CLINICAL PHARMACOLOGY
Mechanism Of Action
Tedizolid phosphate is the prodrug of tedizolid, an
antibacterial agent [see Pharmacokinetics and Microbiology].
Pharmacodynamics
The AUC/minimum inhibitory concentration (MIC) was shown
to best correlate with tedizolid activity in animal infection models.
In the mouse thigh infection model of S. aureus,
antistaphylococcal killing activity was impacted by the presence of
granulocytes. In granulocytopenic mice (neutrophil count <100 cells/mL),
bacterial stasis was achieved at a human-equivalent dose of approximately 2000
mg/day; whereas, in non-granulocytopenic animals, stasis was achieved at a
human-equivalent dose of approximately 100 mg/day. The safety and efficacy of
SIVEXTRO for the treatment of neutropenic patients (neutrophil counts <1000
cells/mm³) have not been evaluated.
Cardiac Electrophysiology
In a randomized, positive-and placebo-controlled
crossover thorough QTc study, 48 enrolled subjects were administered a single
oral dose of SIVEXTRO at a therapeutic dose of 200 mg, SIVEXTRO at a supratherapeutic
dose of 1200 mg, placebo, and a positive control; no significant effects of
SIVEXTRO on heart rate, electrocardiogram morphology, PR, QRS, or QT interval
were detected. Therefore, SIVEXTRO does not affect cardiac repolarization.
Pharmacokinetics
Tedizolid phosphate is a prodrug that is converted by
phosphatases to tedizolid, the microbiologically active moiety, following oral
and intravenous administration. Only the pharmacokinetic profile of tedizolid
is discussed further due to negligible systemic exposure of tedizolid phosphate
following oral and intravenous administration. Following multiple once-daily
oral or intravenous administration, steady-state concentrations are achieved
within approximately three days with tedizolid accumulation of approximately
30% (tedizolid half-life of approximately 12 hours). Pharmacokinetic (PK)
parameters of tedizolid following oral and intravenous administration of 200 mg
once daily tedizolid phosphate are shown in Table 4.
Table 4: Mean (Standard Deviation) Tedizolid
Pharmacokinetic Parameters Following Single and Multiple Oral and Intravenous
Administration of 200 mg Once-Daily Tedizolid Phosphate
Pharmacokinetic Parameters of Tedizolid* |
Oral |
Intravenous |
Single Dose |
Steady State |
Single Dose |
Steady State |
Cmax (mcg/mL) |
2.0 (0.7) |
2.2 (0.6) |
2.3 (0.6) |
3.0 (0.7) |
Tmax (hr)† |
2.5 (1.0 -8.0) |
3.5 (1.0 -6.0) |
1.1 (0.9 -1.5) |
1.2 (0.9 -1.5) |
AUC (mcg-hr/mL)‡ |
23.8 (6.8) |
25.6 (8.4) |
26.6 (5.2) |
29.2 (6.2) |
CL or CL/F (L/hr) |
6.9 (1.7) |
8.4 (2.1) |
6.4 (1.2) |
5.9 (1.4) |
* Cmax , maximum concentration; Tmax , time to reach Cmax
; AUC, area under the concentration-time curve; CL, systemic clearance; CL/F,
apparent oral clearance
† Median (range)
‡ AUC is AUC0-∞ (AUC from time 0 to infinity)
for single-dose administration and AUC0-24 (AUC from time 0 to 24 hours) for
multiple-dose administration |
Absorption
Peak plasma tedizolid
concentrations are achieved within approximately 3 hours following oral
administration under fasting conditions or at the end of the 1 hour intravenous
infusion of tedizolid phosphate. The absolute bioavailability is approximately
91% and no dosage adjustment is necessary between intravenous and oral
administration.
Tedizolid phosphate (oral) may
be administered with or without food as total systemic exposure (AUC0-∞)
is unchanged between fasted and fed (high-fat, high-calorie) conditions.
Distribution
Protein binding of tedizolid to human plasma proteins is
approximately 70 to 90%. The mean steady state volume of distribution of
tedizolid in healthy adults following a single intravenous dose of tedizolid
phosphate 200 mg ranged from 67 to 80 L (approximately twice total body water).
