ABSTRACT

The activities of azithromycin, fusidic acid, vancomycin, doxycycline, and minocycline were evaluated alone and in combination with SPR741. A total of 202 Escherichia coli and 221 Klebsiella pneumoniae isolates were selected, and they included a genome-sequenced subset (n = 267), which was screened in silico for β-lactamase, macrolide-lincosamide-streptogramin (MLS), and tetracycline (tet) genes. Azithromycin (>16 mg/liter), fusidic acid (>64 mg/liter), vancomycin (>16 mg/liter), and SPR741 (>8 mg/liter) showed off-scale MICs when each was tested alone against all isolates. MIC50/90 results of 0.5/8 mg/liter, 4/>32 mg/liter, 16/>16 mg/liter, 2/32 mg/liter, and 0.25/4 mg/liter were obtained for azithromycin-SPR741, fusidic acid-SPR741, vancomycin-SPR741, doxycycline-SPR741 and minocycline-SPR741, respectively, against all isolates. Overall, azithromycin-SPR741 (MIC90, 2 to 4 mg/liter) and minocycline-SPR741 (MIC90, 0.5 to 2 mg/liter) showed the lowest MIC90 values against different subsets of E. coli isolates, except for azithromycin-SPR741 (MIC90, 16 mg/liter) against the AmpC and metallo-β-lactamase subsets. In general, minocycline-SPR741 (MIC90, 2 to 8 mg/liter) had the lowest MIC90 against K. pneumoniae isolates producing different groups of β-lactamases. The azithromycin-SPR741 MIC (MIC50/90, 2/32 mg/liter) was affected by MLS genes (MIC50/90 of 0.25/2 mg/liter against isolates without MLS genes), whereas doxycycline-SPR741 (MIC50/90, 0.5/2 versus 8/32 mg/liter) and minocycline-SPR741 (MIC50/90, 0.25/1 versus 1/8 mg/liter) MIC results were affected when tested against isolates carrying tet genes in general. However, minocycline-SPR741 inhibited 88.2 to 92.9% of tet-positive isolates regardless of the tet gene. The azithromycin-SPR741 MIC results (MIC50/90, 1/16 mg/liter) against isolates with enzymatic MLS mechanisms were lower than against those with ribosomal protection (MIC50/90, 16/>32 mg/liter). SPR741 increased the in vitro activity of tested codrugs at different levels and seemed to be dependent on the species and resistance mechanisms of the respective codrug.

INTRODUCTION

Survey studies have reported that 4 to 6% of patients develop health care-associated infections (HAIs) in the United States, and Enterobacteriaceae isolates account for approximately 27% of infections (1, 2). In addition, the proportion of isolates producing extended-spectrum β-lactamases (ESBLs) have increased since 2000 and are responsible for approximately 197,400 cases and 9,100 deaths per year (3). ESBL-producing Enterobacteriaceae isolates have spread in the nosocomial and community settings, complicating the empirical treatment of infections caused by these organisms (4). The increased frequency of ESBL-producing Enterobacteriaceae isolates may increase the use of more potent antimicrobial agents (5, 6), including carbapenems, and more carbapenem use increases selection pressure for emerging and disseminating carbapenem-resistant Enterobacteriaceae (CRE) isolates (7).
Although CRE isolates are relatively uncommon in the United States, the number of U.S. facilities reporting CRE isolates has risen steadily and includes 4% of acute care hospitals and 18% of long-term acute care facilities. In addition to CRE, organisms such as Acinetobacter baumannii and other nonfermentative isolates can also express a diverse array of antimicrobial resistance mechanisms, which can compromise therapy. These often hard-to-treat infections remain one of the most pressing challenges in the infectious diseases field and drive the development of new therapeutic strategies. SPR741 is a novel polymyxin analog with no direct in vitro activity against Gram-negative bacteria, but it interacts with the outer membrane and compromises the integrity of the lipopolysaccharide. SPR741 increases cell permeability and enables antimicrobial compounds to enter (8).
We recently reported on the activity of SPR741 in combination with a series of β-lactam agents tested against a challenge set of Enterobacteriaceae (9). This study aimed to expand on the previous report and describe the in vitro activity of several more commonly used and clinically available anti-Gram-positive and tetracycline agents tested alone and in combination with SPR741 against the same challenge set of Enterobacteriaceae utilized in the previous study (9). In addition, a subset of pathogens had their genome sequenced and screened in silico for acquired macrolide-lincosamide-streptogramin (MLS) and tetracycline resistance genes for a more granular analysis.

RESULTS

MIC results against resistant subsets of E. coli.

MIC results against resistant Escherichia coli subsets are shown in Table 1. Azithromycin-SPR741 showed MIC90 values of 2 to 4 mg/liter when tested against E. coli subsets, except against isolates carrying AmpC and metallo-β-lactamase (MBL) genes, against which the MIC90 value was 16 mg/liter. These results translated into increased azithromycin potency of at least 8- to 16-fold compared to the drug tested alone (MIC90, >16 mg/liter) against E. coli isolates other than AmpC and MBL producers. SPR741 increased the potency (MIC90) of fusidic acid (MIC90, 4 to 8 mg/liter) at least 16- to 32-fold compared to fusidic acid tested alone (MIC90, >64 mg/liter) against each E. coli subset. In general, although the MIC50 results for vancomycin-SPR741 (MIC50, 8 to 16 mg/liter) against E. coli decreased relative to vancomycin (MIC50, >16 mg/liter), they remained elevated, as did the MIC90 (≥16 mg/liter). Similarly, SPR741 did not decrease the doxycycline MIC values (MIC90, 32 mg/liter) appreciably against E. coli subsets, except against AmpC producers (MIC50/90, 0.12/4 mg/liter). In contrast, the minocycline-SPR741 combination showed MIC90 results of 0.5 to 2 mg/liter against these challenge sets of E. coli, translating into 99.0 to 100.0% susceptibility based on current breakpoints from as low as 40.6% susceptible when minocycline was tested alone. In other words, SPR741 increased the in vitro potency of minocycline 4- to 32-fold compared to minocycline tested alone against E. coli.
TABLE 1
TABLE 1 Antimicrobial activity of SPR741 combinations and other agents against E. coli isolates by β-lactamase groupa
Antimicrobial agent(s) (no. of isolates)b MIC (mg/liter) Interpretationc
50% 90% Range % S % I % R
AmpC (10)            
    Azithromycin-SPR741 0.12 16 0.06 to 16      
    Fusidic acid-SPR741 2 4 0.5 to 4      
    Vancomycin-SPR741 8 16 1 to >16      
    Doxycycline 1 8 0.5 to >64 60.0 30.0 10.0
    Doxycycline-SPR741 0.12 4 0.12 to 32 90.0 0.0 10.0
    Minocycline 1 2 0.5 to 16 90.0 0.0 10.0
    Minocycline-SPR741 0.06 0.5 0.03 to 1 100.0 0.0 0.0
    Polymyxin B ≤0.25 0.5 ≤0.25 to 0.5 100.0   0.0
    Colistin ≤0.12 ≤0.12 ≤0.12 to 0.25 100.0   0.0
    Ceftazidime 32 128 16 to >256 0.0 0.0 100.0
    Meropenem 0.03 0.06 ≤0.015 to 0.12 100.0 0.0 0.0
    Piperacillin-tazobactam 4 16 4 to 128 90.0 0.0 10.0
    Levofloxacin 1 >4 ≤0.06 to >4 40.0 10.0 50.0
 
