National Cancer Institute
U.S. National Institutes of Health | www.cancer.gov

NCI Home
Cancer Topics
Clinical Trials
Cancer Statistics
Research & Funding
News
About NCI
Pain (PDQ®)
Patient Version   Health Professional Version   En español   Last Modified: 07/10/2008



Purpose of This PDQ Summary






Overview






Pain Assessment






Pharmacologic Management






Physical and Psychosocial Interventions






Antineoplastic Interventions






Invasive Interventions






Discharge Planning






Treating Elderly Patients






Get More Information From NCI






Changes to This Summary (07/10/2008)






Questions or Comments About This Summary






More Information



Page Options
Print This Page
Print Entire Document
View Entire Document
E-Mail This Document
Quick Links
Director's Corner

Dictionary of Cancer Terms

NCI Drug Dictionary

Funding Opportunities

NCI Publications

Advisory Boards and Groups

Science Serving People

Español
NCI Highlights
Virtual and Standard Colonoscopy Both Accurate

New Study of Targeted Therapies for Breast Cancer

The Nation's Investment in Cancer Research FY 2009

Cancer Trends Progress Report: 2007 Update

Past Highlights
You CAN Quit Smoking Now!
Pharmacologic Management

Basic Principles of Cancer Pain Management
Acetaminophen and Nonsteroidal Anti-inflammatory Drugs
Opioids
        Opioid types
        Principles of opioid administration
        Route of administration
        Drugs and routes to be avoided
        Side effects of opioids
Adjuvant Drugs



Basic Principles of Cancer Pain Management

The World Health Organization (WHO) has described a three-step analgesic ladder as a framework for pain management.[1] It involves a stepped approach based on the severity of the pain. If the pain is mild, one may begin by prescribing a Step 1 analgesic such as acetaminophen or a nonsteroidal anti-inflammatory drug (NSAID). Potential adverse effects should be noted, particularly the renal and gastrointestinal adverse effects of the NSAIDs. If pain persists or worsens despite appropriate dose increases, a change to a Step 2 or Step 3 analgesic is indicated. Most patients with cancer pain will require a Step 2 or Step 3 analgesic. Step 1 can be skipped in those patients presenting at the onset with moderate-to-severe pain in favor of Step 2 or Step 3. At each step, an adjuvant drug or modality such as radiation therapy may be considered in selected patients. WHO recommendations are based on worldwide availability of drugs and not strictly on pharmacology.

Analgesics should be given “by mouth, by the clock, by the ladder, and for the individual.”[1] This requires regular scheduling of the analgesic, not just as needed. In addition, rescue-doses for breakthrough pain need to be added. The oral route is preferred as long as a patient is able to swallow. Each analgesic regimen should be adjusted for each patient’s individual circumstances and physical condition.

Acetaminophen and Nonsteroidal Anti-inflammatory Drugs

NSAIDs are effective for relief of mild pain and may have an opioid dose–sparing effect that helps reduce side effects when given with opioids for moderate-to-severe pain. Acetaminophen is included with aspirin and other NSAIDs because it has similar analgesic potency, though it lacks peripheral anti-inflammatory activity.[2] Side effects can occur at any time, and patients who take acetaminophen or NSAIDs, especially elderly patients, should be followed carefully.[3-5] There is growing debate about whether NSAIDs are useful and have significant opioid-sparing effects. One meta-analysis [6] suggests that the usefulness of NSAIDs is limited and that they do not significantly spare opioid doses. Another study suggests that NSAIDs are useful and reduce the need for opioid dose increases; however, only patients with pain progression after 1 week of opioid stabilization were selected for the study.[7]

The coxibs are a subclass of NSAIDs designed to selectively inhibit cyclooxygenase-2 (COX-2).[8] Development of these drugs was based on the hypothesis that COX-2 was the source of prostaglandins E2 and I2, which mediate inflammation, and COX-1 was the source of the same prostaglandins in gastric epithelium, with the potential advantage over traditional NSAIDs of less gastrointestinal ulceration and bleeding and the absence of platelet inhibition. Direct comparisons between COX-2 inhibitors are few. A systematic meta-analysis of COX-2 inhibitors compared with traditional NSAIDs or different COX-2 inhibitors for postoperative pain suggests that rofecoxib, 50 mg, and parecoxib, 40 mg, are equipotent to traditional NSAIDs for postoperative pain after minor and major surgical procedures and have a longer duration of action after dental surgery. Rofecoxib was found to provide superior analgesic effect compared with celecoxib, 200 mg. There were insufficient data to comment on toxicity.[9]

There are three coxibs that were approved by the U.S. Food and Drug Administration (FDA): celecoxib, rofecoxib, and valdecoxib. On September 30, 2004, rofecoxib was withdrawn from the market after a study demonstrated that subjects in a colon cancer prevention trial taking the drug at higher-than-typical doses on a long-term basis had a significant increase in the incidence of serious thromboembolic complications. The question that remains unanswered is whether the increased risk applies to all COX-2 inhibitors, with the caution that the burden of proof rests with those who might claim that this is a problem for rofecoxib alone and does not extend to other coxibs.[8,10] On April 7, 2005, valdecoxib was withdrawn from the market. FDA is also asking manufacturers of all marketed prescription NSAIDs, including celecoxib (Celebrex) to revise the labeling (package insert) for their products to include a boxed warning, highlighting the potential for increased risk of cardiovascular events and/or the serious, potentially life-threatening gastrointestinal bleeding associated with use of these drugs.

