throbber
Pharmacokinetics and Dose-Proportionality of
`Oxymorphone Extended Release and Its Metabolites:
`Results of a Randomized Crossover Study
`
`Michael P. Adams, Pharm.D., and Harry Ahdieh, Ph.D.
`
`Study Objective. To evaluate the pharmacokinetics and dose-proportionality
`of four dose strengths (5, 10, 20, and 40 mg) of oxymorphone extended
`release (ER) under both single-dose and steady-state conditions.
`Design. Randomized, three-period, four-sequence, crossover study.
`Setting. Bioavailability clinic.
`Subjects. Twenty-four healthy adult volunteers.
`Intervention. Each subject received three of the four possible doses. The
`three 8-day administration periods were separated by a 7-day washout.
`Plasma was collected for up to 48 hours after a single dose on day 1 and
`during a 12-hour dosage interval at steady state. Naltrexone was
`administered to reduce opioid-related adverse effects.
`Measurements and Main Results. Twenty-three subjects completed at least
`one study period. Dose-proportionality and linearity were confirmed after
`single doses (mean oxymorphone ER area under the concentration versus
`time curve [AUC] 4.54, 8.94, 17.80, and 37.90 ng•hr/ml for 5-, 10-, 20-,
`and 40-mg doses, respectively) and at steady state (mean oxymorphone ER
`AUC 5.60, 9.77, 19.3, and 37.0 ng•hr/ml for 5-, 10-, 20-, and 40-mg doses
`every 12 hrs, respectively). Similar results were found for maximum
`plasma concentration. Metabolite (6-hydroxyoxymorphone and
`oxymorphone-3-glucuronide) plasma levels also increased in a linear
`fashion after single-dose administration and at steady state.
`Conclusion. The pharmacokinetic profile of oxymorphone ER demonstrates
`linearity and dose-proportionality under single-dose and steady-state
`conditions for the parent compound and its metabolites for doses of 5–40 mg.
`Key Words: analgesia, oxymorphone ER, pharmacokinetics, steady state,
`dose-proportionality.
`(Pharmacotherapy 2004;24(4):468–476)
`
`The undertreatment of patients with chronic
`malignant and nonmalignant pain remains an
`important clinical issue.1 Opioid therapy (alone
`or in combination with other analgesic
`approaches) is widely used in patients with
`moderate or severe cancer pain2, 3 and in patients
`with chronic nonmalignant pain when analgesia
`provided by other therapies (e.g., nonsteroidal
`antiinflammatory drugs, transcutaneous electrical
`nerve stimulation) is no longer adequate. 4
`Optimal management of chronic pain requires
`around-the-clock coverage with analgesic agents.
`
`Agents that provide a sustained release of drug
`allow the patient to obtain this baseline analgesia
`with minimal doses/day, thereby improving
`patient compliance and minimizing interference
`with activities of daily living.5 The Agency for
`Healthcare Research and Quality determined that
`parenteral (intramuscular or intravenous)
`administration offers little pain relief advantage
`over enteral administration and that sustained-
`release oral formulations provide an implicit
`advantage by reducing the administration
`frequency.6 A meta-analysis of eight trials that
`
`ENDO - Ex. 2017
`Amneal v. Endo
`IPR2014-00360
`
`

