You are on page 1of 8

PAIN MEDICINE

Volume 10 • Number S1 • 2009

REVIEW ARTICLES

Clinical Pharmacology of Oxymorphone

Howard S. Smith, MD
Anesthesiology and Academic Director of Pain Management, Albany Medical College, Department of Anesthesiology,

Downloaded from https://academic.oup.com/painmedicine/article/10/suppl_1/S3/1915029 by guest on 25 July 2023


Albany, New York, USA

ABSTRACT

Oxymorphone (14-hydroxydihydromorphinone) is primarily a potent m-opioid receptor agonist


with oral immediate-release (IR) and extended-release (ER) formulations approved in the United
States in 2006. The oral oxymorphone formulations are roughly three times more potent than oral
morphine. It is more lipophilic than morphine and, thus, may more easily cross the blood-brain
barrier because it differs from morphine having a ketone-group substituent at the C-6 position.
Oxymorphone IR is indicated for the relief of moderate—to severe pain, while oxymorphone ER is
indicated for persistent pain. Initial doses (opioid-naïve) are 10–20 mg every 4–6 hours (IR) and
5 mg every 12 hours (ER).Oxymorphone was found not to have any clinically significant cyto-
chrome (CYP)3A4, CPY2C9, or CYP2D6 interactions, thus limiting its potential for causing some
of the more common drug–drug interactions via the CYP450 system.The common adverse effects
of oxymorphone are consistent with those commonly seen with other opioid, including nausea/
vomiting, constipation, pruritis, pyrexia, somnolence, and sedation.

Key Words. Opioids; Oxymorphone; Metabolism Pharmacokinetic; Pharmacodynamic Excretion;


Absorption

Introduction Oxymorphone ER is a new sustained-release


tablet formulation of oxymorphone hydrochlo-
O xymorphone (14-hyroxydihydromor-
phinone) is a semisynthetic m-opioid recep-
tor agonist, which entered the United States
ride developed to provide 12 hours of sustained
analgesia [5]. The ER matrix, TIMERx (Penwest
marketplace in 1959, available at that time in only Pharmaceuticals Co., Danbury, CT), alters and
parenteral and rectal formulations. In June 2006, delays drug dissolution and absorption from the
oral oxymorphone immediate-release (IR) and gastrointestinal tract, resulting in a completely dif-
oxymorphone extended-release (ER) tablets were ferent pharmacokinetic profile relative to the IR
approved by the Food and Drug Administration drug formulations. Drug release from the ER
[1,2]. Oxymorphone hydrochloride has a more matrix is controlled by the rate of penetration of
rapid onset of action and several times the analge- water into a hydrophilic matrix (consisting of
sic potency of its parent compound morphine xanthan and locust bean gum) and the subsequent
when administered parenterally [3,4]. expansion of the gel coating. Linearity of pharma-
cokinetic parameters contributes to a predictable
dose. Multiple clinical trials have been performed
evaluating the safety and/or efficacy of oxymor-
Reprint requests to: Howard S. Smith, MD, Anesthesiology phone ER in the management of moderate—to
and Academic Director of Pain Management, Albany severe cancer and noncancer persistent pain
Medical College, Department of Anesthesiology, 47 New
Scotland Avenue, MC-131, Albany, NY 12208, USA. [6–15].
Tel: 518-262-4461; Fax: 518-262-2671; E-mail: smithh@ Oxymorphone is a pyridine-ring unsubstituted
mail.amc.edu. pyridomorphinan (Figure 1) [16]. It differs from

© American Academy of Pain Medicine 1526-2375/09/$15.00/S3 S3–S10 doi:10.1111/j.1526-4637.2009.00594.x


S4 Smith

CH3 and oxycodone to oxymorphone (which do not


take into account incomplete opioid cross-
N CH2 tolerance) of 3:1 and 2:1, respectively [24,26].
These conversion ratios are only estimates, neces-
sitating individualized titration with close moni-
HO • HCl toring of each patient until a dose providing
acceptable efficacy and tolerability is identified.
CH2 The analgesic effects of opioids are mediated
predominantly via m-opioid receptors and may be
modulated by d-opioid receptors (e.g., agonism at

Downloaded from https://academic.oup.com/painmedicine/article/10/suppl_1/S3/1915029 by guest on 25 July 2023