ESBL (97)            
    Azithromycin-SPR741 0.5 2 ≤0.03 to >32      
    Fusidic acid-SPR741 4 8 0.06 to >32      
    Vancomycin-SPR741 16 >16 ≤0.5 to >16      
    Doxycycline 8 >32 0.5 to >32 45.4 19.6 35.1
    Doxycycline-SPR741 4 32 ≤0.015 to >32 71.1 7.2 21.6
    Minocycline 2 16 0.25 to >32 69.1 8.2 22.7
    Minocycline-SPR741 0.25 2 ≤0.015 to 8 99.0 1.0 0.0
    Polymyxin B ≤0.25 2 ≤0.25 to 8 97.9   2.1
    Colistin ≤0.12 0.25 ≤0.12 to 2 100.0   0.0
    Ceftazidime 16 128 ≤2 to >256 28.9 9.3 61.9
    Meropenem 0.03 0.06 ≤0.015 to 1 100.0 0.0 0.0
    Piperacillin-tazobactam 4 32 ≤1 to >256 86.6 9.3 4.1
    Levofloxacin >4 >4 ≤0.06 to >4 14.4 2.1 83.5
 
KPC (46)            
    Azithromycin-SPR741 0.25 4 ≤0.03 to 16      
    Fusidic acid-SPR741 2 4 0.5 to >32      
    Vancomycin-SPR741 8 >16 ≤0.5 to >16      
    Doxycycline 2 32 0.5 to >32 52.2 15.2 32.6
    Doxycycline-SPR741 1 32 0.03 to 32 71.7 10.9 17.4
    Minocycline 2 16 0.25 to >32 82.6 4.3 13.0
    Minocycline-SPR741 0.12 0.5 0.03 to 2 100.0 0.0 0.0
    Polymyxin B ≤0.25 1 ≤0.25 to >8 97.8   2.2
    Colistin ≤0.12 0.25 ≤0.12 to >4 97.8   2.2
    Ceftazidime 64 128 4 to >256 2.2 2.2 95.7
    Meropenem 4 8 0.03 to >32 10.9 17.4 71.7
    Piperacillin-tazobactam 256 >256 32 to >256 0.0 10.9 89.1
    Levofloxacin >4 >4 ≤0.06 to >4 26.1 4.3 69.6
 
MBL (32)            
    Azithromycin-SPR741 2 16 ≤0.03 to >32      
    Fusidic acid-SPR741 2 8 ≤0.03 to 8      
    Vancomycin-SPR741 8 16 ≤0.5 to 16      
    Doxycycline 16 >32 0.5 to >32 25.0 9.4 65.6
    Doxycycline-SPR741 4 32 ≤0.015 to >32 56.2 15.6 28.1
    Minocycline 8 >32 1 to >32 40.6 25.0 34.4
    Minocycline-SPR741 0.25 2 ≤0.015 to 4 100.0 0.0 0.0
    Polymyxin B ≤0.25 0.5 ≤0.25 to 2 100.0   0.0
    Colistin ≤0.12 0.25 ≤0.12 to 0.5 100.00   0.0
    Ceftazidime >256 >256 ≤2 to >256 3.1 0.0 96.9
    Meropenem >32 >32 ≤0.015 to >32 6.2 3.1 90.6
    Piperacillin-tazobactam >256 >256 ≤1 to >256 9.4 3.1 87.5
    Levofloxacin >4 >4 ≤0.06 to >4 9.4 3.1 87.5
 
OXA-48-like (17)            
    Azithromycin-SPR741 0.12 2 ≤0.03 to 4      
    Fusidic acid-SPR741 2 4 0.25 to 4      
    Vancomycin-SPR741 8 16 ≤0.5 to >16      
    Doxycycline 8 >32 0.25 to >32 47.1 11.8 41.2
    Doxycycline-SPR741 2 32 0.06 to >32 58.8 11.8 29.4
    Minocycline 2 16 0.25 to >32 76.5 0.0 23.5
    Minocycline-SPR741 0.12 2 0.06 to 4 100.0 0.0 0.0
    Polymyxin B ≤0.25 0.5 ≤0.25 to 0.5 100.0   0.0
    Colistin ≤0.12 0.25 ≤0.12 to 0.5 100.0   0.0
    Ceftazidime 16 256 ≤2 to 256 47.1 0.0 52.9
    Meropenem 0.5 32 0.12 to >32 70.6 0.0 29.4
    Piperacillin-tazobactam 128 >256 64 to >256 0.0 23.5 76.5
    Levofloxacin >4 >4 ≤0.06 to >4 41.2 0.0 58.8
a
Abbreviations: ESBL, extended-spectrum β-lactamase; KPC, Klebsiella pneumoniae carbapenemase; MBL, metallo-β-lactamase.
b
In each combination, SPR741 was used at a fixed concentration of 8 mg/liter. Elevated MIC results for azithromycin (>16 mg/liter), fusidic acid (>64 mg/liter), and vancomycin (>16 mg/liter) were obtained for these agents when tested alone. MIC results of >8 mg/liter were observed for SPR741 tested alone.
c
MIC results obtained against clinical isolates were interpreted using the CLSI M100 document (25), including for doxycycline-SPR741 and minocycline-SPR741, which used the respective CLSI breakpoints for these tetracyclines for comparison. Polymyxin and colistin had EUCAST breakpoints applied (26). S, susceptible; I, intermediate; R, resistant.

