
Gentamicin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
Alternative Therapies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
The “Endocarditis Team” . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
Choice and Timing of Surgery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
PROGNOSIS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
PREVENTION . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
CHALLENGES AND FUTURE PERSPECTIVES . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 18
REFERENCES . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 19
AUTHOR BIOS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 26

SUMMARY Staphylococcus aureus prosthetic valve endocarditis (PVE) remains among


the most morbid bacterial infections, with mortality estimates ranging from 40% to 80%.
The proportion of PVE cases due to methicillin-resistant Staphylococcus aureus (MRSA)
has grown in recent decades, to account for more than 15% of cases of S. aureus PVE
and 6% of all cases of PVE. Because no large studies or clinical trials for PVE have been
published, most guidelines on the diagnosis and management of MRSA PVE rely upon Citation Galar A, Weil AA, Dudzinski DM,
expert opinion and data from animal models or related conditions (e.g., coagulase- Muñoz P, Siedner MJ. 2019. Methicillin-resistant
Staphylococcus aureus prosthetic valve
negative Staphylococcus infection). We performed a review of the literature on MRSA endocarditis: pathophysiology, epidemiology,
PVE to summarize data on pathogenic mechanisms and updates in epidemiology and clinical presentation, diagnosis, and
therapeutic management and to inform diagnostic strategies and priority areas where management. Clin Microbiol Rev 32:e00041-18.
https://doi.org/10.1128/CMR.00041-18.
additional clinical and laboratory data will be particularly useful to guide therapy. Major
Copyright © 2019 American Society for
updates discussed in this review include novel diagnostics, indications for surgical man- Microbiology. All Rights Reserved.
agement, the utility of aminoglycosides in medical therapy, and a review of newer anti- Address correspondence to Alicia Galar,
staphylococcal agents used for the management of MRSA PVE. alg331@mail.harvard.edu.
Published 13 February 2019
KEYWORDS methicillin-resistant Staphylococcus aureus, prosthetic valve endocarditis

April 2019 Volume 32 Issue 2 e00041-18 Clinical Microbiology Reviews cmr.asm.org 1


Galar et al. Clinical Microbiology Reviews

INTRODUCTION

S taphylococcus aureus prosthetic valve endocarditis (PVE) is a devastating infection.


The mortality rate due to methicillin-resistant Staphylococcus aureus (MRSA) has
climbed in recent decades, reaching more than 15% of cases of S. aureus PVE (12) and
6.6% of cases of PVE (4, 6).
Management for MRSA PVE is complex, and guidelines recommend both a multi-
disciplinary team and an individualized approach to care. Given the lack of clinical trials
testing treatments for MRSA PVE, many aspects of management lack an empirical basis.
For example, the timing and necessity of valve surgery remain unknown. In some
studies, hospital mortality rates for S. aureus PVE were significantly higher in patients
who had not undergone valve surgery (2, 3, 7, 8, 10, 13–15), prompting some investi-
gators to conclude that early valve surgery (EVS) should be considered a standard
treatment for S. aureus PVE, especially in patients with early-onset infection (2, 11).

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest


However, recent literature and the experience of the International Collaboration on
Endocarditis (ICE) have called into question the value of EVS (1, 11, 16–18). Specifically,
work by Hill et al. (16) suggested that uncomplicated S. aureus PVE cases might be
successfully managed without early valve surgery, and a multicenter study reported in
2015 by the ICE found no association between EVS and a reduction of 1-year mortality
rates in patients with S. aureus PVE (11).
Furthermore, although multiple treatment guidelines advocate for incorporating an
aminoglycoside into MRSA PVE therapy (19, 20) to promote sustained susceptibility to
rifampin, these recommendations are largely based on experimental models (21–23)
and the use of aminoglycosides for treatment of coagulase-negative Staphylococcus
(CoNS) PVE (24). Clinical data on the benefit of aminoglycoside combination therapy in
humans for MRSA PVE are lacking. Furthermore, the use of an aminoglycoside for other
causes of endocarditis, including methicillin-sensitive Staphylococcus aureus (MSSA)
native valve endocarditis (NVE) and CoNS PVE, demonstrates either harm or a lack of a
survival benefit (24–27). Although current guidelines discuss this lack of evidence, most
continue to recommend the addition of an aminoglycoside based on expert opinion
(28–31). Trimethoprim-sulfamethoxazole, clindamycin, ceftaroline, daptomycin, lin-
ezolid, telavancin, oritavancin, tigecycline, and combinations that might result in
synergy could have a role in treatment but have not yet been thoroughly studied.

TIMING, PATHOPHYSIOLOGY, PATHOGENESIS, AND HISTOPATHOLOGY


Timing of Infection
MRSA PVE is often dichotomized based on duration of disease (32) into early,
defined as the first year postsurgery, or late, defined as after 1 year postsurgery. These
thresholds have been developed based on the risk of developing PVE and differences
in the microbiology of the disease between periods (33, 34). The risk of PVE is greatest
during the first 3 months after surgery. The risk peaks approximately 15 days after
surgery, during which the PVE risk is estimated to be 45 cases/100,000 patient days (35).
After this time period, it decreases steadily to approximately 1 case/100,000 patient
days from 150 days to 20 years postoperatively (35–39). The cumulative proportions of
patients developing PVE range from 1 to 3% in the first 365 days after surgery according
to several studies with close follow-up and from 3 to 6% in 5 years (35–39). S. aureus is
a frequently encountered pathogen in both early and late PVE cases (6, 16, 40, 41),
accounting for approximately 12 to 36% of early cases (53 to 69% at the first 2 months
from surgery) and 18 to 30% of late cases (6, 41). A large, multicenter, international
study (6) showed that MRSA was the causative microorganism in 18.9% of early cases
of PVE, versus 3.3% of late cases.

Pathophysiology
Early PVE infection is believed to be caused by accidental seeding during surgery or
due to bloodstream dissemination in the first hours to months postoperatively. Early
after surgery, the prosthetic sewing ring and cardiac connection tissue and sutures
have not yet endothelized. Fibronectin and fibrinogen coat these areas and are

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 2


Staphylococcus aureus Prosthetic Valve Endocarditis Clinical Microbiology Reviews

believed to be a possible nidus for infection. In late PVE infection, these cardiac
structures become fully endothelized, and the pathogenesis of disease more resembles
that of NVE (42).
The location and distribution of PVE also differ by the type of prosthetic valve and
the route of infection (Fig. 1). In cases where the infection is introduced via contami-
nation around the surgical site, it typically affects the annulus and sewing ring union,
causing pseudoaneurysms, dehiscence, fistulas, and abscesses around valves (43–45). In
one postmortem study, perivalvular invasion, which is commonly coupled with pros-
thesis dehiscence and paravalvular regurgitation, appeared in approximately 40% of
autopsies performed for patients with PVE, and frank extension into tissue leading to
myocardial abscess was seen in 15% (46). Annular and/or myocardial abscesses were
present in 68% of the 47 patients with PVE in another study (47). Dehiscence of the
prosthesis was described in more than 80% of cases of aortic and mitral prostheses

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest


examined by Ben Ismail et al. (48), and vegetations were seen in 75% (48). More than
1 year postoperatively, infection related to biological PVE is more commonly found at
the prosthesis leaflets. Complications of late PVE include leaflet rupture and perforation
of an aortic valve prosthesis, which can extend through the intervalvar fibrosa and
annulus to cause pericarditis or, more frequently, can spread into the membranous
portion of the interventricular septum and cause arrhythmia (49–51). Large vegetations
may also keep the prosthesis open, causing malfunction or encroachment on the valve
orifice, resulting in functional stenosis or regurgitation via malcoaptation or perforation.

Pathogenesis
Several independent factors are involved in the development of MRSA PVE (Fig. 2).
MRSA has microbial surface components recognizing adhesive matrix molecules
(MSCRAMMs) that recognize and bind to adhesion molecules of the fibrin-platelet
matrices of “nonbacterial thrombotic endocarditis,” such as fibronectin, laminin, and
collagen, and these can also adhere to the normal endothelium or minimally injured
tissue (40, 52, 53). Once the thrombus is colonized by MRSA, this microorganism can
proliferate, creating the characteristic vegetation of infective endocarditis (IE). The
presence of cardiac prostheses introduces an additional variable that favors PVE, since
MRSA can adhere by forming biofilms (Fig. 2 and 3) (40, 53). At initial placement, the
ring-prosthesis interface is not endothelized and favors fibrin-platelet thrombus for-
mation. The suture points where the prostheses are placed constitute a mechanism
whereby MRSA can invade the heart tissue and form abscesses. In addition, the
continuous stress caused by the repetitive movement in the bioprostheses may disrupt
the surface of the leaflets and predispose to infection of the fibrin-platelet thrombus.
MRSA can reach the prosthesis by contamination of the prosthetic valve during surgery
(Fig. 4) or by a hematogenous route via a catheter-related infection, intravenous drug
use, a surgical wound, or pulmonary or urinary tract infection (40, 53, 54). During the
surgical procedure (Fig. 4), the surgical site may be exposed to MRSA from the patient’s
or health care practitioner’s skin. The ability of S. aureus strains to produce biofilms (Fig.
3) in vitro has been linked to clinically persistent MRSA bacteremia (ⱖ7 days) and the
evolution of prosthetic valve vegetation propagation (52, 55, 56).
Nonvalvular invasive infection may also cause bioprosthetic valve endocarditis. For
example, annular and myocardial invasion was observed in 38 of 85 patients (45%) in
one study and was more common among cases of bioprosthetic PVE occurring during
the first year after valve placement than in cases presenting later (59 versus 25%) (57).
Invasive disease was more frequent in patients with early than in those with late
bioprosthetic PVE (79% versus 31%) in another series (58).
A large proportion of cases of PVE is nosocomial and correlates with a high
proportion of MRSA infections (6). An international study including 556 patients with
PVE demonstrated that 36.5% of infections were nosocomially acquired or related to
frequent health care visits (6). Similarly, the use of transcatheter aortic valve implanta-
tion (TAVI) also increased the risk for MRSA PVE (59, 60), as did the use of orotracheal

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 3


Galar et al. Clinical Microbiology Reviews

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest

FIG 1 Prosthetic valves explanted from patients with MRSA prosthetic valve endocarditis, and received at the microbiology laboratory to
perform valve culture and 16S PCR (courtesy of Mercedes Marín, Hospital General Universitario Gregorio Marañón, Madrid, Spain). Shown
are a mechanical valve, a mitral ring, an aortic bioprosthesis, and a mitral bioprosthesis.

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 4


Staphylococcus aureus Prosthetic Valve Endocarditis Clinical Microbiology Reviews

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest


FIG 2 Pathogenesis of MRSA (methicillin-resistant Staphylococcus aureus) PVE. MSCRAMMs, microbial surface components recognizing
adhesive matrix molecules.

intubation and percutaneous self-expandable valves (59). Further studies are needed to
better establish a relationship between MRSA virulence factors observed experimen-
tally in vitro and in animal models (61) and clinical disease in humans.

Histopathology
There is no typical pattern of PVE histological characterization in bioprosthetic
valves. As bioprosthetic valves degenerate, they often create noninfective, calcific,
vegetative-like lesions with inflammatory infiltrates, which can result in a noninfectious
process that can mimic and be misdiagnosed as PVE. A retrospective pathological study
of inflamed bioprosthetic valve tissues from 88 cases of resected bioprosthetic valves
(21 for probable endocarditis and 67 for noninfective dysfunction) was performed to
better define the histological criteria for PVE (62). PVE was histologically characterized
by neutrophil-rich inflammatory infiltrates and the presence of microorganisms. Inflam-
matory infiltrates in valve tissue samples from the noninfective control group consisted
mainly of lymphocytes and macrophages. In that study, having a neutrophil percentage
exceeding 1.5% of the valve surface area was associated with a high specificity (94%)
for infectious PVE (62).

EPIDEMIOLOGY
PVE occurs in 1% to 6% of patients after prosthetic valve placement (63), accom-
panied by an incidence of 0.3% to 1.2% per patient year (6, 64–66) and accounting for
16% to 31% of IE cases in several studies (1, 6, 67–70). The etiology of 146 early-PVE
clinical cases and 140 late-PVE cases was summarized from 17 published reports (32).
In that study, early S. aureus PVE accounted for 19.2% of the cases, and late compli-
cations were less likely with increasing time after surgery, occurring in only 11.4% of the
cases. The incidence of both early and late PVE was correlated with increasing under-
lying comorbidity at the time of valve placement, surgeon experience, extracorporeal
circulation duration, sterility of the heart-lung machine and the operating theater,
extracardiac postoperative infection, and the length of time that the patient was

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 5


Galar et al. Clinical Microbiology Reviews

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest

FIG 3 An original electron scanning microscope image of a methicillin-resistant Staphylococcus aureus


biofilm on a patient’s mechanical heart prosthesis. The image was prepared at both the Clinical
Microbiology and Infectious Diseases Department and the Pathology Department of the Hospital General
Universitario Gregorio Marañón and was taken at the National Center of Electron Microscopy (JSM 6400,
CNME, Madrid, Spain).

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 6


Staphylococcus aureus Prosthetic Valve Endocarditis Clinical Microbiology Reviews

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest


FIG 4 Surgery image (courtesy of Gregorio Cuerpo, Cardiac Surgery, Hospital General Universitario
Gregorio Marañón, Madrid). Shown is mitral prosthetic valve endocarditis caused by S. aureus.

monitored after surgery (32). MRSA accounted for approximately 6.5% of PVE cases
(n ⫽ 556) assessed by the ICE Prospective Cohort Study (6). Figure 5 shows a compar-
ison of cases of S. aureus (P ⫽ 0.003) and MRSA (P ⫽ 0.001) involved in PVE across
geographic regions. The data provided by Wang et al. (6) are consistent with the global
epidemiology of S. aureus (P ⫽ 0.007) and MRSA (P ⫽ 0.001) PVE found and also kindly
shared by Murdoch et al. (4).
In one observational study, the prophylactic use of penicillinase-resistant penicillins
(methicillin and oxacillin) during the perioperative periods reduced early postoperative
S. aureus PVE (71). Unsurprisingly, the incidence of PVE was higher when the valve
replacement occurred in the setting of active or recently treated endocarditis (36, 39,
72, 73). In several observational studies, bioprosthetic valves appear to increase the risk
for infection over mechanical valves after 18 months (37, 72, 73). However, three
randomized trials including 1,418 patients monitored for 8 to 20 years could not
demonstrate a statistically significant difference in PVE occurrence between biological
and mechanical valves (P ⫽ 0.45 [74], P ⫽ 0.71 [75], and P ⫽ 0.70 [76]). Another
observational study that included 38,000 patients ⱖ65 years of age demonstrated a
higher risk of endocarditis after a median of 12 years of follow-up among those with
bioprosthetic valves (2.2% versus 1.4%; unadjusted hazard ratio, 1.69 [95% confidence
interval {CI}, 1.43 to 2.00]) (77). Finally, Calderwood et al. (37) showed a higher risk of
occurrence of PVE among patients (n ⫽ 116 out of 2,608 evaluated) who received
mechanical valves than among those who received bioprosthetic valves at 3 months
postsurgery (P ⫽ 0.02), but in contrast, Grover et al. (73) could not demonstrate any
difference between the valves in 66 patients who developed PVE out of 1,032 observed
during a mean length of follow-up of 7.7 years.

CLINICAL PRESENTATION, ASSESSMENT, AND DIAGNOSIS


PVE signs and symptoms are similar to those of native valve disease. Yet the clinical
presentation of MRSA PVE is often nonspecific, especially soon after surgery, when
inflammation and fever might occur for other reasons. Due to the intracardiac compli-
cations described above, clinical manifestations of PVE frequently include hemolysis,
heart failure, valvular dysfunction, and/or new arrhythmia (13, 78, 79). Calderwood et al.
(78) studied the outcomes of 116 patients with PVE and found that 64% of individuals
with PVE suffered some combination of worsened or new cardiac failure, a changed or
new cardiac murmur, continuous fever, or irregularities upon electrocardiography
(ECG). These complications were more common in the first year after valve replacement
and in aortic valve prosthesis infections (Table 1). New or changing murmurs, heart
failure, and new electrocardiographic conduction disturbances are noted more often in

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 7


Galar et al. Clinical Microbiology Reviews

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest

FIG 5 Global epidemiology of MRSA involved in prosthetic valve endocarditis (PVE). The causative agents of PVE differ geographically
(4, 6). Data from Wang et al. (6) were collected between June 2000 and August 2005 from 556 patients with infective prosthetic valve
endocarditis in 53 sites worldwide (P ⫽ 0.003 for Staphylococcus aureus; P ⫽ 0.001 for MRSA [methicillin-resistant Staphylococcus
aureus]). MSSA, methicillin-sensitive Staphylococcus aureus.

