Linezolid (DB00601): A Comprehensive Pharmacological and Clinical Monograph
Executive Summary
Linezolid is a seminal synthetic antibiotic, representing the first member of the oxazolidinone class to be approved for clinical use in 2000.[1] Its introduction marked a significant therapeutic advance in the global effort to combat infections caused by multidrug-resistant (MDR) Gram-positive pathogens. In recognition of its critical role, the World Health Organization (WHO) has classified Linezolid as "critically important" for human medicine, reserving its use primarily for severe infections that are resistant to other antibiotics, most notably those caused by methicillin-resistant
Staphylococcus aureus (MRSA) and vancomycin-resistant enterococci (VRE).[1]
The pharmacological distinction of Linezolid lies in its unique mechanism of action. Unlike many other ribosome-targeting antibiotics that interfere with later stages of protein production, Linezolid inhibits the very first step—the formation of the 70S initiation complex—thereby preventing bacterial protein synthesis at its origin.[1] This novel target site accounts for its activity against bacteria that have developed resistance to other protein synthesis inhibitors. A cornerstone of its clinical utility is its exceptional pharmacokinetic profile, characterized by approximately 100% oral bioavailability. This property allows for equivalent dosing between intravenous and oral formulations, facilitating a seamless transition of therapy that can shorten hospital stays and reduce overall healthcare costs.[1]
Despite its efficacy, the clinical use of Linezolid is governed by a distinct and significant safety profile. Its primary toxicities are duration-dependent, with myelosuppression (particularly thrombocytopenia) and peripheral and optic neuropathy becoming major concerns with treatment courses extending beyond two to four weeks.[1] Furthermore, Linezolid is a weak, reversible, non-selective inhibitor of monoamine oxidase (MAO), creating a significant risk for potentially life-threatening drug-drug and drug-food interactions, including serotonin syndrome and hypertensive crises.[4]
In the clinical armamentarium against MDR Gram-positive infections, Linezolid is frequently compared with vancomycin and daptomycin. It has demonstrated superior efficacy to vancomycin in the treatment of skin and soft tissue infections and offers a critical advantage in patients with renal impairment, as it does not require dose adjustment.[1] When the total cost of care is considered—including administration, monitoring, and length of hospitalization—Linezolid often emerges as a more cost-effective option than its intravenous-only counterparts.[9] The optimal application of Linezolid requires a careful balance of its potent antimicrobial activity against its significant potential for toxicity and interactions, positioning it as a powerful but highly managed tool in modern antimicrobial stewardship.
Drug Identity and Physicochemical Properties
This section establishes the fundamental chemical and physical identity of Linezolid, providing the foundational data necessary for understanding its pharmacological behavior and ensuring its unambiguous identification across scientific and clinical platforms.
Nomenclature and Identifiers
Linezolid is identified by a variety of names and codes across chemical, pharmacological, and regulatory databases. Its consistent identification is crucial for research, clinical practice, and procurement.
- Generic Name: Linezolid [1]
- Synonyms and Developmental Codes: Common synonyms include linezolide and linezolidum. During its development by Pharmacia & Upjohn (now Pfizer), it was known by the codes PNU-100766 and U-100766.[2]
- Brand Names: The most common brand names under which Linezolid is marketed are Zyvox and Zyvoxam.[1]
- IUPAC Name: The systematic chemical name for the active (S)-enantiomer is (S)-N-({3-[3-fluoro-4-(morpholin-4-yl)phenyl]-2-oxo-1,3-oxazolidin-5-yl}methyl)acetamide.[1]
A consolidated list of key database identifiers is provided in Table 1 to facilitate cross-referencing.
Table 1: Key Physicochemical and Database Identifiers for Linezolid
Attribute | Value | Source(s) |
---|
Generic Name | Linezolid | 1 |
DrugBank ID | DB00601 | 1 |
CAS Number | 165800-03-3 | 1 |
IUPAC Name | (S)-N-({3-[3-fluoro-4-(morpholin-4-yl)phenyl]-2-oxo-1,3-oxazolidin-5-yl}methyl)acetamide | 1 |
Molecular Formula | C16H20FN3O4 | 11 |
Molecular Weight | 337.35 g/mol | 11 |
PubChem CID | 441401 | 1 |
ChEMBL ID | CHEMBL126 | 1 |
KEGG ID | D00947 | 1 |
UNII | ISQ9I6J12J | 1 |
InChIKey | TYZROVQLWOKYKF-ZDUSSCGKSA-N | 1 |
Chemical Structure and Properties
Linezolid is a synthetic small molecule belonging to the oxazolidinone class of antibacterial agents. Its structure incorporates several key functional groups that define its activity and classification.
- Molecular Formula: C16H20FN3O4.[11]
- Molecular Weight: The calculated molecular weight is 337.35 g/mol, with an average weight of 337.3461 Da.[4]
- Chemical Class: Linezolid is classified as an oxazolidinone, an organofluorine compound, a member of morpholines, and a member of acetamides.[11] Its identity as the first clinically approved oxazolidinone is a hallmark of its novelty and importance in overcoming resistance to other antibiotic classes.[11]
- Structural Identifiers: For precise computational and chemical representation, the following identifiers are used:
- InChI: InChI=1S/C16H20FN3O4/c1-11(21)18-9-13-10-20(16(22)24-13)12-2-3-15(14(17)8-12)19-4-6-23-7-5-19/h2-3,8,13H,4-7,9-10H2,1H3,(H,18,21)/t13-/m0/s1.[1]
- InChIKey: TYZROVQLWOKYKF-ZDUSSCGKSA-N.[1]
- SMILES: CC(=O)NC[C@H]1CN(C(=O)O1)C2=CC(=C(C=C2)N3CCOCC3)F.[11]
Formulations, Physical Characteristics, and Stability
Linezolid's clinical flexibility is greatly enhanced by its availability in multiple formulations suitable for both parenteral and oral administration.[6]
- Available Formulations:
- Intravenous (IV) Injection: Supplied as a sterile, isotonic, ready-to-use solution at a concentration of 2 mg/mL in single-use infusion bags (e.g., 100 mL, 300 mL).[6] The solution may exhibit a yellow color that can intensify over time, which does not adversely affect its potency.[16]
- Oral Tablets: Available as film-coated 600 mg tablets.[6] A 400 mg tablet was previously available but has been discontinued.[18]
- Oral Suspension: Provided as an orange-flavored powder for reconstitution by a pharmacist to a final concentration of 100 mg/5 mL.[6] This formulation contains sucrose and aspartame, which yields phenylalanine upon metabolism. This is a critical consideration for patients with phenylketonuria (PKU).[3]
- Physical Appearance: In its pure form, Linezolid is a crystalline solid. In commercial preparations, it may appear as a solid or a lyophilized cake or powder, typically pale yellow to light brown in color.[19]
- Solubility: Linezolid exhibits poor solubility in aqueous solutions, with a reported solubility of only 0.1 mg/mL in phosphate-buffered saline (PBS) at pH 7.2. It is more soluble in organic solvents like dimethyl sulfoxide (DMSO) and ethanol.[12] This inherent low water solubility necessitates the specific isotonic formulation for intravenous use.
- Stability and Storage: Linezolid should be stored at room temperature (25°C or 77°F), protected from light and freezing.[12] The reconstituted oral suspension is stable for 21 days when stored at room temperature and should be discarded after this period, regardless of the remaining volume.[21] Intravenous bags should be kept in their protective overwrap until they are ready for use to maintain sterility and protect from light.[16]
The availability of a highly bioavailable oral formulation is not merely a matter of convenience; it stands as a cornerstone of Linezolid's clinical and pharmacoeconomic value. The pharmacokinetic data confirm that oral administration achieves approximately 100% bioavailability, meaning the systemic exposure from an oral dose is virtually identical to that from an intravenous dose of the same magnitude.[1] This allows for a direct, dose-equivalent switch from IV to oral therapy without the need for complex adjustments. This "IV-to-PO switch" capability is a fundamental tenet of modern antimicrobial stewardship programs. It enables clinicians to transition patients off parenteral therapy sooner, which can facilitate earlier hospital discharge compared to treatment with IV-only antibiotics like vancomycin or daptomycin. This, in turn, leads to substantial reductions in total healthcare costs by minimizing hospital bed days, nursing time for IV administration, and the costs and risks associated with maintaining intravenous access.[9] Therefore, the physicochemical property of high oral bioavailability, combined with its formulation diversity, translates directly into a significant clinical and economic advantage, positioning Linezolid as a strategic tool for optimizing patient care pathways and healthcare resource utilization.
