yingweiwo

Linezolid (PNU-100766)

Alias: PNU 100766; PNU-100766; PNU100766; U 100766; U-100766; U100766; Linezolid; Zyvox; Zyvoxid; Zyvoxam
Cat No.:V2009 Purity: ≥98%
Linezolid (also known as PNU-100766), a synthetic oxazolidinone antimicrobial, is an antibiotic used for the treatment of serious infections.
Linezolid (PNU-100766)
Linezolid (PNU-100766) Chemical Structure CAS No.: 165800-03-3
Product category: Bacterial
This product is for research use only, not for human use. We do not sell to patients.
Size Price Stock Qty
25mg
50mg
100mg
250mg
500mg
1g
Other Sizes

Other Forms of Linezolid (PNU-100766):

  • Linezolid D3
Official Supplier of:
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Alternate Text
Top Publications Citing lnvivochem Products
Purity & Quality Control Documentation

Purity: ≥98%

Product Description

Linezolid (also known as PNU-100766), a synthetic oxazolidinone antimicrobial, is an antibiotic used for the treatment of serious infections. Linezolid shows a wide spectrum against Gram-positive bacteria andmultidrug-resistant bacteria such as anaerobes, methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant enterococci, penicillin-resistant pneumococci and streptococcus. Linezolid inhibits initiation complex formation with either the 30S or the 70S ribosomal subunits from Escherichia coli. Linezolid inhibits complex formation with Staphylococcus aureus 70S tight-couple ribosomes.

Biological Activity I Assay Protocols (From Reference)
Targets
Oxazolidinone antibiotic; bacterial protein synthesis
ln Vitro
Linezolid inhibits initiation complex formation with either the 30S or the 70S ribosomal subunits from Escherichia coli. Linezolid inhibits complex formation with Staphylococcus aureus 70S tight-couple ribosomes. Linezolid is a potent inhibitor of cell-free transcription-translation in E. coli, exhibiting 50% inhibitory concentrations (IC50s) of 1.8 mM. Linezolid is an oxazolidinone, a new class of antibacterial agents with enhanced activity against pathogens. Linezolid MICs vary slightly with the test method, laboratory, and significance attributed to thin hazes of bacterial survival, but all workers find that the susceptibility distributions are narrow and unimodal, with MIC values between 0.5 and 4 mg/L for streptococci, enterococci and staphylococci. Linezolid entails mutation of the 23S rRNA that forms the binding site. Linezolid is an oxazolidinone whose mechanism of action involves inhibition of protein synthesis at a very early stage. Linezolid is added to 7H10 agar medium (Difco) supplemented with OADC (oleic acid, albumin, dextrose, and catalase) at 50°C to 56°C by doubling the dilutions to yield a final concentration of 0.125 μg/mL to 4 μg/mL. Linezolid shows excellent in vitro activity against all the strains tested (MICs ≤ 1 μg/ml), including those resistant to SIRE.
Initiation of translation requires the formation of a ternary complex between tRNAfMet, the 30S or the 70S subunit, and mRNA. This initiation complex can be assayed by measuring the binding of radiolabeled tRNAfMet to either the 30S or the 70S subunit. Figure 1A shows that in the presence of the initiation factors IF1, IF2, and IF3, Linezolid had 50% inhibitory concentrations (IC50s) of 110 μM (37 μg/ml) and 130 μM (44 μg/ml) for E. coli 30S and 70S initiation complex formation, respectively. The integrity of the assay was confirmed by demonstrating that kasugamycin was inhibitory to 70S initiation complex formation, with an IC50 of 154 μM (Fig. 1B). Oxazolidinone inhibition of initiation complex formation was studied further with S. aureus 70S ribosomes (Fig. 2). With a truncated mRNA, an IC50 value of 116 μM was obtained for linezolid. These reactions were performed in the absence of initiation factors and with salt-washed 70S ribosomes. Initiation factors IF1, IF2, and IF3 play important roles in the initiation of translation in bacteria. The tRNAfMet is bound by IF2 and is delivered to the 30S subunit joining IF1, IF3, and the mRNA as part of the initiation complex. Linezolid did not inhibit formation of the IF2-tRNAfMet complex when either 5 or 0.5 pmol of E. coli IF2 was used (Table 1). The role of initiation factors in the mechanism of action of linezolid was further investigated by forming E. coli 70S ribosome initiation complexes in the absence of any of the three factors. Figure 4 demonstrates that an IC50 of 152 μM was obtained for linezolid under these conditions. [1]
The oxazolidinones are a new class of synthetic antibiotics with good activity against gram-positive pathogenic bacteria. Experiments with a susceptible Escherichia coli strain, UC6782, demonstrated that in vivo protein synthesis was inhibited by both eperezolid (formerly U-100592) and Linezolid (formerly U-100766). Both Linezolid and eperezolid were potent inhibitors of cell-free transcription-translation in E. coli, exhibiting 50% inhibitory concentrations (IC50s) of 1.8 and 2.5 microM, respectively. The ability to demonstrate inhibition of in vitro translation directed by phage MS2 RNA was greatly dependent upon the amount of RNA added to the assay. For eperezolid, 128 microg of RNA per ml produced an IC50 of 50 microM whereas a concentration of 32 microg/ml yielded an IC50 of 20 microM. Investigating lower RNA template concentrations in linezolid inhibition experiments revealed that 32 and 8 microg of MS2 phage RNA per ml produced IC50s of 24 and 15 microM, respectively. This phenomenon was shared by the translation initiation inhibitor kasugamycin but not by streptomycin. Neither oxazolidinone inhibited the formation of N-formylmethionyl-tRNA, elongation, or termination reactions of bacterial translation. The oxazolidinones appear to inhibit bacterial translation at the initiation phase of protein synthesis [2].
ln Vivo
Linezolid is fully bioavailable following oral administration, with maximum plasma linezolid concentrations achieved between 1 and 2 hours after oral administration. The elimination half-life of linezolid is 5–7 hours, and twice-daily administration of 400–600mg provides steady-state concentrations in the therapeutic range.
Finally, we compared the activities of Linezolid and vancomycin in pneumonia induced by IRDL-7971 in HLA-DR3 transgenic mice using a previously used dosing regimen (20). As shown in Fig. 5, Linezolid conferred significant protection over vancomycin (P = 0.0002; n = 10 to 14 mice/group) and untreated mice (P = 0.0004; n = 8 to 10 mice/group). Surprisingly, vancomycin failed to confer significant protection from lethal pneumonia (P = 0.50; n = 8 to 14 mice/group). Serum cytokine analyses revealed that compared to untreated and vancomycin-treated mice infected with IDRL-7971, linezolid-treated HLA-DR3 transgenic mice infected with IDRL-7971, had significantly lower levels of IL-2, IL-6, and the chemokine KC (Fig. 6). These results suggest that linezolid played a protective role by attenuating the production of SAg in vivo [4].
Enzyme Assay
Synthesis of [32P]mRNA for ribosome binding studies. [1]
The one-step PCR procedure described by Sandhu et al. (24) was used to synthesize a 200-bp mRNA with a defined sequence. Four adjacent oligonucleotide primers with short overlaps were annealed to each other and were subjected to PCR. The primer sequences were as follows (primer sequences are 5′ to 3′): primer 1, GGGAATTCGCAGGTTTAAAAATGAAAGGTAAAGGTAAAGGTAAA; primer 2, GGTGGTGGCCTGGGCAAAGGTAAAGGT; primer 3, AAAGGT AAAAAAGGTAAAGGTAAAGGTAAAAAAGGTAAAAAAGGTAAAGG TGGTGGTTAATAAAAAAAATAAAAAG; and primer 4, CTAGAGGATCCTTTTTATTTTTTTATTAACCACCAC. Primers 1 and 2 were annealed to the sequence 5′-ACCTTTTTTACCTTTACCTTTACCTTTTTTACCTTTTTTACCTTTACCTTTACCTTATCCTTTACCTTTGCCCAGGCCAC-3′. Primer 3 was annealed to the sequence 5′-ACCTTTGCCCAGGCCACCACCTTTACCTTTTTTACCTTTTTTACCTTTACCTTCACC-3′, and primer 4 was annealed to primer 3. Extension by PCR resulted in the asymmetric synthesis of intermediates which annealed to each other, thereby priming the synthesis of a double-stranded DNA template. This template was subsequently used to produce an mRNA with the sequence 5′-GGGAAUUCGGAGGUUUAAAAAUG-(GGUAAA)33UAAUAA-3′ (the Shine-Dalgarno sequence and the AUG start codon are underlined). The coding sequence contained Gly (GGU) and Lys (AAA) codons followed by tandem stop codons. [32P]mRNA was transcribed with a Ribomax kit and either [32P]CTP or [32P]GTP. RNA was isolated by phenol extraction and chromatography through a Quick Spin G-25 column.
Binding of labeled synthetic mRNA to ribosomes. [1]
Binding of 32P-labeled synthetic mRNA was carried out for 15 min at 24°C in duplicate 50-μl reaction mixtures containing 200 to 400 μg of 70S ribosomes, 20 mM MgCl2, 10 mM Tris-HCl (pH 7.4), 1 mM DTT, 80 mM NH4Cl, and 1 μl (16,000 dpm) of 32P-labeled mRNA. Duplicate reactions were terminated by the addition of 1 ml of ice-cold buffer containing 20 mM Tris (pH 7.4), 20 mM MgCl2, and 100 mM NH4Cl. The mRNA-ribosome complex was then trapped on Millipore HA filters, and the radioactivity was counted after the addition of scintillation fluid.
E. coli initiation complex assay with AUG. [1]
E. coli 70S ribosomes (10 pmol) were incubated with [35S]tRNAfMet (45,000 dpm) in 20-μl reaction mixtures containing 20 mM HEPES (pH 7.6), 3 mM MgCl2, 150 mM NH4Cl, 4 mM DTT, 0.05 mM spermine, 2 mM spermidine, and 0.25 μg of the AUG trinucleotide. Duplicate reaction mixtures were incubated at 37°C for 10 min, and the reactions were stopped by the addition of 2 ml of cold buffer A. Complexes were filtered through Millipore filters (pore size, 0.45 μm) and washed with 50 ml of buffer A, and the radioactivity was counted after the addition of scintillation fluid.
Cell Assay
Linezolid was a potent inhibitor of cell-free transcription-translation in E. coli. IC50 was 1.8 mM. linezolid MICs vary slightly owing to the different test method and laboratory. The MIC values were between 0.5 and 4 mg/L for streptococci, enterococci and staphylococci.
Preparation of E. coli 70S ribosomes.[1]
Ribosomes were prepared by the method of Rheinberger et al. Fifty grams (wet weight) of frozen MRE600 cells was mixed with an equal weight of alumina, and the cells were lysed at 0°C by grinding with a mortar and pestle. Fifty milliliters of buffer A containing 1 μg of DNase per ml was added and the suspension was stirred for 20 min. The alumina, unbroken cells, and cellular debris were removed by two centrifugations at 10,000 × g for 10 min. The supernatant was centrifuged again for 30 min at 30,000 × g, and the upper two-thirds of the resulting supernatant was centrifuged again at 30,000 × g for 16 h (S30 extract). The ribosome pellet was suspended in buffer B and centrifuged at 10,000 × g for 10 min, and the clear supernatant was centrifuged at 105,000 × g for 4 h. The pelleted ribosomes were washed twice more in buffer C while maintaining the ribosomes at 5 to 10 mg/ml (14.4 A260 units = 1 mg/ml), suspended in buffer A at 80 to 100 mg of ribosomes per ml, and stored at −80°C.
Preparation of E. coli ribosomal subunits.[1]
Ribosomal subunits were prepared as described by Staehelin and Maglott, with the following modifications. The S30 extract was prepared as described above by using MRE600 mid-logarithmic-phase cells, and the 30S subunits were stored in liquid nitrogen.
Preparation of S. aureus ribosomes.[1]
S. aureus cells (50 g [wet weight]) were resuspended in 100 ml of lysis buffer (50 mM Tris-HCl [pH 8.0], 100 mM NaCl, 2 mg of lysostaphin per ml, 10,000 U of DNase I) and incubated for 1 h in a 37°C water bath. β-Mercaptoethanol was added to a final concentration of 5 mM, and the lysed cells were centrifuged at 10,000 × g for 10 min to remove unbroken cells and cell fragments. The supernatant was centrifuged at 30,000 × g, and the resulting supernatant was centrifuged at 100,000 × g for 16 h to pellet the ribosomes. The ribosome pellet was resuspended in buffer B and was again centrifuged at 100,000 × g for 16 h. The pellet was resuspended in buffer A, applied to linear 5 to 40% (wt/vol) sucrose gradients prepared in buffer A, and centrifuged for 16 h in a Beckman SW28 rotor. Gradients were fractionated; and the 70S ribosomes were pooled, pelleted at 300,000 × g for 5 h, and resuspended in buffer A before they were stored at −80°C.
Initiation factor assays.[1]
Initiation factors were assayed as described by Hershey et al. Complexes between IF2 and tRNAfMet were formed in the reaction mixtures (final volume, 65 μl) containing 190 mM Tris-HCl (pH 7.4), 19 mM MgCl2, 3.8 mM DTT, 1.9 mM GTP, 540 mM NH4Cl, 5 pmol of IF2, and 2 μl of [35S]tRNAfMet (10,000 dpm). Duplicate reaction mixtures were incubated for 10 min at 37°C, and the reactions were stopped by the addition of 1 ml ice-cold buffer A containing 1% glutaraldehyde. Complexes were trapped on Millipore HA filters (pore size, 0.45 μm), washed with 50 ml of buffer A containing 1% glutaraldehyde, and counted after the addition of liquid scintillation fluid.
Animal Protocol


