Tigecycline is used in multidrug regimens for salvage therapy of Mycobacterium abscessus infections but is often poorly tolerated and has no oral formulation. Here, we report similar in vitro activity of two newly approved tetracycline analogs, omadacycline and eravacycline, against 28 drug-resistant clinical isolates of M. abscessus complex.
KEYWORDS: Mycobacterium abscessus, omadacycline, drug susceptibility assay, eravacycline, tetracyclines, tigecycline
ABSTRACT
Tigecycline is used in multidrug regimens for salvage therapy of Mycobacterium abscessus infections but is often poorly tolerated and has no oral formulation. Here, we report similar in vitro activity of two newly approved tetracycline analogs, omadacycline and eravacycline, against 28 drug-resistant clinical isolates of M. abscessus complex. Since omadacycline and eravacycline appear to be better tolerated than tigecycline and since omadacycline is also formulated for oral dosing, these tetracycline analogs may represent new treatment options for M. abscessus infections.
INTRODUCTION
Mycobacterium abscessus complex, consisting of the subspecies abscessus, massiliense, and bolletii, is a group of rapidly growing, nontuberculous mycobacteria (NTM) known for its extensive intrinsic and acquired drug resistance (1). It can cause treatment-refractory lung infections (especially among cystic fibrosis patients), as well as other serious infections (2). Increasing prevalence of pulmonary NTM infections over the last several decades has been reported from several parts of the world, including the United States and Europe (3–6). M. abscessus complex is now the most common rapid-growing NTM causing lung infection and the second most common among all NTM after Mycobacterium avium complex. It is also the most difficult-to-treat NTM lung infection (3–6). A typical multidrug treatment regimen for cystic fibrosis patients with M. abscessus infection consists of an oral macrolide, intravenous amikacin, along with one or more additional intravenous antibiotics, such as cefoxitin, imipenem, or tigecycline (7). Tigecycline (a glycylcycline of the tetracycline class) is active in vitro against most clinical isolates of M. abscessus and has been used clinically for M. abscessus lung infections with some success, but nausea and vomiting are frequent, often treatment-limiting, adverse effects (8, 9). In addition, tigecycline’s intravenous mode of administration is undesirable for a disease that is often treated for more than a year (7, 9). Therefore, new antibiotics with similar or better efficacy, fewer adverse effects, preferably with oral bioavailability, are desperately needed to improve the treatment of M. abscessus infections.
Omadacycline (an aminomethylcycline) is a new tetracycline analog, approved for the treatment of acute bacterial skin and skin-structure infections (ABSSSI) and community-acquired bacterial pneumonia (CABP). It is available in both intravenous and oral formulations (10, 11). Eravacycline (a fluorocycline) is a new tetracycline analog approved for the treatment of complicated intraabdominal infections in an intravenous formulation (12). In the present study, we evaluated the activity of omadacycline and eravacycline against a panel of drug-resistant M. abscessus complex organisms.
Omadacycline, eravacycline, and tigecycline were purchased from MedChem Express, Monmouth Junction, NJ (purity, >95%). All antimicrobials were received in powdered form, stored at −20°C, and dissolved in dimethyl sulfoxide or deionized water in accordance with the manufacturer’s recommendations. M. abscessus strain ATCC 19977 was purchased from the American Type Culture Collection (Manassas, VA) and used as a reference strain. Twenty-eight unique clinical isolates of M. abscessus complex were obtained from the Johns Hopkins Hospital Clinical Microbiology Laboratory from 2005 to 2015, as described previously (13, 14). Isolates were identifies to the subspecies level based on the length of erm(41), which is truncated in M. abscessus subsp. massiliense, and the rpoB sequence (15–17). Reference genomes for each subspecies were as follows: abscessus strain ATCC 19977 (NCBI accession NC_010397), massiliense strain GO 06 (NCBI accession NC_018150), and bolletii strain CIP 198541 (NCBI accession NZ_JRMF00000000). These isolates are resistant to nearly all drugs used to treat M. abscessus infection (amikacin, clarithromycin, imipenem, sulfamethoxazole/trimethoprim, linezolid, and moxifloxacin). The MICs were determined using the broth microdilution method in cation-adjusted Mueller-Hinton broth (CAMHB) in accordance with Clinical and Laboratory Standards Institute (CLSI) guidelines (18). In brief, CAMHB (100 μl/well) was added in each well of 96-well, U-bottom, polystyrene plates (Corning, Inc., Corning, NY). Serial 2-fold dilutions of compounds were prepared. M. abscessus strains were grown to the mid-log phase. An inoculum adjusted to 1 × 104 to 5 × 104 CFU in a 0.1-ml volume was added in each well except the medium control. Plates were sealed and incubated at 30°C for 3 days. Plates were incubated up to 5 days if the pellet size in control wells without drug was small on days 3 and 4. MICs were determined on the basis of presence or absence of pellet with unaided eyes (13). Drug susceptibility assays were repeated to confirm the MICs.