Dosage

  • Use patient response to determine the effective dosing interval for aspirin, acetaminophen, and other NSAIDs listed in the Dosing Recommendations for Acetaminophen and NSAIDs table. When pain relief is not attained with the maximum dosage of one NSAID, try other drugs within this category before abandoning NSAID therapy.

Route of administration

  • Use readily available oral tablets, capsules, or liquid. During intervals of nausea and vomiting, use suppositories. Ketorolac tromethamine is the only NSAID available for parenteral use.

Contraindications

  • Patients taking NSAIDs are at risk for platelet dysfunction that may impair blood clotting. The table below lists NSAIDs with minimal antiplatelet activity.

Other side effects

  • Follow patients carefully for adverse effects, which range from mild gastrointestinal discomfort to more serious problems including the following:
    • Gastric ulceration.
    • Hepatic dysfunction.
    • Myocardial infarction.
    • Renal failure.

    Because both NSAIDs and other drugs (e.g., warfarin, methotrexate, digoxin, cyclosporine, oral antidiabetic agents, and sulfonamide-containing drugs) are highly protein-bound, there is potential for altered efficacy or toxicity when they are given simultaneously.

Dosing Recommendations for Acetaminophen and NSAIDs
Drug  Usual Dose for Adults and Children ≥50 kg Body Weight  Usual Dose for Adults and Children1 <50 kg Body Weight 
Orally Administered Acetaminophen and Over-the-counter NSAIDs
acetaminophen2 650 mg q 4 h 10–15 mg/kg q 4 h
975 mg q 6 h 15–20 mg/kg q 4 h (rectal)
aspirin3 650 mg q 4 h 10–15 mg/kg q 4 h
975 mg q 6 h 15–20 mg/kg q 4 h (rectal)
ibuprofen (Motrin, Advil) 400–600 mg q 6 h 5–10 mg/kg q 4–6 h
magnesium salicylate (Doan’s, Magan, Mobidin, others) 650 mg q 4 h
naproxen (Naprosyn, Aleve) 250–275 mg q 6–8 h 5 mg/kg q 8 h
naproxen sodium (Anaprox) 275 mg q 6–8 h
Prescription NSAIDs
carprofen (Rimadyl) 100 mg tid
choline magnesium trisalicylate4 (Trilisate) 1,000–1,500 mg q 6–8 h 25 mg/kg q 6–8 h
choline salicylate4 (Arthropan) 870 mg q 3–4 h
diflunisal5 (Dolobid) 500 mg q 12 h
etodolac (Lodine) 200–400 mg q 6–8 h
fenoprofen calcium (Nalfon) 300–600 mg q 6 h
ketoprofen (Orudis) 25–60 mg q 6–8 h
ketorolac tromethamine6 (Toradol) 10 mg q 4–6 h to a maximum of 40 mg/day
IV administration should not exceed 5 days
meclofenamate sodium7 (Meclomen) 50–100 mg q 6 h
mefenamic acid (Ponstel) 250 mg q 6 h
sodium salicylate (Anacin, Bufferin) 325–650 mg q 3–4 h
Parenteral NSAIDs
ketorolac tromethamine6,8 (Toradol) 60 mg initially, then 30 mg q 6 h
IV administration should not exceed 5 days

IV = intravenous; q = every.
1. Acetaminophen and NSAID dosages for adults weighing less than 50 kg should be adjusted for weight.
2. Acetaminophen lacks the peripheral anti-inflammatory and antiplatelet activities of the other NSAIDs.
3. The standard against which other NSAIDs are compared. May inhibit platelet aggregation for longer than 1 week and may cause bleeding. Aspirin is not recommended for pain in children.
4. May have minimal antiplatelet activity.
5. Administration with antacids may decrease absorption.
6. Use limited to 5 days or fewer.
7. Coombs-positive autoimmune hemolytic anemia has been associated with prolonged use.
8. Has the same gastrointestinal toxic effects as oral NSAIDs.
 [Note: Only the above NSAIDs have FDA approval for use as simple analgesics, but clinical experience has been gained with other drugs as well.]

Opioids

Opioids, the major class of analgesics used in management of moderate-to-severe pain, are effective, are easily titrated, and have a favorable benefit-to-risk ratio.

The predictable consequences of long-term opioid administration—tolerance and physical dependence—are often confused with psychological dependence (addiction) that manifests as drug abuse. This misunderstanding can lead to ineffective prescribing, administering, or dispensing of opioids for cancer pain. The result is undertreatment of pain.[11]

Clinicians may be reluctant to give high doses of opioids to patients with advanced disease because of a fear of respiratory depression. Many patients with cancer pain become opioid tolerant during long-term opioid therapy. Therefore, the clinician’s fear of shortening life by increasing opioid doses is usually unfounded.

Opioid types

Opioids are classified as full morphine-like agonists, partial agonists, or mixed agonist-antagonists, depending on the specific receptors to which they bind and their activity at these receptors. The benefits of using opioids and the risks associated with their use vary among individuals.

Morphine is the most commonly used opioid in cancer pain management, largely for reasons of availability and familiarity;[12] however, it is useful to be familiar with more than one type of opioid. Wide interindividual variability in response to both the analgesic and adverse effects of opioids is recognized.[13] Some patients may not experience adequate pain control despite appropriate dose adjustments, while others may develop intolerable adverse effects to one particular opioid (see below). Alternative opioids include hydromorphone, oxycodone, methadone, and fentanyl. Knowledge of several medications and formulations give the caregiver much more flexibility in tailoring a regime to a particular patient’s needs.

