You are on page 1of 19

PART III  Etiologic Agents of Infectious Diseases

SECTION A  Bacteria

115 Staphylococcus aureus


Robert S. Daum

Staphylococcus aureus is the most virulent species of the genus Staphylo-


coccus and the most common pathogen isolated among pediatric patients
MICROBIOLOGY AND PATHOGENESIS
in North America. The pathogenicity of S. aureus reflects its ability to Staphylococci are aerobic or facultatively anaerobic gram-positive cocci
acquire and integrate accessory genetic elements that confer virulence. S. that can persist in distressed environments, such as acidic conditions,
aureus is responsible for community- and healthcare-associated infec- high sodium concentrations, and wide temperature variations. Staphylo-
tions and toxin-mediated diseases, and it is a major cause of mortality. cocci can survive on fomites, in dust, or on clothing for at least several
The species also is adept at evolving strategies to elude antimicrobial days. The defining characteristics of S. aureus are the production of the
therapy, leading to limited therapeutic options. extracellular enzyme coagulase and protein A (Fig. 115.1). Clinical

Fatty acid
modifying Panton–Valentine
IgG enzyme leukocidin
(FAME) (PVL)
Protein A
Lipase Leukocidin R Enterotoxins A, B, C1-3, D, E, H
V8
Microcapsule Toxic shock syndrome toxin-1
protease
Exfoliative toxins A, B
Cell wall
(peptidoglycan)
INVASINS
α-Toxin
β-Hemolysin Capsular
γ-Hemolysin polysaccharide + Clumping factor
δ-Hemolysin types 1, 2, 5, + Fibrinogen-binding protein
Phospholipase C and 8 + Fibronectin-binding protein A
Metalloprotease (elastase) + Fibronectin-binding protein B
Hyaluronidase, hyaluronate lyase + ADHESINS + Collagen-binding protein A
+ Coagulase
+ Polysaccharide/adhesin
Lipoteichoic acid + Polysaccharide intracellular adhesin
Penicillin- + 220kd adhesin
binding
proteins Damaged tissue
β-Lactamase Extracellular matrix (ECM)
Ribitol
teichoic Foreign material
acid
FIGURE 115.1  Virulence factors and relevant surface adhesins of Staphylococcus aureus. (Redrawn from Daum RS. Staphylococcus aureus vaccine. In: Plotkin SA,
Orenstein WA (eds). Vaccines. 7th ed. Philadelphia, Saunders/Elsevier, 2016.)

692
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Staphylococcus aureus 115
manifestations of S. aureus infection can result from tissue invasion,
hematogenous dissemination, or toxin release, which incite inflamma-
tory cascades and tissue necrosis. Isolation of S. aureus from skin and
mucosal sites also may represent asymptomatic colonization because the
organism can be a commensal flora.

Capsule and Cell Wall


Many clinical S. aureus isolates have a polysaccharide capsule; 11 capsular
serotypes have been described, but most experts believe that probably
only 4 exist.1 The high prevalence of two of the capsular polysaccha-
ride types (5 and 8) in almost all collections of clinically important
human and veterinary isolates suggests an important but uncertain role
in pathogenesis. Bloodstream infection (BSI) has also been caused by
unencapsulated organisms; for example, the so-called type 336 isolates
and USA300.2,3 Immunization with the type 5 and 8 polysaccharide
conjugated to Pseudomonas exotoxoid A was the basis for two recent
FIGURE 115.2  Staphylococcal scalded skin syndrome (Ritter disease) in an infant
immunization trials, neither of which met primary end points.4–6
1 week old. (Courtesy of J.H. Brien©.)
The cell wall of S. aureus is composed of peptidoglycan, capsular
polysaccharide when present, ribitol teichoic acid, lipoteichoic acid, and
many surface proteins, including protein A, which binds to the Fc region
of the immunoglobulin G (IgG) molecule.7 Binding of IgG antibody to
the staphylococcal cell surface in this nonphysiologic manner is ineffec- with severe skin infections and lung infections,28 specifically necrotizing
tual and decreases the efficiency with which S. aureus is opsonized and pneumonia.29 The importance of PVL in the pathogenesis of S. aureus
phagocytosed.8–10 infections, a controversial topic because of variation in the susceptibility
of host species, has been clarified in human infections.30–32
Surface Proteins S. aureus causes a variety of so-called toxin-mediated diseases, includ-
ing staphylococcal scalded skin syndrome (SSSS), toxic shock syndrome
Many proteins found on the surface of S. aureus isolates have been (TSS), and staphylococcal food poisoning; all of these are caused by the
implicated in pathogenesis. Adherence of S. aureus to mammalian action of exotoxins and enterotoxins. Exfoliative toxins A (ETA) and B
extracellular matrix components is mediated by a family of adhesins, the (ETB) cleave the glycoprotein desmoglein 1, promoting the spread of
microbial surface components that recognize adhesive matrix molecules S. aureus under the stratum corneum33,34 and resulting in blistering of the
(MSCRAMMs).11 Coagulase is found on the bacterial cell surface and in superficial epidermis, which is characteristic of SSSS and bullous impe-
its environment. Coagulase binds to host prothrombin and forms staphy- tigo (Fig. 115.2). Toxic shock syndrome toxin-1 (TSST-1) and staphylo-
lothrombin, an enzyme that catalyzes the formation of fibrin from coccal enterotoxins B (SEB) and C (SEC) have been implicated in most
fibrinogen.12 The A and B clumping factors are cell surface proteins that cases of TSS. These toxins have superantigen activity; that is, they stimu-
bind to fibrinogen, producing the typical clusters of staphylococci when late T lymphocytes nonspecifically, resulting in cytokine release and
mixed with plasma.13 Coagulase and the clumping factors breach host clinical toxic shock. At a skin or mucosal port of entry TSST-1 can
defenses by causing localized clotting; the clumping factors also may aid interfere with the release of inflammatory mediators locally, which may
adherence to traumatized skin, endothelial structures, and foreign sur- account for a surprisingly benign appearance at the local infective site.
faces. Recognition of the role of S. aureus clumping factors has prompted An expanding family of enterotoxins has been implicated in staphylococ-
investigation into their potential use as vaccine antigens.14 cal food poisoning, but the most frequently implicated molecule is
Iron is an essential component of cytochromes and other redox pro- enterotoxin A (SEA).
teins. A cell wall–bound system of iron acquisition and importation has
been identified in S. aureus and is under investigation to define an appar-
ently essential role in pathogenesis.15 However, a trial of an iron- Genetic Basis and Regulation of Pathogenicity
scavenging protein, ISDB, was terminated prematurely for lack of
demonstrable benefit.16
Factors and Antimicrobial Resistance
The whole genome of S. aureus has been sequenced many times. It has a
Toxins circular chromosome of about 2.8 million base pairs.35 Genes for many
housekeeping functions are highly conserved. The genome also carries
The virulence of S. aureus is due to a combination of variously elaborated mobile genetic elements, such as plasmids, transposons, prophages, and
proteins, including extracellular products (e.g., the α, β, γ, and δ hemo- pathogenicity islands, many of which encode virulence factors or deter-
lysins [also called toxins]), leukocidins, proteases, lipase, deoxyribonucle- minants of antibiotic resistance. These factors represent a subset of a large
ase, a fatty acid–modifying enzyme, and hyaluronidase.17–23 The most class of accessory gene products (including surface proteins, exotoxins,
extensively studied of the exotoxins is α-hemolysin, which causes hemo- and other enzymes) that provide one or more advantages in particular
lysis of erythrocytes, necrosis of the skin, and the release of cytokines and environments, although these accessory products are not required for
eicosanoids that can produce shock. S. aureus is lethal when injected into growth and cell division.
animals. S. aureus mutants lacking α-hemolysin generally are less viru- S. aureus has evolved a remarkable network of regulatory mechanisms
lent,24 although pathogenic S. aureus isolates that do not produce that control subsets of genes that are upregulated or downregulated
α-hemolysin have been identified. S. aureus β-hemolysin is a sphingo- under certain growth and environmental conditions. The most exten-
myelinase that can injure lipid-rich membranes; however, most human sively studied is the agr locus with its two-component signal transduction
isolates do not express β-hemolysin. The high prevalence of γ-leukocidin system and its effector RNAIII molecule. The agr locus upregulates genes
genes in isolates that cause necrotizing skin infections suggests the that encode capsular polysaccharides; α-δ toxins; two-component syn-
importance of this toxin in causing dermonecrosis. S. aureus has several ergohymenotropic toxins; enterotoxins; exfoliatins; and proteases; it
leukocidins, which are synergohymenotropic toxins that damage mem- downregulates other genes, such as those that encode MSCRAMMs,
branes of certain cells as a result of the concerted action of two elaborated, other adhesins, and protein A. Other two-component signal transduction
secreted proteins. The Panton-Valentine leukocidin (PVL) is a pore- systems implicated in pathogenesis are saeRS, srrAB, arlSR, and lytRS.
forming cytotoxin,25 and its toxic effect is said to result from the syner- Protein systems that bind deoxyribonucleic acid (DNA), such as sarA
gistic action of the proteins lukS-PV and lukF-PV.26 PVL has been and its regulatory homologues, also regulate virulence factors. Addition-
associated with severe inflammatory lesions, presumably through the ally, the σ-factor, σB, can combine with the ribonucleic acid (RNA)
activation of granulocytes.27 PVL-producing strains have been associated polymerase core enzyme to form a holoenzyme that can recognize

693
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
PART III  Etiologic Agents of Infectious Diseases
SECTION A  Bacteria

promoter elements for at least 36 S. aureus genes involved in bacterial


stress responses. Taken together, membrane sensing systems (e.g., agr)
Healthcare-Associated Infections
and DNA-binding regulatory systems afford S. aureus a versatile environ- S. aureus can develop resistance to all classes of antimicrobial agents. The
mental response system and confer the capacity to respond to a myriad first MRSA strain was reported less than a year after the introduction of
of environmental stimuli, such as high or low sodium chloride concen- semisynthetic penicillins in the early 1960s, even though these penicil-
trations, subinhibitory concentrations of protein synthesis inhibitors, a lins were active against β-lactamase–producing, so-called methicillin-
low pH, or a condition in which essential nutrients (e.g., amino acids) susceptible S. aureus (MSSA). Multidrug-resistant HA-MRSA strains
are limited. spread worldwide in hospital settings. The National Healthcare Safety
S. aureus can overcome environmental antibiotic pressure through the Network (NHSN) (formerly the National Nosocomial Infections Sur-
acquisition and transmission of resistance genes from other, usually less veillance System), an agency of the Centers for Disease Control and
pathogenic, species and isolates of the same species. Resistance to Prevention (CDC), has reported S. aureus as a major pathogen of HAIs.
β-lactam antibiotics (e.g., penicillins, cephalosporins, and carbapenems) In 2003 MRSA accounted for 64% of nosocomial S. aureus isolates in
is one example. The β-lactam antibiotics bind to S. aureus penicillin- intensive care units (ICUs) in U.S. hospitals, an increase from 36% in
binding proteins (PBPs), thereby inhibiting cell wall synthesis. 1992.48 From January, 2006, to October, 2007, S. aureus was the second
Resistance to penicillin was documented almost immediately after the most common cause of HAIs, exceeded only by coagulase-negative
medication’s introduction into clinical practice in the 1940s. It is medi- staphylococci (CoNS).49
ated by the elaboration of a β-lactamase, which is encoded by a transpo- In pediatric ICUs in the U.S., S. aureus is the major nosocomial
son borne on a plasmid. Almost all clinical isolates of S. aureus can pathogen in a variety of clinical situations; it accounts for 9% of HA
elaborate this enzyme, rendering antibiotics susceptible to β-lactamase bloodstream infections (HA-BSIs), 17% of cases of HA pneumonia, and
hydrolysis clinically ineffective. 20% of surgical site infections.50 Current patterns of HA-MRSA isolates
The development of β-lactamase–resistant semisynthetic penicillins reveal decreasing rates of isolation51 and decreasing resistance to non–β-
temporarily overcame this widely prevalent clinical problem. However, lactam antibiotics; this suggests a shift in nosocomial MRSA epidemiol-
some strains became resistant to semisynthetic compounds by acquiring ogy, probably reflecting the movement of “biologically fit” CA-MRSA
mecA, a gene that elaborates PBP2a, a peptidoglycan-synthesizing enzyme isolates into the hospital.40,52
that has decreased affinity for β-lactam antibiotics.36,37 These resistant In recent years MSSA has become a healthcare-associated pathogen,
strains are called methicillin-resistant Staphylococcus aureus (MRSA) and predominantly over MRSA in some settings, but more frequently associ-
are responsible for nosocomial outbreaks and for the current community- ated with BSI and the requirement for intensive care.
based MRSA epidemic. MRSA strains are resistant to all β-lactams except
ceftaroline.
The mecA resistance gene is located on a mobile genetic element
Community-Associated MRSA Infections
called the staphylococcal chromosome cassette mec (SCCmec),38 which Prevalent MRSA infection outside the healthcare setting was first reported
is present in all MRSA isolates, with occasional exceptions. SCCmec in 1982 in Detroit, Michigan, among intravenous (IV) drug users.53 This
contains a mec complex, comprised of mecA and its variably present and subsequent reports of CA-MRSA were associated with risk factors for
mec1 and mecR1 regulatory genes, and a ccr complex, comprised of infection similar to those for HA-MRSA, including IV drug administra-
genes that mediate insertion and excision of SCCmec from the genome. tion, recent hospitalization or surgery, presence of indwelling catheters
Insertion of SCCmec into the S. aureus genome is the genetic event or devices, dialysis, or residence in a longterm care facility.54–57 In the late
that converts a methicillin-susceptible strain into a methicillin-resistant 1990s CA-MRSA infections emerged; these infections occurred mostly
one. Currently 11 SCCmec elements have been sequenced or partially in children with no identifiable predisposing MRSA risk factors,58–62 and
characterized. In the United States, mobile elements SCCmec types I they commonly resulted in skin and soft tissue infections (SSTIs). Some
to III generally are found in healthcare-associated MRSA (HA-MRSA) serious infections required hospitalization or were fatal.63 The number
isolates, and SCCmec types IV and V generally are found in community- and geographic distribution of CA-MRSA infections grew substantially.
associated MRSA (CA-MRSA) isolates, although these distinctions are Strong evidence supports the de novo rise of MRSA in the community,
blurring.39 rather than movement of HA-MRSA into the community.64,65
Although the mechanism of the movement of SCCmec elements from A CA-MRSA infection can be defined as one that occurs in an outpa-
strain to strain is unknown, the large size of types I to III is believed to tient or that develops within 72 hours of admission to the hospital in a
limit easy transfer of the elements. Types IV and V, however, are small patient without any of the factors used to define risk for HA-MRSA:
and mobile. Types IV and V have been found in multiple S. aureus genetic recent hospitalization or surgery, prolonged antibiotic therapy, underly-
backgrounds, which supports the hypothesis that they are readily trans- ing chronic disease, indwelling catheter or other device, healthcare
ferred from strain to strain.40–42 To date, types VI through XI have been contact, or residence in a longterm facility.66 Molecular definitions also
identified in only a limited number of strains. have been used to distinguish HA-MRSA from CA-MRSA strains.67
Outbreaks of CA-MRSA infections have been reported in group childcare
EPIDEMIOLOGY settings,57,68 sports teams,69,70 correctional facilities,71,72 and military
units,73,74 which suggests that close contact and suboptimal hygiene
Colonization practices play a role in the spread of infection.
CA-MRSA isolates also can be distinguished from HA-MRSA by their
Humans and other mammals are the natural reservoirs for S. aureus. lack of multidrug resistance. Most CA-MRSA isolates are susceptible to
Asymptomatic colonization is frequent in humans, and traditionally the clindamycin, trimethoprim-sulfamethoxazole (TMP-SMX), and doxy-
anterior nares were thought to be a predominant site. However, the skin, cycline, whereas HA-MRSA isolates usually are resistant to these agents.
nails, pharynx, axillae, perineum, throat, rectum, and vagina are common Multilocus sequence typing, SCCmec typing, and pulsed-field gel electro-
sites of colonization. Colonization rates range from 25% to 50%; higher phoresis have elucidated important differences between HA-MRSA and
rates are found in children, in people with dermatologic conditions (e.g., CA-MRSA isolates. In the U.S., HA-MRSA usually contain SCCmec type
eczema), in those who perform needle injections frequently (e.g., intra- II and genes that mediate resistance to several non–β-lactam antibiot-
venous drug abusers), in those with indwelling intravascular devices (e.g., ics. In contrast, CA-MRSA isolates usually contain SCCmec type IV or
patients receiving dialysis), and in healthcare personnel (HCP). Even type V and lack non–β-lactam antibiotic resistance genes.59 Even among
higher rates have been documented with the use of highly sensitive detec- CA-MRSA isolates, however, determinants of non–β-lactam agent
tion methods, leading some to speculate that colonization may be uni- resistance can be found elsewhere in the cell, such as on plasmids or
versal.43 Although it has been held that most people are colonized in the chromosome. For example, erythromycin resistance is common
intermittently (and fewer are persistently or rarely colonized44), scant among both HA-MRSA and CA-MRSA strains and can be mediated
explicit data are available. Traditionally, nasal S. aureus carriage has been by a variety of phenotypic and genetic mechanisms. When the erm
a risk factor for developing an infection,45–47 although most colonized gene that commonly mediates erythromycin resistance is present, the
individuals do not do so. CA-MRSA colonization affects more body sites, constitutive or inducible MLSB phenotype (discussed later in the chapter)
which correlates with an increased risk of infection. is conferred.75,76