Against M. abscessus strain ATCC 19977, the MIC of omadacycline was similar to that of tigecycline (1 μg/ml), whereas the eravacycline MIC was 2-fold lower (Table 1). Likewise, omadacycline and tigecycline had the same MIC50 and MIC90 against 28 drug-resistant clinical isolates (2 μg/ml), while the MIC50 and MIC90 of eravacycline were 2-fold lower. Interestingly, while the present study was under review, a newly published study reported similar MICs for tigecycline and omadacycline against M. abscessus complex clinical isolates (19).
TABLE 1.
MICs of tigecycline, omadacycline, and eravacycline against Mycobacterium abscessus ATCC 19977 and 28 drug-resistant M. abscessus complex clinical isolates in CAMHB
| Isolate or MIC | M. abscessus subspecies | MIC (μg/ml) |
||
|---|---|---|---|---|
| Tigecycline | Omadacycline | Eravacycline | ||
| Isolates | ||||
| Strain ATCC 19977a | abscessus | 1 | 1 | 0.5 |
| 1N | abscessus | 1 | 1 | 0.5 |
| 2N | massiliense-bolletiib | 1 | 1 | 0.25 |
| 3N | abscessus | 2 | 2 | 1 |
| 4N | massiliense | 1 | 1 | 0.25 |
| 5N | massiliense | 1 | 0.5 | 0.25 |
| 6N | abscessus | 2 | 4 | 1 |
| 11N | abscessus | 1 | 2 | 2 |
| 12N | abscessus | 1 | 0.5 | 0.25 |
| 13N | massiliense-bolletii | 1 | 2 | 0.5 |
| 14N | massiliense-bolletii | 2 | 2 | 1 |
| 19N | abscessus | 1 | 0.5 | 0.25 |
| 201 | abscessus | 1 | 0.5 | 0.25 |
| 202 | abscessus | 1 | 2 | 0.5 |
| 203 | massiliense-bolletii | 1 | 2 | 0.5 |
| 204 | massiliense | 1 | 1 | 0.5 |
| 206 | massiliense | 0.5 | 0.5 | 0.125 |
| 208 | massiliense | 2 | 2 | 0.5 |
| 210 | abscessus | 2 | 2 | 0.5 |
| 211 | abscessus | 2 | 2 | 0.5 |
| 212 | massiliense-bolletii | 1 | 1 | 0.25 |
| 214 | massiliense | 1 | 1 | 0.5 |
| 215 | abscessus | 1 | 1 | 0.25 |
| 216 | massiliense | 1 | 1 | 0.25 |
| 218 | abscessus | 4 | 4 | 2 |
| JHH2 | abscessus | 1 | 1 | 0.25 |
| JHH4 | abscessus | 1 | 1 | 0.25 |
| JHH9 | abscessus | 2 | 2 | 0.5 |
| JHHKB | abscessus | 2 | 2 | 0.5 |
| MIC data | ||||
| MIC range | 0.5–4 | 0.5–4 | 0.125–2 | |
| MIC50 | 1 | 1 | 0.5 | |
| MIC90 | 2 | 2 | 1 | |
M. abscessus strain ATCC 19977 is included as a reference strain, and the MIC values for this strain were not included when determining the MIC range, MIC50, and MIC90.
Five isolates had truncated erm(41) genes, indicating subsp. massiliense, but had rpoB sequences matching subsp. bolletii.
