Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Acta Pdiatrica ISSN 0803-5253

REVIEW ARTICLE

Treating paediatric community-acquired pneumonia in the era


of antimicrobial resistance
L Galli, C Montagnani, E Chiappini (elena.chiappini@unifi.it), M de Martino
Department of Health Sciences, University of Florence, Anna Meyer Childrens University-Hospital, Florence, Italy

Keywords
Antimicrobial resistance, Children, Communityacquired pneumonia, Drug-resistant bacteria,
Paediatricians
Correspondence
Elena Chiappini, MD, PhD, Department of Health
Sciences, University of Florence, Anna Meyer
Childrens University Hospital, Viale Pieraccini 24,
I-50139 Florence, Italy.
Tel: +39 055 5662518 |
Fax: +39 055 4221012 |
Email: elena.chiappini@unifi.it

ABSTRACT
Increasing levels of paediatric community-acquired pneumonia (CAP), caused by drugresistant bacteria and antimicrobial resistance, vary with age and countries and, in some
cases, serotypes. When empirical first-line treatment administration fails, paediatricians
should consider second-line treatments based on the prevalence of local resistance. A
more judicious use of antimicrobial agents is also required.
Conclusion: Knowledge of local epidemiology and an appropriate use of antimicrobial
drugs are necessary to treat CAP in this era of antimicrobial resistance.

DOI:10.1111/apa.12503

According to data from the World Health Organization,


pneumonia is a major health problem and is still the main
cause of global child mortality (1).
Despite improvements in diagnostic methods, identifying
the aetiology of community-acquired pneumonia (CAP) in
children is still challenging. Causative pathogens can be
identified in 5481% of cases of CAP (2). Moreover, the
epidemiology of CAP has been changed in the last few
decades as a result of the introduction of the pneumococcal
conjugate and Haemophilus influenzae type B vaccines.
In industrialised countries, viruses account for up to
83% of CAP in children, and the human metapneumovirus
and bocavirus have been described in an increasing
number of cases in recent years (3,4). After the introduction of the pneumococcal conjugate vaccine, Elemraid
et al. (2) detected bacteria in 17.5% of children with CAP,
whereas viruses, either alone or as co-pathogens, were
found in 43%.

Key notes


This review provides an overview on drug resistance


mechanisms and debates therapy options for community-acquired pneumonia (CAP) due to drug-resistant
Streptococcus pneumoniae, Staphylococcus aureus
and Mycoplasma pneumoniae.
It shows that CAP, caused by drug-resistant bacteria and
antimicrobial resistance, varies with age and countries
and, in some cases, serotypes.
Knowledge of local epidemiology and an appropriate
use of antimicrobial drugs are necessary to treat CAP in
this era of antimicrobial resistance.

However, bacterial aetiology plays a significant role in


children with severe pneumonia and even more in fatal
cases (5).
Antimicrobial resistance is an increasing problem worldwide. The impact of antibiotic resistance is more consistent
in hospital settings, where multidrug-resistant and extensive
drug-resistant bacteria are spreading. However, there is
growing evidence of an increasing proportion of CAP cases
being caused by drug-resistant bacteria in the last 10 years,
both in adults and in children (68). The proportion of
isolates resistant to antimicrobials is increasing among
Streptococcus pneumoniae, Staphylococcus aureus and
Mycoplasma pneumoniae, and treatment of infections due
to these pathogens can be a challenge for paediatricians. A
possible role for nontypeable Haemophilus influenzae in
paediatric CAP has been suggested (9), but further studies
are necessary to determine whether it can play a significant
clinical role in CAP in children.
Hence, the aims of this review were to analyse the drug
resistance mechanisms of Streptococcus pneumoniae,
Staphylococcus aureus and Mycoplasma pneumoniae and
discuss therapy options for CAP due to drug-resistant
pathogens.

STREPTOCOCCUS PNEUMONIAE
Antibiotic resistance among Streptococcus pneumoniae
isolates, the major cause of CAP in children, has been
widely documented worldwide. To date, 93 different
Streptococcus pneumoniae serotypes have been identified.
Serotype distribution varies widely by geographical region,
pneumococcal vaccination coverage and age. In the 2000s,
many resource-rich countries introduced the seven-valent

2013 Foundation Acta Pdiatrica. Published by John Wiley & Sons Ltd 2013 102 (Suppl. 465), pp. 2533

25

Treating community-acquired pneumonia

Galli et al.

conjugated vaccine (PC7), containing 4, 6B, 9V, 14, 18C,


19F and 23F serotypes, and this was associated with a
reduction in invasive pneumococcal diseases, particularly
in younger children (1012). PCV7 showed good effectiveness (30%; 95% CI: 10.745.7) in preventing pneumonia in
children under 5 years of age (13), but other studies
reported that PCV7 had no significant efficacy when it
came to preventing complicated pneumonia (14). Of note,
several reports documented an increase in hospitalisations
for complicated pneumonia, particularly for empyema, after
the PCV7 was introduced (15). The reasons for this increase
are not completely understood. A role for ibuprofen has
also been suggested (16), as well as replacing the PCV7
serotypes with other more virulent serotypes, such as 19A,
7, 1, 3 (1517). A further reason for this increase may be the
increasing proportion of antimicrobial-resistant pneumococcal isolates in the 2000s (18,19). A large number of
studies documented growing resistance to b-lactams, macrolides and tetracyclines and found an association between
specific serogroups/serotypes and resistance patterns
(Fig. 1). For example, in a study on multidrug resistance,
Lismond et al. (20) stated that the 19A serotype showed a
73.5% resistance to clarithromycin, about 20% resistance to
amoxicillin, cefuroxime and ceftriaxone, but only 3%
resistance to penicillin G. Meanwhile, other authors have
described an additional 30% resistance to meropenem (21).
The PCV7 vaccine was recently replaced by the PCV13, and
this should be able to reduce the spread of these multidrug18

Dual non-sucepblility

16

Single macrolides nonsuscepble

14

Single penicillin nonsuscepble

Proporon (%)

12

Fully suscepble

10
8
6
4
2

rs
he

15

ot

Serogroups

23

14

22

12

19

Figure 1 Proportion of susceptible pneumococcal isolates in Europe by


serogroups. Data from the Annual Report of the European Antimicrobial
Resistance Surveillance Network (16).