`

`PHARMACOKINETICS OF OXYMORPHONE EXTENDED RELEASE Adams and Ahdieh
`
`469
`
`compared oral sustained-release morphine with
`oral immediate-release morphine solution found
`comparable, but prolonged, pain relief with
`sustained-release delivery.6
`Oxymorphone hydrochloride (14-hydroxy-
`dihydromorphinone) is a semisynthetic µ-opioid
`agonist7 that has a more rapid onset of action and
`several times the analgesic potency of its parent
`compound morphine. 7, 8 Oxymorphone is
`metabolized primarily to 6-hydroxyoxymorphone
`(6-OH-OXM) and oxymorphone-3-glucuronide
`(OXM-3-G) in the liver; less than 2% of parent
`drug is recovered unchanged in urine.9 The
`pharmacologic activity of the glucuronide
`metabolite has not been evaluated; 6-OH-OXM
`has been shown in animal studies to have
`analgesic bioactivity.10
`Oxymorphone extended release (ER) is a new
`sustained-release tablet formulation of oxy-
`morphone hydrochloride developed to provide
`12 hours of sustained analgesia.10 The ER
`matrix, TIMERx (Penwest Pharmaceuticals Co.,
`Danbury, CT), alters and delays drug dissolution
`and absorption from the gastrointestinal tract,
`thereby changing the pharmacokinetic profile
`relative to immediate-release drug formulations.
`Drug release from the ER matrix is controlled by
`the rate of penetration of water into the hydro-
`philic matrix and the subsequent expansion of
`the gel coating. Linearity of pharmacokinetic
`parameters contributes to a predictable dose
`response and aids in the design of clinical trials
`to examine the efficacy and safety of oxy-
`morphone ER in the management of moderate-
`to-severe malignant and nonmalignant pain.
`This study was undertaken to determine single
`and steady-state dose pharmacokinetics and
`dose-proportionality of oxymorphone and its
`metabolites (6-OH-OXM and OXM-3-G) across a
`dose range of 5–40 mg.
`
`From SFBC New Drug Services, Kennett Square,
`Pennsylvania (Dr. Adams); and Clinical Operations, Endo
`Pharmaceuticals Inc., Chadds Ford, Pennsylvania (Dr.
`Ahdieh).
`Supported by Endo Pharmaceuticals Inc., Chadds Ford,
`Pennsylvania, and Penwest Pharmaceuticals Co., Danbury,
`Connecticut.
`Presented in part at the annual meeting of the American
`Pain Society, Chicago, Illinois, March 19–22, 2003.
`Manuscript received July 28, 2003. Accepted pending
`revisions September 17, 2003. Accepted for publication in
`final form January 18, 2004.
`Address reprint requests to Michael P. Adams, Pharm.D.,
`SFBC New Drug Services, Longwood Corporate Center
`South, 415 McFarlan Road, Suite 201, Kennett Square, PA
`19348-2412; e-mail: mpadams@newdrugservices.com.
`
`Methods
`The protocol, informed consent form, and any
`amendments for this study were approved by an
`independent institutional review board. The
`study was conducted and informed consent
`obtained according to the ethical principles
`stated in the Declaration of Helsinki, applicable
`guidelines for good clinical practice, or
`applicable laws or regulations of the United
`States; whichever provided the greatest
`protection of the individual.
`
`Study Design
`This randomized, three-period, four-sequence,
`crossover study assessed the pharmacokinetics of
`four dose levels of oxymorphone ER (5, 10, 20,
`and 40 mg) in 24 healthy volunteers. This was
`an incomplete block design, with each subject
`receiving three of the four dose levels. Based on
`the previously published simulated sample-size
`tables for bioequivalence studies listed,11 the
`sample size (15 patients) was obtained by using
`the design for a four-period study and
`extrapolating to that for a balanced, incomplete,
`three-period, four-treatment design. To fulfill the
`requirements of balancing for treatment and
`sequence, this sample was rounded to 16 and an
`additional 8 subjects were added to improve the
`relative efficiency of the design in case of missing
`values or if patient dropout occurred.
`
`Subjects
`Study subjects were healthy, nonsmokers of
`normal body weight (‡ 50 but ≤ 100 kg and
`within 15% of standard weight) who ranged in
`age from 18–45 years and were men or
`nonpregnant, nonlactating women using
`contraception. Women of childbearing potential
`were required to use a medically acceptable form
`of contraception for the duration of the trial and
`must have had a negative serum pregnancy test at
`screening and a negative urine pregnancy test at
`check-in to the study facility. Exclusion criteria
`included hypersensitivity to oxymorphone or
`naltrexone; positive screen for hepatitis B,
`hepatitis C, or human immunodeficiency virus; a
`history of alcohol abuse, illicit drug use, physical
`dependence on any opioid, or drug abuse or
`addiction; or a positive urine drug screen for
`ethanol, cocaine,
`tetrahydrocannabinol,
`barbiturates, amphetamines, benzodiazepines, or
`opiates. In addition, candidates were excluded
`from entry into the study if they had any disease
`
`

`

`470
`
`PHARMACOTHERAPY Volume 24, Number 4, 2004
`
`or condition (medical or surgical) that might
`compromise the hematologic, cardiovascular,
`pulmonary, renal, gastrointestinal, hepatic, or
`central nervous systems, and/or any condition
`that might interfere with the absorption,
`distribution, metabolism, or excretion of the
`study drug. All candidates underwent liver and
`kidney function tests before enrollment; no
`subject with liver enzymes (aspartate amino-
`transferase or alanine aminotransferase) above
`1.25 times the upper limit of normal, total
`bilirubin above the upper limit of normal, or
`serum creatinine level above the upper limit of
`normal were enrolled in the study.
`
`Procedures
`Subjects were assigned in consecutive order to
`a randomization schedule balanced for sequence
`and period (Figure 1). Based on the randomized
`crossover incomplete block design, subjects
`received three of four possible doses (5, 10, 20,
`and 40 mg) of oxymorphone ER tablets (Endo
`Pharmaceuticals Inc., Chadds Ford, PA). The
`three 8-day administration periods were
`separated by 7-day washout periods. To protect
`against potential opioid-related effects, subjects
`received an opioid antagonist, naltrexone
`hydrochloride (ReVia; DuPont Pharmaceuticals,
`Wilmington, DE) 50 mg once/day, beginning the
`
`evening before the first dose of oxymorphone ER
`on day 1 and continuing until the evening of day
`7. Previous studies have shown that naltrexone
`does not significantly affect the pharmacokinetics
`of oxymorphone and its metabolites.10
`During each administration period, subjects
`received a single tablet of oxymorphone ER 5, 10,
`20, or 40 mg on day 1, the same dose every 12
`hours on days 3–7, and the last multiple dose on
`the morning of day 8. On days 1 and 8, oxy-
`morphone ER was administered after a 10-hour
`overnight fast that continued until 4 hours after
`dose administration. Subjects received no
`xanthine-containing foods or beverages, were to
`abstain from alcoholic beverages beginning 72
`hours before administration of the first dose of
`study drug, and were to refrain from strenuous
`physical activity. Prescription drugs were
`prohibited beginning 2 weeks before the first
`dose of oxymorphone (4 wks for any drug that
`might affect hepatic drug metabolism) until
`discharge from the study. Short-term use of over-
`the-counter agents for a self-limiting indication
`(e.g., aspirin for a headache) required investigator
`authorization and was prohibited within 24
`hours before administration of the first dose of
`test drug. To provide authorization for an over-
`the-counter agent, the investigator was required
`to consider the clinical situation and evaluate the
`
`Screening
`
`Randomization
`
`5-mg single dose on day 1
`10-mg single dose on day 1
`20-mg single dose on day 1
`40-mg single dose on day 1
`
`First crossover:
` Subjects repeated single- and
` multiple-dose phases at a
` randomly assigned second dosage
`
`Days 3–8
`Multiple-dose phase:
` Every 12 hours each subject
` received the same dose level they
` received in the single-dose phase
`
`Second crossover:
` Subjects repeated single- and
` multiple-dose phases at a
` randomly assigned third dosage
`
`Figure 1. Randomization and drug administration.
`
`Washout
`7 days
`
`Washout
`7 days
`
`

