throbber
1428
`
`Comparative Clinical Efficacy and Safety of a Novel
`Controlled-Release Oxycodone Formulation and
`Controlled-Release Hydromorphone in the Treatment
`of Cancer Pain
`
`Neil A. Hagen, M.D., F.R.C.P.1
`Najib Babul, Pharm.D.2
`
`1 Department of Medicine, Tom Baker Cancer
`Centre, and Clinical Neurosciences, The Univer-
`sity of Calgary, Alberta, Canada.
`
`2 Department of Scientific Affairs, Purdue Fred-
`erick, Pickering, Ontario, Canada.
`
`Presented in part at the 8th World Congress
`on Pain, Vancouver, British Columbia, Canada,
`August 18, 1996.
`
`The authors thank Drs. Helen Hays, H. S. Dhali-
`wal, Dwight Moulin, Louise Yelle, Oscar Wong,
`and Deborah Dudgeon for patient recruitment;
`Ms. Iva Ford for study monitoring; and Mr. Zol-
`tan Harsanyi for data analysis.
`
`reprints: Neil A. Hagen, M.D.
`Address for
`F.R.C.P.C., Department of Medicine, Tom Baker
`Cancer Centre, 1331 29 St. NW, Calgary, Al-
`berta, Canada T2N 4N2.
`
`Received September 17, 1996; revision received
`December 2, 1996; accepted December 2, 1996.
`
`q 1997 American Cancer Society
`
`BACKGROUND. The use of oxycodone to treat chronic cancer pain has been ham-
`pered by its short elimination half-life, which necessitates administration every 4
`hours. This study compared the clinical efficacy and safety of a novel oxycodone
`formulation with that of hydromorphone in the treatment of cancer pain.
`METHODS. In a double-blind crossover study, 44 patients with stable cancer pain
`were randomized to controlled-release oxycodone or controlled-release hydromor-
`phone, each given every 12 hours for 7 days. Pain intensity, nausea, and sedation
`were assessed by patients four times daily, and breakthrough analgesia was re-
`corded.
`RESULTS. Thirty-one patients completed the study (18 women, 13 men; mean age,
`56 { 3 years) and received a final controlled-release oxycodone dose of 124 { 22
`mg per day and a final controlled-release hydromorphone dose of 30 { 6 mg per
`day. There were no significant differences between treatments in overall Visual
`Analogue Scale (VAS) pain intensity (VAS 28 { 4 mm vs. 31 { 4 mm), categorical
`pain intensity (1.4 { 0.1 vs. 1.5 { 0.1), daily rescue analgesic consumption (1.4 {
`0.3 vs. 1.6 { 0.3), sedation scores (24 { 4 mm vs. 18 { 3 mm), nausea scores
`(15 { 3 mm vs. 13 { 3 mm), or patient preference. Two patients experienced
`hallucinations on controlled-release hydromorphone, but none did while receiving
`controlled-release oxycodone.
`CONCLUSIONS. Controlled-release oxycodone demonstrated excellent pharmaco-
`dynamic characteristics, analgesic efficacy, and safety as compared with con-
`trolled-release hydromorphone and represents an important new therapeutic op-
`tion for cancer pain management. Cancer 1997;79:1428–37.
`q 1997 American Cancer Society.
`
`KEYWORDS: oxycodone, hydromorphone, controlled release, cancer pain, drug treat-
`ment, drug safety, clinical trial.
`
`O
`
`xycodone is a semisynthetic opioid analgesic with a high oral-to-
`parenteral bioavailability1,2 and a twofold greater oral potency
`than oral morphine.3–6 There are anecdotal data suggesting a lower
`incidence of side effects, such as hallucinations, for oxycodone rela-
`tive to morphine2 and relative to other opioids.7 Use of oxycodone
`for cancer pain has been hampered by its short elimination half-
`life, which necessitates dosing every 4 hours in order to maintain an
`optimal level of analgesia.
`
`/ 7b52$$0984
`
`03-07-97 11:35:20
`
`canas
`
`W: Cancer
`
`1
`
`