ADME/Pharmacokinetics
Absorption, Distribution and Excretion
Linezolid is extensively absorbed following oral administration and has an absolute bioavailability of approximately 100%. Maximum plasma concentrations are reached within approximately 1 to 2 hours after dosing (Tmax) and range from 8.1-12.9 mcg/mL after single doses and 11.0-21.2 mcg/mL after multiple dosing. The absorption of orally administered linezolid is not significantly affected by co-administration with food and it may therefore be given without regard to the timing of meals.
Urinary excretion is the primary means by which linezolid and its metabolic products are excreted. Following the administration of a radiolabeled dose of linezolid under steady-state conditions, approximately 84% of radioactivity was recovered in the urine, of which approximately 30% is unchanged parent drug, 40% is the hydroxyethyl glycine metabolite, and 10% is the aminoethoxyacetic acid metabolite. Fecal elimination is comparatively minor, with no parent drug observed in feces and only 6% and 3% of an administered dose found in the feces as the hydroxyethyl glycine metabolite and the aminoethoxyacetic acid metabolite, respectively.
At steady-state, the volume of distribution of linezolid in healthy adults is approximately 40-50 liters.
Total clearance of linezolid is estimated to be 100-200 mL/min, the majority of which appears to be non-renal. Mean renal clearance is approximately 40 mL/min, which suggests net tubular reabsorption, while non-renal clearance is estimated to account for roughly 65% of total clearance, or 70-150 mL/min on average. Variability in linezolid clearance is high, particularly for non-renal clearance.
Distributed to well-perfused tissues; volume of distribution slightly lower in women than men. VolD (steady state) - 40 to 50 L.
AUC is lower for pediatric patients compared with adults and a wider variability of linezolid AUC cross all pediatric age groups as compared with adults. Most pre-term neonates less than 7 days of age (gestational age less than 34 weeks) have larger AUC values than many full-term neonates and older infants.
Linezolid was rapidly absorbed after p.o. dosing with an p.o. bioavailability of > 95% in rat and dog, and > 70% in mouse. Twenty-eight-day i.v./p.o. toxicokinetic studies in rat (20-200 mg kg(-1) day(-1)) and dog (10-80 mg kg(-1) day(-1)) revealed neither a meaningful increase in clearance nor accumulation upon multiple dosing. Linezolid had limited protein binding (<35%) and was very well distributed to most extravascular sites, with a volume of distribution at steady-state (V(ss)) approximately equal to total body water. Linezolid circulated mainly as parent drug and was excreted mainly as parent drug and two inactive carboxylic acids, PNU-142586 and PNU-142300. Minor secondary metabolites were also characterized. In all species, the clearance rate was determined by metabolism. Radioactivity recovery was essentially complete within 24-48 hr. Renal excretion of parent drug and metabolites was a major elimination route. Parent drug underwent renal tubular reabsorption, significantly slowing parent drug excretion and allowing a slow metabolic process to become rate-limiting in overall clearance. It is concluded that ADME data were relatively consistent across species and supported the rat and dog as the principal non-clinical safety species.
In two randomized, double-blind, placebo-controlled, dose-escalating trials, subjects were exposed either to oral (375, 500 or 625 mg) or intravenous (500 or 625 mg) linezolid or placebo twice daily. Serial blood and urine samples were obtained after the first- and multiple-dose administrations for up to 18 days. Non-compartmental pharmacokinetic analyses were used to describe the disposition of linezolid. Plasma linezolid concentrations and area under the concentration-time curves (AUC) increased proportionally with dose irrespective of the route of administration. Plasma linezolid concentrations remained above the MIC90 for susceptible target pathogens (4.0 mg/L) for the majority of the 12 hr dosing interval. Mean clearance, half-life and volume of distribution were similar irrespective of dose for both the oral and intravenous routes. Linezolid was well tolerated and the frequency of drug-related adverse events was similar between the linezolid and placebo groups. Oral and intravenous linezolid exhibit linear pharmacokinetics, with concentrations remaining above the target MIC90 /minimal inhibitory concentration/ for most of the dosing interval. These results support a twice-daily schedule for linezolid and demonstrate the feasibility of converting from intravenous to oral dosing without a dose adjustment.
For more Absorption, Distribution and Excretion (Complete) data for LINEZOLID (16 total), please visit the HSDB record page.
Metabolism / Metabolites
Linezolid is primarily metabolized to two inactive metabolites: an aminoethoxyacetic acid metabolite (PNU-142300) and a hydroxyethyl glycine metabolite (PNU-142586), both of which are the result of morpholine ring oxidation. The hydroxyethyl glycine metabolite - the most abundant of the two metabolites - is likely generated via non-enzymatic processes, though further detail has not been elucidated. While the specific enzymes responsible for the biotransformation of linezolid are unclear, it does not appear to be subject to metabolism via the CYP450 enzyme system, nor does it meaningfully inhibit or induce these enzymes. Linezolid is, however, a reversible and non-selective inhibitor of monoamine oxidase enzymes.
In vitro studies have not shown that linezolid is metabolized by human cytochrome p450 enzymes. Linezolid does not inhibit the cytochrome p450 enzymes.
Linezolid is primarily metabolized via oxidation of the morpholine ring. Two inactive metabolites are formed: the aminoethoxyacetic acid metabolite and the hydroxyethyl glycine metabolite. The hydroxyethyl glycine metabolite is formed via a non-enzymatic chemical oxidation mechanism in vitro.
The drug is metabolized principally via oxidation to 2 inactive metabolites; an aminoethoxyacetic acid metabolite and a hydroxyethyl glycine metabolite. Linezolid is not metabolized to any measurable extent by the cytochrome p450 (CYP) enzyme system. Linezolid does not inhibit CYP isoenzymes 1A2, 2C9, 2C19, 2D6, 2E1, or 3A4 and is not an enzyme inducer, suggesting that the drug is unlikely to alter the pharmacokinetics of drugs metabolized by these enzymes.
In vitro studies were conducted to identify the hepatic enzyme(s) responsible for the oxidative metabolism of linezolid. In human liver microsomes, linezolid was oxidized to a single metabolite, hydroxylinezolid (M1). Formation of M1 was determined to be dependent upon microsomal protein and NADPH. Over a concentration range of 2 to 700 uM, the rate of M1 formation conformed to first-order (nonsaturable) kinetics. Application of conventional in vitro techniques were unable to identify the molecular origin of M1 based on the following experiments: a) inhibitor/substrates for various cytochrome P-450 (CYP) enzymes were unable to inhibit M1 formation; b) formation of M1 did not correlate (r(2) < 0.23) with any of the measured catalytic activities across a population of human livers (n = 14); c) M1 formation was not detectable in incubations using microsomes prepared from a baculovirus insect cell line expressing CYPs 1A1, 1A2, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, 3A4, 3A5, and 4A11. In addition, results obtained from an in vitro P-450 inhibition screen revealed that linezolid was devoid of any inhibitory activity toward the following CYP enzymes (CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4). Additional in vitro studies excluded the possibility of flavin-containing monooxygenase and monoamine oxidase as potential enzymes responsible for metabolite formation. However, metabolite formation was found to be optimal under basic (pH 9.0) conditions, which suggests the potential involvement of either an uncharacterized P-450 enzyme or an alternative microsomal mediated oxidative pathway.
Linezolid is primarily metabolized by oxidation of the morpholine ring, which results in two inactive ring-opened carboxylic acid metabolites: the aminoethoxyacetic acid metabolite (A), and the hydroxyethyl glycine metabolite (B). Formation of metabolite B is mediated by a non-enzymatic chemical oxidation mechanism in vitro. Linezolid is not an inducer of cytochrome P450 (CYP) in rats, and it has been demonstrated from in vitro studies that linezolid is not detectably metabolized by human cytochrome P450 and it does not inhibit the activities of clinically significant human CYP isoforms (1A2, 2C9, 2C19, 2D6, 2E1, 3A4).
Linezolid is rapidly and extensively absorbed after oral dosing. Maximum plasma concentrations are reached approximately 1 to 2 hours after dosing, and the absolute bioavailability is approximately 100%. Linezolid is primarily metabolized by oxidation of the morpholine ring, which results in two inactive ring-opened carboxylic acid metabolites: the aminoethoxyacetic acid metabolite (A), and the hydroxyethyl glycine metabolite (A308). Half Life: 4.5-5.5 hours.