694
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Staphylococcus aureus 115
Another difference between CA-MRSA and HA-MRSA isolates is their
repertoire of toxin genes.77 The most striking examples are the lukS-PV
and lukF-PV genes that encode for PVL and are transferred from strain
to strain by an as yet unknown mechanism.78 These toxin genes have been
associated with MSSA and MRSA strains that cause SSTIs, necrotizing
pneumonia, and empyema.79–81 More severe infections, such as severe
sepsis, thrombosis, and thromboembolism, also are caused by isolates
containing the PVL genes.30 Interestingly, although SCCmec IV or
SCCmec V and PVL together seem to confer a selective advantage for
spread (i.e., transmission and colonization) and pathogenicity (i.e.,
excess rate of infection per colonized individual), the genes encoding PVL
are transferred separately from the SCCmec element. Most circulating
CA-MRSA organisms in the U.S. belong to a genetic lineage called USA
300 (from a nomenclature scheme based on pulsed-field gel electropho-
retic pattern). These organisms contain an island of foreign DNA called
the arginine catabolic mobile element (ACME)–encoded arc cluster. The
role of ACME is the subject of controversy, but it is believed to confer an
improved survival advantage on skin and spermidine susceptibility.82–84

Vancomycin-Intermediate S. aureus and


Vancomycin-Resistant S. aureus Infections
S. aureus isolates with decreased susceptibility to vancomycin also FIGURE 115.3  Bullous impetigo in a neonate. (Courtesy of J.H. Brien©.)
emerged in the late 1990s. These vancomycin-intermediate S. aureus
(VISA) strains, defined as having a minimal inhibitory concentration
(MIC) of vancomycin greater than 2 µg/mL, typically have been isolated
from patients with underlying medical conditions in whom vancomycin hidradenitis. When the lesion is associated with a hair shaft, the term
therapy has failed85,86; these strains usually also are resistant to teicoplanin furuncle is often used (see Fig. 68.6). Rupture yields a purulent discharge.
(a glycopeptide not licensed in the U.S.). Therefore, the term glycopeptide- A carbuncle is a larger lesion formed by the coalescence of furuncles.
intermediately susceptible S. aureus (GISA) may be more appropriate. In With most small skin abscesses the symptoms pain and erythema are
2002 the first high-level vancomycin-resistant S. aureus (VRSA) strain local, although fever can be present. Systemic signs can accompany larger
was reported (MIC of vancomycin >16 µg/mL); this isolate apparently lesions. In the neonate a skin abscess or boil can progress rapidly to BSI
acquired the vanA vancomycin resistance genes from Enterococcus organ- and clinical sepsis.
isms. To date, 13 VRSA isolates have been reported in the U.S., and all S. aureus is the leading cause of breast abscess in infants, which com-
strains evaluated have carried the vanA gene.87–89 monly occurs within the first 3 weeks of life, when the breasts are enlarged
physiologically (Fig. 115.4) The abscess usually is unilateral and accom-
CLINICAL MANIFESTATIONS panied by erythema and induration. Occasionally fever, leukocytosis, or
BSI can occur. Isolation of the pathogen from the abscess’s discharge aids
Skin and Soft Tissue Infections management.94
Local cleansing and incision and drainage are essential components of
Impetigo the management of abscesses.95,96 Sometimes small abscesses (<5 cm) can
be treated without systemic antimicrobial therapy95; however, in a ran-
S. aureus is a major cause of several infections of the skin and its append- domized, placebo-controlled trial, TMP-SMX had a higher clinical cure
ages. The most superficial is impetigo, which begins as a small, tender, rate.97 Neonates with skin abscesses and infants with a breast abscess
erythematous papule. Often the integument has been interrupted, such should be treated. Systemic antibiotic treatment also is required for any
as by an insect bite, a varicella vesicle, eczema, or a minor abrasion from patient with severe or extensive disease, rapid progression of associated
trauma. Bullous impetigo is a specific S. aureus infection in which cellulitis, systemic illness, associated comorbidity or immunosuppres-
transparent bullae rupture easily, exposing a moist base surrounded by a sion, abscess in an area difficult to drain (e.g., face, genitalia, hand), or
thin rim of scale (Fig. 115.3). Bullous impetigo is mediated by the pro- associated phlebitis.98
duction of ETA and ETB.90,91
Several studies have demonstrated the superiority of topical or sys-
temic antibiotics over simple cleansing for the treatment of impetigo. For
uncomplicated singular sites of impetigo in children outside the neonatal
period, topical mupirocin can be appropriate. Mupirocin ointment
should be applied 3 times daily for 7 to 10 days. Retapamulin recently
was licensed for topical therapy for children 9 months of age or older
who have impetigo caused by Streptococcus pyogenes or MSSA (but not
MRSA).92 This drug’s place in the therapeutic armamentarium has yet to
be determined. Many experts prefer systemic antimicrobial therapy for
the treatment of bullous impetigo.93
Oral or parenteral antimicrobial therapy should be considered if the
impetigo is widespread. Because of the high prevalence of MRSA, a culture
should be performed before the initiation of systemic therapy in most
patients. Neonates with minor bullous impetigo (frequently near the
umbilicus) who are afebrile and well sometimes can be managed with oral
empiric systemic antimicrobial therapy with an agent active against MRSA.

Abscess
S. aureus commonly causes superficial abscesses of the skin. Moist and FIGURE 115.4  Abscess and cellulitis due to community-acquired methicillin-
hairy skin is particularly susceptible to furuncles, boils, folliculitis, and resistant Staphylococcus aureus (CA-MRSA). (Courtesy of J. H. Brien©.)

695
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
PART III  Etiologic Agents of Infectious Diseases
SECTION A  Bacteria

Any abscess can heal with scarring. Some patients may suffer from
recurrent furunculosis, which can be difficult to manage. Systemic and
topical antimicrobial agents, along with careful hand and nail hygiene,
may help reduce recurrences.
Recurring MRSA SSTIs are common and pose a management chal-
lenge. Underlying skin conditions (e.g., eczema, diaper dermatitis)
should be managed aggressively. Attempts to decolonize a patient with
nasal mupirocin or applications of an antiseptic (e.g., chlorhexidine)
often are unsuccessful; in addition, the right time to attempt such decolo-
nization is unclear. Consideration of congenital or acquired immunode-
ficiency may be warranted in patients with recurrent disease, especially
if the response to therapy is slow or healing occurs with remarkable
scarring.99

Cellulitis
Cellulitis is an infection of the subcutaneous tissues and the dermis
that manifests as an area of erythema, warmth, edema, and tenderness. FIGURE 115.5  Preseptal cellulitis due to MRSA infection after minor trauma.
S. aureus is a major cause of the infection in all age groups and at all clinical (Courtesy of J.H. Brien©.)
sites. If systemic signs are present, a blood culture should be performed
before antimicrobial therapy is started. When present, purulent material
should be cultured. Culturing an aspirate or biopsy specimen from a responsible for orbital cellulitis that complicates sinusitis.104 Patients or
patient with nonpurulent cellulitis can yield the infecting bacterium.100 parents note a red, swollen “eye”; the physical examination reveals one
Treatment involves oral or parenteral antimicrobial therapy, depend- or more of the following: decreased or painful movement of the globe,
ing on the extent of disease and whether systemic signs or toxicity is impaired visual acuity, chemosis, or proptosis.103 Complicating cavernous
present. sinus thrombosis should be suspected if the patient has abnormal neu-
rologic findings.105 Treatment requires confirmation of the pathogen and
Wound Infection antimicrobial susceptibility. CT helps distinguish between orbital phleg-
mon, subperiosteal abscess, and orbital abscess; staphylococci are more
The likelihood of a postoperative or posttraumatic wound infection often associated with abscesses. These diagnoses mandate assessment for
increases with the use of sutures, especially in poorly vascularized tissues. surgical drainage in addition to IV antibiotics.106 An extended course of
Measures that often can prevent infections include frequent dressing antimicrobial therapy is given parenterally.
changes, meticulous daily wound care, and strict handwashing. S. aureus S. aureus endophthalmitis is an uncommon infection in children and
wound infections generally are recognized within a few days after trauma usually follows ocular surgery or penetrating trauma to the globe.107
or surgery; inflammation is a local sign, and constitutional signs of illness Symptoms include blurred vision, redness, pain, lid swelling, and the
may be present. The exudate often is cloudy, hemorrhagic, and notably presence of a hypopyon. Systemic signs or fever and lethargy often are
odorless. Extension of a wound infection into deeper tissues can occur, present. Treatment includes antibiotics, both given parenterally and
resulting in cellulitis, lymphadenitis, lymphangitis, or necrotizing fasciitis. instilled directly into the vitreal cavity, and vitrectomy. Visual outcomes
Bursitis usually is the complication of a contiguous skin infection, fre- are poor.108
quently with trauma, and must be differentiated from pyogenic arthritis.
Treatment of a wound infection includes exploration of the wound,
drainage, removal of any foreign material, and optimization of skin
Invasive Infections
hygiene. Topical or systemic antimicrobial therapy may be a useful
adjunct for mild infections. Parenteral antimicrobial therapy should be
Osteomyelitis
considered for clinically serious or extensive infections and for wound S. aureus is the major cause of hematogenous osteomyelitis, accounting
infections on the head and neck. for up to 90% of cases. The metaphyses of long, tubular bones are affected
most frequently, although infection can occur in any bone.109–112 Previous
Ocular Infections minor trauma may predispose a person to infection, probably as the
result of subclinical minor vascular injury or because a small area of bony
S. aureus is an important cause of purulent conjunctivitis. The causative necrosis acts as a nidus for seeding during what otherwise would have
organism can be visualized by Gram stain or isolated from purulent been asymptomatic bacteremia.113 The classic clinical picture includes
discharge. Conjunctivitis usually resolves without treatment, although fever and point tenderness over the affected area of bone (usually at the
topical ophthalmic antimicrobial agents frequently are used. S. aureus is metaphysis), decreased range of motion, and minor local signs of inflam-
the most frequently recognized cause of a stye or a hordeolum, an infec- mation. Infants and toddlers can manifest only irritability, limp, or
tion of the sebaceous gland or the eyelash follicle, respectively. Use of a refusal to bear weight. Diagnosis requires a high degree of suspicion.
topical antimicrobial agent may prevent the spread of infection to adja- Symptoms usually are present 1 week or less before they come to medical
cent hair follicles, but frequent application of warm compresses usually attention, but occasionally they are present for several weeks before
suffices for treatment.101 recognition.114
Preseptal (periorbital) cellulitis can result from extension of sinusitis Evaluation should include a blood culture because BSI is detectable in
or from autoinoculation of skin flora after a break in the skin due to local about 50% of patients.115 At the time of clinical presentation the eryth-
trauma, an insect bite, an abscess, or impetigo.102 (Fig. 115.5). S. aureus rocyte sedimentation rate (ESR) and C-reactive protein (CRP) level
is the leading cause of skin lesion–related preseptal cellulitis. Because usually are elevated, although normal values can be seen, particularly
of the elasticity and thinness of the skin in and around the eyelid, the among neonates or patients with sickle cell disease, distal digital infec-
clinical presentation can be dramatic, with a rapid onset of lid swelling, tion, or subacute cases (e.g., Brodie abscess). Other acute phase reactants
erythema, and tenderness.103 Low-grade fever and leukocytosis can be (e.g., leukocyte count) may be normal.116 A radiograph of the affected
present, although typically the child does not appear systemically ill. bone is likely to have a normal appearance in the first days after the onset
Visual acuity is not affected, and movement of the globe is not painful of symptoms, or it may show only deep soft tissue swelling adjacent to
or limited. Treatment of preseptal cellulitis usually is initiated parenter- the bony metaphysis; periosteal elevation or lucencies within the bone
ally; oral therapy can be substituted after a clinical response has been typically are not apparent until 10 to 21 days after the onset of infection.
demonstrated. A technetium-99 methylene phosphonate scan, performed in three
S. aureus is presumed to be the leading cause of orbital (postseptal) phases, identifies increases in regional perfusion and tracer uptake by
cellulitis after surgical or accidental trauma to the orbit, and it can be inflamed bone. Scintigraphy is especially useful for detecting multifocal

696
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Staphylococcus aureus 115
infection. Although scintigraphy is sensitive, the results can be normal in range of motion of the involved joint. Manifestations of a hip infection
neonates and patients with sickle cell disease with osteomyelitis. MRI is can be more subtle because of the depth of tissue surrounding the joint.
the most accurate diagnostic test, and it also can identify critical fascial In infants a subtle increase in resistance to movement or pain with
and deep muscle phlegmon and abscess.117 movement may be the only clue. An imaging study (e.g., ultrasonogra-
Establishing the causative organism by needle aspiration or biopsy of phy) is valuable.124 Infants with osteomyelitis of the femur or humerus
the bone is important, especially if blood cultures are negative. Surgical especially are prone to pyogenic arthritis of the contiguous joint due to
intervention beyond a diagnostic needle aspiration usually is not required the vascular and capsular anatomy in this region at this age125 (see
for simple acute osteomyelitis, but it can be indicated urgently if extensive Fig. 76.4).
subperiosteal or soft tissue infection or ischemic compartment syndrome If pyogenic arthritis is suspected, blood culture and aspiration of joint
is present, and it must be repeated frequently. Surgery often is required fluid are performed urgently; the hip usually is drained surgically. The
for chronic infection to remove sequestra and to debride affected bone. fluid is evaluated by Gram stain and culture, total and differential cell
IV antibiotics should be initiated and subsequently modified accord- counts, and glucose and protein levels. In occasional cases of S. aureus
ing to microbiology test results. Most experts recommend a course of arthritis, bacteria visualized on Gram stain are not isolated in culture
antimicrobial therapy extending up to 4 weeks or longer for S. aureus because of the bacteriostatic nature of synovial fluid or previous antimi-
osteomyelitis, with parenteral therapy used initially. Deep vein thrombo- crobial therapy.
sis adjacent to the bony site of infection and septic thromboembolism Treatment consists of surgical drainage and antimicrobial therapy.
increasingly have become recognized as complications of S. aureus Antimicrobial therapy should be initiated when clinical suspicion is high
osteomyelitis, especially when they are caused by CA-MRSA or PVL- and is continued for at least 3 weeks. Multiple aspiration procedures may
positive MSSA. These conditions usually develop in the proximal long be necessary. Open surgical drainage is used when serial aspiration does
bones and vertebrae.118 The total duration of the therapy and the transi- not produce a rapid response; as initial management when the hip or
tion to oral therapy is individualized, depending on the extent of infec- shoulder joint is affected; or when establishing effective drainage by
tion, susceptibility test results, the clinical setting and course.114,119,120 One simple aspiration is technically difficult and the risk of longterm compli-
study has shown that a total course of therapy of 3 weeks to be noninfe- cations is high.114 A poor outcome is associated with delay of appropriate
rior,120 and another study has shown that early transition to oral therapy management and concomitant osteomyelitis.126
also is effective, even when CA-MRSA is the cause.121
Techniques for monitoring the therapeutic response vary, and few
explicit data are available. Most experts use a rapid fall in the CRP level
Abscesses of Muscle and Viscera
as evidence of an appropriate antibiotic choice, and an eventual fall in S. aureus is the most common cause of pyomyositis (including iliopsoas
the ESR as a guide to an adequate duration of therapy. In general, serial and obturator internus abscess) and liver abscess, and it is an important
imaging of the infected bone does not provide useful information about cause of renal, perinephric, splenic, and pancreatic abscesses. These are
the duration of therapy. all complications of BSI, which may have been asymptomatic; the patient
S. aureus can cause osteomyelitis secondary to an adjacent deep soft may come to medical attention because of symptoms related to the focal
tissue focus; by concurrent direct inoculation; or by contiguous spread, infection or for fever of unknown origin (FUO) or associated with severe
such as through an accidental puncture wound of the foot or the patella, sepsis. Necrotizing fasciitis, especially due to CA-MRSA, usually occurs
a bite, trauma, an infected paronychia, or a surgical procedure.122 Osteo- as a complication of skin infection but also can occur after seeding of a
myelitis that develops after a puncture wound frequently is called deep site during BSI (see Fig. 75.2). Pyomyositis usually manifests with
osteochondritis, partly to remind clinicians of the clinical and patho- fever and an insidious or acute onset of cramping muscle pain, tender-
physiologic features that distinguish it from hematogenous osteomyelitis. ness and, especially, induration of a large muscle group, such as those
Symptoms and signs of acute hematogenous osteomyelitis usually are near the buttock, thigh, or hip. An obturator internus abscess can cause
absent, although many patients have localized pain, fever, and erythema. a painful limp, pelvic girdle pain, or refusal to walk. An iliopsoas abscess
The blood typically is sterile, and the CRP level can be minimally can cause predominantly flank pain and tenderness. A visceral abscess
increased or normal. Surgical debridement is an important component and nephric abscesses are most likely to manifest as FUO, possibly
of the therapeutic strategy in nonhematogenous osteomyelitis/ accompanied by nonspecific abdominal or flank symptoms. A blood
osteochondritis syndromes; antimicrobial therapy is adjunctive. The culture is positive in fewer than one half of cases that present because of
optimal duration of antimicrobial therapy for osteochondritis is unclear, FUO or localized complaints.
but it frequently can be short (e.g., 7 to 14 days) after adequate surgical CT or MRI is highly likely to identify abnormalities. Management
debridement in selected cases.114 usually entails surgical decompression, debridement and drainage, and
prolonged parenteral antibiotic therapy. Multiple, small visceral abscesses
Diskitis frequently can be treated with antibiotic therapy alone if the pathogen is
identified and susceptibility test results are available. Necrotizing fasciitis
Diskitis, or inflammation of an intervertebral disk, is uncommon and is a surgical emergency and requires decompression and debridement of
occurs primarily in children younger than 5 years of age. Diskitis may be the involved tissue and antimicrobial therapy.
difficult to distinguish from vertebral osteomyelitis, and the two entities
probably have a similar pathogenesis (see chapters 76 and 78). Children
with diskitis usually are not febrile or ill, but they refuse to walk, limp or,
Upper Respiratory Tract Infections
in older children, have back pain. MRI helps distinguish diskitis from
vertebral osteomyelitis. S. aureus is the most frequently identified organ-
Head and Neck Infections
ism in both entities, although specimens are collected more frequently S. aureus is an infrequent cause of acute otitis media or sinusitis, but it
and are positive more frequently in patients with vertebral osteomyelitis. should be considered in a child who fails multiple courses of empiric,
Diskitis may resolve spontaneously without antimicrobial therapy; ver- oral antimicrobial therapy and in chronic or complicated disease. Both
tebral osteomyelitis does not. MSSA and MRSA increasingly have been isolated in cases of otorrhea
Diskitis without adjacent vertebral involvement sometimes can be that develops after tympanostomy tube insertion, and as complications
managed with observation alone. If the adjacent vertebrae are involved, of paranasal sinusitis.104,127,128
vertebral osteomyelitis cannot be excluded, and these patients should S. aureus is the leading cause of nasolacrimal duct abscess, suppurative
receive a course of antimicrobial therapy appropriate for osteomyelitis. parotitis, abscess of a lymph node below the angle of the jaw in very
Follow-up imaging studies are appropriate in this circumstance.123 young infants (Fig. 115.6), and retropharyngeal abscess in children
younger than 3 years of age (see Fig. 28.1). Infection occurs presumably
Pyogenic Arthritis after nasopharyngeal or oropharyngeal colonization, sometimes in
association with a viral infection. In older children and adolescents
Hematogenous seeding of the synovium during S. aureus BSI is the most S. aureus is the leading cause of peritonsillar and parapharyngeal abscess
common pathogenesis of infectious arthritis in children. Pyogenic associated with recurrent pharyngeal infections and suppurative cervical
arthritis usually is heralded by warmth, tenderness, erythema, and limited lymphadenitis.