MIC results against resistant subsets of K. pneumoniae.

MIC results against resistant K. pneumoniae subsets are shown in Table 2. In general, the azithromycin-SPR741, fusidic-acid-SPR741, and vancomycin-SPR741 combinations did not show potent MIC90 results (>16 mg/liter) against K. pneumoniae isolates. Exceptions were observed for azithromycin-SPR741 tested against isolates producing AmpC (MIC50, 0.25 mg/liter), ESBL (MIC50/90, 0.25/4 mg/liter), and OXA-48 (MIC50/90, 2/8 mg/liter). MIC90 values of 32 to 64 and 16 to 32 mg/liter were obtained for doxycycline and doxycycline-SPR741 tested against K. pneumoniae. Minocycline and minocycline-SPR741 had MIC50 values of 8 and 0.5 mg/liter against K. pneumoniae, respectively. Where available, SPR741 decreased minocycline MIC90 results 4- to 8-fold compared to minocycline alone, or to 2 to 8 mg/liter from 16 to 64 mg/liter. This decrease in the minocycline MIC90 values in the presence of SPR741 translated into susceptibility rates of 88.1 to 100.0% from 26.7 to 49.5% obtained for minocycline alone.
TABLE 2
TABLE 2 Antimicrobial activity of SPR741 combinations and other agents against K. pneumoniae isolates by β-lactamase groupa
Antimicrobial agent (no. of isolates)b MIC (mg/liter) Interpretationc
50% 90% Range % S % I % R
AmpC (6)            
    Azithromycin-SPR741 0.25   0.06 to >32      
    Fusidic acid-SPR741 8   4 to >32      
    Vancomycin-SPR741 16   8 to >16      
    Doxycycline 16   0.5 to 32 33.3 0.0 66.7
    Doxycycline-SPR741 8   0.5 to 32 33.3 16.7 50.0
    Minocycline 8   2 to 16 33.3 50.0 16.7
    Minocycline-SPR741 0.5   0.25 to 4 100.0 0.0 0.0
    Polymyxin B ≤0.25     83.3   16.7
    Colistin ≤0.12   ≤0.12 to >4 83.3   16.7
    Ceftazidime 16   16 to 64 0.0 0.0 100.0
    Meropenem 0.03   0.03 to 0.12 100.0 0.0 0.0
    Piperacillin-tazobactam 8   4 to 256 66.7 16.7 16.7
    Levofloxacin ≤0.06   ≤0.06 to >4 50.0 33.3 16.7
 
ESBL (101)            
    Azithromycin-SPR741 0.25 4 0.06 to >32      
    Fusidic acid-SPR741 8 >32 0.12 to >32      
    Vancomycin-SPR741 16 >16 2 to >16      
    Doxycycline 8 >32 0.25 to >32 46.5 12.9 40.6
    Doxycycline-SPR741 2 32 0.25 to >32 55.4 18.8 25.7
    Minocycline 8 >32 0.25 to >32 49.5 20.8 29.7
    Minocycline-SPR741 0.5 8 0.03 to 32 88.1 8.9 3.0
    Polymyxin B 1 2 ≤0.25 to >8 95.0   5.0
    Colistin ≤0.12 0.5 ≤0.12 to >4 97.0   3.0
    Ceftazidime 32 256 ≤2 to >256 15.8 5.0 79.2
    Meropenem 0.03 0.12 ≤0.015 to 16 95.0 2.0 3.0
    Piperacillin-tazobactam 8 >256 ≤1 to >256 65.3 15.8 18.8
    Levofloxacin 2 >4 ≤0.06 to >4 52.5 10.9 36.6
 
KPC (74)            
    Azithromycin-SPR741 1 >32 ≤0.03 to >32      
    Fusidic acid-SPR741 8 >32 0.06 to >32      
    Vancomycin-SPR741 16 >16 2 to >16      
    Doxycycline 4 32 0.5 to >32 63.5 16.2 20.3
    Doxycycline-SPR741 1 16 0.12 to >32 79.7 8.1 12.2
    Minocycline 8 16 1 to >32 44.6 33.8 21.6
    Minocycline-SPR741 0.5 4 0.06 to >32 94.6 1.4 4.1
    Polymyxin B 0.5 >8 ≤0.25 to >8 77.0   23.0
    Colistin ≤0.12 >4 ≤0.12 to >4 77.0   23.0
    Ceftazidime >256 >256 32 to >256 0.0 0.0 100.0
    Meropenem 32 >32 4 to >32 0.0 0.0 100.0
    Piperacillin-tazobactam >256 >256 128 to >256 0.0 0.0 100.0
    Levofloxacin >4 >4 ≤0.06 to >4 10.8 2.7 86.5
 
MBL (25)            
    Azithromycin-SPR741 1 >32 0.12 to >32      
    Fusidic acid-SPR741 16 >32 4 to >32      
    Vancomycin-SPR741 16 >16 4 to >16      
    Doxycycline 16 32 1 to >32 40.0 4.0 56.0
    Doxycycline-SPR741 2 16 0.5 to 32 52.0 12.0 36.0
    Minocycline 8 16 2 to >32 32.0 28.0 40.0
    Minocycline-SPR741 0.5 2 0.25 to 4 100.0 0.0 0.0
    Polymyxin B 1 >8 ≤0.25 to >8 84.0   16.0
    Colistin ≤0.12 >4 ≤0.12 to >4 84.0   16.0
    Ceftazidime >256 >256 128 to >256 0.0 0.0 100.0
    Meropenem >32 >32 0.5 to >32 4.0 4.0 92.0
    Piperacillin-tazobactam >256 >256 32 to >256 0.0 4.0 96.0
    Levofloxacin >4 >4 0.5 to >4 20.0 4.0 76.0
 
