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

Linezolid: Pediatric drug information

Linezolid: Pediatric drug information
(For additional information see "Linezolid: Drug information" and see "Linezolid: Patient drug information")

For abbreviations, symbols, and age group definitions used in Lexicomp (show table)
Brand Names: US
  • Zyvox
Brand Names: Canada
  • APO-Linezolid;
  • JAMP-Linezolid;
  • SANDOZ Linezolid;
  • Zyvoxam
Therapeutic Category
  • Antibiotic, Oxazolidinone
Dosing: Neonatal

Dosage guidance:

Safety: The risk of serious hematologic and neurologic toxicity increases after >2 weeks and >4 weeks of therapy, respectively. When prolonged therapy is required, alternative agents are preferred; if benefits of linezolid outweigh risks and prolonged therapy is used, monitor closely.

General dosing (Ref):

GA <34 weeks:

PNA ≤7 days: Oral, IV: 10 mg/kg/dose every 12 hours.

PNA >7 days: Oral, IV: 10 mg/kg/dose every 8 hours.

GA ≥34 weeks: Oral, IV: 10 mg/kg/dose every 8 hours.

Enterococcus faecium infection

Enterococcus faecium (vancomycin resistant) infection: Note: The manufacturer recommends a duration of 14 to 28 days; however, shorter or longer durations may be appropriate; duration should be individualized based on source and extent of infection, response, etc.

GA <34 weeks:

PNA <7 days: Oral, IV: 10 mg/kg/dose every 12 hours; may be increased to 10 mg/kg/dose every 8 hours if clinical response is inadequate.

PNA ≥7 days: Oral, IV: 10 mg/kg/dose every 8 hours.

GA ≥34 weeks: Oral, IV: 10 mg/kg/dose every 8 hours. Note: Case reports describe the use of higher doses up to 12 to 15 mg/kg/dose every 8 hours to treat vancomycin-resistant enterococcal endocarditis and meningitis/ventriculitis in former premature neonates (n=2; GA: 26 and 35 weeks; age at treatment: Term) (Ref).

Pneumonia

Pneumonia: Note: The manufacturer suggests treating for 10 to 14 days; however, optimal duration of therapy is unknown; shorter durations may be appropriate.

GA <34 weeks:

PNA <7 days: Oral, IV: 10 mg/kg/dose every 12 hours; may be increased to 10 mg/kg/dose every 8 hours if clinical response is inadequate.

PNA ≥7 days: Oral, IV: 10 mg/kg/dose every 8 hours.

GA ≥34 weeks: Oral, IV: 10 mg/kg/dose every 8 hours.

Skin and soft tissue infection

Skin and soft tissue infection: Note: The manufacturer recommends treatment for 10 to 14 days; however, treatment duration depends on several factors including likely pathogen and extent of infection; shorter durations may be appropriate.

GA <34 weeks:

PNA <7 days: Oral, IV: 10 mg/kg/dose every 12 hours; may be increased to 10 mg/kg/dose every 8 hours if clinical response is inadequate.

PNA ≥7 days: Oral, IV: 10 mg/kg/dose every 8 hours.

GA ≥34 weeks: Oral, IV: 10 mg/kg/dose every 8 hours.

Dosing: Pediatric

Dosage guidance:

Safety: The risk of serious hematologic and neurologic toxicity increases after >2 weeks and >4 weeks of therapy, respectively. When prolonged therapy is required, alternative agents are preferred; if benefits of linezolid outweigh risks and prolonged therapy is used, monitor closely.

General dosing (Ref):

Infants and Children <12 years: Oral, IV: 10 mg/kg/dose every 8 hours, maximum dose: 600 mg/dose.

Children ≥12 years and Adolescents: Oral, IV: 600 mg every 12 hours.

Bloodstream infection, resistant gram-positive infection

Bloodstream infection, resistant gram-positive infection (eg, vancomycin-resistant Enterococcus faecium, methicillin-resistant Staphylococcus aureus):

Note: Duration of therapy varies based on various clinical factors including causative pathogen, source of bacteremia, and response (Ref).

Infants and Children <12 years: Oral, IV: 10 mg/kg/dose every 8 hours; maximum dose: 600 mg/dose (Ref).

Children ≥12 years and Adolescents: Oral, IV: 600 mg every 12 hours OR 10 mg/kg/dose every 12 hours; maximum dose: 600 mg/dose (Ref).

Endocarditis; pathogen-directed therapy for vancomycin-resistant Enterococcus faecium

Endocarditis; pathogen-directed therapy for vancomycin-resistant Enterococcus faecium:

Note: Dosing is from manufacturer labeling for vancomycin-resistant E. faecium infections; guidelines do not provide specific recommendations due to lack of pediatric data. Infectious Diseases consultation is recommended, and prolonged therapy (>4 to 6 weeks) is required; prolonged use of linezolid may be limited by toxicity (Ref).

Infants and Children <12 years: Oral, IV: 10 mg/kg/dose every 8 hours; maximum dose: 600 mg/dose.

Children ≥12 years and Adolescents: Oral, IV: 600 mg every 12 hours.

Enterococcus faecium infection

Enterococcus faecium (vancomycin-resistant) infection: Note: The manufacturer recommends a duration of 14 to 28 days; however, shorter or longer durations may be appropriate; duration should be individualized based on source and extent of infection, response, etc.

Infants and Children <12 years: Oral, IV: 10 mg/kg/dose every 8 hours, maximum dose: 600 mg/dose.

Children ≥12 years and Adolescents: Oral, IV: 600 mg every 12 hours.

Exit-site or tunnel infection, peritoneal dialysis catheter

Exit-site or tunnel infection, peritoneal dialysis catheter (Ref): Note: Treat exit-site infection for ≥2 weeks (≥3 weeks for S. aureus) and at least 7 days after complete resolution; treat tunnel infection for 2 to 4 weeks.

Infants and Children <5 years: Oral: 10 mg/kg/dose every 8 hours; maximum dose: 600 mg/dose.

Children 5 to <12 years: Oral: 10 mg/kg/dose every 12 hours; maximum dose: 600 mg/dose.

Children ≥12 years and Adolescents: Oral: 600 mg every 12 hours.

Meningitis, including health care-associated ventriculitis/meningitis

Meningitis, including health care-associated ventriculitis/meningitis (eg, cerebrospinal fluid shunt infection) (alternative agent): Limited data available:

Note: Duration should be individualized based on patient characteristics and response; typical duration is 10 to 14 days; longer durations required for abscess or empyema (Ref).

Infants and Children <12 years: Oral, IV: 10 mg/kg/dose every 8 hours; maximum dose: 600 mg/dose (Ref).

Children ≥12 years and Adolescents: Oral, IV: 10 mg/kg/dose every 12 hours; maximum dose: 600 mg/dose (Ref).

Nontuberculous mycobacterial infection, pulmonary

Nontuberculous mycobacterial infection, pulmonary (eg, Mycobacterium abscessus): Limited data available:

Patients with cystic fibrosis (Ref): Note: Use as part of an appropriate combination regimen until patient is culture negative on therapy for ≥1 year; use caution in patients chronically coinfected with methicillin-resistant Staphylococcus aureus (MRSA) due to potential for resistance development. Prolonged use of linezolid may be limited by toxicity; guidelines suggest adding high-dose pyridoxine to reduce risk of cytopenias.

Infants and Children <12 years: Oral, IV: 10 mg/kg/dose every 8 hours; maximum dose: 600 mg/dose.

Children ≥12 years and Adolescents: Oral, IV: 10 mg/kg/dose every 12 or 24 hours; maximum dose: 600 mg/dose.

Patients without cystic fibrosis (Ref): Note: Use as part of an appropriate combination regimen until patient is culture negative on therapy for ≥1 year. Prolonged use of linezolid may be limited by toxicity. Dosing in BTS guidelines for infants and children is based on WHO recommendations for drug resistant tuberculosis.

Infants and Children <10 years: Oral, IV: 10 mg/kg/dose every 12 hours; maximum dose: 300 mg/dose.

Children ≥10 years: Oral, IV: 10 mg/kg/dose every 24 hours; maximum dose: 600 mg/dose; doses of 300 mg every 24 hours have also been used.

Adolescents: Oral, IV: 600 mg every 12 or 24 hours; consider reducing to 300 mg every 24 hours if serious adverse effects develop.

Osteoarticular infection, acute

Osteoarticular infection, acute (eg, septic [bacterial] arthritis, osteomyelitis]): Limited data available:

Note: Duration should be individualized based on several factors including causative pathogen, response to therapy, and normalization of inflammatory markers. Minimum total duration for septic arthritis is 2 to 3 weeks and for osteomyelitis is 3 to 4 weeks; longer duration commonly necessary, particularly for infections caused by MRSA. Prolonged use of linezolid may be limited by toxicity (Ref).

Infants and Children <12 years: Oral, IV: 10 mg/kg/dose every 8 hours; maximum dose: 600 mg/dose (Ref).

Children ≥12 years and Adolescents: Oral, IV: 10 mg/kg/dose every 12 hours; maximum dose: 600 mg/dose (Ref).

Peritonitis

Peritonitis (peritoneal dialysis): Note: Recommended treatment duration is 2 to 3 weeks depending on pathogen (Ref):

Infants and Children <5 years: Oral: 10 mg/kg/dose every 8 hours; maximum dose: 600 mg/dose.

Children 5 to <12 years: Oral: 10 mg/kg/dose every 12 hours; maximum dose: 600 mg/dose.

Children ≥12 years and Adolescents: Oral: 600 mg every 12 hours.

Pneumonia

Pneumonia: Note: The usual duration of therapy for uncomplicated, community-acquired pneumonia is 10 days; however, shorter duration may be effective for more mild disease; complicated infection may require longer treatment (Ref). For nosocomial pneumonia (eg, hospital-acquired, ventilator-associated), optimal duration is unknown; the manufacturer recommends 10 to 14 days of treatment, but shorter durations of 7 days are recommended in adults (Ref).

Infants and Children <12 years: Oral, IV: 10 mg/kg/dose every 8 hours; maximum dose: 600 mg/dose.

Children ≥12 years and Adolescents: Oral, IV: 600 mg every 12 hours.

Skin and soft tissue infection

Skin and soft tissue infection:

Uncomplicated: Note: Typical duration is 5 days but may be extended if clinical response is inadequate (Ref).

Infants and Children <5 years: Oral: 10 mg/kg/dose every 8 hours.

Children 5 to 11 years: Oral: 10 mg/kg/dose every 12 hours; maximum dose: 600 mg/dose.

Children ≥12 years and Adolescents: Oral: 600 mg every 12 hours.

Complicated: Note: Typical duration is 7 to 14 days and should be individualized based on clinical response; for necrotizing infection, continue until further debridement is not necessary and the patient has improved clinically, including being afebrile for ≥48 hours (Ref).

Infants and Children <12 years: Oral, IV: 10 mg/kg/dose every 8 hours; maximum dose: 600 mg/dose.

Children ≥12 years and Adolescents: Oral, IV: 600 mg every 12 hours.

Tuberculosis, active; treatment

Tuberculosis, active (drug-resistant); treatment: Limited data available: Note: Use as part of an appropriate combination regimen; duration should be individualized based on extent of disease, rapidity of culture conversion, clinical response, and toxicity; use of linezolid for >6 months is recommended for optimal effectiveness. Monitor closely for hematologic and neurologic toxicity (Ref).

Infants and Children <12 years: Note: Dosing for lower weight ranges is based on pharmacokinetic modeling (Ref).

5 to <10 kg: Oral, IV: 15 mg/kg/dose once daily.

10 to 23 kg: Oral, IV: 12 mg/kg/dose once daily.

>23 kg: Oral, IV: 10 mg/kg/dose once daily; maximum dose: 600 mg/dose.

Children ≥12 years and Adolescents: Oral, IV: 10 mg/kg/dose once daily; maximum dose: 600 mg/dose (Ref).

Dosage adjustment for concomitant therapy: Significant drug interactions exist, requiring dose/frequency adjustment or avoidance. Consult drug interactions database for more information.

Dosing: Kidney Impairment: Pediatric

Altered kidney impairment: Infants, Children, and Adolescents: IV, Oral: Any degree of kidney impairment: No adjustment is recommended. In patients with impaired kidney function, the 2 primary metabolites may accumulate and the risk of thrombocytopenia is increased. Monitor closely; dosage adjustment may be necessary, but specific recommendations are not available; utilize therapeutic drug monitoring and monitor efficacy closely (Ref).

Hemodialysis, intermittent: Dialyzable, parent drug and metabolites (~30% to 57% removed via high-flux dialyzer) (Ref).

Infants, Children, and Adolescents: IV, Oral: Administer dose after dialysis session; the risk of thrombocytopenia is increased in patients with impaired kidney function. Monitor closely; dosage adjustment may be necessary, but specific recommendations are not available; utilize therapeutic drug monitoring and monitor efficacy closely (Ref).

Peritoneal dialysis: Infants, Children, and Adolescents: IV, Oral: The risk of thrombocytopenia is increased in patients with impaired kidney function. Specific recommendations are not available; utilize therapeutic drug monitoring and monitor efficacy closely (Ref).

Continuous renal replacement therapy (CRRT): Note: Drug clearance is dependent on the effluent flow rate, filter type, and method of renal replacement. Flow rates vary widely in pediatric patients. Appropriate dosing requires consideration of drug penetration to site of infection, minimum inhibitory concentration (MIC) of bacteria, and severity of illness. Close monitoring of response and adverse reactions due to drug accumulation (eg, hematologic toxicity) is important. Due to minimal data in pediatric patients receiving CRRT, consider monitoring serum concentrations if available.

Infants, Children, and Adolescents: IV, Oral: Based on adult data, dosage adjustment is not necessary; monitor closely (Ref). Note: While pediatric data are lacking, high variability of linezolid pharmacokinetics has been observed in critically ill adult patients and those on renal replacement therapies; monitor patient closely for efficacy and toxicities, and utilize therapeutic drug monitoring when possible (Ref).

Dosing: Hepatic Impairment: Pediatric

Mild to moderate impairment: No adjustment necessary.

Severe hepatic impairment: There are no dosage adjustments provided in the manufacturer's labeling (has not been studied). Based on experience in adult patients, linezolid concentrations and risk of thrombocytopenia may be increased in patients with cirrhosis; use with caution and monitor closely; consider therapeutic drug monitoring when feasible (Ref).

Dosing: Adult

(For additional information see "Linezolid: Drug information")

Dosage guidance:

Safety: Linezolid is not a preferred agent for the treatment of infections requiring prolonged therapy as the risk of serious hematologic and neurologic toxicity increases after >2 weeks and >4 weeks of therapy, respectively.

Anthrax, systemic infection

Anthrax, systemic infection (off-label use): Note: Consult public health officials for event-specific recommendations.

IV: 600 mg every 12 hours as part of an appropriate combination regimen. The duration for non-CNS infection is ≥2 weeks or until clinically stable, whichever is longer. The duration for meningitis is ≥2 to 3 weeks or until clinically stable, whichever is longer (Ref).

Note: Antitoxin should also be administered. Following the course of IV combination therapy for systemic anthrax infection (including meningitis), patients exposed to aerosolized spores require oral monotherapy with an appropriate agent to complete a total antimicrobial course of 60 days (Ref).

Bloodstream infection

Bloodstream infection:

Empiric therapy or pathogen-directed therapy for methicillin-resistant Staphylococcus aureus (MRSA) (alternative agent) (off-label use): Oral, IV: 600 mg every 12 hours (Ref); treat uncomplicated S. aureus bacteremia for ≥14 days from day of first negative blood culture, with longer courses warranted for endocarditis or metastatic sites of infection (Ref).

Empiric therapy or pathogen-directed therapy for vancomycin-resistant enterococci: Oral, IV: 600 mg every 12 hours; treat uncomplicated bacteremia for 7 to 14 days from day of first negative blood culture, with longer courses warranted for endocarditis or metastatic sites of infection (Ref). Some experts recommend a duration of 5 to 7 days for uncomplicated infection with rapid blood culture clearance (within 24 hours) and in the absence of metastatic infection (Ref).

CNS infection, health care–associated

CNS infection, health care–associated (eg, cerebrospinal fluid shunt infection) (alternative agent) (off-label use): Pathogen-directed therapy for Staphylococcus spp., including methicillin-resistant species or Cutibacterium acnes.

Oral, IV: 600 mg every 12 hours; duration of therapy is generally 10 to 14 days (Ref). For staphylococci, usually used in combination with rifampin (eg, in the setting of retained hardware) (Ref), but conflicting data are available on the effect of rifampin on linezolid levels (Ref).

Cystic fibrosis, acute pulmonary exacerbation, moderate to severe

Cystic fibrosis, acute pulmonary exacerbation, moderate to severe (off-label use): Empiric therapy or pathogen-directed therapy for MRSA (Ref).

Oral, IV: 600 mg every 12 hours (Ref). Duration is usually 10 to 14 days depending on clinical response (Ref).

Diabetic foot infection, moderate to severe

Diabetic foot infection, moderate to severe: Empiric therapy or pathogen-directed therapy for MRSA.

Oral, IV: 600 mg every 12 hours; for empiric therapy, use as part of an appropriate combination regimen. Duration is usually 2 to 4 weeks in the absence of osteomyelitis; prolonged use of linezolid may be limited by toxicity (Ref).

Endocarditis, treatment

Endocarditis, treatment (off-label use):

Enterococcus, penicillin-, aminoglycoside-, and vancomycin-resistant:

IV, Oral: 600 mg every 12 hours for >6 weeks; prolonged use of linezolid may be limited by toxicity (Ref).

S. aureus, methicillin-resistant or penicillin-allergy for methicillin-susceptible, oral step-down therapy in patients who inject drugs:

Note: Not first-line therapy; data are limited. Reserve use for patients who inject drugs who had initial clinical improvement with IV treatment for methicillin-resistant S. aureus or penicillin allergy for methicillin-susceptible S. aureus but cannot complete IV standard of care therapy (Ref).

Oral: 600 mg every 12 hours in combination with rifampin for a total duration, including initial IV therapy, of 6 weeks (Ref).

Intracranial abscess and spinal epidural abscess

Intracranial abscess (brain abscess, intracranial epidural abscess) and spinal epidural abscess (alternative agent) (off-label use): Empiric or pathogen-directed therapy of MRSA.

IV, Oral: 600 mg every 12 hours; when used for empiric therapy, use as part of an appropriate combination regimen. Duration generally ranges from 4 to 8 weeks for brain abscess and spinal epidural abscess, and 6 to 8 weeks for intracranial epidural abscess; prolonged use of linezolid may be limited by toxicity (Ref).

Meningitis, bacterial

Meningitis, bacterial (off-label use): As pathogen-directed therapy (eg, penicillin-, ampicillin-, and vancomycin-resistant enterococci; Staphylococcus spp., including MRSA [alternative agent]; C. acnes [alternative agent]).

IV: 600 mg every 12 hours. Treatment duration is usually 10 to 14 days (Ref).

Osteomyelitis and/or discitis

Osteomyelitis and/or discitis (alternative agent) (off-label use):

Pathogen-directed therapy for Staphylococcus spp., including MRSA: Oral, IV: 600 mg every 12 hours (Ref). Some experts combine with rifampin in the presence of retained hardware (Ref), but conflicting data are available on the effect of rifampin on linezolid levels (Ref).

Pathogen-directed therapy for Enterococcus spp.: Oral, IV: 600 mg every 12 hours (Ref).

Duration of therapy: Duration is generally ≥6 weeks; prolonged use of linezolid may be limited by toxicity. Shorter courses are appropriate if the affected bone is completely resected (eg, by amputation) (Ref).

Pneumonia, as a component of empiric therapy or pathogen-directed therapy for methicillin-resistant S. aureus

Pneumonia, as a component of empiric therapy or pathogen-directed therapy for methicillin-resistant S. aureus:

Oral, IV: 600 mg every 12 hours; duration of therapy varies based on disease severity and response to therapy; treatment is typically given for 7 days. When used for empiric therapy, give as part of an appropriate combination regimen (Ref).