Short-acting opioids are generally recommended when opioid therapy is being initiated for the first time or when patients are medically unstable or the pain intensity is highly variable. Once stable, patients can be switched to a controlled-release or slow-release formulation. This is more convenient and promotes compliance. (Refer to the Approximate Dose Equivalents for Opioid Analgesics table in the Principles of Opioid Administration section of this summary.)

Full agonists

  • Morphine, hydromorphone, codeine, oxycodone, hydrocodone, methadone, levorphanol, and fentanyl are classified as full agonists because their effectiveness with increasing doses is not limited by a ceiling. Full agonists will not reverse or antagonize the effects of other full agonists given simultaneously.


Morphine

  • The most commonly used opioid, morphine, is readily available in several forms, including sustained-release (8–24 hours duration of effectiveness) formulations for oral administration.


Other agonists

  • For the patient who experiences dose-limiting side effects with one oral opioid (e.g., hallucinations, nightmares, dysphoria, nausea, or mental clouding), other oral opioids should be tried before abandoning one route in favor of another.


Methadone

  • Methadone has had a revival in interest for the management of cancer pain. Published reports have been in the form of case reports,[14-20] outcome surveys,[21-25] and reviews.[26-28]Success has been reported with oral, intravenous (IV), and suppository methadone use. Subcutaneous methadone has been reported to cause tissue irritation at the injection site but has been used effectively in some patients without clinically significant local toxicity.[29]

    Methadone is a synthetic opioid agonist that has been reported to have a number of unique characteristics. These include excellent oral and rectal absorption, no known active metabolites, prolonged duration of action resulting in longer administration intervals, and lower cost than other opioids. Methadone is available as a pill, an elixir, and for parenteral use. Methadone has an average oral bioavailability of approximately 80% (range, 41%–99%).[30]

    Morphine is the international gold standard for first-line treatment of cancer pain. Methadone, however, can be considerably less expensive than existing rapid-release or sustained-release morphine or other opioid options. A randomized trial of 103 patients compared the effectiveness and side effects of morphine and methadone as first-line treatments for cancer pain. The outcome of successful pain management was similar for both groups; however, there were significantly more opioid-related dropouts in the methadone group. This study did not demonstrate superior analgesic effectiveness or overall tolerability of methadone over morphine as a first-line treatment for cancer pain. Despite this finding, the authors of this report suggested that study limitations did not allow definitive conclusions that methadone could not be a useful first-line opioid. Further research exploring other doses and schedules of methadone should still be explored.[31]

    Because of its long and unpredictable half-life and relatively unknown equianalgesic dose as compared with other opioids, methadone has been generally used by pain specialists with experience in its use. The utility of methadone in cancer pain and difficult cancer pain syndromes such as neuropathic pain has become more widely appreciated and has gained increasing acceptance for use in hospital and hospice settings and by clinicians who are not pain specialists.[32] The methadone preparation widely used in the United States is a racemic mix of the d-isomer and l-isomer of methadone. The d-isomer has antagonist activity at the N-methyl-D-aspartate (NMDA) receptor and may be beneficial in controlling neuropathic pain.

    Another controversy related to methadone concerns possible prolongation of QTc interval, leading to torsades de pointes and ventricular arrhythmia. A number of studies have raised the concern that methadone may be associated with prolonged QT interval and may lead to torsades de pointes. Several retrospective case reports suggest that parenteral methadone or oral methadone in high doses could be associated with this adverse effect.[33-36] Chlorobutanol has been implicated; although this substance is present in parenteral solutions, it is not found in oral formulations.[37] Another series of 132 patients taking methadone revealed statistically significant mean increases in QTc of 10.2 to 13.2 milliseconds, yet no episodes of torsades de pointes were reported.[36] This result raises the issue of the clinical significance of this effect. In another retrospective review of 520 patients treated with methadone for cancer pain, no change in QTc was seen in the 56 patients who had electrocardiograms 3 months before and after starting methadone.[38,39] Avoidance of concomitant medications that prolong QT interval [37] or that share common metabolism pathways with methadone [38] is recommended. In high-risk situations, clinicians could consider electrocardiogram monitoring and other clinical precautions such as correcting electrolyte abnormalities.

    When converting from another opioid to methadone, the calculated equianalgesic dose ratio of methadone varies depending on the oral morphine-equivalent daily dose (MEDD) of the previous opioid.[40,22] One guideline for choosing an appropriate initial dose of methadone based on the oral MEDD of the previous opioid is shown in the table below. For example, a patient who has been using sustained-release morphine at 80 mg every 8 hours (240 mg/day) would be appropriately switched to methadone at a dose of 10 mg every 8 hours (30 mg/day, an 8:1 conversion ratio). In contrast, a patient who is taking sustained-release morphine at a total daily dose of 60 mg/day might be switched to an oral methadone dose of 5 mg every 8 hours (15 mg/day, a 4:1 conversion ratio).

    Method 1: Initial Methadone Dose Based on Oral MEDD*
    Oral MEDD (mg/d)  Initial Dose Ratio (oral morphine:oral methadone) 
    <30 2:1
    30–99 4:1
    100–299 8:1
    300–499 12:1
    500–999 15:1
    >1,000 20:1 or greater**

    *Reprinted with permission from Fisch and Cleeland [41]
    **Great caution must be used when converting to methadone when very high opioid doses have been used. Often, only a portion of the total opioid dose is converted initially, with further conversions taking place over several days to weeks.