PVE than in NVE cases due to the likelihood of invasive infection. ECG (Fig. 6), chest
radiograph, and blood cultures are considered part of the standard work-up if there is
a clinical suspicion of PVE.
Pulmonary, neurological, kidney, and musculoskeletal complications or complica-

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 8


TABLE 1 Key articles on epidemiology, diagnosis, management, conclusions, and prevention of S. aureus PVE and IEa
Author(s) (yr) (reference) Epidemiology Diagnosis Management Conclusion(s) and/or prevention strategy
Béraud et al. (2011) (215) 137 physicians participated in the study Infective endocarditis Endocarditis was treated with gentamicin doses of 3 mg/ Guidelines were not followed by most of the
kg/day by 61% of physicians, 4 mg/kg/day by 22.1%, physicians for gentamicin dosing in these
and 5 mg/kg/day by 16.9% patients; instead, they used validated
regimens from published studies
Bille (1995) (216) Antimicrobial therapy review Endocarditis due to Staphylococcus A combination of 3 antibiotics (vancomycin or oxacillin ⫹ Further studies are needed to incorporate novel
gentamicin and rifampin) is suggested for PVE during treatment options, especially in patients
at least 6 wk affected by MRSA
Calderwood et al. (1985) (37) 2,642 patients who underwent valve replacement 116 patients with PVE (4.4%) At 12 mo, the risk of PVE was 3.1%, and at 60 mo, the risk There were significant differences in the risk of
for the first time were included in the study was 5.7%; porcine valves had a significantly lower risk PVE depending on the type of valve, but no
of PVE during the first 90 days from surgery than significant differences between porcine and
mechanical valves but a significantly higher risk after mechanical valves were observed in the risk
12 mo postsurgery of having PVE after 5 yr
Cervera et al. (2014) (217) Analysis of a study cohort 93 cases of S. aureus infective 57% had a vancomycin MIC of ⬍1.5 ␮g/ml, and 43% had Percentages of in-hospital death varied
endocarditis (left sided) an MIC of ⱖ1.5 ␮g/ml significantly between both groups, at 30 and

April 2019 Volume 32 Issue 2 e00041-18


53%, respectively
Chirouze et al. (2004) (1) Evaluation of mortality risk 61 cases of S. aureus PVE Patients who had their valve replaced early, despite S. aureus PVE is a disease with high morbidity
having heart complications, showed lower mortality and mortality rates (28.6–85.7%)
Staphylococcus aureus Prosthetic Valve Endocarditis

rates (P ⫽ 0.09)
Chirouze et al. (2015) (11) Impact of early valve surgery on clinical outcome of 747 cases of definite left-sided PVE Non-S. aureus PVE caused significantly lower rates of Different factors should be taken into account
S. aureus PVE within the International death after 1 yr than S. aureus PVE; at this time, before deciding on EVS
Collaboration of Endocarditis patients with S. aureus PVE and EVS also had lower
mortality rates (P ⬍ 0.01); EVS did not diminish
mortality at 1 yr
Cosgrove et al. (2009) (218) 236 patients from 44 hospitals and 4 countries were S. aureus bacteremia and native Vancomycin or an antistaphylococcal penicillin ⫹ low- Low-dose gentamicin should not be used
prospectively evaluated valve infective endocarditis dose gentamicin or daptomycin alone was routinely for S. aureus bacteremia and native
administered to patients; renal adverse events were valve infective endocarditis due to the
evaluated nephrotoxicity shown
de Feiter et al. (2005) (219) Fusidic acid, rifampicin, vancomycin, oxacillin, and Patient with Staphylococcus Despite the nonapproval for this indication, linezolid was Patient had a favorable outcome with linezolid
gentamicin treatment failures epidermidis PVE administered to this patient
Del Río et al. (2014) (220) Rescue therapy with imipenem ⫹ fosfomycin Complicated bacteremia and Treatment was successful in 69% of cases; the mortality Combination therapy was safe and effective as
MRSA endocarditis rate due to MRSA was 1/5 (20%) rescue therapy
Fernández Guerrero et al. Incidence of infective endocarditis, epidemiology, Definite S. aureus endocarditis NVE was a less common hospital-acquired infection than Valve replacement significantly improved
(2009) (15) clinical features, prognosis (right sided and left sided) PVE; for both types of endocarditis, renal and cardiac outcomes for patients with PVE
failure and central nervous system complications were
detected
Fowler et al. (2006) (157) Daptomycin vs standard therapy S. aureus bacteremia and Microbiological failure was more common in the group A noninferiority rate was observed in the group
endocarditis treated with daptomycin than in the one with treated with daptomycin compared to the
standard therapy one treated with standard therapy for
bacteremia and right-sided endocarditis
caused by S. aureus
Hasbun et al. (2003) (221) Prognostic factors Left-sided endocarditis (native Factors related to mortality after 6 mo, including 4 groups of patients were identified depending
valve) with complications abnormal mental status, bacterial cause, comorbidities, on the mortality risk 6 mo after baseline
medical treatment, moderate/severe congestive cardiac
failure
Holland et al. (2014) (222) Review on hospital management Bacteremia caused by S. aureus Diagnostic methods and antibiotic treatment strategies There are groups of patients who do not need
TEE
John et al. (1998) (3) Clinical strategies and prognostic factors Definite PVE caused by S. aureus Complications affecting the central nervous system (33%) More patients died due to heart problems than
and heart (67%) were found; the 3-mo mortality rate due to problems affecting the central
was 42% nervous system, but this mortality was
diminished when there was surgery for valve
replacement during antibiotic treatment
Kang et al. (2012) (223) 6-wk occurrence of embolic events and mortality Patients with large vegetations, Patients were randomized into 2 groups, conventional Rates of embolic events and mortality
severe disease in valves, left- treatment or early surgery significantly decreased in the group with
sided endocarditis due to early surgery compared to the group of
infection patients treated conventionally
Karchmer et al. (1983) (24) Retrospective study of 75 PVE cases Staphylococcus epidermidis PVE The gentamicin susceptibility rate was 78%, and those for Antibiotic therapy including vancomycin ⫹
rifampin and vancomycin were 100% for all isolates rifampin or an aminoglycoside increased
tested; dysfunction of valves and tissue progression favorable outcome rates; surgical treatment
were the most common problems, needing surgery in was also important
30 cases
(Continued on next page)

cmr.asm.org 9
Clinical Microbiology Reviews

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest


TABLE 1 (Continued)
Author(s) (yr) (reference) Epidemiology Diagnosis Management Conclusion(s) and/or prevention strategy
Karchmer (1991) (224) Infection control PVE During a year postsurgery, the nosocomial risk of PVE Further studies are needed to detect
Galar et al.

was 1.4–3.0%; the most common reason for hospital- postsurgical causes of hospital-acquired PVE
acquired PVE was methicillin-resistant coagulase- and diminish them
negative Staphylococcus
Le and Bayer (2003) (225) Review on antibiotic treatment for endocarditis Few strategies for in vitro, experimental, and clinical Human clinical data are scarce on combination
caused by frequently detected microorganisms evaluation of enterococcal endocarditis have been antibiotic treatment for infective endocarditis
shown due to S. aureus
Mayer and Schoenbaum Review and approach PVE Higher rates of morbidity and mortality were detected in Factors related to poor outcome were early
(1982) (226) early than in late PVE cases; the etiology often PVE, paravalvular leakage, emboli, persistent
included fungi, staphylococci, and Gram-negative rods fever, nonstreptococcal microorganisms,
in early PVE and streptococci in late PVE nonheterograft aortic valve, congestive
cardiac failure
Muñoz et al. (2015) (12) Epidemiology, clinical features, prognostic factors Infective endocarditis (1,804 cases) Previous cardiac surgery, atrial fibrillation, cardiac The rates of in-hospital and 1-yr deaths were
complications and failure, septic shock, age, elevated, and surgery was the only

April 2019 Volume 32 Issue 2 e00041-18


cerebrovascular complications, or Candida or protective factor
Staphylococcus cause was related to in-hospital deaths
(28.9%); after 1 yr, association was found for cancer,
cardiac failure, age, and renal insufficiency (11.2%)
Murdoch et al. (2009) (4) Global infective causes and clinical features Infective endocarditis Infections of mitral and aortic valves due to S. aureus Risk factors for in-hospital mortality were lung
were the most frequent presentation, also complicated edema, age, prosthetic infection, S. aureus or
with heart failure, stroke, and other emboli and coagulase-negative staphylococcal cause,
abscess in the heart mitral vegetation, and valve problems
Rajashekaraiah et al. (1980) Evaluation of tolerance (MBC/MIC ⬎ 16) S. aureus bacteremia and Tolerant microorganisms were accompanied by more Poor outcomes of endocarditis were more
(26) endocarditis deaths, complications, hospitalization in ICU, common in cases caused by tolerant
prolongation of fever microorganisms than in cases caused by
sensitive ones
Ribera et al. (1996) (27) Cloxacillin vs cloxacillin ⫹ gentamicin during 2 wk S. aureus endocarditis (right sided) Mortality occurred 1 and 2 cases, respectively Combination treatment was not more effective
than the single one
Sohail et al. (2006) (17) Mortality rates in patients who received medical vs S. aureus PVE Mortality rates of 48% and 28%, respectively The no. of deaths was lower in the surgical
surgical treatment group; bioprosthetic valves and ASA class IV
were prognostic factors
Wang et al. (2007) (6) Global infective causes and clinical features PVE The most frequent microorganism was S. aureus; 36.5% of S. aureus is globally the main cause of PVE, and
cases were related to health care; the of in-hospital the presence of complications is an
death rate was 22.8% and was related to health care, important prognostic factor
age, persistent bloodstream infection, S. aureus cause,
and cardiac and CNS problems
Wareham et al. (2005) (227) Cases treated with linezolid MRSE and VRE endocarditis In vitro study of Staphylococcus epidermidis and Outcome was favorable in both cases
Enterococcus faecalis infections treated with linezolid ⫹
gentamicin or vancomycin
Watanakunakorn (1979) (32) Treatment with penicillin vs penicillin ⫹ gentamicin S. aureus endocarditis The death rate was 40% in both groups of patients There is no clinically demonstrated advantage
of the use of combination therapy with
gentamicin
Wilson et al. (1995) (228) Treatment efficacy experience Endocarditis caused by Recommendations of treatment based on previously Literature is scarce
enterococci, staphylococci, reported studies
streptococci, and members of
the HACEK group
Yaw et al. (2014) (229) Clinical outcome evaluation MRSA and MSSA bacteremia Rehospitalization rates associated with infection were MSSA bacteremia outcomes were more
similar in both groups of patients favorable than MRSA bacteremia ones;
patients colonized with MRSA should be
treated carefully
aASA, American Society of Anesthesiologists; EVS, early valve surgery; MBC, minimal bactericidal concentration; MRSA, methicillin-resistant Staphylococcus aureus; MRSE, methicillin-resistant Staphylococcus epidermidis; NVE,
native valve endocarditis; PVE, prosthetic valve endocarditis; TEE, transesophageal echocardiography; VRE, vancomycin-resistant Enterococcus; ICU, intensive care unit; HACEK group, a group of Gram-negative bacilli
consisting of Haemophilus spp., Actinobacillus actinomycetemcomitans, Cardiobacterium hominis, Eikenella corrodens, and Kingella spp.

cmr.asm.org 10
Clinical Microbiology Reviews

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest


Staphylococcus aureus Prosthetic Valve Endocarditis Clinical Microbiology Reviews

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest


FIG 6 Electrocardiogram at 25 mm/s of a patient with aortic MRSA PVE with nonspecific findings, including tachycardia (heart rate of approximately 125 beats
per minute), a first-degree block with a PR interval exceeding a duration of 0.2 s (indicated by the arrow), and ST segment changes (indicated by the circle).

tions associated with systemic infections might also occur, at the same time, in some
PVE cases (80–87). Of note, S. aureus is the pathogen most commonly related to IE
complications compared to others (88). Lung complications are more frequently seen
in patients with right-sided endocarditis, in several cases presenting manifestations
such as pneumonia, abscesses, pleural effusion, and/or atelectasis.
Central nervous system (CNS) complications, primarily embolic infarcts or hemor-
rhage, are also more common in PVE than in NVE cases, ranging from 20 to 40%, with
overt arterial emboli being noted for 40% of patients with PVE (48, 89–91). Similar rates
of embolic stroke (18%) and ischemic stroke (20%) among patients with PVE were
reported by Davenport and Hart (92) and Keyser et al. (89), respectively. Fernández
Guerrero et al. (15) observed that, compared to NVE, S. aureus PVE clinical manifesta-
tions were characterized by less-frequent cardiac murmurs and a shorter symptomatic
prodrome.
In PVE patients with no prior antibiotic therapy, blood cultures are positive in at least
90% of cases (63). Molecular techniques, such as DNA examination by pulsed-field gel
electrophoresis or 16S rRNA sequencing, can be used when standard blood or tissue
cultures have not revealed a pathogen (93, 94).
Transesophageal echocardiography (TEE) has poorer diagnostic validity for PVE
(including MRSA PVE) than for NVE (95). However, TEE with a high-resolution biplane or
multiplane transducer that allows continuous-wave and pulsed-wave Doppler and
color flow imaging can increase the accuracy of diagnosis of PVE (96). TEE is considered
the method of choice for diagnosis of PVE (97), because although transthoracic
echocardiography (TTE) may lead to a diagnosis in some cases, TEE has a higher
sensitivity for PVE (98, 99) (Fig. 7A). This increase in sensitivity is not accompanied by
a loss of specificity and is independent of the valve type or position (98–101). For
suspected PVE, TEE sensitivity and specificity have been estimated at 77 to 90% and
90%, respectively, compared to TTE, with a specificity and sensitivity estimated at 40 to
70% and 90%, respectively (102). Nonetheless, TTE has value for the assessment of
ventricular size and function and severe hemodynamic lesions in valves and can often

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 11


Galar et al. Clinical Microbiology Reviews

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest

FIG 7 Imaging modalities for MRSA prosthetic valve endocarditis diagnosis. (A) Transesophageal echocardiography demonstrating a 1.4- by
1.2-cm highly mobile echodensity at the ventricular side of the bioprosthetic aortic valve (indicated by the arrow), without significant valvular
dysfunction. (B) Transthoracic echocardiogram image showing a parasternal view with the prosthetic aortic valve, right ventricular outflow tract,
and aorta on the top; the left atrium and mitral valve at the bottom; and the left ventricle on the left. The prosthetic aortic valve is not well
visualized, but there is a vegetation on the ventricular side (indicated by the circle) and anterior aortic root thickening (indicated by the arrow),
(Continued on next page)

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 12


Staphylococcus aureus Prosthetic Valve Endocarditis Clinical Microbiology Reviews

detect anterior aortic prosthetic valve abscesses (103) (Fig. 7B). Typically, the ventricular
surfaces of prostheses in the mitral, tricuspid, and aortic positions are better viewed by
TTE, whereas TEE has additional value for viewing the aortic valve surfaces, tricuspid
and mitral valves, and the exit of the prosthesis in the aorta; fistula and abscess
detection; detection of paraprosthetic leaks; and visualization of a mitral valve pros-
thesis (104, 105). However, TEE will miss some cases of prosthetic valve dehiscence. In
a study of 26 patients with PVE, there were 14 (56%) cases of aortic valve dehiscence
intraoperatively, 4 (29%) of which were not detected upon TEE (106). One prospective
study that compared the paired use of TTE with TEE in 114 episodes of clinically
suspected IE (34 PVE and 80 NVE) found that the results of the two tests were
concordant in only 55% of cases (107). TEE prompted reclassification for 34% of patients
with prosthetic valves, compared with 11% of patients with native valves.
The Duke criteria have been described as being less useful for the diagnosis of PVE

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest


because of low sensitivity (108, 109) compared to the sensitivity of 70 to 80% (110, 111)
in the diagnosis of NVE. Because of diagnostic challenges in PVE with standard testing
and clinical scores, other techniques have been assessed. Magnetic resonance imaging
(MRI) and multislice computed tomography (MS-CT) might improve the detection of an
intra- or pericardiac anatomical complication (112). The value of ECG-gated multide-
tector CT angiography (MDCTA) was addressed by three studies (113–115), which
demonstrated a ⬎90% sensitivity for the diagnosis of PVE (113), which was improved
to a sensitivity and a specificity of 100% and 83%, respectively, after completion of
routine testing for endocarditis and resulted in modification of therapy in 25% of cases
(115) (Fig. 7C).
Recently, nuclear imaging has shown promise for improving diagnostics. [18F]fluo-
rodeoxyglucose positron emission tomography electrocardiogram-gated computer to-
mography ([18F]FDG PET/CT) scans have shown utility as an additional diagnostic
criterion for PVE in cases where a diagnosis cannot be made with standard echocar-
diography (116–119) (Fig. 7D). FDG PET/CT detects inflammation early in the infection
process (120). For suspected PVE, [18F]FDG PET/CT demonstrated a 67 to 100% positive
predictive value, a 50 to 100% negative predictive value, 73 to 100% sensitivity, and 71
to 100% specificity (116–119, 121–123). As such, an algorithm reported by Saby et al.
(116) shows that PET/CT is useful to assess probable PVE cases. This algorithm incor-
porates the PET/CT 2013 modified Duke criteria for the diagnosis of possible PVE cases
that do not meet criteria for endocarditis by the modified Duke criteria but remain
under high clinical suspicion. The sensitivity of the modified Duke criteria significantly
increased with the addition of PET imaging to the scoring system, from 70% (95% CI,
52% to 83%) to 97% (95% CI, 83% to 99%) (P ⫽ 0.008). This result was the consequence
of a significant reduction (P ⬍ 0.0001) in the number of possible PVE cases from 56%
to 32% (116).
Three retrospective studies have addressed the value of technetium-99m-
hexamethylpropylene amine oxime (99mTc-HMPAO)-labeled leukocyte scintigraphy
with single-photon emission tomography/computed tomography (SPECT/CT) for the
detection of PVE (119, 124, 125). Probable (125) or definite (119, 124) IE cases were
included. Globally leukocyte scintigraphy showed an 85 to 100% positive predictive
value, a 47 to 81% negative predictive value, 64 to 90% sensitivity, and 36 to 100%
specificity (126). Further evaluations with increased sample sizes will be helpful for
better defining a role and optimal scenario of nuclear imaging in the diagnosis of PVE.

