ABSTRACT
Lung infections caused by Mycobacterium abscessus are emerging as a global threat to individuals with cystic fibrosis and to other patient groups. Recent evidence for human-to-human transmission worsens the situation. M. abscessus is an intrinsically multidrug-resistant pathogen showing resistance to even standard antituberculosis drugs, such as rifampin. Here, our objective was to identify existing drugs that may be employed for the treatment of M. abscessus lung disease. A collection of more than 2,700 approved drugs was screened at a single-point concentration against an M. abscessus clinical isolate. Hits were confirmed with fresh solids in dose-response experiments. For the most attractive hit, growth inhibition and bactericidal activities against reference strains of the three M. abscessus subspecies and a collection of clinical isolates were determined. Surprisingly, the rifampin derivative rifabutin had MICs of 3 ± 2 μM (3 μg/ml) against the screening strain, the reference strains M. abscessus subsp. abscessus ATCC 19977, M. abscessus subsp. bolletii CCUG 50184-T, and M. abscessus subsp. massiliense CCUG 48898-T, as well as against a collection of clinical isolates. Furthermore, rifabutin was active against clarithromycin-resistant strains. In conclusion, rifabutin, in contrast to rifampin, is active against the Mycobacterium abscessus complex bacteria in vitro and may be considered for treatment of M. abscessus lung disease.
KEYWORDS: Mycobacterium abscessus, NTM, rifabutin, repurposing
INTRODUCTION
Infections caused by nontuberculous mycobacteria (NTM) are being recognized as real and rising threats (1–3). Mycobacterium abscessus is the most frequently encountered rapid-growing NTM implicated in human disease (2, 4, 5). The bacterium most commonly causes pulmonary infections in patients with immune deficiencies or with a predisposed lung condition, such as cystic fibrosis or bronchiectasis (1, 6–8). Until recently, it was believed that M. abscessus infections were exclusively acquired by exposure to contaminated soil or water. However, a study by Bryant et al. demonstrated human-to-human transmission in cystic fibrosis patients (5, 9).
The taxonomy of M. abscessus has been highly dynamic (10–12). Recent large-scale genome analyses demonstrated separation of M. abscessus into three divergent subspecies—M. abscessus subsp. abscessus, M. abscessus subsp. bolletii, and M. abscessus subsp. massiliense (9, 12, 13)—challenging an earlier classification of M. abscessus into only two subspecies (10).
M. abscessus infection is notoriously difficult to treat, as the bacterium shows a high level of intrinsic drug resistance to many antibiotics, including those used for treatment of Mycobacterium tuberculosis infections, such as rifampin (14, 15). Standard of care calls for 12 months of negative sputum cultures while on therapy (1, 16), which can result in several years of treatment with a minimum of three antibiotics. Because of the poorly performing chemotherapy, surgical resection of M. abscessus lung lesions is recommended when possible (1, 5). Macrolides, specifically clarithromycin, are part of multidrug regimens which include parenteral drugs, such as amikacin, as well as the beta-lactams imipenem and cefoxitin (1, 2, 7, 16). The already poorly performing treatments for M. abscessus infections are further complicated by the widespread occurrence of strains displaying inducible clarithromycin resistance (17, 18). Inducible clarithromycin resistance is mediated by the erm(41) gene, which encodes a methyl transferase that modifies the ribosomal binding site of clarithromycin (19–21). Although erm(41) sequences appear to be present in all M. abscessus subspecies, inducible macrolide resistance appears to occur mainly in M. abscessus subsp. abscessus and M. abscessus subsp. bolletii but not in M. abscessus subsp. massiliense, which carries deletions in the coding sequence of the gene which render the enzyme nonfunctional (20, 21). Not all M. abscessus subsp. abscessus strains harbor inducible clarithromycin resistance. A subset of clinical isolates of this subspecies carrying a polymorphism within its erm(41) coding sequence does not express inducible clarithromycin resistance (21, 22).
Despite an urgent medical need to discover new antimycobacterials or repurpose existing drugs, there is a distinct lack of activity and progress in drug discovery for treating NTM infections (1, 11, 23–25). Here, we asked whether the repertoire of existing drugs and antibiotics may contain overlooked medicines showing anti-M. abscessus activity for rapid bench-to-bedside translation. Repurposing of drugs has proven effective in finding potential candidates for treatment of bacterial infections caused by methicillin-resistant Staphylococcus aureus, Acinetobacter baumannii, Pseudomonas aeruginosa, M. tuberculosis, and others (26–29). There is precedent for treatment of M. abscessus infection where the authors identified the nitroimidazole metronidazole as a potent antibiotic against M. abscessus (14). However, the result could not be reproduced (30).
We screened 2,720 approved drugs against a clinical isolate of M. abscessus. Surprisingly, we found that rifabutin, a derivative of the poorly active rifampin, was a potent growth inhibitor of the screening strain. In vitro activities of rifabutin against reference strains and clinical isolates were characterized.
RESULTS
Screening of approved drugs identifies rifabutin as a potent inhibitor of M. abscessus Bamboo.
To identify existing drugs that may be repurposed for the treatment of M. abscessus infections, we screened a collection of 2,720 approved drugs at 20 μM for their growth inhibition potential against the clinical isolate M. abscessus Bamboo. The screen revealed 31 primary antibiotic hits (1.1% hit rate) when a cutoff of 80% growth inhibition was applied (Fig. 1). Solids of primary hits were repurchased and tested in dose-response experiments, resulting in 17 confirmed hits (Table 1).
FIG 1.
Primary screen compound–growth inhibition scatter plot. A total of 2,720 drugs at a concentration of 20 μM were screened for growth inhibition against M. abscessus Bamboo. Test drugs are shown in blue, drug-free control is in green, and clarithromycin (the positive drug control) is shown in red. The red line indicates our cutoff point of 80% growth inhibition.
TABLE 1.
Confirmed antibiotic hits active against M. abscessus Bamboo screening strain
| Antibiotic class | Antibiotic | MIC90 (μM) |
|---|---|---|
| Macrolide | Azithromycin | 6 |
| Clarithromycin | 0.4 | |
| Erythromycin | 34 | |
| Aminoglycoside | Amikacin | 14 |
| Gentamicin | 9 | |
| Kanamycin | 11 | |
| Fluoroquinolone | Ciprofloxacin | 8 |
| Gatifloxacin | 5 | |
| Levofloxacin | 18 | |
| Moxifloxacin | 4 | |
| Oxazolidinone | Linezolid | 36 |
| Glycylcycline | Tigecycline | 9 |
| Ketolide | Telithromycin | 4 |
| Glycopeptide | Vancomycin | 12 |
| Teicoplanin | 17 | |
| Ramoplanin | 24 | |
| Rifamycin | Rifabutin | 3 |
Based on literature data, expected hits included the macrolides, aminoglycosides, and fluoroquinolones (15, 31, 32), as well as newer drugs, such as the oxazolidinone linezolid, the glycylcycline tigecycline, and the ketolide telithromycin (1). Novel hits which have, to our knowledge, not been reported to be used for treatment of M. abscessus infection were the glycopeptides vancomycin, teicoplanin, and ramoplanin. However, the potencies of these glycopeptides were only modest (MIC90s, 12, 17, and 24 μM, respectively).