Pharmacology and Mechanism of Action
This section details the molecular interactions of Linezolid with its bacterial target and describes its absorption, distribution, metabolism, and excretion (ADME) profile within the human body.
Pharmacodynamics
Pharmacodynamics describes the biochemical and physiological effects of the drug on the target organism. Linezolid's unique mechanism of action is central to its efficacy against resistant pathogens.
Mechanism of Action
Linezolid is a bacteriostatic or bactericidal agent, depending on the organism, that functions as a protein synthesis inhibitor within the oxazolidinone class.[1] Its mechanism is distinct from that of other ribosome-targeting antibiotics.
The drug exerts its effect by binding to a specific site on the 23S ribosomal RNA (rRNA) component of the bacterial 50S subunit.[3] This binding site is located within the peptidyl transferase center (PTC), the ribosomal catalytic core responsible for peptide bond formation.[23] By occupying this critical location, Linezolid physically obstructs the assembly of the components required for translation to begin. Specifically, it
prevents the formation of a functional 70S initiation complex, which consists of the 30S subunit, the 50S subunit, messenger RNA (mRNA), and the initiator tRNA, formylmethionyl-tRNA (tRNAfMet).[4]
This blockade of the initiation step is a crucial point of differentiation. Most other protein synthesis inhibitors, such as macrolides, aminoglycosides, and tetracyclines, act at later stages of translation, primarily during the elongation phase where the polypeptide chain is built.[1] Because Linezolid's target site and mechanism are unique, there is no cross-resistance with these other antibiotic classes, allowing it to remain effective against bacteria that have developed resistance to them.[24]
Antimicrobial Activity
The clinical effect of Linezolid varies depending on the target pathogen.
- It is generally considered bacteriostatic (inhibits growth) against staphylococci, including MRSA, and enterococci, including VRE.[11]
- In contrast, it exhibits bactericidal (kills bacteria) activity against the majority of streptococcal strains, such as Streptococcus pneumoniae.[11]
Furthermore, Linezolid demonstrates a significant post-antibiotic effect (PAE), meaning its bacterial growth suppression continues for a period even after the drug's concentration in the blood falls below the minimum inhibitory concentration (MIC) for the pathogen.[25] This contributes to the overall efficacy of its dosing regimens.
Pharmacokinetic/Pharmacodynamic (PK/PD) Indices
The efficacy and safety of Linezolid are closely linked to specific exposure-response relationships, known as PK/PD indices.
- Efficacy Target: The primary PK/PD parameter that correlates with clinical and microbiological success is the ratio of the Area Under the Concentration-Time Curve over a 24-hour period to the Minimum Inhibitory Concentration (AUC0–24/MIC). A target AUC0–24/MIC ratio of 80 to 120 is widely accepted as necessary for optimal efficacy.[5] An alternative target, particularly for time-dependent antibiotics, is the percentage of the dosing interval during which the drug concentration remains above the MIC (
T>MIC). A target of >85% T>MIC has also been proposed to maximize efficacy and prevent the emergence of resistance.[5]
- Toxicity Target: The trough plasma concentration (Cmin), or the lowest concentration reached before the next dose, is a critical marker for toxicity. A steady-state Cmin consistently above 7 mg/L has been significantly correlated with an increased risk of adverse events, particularly hematological toxicity like thrombocytopenia.[5] For long-term treatment, such as in multidrug-resistant tuberculosis, an even lower threshold of
Cmin>2 mg/L has been associated with mitochondrial toxicity.[29] Consequently, a therapeutic window for
Cmin of 2 to 7 mg/L is often targeted in clinical practice to balance efficacy and safety.[5]
Pharmacokinetics (ADME Profile)
Pharmacokinetics describes the movement of the drug through the body, encompassing its absorption, distribution, metabolism, and excretion (ADME). Linezolid possesses a predictable and favorable pharmacokinetic profile.
Table 2: Summary of Linezolid Pharmacokinetic Parameters in Adults
Parameter | Value | Source(s) |
---|
Oral Bioavailability | ~100% | 1 |
Time to Peak Concentration (Tmax) | 1–2 hours (oral) | 4 |
Plasma Protein Binding | ~31% | 1 |
Volume of Distribution (Vd) | 40–50 L | 4 |
Metabolism Pathway | Non-enzymatic oxidation of morpholine ring (CYP independent) | 1 |
Major Metabolites | PNU-142586, PNU-142300 (inactive) | 4 |
Primary Route of Excretion | Renal (30% parent drug, 50% metabolites) and Non-renal (65% of total clearance) | 15 |
Elimination Half-life (t1/2) | 3–7 hours | 1 |
Absorption
Linezolid is rapidly and extensively absorbed after oral administration, with an absolute bioavailability of approximately 100%.[1] This near-complete absorption means that oral and intravenous doses are bioequivalent, allowing for interchangeability without dose adjustment. Peak plasma concentrations (
Cmax) are typically reached within 1 to 2 hours following an oral dose.[4] While administration with a high-fat meal may delay the time to peak concentration and slightly reduce the peak level, the total drug exposure (AUC) remains unchanged. Therefore, Linezolid can be administered without regard to the timing of meals, adding to its clinical convenience.[2]
Distribution
Linezolid distributes widely throughout the body. Its volume of distribution (Vd) in healthy adults is approximately 40 to 50 liters, a value close to that of total body water, which is indicative of excellent tissue penetration.[4] This is further facilitated by its low plasma protein binding of about 31%, which is primarily to albumin and is independent of drug concentration.[1] This low binding leaves a high fraction of the drug free to diffuse from the bloodstream into tissues.
Clinical studies have confirmed excellent penetration into various body sites, including bone, fat, muscle, and inflammatory fluid in skin blisters, where concentrations can exceed those in the plasma.[2] Critically for its approved indications, Linezolid achieves high concentrations in the epithelial lining fluid and alveolar macrophages of the lungs, with a fluid-to-plasma ratio reported as high as 3.2:1, supporting its efficacy in treating pneumonia.[2] It also penetrates the central nervous system effectively, achieving a cerebrospinal fluid (CSF) to plasma ratio of approximately 0.7:1 even in the absence of meningeal inflammation, making it a viable option for certain CNS infections.[1]
Metabolism
Linezolid undergoes metabolism primarily through a non-enzymatic chemical oxidation of its morpholine ring.[4] This metabolic pathway is a significant clinical advantage because it is
not dependent on the Cytochrome P450 (CYP) enzyme system.[1] Many commonly used medications are substrates, inhibitors, or inducers of CYP enzymes, making this system a major source of pharmacokinetic drug-drug interactions. By bypassing this pathway, Linezolid avoids a vast category of these interactions, which is particularly beneficial in critically ill or polymedicated patients who often receive complex drug regimens. This metabolic profile makes Linezolid a more predictable agent from a pharmacokinetic standpoint, reducing the risk of unexpected alterations in its own exposure or that of co-administered drugs.
The oxidation process yields two major metabolites, both of which are microbiologically inactive:
- PNU-142586 (hydroxyethyl glycine metabolite)
- PNU-142300 (aminoethoxyacetic acid metabolite).[4]
PNU-142586 is the predominant metabolite found in urine.25
Excretion
Linezolid is eliminated from the body through both renal and non-renal pathways. Non-renal clearance, primarily through metabolism, accounts for the majority of elimination, approximately 65% of the total drug clearance.[15] The remaining 35% is cleared by the kidneys.