OXA-48-like (15)            
    Azithromycin-SPR741 2 8 0.06 to 16      
    Fusidic acid-SPR741 16 >32 2 to >32      
    Vancomycin-SPR741 16 >16 4 to >16      
    Doxycycline 16 32 1 to >32 13.3 13.3 73.3
    Doxycycline-SPR741 8 16 0.5 to 32 33.3 20.0 46.7
    Minocycline 8 16 2 to >32 26.7 33.3 40.0
    Minocycline-SPR741 0.5 2 0.12 to 8 93.3 6.7 0.0
    Polymyxin B 1 8 ≤0.25 to >8 80.0   20.0
    Colistin 0.25 >4 ≤0.12 to >4 80.0   20.0
    Ceftazidime 128 256 4 to 256 6.7 6.7 86.7
    Meropenem 16 32 1 to >32 6.7 13.3 80.0
    Piperacillin-tazobactam >256 >256 64 to >256 0.0 6.7 93.3
    Levofloxacin >4 >4 0.5 to >4 6.7 6.7 86.7
a
Abbreviations: ESBL, extended-spectrum β-lactamase; KPC, Klebsiella pneumoniae carbapenemase; MBL, metallo-β-lactamase.
b
In each combination, SPR741 was used at a fixed concentration of 8 mg/liter. Elevated MIC results for azithromycin (>16 mg/liter), fusidic acid (>64 mg/liter), and vancomycin (>16 mg/liter) were obtained for these agents when tested alone. MIC results of >8 mg/liter were observed for SPR741 tested alone.
c
MIC results obtained against clinical isolates were interpreted using the CLSI M100 document (25), including for doxycycline-SPR741 and minocycline-SPR741, which used the respective CLSI breakpoints for these tetracyclines for comparison. Polymyxin and colistin had EUCAST breakpoints applied (26). S, susceptible; I, intermediate; R, resistant.

MIC results against subsets of MLS and tetracycline-resistant isolates.

Azithromycin-SPR741 showed MIC50 and MIC90 values of 0.25 and 2 mg/liter against E. coli and K. pneumoniae isolates, respectively, that did not carry MLS resistance genes (Table 3), whereas MIC results 8- to 16-fold higher (MIC50/90, 2/32 mg/liter) were observed against isolates that carried those genes. Also, the azithromycin-SPR741 (MIC50/90, 0.12/0.25 mg/liter) MIC results against E. coli isolates that were negative for MLS genes were 2- to 16-fold lower than those observed for K. pneumoniae (Table 3). The azithromycin-SPR741 (MIC50/90, 1/16 mg/liter) MIC results against isolates carrying genes encoding MLS-inactivating enzymes were at least 4-fold lower (MIC50/90, 16/>32 mg/liter) than those obtained against isolates carrying genes encoding ribosomal modifications (RNA methylation) or ribosomal protection (Table 3).
TABLE 3
TABLE 3 Antimicrobial activity of SPR741 combinations, doxycycline, and minocycline against specific isolate subsetsa
Combination and genotype (no. of isolates)b No. (cumulative %) of isolates inhibited at MIC (mg/liter) of: MIC (mg/ml)
≤0.03 0.06 0.12 0.25 0.5 1 2 4 8 16 32 >32 50% 90%
Azithromycin-SPR741                            
    MLS gene negative (115) 2 (1.7) 14 (13.9) 27 (37.4) 35 (67.8) 14 (80.0) 8 (87.0) 4 (90.4) 6 (95.7) 3 (98.3) 0 (98.3) 0 (98.3) 2 (100.0) 0.25 2
        E. coli (28) 2 (7.1) 11 (46.4) 12 (89.3) 1 (92.9) 0 (92.9) 0 (92.9) 0 (92.9) 2 (100.0)         0.12 0.25
        K. pneumoniae (87)   3 (3.5) 15 (20.7) 34 (59.8) 14 (75.9) 8 (85.1) 4 (89.7) 4 (94.3) 3 (97.7) 0 (97.7) 0 (97.7) 2 (100.0) 0.25 4
    MLS gene positive (152)c 2 (1.3) 0 (1.3) 4 (4.0) 8 (9.2) 18 (21.1) 37 (45.4) 38 (70.4) 9 (76.3) 6 (80.3) 13 (88.8) 2 (90.1) 15 (100.0) 2 32
        Enzyme (128)d 2 (1.6) 0 (1.6) 4 (4.7) 6 (9.4) 18 (23.4) 34 (50.0) 35 (77.3) 8 (83.6) 6 (88.3) 6 (93.0) 1 (93.8) 8 (100.0) 1 16
        Ribosomal alteration (24)e       2 (8.3) 0 (8.3) 3 (20.8) 3 (33.3) 1 (37.5) 0 (37.5) 7 (66.7) 1 (70.8) 7 (100.0) 16 >32
 
Doxycycline                            
    tet gene negative (103)       1 (1.0) 3 (3.9) 31 (34.0) 28 (61.2) 20 (80.6) 14 (94.2) 5 (99.0) 0 (99.0) 1 (100.0) 2 8
        E. coli (17)         2 (11.8) 10 (70.6) 2 (82.4) 0 (82.4) 2 (94.1) 0 (94.1) 0 (94.1) 1 (100.0) 1 8
        K. pneumoniae (86)       1 (1.2) 1 (2.3) 21 (26.7) 26 (57.0) 20 (80.2) 12 (94.2) 5 (100.0)     2 8
    tet gene positive (164)f           2 (1.2) 1 (1.8) 12 (9.2) 33 (29.3) 42 (54.9) 37 (77.4) 37 (100.0) 16 >32
 
Doxycycline-SPR741                            
    tet gene negative (103) 3 (2.9) 1 (3.9) 5 (8.7) 12 (20.4) 33 (52.4) 32 (83.5) 10 (93.2) 2 (95.2) 4 (99.0) 0 (99.0) 1 (100.0)   0.5 2
        E. coli (17) 3 (17.7) 1 (23.5) 5 (52.9) 7 (94.1) 0 (94.1) 0 (94.1) 0 (94.1) 0 (94.1) 0 (94.1) 0 (94.1) 1 (100.0)   0.12 0.25
        K. pneumoniae (86)       5 (5.8) 33 (38.4) 32 (81.4) 10 (93.0) 2 (95.4) 4 (100.0)       1 2
    tet gene positive (164)f 1 (0.6) 0 (0.6) 2 (1.8) 0 (1.8) 0 (1.8) 3 (3.7) 12 (11.0) 32 (30.5) 37 (53.1) 32 (72.6) 37 (95.1) 8 (100.0) 8 32
 
Minocycline                            
    tet gene negative (103)       2 (1.9) 2 (3.9) 10 (13.6) 50 (62.1) 4 (66.0) 25 (90.3) 10 (100.0)     2 8
        E. coli (17)         2 (11.8) 10 (70.6) 2 (82.4) 0 (82.4) 0 (82.4) 3 (100.0)     1 16
        K. pneumoniae (86)       2 (2.3) 0 (2.3) 0 (2.3) 48 (58.1) 4 (62.8) 25 (91.9) 7 (100.0)     2 8
    tet gene positive (164)           6 (3.7) 41 (28.7) 9 (34.2) 36 (56.1) 36 (78.1) 4 (80.5) 32 (100.0) 8 >32
 
