Skip to main content
Antimicrobial Agents and Chemotherapy logoLink to Antimicrobial Agents and Chemotherapy
. 2017 Nov 22;61(12):e01537-17. doi: 10.1128/AAC.01537-17

In Vitro Susceptibility Testing of Tedizolid against Isolates of Nocardia

Barbara A Brown-Elliott 1,, Richard J Wallace Jr 1
PMCID: PMC5700301  PMID: 28923878

ABSTRACT

There is a paucity of efficacious antimicrobials (especially oral) against clinically relevant species of Nocardia. To date, all species of Nocardia have been susceptible to linezolid, the first commercially available oxazolidinone. Tedizolid is a new oxazolidinone with previously reported improved in vitro and in vivo (intracellular) potency against multidrug-resistant strains of Mycobacterium sp. and Nocardia brasiliensis. Using the current Clinical and Laboratory Standards Institute-recommended broth microdilution method, 101 isolates of Nocardia spp., including 29 Nocardia cyriacigeorgica, 17 Nocardia farcinica, 13 Nocardia nova complex, 21 Nocardia brasiliensis, 5 Nocardia pseudobrasiliensis, and 5 Nocardia wallacei isolates and 11 isolates of less common species, were tested for susceptibility to tedizolid and linezolid. For the most common clinically significant species of Nocardia, tedizolid MIC50 values were 0.25 μg/ml for N. nova complex, N. brasiliensis, N. pseudobrasiliensis, and N. wallacei, compared to linezolid MIC50 values of 1, 2, 0.5, and 1 μg/ml, respectively. Tedizolid and linezolid MIC90 values were 2 μg/ml for N. nova complex and N. brasiliensis. Tedizolid MIC50 and MIC90 values for both N. cyriacigeorgica and N. farcinica were 0.5 μg/ml and 1 μg/ml, respectively, compared to linezolid MIC50 and MIC90 values of 2 and 4 μg/ml, respectively. Based on MIC90 values, this study showed that tedizolid was 2- to 3-fold more active than linezolid in vitro against most common species of Nocardia, with the exception of the N. nova complex and N. brasiliensis, for which values were the same. These results may warrant evaluation of tedizolid as a potential treatment option for Nocardia infections.

KEYWORDS: Nocardia, susceptibility testing, tedizolid

INTRODUCTION

Nocardia species are Gram-positive branching rods responsible for various types of infections, including respiratory, cutaneous, and systemic infections (1). Some clinically relevant species of Nocardia are multidrug resistant, and there is a paucity of antimicrobials with efficacy against these organisms (1, 2). Thus, new antimicrobials with therapeutic potential are desperately needed.

All strains of Nocardia isolated to date have been susceptible to linezolid (LZD) (13), the first commercially available oxazolidinone (3). Tedizolid (TZD) is a new oxazolidinone with previously reported in vitro and in vivo (intracellular) activity against Mycobacterium tuberculosis, including multidrug-resistant strains (4), and Nocardia brasiliensis (57). A previously published study by Vera-Cabrera et al. showed in vitro activity of TZD against small numbers of Nocardia brasiliensis strains (7).

Other investigators have also reported enhanced in vitro activity of TZD against bacterial strains, including LZD-resistant strains of Streptococcus pneumoniae, in addition to methicillin-susceptible and methicillin-resistant coagulase-negative Staphylococcus, Staphylococcus aureus, Streptococcus pyogenes, and Streptococcus agalactiae (811). With this superior activity in mind, we undertook an evaluation of the in vitro MICs of TZD, compared with those of LZD and other comparator antimicrobials that are routinely tested in clinical laboratories, for several clinically significant species of Nocardia.

(A portion of this study was presented at the 1st ASM Microbe meeting, Boston, MA, 16 to 20 June 2016.)