Biological Half-Life
The elimination half-life is estimated to be between 5 and 7 hours.
... A significant although weak correlation between age and total body clearance was observed. The mean (+ or - SD) values for elimination half-life, total clearance and apparent volume of distribution were 3.0 + or - 1.1 hr, 0.34 + or - 0.15 liter/h/kg and 0.73 + or - 0.18 liter/kg, respectively. ...
The following are elimination half live values of linezolid doses in adults: 400 mg tablet (single dose) - 5.2 hours; 400 mg tablet every 12 hours - 4.69 hours; 600 mg tablet (single dose) - 4.26 hours; 600 mg tablet every 12 hours - 5.4 hours; 600 mg oral suspension (single dose) - 4.6 hours; 600 mg intravenous injection (single dose) - 4.4 hours; 600 mg intravenous injection every 12 hours - 4.8 hours;. Pediatrics ranging in age from greater than 7 days of age to 11 years of age have a shorter half-life compared with adults.
Toxicity/Toxicokinetics
Toxicity Summary
Linezolid targets the large 39S subunit of the mitochondrial ribosome thereby deactivation mitochondrial protein synthesis. As a result Linezolid is cytotoxic to the most metabolically active cells or tissues including the heart, liver, thymus and bone-marrow. (A7823). The likely target of Linezolid is the 16S rRNA molecule in the mitochondrial ribosome, which is analogous to the 23S rRNA in bacterial ribosomes.
Hepatotoxicity
Therapy with linezolid has been associated with mild and transient elevations in serum aminotransferase and alkaline phosphatase levels in 1% to 10% of patients, although similar rates of elevations occur in patients with infections treated with comparable agents, and enzyme elevations were not found in normal volunteers given linezolid for short periods. On the other hand, ALT elevations during therapy have been higher with higher doses of linezolid, but in all instances the elevations occurred without symptoms and resolved with discontinuation of the drug.
Although the agent has been available for a limited time and its use has been restricted, several instances of clinically apparent liver disease with jaundice have been reported with linezolid therapy. A case of a hypersensitivity response with rash, eosinophilia and renal insufficiency (DRESS syndrome) with mild serum enzyme elevations has been reported. More frequently, linezolid has been linked to cases of lactic acidosis, generally arising after 1 to 8 weeks of therapy and sometimes associated with evidence of liver injury and jaundice. Lactic acidosis is usually due to injury and dysfunction of hepatic mitochondria, with resulting microvesicular steatosis and disturbed hepatic function (not necessarily accompanied by jaundice or even ALT or alkaline phosphatase elevations). Other serious side effects associated with mitochondrial damage due to linezolid therapy include peripheral and optic neuropathy, pancreatitis, serotonin syndrome and renal injury. Risk factors for developing lactic acidosis from linezolid include higher doses, longer courses of therapy and underlying chronic liver or renal disease. The mitochondrial injury is believed to be due to the inhibition of mitochondrial ribosomal function that matches the known effect of linezolid on bacterial ribosomal function. Lactic acidosis occurs after 1 to 8 weeks of treatment and can be severe, although it often resolves rapidly with discontinuation. In contrast, the optic and peripheral neuropathy due to linezolid resolves more slowly and can be permanent. Lactic acidosis can be fatal and hepatic dysfunction and jaundice have been mentioned in severe cases of lactic acidosis attributed to linezolid.
Likelihood score: A (well established cause of clinically apparent liver injury usually in association with lactic acidosis).
Effects During Pregnancy and Lactation
◉ Summary of Use during Lactation
Linezolid is excreted into breastmilk in concentrations likely to be effective against staphylococcal strains found in mastitis. Limited data indicate that the maximum dose an infant would receive through breastmilk would be only 6 to 9% of the standard infant dose and that resulting infant serum levels are trivial. If the mother requires linezolid, it is not a reason to discontinue breastfeeding. Monitor the infant for possible effects on the gastrointestinal tract, such as diarrhea and vomiting.
◉ Effects in Breastfed Infants
Relevant published information was not found as of the revision date.
◉ Effects on Lactation and Breastmilk
Relevant published information was not found as of the revision date.
Protein Binding
Plasma protein binding of linezolid is approximately 31% - primarily to serum albumin - and is concentration-dependent.
Interactions
Following coadministration with linezolid, minimal but statistically significant increases were observed in pseudoephedrine and phenylpropanolamine plasma concentrations; a minimal but statistically significant decrease was observed in dextrorphan (the primary metabolite of dextromethorphan) plasma concentrations. Increased blood pressure (BP) was observed following the coadministration of linezolid with either pseudoephedrine or phenylpropanolamine; no significant effects were observed with dextromethorphan. None of these coadministered drugs had a significant effect on linezolid pharmacokinetics. Minimal numbers of adverse events were reported. Potentiation of sympathomimetic activity by linezolid was judged not to be clinically significant, but patients sensitive to the effects of increased BP due to predisposing factors should be treated cautiously. No restrictions are indicated for the coadministration of dextromethorphan and linezolid.
To report 2 cases of serotonin toxicity (ST) associated with concomitant use of linezolid and serotonergic drugs and review previously published case reports. Case 1. A 38-year-old white female with cystic fibrosis treated with venlafaxine 300 mg/day for one year was prescribed linezolid 600 mg intravenously every 12 hours for treatment of methicillin-resistant Staphylococcus aureus (MRSA) pulmonary infection. She displayed symptoms of ST 8 days after the introduction of linezolid. The venlafaxine dosage was decreased to 150 mg/day, and symptoms gradually abated over 36 hours. Case 2. A 37-year-old male with multiple myeloma received citalopram 40 mg/day and trazodone 150 mg/day for anxiety-related disorders. Linezolid treatment with 600 mg orally twice daily was instituted for MRSA cellulitis. The following day, the patient developed anxiety, panic attacks, tremors, tachycardia, and hypertension that persisted throughout linezolid treatment. Symptoms finally waned 5 days after linezolid treatment was stopped.
Potential pharmacologic interaction (serotonin syndrome). Although serotonin syndrome was not reported during clinical trials with linezolid, there have been a limited number of postmarketing case reports of the syndrome in patients who received linezolid concurrently with or shortly after discontinuation of certain selective serotonin-reuptake inhibitors (SSRIs) (e.g., citalopram, paroxetine, sertraline). Clinicians should consider the possibility if signs and symptoms of serotonin syndrome (e.g., hyperpyrexia, cognitive dysfunction) occur in patients receiving such concomitant therapy. Some clinicians suggest that linezolid be used with caution in patients receiving SSRIs, and some suggest that SSRI therapy should be discontinued before linezolid is initiated and not reinitiated until 2 weeks after linezolid therapy is completed.
Toxicity resulting from excessive intra-synaptic serotonin, historically referred to as serotonin syndrome, is now understood to be an intra-synaptic serotonin concentration-related phenomenon. Recent research more clearly delineates serotonin toxicity as a discreet toxidrome characterized by clonus, hyper-reflexia, hyperthermia and agitation. Serotonergic side-effects occur with serotonergic drugs, and overdoses of serotonin re-uptake inhibitors (SRIs) frequently produce marked serotonergic side-effects, and in 15% of cases, moderate serotonergic toxicity, but not to a severe degree, which produces hyperthermia and risk of death. It is only combinations of serotonergic drugs acting by different mechanisms that are capable of raising intra-synaptic serotonin to a level that is life threatening. The combination that most commonly does this is a monoamine oxidase inhibitor (MAOI) drug combined with any SRI. There are a number of lesser-known drugs that are MAOIs, such as linezolid and moclobemide; and some opioid analgesics have serotonergic activity. These properties when combined can precipitate life threatening serotonin toxicity. Possibly preventable deaths are still occurring. Knowledge of the properties of these drugs will therefore help to ensure that problems can be avoided in most clinical situations, and treated appropriately (with 5-HT(2A) antagonists for severe cases) if they occur.
For more Interactions (Complete) data for LINEZOLID (12 total), please visit the HSDB record page.
References