OH O O d-opioid receptors may potentiate m-mediated
Figure 1 Chemical structure of oxymorphone
analgesia and may slow the development of toler-
hydrochloride. ance). The binding affinities of oxymorphone to
these two receptors exceed those of morphine by
2.5-fold and 3.4-fold, respectively [16,27]. Unlike
oxycodone, which has some k-opioid receptor
morphine by having a ketone-group substituent at agonism, oxymorphone has very little binding and
the C-6 position and saturation of the 7,8 double no clinically significant effects at the k-opioid
bond. The ketone-group substituent renders the receptor [28].
molecule more lipid-soluble and makes oxymor- Oxymorphone undergoes extensive hepatic
phone much more structurally related to hydro- metabolism via conjugation with glucuronic acid
morphone. Oxymorphone is available in the freely to produce its primary metabolite, oxymorphone-
water-soluble hydrochloride (HCL) salt form. It 3-glucuronide. Uridine diphosphate glucuronyl
exists as a white or slightly off-white, odorless transferase subtype B27 catalyzes glucuronidation
powder. The HCL salt form is sparingly soluble in at the 3 position [29]. To a lesser extent, it also
alcohol and ether. The molecular weight of the undergoes reduction of the 6 ketone group to
salt form is 337.80. The octanol/water partition form 6-hydroxy (OH)-oxymorphone. Although
coefficient is 0.98 (at 37°C and pH 7.4) [1,2,17]. 6-OH-oxymorphone likely has some analgesic
Oxymorphone is soluble in water, less soluble in
alcohol, and more lipid soluble than morphine or
oxycodone [18,19]. The increased lipophilicity is Table 1 Oral oxymorphone hydrochloride
due to the structural differences between oxymor- pharmacokinetic data
phone and morphine; specifically, oxymorphone is
Absolute oral
a 6-keto-opioid with the ketone group substituted availability 10%
for a hydroxyl group at the C-6 position. Opioids
Tmax Oxymorphone IR—1/2 h
with greater lipid solubility may cross the blood- Oxymorphone ER—2–3 h
brain barrier more readily than less lipophilic Time to Oxymorphone IR—after 3 days
opioids [20]. steady state Oxymorphone ER—after 3 days
Protein binding 10–12%
Oxymorphone is 10% to 12% plasma protein- Absorption Oxymorphone IR—1/2 life-7–10 h
bound [1,2] (See Table 1). Mean oxymorphone Oxymorphone ER—1/2 life-9–12 h
volume of distribution is approximately 3 L/kg Food can ↑ plasma conc. by 50%
Metabolism • 6-hydroxyoxymorphone
[17,21]. Oxymorphone is not extensively protein- AUC 70% of parent compound flowing a
bound compared with other opioids [22,23], which single dose and equivalent of steady state.
may be advantageous in patients who are hypoal- • Oxymorphone-3-glucuronide
The mean plasma AUC of oxymorphone-3-
buminemic and in those receiving polypharmacy glucuronide is 90-fold higher than is the
with other drugs that are highly protein bound. AUC of the parent compound after a single
Clinical trials assessing an advantage in patients dose.
• No clinically significant drug–drug
with low serum albumin have not yet been con- interactions.
ducted. Unlike morphine, oxymorphone does not Excretion <2% excreted unchanged in urine
promote histamine release in human skin mast <1% excreted as 6-hydroxyoxymorphone
Co-ingestion Should be avoided—oxymorphone ER serum
cells [24,25]. of Alcohol levels can ↑ unpredictably (up to 270% in
Oral oxymorphone is more potent (per mg) some patients)
than oral oxycodone or oral morphine with esti- IR = immediate release; ER = extended release; AUC = area under the
mated initial oral conversion ratios for morphine concentration-time curve.
Clinical Pharmacology of Oxymorphone S5

bioactivity and the pharmacologic activity of phone is also metabolized via hydroxylation
oxymorphone-3-glucuronide has not been fully to 6-hydroxy-oxymorphone (OH-6-G), but this
evaluated [1,30], oxymorphone has been consid- reaction does not involve any known cytochrome
ered to have relatively clinically inert metabolites. P450 system enzyme.
No clinically significant cytochrome (CYP)3A4, In pharmacokinetic studies, a high-fat meal
CYP2C9, or CYP2D6 mediated drug–drug inter- increased the Cmax of oxymorphone IR and ER by
actions have been noted with oxymorphone, and, 38% [26] and 50% [24], respectively. The AUC
thus, there is minimal potential for clinically sig- for oxymorphone IR also increased 38% but
nificant drug–drug interactions [31]. Less than 2% remained relatively unchanged for oxymorphone
of the parent compound is recovered in the urine ER [24]. For these reasons, the package insert pru-