`

`PHARMACOKINETICS OF OXYMORPHONE EXTENDED RELEASE Adams and Ahdieh
`
`471
`
`Table 1. Pharmacokinetic Parameters
`Parameter
`Definition
`Area under the concentration versus time curve from time zero to infinity; calculated as AUCt + Ct/lz.
`AUC
`AUC0–12
`AUC from time zero to the end of the 12-hour dosage interval after the first dose.
`AUC from time zero to the end of one dosage interval at steady state (i.e., time 0 to t); calculated using the linear
`AUCss
`trapezoid rule.
`AUC from time zero to the last measured concentration (Ct); calculated using the linear trapezoid rule.
`Average plasma concentration; calculated as AUCss/t.
`Maximum plasma concentration; the highest concentration observed during a dosage interval.
`Minimum plasma concentration; the concentration measured just before dose administration.
`The last measured plasma concentration; the last concentration above the lower limit of quantitation after a dose.
`Oral clearance; calculated as dose/AUC or dose/AUCss.
`Fluctuation index; calculated as (Cmax – Cmin)/Cavg.
`The terminal elimination rate constant; calculated using linear regression on the terminal portion of the log-
`transformed AUC (ln-AUC).
`Accumulation ratio; calculated as AUCss/AUC0–12.
`The duration of one dosage interval in hours.
`The time that Cmax was observed.
`Terminal elimination half-life; calculated as 0.693/lz.
`Effective half-life; calculated as 0.693/–ln(1 – 1/R/t).
`
`AUCt
`Cavg
`Cmax
`Cmin
`Ct
`Cl/F
`FI
`lz
`
`R
`t
`Tmax
`t1/2
`t1/2 effective
`
`potential for masking symptoms of a more severe
`underlying event and whether taking the agent
`would compromise the outcome or validity of the
`study.
`In each administration period, venous blood
`samples were collected in 7-ml tubes (containing
`ethylenediaminetetraacetic acid) immediately
`before the first dose of oxymorphone ER, and at
`0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 10, 12, 16, 24, 36, and
`48 hours after administration. During multiple-
`dose administration, blood samples were
`collected immediately before the morning dose
`on days 6–8 (trough concentrations) and, on day
`8, at 0.5, 1, 1.5, 2, 3, 4, 5, 6, 8, 10, and 12 hours.
`On days 1 and 8, dose administration of study
`drug was to occur after an overnight fast.
`Subjects continued fasting until 4 hours after
`dose administration.
`Physical examination and clinical laboratory
`tests were conducted at screening and repeated
`on conclusion of the study. Routine vital signs,
`including pulse, respiratory rate, and blood
`pressure, were obtained just before administration
`of the first dose of test drug and once each
`morning throughout the administration period.
`Adverse events were recorded and coded per
`Medical Dictionary for Regulatory Activities
`terminology.12
`
`Analytic Methods
`Concentrations of oxymorphone, 6-OH-OXM,
`and OXM-3-G in plasma samples were determined
`with parallel high-performance liquid chromato-
`graphy combined with tandem mass spectrometry
`
`(parallel LC/MS/MS). Two validated methods,
`developed and validated by SFBC Analytical
`Laboratories, Inc. (North Wales, PA), were used.
`Simultaneous determination of oxymorphone
`and 6-OH-OXM used liquid-liquid extraction
`from alkalinized human plasma; the residue was
`reconstituted into a small amount of mobile
`phase. Determination of OXM-3-G used solid
`phase extraction from acidified human plasma;
`the eluent was dried down and reconstituted into
`a small amount of acetonitrile. A SCIEX API
`3000 series LC/MS/MS system (MDI SCIEX,
`Concord, Ontario, Canada) was used for analysis.
`The range of quantitation for oxymorphone and
`6-OH-OXM was 0.1–20 ng/ml plasma and for
`OXM-3-G was 5–250 ng/ml plasma.
`
`Statistical Methods
`The relationship between the pharmacokinetic
`parameters (Table 1) and dose was explored and
`summarized by using appropriate descriptive
`statistics. Continuous variables were compared
`by using analysis of variance, with dose, period,
`sequence, and subject (sequence) included in the
`model, with use of PROC GLM of SAS version
`6.12 (Cary, NC). Proportionality was tested after
`normalization of area under the concentration
`versus time curve (AUC) and maximum plasma
`concentration (Cmax) values to a 20-mg dose.
`Separate analyses were conducted for days 1 and
`8 (steady-state) results. An appropriate non-
`parametric test was applied to the comparison of
`the time to Cmax values and other categorical
`variables. Establishment of steady-state conditions
`
`