[1]. Antimicrob Agents Chemother.1998 Dec;42(12):3251-5.

[2]. Antimicrob Agents Chemother.1997 Oct;41(10):2132-6.

[3]. Drugs. 2000 Apr;59(4):815-27; discussion 828.

[4]. Antimicrob Agents Chemother . 2012 Oct;56(10):5401-5.

Additional Infomation
Therapeutic Uses
Antibacterial
Intravenous and oral linezolid is indicated in the treatment of nosocomial pneumonia caused by methicillin-susceptible and methicillin resistant Staphylococcus aureus or penicillin-susceptible strains of Streptococcus pneumonia. /Included in US product labeling/
Intravenous and oral linezolid is indicated in the treatment of vancomycin-resistant Enterococcus faecium infections. /Included in US product labeling/
Oral linezolid is indicated in the treatment of uncomplicated skin and soft tissue infections caused by methicillin-susceptible strains of Staphylococcus aureus or Streptococcus pyogenes. /Included in US product labeling/
For more Therapeutic Uses (Complete) data for LINEZOLID (16 total), please visit the HSDB record page.
Drug Warnings
Myelosuppression (including anemia, leukopenia, pancytopenia, and thrombocytopenia) has been reported in patients receiving linezolid. In cases where the outcome is known, when linezolid was discontinued, the affected hematologic parameters have risen toward pretreatment levels. Complete blood counts should be monitored weekly in patients who receive linezolid, particularly in those who receive linezolid for longer than two weeks, those with pre-existing myelosuppression, those receiving concomitant drugs that produce bone marrow suppression, or those with a chronic infection who have received previous or concomitant antibiotic therapy. Discontinuation of therapy with linezolid should be considered in patients who develop or have worsening myelosuppression.
Lactic acidosis has been reported with the use of Zyvox. In reported cases, patients experienced repeated episodes of nausea and vomiting. Patients who develop recurrent nausea or vomiting, unexplained acidosis, or a low bicarbonate level while receiving zyvox should receive immediate medical evaluation.
Spontaneous reports of serotonin syndrome associated with the co-administration of Zyvox and serotonergic agents, including antidepressants such as selective serotonin reuptake inhibitors (SSRIs), have been reported. Where administration of Zyvox and concomitant serotonergic agents is clinically appropriate, patients should be closely observed for signs and symptoms of serotonin syndrome such as cognitive dysfunction, hyperpyrexia, hyperreflexia and incoordination. If signs or symptoms occur physicians should consider discontinuation of either one or both agents.
Peripheral and optic neuropathy have been reported in patients treated with Zyvox, primarily those patients treated for longer than the maximum recommended duration of 28 days. In cases of optic neuropathy that progressed to loss of vision, patients were treated for extended periods beyond the maximum recommended duration. Visual blurring has been reported in some patients treated with Zyvox for less than 28 days. If patients experience symptoms of visual impairment, such as changes in visual acuity, changes in color vision, blurred vision, or visual field defect, prompt ophthalmic evaluation is recommended. Visual function should be monitored in all patients taking Zyvox for extended periods (> or = 3 months) and in all patients reporting new visual symptoms regardless of length of therapy with Zyvox. If peripheral or optic neuropathy occurs, the continued use of Zyvox in these patients should be weighed against the potential risks.
For more Drug Warnings (Complete) data for LINEZOLID (18 total), please visit the HSDB record page.
Pharmacodynamics
Linezolid is an oxazolidinone antibacterial agent effective against most strains of aerobic Gram-positive bacteria and mycobacteria. It appears to be bacteriostatic against both staphylococci and enterococci and bactericidal against most isolates of streptococci. Linezolid has shown some _in vitro_ activity against Gram-negative and anaerobic bacteria but is not considered efficacious against these organisms. Linezolid is a reversible and non-selective inhibitor of monoamine oxidase (MAO) enzymes and can therefore contribute to the development of serotonin syndrome when administered alongside serotonergic agents such as selective serotonin re-uptake inhibitors (SSRIs) or tricyclic antidepressants (TCAs). Linezolid should not be used for the treatment of catheter-related bloodstream infections or catheter-site infections, as the risk of therapy appears to outweigh its benefits under these circumstances.
Linezolid is an oxazolidinone antibacterial agent that acts by inhibiting the initiation of bacterial protein synthesis. Cross-resistance between linezolid and other inhibitors of protein synthesis has not been demonstrated. Linezolid has a wide spectrum of activity against gram-positive organisms including methicillin-resistant staphylococci, penicillin-resistant pneumococci and vancomycin-resistant Enterococcus faecalis and E. faecium. Anerobes such as Clostridium spp., Peptostreptococcus spp. and Prevotella spp. are also susceptible to linezolid. Linezolid is bacteriostatic against most susceptible organisms but displays bactericidal activity against some strains of pneumococci, Bacteroides fragilis and C. perfringens. In clinical trials involving hospitalised patients with skin/soft tissue infections (predominantly S. aureus), intravenous/oral linezolid (up to 1250 mg mg/day) produced clinical success in >83% of individuals. In patients with community-acquired pneumonia, success rates were >94%. Preliminary clinical data also indicate that twice daily intravenous/oral linezolid 600 mg is as effective as intravenous vancomycin 1 g in the treatment of patients with hospital-acquired pneumonia and in those with infections caused by methicillin-resistant staphylococci. Moreover, linezolid 600 mg twice daily produced >85% clinical/microbiological cure in vancomycin-resistant enterococcal infections. Linezolid is generally well tolerated and gastrointestinal disturbances are the most commonly occurring adverse events. No clinical evidence of adverse reactions as a result of monoamine oxidase inhibition has been reported.[3]
Superantigens (SAg), the potent activators of the immune system, are important determinants of Staphylococcus aureus virulence and pathogenicity. Superior response to SAg in human leukocyte antigen (HLA)-DR3 transgenic mice rendered them more susceptible than C57BL/6 mice to pneumonia caused by SAg-producing strains of S. aureus. Linezolid, a bacterial protein synthesis inhibitor, was superior to vancomycin in inhibiting SAg production by S. aureus in vitro and conferred greater protection from pneumonia caused by SAg-producing staphylococci.[4]
These protocols are for reference only. InvivoChem does not independently validate these methods.
Physicochemical Properties
Molecular Formula
C16H20FN3O4
Molecular Weight
337.35
Exact Mass
337.143
Elemental Analysis
C, 56.97; H, 5.98; F, 5.63; N, 12.46; O, 18.97
CAS #
165800-03-3
Related CAS #
Linezolid-d3;1127120-38-0
PubChem CID
441401
Appearance
White to off-white solid powder
Density
1.3±0.1 g/cm3
Boiling Point
585.5±50.0 °C at 760 mmHg
Melting Point
176-1780C
Flash Point
307.9±30.1 °C
Vapour Pressure
0.0±1.6 mmHg at 25°C
Index of Refraction
1.554
LogP
0.3
Hydrogen Bond Donor Count
1
Hydrogen Bond Acceptor Count
6
Rotatable Bond Count
4
Heavy Atom Count
24
Complexity
472
Defined Atom Stereocenter Count
1
SMILES
FC1C([H])=C(C([H])=C([H])C=1N1C([H])([H])C([H])([H])OC([H])([H])C1([H])[H])N1C(=O)O[C@@]([H])(C([H])([H])N([H])C(C([H])([H])[H])=O)C1([H])[H]
InChi Key
TYZROVQLWOKYKF-ZDUSSCGKSA-N
InChi Code
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
Chemical Name
N-[[(5S)-3-(3-fluoro-4-morpholin-4-ylphenyl)-2-oxo-1,3-oxazolidin-5-yl]methyl]acetamide
Synonyms
PNU 100766; PNU-100766; PNU100766; U 100766; U-100766; U100766; Linezolid; Zyvox; Zyvoxid; Zyvoxam
HS Tariff Code
2934.99.9001
Storage