Unexpectedly, we found that the rifamycin rifabutin had an MIC90 of 3 μM against the primary screening strain (Table 1). This was surprising because the structurally similar tuberculosis drug rifampin (Fig. 2) is reported to be only poorly active against M. abscessus (33) and therefore is not used in clinical practice (34). Rifabutin is successfully used for treatment of lung infections caused by M. tuberculosis, where it shows attractive pharmacokinetic properties (see Discussion). As M. abscessus causes lung infections with pathologies similar to its slow-growing relative, rifabutin may be a candidate for repurposing. Therefore, we characterized the activity of rifabutin against M. abscessus in more detail.
FIG 2.
Structures of tested rifamycins.
Rifabutin is active against reference strains representing all three subspecies of the M. abscessus complex.
To determine whether rifabutin shows similar attractive potencies across the three subspecies of the M. abscessus complex, we measured its MICs against the reference strains M. abscessus subsp. abscessus ATCC 19977, M. abscessus subsp. bolletii CCUG 50184-T, and M. abscessus subsp. massiliense CCUG 48898-T. Rifabutin showed low MIC90s against all three subspecies (Table 2). These results suggest that rifabutin is active across the phylogenetically divergent M. abscessus complex. Table 2 also shows that rifampin and rifapentine displayed poor potency against the reference strains, as expected (7, 15, 35). The approximately 10-fold difference in potency of the three rifamycins is intriguing, considering the minor structural differences between the three molecules (Fig. 2).
TABLE 2.
Potencies of rifabutin against reference strains representing subspecies of the M. abscessus complex compared to those of clarithromycin and other rifamycinsa
| Strain (erm[41] sequevar)b | MIC90 (μM)c |
MIC90 (μM) after RFB pretreatmentd | MBC90 (μM) |
||||
|---|---|---|---|---|---|---|---|
| RFB | CLR | RIF | RFP | RFB | RFB | CLR | |
| M. abscessus subsp. abscessus ATCC 19977 (T28) | 3 | 3 | 37 | 31 | 4 | 6 | 12.5 |
| M. abscessus subsp. bolletii CCUG 50184-T (T28) | 4 | 5 | >50 | 31 | 4 | 6 | 25 |
| M. abscessus subsp. massiliense CCUG 48898-T (deletion) | 1 | 0.4 | 39 | 13 | 0.7 | 6 | >50 |
In Middlebrook 7H9 broth. The experiments were repeated independently two times and mean values are shown. Standard deviations were ± 50% of the shown values. The rifabutin results shown are from drug purchased from Sigma-Aldrich. The MIC experiments were repeated with drug purchased from a different source, Adooq BioScience, and delivered identical results.
Clarithromycin resistance gene erm(41) sequevars: T28 indicates inducible clarithromycin resistance; deletion of erm(41) indicates a nonfunctional gene and thus a clarithromycin-sensitive strain.
RFB, rifabutin; CLR, clarithromycin; RIF, rifampin; RFP, rifapentine.
After RFB pretreatment, prior to MIC90 determination, cultures were exposed to a subinhibitory concentration of rifabutin to identify possible inducible rifabutin resistance (see Materials and Methods for details).
Taken together, these results show that rifabutin retains its activity across the phylogenetically divergent M. abscessus complex and confirm that the drug is more potent than rifampin and rifapentine.
Exposure of M. abscessus to subinhibitory concentrations of rifabutin does not trigger inducible drug resistance.
To determine whether M. abscessus subspecies may harbor any (unknown) inducible rifabutin resistance mechanisms, we pretreated cultures of the three subspecies reference strains with subinhibitory concentrations of rifabutin and measured the impact of antibiotic preexposure on their MIC90s. Pretreatment of bacteria with the drug did not affect the MIC90s, suggesting that M. abscessus does not harbor inducible rifabutin resistance mechanisms (Table 2).
Rifabutin is bactericidal for all three subspecies of M. abscessus.
To determine whether rifabutin shows bactericidal activity against M. abscessus, cultures were treated with the drug, and the effect on viability was determined by CFU enumeration. The minimum bactericidal concentration (MBC90), or concentration that kills 90% of the bacteria, was approximately 2-fold the MIC90, i.e., rifabutin displayed similar or higher bactericidal activities compared to those of clarithromycin (Table 2).
Rifabutin shows potent activity against clinical isolates of the M. abscessus complex.
Rifabutin showed potent growth inhibition activity against the screening strain, as well as against reference strains representing the three subspecies of M. abscessus. This suggests that most clinical M. abscessus strains may be susceptible to this rifamycin. To provide evidence for widespread susceptibility of M. abscessus to rifabutin, we tested its activity against a collection of clinical isolates covering the various subspecies of the M. abscessus complex, including clarithromycin-resistant and clarithromycin-sensitive strains. All isolates were uniformly susceptible to rifabutin, with MIC90 values ranging from 3 to 5 μM (Table 3). This result indicates that a large fraction of disease-causing M. abscessus strains may be susceptible to rifabutin.
TABLE 3.
Inhibitory potency of rifabutin against clinical isolates of M. abscessusa
| Isolate code | M. abscessus subspeciesb | erm41 sequevarc | Clarithromycin susceptibility | Rifabutin MIC90 (μM) |
|---|---|---|---|---|
| M9 | abscessus | T28 | Resistant | 5 |
| M199 | abscessus | T28 | Resistant | 5 |
| M337 | abscessus | T28 | Resistant | 3 |
| M404 | abscessus | C28 | Sensitive | 4 |
| M421 | abscessus | T28 | Resistant | 3 |
| M422 | abscessus | T28 | Resistant | 3 |
| M232 | bolletii | T28 | Resistant | 3 |
| M506 | bolletii | C28 | Sensitive | 3 |
| M111 | massiliense | deletion | Sensitive | 4 |
The experiments were repeated two times independently, and mean values from those experiments are shown. Standard deviations were ±50% of the values shown.