Prosthetic joint infection

Prosthetic joint infection (alternative agent) (off-label use):

Pathogen-directed therapy for Enterococcus spp. (penicillin-susceptible or penicillin-resistant): Oral, IV: 600 mg every 12 hours for 4 to 6 weeks; prolonged use of linezolid may be limited by toxicity (Ref).

Pathogen-directed therapy for Staphylococcus spp., including MRSA: Oral, IV: 600 mg every 12 hours. Duration ranges from 2 to 6 weeks depending on prosthesis management, use of rifampin, and other patient-specific factors; prolonged use of linezolid may be limited by toxicity (Ref).

Note: In select cases (eg, debridement and retention of prosthesis or one-stage arthroplasty), give oral suppressive antibiotic therapy with an appropriate regimen following completion of initial treatment (Ref).

Septic arthritis

Septic arthritis (alternative agent) (off-label use): Pathogen-directed therapy for MRSA.

Oral, IV: 600 mg every 12 hours. Total treatment duration is 3 to 4 weeks (in the absence of osteomyelitis); prolonged use of linezolid may be limited by toxicity (Ref). Some experts recommend 4 weeks of therapy for patients with concomitant bacteremia (Ref).

Skin and soft tissue infection

Skin and soft tissue infection (alternative agent): Empiric therapy or pathogen-directed therapy for resistant gram-positive organisms (eg, MRSA).

Oral, IV: 600 mg every 12 hours. Total duration of therapy is usually 5 to 14 days; for necrotizing infection, continue until further debridement is not necessary and the patient has improved clinically, including being afebrile for ≥48 hours (Ref). Note: For empiric therapy, give as part of an appropriate combination regimen (Ref).

Toxic shock syndrome

Toxic shock syndrome (alternative agent) (off-label use):

Toxin production suppression due to group A streptococci: Oral, IV: 600 mg every 12 hours as part of an appropriate combination regimen. Duration is until clinically and hemodynamically stable for ≥48 to 72 hours; then discontinue linezolid and give monotherapy with an appropriate antistreptococcal agent (Ref).

Empiric therapy or pathogen-directed therapy for S. aureus, including MRSA and toxin production suppression: Oral, IV: 600 mg every 12 hours. Duration for toxin production is until clinically and hemodynamically stable for ≥48 to 72 hours; then discontinue linezolid and give monotherapy with an appropriate antistaphylococcal agent. If linezolid is given alone to complete therapy, the duration is typically 10 to 14 days in the absence of bacteremia (Ref).

Tuberculosis disease, drug-resistant

Tuberculosis disease, drug-resistant (off-label use): Oral, IV: 600 mg once daily as part of an appropriate combination regimen including pyridoxine (Ref); dose reduction to 300 mg once daily or 600 mg 3 to 4 times weekly may be used for certain regimens or for patients who develop toxicity (Ref). Consider serum monitoring to ensure therapeutic concentration (Ref). Note: For patients <50 kg, an initial dose of 450 mg once daily has been suggested (Ref).

Duration of therapy: Individualize based on rapidity of culture conversion, extent of disease, and patient-specific factors, including clinical response and toxicity (Ref).

Urinary tract infection, complicated

Urinary tract infection, complicated (pyelonephritis or urinary tract infection with systemic signs/symptoms) (alternative agent): Note: Reserve for use as a component of empiric therapy or pathogen-directed therapy for resistant gram-positive pathogens (eg, vancomycin-resistant enterococci) (Ref).

Oral, IV: 600 mg every 12 hours; duration generally ranges from 7 to 10 days depending on clinical response (Ref).

Dosage adjustment for concomitant therapy: Significant drug interactions exist, requiring dose/frequency adjustment or avoidance. Consult drug interactions database for more information.

Dosing: Kidney Impairment: Adult

The renal dosing recommendations are based upon the best available evidence and clinical expertise. Senior Editorial Team: Bruce Mueller, PharmD, FCCP, FASN, FNKF; Jason Roberts, PhD, BPharm (Hons), B App Sc, FSHP, FISAC; Michael Heung, MD, MS

Note: Observational studies suggest an increased incidence of thrombocytopenia in patients with kidney impairment (Ref). Use with caution; utilize therapeutic drug monitoring when available and limit duration of use when possible.

Note: Renally adjusted dose recommendations are based on doses of 600 mg every 12 hours.

Altered kidney function:

Oral, IV:

CrCl 40 to <130 mL/minute: No dosage adjustment necessary.

CrCl <40 mL/minute: No dosage adjustment necessary. Based on Monte Carlo simulations, in clinically stable patients with CrCl <30 mL/minute and an anticipated treatment course >10 days, some experts suggest reducing dose to 300 mg twice daily after 72 hours with therapeutic drug monitoring to reduce the risk of thrombocytopenia (Ref).

Augmented renal clearance (measured urinary CrCl ≥130 mL/minute/1.73 m2):

Note: Augmented renal clearance (ARC) is a condition that occurs in certain critically ill patients without organ dysfunction and with normal serum creatinine concentrations. Young patients (<55 years of age) admitted post trauma or major surgery are at highest risk for ARC, as well as those with sepsis, burns, or hematologic malignancies. An 8- to 24-hour measured urinary CrCl is necessary to identify these patients (Ref).

IV: 450 mg every 8 hours. Utilize therapeutic drug monitoring when available (Ref).

Hemodialysis, intermittent (thrice weekly): Dialyzable, parent drug and metabolites (~30% to 57% removed via high-flux dialyzer (Ref).

Oral, IV: No dosage adjustment necessary; when scheduled doses fall on dialysis days, 1 of the twice-daily doses should be administered after the dialysis session (Ref). Based on Monte Carlo simulations, in clinically stable patients with CrCl <30 mL/minute and an anticipated treatment course >10 days, some experts suggest reducing dose to 300 mg twice daily after 72 hours with therapeutic drug monitoring to reduce the risk of thrombocytopenia (Ref). More frequent monitoring of CBC should be considered due to increased risk of myelosuppression (Ref).

Peritoneal dialysis: Likely to be dialyzable (low protein binding, small volume of distribution) (Ref):

Oral, IV: No dosage adjustment necessary (Ref).

Based on Monte Carlo simulations, in clinically stable patients with CrCl <30 mL/minute and an anticipated treatment course >10 days, some experts suggest reducing dose to 300 mg twice daily after 72 hours with therapeutic drug monitoring to reduce the risk of thrombocytopenia (Ref). More frequent monitoring of CBC should be considered due to increased risk of myelosuppression (Ref).

CRRT:

Note: Drug clearance is dependent on the effluent flow rate, filter type, and method of renal replacement. Recommendations are based on high-flux dialyzers and effluent flow rates of 20 to 25 mL/kg/hour (or ~1,500 to 3,000 mL/hour) unless otherwise noted. Appropriate dosing requires consideration of adequate drug concentrations (eg, site of infection). Close monitoring of response and adverse reactions (eg, hematological toxicity) due to drug accumulation is important.

Oral, IV: No dosage adjustment necessary (Ref).

Note: High variability of linezolid pharmacokinetics has been observed in critically ill patients (Ref) and those on renal replacement therapies (Ref). Standard dose may not be sufficient for bacterial infections with high MIC ≥2 mg/L. Consider an alternative agent or utilize higher doses informed by therapeutic drug monitoring in patients at risk of toxicity or treatment failure (Ref).

PIRRT (eg, sustained, low-efficiency diafiltration):

Note: Drug clearance is dependent on the effluent flow rate, filter type, and method of renal replacement. Appropriate dosing requires consideration of adequate drug concentrations (eg, site of infection). Close monitoring of response and adverse reactions (eg, hematologic toxicity) due to drug accumulation is important.

Oral, IV: No dosage adjustment necessary. One of the twice-daily doses should be administered after PIRRT (Ref).

Note: High variability of linezolid pharmacokinetics has been observed in critically ill patients (Ref) and those on renal replacement therapies (Ref). Standard dose may not be sufficient for bacterial infections with MIC ≥2 mg/L; consider an alternative agent or utilize higher doses informed by therapeutic drug monitoring in patients at risk of toxicity or treatment failure (Ref).

Dosing: Hepatic Impairment: Adult

Mild to moderate impairment (Child-Pugh class A or B): No dosage adjustment necessary.

Severe impairment (Child-Pugh class C): There are no dosage adjustments provided in the manufacturer's labeling. Linezolid concentrations may be increased in patients with cirrhosis and risk of thrombocytopenia may be increased. Use with caution and monitor for thrombocytopenia; consider therapeutic drug monitoring when feasible (Ref).

Adverse Reactions (Significant): Considerations
Clostridioides difficile infection

Clostridioides difficile infection has occurred, including Clostridioides-difficile associated diarrhea and Clostridioides difficile colitis.

Onset: Varied; may start on the first day of antibiotic therapy or up to 3 months post-antibiotic (Ref).

Risk factors:

• Antibiotic exposure (highest risk factor) (Ref)

• Type of antibiotic (Ref); linezolid may pose significantly less risk (Ref)

• Long durations in a hospitalization or other health care setting (recent or current) (Ref)

• Older adults (Ref)

• Immunocompromised conditions (Ref)

• A serious underlying condition (Ref)

• GI surgery/manipulation (Ref)

• Antiulcer medications (eg, proton pump inhibitors and H2 blockers) (Ref)

• Chemotherapy (Ref)

Lactic acidosis

Lactic acidosis has been reported with the use of linezolid. Patients who develop recurrent nausea and vomiting, unexplained acidosis, or low bicarbonate levels need immediate evaluation. Overall incidence is relatively low but can be life-threatening (Ref) with one study reporting an overall 6.8% incidence (Ref). Peak lactate levels ranged between 3 and 38 mmol/L (Ref). Most reports have occurred in adults; however, there are also reports in pediatric patients (Ref). Lactic acidosis is generally reversible within 15 days of discontinuation of linezolid; however, some cases may take longer to resolve or result in death (Ref).

Mechanism: Unclear; proposed mechanism is through inhibition of mitochondrial protein synthesis resulting in limited aerobic energy production. This leads to anaerobic glycolysis and lactate generation (Ref).

Onset: Varied; may occur on first day of therapy up to 109 days on therapy (Ref)

Risk factors:

• Age ≥60 years (Ref)

• Prolonged use (≥28 days) (Ref)

• Cmin (trough) >2 mg/L when given for an extended duration (Ref)

Myelosuppression

Myelosuppression (including anemia, leukopenia, pancytopenia, and thrombocytopenia) have been reported with the use of linezolid (Ref). Thrombocytopenia is the most frequently observed blood dyscrasia and has been reported more often in patients with severe kidney impairment and in patients with moderate to severe hepatic impairment. Myelosuppression is typically reversible with discontinuation of therapy (Ref) and typically takes 1 to 2 weeks to recover (Ref).

Mechanism: Unclear; proposed mechanism for linezolid-induced thrombocytopenia may be immune-mediated platelet destruction and for linezolid-induced anemia is likely due to direct bone marrow suppression via inhibition of mitochondrial respiration (Ref).

Onset: Intermediate; typically occurs ≥14 days of therapy (Ref); although some studies have noted thrombocytopenia occurring within 7 days of therapy initiation (Ref).

Risk factors:

• Prolonged use (≥14 days) (Ref)

• Baseline platelet count ≤200 x 109/L (Ref)

• Higher daily per kg dose (Ref)

• Cmin (trough) >2 mg/L when given for an extended duration (Ref)

• Preexisting myelosuppression

• Severe kidney impairment or moderate to severe hepatic impairment

• Concurrent medications that cause bone marrow suppression

• Chronic infection (previous or concurrent antibiotic therapy)

Neuropathy, peripheral and optic

Peripheral neuropathy and optic neuropathy have been reported in both adults and pediatric patients. Symptoms reported related to peripheral neuropathy include pain, numbness, paresthesia, and weakness (Ref). Symptoms related to optic neuropathy include decreased visual acuity, color vision, and sensation of brightness (Ref). Improvement or complete recovery of optic neuropathy typically occurs with discontinuation of therapy; however, complete recovery of peripheral neuropathy may not occur with discontinuation of therapy (Ref).

Mechanism: Unclear; may be associated with mitochondrial toxicity (Ref).

Onset: Intermediate; typically occurs ≥28 days of therapy (Ref); although, there have been some reports of cases occurring before 28 days of therapy (Ref).

Risk factors:

• Prolonged use (≥28 days) (Ref)

• Cmin (trough) >2 mg/L when given for an extended duration (Ref)

Serotonin syndrome

Symptoms of agitation, confusion, hallucinations, hyperreflexia, myoclonus, shivering, and tachycardia may occur. Typically reversible within 48 hours of discontinuation of linezolid and/or serotonergic agents (Ref); however, some reported cases have resulted in death (Ref).

Mechanism: Non–dose-related; reversible, weak nonselective monoamine oxidase inhibitor, resulting in inhibition of serotonin metabolism (Ref).

Onset: Intermediate; range from 1 to 20 days, median of 4 days (Ref)

Risk factors:

• Concurrent use with serotonergic agents (eg, selective serotonin reuptake inhibitors and serotonin norepinephrine reuptake inhibitors) (Ref)

• Concurrent use with agents which reduce linezolid metabolism

• Carcinoid syndrome

Adverse Reactions

The following adverse drug reactions and incidences are derived from product labeling unless otherwise specified.

>10%:

Gastrointestinal: Diarrhea (8% to 11%)

Hematologic & oncologic: Decreased white blood cell count (neonates, infants, and children: 12%; children, adolescents, and adults: ≤2%)

1% to 10%:

Dermatologic: Pruritus (neonates, infants, children, and adolescents: ≤1%; nonapplication site), skin rash (adults: 1% to 2%)

Endocrine & metabolic: Increased lactate dehydrogenase (adults: ≤2%)

Gastrointestinal: Abdominal pain (≤2%), dysgeusia (adults: 1% to 2%), increased serum amylase (neonates, infants, children, and adults: ≤2%), increased serum lipase (adults: 3% to 4%; children and adolescents: <1%), loose stools (neonates, infants, children, and adolescents: 2%), nausea (2% to 7%), oral candidiasis (adults: ≤2%), tongue discoloration (adults: ≤1%), vomiting (2% to 9%)

Genitourinary: Vulvovaginal candidiasis (adults: 1% to 2%)

Hematologic & oncologic: Anemia (neonates, infants, and children: 6%; adults ≤2%) (table 1), decreased neutrophils (neonates, infants, and children: 6%; children, adolescents, and adults: ≤1%), decreased platelet count (adults: ≤10%) (table 2), eosinophilia (neonates, infants, children, and adolescents: ≤2%), thrombocytopenia (neonates, infants, and children: 5%) (table 3)

Linezolid: Adverse Reaction: Anemia

Drug (Linezolid)

Comparator

Population

Dose

Dosage Form

Indication

Number of Patients (Linezolid)

Number of Patients (Comparator)

Comments

6%

7%

Neonates, infants, and children

10 mg/kg every 8 hours

Oral or IV

N/A

215

101

Comparator: Vancomycin

0%

0%

Children and adolescents

Patients 5 through 11 years of age: 10 mg/kg every 12 hours; patients 12 years or older: 600 mg every 12 hours

Oral

Uncomplicated skin and skin structure infections

248

251

Comparator: Cefadroxil

2%

1%

Adults

600 mg every 12 hours

N/A

N/A

1,498

1,464

Comparator: Cefpodoxime proxetil, ceftriaxone, dicloxacillin, oxacillin, or vancomycin

0.4%

0%

Adults

400 mg every 12 hours

Oral

Uncomplicated skin and skin structure infections

548

537

Comparator: Clarithromycin

Linezolid: Adverse Reaction: Decreased Platelet Count

Drug (Linezolid)

Comparator

Population

0.3% to 10%

0.4% to 7%

Adults

Linezolid: Adverse Reaction: Thrombocytopenia

Drug (Linezolid)

Comparator

Population

Dose

Dosage Form

Indication

Number of Patients (Linezolid)

Number of Patients (Comparator)

Comments

5%

2%

Neonates, infants, and children

10 mg/kg every 8 hours

Oral or IV

N/A

215

101

Comparator: Vancomycin

0%

0%

Children and adolescents

Patients 5 through 11 years of age: 10 mg/kg every 12 hours; patients 12 years or older: 600 mg every 12 hours

Oral

Uncomplicated skin and skin structure infections

248

251

Comparator: Cefadroxil

Hepatic: Abnormal hepatic function tests (adults: ≤2%), increased serum alanine aminotransferase (neonates, infants, children, and adults: 2% to 10%), increased serum alkaline phosphatase (adults: ≤4%), increased serum aspartate aminotransferase (adults: 2% to 5%), increased serum bilirubin (neonates, infants, and children: 6%; adults: <1%)

Infection: Fungal infection (adults: ≤2%)

Nervous system: Dizziness (adults: 2% to 3%), headache (children, adolescents, and adults: 6% to 9%; neonates, infants, and children: <1%), vertigo (children and adolescents: 1%)

Renal: Increased blood urea nitrogen (adults: ≤2%), increased serum creatinine (≤2%)

Postmarketing:

Dermatologic: Bullous skin disease (Chen 2020), Stevens-Johnson syndrome, toxic epidermal necrolysis (Ivic 2014)

Endocrine & metabolic: Hypoglycemia (Johannesmeyer 2017), hyponatremia (Ioannou 2018), lactic acidosis (Im 2015, Mao 2018), SIADH (Baik 2015)

Gastrointestinal: Clostridioides difficile-associated diarrhea, hematemesis (Chitsulo 2023), melanoglossia (Braggio 2018), staining of tooth (Santos 2015)

Hematologic & oncologic: Leukopenia (Bayram 2017), pancytopenia (Leader 2018), pure red cell aplasia (Yang 2022), sideroblastic anemia (Willekens 2013)

Hypersensitivity: Anaphylaxis, angioedema (Yang 2012), hypersensitivity angiitis (Kruzer 2018)

Nervous system: Hallucination (Maqsood 2022), peripheral neuropathy (Vishnu 2016), psychosis (Maqsood 2022), seizure (Cholongitas 2009, Shneker 2009), serotonin syndrome (Karkow 2017)

Ophthalmic: Blurred vision (Lee 2003), optic neuropathy (Lee 2018), vision loss (Lee 2003)

Contraindications

Hypersensitivity to linezolid or any component of the formulation; concurrent use or within 2 weeks of MAO inhibitors.

Significant drug interactions exist, requiring dose/frequency adjustment or avoidance. Consult drug interactions database for more information.

Canadian labeling: Additional contraindications (not in US labeling): Unless monitored for potential increases in blood pressure, linezolid should not be administered to patients with uncontrolled hypertension, pheochromocytoma, thyrotoxicosis and/or patients taking any of the following: sympathomimetic agents (eg, pseudoephedrine, phenylpropanolamine), vasopressive agents (eg, epinephrine, norepinephrine), dopaminergic agents (eg, dopamine, dobutamine). Unless carefully observed for signs and/or symptoms of serotonin syndrome, linezolid should not be administered to patients with carcinoid syndrome and/or patients taking any of the following: SSRIs; TCAs; serotonin 5-HT1 receptor agonists (triptans); opioids, including meperidine; or buspirone.

Warnings/Precautions

Concerns related to adverse effects:

• Superinfection: Prolonged use may result in fungal or bacterial superinfection.

Disease-related concerns:

• Carcinoid syndrome: Use with caution and closely monitor for serotonin syndrome in patients with carcinoid syndrome.

• Hepatic impairment: Use with caution in patients with liver cirrhosis; risk of thrombocytopenia may be increased (Luque 2019; Sasaki 2011).

• Hypertension: Use with caution and closely monitor BP in patients with uncontrolled hypertension.

• Hyperthyroidism: Use with caution and closely monitor BP in patients with untreated hyperthyroidism.

• Pheochromocytoma: Use with caution and closely monitor BP in patients with pheochromocytoma.

