You are on page 1of 18

Antimicrobial Susceptibility Testing:

A Primer for Clinicians


Kristi M. Kuper, Pharm.D., Deborah M. Boles, M.S., John F. Mohr, Pharm.D., and Audrey Wanger, Ph.D.

Appropriate use of antimicrobials in health care continues to be a challenge.


Reliable and reproducible antimicrobial susceptibility testing methods are
necessary to provide the clinician with valuable information that can be
translated into positive clinical outcomes at the bedside. However, there are
nuances with these testing methods that, if unrecognized, could lead to
misinterpretation of results and inappropriate antibiotic selection. This
primer describes the common antimicrobial susceptibility tests used in the
clinical microbiology laboratory and reviews how subtle differences in testing
methods and technique can influence reported results. Clinicians who have a
thorough understanding of qualitative and quantitative methods, automated
susceptibility testing systems, and commonly used screening and confirma-
tory tests for antibiotic-resistant organisms can strengthen institutional
antibiotic stewardship programs and improve patient outcomes.
Key Words: infectious disease, antimicrobial, bacterial resistance, antibiotics,
computer technology, pharmacokinetics, susceptibility, microbiology testing
methods.
(Pharmacotherapy 2009;29(11):1326–1343)

OUTLINE Testing
Basic Principles of Antimicrobial Susceptibility Testing Fastidious Organisms
Qualitative Testing Anaerobes
Disk Diffusion Mycobacteria
Quantitative Testing Specialized Screening and Confirmatory Tests for
Minimum Inhibitory Concentration Resistant Organisms
Etest Interpretation
Automated Systems Agreement Among Methods
Vitek and Vitek 2 Clinical Practice Application
MicroScan WalkAway Conclusion
Phoenix
In January 2007, the Infectious Diseases
Sensititre
Society of America, in conjunction with the
Advantages and Disadvantages
Society for Healthcare Epidemiology, released
Panel and Card Selection
guidelines for developing an institutional
Special Considerations in Antimicrobial Susceptibility
program to enhance antimicrobial stewardship.1
From the Department of Clinical Affairs, Cardinal Health, These guidelines underscore the need for
Houston, Texas (Dr. Kuper); the Department of Pharmacy,
Lowell General Hospital, Lowell, Massachusetts (Dr. Boles);
accurate and reproducible identification of
Cubist Pharmaceuticals, Lexington, Massachusetts (Dr. microorganisms and antibiotic susceptibilities.
Mohr); and the Department of Pathology, University of This is integral to the care of patients with
Texas Medical School, Houston, Texas (Dr. Wanger). infectious diseases and plays a critical role in
For reprints, visit http://www.atypon-link.com/PPI/loi/phco. antimicrobial stewardship and epidemiologic
For questions or comments, contact Kristi M. Kuper,
Pharm.D., BCPS, Department of Clinical Affairs, Cardinal
investigations.
Health, 1330 Enclave Parkway, Houston, TX 77077; e-mail: The clinician who has an advanced knowledge
kristine.kuper@cardinalhealth.com. of antimicrobials, coupled with an understanding
ANTIMICROBIAL SUSCEPTIBILITY TESTING PRIMER Kuper et al 1327
of commonly used microbiology testing methods, points. The term “breakpoint” can be confusing,
can be effective in improving antimicrobial as it may refer to one derived from microbiologic,
utilization and optimizing patient care. These clinical, or pharmacokinetic-pharmacodynamic
skills are particularly relevant today and can help data. Microbiologic breakpoints refer to the MIC
combat increased antimicrobial resistance in the for an antibiotic that distinguishes wild-type
hospital and community settings. The purposes bacterial populations from those that have either
of this review are to help pharmacists gain a basic selected or acquired resistance mechanisms.
understanding of common antimicrobial suscep- These breakpoints are derived from moderate-to-
tibility tests used in the clinical microbiology large numbers of in vitro MIC tests. A clinical
laboratory, and to demonstrate how differences in breakpoint is derived from the antimicrobial
testing can influence therapeutic choices for MICs for the infecting organisms isolated in
treating infectious diseases. prospective clinical trials. Susceptibility in this
context correlates with a high likelihood of
Basic Principles of Antimicrobial Susceptibility clinical success. Finally, a pharmacokinetic-
Testing pharmacodynamic breakpoint predictive of
microbiologic effects is derived from human or
The purpose of performing antimicrobial animal data that are modeled by using statistical
susceptibility testing is to assist clinicians with or mathematic techniques such as a Monte Carlo
the selection of appropriate targeted antibiotic simulation.9
therapy in order to optimize clinical outcomes. Breakpoints for new antibiotics are approved
Infection-related and overall mortality is reduced by the United States Food and Drug Adminis-
when patients are treated expeditiously with an tration (FDA) based on composite data from the
antibiotic to which the organism is susceptible.2 breakpoint methodologies described previously
Two basic methods of antimicrobial suscepti- and are not frequently altered after the product is
bility testing are available to laboratories: quali- released to market. A recent inventory of
tative and quantitative. Disk diffusion (also antibacterial drug labels indicated that among
known as the Kirby-Bauer method) is a quali- more than 100 currently approved drug labels,
tative method of susceptibility testing that may over 70 contained breakpoints that were
be prone to some degree of error depending on outdated.10 Breakpoints may not be available for
the drug and organism being tested.3–7 However, older drugs (e.g., polymyxin or colistin for
it is an acceptable option for testing among Enterobacteriaceae) or organisms that rarely
patients with uncomplicated urinary tract cause human disease such as Flavobacterium
infections or other less severe infections where species.
eradication is augmented by the patient’s immune Ongoing surveillance and laboratory process
system. Because of the high spontaneous cure standardization is governed by a nonprofit global
rates for some mild infections, an organism that standards developing organization known as the
is truly resistant yet reported as susceptible by Clinical and Laboratory Standards Institute
the microbiology laboratory may not always be (CLSI), previously known as the National
clinically relevant.8 Broth microdilution and agar Committee for Clinical Laboratory Standards
dilution methodologies are considered quanti- (NCCLS). The CLSI promotes the development
tative because they can measure the minimum and use of voluntary consensus standards and
inhibitory concentration (MIC). The MIC is guidelines within the health care community and
defined as the lowest concentration of an anti- publishes a series of reference manuals that serve
biotic that inhibits visible growth of a micro- as useful guides for antimicrobial susceptibility
organism. Both quantitative methods are considered testing.11, 12 These manuals are referenced by a
the reference methods for susceptibility testing number-letter combination that represents the
because of their high levels of reproducibility. subject matter and the version. Two manuals of
For convenience, most clinical laboratories use interest to the pharmacist include the M-39 and
an automated system supplemented with one or the M-100. The M-39 is updated every 3 years
more manual methods of susceptibility testing. (current version is M-39A3) and provides
Automated systems must provide susceptibility recommendations for antibiogram preparation.
results that are consistent with a reference The M-100 is updated yearly (current version is
method. Regardless of the testing methodology S19) and contains organism-specific breakpoint
selected, the results are correlated with a set of reference tables for disk diffusion and MIC
standardized interpretations known as break- testing. The breakpoints published in the M-100
1328 PHARMACOTHERAPY Volume 29, Number 11, 2009
Table 1. Examples of Discrepancies Between United States Food and Drug Administration and Clinical and Laboratory
Standards Institute Minimum Inhibitory Concentration Breakpoints12–18
MIC Breakpoints (µg/ml)
FDA Approved CLSI Approved
Organism Drug Susceptible Intermediate Resistant Susceptible Intermediate Resistant
Streptococcus
pneumoniae Ceftriaxone
Nonmeningitis ≤ 0.5 1 ≥ 2a ≤1 2 ≥4
Meningitis ≤ 0.5 1 ≥ 2a ≤ 0.5 1 ≥2
Enterococcus sp Daptomycin ≤ 4b —b —b ≤ 4b —b —b
Enterobacteriaceae Tigecycline ≤2 4 ≥8 None
Doripenem ≤ 0.5 —c —c None
Acinetobacter sp Colistin or None ≤2 — ≥4
polymixin
FDA = U.S. Food and Drug Administration; CLSI = Clinical and Laboratory Standards Institute; MIC = minimum inhibitory concentration.
a
The package insert for ceftriaxone does not make a distinction between nonmeningeal and meningeal isolates of S. pneumoniae.
b
The FDA-approved breakpoint in the daptomycin package insert is for vancomycin-susceptible Enterococcus faecalis strains only, but there is
no FDA-approved breakpoint for vancomycin-susceptible Enterococcus faecium strains. The CLSI-approved breakpoint can be applied to all
Enterococcus species.
c
The FDA approved only a susceptible classification because of the absence of resistant isolates in clinical trials. Drugs with an MIC (or disk
diffusion test) that suggests nonsusceptibility of isolates should undergo additional testing.