Tedizolid penetrates into the interstitial space fluid of adipose and skeletal
muscle tissue with exposure similar to free drug exposure in plasma.
Metabolism
Other than tedizolid, which accounts for approximately
95% of the total radiocarbon AUC in plasma, there are no other significant
circulating metabolites in humans.
There was no degradation of tedizolid in human liver
microsomes indicating tedizolid is unlikely to be a substrate for hepatic
CYP450 enzymes.
In vitro studies showed that conjugation of tedizolid is
mediated via multiple sulfotransferase (SULT) isoforms (SULT1A1, SULT1A2, and SULT2A1).
Excretion
Following single oral administration of 14C-labeled
tedizolid phosphate under fasted conditions, the majority of elimination
occurred via the liver, with 82% of the radioactive dose recovered in feces and
18% in urine, primarily as a non-circulating and microbiologically inactive
sulfate conjugate. Most of the elimination of tedizolid (>85%) occurs within
96 hours. Less than 3% of the tedizolid phosphate-administered dose is excreted
in feces and urine as unchanged tedizolid.
Specific Populations
Based on the population pharmacokinetic analysis, there
are no clinically relevant demographic or clinical patient factors (including
age, gender, race, ethnicity, weight, body mass index, and measures of renal or
liver function) that impact the pharmacokinetics of tedizolid.
Hepatic Impairment
Following administration of a single 200 mg oral dose of
SIVEXTRO, no clinically meaningful changes in mean tedizolid Cmax and AUC0-∞
were observed in patients with moderate (n=8) or severe (n=8) hepatic
impairment (Child-Pugh Class B and C) compared to 8 matched healthy control
subjects. No dose adjustment is necessary for patients with hepatic impairment.
Renal Impairment
Following administration of a single 200 mg intravenous
dose of SIVEXTRO to 8 subjects with severe renal impairment defined as eGFR
<30 mL/min/1.73 m², the Cmax was essentially unchanged and AUC0-∞ was
decreased by less than 10% compared to 8 matched healthy control subjects.
Hemodialysis does not result in meaningful removal of tedizolid from systemic
circulation, as assessed in subjects with end-stage renal disease (eGFR <15
mL/min/1.73 m²). No dosage adjustment is necessary in patients with renal
impairment or patients on hemodialysis.
Geriatric Patients
The pharmacokinetics of tedizolid were evaluated in a
Phase 1 study conducted in elderly healthy volunteers (age 65 years and older,
with at least 5 subjects at least 75 years old; n=14) compared to younger
control subjects (25 to 45 years old; n=14) following administration of a
single oral dose of SIVEXTRO 200 mg. There were no clinically meaningful
differences in tedizolid Cmax and AUC0-∞ between elderly subjects and
younger control subjects. No dosage adjustment of SIVEXTRO is necessary in
elderly patients.
Gender
The impact of gender on the pharmacokinetics of SIVEXTRO
was evaluated in clinical trials of healthy males and females and in a
population pharmacokinetics analysis. The pharmacokinetics of tedizolid were
similar in males and females. No dosage adjustment of SIVEXTRO is necessary
based on gender.
Drug Interaction Studies
Drug Metabolizing Enzymes
Transformation via Phase 1 hepatic oxidative metabolism
is not a significant pathway for elimination of SIVEXTRO. 9
Neither SIVEXTRO nor tedizolid detectably inhibited or
induced the metabolism of selected CYP enzyme substrates, suggesting that
drug-drug interactions based on oxidative metabolism are unlikely.
Membrane Transporters
The potential for tedizolid or tedizolid phosphate to
inhibit transport of probe substrates of important drug uptake (OAT1, OAT3,
OATP1B1, OATP1B3, OCT1, and OCT2) and efflux transporters (P-gp and BCRP) was
tested in vitro. No clinically relevant interactions are expected to occur with
these transporters except BCRP.