At steady state, urine is the major route of excretion for the drug and its metabolites. Approximately 30% of an administered dose is excreted in the urine as unchanged parent drug, while about 40% is excreted as metabolite PNU-142586 and 10% as metabolite PNU-142300.[25] Very little parent drug is recovered from the feces, although small amounts of the metabolites are found there.[15] The elimination half-life (
t1/2) of Linezolid in adults with normal renal function is in the range of 3 to 7 hours.[1]
The delicate balance between the PK/PD targets for efficacy (AUC0–24/MIC of 80-120) and toxicity (Cmin < 7 mg/L) establishes a narrow therapeutic window for Linezolid. This is compounded by evidence of high interindividual pharmacokinetic variability, especially in critically ill patient populations where factors such as renal function, hepatic function, body weight, and severity of illness can significantly alter drug exposure.[5] A standard dosing regimen, such as 600 mg every 12 hours, may therefore result in sub-therapeutic concentrations in some patients, risking treatment failure and the development of resistance, while causing supra-therapeutic, toxic concentrations in others. This "one-size-fits-most" approach is often inadequate for a drug with such characteristics in complex clinical scenarios. This reality builds a powerful case for the routine implementation of Therapeutic Drug Monitoring (TDM). TDM allows for the individualization of dosing regimens based on measured serum concentrations, thereby maximizing the probability of achieving efficacy targets while minimizing the risk of dose-related toxicities. This practice is a key recommendation for antimicrobial stewardship programs aiming to optimize Linezolid therapy.
Clinical Efficacy and Therapeutic Applications
This section outlines the antimicrobial spectrum of Linezolid, its approved and investigational clinical uses, and the mechanisms by which bacteria develop resistance.
Antimicrobial Spectrum
Linezolid's spectrum of activity is focused almost exclusively on Gram-positive bacteria, with particular importance against multidrug-resistant strains.
- Gram-Positive Activity: Linezolid demonstrates potent in vitro activity against a broad range of clinically significant aerobic Gram-positive pathogens.[1] Its spectrum includes:
- Staphylococcus aureus: Active against both methicillin-susceptible (MSSA) and methicillin-resistant (MRSA) isolates.[1]
- Enterococcus faecium: Active against both vancomycin-susceptible and vancomycin-resistant (VRE) isolates.[1] It also has activity against
Enterococcus faecalis.[3]
- Streptococci: Active against Streptococcus pneumoniae (including penicillin-resistant strains), Streptococcus pyogenes (Group A streptococci), and Streptococcus agalactiae (Group B streptococci).[1]
- Coagulase-Negative Staphylococci: Active against species such as Staphylococcus epidermidis and Staphylococcus haemolyticus.[3]
- Mycobacterial Activity: Linezolid possesses significant activity against Mycobacterium tuberculosis (Mtb) and is a core component of WHO-recommended treatment regimens for multidrug-resistant (MDR-TB) and extensively drug-resistant (XDR-TB) tuberculosis.[1] It is also active against several species of non-tuberculous mycobacteria (NTM).[31]
- Anaerobic and Gram-Negative Activity: While some minimal in vitro activity has been observed against certain anaerobic and Gram-negative bacteria, Linezolid is not considered clinically efficacious against these organisms and is not indicated for their treatment.[3] If a mixed infection involving Gram-negative pathogens is documented or suspected, concomitant, specific anti-Gram-negative therapy is required.[7]
FDA-Approved Indications
The U.S. Food and Drug Administration (FDA) has approved Linezolid for the treatment of specific infections in both adult and pediatric populations when caused by susceptible strains of the designated microorganisms.[3]
- Nosocomial Pneumonia (Hospital-Acquired Pneumonia): For infections caused by S. aureus (both MRSA and MSSA) or S. pneumoniae.[1] Its excellent penetration into lung tissues provides a strong rationale for this indication.[1]
- Community-Acquired Pneumonia (CAP): For infections caused by S. pneumoniae (including cases with concurrent bacteremia) or S. aureus (MSSA isolates only).[1] Clinical guidelines recommend reserving its use for cases where MRSA is the confirmed or highly suspected pathogen.[1]
- Complicated Skin and Skin Structure Infections (cSSSI): For infections caused by S. aureus (MRSA and MSSA), S. pyogenes, or S. agalactiae. This indication includes diabetic foot infections, provided there is no concomitant osteomyelitis.[1]
- Vancomycin-Resistant Enterococcus faecium (VRE) Infections: This is a primary and critical indication for Linezolid, and the approval includes cases complicated by concurrent bacteremia.[1]
- Limitations of Use: The FDA label carries important limitations. A mortality imbalance was observed in an investigational study of patients with catheter-related bloodstream infections (CRBSI), and as a result, Linezolid is not indicated for the treatment of CRBSI or catheter-site infections.[3] This finding underscores a crucial principle in antimicrobial therapy:
in vitro susceptibility does not always guarantee clinical success for a specific infection type. The reasons for this failure in CRBSI are not fully understood but may involve factors like poor activity against bacteria within biofilms on catheter surfaces. This highlights that treatment decisions must be guided by high-quality clinical evidence for specific syndromes, not solely by laboratory susceptibility reports. Additionally, the safety and efficacy of Linezolid use for durations longer than 28 days have not been established in controlled clinical trials.[16]
Off-Label and Investigational Uses
Beyond its approved indications, Linezolid is used in several other challenging clinical scenarios, typically when other options have failed or are unsuitable.
- Drug-Resistant Tuberculosis (DR-TB): Linezolid is a cornerstone of modern treatment regimens for MDR-TB and XDR-TB. However, the long treatment courses required (often months) are frequently complicated by its significant duration-dependent toxicities, particularly myelosuppression and neuropathy, which often necessitate dose reduction or discontinuation.[1]
- Bone and Joint Infections: There is accumulating evidence, albeit of low-to-medium quality, supporting its use for difficult-to-treat bone and joint infections, including chronic osteomyelitis.[1] Its good bone penetration is an advantage, but the necessity for long-term therapy raises major concerns about adverse effects.[3]
- Infective Endocarditis: It is considered a reasonable therapeutic option for endocarditis caused by multi-resistant Gram-positive bacteria, such as VRE or MRSA, often as part of salvage therapy. However, high-quality evidence from randomized trials is lacking, and its bacteriostatic activity against staphylococci and enterococci is a theoretical limitation for such a severe, deep-seated infection.[1]
- Central Nervous System (CNS) Infections: Due to its excellent penetration into the CSF, Linezolid is a valuable option for treating CNS infections like meningitis or brain abscesses caused by susceptible Gram-positive organisms, particularly MRSA.[3]
Mechanisms of Bacterial Resistance
Although resistance to Linezolid remains relatively uncommon, its emergence is a growing concern, particularly in settings of prolonged use.[1] The resistance mechanisms predominantly involve alterations at the drug's binding site on the ribosome.
- Target Site Mutations:
- 23S rRNA Gene Mutations: The most frequently identified mechanism is the acquisition of point mutations in the V domain of the 23S rRNA genes, with the G2576T substitution (E. coli numbering) being the most common.[23] Bacteria possess multiple copies of these genes, and the level of resistance often correlates directly with the proportion of mutated copies.[24]
- Ribosomal Protein Mutations: Mutations in the genes encoding ribosomal proteins L3 (gene rplC) and L4 (gene rplD), which are located near the PTC, can also reduce Linezolid susceptibility. The Cys154Arg mutation in the L3 protein is a notable mechanism of resistance in M. tuberculosis.[23]
- Target Site Modification by Cfr Methyltransferase:
- A highly significant and concerning mechanism is the acquisition of the cfr (chloramphenicol-florfenicol resistance) gene. This gene encodes an enzyme, a methyltransferase, that modifies the 23S rRNA at a key nucleotide (adenosine at position 2503) within the Linezolid binding site.[23] This methylation sterically hinders the binding of Linezolid.