by the CLSI are for both branded and generic organism reaches its critical mass, resulting in
antibiotics against different microorganisms and false susceptibility.20
incorporate in vitro microbiologic information, The standardized testing process described
human and animal pharmacokinetic-pharmaco- here allows reproducible and consistent results.
dynamic data, and clinical and bacteriologic Before testing, the microbiologist will prepare a
outcomes from clinical studies.9 These break- fresh agar plate by using an inoculum that
points may be discordant with the FDA-approved contains a predefined organism density. The
values (Table 1),12–18 as well as the values set by inoculum is prepared by suspending isolated
the European Union Committee on Antimicrobial bacterial colonies (taken from an 18–24-hr agar
Susceptibility Testing (known as EUCAST). plate) in broth or saline to a turbidity matching a
0.5 McFarland standard (1 x 108 colony-forming
Qualitative Testing units/ml). This standard is used to measure the
turbidity of the bacteria and ensure a certain
Disk Diffusion number of colony-forming units in the inoculum.12
The principle behind the disk diffusion method The standard concentration of the solution is
is that antibiotic molecules diffuse out from a important because an inoculum that is too high
disk into the agar, creating a dynamically in colony-forming units can result in falsely
changing gradient of antibiotic concentrations smaller zone sizes (i.e., false resistance), whereas
while the organism being tested starts to divide too low of an inoculum can result in falsely larger
and growth progresses toward the critical mass.19 zone sizes (i.e., false susceptibility).21, 22
The zone edge is where the concentration of The inoculum suspension should be used
antibiotic begins to inhibit the organism reaching within 15 minutes of preparation. This is
an overwhelming cell mass. At this point, the particularly important for fastidious organisms
density of cells is high enough to absorb (e.g., Neisseria species, Haemophilus influenzae,
antibiotic in the immediate vicinity, thus and ␤-hemolytic streptococci) that lose their
maintaining concentrations at subinhibitory viability rapidly, resulting in a low inoculum. A
levels, and enabling the test organism to grow. sterile cotton swab is dipped into the suspension
For most rapidly growing aerobic and facultative and pressed firmly on the inside of the tube to
anaerobic bacteria, the critical time it takes for remove excess liquid. The entire dried surface of
the organism to absorb the antibiotic varies from the agar plate is inoculated by streaking the
3–6 hours, but interpretation by the microbiologist surface in three directions. After each streak, the
generally occurs between 18 and 24 hours. For plate is rotated 60 degrees to obtain an even
some slow-growing organisms, such as Bacteroides distribution of the inoculum. After streaking, the
fragilis, the gradient is formed before the plate should be dried for no more than 15
ANTIMICROBIAL SUSCEPTIBILITY TESTING PRIMER Kuper et al 1329
minutes. Once the agar plate is completely dry, tilting the plate should be ignored. Again,
antibiotic disks are applied either manually or inappropriate technique could lead to erroneous
with a dispensing apparatus. For most organisms, susceptibility interpretation.
no more than 12 disks should be placed on a The main advantages of disk diffusion testing
150-mm agar plate or 5 disks on a 90-mm plate. are simplicity, inexpensive equipment, and the
It is best to place disks that have predictably cost-effective and flexible choice of antibiotics for
small zone sizes (e.g., gentamicin) next to disks testing. Disadvantages include the inability to
that give predictably larger zones of inhibition obtain an actual MIC and the additional time
(e.g., cephalosporins) so that zone overlap can be required by the technologist for test preparation
prevented.23 and providing an interpretation of manually
Optimally, disks should be positioned at a determined zone sizes based on standards.
distance of 30 mm apart, and no closer than 24 Commercial zone readers (e.g., BioMICV3; Giles
mm apart when measured center to center, to Scientific, Inc., Santa Barbara, CA) are available
minimize inhibition zone overlap.23 Most dispenser to simplify this process.
devices are self-tamping (disks are tapped or Certain antibiotics can be problematic to test
pressed onto the agar surface) but may need to be with the disk diffusion method because of the
pressed down to make immediate and complete specific physiochemical properties of the
contact with the agar surface. Once in contact molecules. Vancomycin, colistin, and macrolides
with the agar, the disk cannot be moved because have higher molecular weights and therefore
the antibiotics diffuse rapidly once the disk comes diffuse very slowly in agar. 22 The limited
into contact with the agar. The microbiologist’s diffusion and poorly resolved concentration
technique is important because movement can gradient around these disks result in only a few
disrupt the process integrity and lead to inaccurate millimeters of difference in zone sizes between
results. susceptible and resistant strains, which could
Agar plates are incubated in an inverted position result in a potentially ambiguous reading.
(agar side up) under conditions appropriate for Results can also be influenced by the positioning
the test organism. The incubation period is of the light source on the plate. Most plates are
16–18 hours for rapidly growing aerobic bacteria, read with use of reflected light, with the
and longer for fastidious organisms or special exception of linezolid, oxacillin, and vancomycin
resistance conditions. When detecting vancomycin for both S. aureus and Enterococcus species. In
or oxacillin resistance in Staphylococcus aureus, these cases, the zones should be measured by
plates need to be kept for a minimum of 24 hours using transmitted light in order to ensure an
per CLSI testing procedures.23 After incubation, accurate measurement of zone diameter. If
the agar plate is examined to determine if a results are unexpected or borderline, another
semiconfluent and even “lawn” of growth has method of testing may be required or the test
been obtained before reading the plate. If repeated for confirmation.
individual colonies are seen, the inoculum is too
light and the test should be repeated since the Quantitative Testing
large zone sizes are inaccurate. The same holds
true for an excessively heavy inoculum causing Minimum Inhibitory Concentration
overly small zones with very hazy edges.22 Either In critical infections such as bacteremia and
outcome could result in false readings. endocarditis, accurate quantitative determination
If the lawn of growth is satisfactory, the zone of the exact MIC value may be useful for therapy
diameter is read to the nearest millimeter by guidance.24, 25 In these situations, an organism
using a ruler or sliding calipers. The zone with an MIC of 0.016 µg/ml to an antimicrobial
margin is identified as the area where no obvious may have a significantly different therapeutic
visible growth is seen by the naked eye, unless implication from that of the same organism-
otherwise specified. Zone diameters on Mueller- antibiotic combination that is susceptible with an
Hinton agar (without blood supplements) are MIC of 1 µg/ml. The pharmacodynamics of the
read from the back of the plate, whereas blood- drug and the infection site factor into this assess-
containing agars are read from the surface to ment. For example, a vancomycin MIC of 1.5
ensure that the zone of inhibition is read µg/ml or greater was found to be independently
accurately. The line of demarcation for this zone associated with treatment failure in patients with
should be clear; faint growth or microcolonies methicillin-resistant S. aureus (MRSA) bacteremia
detectable only with a magnifying glass or by (adjusted risk ratio 2.6, 95% confidence interval
1330 PHARMACOTHERAPY Volume 29, Number 11, 2009

1.3–5.4, p=0.01).26 Interpreted, this means that Etest


patients with bacterial infections caused by Etest (bioMérieux, Durham, NC) was approved
MRSA strains that have an antibiotic MIC of 1.5 by the FDA in 1991. It is a manual method of
or 2 µg/ml may fail therapy even though the exact MIC testing that uses a gradient technique
isolate is considered to be susceptible based on combining the principles of both disk diffusion
current breakpoints. The clinician who uses the and agar dilution. However, Etest differs from
reported antibiotic MIC for the organism coupled disk diffusion and is applicable for use with a
with pharmacokinetic-pharmacodynamic data of wide range of fastidious and nonfastidious
the antibiotic can customize therapy to improve aerobic and anaerobic organisms, with varying
clinical outcomes and reduce mortality.27–30 growth rates, critical times for antibiotic
Although all of these processes may seem absorption, and testing procedures. Etest offers
straightforward, MICs can vary based on the end convenience for hospital laboratories and is
point, type of media, and conditions of the commonly used to complement other routine
incubation period. For example, end points for automated laboratory antimicrobial susceptibility
bacteriostatic drugs should be read at 80% testing methods.
inhibition (significant reduction in growth) and Etest uses a preformed and predefined gradient
bactericidal drugs at 100% inhibition (complete of varying antibiotic concentrations immobilized
inhibition). If a bacteriostatic drug is evaluated in a dry format onto the surface of a plastic strip.
at 100% inhibition (as opposed to 80% inhibition), The concentration of the gradient is calibrated
the MIC reported will be falsely elevated and may across a continuous MIC range covering 15 2-fold
result in the organism being categorized as dilutions. When applied to the surface of an
resistant, instead of susceptible. In addition, inoculated agar plate, the antibiotic on the plastic
capnophilic (carbon dioxide “loving”) organisms strip is transferred to the agar in the form of a
such as pneumococci, streptococci, gonococci, stable continuous gradient directly beneath and in
and Haemophilus species are incubated in 5% the immediate vicinity of the strip. The stability
carbon dioxide and MICs will vary based on this of this antibiotic gradient is maintained for up to
carbon dioxide level.31, 32 20 hours. After incubation, a parabolic-shaped
Most testing methods used in the hospital inhibition zone centered alongside the test strip
laboratory are correlated with either broth micro- is visible. The MIC is read at the point where the
dilution, broth macrodilution, or agar dilution growth or inhibition margin of the organism
methods. Both broth methods and agar dilution
are quantitative tests that are predominantly
performed in reference or research laboratories.
These tests are considered reference methods
because they are used to gauge the accuracy of
other testing methods. These tests are not easily
automated and require the laboratory to prepare
stock solutions of antimicrobial agents that are
then dispensed into test tubes (macrodilution),
multiple well plates (microdilution; Figure 1), or
an agar medium. Each tube, well, or agar plate
contains a differing standard concentration of
antibiotic that is inoculated with the specimen.
The MIC can then be determined based on the
level of visible growth. The broth dilution and
agar dilution methods can be used to verify the
accuracy of an automated instrument should a
Figure 1. Broth microdilution plate contains standard
discrepancy arise. dilutions of eight bacterial organisms in each row (denoted by
Agar dilution should not be confused with letters A–H). Each column of wells (denoted by numbers
newer colorimetric agar tests. Colorimetric agars 1–11; 12 is a sterile control) contains a standard antibiotic
contain certain chemicals that cause a color concentration that doubles when moving from right to left
change when select organisms are present. These (e.g., row 11 = 0.06 µg/ml, row 10 = 0.12 µg/ml, row 9 = 0.25
µg/ml, etc.). The minimum inhibitory concentration (MIC)
tests are also referred to as agar screening is determined by the first well where there is no visible
methodologies. Such agar-based testing is growth. For example, the MIC of this antibiotic for organism
outside the scope of this review. A is 4 µg/ml, and the MIC for organism B is 8 µg/ml.
ANTIMICROBIAL SUSCEPTIBILITY TESTING PRIMER Kuper et al 1331
intersects the edge of the calibrated strip (Figure molecules, such as vancomycin, and lipophilic
2).33 This point can be easily identified for most compounds, such as amphotericin, that would
organisms. Using a magnifying glass and/or otherwise be a limitation for disk diffusion. 33
tilting the plate can be helpful to visualize Although the Etest complements many of the
microcolonies and hazes or other colonies within laboratory’s functions, it is not used as the sole
the ellipse of inhibition. method of testing. An Etest is not available for
The key advantages of Etest are that it uses a all antimicrobials. Test setup can be labor
stable gradient, it is a convenient agar-based intensive, and there is a propensity for error in
method, and higher inocula can be used to detect interpretation if the point of intersection with the
antibiotic resistance. Furthermore, molecular strip does not appear to be well marginated or
properties of the antibiotic do not affect the microcolonies growing in the inhibition area are
performance of Etest since the gradient is missed on visual inspection.
preformed and not dependent on diffusion. Etest is currently FDA approved for a variety of
Thus, the Etest gradient can accommodate large antibiotics for clinical testing of aerobes, anaerobes,
pneumococci, streptococci, Haemophilus species,
and gonococci.34 Laboratories may choose to use
an Etest to confirm equivocal results with their
routine systems or test organisms with distinctive
resistance mechanisms that can be difficult to
detect with standard methods. Etest may be
helpful in detecting resistance in organisms that
are highly inoculum dependent, as they can be
falsely reported as susceptible in some automated
systems.35