Subspecies were determined by sequencing rpoB and hsp65.
The erm(41) sequevar was determined by sequencing the gene. For all strains, rrl (23S rRNA) was sequenced and found to be wild type (see Materials and Methods for details).
Rifamycin activities shift in Mueller-Hinton medium.
All activity determinations so far were carried out in standard Middlebrook-based mycobacterial growth medium typically used in antimycobacterial drug discovery. Drug susceptibility testing for M. abscessus is carried out mostly in Mueller-Hinton medium (36, 37). To determine whether activities of rifamycins differ in the two broth types, we measured the MICs of the three rifamycins against the type strains representing the three subspecies of M. abscessus and grown in cation-adjusted Mueller-Hinton medium. Table 4 shows that rifabutin MICs showed a 2- to 3-fold increase. Similar shifts were observed for the other rifamycins. These results show that the activities of rifamycins are reduced in Mueller-Hinton medium. The relevance of this discrepancy remains to be determined (see Discussion).
TABLE 4.
Potencies of rifamycins and clarithromycin against reference strains representing subspecies of the M. abscessus complex in Middlebrook 7H9 broth versus cation-adjusted Mueller Hinton brotha
| Strain (erm[41] sequevar)b | MIC90 in 7H9 (μM)c |
MIC90 in CAMH (μM) |
||||||
|---|---|---|---|---|---|---|---|---|
| RFB | CLR | RIF | RFP | RFB | CLR | RIF | RFP | |
| M. abscessus subsp. abscessus ATCC 19977 (T28) | 3 | 3 | 37 | 31 | 6 | 0.7 | 200 | 84 |
| M. abscessus subsp. bolletii CCUG 50184-T (T28) | 4 | 5 | >50 | 31 | 9 | 1 | >200 | 100 |
| M. abscessus subsp. massiliense CCUG 48898-T (deletion) | 1 | 0.4 | 39 | 13 | 3 | 0.3 | >200 | 50 |
The experiments were repeated independently two times, and mean values from those experiments are shown. Standard deviations were ±50% of the values shown. The rifabutin results shown are from drug purchased from Sigma-Aldrich. The MIC experiments were repeated with drug purchased from a different source, Adooq BioScience, and delivered identical results. 7H9, Middlebrook 7H9; CAMH, cation-adjusted Mueller Hinton broth.
Clarithromycin resistance gene erm(41) sequevars: T28 indicates inducible clarithromycin resistance; deletion of erm(41) indicates a nonfunctional gene and thus a clarithromycin-sensitive strain.
RFB, rifabutin; CLR, clarithromycin; RIF, rifampin; RFP, rifapentine.
DISCUSSION
Due to its poor in vitro activity, the antituberculosis drug rifampin is not used in clinical practice for the treatment of lung disease caused by M. abscessus. Here, we confirm the poor activity of rifampin and report the surprising finding that its close derivative rifabutin shows attractive activity against reference strains representing the three subspecies of the M. abscessus complex and against a collection of clinical isolates. Rifabutin showed activity against widely spread clarithromycin-resistant strains, and the bactericidal activity of the drug against the three subspecies was comparable to or better than that of clarithromycin. These results suggest that rifabutin may be considered for treatment of M. abscessus infections, including lung disease. Rifabutin is orally bioavailable and is used successfully in the treatment of tuberculosis lung disease, which causes similar pathologies, defined for the most part as nodular or cavitary diseases (38). A rifamycin in general and rifabutin in particular would be a welcome addition to anti-M. abscessus drug regimens for several reasons: (i) rifamycins are active against M. tuberculosis persisters (39), and this is likely to apply to M. abscessus as well; (ii) rifabutin has a long half-life, it exhibits high intracellular penetration and a high volume of distribution, and lung/plasma concentration ratios measured in resected human lung tissue were around 6 to 7, indicating that adequate concentrations are reached at the site of infection, since steady-state plasma levels in patients receiving the standard dose of 300 mg peak around 600 to 700 ng/ml (40, 41); (iii) rifabutin is less prone to drug-drug interactions than other rifamycins due to its reduced induction of CYP3A4 (42, 43); and (iv) rifabutin is well tolerated by a large proportion of patients who develop rifampin-related adverse events (44). Despite all these potentially positive aspects, it needs to be noted that the MIC values of rifabutin of about 3 μM against strains of the M. abscessus complex are higher than the MIC values of the drug against M. tuberculosis (45, 46) and M. avium (47), i.e., the therapeutic value of this rifamycin for treating M. abscessus disease remains to be determined.
An intriguing result is the difference in activities of the three structurally related RNA polymerase inhibitors, rifabutin, rifampin, and rifapentine (Fig. 2). We are investigating whether the difference in their antibacterial activities is due to differences in intrabacterial pharmacokinetic properties of the rifamycin derivatives, i.e., differences in bacterial drug uptake, efflux, or metabolism (45, 48, 49). Understanding the mechanistic basis of the activity differences may open new avenues to inform medicinal chemistry efforts and discover more potent rifamycins for the treatment of mycobacterial infections. Interestingly, Rominski and colleagues (50) recently showed via elegant genetic studies, including heterologous expression and gene knockout studies, that MAB_0591, encoding a putative rifampin ADP-ribosyltransferase (48, 51–53), is a major contributor to the high level of intrinsic rifampin resistance in M. abscessus subsp. abscessus ATCC 19977 (50). It remains to be determined whether rifabutin is less metabolized by this or other putative rifampin-metabolizing enzymes, such as FAD monooxygenases (51, 54, 55), or whether the differential antibacterial activity of the rifamycins against M. abscessus is due to differences in uptake or efflux.
Why has the activity of rifabutin against M. abscessus been “overlooked” so far? A search of the literature gave few results (56). A study where 31 antimicrobials, including rifabutin, were tested against the reference strain M. abscessus subsp. abscessus ATCC 19977 revealed an MIC of 32 mg/liter for rifabutin (33), clearly higher than the value of 3 mg/liter (3 μM) found in this work. This difference may be due to the differences in assay media and conditions (Mueller-Hinton versus standard mycobacterial Middlebrook 7H9 broth) (39), as well as methods (indirect redox activity-based alamarBlue readout for growth versus direct turbidity measurement) (39). When we replaced Middlebrook 7H9 with cation-adjusted Mueller-Hinton medium in our MIC assay, leaving all other parameters (including the readout) unchanged, we observed a 2- to 3-fold shift of the Middlebrook-based MICs for all three rifamycins, confirming higher activity of rifabutin relative to those of rifampin and rifapentine and indicating a weak effect of medium on activity.
