CLINICAL PHARMACOLOGY
Mechanism Of Action
Daptomycin is an antibacterial drug [see Microbiology].
Pharmacodynamics
Based on animal models of infection, the antimicrobial
activity of daptomycin appears to correlate with the AUC/MIC (area under the
concentration-time curve/minimum inhibitory concentration) ratio for certain
pathogens, including S. aureus. The principal
pharmacokinetic/pharmacodynamic parameter best associated with clinical and
microbiological cure has not been elucidated in clinical trials with CUBICIN.
Pharmacokinetics
CUBICIN Administered Over A 30-Minute Period
The mean and standard deviation (SD) pharmacokinetic
parameters of daptomycin at steady-state following intravenous (IV)
administration of CUBICIN over a 30-minute period at 4 to 12 mg/kg q24h to healthy
young adults are summarized in Table 7.
Table 7: Mean (SD) Daptomycin Pharmacokinetic
Parameters in Healthy Volunteers at Steady-State
Dose*†(mg/kg) |
Pharmacokinetic Parameters‡ |
AUC0-24 (mcg•h/mL) |
t½h |
Vss (L/kg) |
CLT (mL/h/kg) |
Cmax (mcg/mL) |
4 (N=6) |
494 (75) |
8.1 (1.0) |
0.096 (0.009) |
8.3 (1.3) |
57.8 (3.0) |
6 (N=6) |
632 (78) |
7.9 (1.0) |
0.101 (0.007) |
9.1 (1.5) |
93.9 (6.0) |
8 (N=6) |
858 (213) |
8.3 (2.2) |
0.101 (0.013) |
9.0 (3.0) |
123.3 (16.0) |
10 (N=9) |
1039 (178) |
7.9 (0.6) |
0.098 (0.017) |
8.8 (2.2) |
141.1 (24.0) |
12 (N=9) |
1277 (253) |
7.7 (1.1) |
0.097 (0.018) |
9.0 (2.8) |
183.7 (25.0) |
*CUBICIN was administered by IV infusion over a 30-minute
period.
†Doses of CUBICIN in excess of 6 mg/kg have not been approved.
‡AUC0-24, area under the concentration-time curve from 0 to 24 hours; t½,
elimination half-life; VSS, volume of distribution at steady-state; CLT, total
plasma clearance; Cmax, maximum plasma concentration. |
Daptomycin pharmacokinetics were generally linear and
time-independent at CUBICIN doses of 4 to 12 mg/kg q24h administered by IV
infusion over a 30-minute period for up to 14 days. Steady-state trough concentrations
were achieved by the third daily dose. The mean (SD) steady-state trough
concentrations attained following the administration of 4, 6, 8, 10, and 12
mg/kg q24h were 5.9 (1.6), 6.7 (1.6), 10.3 (5.5), 12.9 (2.9), and 13.7 (5.2)
mcg/mL, respectively.
CUBICIN Administered Over A 2-Minute Period
Following IV administration of CUBICIN over a 2-minute
period to healthy volunteers at doses of 4 mg/kg (N=8) and 6 mg/kg (N=12), the
mean (SD) steady-state systemic exposure (AUC) values were 475 (71) and 701
(82) mcg·h/mL, respectively. Values for maximum plasma concentration (Cmax) at
the end of the 2-minute period could not be determined adequately in this
study. However, using pharmacokinetic parameters from 14 healthy volunteers who
received a single dose of CUBICIN 6 mg/kg IV administered over a 30-minute
period in a separate study, steady-state Cmax values were simulated for CUBICIN
4 and 6 mg/kg IV administered over a 2-minute period. The simulated mean (SD)
steady-state Cmax values were 77.7 (8.1) and 116.6 (12.2) mcg/mL, respectively.
Distribution
Daptomycin is reversibly bound to human plasma proteins,
primarily to serum albumin, in a concentration-independent manner. The overall
mean binding ranges from 90 to 93%.
In clinical studies, mean serum protein binding in
subjects with creatinine clearance (CLCR) ≥ 30 mL/min was comparable to
that observed in healthy subjects with normal renal function. However, there
was a trend toward decreasing serum protein binding among subjects with CLCR < 30
mL/min (88%), including those receiving hemodialysis (86%) and continuous
ambulatory peritoneal dialysis (CAPD) (84%). The protein binding of daptomycin
in subjects with moderate hepatic impairment (Child-Pugh Class B) was similar to
that in healthy adult subjects.
The volume of distribution at steady-state (Vss) of
daptomycin in healthy adult subjects was approximately 0.1 L/kg and was
independent of dose.
Metabolism
In in vitro studies, daptomycin was not metabolized by
human liver microsomes.
In 5 healthy adults after infusion of radiolabeled 14C-daptomycin,
the plasma total radioactivity was similar to the concentration determined by
microbiological assay. Inactive metabolites were detected in urine, as
determined by the difference between total radioactive concentrations and
microbiologically active concentrations. In a separate study, no metabolites
were observed in plasma on Day 1 following the administration of CUBICIN at 6
mg/kg to subjects. Minor amounts of three oxidative metabolites and one
unidentified compound were detected in urine. The site of metabolism has not
been identified.