Automated Systems
Before the 1970s, labor-intensive manual
susceptibility testing was the dominant method.
In 1974, the first automated system known as the
Autobac I disk elution system was introduced by
Pfizer Diagnostics.36 Now, approximately 83% of
clinical laboratories report using an automated
instrument for primary susceptibility testing.37
Several automated systems are available in the
United States. They include Phoenix (Becton
Dickinson, Franklin Lakes, NJ), Vitek (bioMérieux),
MicroScan WalkAway (Siemens Healthcare
Diagnostics, Tarrytown, NY), and Sensititre (Trek
Diagnostics, Cleveland, OH). Most systems
contain a computerized algorithm for inter-
preting results and identifying inconsistencies
between organism identification and suscepti-
bility. These automated systems use commercially
available panels that contain added growth
factors to speed organism growth, thereby
providing more rapid results compared with
traditional methods. They also use sophisticated
Figure 2. This Etest strip contains graduated concen-
trations of ampicillin ranging from 0.016 µg/ml (not shown) software to analyze the growth rates and
to 256 µg/ml placed on an agar plate growing Escherichia determine the antibiotic MIC for the organism by
coli. Since the intersection of the growth-inhibition margin using specialized decision technology. These
lies between two minimum inhibitory concentrations systems do not read the actual MIC, but rather
(MICs)—0.38 and 0.5 µg/ml—the test is interpreted at the they use a sophisticated rules system to
highest value (0.5 µg/ml). This organism is defined as
susceptible since the MIC lies below the breakpoint of 8 determine whether to adjust to a higher or lower
µg/ml or lower. MIC such that the results fit into a doubling
1332 PHARMACOTHERAPY Volume 29, Number 11, 2009

dilution result (e.g., 1, 2, 4, 8 µg/ml, etc.). panels are the size of conventional microdilution
Although the general process of identification is trays. Depending on the laboratory’s preference,
similar, there are differences among each system. either a combination identification-susceptibility
panel or separate identification and susceptibility
Vitek and Vitek 2 panels may be used. The WalkAway system
consists of an incubator-reading unit that can
The Vitek was first developed in the 1960s. It read either as a conventional panel or a fluorescent
uses small plastic cards containing multiple wells rapid-read panel. Panels are manually inoculated
filled with either biochemical substrates or by using a proprietary device known as the
antibiotic dilutions. Cards can be used for both RENOK (rehydrator-inoculator device). Once
identification and susceptibility testing for most the panels are placed into the incubator-reader
aerobic gram-positive and gram-negative unit, 41 the remainder of the process is fully
organisms. The system uses a manual filler sealer automated. A bar code reader identifies each
module to inoculate the cards with the organism. panel, incubates it for the appropriate amount of
After inoculation, a reader incubator module uses time, and moves the panels to the reading position.
a single wavelength of light to measure turbidity Organism identification can be determined within
and color changes in the card wells. Robotics an average of 2.5 hours by rapid methods but
technology is used to move the cards every 15 may take 6–18 hours with use of conventional
minutes to different reading areas. The Vitek testing methodologies. Final results for
then uses regression analysis to calculate the susceptibilities take an average of 20 hours, with
MIC.38 a range of 16.8–27.8 hours depending on type of
The Vitek 2 system is the second generation of organism.40, 41
Vitek and offers a more sophisticated model of
data analysis as well as a fully automated process
Phoenix
for card identification, organism suspension
dilution, and card filling. Once these steps are This system uses an optimized colorimetric
complete, the Vitek 2 seals the cards into a oxidation-reduction indicator for susceptibility
chamber to prevent contamination during testing and a variety of fluorometric and colori-
processing. The cards are then loaded into the metric indicators for bacterial identification. The
reader incubator, which ejects them at the end of panels are inoculated manually and require a
testing. The Vitek 2 uses colorimetric technology specific organism dilution for accuracy. If the
with use of three wavelengths of light to provide inoculation is incorrect, the system will auto-
broad profiles for the most clinically significant matically reject the panel from the incubation
organisms. The data analysis is performed by and reading phase. After inoculation, the panels
using algorithms to look at a variety of parameters are loaded into the incubator-reader module, and
and test conditions to ensure accurate results and all subsequent steps are fully automated. The
early detection of resistance mechanisms through Phoenix database then analyzes the kinetic
the use of proprietary software.38 The Vitek 2 measurements of bioreactivity within individual
Compact offers a condensed version of the Vitek wells. The average time to identifi-cation with
2; although it requires more technician involve- the Phoenix system is 4.3 hours. The time to
ment before reading the cards, the compact final susceptibility results ranges from 7.5–16
system is desirable for smaller hospitals or clinics hours, with a mean of 10.5 hours.40, 42 Although
with limited space. Species identification with all the systems offer some basic epidemiologic
these systems is complete in an average of 3 software and data for antibiogram production,
hours with rapid methods, but may take up to the Phoenix database allows for greater flexibility
5.7 hours for slow-growing organisms that and data analysis.42
require colorimetric testing methodology.
Susceptibility results may take up to 15 hours, Sensititre
with a mean of about 9 hours.39, 40
The Sensititre susceptibility system is a version
of the broth dilution method and can provide
MicroScan WalkAway
both in vitro qualitative and quantitative
The MicroScan offers a choice of an overnight susceptibility results in a dried plate format.
panel or the more rapid identification-suscep- Isolated colonies for testing are diluted according
tibility panel that uses fluorescent technology. In to a predefined standard and then used to
contrast to the Vitek system, the MicroScan inoculate a 96-well microdilution plate. A
ANTIMICROBIAL SUSCEPTIBILITY TESTING PRIMER Kuper et al 1333
fluorescent precursor may be added at the same section for a detailed description of types of
time as the test organism. Inoculated panels are errors associated with testing.) The authors
sealed with adhesive film to prevent evaporation concluded that these commercial systems
and then incubated at 34–36°C for 18–24 hours. performed poorly for cystic fibrosis isolates in
During incubation, bacterial surface enzymes will contrast to high result correlations that had been
cleave key bonds on the precursor and trigger the reported previously with reference methods and
release of fluorophores, which emit fluorescence. Etest.47
Unlike the Vitek and Phoenix systems, the
contents of the wells can then be examined Panel and Card Selection
manually for bacterial growth. However,
Sensititre can also be read with an automated All of the automated systems offer several
reading system that is able to detect the fluores- panel choices for the susceptibility portion of the
cence. The amount of fluorescence detected is testing. Panels and cards are separated into
directly related to the activity of bacterial surface gram-positive, gram-negative, and fungal panels
enzymes and, therefore, to the presence of (for select systems).48 Each manufacturer’s Web
bacterial growth and resistance to the antibiotic. site provides examples of the potential panels
Additional interpretations of susceptibility results and cards. The choice of panel should be aligned
are performed by the software using rules derived with the hospital’s antimicrobial formulary.
from FDA standards.43 Pharmacy personnel play an important role in
panel selection by ensuring that formulary
antibiotics are reported on the culture and
Advantages and Disadvantages
sensitivity reports.
One major advantage of automated suscepti- Depending on the system, separate cards may
bility methodologies is a reduction in labor.44–46 need to be purchased for organism identification,
Another advantage is that these systems can as well as gram-positive and gram-negative
provide faster reporting of susceptibility results, susceptibilities. The identification panel may be
potentially leading to the earlier initiation of used alone when susceptibilities are not clinically
appropriate antibiotic therapy. Although the significant. For example, Lactobacillus and
reduction in labor requirements and faster Pasteurella species are not tested because approved
reporting are significant advantages to using susceptibilities are lacking, and Staphylococcus
automation in the microbiology laboratory, saprophyticus will only be identified because
definite disadvantages exist. susceptibilities will not change the course of
First, all automated systems require the manual therapy.
step of preparing an inoculum. This step is Interchangeability between panels is limited by
critical to the accuracy of the results because several factors. Whenever a new panel or card is
over- or underinoculating or mixing cultures can used, a series of quality assurance tests must be
result in false or misleading results. Second, all performed before starting the new panel; this
automated systems can test only a limited range may take an average of 20–30 days and limits the
of organisms. Slow-growing or fastidious number of times that panels can be changed.
organisms must still be tested by alternative Most microbiology laboratories purchase supplies
methods. Mucoid Pseudomonas aeruginosa in bulk to obtain better pricing. In an effort to
isolated from patients with cystic fibrosis also reduce cost, the laboratory may need to finish the
presents a challenge for automated systems existing panel supply before proceeding to a new
because of the thick cell wall caused by one. This can be a challenge when changes are
exopolysaccharide-alginate and its slow growth. made to the antibiotic formulary. If the new
In one study, antibiotic susceptibilities for 498 antibiotic is not on the susceptibility panel, then
strains from patients with cystic fibrosis (one manual tests must be conducted to verify
third of which were mucoid) were performed by susceptibilities to the new agents and the results
using Vitek and MicroScan WalkAway systems, for nonformulary antibiotics need to be
and results were compared with those of a suppressed. Customized testing panels can be
reference broth microdilution method. “Very developed for a specific institution based on their
major” error rates (i.e., erroneously reported as specific formulary or to increase the number of
susceptible, yet determined to be resistant by a dilutions tested to provide a better range of
reference method) were 17% and 10.4–12.4%, MICs. However, this may not be a cost-effective
respectively. (See Agreement Among Methods strategy, especially for institutions with dynamic
1334 PHARMACOTHERAPY Volume 29, Number 11, 2009