    To be conservative, one might estimate that methadone is roughly twice as potent when administered via IV versus oral administration. Thus, a patient with well-controlled pain on a stable oral methadone dose of 10 mg every 8 hours might be given IV methadone at an initial dose of 5 mg every 8 hours if IV use is necessary. Subcutaneous use of methadone may cause skin irritation in some patients but has been used successfully.

    In addition to the method described in the table above, several methods of switching to methadone have been proposed.[42,22,43-46] Some rely on patient-controlled analgesia with fixed doses and flexible intervals, some require fixed intervals and fixed doses, while others stagger the conversion over a few days. Whatever method is chosen, this kind of switch can be safe and effective as long as regular assessments are provided over time, and there is an appreciation of the equianalgesic dose ratio of methadone to morphine in opioid-tolerant patients.

    • Method 2: Staggered or 3-day Switchover

      One approach calls for a gradual switch over 3 to 5 days to decrease the risk of relative overdosing. An equianalgesic dose of methadone is first calculated, using an equianalgesic dose ratio of morphine to methadone of 10:1 (i.e., methadone being approximately ten times more potent than morphine). The caveat in using a ratio of 10:1 is that variations in ratios have been noted, depending on the dose of the previous opioid. The ratio may be much higher (12:1 or even higher) in patients being switched from high doses of morphine to methadone. The following example is given to illustrate this method: A patient who is on the equivalent of 450 mg per day of oral morphine (quick-release morphine 75 mg orally every 4 hours) needs to be switched to methadone. Using a ratio of 10:1, the predicted equivalent daily oral dose of methadone, once the switch is completed, will be 45 mg. On day 1 of the switch, the daily morphine dose is reduced by one third to approximately 300 mg per day (morphine 50 mg orally every 4 hours) and one third of the predicted daily methadone dose is added, divided into three doses per 24 hours (i.e., methadone 5 mg orally every 8 hours). Morphine continues to be given for rescue doses. On day 2 of the switch the patient is reassessed, and if no problems have developed, the morphine dose is reduced by another third (i.e., morphine 25 mg orally every 4 hours) and the methadone dose is increased by another third (i.e., methadone 10 mg orally every 8 hours). On day 3, the patient is reassessed. If there are complications such as significant somnolence, but the pain is still not under good control, the methadone dose is increased to 15 mg every 8 hours and the morphine is discontinued. On day 3, methadone or a short half-life opioid is added as a rescue dose as needed. The rescue dose is calculated at 5% to 15% of the total daily dose. On day 3, if the patient has good pain control but shows signs of relative overdosing such as significant somnolence, the morphine is discontinued without any increase in the methadone dose (i.e., it remains at the day 2 level or may even be decreased if needed).



    • Method 3: Ad Libitum [42]

      This approach calls for the previous dose to be discontinued and a single fixed-dose of methadone to be given at the start, calculated using an equianalgesic dose ratio of morphine to methadone of 10:1 (i.e., morphine 10 mg being roughly equivalent to 1 mg of methadone), but to a maximum of 50 mg of methadone per dose. After the initial single priming dose, the same dose is administered every 3 hours as needed. The clinician observes the patient and when the demand for rescue doses reduces or stabilizes (indicating steady-state being reached), which is usually on day 4 to 7, the daily requirement is calculated and the dose is given every 8 to 12 hours.



    • Method 4: Initial Priming Followed By Variable Conversion [43]

      In this method, an opioid-naïve patient is started on 3 to 5 mg of methadone every 8 hours and a nonnaïve patient is started on a dose of methadone that is equivalent to 50% of the estimated daily morphine dose. These doses are initially given for 3 days. Once the patient has acceptable pain relief for 6 to 8 hours, the dose is changed to a single fixed-dose once a day and rescue doses are given as needed. This method is probably best suited for opioid-naïve patients (in relatively unlikely situations where more frequently used opioids such as morphine are not available) or patients who are, for one reason or another, being switched from relatively low doses of morphine or other opioids.



    • Method 5: German Model [45]

      This method is suggested when patients are being switched from high equivalent daily doses of morphine (>600 mg orally per day). The morphine or other opioid the patient is receiving is stopped. Methadone at a dose of 5 to 10 mg orally is started every 4 hours and rescue-doses of 5 to 10 mg every hour are allowed as needed. On the second to third days of the switch, the methadone dose is increased by up to 30% every 4 hours until sufficient pain relief is achieved and no significant adverse effects are noted. After exactly 72 hours following the switch to methadone, the dose is changed from every 4 hours to every 8 hours, and the interval of rescue doses is increased to every 3 hours as needed at the same single dose as established on days 2 to 3. The dose can then be increased by up to 30% if further upward titration is required.



    In some countries there are restrictions that do not apply to other opioids on the ability of physicians to prescribe methadone. In the United States, this pertains to methadone for maintenance of addiction. Methadone is not restricted when used for pain management; however, physicians should carefully document the use of methadone.[47] It should be noted that ratios are different for switching from methadone to a morphine-like opioid.[22]



Meperidine (Demerol)

  • Useful for brief courses (a few days) to treat acute pain, meperidine is not recommended in treating persistent cancer pain because of its short duration of action (2.5–3.5 hours) and its neurotoxic metabolite, normeperidine. Accumulation of this metabolite, particularly when renal function is impaired, causes central nervous system (CNS) stimulation that may lead to delirium or seizures. Seizures are typically preceded by development of multifocal myoclonus, which can be used as a warning sign.

