ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

Linezolid and tedizolid (oxazolidinones): An overview

Linezolid and tedizolid (oxazolidinones): An overview
Literature review current through: Jan 2024.
This topic last updated: Dec 14, 2023.

INTRODUCTION — Linezolid is a synthetic oxazolidinone with bacteriostatic activity against gram-positive organisms [1]. Tedizolid is a newer drug in the same class with comparable spectrum of activity but with limited US Food and Drug Administration-approved indications. Longer-term use of these drugs has been limited by concern for adverse effects.

This topic will review issues related to the clinical use of linezolid and tedizolid. The clinical indications in which the drug may be used are discussed separately in the appropriate topic reviews.

SPECTRUM AND INDICATIONS — Linezolid and tedizolid demonstrate activity in vitro against a variety of gram-positive bacteria including streptococci, enterococci (including vancomycin-resistant enterococci [VRE]), coagulase-negative staphylococci, methicillin-sensitive Staphylococcus aureus, methicillin-resistant S. aureus (MRSA), Bacillus species, Corynebacterium species, and Listeria monocytogenes [2-9].

Linezolid and tedizolid demonstrate activity in vitro against several Mycobacterium spp, including both Mycobacterium tuberculosis and nontuberculous mycobacterium [10-15]. In addition, linezolid and tedizolid are active in vitro against numerous Nocardia spp [16-18]. Linezolid abolishes toxin production and reduces sporulation in Bacillus anthracis in vitro; therefore, it is a treatment option for select infections due to anthrax [19,20].

Linezolid is approved by the US Food and Drug Administration (FDA) for treatment of adults and children with pneumonia (nosocomial and community-acquired) and skin and skin structure infections (complicated and uncomplicated), including those due to MRSA and VRE [21,22]. (See "Acute cellulitis and erysipelas in adults: Treatment" and "Methicillin-resistant Staphylococcus aureus (MRSA) in adults: Treatment of skin and soft tissue infections" and "Treatment of enterococcal infections".)

Clinical experience with use of linezolid for other forms of infections due to MRSA and VRE has been described, including bacteremia and osteomyelitis [23]. In addition, linezolid has been used for treatment of mycobacterial infections and nocardial infections. (See "Treatment of drug-resistant pulmonary tuberculosis in adults" and "Overview of nontuberculous mycobacterial infections".)

Tedizolid is approved by the FDA for treatment of adults and children ≥12 years with complicated and uncomplicated skin and skin structure infections. Data from randomized trials suggest tedizolid is noninferior to linezolid for the treatment of acute bacterial skin and soft tissue infections [24,25].

MECHANISM OF ACTION — Linezolid and tedizolid are bacteriostatic oxazolidinones that inhibit initiation of bacterial protein synthesis at the 50S ribosome. The 50S subunit contains 5S rRNA, 23S rRNA, and 34 proteins [4-6,26,27]. These agents also suppress production of bacterial toxins such as Panton-Valentine leukocidin, alpha-hemolysin, and toxic shock syndrome toxin-1 [28-30].

RESISTANCE — Among staphylococci, linezolid resistance appears to occur most frequently among coagulase-negative staphylococci isolates, mainly Staphylococcus epidermidis [31]. However, S. aureus linezolid resistance, while infrequent, has also been described [32-36].

Vancomycin-resistant enterococci with linezolid resistance has also been described [37]. Resistance in enterococci is also infrequent, occurring in <1 percent of isolates in large surveillance studies [38]. Resistance to linezolid typically occurs through multiple mutations of the 23S rRNA genes [39].

Resistance also can emerge from mutations or by acquisition from other organisms [40].

A mobile gene, cfr, encoding a methyltransferase that modifies 23S rRNA (causing failure of linezolid binding to its bacterial target site, conferring resistance) has been identified in staphylococci [41-43] and in enterococci [44,45].

In vitro data demonstrating retained tedizolid activity against cfr isolates suggest this may be an effective alternative agent against some linezolid-resistant pathogens, most notably S. aureus [46-48]. Such isolates, although susceptible, may also demonstrate increases in tedizolid minimum inhibitory concentration (MIC) [48].

Other plasmid-borne resistant determinants, optrA and poxtA, encode ribosomal protection factors leading to increased MICs for linezolid and tedizolid [45,49,50]. In one study including 154 linezolid-resistant Enterococcus isolates (Enterococcus faecium and Enterococcus faecalis), 26.3 percent of isolates carrying the poxtA-encoding plasmid also harbored the vanA gene [50].

PHARMACOKINETICS AND PHARMACODYNAMICS

General principles — The systemic absorption of linezolid approaches 100 percent following oral administration [51]. Linezolid binds poorly to serum proteins (31 percent). Therefore, it penetrates well into most body compartments (including bone, alveoli, and the cerebrospinal fluid) [51]. As a result, linezolid has a volume of distribution that is similar to total body water (30 to 50 L). The overall tissue distribution of linezolid is stable and is not adversely affected by sepsis or peripheral vascular disease or obesity [52-58]. The drug undergoes hepatic oxidative metabolism into two inactive metabolites eliminated predominantly in the urine [52]. Linezolid has an elimination half-life of four hours [1].

Tedizolid phosphate (a prodrug) is rapidly converted by phosphatases to the active form of tedizolid after oral or intravenous administration. Similar to linezolid, bioavailability following oral administration is high (approximately 90 percent). In contrast to linezolid, tedizolid is highly bound to plasma proteins (70 to 90 percent). It is extensively distributed (with a volume of distribution of 67 to 80 L). Elimination as inactive sulfate conjugate occurs via the liver, with <3 percent of the dose excreted in feces and urine as unchanged drug [59,60].

Based largely on in vitro modeling, the antimicrobial effectiveness of both linezolid and tedizolid is best predicted by the ratio of the 24-hour area under the time-concentration curve to minimum inhibitory concentration (MIC) [52,61]. Based on these targets, simulations examining target attainment suggest that isolates with an MIC ≥2 mcg/mL may not respond to standard dosing [59,62,63]. Linezolid displays moderate postantibiotic effects against S. aureus [64].

Children — In general, the clearance of linezolid in children is influenced by age, weight, and renal function [65-67]. In studies of linezolid in children with critical illness, drug clearance correlated directly with body weight and inversely with aspartate aminotransferase [68]. While clearance in newborns is comparable with clearance in adults, children <12 years of age exhibit more rapid clearance than adults [69].

Published data for tedizolid pharmacokinetics in pediatric patients are limited to adolescents and were generally comparable with parameters seen in adult patients [70].

Other populations — Significant variability in linezolid trough concentrations have been reported in hospitalized patients [62]. Patients >80 years of age and those with renal dysfunction have demonstrated elevated trough concentrations [71]. In contrast, critical care patients and those with cystic fibrosis or major burns may be at increased risk of subtherapeutic exposure with standard dosing. Pharmacokinetic studies also indicate altered pharmacokinetics of linezolid in patients with obesity and hepatic dysfunction. (See 'Dosing and administration' below and 'Serum concentration monitoring' below.)

Tedizolid pharmacokinetics are not altered in older adult patients [72] or in patients with cystic fibrosis [73].