`

`In contrast, the use of morphine has gained wide-
`spread acceptance due to its global availability, sig-
`nificant pharmacokinetic and pharmacodynamic data,
`and the development of a controlled-release formula-
`tion.8–13 Despite its extensive use, the occurrence of
`adverse effects necessitates discontinuation in some
`patients. Clinicians treating cancer pain with opioids
`report significant variability among patients in efficacy
`and side effects with available opioid analgesics. Pa-
`tients who have poor analgesic efficacy or safety out-
`comes with one opioid will frequently tolerate another
`opioid well.11,13–20 The clinical observation of a differ-
`ential pharmacologic response has led to the growing
`acceptance and clinical practice of opioid rota-
`tion.11,13–20 The recent availability of controlled-release
`formulations of other opioids, such as hydromor-
`phone, provides clinicians with therapeutic options for
`cancer pain management.9,10,21,22 Recently, a con-
`trolled-release oxycodone formulation has been devel-
`oped that provides a biphasic absorption pattern,
`characterized by an initial rapid onset followed by a
`prolonged phase.5,23,24 We compared the clinical effi-
`cacy and safety of this formulation of oxycodone with
`a previously evaluated controlled-release formulation
`of hydromorphone9,10,25 available in Canada and other
`countries.
`
`MATERIALS AND METHODS
`Subjects
`Forty-four patients with chronic cancer pain and
`stable analgesic requirements were selected for par-
`ticipation. Criteria for exclusion included known hy-
`persensitivity to opioid analgesics, intolerance of ox-
`ycodone or hydromorphone, presence of a medical
`or surgical condition likely to interfere with drug
`absorption in the gastrointestinal tract, concurrent
`use of other opioid analgesics during the study pe-
`riod, and presence of intractable nausea or vomiting.
`Patients who had undergone or were expected to
`undergo therapeutic procedures likely to influence
`their pain during the study period were also ex-
`cluded. The study protocol and informed consent
`form received scientific and ethical approval, and
`patients gave written informed consent before par-
`ticipating in the study.
`
`Medications
`Controlled-release oxycodone (Oxycontin; Purdue
`Frederick, Pickering, Ontario, Canada) and controlled-
`release hydromorphone (Hydromorph Contin; Purdue
`Frederick, Pickering, Ontario, Canada) were each ad-
`ministered every 12 hours for 7 days. Further blind-
`label dose changes were permitted during the study;
`
`Oxycodone for Cancer Pain/Hagen and Babul
`
`1429
`
`and in the event of a dose change, the rescue analgesic
`dose was modified correspondingly. Blinding was
`maintained by the double-dummy technique, which
`involved matching placebos. In the active treatment
`phases, patients received either active controlled-re-
`lease oxycodone and placebos matching controlled-
`release hydromorphone or active controlled-release
`hydromorphone and placebos matching controlled-
`release oxycodone.
`Incident and nonincident breakthrough pain was
`treated with immediate-release oxycodone and hydro-
`morphone matching the active opioid analgesic at a
`dosage of approximately 10% of the daily scheduled
`opioid dose. No opioids other than the test and rescue
`analgesic medications were permitted during the
`course of the study. Nonopioid analgesics, such as
`nonsteroidal anti-inflammatory drugs and acetamino-
`phen, and adjuvant drugs, such as antidepressants,
`anticonvulsants,
`corticosteroids, bisphosphonates,
`and psychostimulants, that had been part of the pa-
`tient’s therapy were continued at the same dose level
`throughout the study period.
`
`Study Design
`The study was a double-blind, randomized, two-way
`crossover evaluation of the clinical efficacy and
`safety of controlled-release oxycodone and con-
`trolled-release hydromorphone. Pain was character-
`ized at the start of the study using the Edmonton
`staging system according to pathophysiologic mech-
`anisms (visceral, bone/soft
`tissue, neuropathic,
`mixed, or unknown) and pain characteristics (inci-
`dent or nonincident pain).26 Patients with 3 days of
`stable analgesic requirements on a prestudy opioid
`were randomized to controlled-release oxycodone
`or controlled-release hydromorphone. Stable anal-
`gesia was defined as 2 or fewer rescue doses of opioid
`analgesic per 24-hour period, calculated over 3 or
`more days. Once controlled-release oxycodone and
`controlled-release hydromorphone treatment were
`initiated, further dose adjustments were permitted
`throughout the study period, and breakthrough pain
`was treated with rescue doses of the matching im-
`mediate-release opioid. At the end of Phase I, pa-
`tients were crossed over to the alternative treatment
`in Phase II without an intervening washout period.
`Pain intensity was assessed by the patient 4
`times a day (at 8 a.m., 12 noon, 4 p.m., and 8 p.m.)
`on a 100 mm Visual Analogue Scale (VAS) (anchors:
`no pain to excruciating pain) and on a 5-point cate-
`gorical scale (0 (cid:129) none, 1 (cid:129) mild, 2 (cid:129) moderate, 3
`(cid:129) severe, 4 (cid:129) excruciating). Nausea and sedation
`were also assessed on a 100 mm VAS at the same
`
`/ 7b52$$0984
`
`03-07-97 11:35:20
`
`canas
`
`W: Cancer
`
`2
`
`