Tramadol

  • Tramadol can be considered an atypical opioid analgesic that has a dual action. It is a weak mu opioid agonist that also inhibits the reuptake of norepinephrine and serotonin.[48,49] It is believed that both mechanisms work synergistically to provide analgesic benefit with a potency that is approximately one-tenth that of morphine [50] and approximately equivalent to codeine. The most common side effects reported with tramadol are drowsiness, constipation, dizziness, nausea, and orthostatic hypotension.[48] There is also a risk of precipitating seizures in patients with a previous history or in patients who are receiving medications that could reduce the seizure threshold. The use of other serotonergic medications (e.g., selective serotonin reuptake inhibitors [SSRIs] and serotonin-norepinephrine reuptake inhibitors [SNRIs]) together with tramadol has the potential to increase the risk of the serotonin syndrome. Tramadol is available in short- and long-acting formulations and in fixed combination with acetaminophen. The recommended starting dose of oral tramadol is 50 mg 1 or 2 times a day, with gradual titration up to a maximum of 400 mg per day.[48] There is also the option of using tramadol via the rectal or subcutaneous route in patients who are unable to tolerate oral medication.[51,52]


Partial agonists

  • Partial agonists such as buprenorphine have less effect than full agonists at opioid receptors. They are subject to a ceiling effect and thus are less effective analgesics.


Mixed agonist-antagonists

  • Mixed agonist-antagonists block or are neutral at one type of opioid receptor while activating a different opioid receptor. Mixed agonist-antagonists are contraindicated for use in the patient receiving an opioid agonist because they may precipitate a withdrawal syndrome and increase pain. Mixed agonist-antagonists include pentazocine (Talwin), butorphanol tartrate (Stadol), dezocine (Dalgan), and nalbuphine hydrochloride (Nubain). Their analgesic effectiveness is limited by a dose-related ceiling effect.


Principles of opioid administration

Most patients with cancer pain require fixed-schedule dosing to manage the constant pain and prevent the pain from worsening.[53] An Italian study of patients whose baseline pain was well controlled on morphine when admitted to a palliative care unit found that most episodes of breakthrough pain were rapidly controlled with IV morphine equivalent to 20% of the calculated equianalgesic total daily dose. Adverse effects were uncommon.[54] An as-needed rescue dose (breakthrough dose) should be combined with the regular fixed-schedule opioid to control the episodic exacerbation of pain, often referred to as breakthrough pain. When this pain is elicited by an action such as weight-bearing, breathing, or defecation, it is termed incident pain. Rescue or breakthrough doses can be given hourly or more frequently as needed, depending on route of administration, pharmacokinetic properties of the drug, and presence or absence of side effects. The breakthrough dose is generally calculated to be 10% to 20% of the total dose of the fixed schedule.[55] Adherence rates are improved when patients are prescribed around-the-clock opioids compared with as-needed prescribing.[56] Preliminary data suggest that the intensity of incident pain related to bone metastases may be diminished by increasing the dose of the scheduled opioid above that needed for control of baseline pain, while maintaining it below that associated with the development of limiting side effects.[57]

Dosage

  • The appropriate dosing interval is determined by the opioid and formulation used. The analgesic effects of short-acting oral opioids such as morphine, hydromorphone, codeine, and oxycodone begin within a half hour after administration and last for approximately 4 hours. The dosing interval of these drugs is usually 4 hours. In patients given controlled-release formulations of morphine, hydromorphone, codeine, or oxycodone, relief should begin in 1 hour, peak in 2 to 3 hours, and last for 12 hours (controlled-release hydromorphone and codeine are not available in the United States); these formulations are usually prescribed in 12-hour intervals. A small group of patients, however (10%–20% of those on 12-hour controlled-release formulations), may require administration every 8 hours. The analgesic effect of transdermal fentanyl begins approximately 12 hours after the application of the patch, peaks in 24 to 48 hours, and lasts for approximately 72 hours. Patches are therefore changed every 72 hours. In a select group of patients who consistently experience end-of-dose failure despite increases in the patch doses, the dosing interval can be increased to every 48 hours (<10% of patients on fentanyl patches). Transdermal fentanyl is not recommended for control of acute pain or poorly controlled pain because there is a delayed onset of action until reaching steady-state either with new use or with a change in the dose. Patients receiving transdermal fentanyl may be switched to a continuous IV or subcutaneous infusion of fentanyl using a conversion ratio of 1:1 to facilitate more rapid titration.[58]

Dose titration

  • To date, dose titration is largely patient-driven, as determined by the balance of analgesia with side effects. For example, while morphine dose correlates with peak-and-trough plasma concentrations of a parent drug and its metabolites morphine-3-glucuronide and morphine-6-glucuronide, studies are conflicting with regard to the association between plasma levels of morphine and its metabolites versus analgesia as measured by pain scores.[59] The strong opioid agonists have no maximum dose or ceiling dose. The appropriate dose is the amount of opioid that controls pain with the fewest side effects. Dose titration should continue until good pain relief is achieved or intolerable side effects develop that cannot otherwise be controlled. The goal is to achieve a favorable balance between analgesia and side effects through gradual adjustment of the dose. If analgesic tolerance appears to be occurring, the dose can be increased or consideration given to switching the opioid, especially if higher doses are required.