formularies. Finally, testing space on the antibiotic perform anaerobic susceptibilities and must send
susceptibility cards is not infinite, and therefore the sample out to a reference laboratory if
not all MICs can be tested. Generally, the range requested. Indications for anaerobic susceptibility
of MICs tested are doubling dilutions (e.g., 0.25, testing include the management of patients with
0.5, 1, 2 µg/ml). In some cases, only the break- serious infections requiring long-term therapy
point is reported (e.g., MIC ≤ 2 µg/ml). and selection of appropriate therapy for
organisms that are not predictably susceptible.
Special Considerations in Antimicrobial The CLSI recommends that agar dilution (known
Susceptibility Testing as the Wadsworth method) be used as the
reference method for anaerobic susceptibility
Fastidious Organisms testing.50 A broth microdilution method is also
As their category implies, most fastidious approved but is limited to rapidly growing
organisms do not grow well enough in automated anaerobes (Bacteroides species). Another
antimicrobial testing systems and require some disadvantage is that the method is only described
type of supplementation (e.g., blood). Disk for a limited number of antimicrobials, and the
diffusion was initially developed for susceptibility panels are not commercially available. Because of
testing of rapidly growing aerobic bacteria, these limitations, performing susceptibility
including enterococci, staphylococci, Enterobacte- testing on obligate anaerobes is not a standard
riaceae, and P. aeruginosa. Modifications for procedure. 51 However, due to the increasing
performing disk diffusion testing with certain resistance rates reported among some anaerobes
fastidious organisms such as H. influenzae, in recent years, the CLSI recommends suscep-
Neisseria gonorrhea, Streptococcus pneumoniae, tibility testing for clinically significant anaerobes
and other Streptococcus species have been (e.g., B. fragilis, Prevotella species, and Fusobac-
described by CLSI and include using agar that is terium species) and creating an antibiogram.52–55
enriched with glucose, yeast extract, and other The Etest can be used for testing individual isolates
substances. Modifications for broth microdilution as well as batch-testing for an antibiogram.
can also be used for testing fastidious organisms,
although results must be read manually and not Mycobacteria
by automated instruments.49 Automated testing Because of minimal equipment requirements
methodologies can be developed to correlate for acid-fast bacilli smears and their ability to
results that are obtained with CLSI recommen- produce rapid results, almost all laboratories
dations for testing of fastidious organisms. have the capacity to conduct these tests. Further
Because of the stability of the gradient, Etest identification and susceptibility testing of
can also be used for testing very slow-growing mycobacteria are generally performed in larger
fastidious organisms, as previously mentioned. institutions or reference laboratories with the
Clinical situations often warrant susceptibility equipment necessary to further process these
testing of rarely encountered, unusual, and/or organisms. A negative pressure room and a
opportunistic pathogens for which no CLSI sterile hood are required, along with a specialized
guidelines are available. In these cases, scientific set of materials and techniques specific to the
references can be used to guide the choice of selection and testing of mycobacteria. Clinical
appropriate media, incubation conditions, and laboratories that test mycobacteria use an
antibiotics to test. Because of the lack of automated or semiautomated broth-based system
guidelines, accompanying interpretations may be for growth as well as susceptibility of Mycobac-
difficult to find, so the MIC value is reported terium tuberculosis. Susceptibility testing of
without interpretation. These situations should rapidly growing mycobacteria can be performed
be handled on an individual basis in consultation by either broth microdilution or by Etest.56, 57
with an infectious diseases physician. Factors
that may influence the course of action include Specialized Screening and Confirmatory Tests
the organism(s), site of infection, pharmacokinetics for Resistant Organisms
of the antibiotic, and previous experience with a
similar antibiotic for treating the same condition. Microbiologists can use certain antibiotics to
screen for key resistance patterns among common
bacteria. Screening tests may differ from
Anaerobes
traditional susceptibility methods in some cases
Most hospital laboratories do not routinely because the antibiotic used to detect the resis-
ANTIMICROBIAL SUSCEPTIBILITY TESTING PRIMER Kuper et al 1335
tance may be incongruous with the antibiotic that grow on the agar should be suspected as
selected for treatment. Most screening tests used possible vancomycin-intermediate or vancomycin-
in the clinical laboratory are designed to detect resistant S. aureus and confirmed by using a
methicillin-, oxacillin-, or vancomycin-resistant validated MIC method such as broth dilution,
gram-positive organisms (most commonly MRSA, agar dilution, or Etest, or through approved
vancomycin-resistant Enterococcus, and ␤- antimicrobial susceptibility testing.65 Screening
lactamase–producing gram-negative organisms). and detection of heterogeneous vancomycin-
There are several methods that can be used to intermediate S. aureus presents a significant
detect oxacillin resistance among S. aureus. In challenge in the clinical laboratory. Although
addition to disk diffusion or broth microdilution, methods exist in research, there is no standard-
other methodologies used include the oxacillin ized method for identifying heterogeneous
salt agar screen and the cefoxitin disk screen.12 A vancomycin-intermediate S. aureus consistently
latex agglutination test for penicillin-binding in the clinical microbiology laboratory.66
protein 2a is also available, but this is not Brain-heart infusion agar supplemented with
commonly used in the traditional hospital vancomycin 6 µg/ml can also be used as a screen
microbiology laboratory because of the ease of for vancomycin resistance in enterococci. 67
other methods and cost. All methods vary in Plates are inoculated, incubated, and interpreted
terms of their specificity and sensitivity, and the in the same manner as the MRSA screen agar
results can vary depending on strain type.58, 59 plates described previously. Additional screening
The oxacillin salt agar screen test uses Mueller- agars are also available for testing for high-level
Hinton agar supplemented with 4% sodium aminoglycoside resistance in enterococci by
chloride and oxacillin 6 µg/ml inoculated with a using brain-heart infusion agar supplemented
0.5 McFarland suspension of the organism. The with gentamicin 500 µg/ml and streptomycin
plates are incubated for 24 hours at a maximum 2000 µg/ml. The presence of high-level amino-
temperature of 35 o C. Testing at higher glycoside resistance indicates a lack of synergistic
temperatures may interfere with the detection of effect when an aminoglycoside is combined with
methicillin resistance.23 The appearance of more a cell-wall inhibitor. Gentamicin high-level
than one colony indicates resistance. This screen resistance is associated with high-level resistance
has a high sensitivity for detecting resistant to other aminoglycosides, such as tobramycin,
strains, but sensitivity decreases when very netilmicin, amikacin, and kanamycin. Streptomycin
heteroresistant strains are tested or when the resistance occurs by means of a separate
MIC is borderline.59, 60 Recently, the cefoxitin mechanism. This is the justification for testing
disk screen has been recognized as a better both gentamicin and streptomycin for high-level
predictor of methicillin resistance than many resistance in enterococci.68
other classic methods because cefoxitin is a more Some strains of macrolide-resistant S. aureus
potent inducer of mecA than are penicillins.61 and coagulase-negative Staphylococcus species
The substitution of a cefoxitin disk for an have a transferable resistance mechanism known
oxacillin disk results in an easier test for the as macrolide-lincosamide-streptogramin B
microbiologist to read and has similar sensitivity (MLSB) resistance. This inducible resistance can
and specificity to detect oxacillin resistance in result in clindamycin treatment failures,69, 70 but
Staphylococcus species. The incubation time it can be detected by using a disk approximation
required for testing S. aureus and Staphylococcus test known as the double disk diffusion, which is
lugdunensis is only 16 hours,23 but some studies frequently referred to as the D-test. A 2-µg
have shown that results can be achieved even clindamycin disk is placed 15–26 mm away from
earlier.62 In addition, the cefoxitin disk screen the edge of a 15-µg erythromycin disk on
has equal sensitivity but improved specificity in Mueller-Hinton agar. After incubation, when
coagulase-negative Staphylococcus species. The inducible resistance is not present, the zone of
disadvantage of this test is that it may falsely inhibition around the clindamycin disk will
detect resistance in some mecA-negative strains appear as a concentric circle. However, if the
and may fail to detect resistance in some strains edge of the clindamycin zone closest to the
of mecA-positive Staphylococcus simulans.63 erythromycin disk is flattened (thus causing the
Brain-heart infusion agar supplemented with D shape), then the isolate is reported as
vancomycin 6 µg/ml can be used to screen for clindamycin resistant (Figure 3).12 Although the
vancomycin resistance in S. aureus isolates with a CLSI recommends a distance of up to 26 mm,
vancomycin MIC of 4 µg/ml or more.64 Organisms one recent study found that using a 22-mm
1336 PHARMACOTHERAPY Volume 29, Number 11, 2009