Coadministration of multiple oral doses of SIVEXTRO (200
mg once daily) increased the Cmax and AUC of rosuvastatin (10 mg single oral
dose), a known BCRP substrate, by approximately 55% and 70%, respectively, in
healthy subjects [see DRUG INTERACTIONS].
Monoamine Oxidase Inhibition
Tedizolid is a reversible inhibitor of monoamine oxidase
(MAO) in vitro. The interaction with MAO inhibitors could not be evaluated in
Phase 2 and 3 trials, as subjects taking such medications were excluded from
the trials.
Adrenergic Agents
Two placebo-controlled crossover studies were conducted
to assess the potential of 200 mg oral SIVEXTRO at steady state to enhance
pressor responses to pseudoephedrine and tyramine in healthy individuals. No
meaningful changes in blood pressure or heart rate were seen with
pseudoephedrine. The median tyramine dose required to cause an increase in
systolic blood pressure of ≥ 30 mmHg from pre-dose baseline was 325 mg
with SIVEXTRO compared to 425 mg with placebo. Palpitations were reported in
21/29 (72.4%) subjects exposed to SIVEXTRO compared to 13/28 (46.4%) exposed to
placebo in the tyramine challenge study.
Serotonergic Agents
Serotonergic effects at doses of tedizolid phosphate up
to 30-fold above the human equivalent dose did not differ from vehicle control
in a mouse model that predicts serotonergic activity. In Phase 3 trials,
subjects taking serotonergic agents including antidepressants such as selective
serotonin reuptake inhibitors (SSRIs), tricyclic antidepressants, and serotonin
5-hydroxytryptamine (5-HT1) receptor agonists (triptans), meperidine, or
buspirone were excluded.
Microbiology
Tedizolid belongs to the oxazolidinone class of
antibacterial drugs.
Mechanism Of Action
The antibacterial activity of tedizolid is mediated by
binding to the 50S subunit of the bacterial ribosome resulting in inhibition of
protein synthesis. Tedizolid inhibits bacterial protein synthesis through a
mechanism of action different from that of other nonoxazolidinone class
antibacterial drugs; therefore, cross-resistance between tedizolid and other
classes of antibacterial drugs is unlikely. The results of in vitro time-kill
studies show that tedizolid is bacteriostatic against enterococci, staphylococci,
and streptococci.
Mechanism Of Resistance
Organisms resistant to oxazolidinones via mutations in
chromosomal genes encoding 23S rRNA or ribosomal proteins (L3 and L4) are
generally cross-resistant to tedizolid. In the limited number of Staphylococcus
aureus strains tested, the presence of the chloramphenicol-florfenicol
resistance (cfr) gene did not result in resistance to tedizolid in the absence
of chromosomal mutations.
Frequency Of Resistance
Spontaneous mutations conferring reduced susceptibility
to tedizolid occur in vitro at a frequency rate of approximately 10-10.
Interaction With Other Antimicrobial Drugs
In vitro drug combination studies with tedizolid and
aztreonam, ceftriaxone, ceftazidime, imipenem, rifampin,
trimethoprim/sulfamethoxazole, minocycline, clindamycin, ciprofloxacin,
daptomycin, vancomycin, gentamicin, amphotericin B, ketoconazole, and
terbinafine demonstrate neither synergy nor antagonism.
Spectrum Of Activity
Tedizolid has been shown to be active against most
isolates of the following bacteria, both in vitro and in clinical infections,
as described in Indications and Usage (1).
Aerobic And Facultative Gram-positive Bacteria
Staphylococcus aureus (including
methicillin-resistant [MRSA] and methicillin-susceptible [MSSA] isolates)
Streptococcus pyogenes Streptococcus agalactiae
Streptococcus anginosus Group (including S. anginosus, S.
intermedius, and S. constellatus)
Enterococcus faecalis
The following in vitro data are available, but their
clinical significance has not been established. At least 90% of the following
microorganisms exhibit an in vitro minimum inhibitory concentration (MIC) less
than or equal to 0.5 mcg/mL for tedizolid. However, the safety and
effectiveness of SIVEXTRO in treating clinical infections due to these
microorganisms have not been established in adequate and well-controlled
clinical trials.