- The cfr gene confers a broad multidrug-resistance phenotype known as PhLOPSA, providing resistance to Phenicols, Lincosamides, Oxazolidinones, Pleuromutilins, and Streptogramin A antibiotics.[24]
- The emergence of cfr represents a paradigm shift in the threat of Linezolid resistance. Unlike chromosomal mutations, which are typically passed down "vertically" during bacterial replication and may carry a fitness cost, the cfr gene is often located on mobile genetic elements like plasmids.[23] This allows it to be transferred "horizontally" between bacteria, including across different species. This transforms Linezolid resistance from a sporadic, slowly evolving issue into a potential epidemic threat that can spread rapidly within a healthcare environment. The potential for horizontal transfer of high-level resistance elevates the importance of vigilant infection control and active microbiological surveillance to prevent outbreaks of difficult-to-treat Gram-positive infections.
- Other Mechanisms: Non-ribosomal mechanisms are considered rare but have been described. These include the increased expression of ABC transporter efflux pumps, which actively pump the drug out of the bacterial cell, as seen in some strains of S. pneumoniae.[23]
Dosing and Administration
This section provides a comprehensive guide to the appropriate dosing and administration of Linezolid for its approved indications in various patient populations, based on FDA-approved labeling and clinical guidelines.
Table 3: Dosing Regimens for Linezolid in Adult and Pediatric Populations
Indication | Patient Population | Recommended Dose | Route(s) | Frequency | Usual Duration |
---|
Nosocomial Pneumonia | Adults & Adolescents (≥12 yrs) | 600 mg | IV or Oral | Every 12 hours | 10–14 days |
| Pediatrics (<12 yrs) | 10 mg/kg | IV or Oral | Every 8 hours | 10–14 days |
Community-Acquired Pneumonia | Adults & Adolescents (≥12 yrs) | 600 mg | IV or Oral | Every 12 hours | 10–14 days |
| Pediatrics (<12 yrs) | 10 mg/kg | IV or Oral | Every 8 hours | 10–14 days |
Complicated Skin & Skin Structure Infections (cSSSI) | Adults & Adolescents (≥12 yrs) | 600 mg | IV or Oral | Every 12 hours | 10–14 days |
| Pediatrics (<12 yrs) | 10 mg/kg | IV or Oral | Every 8 hours | 10–14 days |
Uncomplicated Skin & Skin Structure Infections (uSSSI) | Adults | 400 mg | Oral | Every 12 hours | 10–14 days |
| Adolescents (≥12 yrs) | 600 mg | Oral | Every 12 hours | 10–14 days |
| Pediatrics (5–11 yrs) | 10 mg/kg | Oral | Every 12 hours | 10–14 days |
| Pediatrics (<5 yrs) | 10 mg/kg | Oral | Every 8 hours | 10–14 days |
Vancomycin-Resistant E. faecium Infections | Adults & Adolescents (≥12 yrs) | 600 mg | IV or Oral | Every 12 hours | 14–28 days |
| Pediatrics (<12 yrs) | 10 mg/kg | IV or Oral | Every 8 hours | 14–28 days |
Data compiled from.3 | | | | | |
Adult and Adolescent (≥12 years) Dosing Regimens
For most approved indications, including nosocomial pneumonia, community-acquired pneumonia, cSSSI, and VRE infections, the standard dose for adults and adolescents is 600 mg administered every 12 hours (q12h).[6] This dose can be given either intravenously or orally due to the drug's excellent bioavailability. For uncomplicated skin and skin structure infections (uSSSI), a lower dose of
400 mg orally every 12 hours is an approved option for adults, while adolescents should still receive the 600 mg dose.[3] The duration of therapy typically ranges from 10 to 14 days for pneumonia and skin infections, and is extended to 14 to 28 days for VRE infections.[6]
Pediatric (Birth through 11 years) Dosing Regimens
Dosing in pediatric patients is weight-based and administered more frequently than in adults. This adjustment is a direct clinical application of developmental pharmacokinetics. Studies have shown that children have a greater plasma clearance and a larger volume of distribution relative to their body size compared to adults.[2] This means they eliminate the drug more rapidly and require more frequent dosing to achieve and maintain therapeutic drug concentrations necessary for efficacy (i.e., to meet the
AUC/MIC and %T>MIC targets). Applying adult dosing intervals to children would likely result in underdosing and an increased risk of therapeutic failure.
For most serious infections, including pneumonia, cSSSI, and VRE infections, the recommended pediatric dose is 10 mg/kg every 8 hours (q8h), which can be given IV or orally.[6] For uSSSI, the dosing frequency varies by age: children under 5 years receive 10 mg/kg orally q8h, while children aged 5 to 11 years receive 10 mg/kg orally every 12 hours.[3]
Administration Guidelines
Proper administration is key to ensuring the safety and efficacy of Linezolid therapy.
- Intravenous (IV) Administration: The IV solution is supplied in ready-to-use infusion bags and should be administered over a period of 30 to 120 minutes.[3] It should not be mixed with other medications in the same IV line. Prior to administration, the bag should be visually inspected for particulate matter and checked for leaks by squeezing it firmly. If leaks or particulates are found, the solution should be discarded as sterility may be compromised.[16]
- Oral Administration: Both the tablets and the oral suspension can be taken with or without food.[4] Taking the medication with food may help lessen the common side effects of nausea and vomiting.[22] The oral suspension must be constituted by a pharmacist before dispensing. It is important to use an accurate oral dosing syringe to measure the dose, as household spoons are not accurate. The suspension does not need to be shaken before each use. Any unused portion of the reconstituted suspension must be discarded after 21 days.[21]
Safety, Tolerability, and Risk Management
The clinical utility of Linezolid is defined as much by its safety profile and potential for interactions as it is by its efficacy. A thorough understanding of its risks is essential for appropriate patient selection and monitoring.
Adverse Drug Reactions
Linezolid is generally well-tolerated for short courses, but the risk of significant toxicity increases with the duration of therapy.
Common Side Effects (≥2% incidence)
The most frequently reported adverse events associated with short-term Linezolid use are primarily gastrointestinal and neurological. These include diarrhea (reported in 4-11% of patients), nausea (3-7%), headache (2-6%), vomiting, and rash.[1] Tongue discoloration has also been reported.[35]
Serious Adverse Events and Toxicities
The safety profile of Linezolid reveals a clear "duration-toxicity" relationship that fundamentally governs its clinical application. While relatively safe for the 10-14 day courses typical for its primary indications, its most severe toxicities emerge with prolonged use. This positions Linezolid as an optimal agent for acute infections but a high-risk choice for chronic conditions requiring long-term therapy. This risk-benefit calculation is central to responsible antimicrobial stewardship.
- Myelosuppression: This is the most significant duration-dependent toxicity. It manifests as reversible anemia, leukopenia, pancytopenia, and most commonly, thrombocytopenia (low platelet count).[1] The risk of myelosuppression becomes clinically significant in patients receiving treatment for
more than two weeks.[1] The condition is generally reversible upon discontinuation of the drug.[35] Patients with pre-existing renal or hepatic impairment are at a higher risk for developing these hematological complications.[3]
- Peripheral and Optic Neuropathy: This is a serious, and in some cases irreversible, toxicity associated with long-term Linezolid administration, typically with courses lasting longer than 28 days.[1] Peripheral neuropathy usually presents as paresthesia (numbness or tingling) in the extremities. Optic neuropathy can manifest as blurred vision, changes in visual acuity, or defects in the visual field or color vision.[3] Any patient reporting new visual symptoms while on Linezolid requires a prompt ophthalmic evaluation.[3]
- Serotonin Syndrome: Linezolid is a weak, reversible, non-selective inhibitor of monoamine oxidase (MAO) enzymes A and B.[4] This property can lead to an accumulation of serotonin in the central nervous system when Linezolid is co-administered with other serotonergic medications, resulting in the potentially life-threatening condition known as serotonin syndrome. The classic triad of symptoms includes altered mental status (e.g., confusion, agitation), autonomic hyperactivity (e.g., fever, tachycardia, diaphoresis), and neuromuscular abnormalities (e.g., tremor, myoclonus, hyperreflexia).[6] The risk is highest with concurrent use of drugs like selective serotonin reuptake inhibitors (SSRIs), tricyclic antidepressants (TCAs), triptans, and certain opioids like meperidine.[6]
- Lactic Acidosis: A rare but severe metabolic adverse event has been reported. It is believed to be a manifestation of mitochondrial toxicity, as Linezolid can inhibit mitochondrial protein synthesis.[1] Patients who develop recurrent nausea, vomiting, and unexplained acidosis while on therapy should be evaluated for this condition.[7]
- Hypertension: Due to its MAO inhibitory activity, Linezolid can cause a significant increase in blood pressure, particularly when co-administered with sympathomimetic agents (e.g., pseudoephedrine) or when patients ingest large amounts of tyramine-rich foods.[7]
- Clostridioides difficile-Associated Diarrhea (CDAD): As with nearly all broad-spectrum antibiotics, Linezolid alters the normal colonic flora, which can lead to the overgrowth of C. difficile. This can result in a spectrum of illness ranging from mild diarrhea to severe, life-threatening pseudomembranous colitis.[7]
Drug and Food Interactions
Linezolid's MAO-inhibitory property is the source of its most critical and unique interactions.