TREATMENT
The S. aureus PVE mortality rate remains high (25 to 42%) (3, 54) despite advances
in antibiotic treatment (1). Both the American Heart Association (AHA) and its European

FIG 7 Legend (Continued)


suggestive of an aortic root abscess. There is also mild prosthetic aortic valve regurgitation, but it cannot be appreciated in the still image. (C)
Electrocardiogram-gated multidetector CT angiography demonstrating a 4- by 8-mm vegetation on the bioprosthetic aortic valve. (D) PET/CT
image at the posterior prosthetic aortic valve, after 16.29 mCi [18F]fluorodeoxyglucose uptake. The arrow notes an area of hyperintensity,
suggesting a focus of inflammation or infection consistent with prosthetic valve endocarditis.

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 13


Galar et al. Clinical Microbiology Reviews

TABLE 2 International guidelines for therapy of MRSA PVEa


AHA guidelines
ESC guidelines for adults Pediatric Adult
Vancomycin at 30–60 mg/kg Q24h i.v., BID or TID Vancomycin at 40 mg/kg Q24h i.v. (maximum dose, 2 g Q24h), Vancomycin at 30 mg/kg Q24h i.v., BID
during ⱖ6 wk BID or TID during ⱖ6 wk during ⱖ6 wk
Rifampin at 900–1,200 mg Q24h i.v./orally, BID or Rifampin at 20 mg/kg Q24h i.v. (maximum dose, 900 mg Rifampin at 900 mg Q24h i.v./orally, TID
TID during ⱖ6 wk Q24h), TID during ⱖ6 wk during ⱖ6 wk
Gentamicin at 3 mg/kg Q24h i.v./i.m., once a day Gentamicin at 3 to 6 mg/kg Q24h i.v./i.m., TID during the first Gentamicin at 3 mg/kg Q24h i.v./i.m., BID
or BID during the first 2 wk 2 wk or TID during the first 2 wk
aThedoses of these drugs must be adjusted in the setting of renal insufficiency. The intravenous (i.v.) route is preferred, particularly in infants and children. Q24h,
every 24 h; BID, twice a day; TID, three times a day; i.m., intramuscular.

counterpart, the European Society for Cardiology (ESC), suggest a triple-drug regimen
(class I; level of evidence C) with vancomycin and rifampin for ⱖ6 weeks and genta-

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest


micin during the first 2 weeks for the management of MRSA PVE (Table 2) (29, 30).
These recommendations are based on CoNS PVE therapeutic regimen activity, IE
experiments, and retrospective clinical series (57, 127–130). A single retrospective study
of valve cultures from 61 patients with staphylococcal PVE (29 due to S. aureus and 32
due to CoNS) treated surgically showed that those receiving combination therapy were
5.9 times more likely to have culture-negative valves than patients receiving mono-
therapy (adjusted by the length of therapy prior to surgery) (128). In this study, the six
patients treated with a triple-drug regimen including rifampin prior to surgery had
negative valve cultures. Regarding surgical indication, all of the groups of therapy
evaluated for S. aureus NVE and any staphylococcal NVE or PVE were similar (P ⫽ 0.53,
P ⫽ 0.51, and P ⫽ 0.66, respectively). No data on clinical outcomes restricted to those
with S. aureus PVE were reported.
Antibiotic recommendations for treatment of MRSA PVE are also partially based on
ex vivo and animal studies of Staphylococcus epidermidis PVE. A retrospective evaluation
of 23 cases of methicillin-resistant S. epidermidis (MRSE) PVE (129) showed an increase
in serum bactericidal activity when rifampin was added to vancomycin regimens (129).
Studies regarding MRSE endocarditis in rabbit models also demonstrated that genta-
micin, rifampin, and vancomycin in combination increased the efficacy of eradication of
S. epidermidis from vegetations compared to beta-lactam antibiotics alone (131, 132).
Another study (130) found that when rifampin was added to the combination of
vancomycin and gentamicin in broth, there was an enhanced bactericidal effect in
rabbits despite antagonism of the bactericidal rate. Notably, similar data for these
relationships on MRSA are lacking.

Vancomycin
Vancomycin remains the mainstay of therapy for MRSA PVE (29, 30). Vancomycin
dosing should be based on actual body weight and adjusted to achieve troughs of 15
to 20 ␮g/ml (29). Vancomycin binds the terminal D-alanyl-D-alanine moieties of
N-acetylmuramic acid (NAM)/N-acetylglucosamine (NAG) peptides in the bacterial cell
wall and thus prevents the addition of the NAM/NAG peptide subunits into the
peptidoglycan matrix of MRSA. It also acts by altering bacterial cell membrane perme-
ability and RNA synthesis (133). Common vancomycin toxicities include hypersensitivity
(“red man syndrome,” related to the infusion rate) after intravenous administration and
nephrotoxicity when used concomitantly with aminoglycosides (134). Significant
weight gain can also occur during prolonged treatment, particularly in older men (135).

Rifampin
The rifampin mechanism of action is based on the suppression of RNA synthesis
through the inhibition of the bacterial DNA-dependent RNA polymerase. Rifampin is
believed to bind to a pocket of the RNA polymerase ␤-subunit within the DNA/RNA
channel. This noncompetitive inhibitor prevents RNA synthesis by directly blocking RNA
elongation and thus preventing the synthesis of host bacterial proteins (136). Hepatic
and immunoallergic toxicities are the most common rifampin adverse effects. Hepato-

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 14


Staphylococcus aureus Prosthetic Valve Endocarditis Clinical Microbiology Reviews

toxicity usually affects patients with previous liver damage and is associated with the
dose administered. Adverse immunoallergic events might be minor or major and are
predominantly observed after prolonged or intermittent treatment (137).
Animal model experiments, in vitro data, and clinical observations have each dem-
onstrated an outsized beneficial effect of rifampin on foreign-material infections (24, 57,
127, 130, 138–140). Nevertheless, S. aureus has a high intrinsic mutation rate at the rpoB
gene, encoding the rifampin-binding site. When large numbers of S. aureus bacteria are
exposed to rifampin alone or in combination with ineffective antimicrobials, single
mutations in this region allow the rapid selection of a rifampin-resistant subpopulation
(57, 138). Consequently, recommended regimens support the selection of two addi-
tional antimicrobials with rifampin, to protect against the development of resistance to
rifampin. Similarly, many have recommended waiting until vancomycin and gentamicin
have been administered for 3 to 5 days, bacteremia has resolved, and undrained

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest


abscesses or collections have been debrided prior to initiation of rifampin (30). This
approach is supported by the in vitro antagonism demonstrated when rifampin is
exposed to replicating bacteria in combination with other antimicrobials (141) and the
synergism detected when bacteria are instead in a latent state (142). This effect can be
seen in biofilm-mediated foreign-body infections, such as those associated with ortho-
pedic hardware, prostheses, or vascular grafts (143). Lowy et al. (131) prevented the
development of rifampin-resistant S. epidermidis by adding either vancomycin or
gentamicin in a rabbit endocarditis model, but these observations have unclear clinical
utility in treating PVE in humans.
Rifampin is recommended for a minimum of 6 weeks, at a dosing regimen of 300 mg
every 8 h, in combination with gentamicin for the first 2 weeks and vancomycin for the
full treatment course of 6 weeks (29, 31). Notably, some authors alternatively recom-
mend rifampin at 600 mg once daily or 300 to 450 mg every 12 h with another
antistaphylococcal antibiotic for S. aureus infections (31).
The data on synergy between rifampin and other antimicrobials are conflicting (144,
145). Although several studies (146–148) have demonstrated that incorporating rifam-
pin into failing therapies can increase bactericidal rates and the chances of eradication
of serious S. aureus and S. epidermidis infections, others have reported both in vitro
synergy and antagonism for rifampin in combination with beta-lactam agents, vanco-
mycin, or gentamicin against S. aureus (149, 150). Another study concluded that there
was no synergy or antagonism against MRSE regarding the concomitant use of other
antibiotics (cephalothin, nafcillin, vancomycin, or gentamicin) with rifampin (127).
MRSE isolates harboring rifampin mutations appeared to be as virulent as their
rifampin-sensitive antecedents in rabbit endocarditis models, although a similar effect
was not seen in rifampin-resistant S. aureus isolates in a mouse model (151). The
mechanism of decreased virulence may be due to decreased production of toxins by
rifampin-resistant S. aureus (127). The emergence of rifampin resistance was not
prevented in vivo by the combination of rifampin with a beta-lactam antibiotic,
although it was prevented in vitro. There was no decrease in virulence of rifampin-
resistant methicillin-resistant S. epidermidis in comparison to rifampin-sensitive ante-
cedent strains (127). The MRSE study showed good bactericidal efficacy of rifampin in
vitro when there was a prevention of emergent rifampin-resistant mutants by a
beta-lactam antibiotic.

Gentamicin
European and U.S. guidelines suggest intramuscular or intravenous gentamicin at a
dose of 3 mg/kg of body weight every 24 h, once daily or in 2 or 3 divided doses, for
the treatment of MRSA PVE. Gentamicin serum levels and renal function should be
assessed at least once weekly, and more-frequent measurements are suggested in
cases of renal insufficiency. Gentamicin doses as high as 4 mg/kg/day have been
demonstrated to be successful in MRSA PVE treatment without additional toxicity in
both human and animal studies (152, 153). In nonobese adults, the gentamicin dose is
based on ideal body weight. Gentamicin is not distributed into adipose tissue, as it is

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 15


Galar et al. Clinical Microbiology Reviews

highly hydrophilic. Therefore, corrected body weight should be used for dosing calcu-
lations for obese patients, rather than ideal body weight. The gentamicin mechanism
of action is based on irreversible binding to specific proteins on the 30S subunit of the
MRSA ribosome and the decoding site of the 16S rRNA. This leads to a misreading of
the mRNA, the addition of incorrect amino acids in the growing peptide chain, and the
interruption of MRSA protein synthesis (154).
Ototoxicity and nephrotoxicity are important adverse events limiting gentamicin
clinical use. Both side effects are associated with the dose administered and might not
appear until the end of treatment. Kidney damage, in contrast to inner ear damage, is
usually reversible but can be fatal (155, 156). Gentamicin trough and peak values lower
than 1 ␮g/ml and 3 to 4 ␮g/ml, respectively, are recommended when gentamicin is
administered every 8 h. When gentamicin is dosed daily, serum troughs should be
lower than 1 ␮g/ml. ESC guidelines recommend that peaks be assessed once after

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest


infusion, with a goal range of 10 to 12 ␮g/ml (per AHA/IDSA guidelines, there is no role
for measuring peak gentamicin concentrations following single daily dosing).
The risk-to-benefit ratio of gentamicin therapy for MRSA PVE remains controversial.
In a study of 35 ex vivo strains of S. aureus isolated from blood cultures of septic patients
(23), vancomycin or nafcillin combined with tobramycin, gentamicin, or kanamycin had
improved activity against most of the strains. In a rabbit model of S. aureus endocarditis,
nafcillin and gentamicin altogether achieved a faster eradication of S. aureus at the
heart vegetation than nafcillin in monotherapy (22).
Nonetheless, clinical data in support of gentamicin use in this setting are lacking. In
a recent study comparing daptomycin therapy with an antistaphylococcal penicillin or
vancomycin in combination with gentamicin for MSSA or MRSA bacteremia and/or
right-sided endocarditis (157), significantly more patients who received standard ther-
apy with gentamicin suffered nephrotoxicity (18.1% versus 6.7% with renal tubular
necrosis and 46.8% versus 19.8% with worsening creatinine clearance), without an
improvement in clinical outcomes for those who received gentamicin (157). Whether
these results pertain to MRSA PVE is unknown, but this topic warrants urgent study
given the ongoing guideline recommendations for the use of gentamicin in the
treatment of MRSA PVE.

Alternative Therapies
If an isolate is resistant to gentamicin and all available aminoglycosides, a fluoro-
quinolone to which the strain is highly susceptible has been recommended (138–140).
If the patient is treated with a fluoroquinolone instead of an aminoglycoside, a
three-drug regimen for the entire course of treatment is preferred. In cases of resistance
to aminoglycosides and fluoroquinolones, ceftaroline, trimethoprim-sulfamethoxazole,
or linezolid (158) has been proposed as the third agent during the first 2 weeks of
treatment, if the isolate is susceptible in vitro.
For cases of MRSA PVE with reduced vancomycin susceptibility (MIC ⬎ 1.0 ␮g/ml),
substantial toxicity, or failure of vancomycin, the optimal treatment is not established.
Options include high-dose daptomycin (8 to 10 mg/kg once per day, if the isolate is
daptomycin susceptible), linezolid, telavancin, ceftaroline, and daptomycin combined
with ceftaroline, nafcillin, or fosfomycin, combinations that might result in synergy
(159–165). Yet reported clinical experience with these treatments and combination
therapies in MRSA PVE is limited. Daptomycin combination with rifampin and genta-
micin has been recommended in these cases as a second-line therapy for MRSA PVE
(30). Synergy between ␤-lactams and daptomycin is associated with several character-
istics, including increased daptomycin binding and ␤-lactam-mediated potentiation of
innate immunity, but the precise molecular mechanism is unknown (139). Dhand et
al. (166) reported a series of seven cases with rapid clearance of persistent MRSA
bacteremia when high-dose nafcillin was added to high-dose daptomycin. Other
experts (167) suggested the use of high-dose daptomycin combined with fosfomy-
cin for MRSA PVE.

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 16


Staphylococcus aureus Prosthetic Valve Endocarditis Clinical Microbiology Reviews

There are several case reports of ceftaroline being used to successfully treat refrac-
tory or drug-resistant MRSA PVE, either alone (168) or in combination therapy (169). In
one case report, the efficacy and tolerability of ceftaroline were demonstrated in one
patient with osteomyelitis and endocarditis caused by an MRSA strain that was not
susceptible to daptomycin (170). A patient with MRSA aortic PVE was also cured with
prolonged high-dose daptomycin plus ceftaroline after other therapeutic failures (169).
Ceftaroline fosamil was the most active bactericidal drug in a rabbit model of MRSA
endocarditis (171). For each MRSA strain, rabbits were randomized to no therapy
(controls), a ceftaroline fosamil dose equivalent to 10 mg/kg/12 h in humans (600 mg
twice daily), daptomycin at a dose comparable to 6 mg/kg/24 h in humans, or a
tigecycline dose equivalent to 100 mg/24 h in humans plus 50 mg/12 h. Both ceftaro-
line and daptomycin exhibited high bactericidal efficacy based on MRSA vegetation
reduction rates (⬎5 log10 CFU/g), whereas tigecycline did not show bactericidal effi-

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest


cacy, and MRSA vegetation reduction rates were ⬍2 log10 CFU/g in comparison to
controls. However, the MRSA vegetation sterilization rate by ceftaroline was 100%, in
contrast to the rate of 57% reached by daptomycin, with resistant mutants being seen
only in the daptomycin therapy group. Recent clinical data also demonstrate ceftaroline
as an alternative for MRSA bacteremia salvage treatment (170, 172–177). Two obser-
vational studies also suggest that ceftaroline therapy alone (178) or in combination
with trimethoprim-sulfamethoxazole (179) can be used to treat invasive MRSA infec-
tion, although more experience and, if possible, adequately designed clinical studies
are needed before there is widespread recommendation for its use.
Linezolid is an alternate treatment in cases of MRSA PVE complicated by drug allergy
or intolerance, although its use is limited by a relative scarcity of data and side effects
of prolonged use. Among 33 cases of endocarditis treated with linezolid, 21 (63.6%) had
a favorable outcome. PVE accounted for 25% of the reviewed cases, and MRSA
accounted for 24.2% (1 case of MRSA PVE among 8 PVE cases [12.5%]). The one patient
with MRSA PVE treated with linezolid had a favorable outcome. Additional efficacy and
tolerability data are required to better support the use of linezolid for PVE (180).
In an experimental rabbit model, Miró et al. (181) suggested that telavancin could
be as effective as vancomycin in the treatment of endocarditis caused by glycopeptide-
intermediate S. aureus (GISA). Other experimental IE models also showed telavancin
bactericidal activity against different MRSA strains, including daptomycin-resistant S.
aureus, vancomycin-intermediate S. aureus (VISA), and GISA (182–184). Telavancin (185)
and also quinupristin-dalfopristin (186) have been reported as favorable rescue thera-
pies in MRSA IE patients after vancomycin clinical failure.
The efficacy of oritavancin was also recently assessed in animal models of left-sided
MRSA endocarditis. The drug has gained attention for its single intravenous dosing
schedule at 1,200 mg over 3 h, which allows treatment of complicated MRSA infections
without the need for indwelling central venous catheters, a particular concern for
patients with recent or active injection drug use, and has shown promise in other
refractory and drug-resistant cases of PVE (187, 188). A left-sided MRSA endocarditis
rabbit model suggested that oritavancin was superior to vancomycin in resolving
bacteremia and reducing bacterial counts in vegetations and tissues (189). Those
investigators concluded that oritavancin was microbiologically effective and might be
an alternative to vancomycin in treating similar infections in humans (189). Of note,
Stewart et al. (190) reported a case series of 10 patients treated with oritavancin,
including 1 patient with NVE due to group B Streptococcus, who unfortunately failed
treatment. Although oritavancin is currently indicated only for acute bacterial skin and
skin structure infections, it has the potential to play an increasing role as an agent
against MRSA PVE, particularly in light of the ongoing international opioid epidemic
(191, 192).
Tigecycline has activity against MRSA; however, it has been demonstrated to have
a lower efficacy than vancomycin against MRSA strains (193), and its peak serum
concentrations do not exceed 1 ␮g/ml (194, 195). As such, tigecycline is not typically
recommended as an agent for MRSA bacteremia or endocarditis. Similarly, although

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 17


Galar et al. Clinical Microbiology Reviews

clindamycin and trimethoprim-sulfamethoxazole are theoretically alternative options


for MRSA PVE, experience with these agents for this condition is limited, and they are
typically considered only in the case of severe drug intolerance or as considerations in
synergistic regimens (196).