distance was associated with inaccuracies, but frequent, it is safe to assume resistance will be
placing the disks 15 mm apart resulted in 100% present, and treatment with a ␤-lactam–enzyme
sensitivity and specificity.71 inhibitor combination will be necessary.
One of the most frequently used screening tests Hospital microbiology laboratories also have
detects ␤-lactamases, of which the most common the ability to detect extended-spectrum ␤-
among gram-negative bacteria is the enzyme lactamases (ESBLs). Although ESBLs have been
known as TEM-1. This enzyme causes ampicillin identified in many organisms,75 CLSI procedures
and penicillin resistance in H. influenzae and N. for screening and confirmation of ESBLs exist
gonorrhea.72 Most hospitals will perform a ␤- only for Klebsiella pneumoniae, Klebsiella oxytoca,
lactamase screening test for these two organisms Escherichia coli, and clinically relevant Proteus
because these ␤-lactamases may produce low- mirabilis strains (e.g., a bacteremic isolate). 12
level resistance that may go undetected. There Screening for ESBLs in E. coli, K. pneumoniae, or
are three categories of ␤-lactamase tests: K. oxytoca involves performing a preliminary
colorimetric, acidimetric, and iodometric. In the susceptibility test against cefpodoxime, ceftazidime,
hospital microbiology laboratory, the colorimetric aztreonam, cefotaxime, or ceftriaxone. The process
method is the most commonly used and involves is similar for P. mirabilis but does not involve the
sticks or disks impregnated with certain use of aztreonam or ceftriaxone. If the results
compounds such as nitrocefin or cephalosporin- suggest the presence of an ESBL (based on CLSI
pyridinium-2-azo-p-dimethylaniline that produce procedure), a phenotypic confirmatory test is
a color change when the ␤-lactam is hydrolyzed. performed by using either disk diffusion or broth
Acidometric and iodometric tests are referred to microdilution. The disk diffusion confirmatory
as linked detection systems because more than test involves placing a 30-µg ceftazidime disk and
one chemical reaction must occur to produce an a 30-µg cefotaxime disk, both alone and in
interpretive result. 73 These tests are typically conjunction with clavulanic acid 10 µg, on a
performed by using penicillin as a substrate and Mueller-Hinton agar plate. If the difference
therefore may only detect enzymes that hydrolyze between the zone diameter for the single antibiotic
penicillins.74 These ␤-lactamase tests also may be versus the combination with clavulanic acid is 5
performed on Moraxella catarrhalis. However, mm or greater, then the organism is classified as
because ␤-lactamase–positive M. catarrhalis is so ESBL-positive. For broth microdilution, the
ESBL test is positive if a three or more 2-fold
concentration decrease in the MIC occurs between
the original single antibiotic (e.g., ceftazidime or
cefotaxime) and the combination of the reference
antibiotic with clavulanic acid (e.g., going from 8
µg/ml with a single antibiotic to 1 µg/ml or less
with the inhibitor present).12 Etest strips are also
available that contain cefotaxime or ceftazidime
on one end of the strip and cefotaxime or
ceftazidime plus clavulanic acid on the other end
of the strip (Figure 4).33 The interpretation for
the Etest ESBL test is the same as that for broth
microdilution. Several automated systems can
also be used to detect ESBLs. Early and appro-
priate detection of ESBL-producing organisms
allows for better customization of therapy, which
can lead to improvements in morbidity and
mortality.76
In addition to ESBLs, there are several ␤-
lactamase–producing enzymes that may also
affect interpretation and antibiotic selection.
One of the most common is the AmpC type ␤-
Figure 3. Positive double disk diffusion test (D-Test) shows lactamases. The AmpC ␤-lactamases are
induction of clindamycin (CC) resistance by erythromycin
(E) in this methicillin-resistant Staphylococcus aureus
commonly isolated from extended-spectrum,
isolate. This is indicated by the blunting of the clindamycin cephalosporin-resistant, gram-negative bacteria
zone of inhibition, which appears as a D shape. such as Serratia, Enterobacter, and Citrobacter
ANTIMICROBIAL SUSCEPTIBILITY TESTING PRIMER Kuper et al 1337

species. 77 The AmpC enzymes are inhibitor- strip whereby one side contains a cephamycin
resistant ␤-lactamases. They are resistant to (e.g., cefotetan or cefoxitin) alone and the other
clavulanic acid and other ␤-lactamase inhibitors side contains a cephamycin with cloxacillin. A
but susceptible to cloxacillin. High level of positive test for AmpC is noted if the cephamycin
expression of AmpC may prevent recognition of MIC decreases by three or more 2-fold dilutions
an ESBL because of this resistance to clavulanic in the presence of cloxacillin or a deformation of
acid, which is used to detect the presence of the inhibition ellipses around the Etest.78, 79
ESBLs.77 The presence of AmpC ultimately could Organisms with increased carbapenem MICs
lead to a false-negative ESBL screen. One are of great concern. The metallo-␤-lactamases
possible solution is to include cefepime as an are carbapenem-hydrolyzing ␤-lactamases that
ESBL screening agent because high-level AmpC emerged in Japan in the 1990s.80 These enzymes
expression is minimally affected with this drug. effectively hydrolyze both ␤-lactam antibiotics
There is also a commercially available Etest for and carbapenems except aztreonam. Although
the detection of AmpC, but it is not FDA the clinical prevalence of these enzymes is low,
approved. The Etest contains a double-sided they have been reported in Enterobacter and
Pseudomonas species. Because of difficulty of
detection, a screening method for metallo-␤-
lactamases can be used when an increase in the
MIC of carbapenems is observed. 81, 82 The
metallo-␤-lactamase enzymes are zinc mediated
and can be repressed by using ethylene-
diaminetetraacetic acid (EDTA). A commercially
available Etest may be used for metallo-␤-
lactamase detection, but it is not FDA approved.
One half of the Etest strip is impregnated with
imipenem and the other half contains imipenem
plus EDTA. A reduction in the MIC of imipenem
of three or more 2-fold dilutions in the presence
of EDTA represents a positive metallo-␤-
lactamase organism.82, 83
Carbapenemase-producing Enterobacteriaceae
(CPE), such as K. pneumoniae, are endemic in the
northeastern part of the United States 84 and
present a new challenge in antimicrobial
susceptibility testing and breakpoint reporting.
They can be missed by automated testing
systems85 and may require manual testing. Only
recently have formalized screening and
confirmatory standards been developed by the
CLSI. 12 For screening purposes, a zone of
inhibition of 19–21 mm for an ertapenem 10-µg
disk or a zone of 16–21 mm for a meropenem 10-
µg disk suggests a possible CPE. No inter-
pretative standards exist for imipenem since it
performs poorly as a screen for carbapenemases.
Broth microdilution using imipenem, meropenem,
and ertapenem 1 µg/ml can also be used as a
Figure 4. Confirmatory extended-spectrum ␤-lactamase
screen. An MIC of 2–4 µg/ml for imipenem and
(ESBL) with use of the Etest. The strip on the left contains meropenem or an MIC of 2 µg/ml for ertapenem
cefotaxime + clavulanic acid (CTL) and cefotaxime (CT). may indicate the presence of a carbapenemase
The strip on the right contains ceftazidime + clavulanic acid despite being considered susceptible based on
(TZL) and ceftazidime (TZ). The organism is ESBL-positive standard interpretations. To confirm the presence
based on the deformation of the CT ellipse, but may also be
determined by the CT and TZ minimum inhibitory
of a CPE, a modified Hodge test must be
concentrations (MICs) and the CT:CTL MIC ratio or the performed. This test involves streaking CPE-
TZ:TZL MIC ratio.33 positive and -negative reference strains along
1338 PHARMACOTHERAPY Volume 29, Number 11, 2009