Minocycline-SPR741                            
    tet gene negative (103) 4 (3.9) 10 (13.6) 15 (28.2) 33 (60.2) 29 (88.4) 9 (97.1) 3 (100.0)           0.25 1
        E. coli (17) 3 (17.7) 9 (70.6) 4 (94.1) 0 (94.1) 0 (94.1) 1 (100.0)             0.06 0.12
        K. pneumoniae (86) 1 (1.2) 1 (2.3) 11 (15.1) 33 (53.5) 29 (87.2) 8 (96.5) 3 (100.0)           0.25 1
    tet gene positive (164) 1 (0.6) 2 (1.8) 5 (4.9) 31 (23.8) 36 (45.3) 33 (65.9) 17 (76.2) 21 (89.0) 12 (96.3) 3 (98.2) 2 (99.4) 1 (100.0) 1 8
a
Abbreviation: MLS, macrolide-lincosamide-streptogramin.
b
In each combination, SPR741 was used at a fixed concentration of 8 mg/liter.
c
Includes 125 isolates carrying phosphorylase-encoding mph(A) alone, 12 isolates carrying genes encoding posttranslational target site modifications [RNA methylases—erm(42) or erm(B)—alone or in combination with mph(A)], 12 isolates carrying genes encoding ABC-F ribosomal protection [msr(E) in combination with mph(A) and/or mph(E), except for 1 isolate that carried ere(A), mph(E), and msr(E)], and 3 esterase-producing [ere(A) or ere(B)] isolates.
d
Includes isolates producing macrolide-inactivating enzymes: 3 isolates produced esterases [ere(A) or ere(B)] and had MIC values of 0.12 or 0.25 mg/liter, whereas the remaining 125 isolates carried phosphorylase-encoding mph(A).
e
Includes isolates carrying genes that encode ribosomal alterations or ribosomal protection. Twelve isolates carried genes encoding posttranslational target site modifications [RNA methylases—erm(42) or erm(B)—alone or in combination with mph(A)], and 12 isolates carried genes encoding ABC-F ribosomal protection [msr(E) in combination with mph(A) and/or mph(E), except for 1 isolate that carried ere(A), mph(E), and msr(E)].
f
Includes isolates carrying tetracycline efflux pump genes [tet(A), tet(B), tet(D), or tet(G)], except for two isolates carrying genes encoding ribosomal protection [tet(M)] in combination with tet(A) and tet(B) (16).
Doxycycline (MIC50/90, 16/64 mg/liter) and minocycline (MIC50/90, 8/>32 mg/liter) had MIC results against isolates carrying tetracycline resistance genes 4- to 8-fold higher than those obtained against isolates without tetracycline resistance genes (MIC50/90, 2/8 mg/liter for both) (Table 3). Similarly, doxycycline-SPR741 (MIC50/90, 8/32 mg/liter) and minocycline-SPR741 (MIC50/90, 1/8 mg/liter) MIC values against isolates carrying tetracycline resistance genes were 4- to 16-fold higher than those obtained against their counterparts (MIC50/90, 0.5/2 mg/liter; MIC50/90, 0.25/1 mg/liter, respectively) (Table 3). However, SPR741 decreased the doxycycline MIC values 2- to 4-fold relative to doxycycline tested alone, whereas SPR741 decreased the minocycline MIC values 8-fold (Table 3). In addition, SPR741 decreased the doxycycline (MIC50/90, 0.12/0.25 mg/liter) and minocycline (MIC50/90, 0.06/0.12 mg/liter) MIC values 8- to 128-fold relative to those drugs tested alone (MIC50/90, 1/8 and 1/16 mg/liter, respectively) against tet-negative E. coli (Table 3). Similarly, SPR741 decreased the doxycycline (MIC50/90, 1/2 mg/liter) and minocycline (MIC50/90, 0.25/1 mg/liter) MIC values 2- to 8-fold relative to values obtained for those drugs tested alone (MIC50/90, 2/8 mg/liter for both) against tet-negative K. pneumoniae (Table 3).