Powder      -20°C    3 years

                     4°C     2 years

In solvent   -80°C    6 months

                  -20°C    1 month

Note: This product requires protection from light (avoid light exposure) during transportation and storage.
Shipping Condition
Room temperature (This product is stable at ambient temperature for a few days during ordinary shipping and time spent in Customs)
Solubility Data
Solubility (In Vitro)
DMSO : 68~100 mg/mL ( 201.57~296.43 mM )
Ethanol : ~10 mg/mL
Solubility (In Vivo)
Solubility in Formulation 1: ≥ 2.5 mg/mL (7.41 mM) (saturation unknown) in 10% DMSO + 40% PEG300 + 5% Tween80 + 45% Saline (add these co-solvents sequentially from left to right, and one by one), clear solution.
For example, if 1 mL of working solution is to be prepared, you can add 100 μL of 25.0 mg/mL clear DMSO stock solution to 400 μL PEG300 and mix evenly; then add 50 μL Tween-80 to the above solution and mix evenly; then add 450 μL normal saline to adjust the volume to 1 mL.
Preparation of saline: Dissolve 0.9 g of sodium chloride in 100 mL ddH₂ O to obtain a clear solution.

Solubility in Formulation 2: ≥ 2.5 mg/mL (7.41 mM) (saturation unknown) in 10% DMSO + 90% (20% SBE-β-CD in Saline) (add these co-solvents sequentially from left to right, and one by one), clear solution.
For example, if 1 mL of working solution is to be prepared, you can add 100 μL of 25.0 mg/mL clear DMSO stock solution to 900 μL of 20% SBE-β-CD physiological saline solution and mix evenly.
Preparation of 20% SBE-β-CD in Saline (4°C,1 week): Dissolve 2 g SBE-β-CD in 10 mL saline to obtain a clear solution.

View More

Solubility in Formulation 3: ≥ 2.5 mg/mL (7.41 mM) (saturation unknown) in 10% DMSO + 90% Corn Oil (add these co-solvents sequentially from left to right, and one by one), clear solution.
For example, if 1 mL of working solution is to be prepared, you can add 100 μL of 25.0 mg/mL clear DMSO stock solution to 900 μL of corn oil and mix evenly.


Solubility in Formulation 4: 30% PEG400 + 0.5% Tween 80+ 5% propylene glycol: 30mg/ml (88.93mM)

 (Please use freshly prepared in vivo formulations for optimal results.)
Preparing Stock Solutions 1 mg 5 mg 10 mg
1 mM 2.9643 mL 14.8214 mL 29.6428 mL
5 mM 0.5929 mL 2.9643 mL 5.9286 mL
10 mM 0.2964 mL 1.4821 mL 2.9643 mL

*Note: Please select an appropriate solvent for the preparation of stock solution based on your experiment needs. For most products, DMSO can be used for preparing stock solutions (e.g. 5 mM, 10 mM, or 20 mM concentration); some products with high aqueous solubility may be dissolved in water directly. Solubility information is available at the above Solubility Data section. Once the stock solution is prepared, aliquot it to routine usage volumes and store at -20°C or -80°C. Avoid repeated freeze and thaw cycles.

Calculator

Molarity Calculator allows you to calculate the mass, volume, and/or concentration required for a solution, as detailed below:

  • Calculate the Mass of a compound required to prepare a solution of known volume and concentration
  • Calculate the Volume of solution required to dissolve a compound of known mass to a desired concentration
  • Calculate the Concentration of a solution resulting from a known mass of compound in a specific volume
An example of molarity calculation using the molarity calculator is shown below:
What is the mass of compound required to make a 10 mM stock solution in 5 ml of DMSO given that the molecular weight of the compound is 350.26 g/mol?
  • Enter 350.26 in the Molecular Weight (MW) box
  • Enter 10 in the Concentration box and choose the correct unit (mM)
  • Enter 5 in the Volume box and choose the correct unit (mL)
  • Click the “Calculate” button
  • The answer of 17.513 mg appears in the Mass box. In a similar way, you may calculate the volume and concentration.

Dilution Calculator allows you to calculate how to dilute a stock solution of known concentrations. For example, you may Enter C1, C2 & V2 to calculate V1, as detailed below:

What volume of a given 10 mM stock solution is required to make 25 ml of a 25 μM solution?
Using the equation C1V1 = C2V2, where C1=10 mM, C2=25 μM, V2=25 ml and V1 is the unknown:
  • Enter 10 into the Concentration (Start) box and choose the correct unit (mM)
  • Enter 25 into the Concentration (End) box and select the correct unit (mM)
  • Enter 25 into the Volume (End) box and choose the correct unit (mL)
  • Click the “Calculate” button
  • The answer of 62.5 μL (0.1 ml) appears in the Volume (Start) box
g/mol

Molecular Weight Calculator allows you to calculate the molar mass and elemental composition of a compound, as detailed below:

Note: Chemical formula is case sensitive: C12H18N3O4  c12h18n3o4
Instructions to calculate molar mass (molecular weight) of a chemical compound:
  • To calculate molar mass of a chemical compound, please enter the chemical/molecular formula and click the “Calculate’ button.
Definitions of molecular mass, molecular weight, molar mass and molar weight:
  • Molecular mass (or molecular weight) is the mass of one molecule of a substance and is expressed in the unified atomic mass units (u). (1 u is equal to 1/12 the mass of one atom of carbon-12)
  • Molar mass (molar weight) is the mass of one mole of a substance and is expressed in g/mol.
/

Reconstitution Calculator allows you to calculate the volume of solvent required to reconstitute your vial.

  • Enter the mass of the reagent and the desired reconstitution concentration as well as the correct units
  • Click the “Calculate” button
  • The answer appears in the Volume (to add to vial) box
In vivo Formulation Calculator (Clear solution)
Step 1: Enter information below (Recommended: An additional animal to make allowance for loss during the experiment)
Step 2: Enter in vivo formulation (This is only a calculator, not the exact formulation for a specific product. Please contact us first if there is no in vivo formulation in the solubility section.)
+
+
+

Calculation results

Working concentration mg/mL;

Method for preparing DMSO stock solution mg drug pre-dissolved in μL DMSO (stock solution concentration mg/mL). Please contact us first if the concentration exceeds the DMSO solubility of the batch of drug.

Method for preparing in vivo formulation:Take μL DMSO stock solution, next add μL PEG300, mix and clarify, next addμL Tween 80, mix and clarify, next add μL ddH2O,mix and clarify.

(1) Please be sure that the solution is clear before the addition of next solvent. Dissolution methods like vortex, ultrasound or warming and heat may be used to aid dissolving.
             (2) Be sure to add the solvent(s) in order.

Clinical Trial Information
Clinical Trial Testing Whether Targeted Antibiotic Prophylaxis Can Reduce Infections After Cystectomy Compared to Empiric Prophylaxis
CTID: NCT06709196
Phase: Phase 4    Status: Not yet recruiting
Date: 2024-11-29
Trial of Novel Regimens for the Treatment of Pulmonary Tuberculosis
CTID: NCT06192160
Phase: Phase 2    Status: Not yet recruiting
Date: 2024-11-13
Innovating Shorter, All- Oral, Precised Treatment Regimen for Rifampicin Resistant Tuberculosis:BDLL Chinese Cohort
CTID: NCT06649721
Phase: Phase 3    Status: Not yet recruiting
Date: 2024-11-12
Refining MDR-TB Treatment (T) Regimens (R) for Ultra(U) Short(S) Therapy(T)-PLUS
CTID: NCT04717908
Phase: N/A    Status: Completed
Date: 2024-11-12
Linezolid or Vancomycin Surgical Site Infection Prophylaxis
CTID: NCT05571722
Phase: Phase 4    Status: Recruiting
Date: 2024-11-06
View More

MRX-I Versus Linezolid for the Treatment of Acute Bacterial Skin and Skin Structure Infection
CTID: NCT02269319
Phase: Phase 2    Status: Completed
Date: 2024-11-04