Table 4: Clinically Significant Drug and Food Interactions with Linezolid
Interacting Agent/Class | Mechanism of Interaction | Potential Clinical Effect | Management Recommendation | Source(s) |
---|
Serotonergic Agents (SSRIs, TCAs, Triptans, Meperidine, Buspirone) | MAO-A Inhibition → Increased Serotonin Levels | Serotonin Syndrome | Contraindicated or use only if essential. Discontinue serotonergic agent if possible and monitor closely for 2-5 weeks. | 6 |
Adrenergic Agents (Pseudoephedrine, Phenylephrine, Epinephrine, Dopamine) | MAO Inhibition → Potentiation of Pressor Effects | Hypertensive Crisis | Contraindicated or use with extreme caution and blood pressure monitoring. | 6 |
Tyramine-Rich Foods/Beverages (Aged cheeses, cured meats, fermented foods, red wine) | MAO Inhibition → Prevents Tyramine Breakdown | Hypertensive Reaction | Advise patients to avoid consuming large quantities of these items. | 39 |
Hypoglycemic Agents (Insulin, Sulfonylureas) | Unknown; potential for causing hypoglycemia | Hypoglycemia | Use with caution; enhance blood glucose monitoring in diabetic patients. | 3 |
Myelosuppressive Agents (Chemotherapy, Clozapine, Cladribine) | Additive Bone Marrow Suppression | Severe Myelosuppression (Thrombocytopenia, Anemia) | Use with caution; increase frequency of hematological monitoring. | 3 |
Contraindications and Precautions
- Contraindications:
- Known hypersensitivity to Linezolid or any of its excipients.[17]
- Concurrent use of any drug that inhibits monoamine oxidase A or B (e.g., phenelzine, isocarboxazid), or use within two weeks of taking any such drug.[3]
- Precautions/Warnings: Linezolid should be used with special caution, or not at all unless patients can be closely monitored, in those with uncontrolled hypertension, pheochromocytoma, thyrotoxicosis, or carcinoid syndrome.[7]
Monitoring Recommendations
Vigilant monitoring is required to mitigate the risks associated with Linezolid therapy.
- Complete Blood Count (CBC): A CBC with differential and platelet count should be monitored weekly, especially for patients receiving therapy for more than two weeks, those with pre-existing myelosuppression, or those with underlying renal or hepatic impairment.[3]
- Neurological and Visual Function: Patients on long-term therapy should be regularly questioned about and assessed for symptoms of peripheral or optic neuropathy.[3]
- Blood Pressure: Should be monitored in patients with pre-existing hypertension or those receiving concomitant adrenergic agents.[3]
- Serotonin Syndrome: Clinicians must maintain a high index of suspicion for serotonin syndrome in any patient receiving Linezolid with a serotonergic agent. Patients should be educated on the symptoms and instructed to seek immediate medical attention if they occur.[3]
- Blood Glucose: Blood glucose levels should be monitored more frequently in diabetic patients.[3]
Use in Special Populations
The use of Linezolid requires special consideration in certain patient populations due to altered pharmacokinetics or increased risk of toxicity.
Renal and Hepatic Impairment
The official recommendation of "no dose adjustment" for Linezolid in patients with renal impairment is a potentially misleading oversimplification that belies a more complex clinical reality. While pharmacokinetic studies confirm that the clearance of the parent drug is not significantly affected by renal function, these same studies show that its two major, inactive metabolites (PNU-142586 and PNU-142300) accumulate to very high levels in patients with renal insufficiency.[15] There is a clear disconnect between the pharmacokinetic recommendation, which is based solely on the parent drug, and the clinical safety data, which consistently show that patients with renal impairment have a significantly higher incidence of thrombocytopenia.[3] The exact cause of this increased toxicity is not fully elucidated but may be related to the high concentrations of metabolites or other underlying patient factors. This discrepancy means that the simple "no adjustment needed" guideline can provide a false sense of security. A more nuanced and safer clinical approach is warranted: while the parent drug dose may not require initial adjustment, patients with renal impairment must be recognized as a high-risk group requiring heightened clinical vigilance and more frequent hematological monitoring. For prolonged therapy in this population, therapeutic drug monitoring (TDM) or even an empirical dose reduction should be strongly considered to mitigate the elevated risk of toxicity.
- Renal Impairment: No dosage adjustment for Linezolid is recommended based on renal function, including for patients with end-stage renal disease (ESRD) on hemodialysis.[1] However, as noted, the inactive metabolites accumulate, and the risk of thrombocytopenia is increased.[3] For patients undergoing hemodialysis, Linezolid is dialyzable; approximately one-third of a dose is removed during a standard session. Therefore, it is recommended that Linezolid be administered
after the hemodialysis session is complete.[15]
- Hepatic Impairment: No dosage adjustment is required for patients with mild-to-moderate hepatic insufficiency (Child-Pugh class A or B).[1] Data for severe hepatic impairment are limited. Similar to patients with renal dysfunction, those with hepatic impairment are also at an increased risk of developing thrombocytopenia.[3] Animal models suggest that hepatic impairment can increase overall Linezolid exposure by reducing its metabolic clearance.[30]
Geriatric Patients
The pharmacokinetic profile of Linezolid is generally considered to be unaltered in elderly patients, and routine dose adjustment based on age is not recommended.[2] However, clinical experience suggests that geriatric patients may be more sensitive to the effects of the medication.[21] One pharmacokinetic study found that Linezolid trough concentrations were dramatically higher in elderly patients (increasing by approximately 10 mg/L in those aged 65-80), leading to the recommendation that TDM should be considered in this population to avoid accumulation and potential toxicity.[42]
Pregnancy and Lactation
- Pregnancy: Linezolid is classified as US FDA Pregnancy Category C and Australia TGA Category B3, indicating that risk cannot be ruled out.[1] There are no adequate and well-controlled studies in pregnant women. Animal reproduction studies have not shown evidence of teratogenicity, but did demonstrate embryo and fetal toxicities, such as decreased fetal weight and reduced pup survival, at exposure levels that were also toxic to the mother.[43] Therefore, Linezolid should be used during pregnancy only if the potential benefit to the mother clearly justifies the potential risk to the fetus.[3]
- Lactation: Linezolid is known to be excreted into human breast milk.[44] Data from case studies are limited but suggest that a fully breastfed infant would receive a relatively small portion (estimated at 6-16%) of the standard maternal weight-adjusted dose, leading to low or undetectable serum levels in the infant.[44] The general consensus is that maternal need for Linezolid is not an absolute contraindication to breastfeeding. However, the infant should be carefully monitored for potential adverse effects on the gastrointestinal tract, such as diarrhea, vomiting, or candidiasis (thrush). Caution is particularly advised for nursing infants who are premature or younger than one month.[3]
Comparative Analysis: Linezolid vs. Vancomycin and Daptomycin
Linezolid, vancomycin, and daptomycin are three cornerstone antibiotics for the treatment of serious infections caused by resistant Gram-positive bacteria, particularly MRSA and VRE. The choice among these agents is a complex clinical decision that requires a multifactorial assessment of the patient, the pathogen, the site of infection, and the healthcare environment. There is no single "best" drug; rather, the optimal agent is the one that best fits the specific clinical scenario. This selection process represents a masterclass in personalized medicine and antimicrobial stewardship, highlighting the critical role of infectious diseases specialists and clinical pharmacists in guiding therapy. A formulary that unduly restricts access to any of these three agents can significantly hamper a clinician's ability to tailor treatment appropriately. Table 5 provides a comparative overview of their key attributes.