697
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
PART III  Etiologic Agents of Infectious Diseases
SECTION A  Bacteria

about 90% of pulmonary infections; spontaneous pneumothorax


occurs in 25% to 50%. A pneumatocele (an air- or fluid-filled cavitation
resulting from dissolution of intra-alveolar septa) occurs in more than
50% of cases. Most cases occur in previously healthy children, although
predisposing factors (e.g., young age [younger than 1 year], chronic lung
disease, immunosuppression, and presence of a foreign body or skin
infection) may increase the likelihood of S. aureus pneumonia. A seasonal
peak in S. aureus pneumonia is associated with viral respiratory tract
infections such as influenza, presumably because viral illness transiently
alters pulmonary defense mechanisms.130
S. aureus necrotizing community-acquired pneumonia (CAP) has
increased dramatically in immunocompetent children. It can be caused
by both MSSA and MRSA strains, which usually contain the PVL
genes.28,131–134 Leukopenia, and rapidly progressive unilateral or bilateral
diffuse parenchymal pulmonary infiltrates are typical.29 Severe sepsis
syndrome can be present and has a high fatality rate. Histologic findings
at autopsy reveal dense S. aureus with necrosis extending from the tra-
A cheobronchial epithelium to the congested and hemorrhagic parenchyma
of the lung29,80 (Fig. 115.8).
A high index of suspicion for S. aureus is critical in evaluating possible
cases and aggressively treating them with empiric therapy active against
MRSA and MSSA. The diagnosis is confirmed by recovery of the organ-
ism from pleural fluid, blood, or tracheobronchial aspirate. The duration
of parenteral antimicrobial therapy depends on the clinical course. When
compared with nonoperative treatment, early intervention for empyema
with video-assisted thoracoscopic surgery (VATS) or open surgical
debridement reduces mortality and the duration of antimicrobial
therapy.135

Lung Abscess
Lung abscess can result as a complication of aspiration of infected mate-
rial or a foreign body, can develop during pneumonia, or can result from
hematogenous seeding during S. aureus BSI or septic thrombophlebitis.
When bacteriology is proved as the mechanism of infection in children
with aspiration, S. aureus can be recovered with or without facultative
and anaerobic oral bacteria.136,137
Clinical features of S. aureus lung abscess include fever, cough, chest
pain, dyspnea, anorexia, and weight loss.136 CT helps determine the extent
of disease and aids in following abscess resolution. Despite a dramatic
B appearance on radiographs, lung abscesses uncomplicated by pleural
empyema usually can be treated adequately with IV antimicrobial therapy
FIGURE 115.6  Two-month-old infant with necrotizing lymphadenitis due to alone, as evidenced by resolution seen on follow-up chest radiographs.138
MSSA. The infant had no intraoral, skin, or middle-ear abnormality. Note the Surgical drainage occasionally is required.139 In these situations percuta-
location of the mass (A and B) and elevation of the earlobe (A). (Courtesy of J.H. neous drainage is a well-established technique.140–142
Brien©.)
Cardiovascular Infections
Endocarditis
Tracheitis S. aureus is the most common cause of infective endocarditis (IE).143 IE
Bacterial tracheitis caused by S. aureus can occur in an airway inflamed occurs more frequently in patients with congenital heart disease, whether
by a viral process or injured by medical manipulation. Affected patients surgically repaired or not, but can follow S. aureus BSI in any patient; or,
manifest hoarseness and stridor (if the larynx is involved concomitantly) it can be associated with device-associated persistent BSI.144 Children
and can deteriorate suddenly, showing respiratory distress and s high whose cardiac lesions cause high-velocity blood flow and those with
temperature. The diagnosis can be suspected by the results of imaging prosthetic valves or patches have an increased risk of developing S. aureus
studies and is confirmed by direct visualization of thick, purulent secre- IE.145,146
tions in the trachea and subglottic edema, along with a positive Gram Fever, a new or changed heart murmur, and continuous BSI are the
stain of secretions and recovery of S. aureus upon culture. Management hallmarks of IE. The presentation can be indolent or acute; common
includes establishment of an artificial airway and IV antibiotic therapy129 complaints include myalgia, arthralgia, general malaise, weight loss, and
(see Chapter 28). anorexia. Clinical progression can be rapid, with associated lethargy and
cardiac failure, particularly when the aortic valve is involved. Right-sided
Lower Respiratory Tract Infections endocarditis can be the source of emboli to the lungs, causing lung
abscesses. Small vessel embolic phenomena, such as petechiae, splinter
Pneumonia and conjunctival hemorrhages, and Janeway lesions, rarely manifest in
children.145
S. aureus can cause rapidly progressive pneumonia after inhalation of the The diagnosis is straightforward when echocardiography demonstrates
organism or seeding of the lungs during BSI. Clinical features include a vegetation or abscess. However, this evidence may be lacking in children.
fever, cough, grunting, and tachypnea, which can evolve into rapidly pro- A negative finding on a transthoracic or transesophageal echocardiogram
gressive and severe respiratory distress. A chest radiograph demonstrates does not exclude IE.147,148 The modified Duke criteria have been used to
multiple, patchy alveolar infiltrates that tend to coalesce to form large identify children with IE based on major and minor clinical criteria (see
consolidations (Fig. 115.7). Pleural effusion and empyema accompany Chapter 37).

698
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Staphylococcus aureus 115

FIGURE 115.7  Progressive MRSA pneumonia with pneumatoceles in a previously


healthy 9-month-old boy. Chest radiographic findings spanning 4 days showed a
perihilar right lower lobe infiltrate (A) progressing to a worsening infiltrate and large
hydropneumothorax with mediastinal shift (B and C) despite appropriate therapy. Axial
CT of the chest without contrast (lung windows) showed partial loculation of hydro-
pneumothorax, multilobar consolidation, pneumatoceles, and atelectasis (D and E).
Video-assisted thoracoscopic surgery (VATS) was performed, and a chest tube was
E placed for 3 days. MRSA was isolated from pleural fluid. After 2 weeks of clindamycin
therapy the chest radiograph had only minor abnormalities. (Courtesy of S.S. Long.)

699
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
PART III  Etiologic Agents of Infectious Diseases
SECTION A  Bacteria

drainage, and parenteral antimicrobial therapy.153–155 Mortality is due to


cardiac tamponade or the systemic inflammatory response syndrome; the
likelihood of both these complications is decreased by early medical and
surgical treatment. Patients who develop constrictive pericarditis require
pericardiectomy.

Suppurative Phlebitis and Septic Thrombophlebitis


Suppurative phlebitis and septic thrombophlebitis are serious endovas-
cular infections that occur along a continuum of clinical severity; they are
most commonly caused by S. aureus. Suppurative phlebitis usually occurs
in a catheterized vessel wall and is characterized by a fluctuant, palpable
vein that is warm, tender, and erythematous; pus sometimes can be
found at the catheter insertion site or by needle aspiration. Infection can
progress to obstruction of the vessel or dissemination of infected thrombi
A to other sites, or both. The treatment of suppurative phlebitis includes
removal of a device, drainage or resection of the infected segment of the
vessel, and IV antimicrobial therapy.
Septic thrombophlebitis also occurs in association with invasive S.
aureus infection without catheterization of the infected vessel. The veins
involved usually are major deep veins, especially pelvic or lower extremity
vessels, and they commonly are contiguous with an area of osteomyelitis,
pyogenic arthritis, pyomyositis, or other soft tissue infection.118 Longterm
IV antibiotic therapy is warranted for this serious endovascular infection,
with its attendant severe complications and mortality. Anticoagulant
therapy and placement of an inferior vena cava filter are considered when
infected clots disperse septic emboli to the lungs.156

Central Nervous System Infections

B
Meningitis
S. aureus meningitis can occur as a complication of BSI or endocarditis,
FIGURE 115.8  Pathologic findings from a patient with fatal necrotizing pneumonia or from extension of a parameningeal focus, including sinusitis, osteomy-
due to S. aureus show a gross specimen of lung with microabscesses (A), and elitis of the skull bone, subdural empyema, epidural abscess, or rupture
the histologic appearance of a section of lung with S. aureus colonies and hemor- of a brain abscess into the lateral cerebral ventricles or the subarachnoid
rhage (B). (Hematoxylin-eosin stain.) (From Adem PV, et al. Staphylococcus aureus space. Congenital anomalies, such as a dermal sinus or meningomyelo-
sepsis and the Waterhouse-Friderichsen syndrome in children. N Engl J Med cele, can provide a portal of entry. Any neurosurgical procedure increases
2005;353:1245–1251.) the risk of staphylococcal meningitis; insertion of a cerebrospinal fluid
(CSF) shunt is the most common predisposing procedure.157
Treatment consists of parenteral antimicrobial therapy for 3 to 6
weeks, depending on the pathophysiology of the meningitis and removal
Treatment requires 4 to 6 weeks of high-dose bactericidal antimi- of a device when present. (Treatment of device-related infection is dis-
crobial therapy, administered intravenously, because of the relatively cussed later in the chapter.)
avascular nature of valves, the high organism load, and the decreased
ability of polymorphonuclear leukocytes to function in endocarditis
lesions. Guidelines for the treatment of S. aureus endocarditis, includ-
Brain Abscess
ing consideration of combination therapy with aminoglycosides, can A brain abscess caused by S. aureus can result from direct extension of
be found in Baltimore and colleagues149 and Chapter 37. Fever may an adjacent infectious focus (e.g., sinusitis,104 mastoiditis, otitis media);
persist for about 1 week after the start of antimicrobial therapy.150 from inoculation of bacteria during a surgical procedure or as a conse-
Sustained bacteremia that spans several days in usual. Serial blood quence of trauma; or from seeding of the brain during bacteremia. A
cultures should be obtained until sterility is demonstrated. Occasionally, brain abscess more frequently complicates S. aureus BSI than BSI due to
surgical intervention is necessary, particularly when embolic phenomena other pathogens158,159 (see Chapter 46). An S. aureus etiology should be
to major organs, intractable cardiac failure, or aortic insufficiency is considered, especially when brain abscess complicates severe S. aureus
documented, or when BSI persists despite appropriate antimicrobial BSI or the patient has had a neurosurgical procedure or penetrating
therapy, particularly when a prosthetic valve or other foreign material is trauma. Cyanotic congenital heart disease also is a predisposing risk
involved. Prosthetic valve endocarditis caused by S. aureus poses a risk of factor.160
mortality.151,152 Clinical features of brain abscess in children include headache, vomit-
Close follow-up should be maintained after therapy has been con- ing, irritability, focal neurologic defect, seizures, or other manifestations
cluded because some patients can relapse and require definitive surgical of increased intracranial pressure. The onset of symptoms can be insidi-
intervention or removal of foreign material. ous, depending on the pathophysiology; fever can be absent or low grade.
The physical examination findings depend on the location of the abscess.
Pericarditis Signs of meningitis are expected only if the abscess ruptures into a
ventricle.
Bacterial pericarditis in children is a rare complication of S. aureus BSI Imaging techniques are most often definitive in diagnosing a brain
or other focal (usually pleural) infection. Pericarditis should be suspected abscess. Enhanced MRI is superior to CT, although both modalities are
in any patient with BSI and cardiomegaly. Signs and symptoms include useful.161 The principal advantages of MRI are the abilities to distinguish
fever, dyspnea, cough, and precordial chest pain. The chest radiograph the central core of an abscess from a surrounding area of ring enhance-
shows an enlarged heart, and echocardiography demonstrates the pres- ment, identify adjacent areas of cerebral edema, and detect abscesses in
ence of pericardial fluid. The diagnosis is made by examination of the the posterior fossa.
pericardial fluid for inflammatory cells and bacteria. Management Treatment consists of parenteral antimicrobial therapy and surgical
includes urgent pericardial decompression, closed or open pericardial drainage of the abscess, frequently by aspiration. In rare cases enucleation

