Introduction

Tedizolid is a relatively new oxazolidinone antibiotic, active against Gram-positive bacteria, and licenced in the UK for the treatment of acute skin and associated structure bacterial infections (SASBIs). Tedizolid 200 mg once daily for 6 days was shown to be as efficacious as 10 days of linezolid (600 mg twice daily) in a randomised controlled trial [1]. The potential to extend tedizolid use to other indications, however, remains unclear due to lack of data [2]. Data are also needed to demonstrate tolerance compared with linezolid in courses longer than 6 days [2]. A recent series of four patients showed that it was possible to use it successfully for 7 to 14 days [3].

The potential for tedizolid to be used as a switch agent from linezolid in patients who have developed linezolid-associated adverse effects (LAAEs) has not been adequately investigated. Our centre has used tedizolid since 2016, mainly within the complex oral and parenteral antibiotic therapy (COPAT) service, to treat a range of infections other than SASBIs. Patients who have developed LAAEs have commonly been switched to tedizolid and prolonged therapy has been prescribed. Within our institution, tedizolid can only be prescribed by an infection consultant when oxazolidinone therapy is considered optimal with prescriptions approved by at least two infection consultants or within a multidisciplinary meeting. Linezolid has remained the first line oxazolidinone. This service evaluation describes our experiences of using tedizolid to date.

Methods

Our hospital is a 1400-bed teaching hospital with all sub-specialties except transplantation. Patients prescribed linezolid/tedizolid as an outpatient are reviewed weekly in an established COPAT service that manages approximately 300 patients yearly. Consecutive patients prescribed tedizolid according to electronic pharmacy records (May 1, 2016, to December 31, 2018) were included. Hard copy and electronic medical records were reviewed. Demographic and clinical characteristics were transcribed to an Excel (for Windows) spreadsheet. Linezolid contraindicated co-medications (as per the linezolid summary of product characteristics (SPC), UK), diagnoses that resulted in tedizolid prescription, and relevant positive microbiological tests were also recorded [4].

Blood test values at the start and end of linezolid/tedizolid were taken from the sample immediately prior to starting/stopping therapy. For patients who had not had bloods within 7 days, the nearest previous blood tests were used. Haemoglobin (Hb) results were categorised as (1) Critical, Hb of < 90 g/L; (2) Observe, Hb 90–100 g/L (inclusive); and (3) Satisfactory, Hb > 100 g/L. Similarly, platelets were categorised as (1) Critical < 50 × 10^9/L; (2) Observe 50–100 × 10^9/L (inclusive); and (3) Satisfactory > 100 × 10^9/L. White cell counts were defined as (1) Low, < 4.0 × 10^9/L; (2) Normal, 4.0 × 10^9/L to 11.0 × 10^9/L (inclusive); and (3) Elevated, > 11.0 × 10^9.

The longest continuous course of tedizolid for each patient was used for this evaluation. Documented adverse effects/events were recorded regardless of causality. Outcomes at the end of therapy were categorised as improved, no change, or worsened. Improved was defined as clearly documented evidence of clinical improvement in the patient’s case records. No change was if there was no clinical improvement or deterioration. Worsened was if there was a documented deterioration in the patient’s condition.

Statistical analyses were performed using Excel (for Windows). Descriptive statistics, with means, medians, and 95% confidence interval (CI), as appropriate, are presented. By United Kingdom (UK) National Research Ethics Service definitions, this study did not require formal ethical assessment as it was defined as a service evaluation, but it was approved by the hospital’s clinical governance and audit committee (reference 2018.256) prior to commencement. All data were recorded and held according to UK data protection laws.

Results

Sixty-eight patients were identified with eight excluded; four because clinical notes were unavailable, two because tedizolid was never received, and in two it was unclear if they had received tedizolid. Baseline demographics are presented in Table 1. All patients had at least one comorbidity with a mean/median of 3 comorbidities; 7 (12%, N = 60) patients had 6 or more comorbidities.

Table 1 Demographics, co-morbidities, and underlying diagnoses

Twenty patients (33%) had more than one diagnosis leading to tedizolid prescription. Microbiology is shown in the Appendix; two patients, who had Gram-negative bacteria isolated from relevant clinical specimens, received tedizolid despite having no Gram-positive bacteria detected.

Forty-nine (82%) patients received linezolid immediately prior to tedizolid for a mean of 18 days (median = 15 days). Table 2 shows the reasons for linezolid cessation. Eleven patients did not receive linezolid beforehand; 10 were prescribed tedizolid because they were taking linezolid contraindicated co-medications and one by mistake. Tedizolid was prescribed with other antibiotics in 33 patients (55%); most commonly ciprofloxacin in 20 patients (33%) [see Appendix].