Table 5: Comparative Summary of Linezolid, Vancomycin, and Daptomycin
Attribute | Linezolid | Vancomycin | Daptomycin |
---|
Drug Class | Oxazolidinone | Glycopeptide | Cyclic Lipopeptide |
Mechanism of Action | Inhibits protein synthesis at initiation (binds 50S subunit) | Inhibits cell wall synthesis (binds D-Ala-D-Ala) | Disrupts cell membrane function (Ca²⁺-dependent) |
Key Spectrum | MRSA, VRE, S. pneumoniae, MDR-TB | MRSA, MSSA, Streptococci, C. difficile (oral only) | MRSA, VRE, MSSA, Streptococci |
Route(s) of Admin. | IV, Oral | IV (systemic), Oral (gut only) | IV only |
Oral Bioavailability | ~100% | Negligible | Negligible |
Key PK/PD Parameter | AUC0–24/MIC | AUC0–24/MIC | AUC0–24/MIC |
Dosing Frequency (Adult) | Every 12 hours | Every 8–24 hours (variable) | Every 24 hours |
Renal Dose Adjustment | No (but metabolites accumulate) | Yes (mandatory) | Yes |
Key Toxicities | Myelosuppression, Neuropathy, Serotonin Syndrome | Nephrotoxicity, Ototoxicity, Infusion Reactions | Muscle Toxicity (Rhabdomyolysis) |
Required Monitoring | Weekly CBCs, Neuro/Vision exams | TDM (trough or AUC), Renal function | CPK levels, Renal function |
Use in Pneumonia | Yes (effective) | Yes (standard therapy) | No (inactivated by surfactant) |
Cost-Effectiveness | Often superior to vancomycin/daptomycin due to oral switch and reduced monitoring/LOS | Low drug cost but high monitoring and administration costs | High drug cost, less monitoring than vancomycin |
Data compiled from.1 | | | |
Head-to-Head Efficacy
- Complicated Skin & Skin Structure Infections (cSSSI): The evidence strongly supports Linezolid in this setting. Multiple meta-analyses of randomized controlled trials have found that Linezolid is associated with significantly better clinical and microbiological cure rates compared to vancomycin.[1] Economic analyses further suggest that Linezolid is a more cost-effective strategy than both vancomycin and daptomycin for cSSSI, largely due to its oral formulation facilitating earlier discharge.[10]
- Pneumonia (Nosocomial/VAP): The comparative efficacy in pneumonia is less clear-cut. Some studies, particularly in the context of ventilator-associated pneumonia (VAP) caused by MRSA, have suggested that Linezolid is superior to vancomycin. This potential advantage is often attributed to Linezolid's significantly better penetration into bronchial fluids and lung tissue compared to vancomycin.[1] However, other large analyses and meta-analyses have found no significant difference in overall treatment success or mortality rates between the two agents.[1] Current U.S. guidelines reflect this equipoise, recommending either Linezolid or vancomycin as appropriate first-line treatments for confirmed MRSA nosocomial pneumonia.[1] Daptomycin is
contraindicated for pneumonia because it is inactivated by pulmonary surfactant.[39]
- Bacteremia:
- VRE Bacteremia: The optimal treatment for VRE bacteremia is a subject of considerable debate. Several meta-analyses, primarily of retrospective observational studies, have concluded that treatment with Linezolid is associated with lower all-cause and infection-related mortality compared to daptomycin.[53] Conversely, at least one large, multicenter cohort study found that after adjusting for severity of illness, Linezolid was associated with
higher rates of microbiological failure and overall treatment failure compared to daptomycin.[32] The bacteriostatic nature of Linezolid against enterococci is a theoretical concern for severe, endovascular infections, which may favor a bactericidal agent like daptomycin.[32]
- MRSA Bacteremia: For MRSA bacteremia, meta-analyses of available studies have generally shown comparable effectiveness among Linezolid, vancomycin, and daptomycin with respect to clinical cure, microbiological cure, and mortality.[52]
Comparative Safety and Tolerability
The three drugs have distinct and non-overlapping primary toxicity profiles, which heavily influence drug selection based on patient comorbidities.
- Linezolid: The defining toxicities are duration-dependent myelosuppression and neuropathy, and the risk of serotonin syndrome from its MAO-inhibitory properties.[36] This makes it a less favorable choice for patients requiring long-term therapy or those on interfering serotonergic medications.
- Vancomycin: The signature toxicity is nephrotoxicity, or acute kidney injury (AKI). It can also cause ototoxicity and infusion-related reactions ("Red Man Syndrome").[50] Its use is complicated by the absolute requirement for therapeutic drug monitoring (TDM) to minimize toxicity while ensuring efficacy.[56] This makes it a poor choice for patients with pre-existing or unstable renal function.
- Daptomycin: The primary safety concern is muscle toxicity, ranging from myalgia to life-threatening rhabdomyolysis. This necessitates routine monitoring of creatine phosphokinase (CPK) levels.[39] It is generally well-tolerated otherwise and avoids the renal and hematological toxicities of its counterparts.
Pharmacokinetic and Administrative Advantages
- Route of Administration: This is a major point of differentiation. Linezolid is the only one of the three with excellent (~100%) oral bioavailability, which enables a seamless IV-to-oral switch strategy.[47] Vancomycin (for systemic use) and daptomycin are restricted to
intravenous administration only.[49]
- Dosing and Monitoring: Linezolid offers the simplicity of a fixed twice-daily dose for most adults. In contrast, vancomycin requires complex, individualized dosing based on weight and renal function, with mandatory and costly TDM.[55] Daptomycin offers the convenience of once-daily dosing but still requires dose adjustment for significant renal impairment.[57]
- Use in Renal Impairment: Linezolid does not require dose adjustment in renal failure, making it a preferred agent in this population. Both vancomycin and daptomycin require significant dose and/or interval adjustments in patients with impaired renal function.[34]
Cost-Effectiveness Analysis
- Drug Acquisition Cost: Historically and currently, generic intravenous vancomycin has the lowest acquisition cost.[59] Generic oral Linezolid and intravenous daptomycin are considerably more expensive, although their costs have decreased significantly since their generic versions became available.[59]
- Total Cost of Care: When a broader, societal or hospital perspective is taken, the economic picture changes dramatically. Multiple analyses have concluded that Linezolid is often more cost-effective than IV vancomycin, particularly for treating cSSSI.[9] The cost savings are not from the drug itself but are driven by the avoidance of costs associated with prolonged IV therapy. These include:
- Reduced hospital length of stay, enabled by an early switch to oral therapy.
- Avoidance of costs related to IV line placement (e.g., PICC lines) and maintenance.
- Elimination of costs for outpatient parenteral antibiotic therapy (OPAT) services.
- Avoidance of the laboratory and pharmacist costs associated with mandatory vancomycin TDM.
- Linezolid vs. Daptomycin: In head-to-head economic comparisons, Linezolid has generally been found to be the more cost-effective option compared to daptomycin for treating MRSA cSSSI and bacteremia, with lower total direct medical costs.[10]
Conclusion and Expert Recommendations
Linezolid is a potent and clinically vital antibiotic that fundamentally changed the treatment landscape for resistant Gram-positive infections. Its development provided a reliable weapon against pathogens like MRSA and VRE, for which therapeutic options were dwindling. The unique mechanism of action, predictable pharmacokinetics, and exceptional oral bioavailability are profound clinical assets that distinguish it from other agents.