`

`472
`
`PHARMACOTHERAPY Volume 24, Number 4, 2004
`
`was based on analysis of trough (predose)
`concentrations from days 6–8 (after 3–5 days of
`dosing every 12 hrs). The degree of accumulation
`was estimated by comparing AUC from time zero
`to the end of one dosage interval at steady state
`(AUCss) on day 8 with results of AUC from time
`zero to the end of the 12-hour dosage interval
`after the first dose (AUC0–12) on day 1.
`Adverse events were coded and summarized by
`treatment group and according to maximum
`intensity and frequency. Clinical laboratory
`values and other safety variables were sum-
`marized by mean values and changes from
`baseline. Changes from baseline were analyzed
`across treatment groups by using an appropriate
`parametric statistic.
`
`Results
`
`Fourteen men and 10 women (18 white, 4
`Hispanic, 2 black; age range 20–45 yrs, mean
`35.5 yrs) were enrolled and randomized.
`Twenty-one subjects completed the trial (two
`subjects withdrew consent, and one subject
`tested positive on the drug screen before period
`3). Twenty-three subjects completed at least one
`treatment period and were included in the
`pharmacokinetic analyses. Average weight and
`height were 72.6 kg and 168 cm, respectively.
`Most subjects (75%) were of medium build. All
`24 subjects were included in the safety analyses.
`
`Single-Dose Pharmacokinetics
`
`Single-dose administration of oxymorphone ER
`resulted in mean plasma concentrations of
`oxymorphone and its metabolites that increased
`proportionally with increasing dose (Figure 2).
`Mean single-dose pharmacokinetic values are
`presented in Table 2. Mean AUC values for
`oxymorphone, 6-OH-OXM, and OXM-3-G
`increased at a rate very close to a 2-fold (linear)
`progression as the dose increased from 5 to 10,
`20, and 40 mg.
`
`Steady-State Pharmacokinetics
`
`Trough concentrations of oxymorphone, 6-OH-
`OXM, and OXM-3-G, measured before
`administration of the morning dose on days 6–8,
`indicated that steady-state conditions were
`achieved after 3 days of dosing every 12 hours.
`As was the case after single-dose administration,
`steady-state mean plasma concentrations of
`oxymorphone and its metabolites rose with
`increasing dose (Figure 3). Mean steady-state
`
`pharmacokinetic values are presented in Table 3.
`The mean accumulation ratio (a measure of the
`difference between the AUCss and the AUC0–12)
`for oxymorphone was greatest at the 5-mg dose
`
`Figure 2. Mean plasma concentrations of oxymorphone
`(A), 6-hydroxyoxymorphone (6-OH-OXM) (B), and
`oxymorphone-3-glucuronide (OXM-3-G) (C) after
`administration of single doses of oxymorphone extended
`release 5, 10, 20, or 40 mg to healthy volunteers.
`
`