RESULTS

A total of 98/101 (97%) of the Nocardia isolates showed MIC50s of ≤1 μg/ml for TZD and LZD (Table 1). Generally, isolates of Nocardia showed either equivalent or 2- to 3-fold lower TZD MIC90 values, compared with LZD MIC values. Table 1 shows the MICs for species tested with ≥5 isolates. The TZD MIC50 values were 0.5 μg/ml for 29 isolates of Nocardia cyriacigeorgica and 17 isolates of Nocardia farcinica, compared to 2 μg/ml for LZD for both species. For 13 isolates of Nocardia nova complex, 22 isolates of Nocardia brasiliensis, and 5 isolates each of Nocardia pseudobrasiliensis and Nocardia wallacei, the TZD MIC50 values were 0.25 μg/ml, with LZD MIC50 values of 1, 2, 0.5, and 1 μg/ml, respectively. Two isolates each of Nocardia abscessus and Nocardia sp. showed TZD MICs of 0.12 μg/ml, compared to 0.25 to 0.5 μg/ml and 0.5 to 1 μg/ml, respectively, for LZD. Single isolates of Nocardia beijingensis, Nocardia niigatensis, and Nocardia otitidiscaviarum had TZD MICs of 0.25 μg/ml, compared to 1, 1, and 2 μg/ml, respectively, for LZD (data not shown). One isolate each of Nocardia asiatica, Nocardia paucivorans, Nocardia mexicana, and Nocardia testacea showed TZD MICs of 0.12 μg/ml, compared to 0.5 μg/ml for N. asiatica, N. paucivorans, and N. mexicana and 1 μg/ml for N. testacea for LZD (data not shown).

TABLE 1.

MIC ranges and MIC50 and MIC90 values for tedizolid and linezolid against the most commonly encountered clinical isolates of Nocardia

Species (no. of isolates tested) and antimicrobial MIC (μg/ml)
Range MIC50 MIC90
N. cyriacigeorgica (29 isolates)
    Tedizolid 0.12–2 0.5 1
    Linezolida 1–4 2 4
N. farcinica (17 isolates)
    Tedizolid 0.25–2 0.5 1
    Linezolida 1–4 2 4
N. nova complex (13 isolates)
    Tedizolid 0.12–2 0.25 2
    Linezolida 0.25–2 1 2
N. brasiliensis (21 isolates)
    Tedizolid 0.12–0.5 0.25 2
    Linezolida 1–4 2 2
N. pseudobrasiliensis (5 isolates)
    Tedizolid 0.12–0.5 0.25
    Linezolida 0.25–2 0.5
N. wallacei (5 isolates)
    Tedizolid 0.06–0.5 0.25
    Linezolida 0.5–2 1
a

There are no intermediate or resistance breakpoints for LZD for Nocardia. All known species isolated to date have been susceptible to LZD (≤8 μg/ml).

MICs for additional antimicrobials were also assessed, to confirm the susceptibility patterns of some species and to provide comparator data for TZD and LZD. The comparator agents that were most active in vitro (other than LZD, to which all known species of Nocardia are susceptible) (3) for the most commonly seen species (i.e., N. cyriacigeorgica, N. farcinica, N. nova complex, and N. brasiliensis) in this study included amikacin and trimethoprim-sulfamethoxazole (TMP-SMX) for all four species and imipenem for N. nova complex.

The manufacturer's acceptable MIC ranges for Staphylococcus aureus ATCC 29213 and Enterococcus faecalis ATCC 29212 were 0.25 to 1 μg/ml. All 34 isolates of S. aureus ATCC 29213 and 10 isolates of E. faecalis had TZD MICs within the acceptable ranges.

DISCUSSION

Tedizolid phosphate is a novel oxazolidinone prodrug (TR-701) that is orally absorbed and converted in the serum to the active drug, tedizolid (TR-700, formerly DA-7157) (8), with a broad range of activity against Gram-positive microorganisms, including mycobacteria and Nocardia (47). The mechanism of action of TZD is through inhibition of protein synthesis. TZD binds to the 50S ribosome, apparently at a site near the 30S ribosome, which blocks formation of the 70S initiation complex and in turn prevents protein synthesis (8). The supposition is that the major site of action of oxazolidinones is at the ribosomal peptidyltransferase center and that unique, phylogenetically conserved, structural features of this site are likely responsible for reducing the mutational resistance and cross-resistance with other antimicrobial classes (8, 11). Studies of crystallographic structures and cross-linking have seemed to substantiate this supposition (12).

