You are on page 1of 12

Drug Resistance Updates 17 (2014) 1–12

Contents lists available at ScienceDirect

Drug Resistance Updates


journal homepage: www.elsevier.com/locate/drup

Review

Linezolid update: Stable in vitro activity following more than a decade


of clinical use and summary of associated resistance mechanisms
Rodrigo E. Mendes a,∗ , Lalitagauri M. Deshpande a , Ronald N. Jones a,b
a
JMI Laboratories, North Liberty, IA 52317, USA
b
Tufts University School of Medicine, Boston, MA 02111, USA

a r t i c l e i n f o a b s t r a c t

Keywords: Linezolid, approved for clinical use since 2000, has become an important addition to the anti-Gram-
Linezolid positive infection armamentarium. This oxazolidinone drug has in vitro and in vivo activity against
Surveillance essentially all Gram-positive organisms, including methicillin-resistant Staphylococcus aureus (MRSA)
Oxazolidinones
and vancomycin-resistant enterococci (VRE). The in vitro activity of linezolid was well documented
Resistance
prior to its clinical application, and several ongoing surveillance studies demonstrated consistent and
LEADER
ZAAPS potent results during the subsequent years of clinical use. Emergence of resistance has been lim-
ited and associated with invasive procedures, deep organ involvement, presence of foreign material
and mainly prolonged therapy. Non-susceptible organisms usually demonstrate alterations in the 23S
rRNA target, which remain the main resistance mechanism observed in enterococci; although a few
reports have described the detection of cfr-mediated resistance in Enterococcus faecalis. S. aureus isolates
non-susceptible to linezolid remain rare in large surveillance studies. Most isolates harbour 23S rRNA
mutations; however, cfr-carrying MRSA isolates have been observed in the United States and elsewhere.
It is still uncertain whether the occurrences of such isolates are becoming more prevalent. Coagulase-
negative isolates (CoNS) resistant to linezolid were uncommon following clinical approval. Surveillance
data have indicated that CoNS isolates, mainly Staphylococcus epidermidis, currently account for the major-
ity of Gram-positive organisms displaying elevated MIC results to linezolid. In addition, these isolates
frequently demonstrate complex and numerous resistance mechanisms, such as alterations in the ribo-
somal proteins L3 and/or L4 and/or presence of cfr and/or modifications in 23S rRNA. The knowledge
acquired during the past decades on this initially used oxazolidinone has been utilized for developing
new candidate agents, such as tedizolid and radezolid, and as linezolid patents soon begin to expire,
generic brands will certainly become available. These events will likely establish a new chapter for this
successful class of antimicrobial agents.
© 2014 Elsevier Ltd. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Antimicrobial spectrum and activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.1. Pre-FDA approval . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.2. Post-FDA approval . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
3. Resistance development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
4. Resistance mechanisms . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
4.1. Mutations in 23S rRNA and ribosomal proteins L3 and L4 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
4.2. Acquired resistance mechanism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
4.3. Resistance mechanisms other than target site modifications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
5. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
Acknowledgements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9

∗ Corresponding author at: JMI Laboratories, 345 Beaver Kreek Centre, Suite A, North Liberty, IA 52317, USA. Tel.: +1 319 665 3370; fax: +1 319 655 3371.
E-mail address: rodrigo-mendes@jmilabs.com (R.E. Mendes).

http://dx.doi.org/10.1016/j.drup.2014.04.002
1368-7646/© 2014 Elsevier Ltd. All rights reserved.
2 R.E. Mendes et al. / Drug Resistance Updates 17 (2014) 1–12

1. Introduction -resistant), enterococci (vancomycin-susceptible and -resistant),


streptococci, Corynebacterium spp., Mycobacterium tuberculosis and
During the last decades, only a few new antimicrobial agents some species of anaerobic bacteria (Zurenko et al., 1996).
have been approved for clinical use by the United States (USA) Table 1 summarizes the MIC50 and MIC90 values for line-
Food and Drug Administration (FDA), European Medicines Agency zolid when tested against Gram-positive clinical isolates obtained
(EMA) and other regulatory agencies (Gould and Bal, 2013). Among from studies performed prior to the FDA approval. Overall, these
those agents with Gram-positive organism coverage, ceftaroline studies reported linezolid MIC results between 0.25 and 4 mg/L,
and telavancin are improved analogues of known scaffolds, while and non-susceptible results were only initially obtained against
daptomycin and linezolid are its sole representative molecules laboratory-derived mutants after numerous daily passaging exper-
of entirely new classes of antimicrobials (i.e. lipopeptides and iments in drug containing media. During the pre-FDA approval era,
oxazolidinones, respectively). Linezolid received a priority review linezolid demonstrated a normal MIC distribution (i.e. Gauss curve)
by the FDA, which approved this drug for clinical use in March when tested against clinical Gram-positive pathogens, which was
of 2000, and linezolid has become an important addition to the confirmed by several local and multicentre in vitro studies con-
anti-Gram-positive infection armamentarium. Indications consist ducted by investigators on several continents (Zurenko et al.,
of treatment for uncomplicated and complicated skin and skin 1996; Jones et al., 1996; Wise et al., 1998; von Eiff and Peters,
structure infections (cSSSI) and hospital- and community-acquired 1999; Noskin et al., 1999; Johnson et al., 2000; Rybak et al., 1998,
pneumonia caused by Gram-positive pathogens (Zyvox, 2010). 2000).
Linezolid is also indicated for the treatment of vancomycin- Overall, studies evaluating the in vitro activity and spectrum
resistant Enterococcus faecium (VRE) infections (including cases of linezolid have reported MIC50 values of 2–4, 2–4 and 1–2 mg/L
with concurrent bacteremia). when tested against staphylococci, enterococci and streptococci,
Since the ribosome represents the core for protein synthesis in respectively. However, Wise et al. (1998) published linezolid MIC
all living cells, numerous antimicrobial agents targeting the pep- results when tested against staphylococci and enterococci lower
tidyltransferase centre (PTC) of the large ribosomal subunit have than other authors, with highest linezolid values at 1 mg/L (see
been developed. Targeting the PTC remains one of the main advan- Table 1). In addition, Jones et al. (1996) published linezolid MIC50
tages of the oxazolidinones due to the number of rRNA genes, which values (1 mg/L) against enterococci similar to those reported by
minimizes the emergence of resistance (Toh et al., 2007). Oxazolidi- Wise et al. (1998), which were, in general, lower than those from
nones were initially known to affect protein synthesis during the other publications.
initiation phase of translation (Lin et al., 1997; Shinabarger et al., The variations observed in the linezolid MIC results in these
1997). A later study offered additional insights and demonstrated studies can, perhaps, be explained by the different guidelines
that linezolid interacts with the 23S rRNA (A2602; Escherichia coli (EUCAST versus CLSI) and methods (broth microdilution and agar
numbering used throughout) and prevents binding or proper place- dilution versus Etest). Also, some studies utilized agar dilution
ment of aminoacyl-tRNA in the PTC site (Leach et al., 2007). techniques and IsoSensititre media (Wise et al., 1998; von Eiff
However, linezolid also seems to interact with ribosomal protein and Peters, 1999; Johnson et al., 2000), while other investigators
L27, ribosomal-associated protein LepA and tRNA (Colca et al., made use of broth microdilution methods and the Mueller-Hinton
2003). These interactions are still unknown, but could be associated medium (Zurenko et al., 1996; Jones et al., 1996; Noskin et al.,
with ribosome formation and fidelity of translation (Colca et al., 1999; Rybak et al., 1998, 2000). Regardless, the different guidelines,
2003). methods and media utilized only indicate the presence of several
Linezolid is available in intravenous and oral formulations, uncontrolled variables among early studies and imply methodolog-
which have provided this agent as an attractive alternative for ical differences, and there was no correlation of the method applied
treating numerous infection types, including respiratory tract with the MIC results obtained. The MIC endpoint reading itself
infection caused by methicillin-resistant Staphylococcus aureus can be another potential element causing variability. Trailing is a
(MRSA) and other serious multidrug-resistant (MDR) infections due common phenomenon when testing linezolid, requiring additional
to vancomycin-resistant enterococci (VRE). The clinical and com- expertise for determining the correct endpoint value as currently
mercial success of linezolid has prompted many pharmaceutical stated in some international standardized methods (Biedenbach
companies to investigate and develop oxazolidinone-like com- and Jones, 1997, 2001, 2003; Poppe et al., 2006; Worth et al., 1996;
pounds (Michalska et al., 2013). Several molecules have been devel- Tenover et al., 2007).
oped as candidates, but only tedizolid and radezolid have advanced These technical difficulties along with the lack of reading stan-
into clinical trials (Shaw and Barbachyn, 2011). This review pro- dards when testing such drug for susceptibility provide additional
vides a summary update for linezolid with regard to the pre- challenges for clinical microbiologists and research personnel. Dur-
and post-FDA approval study results related to in vitro antimicro- ing the January 2006 Clinical and Laboratory Standards Institute
bial activity and spectrum, and development and dissemination of (CLSI) meeting (Miami, FL), the Staphylococci Working Group pro-
resistance mechanisms. A thorough review on the activity in vitro of posed additional language in the M02, M07 and M100 documents to
other oxazolidinones tested against Gram-positive isolates, includ- provide information detail on how to read MIC results and zones of
ing linezolid-resistant strains, was recently published and this topic inhibition in the presence of so-called “trailing endpoints” (Clinical
will not be addressed this topic (Shaw and Barbachyn, 2011). and Laboratory Standards Institute, 2006). These changes were
proposed to contain the following recommendation: “For some
antimicrobial agents (such as for chloramphenicol, clindamycin,
2. Antimicrobial spectrum and activity erythromycin, linezolid and tetracycline), trailing growth can make
endpoint determination difficult. In such cases, the MIC should be
2.1. Pre-FDA approval read at the first well that shows a prominent reduction in growth.
Tiny buttons of growth should be ignored”. However, these pro-
Investigational studies performed during the development of posed languages have not been incorporated into the M02, M07
linezolid documented a broad and potent antimicrobial activ- or M100 documents by the time this review article was written.
ity against Gram-positive organisms. Linezolid displayed in vitro According to the summary minutes of the 2012 CLSI meeting, this
inhibitory activities against numerous clinically relevant bacte- recommendation will be published in the M07-10 in 2015 (Clinical
rial species, including staphylococci (methicillin-susceptible and and Laboratory Standards Institute, 2012a, 2014).
R.E. Mendes et al. / Drug Resistance Updates 17 (2014) 1–12 3

Summary of linezolid MIC50 and MIC90 values when tested against Gram-positive isolates obtained from investigational studies published prior to FDA approval and those obtained from the most recently reported post-marketing
2.2. Post-FDA approval

Mendes et al.
(2014b)

0.5/1
The activity and spectrum of linezolid has been continually

1/2
1/1

1/2
1/2
1/1
1/1
1/1
monitored via international surveillance programmes, which were
initiated several years prior to product launch with multiple
Zyvox® Antimicrobial Potency Study (ZAPS) (Ballow et al., 2002a,

Mendes et al.
2002b; Bell et al., 2003; Bolmstrom et al., 2002; Jones et al., 2001)
and other programmes (Mutnick et al., 2002). These surveillance
(2014a)

0.5/1
1/2
1/1

1/2
1/2
1/1
1/1
1/1
studies were followed by the Zyvox® Annual Appraisal of Potency
and Spectrum (ZAAPS) Programme developed as a postmarket-
ing risk management during regulatory compliance tool, which
remains active. ZAAPS initially consisted of a worldwide interna-
tional and multicentre surveillance study (Anderegg et al., 2005;
Rybak et al.