700
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Staphylococcus aureus 115
of the abscess is performed. Evolving abscesses in the non-necrotizing Vancomycin is the preferred empiric therapy for suspected S. aureus
cerebritis stage and some small intracerebral abscesses can be treated with catheter-associated infection. Combination bactericidal therapy and an
antimicrobial agents alone.158 Corticosteroid therapy is limited to cases extended course of antimicrobial therapy (e.g., 4 to 6 weeks) frequently
in which increased intracranial pressure is an important component. are recommended because of the known tendency of S. aureus BSI to
Patients typically are treated with antimicrobial agents parenterally seed and form abscesses at multiple sites. This point is controversial
for 6 weeks. Mortality associated with brain abscess is relatively low, because of the lack of specific data (see Chapter 100). Consistent
but up to 60% of patients have neurologic sequelae, most commonly use of a “bundle” of techniques for placing and handling catheters is
seizures.162 among the measures that have been shown to reduce the infection
rate; these measures include skin disinfection with chlorhexidine, an
Spinal Epidural Abscess aseptic placement technique, use of a BioPatch dressing, dedicated
teams for ongoing catheter care, and the earliest possible removal of the
S. aureus is the most common cause of a spinal epidural abscess, which catheter.
is a collection of purulent material external to the dura in the spinal canal,
usually in the posterior aspect of the thoracic or lumbar vertebrae.
Abscesses arise as a result of seeding of the epidural space during BSI or
Cerebrospinal Fluid Shunt Devices
through direct extension of an adjacent focus of infection, such as pyo- A contaminated CSF shunt or subcutaneous reservoir device is the most
myositis or osteomyelitis. Complaints in children can be nonspecific and common predisposing cause of staphylococcal meningitis.157 Presumably
include fever, vomiting, lethargy, and irritability. Localized tenderness S. aureus gains access to the catheter during surgical insertion, although
and refusal to lie prone also can be noted. In older children, localized or the device and CSF can be seeded by a subsequent adjacent primary SSTI
radicular pain can be present. Paresis of limb muscles and loss of bowel or tract infection or during BSI – the presence of the catheter increases
or bladder control are specific and ominous signs because paralysis and the likelihood that the organism will survive and multiply.
loss of sensory modalities at levels below the affected area can ensue Most CSF shunt infections occur within 2 months of shunt placement
rapidly. A history of a clinical illness compatible with bacteremia some- or revision. Low-grade fever and dysfunction of the shunt are variably
times is elicited.163 present. For ventriculoperitoneal (VP) shunts, symptoms and signs
Spinal epidural abscess must be distinguished from diskitis, extradural attributable to peritonitis commonly can be present.167,168 If the patient
neoplasm, hematoma, disk herniation, and vertebral osteomyelitis. MRI had recently undergone surgery, a wound infection may be evident. A
is the diagnostic procedure of choice.164 Lumbar puncture is deferred first episode of an S. aureus shunt infection is predictive of a subsequent
when this diagnosis is suspected; if it is performed, CSF findings are S. aureus shunt infection.169
similar to those in other parameningeal infections (i.e., a variable, Examination of the CSF (typically obtained by percutaneous puncture
modest pleocytosis, normal glucose, and modestly increased protein of the shunt reservoir/tubing) may reveal only mild abnormalities, such
concentration). as modest pleocytosis, a normal glucose level, and a normal or minimally
Therapy includes immediate surgical decompression, debridement of elevated protein concentration. Microorganisms infrequently are visual-
the epidural space, and parenteral antimicrobial therapy, which generally ized on Gram stain. Culture of CSF is the most sensitive test; however,
is administered for 6 weeks.165 Medical management alone is rarely culture of CSF obtained by lumbar puncture frequently is not diagnostic
considered in immunocompetent patients with a normal or stable neu- of a VP shunt infection.
rologic examination and no signs of septicemia. Continuous reassess- Most experts recommend combined medical and surgical treatment
ment is necessary to identify progressive pain or the onset of any for CSF shunt infections. Typically a temporary external ventricular drain
neurologic abnormality that necessitates urgent surgery.166 is used, while IV antimicrobial therapy is administered, until the CSF is
sterile and inflammation subsides, at which time a new shunt can be
Device-Related Infections placed.170,171 The timing of this procedure is important – insertion of the
new catheter into an infected field must be avoided. However, maintain-
Indwelling Vascular Catheters ing an externalized catheter as a drainage device carries its own risk of
infection, which accelerates after 5 to 7 days of use.
Staphylococci are the most common causes of catheter-associated infec-
tions. For peripheral venous catheters, S. aureus is the most common
infecting species; for central venous catheters (CVCs), coagulase-negative
Toxin-Mediated Syndromes
staphylococci are recovered more commonly. Cutaneous colonization at
the insertion site is the most important predictor of catheter-associated
Staphylococcal Food Poisoning
infections related to short-term, percutaneously inserted catheters. For Staphylococcal food poisoning is caused by ingestion of S. aureus entero-
surgically implanted CVCs, organisms introduced through the hub and toxins. It accounts for up to one third of foodborne gastrointestinal ill-
lumen are the most important source of infection. nesses (see Chapter 59). Protein-rich foods (e.g., ham and chicken) often
Local redness, pain, and warmth at the site of a simple peripheral are implicated as the outbreak source, as are dairy products and egg and
catheter suggest local phlebitis, which is managed by removal of the potato salads. The individual who inadvertently inoculates the food with
device. Diagnosing a central line–associated BSI (CLABSI) can be more S. aureus may have a clinical infection but usually is colonized
difficult. Fever and the presence of pain, fluctuance, erythema, purulence asymptomatically.172,173
or soft tissue infection at the site suggest CLABSI. Most often, however, Illness begins shortly after consumption (4 hours on average) because
the catheter entrance site appears normal. ingestion of preformed toxin causes the clinical manifestation. Classic
Blood for culture is taken via the catheter; a positive culture can rep- symptoms are nausea, vomiting, abdominal pain, and diarrhea; headache
resent CLABSI or BSI from another source. Isolation of S. aureus should and prostration are less common. Fever occurs in about 25% of patients.
not be regarded as the result of a procedural contaminant; repeated Hospitalization is required infrequently, and death occurs rarely.
samplings or, occasionally, blood drawn simultaneously by venipuncture S. aureus can be isolated from the vomitus or feces of ill people.
help to clarify the issue. Semiquantitative culture of the catheter tip after Culture of the contaminated food may yield the offending organism.
removal can implicate the catheter as the infectious nidus when the Therapy is supportive; antibiotics are not useful. The syndrome can be
colony count exceeds 15 colonies; a negative culture of the catheter tip prevented by proper handling of food and careful hygiene practices by
does not exclude CLABSI. food handlers. When cases are recognized, the Department of Public
S. aureus exit-site infections, whether in the presence of a tunnel track Health must be notified.
infection or BSI, are managed by removing the catheter and administer-
ing antimicrobial therapy. Because of the propensity of S. aureus BSI to
persist and to cause clinical sepsis or to seed the endocardium, abdominal
Staphylococcal Scalded Skin Syndrome
viscera, brain, lung, or bone, many experts recommend removal of an SSSS is an illness mediated by exfoliative toxins ETA and ETB.90,91 Hema-
intravascular catheter when S. aureus infection is documented, even in togenous spread of the toxins produces fever and widespread erythema
the absence of an exit-site infection. of the skin, which can be tender and quickly forms thin-walled,

701
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
PART III  Etiologic Agents of Infectious Diseases
SECTION A  Bacteria

fluid-containing bullae that rupture, leaving a moist base of skin. Gentle Risk factors for TSS include colonization and disruption of the skin,
friction applied to the skin produces Nikolsky sign, a sloughing of colonization of mucous membranes, or any infection caused by a toxin-
superficial sheets of skin (see Fig. 13.2). Desquamation follows rapidly producing strain of S. aureus. TSS often is associated with surgery or
and can be widespread, especially in the neonate.174,175 trauma, manifesting several days after the event. The sudden onset of
SSSS is diagnosed clinically or by isolation of S. aureus from a local fever, chills, headache, vomiting, sore throat, myalgia, and diarrhea is
site (not necessarily the exfoliating site), ideally with demonstration by followed by hypotension, respiratory distress, edema, and rash within 24
the isolate of toxin production or the toxin gene. Histologic evaluation to 48 hours. Dizziness, listlessness, and disorientation are clues to hypo-
of the skin reveals the separation of skin layers at the granular layer tension and cerebral dysfunction; coma can ensue.
within the epidermis, rather than the separation of the dermis and epi- The initial S. aureus focus can show no signs of inflammation or
dermis seen in drug-induced toxic epidermal necrolysis.176 purulence. Initially the rash can appear like that seen in scarlet fever, but
Management of SSSS is primarily supportive, and careful attention is it classically is described as diffuse erythroderma, or “sunburn.” The rash
paid to electrolyte levels because fluid shifts can occur across the denuded typically desquamates 1 to 2 weeks after the onset of illness, most notably
skin. Antiseptic measures should be taken to prevent infection of the on the palms and soles.
unprotected, affected areas. Parenteral antimicrobial therapy is adminis- Mucous membrane involvement includes conjunctival hyperemia,
tered to reduce the staphylococcal burden. Topical antimicrobial agents strawberry tongue, beefy red oropharyngeal membranes (sometimes
are not helpful.176 with ulcerations), and lesions representing reactivation of the herpes
simplex virus.
Toxic Shock Syndrome A vaginal culture yields the organism in 85% of menstrual TSS cases.
In nonmenstrual TSS cases a nasopharyngeal or a wound culture may
TSS is caused by S. aureus strains that produce TSST-1 or certain entero- yield S. aureus; a blood culture should be obtained (to diagnose concur-
toxins (specifically SEB and SEC).177 It is characterized by fever, rash, rent BSI) but is seldom positive.
hypotension, and multisystem organ dysfunction178 (Box 115.1). The Management consists of treating intravascular volume depletion,
incidence of TSS peaked in the early 1980s; the condition was attributed shock, and respiratory failure, in addition to the S. aureus infection. Any
to the use of superabsorbent tampons by menstruating adolescents and relevant focus of infection should be drained and any foreign body
women.179 Subsequent public education and manufacturing regulations removed if possible. IV antimicrobial therapy, including an antibiotic
helped reduce the number of TSS cases, and now nonmenstrual TSS that inhibits protein synthesis (e.g., clindamycin or linezolid), may be
accounts for approximately one half of all TSS cases.180 preferable. The response to effective therapy can be rapid. For a patient
who does not respond to these interventions, most experts recommend
immune globulin intravenous (IGIV), based on the presence of antibody
to staphylococcal enterotoxins and TSST-1.181,182 End-organ failure, par-
ticularly renal failure and acute respiratory distress syndrome, can occur
within the first few days of illness and are responsible for the attendant
BOX 115.1  Staphylococcal Toxic Shock Syndrome: Clinical Case 3% mortality rate.
Definition
Severe Sepsis Syndrome
An illness with the following clinical manifestations:
Fever: Temperature >38.9°C (102°F) Severe sepsis syndrome is defined as isolation of S. aureus from a clini-
Rash: Diffuse macular erythroderma cally important site, hypotension (i.e., systolic blood pressure less than
Desquamation: 1–2 weeks after onset of illness, particularly on
the 5th percentile for age for children or less than 90 mm Hg for adults),
and respiratory distress syndrome or respiratory failure, in addition to
palms and soles
involvement of the central nervous system (CNS), liver, kidneys, muscles,
Hypotension: Systolic blood pressure ≥90 mm Hg for adults or
or skin; or abnormal hemostasis; or the presence of leukopenia or
less than 5th percentile by age for children <16 years; thrombocytopenia. Severe sepsis syndrome is similar to TSS but does not
orthostatic drop in diastolic blood pressure ≥15 mm Hg from meet all the clinical criteria for TSS, and the S. aureus isolates do not
lying to sitting, orthostatic syncope, or orthostatic dizziness produce TSST-1.81
Multisystem involvement: 3 or more of the following: Increasingly, severe sepsis occurs in children and adolescents with no
1. Gastrointestinal: Vomiting or diarrhea at onset of illness underlying medical conditions or other identifiable risk factors.81,183–185
2. Muscular: Severe myalgia or creatine phosphokinase level Both MSSA and MRSA are implicated; genes encoding PVL frequently
≥2 times the upper limit of normal for laboratory are identified in the infecting strain.
3. Mucous membrane: Vaginal, oropharyngeal, or conjunctival A primary infectious process (e.g., pyogenic arthritis or pneumonia)
hyperemia often is identified before clinical deterioration begins. However, patients
4. Renal: Blood urea nitrogen or creatinine ≥2 times the upper can have an overwhelming infection and experience subsequent rapid
limit of normal for laboratory or urinary sediment with pyuria decline without a primary tissue site of infection.81,183,184 Skin lesions are
(≥5 leukocytes per high-power field) in the absence of common and manifest as erythroderma, pustules, petechiae, and/or
urinary tract infection purpura. Waterhouse-Friderichsen syndrome,186 staphylococcal purpura
5. Hepatic: Total bilirubin, serum glutamic-oxaloacetic fulminans,187 and necrotizing fasciitis have been reported.188
transaminase (SGOT), or serum alanine aminotransferase The diagnosis is made by isolation of S. aureus from a clinically
(ALT) or aspartate aminotransferase (AST) ≥2× the upper important site, such as the blood, a joint space, or respiratory secretions
limit of normal for laboratory
(with concomitant systemic illness). A high index of suspicion is critical
to instituting appropriate parenteral antimicrobial therapy and aggressive
6. Hematologic: Platelets <100,000/mm3
cardiopulmonary support. Mortality rates based on reviews of case
7. CNS: Disorientation or alteration in consciousness without
reports approach 64%.183
focal neurologic signs when fever and hypotension are
absent
Negative results on the following tests, if obtained: MANAGEMENT
1. Blood, throat, or cerebrospinal fluid cultures (for all Treatment of invasive S. aureus infections should be rapid and aggressive.
microorganisms except Staphylococcus aureus) IV antimicrobial therapy should be started promptly, and the source of
2. Rise in antibody titer to Rickettsia or Leptospira species or BSI and sites of possible metastatic infection should be sought aggres-
rubeola sively. Abscesses should be drained, and CVCs or other foreign bodies
Modified from Wharton M, Chorba TL, Vogt RL, et al. Case definitions for public health surveil-
should be removed when possible. Associated endocarditis or other
lance. MMWR Recomm Rep 1990;39:1–43. endovascular infections always should be considered in patients with
S. aureus BSI because this species has a tropism for endothelial cells, and

702
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Staphylococcus aureus 115
organisms with the PVL gene have the propensity to cause thrombosis When it is inducible, however, the isolate tests as resistant to erythromy-
and thromboembolism.118,189,190 cin and susceptible to clindamycin.
If rapid improvement does not occur or BSI continues after more than Clinical laboratories test for inducible clindamycin resistance with the
24 hours of therapy, the therapeutic approach should be carefully disk diffusion D-test or with automated simultaneous broth dilution
reevaluated because irreversible organ damage can progress rapidly. Once testing in the presence of erythromycin (see Chapter 290 and Fig.
the antimicrobial susceptibility of the organism is known, therapy should 290.4).198,198 To date, although the incidence of erythromycin resistance
be tailored to use the appropriate antimicrobial agent or agents with the among CA-MRSA isolates is high, the incidence of clindamycin resistance
narrowest appropriate spectrum of activity. is not. Clinicians should be aware that treatment failure with clindamycin
The CA-MRSA epidemic has refocused attention on the importance is possible if the target organisms have demonstrable inducible resistance
of adjunctive local treatments, such as incision and drainage. For many in vitro.198,199
purulent SSTIs, incision and drainage may suffice. Guidelines have been Clindamycin also is a potent suppressor of bacterial toxin synthesis.
published for management involving antibiotics and drainage.98,191 Clindamycin therapy may be clinically superior in toxin-mediated
Because susceptibility to β-lactam antibiotics cannot be assumed, obtain- staphylococcal syndromes, although explicit data are lacking. When
ing a specimen for culture is essential for patients who require antibiotic clindamycin is used in this setting, combination therapy with oxacillin
treatment, to further guide therapy.192 or nafcillin, or with vancomycin, frequently is used.
Clindamycin is bacteriostatic and does not cross the blood-brain
Antistaphylococcal Agents barrier; it is not used alone in the treatment of endocarditis or other
intravascular or CNS infections that can occur during S. aureus invasive
The compounds presented in the following sections are discussed further infection. The poor taste of clindamycin suspensions can hamper adher-
in Chapter 292. ence to oral therapy in outpatients.
Clindamycin resistance rates among S. aureus isolates, both MRSA and
β-Lactam Antibiotics MSSA, should be monitored locally. In areas where the resistance rates
exceed 10% to 15%, empiric therapy with this agent probably should be
Previously, semisynthetic β-lactam penicillins (nafcillin, oxacillin, avoided.200–202
cloxacillin, and dicloxacillin), were the gold standard for treatment of
staphylococcal infections. These agents inhibit cell wall synthesis and thus
are bactericidal, well tolerated, and have a long record of therapeutic
Vancomycin
effectiveness. When an MSSA strain is isolated, β-lactams are the drugs Vancomycin is a glycopeptide that inhibits synthesis and assembly of the
of choice for serious staphylococcal infections. Parenteral compounds in second stage of cell wall peptidoglycan in gram-positive bacteria. It is
wide use in the U.S. include oxacillin and nafcillin; dicloxacillin is available the therapeutic agent of choice for HA-MRSA infections. However, the
for oral use, although some children find the taste of the suspension emergence of VISA and VRSA isolates, in addition to the so-called MIC
unpleasant. creep to 1 to 2 µg/mL, has the potential to limit the effectiveness of
The combination drugs made up of a β-lactam plus a β-lactamase vancomycin.203–205 Some experts recommend the use of higher doses in
inhibitor (i.e., (ampicillin-sulbactam, amoxicillin-clavulanate, ticarcillin- serious infections to elicit higher serum trough levels of vancomycin;
clavulanate, and piperacillin-tazobactam) are active against MSSA but however, the effectiveness, necessity, and safety of these strategies in
offer no advantage over the semisynthetic penicillins. The antistaphylo- children is controversial.98
coccal activity of the so-called first-generation cephalosporins is superior Vancomycin should be considered for children suspected of having
to that of later generations of cephalosporins. Cefazolin (for parenteral serious CA-MRSA infections, such as SSTIs with systemic manifestations
administration) and cephalexin (for oral administration) are useful class that require hospitalization, severe sepsis, TSS, CNS infection, necrotizing
compound examples; their efficacy is similar to that of the semisynthetic pneumonia, and necrotizing fasciitis. Considerations for empiric use for
penicillins for the treatment of MSSA infections that are not life threaten- osteoarticular infections include the local prevalence of CA-MRSA,
ing, and cefazolin is associated with fewer episodes of premature drug degree of illness, site of infection, and adequacy of culture specimens
discontinuation and drug-emergent events.193 obtained to guide therapy.
Until 2010 none of the β-lactam antibiotics, β-lactamase inhibitors, Because vancomycin is less bactericidal than oxacillin or nafcillin for
or cephalosporins had proved useful for therapy of MRSA infections. MSSA and can result in treatment failure or relapse, it should not be used
However, in 2010 the drug ceftaroline was licensed for treatment of for therapy of MSSA infections unless it is necessary (e.g., the patient has
CAP and complicated SSTIs. Designated a fifth-generation cephalospo- a type 1 allergic reaction to β-lactam agents). In severely ill patients,
rin, ceftaroline is active against MRSA strains and has broad-spectrum semisynthetic penicillin frequently is given in addition to vancomycin
activity against many gram-positive and gram-negative bacteria. Cef- until culture and susceptibility test results are available. Vancomycin is
taroline has been used as salvage therapy in adults with invasive MRSA available only in IV form for use in the therapy of S. aureus infections,
infections who have responded slowly to vancomycin. No data have and the concentration obtained in the alveolar space is poor. Oral van-
been reported on the drug’s use in children,194 although evaluation is comycin is not absorbed.
underway.
Oritavancin
Clindamycin Oritavancin is a semisynthetic lipoglycopeptide that was approved by the
Clindamycin is a lincosamide antibiotic that inhibits protein synthesis at FDA in 2014 for the treatment of SSTIs caused by gram-positive organ-
the chain elongation step by interfering with transpeptidation of the 50S isms. Oritavancin is active against MRSA, VISA, VRSA, daptomycin-
ribosomal subunit. Most HA-MRSA isolates are resistant to clindamycin, nonsusceptible isolates and vancomycin-resistant Enterococcus. Due to a
but most CA-MRSA isolates are susceptible,191 particularly in chil- long serum half-life (393 hours), concentration-dependent bactericidal
dren.60,77,98,195,196 Despite its increasingly frequent use in the treatment of activity, and a three-pronged mechanism of action, a single dose of ori-
CA-MRSA infections, clindamycin is not licensed by the U.S. Food and tivancin has been shown to be noninferior to repeated vancomycin
Drug Administration (FDA) for this purpose. dosing for treatment of SSTI in adults.206,207 Due to slow tissue elimina-
Erythromycin resistance of S. aureus is prevalent and can impair the tion, no oritivancin dosing adjustments are needed in patients with renal
effectiveness of clindamycin. The erythromycin ribosomal methylase or hepatic insufficiency.
(erm) gene codes for the enzyme that modifies the binding site not only In addition to achieving steric inhibition of transglycosylation,
for macrolides, but also for lincosamides and streptogramin B antibiotics. oritavancin can bind pentaglycyl cross-bridges, which inhibits pep-
Consequently, cross-resistance to these three classes of antibiotics, the tidoglycan transpeptidation and cross-linking. It interacts with the
so-called macrolide–lincosamide–streptogramin B (MLSB) phenotype, is bacterial cell membrane, leading to depolarization, increased membrane
common among S. aureus isolates.197 Phenotypic expression of the MLSB permeability, and cell death of both stationary and exponential growth
phenotype can be constitutive or inducible with exposure to a macrolide. phase bacteria. These mechanisms distinguish oritavancin from vanco-
When it is constitutive, the isolate tests as resistant to all MLSB antibiotics. mycin.208 Membrane depolarization also provides effectiveness against