Another study by van Ingen et al. used an agar dilution method (defining MIC as the concentration that inhibits >99% of growth) to determine susceptibility of a large number of NTM species, including 82 clinical M. abscessus isolates, for a number of antibiotics, including rifabutin (47, 57). The authors deemed M. abscessus “resistant” to rifabutin based on the Clinical and Laboratory Standards Institute (CLSI) breakpoint of 2 mg/liter. However, this breakpoint was set based on data from the slow-growing Mycobacterium avium. The authors acknowledged that the selected breakpoint was not based on clinical outcome or presence of mutations in rpoB and may not hold true for rapid-growing mycobacteria (47, 57). The same study determined the median MIC of rifabutin for M. abscessus to be >5 mg/liter, similar to that of rifampin, suggesting that both drugs may have high MIC values (47, 57), and apparently contradicting our data. However, the authors used 5 mg/liter as the maximum concentration tested for both rifabutin and rifampin. Thus, due to the assay conditions chosen and concentration range and breakpoints selected, the authors may have missed the potency difference between the two drugs, which we detected in broth dilution dose-response experiments, where we covered a wider range of drug concentrations.
The current study has a few limitations. Susceptibility testing against a larger collection of clinical isolates need to be carried out. Furthermore, development of resistance needs to be studied, and in vitro and in vivo drug combination studies need to be conducted. Nevertheless, our results support further testing of rifabutin in preclinical pharmacokinetic/pharmacodynamic (PK/PD) models, including mouse models of infection, as well as the characterization of the drug's pharmacokinetics in patients infected with M. abscessus. Taken together, the current report indicates that rifabutin may be useful as an add-on in the treatment of chronic, largely incurable M. abscessus pulmonary disease.
MATERIALS AND METHODS
Compounds.
A collection of 2,662 drugs approved by the U.S. Food and Drug Administration (FDA) was provided by Vincent Smeraglia and David Kimball from Rutgers University's Office of Research and Economic Development. An in-house collection of 58 antibiotics obtained from commercial sources was included in the screen, resulting in a total of 2,720 compounds screened. The compounds from the FDA library were dissolved in 90% dimethyl sulfoxide (DMSO), and the antibiotics from the in-house library were dissolved according to the manufacturers' recommendations. For confirmation of hits identified from screening the FDA drug collection, compounds were purchased from commercial sources. Rifabutin was obtained from two independent sources, Sigma-Aldrich and Adooq BioScience, and dissolved in 90% DMSO.
Bacterial strains and culture media.
For screen and hit confirmation, Mycobacterium abscessus Bamboo was used. M. abscessus Bamboo was isolated from the sputum of a patient with amyotrophic lateral sclerosis and bronchiectasis and was provided by Wei Chang Huang, Taichung Veterans General Hospital, Taichung, Taiwan. M. abscessus Bamboo whole-genome sequencing showed that the strain belongs to M. abscessus subsp. abscessus and harbors an inactive clarithromycin-sensitive erm(41) C28 sequevar (GenBank accession no. MVDX00000000) (21, 64).
For dose response and bactericidal activity determination of subspecies of the M. abscessus complex, the reference strains for the three M. abscessus subspecies were used, Mycobacterium abscessus subsp. abscessus ATCC 19977, harboring the inducible clarithromycin resistance-conferring erm(41) T28 sequevar (51), Mycobacterium abscessus subsp. bolletii CCUG 50184-T, harboring the inducible clarithromycin resistance-conferring erm(41) T28 sequevar (58), and Mycobacterium abscessus subsp. massiliense CCUG 48898-T, harboring the nonfunctional erm(41) deletion sequevar (13). The reference strains were purchased from the American Type Culture Collection (ATCC) and the Culture Collection University of Goteborg (CCUG), respectively.
For the dose-response studies of clinical isolates covering the M. abscessus complex, strains from the clinical microbiology laboratory at the National University Hospital in Singapore were used. The subspecies for each isolate was determined by multilocus sequencing employing the rpoB and hsp65 genes (59, 60). Primer pairs 5′-GACGACATCGACCACTTCGG-3′ and 5′-GGGGTCTCGATCGGGCACAT-3′ (for rpoB) and 5′-ATCGCCAAGGAGATCGAGCT-3′ and 5′-AAGGTGCCGCGGATCTTGTT-3′ (for hsp65) were used for PCR amplification. Amplicon sequencing was performed and the gene sequences compared to those available in the GenBank database using BLASTN. Phylogenetic trees were built for each gene target using MegAlign software (DNASTAR, Madison, WI) and analyzed by bootstrap analysis with 1,000 resamplings and 111 seeds. Reference genes hsp65 and rpoB were derived from whole-genome sequences for M. abscessus subsp. bolletii strain MM1513 (GenBank accession no. CP009447.1), M. abscessus subsp. massiliense strain GO 06 (CP003699.2), and M. abscessus strain FLAC054 (CP014961.1). The erm(41) and rrl genes were analyzed to determine clarithromycin resistance. Primers for erm(41) amplification were 5′-GACCGGGGCCTTCTTCGTGAT-3′ and 5′-GACTTCCCCGCACCGATTCC-3′ (20). The rrl gene was amplified using primers 5′-GTAGCGAAATTCCTTGTCGG-3′ and 5′-TTCCCGCTTAGATGCTTTCAG-3′ (21). For erm(41), the full-length gene sequence of 673 bp was examined for T28C polymorphism and deletions. For the rrl gene, the nucleotide region spanning nucleotides 2058 to 2059 was examined. Mutations at 2058 to 2059 are known to be responsible for constitutive clarithromycin resistance (21, 61).
All liquid bacterial cultures were grown in standard mycobacterium medium, Middlebrook 7H9 broth (BD Difco) supplemented with 0.5% albumin, 0.2% glucose, 0.085% sodium chloride, 0.0003% catalase, 0.2% glycerol, and 0.05% Tween 80. Solid cultures were grown on Middlebrook 7H10 agar (BD Difco) supplemented with 0.5% albumin, 0.2% glucose, 0.085% sodium chloride, 0.5% glycerol, 0.0003% catalase, and 0.006% oleic acid.
Single-point growth inhibition screening assay.
