Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Available online at www.sciencedirect.

com

Diagnostic Microbiology and Infectious Disease 66 (2010) 407 – 418


www.elsevier.com/locate/diagmicrobio

In vitro activity of tigecycline against patient isolates collected during


phase 3 clinical trials for diabetic foot infections
Peter J. Petersen, Alexey Ruzin, Margareta Tuckman, C. Hal Jones⁎
Infectious Diseases, Biosynthetic Chemistry, Wyeth Research, Pearl River, NY 10965, USA
Received 17 September 2009; accepted 22 November 2009

Abstract

The in vitro activity of tigecycline and comparative antimicrobial agents was evaluated against 1828 primary baseline pathogens isolated
from 844 patients enrolled in the phase 3 clinical trials investigating the efficacy of tigecycline in diabetic foot infection (DFI). The trials were
global, enrolling patients in 30 countries. Tigecycline was active against the most prevalent pathogens in DFI, including Gram-positive and
Gram-negative isolates of both aerobic and anaerobic bacteria with 95% of MICs ≤2 μg/mL for the entire collection. The spectrum of activity
of tigecycline included important pathogens for DFI, such as Staphylococcus aureus, Enterococcus faecalis, Streptococcus agalactiae,
Escherichia coli, Enterobacter cloacae, Klebsiella pneumoniae, and Bacteroides fragilis. As reported previously, Pseudomonas aeruginosa
and several pathogens in the Proteeae group were generally less susceptible to tigecycline by comparison to other Gram-negative pathogens.
The excellent in vitro expanded broad-spectrum activity of tigecycline in the clinical isolates confirmed the potential utility of tigecycline for
pathogens associated with DFIs.
© 2010 Elsevier Inc. All rights reserved.

Keywords: Tigecycline; Clinical isolates; In vitro susceptibility; Diabetic foot infection; MIC

1. Introduction tigecycline is not affected by the known tetracycline


resistance determinants encoding monospecific tetracycline
Tigecycline is the first in class “glycylcycline” antibiotic efflux pumps (e.g., Tet(A), Tet(K)) or ribosomal protection
that was developed by Wyeth Pharmaceuticals (2005) proteins (e.g., Tet(M), Tet(O)) (Jones et al., 2006, 2008;
(Collegeville, PA) in response to the worldwide threat of Petersen et al., 1999; Tuckman et al., 2007). Tigecycline
emerging resistance to antibiotics (Jones and Petersen, 2005; demonstrates activity against a broad range of antibiotic-
Sum and Petersen, 1999). Tigecycline is currently approved susceptible and antibiotic-resistant Gram-positive and Gram-
for treatment of complicated skin and skin structure negative aerobes, anaerobes, and “atypical” bacteria (Brad-
infections (cSSSIs), complicated intra-abdominal infections ford et al., 2005; Kenny and Cartwright, 2001; Petersen and
(cIAIs), and community-acquired bacterial pneumonia Bradford, 2005; Roblin and Hammerschlag, 2000).
(CABP) (Babinchak et al., 2005; Bradford et al., 2005, Diabetic foot ulcers are the most common cause of lower
2008; Dukart et al., 2006; Ellis-Grosse et al., 2005). The extremity amputation and are the primary cause of prolonged
mode of action of tigecycline, the 9-t-butylglycylamido hospitalization for diabetic patients (Lipsky et al., 2004;
derivative of minocycline, is through inhibition of protein Tentolouris et al., 2006). Of those diabetic patients who have
translation in bacteria. Similar to the classic tetracyclines, a foot ulcer (15% lifetime risk), between 40% and 80%
tigecycline binds to the 30S ribosomal subunit and blocks become infected (Lipsky et al., 2004). The predominant
access of aminoacyl tRNA molecules to the A site (Olson pathogens in diabetic foot infection (DFI) are aerobic Gram-
et al., 2006). In contrast to the classic tetracyclines, positive cocci (e.g., Staphylococcus aureus); however, in
those patients who have chronic wounds or have received
⁎ Corresponding author. Tel.: +1-845-602-4612; fax: +1-845-602-5671. antibiotic therapy, Gram-negative rods are also frequent
E-mail address: jonesh3@wyeth.com (C.H. Jones). pathogens (Lipsky et al., 2004; Tentolouris et al., 2006).
0732-8893/$ – see front matter © 2010 Elsevier Inc. All rights reserved.
doi:10.1016/j.diagmicrobio.2009.11.009
408 P.J. Petersen et al. / Diagnostic Microbiology and Infectious Disease 66 (2010) 407–418

To evaluate the safety and efficacy of tigecycline in 25285, Bacteroides thetaiotaomicron ATCC 29741, and Eu-
treatment of DFIs a randomized, double-blind trial was bacterium lentum ATCC 43055.
conducted with ertapenem as the active comparator (MB
Sabol et al. Phase 3 study comparing tigecycline (TGC) and 2.3. Confirmation of extended-spectrum β-lactamase
ertapenem (ERT) in patients (pts) with DFIs with and without E-test extended-spectrum β-lactamase (ESBL) strips
osteomyelitis. 47th annual meeting of the Infectious Disease containing either ceftazidime or cefotaxime with and without
Society of America. 2009). Microbiologic cultures from the clavulanic acid were used according to the manufacturer's
patients enrolled in the clinical trials yielded 1828 baseline instructions (AB Biodisk, Solna, Sweden) to confirm the
bacterial isolates from 844 patients and were provided by 110 presence of an ESBL. An isolate was judged to be positive
investigational sites in 30 countries worldwide. This analysis by the E-test confirmatory test if the MIC ratio of either
was conducted to evaluate the susceptibility of the clinical β-lactam (ceftazidime or cefotaxime) alone versus the
isolates to tigecycline and selected comparator agents. β-lactam plus clavulanic acid was ≥8.

2. Materials and methods 2.4. Polymerase chain reaction analysis of


resistance determinants
2.1. Clinical isolates
Methicillin (S. aureus) and tetracycline resistance deter-
Baseline isolates from all patients enrolled (2007–2009) in minants (S. aureus, E. coli) were identified using diagnostic
either the tigecycline or comparator treatment group were used polymerase chain reaction (PCR) assays as previously
in this analysis. Each site laboratory processed patient speci- described (Jones et al., 2006; Tuckman et al., 2007). In
mens and cultured bacterial pathogens according to local addition, confirmed ESBL-containing isolates were further
practices. All bacterial pathogens were sent to a central labo- examined by PCR to determine the class(es) of β-lactamase
ratory for identification and susceptibility testing, which were (e.g., TEM, SHV, CTX, OXA) encoded using protocols
performed in real time for the duration of the clinical trials. previously described (Jones et al., 2009).