• Renal impairment: Use with caution; risk of thrombocytopenia may be increased (Hanai 2016).

• Seizure disorder: Seizures have been reported; use with caution in patients with a history of seizures.

Dosage form specific issues:

• Benzyl alcohol and derivatives: Some dosage forms may contain sodium benzoate/benzoic acid; benzoic acid (benzoate) is a metabolite of benzyl alcohol; large amounts of benzyl alcohol (≥99 mg/kg/day) have been associated with a potentially fatal toxicity (“gasping syndrome”) in neonates; the “gasping syndrome” consists of metabolic acidosis, respiratory distress, gasping respirations, CNS dysfunction (including convulsions, intracranial hemorrhage), hypotension, and cardiovascular collapse (AAP ["Inactive" 1997]; CDC 1982); some data suggests that benzoate displaces bilirubin from protein binding sites (Ahlfors 2001); avoid or use dosage forms containing benzyl alcohol derivative with caution in neonates. See manufacturer's labeling.

• Phenylalanine: Some products may contain phenylalanine.

Other warnings/precautions:

• Appropriate use: Unnecessary use may lead to the development of resistance to linezolid; consider alternatives before initiating outpatient treatment.

• Catheter-related bloodstream infections (CRBSI): Linezolid should not be used in the empiric treatment of CRBSI but may be appropriate for targeted therapy (Mermel 2009).

Dosage Forms: US

Excipient information presented when available (limited, particularly for generics); consult specific product labeling. [DSC] = Discontinued product

Solution, Intravenous:

Generic: 600 mg/300 mL (300 mL)

Solution, Intravenous [preservative free]:

Zyvox: 200 mg/100 mL (100 mL); 600 mg/300 mL (300 mL)

Generic: 600 mg/300 mL (300 mL)

Suspension Reconstituted, Oral:

Zyvox: 100 mg/5 mL (150 mL) [contains aspartame, sodium benzoate]

Zyvox: 100 mg/5 mL (150 mL [DSC]) [contains aspartame, sodium benzoate; orange flavor]

Generic: 100 mg/5 mL (150 mL)

Tablet, Oral:

Zyvox: 600 mg

Generic: 600 mg

Generic Equivalent Available: US

Yes

Pricing: US

Solution (Linezolid in Sodium Chloride Intravenous)

600MG/300ML 0.9% (per mL): $0.28

Solution (Linezolid Intravenous)

600MG/300ML (per mL): $0.05 - $0.37

Solution (Zyvox Intravenous)

200 mg/100 mL (per mL): $0.69

600MG/300ML (per mL): $0.13

Suspension (reconstituted) (Linezolid Oral)

100 mg/5 mL (per mL): $2.69 - $6.38

Suspension (reconstituted) (Zyvox Oral)

100 mg/5 mL (per mL): $5.46

Tablets (Linezolid Oral)

600 mg (per each): $181.83 - $288.00

Tablets (Zyvox Oral)

600 mg (per each): $325.21

Disclaimer: A representative AWP (Average Wholesale Price) price or price range is provided as reference price only. A range is provided when more than one manufacturer's AWP price is available and uses the low and high price reported by the manufacturers to determine the range. The pricing data should be used for benchmarking purposes only, and as such should not be used alone to set or adjudicate any prices for reimbursement or purchasing functions or considered to be an exact price for a single product and/or manufacturer. Medi-Span expressly disclaims all warranties of any kind or nature, whether express or implied, and assumes no liability with respect to accuracy of price or price range data published in its solutions. In no event shall Medi-Span be liable for special, indirect, incidental, or consequential damages arising from use of price or price range data. Pricing data is updated monthly.

Dosage Forms: Canada

Excipient information presented when available (limited, particularly for generics); consult specific product labeling. [DSC] = Discontinued product

Solution, Intravenous:

Zyvoxam: 600 mg/300 mL (300 mL)

Generic: 600 mg/300 mL (300 mL)

Suspension Reconstituted, Oral:

Zyvoxam: 100 mg/5 mL (150 mL) [contains aspartame, sodium benzoate]

Tablet, Oral:

Zyvoxam: 600 mg [DSC] [contains alcohol, usp]

Generic: 600 mg

Administration: Pediatric

Oral: Administer with or without food. With reconstituted suspension, gently invert bottle 3 to 5 times before use. Do not shake.

Parenteral: IV: Check infusion bag for minute leaks and solution for particulate matter prior to administration. Administer without further dilution over 30 to 120 minutes. When the same intravenous line is used for sequential infusion of other medications, flush line with D5W, NS, or LR before and after infusing linezolid. The yellow color of the injection may intensify over time without affecting potency.

Administration: Adult

IV: Administer intravenous infusion over 30 to 120 minutes. When the same intravenous line is used for sequential infusion of other medications, flush line with D5W, NS, or LR before and after infusing linezolid. The yellow color of the injection may intensify over time without affecting potency.

Oral: Administer without regard to meals.

Oral suspension: Invert gently to mix prior to administration, do not shake.

Storage/Stability

Infusion: Store at 25°C (77°F). Protect from light and freezing. Keep infusion bags in overwrap until ready for use.

Oral suspension: Store at 25°C (77°F); following reconstitution store at room temperature and use suspension within 21 days. Protect from light.

Tablet: Store at 25°C (77°F). Protect from light and moisture.

Use

Treatment of community-acquired pneumonia, nosocomial pneumonia (eg, hospital-acquired, ventilator-associated), uncomplicated and complicated skin and soft tissue infections, and vancomycin-resistant Enterococcus faecium infections (All indications: FDA approved in all ages). Has also been used for the treatment of bloodstream infections, osteoarticular infections, meningitis, tuberculosis, Mycobacterium abscessus pulmonary infections, and exit-site or tunnel infections and peritonitis in patients with peritoneal dialysis catheters.

Medication Safety Issues
Sound-alike/look-alike issues:

Zyvox may be confused with Zosyn, Zovirax

Metabolism/Transport Effects

Substrate of CYP2J2 (minor), CYP4F2 (minor); Note: Assignment of Major/Minor substrate status based on clinically relevant drug interaction potential; Inhibits Monoamine Oxidase

Drug Interactions

Note: Interacting drugs may not be individually listed below if they are part of a group interaction (eg, individual drugs within “CYP3A4 Inducers [Strong]” are NOT listed). For a complete list of drug interactions by individual drug name and detailed management recommendations, use the Lexicomp drug interactions program by clicking on the “Launch drug interactions program” link above.

5-Aminosalicylic Acid Derivatives: May enhance the myelosuppressive effect of Myelosuppressive Agents. Risk C: Monitor therapy

Agents with Blood Glucose Lowering Effects: Monoamine Oxidase Inhibitors may enhance the hypoglycemic effect of Agents with Blood Glucose Lowering Effects. Risk C: Monitor therapy

Alcohol (Ethyl): May enhance the adverse/toxic effect of Monoamine Oxidase Inhibitors. Risk X: Avoid combination

Alosetron: May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

Amifampridine: Agents With Seizure Threshold Lowering Potential may enhance the neuroexcitatory and/or seizure-potentiating effect of Amifampridine. Risk C: Monitor therapy

Amisulpride (Oral): Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Amisulpride (Oral). Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Amphetamines: Monoamine Oxidase Inhibitors may enhance the hypertensive effect of Amphetamines. While linezolid and tedizolid may interact via this mechanism, management recommendations differ from other monoamine oxidase inhibitors. Refer to monographs specific to those agents for details. Risk X: Avoid combination

Antiemetics (5HT3 Antagonists): May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

Antipsychotic Agents: Serotonergic Agents (High Risk) may enhance the adverse/toxic effect of Antipsychotic Agents. Specifically, serotonergic agents may enhance dopamine blockade, possibly increasing the risk for neuroleptic malignant syndrome. Antipsychotic Agents may enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Risk C: Monitor therapy

Apraclonidine: Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of Apraclonidine. Monoamine Oxidase Inhibitors may increase the serum concentration of Apraclonidine. Risk X: Avoid combination

ARIPiprazole: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of ARIPiprazole. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

ARIPiprazole Lauroxil: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of ARIPiprazole Lauroxil. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Asenapine: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Asenapine. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Atomoxetine: Monoamine Oxidase Inhibitors may enhance the neurotoxic (central) effect of Atomoxetine. Risk X: Avoid combination

Atropine (Ophthalmic): Monoamine Oxidase Inhibitors may enhance the hypertensive effect of Atropine (Ophthalmic). Risk X: Avoid combination

Bacillus clausii: Antibiotics may diminish the therapeutic effect of Bacillus clausii. Management: Bacillus clausii should be taken in between antibiotic doses during concomitant therapy. Risk D: Consider therapy modification

BCG (Intravesical): Antibiotics may diminish the therapeutic effect of BCG (Intravesical). Risk X: Avoid combination

BCG (Intravesical): Myelosuppressive Agents may diminish the therapeutic effect of BCG (Intravesical). Risk X: Avoid combination

BCG Vaccine (Immunization): Antibiotics may diminish the therapeutic effect of BCG Vaccine (Immunization). Risk C: Monitor therapy

Benperidol: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Benperidol. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Benzhydrocodone: May enhance the serotonergic effect of Monoamine Oxidase Inhibitors. This could result in serotonin syndrome. Management: The use of benzhydrocodone is not recommended for patients taking monoamine oxidase inhibitors (MAOIs) or within 14 days of MAOI discontinuation. If coadministration is required, use test doses and frequent titration of small benzhydrocodone. Risk D: Consider therapy modification

Betahistine: Monoamine Oxidase Inhibitors may increase the serum concentration of Betahistine. Risk C: Monitor therapy

Bezafibrate: Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of Bezafibrate. Risk X: Avoid combination

Blonanserin: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Blonanserin. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Brexpiprazole: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Brexpiprazole. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Brimonidine (Ophthalmic): Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of Brimonidine (Ophthalmic). Monoamine Oxidase Inhibitors may increase the serum concentration of Brimonidine (Ophthalmic). Risk C: Monitor therapy

Brimonidine (Topical): Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of Brimonidine (Topical). Monoamine Oxidase Inhibitors may increase the serum concentration of Brimonidine (Topical). Risk C: Monitor therapy

Bromperidol: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Bromperidol. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Buprenorphine: May enhance the adverse/toxic effect of Monoamine Oxidase Inhibitors. Risk X: Avoid combination

BuPROPion: Monoamine Oxidase Inhibitors may enhance the hypertensive effect of BuPROPion. Risk X: Avoid combination

BusPIRone: May enhance the serotonergic effect of Linezolid. This could result in serotonin syndrome. Risk X: Avoid combination

Butorphanol: May enhance the serotonergic effect of Monoamine Oxidase Inhibitors. This could result in serotonin syndrome. Risk X: Avoid combination

CarBAMazepine: May enhance the adverse/toxic effect of Monoamine Oxidase Inhibitors. Management: Do not use carbamazepine during, or within 14 days of discontinuing, treatment with a monoamine oxidase inhibitor. Risk X: Avoid combination

Carbinoxamine: Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of Carbinoxamine. Specifically, the anticholinergic effects of carbinoxamine may be enhanced and prolonged. Risk X: Avoid combination

Cariprazine: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Cariprazine. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Cerebrolysin: May enhance the adverse/toxic effect of Monoamine Oxidase Inhibitors. Risk C: Monitor therapy

Chloramphenicol (Ophthalmic): May enhance the adverse/toxic effect of Myelosuppressive Agents. Risk C: Monitor therapy

Chloramphenicol (Systemic): Myelosuppressive Agents may enhance the myelosuppressive effect of Chloramphenicol (Systemic). Risk X: Avoid combination

Chlorphenesin Carbamate: May enhance the adverse/toxic effect of Monoamine Oxidase Inhibitors. Risk C: Monitor therapy

ChlorproMAZINE: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of ChlorproMAZINE. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Cholera Vaccine: Antibiotics may diminish the therapeutic effect of Cholera Vaccine. Management: Avoid cholera vaccine in patients receiving systemic antibiotics, and within 14 days following the use of oral or parenteral antibiotics. Risk X: Avoid combination

Cladribine: May enhance the myelosuppressive effect of Myelosuppressive Agents. Risk X: Avoid combination

Clothiapine: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Clothiapine. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

CloZAPine: Myelosuppressive Agents may enhance the adverse/toxic effect of CloZAPine. Specifically, the risk for neutropenia may be increased. Risk C: Monitor therapy

CloZAPine: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of CloZAPine. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Codeine: Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of Codeine. Risk X: Avoid combination

COMT Inhibitors: May enhance the adverse/toxic effect of Monoamine Oxidase Inhibitors. Management: Avoid coadministration of COMT inhibitors and nonselective monoamine oxidase inhibitors (MAOIs) (eg, isocarboxazid, phenelzine, tranylcypromine, linezolid, methylene blue) whenever possible. Risk D: Consider therapy modification

Cyclobenzaprine: May enhance the serotonergic effect of Monoamine Oxidase Inhibitors. This could result in serotonin syndrome. Risk X: Avoid combination

CycloSPORINE (Systemic): May increase the serum concentration of Linezolid. Risk C: Monitor therapy

Cyproheptadine: Monoamine Oxidase Inhibitors may enhance the anticholinergic effect of Cyproheptadine. Cyproheptadine may diminish the serotonergic effect of Monoamine Oxidase Inhibitors. Risk X: Avoid combination

Dapoxetine: May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Management: Do not use serotonergic agents (high risk) with dapoxetine or within 7 days of serotonergic agent discontinuation. Do not use dapoxetine within 14 days of monoamine oxidase inhibitor use. Dapoxetine labeling lists this combination as contraindicated. Risk X: Avoid combination

Deferiprone: Myelosuppressive Agents may enhance the neutropenic effect of Deferiprone. Management: Avoid the concomitant use of deferiprone and myelosuppressive agents whenever possible. If this combination cannot be avoided, monitor the absolute neutrophil count more closely. Risk D: Consider therapy modification

Deutetrabenazine: Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of Deutetrabenazine. Risk X: Avoid combination

Dexmethylphenidate: Monoamine Oxidase Inhibitors may enhance the hypertensive effect of Dexmethylphenidate. Risk X: Avoid combination

Dextromethorphan: May enhance the serotonergic effect of Monoamine Oxidase Inhibitors. This may cause serotonin syndrome. Risk X: Avoid combination

Diamorphine: Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of Diamorphine. Risk X: Avoid combination

Diethylpropion: Monoamine Oxidase Inhibitors may enhance the hypertensive effect of Diethylpropion. Risk X: Avoid combination

Dihydrocodeine: May enhance the adverse/toxic effect of Monoamine Oxidase Inhibitors. Management: Consider avoiding use of dihydrocodeine while the patient is taking monoamine oxidase inhibitors (MAOIs) and for 2 weeks after MAOI discontinuation. Risk D: Consider therapy modification

Diphenoxylate: May enhance the hypertensive effect of Monoamine Oxidase Inhibitors. Risk X: Avoid combination

Dipyrone: May enhance the adverse/toxic effect of Myelosuppressive Agents. Specifically, the risk for agranulocytosis and pancytopenia may be increased Risk X: Avoid combination

Domperidone: Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of Domperidone. Monoamine Oxidase Inhibitors may diminish the therapeutic effect of Domperidone. Domperidone may diminish the therapeutic effect of Monoamine Oxidase Inhibitors. Risk C: Monitor therapy

DOPamine: Monoamine Oxidase Inhibitors may enhance the hypertensive effect of DOPamine. Management: Initiate dopamine at no greater than one-tenth (1/10) of the usual dose in patients who are taking (or have taken within the last 2 to 3 weeks) monoamine oxidase inhibitors. Monitor for an exaggerated hypertensive response to dopamine. Risk D: Consider therapy modification

DroPERidol: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of DroPERidol. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Droxidopa: Monoamine Oxidase Inhibitors may enhance the hypertensive effect of Droxidopa. Risk X: Avoid combination

EPINEPHrine (Oral Inhalation): Monoamine Oxidase Inhibitors may enhance the hypertensive effect of EPINEPHrine (Oral Inhalation). Risk X: Avoid combination

Epinephrine (Racemic): Monoamine Oxidase Inhibitors may enhance the hypertensive effect of Epinephrine (Racemic). Risk C: Monitor therapy

Ergot Derivatives: May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

Esketamine: May enhance the hypertensive effect of Monoamine Oxidase Inhibitors. Risk C: Monitor therapy

Fecal Microbiota (Live) (Oral): May diminish the therapeutic effect of Antibiotics. Risk X: Avoid combination

Fecal Microbiota (Live) (Rectal): Antibiotics may diminish the therapeutic effect of Fecal Microbiota (Live) (Rectal). Risk X: Avoid combination

Fenfluramine: May enhance the serotonergic effect of Linezolid. This could result in serotonin syndrome. Risk X: Avoid combination

Fexinidazole: Myelosuppressive Agents may enhance the myelosuppressive effect of Fexinidazole. Risk X: Avoid combination

Flupentixol: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Flupentixol. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

FluPHENAZine: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of FluPHENAZine. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Gepirone: May enhance the serotonergic effect of Linezolid. This could result in serotonin syndrome. Risk X: Avoid combination

Guanethidine: May enhance the adverse/toxic effect of Monoamine Oxidase Inhibitors. Risk X: Avoid combination

Haloperidol: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Haloperidol. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

HYDROcodone: Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of HYDROcodone. HYDROcodone may enhance the serotonergic effect of Monoamine Oxidase Inhibitors. This could result in serotonin syndrome. Management: Consider alternatives to this combination when possible. If coadministration is required, use test doses, titrate small doses frequently, and monitor patients closely for evidence of serotonergic and opioid toxicities. Risk D: Consider therapy modification

HYDROmorphone: Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of HYDROmorphone. Management: Coadministration of hydromorphone and monoamine oxidase (MAO) inhibitors (or within 14 days of stopping an MAO inhibitor) is not recommended. If required, use test doses, titrate small doses frequently, and monitor for CNS and respitatory depression. Risk D: Consider therapy modification

Iloperidone: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Iloperidone. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Immune Checkpoint Inhibitors (Anti-PD-1, -PD-L1, and -CTLA4 Therapies): Antibiotics may diminish the therapeutic effect of Immune Checkpoint Inhibitors (Anti-PD-1, -PD-L1, and -CTLA4 Therapies). Risk C: Monitor therapy

Indoramin: Monoamine Oxidase Inhibitors may enhance the hypotensive effect of Indoramin. Risk X: Avoid combination

Iobenguane Radiopharmaceutical Products: Monoamine Oxidase Inhibitors may diminish the therapeutic effect of Iobenguane Radiopharmaceutical Products. Management: Discontinue all drugs that may inhibit or interfere with catecholamine transport or uptake for at least 5 biological half-lives before iobenguane administration. Do not administer these drugs until at least 7 days after each iobenguane dose. Risk X: Avoid combination

Iohexol: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Iohexol. Specifically, the risk for seizures may be increased. Management: Discontinue agents that may lower the seizure threshold 48 hours prior to intrathecal use of iohexol. Wait at least 24 hours after the procedure to resume such agents. In nonelective procedures, consider use of prophylactic antiseizure drugs. Risk D: Consider therapy modification

Iomeprol: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Iomeprol. Specifically, the risk for seizures may be increased. Management: Discontinue agents that may lower the seizure threshold 48 hours prior to intrathecal use of iomeprol. Wait at least 24 hours after the procedure to resume such agents. In nonelective procedures, consider use of prophylactic antiseizure drugs. Risk D: Consider therapy modification

Iopamidol: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Iopamidol. Specifically, the risk for seizures may be increased. Management: Discontinue agents that may lower the seizure threshold 48 hours prior to intrathecal use of iopamidol. Wait at least 24 hours after the procedure to resume such agents. In nonelective procedures, consider use of prophylactic antiseizure drugs. Risk D: Consider therapy modification

Isometheptene: Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of Isometheptene. Risk X: Avoid combination

Lactobacillus and Estriol: Antibiotics may diminish the therapeutic effect of Lactobacillus and Estriol. Risk C: Monitor therapy