DOSING AND ADMINISTRATION

General principles — Linezolid may be given orally or parenterally. The adult dose is 600 mg intravenously or orally twice daily [74]. A reduced oral dose (linezolid 400 mg twice daily) can be administered for uncomplicated skin and skin structures in adult patients. The pediatric dose is 10 mg/kg intravenously or orally either every 8 hours (≤12 years of age) or every 12 hours (>12 years of age).

Linezolid 600 mg to 1200 mg daily has been evaluated for treatment of drug-resistant tuberculosis as part of combination therapy (often bedaquiline and pretomanid, with or without moxifloxacin) [75-77].

Tedizolid may be given orally or parenterally as a single daily dose of 200 mg. It is US Food and Drug Administration approved for patients >12 years of age.

Renal insufficiency — No dose adjustments are recommended for patients with renal insufficiency based on the product monograph. However, renal impairment leads to accumulation of linezolid and its metabolites, and observational data has demonstrated increased risk of thrombocytopenia in patients with renal insufficiency [78,79].

Use of serum drug concentration monitoring with dose adjustment may be useful in patients with renal insufficiency [80,81]. (See 'Serum concentration monitoring' below.)

Hepatic insufficiency — Despite reductions in linezolid clearance in patients with cirrhosis [82], dose adjustments for linezolid and tedizolid are not recommended for patients with hepatic insufficiency.  

Use of ECMO — Reduced serum concentrations of linezolid have been observed in patients undergoing extracorporeal membrane oxygenation (ECMO) while receiving standard dosing [83-85]. Use of serum drug concentration monitoring with dose adjustment may be useful in such cases. (See 'Serum concentration monitoring' below.)

Obesity — Standard linezolid dosing may be inadequate for patients who are overweight and patients with obesity [86,87]. In such cases, some favor that linezolid dose adjustments be based on creatinine clearance (estimated utilizing the Chronic Kidney Diseases Epidemiology formula [CrCLCKD-EPI]), rather than based on weight (using body mass index or total body weight) [87]. For patients ≥140 kg with invasive infection due to pathogen with a minimum inhibitory concentration ≥2 mcg/mL in the setting of weight ≥140 kg and CrCLCKD-EPI ≥60 mL/min/1.73 m2, dose escalation (up to 450 mg every 8 hours) may be appropriate.

Duration of treatment — Linezolid and tedizolid are licensed for a maximum treatment duration of 28 days and 6 days, respectively. Excluding its use in mycobacterial infections, clinical studies have reported efficacy and safety data on the use of linezolid for periods beyond 28 days. Only limited data are available to determine the safety of tedizolid for extended periods [88-90].

Coadministration with other agents — When possible, patients on serotonergic agent(s) who require linezolid or tedizolid should discontinue the serotonergic agent at least two weeks prior to beginning oxazolidinone therapy (table 1). However, this approach is not practical for management of acute bacterial infection. (See 'Serotonin syndrome' below.)

Patients on linezolid or tedizolid should avoid tyramine-containing foods (table 2).

PREGNANCY AND BREASTFEEDING — Data on use of linezolid and tedizolid in pregnancy and breastfeeding are limited.

Studies of linezolid in animals have revealed adverse fetal effects (decreased fetal body weight and increased incidence of fusion of costal cartilages) [91]. However, there are no controlled studies in pregnant women. Linezolid and tedizolid should be given to pregnant women only if the potential benefits justify the potential risk to the fetus.

Linezolid concentrations in breast milk are similar to maternal serum concentrations. If linezolid is required by the mother, it is not a reason to discontinue breastfeeding. However, because there is no published experience with linezolid during breastfeeding, an alternate drug may be preferred, especially while nursing a newborn or preterm infant [92].

It is unknown whether tedizolid is excreted in human breast milk [60].

ADVERSE EFFECTS

General principles — In general, safety concerns have limited widespread and extended use of linezolid. The most significant adverse effects include gastrointestinal symptoms, myelosuppression (most commonly thrombocytopenia), neuropathy (peripheral and optic), and lactic acidosis [93-95]. Less frequently observed adverse effects include hepatotoxicity, hypoglycemia, and syndrome of inappropriate antidiuretic hormone secretion [1,96].

Risk factors for adverse effects (notably peripheral neuropathy and myelosuppression) secondary to linezolid administration appear to be both dose and duration related [75,97]. In clinical trials utilizing linezolid 1200 mg/d (as part of combination therapy) for drug-resistant tuberculosis, peripheral neuropathy and myelosuppression developed in 81 percent and 48 percent of study subjects (respectively) [76]. In contrast, rates were substantially lower (24 percent and 13 percent, respectively) when lower doses (600 mg/d) were utilized [75].

Data on tedizolid at higher doses for extended periods are limited. In one report of 60 patients receiving tedizolid for a mean length of 27 days, most (72 percent) finished the course [88]. Another reported tedizolid use for longer durations (median treatment duration of 29 days [interquartile range -IQR- 15 to 44]) in 51 patients for the treatment of osteoarticular infections [90].

Tedizolid has been associated with myelosuppression [98] and neuropathy. Use and experience with tedizolid is limited compared with that of linezolid. Emerging data also suggests the risk of lactic acidosis or serotonin syndrome may be similar with tedizolid as with linezolid [1,99].

Available studies comparing adverse effects linezolid and tedizolid include the following:

In a randomized trial including patients with acute bacterial skin and skin infections treated with tedizolid (200 mg orally once daily for 6 days) or linezolid (600 mg orally every 12 hours for 10 days), the most frequent adverse effects were nausea (8.5 versus 13.4 percent), headache (6.3 versus 5.1 percent), and diarrhea (4.5 versus 5.4 percent) [24].

In a randomized trial including patients with pneumonia on mechanical ventilation treated with tedizolid (200 mg IV once daily for 7 days) or linezolid (600 mg IV every 12 hours for 10 to 14 days), the most frequent adverse effects were anemia (0.6 versus 1.1 percent), thrombocytopenia (0.6 versus 0.8 percent), and diarrhea (1.7 versus 5.5 percent), respectively [100].

A retrospective review of the US Food and Drug Administration (FDA) Adverse Event Reporting System (FAERS) database included 271 and 11,259 adverse events due to tedizolid and linezolid respectively. No difference in the odds of pancytopenia, peripheral neuropathy, serotonin syndrome, and lactic acidosis were observed between oxazolidinones [99].

Myelosuppression — Linezolid has been associated with suppression of all blood cell lineages; the incidence is less than 1 percent and is generally dependent on dose and duration of treatment [101]. Thrombocytopenia (<100,000 platelets/microL) is the most common presentation of linezolid-induced myelosuppression [102].

Risk factors for linezolid-induced myelosuppression include low baseline blood cell counts, renal impairment, and duration of linezolid therapy ≥14 days [78,93,101-106]. Monitoring for myelosuppression during linezolid administration is discussed below. (See 'Monitoring' below.)

Count recovery typically occurs within one to three weeks after discontinuation of linezolid [101,107].

There are no known effective measures for prevention of linezolid-induced myelosuppression. Use of pre-emptive pyridoxine was not associated with prevention of myelosuppression in one study including 24 patients treated with a prolonged course of linezolid [108]. The time to recovery varies with the cell line and degree of suppression [101].

Data comparing the incidence and severity of linezolid-induced thrombocytopenia with that of tedizolid come primarily from trials for treatment of skin and skin structure infections. In these studies, the incidence of tedizolid-associated myelosuppression was similar or lower to that observed with linezolid [109,110].