The drug library and collection of in-house antibiotics was screened in microtiter plates as previously described (62) with minor modifications. Briefly, the screen was carried out in 96-well flat-bottom Corning Costar cell culture plates at a single-point concentration of 20 μM with a starting inoculum of an optical density at 600 nm (OD600) of 0.05 (107 CFU/ml) in a final volume of 200 μl. The culture for the starting inoculum was diluted from a preculture at mid-log phase (OD600, 0.4 to 0.6). The plates were sealed using a Breathe-Easy sealing membrane (Sigma-Aldrich), put in an airtight container with moist tissue, and incubated for 3 days at 37°C on an orbital shaker at 110 rpm. Each plate had a medium-only control and a drug-free control, as well as a positive control, clarithromycin at 20 μM. After 3 days of incubation, the cultures in the wells were manually resuspended before the OD600 was read in a TECAN Infinite Pro 200 plate reader. Compounds were defined as hits if they showed growth inhibition of 80% or more of the treated culture compared to the untreated culture. The experiment was conducted in duplicate, and the results are shown as a scatter plot, with each data point representing the mean of data from the two replicates for each compound (Fig. 1).
Growth inhibition dose-response and bactericidal assays.
MICs in dose-response assays were determined by the broth microdilution method as described previously (63), with some modifications. Briefly, 96-well plates were filled with 100 μl of 7H9 medium in each well. Two times the desired highest final concentration of compound was added to the first well in each row. A 10-point 2-fold serial dilution was carried out. An appropriate dilution of a mid-log-phase culture to an OD600 of 0.1 (final OD600 in all wells was 0.05) was carried out, and 100 μl of the bacterial culture was added to the wells. The plates were incubated at 37°C and 110 rpm on an orbital shaker for 3 days and then manually resuspended, and the OD600 was measured using the plate reader. We report MIC90s, which is the concentration that inhibits 90% of growth compared to the untreated control and corresponds to the standard “no visible growth” MIC. In one growth inhibition experiment (Table 4), cation-adjusted Mueller-Hinton broth (36, 37) was used instead of Middlebrook 7H9. All other parameters were kept constant.
For bactericidal activity determinations, cultures were grown as described for the MIC determinations with the difference that the cultures were plated on agar for CFU enumeration at the end of the experiment. After 3 days of drug exposure, wells were manually resuspended and 10 μl of the cultures from the first clear well onward were plated at different dilutions on 7H10 agar. The plates were incubated at 37°C for 4 days, and then colonies were counted. We report the MBC90, which is the concentration of drug that results in a 90% reduction in CFU/ml of the treated culture compared to the untreated control at time zero.
Rifabutin preexposure assay.
To determine whether preexposure of M. abscessus to subinhibitory concentrations of rifabutin triggers induction of any (unknown) rifabutin resistance mechanisms, rifabutin preexposure experiments were carried out. A mid-log-phase culture was diluted to an OD600 of 0.05 and treated with rifabutin at a subinhibitory concentration of 0.5 μM for M. abscessus subsp. abscessus and M. abscessus subsp. bolletii and 0.1 μM for M. abscessus subsp. massiliense, 4-fold lower than their MIC50s (concentrations that causes 50% growth inhibition). An untreated culture was set up as a control. Cultures were grown to mid-log phase overnight and then subjected to standard dose-response determination as described above.
ACKNOWLEDGMENTS
We thank Vincent Smeraglia and David Kimball, Rutgers University, for distributing the collection of FDA-approved drugs. We are grateful to Wei Chang Huang, Taichung Veterans General Hospital, Taichung, Taiwan, for the M. abscessus Bamboo strain. We thank Hayden Yeo and Joe Liu, National University of Singapore, for help with the screening of the compounds, and Michelle Yee, National University of Singapore, for sharing her unpublished M. abscessus Bamboo whole-genome sequencing data. We thank Roland Jureen and Raymond Lin, National University Hospital Singapore, for discussions on NTM and resistance.
This work was supported by the Singapore Ministry of Health's National Medical Research Council under Translational Clinical Research Flagship grant NMRC/TCR/011-NUHS/2014 and is part of the Singapore Programme of Research Investigating New Approaches to Treatment of Tuberculosis (SPRINT-TB [see www.sprinttb.org]), managed by Kristina Rutkute and led by Nick Paton.
D.B.A., V.D., and T.D. conceived the idea, developed the strategy, and wrote the manuscript. D.B.A., J.L.L., M.-L.W., and M.G. carried out the experiments. J.W.P.T. characterized the clinical isolates.
REFERENCES
- 1.Griffith DE, Aksamit T, Brown-Elliott BA, Catanzaro A, Daley C, Gordin F, Holland SM, Horsburgh R, Huitt G, Iademarco MF, Iseman M, Olivier K, Ruoss S, von Reyn CF, Wallace RJ Jr, Winthrop K, ATS Mycobacterial Diseases Subcommittee. 2007. An official ATS/IDSA statement: diagnosis, treatment, and prevention of nontuberculous mycobacterial diseases. Am J Respir Crit Care Med 175:367–416. doi: 10.1164/rccm.200604-571ST. [DOI] [PubMed] [Google Scholar]
- 2.Aksamit TR, Philley JV, Griffith DE. 2014. Nontuberculous mycobacterial (NTM) lung disease: the top ten essentials. Respir Med 108:417–425. doi: 10.1016/j.rmed.2013.09.014. [DOI] [PubMed] [Google Scholar]
- 3.Cook JL. 2010. Nontuberculous mycobacteria: opportunistic environmental pathogens for predisposed hosts. Br Med Bull 96:45–59. doi: 10.1093/bmb/ldq035. [DOI] [PubMed] [Google Scholar]