PHASE II SINGLE-CENTER, RANDOMIZED, OPEN-LABEL, PROSPECTIVE, STUDY TO DETERMINE THE IMPACT OF SERIAL PROCALCITONIN
CTID: NCT04983901
Phase: Phase 2    Status: Completed
Date: 2024-11-04
Safety and Efficacy Study of Contezolid Acefosamil and Contezolid Compared to Linezolid Administered Intravenously and Orally to Adults With Moderate or Severe Diabetic Foot Infections (DFI)
CTID: NCT05369052
Phase: Phase 3    Status: Recruiting
Date: 2024-11-01
Platform Assessing Regimens and Durations In a Global Multisite Consortium for TB
CTID: NCT06114628
Phase: Phase 2    Status: Recruiting
Date: 2024-10-16
Linezolid Dosing Strategies in Drug-Resistant TB
CTID: NCT05007821
Phase: Phase 2    Status: Active, not recruiting
Date: 2024-10-08
Program for Rifampicin-Resistant Disease With Stratified Medicine for Tuberculosis
CTID: NCT06441006
Phase: Phase 3    Status: Not yet recruiting
Date: 2024-10-04
Phase 2 Trial Assessing TBAJ876 or Bedaquiline, with Pretomanid and Linezolid in Adults with Drug-sensitive Pulmonary Tuberculosis
CTID: NCT06058299
Phase: Phase 2    Status: Active, not recruiting
Date: 2024-09-19
Building Evidence for Advancing New Treatment for Rifampicin Resistant Tuberculosis (RR-TB) Comparing a Short Course of Treatment (Containing Bedaquiline, Delamanid and Linezolid) With the Current South African Standard of Care
CTID: NCT04062201
Phase: Phase 3    Status: Completed
Date: 2024-08-28
Trial of a Six-Month Regimen of High-Dose Rifampicin, High-Dose Isoniazid, Linezolid, and Pyrazinamide Versus a Standard Nine-Month Regimen for the Treatment of Adults and Adolescents With Tuberculous Meningitis
CTID: NCT05383742
Phase: Phase 2    Status: Recruiting
Date: 2024-08-21
A Phase 3 Study Assessing Safety and Efficacy of Bedaquiline Plus PA-824 Plus Linezolid in Subjects With Drug Resistant Pulmonary Tuberculosis
CTID: NCT02333799
Phase: Phase 3    Status: Completed
Date: 2024-08-09
Evaluating Newly Approved Drugs for Multidrug-resistant TB
CTID: NCT02754765
Phase: Phase 3    Status: Completed
Date: 2024-07-24
Evaluating Newly Approved Drugs in Combination Regimens for Multidrug-Resistant TB With Fluoroquinolone Resistance (endTB-Q)
CTID: NCT03896685
Phase: Phase 3    Status: Active, not recruiting
Date: 2024-07-24
Repurposing Clinically Approved Drugs for Yaws With an Insight Into the Cutaneous Ulcer Disease Syndrome (Trep-AByaws)
CTID: NCT05764876
Phase: Phase 3    Status: Active, not recruiting
Date: 2024-07-18
Model-informed Precision Dosing for Linezolid
CTID: NCT06444802
Phase: Phase 3    Status: Not yet recruiting
Date: 2024-06-06
Patient-reported Experiences and Quality of Life Outcomes in the TB-PRACTECAL Clinical Trial
CTID: NCT03942354
Phase:    Status: Completed
Date: 2024-05-29
Alternative Antibiotics for Syphilis
CTID: NCT05069974
Phase: Phase 3    Status: Recruiting
Date: 2024-05-10
Pragmatic Clinical Trial for a More Effective Concise and Less Toxic MDR-TB Treatment Regimen(s)
CTID: NCT02589782
Phase: Phase 2/Phase 3    Status: Completed
Date: 2024-05-01
Economic Evaluation of New MDR TB Regimens
CTID: NCT04207112
Phase: Phase 2/Phase 3    Status: Completed
Date: 2024-04-22
Efficacy and Tolerability of Bedaquiline, Delamanid, Levofloxacin, Linezolid, and Clofazimine to Treat MDR-TB
CTID: NCT03828201
Phase: Phase 2    Status: Recruiting
Date: 2024-04-12
Innovating Shorter, All- Oral, Precised, Individualized Treatment Regimen for Rifampicin Resistant Tuberculosis:Contezolid, Delamanid and Bedaquiline Cohort
CTID: NCT06081361
Phase: Phase 3    Status: Recruiting
Date: 2024-03-15
Modified BPaL Regimen for Managing Pre-XDR TB and MDR (TI/NR) TB in India
CTID: NCT05040126
Phase: Phase 3    Status: Active, not recruiting
Date: 2024-01-03
Adjunctive Clindamycin Versus Linezolid for β-lactam Treated Patients With Invasive Group A Streptococcal Infections
CTID: NCT06126263
Phase:    Status: Active, not recruiting
Date: 2023-11-13
Oral Antibiotic Treatment at Home Instead of Intravenous Treatment in Hospital for Resistant Gram Positive Infections
CTID: NCT00501150
Phase: N/A    Status: Completed
Date: 2023-10-26
Pharmacokinetic Study of Linezolid for TB Meningitis
CTID: NCT03537495
Phase: Phase 2    Status: Completed
Date: 2023-09-06
Two-month Regimens Using Novel Combinations to Augment Treatment Effectiveness for Drug-sensitive Tuberculosis
CTID: NCT03474198
Phase: Phase 2/Phase 3    Status: Completed
Date: 2023-08-14
Finding the Optimal Regimen for Mycobacterium Abscessus Treatment
CTID: NCT04310930
Phase: Phase 2/Phase 3    Status: Recruiting
Date: 2023-08-08
The Incidence of Linezolid-induced Thrombocytopenia (LIT) in Critically Ill Patients, the Risk Factors for LIT
CTID: NCT05944445
Phase:    Status: Recruiting
Date: 2023-07-17
Various Doses and Durations of Linezolid Plus Bedaquiline & Pretomanid in Participants With Drug Resistant Tuberculosis
CTID: NCT03086486
Phase: Phase 3    Status: Completed
Date: 2023-06-29
Imipenem/Cilastatin/Relebactam (MK-7655A) Versus Piperacillin/Tazobactam in Participants With Hospital-Acquired or Ventilator-Associated Bacterial Pneumonia (MK-7655A-016)
CTID: NCT03583333
Phase: Phase 3    Status: Completed
Date: 2023-06-28
Safety and Efficacy of Strategy to Prevent Drug-Induced Nephrotoxicity in High-Risk Patients
CTID: NCT01734694
Phase: Phase 4    Status: Terminated
Date: 2023-05-03
Efficacy and Safety of Continuous Versus Intermittent Linezolid Infusion in Critically Ill Patients With Septic Shock
CTID: NCT05813951
Phase: N/A    Status: Recruiting
Date: 2023-04-14
A Study of an Oral Short-course Regimen Including Bedaquiline for the Treatment of Participants With Multidrug-resistant Tuberculosis in China
CTID: NCT05306223
Phase: Phase 4    Status: Recruiting
Date: 2023-01-18
Contezolid Acefosamil Versus Linezolid for the Treatment of Acute Bacterial Skin and Skin Structure Infection
CTID: NCT03747497
Phase: Phase 2    Status: Completed
Date: 2022-11-08
Antimicrobial Treatment in Patients With Ventilator-associated Tracheobronchitis
CTID: NCT03012360
Phase: Phase 4    Status: Unknown status
Date: 2022-08-17
Pharmacokinetic Study of Antiretroviral Drugs and Related Drugs During and After Pregnancy
CTID: NCT00042289
Phase:    Status: Completed
Date: 2022-07-22
Oral Antimicrobial Treatment vs. Outpatient Parenteral for Infective Endocarditis
CTID: NCT05398679
Phase: Phase 4    Status: Not yet recruiting
Date: 2022-06-01
Delafloxacin IV and OS Administration Compared to Best Available Therapy in Patients With Surgical Site Infections
CTID: NCT04042077
Phase: Phase 3    Status: Terminated
Date: 2022-02-02
Pharmacokinetic and Pharmacodynamic Evaluation of Linezolid Administered Intravenously in MRSA-positive, Morbidly Obese Patients With Pneumonia
CTID: NCT01805284
Phase: Phase 4    Status: Completed
Date: 2021-12-01
An Open-label RCT to Evaluate a New Treatment Regimen for Patients With Multi-drug Resistant Tuberculosis
CTID: NCT02454205
Phase: Phase 2/Phase 3    Status: Completed
Date: 2021-09-29
Linezolid, Aspirin and Enhanced Dose Rifampicin in HIV-TBM
CTID: NCT03927313
Phase: Phase 2    Status: Completed
Date: 2021-09-28
Optimized Antibiotic Therapy in Patients With Subarachnoid Haemorrhage (ES) and Cerebral Haemorrhage (EC)
CTID: NCT04132115
Phase:    Status: Unknown status
Date: 2021-07-21
PRACTECAL-PKPD Sub Study
CTID: NCT04081077
Phase: Phase 2/Phase 3    Status: Unknown status
Date: 2021-05-14
Study the Safety and Efficacy of PTK 0796 in Patients With Complicated Skin and Skin Structure Infection (CSSSI)
CTID: NCT00865280
Phase: Phase 3    Status: Terminated
Date: 2021-03-12
AbioKin - Antibiotic Kinetics
CTID: NCT02609646
Phase:    Status: Completed
Date: 2021-01-19
Clinical Study of Cefiderocol (S-649266) for the Treatment of Nosocomial Pneumonia Caused by Gram-negative Pathogens
CTID: NCT03032380
Phase: Phase 3    Status: Completed
Date: 2020-11-13
Study the Efficacy and Safety of PTK 0796 in Patients With Complicated Skin and Skin Structure Infection (CSSSI)
CTID: NCT03716024
Phase: Phase 2    Status: Completed
Date: 2020-10-12
Staphylococcus Aureus Bacteremia Antibiotic Treatment Options
CTID: NCT01792804
Phase: Phase 3    Status: Completed