with the clinical isolate onto a Mueller-Hinton source of the isolate. For example, S. pneumoniae
agar plate that has already been inoculated with a with a penicillin MIC of 1 µg/ml in a patient with
reference strain of E. coli. An ertapenem disk is meningitis would be defined as resistant since it
placed in the center. If the test is read as positive, is above the defined resistant breakpoint of 0.12
the CLSI suggests that the actual carbapenem µg/ml or greater. The same isolate from a
MIC be reported along with a comment indicating nonmeningeal source, such as sputum, would be
that the isolate demonstrates carbapenemase considered susceptible because the MIC fell
production and that the clinical efficacy of below the susceptible breakpoint of 2 µg/ml or
treating these organisms with carbapenems has lower for nonmeningitis isolates. This
not been established. If the test is negative, the differentiation by site of infection also exists with
CLSI recommends interpreting the carbapenem ␤-lactam–␤-lactamase inhibitors and intravenous
MICs by using current interpretative breakpoints. cephalosporins. Some antibiotic-organism
combinations only have a susceptible and
Interpretation nonsusceptible or susceptible and resistant
categorization. For example, the susceptibility
On completion of antimicrobial susceptibility breakpoint for doripenem tested against
testing and determination of the MIC or zone Enterobacteriaceae is 0.5 µg/ml or lower, whereas
diameter, the organism may be classified into one any MIC above this is only granted a non-
of three interpretative categories: susceptible, susceptible classification. For piperacillin-
intermediate, or resistant. There is an inverse tazobactam and P. aeruginosa, there is no
relationship between results and the inter- intermediate interpretative standard, only
pretative categories when conducting MIC susceptible (MIC ≤ 64/4 µg/ml) or resistant (MIC
testing versus disk diffusion testing. Organisms ≥ 128/4 µg/ml).17, 86
with an MIC less than (or equal to) a certain
concentration (µg/ml) or a zone diameter greater
Agreement Among Methods
than (or equal to) a specific millimeter measure-
ment are considered to be susceptible and will Because of the impracticality of reference
likely respond to treatment with a standard methods for routine susceptibility testing in the
antibiotic dosage as indicated. Organisms are clinical microbiology laboratory, automated
considered to have intermediate susceptibility to susceptibility testing methods and the Etest have
an antibiotic when the MICs approach the top become the microbiologist’s workhorse tools.
end of the usually achievable serum concentrations However, these nonreference method testing
for a standard dose or when the zone diameter systems must show that the results obtained with
falls within a certain range. The intermediate the test system agree with the results obtained
category also serves as a buffer zone to help from a reference method. The FDA sets
prevent major categoric errors caused by slight standards to ensure new devices and new drugs
changes in the zone sizes due to the influence of to be added to the panels of the automated
technical variables. Antibiotics used for the systems are substantially equivalent to a reference
treatment of intermediately susceptible organisms method for determining susceptibility testing.
may achieve clinical success when the antibiotic The reference methods for susceptibility testing
being used concentrates at the site of infection are broth dilution and agar dilution.87
(e.g., fluoroquinolones for urinary tract infec- When conducting these studies (frequently
tions) or when a higher than normal dosage is referred to as a 510[k]), the testing device is
used (e.g., ␤-lactams). If the MIC is greater than being evaluated to determine if the results are the
(or equal to) or the zone diameter is less than (or same as those of a proven reference method. A
equal to) the resistant breakpoint, this implies testing system is said to have “essential agree-
that the infection is not likely to respond to ment” when the device under evaluation has an
therapy because the physiologic concentrations MIC within ± one 2-fold dilution compared with
required to overcome resistance would cause the reference method MIC determination.
toxicity in humans. “Categorical agreement” is when the interpretive
There are some caveats to these interpretative results (susceptible, intermediate, or resistant)
categories. For some organisms, such as P. between a new device under evaluation and a
aeruginosa, there are no interpretative categories standard reference method are the same, by using
for disk diffusion tests, only MIC tests. For S. FDA interpretive criteria as presented in the
pneumoniae, breakpoints vary depending on the FDA-approved pharmaceutical antimicrobial
ANTIMICROBIAL SUSCEPTIBILITY TESTING PRIMER Kuper et al 1339
agent package insert. It is possible for there to be Table 2. Agreement Categories According to Reference
essential agreement with categorical disagree- Method versus Test Method Results
ment. When this occurs, it is considered a “minor Reference Test Method Agreement
error” and frequently results in an intermediate Method Result Result Category
susceptibility with one method and susceptible Resistant Resistant Agreement
Intermediate Minor error
or resistant with another method. For drugs Susceptible Very major error
without an intermediate category, there cannot be Intermediate Resistant Minor error
minor errors. A “major error” occurs when the Intermediate Agreement
testing method determines that the organism is Susceptible Minor error
resistant, but the reference method determines Susceptible Resistant Major error
that the organism is susceptible. Finally, a “very Intermediate Minor error
major error” occurs when the converse is true— Susceptible Agreement
the testing method indicates the organism is
susceptible but the reference method indicates
resistant (Table 2).
The FDA has issued guidance that outlines error rates when testing aztreonam. Clinically,
acceptable performance when susceptibility these discrepancies could manifest as lower-than-
testing is conducted by methods other than a expected susceptibility rates on a hospital
reference method. 87 First, the percentage antibiogram. If there are sudden sharp declines
categorical and essential agreement must be in susceptibility of P. aeruginosa to cefepime,
greater than 89.9%. Categorical agreement less confirmatory testing of a subset of isolates using
than 90% may be acceptable if there is very good another testing method may be prudent.
essential agreement, with the majority of the
discrepancies as minor discrepancies. In Clinical Practice Application
addition, the major error rate must be less than Patients infected with an organism that is
3% of all the susceptible organisms tested. considered susceptible to an antibiotic do not
Finally, the very major error rate based on the respond 100% of the time; conversely, patients
number of resistant organisms tested must who are infected with an organism that is
include an upper 95% confidence limit for the resistant to an antibiotic do not universally fail
true very major error rate of 7.5% or less, and the treatment. Infections caused by susceptible
lower 95% confidence limit for the true very isolates respond to appropriate therapy
major error rate of 1.5% or less. If 100 resistant approximately 90% of the time, whereas
organisms were tested, the very major error rate infections caused by resistant isolates actually
should not be greater than 2/100 (95% respond to the inappropriate antibiotic about
confidence interval 0.24–7.04). 60% of the time—the “90-60” rule.8 Either the
Two studies demonstrate these concepts for susceptibility methodologies or patient factors
gram-negative and gram-positive organisms. In likely explain the variability seen in patient
the first study, the investigators tested 101 outcomes based solely on susceptibility results.
bacteremic isolates of MRSA recovered from Thus, the interpretation should be used as part of
2002–2006.88 Each isolate was tested by using the antibiotic therapy selection process. An
both the broth microdilution and agar dilution understanding of the antibiotic’s pharmacokinetic
methods, and the results were compared with properties, the antibiotic’s MIC for the infecting
those of Etests performed on two different brands organism, and the pharmacodynamic relationship
of Mueller-Hinton agar plates. Vancomycin MIC between the two can help boost the patient’s
results from the Etests were consistently 1–2-fold outcome to the 90% clinical success rate.
higher than the MICs determined with the Although there are hundreds of in vitro
reference methods. In the second study, the microbiology tests that can be performed in the
investigators evaluated the accuracy of three hospital microbiology laboratory, maintaining a
automated testing systems and tested the working knowledge of common clinical
susceptibility of five broad-spectrum ␤-lactams laboratory tests enables pharmacists to become
against 100 strains of P. aeruginosa.89 Although valuable resources in helping to translate bench
the rate of major errors was minimal, the results into bedside application. For example,
investigators noted that two of the systems many practitioners interpret the results of culture
produced minor error rates of 8–32% for and susceptibility tests at “face value” and do not
cefepime and that all three systems had minor understand the discordance between laboratory
1340 PHARMACOTHERAPY Volume 29, Number 11, 2009