Linezolid has been an important addition to the armamentarium of antimicrobials used in the treatment of Nocardia infections (13). The introduction of TZD provides another potential antimicrobial with efficacy against these organisms, and early studies showed that TZD exhibited 4- to 16-fold greater potency than did LZD against some bacteria, including LZD-resistant strains (10, 11, 13, 14). Ninety-seven percent of all of the isolates of Nocardia species studied, including all of the major clinically relevant species, had MICs of ≤1 μg/ml and ≤4 μg/ml for TZD and LZD, respectively.

TZD has high oral bioavailability and a longer half-life (11 h, compared with 5 h for LZD), which allows clinicians to modify the route of administration from intravenous to oral easily and to use once-daily dosing, encouraging greater patient compliance and outpatient usage (15, 16). Moreover, although long-term usage has not been assessed, TZD appears to be better tolerated than LZD, especially in regard to hematological adverse events, including thrombocytopenia (8, 13). In a recent clinical trial by Kim et al. at the National Institute of Allergy and Infectious Diseases, the safety and tolerability among 25 patients treated with TZD were comparable to those for LZD after a median duration of 91 days of therapy. Except for gastrointestinal intolerance, which was 20% with TZD versus 40% with LZD, adverse events, including peripheral neuropathy and thrombocytopenia, were equivalent in occurrence (17). The incidences of anemia were not compared in this trial (17). However, a single case study in Spain showed that a patient (age given as “in his 70s”) with a pulmonary infection with Mycobacterium avium complex and Mycobacterium kansasii that was treated with TZD had stable hemoglobin levels for 42 days, although a subsequent decrease after 58 days of treatment with TZD necessitated the withdrawal of TZD (18). No apparent dose-related toxicity with TZD has been reported (14).

TZD was shown to be more active than LZD in evaluations of the ability to decrease the CFU of bacterial species, including Staphylococcus aureus, Listeria monocytogenes, and Legionella pneumophila, in cultured macrophages or human umbilical vein endothelial cells (9, 19). Furthermore, Vera-Cabrera et al. showed that TZD was more active than LZD in inhibiting the intracellular growth of Nocardia (5). TZD accumulates more rapidly than LZD, and the intracellular concentration of TZD was at least 10- to 15-fold higher than the extracellular concentration, in contrast to the intracellular concentration of LZD, which is equivalent to the extracellular concentration (13, 19, 20). Moreover, previous studies showed excellent penetration of TZD into the epithelial lining fluid of the lungs, suggesting that TZD may be useful in the setting of pneumonia (20), and other studies showed superior distribution of TZD into the interstitial fluid of adipose and muscle tissue, possibly making TZD an attractive therapeutic option for skin and soft tissue infections with Nocardia (13).

Previous studies in healthy adults showed that TZD half-life values are approximately 2-fold greater than those of LZD and TZD is rapidly absorbed, with nearly complete oral bioavailability following 200-mg doses of tedizolid phosphate. Studies also suggested that the 200-mg dose of tedizolid phosphate (150 mg TZD equivalent) has a favorable pharmacokinetic, safety, and efficacy profile, and thus this dose was selected for therapeutic dosing (8, 14, 15, 20). The in vitro MICs of TZD obtained in this and previous studies, along with the once-daily lower dosage for TZD and the potential for fewer and less serious adverse events associated with TZD, compared to LZD, emphasize the potential for TZD in the treatment of infections caused by Nocardia (8, 9, 15, 19, 20).

MATERIALS AND METHODS

Isolates.

A total of 101 isolates of Nocardia sp. that had been submitted to the Mycobacteria/Nocardia Research Laboratory at the University of Texas Health Science Center at Tyler in 2014 and 2015 were tested against TZD, LZD, and other comparative antimicrobials (see Table S1 in the supplemental material). These isolates included 29 N. cyriacigeorgica isolates, 17 N. farcinica isolates, 13 N. nova complex isolates, 21 N. brasiliensis isolates, 5 N. pseudobrasiliensis isolates, 5 N. wallacei isolates, 2 N. abscessus isolates, 2 Nocardia sp. isolates (most closely related to Nocardia carnea), and 1 isolate each of N. beijingensis, N. niigatensis, N. paucivorans, N. asiatica, N. otitidiscaviarum, N. mexicana, and N. testacea.

Identification.

Isolates of Nocardia were identified by sequencing of the secA1 gene and partial 16S rRNA gene as indicated by species and group, following the interpretation criteria published by the Clinical and Laboratory Standards Institute (CLSI) (21) and by Conville et al., as applicable (22). Partial 16S rRNA gene sequencing was performed for species with no available secA1 sequence or isolates for which secA1 sequences were ambiguous (2123).

Antimicrobial susceptibility testing.

Isolates were tested by broth microdilution using customized, frozen, 96-well panels commercially manufactured by Thermo-Fisher Scientific, with doubling dilutions of antimicrobials (TZD concentrations were 0.008 to 32 μg/ml) in cation-adjusted Mueller-Hinton broth, after 3- to 4-day incubations at 35°C, following the CLSI-recommended procedures for Nocardia (24). Comparative antimicrobials included LZD, amikacin, amoxicillin-clavulanic acid, ceftriaxone, ciprofloxacin, clarithromycin, imipenem, minocycline, moxifloxacin, tigecycline, tobramycin, and TMP-SMX (Table S1).

Quality control.

Quality control for susceptibility testing was performed weekly using the CLSI-recommended strain Mycobacterium peregrinum ATCC 700686 for comparative antimicrobials (24) and the strains Staphylococcus aureus ATCC 29213 and Enterococcus faecalis ATCC 29212 for TZD (Table 2).

TABLE 2.

Tedizolid MICs and MIC ranges for reference strains tested

Organism Acceptable MIC
range (μg/ml)
No. of isolates with MIC of:
0.25 μg/ml 0.5 μg/ml 1 μg/ml
Staphylococcus aureus ATCC 29213 0.25–1 8 25 1
Enterococcus faecalis ATCC 29212 0.25–1 0 10 0

Supplementary Material

Supplemental material

ACKNOWLEDGMENTS

We thank Nicholas Parodi, Megan Ashcraft, Amber McKinney, Ravikiran Vasireddy, Sruthi Vasireddy, Elena Iakhiaeva, Katie Shipp, and Terry Smith in the Mycobacteria/Nocardia Laboratory for their excellent laboratory assistance and Joanne Woodring for her superior formatting of the manuscript. We also thank Amy Calamari, Mekki Bensaci, Christopher Tan, and Dominic Wolfe at Merck for their careful review of the manuscript.

Footnotes

Supplemental material for this article may be found at https://doi.org/10.1128/AAC.01537-17.