Downloaded from https://academic.oup.com/painmedicine/article/10/suppl_1/S3/1915029 by guest on 25 July 2023


in unchanged form [32]. Upon absorption, drug dently states that oxymorphone should be taken
concentrations may be graphed (ordinate) against on an empty stomach—at least 1 hour before or 2
time (abscissa). The peak drug concentration hours after a meal. During the clinical studies,
(Cmax) is reached at Tmax and the trough concen- however, participants were not given direction on
tration is Cmin. The area under the concentration- when to take oxymorphone ER relative to meals,
time curve (AUC) is a measure of drug exposure and it appears that regardless of whether it is
that can be calculated (as the sum of trapezoids). administered preprandially vs postprandially, oxy-
At steady state, the AUC for 6-OH-oxymorphone morphone absorption has no clinically meaningful
approximates oxymorphone. For oxymorphone-3- differences [36]. Oxymorphone IR and ER each
glucuronide, 33% to 38% is excreted in the urine provide predictable, dose-proportional plasma
in patients with normal renal and hepatic function, concentrations across their entire dose ranges.
and the AUC is 90-fold higher than oxymorphone Gender does not appear to influence oxymor-
(Endo Pharmaceuticals Inc., Chadds Ford, PA). In phone pharmacokinetics [1,2]. Patient gender does
an early pharmacokinetic study of oxymorphone not affect oxymorphone ER concentrations after
administered as a 10 mg IR tablet, a 10 mg oral adjusting for body weight (gender effects with oxy-
solution, or a 1 mg intravenous bolus, absolute morphone IR have not been studied).
oral bioavailability was determined to be approxi-
mately 11%, a value that is in the ballpark with Oxymorphone IR
that estimated for morphine and other 3-OH In single- and multiple-dose studies of oxymor-
opioids (Endo Pharmaceuticals Inc.). There was a phone IR [37], absorption is rapid and the time of
large difference in terminal elimination half-life maximum plasma concentration (tmax) was 0.5
( t 12 ) for oral vs intravenous administration of hours following oral administration. Steady-state
oxymorphone, 15.2 ⫾ 21.0 vs 2.0 ⫾ 0.6 hours concentrations (CSS) were achieved in 3 days. The
(mean ⫾ standard deviation [SD]), most likely observed tmax is consistent with the rapid onset of
attributable to enterophepatic recirculation fol- analgesia at 0.5–0.75 hours observed in clinical
lowing oral administration. Animal studies of studies with single or multiple doses of oxymor-
6-OH-oxymorphone have shown analgesic effects phone IR 20–30 mg [38,39]. The elimination half-
similar to oxymorphone (Endo Pharmaceuticals life (t1/2b) for a single dose of oxymorphone IR
Inc.). In this respect, oxymorphone may be similar ranges from approximately 7.2 hours for the 5-mg
to morphine because substitution at the 6 position dose to 9.4 hours for the 20-mg dose. The rapid
appears to produce active metabolites with both absorption and prolonged t1/2b of oxymorphone
opioids. At present, it is not clear if substitution IR compare favorably with those of morphine
at the 3 position also produces parallel effects. and oxycodone IR formulations [40–42]. The
Although the 3-glucuronide metabolite of mor- maximum observed concentration (Cmax) and
phine is devoid of clinically meaningful analgesic AUC after a single oxymorphone IR dose at CSS
activity [33–35], the analgesic activity or inactivity increases dose proportionally with increasing
of oxymorphone-3-glucuronide has not been doses from 5 to -20 mg.
established. In a randomized crossover of oxymorphone IR
in healthy volunteers, the single-dose and steady-
Pharmacokinetics state pharmacokinetic profiles of oxymorphone
The bioavailability of oral oxymorphone is were linear and dose-proportional across a dose
approximately 10% [24,26] following extensive range of 5–20 mg [37]. Oxymorphone IR was not
first-pass hepatic metabolism, primarily via phase associated with any clinically significant effects on
II glucuronidation. To a lesser extent, oxymor- laboratory or other safety variables [37].
S6 Smith

After single-dose administration and at steady oxymorphone IR oral dose. Over the subsequent
state, the increases in mean AUC• and AUCSS, 120 hours, the mean percentage of the dose
respectively, and Cmax for oxymorphone followed a eliminated was as follows: free oxymorphone,
twofold progression between doses. Mean oxy- 1.9%; conjugated oxymorphone, 44.1%; free
morphone AUCSS was 4.63, 10.19, and 21.10 fol- 6 b-OH-oxymorphone, 0.3%; conjugated 6-OH-
lowing administration of oxymorphone IR 5, 10, oxymorphone, 2.6%, free 6 a-OH-oxymorphone,
and 20 mg every 6 hours, respectively [37]. These none; and conjugated 6 a-OH-oxymorphone,
data provide important information for the clini- 0.1%. Thus, 49% of the dose was eliminated in the
cian in that an increase in dose in the clinical urine over 120 hours, with 82% of the ultimate
setting will reliably produce predictable increases amount eliminated in the urine being eliminated