Neuropathy — Oxazolidinones have been associated with peripheral neuropathy and optic neuropathy; these effects are time and dose dependent and their incidence is unknown [99,111-114]. The reported duration of use prior to development of neuropathy is 5 to 11 months [115]. The mechanism of neural toxicity is uncertain. Possibilities include impairment of mitochondrial protein synthesis or direct toxic effects [115-117].

Peripheral neuropathy typically presents as a "glove and stocking" sensory impairment. Nerve conduction studies demonstrate a sensory-motor axonal pattern [111,118]. Optic neuropathy presents with diminished visual acuity, development of scotomas, and diminished color perception [112,115].

Oxazolidinones should be discontinued in patients with peripheral or optic neuropathy. There is no specific treatment for oxazolidinones-induced neuropathy.

Optic neuropathy may be reversible. In one report including two patients with linezolid-induced optic neuropathy, visual function gradually recovered three to four months after drug discontinuation [119]. Peripheral neuropathy may be irreversible in some cases [115,120].

Lactic acidosis — Between 2011 and 2016, 90 cases of lactic acidosis associated with linezolid use were reported to the US Food and Drug Administration [121]. Between 2014 and 2020, 154 cases of lactic acidosis due to linezolid and 4 cases due to tedizolid were reported to the US Food and Drug Administration [99]. Lactic acidosis appears to occur more frequently in the setting of prolonged linezolid administration (40 to 50 days) [122]. The mechanism may be attributable to mitochondrial toxicity, given the similarity between human mitochondrial 16S RNA and bacterial 23S rRNA [95,123].

Onset of oxazolidinones-associated lactic acidosis may be 1 to 16 weeks after drug initiation [99,120]. Clinical manifestations may be nonspecific and include abdominal pain, nausea, vomiting, and generalized weakness, in the setting of low serum bicarbonate concentration. Such manifestations should prompt measurement of serum lactate concentration [122].

Oxazolidinones should be discontinued in patients with lactic acidosis [94,95]. Oxazolidinones-induced lactic acidosis has a mortality rate of 25 to 50 percent [99,124]. In patients who recover, lactate levels generally normalize in 2 to 14 days [122]. Issues related to management of lactic acidosis are discussed separately. (See "Approach to the adult with metabolic acidosis" and "Approach to the child with metabolic acidosis".)

DRUG INTERACTIONS — Administration of linezolid with concomitant serotonergic agents (notably selective serotonin-reuptake inhibitors [SSRIs], serotonin norepinephrine reuptake inhibitors [SNRIs]) or agents inhibiting monoamine oxidase have been associated with serotonin syndrome. (See 'Serotonin syndrome' below.)

Linezolid does not interact with the cytochrome P450 oxidative system. Inducers of CYP3A may increase the clearance of linezolid. Concomitant administration of agents that either induce (eg, rifampin, levothyroxine) or inhibit (eg, clarithromycin, select proton pump inhibitors) p-glycoprotein may decrease or increase linezolid concentrations, respectively.

Coadministration of linezolid with warfarin may be associated with increased prothrombin time; the mechanism is not known [62,125].

Plasma concentrations of oral Breast Cancer Resistance Protein (BCRP) substrates may be increased by coadministration with oral tedizolid due to its inhibition of BCRP in the intestine [60]. Coadministration of tedizolid with BCRP substrates with a narrow therapeutic index (such as methotrexate or topotecan) should be avoided if possible.

Concomitant use of linezolid may enhance the hypertensive effect of either directly and indirectly acting sympathomimetic agents (such as pseudoephedrine, epinephrine, norepinephrine) and dopaminergic agents (eg, dopamine, dobutamine).

Additional information on drug interactions may be found using the drug interactions tool within UpToDate.

Serotonin syndrome — Linezolid can reversibly inhibit monoamine oxidase (MAO). Coadministration of linezolid with nonselective MAO inhibitors, SSRIs, SNRIs, or bupropion can precipitate serotonin toxicity (table 1) [1,126-130]. Similarly, ingestion of tyramine-containing foods can cause hypertensive crisis or serotonin syndrome (table 2) [1,131]. (See "Serotonin syndrome (serotonin toxicity)".)

Estimates of risk of serotonin syndrome resulting from concomitant use of linezolid and other serotonergic agents range from 0.2 to 1 percent [1,131-135]. The highest risk appears to be in combination with higher linezolid doses and/or >2 serotonergic agents and/or specific agents (such as citalopram, escitalopram, MAO-Is, and methadone).

The US Food and Drug Administration has warned that linezolid should be used in patients taking SSRIs or SNRIs only when absolutely needed [136]. If linezolid is required, the serotonergic medication should be stopped at least two weeks in advance of linezolid treatment, if feasible. However, abrupt withdrawal of SSRIs or SNRIs can be problematic due to concern for withdrawal. In such situations, dose reduction may be an acceptable alternative. While concomitant use of linezolid with an SSRI or SNRI is not absolutely contraindicated, patients receiving serotonergic agents while receiving linezolid should be monitored closely. Patients receiving concomitant therapy should be educated regarding the signs and symptoms of serotonin syndrome.

Clinical manifestations of serotonin syndrome include a broad range of symptoms, from mild tremor to life-threatening hyperthermia and shock. Onset of symptoms may be hours to days following coadministration of linezolid with serotonergic agents and may include tachycardia, hypertension, hyperthermia, agitation, tremor, myoclonus, hyperreflexia, muscle rigidity, dilated pupils, dry mucus membranes, increased bowel sounds, flushed skin, and diaphoresis. Diagnostic criteria are discussed separately [137]. (See "Serotonin syndrome (serotonin toxicity)".)

The approach to management depends on the severity of illness. In patients with severe serotonergic syndrome, all serotonergic agents should be discontinued. Other aspects of management for serotonin syndrome are discussed further separately. (See "Serotonin syndrome (serotonin toxicity)".)

Symptoms usually resolve within 24 hours of drug discontinuation and initiating care, but drugs with long durations of action or active metabolites may cause prolonged symptoms.

Tedizolid exhibits weak and reversible MAO inhibition [138]. Therefore, the potential for serotonin syndrome is thought to be less than linezolid. In addition, patients at increased risk of serotonin syndrome were specifically excluded from phase 2 and 3 trials of tedizolid. And early published experience with tedizolid was restricted to shorter courses of therapy than linezolid.

Descriptions of prolonged tedizolid therapy for the management of "off-label" indications (such as nontuberculous mycobacterial infections) have been reported [89,99,139,140]. These retrospective reports generally involve a limited number of patients. Between 2014 and 2020, 369 cases of serotonin syndrome due to tedizolid were reported to the US Food and Drug Administration (FDA) [99].

MONITORING

Clinical monitoring — For patients receiving linezolid longer than 28 days, routine ophthalmologic and neurologic assessment should be performed [106].

Patients on serotonergic agents who are receiving linezolid or tedizolid should be monitored for signs and symptoms of serotonin syndrome.

Laboratory monitoring — For patients receiving linezolid or tedizolid for longer than seven days, routine laboratory monitoring includes weekly complete blood count, basic metabolic panel, and liver function panel [102].

In addition, twice-weekly blood count monitoring is warranted for patients with underlying myelosuppression or receiving other potentially myelosuppressive medications.