While no formal susceptibility breakpoint has been established for tigecycline against M. abscessus, breakpoints ranging from 0.5 to 4 μg/ml have been proposed (8, 20). Clinical isolates of rapidly growing mycobacteria are susceptible to tigecycline concentrations of ≤2 μg/ml (21–24), which is the approved susceptibility breakpoint against Enterobacteriaceae (25). The MIC50 and MIC90 of omadacycline reported here are 4- and 2-fold lower, respectively, than the susceptibility breakpoint for Enterobacteriaceae (26). The MIC50 of eravacycline reported here matches its susceptibility breakpoint for Enterobacteriaceae and anaerobes (27). It is noteworthy that steady-state plasma concentrations equivalent to our observed MIC90 for drug-resistant M. abscessus clinical isolates are achievable with intravenous dosing of omadacycline and eravacycline (28–30). At an intravenous omadacycline dose of 100 mg/day (approved marketed dose for CABP and ABSSSI), the steady-state plasma Cmax and AUC0–24 are 2.12 μg/ml and 12.14 μg ⋅ h/ml, respectively, compared to 0.87 μg/ml and 4.7 μg ⋅ h/ml for tigecycline at 50 mg twice daily (29, 31, 32). Oral omadacycline doses of 300 to 450 mg produced Cmax values of 9.52 to 10.8 μg/ml and AUC0–24 values of 11.2 to 13.4 μg ⋅ h/ml, respectively (29). Eravacycline after intravenous dosing 1.0 mg/kg every 12 h produced a plasma Cmax of 1.83 μg/ml and an AUC0–24 of at least 12.6 μg ⋅ h/ml (25). Although there is no marketed oral formulation of eravacycline, a single oral dose of 100 mg produced a Cmax of 0.17 μg/ml and an AUC0–∞ of 2.25 μg ⋅ h/ml (33). In addition, omadacycline has a low protein binding (21%) compared to eravacycline (79 to 87%) and tigecycline (69 to 87%) (28).
The free drug AUC/MIC ratio was the pharmacokinetic/pharmacodynamic parameter most closely correlated with tigecycline activity in an in vitro hollow fiber model of M. abscessus infection (8). Considering the steady-state AUC and protein binding data described above and the MICs obtained in our study against M. abscessus, the free drug AUC/MIC ratios for omadacycline and eravacycline given intravenously are expected to be approximately 8 to 10 times higher and 2 times higher, respectively, compared to tigecycline. This preliminary comparison suggests that eravacycline and, especially, omadacycline could be more efficacious clinically than tigecycline. These hypotheses should be evaluated further in nonclinical models of M. abscessus infection.
Despite tigecycline’s appreciable activity as a component of multidrug regimen for M. abscessus infections, its clinical utility is limited by significant nausea and vomiting (7, 9), especially at the 200-mg daily dose identified as the optimal dose in the hollow fiber infection model (8). Omadacycline and eravacycline appear better tolerated. Omadacycline was associated with significantly less nausea and fewer treatment-emergent adverse events (TEAEs) compared to tigecycline in one study (31). Omadacycline also demonstrated similar safety and side effect profiles to linezolid (for treatment of ABSSI) and moxifloxacin (for CABP) in pivotal trials (10, 11). In IGNITE1 and IGNITE4 trials, eravacycline-treated patients experienced only slightly more TEAEs compared to ertapenem- and meropenem-treated patients (12, 34).
In conclusion, omadacycline and eravacycline may represent new options for treatment of M. abscessus complex infections. The results presented here support further investigation of their efficacy and exposure-response profiles in animal models and clinical trials to better understand their potential clinical utility.
ACKNOWLEDGMENTS
We gratefully acknowledge Gyanu Lamichhane and Maram Naji for assistance in curating and characterizing the M. abscessus clinical isolates.
Funding was provided by the National Institutes of Health (R21AI137814; E.L.N.).