Date: 2020-05-27
Imipenem/Relebactam/Cilastatin Versus Piperacillin/Tazobactam for Treatment of Participants With Bacterial Pneumonia (MK-7655A-014)
CTID: NCT02493764
Phase: Phase 3    Status: Completed
Date: 2020-04-16
Pharmacokinetics of Linezolid in Children With Cystic Fibrosis
CTID: NCT00625703
Phase: Phase 2    Status: Completed
Date: 2020-03-27
Study to Compare Delafloxacin to Moxifloxacin for the Treatment of Adults With Community-acquired Bacterial Pneumonia
CTID: NCT02679573
Phase: Phase 3    Status: Completed
Date: 2020-02-27
Study of Debio 1450 for Bacterial Skin Infections
CTID: NCT02426918
Phase: Phase 2    Status: Completed
Date: 2019-11-13
Asymptomatic Colonization With S. Aureus After Therapy With Linezolid or Clindamycin for Acute Skin Infections
CTID: NCT01619410
Phase: N/A    Status: Terminated
Date: 2019-11-08
Study to Compare Lefamulin to Moxifloxacin (With or Without Linezolid) for the Treatment of Adults With Pneumonia
CTID: NCT02559310
Phase: Phase 3    Status: Completed
Date: 2019-10-23
A Study to Assess Objective Endpoint Measurements of Response in Bacterial Skin Infections
CTID: NCT01283581
Phase: Phase 2    Status: Completed
Date: 2019-10-16
Sivextro in Acute Bacterial Skin and Skin Structure Infection (ABSSSI) in Hospitalized Patients. A Global Observational Study
CTID: NCT02991131
Phase:    Status: Terminated
Date: 2019-10-02
Tedizolid Phosphate (TR-701 FA, MK-1986) vs Linezolid for the Treatment of Nosocomial Pneumonia (MK-1986-002)
CTID: NCT02019420
Phase: Phase 3    Status: Completed
Date: 2019-06-27
Safety and Efficacy of CEM-102 Compared to Linezolid in Acute Bacterial Skin Infections
CTID: NCT00948142
Phase: Phase 2    Status: Completed
Date: 2019-04-19
Oral Sodium Fusidate (CEM-102) Versus Oral Linezolid for the Treatment of Acute Bacterial Skin and Skin Structure Infections
CTID: NCT02570490
Phase: Phase 3    Status: Completed
Date: 2019-04-19
Retrospective Real-word Study of Linezolid for the Treatment of Tuberculous Meningitis
CTID: NCT03898635
Phase:    Status: Unknown status
Date: 2019-04-02
Omadacycline Versus Linezolid for the Treatment of ABSSSI (EudraCT #2013-003644-23)
CTID: NCT02378480
Phase: Phase 3    Status: Completed
Date: 2019-03-21
Linezolid in Healthy Volunteers
CTID: NCT03841721
Phase: Phase 1    Status: Unknown status
Date: 2019-02-15
Treatment Shortening of MDR-TB Using Existing and New Drugs
CTID: NCT02619994
Phase: Phase 2    Status: Unknown status
Date: 2019-02-04
Pharmacokinetic and Therapeutic Adaptation of Linezolid in the Treatment of Multi-Resistant Tuberculosis
CTID: NCT02778828
Phase: N/A    Status: Completed
Date: 2019-02-04
A Phase 2 Trial to Evaluate the Efficacy and Safety of Linezolid in Tuberculosis Patients. (LIN-CL001)
CTID: NCT02279875
Phase: Phase 2    Status: Completed
Date: 2019-01-11
Oral Omadacycline vs. Oral Linezolid for the Treatment of ABSSSI
CTID: NCT02877927
Phase: Phase 3    Status: Completed
Date: 2018-11-30
Early Bactericidal Activity of Linezolid, Gatifloxacin, Levofloxacin, Isoniazid (INH) and Moxifloxacin in HIV Negative Adults With Initial Episodes of Sputum Smear-Positive Pulmonary Tuberculosis
CTID: NCT00396084
Phase: Phase 1/Phase 2    Status: Completed
Date: 2018-11-08
Safety and Efficacy of BAY1192631 in Japanese Patients With Methicillin-resistant Staphylococcus Aureus (MRSA) Infections
CTID: NCT01967225
Phase: Phase 3    Status: Completed
Date: 2018-1
Efficacy and safety of different antimicrobial DURATions for the treatment of infections associated with Osteosynthesis
CTID: null
Phase: Phase 3    Status: Ongoing
Date: 2022-02-14
Oral antimicrobial treatment vs. outpatient parenteral for infective
CTID: null
Phase: Phase 4    Status: Ongoing
Date: 2021-12-15
IMPACT ON THE CLINICAL OUTCOMES AND COST-EFFECTIVENESS OF THE ANTIMICROBIAL THERAPEUTIC MONITORING PROGRAM IN CRITICAL PATIENTS
CTID: null
Phase: Phase 4    Status: Ongoing
Date: 2021-05-17
Study on linezolid use and adverse drug reactions in clinical practice: Follow-up of patients treated in Belgian Hospitals
CTID: null
Phase: Phase 4    Status: Completed
Date: 2021-04-21
Evaluation of the clinical implementation of biofilm susceptibility to antibiotics using Minimum Biofilm Eradication Concentration (MBEC) in addition to Minimum Inhibitory Concentration (MIC) to guide the treatment of periprosthetic joint infections; a prospective randomized clinical trial
CTID: null
Phase: Phase 4    Status: Trial now transitioned
Date: 2020-11-17
Use of repeated Multiple Breath Washout to detect and treat pulmonary exacerbation in children with Cystic Fibrosis, a multicenter randomized controlled study.
CTID: null
Phase: Phase 4    Status: Ongoing, Prematurely Ended
Date: 2020-04-08
Partial oral antibiotic treatment for bacterial brain abscess: An open-label randomised non-inferiority trial (ORAL)
CTID: null
Phase: Phase 4    Status: Trial now transitioned
Date: 2019-10-02
A randomized, observer-blinded, active-controlled, Phase IIIb study to compare IV / Oral delafloxacin fixed-dose monotherapy with best available treatments in a microbiologically enriched population with surgical site infections
CTID: null
Phase: Phase 3    Status: Completed, Prematurely Ended
Date: 2019-08-19
Effects of antibiotics on micobiota, pulmonary immune response and incidence of ventilator-associated infections
CTID: null
Phase: Phase 4    Status: Prematurely Ended
Date: 2019-01-14
Pharmacokinetics of different antibiotics in cerebrospinal fluid in children with malignant brain tumors – a pilot study
CTID: null
Phase: Phase 1    Status: Ongoing
Date: 2018-09-27
Pharmacokinetics of antibiotics in cerebrospinal fluid of children with external ventricular drain
CTID: null
Phase: Phase 4    Status: Ongoing
Date: 2018-09-27
Randomized clinical trial on the need for antibiotic to treat low-risk catheter bacteremia due to coagulase-negative staphylococci.
CTID: null
Phase: Phase 4    Status: Prematurely Ended
Date: 2018-04-25
Efficacy of seven and fourteen days of antibiotic treatment in uncomplicated Staphylococcus aureus bacterermia:
CTID: null
Phase: Phase 4    Status: Ongoing
Date: 2017-10-19
Antimicrobial treatment in patients with ventilator-associated tracheobronchitis: a prospective randomized placebo-controlled double-blind multicenter trial
CTID: null
Phase: Phase 3    Status: Ongoing
Date: 2016-09-06
Linezolid continuous infusion: optimizing the dosage regimen in critically ill patients through a pharmacokinetic / pharmacodynamic analysis
CTID: null
Phase: Phase 3    Status: Ongoing
Date: 2016-02-08
A PHASE 3, MULTICENTER, RANDOMIZED, DOUBLE-BLIND, COMPARATOR-CONTROLLED STUDY TO EVALUATE THE SAFETY AND EFFICACY OF INTRAVENOUS TO ORAL DELAFLOXACIN IN ADULT SUBJECTS WITH COMMUNITY-ACQUIRED BACTERIAL PNEUMONIA
CTID: null
Phase: Phase 3    Status: Completed
Date: 2016-02-04
A Phase 3, Randomized, Double-Blind, Double-Dummy Study to Compare the Efficacy and Safety of Lefamulin (BC 3781) Versus Moxifloxacin (With or Without Adjunctive Linezolid) in Adults With Community-Acquired Bacterial Pneumonia
CTID: null
Phase: Phase 3    Status: Completed
Date: 2016-01-12
A Phase 3 Randomized, Double-Blind, Multi-Center Study to Compare the Safety and Efficacy of Omadacycline IV/PO to Linezolid IV/PO for Treating Adult Subjects with Acute Bacterial Skin and Skin Structure Infection (ABSSSI)
CTID: null
Phase: Phase 3    Status: Completed
Date: 2015-07-02
PHASE 3 STUDY OF IV TO ORAL 6-DAY TEDIZOLID PHOSPHATE COMPARED WITH 10-DAY COMPARATOR IN SUBJECTS
CTID: null
Phase: Phase 3    Status: Prematurely Ended, Completed
Date: 2015-05-06
A Phase 3, double-blinded, randomized, comparator trial of the safety and efficacy of a single dose of dalbavancin to twice daily linezolid for the treatment of community acquired bacterial pneumonia
CTID: null
Phase: Phase 3    Status: Prematurely Ended
Date: 2015-03-30
A prospective, open label, randomised controlled clinical trial, with pharmacokinetic-pharmacodynamic validation, to compare antimicrobial treatment with oral minocycline plus rifampicin to treatment with oral linezolid for complicated skin and skin structure infections (cSSSI) caused by Methicillin resistant Staphylococcus aureus (MRSA).