The “Endocarditis Team”


A multidisciplinary team including specialists from different clinical fields (neurology,
microbiology, infectious diseases, cardiology, imaging, surgery, and congenital heart dis-
ease) (30) has been recommended for the management of PVE (197, 198). Early involve-
ment of all team members is crucial to this strategy. Some studies have reported reductions
in 1-year mortality rates of PVE with the implementation of such teams (199, 200).

Choice and Timing of Surgery


Surgery is recommended for high-risk patients, particularly those with valvular

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest


failures complicated by heart failure, abscess formation, or fistulas and those not
responding to maximally effective antimicrobial therapy (30). The timing of surgery for
PVE is a topic of ongoing debate (2, 7, 16, 17, 201–203) and is a decision based on
surgeon- and patient-specific factors, including complications of disease, such as CNS
emboli and hemorrhage, and overall surgical risk. A recent study of 4,166 cases of
infective PVE and NVE in patients with heart failure found decreases in both hospital
and 1-year deaths with early surgery (during the index hospitalization) (65). More
recently, two prospective cohort studies, reported by Chirouze et al. (11) and Lalani
et al. (204), found a crude reduction in 1-year mortality rates with early versus delayed
surgery but similar rates after consideration of confounding and survivor bias. Thus, the
decision on the timing of therapy continues to largely depend on surgical risk and input
from multidisciplinary teams.

PROGNOSIS
The PVE mortality rate remains high in the current era, ranging from 30 to 80% for
early PVE and 20 to 40% for late PVE (54, 63). The identification of high-risk subgroups
is critical to establishing more-effective treatment strategies (9). Health care-associated
disease, staphylococcal or fungal etiologies, older age, diabetes mellitus, early PVE,
heart failure, CNS embolic disease, and intracardiac abscess are predictive of worse
clinical outcomes (1, 3, 8, 13, 65, 205). Among these, staphylococcal infection (5) and
complicated PVE (78) are the strongest indicators of poor outcomes. Patients with
complications require aggressive management, including antibiotic therapy and, often,
early surgery (30).

PREVENTION
The utility of antimicrobial prophylaxis for prevention of bacteremia and IE in
humans remains unclear (30, 206). Since the relaxation of antibiotic prophylaxis after
the 2007 guideline revisions, no incremental increase in the IE incidence was observed
(207, 208), with the exception of a recent ecological study showing increased rates of
Streptococcus but not staphylococcal IE cases since the guideline change (209). Accord-
ing to current guidelines, antimicrobial prophylaxis is recommended for high-risk
patients (30, 33, 71, 210–213) undergoing high-risk procedures (30), including those
with prosthetic heart valves. Probably as important as antibiotic prophylaxis is careful
attention to skin and dental hygiene. Moreover, procedures affecting gastrointestinal,
respiratory, musculoskeletal, genitourinary, and dermatological systems do not typically
require antimicrobial prophylaxis unless they are invasive (30).

CHALLENGES AND FUTURE PERSPECTIVES


MRSA is an increasingly common cause of PVE and continues to be among the most
morbid infections in the modern era. Recent data have changed our approach to this
disease. Multidisciplinary teams can improve outcomes (200, 214), and specialized
centers are an important aspect of optimizing care. Moreover, both nuclear imaging
modalities and molecular techniques that show promise in improving PVE diagnostic

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 18


Staphylococcus aureus Prosthetic Valve Endocarditis Clinical Microbiology Reviews

sensitivity are emerging. Finally, a number of newer agents that are active against
MRSA have recently been approved and have shown early efficacy in the management
of this condition. However, because prospective and randomized clinical data on the
management of MRSA PVE remain scarce, recently updated international practice
guidelines for IE from the ESC and the American College of Cardiology (30) continue to
rely largely on nonhuman data and expert opinion. As such, important priorities for the
field include (i) validating newer diagnostic modalities, such as advanced cardiac
imaging, that may increase the sensitivity and specificity of PVE diagnosis; (ii) clarifying
the role and optimal timing of valve replacement; (iii) performing comparative effec-
tiveness studies to assess newer alternatives to vancomycin with gentamicin and
rifampin as the mainstays of therapy; and (iv) clarifying the role of aminoglycosides in
MRSA PVE therapy.

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest


REFERENCES
1. Chirouze C, Cabell CH, Fowler VG, Jr, Khayat N, Olaison L, Miro JM, Erpelding M-L, Durante-Mangoni E, Fernández-Hidalgo N, Giannitsioti
Habib G, Abrutyn E, Eykyn S, Corey GR, Selton-Suty C, Hoen B, Inter- E, Hannan MM, Lejko-Zupanc T, Miró JM, Muñoz P, Murdoch DR,
national Collaboration on Endocarditis Study Group. 2004. Prognostic Tattevin P, Tribouilloy C, Hoen B, ICEP Prospective Investigators. 2015.
factors in 61 cases of Staphylococcus aureus prosthetic valve infective Impact of early valve surgery on outcome of Staphylococcus aureus
endocarditis from the International Collaboration on Endocarditis prosthetic valve infective endocarditis: analysis in the International
merged database. Clin Infect Dis 38:1323–1327. https://doi.org/10 Collaboration of Endocarditis-Prospective Cohort Study. Clin Infect Dis
.1086/383035. 60:741–749. https://doi.org/10.1093/cid/ciu871.
2. Habib G, Tribouilloy C, Thuny F, Giorgi R, Brahim A, Amazouz M, Remadi 12. Muñoz P, Kestler M, De Alarcon A, Miro JM, Bermejo J, Rodriguez-Abella
JP, Nadji G, Casalta JP, Coviaux F, Avierinos JF, Lescure X, Riberi A, H, Farinas MC, Cobo Belaustegui M, Mestres C, Llinares P, Goenaga M,
Weiller PJ, Metras D, Raoult D. 2005. Prosthetic valve endocarditis: who Navas E, Oteo JA, Tarabini P, Bouza E, Spanish Collaboration on
needs surgery? A multicentre study of 104 cases. Heart 91:954 –959. Endocarditis-Grupo de Apoyo al Manejo de la Endocarditis Infecciosa
https://doi.org/10.1136/hrt.2004.046177. en España. 2015. Current epidemiology and outcome of infective
3. John MD, Hibberd PL, Karchmer AW, Sleeper LA, Calderwood SB. 1998. endocarditis: a multicenter, prospective, cohort study. Medicine (Balti-
Staphylococcus aureus prosthetic valve endocarditis: optimal manage- more) 94:e1816. https://doi.org/10.1097/MD.0000000000001816.
ment and risk factors for death. Clin Infect Dis 26:1302–1309. 13. Tornos P, Almirante B, Olona M, Permanyer G, Gonzalez T, Carballo J,
4. Murdoch DR, Corey GR, Hoen B, Miro JM, Fowler VG, Jr, Bayer AS, Pahissa A, Soler-Soler J. 1997. Clinical outcome and long-term progno-
Karchmer AW, Olaison L, Pappas PA, Moreillon P, Chambers ST, Chu VH, sis of late prosthetic valve endocarditis: a 20-year experience. Clin
Falco V, Holland DJ, Jones P, Klein JL, Raymond NJ, Read KM, Tripodi Infect Dis 24:381–386.
MF, Utili R, Wang A, Woods CW, Cabell CH, International Collaboration 14. Alonso-Valle H, Fariñas-Álvarez C, García-Palomo JD, Bernal JM, Martín-
on Endocarditis-Prospective Cohort Study Investigators. 2009. Clinical Durán R, Gutiérrez Díez JF, Revuelta JM, Fariñas MC. 2010. Clinical
presentation, etiology, and outcome of infective endocarditis in the course and predictors of death in prosthetic valve endocarditis over a
21st century: the International Collaboration on Endocarditis- 20-year period. J Thorac Cardiovasc Surg 139:887– 893. https://doi.org/
Prospective Cohort Study. Arch Intern Med 169:463– 473. https://doi 10.1016/j.jtcvs.2009.05.042.
.org/10.1001/archinternmed.2008.603. 15. Fernández Guerrero ML, González López JJ, Goyenechea A, Fraile J, de
5. Selton-Suty C, Celard M, Le Moing V, Doco-Lecompte T, Chirouze C, Górgolas M. 2009. Endocarditis caused by Staphylococcus aureus: a
Iung B, Strady C, Revest M, Vandenesch F, Bouvet A, Delahaye F, Alla F, reappraisal of the epidemiologic, clinical, and pathologic manifesta-
Duval X, Hoen B, AEPEI Study Group. 2012. Preeminence of Staphylo- tions with analysis of factors determining outcome. Medicine (Balti-
coccus aureus in infective endocarditis: a 1-year population-based sur- more) 88:1–22. https://doi.org/10.1097/MD.0b013e318194da65.
vey. Clin Infect Dis 54:1230 –1239. https://doi.org/10.1093/cid/cis199. 16. Hill EE, Herregods MC, Vanderschueren S, Claus P, Peetermans WE,
6. Wang A, Athan E, Pappas PA, Fowler VG, Jr, Olaison L, Pare C, Almirante Herijgers P. 2008. Management of prosthetic valve infective
B, Munoz P, Rizzi M, Naber C, Logar M, Tattevin P, Iarussi DL, Selton-Suty endocarditis. Am J Cardiol 101:1174 –1178. https://doi.org/10.1016/
C, Jones SB, Casabe J, Morris A, Corey GR, Cabell CH, International j.amjcard.2007.12.015.
Collaboration on Endocarditis-Prospective Cohort Study Investigators. 17. Sohail MR, Martin KR, Wilson WR, Baddour LM, Harmsen WS, Steckel-
2007. Contemporary clinical profile and outcome of prosthetic valve berg JM. 2006. Medical versus surgical management of Staphylococcus
endocarditis. JAMA 297:1354 –1361. aureus prosthetic valve endocarditis. Am J Med 119:147–154. https://
7. Wang A, Pappas P, Anstrom KJ, Abrutyn E, Fowler VG, Jr, Hoen B, Miro doi.org/10.1016/j.amjmed.2005.09.037.
JM, Corey GR, Olaison L, Stafford JA, Mestres CA, Cabell CH, Interna- 18. Fowler VG, Jr, Sexton DJ. 1998. The role of valve replacement in the
tional Collaboration on Endocarditis Investigators. 2005. The use and treatment of prosthetic valve endocarditis. Clin Infect Dis 26:
effect of surgical therapy for prosthetic valve infective endocarditis: a 1310 –1311.
propensity analysis of a multicenter, international cohort. Am Heart J 19. Baddour LM, Wilson WR, Bayer AS, Fowler VG, Jr, Bolger AF, Levison ME,
150:1086 –1091. https://doi.org/10.1016/j.ahj.2005.01.023. Ferrieri P, Gerber MA, Tani LY, Gewitz MH, Tong DC, Steckelberg JM,
8. Wolff M, Witchitz S, Chastang C, Regnier B, Vachon F. 1995. Prosthetic Baltimore RS, Shulman ST, Burns JC, Falace DA, Newburger JW, Pallasch
valve endocarditis in the ICU. Prognostic factors of overall survival in a TJ, Takahashi M, Taubert KA, Committee on Rheumatic Fever, Endocar-
series of 122 cases and consequences for treatment decision. Chest ditis, and Kawasaki Disease, Council on Cardiovascular Disease in the
108:688 – 694. https://doi.org/10.1378/chest.108.3.688. Young, Councils on Clinical Cardiology, Stroke, and Cardiovascular
9. Sy RW, Chawantanpipat C, Richmond DR, Kritharides L. 2011. Develop- Surgery and Anesthesia, American Heart Association, Infectious Dis-
ment and validation of a time-dependent risk model for predicting eases Society of America. 2005. Infective endocarditis: diagnosis, anti-
mortality in infective endocarditis. Eur Heart J 32:2016 –2026. https:// microbial therapy, and management of complications. A statement for
doi.org/10.1093/eurheartj/ehp085. healthcare professionals from the Committee on Rheumatic Fever,
10. Yu VL, Fang GD, Keys TF, Harris AA, Gentry LO, Fuchs PC, Wagener MM, Endocarditis, and Kawasaki Disease, Council on Cardiovascular Disease
Wong ES. 1994. Prosthetic valve endocarditis: superiority of surgical in the Young, and the Councils on Clinical Cardiology, Stroke, and
valve replacement versus medical therapy only. Ann Thorac Surg 58: Cardiovascular Surgery and Anesthesia, American Heart Association:
1073–1077. endorsed by the Infectious Diseases Society of America. Circulation
11. Chirouze C, Alla F, Fowler VG, Sexton DJ, Corey GR, Chu VH, Wang A, 111:e394 – e434.

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 19


Galar et al. Clinical Microbiology Reviews

20. Habib G, Hoen B, Tornos P, Thuny F, Prendergast B, Vilacosta I, Moreil- Gómez I, Rollán MJ, San Román JA. 2007. Definition, clinical profile,
lon P, de Jesus Antunes M, Thilen U, Lekakis J, Lengyel M, Muller L, microbiological spectrum, and prognostic factors of early-onset
Naber CK, Nihoyannopoulos P, Moritz A, Zamorano JL, ESC Committee prosthetic valve endocarditis. Eur Heart J 28:760 –765. https://doi
for Practice Guidelines. 2009. Guidelines on the prevention, diagnosis, .org/10.1093/eurheartj/ehl486.
and treatment of infective endocarditis (new version 2009): the Task 35. Rutledge R, Kim BJ, Applebaum RE. 1985. Actuarial analysis of the risk
Force on the Prevention, Diagnosis, and Treatment of Infective Endo- of prosthetic valve endocarditis in 1,598 patients with mechanical and
carditis of the European Society of Cardiology (ESC). Endorsed by the bioprosthetic valves. Arch Surg 120:469 – 472.
European Society of Clinical Microbiology and Infectious Diseases 36. Agnihotri AK, McGiffin DC, Galbraith AJ, O’Brien MF. 1995. The preva-
(ESCMID) and the International Society of Chemotherapy (ISC) for lence of infective endocarditis after aortic valve replacement. J Thorac
Infection and Cancer. Eur Heart J 30:2369 –2413. https://doi.org/10 Cardiovasc Surg 110:1708 –1720; discussion, 1720 –1724. https://doi
.1093/eurheartj/ehp285. .org/10.1016/S0022-5223(95)70035-8.
21. Hoeprich PD. 1972. Infective endocarditis, p 1045–1060. In Hoeprich PD 37. Calderwood SB, Swinski LA, Waternaux CM, Karchmer AW, Buckley MJ.
(ed), Infectious diseases. Harper & Row, New York, NY. 1985. Risk factors for the development of prosthetic valve endocarditis.
22. Sande MA, Courtney KB. 1976. Nafcillin-gentamicin synergism in exper- Circulation 72:31–37.
imental staphylococcal endocarditis. J Lab Clin Med 88:118 –124. 38. Horstkotte D, Piper C, Niehues R, Wiemer M, Schultheiss HP. 1995. Late
23. Watanakunakorm C, Glotzbecker C. 1974. Enhancement of the effects prosthetic valve endocarditis. Eur Heart J 16:39 – 47.
of anti-staphylococcal antibiotics by aminoglycosides. Antimicrob 39. Ivert TS, Dismukes WE, Cobbs CG, Blackstone EH, Kirklin JW, Bergdahl