REFERENCES
- 1.Griffith DE. 2019. Mycobacterium abscessus and antibiotic resistance: same as it ever was. Clin Infect Dis doi: 10.1093/cid/ciz071. [DOI] [PubMed] [Google Scholar]
- 2.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]
- 3.Prevots DR, Shaw PA, Strickland D, Jackson LA, Raebel MA, Blosky MA, Montes de Oca R, Shea YR, Seitz AE, Holland SM, Olivier KN. 2010. Nontuberculous mycobacterial lung disease prevalence at four integrated health care delivery systems. Am J Respir Crit Care Med 182:970–976. doi: 10.1164/rccm.201002-0310OC. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Ringshausen FC, Apel RM, Bange FC, de Roux A, Pletz MW, Rademacher J, Suhling H, Wagner D, Welte T. 2013. Burden and trends of hospitalizations associated with pulmonary non-tuberculous mycobacterial infections in Germany, 2005-2011. BMC Infect Dis 13:231. doi: 10.1186/1471-2334-13-231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Thomson RM, NTM Working Group at Queensland TB Control Centre and Queensland Mycobacterial Reference Laboratory. 2010. Changing epidemiology of pulmonary nontuberculous mycobacteria infections. Emerg Infect Dis 16:1576–1583. doi: 10.3201/eid1610.091201. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Adjemian J, Frankland TB, Daida YG, Honda JR, Olivier KN, Zelazny A, Honda S, Prevots DR. 2017. Epidemiology of nontuberculous mycobacterial lung disease and tuberculosis, Hawaii, USA. Emerg Infect Dis 23:439–447. doi: 10.3201/eid2303.161827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.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: executive summary. Thorax 71:88–90. doi: 10.1136/thoraxjnl-2015-207983. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Ferro BE, Srivastava S, Deshpande D, Pasipanodya JG, van Soolingen D, Mouton JW, van Ingen J, Gumbo T. 2016. Tigecycline is highly efficacious against Mycobacterium abscessus pulmonary disease. Antimicrob Agents Chemother 60:2895–2900. doi: 10.1128/AAC.03112-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Wallace RJ Jr, Dukart G, Brown-Elliott BA, Griffith DE, Scerpella EG, Marshall B. 2014. Clinical experience in 52 patients with tigecycline-containing regimens for salvage treatment of Mycobacterium abscessus and Mycobacterium chelonae infections. J Antimicrob Chemother 69:1945–1953. doi: 10.1093/jac/dku062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Stets R, Popescu M, Gonong JR, Mitha I, Nseir W, Madej A, Kirsch C, Das AF, Garrity-Ryan L, Steenbergen JN, Manley A, Eckburg PB, Tzanis E, McGovern PC, Loh E. 2019. Omadacycline for community-acquired bacterial pneumonia. N Engl J Med 380:517–527. doi: 10.1056/NEJMoa1800201. [DOI] [PubMed] [Google Scholar]
- 11.O’Riordan W, Green S, Overcash JS, Puljiz I, Metallidis S, Gardovskis J, Garrity-Ryan L, Das AF, Tzanis E, Eckburg PB, Manley A, Villano SA, Steenbergen JN, Loh E. 2019. Omadacycline for acute bacterial skin and skin-structure infections. N Engl J Med 380:528–538. doi: 10.1056/NEJMoa1800170. [DOI] [PubMed] [Google Scholar]
- 12.Solomkin J, Evans D, Slepavicius A, Lee P, Marsh A, Tsai L, Sutcliffe JA, Horn P. 2017. Assessing the efficacy and safety of eravacycline versus ertapenem in complicated intra-abdominal infections in the Investigating Gram-Negative Infections Treated with Eravacycline (IGNITE 1) Trial: a randomized clinical trial. JAMA Surg 152:224–232. doi: 10.1001/jamasurg.2016.4237. [DOI] [PubMed] [Google Scholar]
- 13.Kaushik A, Gupta C, Fisher S, Story-Roller E, Galanis C, Parrish N, Lamichhane G. 2017. Combinations of avibactam and carbapenems exhibit enhanced potencies against drug-resistant Mycobacterium abscessus. Future Microbiol 12:473–480. doi: 10.2217/fmb-2016-0234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kaushik A, Ammerman NC, Lee J, Martins O, Kreiswirth BN, Lamichhane G, Parrish NM, Nuermberger EL. 2019. In vitro activity of the new β-lactamase inhibitors relebactam and vaborbactam in combination with β-lactams against Mycobacterium abscessus complex clinical isolates. Antimicrob Agents Chemother 63:e02623-18. doi: 10.1128/AAC.02623-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.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]
- 16.Adékambi 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]
- 17.Griffith DE, Brown-Elliott BA, Benwill JL, Wallace RJ Jr. 2015. Mycobacterium abscessus: “pleased to meet you, hope you guess my name…” Annals ATS 12:436–439. doi: 10.1513/AnnalsATS.201501-015OI. [DOI] [PubMed] [Google Scholar]