Aerobic And Facultative Anaerobic Gram-Positive Bacteria
Staphylococcus epidermidis (including
methicillin-susceptible and methicillin-resistant isolates)
Staphylococcus haemolyticus
Staphylococcus lugdunensis
Enterococcus faecium
Susceptibility Test Methods
When available, the clinical microbiology laboratory
should provide cumulative results of the in vitro susceptibility test results
for antimicrobial drugs used in local hospitals and practice areas to the
physician as periodic reports that describe the susceptibility profile of
nosocomial and community-acquired pathogens. These reports should aid the
physician in selecting an effective antibacterial drug for treatment.
Dilution Techniques
Quantitative methods are used to determine antimicrobial
minimum inhibitory concentrations (MICs). These MIC values provide estimates of
the susceptibility of bacteria to antimicrobial compounds. The MIC values
should be determined using a standardized procedure based on dilution methods
(broth, agar, or microdilution) or equivalent using standardized inoculum and concentrations
of tedizolid.1,3 The MIC values should be interpreted according to
the criteria provided in Table 5.
Table 5: Susceptibility Test Interpretive Criteria for
SIVEXTRO
Pathogen |
Minimum Inhibitory Concentrations (mcg/mL) |
Disk Diffusion Zone Diameter (mm) |
S |
I |
R |
S |
I |
R |
Staphylococcus aureus (methicillin-resistant and methicillin-susceptible isolates) |
≤ 0.5 |
1 |
≥ 2 |
≥ 19 |
16 -18 |
≤ 15 |
Streptococcus pyogenes |
≤ 0.5 |
- |
- |
≥ 18 |
- |
- |
Streptococcus agalactiae |
≤ 0.5 |
- |
- |
≥ 18 |
- |
- |
Streptococcus anginosus Group* |
≤ 0.25 |
- |
- |
≥ 17 |
- |
- |
Enterococcus faecalis |
≤ 0.5 |
- |
- |
≥ 19 |
- |
- |
S=susceptible, I=intermediate,
R=resistant
* Includes S. anginosus, S. intermedius, S. constellatus |
Diffusion Techniques
Quantitative methods that require measurement of zone
diameters also provide reproducible estimates of the susceptibility of bacteria
to antimicrobial compounds. The standardized procedure requires the use of
standardized inoculum concentrations.2,3 This procedure uses paper
disks impregnated with 20 mcg tedizolid to test the susceptibility of
microorganisms to tedizolid. Reports from the laboratory providing results of
the standard single-disk susceptibility test with a 20 mcg tedizolid disk
should be interpreted according to the criteria in Table 5.
A report of “Susceptible”
indicates that the antimicrobial drug is likely to inhibit growth of the
pathogen if the antimicrobial drug reaches the concentration usually achievable
at the site of infection. A report of “Intermediate” indicates that the result
should be considered equivocal, and if the microorganism is not fully
susceptible to alternative drugs, the test should be repeated. This category
implies possible clinical efficacy in body sites where the drug is
physiologically concentrated. This category also provides a buffer zone that
prevents small uncontrolled technical factors from causing major discrepancies
in interpretation. A report of “Resistant” indicates that the antimicrobial
drug is not likely to inhibit growth of the pathogen if the antimicrobial drug
reaches the concentrations usually achievable at the infection site; other
therapy should be selected.
Quality Control
Standardized susceptibility test procedures require the
use of laboratory control microorganisms to monitor and ensure the accuracy and
precision of supplies and reagents used in the assay, and the techniques of the
individuals performing the test.1,2,3 Standardized tedizolid powder
should provide the following range of MIC values noted in Table 6. For the
diffusion technique using the 20 mcg tedizolid disk, results within the ranges
specified in Table 6 should be observed.