    The severity of the pain and the opioid formulation chosen determine the rate of titration. The dose of immediate-release formulations can be increased on a daily basis if necessary until pain relief is adequate. Among patients receiving relatively low doses of opioids, those with uncontrolled moderate-intensity pain require daily increases of between 25% and 50% to their previous dose, while patients with severe uncontrolled pain may require a higher increase. At higher opioid doses, increases of 20% to 30% would be more prudent. Rapid dose escalation requires close monitoring for both efficacy and side effects. Preliminary data suggest that titration with sustained-release daily morphine is equivalent to titration with immediate-release morphine administered every 4 hours by an expert group of clinicians, but standard practice is to use a short-acting opioid for initial titration.[60]

    Occasionally, doses may need to be reduced or, rarely, stopped. This may occur when patients become pain free as a result of cancer treatment, including treatments such as nerve blocks and radiation therapy. Another time to consider reducing the dose is when a patient experiences significant opioid-related sedation that is accompanied by good pain control. In situations where interventions achieve complete pain relief, rapid opioid tapering rather than abrupt discontinuation is recommended and usually adequate.



Different types of opioids

  • The debate regarding whether any individual opioid causes fewer side effects or is more effective is characterized by much speculation but little clinical evidence. These inconclusive findings have prompted expert working groups of the European Association of Palliative Care to recommend that there is currently little evidence of the clinical superiority of one opioid over another regarding the side-effect profile and/or analgesia.[12,13] Even constipation and other side effects may be positively affected by a switch. Compared with morphine, fentanyl may cause less constipation.[61,62] Studies suggesting that oxycodone and hydromorphone may cause less nausea and hallucinations than morphine [63] are juxtaposed with other studies that found no significant differences between them.[64-66] One study found that transdermal fentanyl was better tolerated than sustained-release oral morphine and equally effective.[67]

Tolerance

  • Assume that patients actively abusing heroin or prescription opioids (including methadone) have some pharmacologic tolerance that will require higher starting doses and shorter dosing intervals.

Opioid therapy in special populations

  • Health professionals should check current recommendations for opioid use in older people, children, people who are cognitively impaired, and known or suspected drug abusers.

Opioid switching

A series of case reports have demonstrated the clinical problem of inadequate pain control with escalating opioid doses in the presence of dose-limiting toxic effects, including hallucinations, confusion, hyperalgesia, myoclonus, sedation, and nausea.[17,23,68-70] It was suggested that these problems could be managed by switching to an alternative opioid, with the result being improved pain management and decreased toxic effects. The improvement with opioid switching, although predominantly demonstrated initially with morphine, has also been reported with other opioids.[71-74] A retrospective review over a 1-year period in a pediatric oncology center supports efficacy of this technique in children, with resolution of adverse opioid effects, largely pruritus, achieved in 90% of patients, while maintaining pain control.[75]

  • Guidelines for switching from one opioid to another

    Guidelines for opioid switching are intended to reduce the risk of relative overdosing or underdosing as one opioid is replaced by another. These guidelines require a working knowledge of an equianalgesic-dose table.[13,76] The equianalgesic-dose table provides only a broad guide for dose selection when switching from one opioid to another. Wide ranges in interindividual responses to the various opioids have been noted.[76] Therefore, because of incomplete cross-tolerance in most cases, the calculated dose-equivalent of a new drug must be reduced by 25% to 50% to ensure safety. These figures are based on clinical experience rather than empiric data. The selection of an alternative opioid is largely empirical. There is little clinical evidence to indicate that one opioid has therapeutic superiority over another opioid. A patient, for example, who requires a switch from morphine to another opioid can be switched to hydromorphone, oxycodone, fentanyl, or methadone.[77-79] In one prospective study of 186 cancer patients being treated with morphine, 25% did not respond and required switching to another opioid (oxycodone). The primary reasons for switching included pain, confusion, drowsiness, nightmares, and nausea. Of the 47 patients who required switching to an alternative opioid, 37 (79%) obtained good relief. This result provides beginning evidence for the prevalence of the need to switch, as well as determining the success rate once switching occurs.[80] Patients should be followed closely after a switch and should be reassessed, and the new opioid dose should be adjusted according to the intensity of pain and lack or presence of adverse effects.



Note: The values that appear in the table below are NOT recommended starting doses. Opioid doses are highly variable and should be based on the individual’s previous responses and overall condition. Important cautions are contained in the footnotes.

Approximate Dose Equivalents for Opioid Analgesics1
Drug  Oral Dose (mg)  Parenteral Dose2 
Morphine 3 30 10 mg
Codeine4 200 100 mg
Fentanyl5a,b NA 100 μg
Hydrocodone (Vicodin) 30–45 NA
Hydromorphone (Dilaudid)3 8 2 mg
Levorphanol (Levo-Dromoran) 4 2 mg
Methadone6,7 The conversion ratio of methadone is variable. Please refer to the Opioid Types section and Opioid switching section.
Oxycodone (OxyContin)4 20–30 10–15 mg
Oxymorphone
(Opana, Opana ER, and Opana IV3) 10 1 mg