Excretion
Daptomycin is excreted primarily by the kidneys. In a
mass balance study of 5 healthy subjects using radiolabeled daptomycin, approximately
78% of the administered dose was recovered from urine based on total
radioactivity (approximately 52% of the dose based on microbiologically active concentrations),
and 5.7% of the administered dose was recovered from feces (collected for up to
9 days) based on total radioactivity.
Specific Populations
Renal Impairment
Population-derived pharmacokinetic parameters were
determined for infected patients (complicated skin and skin structure
infections [cSSSI] and S. aureus bacteremia) and noninfected subjects
with various degrees of renal function (Table 8). Total plasma clearance (CLT),
elimination half-life (t1/2), and volume of distribution at steady-state (Vss)
in patients with cSSSI were similar to those in patients with S. aureus bacteremia.
Following administration of CUBICIN 4 mg/kg q24h by IV infusion over a 30- minute
period, the mean CLCR was 9%, 22%, and 46% lower among subjects and patients
with mild (CLCR 50–80 mL/min), moderate (CLCR 30– < 50 mL/min), and severe (CLCR
< 30 mL/min) renal impairment, respectively, than in those with normal renal
function (CLCR > 80 mL/min). The mean steadystate systemic exposure (AUC), t1/2,
and Vss increased with decreasing renal function, although the mean AUC for
patients with CLCR 30–80 mL/min was not markedly different from the mean AUC
for patients with normal renal function. The mean AUC for patients with CLCR
< 30 mL/min and for patients on dialysis (CAPD and hemodialysis dosed
post-dialysis) was approximately 2 and 3 times higher, respectively, than for
patients with normal renal function. The mean Cmax ranged from 60 to 70 mcg/mL in
patients with CLCR ≥ 30 mL/min, while the mean Cmax for patients with CLCR
< 30 mL/min ranged from 41 to 58 mcg/mL. After administration of CUBICIN 6
mg/kg q24h by IV infusion over a 30-minute period, the mean Cmax ranged from 80
to 114 mcg/mL in patients with mild to moderate renal impairment and was
similar to that of patients with normal renal function.
Table 8: Mean (SD) Daptomycin Population
Pharmacokinetic Parameters Following Infusion of CUBICIN 4 mg/kg or 6 mg/kg to
Infected Patients and Noninfected Subjects with Various Degrees of Renal
Function
Renal Function |
Pharmacokinetic Parameters* |
t½† (h) 4 mg/kg |
Vss (L/kg)
4 mg/kg |
CLT
† (mL/h/kg) 4 mg/kg |
AUC 0 -∞† (mcg•h/mL)
4 mg/kg |
AUC (mcg•h/mL)
6 mg/kg |
Cmin,ss‡
(mcg/mL) 6 mg/kg |
Normal |
9.39 (4.74) |
0.13 (0.05) |
10.9 (4.0) |
417 (155) |
545 (296) |
6.9 (3.5) |
(CLCR > 80 mL/min) |
N=165 |
N=165 |
N=165 |
N=165 |
N=62 |
N=61 |
Mild Renal Impairment (CLCR50-80 mL/min) |
10.75 (8.36) N=64 |
0.12 (0.05) N=64 |
9.9 (4.0) N=64 |
466 (177) N=64 |
637 (215) N=29 |
12.4 (5.6) N=29 |
Moderate Renal Impairment (CLCR 30- < 50 mL/min) |
14.70 (10.50) N=24 |
0.15 (0.06) N=24 |
8.5 (3.4) N=24 |
560 (258) N=24 |
868 (349) N=15 |
19.0 (9.0) N=14 |
Severe Renal Impairment (CLCR < 30 mL/min) |
27.83 (14.85) N=8 |
0.20 (0.15) N=8 |
5.9 (3.9) N=8 |
925 (467) N=8 |
1050, 892 N=2 |
24.4, 21.4 N=2 |
Hemodialysis |
30.51 (6.51) N=16 |
0.16 (0.04) N=16 |
3.9 (2.1) N=16 |
1193 (399) N=16 |
NA |
NA |
CAPD |
27.56 (4.53) N=5 |
0.11 (0.02) N=5 |
2.9 (0.4) N=5 |
1409 (238) N=5 |
NA |
NA |
Note: CUBICIN was administered over a 30-minute period.
*CLCR, creatinine clearance estimated using the Cockcroft-Gault equation with
actual body weight; CAPD, continuous ambulatory peritoneal dialysis; AUC0-∞,
area under the concentration-time curve extrapolated to infinity; AUCss, area
under the concentration-time curve calculated over the 24 -hour dosing interval
at steadystate; Cmax, trough concentration at steady-state; NA, not applicable.
†Parameters obtained following a single dose from patients with complicated
skin and skin structure infections and healthy subjects.
‡Parameters obtained at steady-state from patients with S. aureus bacteremia. |
Because renal excretion is the primary route of
elimination, adjustment of CUBICIN dosage interval is necessary in patients
with severe renal impairment (CL < 30 mL/min) [see DOSAGE AND
ADMINISTRATION].
Hepatic Impairment
The pharmacokinetics of daptomycin were evaluated in 10
subjects with moderate hepatic impairment (Child-Pugh Class B) and compared
with those in healthy volunteers (N=9) matched for gender, age, and weight. The
pharmacokinetics of daptomycin were not altered in subjects with moderate
hepatic impairment. No dosage adjustment is warranted when CUBICIN is
administered to patients with mild to moderate hepatic impairment. The
pharmacokinetics of daptomycin in patients with severe hepatic impairment
(Child-Pugh Class C) have not been evaluated.