`

`PHARMACOKINETICS OF OXYMORPHONE EXTENDED RELEASE Adams and Ahdieh
`
`473
`
`Table 2. Single-Dose Pharmacokinetic Values
`
`5 mg
`
`Oxymorphone Extended Release Dose
`10 mg
`20 mg
`
`2.13 ± 1.28
`4.54 ± 2.04
`1.74 ± 0.94
`0.27 ± 0.13
`3.00 (1.00–12.00)
`26.36 ± 25.21
`0.1419 ± 0.1502
`11.30 ± 10.81
`
`0.88 ± 1.40
`5.80 ± 4.99
`0.59 ± 0.89
`0.14 ± 0.12
`3.50 (1.00–12.00)
`0.1685 ± 0.2624
`16.96 ± 16.67
`
`6.66 ± 3.55
`8.94 ± 4.16
`4.44 ± 1.70
`0.65 ± 0.29
`3.00 (0.50–6.00)
`22.26 ± 9.27
`0.0869 ± 0.0366
`9.83 ± 5.68
`
`3.64 ± 3.68
`6.94 ± 6.79
`1.95 ± 1.30
`0.37 ± 0.19
`2.00 (0.50–6.00)
`0.2086 ± 0.2728
`13.40 ± 11.53
`
`Parametera
`Oxymorphone
`AUCt (ng•hr/ml)
`AUC (ng•hr/ml)
`AUC0–12 (ng•hr/ml)
`Cmax (ng/ml)
`Tmax (hrs)
`Cl/F (L/min)
`lz (hr-1)
`t1/2 (hrs)
`6-OH-OXM
`AUCt (ng•hr/ml)
`AUC (ng•hr/ml)
`AUC0–12 (ng•hr/ml)
`Cmax (ng/ml)
`Tmax (hrs)
`lz (hr-1)
`t1/2 (hrs)
`OXM-3-G
`1179.63 ± 245.24
`492.81 ± 127.15
`AUCt (ng•hr/ml)
`1298.46 ± 249.06
`589.35 ± 132.83
`AUC (ng•hr/ml)
`AUC0–12 (ng•hr/ml)
`767.18 ± 143.11
`347.23 ± 64.09
`111.79 ± 23.35
`52.98 ± 17.79
`Cmax (ng/ml)
`Tmax (hrs)
`3.00 (1.50–10.00)
`3.00 (1.50–5.00)
`lz (hr-1)
`0.0915 ± 0.0352
`0.0938 ± 0.0307
`t1/2 (hrs)
`8.42 ± 2.56
`8.28 ± 3.17
`6-OH-OXM = 6-hydroxyoxymorphone; OXM-3-G = oxymorphone-3-glucuronide.
`Data are mean ± SD, except Tmax, which is median (range).
`aPharmacokinetic parameters are defined in Table 1.
`
`15.08 ± 6.95
`17.80 ± 7.22
`8.50 ± 4.04
`1.21 ± 0.77
`4.00 (1.00–12.00)
`20.93 ± 6.46
`0.0781 ± 0.0275
`9.89 ± 3.21
`
`11.26 ± 7.19
`16.12 ± 9.47
`4.88 ± 2.12
`0.80 ± 0.33
`1.50 (0.50–12.00)
`0.0482 ± 0.0211
`16.82 ± 6.59
`
`2710.69 ± 676.99
`2866.83 ± 717.49
`1633.37 ± 419.94
`250.08 ± 85.68
`3.00 (1.50–6.00)
`0.0849 ± 0.0189
`8.57 ± 2.07
`
`40 mg
`
`35.37 ± 16.19
`37.90 ± 16.20
`18.47 ± 9.98
`2.59 ± 1.65
`2.50 (0.50–12.00)
`19.44 ± 5.06
`0.0849 ± 0.0408
`9.35 ± 2.94
`
`25.52 ± 13.28
`32.79 ± 18.13
`9.92 ± 5.10
`1.44 ± 0.56
`2.00 (1.00–12.00)
`0.0501 ± 0.0412
`18.44 ± 7.88
`
`5465.32 ± 1185.11
`5669.67 ± 1247.34
`3134.30 ± 712.26
`454.50 ± 105.35
`2.00 (1.50–4.00)
`0.0805 ± 0.0172
`9.03 ± 2.17
`
`taken every 12 hours and for 6-OH-OXM was
`greatest at the 5- and 10-mg doses every 12
`hours. However, because of limitations in the
`assay sensitivity, fewer samples provided values
`above the limit of quantification, adding
`difficulty in estimating the AUC from the low
`plasma concentrations of oxymorphone and 6-
`OH-OXM at the lower dose levels. The mean
`accumulation ratio was greater for 6-OH-OXM
`than for the other analytes at all doses, a
`consequence of its slower elimination. At 40 mg,
`the average accumulation values for multiple-
`versus single-dose administration (AUCss vs
`AUC0–12) were approximately 4-fold higher for 6-
`OH-OXM and approximately 2-fold higher for
`oxymorphone and OXM-3-G, consistent with
`mean single-dose elimination half-life values of
`approximately 18 hours for 6-OH-OXM, and
`approximately 9 hours for oxymorphone and
`OXM-3-G. The mean ± SD effective half-life,
`calculated from the degree of accumulation at a
`dosage of 40 mg every 12 hours, was 29.84 ±
`9.63 hours for 6-OH-OXM, 13.05 ± 4.49 hours
`for oxymorphone, and 10.62 ± 2.64 hours for
`OXM-3-G. Consistent with a linear pharmaco-
`
`kinetic system, the changes in accumulation
`ratios mirror the changes measured for effective
`half-life values.
`The mean fluctuation index was less than 1 for
`all three analytes at all dosages, indicating low
`fluctuation in plasma concentration over the
`dosing period after steady state was achieved.
`Linear pharmacokinetics at steady state are
`supported by the nearly linear progression for all
`concentration-dependent variables (AUCss, Cmax,
`minimum concentration [Cmin], and average
`concentration [Cavg]) as the dosage increased
`from 5 to 40 mg every 12 hours and by the
`absence of any meaningful differences in
`oxymorphone clearance across the four dosages.
`
`Dose-Proportionality
`
`Dose-proportionality was compared after
`normalization of pharmacokinetic parameters to
`a 20-mg dose and logarithmic (ln) transfor-
`mation (Table 4). The absence of statistically
`significant differences in ln-AUC, ln-AUCss, or
`ln-Cmax across doses confirms that the pharmaco-
`kinetics of oxymorphone, 6-OH-OXM, and
`
`