DISCUSSION

Antimicrobial resistance challenges the empirical and guided treatment for bacterial infections and results in the concurrent use of multiple antibiotics and prolonged therapies and hospitalizations. Accordingly, the World Health Organization assembled a list of 20 major threat species (10). A report from the Centers for Disease Control and Prevention in 2013 estimated that in the United States, at least 2 million people were affected by antibiotic-resistant infections, which resulted in at least 23,000 deaths each year (11). These numbers were later confirmed to be underestimated, and revised estimates showed that more than 2.6 million antibiotic-resistant infections and nearly 44,000 deaths occurred each year when the 2013 report was released. A 2019 CDC report indicated that 2.8 million antibiotic-resistant infections occurred in the United States each year, and more than 35,000 people died as a result (3).
The current antimicrobial resistance scenario and the progress toward personalized disease treatment, including infectious diseases (12), generated an urgent need to develop new alternative approaches to treat bacterial infections. The new, expanded-spectrum polymyxin agent SPR741, in development to be used along with drugs in the current available armamentarium, represents an alternative approach for treating Gram-negative infections, including those caused by resistant organisms. Anti-Gram-positive agents, such as azithromycin, fusidic acid, and vancomycin, as tested here, are not active against Gram-negative isolates due to poor penetration through the outer membrane and/or exclusion by efflux pumps (1315). Vancomycin is a large molecule and can only reach its target site in a Gram-negative isolate by crossing the outer membrane; however, it possesses a hydrophilic characteristic that also negates crossing. The results obtained here suggest that although SPR741 decreased the vancomycin MIC value, the cells were not permeabilized enough to reduce the MIC values around therapeutic-level concentrations, as the MIC50 values remained 8 to 16 mg/liter. Azithromycin and fusidic acid also possess large molecular structures, but demonstrate a more lipophilic characteristic than vancomycin; therefore, the lower MIC values noted for azithromycin and fusidic acid when paired with SPR741 compared to vancomycin-SPR741 may be due to the lipophilic nature of the former, which may facilitate their crossing through the damaged outer membrane.
A great number of E. coli and K. pneumoniae isolates carried MLS and/or tetracycline genes, and more granular data can be seen in Table S1 in the supplemental material. The MIC results obtained by the combinations tended to correlate with the presence of these MLS and/or tetracycline genes. Regardless of the increased permeability caused by a “permeabilizer,” it is expected that increased activity by the codrug will not be observed if the bacteria possess resistance mechanisms other than decreased permeability (e.g., target site alterations/modifications). This concept was illustrated here by observing azithromycin-SPR741 MIC values against isolates carrying genes encoding ribosomal protection or posttranslational modification higher than those seen against isolates encoding esterases or phosphorylases. However, isolates carrying ribosomal protection or posttranslational modification genes were less prevalent among the isolates tested here. Similarly, isolates resistant to polymyxins were not expected to benefit from the studied combination approach. Accordingly, isolates displaying colistin MIC values of ≥4 mg/liter had MIC50/90 results for azithromycin-SPR741, fusidic acid-SPR741, doxycycline-SPR741, and minocycline-SPR741 of 16/>32, >32/>32, 4/32, and 2/16 mg/liter, respectively, whereas the susceptible counterparts displayed MIC values of 0.5/4, 4/32, 2/32, and 0.5/2 mg/liter, respectively (Table S1).
In general, doxycycline and minocycline tested alone showed similar MIC results against these E. coli and K. pneumoniae subsets (Tables 1 and 2) and when analyzed by the presence or absence of tet genes (Table 3). Although SPR741 increased the activity of both doxycycline and minocycline against tet-negative isolates, the addition of SPR741 seemed to provide a minocycline activity greater than that observed for doxycycline. Although both molecules have similar structures, minocycline is more lipophilic than doxycycline, and this difference may favor minocycline uptake through the SPR741-damaged outer membrane (16). The presence of tet genes affected the activities of doxycycline-SPR741 and minocycline-SPR741, but the latter (4- to 8-fold) was affected to a lesser extent than doxycycline-SPR741 (16-fold). The reasons for these results are unclear, and additional investigations are needed; however, the further lipophilic nature of minocycline could also provide an advantage over doxycycline against these isolates having an outer membrane damaged by SPR741.
The results obtained here for the SPR741 combinations against E. coli isolates not carrying MLS or tet genes tended to be lower than those for K. pneumoniae, suggesting that E. coli may be more prone to permeabilization with SPR741. When overexpressed, the AcrAB-TolC system intrinsic in both species may confer resistance to a number of structurally unrelated biocides and antibiotics, including tetracyclines and glycylcyclines (1719). In addition, decreased permeability due to alterations in porins could also play a role; however, tetracyclines seem to be less affected by alterations in porin channels (20). Moreover, azithromycin-SPR741 and fusidic acid-SPR741 MIC values tended to be higher in K. pneumoniae than E. coli, and both azithromycin and fusidic acid are recognized by the AcrAB-TolC pump. Thus, similar to tetracyclines, these MIC result differences for azithromycin-SPR741 and fusidic acid-SPR741 between K. pneumoniae and E. coli may be associated with differences in AcrAB expression levels between species. These findings emphasize a greater potential for SPR741 to permeabilize the E. coli cell envelope.
This study provides a detailed in vitro evaluation of the activity of several antimicrobial agents tested in combination with a new polymyxin, SPR741, against a challenge set of E. coli and K. pneumoniae clinical isolates. In addition, a subset of this collection was screened for MLS and tetracycline resistance genes, and these additional data allowed a more granular analysis and interpretation of results. In summary, adding SPR741 increased the in vitro activity of all tested codrugs at different levels, but the resulting activity seemed to be dependent on species, polymyxin resistance, and the biochemical properties and mechanism of resistance associated with the codrugs. However, the minocycline-SPR741 combination provided the lowest MIC90 values and was somehow less affected by species, β-lactamases, or tetracycline resistance mechanisms. These results indicate that the approach evaluated here has potential for treating infections caused by E. coli and K. pneumoniae, including resistant organisms, and deserves further investigations.

MATERIALS AND METHODS

Clinical isolates and susceptibility testing.

This study used a geographically diverse set of 423 Enterobacteriaceae clinical isolates (202 of E. coli and 221 of K. pneumoniae), which were selected by the presence of β-lactamases, which included plasmid-mediated AmpCs (pAmpC), ESBLs, K. pneumoniae carbapenemases, MBLs, and OXA-48-like enzymes. These isolates were part of the SENTRY Antimicrobial Surveillance Program, and additional information, including β-lactamases detected in this set, can be found in an article by Mendes et al. (9). Isolates were tested for susceptibility by broth microdilution using Mueller-Hinton broth following the Clinical and Laboratory Standards Institute (CLSI) M07-A10 document (21). Antimicrobial agents were tested alone and in combination with SPR741 at a fixed concentration of 8 mg/liter. Additional information about the rationale for the SPR741 testing concentration can be found in reference 9. MIC interpretations were based on the CLSI guidelines (22), except for polymyxin and colistin, for which breakpoints from EUCAST were applied (23). The doxycycline and minocycline CLSI breakpoints were utilized for the respective combinations for comparison. Escherichia coli ATCC 25922 and ATCC 35218, Pseudomonas aeruginosa ATCC 27853, Staphylococcus aureus ATCC 29213, and Enterococcus faecalis were used for quality control purposes. Allowable MIC ranges were those published in the CLSI M100 document, as available. Azithromycin (>16 mg/liter [data not shown]), fusidic acid (>64 mg/liter), vancomycin (>16 mg/liter), and SPR741 (>8 mg/liter) have no direct in vitro activity against Gram-negative organisms and showed off-scale MIC results when each was tested alone (Table S1).

Molecular screening for acquired MLS and tetracycline resistance genes.

A subset of 267 isolates was subjected to genome sequencing. Total genomic DNA was used as input material for library construction. DNA libraries were prepared using the Nextera XT library construction protocol and index kit (Illumina, San Diego, CA) and sequenced on a MiSeq sequencer (Illumina). Sequencing reactions were initially performed to achieve DNA read lengths of up to 300 bp and an average genome coverage depth of approximately 30×. Illumina FASTQ-format sequencing files for each isolate were quality trimmed using Sickle v1.33 to ensure high-quality reads of sufficient length after adapter removal (>20 bp) and quality scores (>Q18). FASTQ format sequencing files for each sample set were assembled independently using the de novo assembler SPAdes 3.13.0 and subjected to an in-house-designed software pipeline (JMI Laboratories) to align assembled sequences against known resistance genes (24).