- 4.Brown-Elliott BA, Wallace RJ Jr. 2002. Clinical and taxonomic status of pathogenic nonpigmented or late-pigmenting rapidly growing mycobacteria. Clin Microbiol Rev 15:716–746. doi: 10.1128/CMR.15.4.716-746.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Medjahed H, Gaillard JL, Reyrat JM. 2010. Mycobacterium abscessus: a new player in the mycobacterial field. Trends Microbiol 18:117–123. doi: 10.1016/j.tim.2009.12.007. [DOI] [PubMed] [Google Scholar]
- 6.Qvist T, Pressler T, Hoiby N, Katzenstein TL. 2014. Shifting paradigms of nontuberculous mycobacteria in cystic fibrosis. Respir Res 15:41. doi: 10.1186/1465-9921-15-41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Brown-Elliott BA, Nash KA, Wallace RJ Jr. 2012. Antimicrobial susceptibility testing, drug resistance mechanisms, and therapy of infections with nontuberculous mycobacteria. Clin Microbiol Rev 25:545–582. doi: 10.1128/CMR.05030-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Jeong YJ, Lee KS, Koh WJ, Han J, Kim TS, Kwon OJ. 2004. Nontuberculous mycobacterial pulmonary infection in immunocompetent patients: comparison of thin-section CT and histopathologic findings. Radiology 231:880–886. doi: 10.1148/radiol.2313030833. [DOI] [PubMed] [Google Scholar]
- 9.Bryant JM, Grogono DM, Rodriguez-Rincon D, Everall I, Brown KP, Moreno P, Verma D, Hill E, Drijkoningen J, Gilligan P, Esther CR, Noone PG, Giddings O, Bell SC, Thomson R, Wainwright CE, Coulter C, Pandey S, Wood ME, Stockwell RE, Ramsay KA, Sherrard LJ, Kidd TJ, Jabbour N, Johnson GR, Knibbs LD, Morawska L, Sly PD, Jones A, Bilton D, Laurenson I, Ruddy M, Bourke S, Bowler IC, Chapman SJ, Clayton A, Cullen M, Dempsey O, Denton M, Desai M, Drew RJ, Edenborough F, Evans J, Folb J, Daniels T, Humphrey H, Isalska B, Jensen-Fangel S, Jonsson B, Jones AM, et al. 2016. Emergence and spread of a human-transmissible multidrug-resistant nontuberculous mycobacterium. Science 354:751–757. doi: 10.1126/science.aaf8156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Leao SC, Tortoli E, Euzeby JP, Garcia MJ. 2011. Proposal that Mycobacterium massiliense and Mycobacterium bolletii be united and reclassified as Mycobacterium abscessus subsp. bolletii comb, nov., designation of Mycobacterium abscessus subsp. abscessus subsp. nov. and emended description of Mycobacterium abscessus. Int J Syst Evol Microbiol 61:2311–2313. doi: 10.1099/ijs.0.023770-0. [DOI] [PubMed] [Google Scholar]
- 11.Benwill JL, Wallace RJ Jr. 2014. Mycobacterium abscessus: challenges in diagnosis and treatment. Curr Opin Infect Dis 27:506–510. doi: 10.1097/QCO.0000000000000104. [DOI] [PubMed] [Google Scholar]
- 12.Lee MR, Sheng WH, Hung CC, Yu CJ, Lee LN, Hsueh PR. 2015. Mycobacterium abscessus complex infections in humans. Emerg Infect Dis 21:1638–1646. doi: 10.3201/2109.141634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Cho YJ, Yi H, Chun J, Cho SN, Daley CL, Koh WJ, Shin SJ. 2013. The genome sequence of “Mycobacterium massiliense” strain CIP 108297 suggests the independent taxonomic status of the Mycobacterium abscessus complex at the subspecies level. PLoS One 8:e81560. doi: 10.1371/journal.pone.0081560. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Chopra S, Matsuyama K, Hutson C, Madrid P. 2011. Identification of antimicrobial activity among FDA-approved drugs for combating Mycobacterium abscessus and Mycobacterium chelonae. J Antimicrob Chemother 66:1533–1536. doi: 10.1093/jac/dkr154. [DOI] [PubMed] [Google Scholar]
- 15.Nessar R, Cambau E, Reyrat JM, Murray A, Gicquel B. 2012. Mycobacterium abscessus: a new antibiotic nightmare. J Antimicrob Chemother 67:810–818. doi: 10.1093/jac/dkr578. [DOI] [PubMed] [Google Scholar]
- 16.Floto RA, Olivier KN, Saiman L, Daley CL, Herrmann JL, Nick JA, Noone PG, Bilton D, Corris P, Gibson RL, Hempstead SE, Koetz K, Sabadosa KA, Sermet-Gaudelus I, Smyth AR, van Ingen J, Wallace RJ, Winthrop KL, Marshall BC, Haworth CS. 2016. US Cystic Fibrosis Foundation and European Cystic Fibrosis Society consensus recommendations for the management of non-tuberculous mycobacteria in individuals with cystic fibrosis. Thorax 71(Suppl):i1–i22. doi: 10.1136/thoraxjnl-2015-207360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Choi GE, Shin SJ, Won CJ, Min KN, Oh T, Hahn MY, Lee K, Lee SH, Daley CL, Kim S, Jeong BH, Jeon K, Koh WJ. 2012. Macrolide treatment for Mycobacterium abscessus and Mycobacterium massiliense infection and inducible resistance. Am J Respir Crit Care Med 186:917–925. doi: 10.1164/rccm.201111-2005OC. [DOI] [PubMed] [Google Scholar]
- 18.Maurer FP, Castelberg C, Quiblier C, Bottger EC, Somoskovi A. 2014. Erm(41)-dependent inducible resistance to azithromycin and clarithromycin in clinical isolates of Mycobacterium abscessus. J Antimicrob Chemother 69:1559–1563. doi: 10.1093/jac/dku007. [DOI] [PubMed] [Google Scholar]
- 19.Weisblum B. 1995. Erythromycin resistance by ribosome modification. Antimicrob Agents Chemother 39:577–585. doi: 10.1128/AAC.39.3.577. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Nash KA, Brown-Elliott BA, Wallace RJ Jr. 2009. A novel gene, erm(41), confers inducible macrolide resistance to clinical isolates of Mycobacterium abscessus but is absent from Mycobacterium chelonae. Antimicrob Agents Chemother 53:1367–1376. doi: 10.1128/AAC.01275-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Bastian S, Veziris N, Roux AL, Brossier F, Gaillard JL, Jarlier V, Cambau E. 2011. Assessment of clarithromycin susceptibility in strains belonging to the Mycobacterium abscessus group by erm(41) and rrl sequencing. Antimicrob Agents Chemother 55:775–781. doi: 10.1128/AAC.00861-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Brown-Elliott BA, Vasireddy S, Vasireddy R, Iakhiaeva E, Howard ST, Nash K, Parodi N, Strong A, Gee M, Smith T, Wallace RJ Jr. 2015. Utility of sequencing the erm(41) gene in isolates of Mycobacterium abscessus subsp. abscessus with low and intermediate clarithromycin MICs. J Clin Microbiol 53:1211–1215. doi: 10.1128/JCM.02950-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Soni I, De Groote MA, Dasgupta A, Chopra S. 2016. Challenges facing the drug discovery pipeline for non-tuberculous mycobacteria. J Med Microbiol 65:1–8. doi: 10.1099/jmm.0.000198. [DOI] [PubMed] [Google Scholar]