`

`474
`
`PHARMACOTHERAPY Volume 24, Number 4, 2004
`
`OXM-3-G are both linear and dose-proportional
`after single-dose administration and at steady
`state.
`
`Figure 3. Mean steady-state plasma concentrations of
`oxymorphone (A), 6-hydroxyoxymorphone (6-OH-OXM)
`(B), and oxymorphone-3-glucuronide (OXM-3-G) (C) after
`administration of single doses of oxymorphone extended
`release 5, 10, 20, or 40 mg to healthy volunteers.
`
`Safety
`A summary of adverse events is shown in Table
`5. All adverse events were mild and resolved
`without sequelae. No adverse event was severe
`or life threatening or caused discontinuation
`from the study. No clinically significant changes
`were observed in clinical laboratory test values or
`other safety variables. At each of the three lower
`dose levels, gastrointestinal disorders, such as
`constipation, that typically are associated with
`opioid use were reported by 2–3 subjects/dose
`level. Although subjects received naltrexone to
`help minimize opioid-related adverse events, we
`cannot exclude the possibility that naltrexone
`may have contributed to the adverse events in
`some subjects.
`
`Discussion
`The data obtained from this randomized study
`of oxymorphone ER in healthy adults show that
`the pharmacokinetics of the new oral formulation
`and its major metabolites are linear and dose-
`proportional. Furthermore, all doses were well
`tolerated, with mild adverse events typically
`found with opioids.
`Single-dose and steady-state plasma concen-
`tration data and pharmacokinetic variables (e.g.,
`AUC, AUCss, Cmax, Cmin, Cavg) reflecting drug
`and metabolite concentrations showed essentially
`linear increases of oxymorphone and its
`metabolites (6-OH-OXM and OXM-3-G) as the
`dose of oxymorphone ER was incrementally
`doubled across a dose range of 5–40 mg. The
`absence of statistically significant differences in
`dose-normalized ln-AUC, ln-AUCss, or ln-Cmax
`confirms that the pharmacokinetics of
`oxymorphone, 6-OH-OXM, and OXM-3-G are
`dose proportional after single-dose adminis-
`tration and at steady state. The absence of mean-
`ingful differences in oxymorphone clearance
`across the four dose levels is also consistent with
`a linear pharmacokinetic profile.
`Compared with parenteral formulations, oral
`formulations offer several advantages, including
`more convenient dosing, potentially fewer
`adverse events, and improved compliance and
`quality of life. Compliance and quality of life are
`improved by maintaining clinically effective drug
`concentrations for a longer duration, enabling a
`prolonged and more convenient dosing interval.
`Although opioid dosages typically are titrated to
`provide meaningful pain relief while minimizing
`adverse events, linear pharmacokinetics provide
`confidence to clinicians that doubling a patient’s
`
`