Gender
No clinically significant gender-related differences in
daptomycin pharmacokinetics have been observed. No dosage adjustment is
warranted based on gender when CUBICIN is administered.
Geriatric
The pharmacokinetics of daptomycin were evaluated in 12
healthy elderly subjects ( ≥ 75 years of age) and 11 healthy young controls
(18 to 30 years of age). Following administration of a single 4 mg/kg dose of
CUBICIN by IV infusion over a 30-minute period, the mean total clearance of
daptomycin was approximately 35% lower and the mean AUC0-∞ was
approximately 58% higher in elderly subjects than in healthy young subjects.
There were no differences in Cmax [see Use in Specific Populations].
Obesity
The pharmacokinetics of daptomycin were evaluated in 6
moderately obese (Body Mass Index [BMI] 25 to 39.9 kg/m²) and 6 extremely obese
(BMI ≥ 40 kg/m²) subjects and controls matched for age, gender, and renal
function. Following administration of CUBICIN by IV infusion over a 30-minute
period as a single 4 mg/kg dose based on total body weight, the total plasma
clearance of daptomycin normalized to total body weight was approximately 15%
lower in moderately obese subjects and 23% lower in extremely obese subjects
than in nonobese controls. The AUC0-∞ of daptomycin was approximately 30%
higher in moderately obese subjects and 31% higher in extremely obese subjects than
in nonobese controls. The differences were most likely due to differences in
the renal clearance of daptomycin. No adjustment of CUBICIN dosage is warranted
in obese patients.
Pediatric
The pharmacokinetics of daptomycin in pediatric
populations ( < 18 years of age) have not been established [see Nonclinical
Toxicology].
Drug-Drug Interactions
In Vitro Studies
In vitro studies with human hepatocytes indicate that
daptomycin does not inhibit or induce the activities of the following human
cytochrome P450 isoforms: 1A2, 2A6, 2C9, 2C19, 2D6, 2E1, and 3A4. It is unlikely
that daptomycin will inhibit or induce the metabolism of drugs metabolized by
the P450 system.
Aztreonam
In a study in which 15 healthy adult subjects received a
single dose of CUBICIN 6 mg/kg IV and a combination dose of CUBICIN 6 mg/kg IV
and aztreonam 1 g IV, administered over a 30-minute period, the Cmax and AUC0-∞
of daptomycin were not significantly altered by aztreonam.
Tobramycin
In a study in which 6 healthy adult males received a single
dose of CUBICIN 2 mg/kg IV, tobramycin 1 mg/kg IV, and both in combination,
administered over a 30-minute period, the mean Cmax and AUC0-∞ of daptomycin
were 12.7% and 8.7% higher, respectively, when CUBICIN was coadministered with tobramycin.
The mean Cmax and AUC0-∞ of tobramycin were 10.7% and 6.6% lower,
respectively, when tobramycin was coadministered with CUBICIN. These
differences were not statistically significant. The interaction between
daptomycin and tobramycin with a clinical dose of CUBICIN is unknown.
Warfarin
In 16 healthy subjects, administration of CUBICIN 6 mg/kg
q24h by IV infusion over a 30-minute period for 5 days, with coadministration
of a single oral dose of warfarin (25 mg) on the 5th day, had no significant
effect on the pharmacokinetics of either drug and did not significantly alter
the INR (International Normalized Ratio).
Simvastatin
In 20 healthy subjects on a stable daily dose of
simvastatin 40 mg, administration of CUBICIN 4 mg/kg q24h by IV infusion over a
30-minute period for 14 days (N=10) had no effect on plasma trough concentrations
of simvastatin and was not associated with a higher incidence of adverse
events, including skeletal myopathy, than in subjects receiving placebo once
daily (N=10) [see WARNINGS AND PRECAUTIONS and DRUG INTERACTIONS].
Probenecid
Concomitant administration of probenecid (500 mg 4 times
daily) and a single dose of CUBICIN 4 mg/kg by IV infusion over a 30-minute
period did not significantly alter the Cmax or AUC0-∞ of daptomycin.
Microbiology
Daptomycin belongs to the cyclic lipopeptide class of
antibacterials. Daptomycin has clinical utility in the treatment of infections
caused by aerobic, Gram-positive bacteria. The in vitro spectrum of activity of
daptomycin encompasses most clinically relevant Gram-positive pathogenic
bacteria.
Daptomycin exhibits rapid, concentration-dependent
bactericidal activity against Gram-positive bacteria in vitro. This has been
demonstrated both by time-kill curves and by MBC/MIC (minimum bactericidal concentration/minimum
inhibitory concentration) ratios using broth dilution methodology. Daptomycin maintained
bactericidal activity in vitro against stationary phase S. aureus in
simulated endocardial vegetations. The clinical significance of this is not
known.
Mechanism Of Action
The mechanism of action of daptomycin is distinct from
that of any other antibacterial. Daptomycin binds to bacterial cell membranes
and causes a rapid depolarization of membrane potential. This loss of membrane
potential causes inhibition of DNA, RNA, and protein synthesis, which results
in bacterial cell death.