26

resistant serotypes among children. However, a further


cause of the increase in pneumococcal isolates resistant to
b-lactams and macrolides is antibiotic consumption at a
national level. Data from the European Antimicrobial
Resistance Surveillance System clearly show that antibiotic
resistance in pneumococcal isolates has increased in those
European countries where consumption of b-lactams and
macrolides is higher (22).
Minimum inhibitory concentration (MIC) breakpoints
for defining clinical susceptibility of Streptococcus pneumoniae to b-lactams have been suggested by the European
Committee on Antibiotic Susceptibility Testing (EUCAST):
susceptible is quantified as 0.5 mg/L and resistant is
quantified as >2 mg/L. Antimicrobial susceptibility has
been differently defined by the United States Clinical and
Laboratory Standards Institute (CLSI). Lack of susceptibility to penicillin and other b-lactams is, in fact, associated
with the modification of pneumococcal cell wall penicillinbinding proteins. This results in decreased, but not absent,
affinity with these antibiotics. Therefore, higher b-lactam
concentrations resulting from higher dosages and good
tissue distribution, as in the lung but not in the central
nervous system, may overcome resistance. In 2008, the
CLSI changed its MIC breakpoints for parenterally administered penicillin (2 mg/L: susceptible; 8 mg/L resistant),
after studies reported the clinical effectiveness of penicillin
and aminopenicillins when treating pneumococcal diseases
other than meningitis, even if classified as resistant in vitro
(23). Breakpoints to define nonsusceptibility to b-lactams in
pneumococcal meningitis remain unchanged (0.5 mg/L).
As a consequence, a high proportion of pneumococcal
isolates from CAP, categorised as intermediate to b-lactams
using the EUCAST breakpoint, may be considered susceptible according to CLSI criteria. In clinical practice,
high-dose penicillin or high-dose amoxicillin can be still
considered in the empirical treatment of pneumococcal
CAP, even in those countries where penicillin nonsusceptible isolates are above 25% (22).
Pneumococcal resistance to macrolide antibiotics
(clarithromicin, erythromicin and azithromicin) is the result
of a mutation on the mef (A) gene, whereas a mutation on
the erm (B) gene results in resistance to both macrolides
and lincosamides (24). As the mutations modify the 23S
subunit of the ribosomal RNA, blocking macrolide binding
to the ribosome, differently to resistance to b-lactams,
macrolide resistance cannot be overcome using higher
macrolide dosages. Furthermore, there have been several
reports of clinical failure of macrolide treatment for CAP
when macrolide-resistant (MIC 1 mg/L) Streptococcus
pneumoniae is isolated (25). Therefore, empirical macrolide
monotherapy treatment for CAP in children should be
avoided, and macrolide treatment should be used when
there is a reasonable clinical suspicion of Mycoplasma
pneumoniae aetiology.
In conclusion, studies from United States isolates suggest
that, at present, Streptococcus pneumoniae has the highest
susceptibility to ampicillin, piperacillin, respiratory fluoroquinolones (levofloxacin and moxifloxacin), ertapenem,

2013 Foundation Acta Pdiatrica. Published by John Wiley & Sons Ltd 2013 102 (Suppl. 465), pp. 2533

Galli et al.

linezolid and vancomycin (>99% susceptibility), followed


by penicillin G, ceftriaxone, cefotaxime and cefepime (85
92% susceptibility) and meropenem, imipenem and clindamycin (7081% susceptibility) (6). Only 6065% of isolates
are susceptible to macrolides.

STAPHYLOCOCCUS AUREUS
Methicillin-resistant Staphylococcus aureus (MRSA) is an
increasing cause of CAP in children, mainly associated with
severe infections. MRSA was first reported in 1961, soon
after the clinical introduction of methicillin, and in the
1980s, these strains began to spread worldwide as nosocomial pathogens.
Methicillin resistance is due to the acquisition of the
staphylococcal cassette chromosome mec (SCCmec), which
carries the mec gene for methicillin and resistance to all
other b-lactam drugs (7).
Over the last decade, the epidemiology of MRSA has
been evolving rapidly, with the diffusion of communityacquired MRSA (CA-MRSA) strains, which have different
genetic backgrounds to the healthcare-associated (HA)
strains and are more related to CA methicillin-sensitive
SA (CA-MSSA). CA-MRSA carries smaller SCCmec elements than HA-MRSA, generally type IV, V and VII (26).
Several sequence types of CA-MRSA have been described
worldwide, and their prevalence varies widely from area to
area. USA300/ST8 has become the dominant clone in USA,
whereas ST80 clone is prevalent in Europe, ST59 in Taiwan
and ST93 in Australia (27,28).
In addition, SA strains producing Panton-Valentine
leukocidin (PVL) cause severe necrotising pneumonia in
young healthy people. PVL is a bicomponent toxin that
causes the lysis of leucocytes, forming a pore in their
membrane and representing a main virulence factor of SA,
independent of methicillin resistance (29).
Community-acquired MRSA is typically susceptible to
most non-b-lactam drugs, such as trimethoprim-sulfamethoxazole, clindamycin and tetracycline, even if the ST80
clone is variably resistant to fluoroquinolones. Strains
resistant to erythromycin, and susceptible to clindamycin,
have to be tested for inducible clindamycin resistance (30).
Community-acquired MRSA pneumonia is becoming a
major concern in United States, Taiwan, Canada and
Australia, but there are also increasing reports of CAMRSA and PVL-SA pneumonia in Europe (31). A study
conducted at Texas Childrens Hospital over a 7-year period
showed an increasing rate of SA pneumonia, from 4.81
hospitalisations per 10 000 admissions in 2001 to 9.75 per
10 000 admissions in 2009. MRSA was responsible for 74%
of infections. More than 90% of the MRSA and MSSA
isolates carried PVL genes (32). In Taiwan, more than 50%
of staphylococcal infections in children are caused by
MRSA and these are mainly PVL carried (33). CA-MRSA
and PVL-SA are frequently associated with necrotising
pneumonia and typically affect previously healthy young
people who dont carry any risk factors because of their
exposure to the healthcare system (29). Schultz et al. (34)

Treating community-acquired pneumonia

reported that CA-MRSA has replaced Streptococcus pneumoniae as a more frequent pathogen isolated from children
with complicated pneumonia. Moreover, in a recent series
of 41 French paediatric patients with necrotising pneumonia, PVL-SA was the most commonly isolated pathogen
(35).
Community-acquired MRSA CAP has a rapidly progressive course and a 5663% mortality rate (30). Therefore, as
we recently reported (36), MRSA and PVL-SA infections
should be suspected in all children with severe acquired
pneumonia, characterised by leukopenia, necrotising or
cavitary infiltrates, haemoptysis or empyema, previous
history or family history of recurrent furuncles or skin
abscesses (37), independent of culture results and finally by
intensive care unit admission (31,38).

MYCOPLASMA PNEUMONIAE
Mycoplasma pneumoniae is a common cause of CAP in
children aged five to 14 years, accounting for up to 40% of
bacterial forms (39). In most of cases, infections are mild to
moderate, and signs and symptoms tend to resolve spontaneously without specific therapy.
Due to the absence of a cell wall, Mycoplasma pneumoniae is not sensitive to b-lactams. Macrolides, tetracyclines
and fluoroquinolones are indicated, but only macrolides are
recommended in children (39).
Since 2000, the prevalence of macrolide resistance
Mycoplasma pneumonia (MRMP) has been described with
increasing incidence worldwide. MRMP has become widespread in far East Asia, with up to 90% of Mycoplasma
pneumonia strains being resistant in China (40). Recent
reports have also documented the emergence of MRMP in
the West, but still with lower prevalence: 8.2% in United
States, 0.92.9% in Denmark, 3% in Germany, 10% in
France and 32% in Israel (40,41). Chironna et al. (42)
found that 26% of strains were resistant during a paediatric
outbreak in southern Italy in 2010. Moreover, macrolide
resistance has been always shown to be more frequent in
children than in adults (39).
Macrolide resistance is strongly associated with a specific
point mutation in the V domain of the 23S rRNA gene (39).
A-to-G transition at position 2063 and A-to-C transition at
position 2064 induce a high level of macrolide resistance,
whereas C-to-A or G transition at position 2617 and A-to-T
transition at position 2063 induces a low-level resistance
(43). Some studies have found that macrolide resistance can
be directly induced during the course of infection by the use
of macrolides, even when proper dosages are administered
(42,44). As the Mycoplasma cultures are often difficult to
obtain, several studies on MRMP are based on molecular
diagnosis of resistance. Therefore, the real antimicrobial
pattern of susceptibility is unknown. However, all the
genetically determined MRMP seem to have high resistance
to macrolides, with MICs 16 lg/mL (42,45).
The presence of macrolide resistance does not seem to
influence the presentation of CAP in terms of symptoms,
laboratory results and radiographic findings, but it is

2013 Foundation Acta Pdiatrica. Published by John Wiley & Sons Ltd 2013 102 (Suppl. 465), pp. 2533

27

Treating community-acquired pneumonia

Galli et al.

associated with longer duration of signs and symptoms,


hospitalisation and antibiotic treatment (46). Kawai et al.
(47) reported that microbiological and clinical efficacy of
macrolides were low in patients with MRMP infections.
On the other hand, some authors have reported good
outcomes when children with MRMP pneumonia are
treated with macrolides (48), and they suggest that this is
as a result of the anti-inflammatory activity of macrolides.
As a matter of fact, severe lung disease due to Mycoplasma pneumoniae was presumed to be determined by
overproduction of cytokines and excessive cell-mediated
immune response. Therefore, many investigators suggested
that these cases could benefit from combined therapy with
antimicrobial agents and immune modulators (49). Prednisolone (1 mg/kg/day for 37 days, tapering within
1 week), methylprednisone (2 mg/kg/day or 2030 mg/kg
for 3 days, tapering within 1 month) and intravenous
immunoglobulins (1 g/kg/day, one or two doses) have
therefore been successfully used (49,50).
As a result, even if evidence of a relationship between
lack of clinical efficacy of macrolides and the presence of
MRMP is controversial, molecular characterisation of
Mycoplasma pneumoniae can be useful in all patients with
severe CAP who fail to respond to macrolides.
However, given that Mycoplasma pneumoniae infection
is typically mild and self-limited, the opportunity to provide
alternative therapy in MRMP infections requires careful
evaluation and has to be limited to severe CAP.

ANTIMICROBIAL TREATMENT IN DRUG-RESISTANT CAP


Recommendations from international guidelines on the
treatment of CAP are reported in the systematic review by
Berti et al. in this supplement. Guideline recommendations
are controversial when a penicillin-resistant Streptococcus
pneumoniae is suspected or confirmed. If Streptococcus
pneumoniae isolates have MICs to penicillin 4.0 lg/mL,
the 2011 United States guidelines recommend ceftriaxone
or alternatively ampicillin or levofloxacin or linezolid (51).
Clindamycin or vancomycin may be also prescribed. Oral
levofloxacin or oral linezolid is suggested as a step-down
therapy or for mild infection. Because of limited evidence
on the relationship between penicillin nonsusceptibility and
worse clinical outcome, the 2011 UK guidelines still suggest
oral amoxicillin as the first choice of antibiotic therapy for
all children, alternatively suggesting amoxicillin/clavulanate, cefaclor, erythromycin, azithromycin and clarithromycin (52).
Despite conflicting recommendations by the guidelines
on second-line treatments, all guidelines, including a review
by the European Society of Paediatric Infectious Diseases
(53), suggest oral high-dose amoxicillin (90 mg/kg/day) as
the first-line antimicrobial drug in the empirical treatment
of noncomplicated pneumococcal CAP, even in those
countries where penicillin nonsusceptibility among pneumococcal isolates is increasing.
In our opinion, when parenteral treatment is required
and/or drug-resistant Streptococcus pneumoniae (MICs to

28

penicillin 4.0 lg/mL) is documented, high-dose ampicillin


(300400 mg/kg/die), ceftriaxone (100 mg/kg/day), vancomycin (60 mg/kg/day) or clindamycin are alternatives. In
selected cases, linezolid or levofloxacin is possible alternatives and can also be used for oral step-down treatment.
This approach is quite similar to that suggested in the
United States guidelines (51).
Vancomycin, linezolid and clindamycin are the first-line
antibiotics to treat suspected or confirmed MRSA pneumonia (31,38,51) and should be administered for at least
14 days (51).
Vancomycin is a glycopeptide agent that inhibits cell
wall peptidoglycan synthesis. It has slow bactericidal
activity against gram-positive pathogens. Vancomycin is
associated with nephrotoxicity and ototoxicity, other than
the well known red man syndrome. The pharmacokinetic
and pharmacodynamic literature on vancomycin use in
children is limited. The suggested dose is 4060 mg/kg/
day, divided into 68 h doses (38,51) (Table 1). Therapeutic range monitoring is recommended, with a target
trough serum concentration of 1520 lg/mL or an AUC/
MIC 400 lg/mL (30). Maintaining a trough serum
concentration of 1520 lg/mL does not seem to be
associated with an increased rate of nephrotoxicity (54).
A recent study suggested that vancomycin doses of
70 mg/kg/day are necessary in children <12 years old
with normal renal function and 60 mg/kg/day in children
12 years old. Resistance of SA to vancomycin remains
rare, but when isolates have an MIC of >2 lg/mL, or
doses above 80 mg/kg/day are required to obtain an
AUC/MIC >400, alternative antibiotics to treat MRSA
should be used (55).
Teicoplanin, another member of glycopeptides family, is
widely used in Europe and Japan, but is not approved for
use in the United States (56). It provides bactericidal
activity against gram-positive infections, with similar clinical efficacy to vancomycin (57). However, teicoplanin
seems to be associated with a lower adverse event rate,
except for thrombocytopenia (58,59). Teicoplanin presents
a long elimination half-life, which means that once daily
administration is possible. However, because it takes a long
time to achieve an optimal plasma concentration, an initial
loading-dose regimen has been recommended (56). A
teicoplanin MIC value >1.52 mg/L is associated with an
increased risk of treatment failure among patients with
MRSA pneumonia and bacteraemia (60,61). Teicoplanin
trough concentration >10 lg/mL has been considered
appropriate, but more recent data have encouraged experts
to recommend 1530 lg/mL as the new target trough
concentration (62). Ito et al. (56) found that adequate
serum trough concentrations are rarely reached in children
treated with standard doses. Matsumoto et al. suggested
that an initial loading dose of 1115 mg/kg every 12 h for
three doses is necessary to reach the suggested trough
concentration in critically ill patients. Moreover, toddlers
have a high risk of presenting low trough concentration,
even with three loading dose of 1015 mg/kg (59)
(Table 1).

2013 Foundation Acta Pdiatrica. Published by John Wiley & Sons Ltd 2013 102 (Suppl. 465), pp. 2533

Galli et al.

Treating community-acquired pneumonia

Table 1 Antibiotics used in drug-resistant community-acquired pneumonia


Antibiotics

Dose

Route

Adverse effects

Comments

Species

Vancomycin

15 mg/kg every 68 h

IV

Nephrotoxicity, ototoxicity, red


man syndrome

Slowly bactericidal. Therapeutic


levels to be monitored

Teicoplanin

10 mg/kg every 12 h for three


doses then 10 mg/kg/day
10 mg/kg (max 600 mg) every 8
<12 years; every 12 h
>12 years
610 mg/kg (max 1.2 g) every
6h
10 mg/kg (max 600 mg) every
12 h

IV/IM

Nephrotoxicity, ototoxicity,
thrombocytopenia
Lactic acidosis, myelosuppression,
peripheral and optic neuropathy

Bactericidal. Therapeutic levels to


be monitored
Bacteriostatic. Inhibit toxin
synthesis
Treatment duration <28 days
Bacteriostatic. Inhibit toxin
synthesis
Bactericidal. Colours biological
fluids red. Rapid development of
resistance in monotherapy
Bactericidal. Not approved for
children
Bactericidal. Not approved for
children
Bacteriostatic. Not approved for
children

Streptococcus
pneumoniae
Staphylococcus
aureus
S. pneumoniae
S. aureus
S. pneumoniae
S. aureus

Linezolid

Clindamycin
Rifampicin

IV/PO

IV/PO

Diarrhoea, C. difficile colitis

IV/PO

Hepatotoxicity, anaphylaxis
agranulocytosis
Nausea, vomiting, taste sense
disorders, nephrotoxicity
Rash

Telavancin

10 mg/kg/day

IV

Ceftaroline

600 mg every 12 h

IV

Tigecycline

2 mg/kg stat, then 1 mg/kg

IV

Doxycycline

12 mg/kg (max 200 mg) every


12 h
4 mg/kg stat (max 200 mg) then
2 mg/kg (max 100 mg)

PO/IV

Minocycline

Ciprofloxacin
Levofloxacin
Moxifloxacin

1015 mg/kg every 12 h PO,


every 812 h IV
8 mg/kg (max 500 mg) every
12 h
10 mg/kg/day (max 400 mg)

PO

IV/PO
IV/PO
IV/PO

Diarrhoea, nausea, vomiting,


headache, raised ALT/AST,
pancreatitis
Diarrhoea, Hepatotoxicity, tooth
discolouration in children
Dizziness, headache,
hepatotoxicity, diarrhoea, tooth
discolouration in children
Rupture of tendon. Cartilage
damage reported in animals
Rupture of tendon. Cartilage
damage reported in animals
Rupture of tendon. Cartilage
damage reported in animals

Telavancin is a new glycopeptide approved in Europe for


hospital-acquired pneumonia (30). It presents rapid bactericidal activity against gram-positive pathogens, and once
daily administration is possible, due to its long half-life (63)
(Table 1). It has been shown to have potent activity against
MRSA, but its safety and effectiveness have not been
established in children (64).
Linezolid is the first member of the oxazolidinone family.
It provides bacteriostatic activity against gram-positive
pathogens, including MRSA, and inhibits toxin synthesis.
It inhibits protein synthesis by preventing binding of the
30S and 50S ribosomal subunits (58). Because of this
ability, linezolid is also recommended for PVL-SA CAP.
The recommended paediatric dose is 10 mg/kg every 8 h in
children <12 years old and 10 mg/kg every 12 h in children
12 years old (Table 1). Due to its optimal oral bioavailability, an early switch from intravenous to oral administration is possible (65).
Linezolid is generally safe and well tolerated in children.
Adverse events have been associated with treatment duration
>28 days. The most common adverse events are headache,
diarrhoea and nausea. Lactic acidosis, myelosuppression,

S. pneumoniae
S. aureus
S. aureus

S. pneumoniae
S. aureus
S. pneumoniae
S. aureus
S. pneumoniae
S. aureus

Bacteriostatic. Not recommended


<8 years
Bacteriostatic. Not recommended
<8 years

Mycoplasma
pneumoniae
M. pneumoniae

Bactericidal. Not recommended for


children
Bactericidal. Not recommended for
children
Bactericidal. Not recommended
for children

S. pneumoniae
M. pnuemoniae
S. pneumoniae
M. pnuemoniae
S. pneumoniae
M. pnuemoniae

peripheral and optic neuropathy have also been reported and


need careful monitoring (65).
Wunderink et al. (66) reported that the clinical outcomes
of adult patients with hospital-acquired pneumonia were
significantly better with linezolid than with adjusted-dose
vancomycin, even if 60-day mortality was similar. However,
several meta-analyses have found that linezolid is superior
to glycopeptides for treating hospital-acquired pneumonia.
Similar clinical cure, microbiological eradication and
adverse events rates were found in both therapeutic groups
(6770). Moreover, vancomycin is considered to have a
poor lung penetration, whereas linezolid achieves high
concentration in lung tissue. However, clinical significance
of different levels of pulmonary penetration requires further
clinical studies (71).
Linezolid is an expensive drug, but the early switch from
endovenous to oral therapy could be a cost saving (72).
Cost analysis studies have produced conflicting results (73).
Clindamycin is an effective agent for susceptible MRSA
and Streptococcus pneumoniae in the absence of bacteremia
or intravascular infection (38,51) (Table 1). It inhibits
protein synthesis by blocking the peptidyltransferase

2013 Foundation Acta Pdiatrica. Published by John Wiley & Sons Ltd 2013 102 (Suppl. 465), pp. 2533

29

Treating community-acquired pneumonia

Galli et al.

reaction on the 50S ribosomal subunits (74). It should be


used alone, or in combination with another anti-MRSA
antibiotic, in case of PVL-SA pneumonia, because of its
ability to inhibit toxin synthesis (30). However, it is not
recommended that it is used alone in the case of bacteraemia.
Inducible resistance to clindamycin may be present in the
case of SA pneumonia, and therefore, a D-test should be
performed (51). The most frequent adverse event is diarrhoea, which can be associated with Clostridium difficile
colitis (30).
Rifampicin is often used as an adjunct to vancomycin
therapy in the case of MRSA pneumonia, even if limited
evidence exists to support this practice (75). However, it has
a role in PVL-SA pneumonia, as it inhibits protein synthesis. Rifampicin should not be used alone, because of the risk
of developing resistance (29).
Ceftaroline fosamil is a fifth-generation cephalosporin
with bactericidal activity against gram-positive and gramnegative pathogens. Ceftaroline fosamil is shown to be
active against the resistant gram-positive pathogens
involved in CAP, such as MRSA and MDR Streptococcus
pneumoniae (76). It is generally well tolerated, with an
adverse events profile similar to other cephalosporins (77).
It has been approved for the treatment for CAP in adults,
but it is not recommended in children (77) (Table 1).
Tigecycline is the first member of glycylcyclines, derived by
minocycline (78). It provides bacteriostatic activity against
gram-positive and gram-negative pathogens, including MRSA
and penicillin-resistant Streptococcus pneumoniae. Tigecycline also seems to be active against Mycoplasma pneumoniae, and due to its ability to reduce protein synthesis, it is
effective against PVL-SA (79). It was recently approved in
United States for the treatment for CAP, but there are still no
data to support its use for MRSA pneumonia (30) and it is not
recommended for use in children. Even so, Purdy et al. (80)
reported a dosage of 1.2 mg/kg every 12 h as the most
appropriate dose for children aged eight to 11 years.
However, recent meta-analyses of clinical trials involving
tigecycline monotherapy in adults reported an increasing
mortality rate with this drug (81).
Most studies have shown that fluoroquinolones and
tetracyclines have excellent in vitro antimycoplasma activity in children with MRMP pneumonia (43,45). Several
studies have also reported that children with severe MRMP
CAP have been successfully treated with ciprofloxacin,
levofloxacin, moxifloxacin, minocycline and doxycycline
(40,46,49,82) (Table 1).
However, tetracyclines are not approved for use in
children under 8 years old due to the potential for tooth
discoloration, and fluoroquinolones are not recommended
in children under the age of 18 because of the risk of
cartilaginous abnormalities reported in animal models.
However, they have been successfully prescribed for severe
infection in children, and they do not appear to be
associated with arthropathy (49).
Minocycline and tosufloxacin have been approved in
Japan to treat suspected MRMP pneumonia in children, but
not in Europe or the United States (82).

30

Recent reports suggested that tetracyclines are significantly more effective than fluoroquinolones in treating
children with MRMP pneumonia. Minocycline and doxycycline achieve better clinical improvements and decrease
the Mycoplasma pneumoniae DNA copy number more
effectively than tosufloxacin (41,82,83). Therefore, minocycline and doxycycline are the drugs of choice for MRMP
in children over 8 years old, and levofloxacin, moxifloxacin
or tosufloxacin could be an alternative in adolescents with
skeletal maturity. In children aged <8 years old, tetracyclines and fluoroquinolones should only be used in severe
CAP that does not respond to macrolide treatment, after an
accurate assessment of risk-benefit and for the shortest
possible period. This is because of the possible spontaneous
healing of Mycoplasma pneumonia and the risk of adverse
events related to these drugs.

CONCLUSIONS
Antimicrobial resistance is an increasing problem in the
treatment of CAP, particularly in a paediatric age when
there are limited therapeutic options. Preventing further
increases in resistance is the better strategy, and paediatricians should follow treatment guidelines and recommendations on the judicious use of antimicrobial agents. In
particular, they should avoid using antibiotics in common
respiratory infections, mostly those of viral aetiology.
Surveillance on the prevalence of antimicrobial resistance
at a country level should be implemented. The results
should be disseminated, and educational programmes and
guidelines should be updated in the light of local resistance
patterns. An appropriate therapy should be chosen, based
on knowledge of local epidemiology, an accurate evaluation
of CAP severity and of the risk-benefit ratio, as off-label and
expensive drugs are often required. Efforts should also be
made to obtain a microbiological diagnosis, to limit the use
of antibiotics to bacterial CAP and prevent prescribing
broad-spectrum antibiotics.

CONFLICT OF INTERESTS
The authors have no conflict of interest to declare.

References
1. Liu L, Johnson HL, Cousens S, Perin J, Scott S, Lawn JE, et al.
Global, regional, and national causes of child mortality: an
updated systematic analysis for 2010 with time trends since
2000. Lancet 2012; 379: 215161.
2. Elemraid MA, Sails AD, Eltringham GJ, Perry JD, Rushton
SP, Spencer DA, et al. Aetiology of paediatric pneumonia after
the introduction of pneumococcal conjugate vaccine. Eur
Respir J 2013; Apr 18. DOI: 10.1183/09031936.00199112.
3. Mameli C, Zuccotti GV. The impact of viral infections in
children with community-acquired pneumonia. Curr Infect Dis
Rep 2013; 15: 197202.
4. Edwards KM, Zhu Y, Griffin MR, Weinberg GA, Hall CB,
Szilagyi PG, et al. Burden of human metapneumovirus
infection in young children. N Engl J Med 2013; 368: 63343.

2013 Foundation Acta Pdiatrica. Published by John Wiley & Sons Ltd 2013 102 (Suppl. 465), pp. 2533

Galli et al.

5. Rudan I, OBrien KL, Nair H, Liu L, Theodoratou E, Qazi S,


et al. Epidemiology and etiology of childhood pneumonia in
2010: estimates of incidence, severe morbidity, mortality,
underlying risk factors and causative pathogens for 192
countries. J Glob Health 2013; 3: 10401.
6. Jinno S, Jacobs MR. Pneumonia due to drug-resistant
Streptococcus pneumoniae. Curr Infect Dis Rep 2012; 14: 2929.
7. DeLeo FR, Otto M, Kreiswirth BN, Chambers HF.
Community-associated meticillin-resistant Staphylococcus
aureus. Lancet 2010; 375: 155768.
8. Jones RN, Sader HS, Moet GJ, Farrell DJ. Declining
antimicrobial susceptibility of Streptococcus pneumoniae in the
United States: report from the SENTRY Antimicrobial
Surveillance Program (19982009). Diagn Microbiol Infect Dis
2010; 68: 3346.
9. Morozumi M, Chiba N, Okada T, Sakata H, Matsubara K,
Iwata S, et al. Antibiotic susceptibility in relation to genotype of
Streptococcus pneumonia, Haemophilus influenza, and
Mycoplasma pneumoniae responsible for community-acquired
pneumonia in children. J Infect Chemother 2013; 19: 43240.
10. Whitney CG, Pilishvili T, Farley MM, Schaffner W, Craig AS,
Lynfield R, et al. Effectiveness of seven-valent pneumococcal
conjugate vaccine against invasive pneumococcal disease: a
matched case-control study. Lancet 2006; 368: 1495502.
11. Centers for Disease Control and Prevention (CDC). Invasive
pneumococcal disease in children 5 years after conjugate
vaccine introductioneight states, 19982005. MMWR Morb
Mortal Wkly Rep 2008; 57: 1448.
12. Chapman KE, Wilson D, Gorton R. Serotype dynamics of
invasive pneumococcal disease post-PCV7 and pre-PCV13
introduction in North East England. Epidemiol Infect 2012; 8:
19.
13. Hansen J, Black S, Shinefield H, Cherian T, Benson J, Fireman
B, et al. Effectiveness of heptavalent pneumococcal conjugate
vaccine in children younger than 5 years of age for prevention
of pneumonia: updated analysis using World Health
Organization standardized interpretation of chest radiographs.
Pediatr Infect Dis J 2006; 25: 77981.
14. Cutts FT, Zaman SM, Enwere G, Jaffar S, Levine OS, Okoko
JB, et al. Efficacy of nine-valent pneumococcal conjugate
vaccine against pneumonia and invasive pneumococcal disease
in The Gambia: randomised, double-blind, placebo-controlled
trial. Lancet 2005; 365: 113946.
15. Strachan RE, Snelling TL, Jaffe A. Increased paediatric
hospitalizations for empyema in Australia after introduction of
the 7-valent pneumococcal conjugate vaccine. Bull World
Health Organ 2013; 91: 16773.
16. Byington CL, Spencer LY, Johnson TA, Pavia AT, Allen D,
Mason EO, et al. An epidemiological investigation of a
sustained high rate of paediatric parapneumonic empyema:
risk factors and microbiological associations. Clin Infect Dis
2002; 34: 43440.
17. Jacobs MR, Good CE, Bajaksouzian S, Windau AR. Emergence
of Streptococcus pneumoniae serotypes 19A, 6C, and 22F and
serogroup 15 in Cleveland, Ohio, in relation to introduction of
the protein-conjugated pneumococcal vaccine. Clin Infect Dis
2008; 47: 138895.
18. Hausdorff WP, Feikin DR, Klugman KP. Epidemiological
differences among pneumococcal serotypes. Lancet Infect Dis
2005; 5: 8393.
n M, Gallego M, Belmonte Y, Fontanals D, Valles J, Lisboa
19. Luja
T, et al. Influence of pneumococcal serotype group on outcome
in adults with bacteraemic pneumonia. Eur Respir J 2010; 36:
10739.
20. Lismond A, Carbonnelle S, Verhaegen J, Schatt P, De Bel A,
Jordens P, et al. Antimicrobial susceptibility of Streptococcus

Treating community-acquired pneumonia

21.

22.

23.

24.

25.

26.
27.

28.

29.

30.

31.

32.

33.

34.

35.

36.

pneumoniae isolates from vaccinated and non-vaccinated


patients with a clinically confirmed diagnosis of
community-acquired pneumonia in Belgium. Int J Antimicrob
Agents 2012; 39: 20816.
rquez A, Richardson V, Ortiz-Orozco O,
Bautista-Ma
Luna-Cruz ME, Carnalla-Barajas MN, Echaniz-Avil
es G, et al.
Prevalence of pneumococcal disease, serotype distribution, and
antimicrobial susceptibility in Mexican children younger than
5 years of age. Arch Med Res 2013; 44: 14250.
European Centre for Disease Prevention and Control.
Antimicrobial resistance surveillance in Europe 2011. Annual
Report of the European Antimicrobial Resistance Surveillance
Network (EARS-Net). doi 10.2900/6551.
Clinical and Laboratory Standards Institute. Performance
standards for antimicrobial susceptibility testing: twenty-first
informational supplement. Wayne, PA: Clinical and
Laboratory Standards Institute, 2011.
Widdowson CA, Klugman KP. Molecular mechanisms of
resistance to commonly used non-betalactam drugs in
Streptococcus pneumoniae. Semin Respir Infect 1999; 14: 255
68.
Daneman N, Mc Geer A, Green K, Low DE. Macrolide
resistance in bacteremic pneumococcal disease: implication for
patient management. Clin Infect Dis 2006; 43: 4328.
Pantosti A, Venditti M. What is MRSA? Eur Respir J 2009; 34:
11906.
Otto M. Basis of virulence in community-associated
methicillin-resistant Staphylococcus aureus. Annu Rev
Microbiol 2010; 64: 14362.
David MZ, Daum RS. Community-associated
methicillin-resistant Staphylococcus aureus: epidemiology and
clinical consequences of an emerging epidemic. Clin Microbiol
Rev 2010; 23: 61687.
Gillet Y, Domitrescu O, Tristan A, Dauwalder O, Javouhey E,
Floret D, et al. Pragmatic management of Panton-Valentine
leukocidin-associated staphylococcal diseases. Int J Antimicrob
Agents 2011; 38: 45764.
Karampela I, Poulakou G, Dimopoulos G. Community
acquired methicillin resistant Staphylococcus aureus
pneumonia: an update for the emergency and intensive care
physician. Minerva Anestesiol 2012; 78: 93040.
Nathwani D, Morgan M, Masterton RG, Dryden M, Cookson
BD, French G, et al. Guidelines for UK practice for the
diagnosis and management of methicillin-resistant
Staphylococcus aureus (MRSA) infections presenting in the
community. J Antimicrob Chemoter 2008; 61: 97694.
Carrillo-Marquez MA, Hulten KG, Hammerman W, Lamberth
L, Mason EO, Kaplan SL. Staphylococcus aureus pneumonia
in children in the era of community-acquired
methicillin-resistance at Texas Childrens Hospital. Pediatr
Infect Dis J 2011; 30: 54550.
Huang YC, Chen CJ. Community-associated
meticillin-resistant Staphylococcus aureus in children in
Taiwan, 2000s. Int J Antimicrob Agents 2011; 38: 28.
Schultz KD, Fan LL, Pinsky J, Ochoa L, Smith EO, Kaplan
SL, et al. The changing face of pleural empyemas in children:
epidemiology and management. Pediatrics 2004; 113: 173540.
Lema^tre C, Angoulvant F, Gabor F, Makhoul J, Bonacorsi S,
Naudin J, et al. Necrotizing pneumonia in children: report of
41 cases between 2006 and 2011 in a French tertiary care
center. Pediatr Infect Dis J 2013; 32: 11469.
Montagnani C, Cocchi P, Bianchi L, Resti M, de Martino M,
Galli L. Severe infections caused by Panton-Valentine
leukocidin-positive Staphylococcus aureus in infants: report of
three cases and review of literature. Acta Paediatr 2013; 102:
e2847.

2013 Foundation Acta Pdiatrica. Published by John Wiley & Sons Ltd 2013 102 (Suppl. 465), pp. 2533

31

Treating community-acquired pneumonia

Galli et al.

37. Cocchi P, Taccetti G, Montagnani C, Campana S, Galli L,


Braggion C, et al. Evidence of transmission of a
Panton-Valentine leukocidin-positive community-acquired
methicillin-resistant Staphylococcus aureus clone: a family
affair. Clin Microbiol Infect 2013; Jan 17. doi:10.1111/
1469-0691.12159.
38. Liu C, Bayer A, Cosgrove SE, Daum RS, Fridkin SK, Gorwitz
RJ, et al. Clinical practice guidelines by the Infectious Diseases
Society of America for the treatment of methicillin-resistant
Staphylococcus aureus infections in adults and children:
executive summary. Clin Infect Dis 2011; 52: 28592.
39. Principi N, Esposito S. Macrolide-resistant Mycoplasma
pneumoniae: its role in respiratory infection. J Antimicrob
Chemother 2013; 68: 50611.
40. Atkinson TP, Boppana S, Theos A, Clements LS, Xiao L,
Waites K. Stevens-Johnson syndrome in a boy with
macrolide-resistant Mycoplasma pneumoniae pneumonia.
Pediatrics 2011; 127: e16059.
41. Okada T, Morozumi M, Tajima T, Hasegawa M, Sakata H,
Ohnari S, et al. Rapid effectiveness of minocycline or
doxycycline against macrolide-resistant Mycoplasma
pneumoniae infection in a 2011 outbreak among Japanese
children. Clin Infect Dis 2012; 55: 16429.
42. Chironna M, Sallustio A, Esposito S, Perulli M, Chinellato I, Di
Bari C, et al. Emergence of macrolide-resistant strains during
an outbreak of Mycoplasma pneumoniae infections in
children. J Antimicrob Chemother 2011; 66: 7347.
43. Akaike H, Miyashita N, Kubo M, Kawai Y, Tanaka T, Ogita S,
et al. In vitro activities of 11 antimicrobial agents against
macrolide-resistant Mycoplasma pneumoniae isolates from
pediatric patients: results from a multicenter surveillance study.
Jpn J Infect Dis 2012; 65: 5358.
44. Itagaki T, Suzuki Y, Seto J, Abiko C, Mizuta K, Matsuzaki Y.
Two cases of macrolide resistance in Mycoplasma pneumoniae
acquired during the treatment period. J Antimicrob Chemother
2013; 68: 7245.
45. Zhao F, Lv M, Tao X, Huang H, Zhang B, Zhang Z, et al.
Antibiotic sensitivity of 40 Mycoplasma pneumoniae isolates
and molecular analysis of macrolide-resistant isolates from
Beijing, China. Antimicrob Agents Chemother 2012; 56: 11089.
46. Cardinale F, Chironna M, Chinellato I, Principi N, Esposito S.
Clinical relevance of Mycoplasma pneumoniae macrolide
resistance in children. J Clin Microbiol 2013; 51: 7234.
47. Kawai Y, Miyashita N, Yamaguchi T, Saitoh A, Kondoh E,
Fujimoto H, et al. Clinical efficacy of macrolide antibiotics
against genetically determined macrolide-resistant
Mycoplasma pneumoniae pneumonia in paediatric patients.
Respirology 2012; 17: 35462.
48. Hong JH, Chun JK, Uh Y, Oh KJ, Kim J, Yoon KJ. Two cases
of Mycoplasma pneumoniae pneumonia with A2063G
mutation in the 23S rRNA gene in siblings. Ann Lab Med
2013; 33: 658.
49. Shen Y, Zhang J, Hu Y, Shen K. Combination therapy with
immune-modulators and moxifloxacin on fulminant
macrolide-resistant Mycoplasma pneumoniae infection: a case
report. Pediatr Pulmonol 2013; 48: 51922.
50. Youn YS, Lee KY. Mycoplasma pneumoniae pneumonia in
children. Korean J Pediatr 2012; 55: 427.
51. Bradley JS, Byington CL, Shah SS, Alverson B, Carter ER,
Harrison C, et al. The management of community-acquired
pneumonia in infants and children older than 3 months of age:
clinical practice guidelines by the Pediatric Infectious Diseases
Society and the Infectious Diseases Society of America. Clin
Infect Dis 2011; 53: e2576.
52. Harris M, Clark J, Coote N, Fletcher P, Harnden A, McKean M,
et al. British Thoracic Society guidelines for the management

32

53.

54.

55.

56.
57.

58.

59.

60.

61.

62.

63.

64.

65.

66.

67.

68.

of community acquired pneumonia in children: update 2011.


Thorax 2011; 66: ii123.
Esposito S, Cohen R, Diez Domingo J, Falup Pecurariu O,
Greenberg D, Heininger U, et al. Do we know when, what and
for how long to treat? Antibiotic therapy for pediatric
community-acquired pneumonia. Pediatr Infect Dis J 2012; 31:
e7885.
Cies JJ, Shankar V. Nephrotoxicity in patients with vancomycin
trough concentrations of 1520 lg/ml in a pediatric intensive
care unit. Pharmacotherapy 2013; 33: 392400.
Le J, Bradley JS, Murray W, Romanowski GL, Tran TT, Nguyen
N, et al. Improved vancomycin dosing in children using
area-under-the-curve exposure. Pediatr Infect Dis J 2013; 32:
e15563.
Ito H, Shime N, Kosaka T. Pharmacokinetics of glycopeptide
antibiotics in children. J Infect Chemother 2013; 19: 3525.
Svetitsky S, Leibovici L, Paul M. Comparative efficacy and
safety of vancomycin versus teicoplanin: systematic review and
meta-analysis. Antimicrob Agents Chemother 2009; 53:
406979.
Welte T, Pletz MW. Antimicrobial treatment of nosocomial
meticillin-resistant Staphylococcus aureus (MRSA)
pneumonia: current and future options. Int J Antimicrob
Agents 2010; 36: 391400.
dl S, Ho
nigl M, Raggam R, Seidel MG,
Strenger V, Hofer N, Ro
et al. Age- and gender-related differences in teicoplanin levels
in paediatric patients. J Antimicrob Chemother 2013; 68: 2318
23.
Chen KY, Chang HJ, Hsu PC, Yang CC, Chia JH, Wu TL, et al.
Relationship of teicoplanin MICs to treatment failure in
teicoplanin-treated patients with methicillin-resistant
Staphylococcus aureus pneumonia. J Microbiol Immunol
Infect 2013; 46: 2106.
Chang HJ, Hsu PC, Yang CC, Siu LK, Kuo AJ, Chia JH, et al.
Influence of teicoplanin MICs on treatment outcomes among
patients with teicoplanin-treated methicillin-resistant Staphylococcus aureus bacteraemia: a hospital-based retrospective
study. J Antimicrob Chemother 2012; 67: 73641.
Ueda T, Takesue Y, Nakajima K, Ichki K, Wada Y, Tsuchida T,
et al. Evaluation of teicoplanin dosing designs to achieve a
new target trough concentration. J Infect Chemother 2012; 18:
296302.
Nannini EC, Corey GR, Stryjewski ME. Telavancin for
the treatment of hospital-acquired pneumonia: findings from
the ATTAIN studies. Expert Rev Anti Infect Ther 2012; 10:
84754.
Pfaller MA, Mendes RE, Sader HS, Jones RN. Telavancin
activity against Gram-positive bacteria isolated from
respiratory tract specimens of patients with nosocomial
pneumonia. J Antimicrob Chemother 2010; 65: 2396404.
Chiappini E, Conti C, Galli L, de Martino M. Clinical efficacy
and tolerability of linezolid in pediatric patients: a systematic
review. Clin Ther 2010; 32: 6688.
Wunderink RG, Niederman MS, Kollef MH, Shorr AF, Kunkel
MJ, Baruch A, et al. Linezolid in methicillin-resistant
Staphylococcus aureus nosocomial pneumonia: a randomized,
controlled study. Clin Infect Dis 2012; 54: 6219.
Kalil AC, Murthy MH, Hermsen ED, Neto FK, Sun J, Rupp
ME. Linezolid versus vancomycin or teicoplanin for
nosocomial pneumonia: a systematic review and meta-analysis.
Crit Care Med 2010; 38: 18028.
Walkey AJ, ODonnell MR, Wiener RS. Linezolid vs
glycopeptide antibiotics for the treatment of suspected
methicillin-resistant Staphylococcus aureus nosocomial
pneumonia: a meta-analysis of randomized controlled trials.
Chest 2011; 139: 114855.

2013 Foundation Acta Pdiatrica. Published by John Wiley & Sons Ltd 2013 102 (Suppl. 465), pp. 2533

Galli et al.

69. Fu J, Ye X, Chen C, Chen S. The efficacy and safety of linezolid


and glycopeptides in the treatment of Staphylococcus aureus
infections. PLoS One 2013; 8: e58240.
70. Jiang H, Tang RN, Wang J. Linezolid versus vancomycin or
teicoplanin for nosocomial pneumonia: meta-analysis of
randomised controlled trials. Eur J Clin Microbiol Infect Dis
2013; 32: 11218.
71. Stein GE, Wells EM. The importance of tissue penetration in
achieving successful antimicrobial treatment of nosocomial
pneumonia and complicated skin and soft-tissue infections
caused by methicillin-resistant Staphylococcus aureus:
vancomycin and linezolid. Curr Med Res Opin 2010; 26: 57188.
72. De Cock E, Krueger WA, Sorensen S, Baker T, Hardewig J,
Duttagupta S, et al. Cost-effectiveness of linezolid vs
vancomycin in suspected methicillin-resistant Staphylococcus
aureus nosocomial pneumonia in Germany. Infection 2009;
37: 12332.
73. Watkins RR, Lemonovich TL, File TM Jr. An evidence-based
review of linezolid for the treatment of methicillin-resistant
Staphylococcus aureus (MRSA): place in therapy. Core Evid
2012; 7: 13143.
74. Spzek J, Rezanka T. Lincomycin, clindamycin and their
applications. Appl Microbiol Biotechnol 2004; 64: 45564.
75. Tremblay S, Lau TT, Ensom MH. Addition of rifampin to
vancomycin for methicillin-resistant Staphylococcus aureus
infections: what is the evidence? Ann Pharmacother 2013; 47:
104554.
76. Sader HS, Flamm RK, Farrell DJ, Jones RN. Activity analyses of
staphylococcal isolates from pediatric, adult, and elderly

Treating community-acquired pneumonia

77.

78.
79.

80.

81.

82.

83.

patients: AWARE Ceftaroline Surveillance Program. Clin


Infect Dis 2012; 55: S1816.
File TM Jr, Wilcox MH, Stein GE. Summary of ceftaroline
fosamil clinical trial studies and clinical safety. Clin Infect Dis
2012; 55: S17380.
Stein GE, Babinchak T. Tigecycline: an update. Diagn
Microbiol Infect Dis 2013; 75: 3316.
Townsend ML, Pound MW, Drew RH. Potential role of
tigecycline in the treatment of community-acquired bacterial
pneumonia. Infect Drug Resist 2011; 4: 7786.
Purdy J, Jouve S, Yan JL, Balter I, Dartois N, Cooper CA, et al.
Pharmacokinetics and safety profile of tigecycline in children
aged 8 to 11 years with selected serious infections: a
multicenter, open-label, ascending-dose study. Clin Ther 2012;
34: 496507.
Verde PE, Curcio D. Imbalanced mortality evidence for
tigecycline: 2011, the year of the meta-analysis. Clin Infect Dis
2012; 55: 4712.
Lung DC, Yip EK, Lam DS, Que TL. Rapid defervescence after
doxycycline treatment of macrolide-resistant Mycoplasma
pneumoniae associated community-acquired pneumonia in
children. Pediatr Infect Dis J 2013; 32: 13969.
Kawai Y, Miyashita N, Kubo M, Akaike H, Kato A, Nishizawa
Y, et al. Therapeutic efficacy of macrolides, minocycline, and
tosufloxacin against macrolide-resistant Mycoplasma
pneumoniae pneumonia in pediatric patient. Antimicrob
Agents Chemother 2013; 57: 22528.

2013 Foundation Acta Pdiatrica. Published by John Wiley & Sons Ltd 2013 102 (Suppl. 465), pp. 2533

33

Copyright of Acta Paediatrica. Supplement is the property of Wiley-Blackwell and its content
may not be copied or emailed to multiple sites or posted to a listserv without the copyright
holder's express written permission. However, users may print, download, or email articles for
individual use.

Copyright of Acta Paediatrica is the property of Wiley-Blackwell and its content may not be
copied or emailed to multiple sites or posted to a listserv without the copyright holder's
express written permission. However, users may print, download, or email articles for
individual use.

You might also like