2.2. Susceptibility testing


3. Results
For aerobic organisms, MICs were determined in
Mueller–Hinton II broth (MHB). For streptococcal MIC The most prevalent pathogens isolated from patients
determinations, MHB containing 5% lysed horse blood was during the clinical trail (2007–2009) for DFI are listed in
used. MICs were determined using custom-prepared dehy- Table 1. The distribution of pathogens was representative for
drated microdilution panels (Trek Diagnostics, Westlake,
OH) according to the manufacturer's instructions and Table 1
following reference methodology as described by the Clini- Predominant pathogens collected during phase 3 clinical trials for DFI
cal and Laboratory Standards Institute (CLSI, 2009a, 2009b).
Organism n (%) MDR strains (%)a
Interpretations were made using CLSI (2009a) M100-S19
Gram-positive aerobes 1107 (60.5%) 145 (13%)
breakpoint criteria for all comparator agents tested. Tigecy-
S. aureus 447 (24.5%) 99 (22%)
cline breakpoints were those approved by the US Food and E. faecalis 196 (11%)
Drug Administration (US FDA) (Enterobacteriaceae ≤2, 4, S. agalactiae 135 (7.4%)
≥8 μg/mL [S, I, R]; anaerobes ≤4, 8, ≥16 μg/mL [S, I, R]; S. epidermidis 79 (4.3%) 46 (58%)
staphylococci spp. ≤0.5 μg/mL [S only breakpoint]; Strep- Gram-negative aerobes 607 (33.2%) 79 (13%)
E. coli 88 (4.8%) 14 (16%)
tococcus spp. other than Streptococcus pneumoniae, Enter-
P. mirabilis 74 (4.0%) 4 (5.4%)
ococcus faecalis, and Haemophilus influenzae ≤0.25 μg/mL E. cloacae 68 (3.7%)
[S only breakpoint]; S. pneumoniae ≤0.06 μg/mL [S only P. aeruginosa 58 (3.1%) 19 (32%)
breakpoint]) (Wyeth Pharmaceuticals, 2005). For anaerobes, K. pneumoniae 57 (3.1%) 7 (12%)
MICs were determined by standard agar dilution methodol- A. baumannii/calcoaceticus complex 45 (2.5%) 24 (53%)
K. oxytoca 43 (2.4%)
ogy following CLSI (2007) instructions, M11-A7.
P. vulgaris group 30 (1.6%)
Disk diffusion susceptibility testing for tetracycline was M. morganii 26 (1.4%)
carried out by standard methodology as instructed by the C. freundii 19 (1.0%)
CLSI (2006), M02-A9. Methicillin resistance of staphylo- Anaerobes 114 (6.2%)
cocci was determined by MIC tests for oxacillin supplemen- F. magna 30 (1.6%)
P. bivia 11 (0.6%)
ted with 2% NaCl and interpreted according to CLSI
Prevotella spp. 10 (0.5%)
interpretive criteria (CLSI, 2009a, 2009b). Quality control P. asaccharolyticus 10 (0.5%)
was performed using, S. aureus ATCC 29213, E. faecalis a
MDR strains are MRSA and methicillin-resistant S. epidermis
ATCC 29212, S. pneumoniae ATCC 49619, Escherichia (MRSE) isolates by default and pathogens having resistance to 3 classes
coli ATCC 25922, Pseudomonas aeruginosa ATCC 27853, of antibacterial agent. No entry in a row indicates that no MDR strains were
H. influenzae ATCC 49247, Bacteroides fragilis ATCC identified for the given organism.
P.J. Petersen et al. / Diagnostic Microbiology and Infectious Disease 66 (2010) 407–418 409

the infection type and similar to reports from recent studies Streptococcus spp., the minocycline MIC90 of N32 μg/mL
(Lipsky et al., 2004; Sotto et al., 2007; Tentolouris et al., was above the resistant breakpoint. Similar activity was seen
2006). A summary of the tigecycline susceptibility of the against a small sample (13 isolates) of Streptococcus
baseline isolates obtained from 844 patients in the clinical pyogenes isolates (MIC90, 0.12 μg/mL). Lastly, tigecycline
trial for DFI is presented in Table 2. The most prevalent had potent activity against isolates of the Streptococcus
pathogens isolated were Staphylococci spp. (621 isolates); anginosus group with an MIC range of ≤0.03 to 0.06 μg/mL
there were 99 methicillin-resistant S. aureus (MRSA) and 348 and an MIC90 of 0.06 μg/mL.
methicillin-susceptible S. aureus (MSSA) isolates. In addi- Tigecycline showed good activity against Gram-negative
tion, among Gram-positive pathogens, E. faecalis (196 organisms of which E. coli was the predominant pathogen
isolates) and Streptococcus agalactiae (135 isolates) were isolated. For the 88 baseline patient isolates tested, all were
predominant. E. coli was the most prevalent Gram-negative susceptible to 1 μg/mL tigecycline (MIC range, 0.06–1 μg/
pathogen isolated (88 baseline isolates), followed by Proteus mL), with an MIC90 of 0.5 μg/mL. Fourteen (16%) of these
mirabilis, Enterobacter cloacae, and K. pneumonia (Table 1). isolates were multidrug-resistant (MDR) pathogens showing
As shown in Table 2, 100% of the MRSA and MSSA resistance to ceftriaxone, ciprofloxacin, and tetracycline. The
isolates were susceptible to tigecycline, with MIC90s of MIC90s for ceftriaxone and ciprofloxacin were N64 and
0.25 μg/mL, and all isolates were susceptible at the FDA N4 μg/mL, respectively. The E. coli isolates included 58
assigned susceptible breakpoint of ≤0.5 μg/mL. Most of the tetracycline-resistant isolates, determined by disk diffusion
MRSA isolates were susceptible to minocycline, and the (zone ≤19 mm); 29 of which were also resistant to
4 isolates that were minocycline resistant (MIC, ≥8 μg/mL) minocycline (MIC, ≥16 μg/mL). The tetracycline resistance
were susceptible to tigecycline (MICs, 0.12–0.25 μg/mL) determinants in these isolates were identified by PCR as
(Table 3). Three of the minocycline-resistant isolates previously described (Tuckman et al., 2007). Twenty-three
encoded both tet(M) and tet(K), and a single isolate encoded minocycline-resistant isolates were found to encode tet(B), 4
only the tet(M) gene. Fifty-eight of the minocycline- isolates encoded both tet(A) and tet(B), and a single isolate
susceptible isolates were resistant to tetracycline; of these, encoded tet(A). Twenty-nine isolates were found to be
50 isolates encoded tet(K), 2 isolates encoded tet(L), a single susceptible to minocycline and resistant to tetracycline.
isolate encoded both tet(K) and tet(M), and 5 isolates were Eighteen isolates were found to encode tet(A), and a single
negative for all tested determinants. As expected, a high isolate each encoded tet(B), tet(C), and both the tet(A) and
percentage of the MRSA isolates were resistant to cipro- tet(C) determinants. In the case of 8 tetracycline-resistant
floxacin (MIC90, ≥4 μg/mL) (Table 3). All of the isolates of isolates and 1 minocycline-resistant isolate, a gene(s)
Staphylococcus epidermidis were susceptible to 0.5 μg/mL responsible for the resistance phenotype was not identified.
of tigecycline (MIC90, 0.25 μg/mL) irrespective of the As previously shown, the presence of tetracycline resistance
susceptibility of the organisms to oxacillin. When consider- determinants, specifically monospecific tetracycline efflux
ing all isolates of Staphylococcus spp., N99% of isolates pumps, had no effect on tigecycline susceptibility of the
were tigecycline susceptible, with a single isolate of Sta- isolates (Tuckman et al., 2007).
phylococcus haemolyticus having a tigecycline MIC of Fifteen E. coli isolates were identified as encoding ESBLs
1 μg/mL (susceptible breakpoint ≤0.5 μg/mL). due to a ceftriaxone MIC ≥2 μg/mL and confirmed using
Tigecycline had good activity against all 213 isolates of ESBL E-test strips. As previously described (Jones et al.,
Enterococcus spp. collected (Table 2). The predominant 2009), the class of β-lactamase responsible for the ESBL
species obtained was E. faecalis (196 isolates), and all of the phenotype was determined by PCR. Thirteen of the isolates
isolates were susceptible to ≤0.25 μg/mL, which is the FDA- were found to encode a blaCTX family enzyme with various
assigned breakpoint for E. faecalis. By contrast, the MIC90s combinations of blaTEM, blaSHV, and blaOXA genes. One
for both ciprofloxacin (N4 μg/mL) and minocycline (16 μg/ isolate was found to carry both a blaTEM and blaSHV gene,
mL) were at the resistant breakpoints for the E. faecalis whereas another isolate was negative in all of the PCR
isolates (Table 3). All of the E. faecalis isolates were assays. Further analysis will be required to correlate the
susceptible to vancomycin (MIC90, 4 μg/mL), as were the phenotype and genotype in these isolates. As previously
6 isolates of Enterococcus faecium that were collected. described, E. coli isolates expressing ESBLs are as suscep-
Among the 11 isolates of Enterococcus spp. obtained, there tible as non-ESBL isolates to tigecycline (Jones et al., 2009).
were 3 isolates (2 Enterococcus gallinarum and 1 Entero- When tested against Klebsiella pneumoniae, tigecy-
coccus casseliflavus) that were vancomycin intermediate cline performed well with an MIC range and MIC90 of
(MIC, 8 μg/mL). All 3 isolates were susceptible to 0.25 to 4 and 2 μg/mL, respectively, for the 57 isolates
tigecycline with MICs of 0.12 μg/mL. tested. Whereas in earlier studies, K. pneumoniae had shown
Tigecycline activity was determined against 135 isolates a tendency for elevated tigecycline MICs (Ruzin et al.,
of S. agalactiae and 27 isolates of Streptococcus dysgalac- 2005), only a single isolate in the present study had an MIC
tiae; all of the isolates were inhibited by 0.25 μg/mL (MIC90, of 4 μg/mL, which is above the susceptible breakpoint set
0.06 μg/mL) and 0.5 μg/mL (MIC 90, 0.25 μg/mL) by the FDA (Wyeth Pharmaceuticals, 2005). Seven (12%)
tigecycline, respectively. In the case of both β-hemolytic of the baseline isolates were MDR pathogens, resistant to
410 P.J. Petersen et al. / Diagnostic Microbiology and Infectious Disease 66 (2010) 407–418

Table 2
In vitro activity of tigecycline against bacterial isolates from phase 3 clinical trials for DFI
Organism (no. of strains) (μg/mL) %Sa No. of isolates with MIC (μg/mL) of
MIC50 MIC90 ≤0.03 0.06 0.12 0.25 0.5 1 2 4 8 N8
Gram-negative aerobes
C. freundii complex (19) 0.25 0.5 100 2 10 7
C. koseri (8) 0.25 NAb 100 7 1
E. aerogenes (8) 0.25 NA 100 4 3 1
E. cloacae (68) 0.5 0.5 100 13 51 4
E. coli (88) 0.25 0.5 100 2 37 37 11 1
K. oxytoca (43) 0.25 0.5 100 3 34 4 2
K. pneumoniae (57) 0.5 2 98 10 32 4 10 1
M. morganii (26) 1 2 96 3 12 10 1
P. mirabilis (74) 2 4 65 1 6 41 20 6
P. vulgaris (30) 2 2 97 1 1 7 20 1
Providencia spp.c (10) 2 2 100 1 2 7
Serratia spp.d (14) 0.5 1 100 2 5 6 1
A. calcoaceticus/baumannii complex (45) 0.5 2 NA 5 7 7 8 13 4 1
Acinetobacter lwoffii (5) 0.06 NA NA 2 3
P. aeruginosa (58) 8 32 NA 1 31 26
Stenotrophomonas maltophilia (6) 0.5 NA NA 3 3
Gram-positive aerobes
S. aureus (MRSA) (99) 0.12 0.25 100 2 13 41 38 5
S. aureus (MSSA) (348) 0.12 0.25 100 7 228 109 4
S. epidermidis (MRSE) (46) 0.25 0.5 100 2 12 18 14
S. epidermidis (methicillin susceptible) (33) 0.12 0.25 100 4 14 10 5
S. haemolyticus (16) 0.25 0.5 94 1 4 5 5 1
Staphylococcus hominis (12) 0.12 0.12 100 4 7 1
Staphylococcus lugdunensis (12) 0.06 0.25 100 1 5 3 3
Staphylococcus simulans (14) 0.12 0.12 100 4 10
Coagulase-negative Staphylococcuse (41) 0.12 0.25 100 1 4 20 15 1
E. faecalis (196) 0.12 0.25 100 1 23 101 71
Enterococcus spp.f (17) 0.06 0.12 100 9 6 2
S. agalactiae (135) 0.06 0.06 100 44 85 5 1
S. anginosus (13) ≤0.03 0.06 100 9 4
S. anginosus groupg (20) ≤0.03 0.06 100 15 5
S. dysgalactiae (27) 0.06 0.25 96 7 10 6 3 1
Streptococcus mitis group (7) 0.06 NA 86 4 1 1 1
Streptococcus oralis (14) ≤0.03 0.25 93 8 1 2 2 1
S. pyogenes (13) ≤0.03 0.25 100 7 4 2
Corynebacterium spp.h (31) 0.06 0.25 NA 12 8 6 4 1
Anaerobes
B. fragilis (8) 1 NA 100 2 1 3 1 1
P. bivia (11) 1 1 100 1 4 5 1
Prevotella spp.i (10) 0.12 0.25 100 1 5 3 1
Clostridium perfringens (4) 2 NA 100 1 3
F. magna (30) 0.12 0.25 100 20 10
P. asaccharolyticus (10) 0.12 0.25 100 1 3 3 3
Peptostreptococcus spp.j (7) 0.06 NA 100 5 1 1
Anaerococcus spp.k (6) 0.12 NA 100 5 1
%S is the percentage of strains susceptible at FDA susceptibility breakpoints for tigecycline: ≤2 μg/mL for Enterobacteriaceae, ≤0.5 μg/mL for
a

Staphylococcus spp., ≤0.25 μg/mL for Streptococcus spp., ≤0.25 μg/mL for Enterococcus spp., and ≤4 μg/mL for anaerobes.
b
NA = not applicable due to number of strains or no approved interpretive criteria are available.
c
Providencia rettgeri (9), Providencia stuartii (1).
d
Serratia marcescens (9), Serratia liquefaciens (4), Serratia plymuthica (1).
e
Staphylococcus capitis (8), Staphylococcus caprae (4), Staphylococcus carnosus (1), Staphylococcus cohnii (7), Staphylococcus hyicus (1), Staphylococcus
intermedius (1), Staphylococcus kloosii (2), Staphylococcus pasteuri (1), Staphylococcus sciuri (6), Staphylococcus warneri (8), Staphylococcus xylosus (2).
f
Enterococcus avium (5), E. casseliflavus (3), E. faecium (6), E. gallinarum (2), Enterococcus harae (1).
g
S. anginosus (13), Streptococcus constellatus (5), S. intermedius (2).
h
Corynebacterium glucuronolyticum (1), Corynebacterium accolens (1), Corynebacterium amycolatum (6), Corynebacterium diphtheriae (1),
Corynebacterium macginleyi (1), Corynebacterium minutissimum (4), Corynebacterium striatum (11), Corynebacterium spp. (6).
i
Prevotella buccae (2), Prevotella denticola (1), Prevotella disiens (1), Prevotella intermedia (1), Prevotella melaninogenica (1), Prevotella oralis (1),
Prevotella spp. (3).
j
Peptostreptococcus anaerobius (4), Peptostreptococcus micros (1), Peptostreptococcus tetradius (2).
k
Anaerococcus hydrogenalis (1), Anaerococcus prevotii (5).
P.J. Petersen et al. / Diagnostic Microbiology and Infectious Disease 66 (2010) 407–418 411

β-lactam and at least 2 other classes of agents, in this case, The 58 P. aeruginosa isolates collected during the
ciprofloxacin and minocycline (Table 4). The ESBL status clinical trial had MIC90s in the resistant range for all of the
of 16 isolates was confirmed (ceftriaxone MIC, ≥2 μg/mL, test agents. The tigecycline MIC90 was 32 μg/mL,
and E-test positive) and the class of genetic determinant which is reflective of earlier studies demonstrating reduced
responsible for the ESBL status identified by PCR. Fifteen susceptibility of this organism to tigecycline (Dean et al.,
of 16 isolates encoded a blaSHV gene with 13 of the 15 2003). Like the Proteeae, P. aeruginosa expresses a family
encoding additional determinants in various combinations of multidrug efflux pumps that efficiently remove
of the blaTEM, blaCTX, and blaOXA classes. A single isolate tigecycline from the cytoplasm, reducing its effectiveness
encoded only a blaCTX family β-lactamase. All 43 of (Poole, 2005).
the Klebsiella oxytoca isolates collected were inhibited by Against the anaerobic organisms collected, tigecycline
1 μg/mL tigecycline with MIC90 of 0.5 μg/mL. performed well with all of the isolates susceptible to
As demonstrated in prior studies (Bradford et al., 2005; tigecycline at 4 μg/mL (Table 5). The MIC90s for the most
Ruzin et al., 2005; Visalli et al., 2003) tigecycline MICs for prevalent anaerobes tested were 0.25 μg/mL for Finegoldia
the Proteeae are generally higher than for other Enterobac- magna, Peptoniphilus asaccharolyticus, and Prevotella spp.
teriaceae. Mechanistic studies revealed that a multidrug and 1 μg/mL for Prevotella bivia.
efflux system, AcrAB, is responsible for reduced tigecycline
susceptibility in P. mirabilis and the other Proteeae (Visalli
et al., 2003). Against the collection of P. mirabilis from DFI, 4. Discussion
tigecycline showed results in agreement with what has been
seen in prior studies: MIC range, 0.5 to 8 μg/mL, and MIC90, Tigecycline is an antibacterial agent that has an
4 μg/mL. All of the P. mirabilis isolates were resistant to expanded broad spectrum of activity against many
minocycline (MIC range, 8 to N32 μg/mL) (Table 4). In important bacterial pathogens and has demonstrated
addition, 3 baseline isolates were found to express the ESBL clinical utility for cSSSI, cIAI, and CABP indications
phenotype (ceftriaxone MIC, ≥2 μg/mL, and E-test (Babinchak et al., 2005; Bradford et al., 2005, 2008;
positive), and PCR analysis revealed that 2 of the isolates Dukart et al., 2006; Ellis-Grosse et al., 2005). This agent
encoded blaCTX family enzymes and the third encoded a was specifically designed to overcome the 2 classic tetra-
blaCMY family enzyme. Tigecycline performed well against cycline resistance mechanisms: ribosomal protection and
the 30 Proteus vulgaris group isolates in the study, with an monospecific tetracycline efflux pumps (Petersen et al.,
MIC range and MIC90 of 0.25 to 4 and 2 μg/mL, 1999; Sum and Petersen, 1999). During preclinical
respectively. Similar activity for tigecycline was seen with development, tigecycline was shown to have activity
the 26 isolates of Morganella morganii: MIC range, 0.5 to 4 against a broad range of clinically important pathogens
μg/mL, MIC90, 2 μg/mL. The minocycline MIC90 for (Edlund and Nord, 2000; Jacobus et al., 2004; Kenny and
P. vulgaris was 16 μg/mL, and for M. morganii, it was Cartwright, 2001; Roblin and Hammerschlag, 2000). The
N32 μg/mL. potent antibacterial activity of tigecycline demonstrated in
The activity of tigecycline was evaluated against 68 the present study echoed that seen in earlier clinical studies
E. cloacae isolates, with the result that all isolates were for the approved indications, cIAI, cSSSI, and CABP, as
susceptible to 1 μg/mL with an MIC90 of 0.5 μg/mL. The well as during preclinical development (Babinchak et al.,
findings were similar for the small collection (8) of Entero- 2005; Bradford et al., 2005, 2008; Dukart et al., 2006;
bacter aerogenes isolates: all isolates were susceptible to Ellis-Grosse et al., 2005). For example, the tigecycline
1 μg/mL tigecycline. When tested against 19 isolates of MIC90s for MRSA and E. coli were 0.25 and 0.5 μg/mL,
Citrobacter freundii complex and 8 isolates of Citrobacter respectively, for isolates from earlier clinical trials for
koseri, tigecycline performed well with all isolates suscep- cSSSI and cIAI and the present DFI study. In addition,
tible to 0.5 μg/mL. The MIC90 for the C. freundii complex tigecycline activity in the present study was not impacted
isolates was 0.5 μg/mL. by the presence of the classic tetracycline resistance
Tigecycline demonstrated good activity against 45 mechanisms in E. coli and S. aureus or ESBLs in E. coli
baseline isolates of Acinetobacter baumannii/calcoaceticus and K. pneumoniae.
complex; tigecycline and minocycline were the only agents The isolates from the clinical trials were obtained from
on the panel that showed MIC90s (2 μg/mL for both patients enrolled in 30 countries in North America, Latin
agents) in the susceptible range against this important America, Eastern Europe, Western Europe, Asia, South
pathogen. The MIC90s for cefoxitin, ceftriaxone, cipro- Africa, and Australia. Regional variation supporting a trend
floxacin, ertapenem, and piperacillin/tazobactam were all for differences in the MICs of tigecycline in certain regions
above the resistance breakpoint (CLSI, 2009a) (Table 4). was not noted for isolates from the various regions
Equivalent or better activity for minocycline versus conducting trials. These results are in agreement with results
tigecycline against A. baumannii/calcoaceticus complex obtained previously from a number of large in vitro
has been noted before and is not fully understood (Bradford susceptibility studies that included isolates from North
et al., 2005). America, Latin America, Europe, the Middle East, and
412 P.J. Petersen et al. / Diagnostic Microbiology and Infectious Disease 66 (2010) 407–418

Table 3
In vitro activities of tigecycline and comparative antibacterial agents against Gram-negative clinical isolates from DFI
Organism (no. of isolates tested) Antibiotic MIC (μg/mL) %Sa
Range 50% 90%
C. freundii complex (19) Tigecycline 0.12 to 0.5 0.25 0.5 100
Cefoxitin 16 to N32 N32 N32 0
Ceftriaxone 0.12 to N64 0.25 0.5 95
Ciprofloxacin ≤0.12 to N4 ≤0.12 0.5 89
Ertapenem ≤0.03 to 2 ≤0.03 ≤0.03 100
Minocycline 1 to 8 2 4 89
Piperacillin–tazobactam 1 to 128 2 8 95
C. koseri (8) Tigecycline 0.25 to 0.5 0.25 NAb 100
Cefoxitin 2 to 16 2 NA 88
Ceftriaxone ≤0.03 to 0.25 0.06 NA 100
Ciprofloxacin ≤0.12 ≤0.12 NA 100
Ertapenem ≤0.03 ≤0.03 NA 100
Minocycline 1 to 4 2 NA 100
Piperacillin–tazobactam 2 to 8 2 NA 100
E. aerogenes (8) Tigecycline 0.25 to 1 0.25 NA 100
Cefoxitin N32 N32 NA 0
Ceftriaxone 0.06 to 4 0.12 NA 100
Ciprofloxacin ≤0.12 to 2 ≤0.12 NA 100
Ertapenem ≤0.03 to 0.25 0.06 NA 100
Minocycline 1 to 8 2 NA 88
Piperacillin–tazobactam 2 to 8 4 NA 100
E. cloacae (68) Tigecycline 0.25 to 1 0.5 0.5 100
Cefoxitin 16 to N32 N32 N32 0
Ceftriaxone 0.06 to N64 0.25 2 93
Ciprofloxacin ≤0.12 to N4 ≤0.12 0.5 96
Ertapenem ≤0.03 to 0.5 ≤0.03 0.12 100
Minocycline 2 to 32 4 8 82
Piperacillin–tazobactam 1 to 128 2 4 94
E. coli (88) Tigecycline 0.06 to 1 0.25 0.5 100
Cefoxitin 2 to N32 4 16 88
Ceftriaxone ≤0.03 to N64 0.06 N64 84
Ciprofloxacin ≤0.12 to N4 ≤0.12 N4 65
Ertapenem ≤0.03 to 1 ≤0.03 0.12 100
Minocycline 0.5 to N32 2 32 67
Piperacillin–tazobactam 0.5 to N128 2 16 90
K. oxytoca (43) Tigecycline 0.12 to 1 0.25 0.5 100
Cefoxitin 2 to 16 2 8 98
Ceftriaxone ≤0.03 to 2 0.06 0.12 100
Ciprofloxacin ≤0.12 to 0.25 ≤0.12 ≤0.12 100
Ertapenem ≤0.03 to 0.06 ≤0.03 ≤0.03 100
Minocycline 1 to 32 2 2 95
Piperacillin–tazobactam 1 to 16 2 4 100
K. pneumoniae (57) Tigecycline 0.25 to 4 0.5 2 98
Cefoxitin 1 to N32 4 32 84
Ceftriaxone ≤0.03 to N64 0.06 N64 77
Ciprofloxacin ≤0.12 to N4 ≤0.12 N4 77
Ertapenem ≤0.03 to 4 ≤0.03 0.25 96
Minocycline 1 to N32 4 16 63
Piperacillin–tazobactam 0.5 to N128 4 64 84
M. morganii (26) Tigecycline 0.5 to 4 1 2 96
Cefoxitin 8 to N32 16 16 35
Ceftriaxone ≤0.03 to N64 ≤0.03 4 92
Ciprofloxacin ≤0.12 to N4 ≤0.12 N4 77
Ertapenem ≤0.03 to 0.06 ≤0.03 ≤0.03 100
Minocycline 4 to N32 8 N32 23
Piperacillin–tazobactam ≤0.12 to 32 0.25 2 96
P. mirabilis (74) Tigecycline 0.5 to 8 2 4 65
Cefoxitin 1 to N32 4 4 99
Ceftriaxone ≤0.03 to N64 ≤0.03 0.06 97
Ciprofloxacin ≤0.12 to N4 ≤0.12 2 82
Ertapenem ≤0.03 to 0.25 ≤0.03 ≤0.03 100
Minocycline 8 to N32 32 N32 0
P.J. Petersen et al. / Diagnostic Microbiology and Infectious Disease 66 (2010) 407–418 413

Table 3 (continued)
Organism (no. of isolates tested) Antibiotic MIC (μg/mL) %Sa
Range 50% 90%
Piperacillin–tazobactam ≤0.12 to 32 0.25 0.5 99
P. vulgaris (30) Tigecycline 0.25 to 4 2 2 97
Cefoxitin 2 to 16 4 8 97
Ceftriaxone ≤0.03 to N64 ≤0.03 4 97
Ciprofloxacin ≤0.12 to 4 ≤0.12 0.5 90
Ertapenem ≤0.03 to 0.06 ≤0.03 ≤0.03 100
Minocycline 4 to 32 8 16 17
Piperacillin–tazobactam 0.25 to 128 0.5 1 97
Providencia spp.c (10) Tigecycline 0.5 to 2 2 2 100
Cefoxitin 1 to 8 2 4 100
Ceftriaxone ≤0.03 to 64 0.06 0.25 90
Ciprofloxacin ≤0.12 to N4 ≤0.12 N4 70
Ertapenem ≤0.03 ≤0.03 ≤0.03 100
Minocycline 8 to N32 16 16 0
Piperacillin–tazobactam 0.25 to 128 0.5 128 80
Serratia spp.d (14) Tigecycline 0.25 to 2 0.5 1 100
Cefoxitin 8 to N32 16 N32 14
Ceftriaxone 0.06 to 2 0.25 1 100
Ciprofloxacin ≤0.12 to 0.5 ≤0.12 ≤0.12 100
Ertapenem ≤0.03 to 0.12 ≤0.03 ≤0.03 100
Minocycline 1 to 16 2 16 64
Piperacillin–tazobactam 1 to 8 2 8 100
A. calcoaceticus/baumannii complex (45) Tigecycline 0.06 to 4 0.5 2 NA
Cefoxitin 32 to N32 N32 N32 NA
Ceftriaxone 4 to N64 N64 N64 22
Ciprofloxacin ≤0.12 to N4 N4 N4 NA
Ertapenem 1 to N32 8 N32 NA
Minocycline ≤0.12 to 8 0.25 2 96
Piperacillin–tazobactam ≤0.12 to N128 128 N128 29
A. lwoffii (5) Tigecycline 0.06 to 0.25 0.06 NA NA
Cefoxitin 4 to N32 4 NA NA
Ceftriaxone 2 to 64 4 NA 60
Ciprofloxacin ≤0.12 to 0.5 ≤0.12 NA NA
Ertapenem 0.5 to 8 2 NA NA
Minocycline ≤0.12 to 0.5 ≤0.12 NA 100
Piperacillin–tazobactam ≤0.12 to N128 ≤0.12 NA 60
P. aeruginosa (58) Tigecycline 4 to N32 8 32 NA
Cefoxitin N32 N32 N32 NA
Ceftriaxone 8 to N64 N64 N64 5
Ciprofloxacin ≤0.12 to N4 0.5 N4 60
Ertapenem 1 to N32 8 N32 NA
Minocycline 8 to N32 16 N32 NA
Piperacillin–tazobactam 2 to N128 8 128 86
S. maltophilia (6) Tigecycline 0.5 to 1 0.5 NA NA
Cefoxitin N32 N32 NA NA
Ceftriaxone N64 N64 NA NA
Ciprofloxacin 2 to N4 4 NA NA
Ertapenem 32 to N32 N32 NA NA
Minocycline 0.25 to 1 0.25 NA 100
Piperacillin–tazobactam 16 to N128 N128 NA NA
a
FDA, Tygacil label accessed May 2009 (Wyeth Pharmaceuticals, 2005) or CLSI (2009a), M100-S19, assigned breakpoints were used to determine
percentage susceptible. NA = breakpoints not established.
b
NA = not applicable—less than 10 organisms analyzed.
c
P. rettgeri (9), P. stuartii (1).
d
S. marcescens (9), S. liquefaciens (4), S. plymuthica (1).

Asia (Fritsche et al., 2005; Reinert et al., 2007; Sader et al., Tigecycline has been shown to be safe and effective in
2005). All of these studies reported good activity against a double-blind, multicenter, global clinical trials for cSSSI,
wide variety of pathogens, most of which were inhibited cIAI, and, most recently, CABP (Babinchak et al., 2005;
by ≤2 μg/mL of tigecycline. Dukart et al., 2006; Ellis-Grosse et al., 2005; Wyeth
414 P.J. Petersen et al. / Diagnostic Microbiology and Infectious Disease 66 (2010) 407–418

Table 4
In vitro activities of tigecycline and comparative antibacterial agents against Gram-positive clinical isolates from DFI
Organism Antibiotic MIC (μg/mL) %Sa
(no. of isolates tested)
Range 50% 90%
MRSA (99) Tigecycline 0.06 to 0.5 0.12 0.25 100
Cefoxitin 4 to N32 32 N32 NA
Ceftriaxone 4 to N64 64 N64 NA
Ciprofloxacin ≤0.12 to N4 N4 N4 21
Ertapenem 0.25 to N32 2 N32 NA
Linezolid 1 to 4 2 2 100
Minocycline ≤0.12 to 16 0.25 8 89
Piperacillin–tazobactam 2 to N128 16 128 NA
Vancomycin 0.5 to 2 1 2 100
Oxacillin 8 to N32 N32 N32 0
MSSA (348) Tigecycline ≤0.03 to 0.5 0.12 0.25 100
Cefoxitin ≤0.5 to 32 4 4 NA
Ceftriaxone 1 to 64 4 4 99.7
Ciprofloxacin ≤0.12 to N4 0.5 0.5 92
Ertapenem 0.06 to 4 0.25 0.25 99.7
Linezolid 1 to 4 2 4 100
Minocycline ≤0.12 to 8 ≤0.12 0.25 99
Piperacillin–tazobactam ≤0.12 to 4 1 2 100
Vancomycin 0.5 to 2 1 2 100
Oxacillin ≤0.06 to 2 0.5 1 100
MRSE (46) Tigecycline 0.06 to 0.5 0.25 0.5 100
Cefoxitin 4 to N32 8 32 NA
Ceftriaxone 2 to N64 8 32 NA
Ciprofloxacin ≤0.12 to N4 4 N4 43
Ertapenem 0.25 to N32 2 16 NA
Linezolid 0.5 to 2 1 2 100
Minocycline ≤0.12 to 2 0.5 1 100
Piperacillin–tazobactam 0.25 to 64 1 4 NA
Vancomycin 1 to 4 2 2 100
Oxacillin 0.5 to N32 8 N32 0
Methicillin-susceptible Tigecycline 0.06 to 0.5 0.12 0.25 100
S. epidermidis (33) Cefoxitin 1 to 4 2 4 NA
Ceftriaxone 0.5 to 4 2 2 100
Ciprofloxacin ≤0.12 to N4 0.25 N4 85
Ertapenem 0.06 to 0.5 0.25 0.5 100
Linezolid 0.5 to 2 1 2 100
Minocycline ≤0.12 to 1 ≤0.12 0.5 100
Piperacillin–tazobactam ≤0.12 to 0.5 0.25 0.5 100
Vancomycin 1 to 4 2 2 100
Oxacillin ≤0.06 to 0.25 0.12 0.25 100
S. haemolyticus (16) Tigecycline 0.06 to 1 0.25 0.5 94
Cefoxitin 2 to N32 16 N32 NA
Ceftriaxone 4 to N64 32 N64 25
Ciprofloxacin ≤0.12 to N4 N4 N4 31
Ertapenem 0.5 to N32 4 N32 38
Linezolid 0.5 to 2 1 2 100
Minocycline ≤0.12 to 16 0.5 1 88
Piperacillin–tazobactam 0.5 to N128 8 N128 50
Vancomycin 0.5 to 2 1 2 100
Oxacillin 0.12 to N32 32 N32 19
S. hominis (12) Tigecycline 0.06 to 0.5 0.12 0.12 100
Cefoxitin 2 to 32 4 16 NA
Ceftriaxone 2 to 16 4 8 92
Ciprofloxacin ≤0.12 to 0.5 ≤0.12 0.5 100
Ertapenem 0.12 to 16 0.5 4 83
Linezolid 1 to 2 1 2 100
Minocycline ≤0.12 to 0.5 ≤0.12 0.25 100
Piperacillin–tazobactam 0.25 to 4 0.5 4 100
Vancomycin 0.5 to 2 1 2 100
Oxacillin ≤0.06 to N32 0.12 N32 67
P.J. Petersen et al. / Diagnostic Microbiology and Infectious Disease 66 (2010) 407–418 415

Table 4 (continued)
Organism Antibiotic MIC (μg/mL) %Sa
(no. of isolates tested)
Range 50% 90%
S. lugdunensis (12) Tigecycline ≤0.03 to 0.25 0.06 0.25 100
Cefoxitin 2 to 8 4 4 NA
Ceftriaxone 1 to 4 4 4 100
Ciprofloxacin ≤0.12 to 0.25 0.25 0.25 100
Ertapenem 0.5 to 1 0.5 0.5 100
Linezolid 1 to 2 1 1 100
Minocycline ≤0.12 to 0.25 ≤0.12 0.25 100
Piperacillin–tazobactam 0.25 to 2 1 1 100
Vancomycin 0.5 to 1 1 1 100
Oxacillin 0.25 to 4 0.5 1 92
S. simulans (14) Tigecycline 0.06 to 0.12 0.12 0.12 100
Cefoxitin 2 to 16 2 4 NA
Ceftriaxone 2 to 32 4 8 93
Ciprofloxacin ≤0.12 to N4 ≤0.12 N4 86
Ertapenem 0.12 to 1 0.25 1 100
Linezolid 1 to 2 2 2 100
Minocycline ≤0.12 to 0.25 ≤0.12 ≤0.12 100
Piperacillin–tazobactam 0.25 to 2 0.5 1 100
Vancomycin 0.5 to 1 1 1 100
Oxacillin 0.12 to 16 0.25 4 57
Coagulase-negative Tigecycline ≤0.03 to 0.5 0.12 0.25 100
Staphylococcus spp.b (41) Cefoxitin ≤0.5 to N32 4 32 NA
Ceftriaxone 0.12 to N64 4 N64 68
Ciprofloxacin ≤0.12 to N4 0.25 N4 83
Ertapenem 0.06 to N32 0.5 8 78
Linezolid ≤0.25 to 2 2 2 100
Minocycline ≤0.12 to 8 0.25 0.5 95
Piperacillin–tazobactam ≤0.12 to N128 1 64 80
Vancomycin ≤0.25 to 4 1 2 100
Oxacillin 0.12 to N32 1 N32 29
E. faecalis (196) Tigecycline ≤0.03 to 0.25 0.12 0.25 100
Cefoxitin N32 N32 N32 NA
Ceftriaxone 2 to N64 N64 N64 NA
Ciprofloxacin 0.5 to N4 0.5 N4 64
Ertapenem 4 to 32 8 16 NA
Linezolid 1 to 8 2 2 98
Minocycline ≤0.12 to 32 16 16 25
Piperacillin–tazobactam 0.5 to 32 4 8 NA
Vancomycin 0.5 to 4 2 4 100
Enterococcus spp.c (17) Tigecycline 0.06 to 0.25 0.06 0.12 100
Cefoxitin 16 to N32 32 N32 NA
Ceftriaxone 0.5 to N64 16 N64 NA
Ciprofloxacin 0.5 to N4 2 N4 41
Ertapenem 0.12 to N32 8 N32 NA
Linezolid 1 to 4 2 4 82
Minocycline ≤0.12 to 32 ≤0.12 32 47
Piperacillin–tazobactam 2 to N128 16 N128 NA
Vancomycin 0.5 to 8 1 8 82
S. agalactiae (135) Tigecycline ≤0.03 to 0.25 0.06 0.06 100
Cefoxitin 2 to 32 8 8 NA
Ceftriaxone ≤0.03 to 0.25 0.12 0.12 100
Ciprofloxacin 0.5 to N4 0.5 0.5 NA
Ertapenem ≤0.03 to 0.25 0.06 0.12 100
Linezolid 1 to 2 1 2 100
Minocycline ≤0.12 to N32 16 N32 NA
Piperacillin–tazobactam ≤0.12 to 1 0.5 0.5 NA
Vancomycin 0.5 to 1 0.5 1 100
S. anginosus (13) Tigecycline ≤0.03 to 0.06 ≤0.03 0.06 100
Cefoxitin ≤0.5 to 16 8 8 NA
Ceftriaxone ≤0.03 to 0.5 0.25 0.5 100
Ciprofloxacin 0.25 to 0.5 0.5 0.5 NA
Ertapenem 0.06 to 0.5 0.25 0.5 100
(continued on next page)
416 P.J. Petersen et al. / Diagnostic Microbiology and Infectious Disease 66 (2010) 407–418

Table 4 (continued)
Organism Antibiotic MIC (μg/mL) %Sa
(no. of isolates tested)
Range 50% 90%
Linezolid 1 to 2 1 2 100
Minocycline ≤0.12 to 16 ≤0.12 16 NA
Piperacillin–tazobactam ≤0.12 to 0.5 ≤0.12 0.5 NA
Vancomycin 0.5 to 1 1 1 100
S. anginosus groupd (20) Tigecycline ≤0.03 to 0.06 ≤0.03 0.06 100
Cefoxitin ≤0.5 to 16 8 8 NA
Ceftriaxone ≤0.03 to 0.5 0.25 0.5 100
Ciprofloxacin 0.25 to 0.5 0.5 0.5 NA
Ertapenem ≤0.03 to 1 0.25 0.5 100
Linezolid ≤0.25 to 2 1 2 100
Minocycline ≤0.12 to 16 ≤0.12 8 NA
Piperacillin–tazobactam ≤0.12 to 1 ≤0.12 0.5 NA
Vancomycin 0.5 to 1 1 1 100
S. dysgalactiae (27) Tigecycline ≤0.03 to 0.5 0.06 0.25 96
Cefoxitin ≤0.5 to 2 1 2 NA
Ceftriaxone ≤0.03 to 0.06 ≤0.03 0.06 100
Ciprofloxacin 0.25 to N4 0.5 0.5 NA
Ertapenem ≤0.03 ≤0.03 ≤0.03 100
Linezolid 0.5 to 2 1 2 100
Minocycline ≤0.12 to N32 16 N32 NA
Piperacillin–tazobactam ≤0.12 ≤0.12 ≤0.12 NA
Vancomycin ≤0.25 to 1 0.5 0.5 100
S. mitis group (7) Tigecycline 0.06 to 0.5 0.06 NAe 86
Cefoxitin 1 to 16 2 NA NA
Ceftriaxone ≤0.03 to 0.25 0.06 NA 100
Ciprofloxacin 4 to N4 4 NA NA
Ertapenem ≤0.03 to 0.5 0.06 NA 100
Linezolid 0.5 to 1 1 NA 100
Minocycline 0.25 to 32 1 NA NA
Piperacillin–tazobactam ≤0.12 to 1 ≤0.12 NA NA
Vancomycin 0.5 to 1 0.5 NA 100
Streptococcus oralis (14) Tigecycline ≤0.03 to 0.5 ≤0.03 0.25 93
Cefoxitin 1 to 16 2 4 NA
Ceftriaxone 0.06 to 0.5 0.12 0.5 100
Ciprofloxacin 0.5 to N4 4 N4 NA
Ertapenem ≤0.03 to 0.25 0.06 0.12 100
Linezolid 1 to 2 1 2 100
Minocycline ≤0.12 to 16 ≤0.12 8 NA
Piperacillin–tazobactam ≤0.12 to 2 ≤0.12 0.5 NA
Vancomycin 0.5 to 1 1 1 100
S. pyogenes (13) Tigecycline ≤0.03 to 0.25 ≤0.03 0.25 100
Cefoxitin 1 to 2 2 2 NA
Ceftriaxone ≤0.03 to 0.06 ≤0.03 ≤0.03 100
Ciprofloxacin 0.25 to 0.5 0.5 0.5 NA
Ertapenem ≤0.03 ≤0.03 ≤0.03 100
Linezolid 1 to 2 1 2 100
Minocycline ≤0.12 to 16 ≤0.12 8 NA
Piperacillin–tazobactam ≤0.12 ≤0.12 ≤0.12 NA
Vancomycin ≤0.25 to 1 0.5 0.5 100
Corynebacterium spp.f (31) Tigecycline ≤0.03 to 0.5 0.06 0.25 NA
Cefoxitin ≤0.5 to N32 4 N32 NA
Ceftriaxone 0.12 to N64 4 32 NA
Ciprofloxacin ≤0.12 to N4 2 N4 NA
Ertapenem 0.06 to N32 0.5 2 NA
Linezolid ≤0.25 to 1 0.5 0.5 NA
Minocycline ≤0.12 to 16 0.25 16 NA
Piperacillin–tazobactam ≤0.12 to N128 16 64 NA
Vancomycin ≤0.25 to 1 0.5 1 NA
P.J. Petersen et al. / Diagnostic Microbiology and Infectious Disease 66 (2010) 407–418 417

Table 5
In vitro activities of tigecycline and comparative antibacterial agents against Gram-negative and Gram-positive anaerobic clinical isolates from DFI
Organism Antibiotic MIC (μg/mL) %Sa
(no. of isolates tested)
Range 50% 90%
B. fragilis (8) Tigecycline 0.25 to 4 1 NAb 100
Ceftriaxone 2 to N128 32 NA 13
Clindamycin 1 to N64 2 NA 63
Imipenem 0.12 to 0.25 0.25 NA 100
Metronidazole 1 to 4 1 NA 100
P. bivia (11) Tigecycline 0.25 to 2 1 1 100
Ceftriaxone 0.25 to N128 8 32 64
Clindamycin ≤0.03 to N64 ≤0.03 N64 64
Imipenem ≤0.015 to 0.06 0.03 0.06 100
Metronidazole 0.5 to 16 2 8 91
Prevotella spp.c (10) Tigecycline 0.06 to 2 0.12 0.25 100
Ceftriaxone 0.12 to N128 2 64 70
Clindamycin ≤0.03 to N64 ≤0.03 N64 80
Imipenem 0.03 to 0.12 0.03 0.06 100
Metronidazole 0.5 to 8 1 4 100
C. perfringens (4) Tigecycline 0.12 to 2 2 NA 100
Ceftriaxone 0.06 to 4 0.5 NA 100
Clindamycin 0.06 to 2 0.06 NA 100
Imipenem 0.06 to 0.25 0.06 NA 100
Metronidazole 0.5 to 4 2 NA 100
F. magna (30) Tigecycline 0.12 to 0.25 0.12 0.25 100
Ceftriaxone 1 to 16 8 16 100
Clindamycin 0.06 to N64 2 N64 53
Imipenem ≤0.015 to 0.12 0.06 0.06 100
Metronidazole 0.12 to 2 0.5 1 100
P. asaccharolyticus (10) Tigecycline ≤0.03 to 0.25 0.12 0.25 100
Ceftriaxone 0.12 to 1 0.25 1 100
Clindamycin ≤0.03 to N64 0.5 N64 60
Imipenem ≤0.015 to 0.06 ≤0.015 0.06 100
Metronidazole 0.25 to 2 1 2 100
Peptostreptococcus spp.d (7) Tigecycline 0.06 to 0.5 0.06 NA 100
Ceftriaxone 0.25 to 64 0.5 NA 86
Clindamycin ≤0.03 to N64 0.25 NA 71
Imipenem ≤0.015 to 0.12 0.06 NA 100
Metronidazole ≤0.06 to 2 1 NA 100
Anaerococcus spp.e (6) Tigecycline 0.12 to 0.25 0.12 NA 100
Ceftriaxone 0.12 to 1 0.25 NA 100
Clindamycin ≤0.03 to N64 0.25 NA 83
Imipenem ≤0.015 to 0.25 ≤0.015 NA 100
Metronidazole 1 to 128 2 NA 67
a
FDA, Tygacil label accessed May 2009 (Wyeth Pharmaceuticals, 2005) or CLSI (2007), M11-A7, assigned breakpoints were used to determine percentage
susceptible.
b
NA = not applicable—less than 10 isolates analyzed.
c
P. buccae (2), P. denticola (1), P. disiens (1), P. intermedia (1), P. melaninogenica (1), P. oralis (1), Prevotella spp. (3).
d
P. anaerobius (4), P. micros (1), P. tetradius (2).
e
A. hydrogenalis (1), A. prevotii (5).