Serum concentration monitoring

To reduce toxicity – Serum concentration monitoring of linezolid has been proposed as a potential strategy to reduce toxicity, notably thrombocytopenia [80,141-144]. Serum concentration monitoring may be most useful in patients with high interpatient variability in serum concentrations. These include patients with renal impairment or augmented renal clearance, patients on renal replacement therapy, patients receiving high doses and/or prolonged linezolid therapy (>28 days), children, patients with severe hepatic insufficiency, patients receiving multiple concomitant interacting medications (including clarithromycin, proton pump inhibitors, amiodarone, amlodipine, calcium channel blockers, rifampin, phenobarbital, and levothyroxine), patients with low baseline platelet counts, and patients with extremes of body weight.

However, the role of routine serum concentration monitoring of linezolid is hampered by lack of established peak and trough targets, and absence of a readily available assay. For patients who do undergo linezolid serum concentration monitoring, target serum trough concentrations of 2 to 8 mcg/mL are suggested.

The above approach is supported by a retrospective study including 108 patients (including 21 patients with eGFR ≤60 mL/min) who underwent linezolid therapeutic drug monitoring, lower rates of treatment failure (due to toxicity or persistent infection) were observed among patients who monitored (14 versus 64 percent) [80,144].

To enhance efficacy – The benefits of monitoring to enhance efficacy have not been established, since optimal exposures based on pharmacokinetic/pharmacodynamic targets may vary with pathogen and site of infection. Thus far, clinical trial data for tedizolid have not established a relationship between drug exposure and clinical response [100,145].

Limited clinical data have been published to define optimal efficacy targets for most pathogens. Dose titrations may further be limited by the lack of commercially available dosage forms to provide the desired amount.

SUMMARY

Linezolid is a synthetic oxazolidinone with bacteriostatic activity against gram-positive organisms, including methicillin-resistant Staphylococcus aureus and vancomycin-resistant enterococci. Tedizolid is a newer drug in the same class with comparable spectrum of activity. Use of these drugs has been limited by cost and concern for adverse effects. In general, they are reserved for patients who do not respond to or cannot tolerate other agents. (See 'Spectrum and indications' above.)

Linezolid and tedizolid are bacteriostatic oxazolidinones that inhibit initiation of bacterial protein synthesis at the 50S ribosome. These drugs also suppress production of bacterial toxins. Linezolid failure and resistance have been described. (See 'Mechanism of action' above and 'Resistance' above.)

Linezolid and tedizolid may be administered orally or intravenously. Both drugs exhibit high oral bioavailability and do not require dosing adjustment in patients with hepatic or renal dysfunction. Ideally, patients on serotonergic agent(s) (table 1) who require linezolid or tedizolid should discontinue the serotonergic agent at least two weeks prior to beginning oxazolidinone therapy and avoid tyramine-containing foods (table 2). (See 'Dosing and administration' above.)

Safety concerns limit use of linezolid, particularly for extended durations. Adverse effects include myelosuppression, neuropathy (peripheral and optic), and lactic acidosis. (See 'Adverse effects' above.)

Linezolid-associated myelosuppression can involve suppression of one or more blood cell lineages; it is typically mild and reversible. Thrombocytopenia (<100,000 platelets/microL) is the most common presentation. Risk factors include low baseline blood cell counts, renal impairment, and duration of linezolid therapy ≥14 days. Management consists of drug discontinuation and supportive care. (See 'Myelosuppression' above.)

Linezolid has been associated with peripheral neuropathy and optic neuropathy. The neuropathic effects are time and dose dependent. Peripheral neuropathy typically presents as a "glove and stocking" sensory impairment. Optic neuropathy presents with diminished visual acuity, development of scotomas, and diminished color perception. Management consists of drug discontinuation. (See 'Neuropathy' above.)

The likelihood of linezolid-associated lactic acidosis increases with the duration of administration. Clinical manifestations may be nonspecific and include abdominal pain, nausea, vomiting, and generalized weakness, in the setting of low serum bicarbonate concentration. Management consists of drug discontinuation and supportive care. (See 'Lactic acidosis' above and "Approach to the adult with metabolic acidosis" and "Approach to the child with metabolic acidosis".)

Concomitant administration of linezolid with serotonergic agents (table 1) has been associated with serotonin syndrome. Clinical manifestations include a broad range of symptoms, from mild tremor to life-threatening hyperthermia and shock. The management approach depends on the illness severity. In patients with mild symptoms, clinicians must weigh risks and benefits when determining whether to continue coadministration of linezolid with serotonergic agents. Patients who require continuation of serotonergic agents should receive linezolid only if no other antimicrobial therapies are available; in such cases, patients should be monitored closely. (See 'Serotonin syndrome' above and "Serotonin syndrome (serotonin toxicity)".)

Tedizolid has been associated with myelosuppression and neuropathy. Its association with serotonin syndrome is weak and includes patients treated with prolonged therapy. (See 'Adverse effects' above and 'Drug interactions' above.)

For patients receiving linezolid or tedizolid for longer than seven days, routine laboratory monitoring includes weekly complete blood count, basic metabolic panel, and liver function panel. For patients with underlying myelosuppression or receiving other potentially myelosuppressive medications, twice-weekly blood count monitoring is warranted. For patients receiving linezolid longer than 28 days, routine ophthalmologic and neurologic assessment should be performed. (See 'Monitoring' above.)