- 18.Clinical and Laboratory Standards Institute. 2011. Susceptibility testing of mycobacteria, nocardiae, and other aerobic actinomycetes; approved standard, 2nd ed CLSI document M24-A2; vol 31, no 5. Clinical and Laboratory Standards Institute, Wayne, PA. [PubMed] [Google Scholar]
- 19.Shoen C, Benaroch D, Sklaney M, Cynamon M. 2019. In vitro activities of omadacycline against rapidly growing mycobacteria. Antimicrob Agents Chemother doi: 10.1128/AAC.02522-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Petrini B. 2006. Mycobacterium abscessus: an emerging rapid-growing potential pathogen. APMIS 114:319–328. doi: 10.1111/j.1600-0463.2006.apm_390.x. [DOI] [PubMed] [Google Scholar]
- 21.Wallace RJ Jr, Brown-Elliott BA, Crist CJ, Mann L, Wilson RW. 2002. Comparison of the in vitro activity of the glycylcycline tigecycline (formerly GAR-936) with those of tetracycline, minocycline, and doxycycline against isolates of nontuberculous mycobacteria. Antimicrob Agents Chemother 46:3164–3167. doi: 10.1128/AAC.46.10.3164-3167.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Huang YC, Liu MF, Shen GH, Lin CF, Kao CC, Liu PY, Shi ZY. 2010. Clinical outcome of Mycobacterium abscessus infection and antimicrobial susceptibility testing. J Microbiol Immunol Infect 43:401–406. doi: 10.1016/S1684-1182(10)60063-1. [DOI] [PubMed] [Google Scholar]
- 23.Cavusoglu C, Gurpinar T, Ecemis T. 2012. Evaluation of antimicrobial susceptibilities of rapidly growing mycobacteria by SensiTitre RAPMYCO panel. New Microbiol 35:73–76. [PubMed] [Google Scholar]
- 24.Huang CW, Chen JH, Hu ST, Huang WC, Lee YC, Huang CC, Shen GH. 2013. Synergistic activities of tigecycline with clarithromycin or amikacin against rapidly growing mycobacteria in Taiwan. Int J Antimicrob Agents 41:218–223. doi: 10.1016/j.ijantimicag.2012.10.021. [DOI] [PubMed] [Google Scholar]
- 25.U.S. Food and Drug Administration. 2019. Eravacycline package insert. U.S. FDA, Bethesda, MD: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/211109lbl.pdf. [Google Scholar]
- 26.U.S. Food and Drug Administration. 2019. Omadacycline injection and oral products. U.S. FDA, Bethesda, MD: https://www.fda.gov/Drugs/DevelopmentApprovalProcess/DevelopmentResources/ucm622612.htm. [Google Scholar]
- 27.U.S. Food and Drug Administration. 2019. Eravacycline: injection products. U.S. FDA, Bethesda, MD: https://www.fda.gov/Drugs/DevelopmentApprovalProcess/DevelopmentResources/ucm618563.htm. [Google Scholar]
- 28.Tanaka SK, Steenbergen J, Villano S. 2016. Discovery, pharmacology, and clinical profile of omadacycline, a novel aminomethylcycline antibiotic. Bioorg Med Chem 24:6409–6419. doi: 10.1016/j.bmc.2016.07.029. [DOI] [PubMed] [Google Scholar]
- 29.U.S. Food and Drug Administration. 2019. Omadacycline package insert. U.S. FDA, Bethesda, MD: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/209816_209817lbl.pdf. [Google Scholar]
- 30.Newman JV, Zhou J, Izmailyan S, Tsai L. 2018. Randomized, double-blind, placebo-controlled studies of the safety and pharmacokinetics of single and multiple ascending doses of eravacycline. Antimicrob Agents Chemother 62:e01174-18. doi: 10.1128/AAC.01174-18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Gotfried MH, Horn K, Garrity-Ryan L, Villano S, Tzanis E, Chitra S, Manley A, Tanaka SK, Rodvold KA. 2017. Comparison of omadacycline and tigecycline pharmacokinetics in the plasma, epithelial lining fluid, and alveolar cells of healthy adult subjects. Antimicrob Agents Chemother 61:e01135-17. doi: 10.1128/AAC.01135-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.U.S. Food and Drug Administration. 2019. Tigecycline: injection products, FDA-identified interpretive criteria. U.S. FDA, Bethesda, MD: https://www.fda.gov/Drugs/DevelopmentApprovalProcess/DevelopmentResources/ucm587585.htm. [Google Scholar]
- 33.Zhanel GG, Cheung D, Adam H, Zelenitsky S, Golden A, Schweizer F, Gorityala B, Lagacé-Wiens PR, Walkty A, Gin AS, Hoban DJ, Karlowsky JA. 2016. Review of eravacycline, a novel fluorocycline antibacterial agent. Drugs 76:567–588. doi: 10.1007/s40265-016-0545-8. [DOI] [PubMed] [Google Scholar]
- 34.Solomkin JS, Gardovskis J, Lawrence K, Montravers P, Sway A, Evans D, Tsai L. 2018. IGNITE4: results of a phase 3, randomized, multicenter, prospective trial of eravacycline versus meropenem in the treatment of complicated intra-abdominal infections. Clin Infect Dis doi: 10.1093/cid/ciy1029. [DOI] [PMC free article] [PubMed] [Google Scholar]