Downloaded from https://academic.oup.com/painmedicine/article/10/suppl_1/S3/1915029 by guest on 25 July 2023


in the plasma levels of oxymorphone. This may during the initial 24 hours. The mean ⫾ standard
be especially important for opioids. Individual error of the mean terminal disposition half-life
responses to opioids are variable, and clinicians of unconjugated oxymorphone, calculated using
frequently must titrate opioids across a large mil- urinary excretion data, was 8.9 ⫾ 0.4 hour
ligram dose range to find a dosage that provides (Sigma–Minus method) and 7.6 ⫾ 2.0 hours
adequate analgesia with acceptable opioid-related (urinary excretion rate plots) [32]. Oxymorphone
adverse events. Formulations that provide pre- IR (which comes in two strengths [5 mg, 10 mg])
dictable increases in drug concentrations with can be initiated in opioid-naïve patients at starting
increasing doses provide less uncertainty when doses of 5–10 mg every 4–6 hours depending on
empirically determining a patient’s optimum dose. the individual patient and situation.
Adams and Ahdieh pointed out that a limitation of
their study is that the plasma concentrations for
6-OH-oxymorphone at the 5 and 10 mg doses Oxymorphone ER
were below the lower limit of quantification in The data obtained from Adams and Ahdeih in a
some samples, which may account for some devia- randomized study of oxymorphone ER in healthy
tion in linearity that was observed with the 6-OH adults show that the pharmacokinetics of the ER
metabolite [37]. In support of this interpretation, oral formulation and its major metabolites are also
plasma concentrations for 6-OH-oxymorphone linear and dose-proportional [43]. Furthermore,
were higher and, therefore, more reliably ascer- all doses were well tolerated, with mild adverse
tained at steady state than during single-dose events typically found with opioids [43].
administration. The CSS of 6-OH-oxymorphone Single-dose and CSS data and pharmacokinetic
exhibited less deviation from a linear dose re- variables (e.g., AUC, AUCSS, Cmax, Cmin, Cavg)
sponse (approximately 10%) than during single- reflecting drug and metabolite concentrations
dose administration (approximately 20%) [37]. showed essentially linear increases of oxymor-
An important pharmacokinetic parameter for phone and its metabolites (6-OH-OXM and
an IR analgesic is a short time to peak plasma OXM-3-G) as the dose of oxymorphone ER was
concentration. Adams and Ahdieh demonstrated incrementally doubled across a dose range of
that the median time to Cmax (tmax) was 0.5 hours 5–40 mg [43]. From single dose to steady state, the
across all doses of oxymorphone IR [37]. This plasma concentrations of all three moieties accu-
value compares favorably with IR tablets of mor- mulate to a clinically relevant degree (oxymor-
phine and oxycodone, for which mean tmax has phone 2.1- to 3.4-fold, 6-hydroxy metabolite
been reported to be 1.2 and 1.5 hours, respectively 4.1- to 26.3-fold, and 3-glucuronide metabolite
[40,41]. Although the tmax of oxymorphone pre- 1.8- to 2.1-fold) [1,2,17,43]. Calculated on the
dicts a more rapid onset of analgesia, the t 12 of basis of degree of accumulation after 40 mg every
oxymorphone IR was measured at 7.25–9.43 12 hours, the mean ⫾ SD “effective” terminal dis-
hours, considerably longer than the 3- to 5.7-hour position half-lives of oxymorphone, the 6-hydroxy
values reported for oxycodone [41,42]. However, metabolite, and the 3-glucuronide metabolite
active-controlled clinical trials will be required to are 13.1 ⫾ 4.5 hours, 29.8 ⫾ 9.6 hours, and
determine whether the shorter tmax and longer t 12 10.6 ⫾ 2.6 hours, respectively [17,43]. The fluc-
of oxymorphone IR translates into more rapid and tuation index values of less than one for all three
sustained analgesia compared with other short- moieties at all oxymorphone dosage levels verify
acting opioids [37]. the low degree of fluctuation of plasma concentra-
Urinary excretion was evaluated in six healthy tions over the 12-hour dosing interval of oxymor-
volunteers after administration of a single 10-mg phone ER at steady state [17,43]. The absence
Clinical Pharmacology of Oxymorphone S7

of statistically significant differences in dose- and does not inhibit or induce CYP enzymes
normalized natural logarithm (1n)-AUC, [31,44], resulting in a low interaction potential
(1n)-AUCSS, or (1n)-Cmax confirms that the phar- with drugs that are CYP substrates and CYP
macokinetics of oxymorphone, 6-OH-OXM, and inhibitors or inducers.
OXM-3-G are dose-proportional after single-dose Unpublished in vitro data suggest that oxymor-
administration and at steady state. The absence of phone is unlikely to react with any members of the
meaningful differences in oxymorphone clearance cytochrome P450 (CYP450) family of isoenzymes
across the four-dose levels is also consistent with a at normal analgesic concentrations while produc-
linear pharmacokinetic profile [43]. ing only low levels of change (a 1.2- to 2-fold
Although opioid dosages typically are titrated to induction) in the activity of 2 CYP450 family