`

`PHARMACOKINETICS OF OXYMORPHONE EXTENDED RELEASE Adams and Ahdieh
`
`475
`
`Table 3. Steady-State Pharmacokinetic Values
`
`5 mg
`
`Oxymorphone Extended Release Dose Every 12 Hours
`10 mg
`20 mg
`
`5.60 ± 3.87
`0.70 ± 0.55
`1.50 (0.50–12.00)
`18.18 ± 6.31
`0.41 ± 0.59
`0.47 ± 0.32
`0.71 ± 0.27
`3.43 ± 1.65
`
`5.06 ± 3.77
`0.62 ± 0.46
`1.50 (0.50–12.00)
`0.42 ± 0.45
`0.42 ± 0.31
`0.75 ± 0.80
`26.27 ± 27.76
`
`9.77 ± 3.52
`1.24 ± 0.56
`2.00 (0.50–12.00)
`19.33 ± 7.29
`0.64 ± 0.26
`0.81 ± 0.29
`0.69 ± 0.24
`2.29 ± 0.62
`
`8.87 ± 3.83
`1.05 ± 0.44
`1.00 (0.50–4.00)
`0.70 ± 0.31
`0.74 ± 0.32
`0.48 ± 0.22
`7.05 ± 6.09
`
`Parametera
`Oxymorphone
`AUCss (ng•hr/ml)
`Cmax (ng/ml)
`Tmax (hrs)
`Cl/F (L/min)
`Cmin (ng/ml)
`Cavg (ng/ml)
`FI
`R
`6-OH-OXM
`AUCss (ng•hr/ml)
`Cmax (ng/ml)
`Tmax (hrs)
`Cmin (ng/ml)
`Cavg (ng/ml)
`FI
`R
`OXM-3-G
`AUCss (ng•hr/ml)
`1367.78 ± 272.66
`723.39 ± 298.80
`Cmax (ng/ml)
`175.83 ± 46.08
`94.42 ± 45.14
`2.00 (1.00–5.00)
`2.00 (0.50–4.00)
`Tmax (hrs)
`Cmin (ng/ml)
`79.22 ± 18.95
`45.16 ± 41.85
`Cavg (ng/ml)
`113.98 ± 22.72
`60.28 ± 24.90
`0.82 ± 0.28
`0.85 ± 0.28
`FI
`1.81 ± 0.33
`2.09 ± 0.81
`R
`6-OH-OXM = 6-hydroxyoxymorphone; OXM-3-G = oxymorphone-3-glucuronide.
`Data are mean ± SD, except for Tmax, which is median (range).
`aPharmacokinetic parameters are defined in Table 1.
`
`19.28 ± 8.32
`2.54 ± 1.35
`1.25 (0.50–6.00)
`20.23 ± 7.96
`1.22 ± 0.52
`1.61 ± 0.69
`0.78 ± 0.57
`2.39 ± 0.75
`
`19.24 ± 10.09
`2.26 ± 1.12
`1.50 (0.50–12.00)
`1.48 ± 0.76
`1.60 ± 0.84
`0.50 ± 0.17
`4.23 ± 1.85
`
`2889.53 ± 628.73
`383.74 ± 106.01
`2.00 (1.00–3.00)
`169.27 ± 62.90
`240.79 ± 52.39
`0.92 ± 0.38
`1.82 ± 0.40
`
`40 mg
`
`36.98 ± 13.53
`4.47 ± 1.91
`3.50 (0.50–10.00)
`19.76 ± 5.57
`2.33 ± 1.16
`3.08 ± 1.13
`0.68 ± 0.23
`2.13 ± 0.52
`
`39.70 ± 19.87
`4.55 ± 2.24
`1.25 (0.50–10.00)
`3.17 ± 1.73
`3.31 ± 1.66
`0.44 ± 0.16
`4.11 ± 1.15
`
`5721.60 ± 1318.13
`705.04 ± 169.07
`2.00 (1.00–5.00)
`337.30 ± 142.15
`476.80 ± 109.84
`0.79 ± 0.22
`1.84 ± 0.30
`
`Table 4. Analysis of Dose-Proportionality Normalized to 20 mg and Log Transformed
`Geometric Least Squares
`10 mg
`20 mg
`
`5 mg
`
`15.74 ± 1.12
`1.04 ± 1.11
`21.18 ± 1.12
`
`10.98 ± 1.36
`0.63 ± 1.13
`
`16.53 ± 1.11
`1.12 ± 1.11
`20.16 ± 1.11
`
`8.41 ± 1.23
`0.64 ± 1.12
`
`17.72 ± 1.11
`1.10 ± 1.11
`18.81 ± 1.11
`
`12.73 ± 1.23
`0.74 ± 1.12
`
`2418.7 ± 1.05
`209.7 ± 1.07
`
`2510.3 ± 1.05
`207.9 ± 1.07
`
`2695.5 ± 1.05
`233.3 ± 1.07
`
`19.16 ± 1.09
`2.32 ± 1.10
`17.40 ± 1.09
`
`14.96 ± 1.13
`1.92 ± 1.11
`
`18.16 ± 1.10
`2.14 ± 1.10
`18.36 ± 1.10
`
`16.34 ± 1.13
`1.90 ± 1.11
`
`18.12 ± 1.10
`2.32 ± 1.10
`18.39 ± 1.10
`
`17.56 ± 1.13
`2.07 ± 1.11
`
`Parametera
`Single dose
`Oxymorphone
`ln-AUC (ng•hr/ml)
`ln-Cmax (ng/ml)
`ln-Cl/F (L/min)
`6-OH-OXM
`ln-AUC (ng•hr/ml)
`ln-Cmax (ng/ml)
`OXM-3-G
`ln-AUC (ng•hr/ml)
`ln-Cmax (ng/ml)
`Steady stateb
`Oxymorphone
`ln-AUCss (ng•hr/ml)
`ln-Cmax (ng/ml)
`ln-Cl/F (L/min)
`6-OH-OXM
`ln-AUCss (ng•hr/ml)
`ln-Cmax (ng/ml)
`OXM-3-G
`ln-AUCss (ng•hr/ml)
`2780.6 ± 1.06
`2627.4 ± 1.06
`2770.1 ± 1.06
`ln-Cmax (ng/ml)
`366.1 ± 1.07
`330.1 ± 1.07
`355.9 ± 1.07
`ln = natural logarithm; 6-OH-OXM = 6-hydroxyoxymorphone; OXM-3-G = oxymorphone-3-glucuronide.
`Data are mean ± SE.
`aPharmacokinetic parameters are defined in Table 1.
`bEvery 12 hours x 11 doses.
`
`40 mg
`
`p Value
`
`17.54 ± 1.11
`1.19 ± 1.11
`19.01 ± 1.11
`
`13.80 ± 1.23
`0.68 ± 1.12
`
`2662.1 ± 1.05
`219.5 ± 1.07
`
`18.24 ± 1.10
`2.23 ± 1.10
`18.27 ± 1.10
`
`18.89 ± 1.13
`2.18 ± 1.12
`
`2774.4 ± 1.06
`342.9 ± 1.07
`
`0.7766
`0.5574
`0.7766
`
`0.1590
`0.5031
`
`0.0610
`0.1686
`
`0.9229
`0.8109
`0.9229
`
`0.2132
`0.4104
`
`0.8151
`0.6480
`
`