Ross et al., 2005). In 2004, the number of sites in the USA was
(2000)

expanded and the USA sampling was separated from ZAAPS and
4/4
2/4
2/4
2/4
4/4

designated as the Linezolid Experience and Accurate Determination





of Resistance (LEADER) Programme (Draghi et al., 2005; Jones et al.,
2007a, 2008; Farrell et al., 2009, 2011; Flamm et al., 2012a, 2013a;
Johnson et al.

Mendes et al., 2014a). Participating sites from ex-USA remained


within the ZAAPS Programme (Mendes et al., 2014b; Jones et al.,
(2000)

2/2
2/2

4/4
4/4
1/2
2/2
2/2

2006, 2007b, 2009, 2010; Ross et al., 2007a, 2009; Flamm et al.,
–b

2012b, 2013b).
The ZAAPS and LEADER surveillance programmes have sys-
tematically surveyed for linezolid resistance since 2002 and 2004,
Noskin et al.

respectively; applying reference and centralized broth microdilu-


(1999)

tion MIC testing, following CLSI methods (Clinical and Laboratory


2/4
2/4
2/4
2/4
2/4
1/1

Standards Institute, 2012b) and CLSI and EUCAST MIC interpre-



tative criteria (Clinical and Laboratory Standards Institute, 2013;


EUCAST, 2013). The linezolid MIC distributions obtained during the
Peters (1999)

last three surveillance years for LEADER and ZAAPS are displayed
von Eiff and

in Tables 2 and 3, respectively. The postmarketing MIC results for


both programmes are very comparable, which remain very similar
2/2
2/2
1/1

to those described prior to FDA approval by Wise et al. (1998) and






Jones et al. (1996). Also, Tables 2 and 3 exhibit consistent modal


MIC results (1 mg/L) across all species or groups of organisms and
geography, except for CoNS (modal MIC and MIC50 of 0.5 mg/L for
both programmes).
Rybak et al.
(1998)

2/4
2/4
1/2
2/4
2/2

3. Resistance development



Linezolid MIC50 /MIC90 values in mg/L by reference/method:

The linezolid molecule possesses several advantageous char-


MSSA = methicillin-susceptible S. aureus; MRSA = methicillin-resistant S. aureus.

acteristics, including its synthetic nature, bacterial redundancy of


0.25/0.5
Wise et al.

target site (PTC at the 23S rRNA) and absence of cross-resistance


(1998)

1/1
1/1

1/1
1/1
2/2
1/1
1/1

(more on this later). The synthetic nature of this agent implies a low
probability for naturally-occurring resistance mechanisms, which
explains the in vitro study data described above (Tables 2 and 3)
where the linezolid normal and narrow MIC distribution persist
when tested in large surveys of naive clinical Gram-positive iso-
Jones et al.

lates. In fact, studies pre-FDA approval demonstrated essentially


complete antimicrobial coverage against tested Gram-positive
(1996)

2/2
2/2
1/2
1/1
1/2
1/1
2/4
1/2

pathogens (Ballow et al., 2002a, 2002b; Bolmstrom et al., 2002;


Mutnick et al., 2002).
Serial passaging analyses (selection of spontaneous mutations)
Zurenko et al.

performed during drug development demonstrated that resistance


occurred rarely and at a frequency of <1 resistant mutant per
surveillance programmes in 2012.

(1996)

8 × 1011 CFU of S. aureus ATCC 29213, and with even more diffi-
4/4
4/4
2/2
4/4
2/2
1/1
1/2

culty for enterococci (Zurenko et al., 1996). During Phase 3 clinical


trials, resistance to linezolid developed in six patients infected with


Group A streptococci
Group B streptococci

E. faecium (four patients received 200 mg every 12 h, lower than the


Data not available.
Resistance subsets
Organism/Groupa

recommended dose; and two patients received 600 mg every 12 h)


S. pneumoniae

(Zyvox, 2010). In a compassionate use programme, resistance to


S. epidermidis

linezolid developed in eight patients with E. faecium and in one


E. faecium
E. faecalis

patient with Enterococcus faecalis.


MRSA
MSSA
Table 1

After FDA-approval, case reports describing emergence of resis-


a

tance among clinical isolates were published (Gales et al., 2006;


4 R.E. Mendes et al. / Drug Resistance Updates 17 (2014) 1–12

Table 2
Linezolid MIC distribution when tested against species and groups of Gram-positive cocci isolated from USA hospitals during the LEADER Programme for the 2010–2012
period.

Organisma (no. tested) Number (cumulative %) of isolates inhibited at linezolid MIC in mg/L of:b MIC (mg/L)

≤0.12 0.25 0.5 1 2 4 8 >8 MIC50 MIC90

S. aureus (9110) 1 (<0.1) 12 (0.1) 573 (6.4) 7602 (89.9) 910 (99.9) 6 (>99.9) 5 (>99.9) 1 (100.0) 1 2
Oxacillin-susceptible (4584) 0 (0.0) 5 (0.1) 251 (5.6) 3778 (88.0) 549 (>99.9) 0 (>99.9) 0 (>99.9) 1 (100.0) 1 2
Oxacillin-resistant (4526) 1 (<0.1) 7 (0.2) 322 (7.3) 3824 (91.8) 361 (99.8) 6 (99.9) 5 (100.0) – 1 1
CoNS (2458) 6 (0.2) 222 (9.3) 1645 (76.2) 544 (98.3) 10 (98.7) 1 (98.8) 0 (98.8) 30 (100.0) 0.5 1
Enterococcus spp. (3031) 2 (<0.1) 13 (0.5) 302 (10.5) 2398 (89.6) 298 (99.4) 6 (99.6) 9 (99.9) 3 (100.0) 1 2
E. faecalis (1970) 1 (0.2) 10 (0.6) 206 (11.0) 1557 (90.1) 192 (99.8) 2 (99.9) 1 (>99.9) 1 (100.0) 1 1
VRE (71) 0 (0.0) 0 (0.0) 9 (12.7) 60 (97.2) 0 (97.2) 1 (98.6) 1 (>99.9) 1 (100.0) 1 1
E. faecium (942) 0 (0.0) 3 (0.3) 75 (8.3) 757 (88.6) 93 (98.5) 4 (98.9) 8 (99.8) 2 (100.0) 1 2
VRE (716) 0 (0.0) 0 (0.0) 62 (8.7) 596 (91.9) 48 (98.6) 3 (99.0) 6 (99.9) 1 (100.0) 1 1
S. pneumoniae (3019) 13 (0.4) 74 (2.9) 974 (35.1) 1904 (98.2) 53 (>99.9) 1 (100.0) – – 1 1
VGS (1456) 19 (1.3) 56 (5.2) 566 (44.0) 787 (98.1) 27 (>99.9) 0 (>99.9) 0 (>99.9) 1 (100.0) 1 1
BHS (2459) 2 (<0.1) 8 (0.4) 629 (26.0) 1815 (99.8) 5 (100.0) – – – 1 1
a
CoNS = coagulase-negative staphylococci; VRE = vancomycin-resistant enterococci (MIC, ≥8 mg/L); VGS = viridans group streptococci; and BHS = ␤-haemolytic strepto-
cocci.
b
Linezolid MIC distribution for the LEADER surveillance sampling years of 2010, 2011 and 2012 were adapted from Flamm et al. (2012a, 2013) and Mendes et al. (2014b).

Meka et al., 2004; Tsiodras et al., 2001; Wilson et al., 2003; Johnson linezolid non-susceptibility rates against CoNS exhibited an overall
et al., 2002; Auckland et al., 2002). These clinical isolates had alter- slight increased trend for non-susceptibility during ZAAPS Pro-
ations in the linezolid target site (i.e. PTC at 23S rRNA) and these grammes, while increasing rates were observed during LEADER
sporadic reports corroborate the in vitro early experiments suggest- from 2004 to 2008, and decreasing in the following years. Increased
ing that selection for resistance occurs rarely (Zurenko et al., 1996). non-susceptibility rates against enterococci reached a peak in
These earlier cases of resistance development to linezolid occurred 2006 for both the ZAAPS and LEADER Programmes, and fluctu-
during prolonged therapy (>21 days) (Peeters and Sarria, 2005), ated between 0.4–0.7% and 0.3–1.0%, respectively, in the following
as practice contrary to product indications. In addition, these cases period.
were associated with invasive procedures, deep organ involvement, These reported low non-susceptibility rates for linezolid are
and presence of foreign material. Moreover, all patients infected likely due to the number of bacterial 23S rRNA target sites alle-
by isolates that developed resistance during the Phase 3 trials for les (4–6 copies) present in Gram-positive isolates and resistance
linezolid described above had either unremoved prosthetic devices development requires alteration in several alleles. Further studies
or undrained abscesses (Zyvox, 2010). Therefore, caution should demonstrated that the level of decreased linezolid susceptibility
be exercised during linezolid therapy, especially during extended is associated with the number of alleles mutated (Marshall et al.,
treatment courses to monitor for development of resistance. 2002). However, these alterations in the 23S rRNA cause a consider-
The rare clinical occurrence of linezolid resistance development able bacterial fitness cost, mainly when several alleles are mutated.
also coincide with the LEADER Programme results, which reported Interestingly, it has been reported that isolates containing alter-
non-susceptibility rates between 0.03% and 1.83% when tested ations in 23S rRNA revert to a wildtype genotype and susceptible
against more than 42,419 clinical isolates of staphylococci and ente- phenotype, once removed from selective drug pressure; although
rococci across nine consecutive surveillance years (2004–2012) not completely, since a single allele may remain mutated, which
in the USA (Mendes et al., 2014a). The ZAAPS Programme has provides rapid selection to resistance if selective pressure returns
documented even lower linezolid non-susceptibility rates (≤1.2%) (Tsakris et al., 2007).
among staphylococci and enterococci from non-USA hospitals Most of these study results were obtained from staphylo-
(Mendes et al., 2014b) (Fig. 1). More specifically, non-susceptibility cocci and enterococci and selection for resistance seems to be
rates against S. aureus noted in LEADER increased from 2004 extremely rare among streptococci. Although published reports
to 2009, remaining stable in the latter period (2010–2012). The have described the in vitro selection of laboratory strains of

Table 3
Linezolid MIC distributions when tested against species and groups of Gram-positive cocci isolated from hospitals located in four continents during the ZAAPS Programme
for the 2010–2012 period.