CTID: null
Phase: Phase 4    Status: Completed
Date: 2014-07-11
A Phase 3 Randomised Double-Blind Study Comparing TR-701FA and Linezolid in Ventilated Gram-Positive Nosocomial Pneumonia
CTID: null
Phase: Phase 3    Status: Prematurely Ended, Completed
Date: 2014-04-17
Investigation of pharmacokinetics of Linezolid and Meropenem in patients receiving continuous renal replacement therapy
CTID: null
Phase: Phase 4    Status: Ongoing
Date: 2013-10-16
EARLY ORAL SWITCH THERAPY IN LOW-RISK STAPHYLOCOCCUS AUREUS BLOODSTREAM INFECTION
CTID: null
Phase: Phase 3    Status: Completed
Date: 2013-07-11
Perioperative complications in obese and non-obese patients: Prevention and treatment of wound infections and post-operative pain.
CTID: null
Phase: Phase 3    Status: Completed
Date: 2013-05-13
Investigation on the peritoneal tissue concentrations of antibiotics in surgical patients with peritonitis using microdialysis. Example: linezolide, tigecycline
CTID: null
Phase: Phase 4    Status: Completed
Date: 2013-01-22
Pharmacokinetic and pharmacodynamic evaluation of linezolid administered intravenously in MRSA-positive, morbidly obese patients with pneumonia.
CTID: null
Phase: Phase 3    Status: Completed
Date: 2013-01-11
Prospective, Randomized, open label, European, multicenter study of the efficacy of the linezolid-rifampin combination versus standard of care in the treatment of Gram-positive prosthetic hip joint infection
CTID: null
Phase: Phase 2    Status: Completed
Date: 2012-10-08
Pre-emptive targeted and optimized treatment of critical airway colonization to prevent Ventilator Associated Pneumonia: a randomized controlled study
CTID: null
Phase: Phase 4    Status: Ongoing
Date: 2012-08-22
Tissue concentration of cefazolin and linezolid in sternal spongiosa in elective coronary artery bypass grafting: an in-vivo microdialysis study
CTID: null
Phase: Phase 2    Status: Ongoing
Date: 2012-05-14
A PHASE 3, RANDOMIZED, DOUBLE-BLIND, DOUBLE-DUMMY STUDY TO COMPARE THE EFFICACY AND SAFETY OF DALBAVANCIN TO A COMPARATOR REGIMEN (VANCOMYCIN AND LINEZOLID) FOR THE TREATMENT OF ACUTE BACTERIAL SKIN AND SKIN STRUCTURE INFECTIONS
CTID: null
Phase: Phase 3    Status: Completed
Date: 2012-05-04
Plasma and synovial fluid pharmacokinetics of cefuroxime and linezolid in patients undergoing elective knee arthroscopy after a single intravenous dose of cefuroxime and linezolid: an exploratory microdialysis study
CTID: null
Phase: Phase 4    Status: Completed
Date: 2012-05-04
Assessment of extracellular concentrations of linezolid and other currently approved antibiotics in patients presenting with severe bacterial lung infection
CTID: null
Phase: Phase 4    Status: Ongoing
Date: 2012-01-19
A Phase 3 Randomized, Double-Blind, Multicenter Study Comparing
CTID: null
Phase: Phase 3    Status: Completed
Date: 2011-12-21
Individualizing duration of antibiotic therapy in hospitalized patients with community-acquired pneumonia: a non-inferiority, randomized, controlled trial.
CTID: null
Phase: Phase 4    Status: Ongoing
Date: 2011-12-20
A PHASE 3, RANDOMIZED, DOUBLE-BLIND, DOUBLE-DUMMY STUDY TO COMPARE THE EFFICACY AND SAFETY OF DALBAVANCIN TO A COMPARATOR REGIMEN (VANCOMYCIN AND LINEZOLID) FOR THE
CTID: null
Phase: Phase 3    Status: Completed
Date: 2011-11-23
The pharmacokinetic effect of clarithromycin on the AUC0-12h of linezolid in multidrug-resistant and extensively drug-resistant tuberculosis patients
CTID: null
Phase: Phase 1, Phase 4    Status: Ongoing
Date: 2011-07-27
A prospective single blind randomised controlled study to compare the outcomes of patients with diabetes and clinically non-infected ischaemic and neuropathic foot ulcers treated with and without oral antibiotics
CTID: null
Phase: Phase 4    Status: Prematurely Ended
Date: 2011-07-13
Concentración y actividad antibiótica en soluciones de sellado
CTID: null
Phase: Phase 4    Status: Completed
Date: 2011-06-17
Pk/Pd aspects of linezolid in critically ill patients with late onset Ventilator-Associated Pneumonia. Intermittent versus Continuous Infusion Introduction
CTID: null
Phase: Phase 4    Status: Ongoing
Date: 2011-04-21
Target site pharmacokinetics of linezolid or tigecycline in patients with severe skin and skin structure infections (SSSI) associated with chronic fistulas.
CTID: null
Phase: Phase 4    Status: Prematurely Ended
Date: 2011-03-17
A Phase 3 Randomized, Double-Blind, Multicenter Study Comparing
CTID: null
Phase: Phase 3    Status: Completed
Date: 2011-03-08
treatment of healthcare-associated pneumonia: a prospective, multicenter study
CTID: null
Phase: Phase 4    Status: Ongoing
Date: 2009-03-23
PROSPECTIVE STUDY OF OPHTHALMOLOGIC FUNCTION IN PATIENTS RECEIVING LINEZOLID FOR TWO MONTHS OR GREATER
CTID: null
Phase: Phase 3    Status: Prematurely Ended
Date: 2008-12-17
Target site pharmacokinetics of linezolid after single and multiple doses in diabetes patients with soft tissue infection
CTID: null
Phase: Phase 4    Status: Completed
Date: 2008-04-28
“Necesidad de tratamiento antibiótico de la bacteriemia relacionada con catéteres venosos centrales producida por Staphylococcus coagulasa-negativa tras la retirada del catéter en pacientes de bajo riesgo”
CTID: null
Phase: Phase 4    Status: Ongoing
Date: 2008-04-22
LINEZOLID IN THE TREATMENT OF SUBJECTS WITH NOSOCOMIAL PNEUMONIA PROVEN TO BE DUE TO METHICILLIN-RESISTANT STAPHYLOCOCCUS AUREUS
CTID: null
Phase: Phase 4    Status: Prematurely Ended, Completed
Date: 2007-11-26
Etude de pharmacocinétique de population du linézolide chez des patients de réanimation présentant une infection sévère à Staphylocoque doré méticillino-résistant
CTID: null
Phase: Phase 4    Status: Ongoing
Date: 2007-10-17
LINEZOLID IN THE TREATMENT OF SUBJECTS WITH COMPLICATED SKIN AND SOFT TISSUE INFECTIONS PROVEN TO BE DUE TO METHICILLIN-RESISTANT STAPHYLOCOCCUS AUREUS
CTID: null
Phase: Phase 4    Status: Completed
Date: 2007-08-27
Randomized, Double-Blind, Multicenter Study of Ceftobiprole Medocaril Versus Ceftriaxone with/without Linezolid in Treatment of Subjects Hospitalized With Community-Acquired Pneumonia
CTID: null
Phase: Phase 3    Status: Completed
Date: 2006-10-10
Rescue therapy for indwelling central venous Hickmann-Broviac catheter related infections with antibiotic lock technique
CTID: null
Phase: Phase 2    Status: Completed
Date: 2006-09-06
Rescue therapy of tunnel/pocket infections of Hickman-Broviac/Port indwelling central venous catheters with antibiotic continuous infusion
CTID: null
Phase: Phase 2    Status: Completed
Date: 2006-09-06
A Phase 3 Randomized, Double-Blind Study of Ceftobiprole Medocaril versus Linezolid Plus Ceftazidime in the Treatment of Nosocomial Pneumonia
CTID: null
Phase: Phase 3    Status: Completed
Date: 2006-08-24
Serum concentrations and concentrations in epithelial lining fluid of antibiotics under continious infusion
CTID: null
Phase: Phase 4    Status: Completed
Date: 2006-04-13
Phase 3, randomized, investigator-blind, multi-center study to evaluate efficacy and safety of intravenous iclaprim versus intravenous linezolid in complicated skin and skinstructure infections
CTID: null
Phase: Phase 3    Status: Completed
Date: 2006-01-31
Phase 3, Randomized, Investigator-Blind, Multi-Center Study to Evaluate Efficacy and Safety of Intravenous Iclaprim versus Intravenous Linezolid in Complicated Skin and Skin Structure Infections
CTID: null
Phase: Phase 3    Status: Completed
Date: 2005-06-21
Linezolid vs Vancomycin/Cefazolin in the treatment of hemodialysis patients with catheter-related gram-positive bloodstream infections
CTID: null
Phase: Phase 3    Status: Prematurely Ended, Completed
Date: 2005-04-28
Linezolid vs. Vancomycin/Oxacillin/Dicloxacillin in the Treatment of Catheter-Related Gram Positive Bloodstream Infections.
CTID: null
Phase: Phase 3    Status: Completed
Date: 2004-12-08
A Phase 3, Randomized Double-Blind Study of Ceftobiprole Medocaril Versus Linezolid Plus Ceftazidime in the Treatment of Nosocomial Pneumonia
CTID: null
Phase: Phase 3    Status: Completed
Date: 2004-09-16

Contact Us