Table 6: Acceptable Quality
Control Ranges for Susceptibility Testing
Quality Control Organism |
Minimum Inhibitory Concentrations (mcg/mL) |
Disk Diffusion (zone diameter in mm) |
Staphylococcus aureus ATCC 29213 |
0.12 -1 |
Not Applicable |
Staphylococcus aureus ATCC 25923* |
0.12 - 0.5 |
22 -29 |
Enterococcus faecalis ATCC 29212 |
0.25 -1 |
Not Applicable |
Streptococcus pneumoniae ATCC 49619 |
0.12 -0.5 |
24 -30 |
*QC range for S. aureus ATCC
25923 with tedizolid is 0.12-0.5μg/mL which exhibits less trailing and is
easier to read. This strain is considered supplemental and not required for
routine user QC. |
Animal Toxicology And/Or Pharmacology
Repeated-oral and intravenous
dosing of tedizolid phosphate in rats in 1-month and 3-month toxicology studies
produced dose-and time-dependent bone marrow hypocellularity (myeloid,
erythroid, and megakaryocyte), with associated reduction in circulating RBCs,
WBCs, and platelets. These effects showed evidence of reversibility and
occurred at plasma tedizolid exposure levels (AUC) ≥ 6-fold greater than
the plasma exposure associated with the human therapeutic dose. In a 1-month
immunotoxicology study in rats, repeated oral dosing of tedizolid phosphate was
shown to significantly reduce splenic B cells and T cells and reduce plasma IgG
titers. These effects occurred at plasma tedizolid exposure levels (AUC)
≥ 3-fold greater than the expected human plasma exposure associated with
the therapeutic dose.
Clinical Studies
Acute Bacterial Skin And Skin
Structure Infections
A total of 1333 adults with
acute bacterial skin and skin structure infections (ABSSSI) were randomized in
two multicenter, multinational, double-blind, non-inferiority trials. Both
trials compared SIVEXTRO 200 mg once daily for 6 days versus linezolid 600 mg
every 12 hours for 10 days. In Trial 1, patients were treated with oral
therapy, while in Trial 2, patients could receive oral therapy after a minimum
of one day of intravenous therapy. Patients with cellulitis/erysipelas, major
cutaneous abscess, or wound infection were enrolled in the trials. Patients
with wound infections could have received aztreonam and/or metronidazole as
adjunctive therapy for gram-negative bacterial coverage, if needed. The
intent-to-treat (ITT) patient population included all randomized patients.
In Trial 1, 332 patients with ABSSSI were randomized to
SIVEXTRO and 335 patients were randomized to linezolid. The majority (91%) of
patients treated with SIVEXTRO in Trial 1 were less than 65 years old with a
median age of 43 years (range: 18 to 86 years). Patients treated with SIVEXTRO
were predominantly male (61%) and White (84%); 13% had BMI ≥ 35 kg/m², 8%
had diabetes mellitus, 35% were current or recent intravenous drug users, and
2% had moderate to severe renal impairment. The overall median surface area of
infection was 188 cm². The types of ABSSSI included were cellulitis/erysipelas
(41%), wound infection (29%), and major cutaneous abscess (30%). In addition to
local signs and symptoms of infection, patients were also required to have at
least one regional or systemic sign of infection at baseline, defined as
lymphadenopathy (87% of patients), temperature 38°C or higher (16% of
patients), white blood cell count greater than 10,000 cells/mm³ or less than
4000 cells/mm³ (42%), or 10% or more band forms on white blood cell
differential (4%).
The primary endpoint in Trial 1 was early clinical
response defined as no increase from baseline lesion area at 48-72 hours after
the first dose and oral temperature of ≤ 37.6°C, confirmed by a second
temperature measurement within 24 hours in the ITT population.
In Trial 2, 332 patients with ABSSSI were randomized to
SIVEXTRO and 334 patients were randomized to linezolid. The majority (87%) of
patients treated with SIVEXTRO in Trial 2 were less than 65 years old with a
median age of 46 years (range: 17 to 86 years). Patients treated with SIVEXTRO
were predominantly male (68%) and White (86%); 16% had BMI ≥ 35 kg/m², 10%
had diabetes mellitus, 20% were current or recent intravenous drug users, and
4% had moderate to severe renal impairment. The overall median surface area of
infection was 231 cm². The types of ABSSSI included were cellulitis/erysipelas
(50%), wound infection (30%), and major cutaneous abscess (20%). In addition to
local signs and symptoms of infection, patients were also required to have at
least one regional or systemic sign of infection at baseline, defined as
lymphadenopathy (71% of patients), temperature 38°C or higher (31% of patients),
white blood cell count greater than 10,000 cells/mm³ or less than 4000 cells/mm³
(53%), or 10% or more band forms on white blood cell differential (16%).