Lasmiditan: May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

Levodopa-Foslevodopa: May enhance the adverse/toxic effect of Monoamine Oxidase Inhibitors. Of particular concern is the development of hypertensive reactions when levodopa is used with nonselective MAOI. Risk X: Avoid combination

Levomethadone: May enhance the adverse/toxic effect of Monoamine Oxidase Inhibitors. Risk X: Avoid combination

Levonordefrin: Monoamine Oxidase Inhibitors may enhance the hypertensive effect of Levonordefrin. Risk X: Avoid combination

Lithium: Linezolid may enhance the serotonergic effect of Lithium. This could result in serotonin syndrome. Management: Consider alternatives to this drug combination. If combined, monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes). Risk D: Consider therapy modification

Lorcaserin (Withdrawn From US Market): May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

Loxapine: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Loxapine. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Lumateperone: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Lumateperone. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Lurasidone: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Lurasidone. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Maprotiline: May enhance the hypertensive effect of Monoamine Oxidase Inhibitors. Risk X: Avoid combination

Meptazinol: Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of Meptazinol. Risk X: Avoid combination

Mequitazine: Monoamine Oxidase Inhibitors may enhance the anticholinergic effect of Mequitazine. Risk X: Avoid combination

Metaxalone: May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

Methadone: May enhance the serotonergic effect of Monoamine Oxidase Inhibitors. This could result in serotonin syndrome. Risk X: Avoid combination

Methotrimeprazine: Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of Methotrimeprazine. Specifically, monoamine oxidase inhibitors may enhance dopamine blockade, possible increasing the risk for neuroleptic malignant syndrome. Methotrimeprazine may enhance the serotonergic effect of Monoamine Oxidase Inhibitors. This could result in serotonin syndrome. Risk X: Avoid combination

Methyldopa: Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of Methyldopa. Risk X: Avoid combination

Methylene Blue: May enhance the serotonergic effect of Linezolid. This could result in serotonin syndrome. Risk X: Avoid combination

Methylphenidate: Monoamine Oxidase Inhibitors may enhance the hypertensive effect of Methylphenidate. Risk X: Avoid combination

Metoclopramide: May enhance the hypertensive effect of Monoamine Oxidase Inhibitors. Risk X: Avoid combination

Mianserin: Monoamine Oxidase Inhibitors may enhance the neurotoxic effect of Mianserin. Risk X: Avoid combination

Mivacurium: Monoamine Oxidase Inhibitors may increase the serum concentration of Mivacurium. Risk C: Monitor therapy

Molindone: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Molindone. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Monoamine Oxidase Inhibitors (Antidepressant): Linezolid may enhance the serotonergic effect of Monoamine Oxidase Inhibitors (Antidepressant). This could result in serotonin syndrome. Risk X: Avoid combination

Monoamine Oxidase Inhibitors (Type B): Linezolid may enhance the serotonergic effect of Monoamine Oxidase Inhibitors (Type B). This could result in serotonin syndrome. Risk X: Avoid combination

Morphine (Systemic): Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of Morphine (Systemic). Risk X: Avoid combination

Nalbuphine: Monoamine Oxidase Inhibitors may enhance the CNS depressant effect of Nalbuphine. Nalbuphine may enhance the hypertensive effect of Monoamine Oxidase Inhibitors. Nalbuphine may enhance the serotonergic effect of Monoamine Oxidase Inhibitors. This could result in serotonin syndrome. Management: Use of nalbuphine is not recommended in patients taking MAOIs, or within 14 days of stopping MAOI therapy. If urgent nalbuphine use is needed, use test doses and frequent titration while monitoring blood pressure, CNS depression, and serotonergic toxicity Risk D: Consider therapy modification

Nefazodone: Linezolid may enhance the serotonergic effect of Nefazodone. This could result in serotonin syndrome. Risk X: Avoid combination

Nefopam: Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of Nefopam. Risk X: Avoid combination

Normethadone: Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of Normethadone. Risk X: Avoid combination

OLANZapine: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of OLANZapine. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Olaparib: Myelosuppressive Agents may enhance the myelosuppressive effect of Olaparib. Risk C: Monitor therapy

Ondansetron: May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

Opicapone: May enhance the adverse/toxic effect of Monoamine Oxidase Inhibitors. Risk X: Avoid combination

Opioid Agonists: May enhance the serotonergic effect of Monoamine Oxidase Inhibitors. This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

Opium: Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of Opium. Risk X: Avoid combination

Oxitriptan: Serotonergic Agents (High Risk) may enhance the serotonergic effect of Oxitriptan. This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

OxyCODONE: May enhance the serotonergic effect of Monoamine Oxidase Inhibitors. This could result in serotonin syndrome. Management: Use of oxycodone is not recommended for patients taking MAOIs or within 14 days of MAOI discontinuation. If combined, use test doses and frequent titration of small doses while monitoring blood pressure, CNS depression, and signs of serotonin syndrome. Risk D: Consider therapy modification

OxyMORphone: May enhance the adverse/toxic effect of Monoamine Oxidase Inhibitors. Risk X: Avoid combination

Ozanimod: May enhance the hypertensive effect of Monoamine Oxidase Inhibitors. Risk X: Avoid combination

Paliperidone: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Paliperidone. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Periciazine: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Periciazine. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Perphenazine: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Perphenazine. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Pheniramine: May enhance the anticholinergic effect of Monoamine Oxidase Inhibitors. Risk X: Avoid combination

Pholcodine: May enhance the serotonergic effect of Monoamine Oxidase Inhibitors. This could result in serotonin syndrome. Risk X: Avoid combination

Pimozide: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Pimozide. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Pipamperone [INT]: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Pipamperone [INT]. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Pizotifen: Monoamine Oxidase Inhibitors may enhance the anticholinergic effect of Pizotifen. Risk X: Avoid combination

Polyethylene Glycol-Electrolyte Solution: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Polyethylene Glycol-Electrolyte Solution. Specifically, the risk of seizure may be increased. Risk C: Monitor therapy

Prochlorperazine: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Prochlorperazine. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Promazine: May enhance the myelosuppressive effect of Myelosuppressive Agents. Risk C: Monitor therapy

Promazine: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Promazine. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

QUEtiapine: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of QUEtiapine. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Ramosetron: May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

Reboxetine: Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of Reboxetine. Risk X: Avoid combination

Remifentanil: Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of Remifentanil. Specifically, the risk for opioid toxicity (eg, respiratory depression) may be increased. Remifentanil may enhance the serotonergic effect of Monoamine Oxidase Inhibitors. This could result in serotonin syndrome. Management: The use of remifentanil is not recommended for patients taking monoamine oxidase inhibitors (MAOIs) or within 14 days of MAOI discontinuation. If coadministration is required, use test doses and titrate small doses of remifentanil frequently. Risk D: Consider therapy modification

Reserpine: Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of Reserpine. Existing MAOI therapy can result in paradoxical effects of added reserpine (e.g., excitation, hypertension). Management: Monoamine oxidase inhibitors (MAOIs) should be avoided or used with great caution in patients who are also receiving reserpine. Monitor closely for paradoxical effects of reserpine (eg, excitation, hypertension). Risk D: Consider therapy modification

RisperiDONE: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of RisperiDONE. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Ropeginterferon Alfa-2b: Myelosuppressive Agents may enhance the myelosuppressive effect of Ropeginterferon Alfa-2b. Management: Avoid coadministration of ropeginterferon alfa-2b and other myelosuppressive agents. If this combination cannot be avoided, monitor patients for excessive myelosuppressive effects. Risk D: Consider therapy modification

Selective Serotonin Reuptake Inhibitors: Linezolid may enhance the serotonergic effect of Selective Serotonin Reuptake Inhibitors. This could result in serotonin syndrome. Risk X: Avoid combination

Serotonergic Non-Opioid CNS Depressants: Linezolid may enhance the serotonergic effect of Serotonergic Non-Opioid CNS Depressants. This could result in serotonin syndrome. Risk X: Avoid combination

Serotonergic Opioids (High Risk): Linezolid may enhance the serotonergic effect of Serotonergic Opioids (High Risk). This could result in serotonin syndrome. Management: Consider alternatives to this drug combination. If combined, monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes). Risk D: Consider therapy modification

Serotonin 5-HT1D Receptor Agonists (Triptans): May enhance the serotonergic effect of Monoamine Oxidase Inhibitors. This could result in serotonin syndrome. Monoamine Oxidase Inhibitors may increase the serum concentration of Serotonin 5-HT1D Receptor Agonists (Triptans). Risk X: Avoid combination

Serotonin/Norepinephrine Reuptake Inhibitors: Linezolid may enhance the serotonergic effect of Serotonin/Norepinephrine Reuptake Inhibitors. This could result in serotonin syndrome. Risk X: Avoid combination

Sertindole: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Sertindole. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Sevoflurane: Monoamine Oxidase Inhibitors may enhance the adverse/toxic effect of Sevoflurane. Specifically, the risk of hemodynamic instability may be increased. Risk C: Monitor therapy

Sodium Phosphates: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Sodium Phosphates. Specifically, the risk of seizure or loss of consciousness may be increased in patients with significant sodium phosphate-induced fluid or electrolyte abnormalities. Risk C: Monitor therapy

Sodium Picosulfate: Antibiotics may diminish the therapeutic effect of Sodium Picosulfate. Management: Consider using an alternative product for bowel cleansing prior to a colonoscopy in patients who have recently used or are concurrently using an antibiotic. Risk D: Consider therapy modification

Solriamfetol: Monoamine Oxidase Inhibitors may enhance the hypertensive effect of Solriamfetol. Risk X: Avoid combination

St John's Wort: May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. St John's Wort may decrease the serum concentration of Serotonergic Agents (High Risk). Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

SUFentanil: May enhance the adverse/toxic effect of Monoamine Oxidase Inhibitors. Specifically, the risk for serotonin syndrome or opioid toxicities (eg, respiratory depression, coma) may be increased. Management: Sufentanil should not be used with monoamine oxidase (MAO) inhibitors (or within 14 days of stopping an MAO inhibitor) due to the potential for serotonin syndrome and/or excessive CNS depression. Risk X: Avoid combination

Sulpiride: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Sulpiride. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Sympathomimetics: Linezolid may enhance the hypertensive effect of Sympathomimetics. Management: Consider initial dose reductions of sympathomimetic agents, and closely monitor for enhanced blood pressure elevations, in patients receiving linezolid. Risk D: Consider therapy modification

Syrian Rue: May enhance the serotonergic effect of Serotonergic Agents (High Risk). This could result in serotonin syndrome. Management: Monitor for signs and symptoms of serotonin syndrome/serotonin toxicity (eg, hyperreflexia, clonus, hyperthermia, diaphoresis, tremor, autonomic instability, mental status changes) when these agents are combined. Risk C: Monitor therapy

Tapentadol: May enhance the adverse/toxic effect of Monoamine Oxidase Inhibitors. Specifically, the additive effects of norepinephrine may lead to adverse cardiovascular effects. Tapentadol may enhance the serotonergic effect of Monoamine Oxidase Inhibitors. This could result in serotonin syndrome. Risk X: Avoid combination

Tetrabenazine: May enhance the adverse/toxic effect of Monoamine Oxidase Inhibitors. Risk X: Avoid combination

Tetrahydrozoline (Nasal): Monoamine Oxidase Inhibitors may enhance the hypertensive effect of Tetrahydrozoline (Nasal). Risk X: Avoid combination

Thioridazine: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Thioridazine. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Thiothixene: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Thiothixene. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Tianeptine: May enhance the adverse/toxic effect of Monoamine Oxidase Inhibitors. Risk X: Avoid combination

Tricyclic Antidepressants: Linezolid may enhance the serotonergic effect of Tricyclic Antidepressants. This could result in serotonin syndrome. Risk X: Avoid combination

Trifluoperazine: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Trifluoperazine. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Tryptophan: Linezolid may enhance the serotonergic effect of Tryptophan. This could result in serotonin syndrome. Risk X: Avoid combination

Typhoid Vaccine: Antibiotics may diminish the therapeutic effect of Typhoid Vaccine. Only the live attenuated Ty21a strain is affected. Management: Avoid use of live attenuated typhoid vaccine (Ty21a) in patients being treated with systemic antibacterial agents. Postpone vaccination until 3 days after cessation of antibiotics and avoid starting antibiotics within 3 days of last vaccine dose. Risk D: Consider therapy modification

Tyrosine: May enhance the hypertensive effect of Monoamine Oxidase Inhibitors. Risk X: Avoid combination

Valbenazine: May enhance the adverse/toxic effect of Monoamine Oxidase Inhibitors. Risk X: Avoid combination

Viloxazine: May enhance the hypertensive effect of Monoamine Oxidase Inhibitors. Risk X: Avoid combination

Ziprasidone: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Ziprasidone. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Zuclopenthixol: Agents With Seizure Threshold Lowering Potential may enhance the adverse/toxic effect of Zuclopenthixol. Specifically, the risk of seizures may be increased. Risk C: Monitor therapy

Food Interactions

Concurrent ingestion of foods rich in tyramine, dopamine, tyrosine, phenylalanine, tryptophan, or caffeine may cause sudden and severe high blood pressure (hypertensive crisis or serotonin syndrome). Beverages containing tyramine (eg, hearty red wine and beer) may increase toxic effects. Management: Avoid tyramine-containing foods (aged or matured cheese, air-dried or cured meats including sausages and salamis; fava or broad bean pods, tap/draft beers, Marmite concentrate, sauerkraut, soy sauce, and other soybean condiments). Food’s freshness is also an important concern; improperly stored or spoiled food can create an environment in which tyramine concentrations may increase. Avoid foods containing dopamine, tyrosine, phenylalanine, tryptophan, or caffeine. Avoid beverages containing tyramine.

Dietary Considerations

Some products may contain sodium and/or phenylalanine. Avoid consuming large amounts of tyramine-containing foods/beverages. Some examples include aged or matured cheese, air-dried or cured meats (including sausages and salami), fava or broad bean pods, tap/draft beers, Marmite concentrate, sauerkraut, soy sauce, and other soybean condiments.

Pregnancy Considerations

Information related to linezolid use during pregnancy is limited (Jaspard 2017; Mercieri 2010). Due to pregnancy-induced physiologic changes, some pharmacokinetic properties of linezolid may be altered (van Kampenhout 2017).

Monitoring Parameters

Weekly CBC, particularly in the following patients: Patients at increased risk for bleeding, patients with preexisting myelosuppression, patients receiving concomitant medications that cause bone marrow suppression, patients receiving >2 weeks of therapy, patients with chronic infection who have received previous or concomitant antibiotic therapy; more frequent monitoring may be warranted in certain scenarios based on clinical judgment (Jones 2015; Norris 2019; manufacturer's labeling). Weekly liver function tests with extended use (Norris 2019). Sodium (routinely in patients at risk of hyponatremia [eg, patients receiving diuretics] and/or syndrome of inappropriate antidiuretic hormone secretion [SIADH]). Periodic serum bicarbonate with extended therapy. Consider monitoring lactate in patients with renal or hepatic dysfunction or who are receiving long-term therapy (Su 2011).

Peripheral sensory and visual function with extended therapy (eg, ≥3 months) or in patients with new-onset neuropathic or visual symptoms, regardless of therapy length (any symptoms of visual change or impairment warrant immediate ophthalmic evaluation) (Nambiar 2011; manufacturer's labeling). Monitor for signs and symptoms of lactic acidosis (eg, recurrent nausea and vomiting, unexplained acidosis, low bicarbonate levels) and signs and symptoms of hyponatremia. Monitor for signs and symptoms of serotonin syndrome or neuroleptic malignant syndrome-like reaction (especially in patients on concomitant serotonergic agents). Monitor for diarrhea.

Reference Range

Linezolid concentration monitoring is not routine, but may be considered in certain scenarios (eg, pathogens with minimum inhibitory concentrations [MICs] ≥2 mg/L, kidney impairment, drug-drug interactions, inadequate response to infection, patients at increased risk for adverse effects) (Cojutti 2015; Li 2019; Rao 2020; Thibault 2019).

Gram-positive bacterial infections: Efficacy targets:

AUC24/MIC: ≥80 to 120 (Li 2019; Rao 2020).

Trough concentration: 2 to 7 mg/L (Cojutti 2015; Rao 2020).

Toxicity:

Hematological toxicity may be associated with Cmin (trough) >7 to 10 mg/L (Abdul-Aziz 2020; Cattaneo 2013; Kawasuji 2021; Pea 2012) or AUC >280 to 350 mg•hour/L (Abdul-Aziz 2020; Pea 2012).

Mitochondrial toxicity (peripheral neuropathy, optic neuropathy, lactic acidosis, myelosuppression) may be associated with Cmin (trough) >2 mg/L when given for an extended duration (Song 2015).

Mechanism of Action

Inhibits bacterial protein synthesis by binding to bacterial 23S ribosomal RNA of the 50S subunit. This prevents the formation of a functional 70S initiation complex that is essential for the bacterial translation process. Linezolid is bacteriostatic against enterococci and staphylococci and bactericidal against most strains of streptococci.

Pharmacokinetics (Adult Data Unless Noted)

Absorption: Rapid and extensive.

Distribution:

Vd:

Neonates, GA <34 weeks:

PNA <7 days: 0.81 L/kg.

PNA >7 days: Median 0.99 L/kg; range: 0.7 to 1.45 L/kg (Thibault 2019).

Neonates, GA ≥34 weeks:

PNA <7 days: 0.78 L/kg.

PNA 7 to ≤28 days: 0.66 L/kg.

Infants <3 months of age: 0.79 L/kg.

Infants ≥3 months and Children ≤11 years of age: 0.69 L/kg.

Children ≥12 years and Adolescents: 0.61 L/kg.

Adults: 0.65 L/kg.

Bone (cancellous):serum ratio, steady state, mean: 40% (range: 16% to 53%) (Rana 2002).

CSF:blood ratio: ~60% to 70% (Myrianthefs 2006; Tsona 2010).

Lung penetration:

Epithelial lining fluid:serum ratio, steady state: ≥100%; varies with time (Boselli 2005; Boselli 2012; Conte 2002; Honeybourne 2003).

Tissue:serum ratio, steady state: ~49% (range: 17% to 132%) (Kempker 2018).

Protein binding: Adults: 31%.

Metabolism: Hepatic via oxidation of the morpholine ring, resulting in two inactive metabolites (aminoethoxyacetic acid, hydroxyethyl glycine); minimally metabolized, may be mediated by cytochrome P450.

Bioavailability: Oral: ~100%.

Half-life elimination:

Neonates, GA <34 weeks:

PNA <7 days: 5.6 hours.

PNA >7 days: Median: 3.44 hours; range: 2.44 to 9.77 hours (Thibault 2019).

Neonates, GA ≥ 34 weeks:

PNA <7 days: 3 hours.

PNA 7 to ≤28 days: 1.5 hours.

Infants <3 months of age: 1.8 hours.

Infants ≥3 months and Children ≤11 years of age: 2.9 hours.

Children ≥12 years and Adolescents: 4.1 hours.

Adults: 4.9 hours.

Time to peak: Adults: Oral: 1 to 2 hours.

Excretion: Urine (~30% of total dose as parent drug, ~50% of total dose as metabolites); two metabolites of linezolid may accumulate in patients with severe renal impairment; feces (~9% of total dose as metabolites).

Nonrenal clearance: Adults: ~65%.

Clearance:

Neonates, GA <34 weeks:

PNA <7 days: 2 mL/minute/kg.

PNA >7 days: Median ~2.8 mL/minute/kg; range: ~1.2 to 5.2 mL/minute/kg (Thibault 2019).

Neonates, GA ≥ 34 weeks:

PNA <7 days: 3.8 mL/minute/kg.

PNA 7 to ≤28 days of age: 5.1 mL/minute/kg.

Infants <3 months of age: 5.4 mL/minute/kg.

Infants ≥3 months and Children ≤11 years of age: 3.8 mL/minute/kg.

Children ≥12 years and Adolescents: 2.1 mL/minute/kg.

Adults: 1.7 mL/minute/kg.