703
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
PART III  Etiologic Agents of Infectious Diseases
SECTION A  Bacteria

daptomycin-nonsusceptible bacteria. To date, no oritavancin resistance inferior to vancomycin for MSSA and only equivalent to vancomycin
has been detected in clinical isolates of S. aureus. Lack of evaluation in for MRSA (these data were obtained before the CA-MRSA era221).
children and expense currently limit its use. The observation that thymidine can be released from injured tissue or
bacteria has suggested a possible explanation for discordant in vitro and
Dalbavancin clinical outcomes.222 Additional information is needed to establish the
appropriate role for TMP-SMX in the anti–S. aureus armamentarium.
Dalbavancin is a semisynthetic lipoglycopeptide derivative of teicoplanin.
It contains a long lipophilic side chain that extends its potency and
half-life. Dalbavancin has been approved by the FDA for the treatment
Tetracyclines (Including Tigecycline)
of SSTI caused by gram-positive organisms, and it is dosed once per Tetracyclines inhibit bacterial protein synthesis by binding to the 30S
week. Dalbavancin binds to the D-ala-D-ala residue, destabilizing the cell ribosomal subunit, thereby blocking the attachment of the transfer
membrane and resulting in bacterial cell death. Active against both MSSA RNA amino acid to the ribosome. Tetracyclines such as doxycycline and
and MRSA, dalbavancin is more potent than vancomycin against strains minocycline have emerged as alternative oral antimicrobial agents for
with a vancomycin MIC of 2 µg/mL or less. The kinetics in children 12 to mildly to moderately ill patients with MRSA SSTIs. Most isolates are
17 years of age appear to be similar to those in adults209; no data are avail- susceptible in vitro.191,223
able for younger children. Dalbavancin is noninferior to vancomycin and Tigecycline, a novel glycylcycline that was recently licensed for par-
linezolid. Because of its extended half-life, one or two doses of dalbavancin enteral use, has potent activity against S. aureus in vitro, regardless of
can be used after incision and drainage to treat SSTIs in ambulatory methicillin or vancomycin susceptibility.224,225 However, in 2010 the FDA
patients. The drug is very expensive and must be given parenterally. issued a warning linking tigecycline to an increased mortality risk in
patients being treated for hospital-acquired pneumonia, CAP, compli-
Linezolid and Tedizolid cated intra-abdominal infections, and complicated SSTIs; this suggests
that tigecycline should not be used in the treatment of S. aureus infections.
Linezolid and tedizolid are oxazolidinones that inhibit protein synthesis All tetracyclines are bacteriostatic and should not be used for the
by binding to the 50S ribosomal subunit during the formation of the treatment of endocarditis or other intravascular or CNS infections.
initiation complex.210 They have broad in vitro activity against MRSA, Tetracyclines also should not be used to treat pregnant women.
GISA, VISA, and VRSA isolates. They are well tolerated, and linezolid is as
effective as vancomycin in children with SSTIs and pneumonia caused by
MRSA.211–213 The oral bioavailability of both compounds is almost 100%,
Quinupristin-Dalfopristin
which facilitates transition from IV to oral therapy. The drug Synercid is a combination of two streptogramin antibiotics, one
Linezolid and tedizolid are bacteriostatic against S. aureus and therefore from group B (quinupristin) and one from group A (dalfopristin), in a
are not preferred for the treatment of endocarditis or other intravascular 30/70 ratio. Quinupristin-dalfopristin has good in vitro activity against
infections. If possible, linezolid should be avoided in patients taking S. aureus, including MRSA. The two streptogramins bind to sequential
adrenergic or serotonergic agents.214 Drug interactions may occur less sites on the 50S ribosomal subunit and have a synergistic bactericidal
frequently with tedizolid than with linezolid. Both compounds can be effect that results from inhibition of protein synthesis.226 Recent trials
associated (possibly to a lesser degree for tedizolid) with the development have shown a lower bacteriologic success rate compared with standard
of thrombocytopenia, which can be idiosyncratic and severe; anemia; therapy.227 Quinupristin-dalfopristin is not cross-resistant to S. aureus
and neutropenia.215 The complete blood count should be monitored in isolates with MLSB resistance because the streptogramin A component
patients receiving therapy for longer than 2 weeks, those with underlying remains active; however, few data are available to ensure the clinical
myelosuppression, or those taking other drugs that cause bone marrow validity of this issue. Quinupristin-dalfopristin is available only in IV
suppression. A small number of cases of optic and peripheral neuropathy form, and evaluation in children has been minimal. Infusion site inflam-
and lactic acidosis have been reported in adults during prolonged courses mation, thrombophlebitis, arthralgias, drug-drug interactions, myalgias,
of therapy with linezolid. gastrointestinal toxic effects, and increased serum unconjugated bilirubin
Resistance to linezolid has emerged in patients receiving longterm concentrations have limited widespread use.
therapy. Resistant isolates have mutations in the 23S rRNA gene in the
domain V region, of which S. aureus has five copies.216,217 A mutation in Daptomycin
two or more of the rRNA genes is required for the resistant phenotype.
This kind of resistance probably also occurs with tedizolid. Additional Daptomycin is a novel cyclic lipopeptide with a mechanism of action that
resistance mechanisms have been identified in ribosomal proteins L4 is not completely understood. It appears to bind to the gram-positive
and L3. Plasmid-borne cfr genes have been identified that encode a bacterial cell membrane, which is followed by calcium-dependent inser-
methyltransferase that mediates linezolid resistance by methylation of tion228 that results in a decrease in membrane potential, causing cell
carbon-8 on the 23S rRNA base. cfr genes confer cross-resistance to death by rapidly stopping DNA, RNA, and protein synthesis. It has potent
phenicols, lincosamides, pleuromutilins, and streptogramin A antibiot- bactericidal activity against MRSA.229 Daptomycin was comparable to
ics.218 Available data suggest that isolates with cfr genes are nonetheless vancomycin in the treatment of complicated SSTIs230; however, it showed
responsive to tedizolid therapy.219 decreased effectiveness for the treatment of CAP compared with standard
Both linezolid and tedizolid are available in IV and oral forms. Line- treatment, probably because of its propensity for binding surfactant.231
zolid is available in a convenient pediatric suspension. The compounds Daptomycin has been licensed by the FDA for the treatment of compli-
are extremely expensive, and negotiation with some third-party payers cated SSTIs, BSI, and right-sided endocarditis; however, it is not approved
may be required for each use. Patients also can experience delays in pro- for the treatment of left-sided endocarditis.232 Daptomycin is available
curing linezolid from private pharmacies that do not stock it routinely. A in IV form only. Studies evaluating its use in children suggest good
generic form of linezolid was recently released that costs about 20% less. efficacy.233 When the cell wall is thickened (e.g., in a VISA isolate or after
exposure of an S. aureus isolate to vancomycin), resistance to daptomycin
Trimethoprim-Sulfamethoxazole becomes evident, presumably because the compound cannot reach its
cell membrane target.234,235 Resistance to daptomycin can occur during
TMP-SMX interferes with the biosynthesis of tetrahydrofolic acid, which therapy and was documented in 6 of 120 patients receiving daptomycin
is essential for microbial synthesis of proteins and nucleotides. Although for endocarditis, or BSI.236 In addition, daptomycin-resistant isolates can
most S. aureus isolates are susceptible in vitro, TMP-SMX was seldom be cross-resistant to vancomycin.235
used to treat S. aureus infections until the recent epidemic of CA-MRSA.
Reports now are available on clinical experience using TMP-SMX to treat
CA-MRSA SSTIs220 or more severe S. aureus infections. One study sug-
Telavancin
gested that the efficacy of TMP-SMX was similar to that of clindamycin Telavancin is a lipoglycopeptide that was licensed by the FDA in 2009 for
for skin infections of unspecified cause, including cellulitis.220 For the the treatment of complicated SSTIs. The compound depolarizes bacterial
treatment of invasive S. aureus infection, data show IV TMP-SMX to be membranes and inhibits cell wall formation. No formal evaluation has

704
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Staphylococcus aureus 115
Empiric therapy for S. aureus infection

MILDLY ILL MODERATELY ILL SEVERELY ILL


Afebrile, previously healthy Febrile, previously healthy Febrile, previously healthy,
ill-appearing or
SSTI SSTI immunocompromised
Bone and joint infection
Pulmonary infection SSTI or SSTI complicated
by necrotizing fasciitis
Bone and joint infection
Pulmonary infection including
Incision and drainage necrotizing pneumonia
Hospitalize
+/– Endovascular infection
I&D as appropriate
Oral antimicrobial therapy Severe sepsis
+
Clindamycin Toxic shock syndrome
Parenteral antimicrobial therapy
or Clindamycin (in areas where
TMP-SMX resistance/D-test positivity rate low)
or or Hospitalize
Doxycycline or Vancomycin (in areas where clindamycin I&D as appropriate
Minocycline resistance/D-test positivity rate high) Surgical evaluation and
(>8 years)
intervention if needed
+
Parenteral antimicrobial therapy
Vancomycin
±
Clindamycin
±
Gentamicin
±
IGIV

FIGURE 115.9  Schema for the empiric treatment of community-associated S. aureus infections, determined by the severity of the illness. SSTI, Skin and soft tissue
infection; I&D, incision and drainage; TMP-SMX, trimethoprim-sulfamethoxazole; IGIV, immune globulin for intravenous use.

been done in children. The drug is administered once daily. Telavancin in vitro test results. A study sponsored by the National Institutes of
carries a “black box” warning that it worsens renal dysfunction. As can Health is underway to determine the optimal oral therapy of S. aureus
vancomycin, telavancin can cause red man syndrome. A few resistant large-lesion SSTI in outpatients.220 A similar study comparing clindamy-
isolates have been, obtained although the mechanism is not known. cin, TMP-SMX and placebo therapies for small abscess will be published
(Daum R, et al. In press).
Rifampin
Rifampin is a bactericidal agent that blocks protein synthesis by inhibiting Empiric Therapy for MRSA Infections Requiring
RNA polymerase. It is highly active against almost all S. aureus isolates,
but resistance emerges rapidly when rifampin is used as monotherapy.
Hospitalization
Combination regimens using rifampin with clindamycin, TMP-SMX, or Vancomycin is indicated for empiric therapy of putative S. aureus infec-
doxycycline sometimes are recommended for the treatment of CA-MRSA tions when the patient is severely ill.98,237 VISA isolates have been identified
infections or for treating device- or prosthesis-associated S. aureus infec- that are associated with vancomycin treatment failure.238,239 The Clinical
tions. Whether such regimens are more effective is unclear. Rifampin is and Laboratory Standards Institute defines VISA as an MIC of vancomycin
available in both oral and IV forms, although a suspension suitable for greater than 2 µg/mL. In 2002 the CDC reported the first documented
use in young children is not available. infection caused by S. aureus with high-level resistance (MIC >16 µg/mL)
to vancomycin, or VRSA; 12 additional VRSA strains have been isolated
Aminoglycosides since.87–89,240,241 VISA and VRSA isolates probably occur more frequently
than is documented because optimal tests to detect decreased vancomycin
Aminoglycosides (e.g., gentamicin) are potent bactericidal inhibitors susceptibility often are not performed in clinical laboratories.242
of protein synthesis. Aminoglycosides alone are believed to be poorly Information about parenteral therapy with clindamycin, daptomycin,
efficacious against S. aureus infections, although they are synergistic with tigecycline, linezolid, telavancin, and other agents useful in the therapy
β-lactam antibiotics and vancomycin in vitro. They are commonly used of MRSA infections in hospitalized children can be found in Chapter 292
in combination therapy for severe infections, such as endocarditis, but and in the guidelines established by the Infectious Diseases Society of
whether this offers a clinical advantage is the subject of controversy. America.98,243

Empiric Therapy for Community- PREVENTION


Associated Infections Efforts to prevent S. aureus HAIs have included general measures such as
improved hand hygiene and attention to careful implementation of
CA-MSSA and CA-MRSA skin infections cannot be differentiated on measures aimed at diminishing infections associated with intravascular
clinical grounds.192 In areas where CA-MRSA is prevalent, empiric catheters. Other efforts aimed specifically at reducing infections caused by
β-lactam therapy is not appropriate. Fig. 115.9 suggests an approach to MRSA have included identification of MRSA carriers upon hospitaliza-
the management of these cases. Therapy should be adjusted according to tion, and isolation and cohorting of patients who are colonized. Contact