Downloaded from https://academic.oup.com/painmedicine/article/10/suppl_1/S3/1915029 by guest on 25 July 2023


provide meaningful pain relief while minimizing isoforms—CYP2C9 and CYP3A4—at supraphar-
adverse events, linear pharmacokinetics provide macologic concentrations (plasma concentrations
confidence to clinicians that doubling a patient’s 300–1,000 times those expected in vivo) [45].
dose will predictably double the plasma drug con- Adams and colleagues examined the effects of
centrations and the patient’s extent of exposure ER oxymorphone on CYP2C9 or CYP3A4 meta-
[43]. bolic activities in healthy subjects by performing
Absorption is rapid, even with the ER product, two 14-day, randomized, open-label, parallel-
which has a time-to-peak plasma concentration group studies [31]. On days -1, 7, and 14, subjects
ranging from 2 to 4 hours after single- and received either a CYP2C9 probe (tolbutamide
multiple-dose administration. 500 mg) or CYP3A4 probes (midazolam and [14C
The tmax of oxymorphone ER ranges from 2.5 to N-methyl]-erythromycin for the erythromycin
4.0 hours after a single dose [43]. CSS is achieved breath test) [31]. Subjects were randomized to five
following 3 days of 12-hour daily dosing. At CSS, groups: high-dose oxymorphone ER (3 20 mg
the tmax ranges from 1.5–3.5 hours. The 9- to q12h) + naltrexone (50 mg q24h); low-dose oxy-
11-hour elimination t1/2b of oxymorphone ER morphone ER (10–20 mg q12h [10 mg q12h days
is longer that that observed with morphine- 1–3 and 20 mg q12h days 4–14]); rifampin
controlled release or oxycodone [22,23]. As with (2,300 mg q24h), an inducer of CYP2C9 and
the IR formulation, single-dose and steady-state CYP3A4 activities; naltrexone (50 mg q24h); or
Cmax and AUC increase predictability and dose CYP probes alone (controls). Probe metabolism
proportionally across the entire 5- to 40-mg dose was significantly altered by rifampin on days 7 and
range. 14 (P < 0.05), whereas probe metabolism was not
The manufacturer states that oxymorphone ER significantly affected by low-dose oxymorphone
tablets, such as other ER oral opioids, should be ER or by high-dose oxymorphone ER plus
swallowed whole (i.e., not broken, chewed, dis- naltrexone. Adams et al. concluded that oxymor-
solved, or crushed) to avoid rapid release and phone ER exhibits a minimal potential for causing
absorption of a potentially fatal dose [1,2]. Patients metabolic drug–drug interactions mediated by
should also be cautioned against ingesting alcohol, CYP2C9 or CYP3A4 [31].
including any medication that contains alcohol,
concurrently with oxymorphone to avoid in-
Oxymorphone in Special Populations
creased concentrations. Oxymorphone ER (which
comes in seven strengths: 5 mg, 7.5 mg, 10 mg, The safety and efficacy of oxymorphone in
15 mg, 20 mg, 30 mg, 40 mg), can be initiated in patients less than 18 years of age have not been
opioid-naïve patients at a starting dose of 5 mg established. Oxymorphone is listed as Pregnancy
every 12 hours. Category C, as studies have not yet established
possible adverse effects on fetal development.
Oxymorphone and the CYP450 Systems Additionally, it is unknown whether oxymorphone
Oxymorphone is metabolized hepatically, princi- passes into breast milk [1,2].
pally by phase II metabolism involving conjuga- In the elderly (ⱖ65 years old), plasma oxymor-
tion with glucuronic acid, with only 1–2% of phone, 3-hydroxy metabolite, and 3-glucuronide
unchanged drug excreted by the kidney. The metabolite concentrations were a mean of 40%
primary metabolites are oxymorphone-3- higher than those in younger (18–40 years old)
glucuronide and oxymorphone-6-G. Unlike subjects [1,2]. On average, oxymorphone AUC
oxycodone, fentanyl, and most other opioids, oxy- and Cmax were increased 1.4- and 1.5-fold, respec-
morphone is not metabolized by the CYP pathway tively, in the elderly group. Elderly patients should
S8 Smith