  1. Douros A, Grabowski K, Stahlmann R. Drug-drug interactions and safety of linezolid, tedizolid, and other oxazolidinones. Expert Opin Drug Metab Toxicol 2015; 11:1849.
  2. Gómez-Garcés JL, Alos JI, Tamayo J. In vitro activity of linezolid and 12 other antimicrobials against coryneform bacteria. Int J Antimicrob Agents 2007; 29:688.
  3. Barberis CM, Sandoval E, Rodriguez CH, et al. Comparison between disk diffusion and agar dilution methods to determine in vitro susceptibility of Corynebacterium spp. clinical isolates and update of their susceptibility. J Glob Antimicrob Resist 2018; 14:246.
  4. Weigelt J, Itani K, Stevens D, et al. Linezolid versus vancomycin in treatment of complicated skin and soft tissue infections. Antimicrob Agents Chemother 2005; 49:2260.
  5. Moellering RC. Linezolid: the first oxazolidinone antimicrobial. Ann Intern Med 2003; 138:135.
  6. Wootton M, Howe RA, Walsh TR, et al. In vitro activity of 21 antimicrobials against vancomycin-resistant Staphylococcus aureus (VRSA) and heteroVRSA (hVRSA). J Antimicrob Chemother 2002; 50:760.
  7. Ballow CH, Jones RN, Biedenbach DJ, North American ZAPS Research Group. A multicenter evaluation of linezolid antimicrobial activity in North America. Diagn Microbiol Infect Dis 2002; 43:75.
  8. Pfaller MA, Mendes RE, Streit JM, et al. Five-Year Summary of In Vitro Activity and Resistance Mechanisms of Linezolid against Clinically Important Gram-Positive Cocci in the United States from the LEADER Surveillance Program (2011 to 2015). Antimicrob Agents Chemother 2017; 61.
  9. Noll M, Kleta S, Al Dahouk S. Antibiotic susceptibility of 259 Listeria monocytogenes strains isolated from food, food-processing plants and human samples in Germany. J Infect Public Health 2018; 11:572.
  10. Lifan Z, Sainan B, Feng S, et al. Linezolid for the treatment of extensively drug-resistant tuberculosis: a systematic review and meta-analysis. Int J Tuberc Lung Dis 2019; 23:1293.
  11. Brown-Elliott BA, Ward SC, Crist CJ, et al. In vitro activities of linezolid against multiple Nocardia species. Antimicrob Agents Chemother 2001; 45:1295.
  12. Lebeaux D, Bergeron E, Berthet J, et al. Antibiotic susceptibility testing and species identification of Nocardia isolates: a retrospective analysis of data from a French expert laboratory, 2010-2015. Clin Microbiol Infect 2019; 25:489.
  13. Ruiz P, Causse M, Vaquero M, Casal M. In Vitro Activity of Tedizolid against Mycobacterium tuberculosis. Antimicrob Agents Chemother 2019; 63.
  14. Ruth MM, Koeken VACM, Pennings LJ, et al. Is there a role for tedizolid in the treatment of non-tuberculous mycobacterial disease? J Antimicrob Chemother 2020; 75:609.
  15. Chapagain M, Gumbo T, Heysell SK, Srivastava S. Comparison of a Novel Regimen of Rifapentine, Tedizolid, and Minocycline with Standard Regimens for Treatment of Pulmonary Mycobacterium kansasii. Antimicrob Agents Chemother 2020; 64.
  16. Yi M, Wang L, Xu W, et al. Species Distribution And Antibiotic Susceptibility Of Nocardia Isolates From Yantai, China. Infect Drug Resist 2019; 12:3653.
  17. Moylett EH, Pacheco SE, Brown-Elliott BA, et al. Clinical experience with linezolid for the treatment of nocardia infection. Clin Infect Dis 2003; 36:313.
  18. Brown-Elliott BA, Wallace RJ Jr. In Vitro Susceptibility Testing of Tedizolid against Isolates of Nocardia. Antimicrob Agents Chemother 2017; 61.
  19. Head BM, Alfa M, Sitar DS, et al. In vitro evaluation of the effect of linezolid and levofloxacin on Bacillus anthracis toxin production, spore formation and cell growth. J Antimicrob Chemother 2017; 72:417.
  20. Hendricks KA, Wright ME, Shadomy SV, et al. Centers for disease control and prevention expert panel meetings on prevention and treatment of anthrax in adults. Emerg Infect Dis 2014; 20.
  21. Linezolid (Zyvox) Product Information. Pharmacia and Upjohn, LLC. New York, NY. 1/2017.
  22. Food and Drug Administration (FDA). FDA Approves Zyvox, The First Antimicrobial Drug In A New Class. In: FDA Talk Paper, Rockville, MD 2000.
  23. Hashemian SMR, Farhadi T, Ganjparvar M. Linezolid: a review of its properties, function, and use in critical care. Drug Des Devel Ther 2018; 12:1759.
  24. Prokocimer P, De Anda C, Fang E, et al. Tedizolid phosphate vs linezolid for treatment of acute bacterial skin and skin structure infections: the ESTABLISH-1 randomized trial. JAMA 2013; 309:559.
  25. Lan SH, Lin WT, Chang SP, et al. Tedizolid Versus Linezolid for the Treatment of Acute Bacterial Skin and Skin Structure Infection: A Systematic Review and Meta-Analysis. Antibiotics (Basel) 2019; 8.
  26. Champney WS, Miller M. Linezolid is a specific inhibitor of 50S ribosomal subunit formation in Staphylococcus aureus cells. Curr Microbiol 2002; 44:350.
  27. Wilson DN, Schluenzen F, Harms JM, et al. The oxazolidinone antibiotics perturb the ribosomal peptidyl-transferase center and effect tRNA positioning. Proc Natl Acad Sci U S A 2008; 105:13339.
  28. Stevens DL, Wallace RJ, Hamilton SM, Bryant AE. Successful treatment of staphylococcal toxic shock syndrome with linezolid: a case report and in vitro evaluation of the production of toxic shock syndrome toxin type 1 in the presence of antibiotics. Clin Infect Dis 2006; 42:729.
  29. Stevens DL, Ma Y, Salmi DB, et al. Impact of antibiotics on expression of virulence-associated exotoxin genes in methicillin-sensitive and methicillin-resistant Staphylococcus aureus. J Infect Dis 2007; 195:202.
  30. Micek ST, Dunne M, Kollef MH. Pleuropulmonary complications of Panton-Valentine leukocidin-positive community-acquired methicillin-resistant Staphylococcus aureus: importance of treatment with antimicrobials inhibiting exotoxin production. Chest 2005; 128:2732.
  31. Mendes RE, Deshpande LM, Jones RN. Linezolid update: stable in vitro activity following more than a decade of clinical use and summary of associated resistance mechanisms. Drug Resist Updat 2014; 17:1.
  32. Besier S, Ludwig A, Zander J, et al. Linezolid resistance in Staphylococcus aureus: gene dosage effect, stability, fitness costs, and cross-resistances. Antimicrob Agents Chemother 2008; 52:1570.
  33. Tsiodras S, Gold HS, Sakoulas G, et al. Linezolid resistance in a clinical isolate of Staphylococcus aureus. Lancet 2001; 358:207.
  34. Morales G, Picazo JJ, Baos E, et al. Resistance to linezolid is mediated by the cfr gene in the first report of an outbreak of linezolid-resistant Staphylococcus aureus. Clin Infect Dis 2010; 50:821.
  35. Sánchez García M, De la Torre MA, Morales G, et al. Clinical outbreak of linezolid-resistant Staphylococcus aureus in an intensive care unit. JAMA 2010; 303:2260.