The primary challenge in harnessing Linezolid's power lies in navigating its significant safety profile. The drug's utility is intrinsically linked to its duration of use, with duration-dependent myelosuppression and neuropathy representing its greatest limitations. Furthermore, its weak MAO-inhibitory activity creates a constant risk of serious drug and food interactions. Consequently, the optimal use of Linezolid is for short, targeted courses for its approved indications, where its benefits are maximized and its risks are minimized.
Based on the comprehensive analysis of the available evidence, the following recommendations are made to ensure the judicious and safe use of Linezolid:
- Recommendations for Optimal Use:
- Patient Selection and Stewardship: The use of Linezolid should be reserved for infections proven or strongly suspected to be caused by susceptible MDR Gram-positive organisms, particularly MRSA and VRE.[1] It should not be used empirically when narrower-spectrum agents are likely to be effective. Clinicians should actively leverage its oral formulation to facilitate early de-escalation from IV therapy and prompt hospital discharge, which is a key principle of both antimicrobial and economic stewardship.
- Risk Mitigation and Monitoring: A rigorous approach to risk mitigation is mandatory. This must include weekly monitoring of complete blood counts for all patients, with increased vigilance for those on therapy beyond two weeks or with underlying renal/hepatic disease.[3] A thorough medication reconciliation must be performed before initiation to identify and manage potentially interacting serotonergic and adrenergic agents.[6] Patients must be educated on the symptoms of serotonin syndrome and neuropathy and counseled to avoid large quantities of tyramine-rich foods.[39]
- Therapeutic Drug Monitoring (TDM): For any patient requiring long-term therapy (e.g., for osteomyelitis or MDR-TB) or for critically ill patients with high pharmacokinetic variability, the implementation of TDM is strongly recommended. Monitoring trough concentrations to maintain them within the 2-7 mg/L therapeutic window can help individualize dosing to maximize efficacy while reducing the risk of toxicity.[5]
The story of Linezolid is a powerful illustration of the delicate balance in modern medicine. Its emergence provided solutions, but its use created new challenges. The rise of resistance mechanisms, particularly the transferable cfr gene, serves as a stark reminder of the fragility of our antibiotic arsenal.[23] This underscores the urgent and ongoing need for the development of novel antimicrobials and, perhaps more importantly, the universal and rigorous implementation of antimicrobial stewardship programs to preserve the effectiveness of the critical agents we are fortunate to have.
Works cited
- Linezolid - Wikipedia, accessed July 24, 2025, https://en.wikipedia.org/wiki/Linezolid
- Linezolid CAS#: 165800-03-3 - ChemicalBook, accessed July 24, 2025, https://m.chemicalbook.com/ProductChemicalPropertiesCB1287912_EN.htm
- Linezolid - StatPearls - NCBI Bookshelf, accessed July 24, 2025, https://www.ncbi.nlm.nih.gov/books/NBK539793/
- Linezolid: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed July 24, 2025, https://go.drugbank.com/drugs/DB00601
- A Review of Population Pharmacokinetic Analyses of Linezolid - PMC - PubMed Central, accessed July 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9192929/
- Zyvox (linezolid) dosing, indications, interactions, adverse effects ..., accessed July 24, 2025, https://reference.medscape.com/drug/zyvox-linezolid-342574
- ZYVOX® - Pfizer, accessed July 24, 2025, https://labeling.pfizer.com/ShowLabeling.aspx?id=12211
- Efficacy and safety of linezolid compared with other treatments for ..., accessed July 24, 2025, https://portlandpress.com/bioscirep/article/38/1/BSR20171125/57239/Efficacy-and-safety-of-linezolid-compared-with
- Rehospitalizations and Direct Medical Costs for cSSSI: Linezolid Versus Vancomycin, accessed July 24, 2025, https://www.pharmacytimes.com/view/rehospitalizations-and-direct-medical-costs-for-csssi-linezolid-versus-vancomycin
- Economic burden of inpatient and outpatient antibiotic treatment for ..., accessed July 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3782516/
- Linezolid | C16H20FN3O4 | CID 441401 - PubChem, accessed July 24, 2025, https://pubchem.ncbi.nlm.nih.gov/compound/Linezolid
- Linezolid | CAS#165800-03-3 | Antibiotic - MedKoo Biosciences, accessed July 24, 2025, https://www.medkoo.com/products/8345
- Linezolid, Protein synthesis inhibitor (CAS 165800-03-3) (ab141991) - Abcam, accessed July 24, 2025, https://www.abcam.com/en-us/products/biochemicals/linezolid-protein-synthesis-inhibitor-ab141991
- [Linezolid (200 mg)] - CAS [165800-03-3] - USP Store, accessed July 24, 2025, https://store.usp.org/product/1367561
- Zyvox (linezolid) tablet label - accessdata.fda.gov, accessed July 24, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2010/021130s022lbl.pdf
- LINEZOLID injection, for intravenous use - accessdata.fda.gov, accessed July 24, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2024/206473s010lbl.pdf
- ZYVOX® (linezolid) Injection, Tablets and Oral Suspension - accessdata.fda.gov, accessed July 24, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/021130s032,021131s026,021132s031lbl.pdf
- Generic Zyvox Availability - Drugs.com, accessed July 24, 2025, https://www.drugs.com/availability/generic-zyvox.html
- Linezolid (PNU 100766, CAS Number: 165800-03-3) | Cayman Chemical, accessed July 24, 2025, https://www.caymanchem.com/product/15012/linezolid
- Cubicin, INN-daptomycin - EMA, accessed July 24, 2025, https://www.ema.europa.eu/en/documents/product-information/cubicin-epar-product-information_en.pdf
- Linezolid (oral route) - Side effects & dosage - Mayo Clinic, accessed July 24, 2025, https://www.mayoclinic.org/drugs-supplements/linezolid-oral-route/description/drg-20067254
- What Is Linezolid (Zyvox)? Uses, Side Effects, Dosage - GoodRx, accessed July 24, 2025, https://www.goodrx.com/linezolid/what-is
- Resistance to Linezolid Caused by Modifications at Its Binding Site on the Ribosome - PMC, accessed July 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3264260/
- Linezolid Resistance in Staphylococci - PMC - PubMed Central, accessed July 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4036669/
- Pharmacokinetic and pharmacodynamic profile of linezolid in healthy volunteers and patients with Gram-positive infections, accessed July 24, 2025, https://academic.oup.com/jac/article-pdf/51/suppl_2/ii17/7503019/dkg248.pdf
- Pharmacokinetics of Linezolid in Subjects with Renal Dysfunction - PMC, accessed July 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC182634/
- Population Pharmacokinetics and Dosing Optimization of Linezolid in Pediatric Patients | Antimicrobial Agents and Chemotherapy - ASM Journals, accessed July 24, 2025, https://journals.asm.org/doi/10.1128/aac.02387-18
- Linezolid Pharmacokinetics in Critically Ill Patients: Continuous Versus Intermittent Infusion, accessed July 24, 2025, https://www.mdpi.com/2079-6382/13/10/961
- Pharmacokinetic analysis of linezolid for multidrug resistant tuberculosis at a tertiary care centre in Mumbai, India - Frontiers, accessed July 24, 2025, https://www.frontiersin.org/journals/pharmacology/articles/10.3389/fphar.2022.1081123/full
- Impact of hepatic impairment and renal failure on the pharmacokinetics of linezolid and its metabolites: contribution of hepatic metabolism and renal excretion | Antimicrobial Agents and Chemotherapy - ASM Journals, accessed July 24, 2025, https://journals.asm.org/doi/10.1128/aac.01892-24
- Mechanisms of Linezolid Resistance in Mycobacteria - MDPI, accessed July 24, 2025, https://www.mdpi.com/1424-8247/16/6/784
- Editorial Commentary: Linezolid vs Daptomycin for Vancomycin-Resistant Enterococci: The Evidence Gap Between Trials and Clinical Experience - PMC, accessed July 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4551011/
- What is the recommended dose of linezolid (Zyvox) for adults with normal renal function?, accessed July 24, 2025, https://www.droracle.ai/articles/164776/linozolid-dose-normal-renal-function-
- Linezolid Dosage Guide + Max Dose, Adjustments - Drugs.com, accessed July 24, 2025, https://www.drugs.com/dosage/linezolid.html
- Worldwide Assessment of Linezolid's Clinical Safety and Tolerability ..., accessed July 24, 2025, https://journals.asm.org/doi/10.1128/aac.47.6.1824-1831.2003
- Linezolid: safety and efficacy in special populations | Journal of Antimicrobial Chemotherapy | Oxford Academic, accessed July 24, 2025, https://academic.oup.com/jac/article/66/suppl_4/iv3/729596
- Linezolid and tedizolid adverse effects: a review on serotonin syndrome, myelosuppression, neuropathies, and lactic acidosis | Antimicrobial Stewardship & Healthcare Epidemiology - Cambridge University Press, accessed July 24, 2025, https://www.cambridge.org/core/journals/antimicrobial-stewardship-and-healthcare-epidemiology/article/linezolid-and-tedizolid-adverse-effects-a-review-on-serotonin-syndrome-myelosuppression-neuropathies-and-lactic-acidosis/9A20E63B355E2F557EED525312D7644A
- Linezolid Reviews and User Ratings: Effectiveness, Ease of Use, and Satisfaction - WebMD, accessed July 24, 2025, https://reviews.webmd.com/drugs/drugreview-18168-6154-linezolid-oral
- Zyvox vs. Daptomycin for Skin Infection and Bacterial Infection: Important Differences and Potential Risks. - GoodRx, accessed July 24, 2025, https://www.goodrx.com/compare/zyvox-vs-daptomycin
- Linezolid: Side Effects, Uses, Dosage, Interactions, Warnings - RxList, accessed July 24, 2025, https://www.rxlist.com/linezolid/generic-drug.htm
- www.mayoclinic.org, accessed July 24, 2025, https://www.mayoclinic.org/drugs-supplements/linezolid-oral-route/description/drg-20067254#:~:text=Appropriate%20studies%20performed%20to%20date%20have%20not%20demonstrated%20geriatric%2Dspecific,this%20medicine%20than%20younger%20adults.