receive smaller dosages initially, and dose titration so was not detected in any urine samples analyzed
should proceed slowly with caution. Adverse from patients who administered morphine or
effects seen more often with oxymorphone in the hydromorphone [47]. Thus, a patient who is
elderly include dizziness, somnolence, confusion, taking only oxymorphone ER and/or oxymor-
and nausea [1,2]. phone IR for chronic pain (without any drugs)
There may be significant effects of hepatic should have a urine in which only oxymorphone is
disease on oxymorphone pharmacokinetics. The detected.
bioavailability of oxymorphone was increased by
means of 1.6-fold and 3.7-fold in subjects with
mild (Child-Pugh class A) and moderate (Child- Summary

Downloaded from https://academic.oup.com/painmedicine/article/10/suppl_1/S3/1915029 by guest on 25 July 2023


Pugh class B) hepatic impairment, respectively;
Oxymorphone is a semi-synthetic opioid, which,
compared with healthy controls. In the one subject
like morphine, is predominantly a mopioid recep-
with severe hepatic impairment (Child-Pugh class
tor agonist. Oxymorphone is a World Health
C), the bioavailability was increased by 12.2-fold
Organization Step 3 opioid for moderate to severe
[1,2,17]. No dosage adjustment is needed in mild
pain and has been utilized for cancer and noncan-
hepatic impairment but starting at a low dose and
cer pain. It is more potent and more lipophilic
titrating upwards slowly is prudent. However,
than morphine and, thus, may more easily cross
oxymorphone is relatively contraindicated in
the blood-brain barrier because it differs from
patients with moderately severe to severe hepatic
morphine having a ketone-group substituent at
dysfunction.
the C-6 position. It is currently available in the
Oxymorphone bioavailability was increased by
United States in an oral IR formulation with every
means of 26%, 57%, and 65% in patients with
4- to 6-hour dosing and an ER formulation with
creatinine clearance values of 51–80 mL/min,
every 12-hour dosing. Oxymorphone ER yields a
30–50 mL/min, and below 30 mL/min, respec-
reasonably stable plateau concentration over much
tively, compared with healthy controls [1,2].
of the 12-hour period.
There are virtually no data regarding the dialyz-
Oxymorphone has no reported CYP450 system
ability of oxymorphone. A single case report has
interactions and so few drug–drug interactions for
provided pre- and posthemodialysis plasma con-
patient on multiple medications. The common
centrations obtained at the midpoint of a 4-hour
adverse effects of oxymorphone are consistent
hemodialysis session. Oxymorphone appeared to
with those commonly seen with other opioids,
be removed by hemodialysis, with a predialysis
including nausea/vomiting, constipation, pruritis,
concentration of 4.98 ng/mL and postdialysis
pyrexia, somnolence, and sedation [1,2].
concentration of 2.36 ng/mL. However, further
studies are needed before the dialyzability of
oxymorphone can be completely characterized
Disclosure
[17,46].
There were no grants or other forms of support for this
Oxymorphone in Urine Drug Testing project.
Oxymorphone is not metabolized to other avail-
able opioids or their metabolites; hence, confirma-
tory assays using liquid chromatography/mass References
spectroscopy (the current gold standard for urine 1 Endo Pharamceuticals. Oxymorphone Hydrochlo-
toxicology screening) performed in oxymorphone- ride (Opana) Product Information. Chadds Ford,
treated patients will reveal almost exclusively PA: Author; 2006.
oxymorphone. During the manufacture of oxy- 2 Endo Pharamceuticals. Oxymorphone Hydrochlo-
morphone, a small fraction (1%) of oxycodone is ride, Extended-Release (Opana ER) Product Infor-
present as a manufacturing process contaminant, mation. Chadds Ford, PA: Author; 2006.
3 Sinatra RS, Hyde NH, Harrison DM. Oxymor-
but this would result in merely trace amounts of
phone revisited. Semin Anesth 1998;7:209–15.
oxycodone in the urine. McDonough et al. as well 4 Eddy NB, Lee LEJ. The analgesic equivalence to
as Cone et al. have detected hydromorphone in morphine and relative side action liability of oxy-
the urine of patients who are administering signifi- morphone (4-hydroxy-dihydromorphinon). J Phar-
cant doses of morphine [47,48]. However, Cone macol Exp Ther 1959;125:116–21.
and colleagues concluded that oxymorphone is not 5 Endo Pharmaceuticals. Data on File. Chadds Ford,
a metabolite of morphine or hydromorphone and PA: Author; 2003.
Clinical Pharmacology of Oxymorphone S9