Organisma (no. tested) Number (cumulative %) of isolates inhibited at linezolid MIC in mg/L ofb MIC (mg/L)

≤0.12 0.25 0.5 1 2 4 8 >8 MIC50 MIC90

S. aureus (10836) 2 (<0.1) 26 (0.3) 781 (7.5) 8229 (83.4) 1793 (>99.9) 2 (>99.9) 3 (100.0) – 1 2
Oxacillin-susceptible (7191) 2 (<0.1) 9 (0.2) 374 (5.4) 5408 (80.6) 1397 (>99.9) 1 (100.0) – – 1 2
Oxacillin-resistant (3645) 0 (0.0) 17 (0.5) 407 (11.6) 2821 (89.0) 396 (>99.9) 1 (>99.9) 3 (100.0) – 1 2
CoNS (2718) 5 (0.2) 160 (6.1) 1750 (70.5) 754 (98.2) 19 (98.9) 3 (99.0) 6 (99.2) 21 (100.0) 0.5 1
Enterococcus spp. (2344) 0 (0.0) 8 (0.3) 247 (10.9) 1813 (88.2) 263 (99.4) 4 (99.6) 7 (99.9) 2 (100.0) 1 2
E. faecalis (1376) 0 (0.0) 7 (0.5) 141 (10.8) 1056 (87.5) 166 (99.6) 3 (99.8) 2 (>99.9) 1 (100.0) 1 2
VRE (16) 0 (0.0) 0 (0.0) 2 (12.5) 13 (93.8) 1 (100.0) – – – 1 1
E. faecium (870) 0 (0.0) 1 (0.1) 87 (10.1) 687 (89.1) 89 (99.3) 0 (99.3) 5 (99.9) 1 (100.0) 1 2
VRE (249) 0 (0.0) 0 (0.0) 28 (11.2) 188 (86.7) 29 (98.4) 0 (98.4) 3 (99.6) 1 (100.0) 1 2
S. pneumoniae (3343) 9 (0.3) 100 (3.3) 1239 (40.3) 1942 (98.4) 53 (100.0) – – – 1 1
VGS (1255) 7 (0.6) 50 (4.5) 479 (42.7) 692 (97.8) 27 (100.0) – – – 1 1
BHS (1840) 0 (0.0) 0 (0.0) 342 (18.6) 1491 (99.6) 7 (100.0) – – – 1 1
a
CoNS = coagulase-negative staphylococci; VRE = vancomycin-resistant enterococci (MIC, ≥8 mg/L); VGS = viridans group streptococci; and BHS = ␤-haemolytic strepto-
cocci.
b
Linezolid MIC distribution for the ZAAPS surveillance sampling years of 2010, 2011 and 2012 were adapted from Flamm et al. (2012b, 2013) and Mendes et al. (2014b).
R.E. Mendes et al. / Drug Resistance Updates 17 (2014) 1–12 5

T2406C, G2576T, C2610T) and the L22 protein (N56D), and in con-
trast to other results, this isolate did not revert into a wildtype
phenotype/genotype, despite numerous drug-free daily passages
and high fitness cost associated with several ribosomal mutations.
Another linezolid characteristic has been the absence of cross
resistance, which was demonstrated by an early investigation
utilizing several isolates possessing a variety of resistance mech-
anisms (Fines and Leclercq, 2000). Even isolates resistant to other
ribosomal target agents possessing overlapping target sites with
linezolid, remained susceptible to this drug (Long et al., 2010).
Miller et al. (2008) also subsequently reported that mutant isolates
selected with the pleuromutilin, tiamulin, did not exhibited resis-
tance to linezolid, while those selected with linezolid were resistant
to both linezolid and tiamulin. These findings were explained by the
development of specific resistance mechanisms. Isolates selected
with tiamulin developed L3 alterations, while linezolid promoted
the development of 23S rRNA mutations. Isolates carrying the lat-
ter, more specifically at positions G2447, A2503, T2504, G2505 and
G2576 exhibit linezolid and chloramphenicol resistance, but no
correlation was observed for linezolid, clindamycin and valnemulin
(Long et al., 2010).
These previous studies indicate that the presence of cross resis-
tance remained unidirectional, and isolates resistance to some
ribosomal targeting agents remained susceptible to linezolid. This
phenomenon was most likely due to the unique interactions of the
oxazolidinone with the ribosome PTC cavity, which are specific
and allow drug activity (Lin et al., 1997; Leach et al., 2007; Colca
et al., 2003; Shinabarger et al., 1997b). The lack of cross resistance
remained valid for several years until a new mechanism (cfr) was
detected in staphylococcal isolates initially recovered from animals
(Schwarz et al., 2000). Additional details on cfr are described in the
next section.

4. Resistance mechanisms

4.1. Mutations in 23S rRNA and ribosomal proteins L3 and L4

It has been recognized since the pre-FDA approval of linezolid


that alterations in the 23S rRNA are responsible for resis-
tance development. The PTC binding pocket comprises conserved
nucleotides, which interact directly with linezolid. In addition,
other nucleotides located more distally, also confer decreased
levels of susceptibility. Several previous literature reviews cover-
ing linezolid have specifically described the 23S rRNA alterations
associated with linezolid resistance (Shaw and Barbachyn, 2011;
Fig. 1. Summary of the linezolid susceptibility rates, for a 98.0–100.0% range (CLSI Bozdogan and Appelbaum, 2004; Long and Vester, 2012; Stefani
breakpoint criteria) documented during the last nine-years of LEADER and ZAAPS
et al., 2010; Meka and Gold, 2004). Therefore, these mutations
surveillance programmes (2004–2012). Graph A., B. and C. represent the linezolid
susceptibility rates against S. aureus, CoNS and enterococci, respectively. Graphs will not be detailed here. However, of note, alterations in the 23S
adapted from Mendes et al., 2014 (42, 43). All streptococcal isolates were susceptible rRNA are the most common resistance mechanism detected among
to linezolid, except for one S. pneumoniae isolate (MIC, 4 ␮g/mL) and one S. sanguinis staphylococcal and enterococcal clinical organisms, and mutation
(MIC, 32 ␮g/mL) included in the LEADER Programme for 2010 and 2011, respectively
at the G2576 position is often the only mechanism detected among
(40, 68). Polynomial trendlines are also shown.
enterococci (see Table 4).
In addition to alteration in the 23S rRNA, it has been proposed
streptococci showing decreased susceptibility to linezolid due to more recently that mutations in the L3 and L4 ribosomal proteins
mutations in the PTC (Bozdogan and Appelbaum, 2004; Long and can cause decreased susceptibility to linezolid. In fact, alterations
Vester, 2012), just a few clinical cases of linezolid-resistant strepto- in L3 have long been associated with resistance to pleuromutilins
coccal isolates (Streptococcus pneumoniae and Streptococcus oralis) (tiamulin, valnemulin and retapamulin) (Bosling et al., 2003; Bock
have been described to date (Flamm et al., 2012a, 2013a; Wolter et al., 1982) and several studies have reported similar findings
et al., 2005; Mutnick et al., 2003). Those reports demonstrated (Miller et al., 2008; Gentry et al., 2007; Kosowska-Shick et al., 2006;
that these clinical and laboratory isolates had mutations in the 23S Pringle et al., 2004). During an in vitro selection study of mutants
rRNA or amino acid substitutions in L4. In addition, we recently under high concentrations of linezolid and tedizolid, several L3
described a case of linezolid-resistant Streptococcus sanguinis dis- and L4 mutations were detected and these alterations were sug-
playing a linezolid MIC result of 32 mg/L, also associated with a gested to confer decreased susceptibility to linezolid (Locke et al.,
prolonged therapy course (79 days) (Mendes et al., 2013a). This 2009a, 2009b). However, few studies have confirmed the associa-
isolate demonstrated several mutations in the 23S rRNA (T2211C, tion of each of L3 and/or L4 alterations with the levels of linezolid
6 R.E. Mendes et al. / Drug Resistance Updates 17 (2014) 1–12

Table 4
Summary of clinical isolates displaying elevated linezolid MIC results (≥4 mg/L) that were screened for resistance mechanisms during ZAPS, ZAAPS, LEADER and other
surveillance programmes.

Country Year Organism (no.) Resistance mechanism

23S rRNAa Ribosomal proteins Methyltransferase

L3 L4 cfr

Belgium 2006 S. aureus (1) WT S145 deletion WT +

Brazil 2005 S. aureus (1) G2576T WT WT −


2012 S. aureus (1) WT WT V142I +
2006/2010–2012 S. epidermidis (8) G2576T WT WT −
2011 S. hominis (1) G2576T F147I/M156T S77T −
2011 S. lugdunensis (1) G2576T WT WT −
2011 S. haemolyticus (1) G2576T WT WT −
2007/2011 E. faecalis (2) G2576T WT WT −
2008/2010 E. faecium (2) G2576T WT WT −

France 2010 S. aureus (1) WT WT WT +


2008 S. epidermidis (1) G2576T G137V WT −
2011 S. epidermidis (1) G2576T WT WT +
2011 S. epidermidis (1) WT WT WT +

Germany 2010 S. hominis (1) G2576T M156T WT −


2006–2012 E. faecium (11) G2576T WT WT −

Greece 2003 S. aureus (1) G2576T WT WT −


2011 S. capitis (1) G2576T T83A WT −

Hong Kong 2012 S. aureus (1) G2447T WT WT −

Ireland 2007 S. aureus (1) G2576T WT WT −


2011 E. faecium (1) G2576T WT WT −

Italy 2012 S. aureus (1) G2576T (1) WT WT −


2012 S. aureus (1) WT Q136H/H146 deletion G69R/T70P/G71S −
2006–2007 S. epidermidis (8) WT F147L/A157R WT −
2007 S. epidermidis (1) WT F147L/A157R K68R −
2008–2009 S. epidermidis (1) WT F147L/A157R WT +
2008 S. epidermidis (1) G2447T WT WT −
2010/2012 S. epidermidis (3) G2576T WT WT −
2011 S. epidermidis (1) G2576T F147I WT −
2011–2012 S. epidermidis (2) WT F147L WT +
2012 S. epidermidis (1) WT H146Q/V154L/A157R 71G72 insert −
2012 S. epidermidis (1) C2319T H146Q/V154L 71G72 insert −

Korea 2009 E. faecium (1) G2576T WT WT −

Mexico 2009 S. epidermidis (2) WT S158Y/D159Y WT +


2010 S. epidermidis (2) T2504/C2534T G152D/D159Y WT −
2011 S. epidermidis (1) WT WT WT +
2009/2012 S. cohnii (2) WT S158F/D159Y A133T/V155I +
2009 S. haemolyticus (1) WT WT WT +
2011 S. haemolyticus (1) WT M156T WT +
2010 S. capitis (1) WT G152D N158S −

PR China 2006–2010 E. faecalis (8) WT WT WT −

Panama 2011 E. faecalis (2) WT WT WT +

Poland 2012 E. avium (1) WT WT P171S −


2012 E. faecalis (1) G2576T WT WT −

Romania 2010 S. cohnii (1) WT D108E/S158Y/D159Y A133T/V155I +


2010 S. simulans (1) WT A143R/S144F/D145Y WT −
2011 S. simulans (1) WT N130D/F147S/G152A/A157R WT −

Spain 2010 S. epidermidis (1) G2576T WT WT +


2010 S. epidermidis (1) G2576T WT WT −
2011 S. epidermidis (1) WT G152S WT +

Sweden 2002/2008 E. faecalis (2) WT WT WT −

Taiwan 2012 E. faecalis (1) WT WT WT −

Thailand 2010 E. faecalis (1) WT WT WT +

Turkey 2011 E. faecium (1) G2576T WT WT −

Wales 2008 E. faecalis (1) G2576T WT WT −

USA 2002 S. aureus (1) G2576T WT A118V −


2002/2006–2011 S. aureus (9) G2576T WT WT −
2007/2009–2012 S. aureus (12) WT WT WT +
2008 S. aureus (1) WT G152D WT −
2009/2011 S. aureus (2) WT S145 deletion WT −
R.E. Mendes et al. / Drug Resistance Updates 17 (2014) 1–12 7

Table 4 (Continued)