ACKNOWLEDGMENTS

We express appreciation to the following persons for significant contributions to this article: H. L. Huyhn, A. Davis, L. Deshpande, T. B. Doyle, L. Flanigan, M. Janechek, J. Oberholser, and L. N. Woosley for technical support and/or assistance with manuscript preparation.
The microbiology studies were funded by Spero Therapeutics (Cambridge, MA). JMI Laboratories also received compensation fees for services for manuscript preparation, which was also funded by Spero Therapeutics. Research reported in this publication was partially supported by BARDA, the Department of Health and Human Services Office of the Assistant Secretary for Preparedness and Response under the Cooperative Agreement no. IDSEP160030, and by an award from the Wellcome Trust as administered by CARB-X. The U.S. Army Medical Research Acquisition Activity, Fort Detrick, MD, is the awarding and administering acquisition office. This work was supported by the Office of the Assistant Secretary of Defense for Health Affairs, through the Peer Reviewed Medical Research Program under award no. W81XWH-16-2-0019.
JMI Laboratories contracted to perform services in 2019 for Achaogen, Inc., Albany College of Pharmacy and Health Sciences, Allecra Therapeutics, Allergan, AmpliPhi Biosciences Corp., Amicrobe Advanced Biomaterials, Amplyx, Antabio, American Proficiency Institute, Arietis Corp., Arixa Pharmaceuticals, Inc., Astellas Pharma, Inc., Athelas, Basilea Pharmaceutica, Ltd., Bayer AG, Becton, Dickinson and Company, bioMérieux SA, Boston Pharmaceuticals, Bugworks Research, Inc., CEM-102 Pharmaceuticals, Cepheid, Cidara Therapeutics, Inc., CorMedix, Inc., DePuy Synthes, Destiny Pharma, Discuva, Ltd., Dr. Falk Pharma GmbH, Emery Pharma, Entasis Therapeutics, Eurofarma Laboratorios SA, U.S. Food and Drug Administration, Fox Chase Chemical Diversity Center, Inc., Gateway Pharmaceutical, LLC, GenePOC, Inc., Geom Therapeutics, Inc., GlaxoSmithKline, plc, Harvard University, Helperby, HiMedia Laboratories, F. Hoffmann-La Roche, Ltd., ICON, plc, Idorsia Pharmaceuticals, Ltd., Iterum Therapeutics plc, Laboratory Specialists, Inc., Melinta Therapeutics, Inc., Merck & Co., Inc., Microchem Laboratory, Micromyx, MicuRx Pharmaceuticals, Inc., Mutabilis Co., Nabriva Therapeutics, plc, NAEJA-RGM, Novartis AG, Oxoid, Ltd., Paratek Pharmaceuticals, Inc., Pfizer, Inc., Polyphor, Ltd., Pharmaceutical Product Development, LLC, Prokaryotics, Inc., Qpex Biopharma, Inc., Roivant Sciences, Ltd., Safeguard Biosystems, Scynexis, Inc., SeLux Diagnostics, Inc., Shionogi and Co., Ltd., SinSa Labs, Spero Therapeutics, Summit Pharmaceuticals International Corp., Synlogic, T2 Biosystems, Inc., Taisho Pharmaceutical Co., Ltd., TenNor Therapeutics, Ltd., Tetraphase Pharmaceuticals, Theravance Biopharma, University of Colorado, University of Southern California—San Diego, University of North Texas Health Science Center, VenatoRx Pharmaceuticals, Inc., Viosera Therapeutics, Vyome Therapeutics Inc., Wockhardt, Yukon Pharmaceuticals, Inc., Zai Lab, and Zavante Therapeutics, Inc. There are no speakers’ bureaus or stock options to declare. Troy Lister, Nicole Cotroneo, and Thomas R. Parr are employees of Spero Therapeutics, Inc., and may hold stock shares or stock options.
Opinions, interpretations, conclusions, and recommendations are those of the authors and are not necessarily endorsed by the Department of Defense. The contents of the article are solely the responsibility of the authors and do not necessarily represent the official views of CARB-X, the HHS Office of the Assistant Secretary for Preparedness and Response, the National Institutes of Health, or the Wellcome Trust.

Supplemental Material

File (aac.00742-20-s0001.xlsx)
ASM does not own the copyrights to Supplemental Material that may be linked to, or accessed through, an article. The authors have granted ASM a non-exclusive, world-wide license to publish the Supplemental Material files. Please contact the corresponding author directly for reuse.