Pharmacokinetics: Additional Considerations (Adult Data Unless Noted)

Altered kidney function: Data are conflicting regarding the contribution of renal dysfunction to the elimination of linezolid. Several studies indicate that linezolid exposure is increased in patients with significant renal dysfunction (Cattaneo 2016; Gervasoni 2015; Ide 2018; Matsumoto 2010; Nukui 2013; Sasaki 2011; Tsuji 2017) although this may be reduced by a compensatory increase in non-renal elimination (Brier 2003; El-Assal 2014). Metabolites A and B accumulate in patients with renal insufficiency; the significance of this accumulation is not known.

Anti-infective considerations:

Parameters associated with efficacy:

Gram-positive bacterial infection: AUC24/minimum inhibitory concentration (MIC), goal: ≥80 to 120; %T > MIC, goal: ≥ ~85% (Abdul-Aziz 2020; Rayner 2003); lower AUC targets may be appropriate for streptococcal infections and higher targets may be needed for bone infection (Andes 2002; Rayner 2003).

Mycobacterium tuberculosis: %T > MIC, goal: 100% (1-log kill, acute phase); %T > MIC, goal: 77% (1-log kill, chronic phase); AUC24/MIC, goal: 1,000 (1-log kill, chronic phase) (Bigelow 2020).

Expected drug exposure in patients with normal renal function:

Cmax (peak) serum concentrations:

600 mg once daily: ~13 mg/L (Kempker 2018).

600 mg twice daily: 18 to 23 mg/L, steady state (Boselli 2005; Myrianthefs 2006; Rana 2002; manufacturer's labeling).

AUC:

Children 2 to 11 years of age: AUC24: ~10 mg/kg 3 times daily: ~240 mg•hour/L (IQR 174 to 395) (Cojutti 2015).

Children 12 years of age and adolescents: AUC24: ~10 mg/kg twice daily: ~300 mg•hour/L (IQR 204 to 655) (Cojutti 2015).

Adults: AUC24: 600 mg twice daily: 154 to 276 mg•hour/L (Boselli 2005; manufacturer's labeling).

Postantibiotic effect:

Methicillin-susceptible S. aureus: ~1 to 3 hours; S. pneumoniae: None (Andes 2002; Chen 2018).

Parameters associated with toxicity:

Hematological toxicity: Cmin (trough) >7 to 10 mg/L (Abdul-Aziz 2020; Cattaneo 2013; Pea 2012); AUC >280 to 350 mg•hour/L (Abdul-Aziz 2020; Pea 2012).

Mitochondrial toxicity (peripheral neuropathy, optic neuropathy, lactic acidosis, myelosuppression): Cmin (trough) >2 mg/L when given for an extended duration (Song 2015).

Brand Names: International
International Brand Names by Country
For country code abbreviations (show table)