- 24.Olivier KN, Shaw PA, Glaser TS, Bhattacharyya D, Fleshner M, Brewer CC, Zalewski CK, Folio LR, Siegelman JR, Shallom S, Park IK, Sampaio EP, Zelazny AM, Holland SM, Prevots DR. 2014. Inhaled amikacin for treatment of refractory pulmonary nontuberculous mycobacterial disease. Ann Am Thorac Soc 11:30–35. doi: 10.1513/AnnalsATS.201307-231OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Ryu YJ, Koh WJ, Daley CL. 2016. Diagnosis and treatment of nontuberculous mycobacterial lung disease: clinicians' perspectives. Tuberc Respir Dis (Seoul) 79:74–84. doi: 10.4046/trd.2016.79.2.74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Chopra S, Torres-Ortiz M, Hokama L, Madrid P, Tanga M, Mortelmans K, Kodukula K, Galande AK. 2010. Repurposing FDA-approved drugs to combat drug-resistant Acinetobacter baumannii. J Antimicrob Chemother 65:2598–2601. doi: 10.1093/jac/dkq353. [DOI] [PubMed] [Google Scholar]
- 27.Carlson-Banning KM, Chou A, Liu Z, Hamill RJ, Song Y, Zechiedrich L. 2013. Toward repurposing ciclopirox as an antibiotic against drug-resistant Acinetobacter baumannii, Escherichia coli, and Klebsiella pneumoniae. PLoS One 8:e69646. doi: 10.1371/journal.pone.0069646. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Thangamani S, Younis W, Seleem MN. 2015. Repurposing clinical molecule ebselen to combat drug resistant pathogens. PLoS One 10:e0133877. doi: 10.1371/journal.pone.0133877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Palomino JC, Martin A. 2013. Is repositioning of drugs a viable alternative in the treatment of tuberculosis? J Antimicrob Chemother 68:275–283. doi: 10.1093/jac/dks405. [DOI] [PubMed] [Google Scholar]
- 30.Mukherjee T, Boshoff H, Barry CE 3rd. 2012. Comment on: Identification of antimicrobial activity among FDA-approved drugs for combating Mycobacterium abscessus and Mycobacterium chelonae. J Antimicrob Chemother 67:252–253. doi: 10.1093/jac/dkr418. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Choi GE, Min KN, Won CJ, Jeon K, Shin SJ, Koh WJ. 2012. Activities of moxifloxacin in combination with macrolides against clinical isolates of Mycobacterium abscessus and Mycobacterium massiliense. Antimicrob Agents Chemother 56:3549–3555. doi: 10.1128/AAC.00685-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Beltrame A, Cattani G, Brillo F, Merelli M, Scarparo C, Como G, Screm MC, Tortoli E, Matteell A, Bassetti M. 2013. Successful antibiotic treatment of Mycobacterium abscessus pulmonary disease in an immunocompetent individual. J Infect Dis Ther 1:103. [Google Scholar]
- 33.Pang H, Li G, Zhao X, Liu H, Wan K, Yu P. 2015. Drug susceptibility testing of 31 antimicrobial agents on rapidly growing mycobacteria isolates from China. Biomed Res Int 2015:419392. doi: 10.1155/2015/419392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Kaushik A, Makkar N, Pandey P, Parrish N, Singh U, Lamichhane G. 2015. Carbapenems and rifampin exhibit synergy against Mycobacterium tuberculosis and Mycobacterium abscessus. Antimicrob Agents Chemother 59:6561–6567. doi: 10.1128/AAC.01158-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Fernandez-Roblas R, Esteban J, Cabria F, Lopez JC, Jimenez MS, Soriano F. 2000. In vitro susceptibilities of rapidly growing mycobacteria to telithromycin (HMR 3647) and seven other antimicrobials. Antimicrob Agents Chemother 44:181–182. doi: 10.1128/AAC.44.1.181-182.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Broda A, Jebbari H, Beaton K, Mitchell S, Drobniewski F. 2013. Comparative drug resistance of Mycobacterium abscessus and M. chelonae isolates from patients with and without cystic fibrosis in the United Kingdom. J Clin Microbiol 51:217–223. doi: 10.1128/JCM.02260-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Pang H, Li G, Wan L, Jiang Y, Liu H, Zhao X, Zhao Z, Wan K. 2015. In vitro drug susceptibility of 40 international reference rapidly growing mycobacteria to 20 antimicrobial agents. Int J Clin Exp Med 8:15423–15431. [PMC free article] [PubMed] [Google Scholar]
- 38.Wallace RJ Jr, Glassroth J, Griffith DE, Olivier KN, Cook JL, Gordin F; American Thoracic Society; Medical Section of the American Lung Association. 1997. Diagnosis and treatment of disease caused by nontuberculous mycobacteria. Am J Respir Crit Care Med 156:S1–S25. doi: 10.1164/ajrccm.156.2.atsstatement. [DOI] [PubMed] [Google Scholar]
- 39.Lakshminarayana SB, Huat TB, Ho PC, Manjunatha UH, Dartois V, Dick T, Rao SP. 2015. Comprehensive physicochemical, pharmacokinetic and activity profiling of anti-TB agents. J Antimicrob Chemother 70:857–867. doi: 10.1093/jac/dku457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Blaschke TF, Skinner MH. 1996. The clinical pharmacokinetics of rifabutin. Clin Infect Dis 22(Suppl 1):S15–S21; discussion S21–S22. doi: 10.1093/clinids/22.Supplement_1.S15. [DOI] [PubMed] [Google Scholar]
- 41.Van der Auwera P, Matsumoto T, Husson M. 1988. Intraphagocytic penetration of antibiotics. J Antimicrob Chemother 22:185–192. doi: 10.1093/jac/22.2.185. [DOI] [PubMed] [Google Scholar]
- 42.Li AP, Reith MK, Rasmussen A, Gorski JC, Hall SD, Xu L, Kaminski DL, Cheng LK. 1997. Primary human hepatocytes as a tool for the evaluation of structure-activity relationship in cytochrome P450 induction potential of xenobiotics: evaluation of rifampin, rifapentine and rifabutin. Chem Biol Interact 107:17–30. doi: 10.1016/S0009-2797(97)00071-9. [DOI] [PubMed] [Google Scholar]
- 43.Baciewicz AM, Chrisman CR, Finch CK, Self TH. 2008. Update on rifampin and rifabutin drug interactions. Am J Med Sci 335:126–136. doi: 10.1097/MAJ.0b013e31814a586a. [DOI] [PubMed] [Google Scholar]
- 44.Horne DJ, Spitters C, Narita M. 2011. Experience with rifabutin replacing rifampin in the treatment of tuberculosis. Int J Tuberc Lung Dis 15:1485–1489, i. doi: 10.5588/ijtld.11.0068. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Kunin CM. 1996. Antimicrobial activity of rifabutin. Clin Infect Dis 22(Suppl 1):S3–S13; discussion S13–S14. doi: 10.1093/clinids/22.Supplement_1.S3. [DOI] [PubMed] [Google Scholar]
- 46.Barluenga J, Aznar F, Garcia AB, Cabal MP, Palacios JJ, Menendez MA. 2006. New rifabutin analogs: synthesis and biological activity against Mycobacterium tuberculosis. Bioorg Med Chem Lett 16:5717–5722. doi: 10.1016/j.bmcl.2006.08.090. [DOI] [PubMed] [Google Scholar]
- 47.van Ingen J, van der Laan T, Dekhuijzen R, Boeree M, van Soolingen D. 2010. In vitro drug susceptibility of 2275 clinical non-tuberculous Mycobacterium isolates of 49 species in The Netherlands. Int J Antimicrob Agents 35:169–173. doi: 10.1016/j.ijantimicag.2009.09.023. [DOI] [PubMed] [Google Scholar]
- 48.Quan S, Venter H, Dabbs ER. 1997. Ribosylative inactivation of rifampin by Mycobacterium smegmatis is a principal contributor to its low susceptibility to this antibiotic. Antimicrob Agents Chemother 41:2456–2460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Baysarowich J, Koteva K, Hughes DW, Ejim L, Griffiths E, Zhang K, Junop M, Wright GD. 2008. Rifamycin antibiotic resistance by ADP-ribosylation: structure and diversity of Arr. Proc Natl Acad Sci U S A 105:4886–4891. doi: 10.1073/pnas.0711939105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Rominski A, Roditscheff A, Selchow P, Bottger EC, Sander P. 2016. Intrinsic rifamycin resistance of Mycobacterium abscessus is mediated by ADP-ribosyltransferase MAB_0591. J Antimicrob Chemother 72:376–384. doi: 10.1093/jac/dkw466. [DOI] [PubMed] [Google Scholar]
- 51.Ripoll F, Pasek S, Schenowitz C, Dossat C, Barbe V, Rottman M, Macheras E, Heym B, Herrmann JL, Daffe M, Brosch R, Risler JL, Gaillard JL. 2009. Non mycobacterial virulence genes in the genome of the emerging pathogen Mycobacterium abscessus. PLoS One 4:e5660. doi: 10.1371/journal.pone.0005660. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Quan S, Imai T, Mikami Y, Yazawa K, Dabbs ER, Morisaki N, Iwasaki S, Hashimoto Y, Furihata K. 1999. ADP-ribosylation as an intermediate step in inactivation of rifampin by a mycobacterial gene. Antimicrob Agents Chemother 43:181–184. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Combrink KD, Denton DA, Harran S, Ma Z, Chapo K, Yan D, Bonventre E, Roche ED, Doyle TB, Robertson GT, Lynch AS. 2007. New C25 carbamate rifamycin derivatives are resistant to inactivation by ADP-ribosyl transferases. Bioorg Med Chem Lett 17:522–526. doi: 10.1016/j.bmcl.2006.10.016. [DOI] [PubMed] [Google Scholar]
- 54.Abdelwahab H, Martin Del Campo JS, Dai Y, Adly C, El-Sohaimy S, Sobrado P. 2016. Mechanism of rifampicin inactivation in Nocardia farcinica. PLoS One 11:e0162578. doi: 10.1371/journal.pone.0162578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Hoshino Y, Fujii S, Shinonaga H, Arai K, Saito F, Fukai T, Satoh H, Miyazaki Y, Ishikawa J. 2010. Monooxygenation of rifampicin catalyzed by the rox gene product of Nocardia farcinica: structure elucidation, gene identification and role in drug resistance. J Antibiot (Tokyo) 63:23–28. doi: 10.1038/ja.2009.116. [DOI] [PubMed] [Google Scholar]
- 56.Cremades R, Santos A, Rodriguez JC, Garcia-Pachon E, Ruiz M, Royo G. 2009. Mycobacterium abscessus from respiratory isolates: activities of drug combinations. J Infect Chemother 15:46–48. doi: 10.1007/s10156-008-0651-Y. [DOI] [PubMed] [Google Scholar]
- 57.Crabol Y, Catherinot E, Veziris N, Jullien V, Lortholary O. 2016. Rifabutin: where do we stand in 2016? J Antimicrob Chemother 71:1759–1771. doi: 10.1093/jac/dkw024. [DOI] [PubMed] [Google Scholar]
- 58.Choi GE, Cho YJ, Koh WJ, Chun J, Cho SN, Shin SJ. 2012. Draft genome sequence of Mycobacterium abscessus subsp. bolletii BD(T). J Bacteriol 194:2756–2757. doi: 10.1128/JB.00354-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Adekambi T, Berger P, Raoult D, Drancourt M. 2006. rpoB gene sequence-based characterization of emerging non-tuberculous mycobacteria with descriptions of Mycobacterium bolletii sp. nov., Mycobacterium phocaicum sp. nov. and Mycobacterium aubagnense sp. nov. Int J Syst Evol Microbiol 56:133–143. doi: 10.1099/ijs.0.63969-0. [DOI] [PubMed] [Google Scholar]
- 60.Kim H, Kim SH, Shim TS, Kim MN, Bai GH, Park YG, Lee SH, Chae GT, Cha CY, Kook YH, Kim BJ. 2005. Differentiation of Mycobacterium species by analysis of the heat-shock protein 65 gene (hsp65). Int J Syst Evol Microbiol 55:1649–1656. doi: 10.1099/ijs.0.63553-0. [DOI] [PubMed] [Google Scholar]
- 61.Wallace RJ Jr, Meier A, Brown BA, Zhang Y, Sander P, Onyi GO, Bottger EC. 1996. Genetic basis for clarithromycin resistance among isolates of Mycobacterium chelonae and Mycobacterium abscessus. Antimicrob Agents Chemother 40:1676–1681. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Moreira W, Lim JJ, Yeo SY, Ramanujulu PM, Dymock BW, Dick T. 2016. Fragment-based whole cell screen delivers hits against M. tuberculosis and non-tuberculous mycobacteria. Front Microbiol 7:1392. doi: 10.3389/fmicb.2016.01392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Moreira W, Aziz DB, Dick T. 2016. Boromycin kills mycobacterial persisters without detectable resistance. Front Microbiol 7:199. doi: 10.3389/fmicb.2016.0019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Yee M, Klinzing D, Wei J-R, Gengenbacher M, Rubin E, Dick T. Draft genome sequence of Mycobacterium abscessus Bamboo. Genome Announc, in press. [DOI] [PMC free article] [PubMed] [Google Scholar]