  36. Bonilla H, Huband MD, Seidel J, et al. Multicity outbreak of linezolid-resistant Staphylococcus epidermidis associated with clonal spread of a cfr-containing strain. Clin Infect Dis 2010; 51:796.
  37. Dobbs TE, Patel M, Waites KB, et al. Nosocomial spread of Enterococcus faecium resistant to vancomycin and linezolid in a tertiary care medical center. J Clin Microbiol 2006; 44:3368.
  38. Bi R, Qin T, Fan W, et al. The emerging problem of linezolid-resistant enterococci. J Glob Antimicrob Resist 2018; 13:11.
  39. Meka VG, Gold HS. Antimicrobial resistance to linezolid. Clin Infect Dis 2004; 39:1010.
  40. Brenciani A, Morroni G, Schwarz S, Giovanetti E. Oxazolidinones: mechanisms of resistance and mobile genetic elements involved. J Antimicrob Chemother 2022; 77:2596.
  41. Long KS, Poehlsgaard J, Kehrenberg C, et al. The Cfr rRNA methyltransferase confers resistance to Phenicols, Lincosamides, Oxazolidinones, Pleuromutilins, and Streptogramin A antibiotics. Antimicrob Agents Chemother 2006; 50:2500.
  42. Toh SM, Xiong L, Arias CA, et al. Acquisition of a natural resistance gene renders a clinical strain of methicillin-resistant Staphylococcus aureus resistant to the synthetic antibiotic linezolid. Mol Microbiol 2007; 64:1506.
  43. Mendes RE, Deshpande LM, Castanheira M, et al. First report of cfr-mediated resistance to linezolid in human staphylococcal clinical isolates recovered in the United States. Antimicrob Agents Chemother 2008; 52:2244.
  44. Arias CA, Torres HA, Singh KV, et al. Failure of daptomycin monotherapy for endocarditis caused by an Enterococcus faecium strain with vancomycin-resistant and vancomycin-susceptible subpopulations and evidence of in vivo loss of the vanA gene cluster. Clin Infect Dis 2007; 45:1343.
  45. Carvalhaes CG, Sader HS, Flamm RK, et al. Assessment of Tedizolid In Vitro Activity and Resistance Mechanisms against a Collection of Enterococcus spp. Causing Invasive Infections, Including Isolates Requiring an Optimized Dosing Strategy for Daptomycin from U.S. and European Medical Centers, 2016 to 2018. Antimicrob Agents Chemother 2020; 64.
  46. Rybak JM, Roberts K. Tedizolid Phosphate: a Next-Generation Oxazolidinone. Infect Dis Ther 2015.
  47. Shaw KJ, Poppe S, Schaadt R, et al. In vitro activity of TR-700, the antibacterial moiety of the prodrug TR-701, against linezolid-resistant strains. Antimicrob Agents Chemother 2008; 52:4442.
  48. Rodríguez-Avial I, Culebras E, Betriu C, et al. In vitro activity of tedizolid (TR-700) against linezolid-resistant staphylococci. J Antimicrob Chemother 2012; 67:167.
  49. Wang Y, Lv Y, Cai J, et al. A novel gene, optrA, that confers transferable resistance to oxazolidinones and phenicols and its presence in Enterococcus faecalis and Enterococcus faecium of human and animal origin. J Antimicrob Chemother 2015; 70:2182.
  50. Egan SA, Shore AC, O'Connell B, et al. Linezolid resistance in Enterococcus faecium and Enterococcus faecalis from hospitalized patients in Ireland: high prevalence of the MDR genes optrA and poxtA in isolates with diverse genetic backgrounds. J Antimicrob Chemother 2020; 75:1704.
  51. Stalker DJ, Jungbluth GL. Clinical pharmacokinetics of linezolid, a novel oxazolidinone antibacterial. Clin Pharmacokinet 2003; 42:1129.
  52. MacGowan AP. Pharmacokinetic and pharmacodynamic profile of linezolid in healthy volunteers and patients with Gram-positive infections. J Antimicrob Chemother 2003; 51 Suppl 2:ii17.
  53. Stein GE, Schooley S, Peloquin CA, et al. Linezolid tissue penetration and serum activity against strains of methicillin-resistant Staphylococcus aureus with reduced vancomycin susceptibility in diabetic patients with foot infections. J Antimicrob Chemother 2007; 60:819.
  54. Thallinger C, Buerger C, Plock N, et al. Effect of severity of sepsis on tissue concentrations of linezolid. J Antimicrob Chemother 2008; 61:173.
  55. Lovering AM, Zhang J, Bannister GC, et al. Penetration of linezolid into bone, fat, muscle and haematoma of patients undergoing routine hip replacement. J Antimicrob Chemother 2002; 50:73.
  56. Myrianthefs P, Markantonis SL, Vlachos K, et al. Serum and cerebrospinal fluid concentrations of linezolid in neurosurgical patients. Antimicrob Agents Chemother 2006; 50:3971.
  57. Bhalodi AA, Papasavas PK, Tishler DS, et al. Pharmacokinetics of intravenous linezolid in moderately to morbidly obese adults. Antimicrob Agents Chemother 2013; 57:1144.
  58. Adembri C, Fallani S, Cassetta MI, et al. Linezolid pharmacokinetic/pharmacodynamic profile in critically ill septic patients: intermittent versus continuous infusion. Int J Antimicrob Agents 2008; 31:122.
  59. Roger C, Roberts JA, Muller L. Clinical Pharmacokinetics and Pharmacodynamics of Oxazolidinones. Clin Pharmacokinet 2018; 57:559.
  60. Sivextro [package insert]. Whitehouse Station, NJ: Merck & Co, Inc; 2015.
  61. Andes D, van Ogtrop ML, Peng J, Craig WA. In vivo pharmacodynamics of a new oxazolidinone (linezolid). Antimicrob Agents Chemother 2002; 46:3484.
  62. Rao GG, Konicki R, Cattaneo D, et al. Therapeutic Drug Monitoring Can Improve Linezolid Dosing Regimens in Current Clinical Practice: A Review of Linezolid Pharmacokinetics and Pharmacodynamics. Ther Drug Monit 2020; 42:83.
  63. Lodise TP, Drusano GL. Use of pharmacokinetic/pharmacodynamic systems analyses to inform dose selection of tedizolid phosphate. Clin Infect Dis 2014; 58 Suppl 1:S28.
  64. Rybak MJ, Cappelletty DM, Moldovan T, et al. Comparative in vitro activities and postantibiotic effects of the oxazolidinone compounds eperezolid (PNU-100592) and linezolid (PNU-100766) versus vancomycin against Staphylococcus aureus, coagulase-negative staphylococci, Enterococcus faecalis, and Enterococcus faecium. Antimicrob Agents Chemother 1998; 42:721.
  65. Li SC, Ye Q, Xu H, et al. Population Pharmacokinetics and Dosing Optimization of Linezolid in Pediatric Patients. Antimicrob Agents Chemother 2019; 63.
  66. Ogami C, Tsuji Y, To H, Yamamoto Y. Pharmacokinetics, toxicity and clinical efficacy of linezolid in Japanese pediatric patients. J Infect Chemother 2019; 25:979.
  67. Thibault C, Kassir N, Goyer I, et al. Population Pharmacokinetics of Intravenous Linezolid in Premature Infants. Pediatr Infect Dis J 2019; 38:82.
  68. Yang M, Zhao L, Wang X, et al. Population Pharmacokinetics and Dosage Optimization of Linezolid in Critically Ill Pediatric Patients. Antimicrob Agents Chemother 2023; 95.
  69. Jungbluth GL, Welshman IR, Hopkins NK. Linezolid pharmacokinetics in pediatric patients: an overview. Pediatr Infect Dis J 2003; 22:S153.
  70. Bradley JS, Flanagan SD, Arrieta AC, et al. Pharmacokinetics, Safety and Tolerability of Single Oral or Intravenous Administration of 200 mg Tedizolid Phosphate in Adolescents. Pediatr Infect Dis J 2016; 35:628.