  • (AE) United Arab Emirates: Linezolid denk | Linxole | Zenix | Zolid | Zyvox;
  • (AR) Argentina: Linezolid norgreen | Linezolid richet | Litrecan | Zyvox;
  • (AT) Austria: Linezolid accord | Linezolid hcs | Linezolid kabi | Linezolid panpharma | Linezolid pfizer | Linezolid ratiopharm | Linezolid sandoz | Linezolid stada | Zyvoxid;
  • (AU) Australia: Apo-linezolid | Linevox | Linezolid kabi | Linezolid myx | Pharmacor linezolid | Zyvox;
  • (BD) Bangladesh: Arlin | Ezolid | Linexil | Linez | Linlid | Linozid | Linzolid | Zolivox;
  • (BE) Belgium: Gramposimide | Linezolid accord | Linezolid sandoz | Zoligram | Zyvoxid;
  • (BG) Bulgaria: Apel | Linezolid kabi | Linezolid krka | Linezolid polpharma | Linezolid sandoz | Linezolid Tchaikapharma | Pneumolid | Zolinid | Zyvoxid;
  • (BR) Brazil: Adiloz | Linezolida | Lizred | Lynoz | Zyvox;
  • (CH) Switzerland: Linezolid Mylan | Linezolid pfizer | Linezolid sandoz | Zyvoxid;
  • (CL) Chile: Linezet | Linezolida | Litasina | Zyvox;
  • (CN) China: Zyvox;
  • (CO) Colombia: Biozolida | Dilozeld uf | Envyliz | Glenzolid | Kabizolid | Linexa | Linezole | Linoid | Linoted | Liolid | Litasina | Lizolid | Lozyd | Mezolide | Nirzolid | Vozolid | Zolidone | Zyvox | Zyvoxid;
  • (CZ) Czech Republic: Dilizolen | Gramposimide | Lineza | Linezolid accord | Linezolid aurovitas | Linezolid kabi | Linezolid krka | Linezolid Mylan | Linezolid olikla | Linezolid sandoz | Linezolid Teva | Linezolid zentiva | Zoxilid | Zyvoxid;
  • (DE) Germany: Linezolid 1a pharma | Linezolid accord | Linezolid al | Linezolid amarox | Linezolid amneal | Linezolid ascend | Linezolid beta | Linezolid demo | Linezolid denk | Linezolid eberth | Linezolid fresenius kabi | Linezolid glenmark | Linezolid hexal | Linezolid hikma | Linezolid inresa | Linezolid kabi | Linezolid Mylan | Linezolid normon | Linezolid panpharma | Linezolid puren | Linezolid ratiopharm | Linezolid rotexmedica | Linezolid stada | Linezolid tad | Linezolid zentiva | Zyvoxid;
  • (DO) Dominican Republic: Litasina | Zyvox;
  • (EC) Ecuador: Tymbro | Zolidone | Zyvox;
  • (EE) Estonia: Apel | Linezolid accord | Linezolid krka | Linezosel | Zyvoxid;
  • (EG) Egypt: Averozolid | Awazi | Debacozolin | Linez | Linezox | Oxazolid | Respenzo | Vilzolid | Voxazoldin | Zyvox | Zyvoxid;
  • (ES) Spain: Apel | Linezolid accord | Linezolid amneal | Linezolid aurovitas spain | Linezolid b. braun | Linezolid g.e.s. | Linezolid glenmark | Linezolid kabi | Linezolid kern pharma | Linezolid krka | Linezolid Mylan | Linezolid normon | Linezolid sala | Linezolid sandoz | Linezolid Teva | Linezolid teva pharma | Zyvoxid;
  • (ET) Ethiopia: Linezolid denk | Nirzolid | Polinoksid;
  • (FI) Finland: Linezolid accord | Linezolid krka | Linezolid Mylan | Linezolid orion | Linezolid pfizer | Linezolid sandoz | Zolic | Zyvoxid;
  • (FR) France: Grampolid | Linezolide accord | Linezolide arrow | Linezolide eg | Linezolide kabi | Linezolide mylan | Linezolide ohre pharma | Linezolide panpharma | Linezolide sandoz | Synzolid | Zyvoxid;
  • (GB) United Kingdom: Linezolid Dr. Reddy's | Zyvox;
  • (GR) Greece: Antizolid | Linesol | Linezan | Linezolid kabi | Zetalid | Zyvoxid;
  • (HK) Hong Kong: Zyvox;
  • (HR) Croatia: Lynz | Zyvoxid;
  • (HU) Hungary: Anozilad | Dilizolen | Linesan | Linezolid hikma | Linezolid panpharma | Linezolid rotexmedica | Zetalid | Zyvox | Zyvoxid;
  • (ID) Indonesia: Zyvox;
  • (IE) Ireland: Linezolid clonmel | Linezolid krka | Linezolid rowex | Linezolid Teva | Zyvox;
  • (IL) Israel: Zyvox;
  • (IN) India: Adaliz | Alinzo | Alzolid | Anzolid | Cadlizo | Capzolid | Entavar | Genolid | Hospilid | Ibizolid | Inzolid | Kabizolid | Kezolid | Lancure | Lenzobact | Lexilid | Limo | Line | Linegold | Linid | Linokem | Linoplus | Linosept | Linospan | Linowin | Linox | Linozid | Linstar | Lintran | Linwell | Linzest | Linzid | Lizbid | Lizemox | Lizobact | Lizofly | Lizoforce | Lizok | Lizolid | Lizomac | Lizomed | Lizoran | Lizosan | Lnz | Lynomarc | Maxolid | Megazolid | Orlina | Ozolid sr | Pedizo | Plusline | Rholid | Ristor | Silid | Solid | Turbo | Voxylid | Wenloid | Winzolid | Xolid | Zifi turbo 600 | Zodlin;
  • (IT) Italy: Linezolid accord | Linezolid Aurobindo | Linezolid Aurobindo Italia | Linezolid b. braun | Linezolid Dr. Reddy's | Linezolid fresenius kabi | Linezolid kabi | Linezolid krka | Linezolid Mylan | Linezolid sandoz | Linezolid sandoz gmbh | Linezolid Teva | Linezolid teva italia | Zyvoxid;
  • (JO) Jordan: Deteria | Zyvox;
  • (JP) Japan: Linezolid hk | Linezolid sawai | Zyvox;
  • (KE) Kenya: Arlin | Axazolid | Bactolid | Linospa | Linzo | Lizolid | Lizomac | Lizomed | Proliz;
  • (KR) Korea, Republic of: Cinezolid | Hanall linezolid | Jw linezolid | Linezolin | Linezoline | Linoxazol | Lizorid | Zyvox;
  • (KW) Kuwait: Linezolid kabi | Zyvox;
  • (LB) Lebanon: Linospan | Zyvox;
  • (LT) Lithuania: Disenil | Linezolid accord | Linezolid hikma | Linezolid krka | Linezolid Mylan | Pneumolid | Zyvoxid;
  • (LV) Latvia: Apel | Linezolid accord | Zyvox | Zyvoxid;
  • (MX) Mexico: Bacvan | Bogadril | Dilfhoz | Elixim | Furano | Invionezo | Lesinol | Lezopisa | Milbrar | Nezovan | Redultrin | Renyvozon | Rodimix | Tymbro | Yaprinca | Zelyxan | Zyvoxam;
  • (MY) Malaysia: Linezolid sandoz | Zyvox;
  • (NL) Netherlands: Linezolid accord | Linezolid Mylan | Linezolid sandoz | Linezolid Teva | Zyvoxid;
  • (NO) Norway: Linezolid accord | Linezolid fresenius kabi | Linezolid krka | Linezolid orion | Linezolid sandoz | Zyvoxid;
  • (NZ) New Zealand: Linezolid kabi | Zyvox;
  • (PE) Peru: Litasina | Nirzolid | Tymbro | Zyvox;
  • (PH) Philippines: Axazolid | Linelid | Linetero | Linzowel | Loxenil | Zyvox;
  • (PK) Pakistan: Adyzil | Akzolid | Avotrexid | Awazid | Barizold | Cazid | Denzodan | Dilozen | Dinoza | Dyzolid | Ecalas | Ecasil | Enzid | Ezolid | Genzolid | Gt line | Guflinz | Hospilid | Invid | Inzox | J linz | Jazolin | Jython | Komlinz | L zole | Ledopif | Lenozo | Lenzonon | Lenzovel | Leozo | Linez | Linezert | Linlid | Linolid | Linospan | Linowin | Linox | Linwrd | Linze | Linzo | Linzod | Linzodin | Linzogen | Linzol | Linzolet | Linzomed | Linzopearl | Linzovel | Linzy | Linzyle | Lizobact | Lizolid | Lizotek | Maclid | Nezkil | Nezocin | Nezolid | Novozid | Obzitic | Odoinz | Oxalid | Ozlin | Radial | Rekto | Resbact | Vaczid | Volinza | Voxylid | Wellinez | Xolid | Zamlide | Zevoxid | Zody | Zoldap | Zolinf | Zolrest | Zolsyn | Zovipet | Zyspan | Zyxov;
  • (PL) Poland: Anozilad | Dilizolen | Linezolid accord | Linezolid adamed | Linezolid aurovitas | Linezolid kabi | Linezolid polpharma | Linezolid sandoz | Linezolid zentiva | Linzomed | Pneumolid | Polinoksid | Zyvoxid;
  • (PR) Puerto Rico: Zyvox;
  • (PT) Portugal: Apel | Linezolida | Linezolida accord | Linezolida accordpharma | Linezolida aurovitas | Linezolida Farmoz | Linezolida Normon | Linezolida sandoz | Linezolida teva | Linezolida Zentiva | Zyvoxid;
  • (PY) Paraguay: Linezolid eurofarma | Linezolida quimfa | Nirzolid | Zyvox;
  • (QA) Qatar: Lindec | Linezolid (Sandoz) | Zersa | Zyvox;
  • (RO) Romania: Linezolid accord | Linezolid Aurobindo | Linezolid fresenius kabi | Linezolid infomed | Linezolid kabi | Linezolid krka | Linezolid sandoz | Linezolid Teva | Pneumolid | Zyvoxid;
  • (RU) Russian Federation: Amizolid | Bactolin | Infilinez | Linegen | Linezolid akri | Linezolid canon | Linezolid krka | Linezolid Teva | Linezolid vial | Roulin routech | Roulin rowtech | Selezolid | Zenix | Zyvox;
  • (SA) Saudi Arabia: Deteria | Dilizolen | Kinzado | Linezolid kabi | Linid | Zersa | Zillion | Zyvox;
  • (SE) Sweden: Linezolid accord | Linezolid actavis | Linezolid glenmark | Linezolid krka | Linezolid Mylan | Linezolid orion | Linezolid reig jofre | Linezolid sandoz | Linezolid Teva | Zyvoxid;
  • (SG) Singapore: Linezolid sandoz | Zyvox;
  • (SI) Slovenia: Linezolid accord | Linezolid actavis | Linezolid kabi | Linezolid krka | Linezolid sandoz | Zyvoxid;
  • (SK) Slovakia: Apel | Dilizolen | Linezolid accord | Linezolid kabi | Linezolid krka | Linezolid Mylan | Linezolid sandoz | Zyvoxid;
  • (TH) Thailand: Zyvox;
  • (TN) Tunisia: Zyvoxid;
  • (TR) Turkey: Limexid | Linedor | Lineject | Linexolid | Linezone | Linoxid | Lizorex | Lynozid | Multiflex linezosel | Nodizil | Polinoksid | Zizolid | Zyvoxid;
  • (TW) Taiwan: Linetero | Prezolid | Zyvox;
  • (UA) Ukraine: Infuzolid | Laen | Linebiotic | Linelid | Linesolf | Linessa | Linezid | Linezolid alvogen | Linezolid krka | Linezolidin | Linozidum | Lizolid | Lizomac | Nezolid | Zyvox;
  • (UG) Uganda: Lizolid | Lizomac | Zolid;
  • (UY) Uruguay: Linezolid richet | Linezolida | Zyvox;
  • (VE) Venezuela, Bolivarian Republic of: Dizolid | Linezid | Prazolid | Zyvox;
  • (VN) Viet Nam: Inlezone | Lichaunox;
  • (ZA) South Africa: Aklid | Alezid | Elturin | Linezolid cipla | Linezolid fresenius kabi | Linezolid hetero | Linezolid specpharm | Linezolid Teva | Linokem | Linosis | Linoxid | Linozid | Voxilid | Voxwin | Voxxzy | Zenilid | Zyvoxid;
  • (ZW) Zimbabwe: Lizomac | Zolid
  1. Abdul-Aziz MH, Alffenaar JC, Bassetti M, et al; Infection Section of European Society of Intensive Care Medicine (ESICM); Pharmacokinetic/pharmacodynamic and Critically Ill Patient Study Groups of European Society of Clinical Microbiology and Infectious Diseases (ESCMID); Infectious Diseases Group of International Association of Therapeutic Drug Monitoring and Clinical Toxicology (IATDMCT); Infections in the ICU and Sepsis Working Group of International Society of Antimicrobial Chemotherapy (ISAC). Antimicrobial therapeutic drug monitoring in critically ill adult patients: a position paper. Intensive Care Med. 2020;46(6):1127-1153. doi:10.1007/s00134-020-06050-1 [PubMed 32383061]
  2. Agyeman AA, Ofori-Asenso R. Efficacy and safety profile of linezolid in the treatment of multidrug-resistant (MDR) and extensively drug-resistant (XDR) tuberculosis: a systematic review and meta-analysis. Ann Clin Microbiol Antimicrob. 2016;15(1):41. doi:10.1186/s12941-016-0156-y [PubMed 27334498]
  3. Ahlfors CE. Benzyl alcohol, kernicterus, and unbound bilirubin. J Pediatr. 2001;139(2):317-319. [PubMed 11487763]
  4. Alsultan A, Peloquin CA. Therapeutic drug monitoring in the treatment of tuberculosis: an update. Drugs. 2014;74(8):839-854. doi:10.1007/s40265-014-0222-8 [PubMed 24846578]
  5. American Academy of Pediatrics (AAP). In: Kimberlin DW, Barnett ED, Lynfield R, Sawyer MH, eds. Red Book: 2021-2024 Report of the Committee on Infectious Diseases. 32nd ed. American Academy of Pediatrics; 2021.
  6. Anderson PO, Sauberan JB. Modeling drug passage into human milk. Clin Pharmacol Ther. 2016;100(1):42-52. [PubMed 27060684]
  7. Andes D, van Ogtrop ML, Peng J, Craig WA. In vivo pharmacodynamics of a new oxazolidinone (linezolid). Antimicrob Agents Chemother. 2002;46(11):3484-3489. doi:10.1128/aac.46.11.3484-3489.2002 [PubMed 12384354]
  8. Ang JY, Lua JL, Turner DR, et al. Vancomycin-Resistant Enterococcus faecium Endocarditis in a Premature Infant Successfully Treated With Linezolid. Pediatr Infect Dis J. 2003;22(12):1101-1103. [PubMed 14688576]
  9. Appalaraju B, Mathews AA, Bhaskaran AC, Arunachalam P. Breast abscess caused by penicillin resistant Pneumococci. J Family Community Med. 2011;18(2):85-86. [PubMed 21897917]
  10. Baddour LM, Chen AF. Prosthetic joint infection: Treatment. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed May 16, 2022.
  11. Baddour LM, Wilson WR, Bayer AS, et al; American Heart Association Committee on Rheumatic Fever, Endocarditis, and Kawasaki Disease of the Council on Cardiovascular Disease in the Young, Council on Clinical Cardiology, Council on Cardiovascular Surgery and Anesthesia, and Stroke Council. Infective endocarditis in adults: diagnosis, antimicrobial therapy, and management of complications: a scientific statement for healthcare professionals from the American Heart Association. Circulation. 2015;132(15):1435-1486. [PubMed 26373316]
  12. Baddour LM, Weimer MB, Wurcel AG, et al; American Heart Association Rheumatic Fever, Endocarditis and Kawasaki Disease Committee of the Council on Lifelong Congenital Heart Disease and Heart Health in the Young; Council on Cardiovascular Surgery and Anesthesia; Council on Cardiovascular and Stroke Nursing; Council on Clinical Cardiology; and Council on Peripheral Vascular Disease. Management of infective endocarditis in people who inject drugs: a scientific statement from the American Heart Association. Circulation. 2022a;146(14):e187-e201. doi:10.1161/CIR.0000000000001090 [PubMed 36043414]
  13. Baltimore RS, Gewitz M, Baddour LM, et al; American Heart Association Rheumatic Fever, Endocarditis, and Kawasaki Disease Committee of the Council on Cardiovascular Disease in the Young and the Council on Cardiovascular and Stroke Nursing. Infective endocarditis in childhood: 2015 update: a scientific statement from the American Heart Association. Circulation. 2015;132(15):1487-1515. doi:10.1161/CIR.0000000000000298 [PubMed 26373317]
  14. Baik SH, Choi YK, Kim HS, Yoon YK, Sohn JW, Kim MJ. A probable case of syndrome of inappropriate antidiuretic hormone secretion associated with linezolid. Am J Health Syst Pharm. 2015;72(21):1865-1869. doi:10.2146/ajhp150208 [PubMed 26490820]
  15. Barrasa H, Soraluce A, Isla A, et al. Pharmacokinetics of linezolid in critically ill patients on continuous renal replacement therapy: influence of residual renal function on PK/PD target attainment. J Crit Care. 2019;50:69-76. doi:10.1016/j.jcrc.2018.11.016 [PubMed 30496913]
  16. Bayram N, Düzgöl M, Kara A, Özdemir FM, Devrim İ. Linezolid-related adverse effects in clinical practice in children. Arch Argent Pediatr. 2017;115(5):470-475. English, Spanish. doi:10.5546/aap.2017.eng.470 [PubMed 28895694]
  17. Berbari EF, Kanj SS, Kowalski TJ, et al; Infectious Diseases Society of America. 2015 Infectious Diseases Society of America (IDSA) clinical practice guidelines for the diagnosis and treatment of native vertebral osteomyelitis in adults. Clin Infect Dis. 2015;61(6):e26-e46. doi:10.1093/cid/civ482 [PubMed 26229122]
  18. Bernard L, Stern R, Lew D, Hoffmeyer P. Serotonin syndrome after concomitant treatment with linezolid and citalopram. Clin Infect Dis. 2003;36(9):1197. doi:10.1086/374558
  19. Bernstein WB, Trotta RF, Rector JT, Tjaden JA, Barile AJ. Mechanisms for linezolid-induced anemia and thrombocytopenia. Ann Pharmacother. 2003;37(4):517-520. doi:10.1345/aph.1C361 [PubMed 12659607]
  20. Bhalodi AA, Papasavas PK, Tishler DS, Nicolau DP, Kuti JL. Pharmacokinetics of intravenous linezolid in moderately to morbidly obese adults. Antimicrob Agents Chemother. 2013;57(3):1144-1149. doi:10.1128/AAC.01453-12 [PubMed 23254421]
  21. Bigelow KM, Deitchman AN, Li SY, et al. Pharmacodynamic correlates of linezolid activity and toxicity in murine models of tuberculosis. J Infect Dis. Published online January 29, 2020. doi:10.1093/infdis/jiaa016 [PubMed 31993638]
  22. Bilbao-Meseguer I, Rodríguez-Gascón A, Barrasa H, Isla A, Solinís MÁ. Augmented renal clearance in critically ill patients: a systematic review. Clin Pharmacokinet. 2018;57(9):1107-1121. doi:10.1007/s40262-018-0636-7 [PubMed 29441476]
  23. Birmingham MC, Rayner CR, Meagher AK, Flavin SM, Batts DH, Schentag JJ. Linezolid for the treatment of multidrug-resistant, gram-positive infections: experience from a compassionate-use program. Clin Infect Dis. 2003;36(2):159-168. doi:10.1086/345744 [PubMed 12522747]
  24. Bishop E, Melvani S, Howden BP, Charles PG, Grayson ML. 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(4):1599-1602. doi:10.1128/AAC.50.4.1599-1602.2006 [PubMed 16569895]
  25. 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(2):e01619-20. doi:10.1128/AAC.01619-20 [PubMed 33257446]
  26. Blassmann U, Roehr AC, Frey OR, et al. Decreased linezolid serum concentrations in three critically ill patients: clinical case studies of a potential drug interaction between linezolid and rifampicin. Pharmacology. 2016;98(1-2):51-55. doi:10.1159/000445194 [PubMed 27046487]
  27. Boak LM, Li J, Spelman D, du Cros P, Nation RL, Rayner CR. Successful treatment and cerebrospinal fluid penetration of oral linezolid in a patient with coagulase-negative Staphylococcus ventriculitis. Ann Pharmacother. 2006;40(7-8):1451-1455. doi:10.1345/aph.1H029 [PubMed 16772405]
  28. Bodilsen J, Brouwer MC, Nielsen H, Van De Beek D. Anti-infective treatment of brain abscess. Expert Rev Anti Infect Ther. 2018;16(7):565-578. doi:10.1080/14787210.2018.1489722 [PubMed 29909695]
  29. Bolhuis MS, Tiberi S, Sotgiu G, et al. Is there still room for therapeutic drug monitoring of linezolid in patients with tuberculosis? Eur Respir J. 2016;47(4):1288-1290. doi:10.1183/13993003.02185-2015 [PubMed 27037319]
  30. Boselli E, Breilh D, Caillault-Sergent A, et al. Alveolar diffusion and pharmacokinetics of linezolid administered in continuous infusion to critically ill patients with ventilator-associated pneumonia. J Antimicrob Chemother. 2012;67(5):1207-1210. doi:10.1093/jac/dks022 [PubMed 22351682]
  31. Boselli E, Breilh D, Rimmelé T, et al. Pharmacokinetics and intrapulmonary concentrations of linezolid administered to critically ill patients with ventilator-associated pneumonia. Crit Care Med. 2005;33(7):1529-1533. doi:10.1097/01.ccm.0000168206.59873.80 [PubMed 16003058]
  32. Bradley JS, Byington CL, Shah SS, et al. The Management of Community-Acquired Pneumonia in Infants and Children Older Than 3 Months of Age: Clinical Practice Guidelines by the Pediatric Infectious Diseases Society and the Infectious Diseases Society of America. Clin Infect Dis. 2011;53(7):e25-e76. [PubMed 21880587]
  33. Bradley JS, Nelson JD, Barnett ED, et al, eds. Nelson's Pediatric Microbial Therapy. 27th ed. American Academy of Pediatrics; 2021.
  34. Braggio C, Bocchialini G, Ventura L, Carbognani P, Rusca M, Ampollini L. Linezolid-induced black hairy tongue. Acta Biomed. 2018;89(3):408-410. doi:10.23750/abm.v89i3.7060 [PubMed 30333468]
  35. Bressler AM, Zimmer SM, Gilmore JL, Somani J. Peripheral neuropathy associated with prolonged use of linezolid. Lancet Infect Dis. 2004;4(8):528-531. doi:10.1016/S1473-3099(04)01109-0 [PubMed 15288827]
  36. Brier ME, Stalker DJ, Aronoff GR, et al. Pharmacokinetics of linezolid in subjects with renal dysfunction. Antimicrob Agents Chemother. 2003;47(9):2775-2780. [PubMed 12936973]
  37. Brown KA, Khanafer N, Daneman N, Fisman DN. Meta-analysis of antibiotics and the risk of community-associated Clostridium difficile infection. Antimicrob Agents Chemother. 2013;57(5):2326-2332. doi:10.1128/AAC.02176-12 [PubMed 23478961]
  38. Brown P, Battistella M. Principles of drug dosing in sustained low efficiency dialysis (SLED) and review of antimicrobial dosing literature. Pharmacy (Basel). 2020;8(1):33. doi:10.3390/pharmacy8010033 [PubMed 32182835]
  39. Caminero JA, Piubello A, Scardigli A, Migliori GB. Proposal for a standardised treatment regimen to manage pre- and extensively drug-resistant tuberculosis cases. Eur Respir J. 2017;50(1):1700648. doi:10.1183/13993003.00648-2017 [PubMed 28679614]
  40. Castro P, Soriano A, Escrich C, Villalba G, Sarasa M, Mensa J. Linezolid treatment of ventriculoperitoneal shunt infection without implant removal. Eur J Clin Microbiol Infect Dis. 2005;24(9):603-606. doi:10.1007/s10096-005-0015-9 [PubMed 16187055]
  41. Cattaneo D, Gervasoni C, Cozzi V, Castoldi S, Baldelli S, Clementi E. Therapeutic drug management of linezolid: a missed opportunity for clinicians? Int J Antimicrob Agents. 2016;48(6):728-731. doi:10.1016/j.ijantimicag.2016.08.023 [PubMed 27769709]
  42. Cattaneo D, Orlando G, Cozzi V, et al. Linezolid plasma concentrations and occurrence of drug-related haematological toxicity in patients with gram-positive infections. Int J Antimicrob Agents. 2013;41(6):586-589. doi:10.1016/j.ijantimicag.2013.02.020 [PubMed 23562639]
  43. Centers for Disease Control and Prevention (CDC). FAQs for clinicians about C. diff. March 27, 2020. Accessed May 4, 2020. https://www.cdc.gov/cdiff/clinicians/faq.html.
  44. Centers for Disease Control and Prevention (CDC). Neonatal deaths associated with use of benzyl alcohol—United States. MMWR Morb Mortal Wkly Rep. 1982;31(22):290-291. http://www.cdc.gov/mmwr/preview/mmwrhtml/00001109.htm. [PubMed 6810084]
  45. Chen C, Zhang M, Chen H, Fang J. Subtherapeutic linezolid concentration in a patient with bullous pemphigoid complicated by methicillin-resistant Staphylococcus aureus infection: A case study. Ther Drug Monit. 2020;42(4):515-517. doi:10.1097/FTD.0000000000000758 [PubMed 32217880]
  46. Chen H, Li L, Liu Y, et al. In vitro activity and post-antibiotic effects of linezolid in combination with fosfomycin against clinical isolates of Staphylococcus aureus. Infect Drug Resist. 2018;11:2107-2115. doi:10.2147/IDR.S175978 [PubMed 30464553]
  47. Chitsulo S, Omotayo D, Mbendera K, Sinyiza FW, Chisale M, Mbakaya BC. Linezolid-induced hematemesis, a rare and life-threatening adverse reaction. A case report of Karonga district in Malawi. Oxf Med Case Reports. 2023;2023(3):omad020. doi:10.1093/omcr/omad020 [PubMed 36993836]
  48. Chmiel JF, Aksamit TR, Chotirmall SH, et al. Antibiotic management of lung infections in cystic fibrosis. I. The microbiome, methicillin-resistant Staphylococcus aureus, gram-negative bacteria, and multiple infections. Ann Am Thorac Soc. 2014;11(7):1120-1129. doi:10.1513/AnnalsATS.201402-050AS [PubMed 25102221]