tests and clinical interpretation. Some practi- Financial savings


tioners associate a lower MIC with increased • Researching alternative sources for manual
efficacy. However, this is erroneous because each susceptibility testing from pharmaceutical
organism-drug combination varies and the MIC manufacturers
does not reflect pharmacodynamics or patient- • Reducing total health care resource con-
specific factors such as renal function, site of sumption through early and appropriate
infection, or volume of distribution. Pharmacists antimicrobial therapy
can translate these objective measurements into
meaningful decisions for clinicians who deliver Conclusion
patient care.
Maintaining an active knowledge base of the
Collective susceptibility testing results, compiled
various antimicrobial susceptibility testing
as an antibiogram, allow hospitals to use local
methods and their limitations can be challenging.
susceptibility data to guide empiric therapy
Qualitative testing methods are helpful and easily
before culture and susceptibility results. This
reproduced in the clinical laboratory setting with
enables the clinician to preliminarily “predict”
minimal investment in equipment and personnel.
the susceptibility of the infecting organism and
However, errors in specimen preparation, disk
ensure the early, appropriate therapy associated
placement, and zone interpretation can lead to
with improved patient outcomes.90–92 The ability
inaccurate results. Quantitative methods are
to recognize erroneous results in an antibiogram more accurate and can provide specific MICs that
is important. As previously mentioned, there are can be used to optimize the pharmacodynamics
certain known limitations with both automated of antibiotic therapy. Agar and broth dilution
and manual systems that can influence results. methods are the most accurate, but are difficult
Under-standing these discrepancies can help to perform in a busy hospital microbiology
institutions distinguish between true resistance laboratory. The development of Etests and the
issues and laboratory testing errors. Advances in deployment of automated testing systems in most
rapid diagnostic testing will undoubtedly hospital laboratories have allowed for faster
influence antibiotic utilization and ultimately result interpretation and earlier initiation of
patient outcomes. The higher cost of the tests appropriate therapy. However, clinicians must be
and laboratory resources may be offset by aware of the potential for major and minor errors
reductions in unnecessary drug therapies. that can occur between these systems.
The benefits of applied microbiology principles Numerous screening tests exist and can help
in the clinical pharmacy setting are numerous the clinician identify key resistance issues
and are summarized as follows: secondary to the presence of certain enzymes.
Results interpretation These tests are increasingly valuable as resistance
• Detecting antibiogram inaccuracies patterns continue to emerge. The development
• Translating how changes in CLSI or FDA of molecular-based technologies will allow for
breakpoints affect antibiotic susceptibility faster identification of select resistant organisms,
trending thus shortening the duration of time to appro-
• Recognizing the influence of human factors priate, targeted therapy. Clinicians who have a
on test results thorough understanding of the strengths and
• Recognizing organism–antibiotic testing weaknesses of these various testing methodologies
mismatches can successfully apply this information to benefit
• Assisting in the selection of antimicrobial both individual patient care and global antibiotic
susceptibility testing panels based on stewardship within their institution.
formulary
Clinical therapeutics Acknowledgments
• Pharmacodynamic dosing based on patient- The authors would like to thank Sheila Maness,
level MIC data M.T., for assisting with content development and
• Antimicrobial stewardship Allison Krug, M.P.H., of Artemis Biomedical
• Translating susceptibilities into empiric Communications, LLC, for assistance with editing the
therapy guidelines final manuscript.
• Educating prescribers to optimize antimicrobial
selection References
• Reducing patient morbidity and mortality 1. Dellit TH, Owens RC, McGowan JE Jr, et al. Infectious
ANTIMICROBIAL SUSCEPTIBILITY TESTING PRIMER Kuper et al 1341
Diseases Society of America and the Society for Healthcare the technique. Scand J Clin Lab Invest 1960;12:408–13.
Epidemiology of America guidelines for developing an 23. Clinical and Laboratory Standards Institute. Performance
institutional program to enhance antimicrobial stewardship. standards for antimicrobial disk susceptibility tests; approved
Clin Infect Dis 2007;44:159–77. standard—tenth edition, M02-A10. Wayne, PA: Clinical and
2. Kollef MH. Inadequate antimicrobial treatment: an important Laboratory Standards Institute, 2009.
determinant of outcome for hospitalized patients. Clin Infect 24. Baddour LM, Wilson WR, Bayer AS, et al. Infective
Dis 2000;31(suppl 4):S131–8. endocarditis: diagnosis, antimicrobial therapy, and management
3. Biedenbach DJ, Jones RN, Erwin ME. Interpretive accuracy of of complications: a statement for healthcare professionals from
the disk diffusion method for testing newer orally administered the committee on rheumatic fever, endocarditis, and Kawasaki
cephalosporins against Morganella morganii. J Clin Microbiol disease, council on cardiovascular disease in the young, and the
1993;31:2828–30. councils on clinical cardiology, stroke, and cardiovascular
4. Jevitt LA, Thorne GM, Traczewski MM, et al. Multicenter surgery and anesthesia, American Heart Association, endorsed
evaluation of the Etest and disk diffusion methods for by the Infectious Diseases Society of America. Circulation
differentiating daptomycin-susceptible from non–daptomycin- 2005;111:e394–434.
susceptible Staphylococcus aureus isolates. J Clin Microbiol 25. Soriano A, Marco F, Martínez JA, et al. Influence of
2006;44:3098–104. vancomycin minimum inhibitory concentration on the
5. Lo-Ten-Foe JR, de Smet AM, Diederen BM, Kluytmans JA, van treatment of methicillin-resistant Staphylococcus aureus
Keulen PH. Comparative evaluation of the VITEK 2, disk bacteremia. Clin Infect Dis 2008;46:193–200.
diffusion, Etest, broth microdilution, and agar dilution 26. Lodise TP, Graves J, Graffunder E, et al. Relationship between
susceptibility testing methods for colistin in clinical isolates, vancomycin MIC and failure among patients with methicillin-
including heteroresistant Enterobacter cloacae and Acinetobacter resistant Staphylococcus aureus bacteremia treated with
baumannii strains. Antimicrob Agents Chemother vancomycin. Antimicrob Agents Chemother 2008;52:3315–20.
2007;51:3726–30. 27. Forrest A, Nix DE, Ballow CH, Goss TF, Birmingham MC,
6. Tenover FC, Williams PP, Stocker S, et al. Accuracy of six Schentag JJ. Pharmacodynamics of intravenous ciprofloxacin in
antimicrobial susceptibility methods for testing linezolid seriously ill patients. Antimicrob Agents Chemother
against staphylococci and enterococci. J Clin Microbiol 1993;37:1073–81.
2007;45:2917–22. 28. Mohr JF, Wanger A, Rex JH. Pharmacokinetic/pharmacodynamic
7. Wiegand I, Geiss HK, Mack D, Sturenburg E, Seifert H. modeling can help guide targeted antimicrobial therapy for
Detection of extended-spectrum ␤-lactamases among nosocomial gram-negative infections in critically ill patients. Diagn
Enterobacteriaceae by use of semiautomated microbiology Microbiol Infect Dis 2004;48:125–30.
systems and manual detection procedures. J Clin Microbiol 29. Rafati MR, Rouini MR, Mojtahedzadeh M, et al. Clinical
2007;45:1167–74. efficacy of continuous infusion of piperacillin compared with
8. Rex JH, Pfaller MA. Has antifungal susceptibility testing come intermittent dosing in septic critically ill patients. Int J
of age? Clin Infect Dis 2002;35:982–9. Antimicrob Agents 2006;28:122–7.
9. Turnidge J, Paterson DL. Setting and revising antibacterial 30. Tam VH, McKinnon PS, Akins RL, Rybak MJ, Drusano GL.
susceptibility breakpoints. Clin Microbiol Rev 2007;20: Pharmacodynamics of cefepime in patients with gram-negative
391–408. infections. J Antimicrob Chemother 2002;50:425–8.
10. Infectious Diseases Society of America. Statement of the 31. Hamilton-Miller JM, Shah S, Loebenberg D. Susceptibility of
Infectious Diseases Society of America before the Food and pneumococci to evernimicin: effect of CO 2 and different
Drug Administration part 15 hearing panel on antimicrobial methodologies. Clin Microbiol Infect 2001;7:339–40.
resistance. April 28,2008. Arlington, VA: Infectious Diseases 32. Johnson J, Bouchillon S, Pontani D. The effect of carbon
Society of America, 2008. dioxide on susceptibility testing of azithromycin,
11. Clinical and Laboratory Standards Institute. Our history. clarithromycin and roxithromycin against clinical isolates of
Available from http://www.clsi.org. Accessed April 13, 2009. Streptococcus pneumoniae and Streptococcus pyogenes by broth
12. Clinical and Laboratory Standards Institute. Performance microdilution and the Etest: Artemis project—first-phase study.
standards for antimicrobial susceptibility testing; 19th Clin Microbiol Infect 1999;5:327–30.
informational supplement. Approved standard M100-S19. 33. AB Biodisk. Etest technical manual. Available from
Wayne, PA: Clinical and Laboratory Standards Institute, 2009. http://www.abbiodisk.com/pdf/etm_html/01_etm.htm. Accessed
13. JHP Pharmaceuticals. Coly-Mycin M parenteral (colistemethate April 13, 2009.
sodium) package insert. Rochester, MI; 2007. 34. AB Biodisk. Product list for the USA 2008. Available from
14. Roche Laboratories, Inc. Rocephin (ceftriaxone sodium) http://www.abbiodisk.com/pdf/pricelist/M0000549.pdf.
package insert. Nutley, NJ; 2009. Accessed April 13, 2009.
15. Cubist Pharmaceuticals, Inc. Cubicin (daptomycin) package 35. Jones RN, Biedenbach DJ, Marshall SA, Pfaller MA, Doern
insert. Lexington, MA; 2008. GV. Evaluation of the Vitek system to accurately test the
16. Wyeth Pharmaceuticals, Inc. Tygacil (tigecycline) package susceptibility of Pseudomonas aeruginosa clinical isolates against
insert. Philadelphia, PA; 2009. cefepime. Diagn Microbiol Infect Dis 1998;32:107–10.
17. Ortho-McNeil Pharmaceutical, Inc. Doribax (doripenem) 36. Felmingham D, Brown DF. Instrumentation in antimicrobial
package insert. Raritan, NJ; 2009. susceptibility testing. J Antimicrob Chemother 2001;48(suppl
18. Abraxis Pharmaceutical Products. Polymixin B package insert. 1):81–5.
Schaumburg, IL; 2008. 37. Baselski V, Buchanan K, Carey R, Clarridge J, Weissfeld A.
19. Bauer AW, Kirby WM, Sherris JC, Turck M. Antibiotic Survey of clinical microbiology laboratory workloads,
susceptibility testing by a standardized single disk method. Am productivity rates, and staffing vacancies. Washington, DC:
J Clin Pathol 1966;45:493–6. American Society for Microbiology Office of Public and
20. Sutter VL, Kwok Y, Finegold SM. Susceptibility of Bacteroides Scientific Affairs, 2005.
fragilis to six antibiotics determined by standardized 38. Sanders CC, Peyret M, Moland ES, et al. Potential impact of
antimicrobial disc susceptibility testing. Antimicrob Agents the VITEK 2 system and the advanced expert system on the
Chemother 1973;3:188–93. clinical laboratory of a university-based hospital. J Clin
21. Laverdiere M, Welter D, Sabath LD. Use of a heavy inoculum Microbiol 2001;39:2379–85.
in the in vitro evaluation of the anti-staphylococcal activity of 39. Eigner U, Schmid A, Wild U, Bertsch D, Fahr AM. Analysis of