  71. Cattaneo D, Fusi M, Cozzi V, et al. Supra-therapeutic Linezolid Trough Concentrations in Elderly Patients: A Call for Action? Clin Pharmacokinet 2021; 60:603.
  72. Flanagan SD, Minassian SL, Prokocimer P. Pharmacokinetics, Safety, and Tolerability of Tedizolid Phosphate in Elderly Subjects. Clin Pharmacol Drug Dev 2018; 7:788.
  73. Park AYJ, Wang J, Jayne J, et al. Pharmacokinetics of Tedizolid in Plasma and Sputum of Adults with Cystic Fibrosis. Antimicrob Agents Chemother 2018; 62.
  74. Stein GE, Schooley SL, Peloquin CA, et al. Pharmacokinetics and pharmacodynamics of linezolid in obese patients with cellulitis. Ann Pharmacother 2005; 39:427.
  75. Conradie F, Bagdasaryan TR, Borisov S, et al. Bedaquiline-Pretomanid-Linezolid Regimens for Drug-Resistant Tuberculosis. N Engl J Med 2022; 387:810.
  76. Conradie F, Diacon AH, Ngubane N, et al. Treatment of Highly Drug-Resistant Pulmonary Tuberculosis. N Engl J Med 2020; 382:893.
  77. Esmail A, Oelofse S, Lombard C, et al. An All-Oral 6-Month Regimen for Multidrug-Resistant Tuberculosis: A Multicenter, Randomized Controlled Clinical Trial (the NExT Study). Am J Respir Crit Care Med 2022; 205:1214.
  78. Crass RL, Cojutti PG, Pai MP, Pea F. Reappraisal of Linezolid Dosing in Renal Impairment To Improve Safety. Antimicrob Agents Chemother 2019; 63.
  79. Shi C, Xia J, Ye J, et al. Effect of renal function on the risk of thrombocytopaenia in patients receiving linezolid therapy: A systematic review and meta-analysis. Br J Clin Pharmacol 2022; 88:464.
  80. Kawasuji H, Tsuji Y, Ogami C, et al. Proposal of initial and maintenance dosing regimens with linezolid for renal impairment patients. BMC Pharmacol Toxicol 2021; 22:13.
  81. Cheng CN, Wu CC, Kuo CH, et al. Impact of high plasma concentrations of linezolid in Taiwanese adult patients- therapeutic drug monitoring in improving adverse drug reactions. J Formos Med Assoc 2021; 120:466.
  82. Luque S, Muñoz-Bermudez R, Echeverría-Esnal D, et al. Linezolid Dosing in Patients With Liver Cirrhosis: Standard Dosing Risk Toxicity. Ther Drug Monit 2019; 41:732.
  83. Kühn D, Metz C, Seiler F, et al. Antibiotic therapeutic drug monitoring in intensive care patients treated with different modalities of extracorporeal membrane oxygenation (ECMO) and renal replacement therapy: a prospective, observational single-center study. Crit Care 2020; 24:664.
  84. Nikolos P, Osorio J, Mohrien K, Rose C. Pharmacokinetics of linezolid for methicillin-resistant Staphylococcus aureus pneumonia in an adult receiving extracorporeal membrane oxygenation. Am J Health Syst Pharm 2020; 77:877.
  85. De Rosa FG, Corcione S, Baietto L, et al. Pharmacokinetics of linezolid during extracorporeal membrane oxygenation. Int J Antimicrob Agents 2013; 41:590.
  86. Cojutti P, Pai MP, Pea F. Population Pharmacokinetics and Dosing Considerations for the Use of Linezolid in Overweight and Obese Adult Patients. Clin Pharmacokinet 2018; 57:989.
  87. Blackman AL, Jarugula P, Nicolau DP, et al. Evaluation of Linezolid Pharmacokinetics in Critically Ill Obese Patients with Severe Skin and Soft Tissue Infections. Antimicrob Agents Chemother 2021; 65.
  88. York JA, Adams K, Cullen L, et al. Tedizolid: a service evaluation in a large UK teaching hospital. Eur J Clin Microbiol Infect Dis 2021; 40:397.
  89. Mensa Vendrell M, Tasias Pitarch M, Salavert Lletí M, et al. Safety and Tolerability of More than Six Days of Tedizolid Treatment. Antimicrob Agents Chemother 2020; 64.
  90. Benavent E, Morata L, Escrihuela-Vidal F, et al. Long-Term Use of Tedizolid in Osteoarticular Infections: Benefits among Oxazolidinone Drugs. Antibiotics (Basel) 2021; 10.
  91. US Food and Drug Administration. Zyvox label. Available at: http://www.accessdata.fda.gov/drugsatfda_docs/label/2008/021130s016,021131s013,021132s014lbl.pdf (Accessed January 11, 2010).
  92. Drugs and Lactation Database (LactMed), National Library of Medicine (US), Bethesda (MD) 2006.
  93. Wu VC, Wang YT, Wang CY, et al. High frequency of linezolid-associated thrombocytopenia and anemia among patients with end-stage renal disease. Clin Infect Dis 2006; 42:66.
  94. Senneville E, Legout L, Valette M, et al. Effectiveness and tolerability of prolonged linezolid treatment for chronic osteomyelitis: a retrospective study. Clin Ther 2006; 28:1155.
  95. Palenzuela L, Hahn NM, Nelson RP Jr, et al. Does linezolid cause lactic acidosis by inhibiting mitochondrial protein synthesis? Clin Infect Dis 2005; 40:e113.
  96. Baik SH, Choi YK, Kim HS, et al. A probable case of syndrome of inappropriate antidiuretic hormone secretion associated with linezolid. Am J Health Syst Pharm 2015; 72:1865.
  97. Thwaites G, Nguyen NV. Linezolid for Drug-Resistant Tuberculosis. N Engl J Med 2022; 387:842.
  98. Lee EY, Caffrey AR. Thrombocytopenia with Tedizolid and Linezolid. Antimicrob Agents Chemother 2018; 62.
  99. Gatti M, Fusaroli M, Raschi E, et al. Serious adverse events with tedizolid and linezolid: pharmacovigilance insights through the FDA adverse event reporting system. Expert Opin Drug Saf 2021; 20:1421.
  100. Wunderink RG, Roquilly A, Croce M, et al. A Phase 3, Randomized, Double-Blind Study Comparing Tedizolid Phosphate and Linezolid for Treatment of Ventilated Gram-Positive Hospital-Acquired or Ventilator-Associated Bacterial Pneumonia. Clin Infect Dis 2021; 73:e710.
  101. Gerson SL, Kaplan SL, Bruss JB, et al. Hematologic effects of linezolid: summary of clinical experience. Antimicrob Agents Chemother 2002; 46:2723.
  102. Attassi K, Hershberger E, Alam R, Zervos MJ. Thrombocytopenia associated with linezolid therapy. Clin Infect Dis 2002; 34:695.
  103. Lin YH, Wu VC, Tsai IJ, et al. High frequency of linezolid-associated thrombocytopenia among patients with renal insufficiency. Int J Antimicrob Agents 2006; 28:345.
  104. Mateu de Antonio J, Grau S, Morales-Molina JA, Marín-Casino M. Thrombocytopenia and anemia associated with linezolid in patients with kidney failure. Clin Infect Dis 2006; 42:1500; author reply 1501.
  105. Rao N, Ziran BH, Wagener MM, et al. Similar hematologic effects of long-term linezolid and vancomycin therapy in a prospective observational study of patients with orthopedic infections. Clin Infect Dis 2004; 38:1058.
  106. Bishop E, Melvani S, Howden BP, et al. Good clinical outcomes but high rates of adverse reactions during linezolid therapy for serious infections: a proposed protocol for monitoring therapy in complex patients. Antimicrob Agents Chemother 2006; 50:1599.
  107. Green SL, Maddox JC, Huttenbach ED. Linezolid and reversible myelosuppression. JAMA 2001; 285:1291.