REFERENCES

  • 1.Brown-Elliott BA, Brown JM, Conville PS, Wallace RJ Jr. 2006. Clinical and laboratory features of the Nocardia spp. based on current molecular taxonomy. Clin Microbiol Rev 19:259–282. doi: 10.1128/CMR.19.2.259-282.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Moylett EH, Pacheco SE, Brown-Elliott BA, Perry TR, Buescher ES, Birmingham MC, Schentag JJ, Gimbel JF, Apodaca A, Schwartz MA, Rakita RM, Wallace RJ Jr. 2003. Clinical experience with linezolid for the treatment of Nocardia infection. Clin Infect Dis 36:313–318. doi: 10.1086/345907. [DOI] [PubMed] [Google Scholar]
  • 3.Brown-Elliott BA, Ward SC, Crist CJ, Mann L, Wilson RW, Wallace RJ Jr. 2001. In vitro activities of linezolid against multiple Nocardia species. Antimicrob Agents Chemother 45:1295–1297. doi: 10.1128/AAC.45.4.1295-1297.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Molina-Torres CA, Barba-Marines A, Valles-Guerra O, Ocampo-Candiani J, Cavazos-Rocha N, Pucci MJ, Castro-Garza J, Vera-Cabrera L. 2014. Intracellular activity of tedizolid phosphate and ACH-702 versus Mycobacterium tuberculosis infected macrophages. Ann Clin Microbiol Antimicrob 13:13. doi: 10.1186/1476-0711-13-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Vera-Cabrera L, Espinoza-Gonzalez NA, Welsh O, Ocampo-Candiani J, Castro-Garza J. 2009. Activity of novel oxazolidinones against Nocardia brasiliensis growing within THP-1 macrophages. J Antimicrob Chemother 64:1013–1017. doi: 10.1093/jac/dkp314. [DOI] [PubMed] [Google Scholar]
  • 6.Espinoza-Gonzalez NA, Welsh O, Ocampo-Candiani J, Said-Fernandez S, Lozano-Garza G, Choi SH, Vera-Cabrera L. 2010. Evaluation of the combined therapy of DA-7218, a new oxazolidinone, and trimethoprim/sulfamethoxazole in the treatment of experimental actinomycetoma by Nocardia brasiliensis. Curr Drug Deliv 7:225–229. doi: 10.2174/156720110791560946. [DOI] [PubMed] [Google Scholar]
  • 7.Vera-Cabrera L, Gonzalez E, Rendon A, Ocampo-Candiani J, Welsh O, Velazquez-Moreno VM, Choi SH, Molina-Torres CA. 2006. In vitro activities of DA-7157 and DA-7218 against Mycobacterium tuberculosis and Nocardia brasiliensis. Antimicrob Agents Chemother 50:3170–3172. doi: 10.1128/AAC.00571-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Kanafani ZA, Corey GR. 2012. Tedizolid (TR-701): a new oxazolidinone with enhanced potency. Expert Opin Invest Drugs 21:515–522. doi: 10.1517/13543784.2012.660250. [DOI] [PubMed] [Google Scholar]
  • 9.Locke JB, Zurenko GE, Shaw KJ, Bartizal K. 2014. Tedizolid for the management of human infections: in vitro characteristics. Clin Infect Dis 58(Suppl 1):S35–S42. doi: 10.1093/cid/cit616. [DOI] [PubMed] [Google Scholar]
  • 10.Rodríguez-Avial I, Culebras E, Betriu C, Morales G, Pena I, Picazo JJ. 2012. In vitro activity of tedizolid (TR-700) against linezolid-resistant staphylococci. J Antimicrob Chemother 67:167–169. doi: 10.1093/jac/dkr403. [DOI] [PubMed] [Google Scholar]
  • 11.Shaw KJ, Poppe S, Schaadt R, Brown-Driver V, Finn J, Pillar CM, Shinabarger D, Zurenko G. 2008. In vitro activity of TR-700, the antibacterial moiety of the prodrug TR-701, against linezolid-resistant strains. Antimicrob Agents Chemother 52:4442–4447. doi: 10.1128/AAC.00859-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Colca JR, McDonald WG, Waldon DJ, Thomasco LM, Gadwood RC, Lund ET, Cavey GS, Mathews WR, Adams LD, Xiong L, Mankin AS. 2003. Cross-linking in the living cell locates the site of action of oxazolidinone antibiotics. J Biol Chem 278:21972–21979. doi: 10.1074/jbc.M302109200. [DOI] [PubMed] [Google Scholar]
  • 13.Sahre M, Sabarinath S, Grant M, Seubert C, DeAnda C, Prokocimer PG, Derendorf H. 2012. Skin and soft tissue concentrations of tedizolid (formerly torezolid), a novel oxazolidinone, following a single oral dose in healthy volunteers. Int J Antimicrob Agents 40:51–54. doi: 10.1016/j.ijantimicag.2012.03.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Prokocimer P, Bien P, Surber J, Mehra P, DeAnda C, Bulitta JB, Corey GR. 2011. Phase 2, randomized, double-blind, dose-ranging study evaluating the safety, tolerability, population pharmacokinetics, and efficacy of oral torezolid phosphate in patients with complicated skin and skin structure infections. Antimicrob Agents Chemother 55:583–592. doi: 10.1128/AAC.00076-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Flanagan SD, Bien PA, Munõz KA, Minassian SL, Prokocimer PG. 2014. Pharmacokinetics of tedizolid following oral administration: single and multiple dose, effect of food, and comparison of two solid forms of the prodrug. Pharmacotherapy 34:240–250. doi: 10.1002/phar.1337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Welshman IR, Sisson TA, Jungbluth GL, Stalker DJ, Hopkins NK. 2001. Linezolid absolute bioavailability and the effect of food on oral bioavailability. Biopharm Drug Dispos 22:91–97. doi: 10.1002/bdd.255. [DOI] [PubMed] [Google Scholar]
  • 17.Kim T, Wills AB, Markus B, Daniel-Wayman S, Prevots DR, Olivier KN. 2016. Safety and tolerability of long term use of tedizolid for treatment of nontuberculous mycobacterial infections, poster 577. ID Week, New Orleans, LA, 26 to 30 October 2016 https://idsa.confex.com/idsa/2016/webprogram/Handout/id5538/POSTER69_577.pdf. [Google Scholar]
  • 18.Yuste JR, Bertó J, Del Pozo JL, Leiva J. 2017. Prolonged use of tedizolid in a pulmonary non-tuberculous mycobacterial infection after linezolid-induced toxicity. J Antimicrob Chemother 72:625–628. doi: 10.1093/jac/dkw484. [DOI] [PubMed] [Google Scholar]
  • 19.Lemaire S, Van Bambeke F, Appelbaum PC, Tulkens PM. 2009. Cellular pharmacokinetics and intracellular activity of torezolid (TR-700): studies with human macrophage (THP-1) and endothelial (HUVEC) cell lines. J Antimicrob Chemother 64:1035–1043. doi: 10.1093/jac/dkp267. [DOI] [PubMed] [Google Scholar]
  • 20.Lodise TP, Drusano GL. 2014. Use of pharmacokinetic/pharmacodynamic systems analyses to inform dose selection of tedizolid phosphate. Clin Infect Dis 58(Suppl 1):S28–S34. doi: 10.1093/cid/cit615. [DOI] [PubMed] [Google Scholar]
  • 21.Clinical and Laboratory Standards Institute. 2008. Interpretive criteria for identification of bacteria and fungi by DNA target sequencing; approved guideline. CLSI document MM18-A Clinical and Laboratory Standards Institute, Wayne, PA. [Google Scholar]
  • 22.Conville PS, Zelazny AM, Witebsky FG. 2006. Analysis of secA1 gene sequences for identification of Nocardia species. J Clin Microbiol 44:2760–2766. doi: 10.1128/JCM.00155-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Edwards U, Rogall T, Blöcker H, Emde M, Böttger EC. 1989. Isolation and direct complete nucleotide determination of entire genes: characterization of a gene coding for 16S ribosomal RNA. Nucleic Acid Res 17:7843–7853. doi: 10.1093/nar/17.19.7843. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Clinical and Laboratory Standards Institute. 2011. Susceptibility testing of mycobacteria, nocardiae, and other aerobic actinomycetes; approved standard—2nd ed. CLSI document M24-A2 Clinical and Laboratory Standards Institute, Wayne, PA. [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental material

Articles from Antimicrobial Agents and Chemotherapy are provided here courtesy of American Society for Microbiology (ASM)

RESOURCES