Notes to Table 4:
a
FDA, Tygacil label accessed May 2009 (Wyeth Pharmaceuticals, 2005) or CLSI (2009a), M100-S19, assigned breakpoints were used to determine
percentage susceptible. NA = breakpoints not established.
b
S. capitis (8), S. caprae (4), S. carnosus (1), S. cohnii (7), S. hyicus (1), S. intermedius (1), S. kloosii (2), S. pasteuri (1), S. sciuri (6), S. warneri (8),
S. xylosus (2).
c
E. avium (5), E. casseliflavus (3), E. faecium (6), E. gallinarum (2), E. harae (1).
d
S. anginosus (13), S. constellatus (5), S. intermedius (2).
e
NA = not applicable—less than 10 isolates analyzed.
f
C. glucuronolyticum (1), C. accolens (1), C. amycolatum (6), C. diphtheriae (1), C. macginleyi (1), C. minutissimum (4), C. striatum (11), Coryne-
bacterium spp. (6).
418 P.J. Petersen et al. / Diagnostic Microbiology and Infectious Disease 66 (2010) 407–418

Pharmaceuticals, 2005). In summary, the in vitro activity aureus isolates from phase 3 clinical trials of tigecycline for complicated
of tigecycline against a broad spectrum of Gram-positive skin and skin structure infections. Antimicrob Agents Chemother 50:
505–510.
and Gram-negative pathogens isolated from patients Jones CH, et al. (2008) Genetic determinants of tetracycline resistance and
enrolled in phase 3 clinical trials conducted worldwide for their effect on tetracycline and glycylcycline antibiotics. Anti-Infect
DFI showed an excellent susceptibility profile and suggests Agents Med Chem 7:84–96.
utility in the treatment of patients with this disease. Jones CH, et al. (2009) Characterization and sequence analysis of extended-
spectrum-{beta}-lactamase–encoding genes from Escherichia coli,
Klebsiella pneumoniae, and Proteus mirabilis isolates collected during
References tigecycline phase 3 clinical trials. Antimicrob Agents Chemother 53:
465–475.
Babinchak T, et al. (2005) The efficacy and safety of tigecycline in the Kenny GE, Cartwright FD (2001) Susceptibilities of Mycoplasma hominis,
treatment of complicated intra-abdominal infections: analysis of pooled M. pneumoniae, and Ureaplasma urealyticum to GAR-936, dalfopristin,
clinical trial data. Clin Infect Dis 41(Suppl 5):S354–S367. dirithromycin, evernimicin, gatifloxacin, linezolid, moxifloxacin, qui-
Bradford PA, et al. (2005) In vitro activity of tigecycline against isolates nupristin–dalfopristin and telithromycin compared to their susceptibil-
from patients enrolled in phase 3 clinical trials for complicated skin and ities to reference macrolides, tetracyclines and quinolones. Antimicrob
skin structure infections and complicated intra-abdominal infections. Agents Chemother 45:2604–2608.
Clin Infect Dis 41(Suppl 5):S315–S332. Lipsky BA, et al. (2004) Diagnosis and treatment of diabetic foot infections.
Bradford PA, et al. (2008) In vitro activity of tigecycline and occurrence of Clin Infect Dis 39:885–910.
tetracycline resistance determinants in isolates from patients enrolled in Olson MW, et al. (2006) Functional, biophysical, and structural bases for
phase 3 clinical trials for community acquired pneumonia. Clin antibacterial activity of tigecycline. Antimicrob Agents Chemother 50:
Microbiol Infect 14:882–886. 2156–2166.
Clinical and Laboratory Standards Institute (CLSI) (2006) Performance Petersen PJ, Bradford PA (2005) Effect of medium age and supplementation
standards for antimicrobial disk susceptibility tests; approved standard— with the biocatalytic oxygen-reducing reagent oxyrase on in vitro
ninth edition, M02-A9, Vol. 26. 2006. Wayne (PA): CLSI. activities of tigecycline against recent clinical isolates. Antimicrob
Clinical and Laboratory Standards Institute (CLSI) (2007) Methods for Agents Chemother 49:3910–3918.
antimicrobial susceptibility testing of anaerobic bacteria: approved Petersen PJ, et al. (1999) In vitro and In vivo antimicrobial activities of a
standard—seventh edition, M11-A7, Vol. 27. 2007. Wayne (PA): CLSI. novel glycylcycline, the 9-t-butylglycylamido derivative of minocycline
Clinical and Laboratory Standards Institute (CLSI) (2009a) Performance (GAR-936). Antimicrob Agents Chemother 43:738–744.
standards for antimicrobial susceptibility testing: 19th informational Poole K (2005) Efflux-mediated antimicrobial resistance. J Antimicrob
supplement (M100-S19), Vol. 29. 2009. Wayne (PA): CLSI. Chemother 56:20–51.
Clinical and Laboratory Standards Institute (CLSI) (2009b) Methods for Reinert RR, et al. (2007) Antimicrobial susceptibility among organisms
dilution antimicrobial susceptibility tests for bacteria that grow from the Asia/Pacific Rim, Europe and Latin and North America
aerobically: approved standard—eighth edition, M07-A8, Vol. 29. collected as part of TEST and the in vitro activity of tigecycline.
2009. Wayne (PA): CLSI. J Antimicrob Chemother 60:1018–1029.
Dean CR, et al. (2003) Efflux-mediated resistance to tigecycline (GAR-936) Roblin PM, Hammerschlag MR (2000) In vitro activity of GAR-936 against
in Pseudomonas aeruginosa PAO1. Antimicrob Agents Chemother 47: Chlamydia pneumoniae and Chlamydia trachomatis. Int J Antimicrob
972–978. Agents 16:61–63.
Dukart G, et al. (2006) Integrated results of 2 phase 3 studies comparing Ruzin A, et al. (2005) Influence of transcriptional activator RamA on expres-
tigecycline (TGC) with levofloxacin (LEV) in patients (Pts) with sion of multidrug efflux pump AcrAB and tigecycline susceptibility in
community-acquired pneumonia (CAP). 2006. Abstr. L-1450. 46th Klebsiella pneumoniae. Antimicrob Agents Chemother 49:1017–1022.
Interscience Conference on Antimicrobial Agents and Chemotherapy. Sader HS, et al. (2005) Tigecycline activity tested against 26,474
San Francisco: American Society for Microbiology. bloodstream infection isolates: a collection from 6 continents. Diagn
Edlund C, Nord CE (2000) In-vitro susceptibility of anaerobic bacteria to Microbiol Infect Dis 52:181–186.
GAR-936, a new glycylcycline. Clin Microbiol Infect 6:158–163. Sotto A, et al. (2007) In vitro activity of tigecycline against strains isolated
Ellis-Grosse E, et al. (2005) The efficacy and safety of tigecycline in the from diabetic foot ulcers. Pathol Biol (Paris) 55:398–406.
treatment of skin and skin structure infections: results of two double- Sum PE, Petersen P (1999) Synthesis and structure–activity relationship of
blind phase 3 comparison studies with vancomycin/aztreonam. Clin novel glycylcycline derivatives leading to the discovery of GAR-936.
Infect Dis 41(Suppl 5):S341–S353. Bioorg Med Chem Lett 9:1459–1462.
Fritsche TR, et al. (2005) Potency and spectrum of tigecycline tested against Tentolouris N, et al. (2006) Prevalence of methicillin-resistant Staphylo-
an international collection of bacterial pathogens associated with skin and coccus aureus in infected and uninfected diabetic foot ulcers. Clin
soft tissue infections (2000–2004). Diagn Microbiol Infect Dis 52: Microbiol Infect 12:186–189.
195–201. Tuckman M, et al. (2007) Occurrence of tetracycline resistance genes among
Jacobus NV, et al. (2004) In vitro activities of tigecycline against the Bac- Escherichia coli isolates from the phase 3 clinical trials for tigecycline.
teroides fragilis group. Antimicrob Agents Chemother 48:1034–1036. Antimicrob Agents Chemother 51:3205–3211.
Jones C, Petersen P (2005) Tigecycline: a review of preclinical and clinical Visalli MA, et al. (2003) AcrAB multidrug efflux pump is associated with
studies of the first-in-class glycylcycline antibiotic. Drugs Today 41: reduced levels of susceptibility to tigecycline (GAR-936) in Proteus
637–659. mirabilis. Antimicrob Agents Chemother 47:665–669.
Jones CH, et al. (2006) Diagnostic PCR analysis of the occurrence of Wyeth Pharmaceuticals (2005) Tygacil™ [package insert]. Collegeville
methicillin and tetracycline resistance genes among Staphylococcus (PA): Wyeth Pharmaceuticals Inc. Vol. Accessed May 15, 2009.

You might also like