IV = intravenous; NA = not available.
1. Published tables vary in the suggested doses that are equianalgesic to morphine. Many of these doses are based on clinical consensus rather than well-controlled trials. Clinical response is the criterion that must be applied for each patient; titration to clinical response is necessary. Because there is not complete cross-tolerance among these drugs, it is usually necessary to use a lower-than-equianalgesic dose when changing drugs and retitrate according to response.
2. Parenteral dosing includes IV and subcutaneous administration. Onset and duration may vary slightly between these routes; however, doses remain approximately equal. The intramuscular route is not recommended because of variability in uptake of the drug and painful injection.
3. Caution: For morphine, hydromorphone, and oxymorphone, rectal administration is an alternate route for patients unable to take oral medications. Equianalgesic doses may differ from oral to parenteral doses because of pharmacokinetic differences. Note: A short-acting opioid should normally be used for initial therapy of moderate-to-severe pain.
4. Caution: Doses of aspirin and acetaminophen in combination opioid/NSAID preparations must be adjusted to the patient’s body weight.
5a. Transdermal fentanyl is an alternative. Transdermal fentanyl dosage is not calculated as equianalgesic to a single morphine dosage but is calculated based on a 24-hour opioid dose. See package insert for dosing calculations. Transdermal fentanyl should not be used in opioid-naive patients. 5b. Transmucosal and buccal fentanyl are also available and indicated for breakthrough pain, although they are not bioequivalent. Titration of either should be conducted gradually; neither should be used in opioid-naive patients.
6. Caution: Methadone is much more potent than indicated in older published literature. On average, it is ten times more potent than morphine. However, its potency relative to morphine is not linear. When morphine at lower doses (e.g., 30–60 mg/d orally) is switched to methadone, the potency may be 3 to 5 times; when switched from high doses (e.g., >300 mg/d orally), the potency may be 12 times or even higher.
7. Caution: The oral to IV dose ratio of methadone is not well established. The IV route is very seldom used, except in cancer centers with pain service familiar with parenteral methadone. Intravenous use of methadone in combination with chlorobutanol is associated with QTc wave prolongation.[37] Subcutaneous administration may cause irritation.

It has been suggested that a less complicated approach than opioid switching would be reassessment of the clinical situation and use of adjuvant analgesics, decreasing the opioid dose if possible, use of medical management for opioid-related side effects, and correction of any contributing metabolic abnormalities.[81,82] Nevertheless, there does appear to be an emerging consensus that opioid switching does have a useful role when pain control remains inadequate with escalating opioid doses and opioid use results in unacceptable opioid-related side effects.[81-83]

Morphine, as the strong opioid of choice for the management of cancer pain, was used increasingly during the 1970s and 1980s.[84] Associated with this increasing experience was the clinical observation of the risk of accumulation of morphine metabolites, particularly in the presence of renal impairment. Morphine-6-glucuronide, an analgesic metabolite, was recognized as having a useful role in enhancing analgesia. A number of reports, however, have described seizures, cognitive impairment, nausea, and problems of myoclonus that were associated with accumulation of morphine-6-glucuronide.[84-91]

The potential role of morphine metabolites, in particular the ratio of 3-glucuronide to 6-glucuronide in the development of opioid-related toxicity, has been reported. The literature on this issue has been somewhat controversial. There is no disagreement that morphine metabolites increase in the presence of deteriorating renal function; however, there has been conflicting evidence regarding the role and ratios of the metabolites in patients exhibiting both a poor response to increasing morphine doses and associated toxicity.[92-96]

Switching from one opioid to another requires familiarity with a range of opioids and the use of opioid dose-conversion tables.[13,76] When using these ratios, it must be understood that the guidelines should be reviewed and the patients should be monitored more closely during the switching phase. A recent review has highlighted some important issues related to these tables.[76] Wide ranges in ratios are noted. In the case of methadone, it is much more potent than previously thought (on average ten times more potent), and its equianalgesic dose-ratio compared to other opioids changes according to the dose of the previous opioid; the higher the dose, the higher the ratio. (Note that potency does not denote more effectiveness but denotes the equivalent dose required to obtain the same effect.)

Route of administration

Oral administration is preferred in patients with intact gastrointestinal tracts because it is convenient and usually inexpensive. When patients cannot take oral medications, other less invasive routes (e.g., rectal or transdermal) should be offered. Parenteral methods should be used only when simpler, less demanding, and less costly methods are inappropriate, ineffective, or unacceptable to the patient. In general, assessing the patient’s response to several different oral opioids is advisable before abandoning the oral route in favor of anesthetic, neurosurgical, or other invasive approaches.

Rectal

  • Use this safe, inexpensive, effective route for delivery of opioids as well as nonopioids when patients have nausea or vomiting. Rectal administration is inappropriate for the patient who has diarrhea, anal/rectal lesions, mucositis, thrombocytopenia, or neutropenia. The use of suppositories is not always culturally acceptable and may not be practical for patients who are obese, have fractures, are physically unable to place the suppository in the rectum, or prefer other routes. When changing from the oral to the rectal route, begin with the same dosage as had been given orally, then titrate as needed.

Transdermal (fentanyl)

  • Patches currently available are formulated to provide analgesia lasting up to 72 hours. This preparation is not suitable for rapid dose titration and should be used for relatively stable analgesic requirements when rapid increases or decreases in dosage are not likely to be needed.[97,98] In the chronic setting, considerable inter- and intraindividual variability may exist in the rate of absorption of fentanyl from transdermal patches in patients receiving a stable dose of transdermal fentanyl.[99] Based on a case series, it has been proposed that conversion from transdermal to IV fentanyl using a 1:1 conversion ratio can be safe and effective during acute exacerbations of cancer pain.[58] Although other opioids are sometimes compounded into gel form for transdermal application, there is insufficient evidence to support this practice.