The primary endpoint in Trial 2 was early clinical
response defined as at least a 20% decrease from baseline lesion area at 48-72
hours after the first dose in the ITT population (Table 7).
Table 7: Early Clinical Response in the ITT Patient
Population
|
SIVEXTRO (200 mg) |
Linezolid (1200 mg) |
Treatment Difference (2-sided 95% CI) |
No increase in lesion surface area from baseline and oral temperature of ≤ 37.6°C, confirmed by a second temperature
measurement within 24 hours at 48-72 hours* |
Trial 1, N |
332 |
335 |
|
Responder, n (%) |
264 (79.5) |
266 (79.4) |
0.1 (-6.1, 6.2) |
Trial 2, N |
332 |
334 |
|
Responder, n (%) |
286 (86.1) |
281 (84.1) |
2.0 (-3.5, 7.3) |
At least a 20% decrease from baseline in lesion area at 48-72 hours†. |
Trial 1, N |
332 |
335 |
|
Responder, n (%) |
259 (78.0) |
255 (76.1) |
1.9 (-4.5, 8.3) |
Trial 2, N |
332 |
334 |
|
Responder, n (%) |
283 (85.2) |
276 (82.6) |
2.6 (-3.0, 8.2) |
CI=confidence interval
* Primary endpoint for Trial 1; sensitivity analysis for Trial 2
† Primary endpoint for Trial 2; sensitivity analysis for Trial 1 |
An investigator assessment of
clinical response was made at the post-therapy evaluation (PTE) (7 -14 days
after the end of therapy) in the ITT and CE (Clinically Evaluable) populations.
Clinical success was defined as resolution or near resolution of most
disease-specific signs and symptoms, absence or near resolution of systemic
signs of infection if present at baseline (lymphadenopathy, fever, >10%
immature neutrophils, abnormal WBC count), and no new signs, symptoms, or
complications attributable to the ABSSSI requiring further treatment of the
primary lesion (Table 8).
Table 8:
Investigator-Assessed Clinical Response at Post-therapy Evaluation in ITT and
CE Patient Populations from Two Phase 3 ABSSSI Trials
|
SIVEXTRO (200 mg)
n/N (%) |
Linezolid (1200 mg)
n/N (%) |
Treatment Difference (2-sided 95% CI) |
Trial 1 |
ITT |
284/332 (85.5) |
288/335 (86.0) |
-0.5 (-5.8, 4.9) |
CE |
264/279 (94.6) |
267/280 (95.4) |
-0.8 (-4.6, 3.0) |
Trial 2 |
ITT |
292/332 (88.0) |
293/334 (87.7) |
0.3 (-4.8, 5.3) |
CE |
268/290 (92.4) |
269/280 (96.1) |
-3.7 (-7.7, 0.2) |
CI=confidence interval;
ITT=intent-to-treat; CE=clinically evaluable |
Clinical success by baseline
pathogens from the primary infection site or blood cultures for the
microbiological intent-to-treat (MITT) patient population for two integrated
Phase 3 ABSSSI studies are presented in Table 9 and Table 10.