6 Beaver WT, Wallenstein SL, Houde RW, Rogers A. 18 O’Neil MJ. The Merck Index, 13th edition. White-
Comparisons of the analgesic effects of oral and house Station, NJ: Merck & Co.; 2001.
intramuscular oxymorphone and morphine in 19 Poyhia R, Seppala T. Liposolubility and protein
patients with cancer. J Clin Pharmacol 1997;17: binding of oxycodone in vitro. Pharmacol Toxicol
186–98. 1994;74:23–7.
7 Gabrail NY, Dvergsten C, Ahdieh H. Establishing 20 Fand I, Sinatra R, Clouet D. Whole body autora-
the dosage equivalency of oxymorphone extended diographic localization of C[14] labeled morphine
release and oxycodone controlled release in patients and nalorphine. Anat Rec 1978;190:392.
with cancer pain: A randomized controlled study. 21 Drug facts and comparisons. 2006.
Curr Med Res Opin 2004;20:911–8. 22 Alpharma. Kadian® (Morphine Sulfate Sustained

Downloaded from https://academic.oup.com/painmedicine/article/10/suppl_1/S3/1915029 by guest on 25 July 2023


8 Sloan P, Slatkin N, Ahdieh H. Effectiveness and Release Capsules). Full Prescribing Information.
safety of oral extended-release oxymorphone for the Piscataway, NH: Author; 2004.
treatment of cancer pain: A pilot study. Support 23 Purdue Pharma L.P. OxyContin® (Oxycodone
Care Cancer 2005;13:57–65. Epub Nov 9, 2004. Hydrochloride Extended-Release Capsules). Full
9 Gallagher RM, Weiz-Bosna M, Gammaitoni A. Prescribing Information. Stanford, CT: Author;
Assessment of dosing frequency of sustained-release 2005.
opioid preparations in patients with chronic nonma- 24 Alpharma. OPANA® ER (Oxymorphone HCI
lignant pain. Pain Med 2007;8:71–4. Controlled-Release Tablets). Full Prescribing Infor-
10 Hale ME, Dvergsten C, Gimbel J. Efficacy and mation. Piscataway, NJ: Author; 2004.
safety of oxymorphone extended-release in chronic 25 Hermens JM, Ebertz JM, Hanifin JM, Hirshman
low back pain: Results of a randomized, double- CA. Comparison of histamine release in human skin
blind, pkacebo- and active-controlled study. J Pain mast cells induced by morphine, fentanyl, and oxy-
2005;6:21–8. morphone. Anesthesiology 1985;62:124–9.
11 Hale ME, Ahdieh H, Ma T, Rauck R. Efficacy and 26 Endo Pharmaceuticals Inc. OPANA® (Oxymor-
safety of OPANA® ER (oxymorphone extended phone Hydrochloride). Full Prescribing Informa-
release) for relief of moderate to severe chronic tion. Chadds Ford, PA: Author; 2007.
low back pain in opioid-experienced patients: A 27 Cheng CY, Hsin LW, Lin YP, et al. N-cubylmethyl
12-week, randomized, placebo-controlled study. substituted morphinoids as novel narcotic antago-
J Pain 2007;8:175–84. nists. Bioorg Med Chem 1996;4:73–80.
12 Katz N, Rauck R, Ahdieh H, Ma T. Gerritsen van 28 Steinfels GF, Cook L. Antinociceptive profiles of
der Hoop R, Kerwin R, Podolsky G. A 12-week, Mu and Kappa opioid agonists in a rat tooth pulp
randomized, placebo-controlled trial assessing the stimulation procedure. J Pharmacol Exp Ther
safety and efficacy of oxymorphone extended release 1986;236:111–7.
for opioid-naïve patients with chronic low back 29 American Hospital Formulary Service. Bethesda,
pain. Curr Med Res Opin 2007;23:117–28. MD: American Society of Health-System Pharma-
13 Kivitz A, Ma C, Ahdieh H, Galer B. A 2-week, cists; 2006.
multicenter, randomized, double-blind, placebo- 30 Armstrong SC, Cozza KL. Pharmacokinetic drug
controlled dose-ranging, phase III trial comparing interactions of morphine, codeine, and their deriva-
the efficacy of oxymoprohone extended release and tives: Theory and clinical reality, part I. Psychoso-
placebo in adults with pain associated with osteoar- matics 2003;44:167–71.
thritis of the hip or knee. Clin Ther 2006;3:352–64. 31 Adams M, Pieniaszek HJ Jr, Gammanoni AR, et al.
14 Matsumoto AK, Babul N, Ahdieh H. Oxymorphone Oxymorphone extended release does not affect
extended-release tablets relieve moderate to severe CYP2C9 or CYP3A4 metabolic pathways. J Clin
pain and improve physical function in osteoarthritis: Pharmacol 2005;45:337–45.
Results of a randomized, double-blind, placebo- and 32 Cone EJ, Darwin WD, Buchwald WF, Gorodetzky
active-controlled phase III trial. Pain Med 2005;6: CW. Oxymorphone metabolism and urinary excre-
357–66. tion in human, rat, guinea pig, rabbit, and dogs.
15 McIlwain H, Ahdieh H. Safety, tolerability, and Drug Metab Dispos 1983;11:446–50.
effective ness of oxymorphone extended release for 33 Hashiguchi Y, Molina PE, Abumrad NN.
moderate to severe osteoarthritis pain: A one-year Morphine-3-glucuronide: Hyperglycemic and neu-
study. Am J Ther 2005;12:106–12. roendocrine potentiating effects. Brain Res 1995;
16 Ananthan S, Khare NK, Saini SK, et al. Identifica- 694:13–20.
tion of opioid ligands possessing mixed micro 34 Ulens C, Baker L, Ratka A, Waumans D, Tytgat J.
agonist/delta antagonist activity among pyridomor- Morphine-6beta-glucuronide and morphine-3-
phinans derived from naloxone, and hydromor- glucuronide, opioid receptor agonists with different
phone. J Med Chem 2004;47:1400–12. potencies. Biochem Pharmacol 2001;62:1273–82.
17 Guay DR. Use of oxymorphone in the elderly. 35 Christrup LL. Morphine metabolites. Acta Anaes-
Consult Pharm 2007;22:417–30. thesiol Scand 1997;41:116–22.
S10 Smith