  49. Choi GW, Lee JY, Chang MJ, et al. Risk factors for linezolid-induced thrombocytopenia in patients without haemato-oncologic diseases. Basic Clin Pharmacol Toxicol. 2019;124(2):228-234. doi:10.1111/bcpt.13123 [PubMed 30171804]
  50. Cholongitas E, Karatzi C, Spyrou S, Georgousaki C, Dasenaki M. Linezolid-induced complex partial seizures in a patient with epilepsy. Scand J Infect Dis. 2009;41(6-7):540-541. doi:10.1080/00365540902896087 [PubMed 19382002]
  51. Chu VC. Staphylococcal toxic shock syndrome. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed March 23, 2021.
  52. Clark DB, Andrus MR, Byrd DC. Drug interactions between linezolid and selective serotonin reuptake inhibitors: case report involving sertraline and review of the literature. Pharmacotherapy. 2006;26(2):269-276. doi:10.1592/phco.26.2.269 [PubMed 16466332]
  53. Cojutti P, Maximova N, Crichiutti G, Isola M, Pea F. Pharmacokinetic/pharmacodynamic evaluation of linezolid in hospitalized paediatric patients: a step toward dose optimization by means of therapeutic drug monitoring and Monte Carlo simulation. J Antimicrob Chemother. 2015;70(1):198-206. doi:10.1093/jac/dku337 [PubMed 25182066]
  54. 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(8):989-1000. doi:10.1007/s40262-017-0606-5 [PubMed 29080937]
  55. Collins CE, Ayturk MD, Flahive JM, Emhoff TA, Anderson FA Jr, Santry HP. Epidemiology and outcomes of community-acquired Clostridium difficile infections in Medicare beneficiaries. J Am Coll Surg. 2014;218(6):1141-1147.e1. doi:10.1016/j.jamcollsurg.2014.01.053 [PubMed 24755188]
  56. Conradie F, Bagdasaryan TR, Borisov S, et al; ZeNix Trial Team. Bedaquiline-pretomanid-linezolid regimens for drug-resistant tuberculosis. N Engl J Med. 2022;387(9):810-823. doi:10.1056/NEJMoa2119430 [PubMed 36053506]
  57. Conradie F, Diacon AH, Ngubane N, et al; Nix-TB Trial Team. Treatment of highly drug-resistant pulmonary tuberculosis. N Engl J Med. 2020;382(10):893-902. doi:10.1056/NEJMoa1901814 [PubMed 32130813]
  58. Conte JE Jr, Golden JA, Kipps J, Zurlinden E. Intrapulmonary pharmacokinetics of linezolid. Antimicrob Agents Chemother. 2002;46(5):1475-1480. doi:10.1128/aac.46.5.1475-1480.2002 [PubMed 11959585]
  59. Crass RL, Cojutti PG, Pai MP, Pea F. Reappraisal of linezolid dosing in renal impairment to improve safety. Antimicrob Agents Chemother. 2019;63(8):e00605-19. doi:10.1128/AAC.00605-19 [PubMed 31109977]
  60. Cremaschi E, Maggiore U, Maccari C, Cademartiri C, Andreoli R, Fiaccadori E. Linezolid levels in a patient with biliary tract sepsis, severe hepatic failure and acute kidney injury on sustained low-efficiency dialysis (SLED). Minerva Anestesiol. 2010;76(11):961-964. [PubMed 21102392]
  61. Dai Y, Wang Y, Zeng Y, Zhang C, Zhou Z, Shi D. Linezolid and the risk of lactic acidosis: Data mining and analysis of the FDA Adverse Event Reporting System. J Clin Pharm Ther. 2020;45(6):1422-1426. doi:10.1111/jcpt.13245 [PubMed 32776380]
  62. Deshpande A, Pasupuleti V, Thota P, et al. Community-associated Clostridium difficile infection and antibiotics: a meta-analysis. J Antimicrob Chemother. 2013;68(9):1951-1961. doi:10.1093/jac/dkt129 [PubMed 23620467]
  63. Donaldson N, Sanders J, Child J, Parker S. Acute hematogenous bacterial osteoarticular infections in children. Pediatr Rev. 2020;41(3):120-136. doi:10.1542/pir.2018-0201 [PubMed 32123023]
  64. Dong H, Wang X, Dong Y, et al. Clinical pharmacokinetic/pharmacodynamic profile of linezolid in severely ill intensive care unit patients. Int J Antimicrob Agents. 2011;38(4):296-300. doi:10.1016/j.ijantimicag.2011.05.007 [PubMed 21741222]
  65. Eimer J, Fréchet-Jachym M, Le Dû D, et al L; LZDM group. Association between increased linezolid plasma concentrations and the development of severe toxicity in multidrug-resistant tuberculosis treatment. Clin Infect Dis. 2023;76(3):e947-e956. doi:10.1093/cid/ciac485 [PubMed 35717636]
  66. El-Assal MI, Helmy SA. Single-dose linezolid pharmacokinetics in critically ill patients with impaired renal function especially chronic hemodialysis patients. Biopharm Drug Dispos. 2014;35(7):405-416. doi:10.1002/bdd.1910 [PubMed 25044219]
  67. Expert opinion. Senior Obesity Editorial Team: Jeffrey F. Barletta, PharmD, FCCM; Manjunath P. Pai, PharmD, FCP; Jason A. Roberts, PhD, BPharm (Hons), B App Sc, FSHP, FISAC.
  68. Expert opinion. Senior Renal Editorial Team: Bruce Mueller, PharmD, FCCP, FASN, FNKF; Jason A. Roberts, PhD, BPharm (Hons), B App Sc, FSHP, FISAC; Michael Heung, MD, MS.
  69. Fiaccadori E, Maggiore U, Rotelli C, et al. Does haemodialysis significantly affect serum linezolid concentrations in critically ill patients with renal failure? A pilot investigation. Nephrol Dial Transplant. 2006;21(5):1402-1406. doi:10.1093/ndt/gfl048 [PubMed 16504979]
  70. Fiaccadori E, Maggiore U, Rotelli C, et al. Removal of linezolid by conventional intermittent hemodialysis, sustained low-efficiency dialysis, or continuous venovenous hemofiltration in patients with acute renal failure. Crit Care Med. 2004;32(12):2437-2442. doi:10.1097/01.ccm.0000147687.06808.92 [PubMed 15599148]
  71. Floto RA, Olivier KN, Saiman L, et al; US Cystic Fibrosis Foundation and European Cystic Fibrosis Society. US Cystic Fibrosis Foundation and European Cystic Fibrosis Society consensus recommendations for the management of non-tuberculous mycobacteria in individuals with cystic fibrosis. Thorax. 2016;71(suppl 1):1-22. [PubMed 26666259]
  72. Flume PA, Mogayzel PJ Jr, Robinson KA, et al; Clinical Practice Guidelines for Pulmonary Therapies Committee. Cystic fibrosis pulmonary guidelines: treatment of pulmonary exacerbations. Am J Respir Crit Care Med. 2009;180(9):802-808. doi:10.1164/rccm.200812-1845PP [PubMed 19729669]
  73. Galli L, Venturini E, Bassi A, et al. Common community-acquired bacterial skin and soft-tissue infections in children: an intersociety consensus on impetigo, abscess, and cellulitis treatment. Clin Ther. 2019;41(3):532-551.e17. doi:10.1016/j.clinthera.2019.01.010 [PubMed 30777258]
  74. Gerson SL, Kaplan SL, Bruss JB, et al. Hematologic effects of linezolid: summary of clinical experience. Antimicrob Agents Chemother. 2002;46(8):2723-2726. doi:10.1128/aac.46.8.2723-2726.2002 [PubMed 12121967]
  75. Gervasoni C, Bergia R, Cozzi V, Clementi E, Cattaneo D. Is it time to revise linezolid doses in peritoneal dialysis patients? A case series. J Antimicrob Chemother. 2015;70(10):2918-2920. doi:10.1093/jac/dkv184 [PubMed 26142409]
  76. Giunio-Zorkin N, Brown G. Real-life frequency of new-onset thrombocytopenia during linezolid treatment. Can J Hosp Pharm. 2019;72(2):133-138. [PubMed 31036974]
  77. Goldenberg DL, Sexton DJ. Septic arthritis in adults. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed October 24, 2019.
  78. Gómez J, Canovas E, Baños V, et al. Linezolid plus rifampin as a salvage therapy in prosthetic joint infections treated without removing the implant. Antimicrob Agents Chemother. 2011;55(9):4308-4310. doi:10.1128/AAC.00352-11 [PubMed 21690277]
  79. Goss CH, Heltshe SL, West NE, et al; STOP2 Investigators. A randomized clinical trial of antimicrobial duration for cystic fibrosis pulmonary exacerbation treatment. Am J Respir Crit Care Med. 2021;204(11):1295-1305. doi:10.1164/rccm.202102-0461OC [PubMed 34469706]
  80. Gupta K. Acute complicated urinary tract infection (including pyelonephritis) in adults. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed March 20, 2021.
  81. Hanai Y, Matsuo K, Ogawa M, et al. A retrospective study of the risk factors for linezolid-induced thrombocytopenia and anemia. J Infect Chemother. 2016;22(8):536-542. doi:10.1016/j.jiac.2016.05.003 [PubMed 27321773]
  82. Haworth CS, Banks J, Capstick T, et al. British Thoracic Society guidelines for the management of non-tuberculous mycobacterial pulmonary disease (NTM-PD). Thorax. 2017;72(suppl 2):ii1-ii64. doi:10.1136/thoraxjnl-2017-210927 [PubMed 29054853]
  83. Heintz BH, Matzke GR, Dager WE. Antimicrobial Dosing Concepts and Recommendations for Critically Ill Adult Patients Receiving Continuous Renal Replacement Therapy or Intermittent Hemodialysis. Pharmacotherapy. 2009;29(5):562-577. [PubMed 19397464]
  84. Hendricks KA, Wright ME, Shadomy SV, et al; Workgroup on Anthrax Clinical Guidelines. Centers for Disease Control and Prevention expert panel meetings on prevention and treatment of anthrax in adults. Emerg Infect Dis. 2014;20(2). doi:10.3201/eid2002.130687 [PubMed 24447897]
  85. Hensgens MP, Goorhuis A, Dekkers OM, Kuijper EJ. Time interval of increased risk for Clostridium difficile infection after exposure to antibiotics. J Antimicrob Chemother. 2012;67(3):742-748. doi:10.1093/jac/dkr508 [PubMed 22146873]
  86. Hiraki Y, Tsuji Y, Hiraike M, et al. Correlation between serum linezolid concentration and the development of thrombocytopenia. Scand J Infect Dis. 2012;44(1):60-64. doi:10.3109/00365548.2011.608712 [PubMed 21899392]
  87. Honeybourne D, Tobin C, Jevons G, Andrews J, Wise R. Intrapulmonary penetration of linezolid. J Antimicrob Chemother. 2003;51(6):1431-1434. doi:10.1093/jac/dkg262 [PubMed 12746375]
  88. Hoyo I, Martínez-Pastor J, Garcia-Ramiro S, et al. Decreased serum linezolid concentrations in two patients receiving linezolid and rifampicin due to bone infections. Scand J Infect Dis. 2012;44(7):548-550. doi:10.3109/00365548.2012.663931 [PubMed 22385321]
  89. Huang V, Gortney JS. Risk of serotonin syndrome with concomitant administration of linezolid and serotonin agonists. Pharmacotherapy. 2006;26(12):1784-1793. doi:10.1592/phco.26.12.1784 [PubMed 17125439]
  90. Ide T, Takesue Y, Ikawa K, et al. Population pharmacokinetics/pharmacodynamics of linezolid in sepsis patients with and without continuous renal replacement therapy. Int J Antimicrob Agents. 2018;51(5):745-751. doi:10.1016/j.ijantimicag.2018.01.021 [PubMed 29425834]
  91. Im JH, Baek JH, Kwon HY, Lee JS. Incidence and risk factors of linezolid-induced lactic acidosis. Int J Infect Dis. 2015;31:47-52. doi:10.1016/j.ijid.2014.12.009 [PubMed 25499040]
  92. "Inactive" ingredients in pharmaceutical products: update (subject review). American Academy of Pediatrics (AAP) Committee on Drugs. Pediatrics. 1997;99(2):268-278. [PubMed 9024461]
  93. Ioannou P, Stavroulaki M, Mavrikaki V, Papakitsou I, Panagiotakis S. A case of severe hyponatremia due to linezolid-induced SIADH. J Clin Pharm Ther. 2018;43(3):434-436. doi:10.1111/jcpt.12681 [PubMed 29542179]
  94. Ito S. Drug therapy for breast-feeding women. N Engl J Med. 2000;343(2):118-126. [PubMed 10891521]
  95. Iversen K, Ihlemann N, Gill SU, et al. Partial oral versus intravenous antibiotic treatment of endocarditis. N Engl J Med. 2019;380(5):415-424. doi:10.1056/NEJMoa1808312 [PubMed 30152252]
  96. Ivic D, Marjanovic K, Ivic J, Došen G. Toxic epidermal necrolysis after linezolid administration: Case report. Signa Vitae. 2014;9(2):65-67.
  97. Jamp Linezolid (linezolid) [product monograph]. Boucherville, Quebec, Canada: Jamp Pharma Corporation; September 2021.
  98. Jantausch BA, Deville J, Adler S, et al. Linezolid for the treatment of children with bacteremia or nosocomial pneumonia caused by resistant gram-positive bacterial pathogens. Pediatr Infect Dis J. 2003;22(9 Suppl):S164-S171. doi:10.1097/01.inf.0000086956.45566.55 [PubMed 14520142]
  99. Jaspard M, Elefant-Amoura E, Melonio I, De Montgolfier I, Veziris N, Caumes E. Bedaquiline and linezolid for extensively drug-resistant tuberculosis in pregnant woman. Emerg Infect Dis. 2017;23(10). [PubMed 28792382]
  100. Johannesmeyer HJ, Bhakta S, Morales F. Linezolid-associated hypoglycemia. Drug Saf Case Rep. 2017;4(1):18. doi:10.1007/s40800-017-0061-0 [PubMed 29124464]
  101. Jones SJ, Nichols KR, DeYoung HL, Cox EG, Knoderer CA. Linezolid-Associated thrombocytopenia in children with renal impairment. J Pediatric Infect Dis Soc. 2015;4(3):272-275. doi:10.1093/jpids/piu035 [PubMed 26407433]
  102. Joshi L, Taylor SR, Large O, Yacoub S, Lightman S. A case of optic neuropathy after short-term linezolid use in a patient with acute lymphocytic leukemia. Clin Infect Dis. 2009;48(7):e73-e74. doi:10.1086/597298 [PubMed 19231981]
  103. Kalil AC, Metersky ML, Klompas M, et al. Management of adults with hospital-acquired and ventilator-associated pneumonia: 2016 clinical practice guidelines by the Infectious Diseases Society of America and the American Thoracic Society. Clin Infect Dis. 2016;63(5):e61-e111. doi:10.1093/cid/ciw353 [PubMed 27418577]
  104. Kamp J, Bolhuis MS, Tiberi S, et al. Simple strategy to assess linezolid exposure in patients with multi-drug-resistant and extensively-drug-resistant tuberculosis. Int J Antimicrob Agents. 2017;49(6):688-694. doi:10.1016/j.ijantimicag.2017.01.017 [PubMed 28389352]
  105. Kanafani ZA. Invasive Cutibacterium (formerly Propionibacterium) infections. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed September 20, 2020.
  106. 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(5):518-523. doi:10.1097/JCP.0000000000000751 [PubMed 28796019]
  107. 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(1):13. doi:10.1186/s40360-021-00479-w [PubMed 33663616]
  108. Kaya Kılıç E, Bulut C, Sönmezer MÇ, et al. Risk factors for linezolid-associated thrombocytopenia and negative effect of carbapenem combination. J Infect Dev Ctries. 2019;13(10):886-891. doi:10.3855/jidc.10859 [PubMed 32084018]
  109. Kearns GL, Jungbluth GL, Abdel-Rahman SM, et al. Impact of ontogeny on linezolid disposition in neonates and infants. Clin Pharmacol Ther. 2003;74(5):413-422. doi:10.1016/S0009-9236(03)00226-1 [PubMed 14586382]
  110. Keel RA, Schaeftlein A, Kloft C, et al. Pharmacokinetics of intravenous and oral linezolid in adults with cystic fibrosis. Antimicrob Agents Chemother. 2011;55(7):3393-3398. doi:10.1128/AAC.01797-10 [PubMed 21518837]
  111. Kempker RR, Heinrichs MT, Nikolaishvili K, et al. A comparison of linezolid lung tissue concentrations among patients with drug-resistant tuberculosis. Eur Respir J. 2018;51(2). doi:10.1183/13993003.02166-2017 [PubMed 29437945]
  112. Kraft MD, Pasko DA, DePestel DD, Ellis JJ, Peloquin CA, Mueller BA. Linezolid clearance during continuous venovenous hemodiafiltration: a case report. Pharmacotherapy. 2003;23(8):1071-1075. doi:10.1592/phco.23.8.1071.32874 [PubMed 12921253]
  113. Kruzer K, Garner W, Honda K, Packer CD. Linezolid-induced leukocytoclastic vasculitis. Ann Pharmacother. 2018;52(12):1263-1264. doi:10.1177/1060028018793246 [PubMed 30066575]
  114. Kumar S, Kohlhoff S, Valencia G, et al. Treatment of Vancomycin-Resistant Enterococcus faecium Ventriculitis in a Neonate. Int J Antimicrob Agents. 2007;29(6):740-741. [PubMed 17395431]
  115. Leader R, Hackett J, Allan A, Carter P. Linezolid-induced pancytopenia. BMJ Case Rep. 2018;2018:bcr2018225480. doi:10.1136/bcr-2018-225480 [PubMed 30054325]
  116. Lee E, Burger S, Shah J, et al. Linezolid-associated toxic optic neuropathy: a report of 2 cases. Clin Infect Dis. 2003;37(10):1389-1391. doi:10.1086/379012 [PubMed 14583875]
  117. Lee M, Lee J, Carroll MW, et al. Linezolid for treatment of chronic extensively drug-resistant tuberculosis. N Engl J Med. 2012;367(16):1508-1518. doi:10.1056/NEJMoa1201964 [PubMed 23075177]
  118. Lee S, Kang BH, Ryu WY, Um SJ, Roh MS, Son C. Is severe and long-lasting linezolid-induced optic neuropathy reversible? Intern Med. 2018;57(24):3611-3613. doi:10.2169/internalmedicine.1344-18 [PubMed 30101940]
  119. Li PK, Szeto CC, Piraino B, et al. ISPD peritonitis recommendations: 2016 update on prevention and treatment. Perit Dial Int. 2016;36(5):481-508. doi:10.3747/pdi.2016.00078 [PubMed 27282851]
  120. Li SC, Ye Q, Xu H, Zhang L, Wang Y. Population pharmacokinetics and dosing optimization of linezolid in pediatric patients. Antimicrob Agents Chemother. 2019;63(4):e02387-18. doi:10.1128/AAC.02387-18 [PubMed 30642929]
  121. Lim FH, Lovering AM, Currie A, Jenkins DR. Linezolid and lactation: measurement of drug levels in breast milk and the nursing infant. J Antimicrob Chemother. 2017;72(9):2677-2678. [PubMed 28541475]
  122. 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(4):345-351. doi:10.1016/j.ijantimicag.2006.04.017 [PubMed 16935472]
  123. Linezolid injection [prescribing information]. Lake Forest, IL: Hospira Inc; September 2023.
  124. Linezolid powder for suspension [prescribing information]. Eatontown, NJ: West-Ward Pharmaceuticals Corp; September 2020.
  125. Linezolid tablet [prescribing information]. Pulaski, TN: AvKARE Inc; January 2016.
  126. Lipsky BA, Berendt AR, Cornia PB, et al; Infectious Diseases Society of America. 2012 Infectious Diseases Society of America clinical practice guideline for the diagnosis and treatment of diabetic foot infections. Clin Infect Dis. 2012;54(12):e132-e173. doi:10.1093/cid/cis346 [PubMed 22619242]
  127. Lipsky BA, Itani K, Norden C; Linezolid Diabetic Foot Infections Study Group. Treating foot infections in diabetic patients: a randomized, multicenter, open-label trial of linezolid versus ampicillin-sulbactam/amoxicillin-clavulanate. Clin Infect Dis. 2004;38(1):17-24. doi:10.1086/380449 [PubMed 14679443]
  128. Liu C, Bayer A, Cosgrove SE, et al. Clinical practice guidelines by the Infectious Diseases Society of America for the treatment of methicillin-resistant Staphylococcus aureus infections in adults and children: executive summary. Clin Infect Dis. 2011;52(3):285-292. doi:10.1093/cid/cir034 [PubMed 21217178]
  129. Lowy FD. Methicillin-resistant Staphylococcus aureus (MRSA) in adults: treatment of bacteremia. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed January 21, 2022.
  130. 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(6):732-739. doi:10.1097/FTD.0000000000000665 [PubMed 31259884]
  131. Ma TK, Leung CB, Chow KM, Kwan BC, Li PK, Szeto CC. Newer antibiotics for the treatment of peritoneal dialysis-related peritonitis. Clin Kidney J. 2016;9(4):616-623. doi:10.1093/ckj/sfw059 [PubMed 27478608]
  132. 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(36):e12114. doi:10.1097/MD.0000000000012114 [PubMed 30200095]
  133. Maqsood H, Sajjad S, Aslam S, Younus S, Naveed S. Linezolid induced psychosis and hallucination: case report and literature review. Ann Med Surg (Lond). 2022;83:104654. doi:10.1016/j.amsu.2022.104654 [PubMed 36389201]
  134. Matsumoto K, Takeda Y, Takeshita A, et al. Renal function as a predictor of linezolid-induced thrombocytopenia. Int J Antimicrob Agents. 2009;33(1):98-99. doi:10.1016/j.ijantimicag.2008.07.002 [PubMed 18790615]
  135. Matsumoto K, Takeshita A, Ikawa K, et al. Higher linezolid exposure and higher frequency of thrombocytopenia in patients with renal dysfunction. Int J Antimicrob Agents. 2010;36(2):179-181. doi:10.1016/j.ijantimicag.2010.02.019 [PubMed 20392606]
  136. McDonald LC, Gerding DN, Johnson S, et al. Clinical practice guidelines for Clostridium difficile infection in adults and children: 2017 update by the Infectious Diseases Society of America (IDSA) and Society for Healthcare Epidemiology of America (SHEA). Clin Infect Dis. 2018;66(7):e1-e48. doi:10.1093/cid/cix1085 [PubMed 29462280]
  137. Mercieri M, Di Rosa R, Pantosti A, et al. Critical Pneumonia Complicating Early-Stage Pregnancy. Anesth Analg. 2010;110(3):852-854. [PubMed 20185661]
  138. Mermel LA, Allon M, Bouza E, et al. Clinical practice guidelines for the diagnosis and management of intravascular catheter-related infection: 2009 update by the Infectious Diseases Society of America. Clin Infect Dis. 2009;49(1):1-45. doi:10.1086/599376 [PubMed 19489710]
  139. Metlay JP, Waterer GW, Long AC, et al. Diagnosis and treatment of adults with community-acquired pneumonia. An official clinical practice guideline of the American Thoracic Society and Infectious Diseases Society of America. Am J Resp Crit Care Med. 2019;200(7):e45-e67. doi:10.1164/rccm.201908-1581ST [PubMed 31573350]
  140. Meyer B, Kornek GV, Nikfardjam M, et al. Multiple-dose pharmacokinetics of linezolid during continuous venovenous haemofiltration. J Antimicrob Chemother. 2005;56(1):172-179. doi:10.1093/jac/dki133 [PubMed 15905303]
  141. Millard J, Pertinez H, Bonnett L, et al. Linezolid pharmacokinetics in MDR-TB: a systematic review, meta-analysis and Monte Carlo simulation. J Antimicrob Chemother. 2018;73(7):1755-1762. doi:10.1093/jac/dky096 [PubMed 29584861]
  142. Mok J, Lee M, Kim DK, et al; MDR-END investigators. 9 months of delamanid, linezolid, levofloxacin, and pyrazinamide versus conventional therapy for treatment of fluoroquinolone-sensitive multidrug-resistant tuberculosis (MDR-END): a multicentre, randomised, open-label phase 2/3 non-inferiority trial in South Korea. Lancet. 2022;400(10362):1522-1530. doi:10.1016/S0140-6736(22)01883-9 [PubMed 36522208]
  143. Moise PA, Forrest A, Birmingham MC, Schentag JJ. The efficacy and safety of linezolid as treatment for Staphylococcus aureus infections in compassionate use patients who are intolerant of, or who have failed to respond to, vancomycin. J Antimicrob Chemother. 2002;50(6):1017-1026. doi:10.1093/jac/dkf215 [PubMed 12461026]
  144. Mori N, Kamimura Y, Kimura Y, Hirose S, Aoki Y, Bito S. Comparative analysis of lactic acidosis induced by linezolid and vancomycin therapy using cohort and case-control studies of incidence and associated risk factors. Eur J Clin Pharmacol. 2018;74(4):405-411. doi:10.1007/s00228-017-2377-1 [PubMed 29222713]
  145. Murray BE, Miller WR. Treatment of enterococcal infections. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed October 8, 2021.