705
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Precautions are used in the care of patients with a MRSA infection. Some 4-component vaccine, including the capsular antigens manufactured by
states have legislated mandatory screening for MRSA upon admission to Pfizer, currently is enrolling adults undergoing elective spinal surgery.
the hospital or, in some cases, to an intensive care unit. However, such a In an initial trial, attempts to protect neonates passively with a high-
screening approach has several problems: high cost, focusing finite titer, anti–clumping factor A polyclonal antibody product (Veronate)
resources on a single pathogen, lack of agreement on best practices to appeared to have promising results; however, a confirmatory trial did not
determine MRSA carriage, lack of evidence-based management when confirm efficacy (Hetherington S. Personal communication, 2006.). A
colonization is found, and the insignificant impact of an effective clinical trial using a monoclonal antibody (pagibaximab) directed against
institution-based strategy on MRSA infections in the community.243 lipoteichoic acid was conducted among small premature infants in an
In the community setting, some sports teams have increased efforts to attempt to prevent infections caused by S. aureus and coagulase-negative
schedule equipment washing, discourage towel sharing, restrict infected staphylococci; however, efficacy was lacking.
members from play until lesions have healed, and encourage good Recent attention has focused on the role of the Th17/IL-17 lymphocyte
hygiene among team members.244,245 Several correctional facilities have pathway.255 One candidate vaccine (rAls3p-N) derived from a Candida
introduced practices to reduce the spread of CA-MRSA, as recommended adhesin protected mice against bacteremic challenge. Efficacy was
by the Federal Bureau of Prisons.72 Chlorhexidine-impregnated cloths retained in mice that were B-lymphocyte deficient but not in those that
were used successfully in prisons to reduce the density of skin coloniza- were T-lymphocyte deficient. Adoptive transfer of CD4+ lymphocytes,
tion,246 and they are frequently are used preoperatively; however, this but not B220+ lymphocytes, transferred immunity. The vaccine appar-
approach was unsuccessful in preventing SSTIs among military recruits.247 ently stimulates proinflammatory Th1, Th17, and Th1/Th17 lympho-
MRSA decolonization has been attempted in outbreak situations and cytes, and these cytokines enhance neutrophil recruitment and activation.
to prevent recurrent SSTI, with little success. Few randomized, controlled As of February, 2015, the manufacturer had yet to decide whether to
trials have evaluated the efficacy of topical antibiotics and skin antisepsis evaluate this vaccine against S. aureus in an efficacy trial.
in the eradication of S. aureus carriage.99,248–251 Intranasal mupirocin
ointment appears to have the most success in reducing previous All references are available online at www.expertconsult.com.
HA-MRSA nasal colonization, and it has shown a subsequent decrease
in carriage at other body sites. A 5-day application of intranasal mupi-
rocin resulted in decreased short-term rates of colonization, but recolo- KEY REFERENCES
nization occurred in at least one half of the treated subjects, usually 43. Kumar N, David MZ, Boyle-Vavra S, et al. High Staphylococcus aureus colonization
within a few months.99 Unacceptably high recolonization rates, concern prevalence among patients with skin and soft tissue infections and controls in an
about mupirocin resistance,252,253 and limited data about the drug’s urban emergency department. J Clin Microbiol 2015;53:810–815.
effectiveness against CA-MRSA prevent intranasal mupirocin from being 97. Talan DA, Mower WR, Krishnahasan F, et al. Trimethoprim-sulfamethoxazole
recommended for routine use. Focusing on reducing the density of colo- versus placebo for uncomplicated skin abscess. N Engl J Med 2016;374:823–832.
120. Peltola H, Pääkkönen M. Acute osteomyelitis in children. N Engl J Med
nization and environmental contamination, as well as improving
2014;370:352–360.
underlying skin conditions, may be more prudent. 121. Keren R, Shah SS, Srivastava R, et al. Comparative effectiveness of intravenous
vs oral antibiotics for postdischarge treatment of acute osteomyelitis in children.
Vaccine Development JAMA Pediatr 2015;169:120–128.
149. Baltimore RS, Gewitz M, Baddour LM, et al. Infective endocarditis in childhood
A vaccine to prevent S. aureus infection remains a formidable challenge. 2015. Update: a scientific statement from the American Heart Association. Circula-
tion 2015;132:1487–1515.
Experts disagree on whether S. aureus infections can be prevented by 192. Youngster I, Shenoy ES, Hooper ES, et al. Comparative evaluation of the tolerability
vaccination and, if so, whether such a vaccine should be deployed uni- of cefazolin and nafcillin for treatment of methicillin-susceptible Staphylococcus
versally or targeted to high-risk groups. aureus infections in the outpatient setting. Clin Infect Dis 2014;59:369–375.
The traditional idea that production of an opsonophagocytic antibody 209. Bradley JS, Puttagunta S, Rubino CM, et al. Pharmacokinetics, safety and toler-
(or antibodies) correlates with protection has been challenged by available ability of single dose dalbavancin in children 12–17 years of age. Pediatr Infect Dis
J 2015;34:748–752.
data. In two vaccine trials of types 5 and 8 polysaccharide conjugate 220. Miller LG, Daum RS, Creech CB, et al. Clindamycin versus trimethoprim-
vaccine, immunization failed to provide protection against BSI.254 Recently sulfamethoxazole for uncomplicated skin infections. N Engl J Med
a protein vaccine containing an iron-scavenging, cell wall–anchored 2015;372:1093–1103.
protein (ISDB) was developed. Enrollees in the study of this vaccine were 243. Stevens DL, Bisno AL, Chambers HF. Practice guidelines for the diagnosis and
adults undergoing elective cardiac surgery that required a sternotomy. The management of skin and soft tissue infections: 2014 update by the Infectious
Diseases Society of America. Clin Infect Dis 2014;59:147–159.
clinical end point was the occurrence of S. aureus mediastinitis or bactere-
246. David MZ, Siegel JD, Henderson J, et al. A randomized, controlled trial of
mia. However, the study was halted for lack of efficacy and because of chlorhexidine-soaked cloths to reduce methicillin-resistant and methicillin-
concern that vaccine recipients who developed S. aureus invasive infection susceptible Staphylococcus aureus carriage prevalence in an urban jail. Infect
had a worse outcome than placebo recipients.16 A clinical trial of a Control Hosp Epidemiol 2014;35:1466–1473.

Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Staphylococcus aureus 115
REFERENCES 34. Lillibridge CB, Melish ME, Glasgow LA. Site of action of exfoliative toxin in the
staphylococcal scalded-skin syndrome. Pediatrics 1972;50:728–738.
1. O’Riordan K, Lee JC. Staphylococcus aureus capsular polysaccharides. Clin Micro- 35. Baba T, et al. Genome and virulence determinants of high virulence community-
biol Rev 2004;17:218–234. acquired MRSA. Lancet 2002;359:1819–1827.
2. David MZ, Daum RS. Community associated methicillin-resistant Staphylococcus 36. Chambers HF. Methicillin-resistant staphylococci. Clin Microbiol Rev
aureus: epidemiology and clinical consequences of an emerging epidemic. Clin 1988;1:173–186.
Microbiol Rev 2010;23:616–687. 37. Pinho MG, de Lencastre H, Tomasz A. An acquired and a native penicillin-binding
3. Roghmann M, et al. Epidemiology of capsular and surface polysaccharide in Staph- protein cooperate in building the cell wall of drug-resistant staphylococci. Proc Natl
ylococcus aureus infections complicated by bacteremia. J Hosp Infect 2005;59:27–32. Acad Sci USA 2001;98:10886–10891.
4. Shinefield H, et al. Use of a Staphylococcus aureus conjugate vaccine in patients 38. Chongtrakool P, Ito T, Ma XX, et al. Staphylococcal cassette chromosome mec
receiving hemodialysis. N Engl J Med 2002;346:491–496. (SCCmec) typing of methicillin-resistant Staphylococcus aureus strains isolated in
5. Shinefield HR, Black S. Prospects for active and passive immunization against 11 Asian countries: a proposal for a new nomenclature for SCCmec elements.
Staphylococcus aureus. Pediatr Infect Dis J 2006;25:167–168. Antimicrob Agents Chemother 2006;50:1001–1012.
6. Fattom A, et al. Efficacy profile of a bivalent Staphylococcus aureus glycoconjugated 39. Maree CL, Daum RS, Boyle-Vavra S, et al. Community-associated methicillin-
vaccine in adults on hemodialysis: phase III randomized study. Hum Vaccin Immu- resistant Staphylococcus aureus isolates causing healthcare-associated infections.
nother 2015;11:632–641. Emerg Infect Dis 2007;13:236–242.
7. Cheung AL, et al. Analysis by gel electrophoresis, Western blot, and peptide 40. Daum RS, et al. A novel methicillin-resistance cassette in community-acquired
mapping of protein A heterogeneity in Staphylococcus aureus strains. Infect Immun methicillin-resistant Staphylococcus aureus isolates of diverse genetic backgrounds.
1987;55:843–847. J Infect Dis 2002;186:1344–1347.
8. Peterson PK, et al. Effect of protein A on staphylococcal opsonization. Infect 41. Okuma K, et al. Dissemination of new methicillin-resistant Staphylococcus aureus
Immun 1977;15:760–764. clones in the community. J Clin Microbiol 2002;40:4289–4294.
9. Verbrugh HA, et al. The role of Staphylococcus aureus cell-wall peptidoglycan, 42. Robinson DA, Enright MC. Evolutionary models of the emergence of methicillin-
teichoic acid and protein A in the processes of complement activation and opso- resistant Staphylococcus aureus. Antimicrob Agents Chemother 2003;47:3926–3934.
nization. Immunology 1979;37:615–621. 43. Kumar N, David MZ, Boyle-Vavra S, et al. High Staphylococcus aureus colonization
10. Verhoef J, Peterson PK, Verbrugh HA. Host-parasite relationship in staphylococcal prevalence among patients with skin and soft tissue infections and controls in an
infections: the role of the staphylococcal cell wall during the process of phagocy- urban emergency department. J Clin Microbiol 2015;53:810–815.
tosis. Antonie Van Leeuwenhoek 1979;45:49–53. 44. Kluytmans J, van Belkum A, Verbrugh H. Nasal carriage of Staphylococcus aureus:
11. Wann ER, Gurusiddappa S, Hook M. The fibronectin-binding MSCRAMM FnbpA epidemiology, underlying mechanisms, and associated risks. Clin Microbiol Rev
of Staphylococcus aureus is a bifunctional protein that also binds to fibrinogen. J 1997;10:505–520.
Biol Chem 2000;275:13863–13871. 45. Davis KA, et al. Methicillin-resistant Staphylococcus aureus (MRSA) nares coloniza-
12. Cheung AI, et al. Cloning, expression, and nucleotide sequence of a Staphylococ- tion at hospital admission and its effect on subsequent MRSA infection. Clin Infect
cus aureus gene (fbpA) encoding a fibrinogen-binding protein. Infect Immun Dis 2004;39:776–782.
1995;63:1914–1920. 46. Huang SS, Platt R. Risk of methicillin-resistant Staphylococcus aureus infection after
13. McDevitt D, et al. Molecular characterization of the clumping factor (fibrinogen previous infection or colonization. Clin Infect Dis 2003;36:281–285.
receptor) of Staphylococcus aureus. Mol Microbiol 1994;11:237–248. 47. Williams RE. Healthy carriage of Staphylococcus aureus: its prevalence and impor-
14. Loffler B, Hussain M, Grundmeier M, et al. Staphylococcus aureus Panton-Valentine tance. Bacteriol Rev 1963;27:56–71.
leukocidin is a very potent cytotoxic factor for human neutrophils. PLoS Pathog 48. Klevens RM, et al. Changes in the epidemiology of methicillin-resistant Staphy-
2010;6:e1000715. lococcus aureus in intensive care units in US hospitals, 1992–2003. Clin Infect Dis
15. Skaar EP, et al. Iron-source preference of Staphylococcus aureus infections. Science 2006;42:389–391.
2004;305:1626–1628. 49. Hidron AL, Edwards JR, Patel J, et al. National Healthcare Safety Network Team;
16. Fowler VG, Allen KB, Moreira ED, et al. Effect of an investigational vaccine for Participating National Healthcare Safety Network Facilities. NHSN annual update:
preventing Staphylococcus aureus infections after cardiothoracic surgery: a random- antimicrobial-resistant pathogens associated with healthcare-associated infections:
ized trial. JAMA 2013;309:1368–1378. annual summary of data reported to the National Healthcare Safety Network at
17. Bhakdi S, Tranum-Jensen J. Alpha-toxin of Staphylococcus aureus. Microbiol Rev the Centers for Disease Control and Prevention, 2006–2007. Infect Control Hosp
1991;55:733–751. Epidemiol 2008;29:996–1011.
18. Finck-Barbancon V, et al. Pore formation by a two-component leukocidin from 50. Richards MJ, et al. Nosocomial infections in pediatric intensive care units in the
Staphylococcus aureus within the membrane of human polymorphonuclear leuko- United States. National Nosocomial Infections Surveillance System. Pediatrics
cytes. Biochim Biophys Acta 1993;1182:275–282. 1999;103:e39.
19. Goguen JD, Hoe NP, Subrahmanyam YV. Proteases and bacterial virulence: a view 51. Kennedy LA, Gill JA, Schultz ME, et al. Inside out: the changing epidemiology
from the trenches. Infect Agents Dis 1995;4:47–54. of methicillin-resistant Staphylococcus aureus. Infect Control Hosp Epidemiol
20. Kapral FA, Smith S, Lal D. The esterification of fatty acids by Staphylococcus aureus 2010;31:983–985.
fatty acid modifying enzyme (FAME) and its inhibition by glycerides. J Med 52. Kallen AJ, Mu Y, Bulens S, et al. Health care-associated invasive MRSA infections,
Microbiol 1992;37:235–237. 2005–2008. JAMA 2010;304:641–648.
21. Lee CY, Iandolo JJ. Lysogenic conversion of staphylococcal lipase is caused by inser- 53. Saravolatz LD, et al. Methicillin-resistant Staphylococcus aureus: epide-
tion of the bacteriophage L54a genome into the lipase structural gene. J Bacteriol miologic observations during a community-acquired outbreak. Ann Intern Med
1986;166:385–391. 1982;96:11–16.
22. Mortensen JE, Shryock TR, Kapral FA. Modification of bactericidal fatty acids by 53. Craven DE, et al. Methicillin-resistant Staphylococcus aureus bacteremia linked
an enzyme of Staphylococcus aureus. J Med Microbiol 1992;36:293–298. to intravenous drug abusers using a “shooting gallery”. Am J Med 1986;80:
23. Wadstrom T, Mollby R. Studies on extracellular proteins from Staphylococcus 770–776.
aureus. VII. Studies on ß-haemolysin. Biochim Biophys Acta 1971;242:308–320. 55. Levine DP, et al. Community-acquired methicillin-resistant Staphylococcus aureus
24. O’Reilly M, et al. Inactivation of the alpha-haemolysin gene of Staphylococcus endocarditis in the Detroit Medical Center. Ann Intern Med 1982;97:330–338.
aureus 8325–4 by site-directed mutagenesis and studies on the expression of its 56. Saravolatz LD, Pohlod DJ, Arking LM. Community-acquired methicillin-resistant
haemolysins. Microb Pathog 1986;1:125–138. Staphylococcus aureus infections: a new source for nosocomial outbreaks. Ann
25. Prevost G, et al. Staphylococcal pore-forming toxins. Curr Top Microbiol Immunol Intern Med 1982;97:325–329.
2001;257:53–83. 57. Shahin R, et al. Methicillin-resistant Staphylococcus aureus carriage in a child care
26. Prevost G, et al. Panton-Valentine leucocidin and gamma-hemolysin from Staphy- center following a case of disease. Toronto Child Care Center Study Group. Arch
lococcus aureus ATCC 49775 are encoded by distinct genetic loci and have different Pediatr Adolesc Med 1999;153:864–868.
biological activities. Infect Immun 1995;63:4121–4129. 58. Chambers HF. The changing epidemiology of Staphylococcus aureus? Emerg Infect
27. Konig B, et al. Effects of Staphylococcus aureus leukocidins on inflammatory media- Dis 2001;7:178–182.
tor release from human granulocytes. J Infect Dis 1995;171:607–613. 59. Gorak EJ, Yamada SM, Brown JD. Community-acquired methicillin-resistant
28. Lina G, et al. Involvement of Panton-Valentine leukocidin–producing Staphylococcus aureus in hospitalized adults and children without known risk
Staphylococcus aureus in primary skin infections and pneumonia. Clin Infect Dis factors. Clin Infect Dis 1999;29:797–800.
1999;29:1128–1132. 60. Herold BC, et al. Community-acquired methicillin-resistant Staphylococcus aureus
29. Gillet Y, et al. Association between Staphylococcus aureus strains carrying gene for in children with no identified predisposing risk. JAMA 1998;279:593–598.
Panton-Valentine leukocidin and highly lethal necrotizing pneumonia in young 61. Hussain FM, et al. Current trends in community-acquired methicillin-resistant
immunocompetent patients. Lancet 2002;359:753–759. Staphylococcus aureus at a tertiary care pediatric facility. Pediatr Infect Dis J
30. Voyich JM, Otto M, Mathema B, et al. Is Panton-Valentine leukocidin the major 2000;19:1163–1166.
virulence determinant in community-associated methicillin-resistant Staphylococ- 62. Hussain FM, Boyle-Vavra S, Daum RS. Community-acquired methicillin-resistant
cus aureus disease? J Infect Dis 2006;194:1761–1770. Staphylococcus aureus colonization in healthy children attending an outpatient
31. Labandeira-Rey M, Cavzon F, Buisset S, et al. Staphylococcus aureus Panton- pediatric clinic. Pediatr Infect Dis J 2001;20:763–767.
Valentine leukocidin causes necrotizing pneumonia. Science 2007;315:1130–1133. 63. Centers for Disease Control and Prevention. Four pediatric deaths from
32. Boyle-Vavra S, Daum RS. Community-acquired methicillin-resistant Staphylococ- community-acquired methicillin-resistant Staphylococcus aureus – Minnesota and
cus aureus: the role of Panton-Valentine leukocidin. Lab Invest 2007;87:3–9. North Dakota, 1997–1999. JAMA 1999;282:1123–1125.
33. Hanakawa Y, Stanley JR. Mechanisms of blister formation by staphylococcal toxins. 64. Fridkin SK, et al. Methicillin-resistant Staphylococcus aureus disease in three com-
J Biochem 2004;136:747–750. munities. N Engl J Med 2005;352:1436–1444.

706.e1
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
PART III  Etiologic Agents of Infectious Diseases
SECTION A  Bacteria

65. Kowalski TJ, Berbari EF, Osmon DR. Epidemiology, treatment, and prevention of 99. Laupland KB, Conly JM. Treatment of Staphylococcus aureus colonization and
community-acquired methicillin-resistant Staphylococcus aureus infections. Mayo prophylaxis for infection with topical intranasal mupirocin: an evidence-based
Clin Proc 2005;80:1201–1207, quiz, 1208. review. Clin Infect Dis 2003;37:933–938.
66. Salgado CD, Farr BM, Calfee DP. Community-acquired methicillin-resistant 100. Swartz MN. Cellulitis. N Eng J Med 2004;39:904–912.
Staphylococcus aureus: a meta-analysis of prevalence and risk factors. Clin Infect 101. Wald ER. Periorbital and orbital infections. Pediatr Rev 2004;25:312–320.
Dis 2003;36:131–139. 102. Smith TF, O’Day D, Wright PF. Clinical implications of preseptal (periorbital)
67. David MZ, Glikman D, Crawford SE, et al. What is community-associated cellulitis in childhood. Pediatrics 1978;62:1006–1009.
methicillin-resistant Staphylococcus aureus? J Infect Dis 2008;197:1235–1243. 103. Lessner A, Stern GA. Preseptal and orbital cellulitis. Infect Dis Clin North Am
68. Adcock PM, et al. Methicillin-resistant Staphylococcus aureus in two child care 1992;6:933–952.
centers. J Infect Dis 1998;178:577–580. 104. Goytia VK, Giannoni CM, Edwards MS. Intraorbital and intracranial extension of
69. Kazakova SV, et al. A clone of methicillin-resistant Staphylococcus aureus among sinusitis: comparative morbidity. J Pediatr 2011;158:486–491.
professional football players. N Engl J Med 2005;352:468–475. 105. Price CD, Hameroff SB, Richards RD. Cavernous sinus thrombosis and orbital
70. Lindenmayer JM, et al. Methicillin-resistant Staphylococcus aureus in a high cellulitis. South Med J 1971;64:1243–1247.
school wrestling team and the surrounding community. Arch Intern Med 106. Moloney JR, Badham NJ, McRae A. The acute orbit: preseptal (periorbital) cellulitis,
1998;158:895–899. subperiosteal abscess and orbital cellulitis due to sinusitis. J Laryngol Otol Suppl
71. Pan ES, et al. Increasing prevalence of methicillin-resistant Staphylococcus aureus 1987;12:1–18.
infection in California jails. Clin Infect Dis 2003;37:1384–1388. 107. Shrader SK, et al. The clinical spectrum of endophthalmitis: incidence, predispos-
72. Centers for Disease Control and Prevention. Methicillin-resistant Staphylococ- ing factors, and features influencing outcome. J Infect Dis 1990;162:115–120.
cus aureus infections in correctional facilities – Georgia, California, and Texas, 108. Recchia FM, et al. Endophthalmitis after pediatric strabismus surgery. Arch Oph-
2001–2003. MMWR Morb Mortal Wkly Rep 2003;52:992–996. thalmol 2000;118:939–944.
73. Campbell KM, et al. Risk factors for community-associated methicillin-resistant 109. Waldvogel FA, Medoff G, Swartz MN. Osteomyelitis: a review of clinical features,
Staphylococcus aureus infections in an outbreak of disease among military trainees therapeutic considerations and unusual aspects. Part 3. Osteomyelitis associated
in San Diego, California, in 2002. J Clin Microbiol 2004;42:4050–4053. with vascular insufficiency. N Engl J Med 1970;282:316–322.
74. Zinderman CE, et al. Community-acquired methicillin-resistant Staphylococcus 110. Waldvogel FA, Medoff G, Swartz MN. Osteomyelitis: a review of clinical
aureus among military recruits. Emerg Infect Dis 2004;10:941–944. features, therapeutic considerations and unusual aspects. Part 2. N Engl J Med
75. Buckingham SC, et al. Emergence of community-associated methicillin-resistant 1970;282:260–266.
Staphylococcus aureus at a Memphis, Tennessee, children’s hospital. Pediatr Infect 111. Waldvogel FA, Medoff G, Swartz MN. Osteomyelitis: a review of clinical features,
Dis J 2004;23:619–624. therapeutic considerations and unusual aspects. N Engl J Med 1970;282:198–206.
76. Chavez-Bueno S, et al. Inducible clindamycin resistance and molecular epidemio- 112. Waldvogel FA, Papageorgiou PS. Osteomyelitis: the past decade. N Engl J Med
logic trends of pediatric community-acquired methicillin-resistant Staphylococcus 1980;303:360–370.
aureus in Dallas, Texas. Antimicrob Agents Chemother 2005;49:2283–2288. 113. Kahn DS, Pritzker KP. The pathophysiology of bone infection. Clin Orthop Relat
77. Naimi TS, et al. Comparison of community-and health care-associated methicillin- Res 1973;12–19.
resistant Staphylococcus aureus infection. JAMA 2003;290:2976–2984. 114. Frank G, Mahoney HM, Eppes SC. Musculoskeletal infections in children. Pediatr
78. Narita S, et al. Phage conversion of Panton-Valentine leukocidin in Staphy- Clin North Am 2005;52:1083–1106.
lococcus aureus: molecular analysis of a PVL-converting phage, phiSLT. Gene 115. Bonhoeffer J, et al. Diagnosis of acute haematogenous osteomyelitis and septic
2001;268:195–206. arthritis: 20 years experience at the University Children’s Hospital Basel. Swiss Med
79. Francis JS, et al. Severe community-onset pneumonia in healthy adults caused by Wkly 2001;131:575–581.
methicillin-resistant Staphylococcus aureus carrying the Panton-Valentine leukoci- 116. Unkila-Kallio L, et al. Serum C-reactive protein, erythrocyte sedimentation rate,
din genes. Clin Infect Dis 2005;40:100–107. and white blood cell count in acute hematogenous osteomyelitis of children.
80. Gonzalez BE, et al. Pulmonary manifestations in children with invasive community- Pediatrics 1994;93:59–62.
acquired Staphylococcus aureus infection. Clin Infect Dis 2005;41:583–590. 117. Elgazzar AH, et al. Multimodality imaging of osteomyelitis. Eur J Nucl Med
81. Mongkolrattanothai K, et al. Severe Staphylococcus aureus infections caused by 1995;22:1043–1063.
clonally related community-acquired methicillin-susceptible and methicillin- 118. Crary SE, Buchanan GR, Drake CE, et al. Venous thrombosis and thromboembo-
resistant isolates. Clin Infect Dis 2003;37:1050–1058. lism in children with osteomyelitis. J Pediatr 2006;149:537–541.
82. Thurlow LR, Joshi GS, Clark JR, et al. Functional modularity of the arginine 119. Le Saux N, et al. Shorter courses of parenteral antibiotic therapy do not appear
catabolic mobile element contributes to the success of USA300 methicillin-resistant to influence response rates for children with acute hematogenous osteomyelitis: a
Staphylococcus aureus. Cell Host Microbe 2013;13:100–107. systematic review. BMC Infect Dis 2002;2:16.
83. Joshi GS, Spontak JS, Klapper DG, Richardson AR. Arginine catabolic mobile 120. Peltola H, Pääkkönen M. Acute osteomyelitis in children. N Engl J Med
element encoded speG abrogates the unique hypersensitivity of Staphylococcus 2014;370:352–360.
aureus to exogenous polyamines. Mol Microbiol 2011;82:9–20. 121. Keren R, Shah SS, Srivastava R, et al. Comparative effectiveness of intravenous
84. Montgomery CP, Boyle-Vavra S, Daum RS. The arginine catabolic mobile element vs oral antibiotics for postdischarge treatment of acute osteomyelitis in children.
is not associated with enhanced virulence in experimental invasive disease caused JAMA Pediatr 2015;169:120–128.
by the community-associated methicillin-resistant Staphylococcus aureus USA300 122. Wald ER. Risk factors for osteomyelitis. Am J Med 1985;78:206–212.
genetic background. Infect Immun 2009;77:2650–2656. 123. Fernandez M, Carrol CL, Baker CJ. Discitis and vertebral osteomyelitis in children:
85. Appelbaum PC. The emergence of vancomycin-intermediate and vancomycin- an 18-year review. Pediatrics 2000;105:1299–1304.
resistant Staphylococcus aureus. Clin Microbiol Infect 2006;12(suppl 1):16–23. 124. Morrissy RT. Bone and joint infection in the neonate. Pediatr Ann 1989;18:33–34,
86. Linares J. The VISA/GISA problem: therapeutic implications. Clin Microbiol Infect 36–38, 40–44.
2001;7(suppl 4):8–15. 125. Ogden JA, Lister G. The pathology of neonatal osteomyelitis. Pediatrics
87. Centers for Disease Control and Prevention. Staphylococcus aureus resistant to van- 1975;55:474–478.
comycin – United States, 2002. MMWR Morb Mortal Wkly Rep 2002;51:565–567. 126. Chen CE, et al. Acute septic arthritis of the hip in children. Arch Orthop Trauma
88. Centers for Disease Control and Prevention. Vancomycin-resistant Staphylococcus Surg 2001;121:521–526.
aureus – Pennsylvania, 2002. MMWR Morb Mortal Wkly Rep 2002;51:902. 127. Coticchia JM, Dohar JE. Methicillin-resistant Staphylococcus aureus otor-
89. Centers for Disease Control and Prevention. Vancomycin-resistant Staphylococcus rhea after tympanostomy tube placement. Arch Otolaryngol Head Neck Surg
aureus – New York, 2004. MMWR Morb Mortal Wkly Rep 2004;53:322–323. 2005;131:868–873.
90. Melish ME, Glasgow LA, Turner MD. The staphylococcal scalded-skin syndrome: 128. Hwang JH, Chu CK, Liu TC. Changes in bacteriology of discharging ears. J Laryngol
isolation and partial characterization of the exfoliative toxin. J Infect Dis Otol 2002;116:686–689.
1972;125:129–140. 129. Donnelly BW, McMillan JA, Weiner LB. Bacterial tracheitis: report of eight new
91. Yamasaki O, et al. Clinical manifestations of staphylococcal scalded-skin syndrome cases and review. Rev Infect Dis 1990;12:729–735.
depend on serotypes of exfoliative toxins. J Clin Microbiol 2005;43:1890–1893. 130. Nickerson CL, Jakab GJ. Pulmonary antibacterial defenses during mild and severe
92. Daum RS, Kar S, Kirkpatrick P. Retapamulin. Nat Rev Drug Discov 2007;6:865–866. influenza virus infection. Infect Immun 1990;58:2809–2814.
93. Koning S, et al. Interventions for impetigo. Cochrane Database Syst Rev 131. Holmes A, et al. Staphylococcus aureus isolates carrying Panton-Valentine leucoci-
2004;(1):CD003261. din genes in England and Wales: frequency, characterization, and association with
94. Rudoy RC, Nelson JD. Breast abscess during the neonatal period: a review. Am J clinical disease. J Clin Microbiol 2005;43:2384–2390.
Dis Child 1975;129:1031–1034. 132. Peleg AY, Munckhof WJ. Fatal necrotising pneumonia due to community-acquired
95. Lee MC, et al. Management and outcome of children with skin and soft tissue methicillin-resistant Staphylococcus aureus (MRSA). Med J Aust 2004;181:
abscesses caused by community-acquired methicillin-resistant Staphylococcus 228–229.
aureus. Pediatr Infect Dis J 2004;23:123–127. 133. Petros S, et al. Severe community acquired pneumonia due to Staphylococcus
96. Llera JL, Levy RC. Treatment of cutaneous abscess: a double-blind clinical study. aureus. Intensive Care Med 1998;24:189.
Ann Emerg Med 1985;14:15–19. 134. Tseng MH, et al. Fatal sepsis and necrotizing pneumonia in a child due to
97. Talan DA, Mower WR, Krishnahasan F, et al. Trimethoprim-sulfamethoxazole community-acquired methicillin-resistant Staphylococcus aureus: case report and
versus placebo for uncomplicated skin abscess. N Engl J Med 2016;374:823–832. literature review. Scand J Infect Dis 2005;37:504–507.
98. Liu C, Bayer A, Cosgrove SE, et al. Clinical practice guidelines by the Infectious 135. Avansino JR, et al. Primary operative versus nonoperative therapy for pediatric
Diseases Society of America for the treatment of methicillin-resistant Staphylococ- empyema: a meta-analysis. Pediatrics 2005;115:1652–1659.
cus aureus infections in adults and children: executive summary. Clin Infect Dis 136. Chan PC, et al. Clinical management and outcome of childhood lung abscess: a
2011;52:285–292. 16-year experience. J Microbiol Immunol Infect 2005;38:183–188.

706.e2
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
Staphylococcus aureus 115
137. Emanuel B, Shulman ST. Lung abscess in infants and children. Clin Pediatr (Phila) 178. Todd J, et al. Toxic-shock syndrome associated with phage-group-I staphylococci.
1995;34:2–6. Lancet 1978;2:1116–1118.
138. Asher MI, et al. Primary lung abscess in childhood: the long-term outcome of 179. Davis JP, et al. Toxic-shock syndrome: epidemiologic features, recurrence, risk
conservative management. Am J Dis Child 1982;136:491–494. factors, and prevention. N Engl J Med 1980;303:1429–1435.
139. Cowles RA, et al. Lung resection in infants and children with pulmonary infections 180. Chi CY, et al. A clinical and microbiological comparison of Staphylococcus aureus
refractory to medical therapy. J Pediatr Surg 2002;37:643–647. toxic shock and scalded skin syndromes in children. Clin Infect Dis 2006;42:181–185.
140. Al-Salem AH, Ali EA. Computed tomography–guided percutaneous needle aspira- 181. Darenberg J, et al. Differences in potency of intravenous polyspecific immuno-
tion of lung abscesses in neonates and children. Pediatr Surg Int 1997;12:417–419. globulin G against streptococcal and staphylococcal superantigens: implications
141. Cuestas RA, Kienzle GD, Armstrong JD II. Percutaneous drainage of lung abscesses for therapy of toxic shock syndrome. Clin Infect Dis 2004;38:836–842.
in infants. Pediatr Infect Dis J 1989;8:390–392. 182. Schlievert PM. Use of intravenous immunoglobulin in the treatment of staphylo-
142. Lee SK, Morris RF, Cramer B. Percutaneous needle aspiration of neonatal lung coccal and streptococcal toxic shock syndromes and related illnesses. J Allergy Clin
abscesses. Pediatr Radiol 1991;21:254–257. Immunol 2001;108:S107–S110.
143. Fowler VG Jr, et al. Staphylococcus aureus endocarditis: a consequence of medical 183. Crum NF. The emergence of severe, community-acquired methicillin-resistant
progress. JAMA 2005;293:3012–3021. Staphylococcus aureus infections. Scand J Infect Dis 2005;37:651–656.
144. Valente AM, et al. Frequency of infective endocarditis among infants and children 184. Gonzalez BE, et al. Severe staphylococcal sepsis in adolescents in the era of
with Staphylococcus aureus bacteremia. Pediatrics 2005;115:e15–e19. community-acquired methicillin-resistant Staphylococcus aureus. Pediatrics
145. Ferrieri P, et al. Unique features of infective endocarditis in childhood. Pediatrics 2005;115:642–648.
2002;109:931–943. 185. Shulman ST, Ayoub EM. Severe staphylococcal sepsis in adolescents. Pediatrics
146. Saiman L, Prince A, Gersony WM. Pediatric infective endocarditis in the modern 1976;58:59–66.
era. J Pediatr 1993;122:847–853. 186. Adem PV, et al. Staphylococcus aureus sepsis and the Waterhouse-Friderichsen
147. Bayer AS. Infective endocarditis. Clin Infect Dis 1993;17:313–320, quiz, 321–322. syndrome in children. N Engl J Med 2005;353:1245–1251.
148. Bayer AS, et al. Diagnosis and management of infective endocarditis and its 187. Kravitz GR, et al. Purpura fulminans due to Staphylococcus aureus. Clin Infect Dis
complications. Circulation 1998;98:2936–2948. 2005;40:941–947.
149. Baltimore RS, Gewitz M, Baddour LM, et al. American Heart Association Rheu- 188. Miller LG, et al. Necrotizing fasciitis caused by community-associated methicillin-
matic Fever, Endocarditis, and Kawasaki Disease Committee of the Council on resistant Staphylococcus aureus in Los Angeles. N Engl J Med 2005;352:1445–1453.
Cardiovascular Disease in the Young; the Council on Cardiovascular and Stroke 189. Peacock SJ, et al. Bacterial fibronectin-binding proteins and endothelial cell surface
Nursing. Infective endocarditis in childhood – 2015 update: a scientific statement fibronectin mediate adherence of Staphylococcus aureus to resting human endothe-
from the American Heart Association. Circulation 2015;132:1487–1515. lial cells. Microbiology 1999;145:3477–3486.
150. Martin JM, Neches WH, Wald ER. Infective endocarditis: 35 years of experience at 190. Weidenmaier C, et al. Lack of wall teichoic acids in Staphylococcus aureus leads to
a children’s hospital. Clin Infect Dis 1997;24:669–675. reduced interactions with endothelial cells and to attenuated virulence in a rabbit
151. John MD, et al. Staphylococcus aureus prosthetic valve endocarditis: optimal model of endocarditis. J Infect Dis 2005;191:1771–1777.
management and risk factors for death. Clin Infect Dis 1998;26:1302–1309. 191. Daum RS. Skin and soft-tissue infections caused by methicillin-resistant S. aureus.
152. Kuyvenhoven JP, et al. Prosthetic valve endocarditis: analysis of risk factors for N Engl J Med 2007;357:1357.
mortality. Eur J Cardiothorac Surg 1994;8:420–424. 192. Youngster I, Shenoy ES, Hooper ES, et al. Comparative evaluation of the tolerability
153. Cakir O, et al. Purulent pericarditis in childhood: ten years of experience. J Pediatr of cefazolin and nafcillin for treatment of methicillin-susceptible Staphylococcus
Surg 2002;37:1404–1408. aureus infections in the outpatient setting. Clin Infect Dis 2014;59:369–375.
154. Roodpeyma S, Sadeghian N. Acute pericarditis in childhood: a 10-year experience. 193. Mongkolrattanothai K, Daum RS. Impact of community-associated, methicillin-
Pediatr Cardiol 2000;21:363–367. resistant Staphylococcus aureus on management of the skin and soft tissue infections
155. Thebaud B, Sidi D, Kachaner J. [Purulent pericarditis in children: a 15 year- in children. Curr Infect Dis Rep 2005;7:381–389.
experience]. Arch Pediatr 1996;3:1084–1090. 194. Zhanel GG, Sniezek G, Schweizer F, et al. Ceftaroline: a novel broad-spectrum
156. Gonzalez BE, et al. Venous thrombosis associated with staphylococcal osteomyelitis cephalosporin with activity against methicillin-resistant Staphylococcus aureus.
in children. Pediatrics 2006;117:1673–1679. Drugs 2009;69:809–831.
157. Givner LB, Kaplan SL. Meningitis due to Staphylococcus aureus in children. Clin 195. Groom AV, et al. Community-acquired methicillin-resistant Staphylococcus aureus
Infect Dis 1993;16:766–771. in a rural American Indian community. JAMA 2001;286:1201–1205.
158. Brook I. Brain abscess in children: microbiology and management. J Child Neurol 196. Naimi TS, et al. Epidemiology and clonality of community-acquired methicillin-
1995;10:283–288. resistant Staphylococcus aureus in Minnesota, 1996–1998. Clin Infect Dis
159. Vartzelis G, et al. Brain abscesses complicating Staphylococcus aureus sepsis in a 2001;33:990–996.
premature infant. Infection 2005;33:36–38. 197. Leclercq R. Mechanisms of resistance to macrolides and lincosamides: nature of the
160. Tekkok IH, Erbengi A. Management of brain abscess in children: review of 130 cases resistance elements and their clinical implications. Clin Infect Dis 2002;34:482–492.
over a period of 21 years. Childs Nerv Syst 1992;8:411–416. 198. Fiebelkorn KR, et al. Practical disk diffusion method for detection of inducible
161. Saez-Llorens X. Brain abscess in children. Semin Pediatr Infect Dis 2003;14:108–114. clindamycin resistance in Staphylococcus aureus and coagulase-negative staphylo-
162. Harris LF, et al. Brain abscess: recent experience at a community hospital. South cocci. J Clin Microbiol 2003;41:4740–4744.
Med J 1985;78:704–707. 199. Zelazny AM, et al. Selection of strains for quality assessment of the disk induction
163. Auletta JJ, John CC. Spinal epidural abscesses in children: a 15-year experience and method for detection of inducible clindamycin resistance in staphylococci: a CLSI
review of the literature. Clin Infect Dis 2001;32:9–16. collaborative study. J Clin Microbiol 2005;43:2613–2615.
164. Rothman SL. The diagnosis of infections of the spine by modern imaging tech- 200. Boyle-Vavra S, Ereshefsky B, Wang CC, Daum RS. Successful multiresistant
niques. Orthop Clin North Am 1996;27:15–31. community-associated methicillin-resistant Staphylococcus aureus lineage from
165. Martin RJ, Yuan HA. Neurosurgical care of spinal epidural, subdural, and intra- Taipei, Taiwan, that carries either the novel staphylococcal chromosome cassette
medullary abscesses and arachnoiditis. Orthop Clin North Am 1996;27:125–136. mec (SCCmec) type VT or SCCmec type IV. J Microbiol 2005;43:4719–4730.
166. Savage K, Holtom PD, Zalavras CG. Spinal epidural abscess: early clinical outcome [Erratum, J Clin Microbiol 2005;43:6223].
in patients treated medically. Clin Orthop Relat Res 2005;439:56–60. 201. Szumowski JD, Cohen DE, Kanaya F, Mayer KH. Treatment and outcomes of
167. Wang KW, et al. Infection of cerebrospinal fluid shunts: causative pathogens, clini- infections by methicillin-resistant Staphylococcus aureus at an ambulatory clinic.
cal features, and outcomes. Jpn J Infect Dis 2004;57:44–48. Antimicrob Agents Chemother 2007;51:423–428.
168. Williams DG, Hayes J, McCool S. Shunt infections in children: presentation and 202. Hulten KG, Kaplan SL, Gonzalez BE, et al. Three-year surveillance of community
management. J Neurosci Nurs 1996;28:155–162. onset health care-associated Staphylococcus aureus infections in children. Pediatr
169. McGirt MJ, et al. Risk factors for pediatric ventriculoperitoneal shunt infection and Infect Dis J 2005;40:1785–1791.
predictors of infectious pathogens. Clin Infect Dis 2003;36:858–862. 203. Wang G, Hindler JF, Ward KW, Bruckner DA. Increased vancomycin MICs for
170. James HE, et al. Prospective randomized study of therapy in cerebrospinal fluid Staphylococcus aureus clinical isolates from a university hospital during a 5-year
shunt infection. Neurosurgery 1980;7:459–463. period. J Clin Microbiol 2006;44:3883–3886.
171. Whitehead WE, Kestle JR. The treatment of cerebrospinal fluid shunt infections. 204. Kapadia M, Coyle E, Prince R, et al. Declining in vitro activity of vancomycin against
Results from a practice survey of the American Society of Pediatric Neurosurgeons. Staphylococcus aureus isolates from cancer patients. In: Programs and Abstracts of
Pediatr Neurosurg 2001;35:205–210. the 45th Interscience Conference on Antimicrobial Agents and Chemotherapy.
172. Michino H, Otsuki K. Risk factors in causing outbreaks of food-borne Washington, DC, American Society of Microbiology, 2001 (abstract E-807).
illness originating in school lunch facilities in Japan. J Vet Med Sci 2000;62: 205. Golan Y, Baiez-Giangreco C, O’Sullivan C, Snydman DR. Trends in vancomycin
557–560. susceptibility among consecutive MRSA bacteremia isolates. In: Programs and
173. Richards MS, et al. Investigation of a staphylococcal food poisoning outbreak in a Abstracts of the 44th Annual Meeting of the Infectious Diseases Society of America.
centralized school lunch program. Public Health Rep 1993;108:765–771. Toronto, 2006, Infectious Diseases Society of America (abstract LB-11).
174. Melish ME, Glasgow LA. The staphylococcal scalded-skin syndrome. N Engl J Med 206. Saravolatz LD, Stein GE. Oritavancin: a long-half-life lipoglycopeptide. Clin Infect
1970;282:1114–1119. Dis 2015;61:627–632.
175. Melish ME, Glasgow LA. Staphylococcal scalded skin syndrome: the expanded 207. Mitra S, Saeed U, Havlichek DH, Stein GE. Profile of oritavancin and its potential
clinical syndrome. J Pediatr 1971;78:958–967. in the treatment of acute bacterial skin structure infections. Infect Drug Resist
176. Patel GK, Finlay AY. Staphylococcal scalded skin syndrome: diagnosis and manage- 2015;8:189–197.
ment. Am J Clin Dermatol 2003;4:165–175. 208. Roberts KD, Sulaiman RM, Rybak MJ. Dalbavancin and oritavancin” an inno-
177. Schlievert PM. Staphylococcal enterotoxin B and toxic-shock syndrome toxin-1 are vative approach to treatment of gram positive infections. Pharmacotherapy
significantly associated with nonmenstrual TSS. Lancet 1986;1:1149–1150. 2015;35:935–948.

706.e3
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.
PART III  Etiologic Agents of Infectious Diseases
SECTION A  Bacteria

209. Bradley JS, Puttagunta S, Rubino CM, et al. Pharmacokinetics, safety and toler- 233. Ardura MI, Mejías A, Katz KS, et al. Daptomycin therapy for invasive gram-positive
ability of single dose dalbavancin in children 12-17 years of age. Pediatr Infect Dis bacterial infections in children. Pediatr Infect Dis J 2007;26:1128–1132.
J 2015;34:748–752. 234. Sakoulas G, et al. Induction of daptomycin heterogeneous susceptibility in
210. Ford CW, Zurenko GE, Barbachyn MR. The discovery of linezolid, the first oxa- Staphylococcus aureus by exposure to vancomycin. Antimicrob Agents Chemother
zolidinone antibacterial agent. Curr Drug Targets Infect Disord 2001;1:181–199. 2006;50:1581–1585.
211. Deville JG, et al. Linezolid versus vancomycin in the treatment of known or 235. Cui L, et al. Correlation between reduced daptomycin susceptibility and vanco-
suspected resistant gram-positive infections in neonates. Pediatr Infect Dis J mycin resistance in vancomycin-intermediate Staphylococcus aureus. Antimicrob
2003;22:S158–S163. Agents Chemother 2006;50:1079–1082.
212. Kaplan SL, et al. Linezolid versus vancomycin for treatment of resistant gram- 236. Fowler VG, Boucher HW, Corey GR, et al. Daptomycin versus standard therapy
positive infections in children. Pediatr Infect Dis J 2003;22:677–686. for bacteremia and endocarditis caused by Staphylococcus aureus. N Engl J Med
213. Yogev R, et al. Linezolid for the treatment of complicated skin and skin structure 2006;355:653–665.
infections in children. Pediatr Infect Dis J 2003;22:S172–S177. 237. Kline MW, Mason EO Jr. Methicillin-resistant Staphylococcus aureus: pediatric
214. Taylor JJ, Wilson JW, Estes LL. Linezolid and serotonergic drug interactions: a perspective. Pediatr Clin North Am 1988;35:613–624.
retrospective survey. Clin Infect Dis 2006;43:180–187. 238. Update. Staphylococcus aureus with reduced susceptibility to vancomycin – United
215. French G. Safety and tolerability of linezolid. J Antimicrob Chemother States, 1997. MMWR Morb Mortal Wkly Rep 1997;46:813–815.
2003;51:ii45–ii53. 239. Smith TL, et al. Emergence of vancomycin resistance in Staphylococcus aureus.
216. Meka VG, et al. Linezolid resistance in sequential Staphylococcus aureus isolates Glycopeptide-Intermediate Staphylococcus aureus Working Group. N Engl J Med
associated with a T2500A mutation in the 23S rRNA gene and loss of a single copy 1999;340:493–501.
of rRNA. J Infect Dis 2004;190:311–317. 240. Chang S, et al. Infection with vancomycin-resistant Staphylococcus aureus contain-
217. Pillai SK, et al. Linezolid resistance in Staphylococcus aureus: characterization and ing the vanA resistance gene. N Engl J Med 2003;348:1342–1347.
stability of resistant phenotype. J Infect Dis 2002;186:1603–1607. 241. Whitener CJ, et al. Vancomycin-resistant Staphylococcus aureus in the absence of
218. Locke JB, Morales G, Hilgers M, et al. Elevated linezolid resistance in clinical c/r- vancomycin exposure. Clin Infect Dis 2004;38:1049–1055.
positive Staphylococcus aureus isolates is associated with co-occurring mutations in 242. Fridkin SK, et al. Epidemiological and microbiological characterization of infec-
ribosomal protein L3. Antimicrob Agents Chemother 2010;54:5352–5355. tions caused by Staphylococcus aureus with reduced susceptibility to vancomycin,
219. Zhanel GG, Love R, Adam H, et al. Tedizolid: a novel oxazolidinone with United States, 1997–2001. Clin Infect Dis 2003;36:429–439.
potent activity against multidrug-resistant gram positive pathogens. Drugs 243. Stevens DL, Bisno AL, Chambers HF. Practice guidelines for the diagnosis and
2015;75:253–270. management of skin and soft tissue infections: 2014 update by the Infectious
220. Miller LG, Daum RS, Creech CB, et al. Clindamycin versus trimethoprim- Diseases Society of America. Clin Infect Dis 2014;59:147–159.
sulfamethoxazole for uncomplicated skin infections. N Engl J Med 244. Nguyen DM, Mascola L, Brancoft E. Recurring methicillin-resistant Staphylococcus
2015;372:1093–1103. aureus infections in a football team. Emerg Infect Dis 2005;11:526–532.
221. Markowitz N, Quinn EL, Saravolatz LD. Trimethoprim-sulfamethoxazole com- 245. Centers for Disease Control and Prevention. Methicillin-resistant Staphylococcus
pared with vancomycin for the treatment of Staphylococcus aureus infection. Ann aureus infections among competitive sports participants – Colorado, Indiana,
Intern Med 1992;117:390–398. Pennsylvania, and Los Angeles County, 2000–2003. MMWR Morb Mortal Wkly
222. Proctor RA. Role of folate in the treatment of methicillin-resistant Staphylococcus Rep 2003;52:793–795.
aureus infection. Clin Infect Dis 2008;15:584–593. 246. David MZ, Siegel JD, Henderson J, et al. A randomized, controlled trial of
223. Ruhe JJ, et al. Use of long-acting tetracyclines for methicillin-resistant Staphylo- chlorhexidine-soaked cloths to reduce methicillin-resistant and methicillin-
coccus aureus infections: case series and review of the literature. Clin Infect Dis susceptible Staphylococcus aureus carriage prevalence in an urban jail. Infect
2005;40:1429–1434. Control Hosp Epidemiol 2014;35:1466–1473.
224. Bouchillon SK, et al. In vitro activity of tigecycline against 3989 gram-negative 247. Whitman TJ, Herlihy RK, Schlett CD, et al. Chlorhexidine-impregnated cloths to
and gram-positive clinical isolates from the United States Tigecycline Evalua- prevent skin and soft tissue infections in Marine recruits: a cluster-randomized,
tion and Surveillance Trial (TEST Program, 2004). Diagn Microbiol Infect Dis double-blind, controlled effectiveness trial. Infect Control Hosp Epidemiol
2005;52:173–179. 2010;31:1207–1215.
225. Zhanel GG, et al. Tigecycline: a novel glycylcycline antibiotic. Expert Rev Anti Infect 248. Boyce JM. MRSA patients: proven methods to treat colonization and infection. J
Ther 2006;4:9–25. Hosp Infect 2001;48(suppl A):S9–S14.
226. Allington DR, Rivey MP. Quinupristin/dalfopristin: a therapeutic review. Clin Ther 249. Loeb M, et al. Antimicrobial drugs for treating methicillin-resistant Staphylococcus
2001;23:24–44. aureus colonization. Cochrane Database Syst Rev 2003;(4):CD003340.
227. Nichols RL, et al. Treatment of hospitalized patients with complicated gram- 250. Kampf G. The value of using chlorhexidine soap in a controlled trial to eradicate
positive skin and skin structure infections: two randomized, multicentre studies MRSA in colonized patients. J Hosp Infect 2004;58:86–87.
of quinupristin/dalfopristin versus cefazolin, oxacillin or vancomycin. Synercid 251. Kampf G, Jarosch R, Ruden H. Limited effectiveness of chlorhexidine based hand
Skin and Skin Structure Infection Group. J Antimicrob Chemother 1999;44: disinfectants against methicillin-resistant Staphylococcus aureus (MRSA). J Hosp
263–273. Infect 1998;38:297–303.
228. Lakey JH, Ptak M. Fluorescence indicates a calcium-dependent interaction between 252. Miller MA, et al. Development of mupirocin resistance among methicillin-resistant
the lipopeptide antibiotic LY146032 and phospholipid membranes. Biochemistry Staphylococcus aureus after widespread use of nasal mupirocin ointment. Infect
1988;27:4639–4645. Control Hosp Epidemiol 1996;17:811–813.
229. Critchley IA, et al. Baseline study to determine in vitro activities of daptomycin 253. Cadilla A, David MZ, Daum RS, Boyle-Vavra S. Association of high level mupirocin
against gram-positive pathogens isolated in the United States in 2000–2001. resistant Staphylococcus aureus at an academic center in the Midwestern United
Antimicrob Agents Chemother 2003;47:1689–1693. States. J Clin Microbiol 2011;49:95–100.
230. Arbeit RD, et al. The safety and efficacy of daptomycin for the treatment of com- 254. Maira-Litran T, et al. Comparative opsonic and protective activities of Staphylo-
plicated skin and skin-structure infections. Clin Infect Dis 2004;38:1673–1681. coccus aureus conjugate vaccines containing native or deacetylated staphylococcal
231. Silverman JA, Mortin LI, Vanpraagh AD, et al. Inhibition of daptomycin by poly-N-acetyl-beta-(1–6)-glucosamine. Infect Immun 2005;73:6752–6762.
pulmonary surfactant: in vitro modeling and clinical impact. J Infect Dis 255. Lin L, Ibrahim AS, Xu X, et al. Th1-Th17 cells mediate protective adaptive immu-
2005;191:2149–2152. nity against Staphylococcus aureus and Candida albicans infection in mice. PLoS
232. Levine D. Daptomycin for infective endocarditis. Curr Infect Dis Rep 2006;8:263–264. Pathog 2009;5:e1000703.

706.e4
Descargado para Anonymous User (n/a) en National Autonomous University of Mexico de ClinicalKey.es por Elsevier en septiembre 21, 2021. Para
uso personal exclusivamente. No se permiten otros usos sin autorización. Copyright ©2021. Elsevier Inc. Todos los derechos reservados.

You might also like