`

`Characteristic
`
`Men/Women
`Mean age (yrs), (mean { SE)
`Primary tumor
`Breast
`Colorectal
`Lung
`Urologic/prostate
`CNS
`Unknown primary site
`Other
`Total
`
`CNS: central nervous system; SE: standard error.
`
`No. of patients
`
`13/18
`56 { 3
`
`7
`5
`1
`5
`4
`2
`7
`31
`
`1430
`
`CANCER April 1, 1997 / Volume 79 / Number 7
`
`TABLE 1
`Characteristics of Study Patients (n (cid:129) 31)
`
`times (nausea anchors: no nausea to severe nausea;
`sedation anchors: no sedation to extreme sedation).
`Spontaneously reported, investigator-observed, and
`elicited adverse events were recorded at the end of
`each phase, and patient and investigator treatment
`preferences were recorded on completion of both
`treatments. At the end of the double-blind study,
`patients who requested continued treatment with
`controlled-release oxycodone received the drug in
`an open-label, longitudinal evaluation.
`The primary measures of efficacy were the 100
`mm pain intensity VAS and the 5-point categorical
`intensity assessment. Previous studies of cancer
`pain have suggested that standard deviations of 17
`mm (pain VAS)27 and 0.6 units (categorical)28 could
`be expected, and we adopted these values for sample
`size estimation. With each patient serving as his or
`her own control, and assuming low correlation be-
`tween responses in a single subject in the 2 treat-
`was determined, and the average number of doses
`ment periods, 24 completed patients (12 patients per
`per time segment was computed. Multivariate re-
`sequence) were calculated to provide 80% power to
`peated measure analysis was used to test for effects
`detect a difference of 15 mm pain intensity VAS and
`of drug, time segment, and their interaction. Data
`0.6 units categorical pain intensity at a statistical
`on rescue consumption from 2 12-hour periods be-
`significance of 0.05.
`ginning at 8 a.m. and 8 p.m. were combined to ob-
`Analysis of treatment sequence revealed no sig-
`tain an overall estimate of rescue use during succes-
`nificant carryover effects. Therefore, differences in
`sive 4-hour intervals during a 12-hour period corre-
`treatment effects for a two-period crossover design
`sponding to the dosing interval of controlled-release
`were evaluated. The pain intensity (VAS and categor-
`oxycodone and controlled-release hydromorphone.
`ical), nausea (VAS), and sedation (VAS) scores were Multivariate repeated measures analysis was used to
`summarized across days and times as well as overall.
`test for the effects of drug, time segment, and their
`interaction.
`The scores for each patient were averaged as follows:
`(1) over 4 timepoints for each of 7 days, (2) over 7
`In addition to analysis of nausea and sedation
`scores, adverse events were analyzed in terms of the
`days for each of 4 timepoints, and (3) over all days
`and times. Three-way analysis of variance was used
`number of patients reporting each event. Fisher’s
`exact test was used to determine the significance of
`to test for the effect of drug and treatment sequence
`and for phase, using data from the overall mean
`differences in the frequency of side effects. Differ-
`ences in patients’ and investigators’ assessment of
`scores. Multivariate repeated measures analysis was
`used to test for effect of drug, day, time of day, and
`treatment effectiveness were compared using the chi
`square test. Treatment preferences were compared
`their interactions. Two separate repeated measures
`analyses were performed: (1) test for drug, day, and
`using normal approximation to the binomial.
`drug 1 day interaction and (2) test for drug, time of
`Statistical significance was defined as P (cid:155) 0.05
`day, and drug 1 time-of-day interaction.
`for a two-tailed hypothesis. All means are presented
`with the corresponding standard error.
`Rescue analgesic use was determined for each
`patient as the total number of rescue doses per day
`of study. Three-way analysis of variance was used to
`test for the effect of drug, treatment sequence, and
`phase, using the data from the overall mean scores.
`Rescue use by time of day was computed by catego-
`rizing time of rescue into 1 of 6 4-hour segments.
`Because the first dose of each phase began at 8 p.m.,
`the interval from 8 p.m. to 12 midnight was desig-
`nated the first 4-hour segment. The number of doses
`for each patient during each of these time segments
`
`RESULTS
`Forty-four patients were enrolled and 31 patients
`completed the study (18 women and 13 men; mean
`age, 56 { 3 years). Reasons for premature withdrawal
`from the study included adverse events (8 patients),
`inadequate pain control (3 patients), intercurrent ill-
`ness (1 patient), and voluntary withdrawal (1 pa-
`tient). Failure to complete both phases of the study
`did not appear to be related to toxicity of one of the
`
`/ 7b52$$0984
`
`03-07-97 11:35:20
`
`canas
`
`W: Cancer
`
`3
`
`

`

`Oxycodone for Cancer Pain/Hagen and Babul
`
`1431
`
`TABLE 2
`Comparison of Clinical Efficacy and Safety Variables by Day of Treatment after Drug Administrationa
`
`CR oxycodone
`
`CR hydromorphone
`
`Variable
`
`Day 1
`
`Day 2
`
`Day 3
`
`Day 4
`
`Day 5
`
`Day 6
`
`Day 7
`
`Overall
`
`Day 1
`
`Day 2
`
`Day 3
`
`Day 4
`
`Day 5
`
`Day 6
`
`Day 7
`
`Overall
`
`Pain intensity VAS
`(0–100 mm)
`Pain intensity ordinal
`(0–5)
`Rescue analgesic use
`(doses/day)
`Sedation VAS (0–100 mm)
`Nausea VAS (0–100 mm)
`
`28.4
`
`1.4
`
`1.3
`23.4
`16.1
`
`27.5
`
`1.3
`
`1.2
`23.5
`15.4
`
`29.7
`
`1.5
`
`1.5
`25.4
`14.3
`
`29.3
`
`1.4
`
`1.6
`24.4
`17.1
`
`28.6
`
`1.4
`
`1.5
`24.1
`15.5
`
`27.4
`
`1.3
`
`1.1
`22.4
`15.7
`
`25.2
`
`1.3
`
`1.3
`22.4
`14.1
`
`28.0
`
`1.4
`
`1.4
`23.6
`15.5
`
`29.7
`
`1.5
`
`2.0
`18.0
`11.8
`
`29.5
`
`1.6
`
`1.5
`18.1
`13.8
`
`32.1
`
`1.5
`
`1.5
`18.0
`12.8
`
`30.6
`
`1.5
`
`1.6
`18.3
`11.8
`
`33.3
`
`1.6
`
`1.5
`16.6
`15.5
`
`31.3
`
`1.5
`
`1.4
`20.6
`14.4
`
`27.7
`
`1.3
`
`1.5
`17.6
`12.2
`
`30.6
`
`1.5
`
`1.6
`18.2
`13.1
`
`CR: controlled-release; VAS: Visual Analogue Scale.
`a Data represent mean daily scores over 4 assessment periods (8 a.m., 12 noon, 4 p.m., and 8 p.m.); overall score represents mean over all days and times.
`
`TABLE 3
`Comparison of Clinical Efficacy and Safety Variables by Time of Day after Drug Administrationa
`
`CR oxycodone
`
`CR hydromorphone
`
`8:00
`a.m.
`
`12:00
`a.m.
`
`4:00
`p.m.
`
`8:00
`p.m.
`
`Overallb
`
`8:00
`a.m.
`
`12:00
`a.m.
`
`4:00
`p.m.
`
`8:00
`p.m.
`
`Overallb
`
`30.7
`
`23.8
`
`27.3
`
`30.3
`
`1.5
`
`0.2
`
`22.9
`
`16.0
`
`1.2
`
`0.4
`
`23.8
`
`15.2
`
`1.4
`
`0.3
`
`24.1
`
`14.7
`
`1.5
`
`0.1
`
`23.9
`
`15.9
`
`28.0
`
`1.4
`
`23.6
`
`15.5
`
`32.9
`
`27.4
`
`29.6
`
`32.6
`
`1.6
`
`0.3
`
`19.8
`
`14.5
`
`1.4
`
`0.4
`
`17.9
`
`13.0
`
`1.5
`
`0.3
`
`16.8
`
`12.3
`
`1.6
`
`0.2
`
`18.2
`
`13.0
`
`30.6
`
`1.5
`
`18.2
`
`13.2
`
`Variable
`
`Pain intensity VAS
`(0–100 mm)
`Pain intensity ordinal
`(0–5)
`Rescue analgesic use
`(dose/4 hrs)
`Sedation VAS
`(0–100 mm)
`Nausea VAS
`(0–100 mm)
`
`CR: controlled-released; VAS: Visual Analogue Scale.
`a Data represent mean score by time of day over 7 days.
`b Overall score represents mean over all days and times.
`
`study drugs over another. The analyses of all efficacy
`outcome variables, including VAS and categorical
`pain intensity, sedation VAS, and nausea VAS were
`restricted to patients completing both phases of the
`study. Spontaneously reported safety variables were
`analyzed for all patients enrolled. The mean daily
`initial controlled-release oxycodone and controlled-
`release hydromorphone doses were 120 { 22 mg and
`24 { 4 mg, respectively. The mean final controlled-
`release oxycodone and controlled-release hydro-
`morphone doses were 124 { 22 and 30 { 6 mg, re-
`spectively. There were 0.65 dose changes during the
`controlled-release oxycodone phase and 0.81 dose
`changes during the controlled-release hydromor-
`phone phase. Sixty-one percent of patients had bone
`
`pain, 29% had soft tissue pain, 23% had visceral pain,
`and 45% had neuropathic pain. Sixteen percent de-
`scribed pain as lancinating. Sixty-one percent of pa-
`tients had steady pain and 52% experienced incident
`pain with or without steady pain. Table 1 provides
`characteristics of evaluable patients.
`The smallest treatment differences that could be
`detected for overall pain intensity using the VAS and
`categorical scales were estimated assuming a type 1
`error rate (alpha) of 0.05 and type 2 error rate (beta)
`of 0.2. Using the sample size of 31, intrasubject cor-
`relations for pain intensity, and the variance esti-
`mates from the data, differences of 4.4 mm on the
`100 mm VAS and 0.2 points on the 5-point categori-
`cal scale were detectable with 80% power.
`
`/ 7b52$$0984
`
`03-07-97 11:35:20
`
`canas
`
`W: Cancer
`
`4
`
`

`

`1432
`
`CANCER April 1, 1997 / Volume 79 / Number 7
`
`FIGURE 1. Mean pain intensity Visual Analogue Scale (VAS) by time of
`day, after administration of controlled-release (CR) oxycodone and CR
`hydromorphone; h (cid:129) hours.
`
`FIGURE 3. Mean rescue analgesic use at successive 4-hour intervals,
`after administration of controlled-release (CR) oxycodone and CR hydro-
`morphone; h (cid:129) hours (time of day).
`
`treatments in pain intensity on the categorical scale
`when tested by day of treatment (P (cid:129) 0.1009) and
`by time of day (P (cid:129) 0.0960). The overall mean cate-
`gorical pain intensity score across all days was 1.4
`{ 0.1 for controlled-release oxycodone and 1.5 { 0.1
`for controlled-release hydromorphone (P (cid:129) 0.0960).
`For both treatment groups, there were significant
`changes in pain intensity VAS (P (cid:129) 0.0018) and cate-
`gorical (P (cid:129) 0.0001) over the course of a day, with
`less pain during the night.
`The frequency of rescue analgesic use is shown in
`6 4-hour time segments in Figure 3 and by day of
`treatment and time of day Tables 2 and 3, respectively.
`There were no significant differences in daily rescue
`analgesic use between controlled-release oxycodone
`and controlled-release hydromorphone (1.4 { 0.3 vs.
`1.6 { 0.3, P (cid:129) 0.1906). Repeated measures analysis
`of variance by time of day indicated no significant
`differences between the treatments in the pattern of
`rescue analgesic use over 6 4-hour time segments (P
`(cid:129) 0.2258), although for both formulations there was a
`significant difference (P (cid:129) 0.0066) in the pattern of
`rescue analgesic use over each day, with fewer doses
`taken during the night. The percentage of rescue anal-
`gesic use in the first, second, and third 4-hour period
`representing the combined 12-hour dosing frequency
`of controlled-release oxycodone (8 a.m. to 8 p.m. and
`8 p.m. to 8 a.m.) showed no significant differences
`between treatments (P (cid:129) 0.3929) and was 23.8%,
`42.8%, and 33.3% for controlled-release oxycodone
`and 33.3%, 37.5%, and 29.2% for controlled-release
`hydromorphone.
`At the completion of the study, patients and inves-
`tigators were asked to identify their treatment prefer-
`ence while blinded to the treatment assignment.
`
`FIGURE 2. Mean categorical pain intensity by day of treatment, after
`administration of controlled-release (CR) oxycodone and CR hydromor-
`phone.
`
`The mean pain intensity VAS scores for con-
`trolled-release oxycodone and controlled-release
`hydromorphone are given in Table 2 by day of treat-
`ment and in Table 3 and Figure 1 by time of day.
`There were no significant differences between treat-
`ments in pain intensity VAS when tested by day of
`treatment (P (cid:129) 0.1091) and by time of day (P (cid:129)
`0.1091). The overall mean pain intensity VAS across
`all days was 28 { 4 mm for controlled-release oxyco-
`done and 31 { 4 mm for controlled-release hydro-
`morphone (P (cid:129) 0.1119). The mean pain intensity
`scores on the categorical scale after administration
`of controlled-release oxycodone and controlled-re-
`lease hydromorphone are given in Table 2 and Fig-
`ure 2 by day of treatment and in Table 3 by time of
`day. There were no significant differences between
`
`/ 7b52$$0984
`
`03-07-97 11:35:20
`
`canas
`
`W: Cancer
`
`5
`
`

`

`Oxycodone for Cancer Pain/Hagen and Babul
`
`1433
`
`FIGURE 4. Mean sedation Visual Analogue Scale (VAS) score by time
`of day, after administration of controlled-release (CR) oxycodone and CR
`hydromorphone, is given; h (cid:129) hours.
`
`FIGURE 5. Mean nausea Visual Analogue Scale (VAS) score by time of
`day, after administration of controlled-release (CR) oxycodone and CR
`hydromorphone, is given; h (cid:129) hours.
`
`Among patients, 25.8% indicated no preference, 35.5%
`preferred controlled-release oxycodone, and 38.7%
`preferred controlled-release hydromorphone. Among
`investigators, 50% indicated no preference, 26.7% pre-
`ferred controlled-release oxycodone, and 23.3% pre-
`ferred controlled-release hydromorphone. Among pa-
`tients and investigators, the proportions expressing a
`preference for controlled-release oxycodone and hy-
`dromorphone were not significantly different (P (cid:129)
`0.2980 and P (cid:129) 0.6290, respectively). There were no
`significant differences in overall effectiveness ratings
`by patients (2.1 { 0.1 vs. 2.1 { 0.1, P (cid:129) 0.9275) and
`by investigators (2.0 { 0.2 vs. 1.9 { 0.1, P (cid:129) 0.9510).
`The mean sedation VAS scores for each treatment
`are given in Table 2 by day of treatment and in Table 3
`and Figure 4 by time of day. There were no significant
`differences between treatments in sedation VAS when
`tested by day of treatment (P (cid:129) 0.1114) and by time
`of day (P (cid:129) 0.1114). The overall mean sedation VAS
`scores across all days was 24 { 4 mm for controlled-
`release oxycodone and 18 { 3 mm for controlled-re-
`lease hydromorphone (P (cid:129) 0.0843).
`Nausea VAS scores for each treatment are given
`in Table 2 by day of treatment and in Table 3 and
`Figure 5 by time of day. There were no significant
`differences between treatments in nausea scores when
`tested by day of treatment (P (cid:129) 0.3807) and by time
`of day (P (cid:129) 0.3807). The overall mean nausea VAS
`score across all days was 15 { 3 mm for controlled-
`release oxycodone and 13 { 3 mm for controlled-re-
`lease hydromorphone (P (cid:129) 0.1958). The overall fre-
`quency of adverse events with controlled-release oxy-
`codone and controlled-release hydromorphone, re-
`corded from spontaneous reports and through the use
`of a nondirected adverse events questionnaire, were
`
`not significantly different (P (cid:129) 0.8250) and generally
`consistent with the use of opioid analgesics to treat
`patients with advanced cancer. Among patients com-
`pleting the study, there were no significant differences
`in the frequency of elicited adverse events, except with
`regard to drowsiness, which was reported more fre-
`quently with oxycodone than with hydromorphone (28
`vs. 19 patients, P (cid:129) 0.0160). Two patients developed
`hallucinations with controlled-release hydromor-
`phone, but none did with controlled-release oxyco-
`done.
`At the completion of the double-blind evaluation,
`13 patients asked to participate in long term, open-
`label treatment with controlled-release oxycodone.
`The starting dose of controlled-release oxycodone was
`111 { 27 mg (range, 20–360 mg), and the daily final
`dose was 149 { 27 mg (range, 20–360), taken for a
`mean duration of 149 days (range, 3–373 days). Pain
`was generally well controlled, although most patients
`required dose escalation at some point in the trajec-
`tory of their illness.
`
`DISCUSSION
`This is the first randomized clinical trial to compare
`the efficacy and safety of hydromorphone and oxy-
`codone, two potent opioid agonists, in the treatment
`of chronic severe cancer pain.
`Oral morphine is widely utilized for the manage-
`ment of chronic severe cancer pain, and it has been
`considered the opioid of choice by many clinicians.
`Unfortunately, the occurrence of unmanageable ad-
`verse effects requires its discontinuation in some pa-
`tients. Recent clinical experience suggests that pa-
`tients who have failed to obtain adequate analgesia
`
`/ 7b52$$0984
`
`03-07-97 11:35:20
`
`canas
`
`W: Cancer
`
`6
`
`

`

`1434
`
`CANCER April 1, 1997 / Volume 79 / Number 7
`
`due to intolerable and unmanageable side effects
`while taking one opioid may benefit from switching
`to an alternative opioid.11,13 – 20 Maddocks et al. have
`shown that in patients with morphine-induced delir-
`ium, the substitution of morphine with oxycodone
`results in significant improvement in mental status,
`nausea, and vomiting.20
`There is extensive clinical experience with the
`use of hydromorphone as an alternative to mor-
`phine. The recent availability of a controlled-release
`formulation of hydromorphone9,10,25 has provided an
`opportunity for an expanded role for this semisyn-
`thetic congener of morphine.
`In contrast, oxycodone is an opioid analgesic
`that has traditionally been utilized in the treatment
`of moderate pain. Oxycodone was first introduced
`in North America as a fixed dose opioid/non-opioid
`combination, and it has generally been used for the
`same indications as codeine or propoxyphene com-
`binations. Although there is convincing evidence
`that oral oxycodone is twice as potent as oral mor-
`phine3 – 6 and equally effective,1,2,29 its widespread use
`has been hampered in part by a need for dosing
`every 4 hours. Ferrell et al. have demonstrated that
`compliance with opioid analgesics increases as the
`required dosing frequency decreases and that non-
`compliance results in suboptimal pain control and
`poor quality of life.30 Therefore, availability of a con-
`trolled-release formulation of oxycodone could en-
`hance its utility as a strong opioid alternative to oral
`morphine for severe cancer pain.
`Opioid rotation has been widely utilized as a
`strategic intervention for dose-limiting toxicities to
`opioids, and it has been recommended in clinical
`practice guidelines for cancer pain management.11
`There is published evidence from case series that
`switching to hydromorphone, oxycodone, or other
`morphine alternatives can be an effective strategy
`for the analgesic management of patients who de-
`velop encephalopathy, sedation, myoclonus, nausea
`and vomiting, hyperalgesia, and analgesic fail-
`ure.11,14 – 20 Variations
`in pharmacodynamic
`re-
`sponses to different opioids have been observed dur-
`ing treatment with drugs that appear to have phar-
`macologically similar sites of activity.14 – 20 These
`reports contradict
`the hypothesis that any one
`strong opioid is uniquely more efficacious than an-
`other. Recent evidence suggests that in addition to m
`receptor – mediated analgesia, oxycodone possesses
`intrinsic activity at the k receptor,31 providing fur-
`ther rationale for its use in treating patients unable
`to obtain an optimal analgesic or side effect profile
`with morphine or hydromorphone. Clinicians can
`
`exploit this variability in drug response by empiri-
`cally offering sequential trials of different opioids
`in order to optimize analgesia and minimize side
`effects.
`In selecting opioid analgesics for cancer pain,
`clinicians need to be mindful of the high prevalence
`of organ impairment and substantial accumulation
`of opioid metabolites, such as morphine-3-glucuro-
`nide and morphine-6-glucuronide.32,33 Morphine-3-
`glucuronide accumulation has been implicated in
`hyperalgesia, respiratory stimulation, and behav-
`ioral excitatory properties through nonopioid recep-
`tor mechanisms.34 – 39 Morphine-6-glucuronide accu-
`mulation has been implicated in increasing levels of
`nausea and sedation in patients with renal impair-
`ment.40 Furthermore, morphine and its principle
`metabolites may also accumulate in patients with
`hepatic dysfunction.41 In contrast, the major metab-
`olite of oxycodone, noroxycodone, is less than 1/
`100th as potent as the parent compound;42 oxyco-
`done administered to patients with renal or hepatic
`impairment only slightly alters the pharmacokinet-
`ics of oxycodone and its metabolites.43,44 Oxymor-
`phone, another metabolite of oxycodone, is pharma-
`cologically active but is produced in such small con-
`centrations that it is unlikely to be of consequence
`in most clinical settings.44
`The results of our study demonstrate that
`chronic severe cancer pain can be well controlled
`with a regular regimen of controlled-release oxyco-
`done given every 12 hours and that the efficacy of
`this regimen is at least equal to that of controlled-
`release hydromorphone.9,10,25 The duration of anal-
`gesia provided by controlled-release oxycodone and
`controlled-release hydromorphone was compared
`through analysis of pain intensity scores and rescue
`analgesic use during three equal 4-hour periods con-
`stituting the 12-hour dosing interval. Analgesics last-
`ing less than 12 hours would be expected to result
`in an increase in pain intensity or rescue analgesic
`use during the period immediately before the next
`dose. The lack of such an end of dose effect in the
`current study provides pharmacodynamic confir-
`mation of the previously documented sustained-re-
`lease pharmacokinetic profile of controlled-release
`oxycodone.45,46
`There were no significant differences in the fre-
`quency of spontaneously reported and elicited ad-
`verse events in the current study, with the exception
`of drowsiness, which occurred more frequently dur-
`ing treatment with oxycodone. However, this finding
`was not consistent with the results of more system-
`
`/ 7b52$$0984
`
`03-07-97 11:35:20
`
`canas
`
`W: Cancer
`
`7
`
`