36 Sloan PA, Barkin RL. Oxymorphone and oxymor- 43 Adams MP, Ahdieh H. Pharmacokinetics and dose-
phone extended release: A pharmacotherapeutic proportionality of oxymorphone extended release
review. J Opioid Manag 2008;4:131–44. and its metabolites: results of a randomized cross-
37 Adams MP, Ahdieh H. Single- and multiple-dose over study. Pharmacotherapy 2004;24:468–76.
pharmacokinetic and dose-proportionality study of 44 Barkin RI, Fusco AM, Barkin SJ. Opioids used in
oxymorphone immediate-release tablets. Drugs RD primary care for the management of pain: A phar-
2005;6:91–9. amocologic, pharmacotherapeutic, and pharmaco-
38 Aqua K, Gimbel J, Singla N, et al. Efficacy and dynamic overview. In: Boswell MV, Cole BE, eds.
tolerability of oxymorphone immediate release for Wieiner’s Pain Management: A Practical Guide for
acute postoperative pain after abnormal surgery: A Clinicians, 7th edition. New York: Taylor & Francis;

Downloaded from https://academic.oup.com/painmedicine/article/10/suppl_1/S3/1915029 by guest on 25 July 2023


randomized, double-blind, active and placebo- 2006:789–804.
controlled, parallel-group trial. Clin Ther 2007;25: 45 Endo Pharmaceuticals Inc. Data on File. Chadds
1008–12. Ford, PA: Author; 2002.
39 Gimbel J, Ahdieh H. The efficacy and safety of oral 46 Lee MA, Leng MEF, Cooper RM. Measurements
immediate-release oxymorphone for postsurgical of plasma oxycodone, noroxycodone, and oxymor-
pain. Anesth Analg 2004;99:1472–7. phone levels in a patient with bilateral nephrectomy
40 Thirlwell MP, Sloan PA, Maroun JA, et al. Pharma- who is undergoing haemodialysis. Pall Med
cokinetics and clinical efficacy of oral morphine 2005;19:259–60.
solution and controlled-release morphine tablets in 47 Cone EJ, Caplan YH, Moser F, Robert T, Black D.
cancer patients. Cancer 1989;63:2275–83. Evidence that morphine is metabolized to hydro-
41 Leow KP, Smith MT, Watt JA, Williams BE, morphone but not to oxymorphone. J Anal Toxicol
Cramond T. Comparative oxycodone pharmacoki- 2008;32:319–23.
netics in humans after intravenous, oral, and rectal 48 McDonough PC, Levine B, Vorce S, Jufer RA,
administration. Ther Drug Monit 1992;14:479–84. Fowler D. The detection of hydromorphone in
42 Leow KP, Smith MT, Williams B, Cramond T. urine specimens with high morphine concentra-
Single-dose and steady-state pharmacokinetics and tions. J Forensic Sci 2008;53:752–4.
pharmacodynamics of oxycodone in patients with
caner. Clin Pharmacol Ther 1992;52:487–95.

You might also like