REFERENCES

1.
Magill SS, Edwards JR, Bamberg W, Beldavs ZG, Dumyati G, Kainer MA, Lynfield R, Maloney M, McAllister-Hollod L, Nadle J, Ray SM, Thompson DL, Wilson LE, Fridkin SK, Emerging Infections Program Healthcare-Associated Infections and Antimicrobial Use Prevalence Survey Team. 2014. Multistate point-prevalence survey of health care-associated infections. N Engl J Med 370:1198–1208.
2.
Magill SS, Hellinger W, Cohen J, Kay R, Bailey C, Boland B, Carey D, de Guzman J, Dominguez K, Edwards J, Goraczewski L, Horan T, Miller M, Phelps M, Saltford R, Seibert J, Smith B, Starling P, Viergutz B, Walsh K, Rathore M, Guzman N, Fridkin S. 2012. Prevalence of healthcare-associated infections in acute care hospitals in Jacksonville, Florida. Infect Control Hosp Epidemiol 33:283–291.
3.
CDC. 2019. Antibiotic resistance threats in the United States. https://www.cdc.gov/drugresistance/pdf/threats-report/2019-ar-threats-report-508.pdf. Accessed November 2019.
4.
Doi Y, Park YS, Rivera JI, Adams-Haduch JM, Hingwe A, Sordillo EM, Lewis JS, Howard WJ, Johnson LE, Polsky B, Jorgensen JH, Richter SS, Shutt KA, Paterson DL. 2013. Community-associated extended-spectrum beta-lactamase-producing Escherichia coli infection in the United States. Clin Infect Dis 56:641–648.
5.
Patel G, Bonomo RA. 2013. “Stormy waters ahead”: global emergence of carbapenemases. Front Microbiol 4:48.
6.
Codjoe FS, Donkor ES. 2017. Carbapenem resistance: a review. Med Sci (Basel) 6:1.
7.
Meletis G. 2016. Carbapenem resistance: overview of the problem and future perspectives. Ther Adv Infect Dis 3:15–21.
8.
Vaara M. 2013. Novel derivatives of polymyxins. J Antimicrob Chemother 68:1213–1219.
9.
Mendes RE, Rhomberg PR, Lister T, Cotroneo N, Rubio A, Flamm RK. 2020. Evaluation of antimicrobial effects of a new polymyxin molecule (SPR741) when tested in combination with a series of beta-lactam agents against a challenge set of Gram-negative pathogens. Microb Drug Resist 26:319–328.
10.
Tacconelli E, Carrara E, Savoldi A, Harbarth S, Mendelson M, Monnet DL, Pulcini C, Kahlmeter G, Kluytmans J, Carmeli Y, Ouellette M, Outterson K, Patel J, Cavaleri M, Cox EM, Houchens CR, Grayson ML, Hansen P, Singh N, Theuretzbacher U, Magrini N, WHO Pathogens Priority List Working Group. 2018. Discovery, research, and development of new antibiotics: the WHO priority list of antibiotic-resistant bacteria and tuberculosis. Lancet Infect Dis 18:318–327.
11.
CDC. 2013. Antibiotic resistance threats in the United States, 2013. http://www.cdc.gov/drugresistance/pdf/ar-threats-2013-508.pdf. Accessed May 2019.
12.
de la Fuente-Nunez C, Torres MD, Mojica FJ, Lu TK. 2017. Next-generation precision antimicrobials: towards personalized treatment of infectious diseases. Curr Opin Microbiol 37:95–102.
13.
Uppu D, Konai MM, Sarkar P, Samaddar S, Fensterseifer ICM, Farias-Junior C, Krishnamoorthy P, Shome BR, Franco OL, Haldar J. 2017. Membrane-active macromolecules kill antibiotic-tolerant bacteria and potentiate antibiotics towards Gram-negative bacteria. PLoS One 12:e0183263.
14.
Li XZ, Plesiat P, Nikaido H. 2015. The challenge of efflux-mediated antibiotic resistance in Gram-negative bacteria. Clin Microbiol Rev 28:337–418.
15.
Chollet R, Chevalier J, Bryskier A, Pages JM. 2004. The AcrAB-TolC pump is involved in macrolide resistance but not in telithromycin efflux in Enterobacter aerogenes and Escherichia coli. Antimicrob Agents Chemother 48:3621–3624.
16.
Chopra I, Roberts M. 2001. Tetracycline antibiotics: mode of action, applications, molecular biology, and epidemiology of bacterial resistance. Microbiol Mol Biol Rev 65:232–260.
17.
Chopra I. 2002. New developments in tetracycline antibiotics: glycylcyclines and tetracycline efflux pump inhibitors. Drug Resist Updat 5:119–125.
18.
Iyer R, Moussa SH, Tommasi R, Miller AA. 2019. Role of the Klebsiella pneumoniae TolC porin in antibiotic efflux. Res Microbiol 170:112–116.
19.
Keeney D, Ruzin A, McAleese F, Murphy E, Bradford PA. 2008. MarA-mediated overexpression of the AcrAB efflux pump results in decreased susceptibility to tigecycline in Escherichia coli. J Antimicrob Chemother 61:46–53.
20.
Mortimer PGS, Piddok LJV. 1993. The accumulation of five antibacterial agents in porin-deficient mutants of Escherichia coli. J Antimicrob Chemother 32:195–213.
21.
CLSI. 2018. M07Ed11. Methods for dilution antimicrobial susceptibility tests for bacteria that grow aerobically, eleventh edition. Clinical and Laboratory Standards Institute, Wayne, PA.
22.
CLSI. 2018. M100Ed28. Performance standards for antimicrobial susceptibility testing: 28th informational supplement. Clinical and Laboratory Standards Institute, Wayne, PA.
23.
EUCAST. 2018. Breakpoint tables for interpretation of MIC's and zone diameters. Version 8.1, May 2018. http://www.eucast.org/fileadmin/src/media/PDFs/EUCAST_files/Breakpoint_tables/v_8.0_Breakpoint_Tables.pdf. Accessed January 2018.
24.
Mendes RE, Jones RN, Woosley LN, Cattoir V, Castanheira M. 2019. Application of next-generation sequencing for characterization of surveillance and clinical trial isolates: analysis of the distribution of beta-lactamase resistance genes and lineage background in the United States. Open Forum Infect Dis 6:S69–S78.
25.
CLSI. 2019. M100Ed29. Performance standards for antimicrobial susceptibility testing: 29th informational supplement. Clinical and Laboratory Standards Institute, Wayne, PA.
26.
EUCAST. 2019. Breakpoint tables for interpretation of MICs and zone diameters. Version 9.0, January 2019. http://www.eucast.org/fileadmin/src/media/PDFs/EUCAST_files/Breakpoint_tables/v_9.0_Breakpoint_Tables.pdf. Accessed January 2019.

Information & Contributors

Information

Published In

cover image Antimicrobial Agents and Chemotherapy
Antimicrobial Agents and Chemotherapy
Volume 65Number 116 December 2020
eLocator: 10.1128/aac.00742-20

History

Received: 16 April 2020
Returned for modification: 16 May 2020
Accepted: 9 October 2020
Published online: 16 December 2020

Permissions

Request permissions for this article.

Keywords

  1. minocycline
  2. potentiator
  3. resistance
  4. tetracyclines

Contributors

Authors

JMI Laboratories, North Liberty, Iowa, USA
JMI Laboratories, North Liberty, Iowa, USA
Troy Lister
Spero Therapeutics, Cambridge, Massachusetts, USA
Spero Therapeutics, Cambridge, Massachusetts, USA
Thomas R. Parr
Spero Therapeutics, Cambridge, Massachusetts, USA
JMI Laboratories, North Liberty, Iowa, USA

Notes

Address correspondence to Rodrigo E. Mendes, [email protected].

Metrics & Citations

Metrics

Note:

  • For recently published articles, the TOTAL download count will appear as zero until a new month starts.
  • There is a 3- to 4-day delay in article usage, so article usage will not appear immediately after publication.
  • Citation counts come from the Crossref Cited by service.

Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. For an editable text file, please select Medlars format which will download as a .txt file. Simply select your manager software from the list below and click Download.

View Options

Figures and Media

Figures

Media

Tables

Share

Share

Share the article link

Share with email

Email a colleague

Share on social media

American Society for Microbiology ("ASM") is committed to maintaining your confidence and trust with respect to the information we collect from you on websites owned and operated by ASM ("ASM Web Sites") and other sources. This Privacy Policy sets forth the information we collect about you, how we use this information and the choices you have about how we use such information.
FIND OUT MORE about the privacy policy