Table 2 Reasons for stopping linezolid therapy

Outcomes

Most patients (72%) had clearly documented improvement whilst taking tedizolid. Eighteen per cent had no change in their clinical condition and required surgery or alternative antibiotic therapy and 10% deteriorated [see Appendix].

Adverse effects and events

Tedizolid was prescribed for a mean of 27 days (95% CI 22–32, median 21, range 106); 19 mean days (95% CI 9–28, median 15) in those who had not received linezolid prior and 29 mean days (95% CI 23–34, median 22) in those who had. Blood results at the start/end of linezolid/tedizolid are shown in Table 3. The mean difference in platelets at the start and end of therapy was − 131 × 10^9/L (95% CI 166 to − 36) for linezolid and + 5.4 × 10^9/L for tedizolid (95% CI 23 to 33).

Table 3 Blood parameters at the beginning and end of Linezolid and tedizolid therapy, and the unadjusted changes

Table 4 shows tedizolid associated adverse effects and events, course length and clinical outcomes at stopping therapy. Most patients (72%) completed the planned course of tedizolid; 82% in those who had not received linezolid prior and 73% in those who had. Course length in those who stopped early was a mean of 23 days (95% CI 14–31, median 18, range 62), versus 28 days (95% CI 22–34, median 21, range 105) in those who completed. Many patients (31, 52%) had a possible adverse effect whilst taking tedizolid, most commonly nausea in 9 (15%) patients. See Table 4 and Appendix for further details of adverse effects and events.

Table 4 Number of patients stopping tedizolid therapy early and the reasons why, and the final outcomes on stopping tedizolid

Discussion

Whilst this is a small descriptive study, there are currently limited published data on the tolerance and efficacy of tedizolid with prolonged use and for indications other than SASBIs. To our knowledge, our study represents the largest to date, and may be of use to clinicians considering tedizolid for their patients or their institutions antimicrobial formulary.

With a mean course length of 27 days, and most patients completing the planned course (72%), in a cohort of patients within which a high proportion had stopped linezolid due to adverse effects immediately prior to tedizolid, it would appear that tedizolid was well tolerated despite approximately one in two patients suffering an apparent adverse effect (see Appendix). Nausea, fatigue, and loose stools were the most common adverse effects likely to be due to tedizolid, consistent with existing literature [5].

Most patients had complex, often polymicrobial, infections, particularly diabetic foot and bone/joint; required prolonged antimicrobial therapy; and had multiple co-morbidities. Within this context, it is very challenging to accurately attribute the contribution of tedizolid to clinical outcomes, but most patients had clearly documented clinical improvement. Our findings may therefore suggest a wider role for tedizolid than current, supporting recent in vitro data indicating activity against a wide variety of Gram-positive isolates [6]. One must be mindful that prior antibiotic therapy, however, may have been the predominant influencer of clinical outcomes.

Tedizolid penetration in diabetic foot infections was investigated in vivo by Stainton et al. who found levels in soft tissue extracellular fluid of the lower limb adequate for a “high probability of bacterial kill” [7]. This supports our findings that tedizolid may be useful in managing diabetic foot infections. In bone and joint infections, however, Abad et al. found that whilst tedizolid can prevent biofilm formation, it is inactive against “biofilm-embedded S. aureus” [8]. In our cohort, 9 patients with prosthetic joint infections (PJI) were prescribed tedizolid monotherapy; 6 of these showed clear improvement, whilst 3 showed no change. Overall, of 15 patients with PJI 10 improved whilst taking tedizolid.

There remains a debate about the tolerance of tedizolid versus linezolid. Pooled analyses of randomised trials (ESTABLISH-1 and ESTABLISH-2) showed fewer episodes of thrombocytopenia and gastrointestinal adverse effects with tedizolid, although tedizolid was prescribed for a shorter duration [9]. A review of FDA reported data indicated similar risks of thrombocytopenia with both medications [10]. Our evaluation suggests that patients who do not tolerate linezolid can switch to tedizolid and subsequently often tolerate prolonged therapy; haemoglobin and platelet counts did not deteriorate during prolonged tedizolid. Indeed, in our limited data set, average platelet counts and the number of patients with a platelet count in the “observe” category improved with tedizolid, but not linezolid. There is existing literature that suggests that switching to tedizolid in cases of linezolid related myelotoxicity can result in improved blood counts [11]. The apparently high occurrence of adverse effects associated with concomitant use of ciprofloxacin (see Appendix) is of interest and in keeping with recent concerns regarding its use and our clinical experience with linezolid plus ciprofloxacin combination.

In summary, our evaluation suggests that the prolonged use of tedizolid when clinically indicated is safe. Tedizolid may also be a useful switch agent for patients with LAAEs when ongoing oxazolidinone therapy is felt to be clinically important. Further evaluation in larger cohorts of patients is required.