  146. Myrianthefs P, Markantonis SL, Vlachos K, et al. Serum and cerebrospinal fluid concentrations of linezolid in neurosurgical patients. Antimicrob Agents Chemother. 2006;50(12):3971-3976. doi:10.1128/AAC.00051-06 [PubMed 16982782]
  147. Nagashima G, Okamoto N, Okuda M, et al. Effect of linezolid against postneurosurgical meningitis caused by methicillin-resistant Staphylococcus epidermidis: case report. J Infect Chemother. 2008;14(2):147-150. doi:10.1007/s10156-007-0582-z [PubMed 18622679]
  148. Nahid P, Mase SR, Migliori GB, et al. Treatment of drug-resistant tuberculosis. An official ATS/CDC/ERS/IDSA clinical practice guideline. Am J Respir Crit Care Med. 2019;200(10):e93-e142. doi:10.1164/rccm.201909-1874ST [PubMed 31729908]
  149. Nambiar S, Rellosa N, Wassel RT, Borders-Hemphill V, Bradley JS. Linezolid-associated peripheral and optic neuropathy in children. Pediatrics. 2011;127(6):e1528-e1532. doi:10.1542/peds.2010-2125 [PubMed 21555496]
  150. Narita M, Tsuji BT, Yu VL. Linezolid-associated peripheral and optic neuropathy, lactic acidosis, and serotonin syndrome. Pharmacotherapy. 2007;27(8):1189-1197. doi:10.1592/phco.27.8.1189 [PubMed 17655517]
  151. Natsumoto B, Yokota K, Omata F, Furukawa K. Risk factors for linezolid-associated thrombocytopenia in adult patients. Infection. 2014;42(6):1007-1012. doi:10.1007/s15010-014-0674-5 [PubMed 25119433]
  152. Norris AH, Shrestha NK, Allison GM, et al. 2018 Infectious Diseases Society of America clinical practice guideline for the management of outpatient parenteral antimicrobial therapy. Clin Infect Dis. 2019;68(1):1-4. doi:10.1093/cid/ciy867 [PubMed 30551156]
  153. Ntziora F, Falagas ME. Linezolid for the treatment of patients with central nervous system infection. Ann Pharmacother. 2007;41(2):296-308. doi:10.1345/aph.1H307 [PubMed 17284501]
  154. Nukui Y, Hatakeyama S, Okamoto K, et al. High plasma linezolid concentration and impaired renal function affect development of linezolid-induced thrombocytopenia. J Antimicrob Chemother. 2013;68(9):2128-2133. doi:10.1093/jac/dkt133 [PubMed 23625638]
  155. Nyang'wa BT, Berry C, Kazounis E, et al; TB-PRACTECAL Study Collaborators. A 24-week, all-oral regimen for rifampin-resistant tuberculosis. N Engl J Med. 2022;387(25):2331-2343. doi:10.1056/NEJMoa2117166 [PubMed 36546625]
  156. O'Driscoll T, Crank C. Vancomycin-resistant enterococcal infections: epidemiology, clinical manifestations, and optimal management. Inf and Drug Res. 2015;8:217-230. [PubMed 26244026]
  157. Osmon DR, Berbari EF, Berendt AR, et al. Diagnosis and management of prosthetic joint infection: clinical practice guidelines by the Infectious Diseases Society of America. Clin Infect Dis. 2013;56(1):e1-e25. doi:10.1093/cid/cis803 [PubMed 23223583]
  158. Osmon DR, Tande AJ. Osteomyelitis in adults: treatment. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed July 8, 2019.
  159. Ozkaya-Parlakay A, Kara A, Celik M, et al. Early lactic acidosis associated with linezolid therapy in paediatric patients. Int J Antimicrob Agents. 2014;44(4):334-336. doi:10.1016/j.ijantimicag.2014.06.017 [PubMed 25182713]
  160. Pea F, Cojutti PG, Baraldo M. A 10-year experience of therapeutic drug monitoring (TDM) of linezolid in a hospital-wide population of patients receiving conventional dosing: is there enough evidence for suggesting TDM in the majority of patients? Basic Clin Pharmacol Toxicol. 2017;121(4):303-308. doi:10.1111/bcpt.12797 [PubMed 28419737]
  161. Pea F, Furlanut M, Cojutti P, et al. Therapeutic drug monitoring of linezolid: a retrospective monocentric analysis. Antimicrob Agents Chemother. 2010;54(11):4605-4610. doi:10.1128/AAC.00177-10 [PubMed 20733043]
  162. Pea F, Viale P, Cojutti P, Del Pin B, Zamparini E, Furlanut M. Therapeutic drug monitoring may improve safety outcomes of long-term treatment with linezolid in adult patients. J Antimicrob Chemother. 2012;67(8):2034-2042. doi:10.1093/jac/dks153 [PubMed 22553142]
  163. Pea F, Viale P, Lugano M, et al. Linezolid disposition after standard dosages in critically ill patients undergoing continuous venovenous hemofiltration: a report of 2 cases. Am J Kidney Dis. 2004;44(6):1097-1102. doi:10.1053/j.ajkd.2004.08.032 [PubMed 15558532]
  164. Pernica JM, Harman S, Kam AJ, et al. Short-course antimicrobial therapy for pediatric community-acquired pneumonia: The SAFER randomized clinical trial. JAMA Pediatr. 2021;175(5):475-482. doi:10.1001/jamapediatrics.2020.6735 [PubMed 33683325]
  165. Pontefract BA, Rovelsky SA, Madaras-Kelly KJ. Linezolid to treat urinary tract infections caused by vancomycin-resistant Enterococcus. SAGE Open Med. 2020;8:2050312120970743. doi:10.1177/2050312120970743 [PubMed 33209303]
  166. Pries-Heje MM, Wiingaard C, Ihlemann N, et al. Five-year outcomes of the partial oral treatment of endocarditis (POET) trial. N Engl J Med. 2022;386(6):601-602. doi:10.1056/NEJMc2114046 [PubMed 35139280]
  167. Rabon AD, Fisher JP, MacVane SH. Incidence and risk factors for development of thrombocytopenia in patients treated with linezolid for 7 days or greater. Ann Pharmacother. 2018;52(11):1162-1164. doi:10.1177/1060028018783498 [PubMed 29911390]
  168. Rac H, Bojikian KD, Lucar J, Barber KE. Successful treatment of necrotizing fasciitis and streptococcal toxic shock syndrome with the addition of linezolid. Case Rep Infect Dis. 2017;2017:5720708. doi:10.1155/2017/5720708 [PubMed 28299216]
  169. Rana B, Butcher I, Grigoris P, Murnaghan C, Seaton RA, Tobin CM. Linezolid penetration into osteo-articular tissues. J Antimicrob Chemother. 2002;50(5):747-750. doi:10.1093/jac/dkf207 [PubMed 12407135]
  170. 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(1):83-92. doi:10.1097/FTD.0000000000000710 [PubMed 31652190]
  171. Rao N, Ziran BH, Hall RA, Santa ER. Successful treatment of chronic bone and joint infections with oral linezolid. Clin Orthop Relat Res. 2004;(427):67-71. [PubMed 15552139]
  172. Rayner CR, Forrest A, Meagher AK, Birmingham MC, Schentag JJ. Clinical pharmacodynamics of linezolid in seriously ill patients treated in a compassionate use programme. Clin Pharmacokinet. 2003;42(15):1411-1423. doi:10.2165/00003088-200342150-00007 [PubMed 14674791]
  173. Refer to manufacturer's labeling.
  174. Rho JP, Sia IG, Crum BA, Dekutoski MB, Trousdale RT. Linezolid-associated peripheral neuropathy. Mayo Clin Proc. 2004;79(7):927-930. doi:10.4065/79.7.927 [PubMed 15244392]
  175. Roger C, Muller L, Wallis SC, et al. Population pharmacokinetics of linezolid in critically ill patients on renal replacement therapy: comparison of equal doses in continuous venovenous haemofiltration and continuous venovenous haemodiafiltration. J Antimicrob Chemother. 2016;71(2):464-470. doi:10.1093/jac/dkv349 [PubMed 26538503]
  176. Roger C, Roberts JA, Muller L. Clinical pharmacokinetics and pharmacodynamics of oxazolidinones. Clin Pharmacokinet. 2018;57(5):559-575. doi:10.1007/s40262-017-0601-x [PubMed 29063519]
  177. Rowe HE, Felkins K, Cooper SD, Hale TW. Transfer of linezolid into breast milk. J Hum Lact. 2014;30(4):410-412. [PubMed 25098610]
  178. Saavedra-Lozano J, Falup-Pecurariu O, Faust SN, et al. Bone and joint infections. Pediatr Infect Dis J. 2017;36(8):788-799. doi:10.1097/INF.0000000000001635 [PubMed 28708801]
  179. Sagirli O, Onal A, Toker S, et al. Determination of Linezolid in Human Breast Milk by High-Performance Liquid Chromatography With Ultraviolet Detection. J AOAC Int. 2009;92(6):1658-1662. [PubMed 20166583]
  180. Same RG, Amoah J, Hsu AJ, et al. The association of antibiotic duration with successful treatment of community-acquired pneumonia in children. J Pediatric Infect Dis Soc. 2021;10(3):267-273. doi:10.1093/jpids/piaa055 [PubMed 32525203]
  181. Santini A, Ronchi D, Garbellini M, Piga D, Protti A. Linezolid-induced lactic acidosis: the thin line between bacterial and mitochondrial ribosomes. Expert Opin Drug Saf. 2017;16(7):833-843. doi:10.1080/14740338.2017.1335305 [PubMed 28538105]
  182. Santos JA, Varandas L, Gouveia C. Reversible teeth discoloration in children: A linezolid therapy side effect. Clin Pediatr (Phila). 2015;54(8):809. doi:10.1177/0009922814562558 [PubMed 25500501]
  183. Sasaki T, Takane H, Ogawa K, et al. Population pharmacokinetic and pharmacodynamic analysis of linezolid and a hematologic side effect, thrombocytopenia, in Japanese patients. Antimicrob Agents Chemothe. 2011;55(5):1867-1873. doi:10.1128/AAC.01185-10 [PubMed 21357301]
  184. Schluger NW, Heysell SK, Friedland G. Treatment of drug-resistant pulmonary tuberculosis in adults. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed May 25, 2022.
  185. Sexton DJ, Sampson JH. Intracranial epidural abscess. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed February 21, 2022.
  186. Sexton DJ, Sampson JH. Spinal epidural abscess. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed October 24, 2019b.
  187. Shane AL, Mody RK, Crump JA, et al. 2017 Infectious Diseases Society of America Clinical practice guidelines for the diagnosis and management of infectious diarrhea. Clin Infect Dis. 2017;65(12):e45-e80. doi:10.1093/cid/cix669 [PubMed 29053792]
  188. Shi C, Xia J, Ye J, et al. Effect of renal function on the risk of thrombocytopenia in patients receiving linezolid therapy: a systematic review and meta-analysis. Br J Clin Pharmacol. Published online October 10, 2021. doi:10.1111/bcp.14965 [PubMed 34192814]
  189. Shneker BF, Baylin PD, Nakhla ME. Linezolid inducing complex partial status epilepticus in a patient with epilepsy. Neurology. 2009;72(4):378-379. doi:10.1212/01.wnl.0000341309.88093.3c [PubMed 19171838]
  190. Sicard M, Launay E, Caillon J, et al. Pharmacokinetics of linezolid treatment using intravenous and oral administrations in extremely premature infants. Eur J Clin Pharmacol. 2015;71(5):611-615. doi:10.1007/s00228-015-1813-3 [PubMed 25740677]
  191. Sicard M, Moussa A, Barrington K, et al. Neonatal and neurodevelopmental outcomes following linezolid for coagulase-negative Staphylococcal Infection: Real world evidence. Pediatr Infect Dis J. 2020;39(7):598-603. doi:10.1097/INF.0000000000002619 [PubMed 32091496]
  192. Simon RH. Cystic fibrosis: Management of pulmonary exacerbations. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed January 20, 2022.
  193. Sipahi OR, Bardak-Ozcem S, Turhan T, et al. Vancomycin versus linezolid in the treatment of methicillin-resistant Staphylococcus aureus meningitis. Surg Infect (Larchmt). 2013;14(4):357-362. doi:10.1089/sur.2012.091 [PubMed 23672240]
  194. Song T, Lee M, Jeon HS, et al. Linezolid trough concentrations correlate with mitochondrial toxicity-related adverse events in the treatment of chronic extensively drug-resistant tuberculosis. EBioMedicine. 2015;2(11):1627-1633. doi:10.1016/j.ebiom.2015.09.051 [PubMed 26870788]
  195. Soraluce A, Barrasa H, Asín-Prieto E, et al. Novel population pharmacokinetic model for linezolid in critically ill patients and evaluation of the adequacy of the current dosing recommendation. Pharmaceutics. 2020;12(1):54. doi:10.3390/pharmaceutics12010054 [PubMed 31936614]
  196. Southwick FS. Treatment and prognosis of bacterial brain abscess. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed June 23, 2022.
  197. Souza E, Crass RL, Felton J, Hanaya K, Pai MP. Accumulation of major linezolid metabolites in patients with renal impairment. Antimicrob Agents Chemother. 2020;64(5):e00027-20. doi:10.1128/AAC.00027-20 [PubMed 32152085]
  198. Srivastava S, Magombedze G, Koeuth T, et al. Linezolid dose that maximizes sterilizing effect while minimizing toxicity and resistance emergence for tuberculosis. Antimicrob Agents Chemother. 2017;61(8):e00751-17. doi:10.1128/AAC.00751-17 [PubMed 28584143]
  199. Stevens DL. Invasive group A streptococcal infection and toxic shock syndrome: treatment and prevention. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed July 21, 2021.
  200. Stevens DL, Bisno AL, Chambers HF, et al; Infectious Diseases Society of America. Practice guidelines for the diagnosis and management of skin and soft tissue infections: 2014 update by the Infectious Diseases Society of America. Clin Infect Dis. 2014;59(2):e10-e52. doi:10.1093/cid/ciu444 [PubMed 24973422]
  201. 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(5):729-730. doi:10.1086/500265 [PubMed 16447124]
  202. Su E, Crowley K, Carcillo JA, Michaels MG. Linezolid and lactic acidosis: a role for lactate monitoring with long-term linezolid use in children. Pediatr Infect Dis J. 2011;30(9):804-806. doi:10.1097/INF.0b013e3182186035 [PubMed 21852764]
  203. Swoboda S, Ober MC, Lichtenstern C, et al. Pharmacokinetics of linezolid in septic patients with and without extended dialysis. Eur J Clin Pharmacol. 2010;66(3):291-298. doi:10.1007/s00228-009-0766-9 [PubMed 20013257]
  204. Takahashi Y, Takesue Y, Nakajima K, et al. Risk factors associated with the development of thrombocytopenia in patients who received linezolid therapy. J Infect Chemother. 2011;17(3):382-387. doi:10.1007/s10156-010-0182-1 [PubMed 21127934]
  205. Tang S, Yao L, Hao X, et al. Efficacy, safety and tolerability of linezolid for the treatment of XDR-TB: a study in China. Eur Respir J. 2015;45(1):161-170. doi:10.1183/09031936.00035114 [PubMed 25234807]
  206. Thibault C, Kassir N, Goyer I, et al. Population pharmacokinetics of intravenous linezolid in premature infants. Pediatr Infect Dis J. 2019;38(1):82-88. doi:10.1097/INF.0000000000002067 [PubMed 29634620]
  207. Trotman RL, Williamson JC, Shoemaker DM, et al. Antibiotic Dosing in Critically Ill Adult Patients Receiving Continuous Renal Replacement Therapy. Clin Infect Dis. 2005;41(8):1159-1166. [PubMed 16163635]
  208. Tsona A, Metallidis S, Foroglou N, et al. Linezolid penetration into cerebrospinal fluid and brain tissue. J Chemother. 2010;22(1):17-19. doi:10.1179/joc.2010.22.1.17 [PubMed 20227987]
  209. Tsuji Y, Holford NHG, Kasai H, et al. Population pharmacokinetics and pharmacodynamics of linezolid-induced thrombocytopenia in hospitalized patients. Br J Clin Pharmacol. 2017;83(8):1758-1772. doi:10.1111/bcp.13262 [PubMed 28186644]
  210. Tunkel AR, Hartman BJ, Kaplan SL, et al. Practice guidelines for the management of bacterial meningitis. Clin Infect Dis. 2004;39(9):1267-1284. doi:10.1086/425368 [PubMed 15494903]
  211. Tunkel AR, Hasbun R, Bhimraj A, et al. 2017 Infectious Diseases Society of America's clinical practice guidelines for healthcare-associated ventriculitis and meningitis. Clin Infect Dis. 2017;64(6):e34-e65. doi:10.1093/cid/ciw861 [PubMed 28203777]
  212. Udy AA, Roberts JA, Boots RJ, Paterson DL, Lipman J. Augmented renal clearance: implications for antibacterial dosing in the critically ill. Clin Pharmacokinet. 2010;49(1):1-16. doi:10.2165/11318140-000000000-00000 [PubMed 20000886]
  213. Valerio M, Pedromingo M, Muñoz P, et al. Potential protective role of linezolid against Clostridium difficile infection. Int J Antimicrob Agents. 2012;39(5):414-419. doi:10.1016/j.ijantimicag.2012.01.005 Erratum in: Int J Antimicrob Agents. 2012;40(1):94. [PubMed 22445203]
  214. Van Kampenhout E, Bolhuis MS, Alffenaar JC, et al. Pharmacokinetics of moxifloxacin and linezolid during and after pregnancy in a patient with multidrug-resistant tuberculosis. Eur Respir J. 2017;49(3). [PubMed 28331037]
  215. Vardakas KZ, Trigkidis KK, Boukouvala E, Falagas ME. Clostridium difficile infection following systemic antibiotic administration in randomised controlled trials: a systematic review and meta-analysis. Int J Antimicrob Agents. 2016;48(1):1-10. doi:10.1016/j.ijantimicag.2016.03.008 [PubMed 27216385]
  216. Villa G, Di Maggio P, De Gaudio AR, et al. Effects of continuous renal replacement therapy on linezolid pharmacokinetic/pharmacodynamics: a systematic review. Crit Care. 2016;20(1):374. doi:10.1186/s13054-016-1551-7 [PubMed 27863531]
  217. Vishnu VY, Modi M, Goyal MK, Lal V. Linezolid induced reversible peripheral neuropathy. Am J Ther. 2016;23(6):e1839-e1841. doi:10.1097/MJT.0000000000000359 [PubMed 26741956]
  218. Warady BA, Bakkaloglu S, Newland J, et al. Consensus guidelines for the prevention and treatment of catheter-related infections and peritonitis in pediatric patients receiving peritoneal dialysis: 2012 update. Perit Dial Int. 2012;(32)(suppl 2):S32-S86. [PubMed 22851742]
  219. Wasserman S, Denti P, Brust JCM, et al. Linezolid pharmacokinetics in South African patients with drug-resistant tuberculosis and a high prevalence of HIV coinfection. Antimicrob Agents Chemother. 2019;63(3):e02164-18. doi:10.1128/AAC.02164-18 [PubMed 30617089]
  220. Watson T, Hickok J, Fraker S, Korwek K, Poland RE, Septimus E. Evaluating the risk factors for hospital-onset Clostridium difficile infections in a large healthcare system. Clin Infect Dis. 2018;66(12):1957-1959. doi:10.1093/cid/cix1112 [PubMed 29272341]
  221. Weintrob AC, Sexton DJ. Clinical manifestations, diagnosis, and management of diabetic infections of the lower extremities. Post TW, ed. UpToDate. Waltham, MA: UpToDate Inc. http://www.uptodate.com. Accessed November 24, 2020.
  222. WHO consolidated guidelines on tuberculosis: Module 4: Treatment - Drug-resistant tuberculosis treatment. Geneva: World Health Organization; 2020. [PubMed 32603040]
  223. Wildenthal JA, Atkinson A, Lewis S, et al. Outcomes of partial oral antibiotic treatment for complicated Staphylococcus aureus bacteremia in people who inject drugs. Clin Infect Dis. 2023;76(3):487-496. doi:10.1093/cid/ciac714 [PubMed 32603040]
  224. Willekens C, Dumezy F, Boyer T, et al. Linezolid induces ring sideroblasts. Haematologica. 2013;98(11):e138-40. doi:10.3324/haematol.2013.092395 [PubMed 24186315]
  225. Woods CR, Bradley JS, Chatterjee A, et al. Clinical practice guideline by the Pediatric Infectious Diseases Society and the Infectious Diseases Society of America: 2021 guideline on diagnosis and management of acute hematogenous osteomyelitis in pediatrics. J Pediatric Infect Dis Soc. 2021;10(8):801-844. doi:10.1093/jpids/piab027 [PubMed 34350458]
  226. World Health Organization (WHO). Breastfeeding and maternal medication, recommendations for drugs in the Eleventh WHO Model List of Essential Drugs. 2002. http://www.who.int/maternal_child_adolescent/documents/55732/en/. [PubMed 23215911]
  227. World Health Organization (WHO). WHO consolidated guidelines on drug-resistant tuberculosis treatment. Geneva: World Health Organization; 2019. https://www.who.int/tb/publications/2019/consolidated-guidelines-drug-resistant-TB-treatment/en/.
  228. World Health Organization (WHO). WHO consolidated guidelines on tuberculosis. Module 4: treatment – drug-resistant treatment, 2022 update. Geneva: World Health Organization; 2022. https://www.who.int/publications/i/item/9789240063129.
  229. 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(1):66-72. doi:10.1086/498509 [PubMed 16323094]
  230. Yagi T, Naito T, Doi M, et al. Plasma exposure of free linezolid and its ratio to minimum inhibitory concentration varies in critically ill patients. Int J Antimicrob Agents. 2013;42(4):329-334. doi:10.1016/j.ijantimicag.2013.06.015 [PubMed 23988716]
  231. Yang M, Xu M. Linezolid-induced angioedema and urticaria in a patient with renal failure. Braz J Infect Dis. 2012;16(6):606-607. doi:10.1016/j.bjid.2012.07.007 [PubMed 23141989]
  232. Yang XY, Chen L, Gu JN, Zeng CJ, Pan DM. Linezolid-induced pure red cell aplasia: a case report. Infect Drug Resist. 2022;15:3847-3856. doi:10.2147/IDR.S362358 [PubMed 35899083]
  233. Yasar KK, Pehlivanoglu F, Sengoz G, Cabioglu N. Successfully treated Mycobacterium abscessus mastitis: a rare cause of breast masses. Indian J Med Microbiol. 2011;29(4):425-427. [PubMed 22120809]
  234. Zhao M, Liang L, Ji L, et al. Similar efficacy and safety of daptomycin versus linezolid for treatment of vancomycin-resistant enterococcal bloodstream infections: a meta-analysis. Int J Antimicrob Agents. 2016;48(3):231-238. doi:10.1016/j.ijantimicag.2016.06.010 [PubMed 27475877]
  235. Zhang SH, Zhu ZY, Chen Z, et al. Population pharmacokinetics and dosage optimization of linezolid in patients with liver dysfunction. Antimicrob Agents Chemother. 2020;64(6):e00133-20. doi:10.1128/AAC.00133-20 [PubMed 32253210]
  236. Zheng J, Sun Z, Sun L, et al. Pharmacokinetics and pharmacodynamics of linezolid in patients with sepsis receiving continuous venovenous hemofiltration and extended daily hemofiltration. J Infect Dis. 2020;221(suppl 2):S279-S287. doi:10.1093/infdis/jiz566 [PubMed 32176792]
  237. Zhou W, Nie W, Wang Q, et al. Linezolid pharmacokinetics/pharmacodynamics-based optimal dosing for multidrug-resistant tuberculosis. Int J Antimicrob Agents. 2022;59(6):106589. doi:10.1016/j.ijantimicag.2022.106589 [PubMed 35405268]
  238. Zoller M, Maier B, Hornuss C, et al. Variability of linezolid concentrations after standard dosing in critically ill patients: a prospective observational study. Crit Care. 2014;18(4):R148. doi:10.1186/cc13984 [PubMed 25011656]
  239. Zyvox (linezolid) [prescribing information]. New York, NY: Pharmacia and Upjohn; November 2021.
  240. Zyvox (linezolid) [prescribing information]. New York, NY: Pharmacia and Upjohn; August 2022.
  241. Zyvox (linezolid) [prescribing information]. New York, NY: Pharmacia and Upjohn; August 2023.
  242. Zyvoxam (linezolid) [product monograph]. Kirkland, Quebec, Canada: Pfizer Canada ULC; November 2022.
Topic 12555 Version 483.0

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