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest


Agents Chemother 6:802– 806. LA. 1984. Prosthetic valve endocarditis. Circulation 69:223–232.
24. Karchmer AW, Archer GL, Dismukes WE. 1983. Staphylococcus epider- 40. Karchmer AW, Longworth DL. 2002. Infections of intracardiac devices.
midis causing prosthetic valve endocarditis: microbiologic and clinical Infect Dis Clin North Am 16:477–505.
observations as guides to therapy. Ann Intern Med 98:447– 455. 41. Lee JH, Burner KD, Fealey ME, Edwards WD, Tazelaar HD, Orszulak
25. Watanakunakorn C, Baird IM. 1977. Prognostic factors in Staphylococcus TA, Wright AJ, Baddour LM. 2011. Prosthetic valve endocarditis:
aureus endocarditis and results of therapy with a penicillin and genta- clinicopathological correlates in 122 surgical specimens from 116
micin. Am J Med Sci 273:133–139. patients (1985–2004). Cardiovasc Pathol 20:26 –35. https://doi.org/
26. Rajashekaraiah KR, Rice T, Rao VS, Marsh D, Ramakrishna B, Kallick CA. 10.1016/j.carpath.2009.09.006.
1980. Clinical significance of tolerant strains of Staphylococcus aureus in 42. Karchmer AW, Chu VH. 2018. Prosthetic valve endocarditis: epidemiol-
patients with endocarditis. Ann Intern Med 93:796 – 801. ogy, clinical manifestations, and diagnosis. UpToDate. https://www
27. Ribera E, Gómez-Jimenez J, Cortes E, del Valle O, Planes A, Gonzalez- .uptodate.com/contents/prosthetic-valve-endocarditis-epidemiology
Alujas T, Almirante B, Ocaña I, Pahissa A. 1996. Effectiveness of cloxa- -clinical-manifestations-and-diagnosis. Accessed 21 March 2018.
cillin with and without gentamicin in short-term therapy for right-sided 43. Mahesh B, Angelini G, Caputo M, Jin XY, Bryan A. 2005. Prosthetic valve
Staphylococcus aureus endocarditis. A randomized, controlled trial. Ann endocarditis. Ann Thorac Surg 80:1151–1158. https://doi.org/10.1016/
Intern Med 125:969 –974.
j.athoracsur.2004.11.001.
28. Baltimore RS, Gewitz M, Baddour LM, Beerman LB, Jackson MA, Lock-
44. Moreillon P, Que YA. 2004. Infective endocarditis. Lancet 363:139 –149.
hart PB, Pahl E, Schutze GE, Shulman ST, Willoughby R, Jr. 15 Septem-
https://doi.org/10.1016/S0140-6736(03)15266-X.
ber 2015. Infective endocarditis in childhood: 2015 update. A scientific
45. Piper C, Korfer R, Horstkotte D. 2001. Prosthetic valve endocarditis.
statement from the American Heart Association. Circulation https://doi
Heart 85:590 –593. https://doi.org/10.1136/heart.85.5.590.
.org/10.1161/CIR.0000000000000298.
46. Dismukes WE, Karchmer AW, Buckley MJ, Austen WG, Swartz MN. 1973.
29. Baddour LM, Wilson WR, Bayer AS, Fowler VG, Jr, Tleyjeh IM, Rybak MJ,
Prosthetic valve endocarditis. Analysis of 38 cases. Circulation 48:
Barsic B, Lockhart PB, Gewitz MH, Levison ME, Bolger AF, Steckelberg
365–377.
JM, Baltimore RS, Fink AM, O’Gara P, Taubert KA. 15 September 2015.
47. Richardson JV, Karp RB, Kirklin JW, Dismukes WE. 1978. Treatment of
Infective endocarditis in adults: diagnosis, antimicrobial therapy, and
infective endocarditis: a 10-year comparative analysis. Circulation 58:
management of complications. A scientific statement for healthcare
589 –597.
professionals from the American Heart Association. Circulation https://
48. Ben Ismail M, Hannachi N, Abid F, Kaabar Z, Rougé JF. 1987. Prosthetic
doi.org/10.1161/CIR.0000000000000296.
30. Habib G, Lancellotti P, Antunes MJ, Bongiorni MG, Casalta JP, Del Zotti valve endocarditis. A survey. Br Heart J 58:72–77.
F, Dulgheru R, El Khoury G, Erba PA, Iung B, Miro JM, Mulder BJ, 49. Anderson DJ, Bulkley BH, Hutchins GM. 1977. A clinicopathologic study
Plonska-Gosciniak E, Price S, Roos-Hesselink J, Snygg-Martin U, Thuny F, of prosthetic valve endocarditis in 22 patients: morphologic basis for
Tornos Mas P, Vilacosta I, Zamorano JL, ESC Scientific Document Group. diagnosis and therapy. Am Heart J 94:325–332.
2015. 2015 ESC guidelines for the management of infective endocar- 50. Arnett EN, Roberts WC. 1976. Prosthetic valve endocarditis: clinicopath-
ditis. The Task Force for the Management of Infective Endocarditis of ologic analysis of 22 necropsy patients with comparison observations
the European Society of Cardiology (ESC) endorsed by: European As- in 74 necropsy patients with active infective endocarditis involving
sociation for Cardio-Thoracic Surgery (EACTS), the European Associa- natural left-sided cardiac valves. Am J Cardiol 38:281–292.
tion of Nuclear Medicine (EANM). Eur Heart J 36:3075–3128. https://doi 51. Rose AG. 1986. Prosthetic valve endocarditis. A clinicopathological
.org/10.1093/eurheartj/ehv319. study of 31 cases. S Afr Med J 69:441– 445.
31. Liu C, Bayer A, Cosgrove SE, Daum RS, Fridkin SK, Gorwitz RJ, Kaplan SL, 52. Holland TL, Baddour LM, Bayer AS, Hoen B, Miro JM, Fowler VG, Jr. 2016.
Karchmer AW, Levine DP, Murray BE, Rybak MJ, Talan DA, Chambers HF, Infective endocarditis. Nat Rev Dis Primers 2:16059. https://doi.org/10
Infectious Diseases Society of America. 2011. Clinical practice guide- .1038/nrdp.2016.59.
lines by the Infectious Diseases Society of America for the treatment of 53. Sampedro MF, Patel R. 2007. Infections associated with long-term
methicillin-resistant Staphylococcus aureus infections in adults and chil- prosthetic devices. Infect Dis Clin North Am 21:785– 819. https://doi
dren. Clin Infect Dis 52:e18 – e55. https://doi.org/10.1093/cid/ciq146. .org/10.1016/j.idc.2007.07.001.
32. Watanakunakorn C. 1979. Prosthetic valve infective endocarditis. Prog 54. Habib G, Thuny F, Avierinos JF. 2008. Prosthetic valve endocarditis:
Cardiovasc Dis 22:181–192. current approach and therapeutic options. Prog Cardiovasc Dis 50:
33. Horstkotte D, Follath F, Gutschik E, Lengyel M, Oto A, Pavie A, Soler- 274 –281. https://doi.org/10.1016/j.pcad.2007.10.007.
Soler J, Thiene G, von Graevenitz A, Priori SG, Garcia MA, Blanc JJ, Budaj 55. Seidl K, Bayer AS, Fowler VG, Jr, McKinnell JA, Abdel Hady W, Sakoulas
A, Cowie M, Dean V, Deckers J, Fernandez Burgos E, Lekakis J, Lindahl G, Yeaman MR, Xiong YQ. 2011. Combinatorial phenotypic signatures
B, Mazzotta G, Morais J, Oto A, Smiseth OA, Lekakis J, Vahanian A, distinguish persistent from resolving methicillin-resistant Staphylococ-
Delahaye F, Parkhomenko A, Filipatos G, Aldershvile J, Vardas P, Task cus aureus bacteremia isolates. Antimicrob Agents Chemother 55:
Force Members on Infective Endocarditis of the European Society of 575–582. https://doi.org/10.1128/AAC.01028-10.
Cardiology, ESC Committee for Practice Guidelines, Document Review- 56. Xiong YQ, Fowler VG, Yeaman MR, Perdreau-Remington F, Kreiswirth
ers. 2004. Guidelines on prevention, diagnosis and treatment of infec- BN, Bayer AS. 2009. Phenotypic and genotypic characteristics of per-
tive endocarditis executive summary; the Task Force on Infective En- sistent methicillin-resistant Staphylococcus aureus bacteremia in vitro
docarditis of the European Society of Cardiology. Eur Heart J 25: and in an experimental endocarditis model. J Infect Dis 199:201–208.
267–276. https://doi.org/10.1016/j.ehj.2003.11.008. https://doi.org/10.1086/595738.
34. López J, Revilla A, Vilacosta I, Villacorta E, González-Juanatey C, 57. Karchmer AW. 2000. Infections of prosthetic heart valves, p 145–172. In

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 20


Staphylococcus aureus Prosthetic Valve Endocarditis Clinical Microbiology Reviews

Waldvogel FA, Bisno AL (ed), Infections associated with indwelling Affairs randomized trial. J Am Coll Cardiol 36:1152–1158. https://doi
medical devices, 3rd ed. ASM Press, Washington, DC. .org/10.1016/S0735-1097(00)00834-2.
58. Lytle BW, Priest BP, Taylor PC, Loop FD, Sapp SK, Stewart RW, McCarthy 75. Oxenham H, Bloomfield P, Wheatley DJ, Lee RJ, Cunningham J, Prescott
PM, Muehrcke D, Cosgrove DM, III, 1996. Surgical treatment of pros- RJ, Miller HC. 2003. Twenty year comparison of a Bjork-Shiley mechan-
thetic valve endocarditis. J Thorac Cardiovasc Surg 111:198 –207; dis- ical heart valve with porcine bioprostheses. Heart 89:715–721.
cussion, 207–110. https://doi.org/10.1016/S0022-5223(96)70417-8. 76. Stassano P, Di Tommaso L, Monaco M, Iorio F, Pepino P, Spampinato N,
59. Amat-Santos IJ, Messika-Zeitoun D, Eltchaninoff H, Kapadia S, Lerakis S, Vosa C. 2009. Aortic valve replacement: a prospective randomized
Cheema AN, Gutiérrez-Ibanes E, Munoz-Garcia AJ, Pan M, Webb JG, evaluation of mechanical versus biological valves in patients ages 55 to
Herrmann HC, Kodali S, Nombela-Franco L, Tamburino C, Jilaihawi H, 70 years. J Am Coll Cardiol 54:1862–1868. https://doi.org/10.1016/j.jacc
Masson J-B, de Brito FS, Jr, Ferreira MC, Lima VC, Mangione JA, Iung B, .2009.07.032.
Vahanian A, Durand E, Tuzcu EM, Hayek SS, Angulo-Llanos R, Gómez- 77. Brennan JM, Edwards FH, Zhao Y, O’Brien S, Booth ME, Dokholyan RS,
Doblas JJ, Castillo JC, Dvir D, Leon MB, Garcia E, Cobiella J, Vilacosta I, Douglas PS, Peterson ED, DEcIDE AVR Research Team. 2013. Long-term
Barbanti M, Makkar RR, Ribeiro HB, Urena M, Dumont E, Pibarot P, Lopez safety and effectiveness of mechanical versus biologic aortic valve
J, San Roman A, Rodés-Cabau J. 2015. Infective endocarditis after trans- prostheses in older patients: results from the Society of Thoracic Sur-
catheter aortic valve implantation: results from a large multicenter registry. geons Adult Cardiac Surgery National Database. Circulation 127:
Circulation 131:1566–1574. https://doi.org/10.1161/CIRCULATIONAHA.114 1647–1655. https://doi.org/10.1161/CIRCULATIONAHA.113.002003.
.014089. 78. Calderwood SB, Swinski LA, Karchmer AW, Waternaux CM, Buckley MJ.

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest


60. Pericas JM, Llopis J, Cervera C, Sacanella E, Falces C, Andrea R, Garcia de 1986. Prosthetic valve endocarditis. Analysis of factors affecting out-
la Maria C, Ninot S, Vidal B, Almela M, Pare JC, Sabate M, Moreno A, come of therapy. J Thorac Cardiovasc Surg 92:776 –783.
Marco F, Mestres CA, Miro JM, Hospital Clinic Endocarditis Study Group. 79. Karchmer AW, Dismukes WE, Buckley MJ, Austen WG. 1978. Late pros-
2015. Infective endocarditis in patients with an implanted transcatheter thetic valve endocarditis: clinical features influencing therapy. Am J
aortic valve: clinical characteristics and outcome of a new entity. J Med 64:199 –206.
Infect 70:565–576. https://doi.org/10.1016/j.jinf.2014.12.013. 80. Conlon PJ, Jefferies F, Krigman HR, Corey GR, Sexton DJ, Abramson MA.
61. Tong SY, Davis JS, Eichenberger E, Holland TL, Fowler VG, Jr. 2015. 1998. Predictors of prognosis and risk of acute renal failure in bacterial
Staphylococcus aureus infections: epidemiology, pathophysiology, clin- endocarditis. Clin Nephrol 49:96 –101.
ical manifestations, and management. Clin Microbiol Rev 28:603– 661. 81. Speechly-Dick ME, Swanton RH. 1994. Osteomyelitis and infective en-
https://doi.org/10.1128/CMR.00134-14. docarditis. Postgrad Med J 70:885– 890.
62. Lepidi H, Casalta JP, Fournier PE, Habib G, Collart F, Raoult D. 2006. 82. Sexauer WP, Quezado Z, Lippmann ML, Goldberg SK. 1992. Pleural
Quantitative histological examination of bioprosthetic heart valves. effusions in right-sided endocarditis: characteristics and pathophysiol-
Clin Infect Dis 42:590 –596. https://doi.org/10.1086/500135. ogy. South Med J 85:1176 –1180.
63. Vongpatanasin W, Hillis LD, Lange RA. 1996. Prosthetic heart valves. N Engl 83. Røder BL, Wandall DA, Espersen F, Frimodt-Møller N, Skinhøj P, Rosdahl
VT. 1997. Neurologic manifestations in Staphylococcus aureus
J Med 335:407–416. https://doi.org/10.1056/NEJM199608083350607.
endocarditis: a review of 260 bacteremic cases in nondrug addicts. Am
64. Bashore TM, Cabell C, Fowler V, Jr. 2006. Update on infective endocar-
J Med 102:379 –386.
ditis. Curr Probl Cardiol 31:274 –352. https://doi.org/10.1016/j.cpcardiol
84. Merkler AE, Chu SY, Lerario MP, Navi BB, Kamel H. 2015. Temporal
.2005.12.001.
relationship between infective endocarditis and stroke. Neurology 85:
65. Kiefer T, Park L, Tribouilloy C, Cortes C, Casillo R, Chu V, Delahaye F,
512–516. https://doi.org/10.1212/WNL.0000000000001835.
Durante-Mangoni E, Edathodu J, Falces C, Logar M, Miro JM, Naber C,
85. Garcia-Cabrera E, Fernandez-Hidalgo N, Almirante B, Ivanova-Georgieva
Tripodi MF, Murdoch DR, Moreillon P, Utili R, Wang A. 2011. Association
R, Noureddine M, Plata A, Lomas JM, Galvez-Acebal J, Hidalgo-Tenorio
between valvular surgery and mortality among patients with infective
C, Ruiz-Morales J, Martinez-Marcos FJ, Reguera JM, de la Torre-Lima J,
endocarditis complicated by heart failure. JAMA 306:2239 –2247.
de Alarcon Gonzalez A, Group for the Study of Cardiovascular Infec-
https://doi.org/10.1001/jama.2011.1701.
tions of the Andalusian Society of Infectious Diseases, Spanish Network
66. Moreillon P, Wilson WR, Leclercq R, Entenza JM. 2007. Single-dose oral
for Research in Infectious Diseases. 2013. Neurological complications of
amoxicillin or linezolid for prophylaxis of experimental endocarditis
infective endocarditis: risk factors, outcome, and impact of cardiac
due to vancomycin-susceptible and vancomycin-resistant Enterococcus surgery: a multicenter observational study. Circulation 127:2272–2284.
faecalis. Antimicrob Agents Chemother 51:1661–1665. https://doi.org/ https://doi.org/10.1161/CIRCULATIONAHA.112.000813.
10.1128/AAC.00744-06. 86. Jones HR, Jr, Siekert RG. 1989. Neurological manifestations of infective
67. Fefer P, Raveh D, Rudensky B, Schlesinger Y, Yinnon AM. 2002. Chang- endocarditis. Review of clinical and therapeutic challenges. Brain 112:
ing epidemiology of infective endocarditis: a retrospective survey of 1295–1315.
108 cases, 1990 –1999. Eur J Clin Microbiol Infect Dis 21:432– 437. 87. Snygg-Martin U, Gustafsson L, Rosengren L, Alsio A, Ackerholm P, Ander-
68. Hoen B, Alla F, Selton-Suty C, Beguinot I, Bouvet A, Briancon S, Casalta sson R, Olaison L. 2008. Cerebrovascular complications in patients with
JP, Danchin N, Delahaye F, Etienne J, Le Moing V, Leport C, Mainardi JL, left-sided infective endocarditis are common: a prospective study using
Ruimy R, Vandenesch F, Association pour l’Etude et la Prevention de magnetic resonance imaging and neurochemical brain damage markers.
l’Endocardite Infectieuse Study Group. 2002. Changing profile of infec- Clin Infect Dis 47:23–30. https://doi.org/10.1086/588663.
tive endocarditis: results of a 1-year survey in France. JAMA 288:75– 81. 88. Fowler VG, Jr, Miro JM, Hoen B, Cabell CH, Abrutyn E, Rubinstein E,
69. Netzer RO, Zollinger E, Seiler C, Cerny A. 2000. Infective endocarditis: Corey GR, Spelman D, Bradley SF, Barsic B, Pappas PA, Anstrom KJ, Wray
clinical spectrum, presentation and outcome. An analysis of 212 cases D, Fortes CQ, Anguera I, Athan E, Jones P, van der Meer JTM, Elliott TSJ,
1980-1995. Heart 84:25–30. https://doi.org/10.1136/heart.84.1.25. Levine DP, Bayer AS, ICE Investigators. 2005. Staphylococcus aureus
70. Tornos P, Iung B, Permanyer-Miralda G, Baron G, Delahaye F, Gohlke- endocarditis: a consequence of medical progress. JAMA 293:
Bärwolf C, Butchart EG, Ravaud P, Vahanian A. 2005. Infective endocar- 3012–3021. https://doi.org/10.1001/jama.293.24.3012.
ditis in Europe: lessons from the Euro heart survey. Heart 91:571–575. 89. Keyser DL, Biller J, Coffman TT, Adams HP, Jr. 1990. Neurologic com-
https://doi.org/10.1136/hrt.2003.032128. plications of late prosthetic valve endocarditis. Stroke 21:472– 475.
71. Stein PD, Harken DE, Dexter L. 1966. The nature and prevention of https://doi.org/10.1161/01.STR.21.3.472.
prosthetic valve endocarditis. Am Heart J 71:393– 407. 90. Masur H, Johnson WD, Jr. 1980. Prosthetic valve endocarditis. J Thorac
72. Arvay A, Lengyel M. 1988. Incidence and risk factors of prosthetic valve Cardiovasc Surg 80:31–37.
endocarditis. Eur J Cardiothorac Surg 2:340 –346. 91. Tornos P, Sanz E, Permanyer-Miralda G, Almirante B, Planes AM, Soler-
73. Grover FL, Cohen DJ, Oprian C, Henderson WG, Sethi G, Hammermeis- Soler J. 1992. Late prosthetic valve endocarditis. Immediate and long-
ter KE. 1994. Determinants of the occurrence of and survival from term prognosis. Chest 101:37– 41.
prosthetic valve endocarditis. Experience of the Veterans Affairs Coop- 92. Davenport J, Hart RG. 1990. Prosthetic valve endocarditis 1976-1987.
erative Study on Valvular Heart Disease. J Thorac Cardiovasc Surg Antibiotics, anticoagulation, and stroke. Stroke 21:993–999. https://doi
108:207–214. .org/10.1161/01.STR.21.7.993.
74. Hammermeister K, Sethi GK, Henderson WG, Grover FL, Oprian C, 93. Breen JD, Karchmer AW. 1994. Usefulness of pulsed-field gel electro-
Rahimtoola SH. 2000. Outcomes 15 years after valve replacement with phoresis in confirming endocarditis due to Staphylococcus lugdunensis.
a mechanical versus a bioprosthetic valve: final report of the Veterans Clin Infect Dis 19:985–986. https://doi.org/10.1093/clinids/19.5.985.

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 21


Galar et al. Clinical Microbiology Reviews

94. Goldenberger D, Kunzli A, Vogt P, Zbinden R, Altwegg M. 1997. Mo- Habib G. 2013. Imaging investigations in infective endocarditis: current
lecular diagnosis of bacterial endocarditis by broad-range PCR ampli- approach and perspectives. Arch Cardiovasc Dis 106:52– 62. https://doi
fication and direct sequencing. J Clin Microbiol 35:2733–2739. .org/10.1016/j.acvd.2012.09.004.
95. Habib G. 2006. Management of infective endocarditis. Heart 92: 113. Fagman E, Perrotta S, Bech-Hanssen O, Flinck A, Lamm C, Olaison L,
124 –130. https://doi.org/10.1136/hrt.2005.063719. Svensson G. 2012. ECG-gated computed tomography: a new role for
96. Cheitlin MD, Armstrong WF, Aurigemma GP, Beller GA, Bierman FZ, patients with suspected aortic prosthetic valve endocarditis. Eur Radiol
Davis JL, Douglas PS, Faxon DP, Gillam LD, Kimball TR, Kussmaul WG, 22:2407–2414. https://doi.org/10.1007/s00330-012-2491-5.
Pearlman AS, Philbrick JT, Rakowski H, Thys DM. 2003. ACC/AHA/ASE 114. Feuchtner GM, Stolzmann P, Dichtl W, Schertler T, Bonatti J, Scheffel H,
2003 guideline update for the clinical application of echocardiogra- Mueller S, Plass A, Mueller L, Bartel T, Wolf F, Alkadhi H. 2009. Multislice
phy—summary article: a report of the American College of Cardiology/ computed tomography in infective endocarditis: comparison with
American Heart Association Task Force on Practice Guidelines (ACC/ transesophageal echocardiography and intraoperative findings. J Am
AHA/ASE Committee To Update the 1997 Guidelines for the Clinical Coll Cardiol 53:436 – 444. https://doi.org/10.1016/j.jacc.2008.01.077.
Application of Echocardiography). J Am Coll Cardiol 42:954 –970. 115. Habets J, Tanis W, van Herwerden LA, van den Brink RB, Mali WP, de
https://doi.org/10.1016/S0735-1097(03)01065-9. Mol BA, Chamuleau SA, Budde RP. 2014. Cardiac computed tomogra-
97. Bruun NE, Habib G, Thuny F, Sogaard P. 2014. Cardiac imaging in phy angiography results in diagnostic and therapeutic change in pros-
infectious endocarditis. Eur Heart J 35:624 – 632. https://doi.org/10 thetic heart valve endocarditis. Int J Cardiovasc Imaging 30:377–387.
.1093/eurheartj/eht274. https://doi.org/10.1007/s10554-013-0335-2.

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest


98. Daniel WG, Mugge A, Grote J, Hausmann D, Nikutta P, Laas J, Lichtlen 116. Saby L, Laas O, Habib G, Cammilleri S, Mancini J, Tessonnier L, Casalta
PR, Martin RP. 1993. Comparison of transthoracic and transesophageal JP, Gouriet F, Riberi A, Avierinos JF, Collart F, Mundler O, Raoult D,
echocardiography for detection of abnormalities of prosthetic and Thuny F. 2013. Positron emission tomography/computed tomography
bioprosthetic valves in the mitral and aortic positions. Am J Cardiol for diagnosis of prosthetic valve endocarditis: increased valvular 18F-
71:210 –215. fluorodeoxyglucose uptake as a novel major criterion. J Am Coll Cardiol
99. Morguet AJ, Werner GS, Andreas S, Kreuzer H. 1995. Diagnostic value of 61:2374 –2382. https://doi.org/10.1016/j.jacc.2013.01.092.
transesophageal compared with transthoracic echocardiography in 117. Fagman E, van Essen M, Freden Lindqvist J, Snygg-Martin U, Bech-
suspected prosthetic valve endocarditis. Herz 20:390 –398. Hanssen O, Svensson G. 2016. (18)F-FDG PET/CT in the diagnosis of
100. Khandheria BK. 1995. Transesophageal echocardiography in the eval- prosthetic valve endocarditis. Int J Cardiovasc Imaging 32:679 – 686.
uation of prosthetic valves. Am J Card Imaging 9:106 –114. https://doi.org/10.1007/s10554-015-0814-8.
101. Stewart WJ, Shan K. 1995. The diagnosis of prosthetic valve endocar- 118. Pizzi MN, Roque A, Fernández-Hidalgo N, Cuéllar-Calabria H, Ferreira-
ditis by echocardiography. Semin Thorac Cardiovasc Surg 7:7–12. González I, Gonzàlez-Alujas MT, Oristrell G, Gracia-Sánchez L, González
102. Habets J, Tanis W, Reitsma JB, van den Brink RB, Mali WP, Chamuleau JJ, Rodríguez-Palomares J, Galiñanes M, Maisterra-Santos O, Garcia-
SA, Budde RP. 2015. Are novel non-invasive imaging techniques Dorado D, Castell-Conesa J, Almirante B, Aguadé-Bruix S, Tornos P.
needed in patients with suspected prosthetic heart valve endocarditis? 2015. Improving the diagnosis of infective endocarditis in prosthetic
A systematic review and meta-analysis. Eur Radiol 25:2125–2133. valves and intracardiac devices with 18F-fluordeoxyglucose positron
https://doi.org/10.1007/s00330-015-3605-7. emission tomography/computed tomography angiography: initial re-
103. Cahill TJ, Baddour LM, Habib G, Hoen B, Salaun E, Pettersson GB, sults at an infective endocarditis referral center. Circulation 132:
Schafers HJ, Prendergast BD. 2017. Challenges in infective endocar- 1113–1126. https://doi.org/10.1161/CIRCULATIONAHA.115.015316.
ditis. J Am Coll Cardiol 69:325–344. https://doi.org/10.1016/j.jacc 119. Rouzet F, Chequer R, Benali K, Lepage L, Ghodbane W, Duval X, Iung B,
.2016.10.066. Vahanian A, Le Guludec D, Hyafil F. 2014. Respective performance of
104. Daniel WG, Mugge A, Martin RP, Lindert O, Hausmann D, Nonnast-Daniel 18F-FDG PET and radiolabeled leukocyte scintigraphy for the diagnosis
B, Laas J, Lichtlen PR. 1991. Improvement in the diagnosis of abscesses of prosthetic valve endocarditis. J Nucl Med 55:1980 –1985. https://doi
associated with endocarditis by transesophageal echocardiography. N .org/10.2967/jnumed.114.141895.
Engl J Med 324:795– 800. https://doi.org/10.1056/NEJM199103213241203. 120. Chen W, Kim J, Molchanova-Cook OP, Dilsizian V. 2014. The potential of
105. De Castro S, Cartoni D, d’Amati G, Beni S, Yao J, Fiorell M, Gallo P, FDG PET/CT for early diagnosis of cardiac device and prosthetic valve
Fedele F, Pandian NG. 2000. Diagnostic accuracy of transthoracic and infection before morphologic damages ensue. Curr Cardiol Rep 16:459.
multiplane transesophageal echocardiography for valvular perforation https://doi.org/10.1007/s11886-013-0459-y.
in acute infective endocarditis: correlation with anatomic findings. Clin 121. Ricciardi A, Sordillo P, Ceccarelli L, Maffongelli G, Calisti G, Di Pietro B,
Infect Dis 30:825– 826. https://doi.org/10.1086/313762. Caracciolo CR, Schillaci O, Pellegrino A, Chiariello L, Andreoni M, Sarmati L.
106. Hill EE, Herijgers P, Claus P, Vanderschueren S, Peetermans WE, Herre- 2014. 18-Fluoro-2-deoxyglucose positron emission tomography-
gods MC. 2007. Abscess in infective endocarditis: the value of trans- computed tomography: an additional tool in the diagnosis of prosthetic
esophageal echocardiography and outcome: a 5-year study. Am Heart valve endocarditis. Int J Infect Dis 28:219–224. https://doi.org/10.1016/j.ijid
J 154:923–928. https://doi.org/10.1016/j.ahj.2007.06.028. .2014.04.028.
107. Roe MT, Abramson MA, Li J, Heinle SK, Kisslo J, Corey GR, Sexton DJ. 122. Salomaki SP, Saraste A, Kemppainen J, Bax JJ, Knuuti J, Nuutila P,
2000. Clinical information determines the impact of transesophageal Seppanen M, Roivainen A, Airaksinen J, Pirila L, Oksi J, Hohenthal U.
echocardiography on the diagnosis of infective endocarditis by the 2017. 18F-FDG positron emission tomography/computed tomography
Duke criteria. Am Heart J 139:945–951. https://doi.org/10.1067/mhj in infective endocarditis. J Nucl Cardiol 24:195–206. https://doi.org/10
.2000.104762. .1007/s12350-015-0325-y.
108. Lamas CC, Eykyn SJ. 1997. Suggested modifications to the Duke criteria 123. Bartoletti M, Tumietto F, Fasulo G, Giannella M, Cristini F, Bonfiglioli R,
for the clinical diagnosis of native valve and prosthetic valve Raumer L, Nanni C, Sanfilippo S, Di Eusanio M, Scotton PG, Graziosi M,
endocarditis: analysis of 118 pathologically proven cases. Clin Infect Dis Rapezzi C, Fanti S, Viale P. 2014. Combined computed tomography and
25:713–719. fluorodeoxyglucose positron emission tomography in the diagnosis of
109. Pérez-Vázquez A, Fariñas MC, García-Palomo JD, Bernal JM, Revuelta prosthetic valve endocarditis: a case series. BMC Res Notes 7:32. https://
JM, González-Macías J. 2000. Evaluation of the Duke criteria in 93 doi.org/10.1186/1756-0500-7-32.
episodes of prosthetic valve endocarditis: could sensitivity be im- 124. Erba PA, Conti U, Lazzeri E, Sollini M, Doria R, De Tommasi SM, Bandera
proved? Arch Intern Med 160:1185–1191. F, Tascini C, Menichetti F, Dierckx RA, Signore A, Mariani G. 2012. Added
110. Durack DT, Lukes AS, Bright DK. 1994. New criteria for diagnosis of value of 99mTc-HMPAO-labeled leukocyte SPECT/CT in the character-
infective endocarditis: utilization of specific echocardiographic find- ization and management of patients with infectious endocarditis. J
ings. Duke Endocarditis Service. Am J Med 96:200 –209. Nucl Med 53:1235–1243. https://doi.org/10.2967/jnumed.111.099424.
111. Habib G, Derumeaux G, Avierinos JF, Casalta JP, Jamal F, Volot F, Garcia 125. Hyafil F, Rouzet F, Lepage L, Benali K, Raffoul R, Duval X, Hvass U, Iung
M, Lefevre J, Biou F, Maximovitch-Rodaminoff A, Fournier PE, Ambrosi B, Nataf P, Lebtahi R, Vahanian A, Le Guludec D. 2013. Role of radio-
P, Velut JG, Cribier A, Harle JR, Weiller PJ, Raoult D, Luccioni R. 1999. labelled leucocyte scintigraphy in patients with a suspicion of pros-
Value and limitations of the Duke criteria for the diagnosis of infective thetic valve endocarditis and inconclusive echocardiography. Eur Heart
endocarditis. J Am Coll Cardiol 33:2023–2029. https://doi.org/10.1016/ J Cardiovasc Imaging 14:586 –594. https://doi.org/10.1093/ehjci/jet029.
S0735-1097(99)00116-3. 126. Gomes A, Glaudemans AW, Touw DJ, van Melle JP, Willems TP, Maass
112. Thuny F, Gaubert JY, Jacquier A, Tessonnier L, Cammilleri S, Raoult D, AH, Natour E, Prakken NH, Borra RJ, van Geel PP, Slart RH, van Assen S,

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 22


Staphylococcus aureus Prosthetic Valve Endocarditis Clinical Microbiology Reviews

Sinha B. 2017. Diagnostic value of imaging in infective endocarditis: a artificial devices. JAMA 240:751–753. https://doi.org/10.1001/jama
systematic review. Lancet Infect Dis 17:e1– e14. https://doi.org/10 .1978.03290080041021.
.1016/S1473-3099(16)30141-4. 147. Faville RJ, Jr, Zaske DE, Kaplan EL, Crossley K, Sabath LD, Quie PG. 1978.
127. Archer GL, Johnston JL, Vazquez GJ, Haywood HB, III. 1983. Efficacy of Staphylococcus aureus endocarditis. Combined therapy with vancomy-
antibiotic combinations including rifampin against methicillin-resistant cin and rifampin. JAMA 240:1963–1965.
Staphylococcus epidermidis: in vitro and in vivo studies. Rev Infect Dis 148. Massanari RM, Donta ST. 1978. The efficacy of rifampin as adjunctive
5:S538 –S542. https://doi.org/10.1093/clinids/5.Supplement_3.S538. therapy in selected cases of staphylococcal endocarditis. Chest 73:
128. Drinković D, Morris AJ, Pottumarthy S, MacCulloch D, West T. 2003. 371–375.
Bacteriological outcome of combination versus single-agent treatment 149. Tuazon CU, Lin MY, Sheagren JN. 1978. In vitro activity of rifampin
for staphylococcal endocarditis. J Antimicrob Chemother 52:820 – 825. alone and in combination with nafcillin and vancomycin against patho-
https://doi.org/10.1093/jac/dkg440. genic strains of Staphylococcus aureus. Antimicrob Agents Chemother
129. Karchmer AW, Archer GL, Dismukes WE. 1983. Rifampin treatment of 13:759 –761.
prosthetic valve endocarditis due to Staphylococcus epidermidis. Rev 150. Zinner SH, Lagast H, Klastersky J. 1981. Antistaphylococcal activity of
Infect Dis 5:S543–S548. https://doi.org/10.1093/clinids/5.Supplement_3 rifampin with other antibiotics. J Infect Dis 144:365–371.
.S543. 151. Moorman DR, Mandell GL. 1981. Characteristics of rifampin-resistant
130. Kobasa WD, Kaye KL, Shapiro T, Kaye D. 1983. Therapy for experi- variants obtained from clinical isolates of Staphylococcus aureus. Anti-
mental endocarditis due to Staphylococcus epidermidis. Rev Infect microb Agents Chemother 20:709 –713.

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest


Dis 5:S533–S537. 152. Tsuji BT, Rybak MJ. 2005. Short-course gentamicin in combination with
131. Lowy FD, Wexler MA, Steigbigel NH. 1982. Therapy of methicillin- daptomycin or vancomycin against Staphylococcus aureus in an in vitro
resistant Staphylococcus epidermidis experimental endocarditis. J Lab pharmacodynamic model with simulated endocardial vegetations. An-
Clin Med 100:94 –104. timicrob Agents Chemother 49:2735–2745. https://doi.org/10.1128/
132. Vazquez GJ, Archer GL. 1980. Antibiotic therapy of experimental Staph- AAC.49.7.2735-2745.2005.
ylococcus epidermidis endocarditis. Antimicrob Agents Chemother 17: 153. Nicolau DP, Freeman CD, Belliveau PP, Nightingale CH, Ross JW, Quin-
280 –285. tiliani R. 1995. Experience with a once-daily aminoglycoside program
133. Watanakunakorn C. 1984. Mode of action and in-vitro activity of van- administered to 2,184 adult patients. Antimicrob Agents Chemother
comycin. J Antimicrob Chemother 14(Suppl D):7–18. 39:650 – 655.
134. Lexicomp. 2016. Vancomycin. In Lexi-drugs online. Lexicomp, Hudson, 154. Schroeder R, Waldsich C, Wank H. 2000. Modulation of RNA function by
OH. http://online.lexi.com. Accessed 21 March 2018. aminoglycoside antibiotics. EMBO J 19:1–9. https://doi.org/10.1093/
135. Thuny F, Richet H, Casalta JP, Angelakis E, Habib G, Raoult D. 2010. emboj/19.1.1.
Vancomycin treatment of infective endocarditis is linked with recently 155. Hutchin T, Cortopassi G. 1994. Proposed molecular and cellular mech-
acquired obesity. PLoS One 5:e9074. https://doi.org/10.1371/journal anism for aminoglycoside ototoxicity. Antimicrob Agents Chemother
.pone.0009074. 38:2517–2520.
136. Campbell EA, Korzheva N, Mustaev A, Murakami K, Nair S, Goldfarb A, 156. Mingeot-Leclercq MP, Tulkens PM. 1999. Aminoglycosides: nephrotox-
Darst SA. 2001. Structural mechanism for rifampicin inhibition of bac- icity. Antimicrob Agents Chemother 43:1003–1012.
terial RNA polymerase. Cell 104:901–912. 157. Fowler VG, Jr, Boucher HW, Corey GR, Abrutyn E, Karchmer AW, Rupp
137. Grosset J, Leventis S. 1983. Adverse effects of rifampin. Rev Infect Dis ME, Levine DP, Chambers HF, Tally FP, Vigliani GA, Cabell CH, Link AS,
5:S440 –S450. DeMeyer I, Filler SG, Zervos M, Cook P, Parsonnet J, Bernstein JM, Price
138. Chuard C, Herrmann M, Vaudaux P, Waldvogel FA, Lew DP. 1991. CS, Forrest GN, Fatkenheuer G, Gareca M, Rehm SJ, Brodt HR, Tice A,
Successful therapy of experimental chronic foreign-body infection due Cosgrove SE, S. aureus Endocarditis and Bacteremia Study Group. 2006.
to methicillin-resistant Staphylococcus aureus by antimicrobial combi- Daptomycin versus standard therapy for bacteremia and endocarditis
nations. Antimicrob Agents Chemother 35:2611–2616. caused by Staphylococcus aureus. N Engl J Med 355:653– 665. https://
139. Lucet JC, Herrmann M, Rohner P, Auckenthaler R, Waldvogel FA, Lew doi.org/10.1056/NEJMoa053783.
DP. 1990. Treatment of experimental foreign body infection caused by 158. Souli M, Pontikis K, Chryssouli Z, Galani I, Giamarellou H. 2005. Suc-
methicillin-resistant Staphylococcus aureus. Antimicrob Agents Che- cessful treatment of right-sided prosthetic valve endocarditis due to
mother 34:2312–2317. methicillin-resistant teicoplanin-heteroresistant Staphylococcus aureus
140. Rouse MS, Wilcox RM, Henry NK, Steckelberg JM, Wilson WR. 1990. with linezolid. Eur J Clin Microbiol Infect Dis 24:760 –762. https://doi
Ciprofloxacin therapy of experimental endocarditis caused by .org/10.1007/s10096-005-0023-9.
methicillin-resistant Staphylococcus epidermidis. Antimicrob Agents 159. Dhand A, Sakoulas G. 2014. Daptomycin in combination with other
Chemother 34:273–276. antibiotics for the treatment of complicated methicillin-resistant Staph-
141. Miro JM, Garcia-de-la-Maria C, Armero Y, Soy D, Moreno A, del Rio A, ylococcus aureus bacteremia. Clin Ther 36:1303–1316. https://doi.org/
Almela M, Sarasa M, Mestres CA, Gatell JM, Jimenez de Anta MT, Marco 10.1016/j.clinthera.2014.09.005.
F, Hospital Clinic Experimental Endocarditis Study Group. 2009. Addi- 160. Howden BP, Ward PB, Charles PG, Korman TM, Fuller A, du Cros P,
tion of gentamicin or rifampin does not enhance the effectiveness of Grabsch EA, Roberts SA, Robson J, Read K, Bak N, Hurley J, Johnson PD,
daptomycin in treatment of experimental endocarditis due to Morris AJ, Mayall BC, Grayson ML. 2004. Treatment outcomes for
methicillin-resistant Staphylococcus aureus. Antimicrob Agents Che- serious infections caused by methicillin-resistant Staphylococcus aureus
mother 53:4172– 4177. https://doi.org/10.1128/AAC.00051-09. with reduced vancomycin susceptibility. Clin Infect Dis 38:521–528.
142. Garrigos C, Murillo O, Lora-Tamayo J, Verdaguer R, Tubau F, Cabellos C, https://doi.org/10.1086/381202.
Cabo J, Ariza J. 2013. Fosfomycin-daptomycin and other fosfomycin 161. Mohan SS, McDermott BP, Cunha BA. 2005. Methicillin-resistant Staph-
combinations as alternative therapies in experimental foreign-body ylococcus aureus prosthetic aortic valve endocarditis with paravalvular
infection by methicillin-resistant Staphylococcus aureus. Antimicrob abscess treated with daptomycin. Heart Lung 34:69 –71. https://doi
Agents Chemother 57:606 – 610. https://doi.org/10.1128/AAC.01570-12. .org/10.1016/j.hrtlng.2004.07.009.
143. Riedel DJ, Weekes E, Forrest GN. 2008. Addition of rifampin to standard 162. Moore CL, Osaki-Kiyan P, Haque NZ, Perri MB, Donabedian S, Zervos MJ.
therapy for treatment of native valve infective endocarditis caused by 2012. Daptomycin versus vancomycin for bloodstream infections due
Staphylococcus aureus. Antimicrob Agents Chemother 52:2463–2467. to methicillin-resistant Staphylococcus aureus with a high vancomycin
https://doi.org/10.1128/AAC.00300-08. minimum inhibitory concentration: a case-control study. Clin Infect Dis
144. Levine DP, Fromm BS, Reddy BR. 1991. Slow response to vancomycin or 54:51–58. https://doi.org/10.1093/cid/cir764.
vancomycin plus rifampin in methicillin-resistant Staphylococcus aureus 163. Sakoulas G, Moise PA, Casapao AM, Nonejuie P, Olson J, Okumura CY,
endocarditis. Ann Intern Med 115:674 – 680. Rybak MJ, Kullar R, Dhand A, Rose WE, Goff DA, Bressler AM, Lee Y,
145. Simon GL, Smith RH, Sande MA. 1983. Emergence of rifampin-resistant Pogliano J, Johns S, Kaatz GW, Ebright JR, Nizet V. 2014. Antimicrobial
strains of Staphylococcus aureus during combination therapy with van- salvage therapy for persistent staphylococcal bacteremia using dapto-
comycin and rifampin: a report of two cases. Rev Infect Dis mycin plus ceftaroline. Clin Ther 36:1317–1333. https://doi.org/10
5:S507–S508. https://doi.org/10.1093/clinids/5.Supplement_3.S507. .1016/j.clinthera.2014.05.061.
146. Archer GL, Tenenbaum MJ, Haywood HB, III. 1978. Rifampin therapy 164. Cosimi RA, Beik N, Kubiak DW, Johnson JA. 2017. Ceftaroline for severe
of Staphylococcus epidermidis. Use in infections from indwelling methicillin-resistant Staphylococcus aureus infections: a systematic re-

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 23


Galar et al. Clinical Microbiology Reviews

view. Open Forum Infect Dis 4:ofx084. https://doi.org/10.1093/ofid/ 180. Falagas ME, Manta KG, Ntziora F, Vardakas KZ. 2006. Linezolid for the
ofx084. treatment of patients with endocarditis: a systematic review of the
165. Kullar R, Casapao AM, Davis SL, Levine DP, Zhao JJ, Crank CW, Segreti published evidence. J Antimicrob Chemother 58:273–280. https://doi
J, Sakoulas G, Cosgrove SE, Rybak MJ. 2013. A multicentre evaluation of .org/10.1093/jac/dkl219.
the effectiveness and safety of high-dose daptomycin for the treatment 181. Miró JM, Garcia-de-la-Maria C, Armero Y, de-Lazzari E, Soy D, Moreno A,
of infective endocarditis. J Antimicrob Chemother 68:2921–2926. del Rio A, Almela M, Mestres CA, Gatell JM, Jimenez-de-Anta MT, Marco
https://doi.org/10.1093/jac/dkt294. F, Hospital Clinic Experimental Endocarditis Study Group. 2007. Efficacy
166. Dhand A, Bayer AS, Pogliano J, Yang SJ, Bolaris M, Nizet V, Wang G, of telavancin in the treatment of experimental endocarditis due to
Sakoulas G. 2011. Use of antistaphylococcal beta-lactams to increase glycopeptide-intermediate Staphylococcus aureus. Antimicrob Agents
daptomycin activity in eradicating persistent bacteremia due to Chemother 51:2373–2377. https://doi.org/10.1128/AAC.01266-06.
methicillin-resistant Staphylococcus aureus: role of enhanced daptomycin 182. Abdelhady W, Bayer AS, Gonzales R, Li L, Xiong YQ. 2017. Telavancin is
binding. Clin Infect Dis 53:158–163. https://doi.org/10.1093/cid/cir340. active against experimental aortic valve endocarditis caused by
167. Miro JM, Entenza JM, Del Rio A, Velasco M, Castaneda X, Garcia de la Maria daptomycin- and methicillin-resistant Staphylococcus aureus strains.
C, Giddey M, Armero Y, Pericas JM, Cervera C, Mestres CA, Almela M, Falces Antimicrob Agents Chemother 61:e01877-16. https://doi.org/10.1128/
C, Marco F, Moreillon P, Moreno A, Hospital Clinic Experimental Endocar- AAC.01877-16.
ditis Study Group. 2012. High-dose daptomycin plus fosfomycin is safe and 183. Madrigal AG, Basuino L, Chambers HF. 2005. Efficacy of telavancin in a
effective in treating methicillin-susceptible and methicillin-resistant Staph- rabbit model of aortic valve endocarditis due to methicillin-resistant

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest


ylococcus aureus endocarditis. Antimicrob Agents Chemother 56: Staphylococcus aureus or vancomycin-intermediate Staphylococcus au-
4511–4515. https://doi.org/10.1128/AAC.06449-11. reus. Antimicrob Agents Chemother 49:3163–3165. https://doi.org/10
168. Pagani L, Bonnin P, Janssen C, Desjoyaux E, Vitrat V, Bru JP. 2014. .1128/AAC.49.8.3163-3165.2005.
Ceftaroline for the treatment of prosthetic valve endocarditis due to 184. Xiong YQ, Abdelhady W, Tang C, Bayer AS. 2016. Comparative efficacy
methicillin-resistant Staphylococcus aureus. J Heart Valve Dis 23: of telavancin and daptomycin in experimental endocarditis due to
219 –221. multi-clonotype MRSA strains. J Antimicrob Chemother 71:2890 –2894.
169. Cunha BA, Gran A. 2015. Successful treatment of meticillin-resistant Staph- https://doi.org/10.1093/jac/dkw249.
ylococcus aureus (MRSA) aortic prosthetic valve endocarditis with pro- 185. Nace H, Lorber B. 2010. Successful treatment of methicillin-resistant
longed high-dose daptomycin plus ceftaroline therapy. Int J Antimicrob Staphylococcus aureus endocarditis with telavancin. J Antimicrob Che-
Agents 46:225–226. https://doi.org/10.1016/j.ijantimicag.2015.04.006. mother 65:1315–1316. https://doi.org/10.1093/jac/dkq113.
170. Jongsma K, Joson J, Heidari A. 2013. Ceftaroline in the treatment of 186. Sander A, Beiderlinden M, Schmid EN, Peters J. 2002. Clinical experi-
concomitant methicillin-resistant and daptomycin-non-susceptible Staph- ence with quinupristin-dalfopristin as rescue treatment of critically ill
ylococcus aureus infective endocarditis and osteomyelitis: case report. J patients infected with methicillin-resistant staphylococci. Intensive
Antimicrob Chemother 68:1444–1445. https://doi.org/10.1093/jac/dkt009. Care Med 28:1157–1160. https://doi.org/10.1007/s00134-002-1358-7.
171. Jacqueline C, Amador G, Batard E, Le Mabecque V, Miegeville AF, Biek D, 187. Brade KD, Rybak JM, Rybak MJ. 2016. Oritavancin: a new lipoglycopep-
Caillon J, Potel G. 2011. Comparison of ceftaroline fosamil, daptomycin and tide antibiotic in the treatment of Gram-positive infections. Infect Dis
tigecycline in an experimental rabbit endocarditis model caused by Ther 5:1–15. https://doi.org/10.1007/s40121-016-0103-4.
methicillin-susceptible, methicillin-resistant and glycopeptide- 188. Johnson JA, Feeney ER, Kubiak DW, Corey GR. 2015. Prolonged use of
intermediate Staphylococcus aureus. J Antimicrob Chemother 66:863–866. oritavancin for vancomycin-resistant Enterococcus faecium prosthetic
https://doi.org/10.1093/jac/dkr019. valve endocarditis. Open Forum Infect Dis 2:ofv156. https://doi.org/10
172. Ho TT, Cadena J, Childs LM, Gonzalez-Velez M, Lewis JS, II. 2012. .1093/ofid/ofv156.
Methicillin-resistant Staphylococcus aureus bacteraemia and endocar- 189. Kaatz GW, Seo SM, Aeschlimann JR, Houlihan HH, Mercier RC, Rybak MJ.
ditis treated with ceftaroline salvage therapy. J Antimicrob Chemother 1998. Efficacy of LY333328 against experimental methicillin-resistant
67:1267–1270. https://doi.org/10.1093/jac/dks006. Staphylococcus aureus endocarditis. Antimicrob Agents Chemother 42:
173. Kullar R, Sakoulas G, Deresinski S, van Hal SJ. 2016. When sepsis 981–983.
persists: a review of MRSA bacteraemia salvage therapy. J Antimicrob 190. Stewart CL, Turner MS, Frens JJ, Snider CB, Smith JR. 2017. Real-world
Chemother 71:576 –586. https://doi.org/10.1093/jac/dkv368. experience with oritavancin therapy in invasive Gram-positive infections.
174. Lin JC, Fierer J, Aung G, Johns S, Thomas A, Jahng M, Johns S, Fierer J. Infect Dis Ther 6:277–289. https://doi.org/10.1007/s40121-017-0156-z.
2013. The use of ceftaroline fosamil in methicillin-resistant Staphylo- 191. Fleischauer AT, Ruhl L, Rhea S, Barnes E. 2017. Hospitalizations for endo-
coccus aureus endocarditis and deep-seated MRSA infections: a retro- carditis and associated health care costs among persons with diagnosed
spective case series of 10 patients. J Infect Chemother 19:42– 49. drug dependence—North Carolina, 2010-2015. MMWR Morb Mortal Wkly
https://doi.org/10.1007/s10156-012-0449-9. Rep 66:569–573. https://doi.org/10.15585/mmwr.mm6622a1.
175. Paladino JA, Jacobs DM, Shields RK, Taylor J, Bader J, Adelman MH, 192. Lagisetty P, Bohnert A. 2017. Web exclusives. Annals for hospitalists
Wilton GJ, Crane JK, Schentag JJ. 2014. Use of ceftaroline after glyco- inpatient notes. The opioid epidemic—what’s a hospitalist to do? Ann
peptide failure to eradicate meticillin-resistant Staphylococcus aureus Intern Med 167:HO2–HO3. https://doi.org/10.7326/M17-1564.
bacteraemia with elevated vancomycin minimum inhibitory concen- 193. Drew RH. 2007. Emerging options for treatment of invasive, multidrug-
trations. Int J Antimicrob Agents 44:557–563. https://doi.org/10.1016/j resistant Staphylococcus aureus infections. Pharmacotherapy 27:
.ijantimicag.2014.07.024. 227–249. https://doi.org/10.1592/phco.27.2.227.
176. Polenakovik HM, Pleiman CM. 2013. Ceftaroline for meticillin-resistant 194. Paterson DL. 2006. Clinical experience with recently approved antibi-
Staphylococcus aureus bacteraemia: case series and review of the liter- otics. Curr Opin Pharmacol 6:486 – 490. https://doi.org/10.1016/j.coph
ature. Int J Antimicrob Agents 42:450 – 455. https://doi.org/10.1016/j .2006.07.001.
.ijantimicag.2013.07.005. 195. Cosgrove SE, Fowler VG, Jr. 2007. Optimizing therapy for methicillin-
177. Tattevin P, Boutoille D, Vitrat V, Van Grunderbeeck N, Revest M, Dupont resistant Staphylococcus aureus bacteremia. Semin Respir Crit Care Med
M, Alfandari S, Stahl JP. 2014. Salvage treatment of methicillin-resistant 28:624 – 631. https://doi.org/10.1055/s-2007-996409.
staphylococcal endocarditis with ceftaroline: a multicentre observa- 196. VanEperen AS, Segreti J. 2016. Empirical therapy in methicillin-resistant
tional study. J Antimicrob Chemother 69:2010 –2013. https://doi.org/ Staphylococcus aureus infections: an up-to-date approach. J Infect Che-
10.1093/jac/dku085. mother 22:351–359. https://doi.org/10.1016/j.jiac.2016.02.012.
178. Casapao AM, Davis SL, Barr VO, Klinker KP, Goff DA, Barber KE, Kaye KS, 197. Vahanian A, Alfieri O, Andreotti F, Antunes MJ, Baron-Esquivias G,
Mynatt RP, Molloy LM, Pogue JM, Rybak MJ. 2014. Large retrospective Baumgartner H, Borger MA, Carrel TP, De Bonis M, Evangelista A, Falk
evaluation of the effectiveness and safety of ceftaroline fosamil ther- V, Lung B, Lancellotti P, Pierard L, Price S, Schafers HJ, Schuler G,
apy. Antimicrob Agents Chemother 58:2541–2546. https://doi.org/10 Stepinska J, Swedberg K, Takkenberg J, Von Oppell UO, Windecker S,
.1128/AAC.02371-13. Zamorano JL, Zembala M, ESC Committee for Practice Guidelines, Joint
179. Fabre V, Ferrada M, Buckel WR, Avdic E, Cosgrove SE. 2014. Ceftaroline Task Force on the Management of Valvular Heart Disease of the
in combination with trimethoprim-sulfamethoxazole for salvage ther- European Society of Cardiology, European Association for Cardio-
apy of methicillin-resistant Staphylococcus aureus bacteremia and en- Thoracic Surgery. 2012. Guidelines on the management of valvular
docarditis. Open Forum Infect Dis 1:ofu046. https://doi.org/10.1093/ heart disease (version 2012): the Joint Task Force on the Management
ofid/ofu046. of Valvular Heart Disease of the European Society of Cardiology (ESC)

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 24


Staphylococcus aureus Prosthetic Valve Endocarditis Clinical Microbiology Reviews

and the European Association for Cardio-Thoracic Surgery (EACTS). Eur 213. Horstkotte D, Weist K, Ruden H. 1998. Better understanding of the
J Cardiothorac Surg 42:S1–S44. https://doi.org/10.1093/ejcts/ezs455. pathogenesis of prosthetic valve endocarditis—recent perspectives for
198. Lancellotti P, Rosenhek R, Pibarot P, Iung B, Otto CM, Tornos P, Donal prevention strategies. J Heart Valve Dis 7:313–315.
E, Prendergast B, Magne J, La Canna G, Pierard LA, Maurer G. 2013. ESC 214. Chirillo F, Scotton P, Rocco F, Rigoli R, Borsatto F, Pedrocco A, De Leo
Working Group on Valvular Heart Disease position paper— heart valve A, Minniti G, Polesel E, Olivari Z. 2013. Impact of a multidisciplinary
clinics: organization, structure, and experiences. Eur Heart J 34: management strategy on the outcome of patients with native valve
1597–1606. https://doi.org/10.1093/eurheartj/ehs443. infective endocarditis. Am J Cardiol 112:1171–1176. https://doi.org/10
199. Thuny F, Grisoli D, Collart F, Habib G, Raoult D. 2012. Management of .1016/j.amjcard.2013.05.060.
infective endocarditis: challenges and perspectives. Lancet 379: 215. Béraud G, Le Moal G, Elsendoorn A, Tattevin P, Godet C, Alfandari S,
965–975. https://doi.org/10.1016/S0140-6736(11)60755-1. Couet W, Roblot P, Roblot F. 2012. A survey on the use of gentamicin
200. Botelho-Nevers E, Thuny F, Casalta JP, Richet H, Gouriet F, Collart F, Riberi in infective endocarditis. Eur J Clin Microbiol Infect Dis 31:1413–1418.
A, Habib G, Raoult D. 2009. Dramatic reduction in infective endocarditis- https://doi.org/10.1007/s10096-011-1458-9.
related mortality with a management-based approach. Arch Intern Med 216. Bille J. 1995. Medical treatment of staphylococcal infective endo-
169:1290–1298. https://doi.org/10.1001/archinternmed.2009.192. carditis. Eur Heart J 16:80 – 83. https://doi.org/10.1093/eurheartj/16
201. Bonow RO, Carabello BA, Chatterjee K, de Leon AC, Jr, Faxon DP, Freed .suppl_B.80.
MD, Gaasch WH, Lytle BW, Nishimura RA, O’Gara PT, O’Rourke RA, Otto 217. Cervera C, Castaneda X, de la Maria CG, del Rio A, Moreno A, Soy D,
CM, Shah PM, Shanewise JS, American College of Cardiology/American Pericas JM, Falces C, Armero Y, Almela M, Ninot S, Pare JC, Mestres CA,
Heart Association Task Force on Practice Guidelines. 2008. 2008 fo-

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest


Gatell JM, Marco F, Miro JM, Hospital Clinic Endocarditis Study Group.
cused update incorporated into the ACC/AHA 2006 guidelines for the 2014. Effect of vancomycin minimal inhibitory concentration on the
management of patients with valvular heart disease: a report of the outcome of methicillin-susceptible Staphylococcus aureus endocarditis.
American College of Cardiology/American Heart Association Task Force Clin Infect Dis 58:1668 –1675. https://doi.org/10.1093/cid/ciu183.
on Practice Guidelines (Writing Committee to revise the 1998 guide- 218. Cosgrove SE, Vigliani GA, Campion M, Fowler VG, Jr, Abrutyn E, Corey
lines for the management of patients with valvular heart disease). GR, Levine DP, Rupp ME, Chambers HF, Karchmer AW, Boucher HW.
Endorsed by the Society of Cardiovascular Anesthesiologists, Society 2009. Initial low-dose gentamicin for Staphylococcus aureus bacteremia
for Cardiovascular Angiography and Interventions, and Society of Tho- and endocarditis is nephrotoxic. Clin Infect Dis 48:713–721. https://doi
racic Surgeons. J Am Coll Cardiol 52:e1– e142. https://doi.org/10.1016/ .org/10.1086/597031.
j.jacc.2008.05.007. 219. de Feiter PW, Jacobs JA, Jacobs MJHM, Vink C, van der Geest S. 2005.
202. Gordon SM, Serkey JM, Longworth DL, Lytle BW, Cosgrove DM, III. 2000. Successful treatment of Staphylococcus epidermidis prosthetic valve
Early onset prosthetic valve endocarditis: the Cleveland Clinic experi-
endocarditis with linezolid after failure of treatment with oxacillin,
ence 1992-1997. Ann Thorac Surg 69:1388 –1392. https://doi.org/10
gentamicin, rifampicin, vancomycin, and fusidic acid regimens. Scand J
.1016/S0003-4975(00)01135-8.
Infect Dis 37:173–176. https://doi.org/10.1080/00365540410021018.
203. Truninger K, Attenhofer Jost CH, Seifert B, Vogt PR, Follath F, Schaffner
220. Del Río A, Gasch O, Moreno A, Peña C, Cuquet J, Soy D, Mestres CA,
A, Jenni R. 1999. Long term follow up of prosthetic valve endocarditis:
Suárez C, Pare JC, Tubau F, De La Mària CG, Marco F, Carratalà J, Gatell
what characteristics identify patients who were treated successfully
JM, Gudiol F, Miró JM, Pericas JM, Cervera C, Armero Y, Almela M, Fuster
with antibiotics alone? Heart 82:714 –720.
D, Cartañá R, Ninot S, Azqueta M, Sitges M, Heras M, Pomar JL, Ramírez
204. Lalani T, Chu VH, Park LP, Cecchi E, Corey GR, Durante-Mangoni E, Fowler
J, Brunet M, Llopis J, Pujol M, Ariza J, Martí C, Mijana M. 2014. Efficacy
VG, Jr, Gordon D, Grossi P, Hannan M, Hoen B, Munoz P, Rizk H, Kanj SS,
and safety of fosfomycin plus imipenem as rescue therapy for compli-
Selton-Suty C, Sexton DJ, Spelman D, Ravasio V, Tripodi MF, Wang A,
cated bacteremia and endocarditis due to methicillin-resistant Staph-
International Collaboration on Endocarditis-Prospective Cohort Study In-
ylococcus aureus: a multicenter clinical trial. Clin Infect Dis 59:
vestigators. 2013. In-hospital and 1-year mortality in patients undergoing
1105–1112. https://doi.org/10.1093/cid/ciu580.
early surgery for prosthetic valve endocarditis. JAMA Intern Med 173:
1495–1504. https://doi.org/10.1001/jamainternmed.2013.8203. 221. Hasbun R, Vikram HR, Barakat LA, Buenconsejo J, Quagliarello VJ. 2003.
205. Akowuah EF, Davies W, Oliver S, Stephens J, Riaz I, Zadik P, Cooper G. Complicated left-sided native valve endocarditis in adults: risk classifi-
2003. Prosthetic valve endocarditis: early and late outcome following cation for mortality. JAMA 289:1933–1940. https://doi.org/10.1001/
medical or surgical treatment. Heart 89:269 –272. https://doi.org/10 jama.289.15.1933.
.1136/heart.89.3.269. 222. Holland TL, Arnold C, Fowler VG, Jr. 2014. Clinical management of
206. Lockhart PB, Brennan MT, Sasser HC, Fox PC, Paster BJ, Bahrani- Staphylococcus aureus bacteremia: a review. JAMA 312:1330 –1341.
Mougeot FK. 2008. Bacteremia associated with toothbrushing and https://doi.org/10.1001/jama.2014.9743.
dental extraction. Circulation 117:3118 –3125. https://doi.org/10.1161/ 223. Kang DH, Kim YJ, Kim SH, Sun BJ, Kim DH, Yun SC, Song JM, Choo SJ,
CIRCULATIONAHA.107.758524. Chung CH, Song JK, Lee JW, Sohn DW. 2012. Early surgery versus
207. Desimone DC, Tleyjeh IM, Correa de Sa DD, Anavekar NS, Lahr BD, Sohail conventional treatment for infective endocarditis. N Engl J Med 366:
MR, Steckelberg JM, Wilson WR, Baddour LM, Mayo Cardiovascular Infec- 2466 –2473. https://doi.org/10.1056/NEJMoa1112843.
tions Study Group. 2012. Incidence of infective endocarditis caused by 224. Karchmer AW. 1991. Prosthetic valve endocarditis: a continuing chal-
viridans group streptococci before and after publication of the 2007 lenge for infection control. J Hosp Infect 18:355–366. https://doi.org/
American Heart Association’s endocarditis prevention guidelines. Circula- 10.1016/0195-6701(91)90042-7.
tion 126:60–64. https://doi.org/10.1161/CIRCULATIONAHA.112.095281. 225. Le T, Bayer AS. 2003. Combination antibiotic therapy for infective
208. Pasquali SK, He X, Mohamad Z, McCrindle BW, Newburger JW, Li JS, endocarditis. Clin Infect Dis 36:615– 621. https://doi.org/10.1086/
Shah SS. 2012. Trends in endocarditis hospitalizations at US children’s 367661.
hospitals: impact of the 2007 American Heart Association antibiotic 226. Mayer KH, Schoenbaum SC. 1982. Evaluation and management of
prophylaxis guidelines. Am Heart J 163:894 – 899. https://doi.org/10 prosthetic valve endocarditis. Prog Cardiovasc Dis 25:43–54.
.1016/j.ahj.2012.03.002. 227. Wareham DW, Abbas H, Karcher AM, Das SS. 2006. Treatment of
209. Pant S, Patel NJ, Deshmukh A, Golwala H, Patel N, Badheka A, Hirsch GA, prosthetic valve infective endocarditis due to multi-resistant Gram-
Mehta JL. 2015. Trends in infective endocarditis incidence, microbiology, positive bacteria with linezolid. J Infect 52:300 –304. https://doi.org/10
and valve replacement in the United States from 2000 to 2011. J Am Coll .1016/j.jinf.2005.05.022.
Cardiol 65:2070–2076. https://doi.org/10.1016/j.jacc.2015.03.518. 228. Wilson WR, Karchmer AW, Dajani AS, Taubert KA, Bayer A, Kaye D, Bisno
210. Bayer AS, Nelson RJ, Slama TG. 1990. Current concepts in prevention of AL, Ferrieri P, Shulman ST, Durack DT. 1995. Antibiotic treatment of
prosthetic valve endocarditis. Chest 97:1203–1207. adults with infective endocarditis due to streptococci, enterococci,
211. Freeman R. 1995. Prevention of prosthetic valve endocarditis. J Hosp staphylococci, and HACEK microorganisms. JAMA 274:1706 –1713.
Infect 30:44 –53. 229. Yaw LK, Robinson JO, Ho KM. 2014. A comparison of long-term out-
212. Khor E, Tay LF, Goh KS, Lee CN. 1996. Prevention of prosthetic valve comes after meticillin-resistant and meticillin-sensitive Staphylococcus
endocarditis by impregnation of gentamicin into surgical pledgets. aureus bacteraemia: an observational cohort study. Lancet Infect Dis
Biomaterials 17:1631–1637. 14:967–975. https://doi.org/10.1016/S1473-3099(14)70876-X.

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 25


Galar et al. Clinical Microbiology Reviews

Alicia Galar, Pharm.D., Ph.D., studied Phar- Patricia Muñoz, M.D., Ph.D., studied Medi-
macy at the University of Navarra and did cine at the Complutense University of Ma-
both her residency in Clinical Microbiology drid (UCM) and trained at the Hospital
and Parasitology and Ph.D. at the Clínica Clínico San Carlos. She was deputy editor of
Universidad de Navarra. She was a Postdoc- Clinical Microbiology and Infection and was
toral Clinical Research Fellow/Specialist at awarded the Young Investigator Award of
Brigham and Women’s Hospital and Harvard the European Society of Clinical Microbiol-
Medical School. Dr. Galar worked at the ogy and Infectious Diseases. She is currently
World Health Organization (WHO) Collabo- Professor in Medicine in Clinical Microbiol-
rating Centre for Surveillance of Antimicro- ogy at UCM and Head of the Section of
bial Resistance and joined the Transplant Clinical Microbiology and Infectious Diseases
and Oncology Team at the Infectious Diseases Division of Brigham and at the Hospital General Universitario Gregorio Marañón (HGUGM). Dr.
Women’s Hospital/Dana-Farber Cancer Institute/Massachusetts General Muñoz’s research interests include fungal infections; infective endocar-
Hospital. She completed her education with several courses at the ditis; infections in solid-organ transplant recipients, immunocompro-
Harvard School of Public Health, Massachusetts General Hospital, and mised hosts, and heart surgery patients; and nosocomially acquired
Harvard University. She is currently working at the Department of infectious diseases. She is an active member of the European Study

Downloaded from http://cmr.asm.org/ on May 6, 2020 by guest


Clinical Microbiology and Infectious Diseases of the Hospital General Groups for Nosocomial Infections and Infection in Compromised Host
Universitario Gregorio Marañón in Madrid. Dr. Galar’s main research and the Spanish Network of Infection in Transplantation. She is the
interests include antimicrobial resistance, therapeutic drug monitoring, Secretary of the Group for the Management of Infective Endocarditis at
pharmacokinetic/pharmacodynamic (PK/PD) strategies, antimicrobial HGUGM and has been President of the Spanish Society for Cardiovas-
stewardship, vaccines, prosthetic-device-related infections, infective en- cular Infections.
docarditis, and infections in patients with heart diseases.

Mark J. Siedner is an Associate Professor of


Ana A. Weil holds an M.P.H. from the Johns Medicine at Harvard Medical School and
Hopkins School of Public Health and an M.D. holds an M.D. from the Johns Hopkins Uni-
at Tufts University. She trained in internal versity School of Medicine and an M.P.H.
medicine and infectious diseases at Massa- from the Johns Hopkins Bloomberg School
chusetts General Hospital (MGH), where she of Public Health. He trained in internal med-
also served as a chief resident. Dr. Weil is icine at Columbia University Medical Center
currently a physician-scientist in the Infec- and in infectious diseases at Massachusetts
tious Diseases Division at MGH. The Weil General Hospital (MGH). His clinical work fo-
laboratory is focused on understanding the cuses on clinical infectious disease both at
influence of the gut microbiome in suscep- MGH and as an HIV care provider in south-
tibility to enteric infections and pathogen- western Uganda and Kwazulu-Natal, South Africa. He leads a research
gut microbe interactions at the mucosal surface, including identifying program in Uganda and South Africa in partnership with MGH and the
gut species that may protect against infection and relationships be- Africa Health Research Institute, aimed at mitigating the causes of
tween gut microbial species and mucosal immune responses. Dr. Weil morbidity and mortality among people living with HIV in low-income
also treats general medicine and infectious disease patients at MGH and countries.
has held several leadership positions in medical education, including as
a faculty advisor for an elective course at Harvard Medical School.

David M. Dudzinski studied Medicine at


Harvard Medical School and trained at Mas-
sachusetts General Hospital, where he was a
chief resident. He is board certified in inter-
nal medicine, cardiovascular diseases, nu-
clear cardiology, adult comprehensive echo-
cardiography, and critical care medicine. Dr.
Dudzinski is a cardiologist, cardiac intensiv-
ist, and echocardiographer at Harvard Med-
ical School and Massachusetts General Hos-
pital and serves as a staff cardiac intensivist
in the cardiac and cardiac surgical intensive care units, with a clinical
expertise centered on the nascent field of critical care cardiology. Dr.
Dudzinski’s academic interests are in pulmonary embolism, right ven-
tricle function, procedural echocardiography, aortic disease, quality
improvement, medical education, resuscitation science and emergency
cardiovascular care, and critical care cardiology. Dr. Dudzinski is also an
attorney with expertise in the scientific and legal aspects of the regu-
lation of protein-based therapeutics, and he serves on the American
College of Cardiology’s national medical professional liability working
group.

April 2019 Volume 32 Issue 2 e00041-18 cmr.asm.org 26

You might also like