  108. Plachouras D, Giannitsioti E, Athanassia S, et al. No effect of pyridoxine on the incidence of myelosuppression during prolonged linezolid treatment. Clin Infect Dis 2006; 43:e89.
  109. Hardalo C, Lodise TP, De Anda C. Myelosuppression with Oxazolidinones: Are There Differences? Antimicrob Agents Chemother 2019; 63.
  110. Lodise TP, Fang E, Minassian SL, Prokocimer PG. Platelet profile in patients with acute bacterial skin and skin structure infections receiving tedizolid or linezolid: findings from the Phase 3 ESTABLISH clinical trials. Antimicrob Agents Chemother 2014; 58:7198.
  111. Legout L, Senneville E, Gomel JJ, et al. Linezolid-induced neuropathy. Clin Infect Dis 2004; 38:767.
  112. Lee E, Burger S, Shah J, et al. Linezolid-associated toxic optic neuropathy: a report of 2 cases. Clin Infect Dis 2003; 37:1389.
  113. Chao CC, Sun HY, Chang YC, Hsieh ST. Painful neuropathy with skin denervation after prolonged use of linezolid. J Neurol Neurosurg Psychiatry 2008; 79:97.
  114. Bressler AM, Zimmer SM, Gilmore JL, Somani J. Peripheral neuropathy associated with prolonged use of linezolid. Lancet Infect Dis 2004; 4:528.
  115. Rucker JC, Hamilton SR, Bardenstein D, et al. Linezolid-associated toxic optic neuropathy. Neurology 2006; 66:595.
  116. De Vriese AS, Coster RV, Smet J, et al. Linezolid-induced inhibition of mitochondrial protein synthesis. Clin Infect Dis 2006; 42:1111.
  117. Javaheri M, Khurana RN, O'hearn TM, et al. Linezolid-induced optic neuropathy: a mitochondrial disorder? Br J Ophthalmol 2007; 91:111.
  118. Zivkovic SA, Lacomis D. Severe sensory neuropathy associated with long-term linezolid use. Neurology 2005; 64:926.
  119. McKinley SH, Foroozan R. Optic neuropathy associated with linezolid treatment. J Neuroophthalmol 2005; 25:18.
  120. Narita M, Tsuji BT, Yu VL. Linezolid-associated peripheral and optic neuropathy, lactic acidosis, and serotonin syndrome. Pharmacotherapy 2007; 27:1189.
  121. Santini A, Ronchi D, Garbellini M, et al. Linezolid-induced lactic acidosis: the thin line between bacterial and mitochondrial ribosomes. Expert Opin Drug Saf 2017; 16:833.
  122. Wiener M, Guo Y, Patel G, Fries BC. Lactic acidosis after treatment with linezolid. Infection 2007; 35:278.
  123. Apodaca AA, Rakita RM. Linezolid-induced lactic acidosis. N Engl J Med 2003; 348:86.
  124. Mao Y, Dai D, Jin H, Wang Y. The risk factors of linezolid-induced lactic acidosis: A case report and review. Medicine (Baltimore) 2018; 97:e12114.
  125. Sakai Y, Naito T, Arima C, et al. Potential drug interaction between warfarin and linezolid. Intern Med 2015; 54:459.
  126. Antal EJ, Hendershot PE, Batts DH, et al. Linezolid, a novel oxazolidinone antibiotic: assessment of monoamine oxidase inhibition using pressor response to oral tyramine. J Clin Pharmacol 2001; 41:552.
  127. Taylor JJ, Wilson JW, Estes LL. Linezolid and serotonergic drug interactions: a retrospective survey. Clin Infect Dis 2006; 43:180.
  128. Ramsey TD, Lau TT, Ensom MH. Serotonergic and adrenergic drug interactions associated with linezolid: a critical review and practical management approach. Ann Pharmacother 2013; 47:543.
  129. Marcucci C, Sandson NB, Dunlap JA. Linezolid-bupropion interaction as possible etiology of severe intermittent intraoperative hypertension? Anesthesiology 2004; 101:1487.
  130. Lawrence KR, Adra M, Gillman PK. Serotonin toxicity associated with the use of linezolid: a review of postmarketing data. Clin Infect Dis 2006; 42:1578.
  131. Karkow DC, Kauer JF, Ernst EJ. Incidence of Serotonin Syndrome With Combined Use of Linezolid and Serotonin Reuptake Inhibitors Compared With Linezolid Monotherapy. J Clin Psychopharmacol 2017; 37:518.
  132. Butterfield JM, Lawrence KR, Reisman A, et al. Comparison of serotonin toxicity with concomitant use of either linezolid or comparators and serotonergic agents: an analysis of Phase III and IV randomized clinical trial data. J Antimicrob Chemother 2012; 67:494.
  133. Bai AD, McKenna S, Wise H, et al. Association of Linezolid With Risk of Serotonin Syndrome in Patients Receiving Antidepressants. JAMA Netw Open 2022; 5:e2247426.
  134. Gatti M, Raschi E, De Ponti F. Serotonin syndrome by drug interactions with linezolid: clues from pharmacovigilance-pharmacokinetic/pharmacodynamic analysis. Eur J Clin Pharmacol 2021; 77:233.
  135. Kufel WD, Parsels KA, Blaine BE, et al. Real-world evaluation of linezolid-associated serotonin toxicity with and without concurrent serotonergic agents. Int J Antimicrob Agents 2023; 62:106843.
  136. US Food and Drug Administration. FDA Drug Safety Communication: Updated information about the drug interaction between linezolid (Zyvox) and serotonergic psychiatric medications. https://www.fda.gov/drugs/drug-safety-and-availability/fda-drug-safety-communication-updated-information-about-drug-interaction-between-linezolid-zyvox-and (Accessed on October 16, 2020).
  137. Woytowish MR, Maynor LM. Clinical relevance of linezolid-associated serotonin toxicity. Ann Pharmacother 2013; 47:388.
  138. Flanagan S, Bartizal K, Minassian SL, et al. In vitro, in vivo, and clinical studies of tedizolid to assess the potential for peripheral or central monoamine oxidase interactions. Antimicrob Agents Chemother 2013; 57:3060.
  139. Poon YK, La Hoz RM, Hynan LS, et al. Tedizolid vs Linezolid for the Treatment of Nontuberculous Mycobacteria Infections in Solid Organ Transplant Recipients. Open Forum Infect Dis 2021; 8:ofab093.
  140. Kim T, Wills A, Markus A, et al. Safety and Tolerability of Long Term Use of Tedizolid for Treatment of Nontuberculous Mycobacterial Infections. Open Forum Infect Dis 2016; 3:577.
  141. Luque S, Hope W, Sorli L, et al. Dosage Individualization of Linezolid: Precision Dosing of Linezolid To Optimize Efficacy and Minimize Toxicity. Antimicrob Agents Chemother 2021; 65.
  142. Dong HY, Xie J, Chen LH, et al. Therapeutic drug monitoring and receiver operating characteristic curve prediction may reduce the development of linezolid-associated thrombocytopenia in critically ill patients. Eur J Clin Microbiol Infect Dis 2014; 33:1029.
  143. Pea F, Viale P, Cojutti P, et al. Therapeutic drug monitoring may improve safety outcomes of long-term treatment with linezolid in adult patients. J Antimicrob Chemother 2012; 67:2034.
  144. Xu J, Lu J, Yuan Y, et al. Establishment and validation of a risk prediction model incorporating concentrations of linezolid and its metabolite PNU142300 for linezolid-induced thrombocytopenia. J Antimicrob Chemother 2023; 78:1974.
  145. Flanagan S, Passarell J, Lu Q, et al. Tedizolid population pharmacokinetics, exposure response, and target attainment. Antimicrob Agents Chemother 2014; 58:6462.
Topic 129184 Version 14.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