`

`476
`
`PHARMACOTHERAPY Volume 24, Number 4, 2004
`
`Table 5. Summary of Adverse Events
`
`Oxymorphone Extended Release Dose
`10 mg
`20 mg
`17
`17
`5 (29.4)
`4 (23.5)
`
`5 mg
`17
`3 (17.6)
`
`Variable
`Total no. of subjects
`Subjects with at least one event, no. (%)
`Adverse event, no. (%)
`Palpitations
`Gastrointestinal disorders
`Flatulence
`Constipation
`Nausea
`Sore throat
`Musculoskeletal, connective tissue,
`and bone disorders
`Elbow pain
`Hand pain
`Headache
`aPossibly related to drug.
`bUnlikely to be related to drug.
`cOne was possibly related to drug, whereas the other was not.
`
`1 (5.9)a
`2 (11.8)
`0 (0.0)
`2 (11.8)a
`0 (0.0)
`0 (0.0)
`
`0 (0.0)
`0 (0.0)
`0 (0.0)
`0 (0.0)
`
`40 mg
`16
`1 (6.3)
`
`0 (0.0)
`0 (0.0)
`0 (0.0)
`0 (0.0)
`0 (0.0)
`0 (0.0)
`
`0 (0.0)
`0 (0.0)
`0 (0.0)
`1 (6.3)a
`
`1 (5.9)a
`3 (17.6)
`0 (0.0)
`2 (11.8)a
`1 (5.9)a
`0 (0.0)
`
`1 (5.9)
`1 (5.9)b
`1 (5.9)b
`2 (11.8)c
`
`0 (0.0)
`4 (23.5)
`1 (5.9)a
`2 (11.8)a
`0 (0.0)
`1 (5.9)b
`
`0 (0.0)
`0 (0.0)
`0 (0.0)
`0 (0.0)
`
`dose will predictably double the plasma drug
`concentrations and the patient’s extent of
`exposure.
`These results support the predictability of dose
`response from the four different dose-level
`formulations of oxymorphone ER tablets.
`Multiple doses provide the flexibility to start
`therapy at a dose that is appropriate to an
`individual patient’s needs and to titrate linearly to
`reach a stable dosage that provides adequate pain
`relief. Furthermore, the low fluctuation in
`plasma oxymorphone concentration over the
`dosing period once steady state was achieved
`confirms the likely potential for maintenance of
`analgesic efficacy throughout the 12-hour dosing
`interval.
`
`Conclusion
`
`The pharmacokinetic profile of oxymorphone
`ER demonstrates linearity and dose-propor-
`tionality for oxymorphone and its metabolites, 6-
`OH-OXM and OXM-3-G, under single-dose and
`steady-state (12-hr) conditions across the 5–40-
`mg range of tablet formulations. Based on the
`pharmacokinetic characteristics of oxymorphone
`ER observed in this study, the administration of
`oxymorphone ER would be expected to provide a
`predictable response from a given dose, enabling
`clinicians to reliably titrate and taper the dosage.
`However, the study was not designed to evaluate
`efficacy, and further investigations are needed to
`
`determine whether the dose-linearity of
`oxymorphone ER correlates with a linear
`analgesic dose response.
`
`References
`1. Resnik DB, Rehm M, Minard RB. The undertreatment of pain:
`scientific, clinical, cultural, and philosophical factors. Med
`Health Care Philos 2001;4:277–88.
`2. World Health Organization Expert Committee. Cancer pain
`relief and palliative care. Technical report series, no. 804.
`Geneva, Switzerland: World Health Organization, 1990.
`3. American Society of Anesthesiologists. A report by the
`American Society of Anesthesiologists Task Force on Pain
`Management, cancer pain section. Anesthesiology 1996;84:
`1243–57.
`4. American Society of Anesthesiologists. A report by the
`American Society of Anesthesiologists Task Force on Pain
`Management, chronic pain section. Anesthesiology 1997;86:
`995–1004.
`5. Reder RF. Opioid formulations: tailoring to the needs in
`chronic pain. Eur J Pain 2001;5(suppl A):109–11.
`6. Agency for Healthcare Research and Quality. Management of
`cancer pain. Summary, evidence report/technology assessment:
`number 35. AHRG publication no. 01-E033. Rockville, MD:
`Agency for Healthcare Research and Quality, January 2001.
`7. Sinatra RS, Hyde NH, Harrison DM. Oxymorphone revisited.
`Semin Anesth 1988;7:209–15.
`8. Eddy NB, Lee LEJ. The analgesic equivalence to morphine and
`relative side action liability of oxymorphone (4-hydroxy-
`dihydromorphinone). J Pharmacol Exp Ther 1959;125:116–21.
`9. Cone EJ, Darwin WD, Buchwald WF, Gorodetzky CW.
`Oxymorphone metabolism and urinary excretion in human,
`rat, guinea pig, rabbit, and dog. Drug Metab Dispos 1983;11:
`446–50.
`10. Endo Pharmaceuticals. Data on file. Chadds Ford, PA; 2003.
`11. Liu JP. Use of the repeated cross-over designs in assessing
`bioequivalence. Stat Med 1995;14:1067–78; discussion 79–80.
`12. Food and Drug Administration. MedDRA: medical dictionary
`for regulatory activities. Rockville, MD: United States Food and
`Drug Administration, 1997.
`
`

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket