WO2000025769A1 - Analgesic regimen - Google Patents

Analgesic regimen Download PDF

Info

Publication number
WO2000025769A1
WO2000025769A1 PCT/US1999/023513 US9923513W WO0025769A1 WO 2000025769 A1 WO2000025769 A1 WO 2000025769A1 US 9923513 W US9923513 W US 9923513W WO 0025769 A1 WO0025769 A1 WO 0025769A1
Authority
WO
WIPO (PCT)
Prior art keywords
tramadol
days
day
titration
regimen
Prior art date
Application number
PCT/US1999/023513
Other languages
French (fr)
Inventor
Marc Kamin
William Olson
Original Assignee
Ortho-Mcneil Pharmaceutical, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ortho-Mcneil Pharmaceutical, Inc. filed Critical Ortho-Mcneil Pharmaceutical, Inc.
Priority to AU64224/99A priority Critical patent/AU763273B2/en
Priority to CA002348907A priority patent/CA2348907A1/en
Priority to EA200100501A priority patent/EA003557B1/en
Priority to BR9914981-8A priority patent/BR9914981A/en
Priority to EP99951876A priority patent/EP1126834A1/en
Priority to PL99348808A priority patent/PL348808A1/en
Publication of WO2000025769A1 publication Critical patent/WO2000025769A1/en
Priority to NO20012133A priority patent/NO20012133L/en
Priority to HR20010311A priority patent/HRP20010311A2/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/13Amines
    • A61K31/135Amines having aromatic rings, e.g. ketamine, nortriptyline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]

Definitions

  • This invention relates to a dosing regimen for the administration of the analgesic tramadol.
  • the dosing regimen achieves the desired analgesic effect while reducing or delaying the on-set of the side effects generally associated with the administration of tramadol.
  • Tramadol the chemical name for which is 2- [ (dimethylamino) methyl] -1- (3-methoxyphenyl) cyclohexanol, is a synthetic, centrally-acting analgesic that is effective for the treatment of moderate to moderately- severe chronic pain. It has been marketed under the trade name TramalTM since 1977 in the dosage forms of capsules, injections, suppositories and drops.
  • the compound can be employed as the free base or its pharmaceutically acceptable salts, stereo isomers and solvates. It is generally supplied in the form of its hydrochloride salt and over 400 million doses of tramadol have been administered since its introduction in Germany.
  • analgesic therapeutic regimen that is both effective and well tolerated.
  • the two traditional categories of analgesics i.e. opioids and nonsteroidal anti- inflammatory drugs (NSAIDs)
  • opioids and nonsteroidal anti- inflammatory drugs are both effective but are associated with potentially serious side effects.
  • Concerns regarding tolerance and dependence minimize the chronic use of narcotics such as morphine and codeine for the treatment of chronic pain.
  • Patients on chronic NSAID therapy risk severe gastrointestinal symptoms, including ulceration and bleeding which have been estimated to result in up to 20,000 deaths each year.
  • An alternative to this dilemma is tramadol, a non-narcotic, non-NSAID analgesic which is indicated for the management of moderate to moderately-severe pain.
  • tramadol After oral administration, tramadol is rapidly and almost completely absorbed and is extensively metabolized. The major metabolic pathways appear to be N- and O- demethylation and glucuronidation or sulfation in the liver. Only one metabolite, i.e. mono-O- desmethyltramadol, has been found to be pharmacologically active .
  • peak plasma concentrations of tramadol hydrochloride occurring two hours after administration are 308+78 ng/ml mean+standard deviation) .
  • Peak plasma concentrations of mono-O-desmethyl tramadol, the active metabolite of tramadol are 55+20 ng/ml, occurring approximately three hours after administration.
  • the terminal plasma elimination half-lives of tramadol hydrochloride and its active metabolite are 6.3 ⁇ 11.4 hours and 7.4+1.4 hours respectively.
  • Tramadol is poorly bound to plasma proteins (20.2%) thus decreasing the potential for drug interactions with highly protein-bound agents.
  • tramadol is not completely understood, but in animal models at least two complementary mechanisms appear to be involved and they are 1) weak binding to the ⁇ opioid receptors and 2) weak inhibition of the reuptake of norepinephrine and serotonin.
  • Tramadol is not chemically related to opiates, but its actions are similar to those of opioid (narcotic) analgesics.
  • the opioid activity of tramadol results from the low-affinity binding of tramadol hydrochloride and the higher affinity binding of the metabolite to ⁇ receptors; however, its induced antinociception is only partially antagonized by the opiate antagonist naloxone in several animal tests.
  • the inhibition of norepinephrine and serotonin reuptake which has been demonstrated in vi tro, is postulated to contribute independently to the overall analgesic profile of tramadol hydrochloride.
  • Tramadol is well tolerated, however, nuisance adverse events such as drowsiness, vomiting and dizziness can occur during the initiation of treatment which may lead to early discontinuation of the treatment.
  • the most frequently reported adverse events observed in clinical trials of tramadol hydrochloride are constipation, nausea, dizziness/vertigo, headache, somnolence, and vomiting.
  • the efficacy, safety, and pharmacokinetic profile of tramadol hydrochloride indicate that the drug may be useful in treating chronic pain.
  • An object of the present invention is to demonstrate that the frequency of nausea and vomiting, two of the most frequently reported adverse events and the events most commonly associated with discontinuation of treatment, as well other adverse events, can be reduced using a lower dosage titration scheme without diminishing the efficacy of the compound.
  • the present invention relates to a dosage regimen which consists of slowing the titration rate for tramadol which results in a reduction of the incidence of discontinuation due to side effects such as nausea and vomiting.
  • the present invention relates to a dosage regimen for tramadol which involves a slower titration rate than that currently prescribed.
  • the slower rate of titration of tramadol therapy results in improved tolerability of the drug.
  • the novel regimen results in a significant reduction in discontinuations due to a lower incidence or severity of side effects.
  • tramadol is intended to include its pharmaceutically acceptable salts, stereo isomers and solvates thereof.
  • Tramadol is indicated for the treatment of moderate to moderately-severe pain and its typical dosing regimen is 50-100 mg every 4 to 6 hours. About 200 mg/day is considered to be a normal initial dose. Clinical studies have shown tramadol to be an effective treatment for chronic joint pain. Tramadol is well tolerated, however, nuisance adverse events can occur during initiation of treatment with tramadol. These side effects may lead to early discontinuation of tramadol therapy.
  • Slow titration of a therapeutic agent is often used by practicing clinicians to minimize adverse events associated with centrally-acting agents such as antidepressants, analgesics and anticonvulsants. Although slow titration may minimize the adverse side effects associated with a particular agent, it may also delay the onset of the effect of the agent as well. It has now been discovered that initiating tramadol therapy using slow titration rates according to the regimen of this invention minimizes discontinuations due to adverse side effects associated with tramadol while maintaining its therapeutic effectiveness which results in a greater tolerance of the drug during therapy.
  • the regimen which is the basis of the present invention is a 1-28 day regimen.
  • tramadol is administered over a ten-28 day period starting on day one in a pharmaceutical composition containing from about 10 - 50 mg of tramadol and the amount of drug is increased incrementally over the next 9-28 days until the target dose of about 200-400 mg/day is reached.
  • Many patients find effective pain relief at 200 mg/day, however, some patients may require doses of up to 400 mg/day in order to achieve the desired relief.
  • tramadol in the form of the free base or its pharmaceutically acceptable salt, is administered at a dose of about 10-50 mg .
  • tramadol On days 4-6 of the regimen tramadol is administered at a dose in the range of about 20-100 mg. On days 7-9 tramadol is administered at a dose in the range of about 30-150 mg and on days 10-28 and thereafter at a dose of about 40-400 mg. At the end of the period the therapy is continued at the target dose which may be anywhere from 200 to about 400 mg of tramadol.
  • tramadol is administered in a regimen which comprises administering tramadol at the rate of about 25 mg of tramadol on days 1-3; 50 mg of tramadol on days 4-6; 75 mg of tramadol on days 7-9; 100 mg of tramadol on days 10-12; 150 mg of tramadol on days 13-15; and 200 mg of tramadol on days 16-28 and thereafter.
  • tramadol is administered in a regimen which comprises administering tramadol at the rate of 50 mg of tramadol on days 1-3; 100 mg of tramadol on days 4-6; 150 mg of tramadol on days 7-9; and 200 mg of tramadol on day 10 and thereafter.
  • the drug is generally administered in the form of its pharmaceutically salt.
  • suitable pharmaceutically acceptable salts include salts of inorganic acids such as hydrochloric and hydrobromic acid.
  • the preferred salt is the hydrochloride salt .
  • the slower initial titration rate of tramadol is effective in reducing discontinuations due to adverse effects while maintaining the analgesic properties of the compound. This is particularly true in the case of patients who previously had difficulty tolerating an analgesic because of side effects such as nausea and/or vomiting. This result is based on the cumulative proportion of patients who discontinued use of the agent due to adverse side effects.
  • EXAMPLE 1 Tramadol, in the form of its hydrochloride salt, was studied in a multicenter, outpatient, randomized, double- blind parallel study that compared the effect of different titration rates of tramadol versus placebo on the incidence of discontinuations resulting from adverse events in patients with chronic joint pain.
  • a total of 465 patients with chronic joint pain were enrolled in the study and randomized into one of four treatment groups for 14 days. Patients continued on their prestudy dose of NSAIDs while concurrently receiving tramadol or placebo. Tramadol groups were titrated at three different rates to achieve the study target dose of 200 mg/day. Each group was examined to determine if a slower titration resulted in a statistically significant (p ⁇ 0.05) trend towards fewer discontinuations due to nausea and/or vomiting and dizziness and/or vertigo. Discontinuation due to any adverse event was similarly analyzed. If the trend was statistically significant, pairwise comparisons were performed to determine the statistical significance between titration rates.
  • the study protocol was approved by an Institutional Review Board at each study site and informed consent regarding the risks and benefits of participation was obtained for all patients.
  • Double-blind therapy began on Day 1, with patients receiving either placebo or one of three titration regimens, ultimately reaching the study target dose of 200 mg/day of tramadol in either 1 day, 4 days (increasing by 50 mg increments each day; Figures la, lb and lc) , or 10 days (increasing by 50 mg increments every 3 days) Patients continued to take their stable dose of NSAID throughout the double- blind phase, in addition to the study drug or placebo. On Day 14, all patients underwent a physical examination with clinical laboratory tests, as well as adverse event assessment Patients could be discontinued from the study as the result of adverse events, treatment failure, significant protocol violation, development of an intercurrent illness or at their own request.
  • Tramadol in the form of its hydrochloride salt, was studied in a multicenter, outpatient, randomized, double-blind parallel study comprised of two phases: a screening/open-label run-in study and a double-blind phase.
  • Subjects with chronic pain e.g. musculoskeletal , neuropathic, joint etc.
  • Subjects with chronic pain e.g. musculoskeletal , neuropathic, joint etc.
  • a daily NSAID dose for at least 30 days prior to the study, who required additional relief of their chronic pain, and who completed the screening evaluations were enrolled in the open-label phase on Day 0 and began open- label study medication on Day 1.
  • Tramadol hydrochloride was titrated in 50 mg/day increments to 200 mg/day over four days. Subjects continued on the 200 mg/day dosage for up to an additional 10 days.
  • Subjects were randomized on Day 0 and began double-blind therapy on Day 1 with one of three dosage regimens of tramadol hydrochloride that employed either a 10-, 16- or 13 -day titration schedule in order to achieve a maximum dose of either 200 mg/day for the 10- and 16-day regimen or 150 mg/day for the 13 -day regimen.
  • the subjects took double-blind study medication consisting of either 25 mg of tramadol hydrochloride or matching placebo (two capsules q.i.d.).
  • the three tramadol hydrochloride dosage regimens were designed to achieve a maximum dose (200 mg/day or 150 mg/day) at different rates of titration (10- , 16- or 13-day) .
  • Subjects assigned to the 10 -day titration group received tramadol hydrochloride at 50 mg q.d. on Days 1-3, 50 mg b . i . d . on Days 4-6, 50 mg t.i.d. on Days 7-9 and 50 mg q.i.d. on Days 10-28; subjects assigned to the 16-day titration group received tramadol hydrochloride at 25 mg q.d. on Days 1-3, 25 mg b.i.d. on Days 4-6, 25 mg t.i.d. on Days 7-9, 25 mg q.i.d. on Days 10-12, 50 mg t.i.d. on Days 13-15 and 50 mg q.i.d.
  • Efficacy evaluations were performed at all visits. These evaluations included a subject assessment of pain using a 10 cm pain visual analogue (PVA) scale and overall assessments of the study medication made by the subject and the investigator. Safety evaluations were performed at screening, at the end of the open-label/run-in phase and at the end of the double-blind phase and included assessments of the occurrence of adverse events, vital signs and body weight measurements and physical examinations .
  • PVA pain visual analogue
  • the primary analysis group included 167 subjects, including 54, 59 and 54 subjects in the tramadol hydrochloride treatment groups that employed 10-, 16- and 13-day titration periods, respectively.
  • Chronic low-back syndrome and osteoarthritis were the most common chronic painful conditions reported for the overall population (each represented 28.1% of subjects). The relative proportions of other chronic painful conditions varied somewhat across treatment groups.
  • the mean reduction from baseline PVA scores for subjects in the open-label/run-in phase was 2.1 cm.
  • the mean reduction from baseline PVA scores was highest in the 13 -day tramadol hydrochloride titration group (1.6 cm), followed by the 16-day titration group (1.5 cm); and the 10-day titration group (1.4 cm) .
  • the survival curve plots the relationship between the cumulative probability of discontinuation and the length of exposure. Examination of the curves ( Figure 2) shows the cumulative probability of discontinuation due to nausea and/or vomiting in the three titration groups to be similar through the first five days of titration.
  • the survival curve plots the relationship between the cumulative probability of discontinuation and length of exposure. Examination of the curves ( Figure 3) shows the cumulative probability of discontinuation due to any adverse event in the three titration groups to be similar through the first five days of titration.
  • the cumulative probability of discontinuation due to any adverse event in the 10-day and 16-day titration groups continue to increase while the survival curves of the 13- day titration group begins to fall below them.
  • the survival curves of the 16-day and 13 day titration groups begin to plateau while the cumulative probability of discontinuation in the 10 -day titration group continues to increase.
  • the median time to discontinuation due to any adverse event was shorter for the 16 -day tramadol hydrochloride titration group (6.0 days; Figure 5) followed by the 13- day group (6.5 days) and the 10-day group (9.0 days) .

Abstract

A regimen for the administration of tramadol for the treatment of analgesia is described. The regimen involves a slower initial titration rate of tramadol which results in a significantly lower percentage of discontinuations of therapy due to a lower incidence and severity of side effects.

Description

ANALGESIC REGIMEN
FIELD OF THE INVENTION
This invention relates to a dosing regimen for the administration of the analgesic tramadol. The dosing regimen achieves the desired analgesic effect while reducing or delaying the on-set of the side effects generally associated with the administration of tramadol.
BACKGROUND OF THE INVENTION
Tramadol, the chemical name for which is 2- [ (dimethylamino) methyl] -1- (3-methoxyphenyl) cyclohexanol, is a synthetic, centrally-acting analgesic that is effective for the treatment of moderate to moderately- severe chronic pain. It has been marketed under the trade name Tramal™ since 1977 in the dosage forms of capsules, injections, suppositories and drops. The compound can be employed as the free base or its pharmaceutically acceptable salts, stereo isomers and solvates. It is generally supplied in the form of its hydrochloride salt and over 400 million doses of tramadol have been administered since its introduction in Germany.
Patients experiencing chronic pain require an analgesic therapeutic regimen that is both effective and well tolerated. The two traditional categories of analgesics, i.e. opioids and nonsteroidal anti- inflammatory drugs (NSAIDs) , are both effective but are associated with potentially serious side effects. Concerns regarding tolerance and dependence minimize the chronic use of narcotics such as morphine and codeine for the treatment of chronic pain. Patients on chronic NSAID therapy risk severe gastrointestinal symptoms, including ulceration and bleeding which have been estimated to result in up to 20,000 deaths each year. An alternative to this dilemma is tramadol, a non-narcotic, non-NSAID analgesic which is indicated for the management of moderate to moderately-severe pain.
After oral administration, tramadol is rapidly and almost completely absorbed and is extensively metabolized. The major metabolic pathways appear to be N- and O- demethylation and glucuronidation or sulfation in the liver. Only one metabolite, i.e. mono-O- desmethyltramadol, has been found to be pharmacologically active .
After a single 100 mg oral dose in healthy subjects, peak plasma concentrations of tramadol hydrochloride occurring two hours after administration are 308+78 ng/ml mean+standard deviation) . Peak plasma concentrations of mono-O-desmethyl tramadol, the active metabolite of tramadol, are 55+20 ng/ml, occurring approximately three hours after administration. The terminal plasma elimination half-lives of tramadol hydrochloride and its active metabolite are 6.3±11.4 hours and 7.4+1.4 hours respectively. Tramadol is poorly bound to plasma proteins (20.2%) thus decreasing the potential for drug interactions with highly protein-bound agents.
The mode of action of tramadol is not completely understood, but in animal models at least two complementary mechanisms appear to be involved and they are 1) weak binding to the μ opioid receptors and 2) weak inhibition of the reuptake of norepinephrine and serotonin. Tramadol is not chemically related to opiates, but its actions are similar to those of opioid (narcotic) analgesics. The opioid activity of tramadol results from the low-affinity binding of tramadol hydrochloride and the higher affinity binding of the metabolite to μ receptors; however, its induced antinociception is only partially antagonized by the opiate antagonist naloxone in several animal tests. The inhibition of norepinephrine and serotonin reuptake, which has been demonstrated in vi tro, is postulated to contribute independently to the overall analgesic profile of tramadol hydrochloride.
Tramadol is well tolerated, however, nuisance adverse events such as drowsiness, vomiting and dizziness can occur during the initiation of treatment which may lead to early discontinuation of the treatment. The most frequently reported adverse events observed in clinical trials of tramadol hydrochloride are constipation, nausea, dizziness/vertigo, headache, somnolence, and vomiting. Taken together, the efficacy, safety, and pharmacokinetic profile of tramadol hydrochloride indicate that the drug may be useful in treating chronic pain.
An object of the present invention is to demonstrate that the frequency of nausea and vomiting, two of the most frequently reported adverse events and the events most commonly associated with discontinuation of treatment, as well other adverse events, can be reduced using a lower dosage titration scheme without diminishing the efficacy of the compound. The present invention relates to a dosage regimen which consists of slowing the titration rate for tramadol which results in a reduction of the incidence of discontinuation due to side effects such as nausea and vomiting.
SUMMARY OF THE INVENTION
The present invention relates to a dosage regimen for tramadol which involves a slower titration rate than that currently prescribed. The slower rate of titration of tramadol therapy results in improved tolerability of the drug. The novel regimen results in a significant reduction in discontinuations due to a lower incidence or severity of side effects. As used hereinafter, the word tramadol is intended to include its pharmaceutically acceptable salts, stereo isomers and solvates thereof.
DETAILED DESCRIPTION OF THE INVENTION
Tramadol is indicated for the treatment of moderate to moderately-severe pain and its typical dosing regimen is 50-100 mg every 4 to 6 hours. About 200 mg/day is considered to be a normal initial dose. Clinical studies have shown tramadol to be an effective treatment for chronic joint pain. Tramadol is well tolerated, however, nuisance adverse events can occur during initiation of treatment with tramadol. These side effects may lead to early discontinuation of tramadol therapy.
Slow titration of a therapeutic agent is often used by practicing clinicians to minimize adverse events associated with centrally-acting agents such as antidepressants, analgesics and anticonvulsants. Although slow titration may minimize the adverse side effects associated with a particular agent, it may also delay the onset of the effect of the agent as well. It has now been discovered that initiating tramadol therapy using slow titration rates according to the regimen of this invention minimizes discontinuations due to adverse side effects associated with tramadol while maintaining its therapeutic effectiveness which results in a greater tolerance of the drug during therapy.
The regimen which is the basis of the present invention is a 1-28 day regimen. In practice, tramadol is administered over a ten-28 day period starting on day one in a pharmaceutical composition containing from about 10 - 50 mg of tramadol and the amount of drug is increased incrementally over the next 9-28 days until the target dose of about 200-400 mg/day is reached. Many patients find effective pain relief at 200 mg/day, however, some patients may require doses of up to 400 mg/day in order to achieve the desired relief. Generally, on days 1-3 of the regimen tramadol, in the form of the free base or its pharmaceutically acceptable salt, is administered at a dose of about 10-50 mg . On days 4-6 of the regimen tramadol is administered at a dose in the range of about 20-100 mg. On days 7-9 tramadol is administered at a dose in the range of about 30-150 mg and on days 10-28 and thereafter at a dose of about 40-400 mg. At the end of the period the therapy is continued at the target dose which may be anywhere from 200 to about 400 mg of tramadol.
In a preferred embodiment of the invention tramadol is administered in a regimen which comprises administering tramadol at the rate of about 25 mg of tramadol on days 1-3; 50 mg of tramadol on days 4-6; 75 mg of tramadol on days 7-9; 100 mg of tramadol on days 10-12; 150 mg of tramadol on days 13-15; and 200 mg of tramadol on days 16-28 and thereafter.
In another preferred embodiment of the invention tramadol is administered in a regimen which comprises administering tramadol at the rate of 50 mg of tramadol on days 1-3; 100 mg of tramadol on days 4-6; 150 mg of tramadol on days 7-9; and 200 mg of tramadol on day 10 and thereafter.
The drug is generally administered in the form of its pharmaceutically salt. Suitable pharmaceutically acceptable salts include salts of inorganic acids such as hydrochloric and hydrobromic acid. The preferred salt is the hydrochloride salt .
The slower initial titration rate of tramadol is effective in reducing discontinuations due to adverse effects while maintaining the analgesic properties of the compound. This is particularly true in the case of patients who previously had difficulty tolerating an analgesic because of side effects such as nausea and/or vomiting. This result is based on the cumulative proportion of patients who discontinued use of the agent due to adverse side effects.
The following examples describe the invention in greater detail and are intended to illustrate the invention but not to limit it.
EXAMPLE 1 Tramadol, in the form of its hydrochloride salt, was studied in a multicenter, outpatient, randomized, double- blind parallel study that compared the effect of different titration rates of tramadol versus placebo on the incidence of discontinuations resulting from adverse events in patients with chronic joint pain.
A total of 465 patients with chronic joint pain were enrolled in the study and randomized into one of four treatment groups for 14 days. Patients continued on their prestudy dose of NSAIDs while concurrently receiving tramadol or placebo. Tramadol groups were titrated at three different rates to achieve the study target dose of 200 mg/day. Each group was examined to determine if a slower titration resulted in a statistically significant (p<0.05) trend towards fewer discontinuations due to nausea and/or vomiting and dizziness and/or vertigo. Discontinuation due to any adverse event was similarly analyzed. If the trend was statistically significant, pairwise comparisons were performed to determine the statistical significance between titration rates.
The study protocol was approved by an Institutional Review Board at each study site and informed consent regarding the risks and benefits of participation was obtained for all patients. Patients who were (1) 45 years of age or older; (2) had a diagnosis of symptomatic chronic joint pain, as confirmed by x-ray; (3) were in otherwise good general health; (4) were on a stable dose of NSAID for at least 30 days; and (5) required additional pain relief were eligible to enroll in the study. Patients with chronic joint pain who had been receiving a maintenance dose (+ 25%) of NSAID for at least 30 days, and who required additional pain relief, were randomized into three titration groups and a placebo group using a 2:2:2:1 randomization schedule. Double-blind therapy began on Day 1, with patients receiving either placebo or one of three titration regimens, ultimately reaching the study target dose of 200 mg/day of tramadol in either 1 day, 4 days (increasing by 50 mg increments each day; Figures la, lb and lc) , or 10 days (increasing by 50 mg increments every 3 days) Patients continued to take their stable dose of NSAID throughout the double- blind phase, in addition to the study drug or placebo. On Day 14, all patients underwent a physical examination with clinical laboratory tests, as well as adverse event assessment Patients could be discontinued from the study as the result of adverse events, treatment failure, significant protocol violation, development of an intercurrent illness or at their own request.
Patients were randomly assigned according to a central computer-generated schedule to receive placebo or one of three tramadol dosage regimens that involved either a 1-, 4- or 10-day titration schedule to attain the study target dose of 200 mg/day. Study medication or placebo was administered four times a day using a double-dummy technique to ensure blinding throughout the full titration schedule. Medication distribution was blinded and controlled through the use of blister packs which contained medication with the appropriate number of active tablets and placebos for the 14-day study period, plus 2 additional days of therapy. Three hundred and fifty-two (352) patients completed the study. Reasons leading to discontinuation included adverse events, lack of drug effectiveness, intercurrent illness, protocol violation and patient choice.
The results of the study showed that a slower initial titration of tramadol is effective in reducing discontinuations due to all adverse effects and, in particular, dizziness and nausea.
Among the three tramadol titration groups, patients in the 10 -day titration group experienced the fewest discontinuations due to dizziness and/or vertigo, nausea and/or vomiting, and any other adverse event. The 10-day titration rate was statistically significantly different (<0.05) from both the 1-day rate and the 4-day rate for discontinuations due to dizziness and/or vertigo and any other adverse event . The study demonstrated that a slower rate of initiation of tramadol therapy (i.e. 50 mg increments every 3 days) will result in improved tolerability because of significantly fewer discontinuations due the occurrence of adverse events.
EXAMPLE 2
Tramadol, in the form of its hydrochloride salt, was studied in a multicenter, outpatient, randomized, double-blind parallel study comprised of two phases: a screening/open-label run-in study and a double-blind phase. Subjects with chronic pain (e.g. musculoskeletal , neuropathic, joint etc.) for at least three months prior to the study, who had been receiving a daily NSAID dose for at least 30 days prior to the study, who required additional relief of their chronic pain, and who completed the screening evaluations were enrolled in the open-label phase on Day 0 and began open- label study medication on Day 1. Tramadol hydrochloride was titrated in 50 mg/day increments to 200 mg/day over four days. Subjects continued on the 200 mg/day dosage for up to an additional 10 days.
Subjects who experienced nausea and/or vomiting within the 14 -day open-label period severe enough for the subjects to discontinue tramadol hydrochloride treatment had the opportunity to enter the double-blind phase. Approximately 150 adult male and female subjects who discontinued the open-label phase due to nausea and/or vomiting were randomized in the double-blind phase to one of three tramadol hydrochloride treatment regimens . Subjects who entered the double-blind phase were assigned in an even-distribution, randomized, double-blind fashion to one of three treatment regimens 10 days after discontinuing open-label tramadol hydrochloride. Subjects were randomized on Day 0 and began double-blind therapy on Day 1 with one of three dosage regimens of tramadol hydrochloride that employed either a 10-, 16- or 13 -day titration schedule in order to achieve a maximum dose of either 200 mg/day for the 10- and 16-day regimen or 150 mg/day for the 13 -day regimen. On Days 1-28, the subjects took double-blind study medication consisting of either 25 mg of tramadol hydrochloride or matching placebo (two capsules q.i.d.). The three tramadol hydrochloride dosage regimens were designed to achieve a maximum dose (200 mg/day or 150 mg/day) at different rates of titration (10- , 16- or 13-day) .
Subjects assigned to the 10 -day titration group received tramadol hydrochloride at 50 mg q.d. on Days 1-3, 50 mg b . i . d . on Days 4-6, 50 mg t.i.d. on Days 7-9 and 50 mg q.i.d. on Days 10-28; subjects assigned to the 16-day titration group received tramadol hydrochloride at 25 mg q.d. on Days 1-3, 25 mg b.i.d. on Days 4-6, 25 mg t.i.d. on Days 7-9, 25 mg q.i.d. on Days 10-12, 50 mg t.i.d. on Days 13-15 and 50 mg q.i.d. on Days 16-28; and subjects assigned to the 13 -day titration group received tramadol hydrochloride at 25 mg q.d. on Days 1-3, 25 mg b.i.d. on Days 4-6, 25 mg t.i.d. on Days 7-9, 25 mg q.i.d. on Days 10-12, and 50 mg t.i.d. on Days 13-28. Subjects who did not experience nausea and/or vomiting severe enough to discontinue tramadol hydrochloride treatment by the end of the open- label run-in phase were discontinued from the study. Subjects continued taking their daily dose of NSAID throughout both the open-label/run-in and double- blind phases of the study. At the completion of the double-blind phase or at the time of premature discontinuation, subjects returned to the invesitgational site for follow-up efficacy and safety evaluations.
Efficacy evaluations were performed at all visits. These evaluations included a subject assessment of pain using a 10 cm pain visual analogue (PVA) scale and overall assessments of the study medication made by the subject and the investigator. Safety evaluations were performed at screening, at the end of the open-label/run-in phase and at the end of the double-blind phase and included assessments of the occurrence of adverse events, vital signs and body weight measurements and physical examinations .
Analyses and summaries were performed for all subjects who discontinued the screening/open-label/run-in phase due to nausea and/or vomiting, who were then randomized to one of the three double-blind titration groups, who took at least one dose of the study medication, and who provided post baseline information. Subjects who participated in the open-label/run-in phase but who either did not qualify for or chose not to participate in the double-blind group are included only in the overall accounting of subjects entering the open- label/run- in phase.
A total of 931 subjects were enrolled in the study at 29 centers for the open-label/run-in phase. There were no apparent differences between the non-randomized and randomized population in terms of the demographic attributes of race, age, chronic painful condition, or time since diagnosis; however, a slightly higher percentage of women were randomized into the double-blind phase of the study. Osteoarthritis and chronic low-back syndrome were the most common chronic painful conditions reported for the overall population (29.3% and 28.8% of subjects, respectively).
The primary analysis group included 167 subjects, including 54, 59 and 54 subjects in the tramadol hydrochloride treatment groups that employed 10-, 16- and 13-day titration periods, respectively. Chronic low-back syndrome and osteoarthritis were the most common chronic painful conditions reported for the overall population (each represented 28.1% of subjects). The relative proportions of other chronic painful conditions varied somewhat across treatment groups.
The mean reduction from baseline PVA scores for subjects in the open-label/run-in phase was 2.1 cm. In the double-blind phase, the mean reduction from baseline PVA scores was highest in the 13 -day tramadol hydrochloride titration group (1.6 cm), followed by the 16-day titration group (1.5 cm); and the 10-day titration group (1.4 cm) .
The pairwise log-rank test revealed statistically significant differences (p=0.006) between the Kaplan-Meier survival curve of the 10 -day titration group and the survival curves of both the 16-day (p=0.007) and the 13- day titration group (p=0.006) with respect to discontinuations from nausea and/or dizziness. The survival curve plots the relationship between the cumulative probability of discontinuation and the length of exposure. Examination of the curves (Figure 2) shows the cumulative probability of discontinuation due to nausea and/or vomiting in the three titration groups to be similar through the first five days of titration. After day 5, the cumulative probability of discontinuation due to nausea and/or vomiting in the 10-day titration group continues to increase at essentially the same rate as seen in the first five days while the survival curves of the two slower titration groups plateau. The pairwise comparison between the survival curves of the 16-day and 13 -day titration groups was not statistically significant (p=0.94) .
The median time to discontinuation due to nausea and/or vomiting was shorter for the 16 -day tramadol hydrochloride titration group (4.0 days; (Figure 2) followed by the 13 -day group (5.5 days) and the 10-day group (9.0 days) .
The pairwise log-rank test revealed statistically significant differences between the Kaplan-Meier survival curve of the 10-day titration group and the survival curves of both the 16-day titration group (p=0.030) and the 13 -day titration group (p=0.010) with respect to discontinuations because of any adverse effect. The survival curve plots the relationship between the cumulative probability of discontinuation and length of exposure. Examination of the curves (Figure 3) shows the cumulative probability of discontinuation due to any adverse event in the three titration groups to be similar through the first five days of titration. After day 5, the cumulative probability of discontinuation due to any adverse event in the 10-day and 16-day titration groups continue to increase while the survival curves of the 13- day titration group begins to fall below them. After 10 days of titration, the survival curves of the 16-day and 13 day titration groups begin to plateau while the cumulative probability of discontinuation in the 10 -day titration group continues to increase. The pairwise comparison between the survival curves of the 13 -day and the 16-day groups was not statistically significant (p=0.620) .
The median time to discontinuation due to any adverse event was shorter for the 16 -day tramadol hydrochloride titration group (6.0 days; Figure 5) followed by the 13- day group (6.5 days) and the 10-day group (9.0 days) .
The studies demonstrated that a slower initial titration rate of tramadol hydrochloride reduced the incidence of discontinuation due to nausea and/or vomiting in subjects with chronic pain who previously had difficulty tolerating tramadol hydrochloride because of nausea and/or vomiting. This is based on the cumulative proportion of subjects who discontinued due to nausea and/or vomiting, with the group that titrates to 200 mg/day over 10 days showing a 20 percentage advantage over the groups that titrate to 200 mg/day over 16 days.

Claims

WHAT IS CLAIMED IS:
1. A regimen for the treatment of analgesia which comprises administering a pharmaceutical composition containing tramadol as the active ingredient which comprises: about 10-50 mg of tramadol on days 1-3; about 20-100 mg of tramadol on days 4-6; about 30-150 mg of tramadol on days 7- 9; and about 40-400 mg of tramadol on days 10-28 and thereafter.
2. A regimen for the treatment of analgesia according to claim 1 which comprises administering a pharmaceutical composition containing tramadol as the active ingredient which comprises: about 10-50 mg of tramadol on days 1-3; about 20-100 mg of tramadol on days 4- 6; about 30-150 mg of tramadol on days 7-9; and about 40-200 mg of tramadol on days 10-28 and thereafter.
3. A regimen for the treatment of analgesia according to claim 1 which comprises administering a pharmaceutical composition containing tramadol as the active ingredient which comprises: 25 mg of tramadol on days 1-3; 50 mg of tramadol on days 4-6; 75 mg of tramadol on days 7-9; 100 mg of tramadol on days 10-12; 150 mg of tramadol on days 13-15; and 200 mg of tramadol on days 16-28 and thereafter.
4. A regimen for the treatment of analgesia according to claim 1 which comprises administering a pharmaceutical composition containing tramadol as the active ingredient which comprises: 50 mg of tramadol on days 1-3; 100 mg of tramadol on days 4-6; 150 mg of tramadol on days 7-9; and 200 mg of tramadol on day 10 and thereafter.
5. A regimen for the treatment of analgesia according to claim 1 which comprises administering a pharmaceutical composition containing tramadol as the active ingredient which comprises: 25 mg of tramadol on days 1-3; 50 mg of tramadol on days 4-6; 75 mg of tramadol on days 7-9; 100 mg of tramadol on days 10-12; 150 mg of tramadol on days 13-15; and 200 mg of tramadol on day 16 and thereafter.
6. A regimen for the treatment of analgesia according to claim 1 which comprises administering a pharmaceutical composition containing tramadol as the active ingredient which comprises: 25 mg of tramadol q.d. on days 1-3, 25 of mg of tramadol b.i.d. on days 4-
6, 25 mg of tramadol t.i.d. on days 7-9, 25 mg of tramadol q.i.d. on days 10-12, and 50 mg of tramadol t.i.d. on days 13-28.
7. The method of claim 1 wherein tramadol is administered in the form of tramadol hydrochloride.
PCT/US1999/023513 1998-11-02 1999-10-12 Analgesic regimen WO2000025769A1 (en)

Priority Applications (8)

Application Number Priority Date Filing Date Title
AU64224/99A AU763273B2 (en) 1998-11-02 1999-10-12 Analgesic regimen
CA002348907A CA2348907A1 (en) 1998-11-02 1999-10-12 Analgesic regimen
EA200100501A EA003557B1 (en) 1998-11-02 1999-10-12 Analgesic regimen
BR9914981-8A BR9914981A (en) 1998-11-02 1999-10-12 Analgesic regime
EP99951876A EP1126834A1 (en) 1998-11-02 1999-10-12 Analgesic regimen
PL99348808A PL348808A1 (en) 1998-11-02 1999-10-12 Analgesic regimen
NO20012133A NO20012133L (en) 1998-11-02 2001-04-30 Painkillers cure
HR20010311A HRP20010311A2 (en) 1998-11-02 2001-05-02 Analgesic regimen

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US10656798P 1998-11-02 1998-11-02
US60/106,567 1998-11-02

Publications (1)

Publication Number Publication Date
WO2000025769A1 true WO2000025769A1 (en) 2000-05-11

Family

ID=22312128

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/023513 WO2000025769A1 (en) 1998-11-02 1999-10-12 Analgesic regimen

Country Status (13)

Country Link
EP (1) EP1126834A1 (en)
CN (1) CN1346266A (en)
AR (1) AR021054A1 (en)
AU (1) AU763273B2 (en)
BR (1) BR9914981A (en)
CA (1) CA2348907A1 (en)
CZ (1) CZ20011535A3 (en)
EA (1) EA003557B1 (en)
HR (1) HRP20010311A2 (en)
HU (1) HUP0104260A3 (en)
NO (1) NO20012133L (en)
PL (1) PL348808A1 (en)
WO (1) WO2000025769A1 (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004080447A1 (en) * 2003-03-11 2004-09-23 Euro-Celtique, S.A. Titration dosing regimen for controlled release tramadol
EP1576986A2 (en) * 2003-10-10 2005-09-21 Labopharm Inc. Sustained-release tramadol formulations with 24-hour clinical efficacy
EP1905435A3 (en) * 2003-03-11 2008-05-14 Euro-Celtique S.A. Titration dosing regimen for controlled release tramadol
US8962019B2 (en) 2005-09-09 2015-02-24 Angelini Pharma, Inc. Sustained drug release composition
WO2017148595A1 (en) * 2016-02-29 2017-09-08 Grünenthal GmbH Titration of cebranopadol

Non-Patent Citations (7)

* Cited by examiner, † Cited by third party
Title
BARKIN ET AL.: "Alternative dosing for tramadol aids effectiveness", FORMULARY, vol. 30, no. 9, 1995, pages 542 - 543, XP000874395 *
CHERNY N.I.: "Opioid analgesics. Comparative features and prescribing guidelines", DRUGS_(_DRUGS_), 1996, 51/5 (713-737), New Zealand, XP000869772 *
KATZ ET AL.: "The role of tramadol in the management of musculoskeletal pain", TODAY'S THERAP. TRENDS, vol. 13, no. 3, 1995, pages 177 - 186, XP000874368 *
KATZ WA: "Pharmacology and clinical experience with #tramadol# in osteoarthritis.", DRUGS, 1996, 52 SUPPL 3 P39-47, NEW ZEALAND, XP000869774 *
PETRONE D ET AL: "Slowing the titration rate of #tramadol# HCl reduces the incidence of discontinuation due to nausea and/or vomiting: a double-blind randomized trial.", J CLIN PHARM THER, APR 1999, 24 (2) P115-23, ENGLAND, XP000869773 *
RUOFF GE: "Slowing the initial titration rate of #tramadol# improves tolerability.", PHARMACOTHERAPY, JAN 1999, 19 (1) P88-93, UNITED STATES, XP000869656 *
TANIGUCHI G. ET AL: "Criteria for use of #tramadol# hydrochloride in adult inpatients and outpatients", AMERICAN JOURNAL OF HEALTH-SYSTEM PHARMACY_(_AM. J. HEALTH-SYST. PHARM._), 1997, 54/6 (696-697), United States, XP000869770 *

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004080447A1 (en) * 2003-03-11 2004-09-23 Euro-Celtique, S.A. Titration dosing regimen for controlled release tramadol
EP1905435A3 (en) * 2003-03-11 2008-05-14 Euro-Celtique S.A. Titration dosing regimen for controlled release tramadol
EP2258362A3 (en) * 2003-03-11 2011-06-22 Euro-Celtique S.A. Titration dosing regimen for controlled release tramadol
US8722087B2 (en) 2003-03-11 2014-05-13 Purdue Pharma L.P. Titration dosing regimen for controlled release tramadol
EP1576986A2 (en) * 2003-10-10 2005-09-21 Labopharm Inc. Sustained-release tramadol formulations with 24-hour clinical efficacy
EP1576986A3 (en) * 2003-10-10 2006-10-25 Labopharm Inc. Sustained-release tramadol formulations with 24-hour clinical efficacy
US8962019B2 (en) 2005-09-09 2015-02-24 Angelini Pharma, Inc. Sustained drug release composition
US9439866B2 (en) 2005-09-09 2016-09-13 Angelini Pharma, Inc. Trazodone composition for once a day administration
WO2017148595A1 (en) * 2016-02-29 2017-09-08 Grünenthal GmbH Titration of cebranopadol
AU2017226830B2 (en) * 2016-02-29 2021-06-24 Grünenthal GmbH Titration of cebranopadol
US11229625B2 (en) 2016-02-29 2022-01-25 Park Therapeutics, Inc. Titration of Cebranopadol

Also Published As

Publication number Publication date
AR021054A1 (en) 2002-06-12
EA003557B1 (en) 2003-06-26
CZ20011535A3 (en) 2002-08-14
NO20012133D0 (en) 2001-04-30
CA2348907A1 (en) 2000-05-11
AU6422499A (en) 2000-05-22
EA200100501A1 (en) 2001-10-22
BR9914981A (en) 2001-10-30
AU763273B2 (en) 2003-07-17
HUP0104260A2 (en) 2002-08-28
HRP20010311A2 (en) 2002-06-30
PL348808A1 (en) 2002-06-17
HUP0104260A3 (en) 2002-11-28
CN1346266A (en) 2002-04-24
NO20012133L (en) 2001-07-02
EP1126834A1 (en) 2001-08-29

Similar Documents

Publication Publication Date Title
US6339105B1 (en) Analgesic regimen
Gibson Pharmacokinetics, efficacy, and safety of analgesia with a focus on tramadol HCl
JP6097859B2 (en) Pharmaceutical composition comprising dextromethorphan and quinidine for the treatment of neurological diseases
KR101618929B1 (en) A combination of an opioid agonist and an opioid antagonist in the treatment of parkinson&#39;s disease
KR101864559B1 (en) Administration of intravenous ibuprofen
AU763273B2 (en) Analgesic regimen
JPH10505087A (en) Pain relieving composition containing non-narcotic analgesic and painless enhancer
Sorge et al. Comparison of the analgesic efficacy and tolerability of tramadol 100mg sustained-release tablets and tramadol 50mg capsules for the treatment of chronic low back pain
Abel Tramadol: an alternative analgesic to traditional opioids and NSAIDs
EP3328383A1 (en) Antitussive compositions and methods
CN112672743A (en) Treatment of pruritus symptoms of liver disease
US20170087104A1 (en) Medicament
WO2014120021A1 (en) A combination medicament comprising phenylephrine and paracetamol
CN116808038A (en) Combination of opioids and N-acylethanolamines
MXPA01004414A (en) Analgesic regimen
US20040167146A1 (en) Method of treatment
EA011926B1 (en) Oral antidepressant formulation comprising acetylsalicylic acid to accelerate onset of action
Ogata et al. N‐Methyl‐D‐Aspartate (NMDA) Receptor Antagonists
KR20230074469A (en) oral dosage form
US20080125414A1 (en) Method of Treatment
JP2005533046A (en) Use of devazepide in combination with opioid analgesics to enhance the action of analgesics
Analgesic et al. NOTES OF INTEREST
Babul et al. SINGLE-DOSE PHARMACOKINETICS OF ABUSE DETERRENT, EXTENDED RELEASE ONCE-A-DAY DOSAGE FORMS OF LEVORPHANOL: A 15-SUBJECT, 5-WAY CROSSOVER STUDY

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 99814987.X

Country of ref document: CN

ENP Entry into the national phase

Ref document number: 1999 64224

Country of ref document: AU

Kind code of ref document: A

AK Designated states

Kind code of ref document: A1

Designated state(s): AE AL AM AT AU AZ BA BB BG BR BY CA CH CN CR CU CZ DE DK EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG UZ VN YU ZA ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SL SZ TZ UG ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
WWE Wipo information: entry into national phase

Ref document number: 64224/99

Country of ref document: AU

WWE Wipo information: entry into national phase

Ref document number: PV2001-1535

Country of ref document: CZ

Ref document number: 511401

Country of ref document: NZ

ENP Entry into the national phase

Ref document number: 2348907

Country of ref document: CA

Ref document number: 2348907

Country of ref document: CA

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: P20010311A

Country of ref document: HR

Ref document number: PA/a/2001/004414

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 1999951876

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 200100501

Country of ref document: EA

WWP Wipo information: published in national office

Ref document number: 1999951876

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

WWP Wipo information: published in national office

Ref document number: PV2001-1535

Country of ref document: CZ

WWG Wipo information: grant in national office

Ref document number: 64224/99

Country of ref document: AU

WWW Wipo information: withdrawn in national office

Ref document number: 1999951876

Country of ref document: EP