Mechanism Of Resistance
The mechanism(s) of daptomycin resistance is not fully
understood. Currently, there are no known transferable elements that confer
resistance to daptomycin.
Complicated Skin And Skin Structure Infection (cSSSI)
Trials
The emergence of daptomycin non-susceptible isolates
occurred in 2 infected patients across the set of Phase 2 and pivotal Phase 3
clinical trials of cSSSI. In one case, a non-susceptible S. aureus was isolated
from a patient in a Phase 2 trial who received CUBICIN at less than the
protocol-specified dose for the initial 5 days of therapy. In the second case,
a non-susceptible Enterococcus faecalis was isolated from a patient with an
infected chronic decubitus ulcer who was enrolled in a salvage trial.
S. Aureus Bacteremia/Endocarditis And Other
Post-Approval Trials
In subsequent clinical trials, non-susceptible isolates
were recovered. S. aureus was isolated from a patient in a
compassionate-use trial and from 7 patients in the S. aureus bacteremia/endocarditis
trial [see Clinical Trials]. An E. faecium was isolated from a
patient in a vancomycin-resistant enterococci trial.
Interactions With Other Antibacterials
In vitro studies have investigated daptomycin
interactions with other antibacterials. Antagonism, as determined by kill curve
studies, has not been observed. In vitro synergistic interactions of daptomycin
with aminoglycosides, β-lactam antibacterials, and rifampin have been
shown against some isolates of staphylococci (including some
methicillin-resistant isolates) and enterococci (including some vancomycin-resistant
isolates).
Activity In Vitro And In Vivo
Daptomycin has been shown to be active against most
isolates of the following Gram-positive bacteria both in vitro and in clinical
infections, as described in Indications and Usage (1).
Gram-Positive Bacteria
Enterococcus faecalis (vancomycin-susceptible
isolates only)
Staphylococcus aureus (including
methicillin-resistant isolates)
Streptococcus agalactiae
Streptococcus dysgalactiae subsp. equisimilis
Streptococcus pyogenes
The following in vitro data are available, but their
clinical significance is unknown. At least 90% of the following
Gram-positive bacteria exhibit an in vitro minimum inhibitory concentration
(MIC) less than or equal to the susceptible breakpoint for daptomycin versus
the bacterial genus (Table 9). However, the efficacy of CUBICIN in treating
clinical infections due to these bacteria has not been established in adequate
and well-controlled clinical trials.
Gram-Positive Bacteria
Corynebacterium jeikeium
Enterococcus faecalis (vancomycin-resistant
isolates)
Enterococcus faecium (including
vancomycin-resistant isolates)
Staphylococcus epidermidis (including
methicillin-resistant isolates)
Staphylococcus haemolyticus
Susceptibility Testing Methods
When available, the clinical microbiology laboratory
should provide the results of in vitro susceptibility tests for antimicrobial
drug products used in resident hospitals 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 antibacterial drug product
for treatment.
Dilution Techniques
Quantitative methods are used to determine antimicrobial
minimum inhibitory concentrations (MICs). These MICs provide estimates of the
susceptibility of bacteria to antimicrobial compounds. The MICs should be
determined using a standardized broth test method with the broth adjusted to a
calcium content of 50 mg/L. The use of the agar dilution method is not
recommended with daptomycin2,3. The MICs should be interpreted
according to the criteria listed in Table 9.
Table 9: Susceptibility Interpretive Criteria for
Daptomycin
Pathogen |
Broth Dilution MIC* (mcg/mL) |
S |
I |
R |
Staphylococcus aureus (methicillin-susceptible and methicillin-resistant) |
≤ 1 |
(†) |
(†) |
Streptococcus pyogenes, Streptococcus agalactiae, and Streptococcus dysgalactiae subsp. equisimilis |
≤ 1 |
(†) |
(†) |
Enterococcus faecalis (vancomycin-susceptible only) |
≤ 4 |
(†) |
(†) |
Note: S, Susceptible; I, Intermediate; R, Resistant.
*The MIC interpretive criteria for S. aureus and E. faecalis are
applicable only to tests performed by broth dilution using Mueller-Hinton broth
adjusted to a calcium content of 50 mg/L; the MIC interpretive criteria for Streptococcus spp. other than S. pneumoniae are applicable only to tests performed by
broth dilution using Mueller- Hinton broth adjusted to a calcium content of 50
mg/L, supplemented with 2 to 5% lysed horse blood, inoculated with a direct
colony suspension and incubated in ambient air at 35° C for 20 to 24 hours.
†The current absence of data on daptomycin-resistant isolates precludes
defining any categories other than “Susceptible.” Isolates yielding
test results suggestive of a “Non-Susceptible” category should be
retested, and if the result is confirmed, the isolate should be submitted to a
reference laboratory for further testing. |
A report of “Susceptible” indicates that the antimicrobial
is likely to inhibit the growth of the pathogen if the antimicrobial compound
reaches the concentration at the infection site necessary to inhibit growth of
the pathogen.
Diffusion Technique
Quantitative methods that require measurement of zone
diameters have not been shown to provide reproducible estimates of the
susceptibility of bacteria to daptomycin. The use of the disk diffusion method
is not recommended with daptomycin3,4.
Quality Control
Standardized susceptibility test procedures require the
use of laboratory controls to monitor and ensure  the accuracy and precision of
supplies and reagents used in the assay, and the techniques of the individuals
performing the test2,3. Standard daptomycin powder should provide
the ranges of MIC values noted in Table 10.
Table 10: Acceptable Quality Control Ranges for
Daptomycin to Be Used in Validation of Susceptibility Test Results
Quality Control Strain |
Broth Dilution MIC Range* (mcg/mL) |
Enterococcus faecalis ATCC 29212 |
1-4 |
Staphylococcus aureus ATCC 29213 |
0.12-1 |
Streptococcus pneumoniae ATCC 49619† |
0.06-0.5 |
*The quality control ranges for S. aureus and E.
faecalis are applicable only to tests performed by broth dilution using
Mueller-Hinton broth adjusted to a calcium content of 50 mg/L; the quality
control range for Streptococcus pneumoniae is applicable only to tests performed
by broth dilution using Mueller-Hinton broth adjusted to a calcium content of 50
mg/L, supplemented with 2 to 5% lysed horse blood, inoculated with a direct
colony suspension and incubated in ambient air at 35° C for 20 to 24 hours. †This strain may be used for validation of susceptibility test results when
testing Streptococcus spp. other than S. pneumoniae. |
Animal Toxicology And/Or Pharmacology
Adult Animals
In animals, daptomycin administration has been associated
with effects on skeletal muscle. However, there were no changes in cardiac or
smooth muscle. Skeletal muscle effects were characterized by microscopic
degenerative/regenerative changes and variable elevations in creatine
phosphokinase (CPK). No fibrosis or rhabdomyolysis was evident in repeat-dose
studies up to the highest doses tested in rats (150 mg/kg/day) and dogs (100
mg/kg/day). The degree of skeletal myopathy showed no increase when treatment
was extended from 1 month to up to 6 months. Severity was dose-dependent. All
muscle effects, including microscopic changes, were fully reversible within 30
days following the cessation of dosing.
In adult animals, effects on peripheral nerve
(characterized by axonal degeneration and frequently accompanied by significant
losses of patellar reflex, gag reflex, and pain perception) were observed at daptomycin
doses higher than those associated with skeletal myopathy. Deficits in the
dogs' patellar reflexes were seen within 2 weeks after the start of treatment
at 40 mg/kg/day (9 times the human Cmax at the 6 mg/kg/day dose), with some
clinical improvement noted within 2 weeks after the cessation of dosing.
However, at 75 mg/kg/day for 1 month, 7 of 8 dogs failed to regain full
patellar reflex responses within a 3-month recovery period. In a separate study
in dogs receiving doses of 75 and 100 mg/kg/day for 2 weeks, minimal residual
histological changes were noted at 6 months after the cessation of dosing.
However, recovery of peripheral nerve function was evident.
Tissue distribution studies in rats showed that
daptomycin is retained in the kidney but appears to penetrate the blood-brain
barrier only minimally following single and multiple doses.
Juvenile Animals
Target organs of daptomycin-related effects in 7-week-old
juvenile dogs were skeletal muscle and nerve, the same target organs as in
adult dogs. In juvenile dogs, nerve effects were noted at lower daptomycin
blood concentrations than in adult dogs following 28 days of dosing. In
contrast to adult dogs, juvenile dogs also showed evidence of effects in nerves
of the spinal cord as well as peripheral nerves after 28 days of dosing. No
nerve effects were noted in juvenile dogs following 14 days of dosing at doses
up to 75 mg/kg/day.
Administration of daptomycin to 7-week-old juvenile dogs
for 28 days at doses of 50 mg/kg/day produced minimal degenerative effects on
the peripheral nerve and spinal cord in several animals, with no corresponding
clinical signs. A dose of 150 mg/kg/day for 28 days produced minimal
degeneration in the peripheral nerve and spinal cord as well as minimal to mild
degeneration of the skeletal muscle in a majority of animals, accompanied by
slight to severe muscle weakness evident in most dogs. Following a 28-day
recovery phase, microscopic examination revealed recovery of the skeletal
muscle and the ulnar nerve effects, but nerve degeneration in the sciatic nerve
and spinal cord was still observed in all 150 mg/kg/day dogs.
Following once-daily administration of daptomycin to
juvenile dogs for 28 days, microscopic effects in nerve tissue were noted at a
Cmax value of 417 mcg/mL, which is approximately 3-fold less than the Cmax
value associated with nerve effects in adult dogs treated once daily with
daptomycin for 28 days (1308 mcg/mL).
Neonatal Animals
Neonatal dogs (4 to 31 days old) were more sensitive to
daptomycin-related adverse nervous system and/or muscular system effects than
either juvenile or adult dogs. In neonatal dogs, adverse nervous system and/or
muscular system effects were associated with a Cmax value approximately 3-fold
less than the Cmax in juvenile dogs, and 9-fold less than the Cmax in adult
dogs following 28 days of dosing. At a dose of 25 mg/kg/day with associated Cmax
and AUCinf values of 147 mcg/mL and 717 mcg·h/mL, respectively (1.6 and
1.0-fold the adult human Cmax and AUC, respectively, at the 6 mg/kg/day dose), mild
clinical signs of twitching and one incidence of muscle rigidity were observed
with no corresponding effect on body weight. These effects were found to be
reversible within 28 days after treatment had stopped.
At higher dose levels of 50 and 75 mg/kg/day with
associated Cmax and AUCinf values of ≥ 321 mcg/mL and ≥ 1470
mcg·h/mL, respectively, marked clinical signs of twitching, muscle rigidity in
the limbs, and impaired use of limbs were observed. Resulting decreases in body
weights and overall body condition at doses ≥ 50 mg/kg/day necessitated
early discontinuation by PND19.
Histopathological assessment did not reveal any
daptomycin-related changes in the peripheral and central nervous system tissue,
as well as in the skeletal muscle or other tissues assessed, at any dose level.
No adverse effects were observed in the dogs that
received daptomycin at 10 mg/kg/day, the NOAEL, with associated Cmax and AUCinf
values of 62 mcg/mL and 247 mcg·h/mL, respectively (or 0.6 and 0.4- fold the
adult human Cmax and AUC, respectively at the 6 mg/kg dose).
Clinical Trials
Complicated Skin And Skin Structure Infections
Adult patients with clinically documented complicated
skin and skin structure infections (cSSSI) (Table 11) were enrolled in two
randomized, multinational, multicenter, investigator-blinded trials comparing CUBICIN
(4 mg/kg IV q24h) with either vancomycin (1 g IV q12h) or an
anti-staphylococcal semisynthetic penicillin (i.e., nafcillin, oxacillin,
cloxacillin, or flucloxacillin; 4 to 12 g IV per day). Patients could switch to
oral therapy after a minimum of 4 days of IV treatment if clinical improvement
was demonstrated. Patients known to have bacteremia at baseline were excluded.
Patients with creatinine clearance (CLCR ) between 30 and 70 mL/min were to receive
a lower dose of CUBICIN as specified in the protocol; however, the majority of
patients in this subpopulation did not have the dose of CUBICIN adjusted.
Table 11: Investigator's Primary Diagnosis in the
cSSSI Trials (Population: Intent-to-Treat)
Primary Diagnosis |
Patients
(CUBICIN / Comparator*) |
Study 9801
N=264 / N=266 |
Study 9901
N=270 / N=292 |
Pooled
N=534 / N=558 |
Wound Infection |
99 (38%) / 116 (44%) |
102 (38%) / 108 (37%) |
201 (38%) / 224 (40%) |
Major Abscess |
55 (21%) / 43 (16%) |
59 (22%) / 65 (22%) |
114 (21%) / 108 (19%) |
Ulcer Infection |
71 (27%) / 75 (28%) |
53 (20%) / 68 (23%) |
124 (23%) / 143 (26%) |
Other Infection† |
39 (15%) / 32 (12%) |
56 (21%) / 51 (18%) |
95 (18%) / 83 (15%) |
*Comparator: vancomycin (1 g IV q12h) or an
anti-staphylococcal semi-synthetic penicillin (i.e., nafcillin, oxacillin, cloxacillin,
or flucloxacillin; 4 to 12 g/day IV in divided doses).
†The majority of cases were subsequently categorized as complicated cellulitis,
major abscesses, or traumatic wound infections. |
One trial was conducted primarily in the United States
and South Africa (study 9801), and the second was conducted at non-US sites
only (study 9901). The two trials were similar in design but differed in patient
characteristics, including history of diabetes and peripheral vascular disease.
There were a total of 534 patients treated with CUBICIN and 558 treated with
comparator in the two trials. The majority (89.7%) of patients received IV
medication exclusively.
The efficacy endpoints in both trials were the clinical
success rates in the intent-to-treat (ITT) population and in the clinically
evaluable (CE) population. In study 9801, clinical success rates in the ITT
population were 62.5% (165/264) in patients treated with CUBICIN and 60.9%
(162/266) in patients treated with comparator drugs. Clinical success rates in
the CE population were 76.0% (158/208) in patients treated with CUBICIN and
76.7% (158/206) in patients treated with comparator drugs. In study 9901,
clinical success rates in the ITT population were 80.4% (217/270) in patients treated
with CUBICIN and 80.5% (235/292) in patients treated with comparator drugs.
Clinical success rates in the CE population were 89.9% (214/238) in patients
treated with CUBICIN and 90.4% (226/250) in patients treated with comparator
drugs.
The success rates by pathogen for microbiologically
evaluable patients are presented in Table 12.
Table 12: Clinical Success Rates by Infecting Pathogen
in the cSSSI Trials (Population: Microbiologically Evaluable)
Pathogen |
Success Rate n/N (%) |
CUBICIN |
Comparator* |
Methicillin-susceptible Staphylococcus aureus (MSSA)† |
170/198 (86%) |
180/207 (87%) |
Methicillin-resistant Staphylococcus aureus (MRSA)† |
21/28 (75%) |
25/36 (69%) |
Streptococcus pyogenes |
79/84 (94%) |
80/88 (91%) |
Streptococcus agalactiae |
23/27 (85%) |
22/29 (76%) |
Streptococcus dysgalactiae subsp. equisimilis |
8/8 (100%) |
9/11 (82%) |
Enterococcus faecalis (vancomycin-susceptible only) |
27/37 (73%) |
40/53 (76%) |
*Comparator: vancomycin (1 g IV q12h) or an
anti-staphylococcal semi-synthetic penicillin (i.e., nafcillin, oxacillin, cloxacillin,
or flucloxacillin; 4 to 12 g/day IV in divided doses).
†As determined by the central laboratory. |
S. Aureus Bacteremia/Endocarditis
The efficacy of CUBICIN in the treatment of patients with
S. aureus bacteremia was demonstrated in a randomized, controlled,
multinational, multicenter, open-label trial. In this trial, adult patients with
at least one positive blood culture for S. aureus obtained within 2
calendar days prior to the first dose of study drug and irrespective of source
were enrolled and randomized to either CUBICIN (6 mg/kg IV q24h) or standard of
care [an anti-staphylococcal semi-synthetic penicillin 2 g IV q4h (nafcillin, oxacillin,
cloxacillin, or flucloxacillin) or vancomycin 1 g IV q12h, each with initial
gentamicin 1 mg/kg IV every 8 hours for first 4 days]. Of the patients in the
comparator group, 93% received initial gentamicin for a median of 4 days,
compared with 1 patient ( < 1%) in the CUBICIN group. Patients with prosthetic
heart valves, intravascular foreign material that was not planned for removal
within 4 days after the first dose of study medication, severe neutropenia,
known osteomyelitis, polymicrobial bloodstream infections, creatinine clearance
< 30 mL/min, and pneumonia were excluded.
Upon entry, patients were classified for likelihood of
endocarditis using the modified Duke criteria (Possible, Definite, or Not
Endocarditis). Echocardiography, including a transesophageal echocardiogram
(TEE), was performed within 5 days following study enrollment. The choice of comparator
agent was based on the oxacillin susceptibility of the S. aureus isolate.
The duration of study treatment was based on the investigator's clinical
diagnosis. Final diagnoses and outcome assessments at Test of Cure (6 weeks
after the last treatment dose) were made by a treatment-blinded
Adjudication Committee, using protocol-specified clinical
definitions and a composite primary efficacy endpoint (clinical and
microbiological success) at the Test of Cure visit. A total of 246 patients
≥ 18 years of age (124 CUBICIN, 122 comparator) with S. aureus bacteremia
were randomized from 48 centers in the US and Europe. In the ITT population,
120 patients received CUBICIN and 115 received comparator (62 received an
anti-staphylococcal semi-synthetic penicillin and 53 received vancomycin).
Thirty-five patients treated with an anti-staphylococcal semi-synthetic penicillin
received vancomycin initially for 1 to 3 days, pending final susceptibility
results for the S. aureus isolates. The median age among the 235 patients in
the ITT population was 53 years (range: 21 to 91 years); 30/120 (25%) in the
CUBICIN group and 37/115 (32%) in the comparator group were ≥ 65 years of
age. Of the 235 ITT patients, there were 141 (60%) males and 156 (66%)
Caucasians across the two treatment groups. In addition, 176 (75%) of the ITT
population had systemic inflammatory response syndrome (SIRS) at baseline and
85 (36%) had surgical procedures within 30 days prior to onset of the S.
aureus bacteremia. Eighty-nine patients (38%) had bacteremia caused by
methicillinresistant S. aureus (MRSA). Entry diagnosis was based on the
modified Duke criteria and comprised 37 (16%) Definite, 144 (61%) Possible, and
54 (23%) Not Endocarditis. Of the 37 patients with an entry diagnosis of
Definite Endocarditis, all (100%) had a final diagnosis of infective
endocarditis, and of the 144 patients with an entry diagnosis of Possible
Endocarditis, 15 (10%) had a final diagnosis of infective endocarditis as
assessed by the Adjudication Committee. Of the 54 patients with an entry diagnosis
of Not Endocarditis, 1 (2%) had a final diagnosis of infective endocarditis as
assessed by the Adjudication Committee.
In the ITT population, there were 182 patients with
bacteremia and 53 patients with infective endocarditis as assessed by the
Adjudication Committee, including 35 with right-sided endocarditis and 18 with
left-sided endocarditis. The 182 patients with bacteremia comprised 121 with
complicated S. aureus bacteremia and 61 with uncomplicated S. aureus bacteremia.
Complicated bacteremia was defined as S. aureus isolated
from blood cultures obtained on at least 2 different calendar days, and/or
metastatic foci of infection (deep tissue involvement), and classification of
the patient as not having endocarditis according to the modified Duke criteria.
Uncomplicated bacteremia was defined as S. aureus isolated from blood
culture(s) obtained on a single calendar day, no metastatic foci of infection,
no infection of prosthetic material, and classification of the patient as not having
endocarditis according to the modified Duke criteria. The definition of
right-sided infective endocarditis (RIE) used in the clinical trial was
Definite or Possible Endocarditis according to the modified Duke criteria and
no echocardiographic evidence of predisposing pathology or active involvement
of either the mitral or aortic valve. Complicated RIE comprised patients who
were not intravenous drug users, had a positive blood culture for MRSA, serum
creatinine ≥ 2.5 mg/dL, or evidence of extrapulmonary sites of infection.
Patients who were intravenous drug users, had a positive blood culture for
methicillin-susceptible S. aureus (MSSA), had serum creatinine < 2.5
mg/dL, and were without evidence of extrapulmonary sites of infection were
considered to have uncomplicated RIE.
The coprimary efficacy endpoints in the trial were the
Adjudication Committee success rates at the Test of Cure visit (6 weeks after
the last treatment dose) in the ITT and Per Protocol (PP) populations. The overall
Adjudication Committee success rates in the ITT population were 44.2% (53/120)
in patients treated with CUBICIN and 41.7% (48/115) in patients treated with
comparator (difference = 2.4% [95% CI -10.2, 15.1]). The success rates in
the PP population were 54.4% (43/79) in patients treated with CUBICIN and 53.3%
(32/60) in patients treated with comparator (difference = 1.1% [95% CI
-15.6, 17.8]).
Adjudication Committee success rates are shown in Table
13.
Table 13: Adjudication Committee Success Rates at Test
of Cure in the S. aureus Bacteremia/Endocarditis Trial (Population: ITT)
Population |
Success Rate
n/N (%) |
Difference: CUBICIN-Comparator (Confidence Interval) |
CUBICIN 6 mg/kg |
Comparator* |
Overall |
53/120 (44%) |
48/115 (42%) |
2.4% (-10.2, 15.1)† |
Baseline Pathogen |
Methicillin-susceptible S. aureus |
33/74 (45%) |
34/70 (49%) |
-4.0% (-22.6, 14.6)‡ |
Methicillin-resistant S. aureus |
20/45 (44%) |
14/44 (32%) |
12.6% (-10.2, 35.5)† |
Entry Diagnosis§ |
Definite or Possible Infective Endocarditis |
41/90 (46%) |
37/91 (41%) |
4.9% (-11.6, 21.4)‡ |
Not Infective Endocarditis |
12/30 (40%) |
11/24 (46%) |
-5.8% (-36.2, 24.5)‡ |
Final Diagnosis |
Uncomplicated Bacteremia |
18/32 (56%) |
16/29 (55%) |
1.1% (-31.7, 33.9); |
Complicated Bacteremia |
26/60 (43%) |
23/61 (38%) |
5.6% (-17.3, 28.6)¶ |
Right-Sided Infective Endocarditis |
8/19 (42%) |
7/16 (44%) |
-1.6% (-44.9, 41.6)¶ |
Uncomplicated Right-Sided Infective Endocarditis |
3/6 (50%) |
1/4 (25%) |
25.0% (-51.6, 100.0)¶ |
Complicated Right-Sided Infective Endocarditis |
5/13 (39%) |
6/12 (50%) |
-11.5% (-62.4, 39.4)¶ |
Left-Sided Infective Endocarditis |
1/9 (11%) |
2/9 (22%) |
-11.1% (-55.9, 33.6)¶ |
*Comparator: vancomycin (1 g IV q12h) or an
anti-staphylococcal semi-synthetic penicillin (i.e., nafcillin, oxacillin, cloxacillin,
or flucloxacillin; 2 g IV q4 h), each with initial low-dose gentamicin.
†95% Confidence Interval
‡97.5% Confidence Interval (adjusted for multiplicity)
§According to the modified Duke criteria5
¶99% Confidence Interval (adjusted for multiplicity) |
Eighteen (18/120) patients in the CUBICIN arm and 19/116
patients in the comparator arm died during the trial. These comprise 3/28
CUBICIN-treated patients and 8/26 comparator-treated patients with endocarditis,
as well as 15/92 CUBICIN-treated patients and 11/90 comparator-treated patients
with bacteremia. Among patients with persisting or relapsing S. aureus infections,
8/19 CUBICIN-treated patients and 7/11 comparator-treated patients died.
Overall, there was no difference in time to clearance of S.
aureus bacteremia between CUBICIN and comparator. The median time to
clearance in patients with MSSA was 4 days and in patients with MRSA was 8
days.
Failure of treatment due to persisting or relapsing S.
aureus infections was assessed by the Adjudication Committee in 19/120
(16%) CUBICIN-treated patients (12 with MRSA and 7 with MSSA) and 11/115 (10%)
comparator-treated patients (9 with MRSA treated with vancomycin and 2 with
MSSA treated with an anti-staphylococcal semi-synthetic penicillin). Among all
failures, isolates from 6 CUBICIN-treated patients and 1 vancomycin-treated
patient developed increasing MICs (reduced susceptibility) by central laboratory
testing during or following therapy. Most patients who failed due to persisting
or relapsing S. aureus infection had deep-seated infection and did not
receive necessary surgical intervention [see WARNINGS AND PRECAUTIONS].
REFERENCES
2. Clinical and Laboratory Standards Institute (CLSI).
Methods for dilution antimicrobial susceptibility tests for bacteria that grow
aerobically; approved standard—ninth edition. CLSI Document M07-A9; Wayne, PA.
2012.
3. Clinical and Laboratory Standards Institute (CLSI).
Performance standards for antimicrobial susceptibility testing; twenty-second
informational supplement. CLSI Document M100-S22; Wayne, PA. 2012.
4. Clinical and Laboratory Standards Institute (CLSI).
Performance standards for antimicrobial disk susceptibility tests; approved
standard—eleventh edition. CLSI Document M02-A11; Wayne, PA. 2012.
5. Li JS, Sexton DJ, Mick N, Nettles R, Fowler VG Jr,
Ryan T, Bashore T, Corey GR. Proposed modifications to the Duke criteria for
the diagnosis of infective endocarditis. Clin Infect Dis 2000;30:633–638.