- Therapeutic drug monitoring of linezolid and exploring optimal regimens and a toxicity-related nomogram in elderly patients: a multicentre, prospective, non-interventional study | Journal of Antimicrobial Chemotherapy | Oxford Academic, accessed July 24, 2025, https://academic.oup.com/jac/article-abstract/79/8/1938/7693234
- What are the effects of Linezolid (oxazolidinone antibiotic) on pregnancy? - Dr.Oracle AI, accessed July 24, 2025, https://www.droracle.ai/articles/70475/linezolid-in-pregnancy-
- Linezolid (Zyvox) Use During Pregnancy - Drugs.com, accessed July 24, 2025, https://www.drugs.com/pregnancy/linezolid.html
- www.merckmanuals.com, accessed July 24, 2025, https://www.merckmanuals.com/professional/infectious-diseases/bacteria-and-antibacterial-medications/linezolid-and-tedizolid#:~:text=Use%20of%20Linezolid%20During%20Pregnancy%20and%20Breastfeeding&text=Linezolid%20should%20be%20used%20during,use%20during%20breastfeeding%20is%20unknown.
- Linezolid - Drugs and Lactation Database (LactMed®) - NCBI Bookshelf, accessed July 24, 2025, https://www.ncbi.nlm.nih.gov/books/NBK501700/
- pmc.ncbi.nlm.nih.gov, accessed July 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3352102/#:~:text=The%20first%20major%20difference%20between,are%20limited%20to%20parenteral%20use.
- Cost-effectiveness analysis of linezolid, daptomycin, and vancomycin in methicillin-resistant Staphylococcus aureus: complicated skin and skin structure infection using Bayesian methods for evidence synthesis - PubMed, accessed July 24, 2025, https://pubmed.ncbi.nlm.nih.gov/21839399/
- Comparison of the pharmacokinetic properties of vancomycin, linezolid, tigecyclin, and daptomycin - PMC, accessed July 24, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3352102/
- A Review of Daptomycin vs. Vancomycin for Susceptible Infections: Is One Superior for - National Home Infusion Association, accessed July 24, 2025, https://nhia.org/wp-content/uploads/2023/04/InfusionJournalMarApr2023ReviewofDaptomycinvsVancomycin.pdf
- Daptomycin - StatPearls - NCBI Bookshelf, accessed July 24, 2025, https://www.ncbi.nlm.nih.gov/books/NBK470407/
- (PDF) Effectiveness and Safety of Linezolid Versus Vancomycin, Teicoplanin, or Daptomycin against Methicillin-Resistant Staphylococcus aureus Bacteremia: A Systematic Review and Meta-Analysis - ResearchGate, accessed July 24, 2025, https://www.researchgate.net/publication/369783075_Effectiveness_and_Safety_of_Linezolid_Versus_Vancomycin_Teicoplanin_or_Daptomycin_against_Methicillin-Resistant_Staphylococcus_aureus_Bacteremia_A_Systematic_Review_and_Meta-Analysis
- Systematic Review and Meta-Analysis of Linezolid versus Daptomycin for Treatment of Vancomycin-Resistant Enterococcal Bacteremia | Antimicrobial Agents and Chemotherapy - ASM Journals, accessed July 24, 2025, https://journals.asm.org/doi/10.1128/aac.01289-13
- Systematic Review and Meta-Analysis of Linezolid versus Daptomycin for Treatment of Vancomycin-Resistant Enterococcal Bacteremia | Antimicrobial Agents and Chemotherapy - ASM Journals, accessed July 24, 2025, https://journals.asm.org/doi/abs/10.1128/aac.01289-13
- Intravenous Vancomycin | Dosing Guidelines, Trough Levels, Side Effects | DoseMeRx, accessed July 24, 2025, https://doseme-rx.com/vancomycin/articles/intravenous
- Clinical Practice Guidelines : Vancomycin - The Royal Children's Hospital, accessed July 24, 2025, https://www.rch.org.au/clinicalguide/guideline_index/vancomycin/
- CUBICIN (daptomycin for injection) for Intravenous Use - accessdata.fda.gov, accessed July 24, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/021572s055lbl.pdf
- CUBICIN (daptomycin for injection) for Intravenous Use - accessdata.fda.gov, accessed July 24, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2015/021572s050lbl.pdf
- Cost Comparison of Oral Linezolid and Intravenous Vancomycin for Empiric Treatment of Suspected MRSA Pneumonia - Allen Press, accessed July 24, 2025, https://meridian.allenpress.com/jcphp/article-pdf/70/2/14/3095269/i2573-2765-70-2-14.pdf
- Counting the Cost of Daptomycin Versus Vancomycin in Hospitalized Patients: A Cost Minimization Analysis | Open Forum Infectious Diseases | Oxford Academic, accessed July 24, 2025, https://academic.oup.com/ofid/article/11/5/ofae217/7647033
- Comparing the Cost of Administering and Monitoring Vancomycin and Daptomycin Through a Home Infusion Pharmacy, accessed July 24, 2025, https://nhia.org/comparing-the-cost-of-administering-and-monitoring-vancomycin-and-daptomycin-through-a-home-infusion-pharmacy/
- 286. Exploratory Cost-Effectiveness Analysis for Treatment of Methicillin-Resistant Staphylococcus aureus Bloodstream Infections: Are Daptomycin and Linezolid Favored over Vancomycin and Other Antibiotics? - Bohrium, accessed July 24, 2025, https://www.bohrium.com/paper-details/286-exploratory-cost-effectiveness-analysis-for-treatment-of-methicillin-resistant-staphylococcus-aureus-bloodstream-infections-are-daptomycin-and-linezolid-favored-over-vancomycin-and-other-antibiotics/812519544630280197-7613