`

`atic evaluation of sedation using a VAS four times a
`day.
`The mean final daily controlled-release oxyco-
`done dose of 124 mg in our study (equivalent to
`approximately 250 mg of oral morphine) is compara-
`ble to the dose of controlled-release morphine used
`in previous randomized trials in cancer pain.27,28 The
`dose of oral oxycodone required to provide optimal
`analgesia without intolerable side effects ranged
`from 20 – 550 mg per day. This wide variability
`among patients is consistent with the results of pre-
`vious studies with controlled-release morphine27,28
`and controlled-release hydromorphone9,10,25 and is
`well within the range of oxycodone doses used in
`the management of cancer pain.1,2 Differences in an-
`algesic requirements may result from the pharmaco-
`kinetic variability observed after oral administration
`of oxycodone.1,45 However, variability in pharmaco-
`kinetics is probably not sufficient to explain the wide
`range of oxycodone doses required in the current
`study. A variety of factors may be important determi-
`nants of opioid requirement, including prior opioid
`exposure, pain mechanism, predisposition to side
`effects, and psychologic distress.47
`Breakthrough pain is reported by about two-
`thirds of patients with well-controlled cancer pain
`and is generally managed by titration of the sched-
`uled dose of controlled release opioids to manage
`baseline pain, together with administration of im-
`mediate-release opioids as required. It is notewor-
`thy that the pattern of steady pain and break-
`through pain in our patients was dynamic despite
`relatively stable analgesic requirements. When we
`evaluated pain intensity on VAS and categorical
`scales by time of day, it was apparent that pain
`was at its worst for both treatments during daytime
`hours. This period of increased pain intensity also
`coincided with a rise in rescue analgesic use. In our
`study, the most likely cause of circadian worsening
`of pain appeared to be due to an increase of patient
`activity in the daytime (incident pain).
`At the start of this study, 45% of our patients had
`at least some neuropathic component to their pain.
`Although the efficacy of opioid analgesics in neuro-
`pathic pain syndromes remains a polarized area of
`clinical practice, data from well-controlled clinical tri-
`als indicate that pain of neuropathic origin can be at
`least partially responsive to opioid analgesics.48–50 The
`mean daily oxycodone dose given to patients with
`neuropathic pain was only slightly higher than that
`given to patients with nonneuropathic pain (136 { 36
`mg vs. 114 { 26 mg, respectively).
`
`Oxycodone for Cancer Pain/Hagen and Babul
`
`1435
`
`CONCLUSIONS
`We conclude that the recently available controlled-
`release oxycodone formulation, with its biphasic ab-
`sorption profile, is highly effective in the management
`of chronic severe cancer pain when administered ev-
`ery 12 hours. Our experience suggests that oxycodone
`can be successfully used for the treatment of severe
`cancer pain, adding to extensive experience of others
`in the use of oxycodone for moderate cancer pain.
`
`REFERENCES
`1.
`Po¨yho¨a R, Vainio A, Kalso E. A review of oxycodone’s clinical
`pharmacokinetics and pharmacodynamics. J Pain Symptom
`Manage 1993;8:63–7.
`2. Kalso E, Vainio A. Morphine and oxycodone hydrochloride
`in the management of cancer pain. Clin Pharmacol Ther
`1990;47:639–46.
`3. Beaver WT, Wallenstein SL, Rogers A, Houde RW. A

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