Country Year Organism (no.) Resistance mechanism

23S rRNAa Ribosomal proteins Methyltransferase

L3 L4 cfr

2010 S. aureus (1) WT G155D/S145 deletion WT −


2011 S. aureus (1) G2576T WT WT +
2012 S. aureus (1) G2576T S145 deletion G69A/T70P/G71S −
2002 S. epidermidis (1) C2534T/G2576T WT WT −
2005–2006/2008–2010 S. epidermidis (10) G2576T H146R/M156T 71G72 insert −
2005/2008 S. epidermidis (2) G2576T M156T WT −
2006 S. epidermidis (1) WT H146Q/F147I WT −
2006/2008 S. epidermidis (2) G2447T WT WT −
2006/2010 S. epidermidis (3) WT A157R WT −
2006–2007/2009/2011 S. epidermidis (12) G2576T WT WT −
2006–2008/2011–2012 S. epidermidis (5) G2576T H146R/V154L/M156T 71G72 insert −
2006–2008 S. epidermidis (3) G2576T F147I WT −
2006 S. epidermidis (1) G2447T A157R WT −
2007 S. epidermidis (1) G2576T H146P/Q136L WT −
2007 S. epidermidis (1) G2447T G152D/D159E WT −
2007 S. epidermidis (1) C2534T H146Q/V154L/A157R 71G72 insert +
2008 S. epidermidis (3) G2576T F147L/M156T 71G72 insert −
2008 S. epidermidis (1) T2504A WT WT −
2008 S. epidermidis (1) G2576T WT N130K −
2008 S. epidermidis (1) WT H146deletion/P151Q WT −
2008/2010–2011 S. epidermidis (3) WT H146Q/V154L/A157R 71G72 insert +
2008 S. epidermidis (1) G2576T G137S/H146P 71G72 insert −
2008–2009/2012 S. epidermidis (4) G2576T G137S/H146P/M156T 71G72 insert −
2008 S. epidermidis (1) G2576T F147L/M156T WT −
2009/2012 S. epidermidis (2) WT V154L/A157R 71G72 insert −
2009 S. epidermidis (1) WT H146Q WT −
2009 S. epidermidis (1) WT V154L/A157R P171S −
2009 S. epidermidis (1) WT H146Q/A157R 71G72 insert +
2009 S. epidermidis (1) WT F147L/M157R WT −
2009 S. epidermidis (1) WT H146Q 71G72 insert −
2009–2012 S. epidermidis (4) G2576T G137D/H146R/V154L/M156T 71G72 insert −
2010 S. epidermidis (1) G2576T V154L/M156T WT −
2010 S. epidermidis (1) G2576T G137D/H146P/M156R G69R/71G72 insert −
2010 S. epidermidis (1) G2576T H146P/M156T WT −
2010 S. epidermidis (1) WT A157R WT +
2010 S. epidermidis (1) WT V154L/A157R WT +
2011 S. epidermidis (1) G2576T Q136L/G137S/H146P 71G72 insert −
2011 S. epidermidis (1) G2576T G137S/H146P/M156T WT −
2011 S. epidermidis (2) G2576T H146R/V154L/M156T WT −
2011 S. epidermidis (1) WT A143R/H146Q/V154L 71G72 insert −
2011–2012 S. epidermidis (2) G2576T G137S/H146P/F147Y/M156T 71G72 insert −
2012 S. epidermidis (1) T2531A G152D/D159K WT −
2012 S. epidermidis (1) C2561T H146Q/V154L/A157R 71G72 insert −
2012 S. epidermidis (1) WT H146Q/V154L/A157R 71G72 insert −
2005–2008 S. haemolyticus (4) G2576T M156T WT −
2006 S. capitis (1) G2576T T83A WT −
2007 S. capitis (1) G2576T WT WT −
2009 S. capitis (1) WT WT WT +
2007 S. simulans (1) G2576T WT WT −
2007–2008 S. hominis (2) G2576T WT WT −
2001–2006 E. faecalis (15) G2576T WT WT −
2008/2010–2012 E. faecalis (5) G2576T WT WT −
2001–2012 E. faecium (91) G2576T WT WT −
2012 E. faecium (1) G2576T WT WT +
2003 E. faecium (2) G2576T WT N130K −
2003 E. faecium (1) WT WT N130K −
2002 S. oralis (1) G2576T WT WT −
2010 S. pneumoniae (1) WT WT Q67K/G69V −
2011 S. sanguinisb (1) G2576T/C2610T WT WT −

WT = wildtype nucleotide or amino acid sequencing.


a
E. coli numbering.
b
Alterations also observed in L22 (N56D, I29V and V110A).

resistance. Wolter et al. demonstrated that deletions in L4 (65WR66 acids 127 and 174 of L3 protein and amino acids 65 and 72 of L4 pro-
and 68KG69 deletions) were responsible for a four-fold increased tein (Kosowska-Shick et al., 2010; Locke et al., 2010; Mendes et al.,
in the linezolid MIC result (Wolter et al., 2005). 2010a, 2010b, 2012, 2013b; Endimiani et al., 2011; Beckert et al.,
Since the recognized association of L3 and L4 with linezolid 2012; LaMarre et al., 2013; Pournaras et al., 2013; Cui et al., 2013;
decreased susceptibility, several studies have reported the pres- de Almeida et al., 2013; Baos et al., 2013). Table 4 describes the
ence of L3 and/or L4 mutations with or without 23S rRNA or the linezolid resistance mechanisms observed among clinical isolates
acquired cfr gene. The vast majority of these publications described included in several surveillance programmes, including L3 and/or
alterations among staphylococcal clinical isolates within the amino L4 alterations. Mutations in ribosomal proteins, mainly L3, were
8 R.E. Mendes et al. / Drug Resistance Updates 17 (2014) 1–12

detected in staphylococci collected as early as 2005 in the USA, and Chen et al., 2013; Febler et al., 2013; Gopegui et al., 2012; Quiles-
2006 in Belgium and Italy. A few isolates screened in earlier years Melero et al., 2013; Shore et al., 2010).
(i.e. 2002, 2003 and 2005–2006 in USA, Greece and Brazil) did not All clinical isolates submitted as part of the SENTRY Antimi-
show ribosomal protein alterations, except for one S. aureus (A118V crobial Surveillance Programme, which includes the LEADER and
in L4) collected in 2002 from the USA. It is not certain whether ZAAPS studies, displaying a linezolid MIC result at ≥4 mg/L have
this L4 modification alone confers decreased linezolid susceptibil- been screened for cfr, and the presence of mutations in 23s rRNA
ity. However, these results do indicate that alterations in L3 and L4 and L3 and L4 ribosomal mutations since 2002 (Table 4). The first
as a resistance mechanism appeared later in time and the complex- detection of cfr occurred in a MRSA recovered from blood cultures
ity and number of such alterations certainly increased in CoNS in in a hospitalized patient in Belgium in August 2006 (unpublished
the last six to eight years. data). This case was followed by the detection of cfr in staphylococci
It is interesting to note that mutations in L3 and L4 proteins from the USA in 2007 (Mendes et al., 2008).
are commonly detected among CoNS clinical isolates, while a small The cfr gene has been, with rare exception (Toh et al., 2007),
number of S. aureus and enterococci have demonstrated such muta- plasmid-located and associated with a range of mobile genetic ele-
tions. Moreover, L3 proteins show amino acid sequences with a ments, such as transposons and insertion sequences. These mobile
diversity greater than L4, likely due to the fact that L3 residues structures have been detected among several Gram-positive iso-
are in close proximity to the PTC (Locke et al., 2009a, 2009b). Other lates other than staphylococci, such as E. faecalis (Diaz et al., 2012;
studies have suggested that L3 alterations may decrease the fitness- Liu et al., 2012), Macrococcus caseolyticus and Jeotgalicoccus pinni-
cost associate with 23S rRNA mutations, when the latter is present. pedialis (Wang et al., 2012a), Bacillus spp. (Zhang et al., 2011; Wang
More specifically, Billal et al. (2011) reported that a mutation at et al., 2012b; Dai et al., 2010), Streptococcus suis (Wang et al., 2013),
position Y137 in S. pneumoniae L3 (which corresponds to amino as well as in Gram-negative isolates of E. coli (Zhang et al., 2014)
acid F147 in Staphylococcus epidermidis) restores the fitness cost and Proteus vulgaris (Wang et al., 2011) recovered from several dif-
associated with G2576T. Other authors suggested that L3 and 23S ferent specimen sources collected from animals. However, recent
rRNA alterations may act synergistically, requiring fewer mutated data has demonstrated the Bacillales order as the natural reservoirs
copies of 23S rRNA alleles; therefore minimizing the fitness costs for the cfr genes (Hansen et al., 2012).
associated with 23S rRNA mutations (Mendes et al., 2012). The diversity of organisms where cfr has been detected and the
It is important to mention that some alterations in L3 and association with mobile genetic structures indicates its propensity
L4 have been observed among linezolid-susceptible isolates, such for genetic mobilization. A complete review on the genetic envi-
as N158S in L4 (Wong et al., 2010). In addition, our group has ronment of cfr has been recently published (Shen et al., 2013); and
previously observed several amino acid alterations in L3 and L4 pro- therefore will not be represented in this review. Interspecies dis-
tein among linezolid-susceptible Gram-positive clinical organisms, semination of cfr has been previously documented (Mendes et al.,
when studying resistance mechanisms other than those associated 2010b, 2013b), as well as outbreaks caused by either Cfr-producing
with linezolid. Among these amino acid modifications, the follow- S. epidermidis or S. aureus (Bonilla et al., 2010; Cai et al., 2012;
ing were observed: A82, A83, G152, V188 and E191 in L3 protein Morales et al., 2010). Following the first outbreak caused by cfr-
among staphylococci; A50 and V142 in S. aureus L4; and N130 and carrying S. aureus in the ICU of Hospital Clínico San Carlos in 2008
G166 in E. faecium L4 protein (unpublished data). (Morales et al., 2010), Baos et al. (2013) monitored the presence
of linezolid-resistant staphylococci from 2008 through 2011. The
authors did not recover linezolid-resistant S. aureus isolates dur-
4.2. Acquired resistance mechanism ing the study period. However, the number of linezolid-resistant S.
epidermidis (58% cfr-positive) increase to 25% by 2010, when rates
Linezolid binding site alterations remain the main mechanism decreased slightly to 20% in the next year. These increased rates of
of resistance detected among Gram-positive clinical organisms. linezolid-resistant S. epidermidis were observed despite implemen-
Although theoretically possible, transfer of ribosomal mutations tation of infection control interventions.
between pathogenic species has not been reported. However, a Through the LEADER and ZAAPS Programme experiences, the
more recent plasmid encoded resistance mechanism (transfer- detection of cfr among Gram-positive clinical isolates remains spo-
able), namely Cfr, has been observed. Cfr is closely related to radic with no apparent increase across the years. In addition, most
RlmN methyltransferase, which belongs to the radical S-adenosyl- isolates seem to be endemic in specific hospitals in Arizona (Mendes
l-methionine (SAM) enzyme superfamily (Yan et al., 2010). RlmN et al., 2008), Ohio (Mendes et al., 2008) Italy (Flamm et al., 2013b;
is an indigenous cellular enzyme that mono-methylates the C2 Mendes et al., 2010a) and Mexico (Flamm et al., 2013b; Mendes
atom of A2503 at 23S rRNA. On the other hand, Cfr catalyzes the et al., 2010b). Other cfr-positive strains have originated from ran-
post-transcriptional methylation of the C8 atom at the same posi- dom sites in the USA (10 S. aureus, 2009–2012; two S. epidermidis,
tion (Toh et al., 2008; Atkinson et al., 2013). This modification 2010; one Staphylococcus capitis, 2009; and one E. faecium, 2012),
confers a MDR phenotype, affecting the susceptibility to pheni- Brazil (one S. aureus, 2012), France (one S. aureus, 2010; two S. epi-
col, lincosamide, oxazolidinone, pleuromutilin, and streptogramin dermidis, 2011), Spain (two S. epidermidis, 2010 and 2011), Panama
A (PhLOPSA ) compounds (Long et al., 2006). (two E. faecalis, 2011), Romania (one Staphylococcus cohnii, 2010)
The Cfr-encoding gene was initially described in 2000 in a and Thailand (one E. faecalis, 2010) (Jones et al., 2006, 2007a, 2008,
17,108 bp 16.5-kb transferable plasmid (pSCFS1) from an animal 2009, 2010; Farrell et al., 2009, 2011; Flamm et al., 2012a, 2012b,
isolate of Staphylococcus sciuri (Schwarz et al., 2000; Kehrenberg 2013a, 2013b; Mendes et al., 2014b; Ross et al., 2007a, 2007b, 2009;
et al., 2004), and was detected in several other staphylococcal Diaz et al., 2012; Deshpande et al., 2013).
organisms also from animal origin in Europe during investigations In a more recent investigation, Quiles-Melero et al. (2013) fur-
associated with resistance to phenicols (florphenicol and chloram- ther investigated linezolid-resistant staphylococci clinical isolates
phenicol) (Kehrenberg and Schwarz, 2006; Kehrenberg et al., 2009). recovered between 2005 and 2009 in a tertiary hospital in Madrid,
In 2005, the first cfr-carrying S. aureus was recovered from a respi- Spain. The authors included 256 isolates (2.5% of total) and all
ratory tract specimen in a patient from Colombia (Toh et al., 2007). seven linezolid-resistant S. aureus (1.0% of total) included were cfr-
This finding was followed by several other reports describing the positive. Six isolates had spa t067 and were recovered during 2008.
detection of cfr among staphylococci recovered from human spec- In the same study, cfr was detected among eight (8/125; 6.4%) of
imens (Mendes et al., 2008, 2009, 2010a, 2010b; Cui et al., 2013; 125 (125/2183; 4.0%) linezolid-resistant S. epidermidis. Sierra et al.
R.E. Mendes et al. / Drug Resistance Updates 17 (2014) 1–12 9

(2013), who evaluate the prevalence of cfr among 2215 bacteremic observe the changes associated with linezolid resistance mecha-
MRSA clinical isolates in Hospital Universitari de Bellvitge (HUB) nisms. In the early 2000s, non-susceptible clinical isolates initially
from 1999 to 2010, observed only a single Cfr-producing MRSA harboured (almost exclusively) alterations in the 23S rRNA target,
(0.05% of total) (Sierra et al., 2013). which is still observed, and has remained the dominant mechanism
The cfr gene has a great potential for dissemination due to its detected among enterococci. However, a limited number of clinical
association with mobile elements (Shen et al., 2013) and low fit- isolates also carrying cfr.
ness cost (LaMarre et al., 2011). In addition, the MDR phenotype In contrast, the scenario has changed significantly among S.
conferred by cfr provides multiple possibilities for in vivo selec- epidermidis and other species of CoNS, which were initially less
tion during antimicrobial therapy and several studies have reported prevalent, and when detected presented the usual alterations in
the detection of cfr-carrying isolates even without prior exposure 23S rRNA. These isolates currently exhibit more diverse and com-
to linezolid (Baos et al., 2013; Deshpande et al., 2013; Gales et al., plex mechanisms of resistance, which consist of a greater number
2014). Moreover, cfr is often associated with other resistance genes, of mutations in ribosomal proteins in conjunction with modifica-
such as erm, fexA, lsa(B) and tet(L), which can assist co-selecting tions in 23S rRNA and/or presence of cfr. Further investigations
the cfr gene and its spread (Shen et al., 2013; Gales et al., 2014; will be required to understand whether these isolates now demon-
Smith and Mankin, 2008). Also, several cfr-carrying isolates sub- strating these complexes DNA alterations and presence of foreign
mitted though the SENTRY Programme, mainly S. aureus, exhibit DNA belong to certain lineages that possess greater potential for
a linezolid MIC value at the extreme of the wildtype distribution adapting to the nosocomial environment and antimicrobial selec-
(i.e. 4 mg/L) and would be considered susceptible when applying tive pressure. The presence of linezolid resistance mechanisms in
the current breakpoints (≤4 mg/L for susceptible). These charac- CoNS may become a concern, since these organisms have gained
teristics warrant active surveillance at local and national levels for clinical relevance. Moreover, these CoNS can serve as a reservoir
early detection and implementation of infection control policies. for cfr.
S. aureus fortunately does not seem to behave like S. epidermidis
and other species of CoNS with regard to resistance mechanisms.
4.3. Resistance mechanisms other than target site modifications
Among linezolid non-susceptible or resistant clinical isolates, S.
aureus remains rare in the original surveillance programmes. In
We have observed that some enterococci displaying elevated
addition, and different from CoNS, most isolates demonstrate a sin-
MIC results for linezolid (MIC, 4–8 mg/L) included in the LEADER
gle linezolid resistance mechanism (cfr). However, this feature has
and ZAAPS Programmes did not show any target site modifica-
often produced isolates with linezolid MIC results at the end of
tions associated with linezolid resistance (i.e. wildtype nucleotide
the wildtype distribution and borderline for susceptibility (i.e. MIC,
sequences for 23S rRNA and amino acid sequences for L3, L4 and
4 mg/L), which may complicate detection.
L22) or presence cfr. Isolates demonstrating such phenotypic and
genotypic characteristics have been collected from China, Panama,
Sweden and Taiwan (Table 4). Recent studies published in the lit-
Acknowledgements
erature, which evaluated the linezolid resistance mechanisms in
Gram-positive isolates from Canada and China, have also reported
The authors wish to thank the following staff members at JMI
similar results (Chen et al., 2013; Patel et al., 2013). Previous studies
Laboratories (North Liberty, Iowa, USA): S. Benning, M. Castanheira,
have demonstrated that linezolid can be recognized as a substrate
P. Clark, A. Costello, D. Farrell, S. Farrell, R. Flamm, M. Konrardy, P.
of efflux-pump systems, which can extrude a wide range of struc-
Romberg, J. Ross, H. Sader, M. Stilwell, J. Streit and L. Woosley for
turally dissimilar substrates (Sierra et al., 2009; Floyd et al., 2010).
technical support and manuscript assistance.
Although the isolates above described were not evaluated for the
presence of efflux-pump or their expression levels, it is tempt-
ing to associate these low-level resistance phenotypes with the
over-expression of such MDR pumps. Further investigations should References
determine the role of efflux-pump systems in these very rare but
Anderegg, T.R., Sader, H.S., Fritsche, T.R., Ross, J.E., Jones, R.N., 2005. Trends in
geographically diverse cases of linezolid decreased susceptibility. linezolid susceptibility patterns: report from the 2002–2003 worldwide Zyvox
Annual Appraisal of Potency and Spectrum (ZAAPS) Program. Int. J. Antimicrob.
Agents 26, 13–21.
5. Conclusions Atkinson, G.C., Hansen, L.H., Tenson, T., Rasmussen, A., Kirpekar, F., Vester, B., 2013.
Distinction between the Cfr methyltransferase conferring antibiotic resistance
and the housekeeping RlmN methyltransferase. Antimicrob. Agents Chemother.
Linezolid has become and remains an important agent for 57, 4019–4026.
treating serious infections caused by Gram-positive organisms, Auckland, C., Teare, L., Cooke, F., Kaufmann, M.E., Warner, M., Jones, G., Bamford, K.,
including MDR isolates. This oxazolidinone has been well studied Ayles, H., Johnson, A.P., 2002. Linezolid-resistant enterococci: report of the first
isolates in the United Kingdom. J. Antimicrob. Chemother. 50, 743–746.
and much is currently known about its clinical applications, adverse Ballow, C.H., Biedenbach, D.J., Rossi, F., Jones, R.N., 2002a. Multicenter assessment of
effects, mode of action, spectrum of activity, risks for resistance the linezolid spectrum and activity using the disk diffusion and Etest methods:
development and resistance mechanisms. The knowledge acquired report of the Zyvox(R) Antimicrobial Potency Study in Latin America (LA-ZAPS).
Braz. J. Infect. Dis. 6, 100–109.
during the past thirteen years has been applied for developing new Ballow, C.H., Jones, R.N., Biedenbach, D.J., 2002b. A multicenter evaluation of line-
oxazolidinone agents, which have completed Phase 2 and 3 clinical zolid antimicrobial activity in North America. Diagn. Microbiol. Infect. Dis. 43,
trials (Shaw and Barbachyn, 2011; Prokocimer et al., 2013). In addi- 75–83.
Baos, E., Candel, F.J., Merino, P., Pena, I., Picazo, J.J., 2013. Characterization and
tion, the linezolid patents in the USA and abroad will soon expire
monitoring of linezolid-resistant clinical isolates of Staphylococcus epider-
and generic brands will certainly become available. These events midis in an intensive care unit 4 years after an outbreak of infection by
will likely establish a new era for this class of antimicrobial agents. cfr-mediated linezolid-resistant Staphylococcus aureus. Diagn. Microbiol. Infect.
Dis. 76, 325–329.
The occurrences of linezolid non-susceptible or resistant iso-
Beckert, P., Hillemann, D., Kohl, T.A., Kalinowski, J., Richter, E., Niemann, S.,
lates have remained relatively low since the drug introduction into Feuerriegel, S., 2012. rplC T460C identified as a dominant mutation in linezolid-
clinical use. However, sporadic cases of linezolid-resistant isolates resistant Mycobacterium tuberculosis strains. Antimicrob. Agents Chemother. 56,
do occur, as well as epidemic outbreaks, emphasizing the impor- 2743–2745.
Bell, J.M., Turnidge, J.D., Ballow, C.H., Jones, R.N., 2003. Multicentre evaluation of the
tance for antimicrobial stewardship and surveillance for resistance. in vitro activity of linezolid in the Western Pacific. J. Antimicrob. Chemother. 51,
As this review has described, it has certainly been interesting to 339–345.
10 R.E. Mendes et al. / Drug Resistance Updates 17 (2014) 1–12

Biedenbach, D.J., Jones, R.N., 1997. Disk diffusion test interpretive criteria and quality Farrell, D.J., Mendes, R.E., Ross, J.E., Jones, R.N., 2009. Linezolid surveillance program
control recommendations for testing linezolid (U-100766) and eperezolid (U- results for 2008 (LEADER Program for 2008). Diagn. Microbiol. Infect. Dis. 65,
100592) with commercially prepared reagents. J. Clin. Microbiol. 35, 3198–3202. 392–403.
Biedenbach, D.J., Jones, R.N., 2001. In vitro activity of linezolid (U-100766) against Farrell, D.J., Mendes, R.E., Ross, J.E., Sader, H.S., Jones, R.N., 2011. LEADER Program
Haemophilus influenzae measured by three different susceptibility testing meth- results for 2009: an activity and spectrum analysis of linezolid using 6,414 clin-
ods. Diagn. Microbiol. Infect. Dis. 39, 49–53. ical isolates from the United States (56 Medical Centers). Antimicrob. Agents
Biedenbach, D.J., Jones, R.N., 2003. Revision of linezolid disk diffusion quality con- Chemother. 55, 3684–3690.
trol guidelines for testing Staphylococcus aureus ATCC, 25923. An independent Febler, A.T., Calvo, N., Gutierrez, N., Munoz Bellido, J.L., Fajardo, M., Garduno, E.,
seven-laboratory trial. Clin. Microbiol. Infect. 9, 1035–1037. Monecke, S., Ehricht, R., Kadlec, K., Schwarz, S., 2013. Cfr-mediated linezolid
Billal, D.S., Feng, J., Leprohon, P., Legare, D., Ouellette, M., 2011. Whole genome anal- resistance in methicillin-resistant Staphylococcus aureus and Staphylococcus
ysis of linezolid resistance in Streptococcus pneumoniae reveals resistance and haemolyticus associated with clinical infections in humans: two case reports.
compensatory mutations. BMC Genomics 12, 512. J. Antimicrob. Chemother. 69, 268–270.
Bock, A., Turnowsky, F., Hogenauer, G., 1982. Tiamulin resistance mutations in Fines, M., Leclercq, R., 2000. Activity of linezolid against Gram-positive cocci pos-
Escherichia coli. J. Bacteriol. 151, 1253–1260. sessing genes conferring resistance to protein synthesis inhibitors. J. Antimicrob.
Bolmstrom, A., Ballow, C.H., Qwarnstrom, A., Biedenbach, D.J., Jones, R.N., 2002. Mul- Chemother. 45, 797–802.
ticentre assessment of linezolid antimicrobial activity and spectrum in Europe: Flamm, R.K., Farrell, D.J., Mendes, R.E., Ross, J.E., Sader, H.S., Jones, R.N., 2012a.
report from the Zyvox antimicrobial potency study (ZAPS-Europe). Clin. Micro- LEADER surveillance program results for 2010: an activity and spectrum anal-
biol. Infect. 8, 791–800. ysis of linezolid using 6801 clinical isolates from the United States (61 medical
Bonilla, H., Huband, M.D., Seidel, J., Schmidt, H., Lescoe, M., McCurdy, S.P., Lemmon, centers). Diagn. Microbiol. Infect. Dis. 74, 54–61.
M.M., Brennan, L.A., Tait-Kamradt, A., Puzniak, L., Quinn, J.P., 2010. Multicity Flamm, R.K., Farrell, D.J., Mendes, R.E., Ross, J.E., Sader, H.S., Jones, R.N., 2012b.
outbreak of linezolid-resistant Staphylococcus epidermidis associated with clonal ZAAPS program results for 2010: an activity and spectrum analysis of linezolid
spread of a cfr-containing strain. Clin. Infect. Dis. 51, 796–800. using clinical isolates from 75 medical centers in 24 countries. J. Chemother. 24,
Bosling, J., Poulsen, S.M., Vester, B., Long, K.S., 2003. Resistance to the peptidyl 328–337.
transferase inhibitor tiamulin caused by mutation of ribosomal protein L3. Flamm, R.K., Mendes, R.E., Ross, J.E., Sader, H.S., Jones, R.N., 2013a. Linezolid
Antimicrob. Agents Chemother. 47, 2892–2896. surveillance results for the United States: LEADER Surveillance Program 2011.
Bozdogan, B., Appelbaum, P.C., 2004. Oxazolidinones: activity, mode of action, and Antimicrob. Agents Chemother. 57, 1077–1081.
mechanism of resistance. Int. J. Antimicrob. Agents 23, 113–119. Flamm, R.K., Mendes, R.E., Ross, J.E., Sader, H.S., Jones, R.N., 2013b. An international
Cai, J.C., Hu, Y.Y., Zhang, R., Zhou, H.W., Chen, G.X., 2012. Linezolid-resistant clinical activity and spectrum analysis of linezolid: ZAAPS program results for 2011.
isolates of meticillin-resistant coagulase-negative staphylococci and Enterococ- Diagn. Microbiol. Infect. Dis. 76, 206–213.
cus faecium from China. J. Med. Microbiol. 61, 1568–1573. Floyd, J.L., Smith, K.P., Kumar, S.H., Floyd, J.T., Varela, M.F., 2010. LmrS is a multidrug
Chen, H., Wu, W., Ni, M., Liu, Y., Zhang, J., Xia, F., He, W., Wang, Q., Wang, Z., Cao, B., efflux pump of the major facilitator superfamily from Staphylococcus aureus.
Wang, H., 2013. Linezolid-resistant clinical isolates of enterococci and Staphy- Antimicrob. Agents Chemother. 54, 5406–5412.
lococcus cohnii from a multicentre study in China: molecular epidemiology and Gales, A.C., Sader, H.S., Andrade, S.S., Lutz, L., Machado, A., Barth, A.L., 2006.
resistance mechanisms. Int. J. Antimicrob. Agents 42, 317–321. Emergence of linezolid-resistant Staphylococcus aureus during treatment of pul-
Clinical and Laboratory Standards Institute, 2006. CLSI Subcommittee on Antimicro- monary infection in a patient with cystic fibrosis. Int. J. Antimicrob. Agents 27,
bial Testing Meeting Minutes, January 2006. Clinical and Laboratory Standards 300–302.
Institute, Miami, FL, USA. Gales, A.C., Deshpande, L.M., Jones, R.N., De Souza, A.G., Pignatari, A.C., Mendes,
Clinical and Laboratory Standards Institute, 2012a. CLSI Subcommittee on Antimi- R.E., 2014. Fatal pneumonia due to methicillin-susceptible Staphylococcus aureus
crobial Susceptibility Testing Meeting Minutes, January 2012. Clinical and ST398/t034 carrying cfr and erm(B) genes: first report in Brazil. J. Antimicrob.
Laboratory Standards Institute, Tempe, AZ, USA. Chemother. (in press).
Clinical and Laboratory Standards Institute, 2012b. M07-A9. Methods for Dilution Gentry, D.R., Rittenhouse, S.F., McCloskey, L., Holmes, D.J., 2007. Stepwise exposure
Antimicrobial Susceptibility Tests for Bacteria that Grow Aerobically; Approved of Staphylococcus aureus to pleuromutilins is associated with stepwise acqui-
Standard, 9th edition. Clinical and Laboratory Standards Institute, Wayne, PA, sition of mutations in rplC and minimally affects susceptibility to retapamulin.
USA. Antimicrob. Agents Chemother. 51, 2048–2052.
Clinical and Laboratory Standards Institute, 2013. M100-S23. Performance Stan- Gopegui, E.R., Juan, C., Zamorano, L., Perez, J.L., Oliver, A., 2012. Transferable mul-
dards for Antimicrobial Susceptibility Testing: 23rd Informational Supplement. tidrug resistance plasmid carrying cfr associated with tet(L), ant(4 )-Ia, and dfrK
Clinical and Laboratory Standards Institute, Wayne, PA, USA. genes from a clinical methicillin-resistant Staphylococcus aureus ST125 strain.
Clinical and Laboratory Standards Institute, 2014. M07-A10. Methods for Dilution Antimicrob. Agents Chemother. 56, 2139–2142.
Antimicrobial Susceptibility Tests for Bacteria that Grow Aerobically; Approved Gould, I.M., Bal, A.M., 2013. New antibiotic agents in the pipeline and
Standard, 10th ed. Clinical and Laboratory Standards Institute, Wayne, PA, USA. how they can help overcome microbial resistance. Virulence 4, 185–
Colca, J.R., McDonald, W.G., Waldon, D.J., Thomasco, L.M., Gadwood, R.C., Lund, E.T., 191.
Cavey, G.S., Mathews, W.R., Adams, L.D., Cecil, E.T., Pearson, J.D., Bock, J.H., Mott, Hansen, L.H., Planellas, M.H., Long, K.S., Vester, B., 2012. The order Bacillales hosts
J.E., Shinabarger, D.L., Xiong, L., Mankin, A.S., 2003. Cross-linking in the living functional homologs of the worrisome cfr antibiotic resistance gene. Antimicrob.
cell locates the site of action of oxazolidinone antibiotics. J. Biol. Chem. 278, Agents Chemother. 56, 3563–3567.
21972–21979. Johnson, A.P., Warner, M., Livermore, D.M., 2000. Activity of linezolid against multi-
Cui, L., Wang, Y., Li, Y., He, T., Schwarz, S., Ding, Y., Shen, J., Lv, Y., 2013. Cfr-mediated resistant Gram-positive bacteria from diverse hospitals in the United Kingdom.
linezolid-resistance among methicillin-resistant coagulase-negative Staphylo- J. Antimicrob. Chemother. 45, 225–230.
cocci from infections of humans. PLoS ONE 8, e57096. Johnson, A.P., Tysall, L., Stockdale, M.V., Woodford, N., Kaufmann, M.E., Warner, M.,
Dai, L., Wu, C.M., Wang, M.G., Wang, Y., Wang, Y., Huang, S.Y., Xia, L.N., Li, B.B., Shen, Livermore, D.M., Asboth, F., Allerberger, F.J., 2002. Emerging linezolid-resistant
J.Z., 2010. First report of the multidrug resistance gene cfr and the phenicol Enterococcus faecalis and Enterococcus faecium isolated from two Austrian
resistance gene fexA in a Bacillus strain from swine feces. Antimicrob. Agents patients in the same intensive care unit. Eur. J. Clin. Microbiol. Infect. Dis. 21,
Chemother. 54, 3953–3955. 751–754.
de Almeida, L.M., de Araujo, M.R., Sacramento, A.G., Pavez, M., de Souza, A.G., Jones, R.N., Johnson, D.M., Erwin, M.E., 1996. In vitro antimicrobial activities and
Rodrigues, F., Gales, A.C., Lincopan, N., Sampaio, J.L., Mamizuka, E.M., 2013. spectra of U-100592 and U-100766, two novel fluorinated oxazolidinones.
Linezolid resistance in Brazilian Staphylococcus hominis strains is associated Antimicrob. Agents Chemother. 40, 720–726.
with L3 and 23S rRNA ribosomal mutations. Antimicrob. Agents Chemother. Jones, R.N., Ballow, C.H., Biedenbach, D.J., 2001. Multi-laboratory assessment of the
57, 4082–4083. linezolid spectrum of activity using the Kirby-Bauer disk diffusion method:
Deshpande, L.M., Vega, S., Ross, J.E., Jones, R.N., Mendes, R.N., 2013. Detection of report of the Zyvox Antimicrobial Potency Study (ZAPS) in the United States.
plasmid-encoded Cfr in clinical isolates of Enterococcus faecalis from Panama Diagn. Microbiol. Infect. Dis. 40, 59–66.
City: report from the SENTRY Antimicrobial Surveillance Programme. In: Jones, R.N., Ross, J.E., Fritsche, T.R., Sader, H.S., 2006. Oxazolidinone susceptibility
Abstract P1405. 23rd ECCMID, April 27–30, 2013, Berlin, Germany. patterns in 2004: report from the Zyvox® Annual Appraisal of Potency and
Diaz, L., Kiratisin, P., Mendes, R., Panesso, D., Singh, K.V., Arias, C.A., 2012. Trans- Spectrum (ZAAPS) Program assessing isolates from 16 nations. J. Antimicrob.
ferable plasmid-mediated resistance to linezolid due to cfr in a human clinical Chemother. 57, 279–287.
isolate of Enterococcus faecalis. Antimicrob. Agents Chemother. 56, 3917–3922. Jones, R.N., Fritsche, T.R., Sader, H.S., Ross, J.E., 2007a. LEADER surveillance program
Draghi, D.C., Sheehan, D.J., Hogan, P., Sahm, D.F., 2005. In vitro activity of linezolid results for 2006: an activity and spectrum analysis of linezolid using clinical
against key Gram-positive organisms isolated in the United States: results of isolates from the United States (50 medical centers). Diagn. Microbiol. Infect.
the LEADER 2004 surveillance program. Antimicrob. Agents Chemother. 49, Dis. 59, 309–317.
5024–5032. Jones, R.N., Fritsche, T.R., Sader, H.S., Ross, J.E., 2007b. Zyvox® Annual Appraisal
Endimiani, A., Blackford, M., Dasenbrook, E.C., Reed, M.D., Bajaksouszian, S., Hujer, of Potency and Spectrum Program results for 2006: an activity and spectrum
A.M., Rudin, S.D., Hujer, K.M., Perreten, V., Rice, L.B., Jacobs, M.R., Konstan, M.W., analysis of linezolid using clinical isolates from 16 countries. Diagn. Microbiol.
Bonomo, R.A., 2011. Emergence of linezolid-resistant Staphylococcus aureus after Infect. Dis. 59, 199–209.
prolonged treatment of cystic fibrosis patients in Cleveland, Ohio. Antimicrob. Jones, R.N., Ross, J.E., Castanheira, M., Mendes, R.E., 2008. United States resistance
Agents Chemother. 55, 1684–1692. surveillance results for linezolid (LEADER Program for 2007). Diagn. Microbiol.
EUCAST, 2013. Breakpoint tables for interpretation of MICs and zone diam- Infect. Dis. 62, 416–426.
eters. Version 3.1, February 2013., available at http://www.eucast. Jones, R.N., Kohno, S., Ono, Y., Ross, J.E., Yanagihara, K., 2009. ZAAPS Interna-
org/clinical breakpoints/ (accessed August 2013). tional Surveillance Program (2007) for linezolid resistance: results from 5591
R.E. Mendes et al. / Drug Resistance Updates 17 (2014) 1–12 11

Gram-positive clinical isolates in 23 countries. Diagn. Microbiol. Infect. Dis. 64, Mendes, R.E., Deshpande, L.M., Costello, A., Farrell, D.J., 2012. Molecular epi-
191–201. demiology of Staphylococcus epidermidis clinical isolates from USA hospitals.
Jones, R.N., Mendes, R.E., Farrell, D.J., Sader, H.S., Ross, J.E., 2010. Summary of Antimicrob. Agents Chemother. 56, 4656–4661.
2009 oxazolidinone resistance (R) surveillance studies worldwide (LEADER and Mendes, R.E., Deshpande, L.M., Kim, J., Myers, D.S., Ross, J.E., Jones, R.N., 2013a. Strep-
ZAAPS Programs). In: Abstract 263. 48th IDSA, October 21–24, 2010, Vancouver, tococcus sanguinis displaying a cross resistance phenotype to several ribosomal
B.C., Canada. RNA targeting agents. J. Clin. Microbiol. 51, 2728–2731.
Kehrenberg, C., Schwarz, S., 2006. Distribution of florfenicol resistance genes fexA Mendes, R.E., Deshpande, L.M., Bonilla, H.F., Schwarz, S., Huband, M.D., Jones, R.N.,
and cfr among chloramphenicol-resistant Staphylococcus spp. isolates. Antimi- Quinn, J.P., 2013b. Dissemination of a pSCFS3-like cfr-carrying plasmid in Staphy-
crob. Agents Chemother. 50, 1156–1163. lococcus aureus and Staphylococcus epidermidis clinical isolates recovered from
Kehrenberg, C., Ojo, K.K., Schwarz, S., 2004. Nucleotide sequence and organization hospitals in Ohio. Antimicrob. Agents Chemother. 57, 2923–2928.
of the multiresistance plasmid pSCFS1 from Staphylococcus sciuri. J. Antimicrob. Mendes, R.E., Flamm, R.K., Hogan, P.A., Ross, J.E., Jones, R.N., 2014a. Summary of
Chemother. 54, 936–939. linezolid activity and resistance mechanisms detected during the 2012 LEADER
Kehrenberg, C., Cuny, C., Strommenger, B., Schwarz, S., Witte, W., 2009. Methicillin- surveillance program for the United States. Antimicrob. Agents Chemother. 58,
resistant and -susceptible Staphylococcus aureus strains of clonal lineages ST398 1243–1247.
and ST9 from swine carry the multidrug resistance gene cfr. Antimicrob. Agents Mendes, R.E., Hogan, P.A., Streit, J.M., Jones, R.N., Flamm, R.K., 2014b. Zyvox® Annual
Chemother. 53, 779–781. Appraisal of Potency and Spectrum (ZAAPS) Program: report of linezolid activity
Kosowska-Shick, K., Clark, C., Credito, K., McGhee, P., Dewasse, B., Bogdanovich, T., over nine years (2004–2012). J. Antimicrob. Chemother. (in press).
Appelbaum, P.C., 2006. Single- and multistep resistance selection studies on the Michalska, K., Karpiuk, I., Krol, M., Tyski, S., 2013. Recent development of potent
activity of retapamulin compared to other agents against Staphylococcus aureus analogues of oxazolidinone antibacterial agents. Bioorg. Med. Chem. Lett. 21,
and Streptococcus pyogenes. Antimicrob. Agents Chemother. 50, 765–769. 577–591.
Kosowska-Shick, K., Julian, K.G., McGhee, P.L., Appelbaum, P.C., Whitener, C.J., 2010. Miller, K., Dunsmore, C.J., Fishwick, C.W., Chopra, I., 2008. Linezolid and tia-
Molecular and epidemiologic characteristics of linezolid-resistant coagulase- mulin cross-resistance in Staphylococcus aureus mediated by point mutations
negative staphylococci at a tertiary care hospital. Diagn. Microbiol. Infect. Dis. in the peptidyl transferase center. Antimicrob. Agents Chemother. 52, 1737–
68, 34–39. 1742.
LaMarre, J.M., Locke, J.B., Shaw, K.J., Mankin, A.S., 2011. Low fitness cost of the mul- Morales, G., Picazo, J.J., Baos, E., Candel, F.J., Arribi, A., Pelaez, B., Andrade, R., de
tidrug resistance gene cfr. Antimicrob. Agents Chemother. 55, 3714–3719. la Torre, M.A., Fereres, J., Sanchez-Garcia, M., 2010. Resistance to linezolid is
LaMarre, J., Mendes, R.E., Szal, T., Schwarz, S., Jones, R.N., Mankin, A.S., 2013. The mediated by the cfr gene in the first report of an outbreak of linezolid-resistant
genetic environment of the cfr gene and the presence of other mechanisms Staphylococcus aureus. Clin. Infect. Dis. 50, 821–825.
account for the very high linezolid resistance of the Staphylococcus epidermidis Mutnick, A.H., Biedenbach, D.J., Turnidge, J.D., Jones, R.N., 2002. Spectrum and
isolate 426-3147L. Antimicrob. Agents Chemother. 57, 1173–1179. potency evaluation of a new oxazolidinone, linezolid: report from the SENTRY
Leach, K.L., Swaney, S.M., Colca, J.R., McDonald, W.G., Blinn, J.R., Thomasco, L.M., Antimicrobial Surveillance Program, 1998–2000. Diagn. Microbiol. Infect. Dis.
Gadwood, R.C., Shinabarger, D., Xiong, L., Mankin, A.S., 2007. The site of action 43, 65–73.
of oxazolidinone antibiotics in living bacteria and in human mitochondria. Mol. Mutnick, A.H., Enne, V., Jones, R.N., 2003. Linezolid resistance since 2001: SENTRY
Cell 26, 393–402. Antimicrobial Surveillance Program. Ann. Pharmacother. 37, 769–774.
Lin, A.H., Murray, R.W., Vidmar, T.J., Marotti, K.R., 1997. The oxazolidinone eperezolid Noskin, G.A., Siddiqui, F., Stosor, V., Hacek, D., Peterson, L.R., 1999. In vitro
binds to the 50S ribosomal subunit and competes with binding of chlorampheni- activities of linezolid against important Gram-positive bacterial pathogens
col and lincomycin. Antimicrob. Agents Chemother. 41, 2127–2131. including vancomycin-resistant enterococci. Antimicrob. Agents Chemother. 43,
Liu, Y., Wang, Y., Wu, C., Shen, Z., Schwarz, S., Du, X.D., Dai, L., Zhang, W., Zhang, Q., 2059–2062.
Shen, J., 2012. First report of the multidrug resistance gene cfr in Enterococcus Patel, S.N., Memari, N., Shahinas, D., Toye, B., Jamieson, F.B., Farrell, D.J., 2013.
faecalis of animal origin. Antimicrob. Agents Chemother. 56, 1650–1654. Linezolid resistance in Enterococcus faecium isolated in Ontario, Canada. Diagn.
Locke, J.B., Hilgers, M., Shaw, K.J., 2009a. Mutations in ribosomal protein L3 are Microbiol. Infect. Dis. 77, 350–353.
associated with oxazolidinone resistance in staphylococci of clinical origin. Peeters, M.J., Sarria, J.C., 2005. Clinical characteristics of linezolid-resistant Staphy-
Antimicrob. Agents Chemother. 53, 5275–5278. lococcus aureus infections. Am. J. Med. Sci. 330, 102–104.
Locke, J.B., Hilgers, M., Shaw, K.J., 2009b. Novel ribosomal mutations in Staphylococ- Poppe, S., Schaadt, R., Sheehan, D., Sahm, D., Zurenko, G., Shinabarger, D., 2006. Muta-
cus aureus strains identified through selection with the oxazolidinones linezolid tion analysis of Staphylococcus aureus isolates demonstrating trailing linezolid
and torezolid (TR-700). Antimicrob. Agents Chemother. 53, 5265–5274. (LZD) endpoints in microdilution MIC assays. In: Abstr. A-088. 106th American
Locke, J.B., Morales, G., Hilgers, M.G., Rahawi, C.K., Jose Picazo, S., Shaw, J., Stein, Society for Microbiology, Orlando, FL, USA.
K.J.J.L., 2010. Elevated linezolid resistance in clinical cfr-positive Staphylococcus Pournaras, S., Ntokou, E., Zarkotou, O., Ranellou, K., Themeli-Digalaki, K., Stathopou-
aureus isolates is associated with co-occurring mutations in ribosomal protein los, C., Tsakris, A., 2013. Linezolid dependence in Staphylococcus epidermidis
L3. Antimicrob. Agents Chemother. 54, 5352–5355. bloodstream isolates. Emerg. Infect. Dis. 19, 129–132.
Long, K.S., Vester, B., 2012. Resistance to linezolid caused by modifications at its Pringle, M., Poehlsgaard, J., Vester, B., Long, K.S., 2004. Mutations in ribosomal pro-
binding site on the ribosome. Antimicrob. Agents Chemother. 56, 603–612. tein L3 and 23S ribosomal RNA at the peptidyl transferase centre are associated
Long, K.S., Poehlsgaard, J., Kehrenberg, C., Schwarz, S., Vester, B., 2006. The cfr with reduced susceptibility to tiamulin in Brachyspira spp. isolates. Mol. Micro-
rRNA methyltransferase confers resistance to phenicols, lincosamides, oxazo- biol. 54, 1295–1306.
lidinones, pleuromutilins, and streptogramin A antibiotics. Antimicrob. Agents Prokocimer, P., De Anda, C., Fang, E., Mehra, P., Das, A., 2013. Tedizolid phosphate vs
Chemother. 50, 2500–2505. linezolid for treatment of acute bacterial skin and skin structure infections: the
Long, K.S., Munck, C., Andersen, T.M., Schaub, M.A., Hobbie, S.N., Bottger, E.C., ESTABLISH-1 randomized trial. J. Am. Med. Assoc. 309, 559–569.
Vester, B., 2010. Mutations in 23S rRNA at the peptidyl transferase center and Quiles-Melero, I., Gomez-Gil, R., Romero-Gomez, M.P., Sanchez-Diaz, A.M., de Pab-
their relationship to linezolid binding and cross-resistance. Antimicrob. Agents los, M., Garcia-Rodriguez, J., Gutierrez, A., Mingorance, J., 2013. Mechanisms of
Chemother. 54, 4705–4713. linezolid resistance among staphylococci in a tertiary hospital. J. Clin. Microbiol.
Marshall, S.H., Donskey, C.J., Hutton-Thomas, R., Salata, R.A., Rice, L.B., 2002. Gene 51, 998–1001.
dosage and linezolid resistance in Enterococcus faecium and Enterococcus faecalis. Ross, J.E., Anderegg, T.R., Sader, H.S., Fritsche, T.R., Jones, R.N., 2005. Trends in line-
Antimicrob. Agents Chemother. 46, 3334–3336. zolid susceptibility patterns in 2002: report from the worldwide Zyvox Annual
Meka, V.G., Gold, H.S., 2004. Antimicrobial resistance to linezolid. Clin. Infect. Dis. Appraisal of Potency and Spectrum Program. Diagn. Microbiol. Infect. Dis. 52,
39, 1010–1015. 53–58.
Meka, V.G., Pillai, S.K., Sakoulas, G., Wennersten, C., Venkataraman, L., DeGirolami, Ross, J.E., Fritsche, T.R., Sader, H.S., Jones, R.N., 2007a. Oxazolidinone susceptibility
P.C., Eliopoulos, G.M., Moellering Jr., R.C., Gold, H.S., 2004. Linezolid resistance in patterns for 2005: international report from the Zyvox® Annual Appraisal of
sequential Staphylococcus aureus isolates associated with a T2500A mutation in Potency and Spectrum Study. Int. J. Antimicrob. Agents 29, 295–301.
the 23S rRNA gene and loss of a single copy of rRNA. J. Infect. Dis. 190, 311–317. Ross, J.E., Jones, R.N., Hogan, P.A., Sheehan, D.J., 2007b. Initial occurrences of linezolid
Mendes, R.E., Deshpande, L.M., Castanheira, M., DiPersio, J., Saubolle, M.A., Jones, (LZD) resistance (R) from the Zyvox Annual Appraisal of Potency and Spectrum
R.N., 2008. First report of cfr-mediated resistance to linezolid in human staphy- (ZAAPS) Program (Europe, Latin, America, Asia Pacific). In: Abstract C2-865. 47th
lococcal clinical isolates recovered in the United States. Antimicrob. Agents ICAAC, September 17–20, 2007, Chicago, IL, USA.
Chemother. 52, 2244–2246. Ross, J.E., Utsuki, U., Fumiaki, I., Kobayashi, I., Jones, R.N., Hogan, P.A., 2009. Zyvox®
Mendes, R.E., Sanchez, M., Deshpande, L.M., De la Torre, M.A., Jones, R.N., 2009. Detec- Annual Appraisal of Potency and Spectrum (ZAAPS) Program 2008 (Europe, Latin
tion of cfr-carrying Staphylococcus spp. isolates recovered from blood cultures America, Canada, Asia Pacific): linezolid global in vitro susceptibility analyses.
in a Spanish hospital during a Phase III clinical trial of tropical omiganan 1% gel In: Abstracts A-078. 109th ASM, May 17–21, 2009, Philadelphia, PA, USA.
versus 10% povidone iodine. In: Abstract 928. 19th ECCMID, May 16–19, 2009, Rybak, M.J., Cappelletty, D.M., Moldovan, T., Aeschlimann, J.R., Kaatz, G.W., 1998.
Helsinki, Finland. Comparative in vitro activities and postantibiotic effects of the oxazolidi-
Mendes, R.E., Deshpande, L.M., Farrell, D.J., Spanu, T., Fadda, G., Jones, R.N., 2010a. none compounds eperezolid (PNU-100592) and linezolid (PNU-100766) versus
Assessment of linezolid resistance mechanisms among Staphylococcus epi- vancomycin against Staphylococcus aureus, coagulase-negative staphylococci,
dermidis causing bacteraemia in Rome, Italy. J. Antimicrob. Chemother. 65, Enterococcus faecalis, and Enterococcus faecium. Antimicrob. Agents Chemother.
2329–2335. 42, 721–724.
Mendes, R.E., Deshpande, L., Rodriguez-Noriega, E., Ross, J.E., Jones, R.N., Morfin- Rybak, M.J., Hershberger, E., Moldovan, T., Grucz, R.G., 2000. In vitro activities
Otero, R., 2010b. First report of staphylococcal clinical isolates in Mexico with of daptomycin, vancomycin, linezolid, and quinupristin-dalfopristin against
linezolid resistance caused by cfr: evidence of in vivo cfr mobilization. J. Clin. Staphylococci and Enterococci, including vancomycin-intermediate and -
Microbiol. 48, 3041–3043. resistant strains. Antimicrob. Agents Chemother. 44, 1062–1066.
12 R.E. Mendes et al. / Drug Resistance Updates 17 (2014) 1–12

Schwarz, S., Werckenthin, C., Kehrenberg, C., 2000. Identification of a plasmid-borne Wang, Y., Wang, Y., Wu, C.M., Schwarz, S., Shen, Z., Zhang, W., Zhang, Q., Shen, J.Z.,
chloramphenicol-florfenicol resistance gene in Staphylococcus sciuri. Antimi- 2011. Detection of the staphylococcal multiresistance gene cfr in Proteus vulgaris
crob. Agents Chemother. 44, 2530–2533. of food animal origin. J. Antimicrob. Chemother. 66, 2521–2526.
Shaw, K.J., Barbachyn, M.R., 2011. The oxazolidinones: past, present, and future. Ann. Wang, Y., Wang, Y., Schwarz, S., Shen, Z., Zhou, N., Lin, J., Wu, C., Shen, J., 2012a.
N.Y. Acad. Sci. 1241, 48–70. Detection of the staphylococcal multiresistance gene cfr in Macrococcus case-
Shen, J., Wang, Y., Schwarz, S., 2013. Presence and dissemination of the multire- olyticus and Jeotgalicoccus pinnipedialis. J. Antimicrob. Chemother. 67, 1824–
sistance gene cfr in Gram-positive and Gram-negative bacteria. J. Antimicrob. 1827.
Chemother. 68, 1697–1706. Wang, Y., Schwarz, S., Shen, Z., Zhang, W., Qi, J., Liu, Y., He, T., Shen, J., Wu, C., 2012b.
Shinabarger, D.L., Marotti, K.R., Murray, R.W., Lin, A.H., Melchior, E.P., Swaney, Co-location of the multiresistance gene cfr and the novel streptomycin resis-
S.M., Dunyak, D.S., Demyan, W.F., Buysse, J.M., 1997. Mechanism of action of tance gene aadY on a small plasmid in a porcine Bacillus strain. J. Antimicrob.
oxazolidinones: effects of linezolid and eperezolid on translation reactions. Chemother. 67, 1547–1549.
Antimicrob. Agents Chemother. 41, 2132–2136. Wang, Y., Li, D., Song, L., Liu, Y., He, T., Liu, H., Wu, C., Schwarz, S., Shen, J., 2013. First
Shore, A.C., Brennan, O.M., Ehricht, R., Monecke, S., Schwarz, S., Slickers, P., report of the multiresistance gene cfr in Streptococcus suis. Antimicrob. Agents
Coleman, D.C., 2010. Identification and characterization of the multidrug resis- Chemother. 57, 4061–4063.
tance gene cfr in a Panton-Valentine leukocidin-positive sequence type 8 Wilson, P., Andrews, J.A., Charlesworth, R., Walesby, R., Singer, M., Farrell, D.J., Rob-
methicillin-resistant Staphylococcus aureus IVa (USA300) isolate. Antimicrob. bins, M., 2003. Linezolid resistance in clinical isolates of Staphylococcus aureus.
Agents Chemother. 54, 4978–4984. J. Antimicrob. Chemother. 51, 186–188.
Sierra, J.M., Ortega, M., Tarrago, C., Albet, C., Vila, J., Terencio, J., Guglietta, A., 2009. Wise, R., Andrews, J.M., Boswell, F.J., Ashby, J.P., 1998. The in vitro activity of line-
Decreased linezolid uptake in an in vitro-selected linezolid-resistant Staphylo- zolid (U-100766) and tentative breakpoints. J. Antimicrob. Chemother. 42, 721–
coccus epidermidis mutant. J. Antimicrob. Chemother. 64, 990–992. 728.
Sierra, J.M., Camoez, M., Tubau, F., Gasch, O., Pujol, M., Martin, R., Dominguez, M.A., Wolter, N., Smith, A.M., Farrell, D.J., Schaffner, W., Moore, M., Whitney, C.G.,
2013. Low prevalence of Cfr-mediated linezolid resistance among methicillin- Jorgensen, J.H., Klugman, K.P., 2005. Novel mechanism of resistance to oxa-
resistant Staphylococcus aureus in a Spanish hospital: case report on linezolid zolidinones, macrolides, and chloramphenicol in ribosomal protein L4 of the
resistance acquired during linezolid therapy. PLoS ONE 8, e59215. pneumococcus. Antimicrob. Agents Chemother. 49, 3554–3557.
Smith, L.K., Mankin, A.S., 2008. Transcriptional and translational control of the mlr Wong, A., Reddy, S.P., Smyth, D.S., Aguero-Rosenfeld, M.E., Sakoulas, G., Robinson,
operon, which confers resistance to seven classes of protein synthesis inhibitors. D.A., 2010. Polyphyletic emergence of linezolid-resistant staphylococci in the
Antimicrob. Agents Chemother. 52, 1703–1712. United States. Antimicrob. Agents Chemother. 54, 742–748.
Stefani, S., Bongiorno, D., Mongelli, G., Campanile, F., 2010. Linezolid resistance in Worth, S., Erwin, M.E., Jones, R.N., 1996. Quality control guidelines for amoxicillin,
staphylococci. Pharmaceuticals 3, 1–18. amoxicillin-clavulanate, azithromycin, piperacillin-tazobactam, roxithromycin,
Tenover, F.C., Williams, P.P., Stocker, S., Thompson, A., Clark, L.A., Limbago, B., Carey, ticarcillin, ticarcillin-clavulanate, trovafloxacin (CP 99,219), U-100592, and
R.B., Poppe, S.M., Shinabarger, D., McGowan Jr., J.E., 2007. Accuracy of six antimi- U-100766 for various National Committee for Clinical Laboratory Standards sus-
crobial susceptibility methods for testing linezolid against staphylococci and ceptibility testing methods. Results from multicenter trials. Diagn. Microbiol.
enterococci. J. Clin. Microbiol. 45, 2917–2922. Infect. Dis. 24, 87–91.
Toh, S.M., Xiong, L., Arias, C.A., Villegas, M.V., Lolans, K., Quinn, J., Mankin, A.S., 2007. Yan, F., LaMarre, J.M., Rohrich, R., Wiesner, J., Jomaa, H., Mankin, A.S., Fujimori,
Acquisition of a natural resistance gene renders a clinical strain of methicillin- D.G., 2010. RlmN and Cfr are radical SAM enzymes involved in methylation of
resistant Staphylococcus aureus resistant to the synthetic antibiotic linezolid. ribosomal RNA. J. Am. Chem. Soc. 132, 3953–3964.
Mol. Microbiol. 64, 1506–1514. Zhang, W.J., Wu, C.M., Wang, Y., Shen, Z.Q., Dai, L., Han, J., Foley, S.L., Shen, J.Z., Zhang,
Toh, S.M., Xiong, L., Bae, T., Mankin, A.S., 2008. The methyltransferase YfgB/RlmN is Q., 2011. The new genetic environment of cfr on plasmid pBS-02 in a Bacillus
responsible for modification of adenosine 2503 in 23S rRNA. RNA 14, 98–106. strain. J. Antimicrob. Chemother. 66, 1174–1175.
Tsakris, A., Pillai, S.K., Gold, H.S., Thauvin-Eliopoulos, C., Venkataraman, L., Wenner- Zhang, W.J., Xu, X.R., Schwarz, S., Wang, X.M., Dai, L., Zheng, H.J., Liu, S., 2014.
sten, C., Moellering Jr., R.C., Eliopoulos, G.M., 2007. Persistence of rRNA operon Characterization of the IncA/C plasmid pSCEC2 from Escherichia coli of swine
mutated copies and rapid re-emergence of linezolid resistance in Staphylococcus origin that harbours the multiresistance gene cfr. J. Antimicrob. Chemother. 69,
aureus. J. Antimicrob. Chemother. 60, 649–651. 385–389.
Tsiodras, S., Gold, H.S., Sakoulas, G., Eliopoulos, G.M., Wennersten, C., Venkataraman, Zurenko, G.E., Yagi, B.H., Schaadt, R.D., Allison, J.W., Kilburn, J.O., Glickman, S.E.,
L., Moellering, R.C., Ferraro, M.J., 2001. Linezolid resistance in a clinical isolate Hutchinson, D.K., Barbachyn, M.R., Brickner, S.J., 1996. In vitro activities of U-
of Staphylococcus aureus. Lancet 358, 207–208. 100592 and U-100766, novel oxazolidinone antibacterial agents. Antimicrob.
von Eiff, C., Peters, G., 1999. Comparative in-vitro activities of moxifloxacin, Agents Chemother. 40, 839–845.
trovafloxacin, quinupristin/dalfopristin and linezolid against staphylococci. J. Zyvox, 2010. Zyvox Package Insert, available at http://www.pfizer.com/products/
Antimicrob. Chemother. 43, 569–573. rx/rx product zyvox.jsp (accessed January 2013).

You might also like