19 cephalosporins. Antimicrob Agents Chemother the comparative workflow and performance characteristics of
1978;13:669–75. the VITEK 2 and Phoenix systems. J Clin Microbiol
22. Ericsson H. The paper disc method for determination of 2005;43:3829–34.
bacterial sensitivity to antibiotics: studies on the accuracy of 40. Sellenriek P, Holmes J, Ferrett R, Drury R, Storch GA.
1342 PHARMACOTHERAPY Volume 29, Number 11, 2009
Comparison of MicroScan Walk-Away, Phoenix, and VITEK 2 Microbiology; 2007:543–72.
microbiology systems used in the identification and 58. Swenson JM, Lonsway D, McAllister S, et al. Detection of
susceptibility testing of bacteria. Presented at the 105th mecA-mediated resistance using reference and commercial
meeting of the American Society of Microbiology, Atlanta, testing methods in a collection of Staphylococcus aureus
Georgia, June 5–9, 2005. Available from http://www.bd.com/ds/ expressing borderline oxacillin MICs. Diagn Microbiol Infect
technicalCenter/whitepapers/lr900.pdf. Accessed April 13, Dis 2007;58:33–9.
2009. 59. Swenson JM, Williams PP, Killgore G, O’Hara CM, Tenover
41. Chapin KC, Musgnug MC. Validation of the automated reading FC. Performance of eight methods, including two new rapid
and incubation system with Sensititre plates for antimicrobial methods, for detection of oxacillin resistance in a challenge set
susceptibility testing. J Clin Microbiol 2003;41:1951–6. of Staphylococcus aureus organisms. J Clin Microbiol 2001;39:
42. Donay JL, Mathieu D, Fernandes P, et al. Evaluation of the 3785–8.
automated Phoenix system for potential routine use in the 60. Louie L, Matsumura SO, Choi E, Louie M, Simor AE.
clinical microbiology laboratory. J Clin Microbiol 2004;42: Evaluation of three rapid methods for detection of methicillin
1542–6. resistance in Staphylococcus aureus. J Clin Microbiol 2000;38:
43. Trek Diagnostic Systems, Inc. Sensititre 510(k) substantial 2170–3.
equivalence determination decision summary; assay only 61. McKinney TK, Shar ma VK, Craig WA, Archer GL.
template. K052091. Available from http://www.fda.gov/cdrh/ Transcription of the gene mediating methicillin resistance in
reviews/K052091.pdf. Accessed April 13, 2009. Staphylococcus aureus (mecA) is corepressed but not coinduced
44. Camporese A. The impact of automation on organizational by cognate mecA and ␤-lactamase regulators. J Bacteriol
changes in a community hospital clinical microbiology 2001;183:6862–8.
laboratory. Infect Med 2004;12:118–25. 62. Bennett K, Sharp SE. Rapid differentiation of methicillin-
45. Cartwright RY, Davies JR, Dulake C, Hart RJ, Morris CA, resistant Staphylococcus aureus and methicillin-susceptible
Wilkinson PJ. A study of workload units in five microbiology Staphylococcus aureus from blood cultures by use of a direct
laboratories. J Clin Pathol 1985;38:208–14. cefoxitin disk diffusion test. J Clin Microbiol 2008;46:3836–8.
46. Freeman R. Microbiology workload [editorial]. J Clin Pathol 63. Swenson JM, Tenover FC. Results of disk diffusion testing with
2002;55:734. cefoxitin correlate with presence of mecA in Staphylococcus spp.
47. Burns JL, Saiman L, Whittier S, et al. Comparison of two J Clin Microbiol 2005;43:3818–23.
commercial systems (Vitek and MicroScan-WalkAway) for 64. Hageman JC, Patel JB, Carey RC, Tenover FC, McDonald LC.
antimicrobial susceptibility testing of Pseudomonas aeruginosa Investigation and control of vancomycin-intermediate and -
isolates from cystic fibrosis patients. Diagn Microbiol Infect Dis resistant Staphylococcus aureus: a guide for health departments and
2001;39:257–60. infection control personnel. Atlanta, GA, 2006. Available from
48. Pfaller MA, Diekema DJ, Procop GW, Rinaldi MG. Multicenter http://www.cdc.gov/ncidod/dhqp/pdf/ar/visa_vrsa_guide.pdf.
comparison of the VITEK 2 antifungal susceptibility test with Accessed April 10, 2009.
the CLSI broth microdilution reference method for testing 65. Centers for Disease Control and Prevention. Algorithm for
amphotericin B, flucytosine, and voriconazole against Candida testing S. aureus with vancomycin. Available from http://www.
spp. J Clin Microbiol 2007;45:3522–8. cdc.gov/ncidod/dhqp/pdf/ar/VRSA_testing_algo09v4.pdf.
49. Jorgensen JH, Hindler JF. New consensus guidelines from the Accessed April 10, 2009.
Clinical and Laboratory Standards Institute for antimicrobial 66. Liu C, Chambers HF. Staphylococcus aureus with hetero-
susceptibility testing of infrequently isolated or fastidious geneous resistance to vancomycin: epidemiology, clinical
bacteria. Clin Infect Dis 2007;44:280–6. significance, and critical assessment of diagnostic methods.
50. Clinical and Laboratory Standards Institute. Methods for Antimicrob Agents Chemother 2003;47:3040–5.
antimicrobial susceptibility testing of anaerobic bacteria; 67. Swenson JM, Clark NC, Ferraro MJ, et al. Development of a
approved standard M11-A7. Wayne, PA: Clinical and standardized screening method for detection of vancomycin-
Laboratory Standards Institute, 2007. resistant enterococci. J Clin Microbiol 1994;32:1700–4.
51. Jorgensen JH, Ferraro MJ. Antimicrobial susceptibility testing: 68. Cetinkaya Y, Falk P, Mayhall CG. Vancomycin-resistant
special needs for fastidious organisms and difficult-to-detect enterococci. Clin Microbiol Rev 2000;13:686–707.
resistance mechanisms. Clin Infect Dis 2000;30:799–808. 69. Sidberr y GK, Tekle T, Carroll K, Dick J. Failure of
52. Aldridge KE, Ashcraft D, Cambre K, Pierson CL, Jenkins SG, clindamycin treatment of methicillin-resistant Staphylococcus
Rosenblatt JE. Multicenter survey of the changing in vitro aureus expressing inducible clindamycin resistance in vitro.
antimicrobial susceptibilities of clinical isolates of Bacteroides Clin Infect Dis 2003;37:1257–60.
fragilis group, Prevotella, Fusobacterium, Porphyromonas, and 70. Drinkovic D, Fuller ER, Shore KP, Holland DJ, Ellis-Pegler R.
Peptostreptococcus species. Antimicrob Agents Chemother Clindamycin treatment of Staphylococcus aureus expressing
2001;45:1238–43. inducible clindamycin resistance. J Antimicrob Chemother.
53. Lassmann B, Gustafson DR, Wood CM, Rosenblatt JE. Aug 2001;48:315–16.
Reemergence of anaerobic bacteremia. Clin Infect Dis 71. O’Sullivan MV, Cai Y, Kong F, Zeng X, Gilbert GL. Influence of
2007;44:895–900. disk separation distance on accuracy of the disk approximation
54. Snydman DR, Jacobus NV, McDermott LA, et al. National test for detection of inducible clindamycin resistance in
survey on the susceptibility of Bacteroides fragilis group: report Staphylococcus spp. J Clin Microbiol 2006;44:4072–6.
and analysis of trends in the United States from 1997 to 2004. 72. Bradford PA. Extended-spectrum ␤-lactamases in the 21st
Antimicrob Agents Chemother 2007;51:1649–55. century: characterization, epidemiology, and detection of this
55. Clinical and Laboratory Standards Institute. Analysis and important resistance threat. Clin Microbiol Rev 2001;14:
presentation of cumulative antimicrobial susceptibility test 933–51.
data; approved guideline—third edition: approved standard 73. Livermore DM. ␤-Lactamases in laboratory and clinical
M39-A3. Wayne, PA: Clinical and Laboratory Standards resistance. Clin Microbiol Rev 1995;8:557–84.
Institute, 2009. 74. BD Diagnostic Systems. BBL paper discs for the detection of ␤-
56. Clinical and Laboratory Standards Institute. Laboratory lactamase enzymes: Cefinase discs package insert. Franklin
detection and identification of mycobacteria: approved standard Lakes, NJ; 2004. Available from http://www.bd.com/ds/
M48A. Wayne, PA: Clinical and Laboratory Standards Institute, productCenter/231650.asp. Accessed July 22, 2008.
2007. 75. Schwaber MJ, Raney PM, Rasheed JK, et al. Utility of National
57. Pfyffer GE. Mycobacterium: general characteristics, laboratory Committee for Clinical Laboratory Standards guidelines for
detection, and staining procedure. In: Murray PR, Baron EJ, identifying extended-spectrum ␤-lactamases in non–Escherichia
Jorgensen JH, Landry ML, Pfaller MA, eds. Manual of clinical coli and non–Klebsiella spp. of Enterobacteriaceae. J Clin
microbiology, 9th ed. Washington, DC: American Society of Microbiol 2004;42:294–8.
ANTIMICROBIAL SUSCEPTIBILITY TESTING PRIMER Kuper et al 1343
76. Hyle EP, Lipworth AD, Zaoutis TE, Nachamkin I, Bilker WB, Thomson KS. Occurrence of newer ␤-lactamases in Klebsiella
Lautenbach E. Impact of inadequate initial antimicrobial pneumoniae isolates from 24 U.S. hospitals. Antimicrob Agents
therapy on mortality in infections due to extended-spectrum ␤- Chemother 2002 46:3837–42.
lactamase–producing Enterobacteriaceae. Arch Intern Med 85. Tenover FC, Kalsi RK, Williams PP, et al. Carbapenem
2005;165:1375–80. resistance in Klebsiella pneumoniae not detected by automated
77. Philippon A, Arlet G, Jacoby GA. Plasmid-determined AmpC- susceptibility testing. Emerg Inf Dis 2006:12:1209–13.
type ␤-lactamases. Antimicrob Agents Chemother 2002;46: 86. Wyeth Pharmaceuticals Inc. Zosyn (piperacillin-tazobactam)
1–11. package insert. Philadelphia, PA; 2007.
78. Bolmström A, Engelhardt A, Bylund L, Ho T, Walsh TR. 87. U.S. Food and Drug Administration. Class II special controls
Evaluation of two new Etest strips for AmpC detection guidance document: antimicrobial susceptibility test (AST)
[abstract]. Abstr Intersci Conf Antimicrob Agents Chemother systems; guidance for industry and FDA. Issued March 5, 2007.
2006;46:145. Rockville, MD: U.S. Food and Drug Administration, 2007.
79. Engelhardt A, Yusof A, Ho P, Johansson C, Sjöström K. 88. Prakash V, Lewis JS, Jorgensen JH. Vancomycin MICs for
Evaluation of a new Etest strip for detection of plasmidic methicillin-resistant Staphylococcus aureus isolates differ based
AmpC. Presented at the 18th European congress of clinical on the susceptibility test method used [letter]. Antimicrob
microbiology and infectious diseases, Barcelona, Spain, April Agents Chemother 2008;52:4528.
19–22, 2008. 89. Sader HS, Fritsche TR, Jones RN. Accuracy of three automated
80. Rasmussen BA, Bush K. Carbapenem-hydrolyzing ␤- systems (MicroScan WalkAway, VITEK, and VITEK 2) for
lactamases. Antimicrob Agents Chemother 1997;41:223–32. susceptibility testing of Pseudomonas aeruginosa against five
81. Picão RC, Andrade SS, Nicoletti AG, et al. Metallo-␤- broad-spectrum ␤-lactam agents. J Clin Microbiol 2006;44:
lactamase detection: comparative evaluation of double-disk 1101–4.
synergy versus combined disk tests for IMP, GIM, SIM, SPM, or 90. Ibrahim EH, Sherman G, Ward S, Fraser VJ, Kollef MH. The
VIM producing organisms. Clin Microbiol 2008;46:2028–37. influence of inadequate antimicrobial treatment of bloodstream
82. Galani I, Rekatsina PD, Hatzaki D, Plachouras D, Souli M, infections on patient outcomes in the ICU setting. Chest
Giamarellou H. Evaluation of different laboratory tests for the 2000;118:146–55.
detection of metallo-␤-lactamase production in Enterobac- 91. Lodise TP, McKinnon PS, Swiderski L, Rybak MJ. Outcomes
teriaceae. J Antimicrob Chemother 2008;61:548–53. analysis of delayed antibiotic treatment for hospital-acquired
83. Bolmström A, Qwarnstrom A, Gales A, Walsh TR. Evaluation Staphylococcus aureus bacteremia. Clin Infect Dis 2003;36:
of new Etest to detect metallo-␤-lactamases (MBL) in routine 1418–23.
clinical testing [abstract]. Abstr Intersci Conf Antimicrob 92. Luna CM, Vujacich P, Niederman MS, et al. Impact of BAL
Agents Chemother 2000;40:154. data on the therapy and outcome of ventilator-associated
84. Moland ES, Black JA, Ourada J, Reisbig MD, Hanson ND, pneumonia. Chest 1997;111:676–85.

You might also like