Table 9: Early Clinical
Response by Baseline Pathogen from Two Phase 3 ABSSSI Trials (MITT Population)
Pathogen |
No increase in lesion surface area from baseline and oral temperature of ≤ 37.6°C* |
At least a 20% decrease from baseline in lesion area† |
SIVEXTRO (200 mg)
n/N (%) |
Linezolid (1200 mg)
n/N (%) |
SIVEXTRO (200 mg)
n/N (%) |
Linezolid (1200 mg)
n/N (%) |
Staphylococcus aureus |
276/329 (83.9) |
278/342 (81.3) |
280/329 (85.1) |
276/342 (80.7) |
Methicillin-resistant S. aureus |
112/141 (79.4) |
113/146 (77.4) |
114/141 (80.9) |
111/146 (76.0) |
Methicillin-susceptible S. aureus |
164/188 (87.2) |
167/198 (84.3) |
166/188 (88.3) |
167/198 (84.3) |
Streptococcus pyogenes |
27/33 (81.8) |
18/20 (90.0) |
25/33 (75.8) |
16/20 (80.0) |
Streptococcus anginosus Group |
22/30 (73.3) |
26/28 (92.9) |
22/30 (73.3) |
25/28 (89.3) |
Streptococcus agalactiae |
6/9 (66.7) |
8/10 (80.0) |
6/9 (66.7) |
7/10 (70.0) |
Enterococcus faecalis |
7/10 (70.0) |
3/4 (75.0) |
6/10 (60.0) |
1/4 (25.0) |
Pooled analysis; n=number of patients in the specific
category; N=Number of patients with the specific pathogen isolated from the
ABSSSI
* Primary endpoint of Trial 1
† Primary endpoint of Trial 2
Baseline bacteremia in the tedizolid arm with relevant pathogens included two
subjects with MRSA, four subjects with MSSA, two subjects with S. pyogenes, one
subject with S. agalactiae, and one subject with S. constellatus. All of these
subjects were Responders at the 48-72 hour evaluation. At the Post-therapy
Evaluation (PTE), 8 of 10 subjects were considered clinical successes. |
Table 10: Clinical Response
at PTE by Baseline Pathogen from Two Phase 3 ABSSSI Trials (MITT Population)
Pathogen |
Clinical Response at PTE |
SIVEXTRO (200 mg)
n/N (%) |
Linezolid (1200 mg)
n/N (%) |
Staphylococcus aureus |
291/329 (88.5) |
303/342 (88.6) |
Methicillin-resistant S. aureus |
118/141 (83.7) |
119/146 (81.5) |
Methicillin-susceptible S. aureus |
173/188 (92.0) |
186/198 (93.9) |
Streptococcus pyogenes |
30/33 (90.9) |
19/20 (95.0) |
Streptococcus anginosus Group |
21/30 (70.0) |
25/28 (89.3) |
Streptococcus agalactiae |
8/9 (88.9) |
8/10 (80.0) |
Enterococcus faecalis |
7/10 (70.0) |
4/4 (100.0) |
Pooled analysis; n=number of
patients in the specific category; N=Number of patients with the specific
pathogen isolated from the ABSSSI
Baseline bacteremia in the tedizolid arm with relevant pathogens included two
subjects with MRSA, four subjects with MSSA, two subjects with S. pyogenes, one
subject with S. agalactiae, and one subject with S. constellatus. All of these
subjects were Responders at the 48-72 hour evaluation. At the Post-therapy
Evaluation (PTE) 8 of 10 subjects were considered clinical successes. |
REFERENCES
1. Clinical and Laboratory Standards Institute (CLSI).
Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria that Grow Aerobically;
Approved Standard - 10th ed., CLSI document M7 A10. Wayne, PA: Clinical and
Laboratory Standards Institute; 2015.
2. Clinical and Laboratory Standards Institute (CLSI).
Performance Standards for Antimicrobial Disk Susceptibility Tests, Approved
Standard - 12th ed. CLSI document M2 A12 (ISBN 1-56238-781-2 [Print]; ISBN
1-56238-782-0 [Electronic]). Clinical and Laboratory Standards Institute, 950
West Valley Road, Suite 2500, Wayne, Pennsylvania 19087 USA, 2015.
3. Clinical and Laboratory Standards Institute (CLSI).
Performance Standards for Antimicrobial Susceptibility Testing - 27th
Informational Supplement. CLSI document M100 S27 (ISBN 1-56238-865-7 [Print];
ISBN 1-56238-866-5 [Electronic]). Clinical and Laboratory Standards Institute,
950 West Valley Road, Suite 2500, Wayne, Pennsylvania 19087 USA, 2017.