Transmucosal/Buccal (fentanyl)

  • Oral transmucosal fentanyl citrate is used for the relief of breakthrough pain. The lipid solubility of fentanyl allows rapid onset of pain relief. In open-label studies, 72% to 92% of patients found a dose that provided relief from breakthrough pain. Side effects in these studies were consistent with other opioid therapies, including sedation, constipation, stomatitis, and nausea.[100,101] There is growing interest in the use of rapidly acting, highly lipophilic opioids such as fentanyl for the management of difficult breakthrough pain syndromes. An oral transmucosal fentanyl citrate compound for buccal administration has become available for this purpose.[102,103] A double-blind, randomized, placebo-controlled study included 77 patients assigned to dose sequences of fentanyl buccal tablets (FBT). Results demonstrated that FBT was efficacious and safe in treating cancer-related breakthrough pain.[104] Other opioids such as morphine, hydromorphone, and oxycodone are not very lipophilic and therefore not suited for buccal or sublingual administration. In the home setting, opioids are sometimes administered buccally or sublingually with erratic absorption that is likely via the lower gastrointestinal tract.

Parenteral: IV and subcutaneous

  • IV administration provides a rapid onset of analgesia within 2 to 10 minutes. The duration of action after a bolus dose may be shorter than with other routes. This route may be useful if a patient cannot swallow and IV access is established.


  • The subcutaneous route is as effective as the IV route.[12,105] In some situations, it may even be more convenient, especially if patients are being cared for at home or in a hospice. To facilitate administration via this route, a 25- or 27-gauge butterfly needle can be inserted subcutaneously and left in place for up to 7 days at a time. The anterior thighs, abdomen, upper arms, subclavicular area, and upper back are possible areas for needle insertion. The site should be monitored for signs of infection or irritation and should be changed if these are noted.


  • The bioavailability of parenterally administered opioids (morphine, hydromorphone, oxycodone, and codeine) is generally 2 to 3 times that of the oral route. The dose therefore needs to be halved or decreased by a third when switching from the oral to the subcutaneous and IV routes, respectively (refer to the Approximate Dose Equivalents for Opioid Analgesics table). Opioids administered parenterally may be given either intermittently (usually every 4 hours) or by a continuous infusion. With some exceptions, these two methods appear to be similarly effective.


Other routes

  • Some studies suggest that the use of inhaled opioids for the management of pain and cancer-related shortness of breath are, with some exceptions, not more effective than systemic administration.[106,107] Their absorption via this route is unpredictable.


  • The intramuscular administration of opioids is not recommended.


Patient-Controlled Analgesia

  • Patient-controlled analgesia (PCA) may be used to determine the opioid dose needs when initiating opioid therapy. Once the pain is well controlled, a regular opioid dose can be instituted on the basis of the PCA doses required. This method is contraindicated in patients with cognitive impairment or patients with significant psychological undercurrents to their pain experience.

Intraspinal

  • The intraspinal administration of opioids (epidural or intrathecal), especially when combined with a local anesthetic, can be helpful in a very small select group of patients with intractable pain. Use of the epidural or intrathecal route requires skill and expertise that may not be available in all settings. The table below presents the advantages and disadvantages of intraspinal administration. Although intrathecal opioid therapy has been FDA approved since 1991, the utility of an implantable drug delivery system (IDDS) to deliver spinal opioids was only recently compared with comprehensive medical management (CMM) (based on the Agency for Health Care Policy and Research 1994 cancer pain management guidelines) in a randomized trial. There were 202 patients enrolled in this unblinded study. Of the 101 patients randomized to the IDDS, 51 actually received this therapy. Sixteen of these patients (31%) had serious adverse effects. Patients using the IDDS experienced more than 20% reduction in both pain and opioid toxicity more often than the CMM group (P = .02). These data and further analysis in follow-up reports [108,109] suggest that the use of an IDDS delivery system may offer benefit for some cancer patients. More research is needed to determine which subsets of patients will benefit the most from this device, and what the proper timing should be for a trial of intrathecal opioids.[110,111] An open-label study demonstrated that patients with refractory cancer pain experienced better pain relief, fewer opioid-associated side effects, and decreased systemic opioid use when managed with patient-activated intrathecal delivery of morphine via an implanted delivery system. The device was implanted in 119 patients. There were 7 serious adverse events related to the device and 55 serious adverse events related to the implant and delivery-system refill procedures. The FDA denied the application for market approval of this system.[78]
Advantages and Disadvantages of Intraspinal Drug Administration
System   Advantages   Disadvantages 
Percutaneous temporary catheter Used extensively both intraoperatively and postoperatively. Mechanical problems include catheter dislodgment, kinking, or migration.
Useful when prognosis is limited (<1 month). Increased risk of infection.
Permanent silicone-rubber epidural Catheter implantation is a minor procedure.
Dislodgment and infection less common than with temporary catheters.
Can deliver bolus injections, continuous infusions, or PCA (with or without continuous delivery).
Subcutaneous implanted injection port Increased stability, less risk of dislodgment. Implantation more invasive than external catheters.
Can deliver bolus injections or continuous infusions (with or without PCA). Approved only for epidural catheter in United States.
Potential for infection increases with frequent injections.
Subcutaneous reservoir Potentially reduced infection in comparison with external system. Difficult to access, and fibrosis may occur after repeated injection.
Implanted pumps (continuous and programmable) Potentially decreased risk of infection. Need for more extensive operative procedure.
Need for specialized equipment with programmable systems.

Drugs and routes to be avoided

The following two tables present data on drugs and routes of administration not recommended for the management of cancer pain.

Drugs To Be Avoided for Treatment of Cancer Pain
Class  Drug  Rationale for NOT Recommending 
Opioids meperidine (Demerol) Short duration (2–3 h) of analgesia.
Repeated administration may lead to CNS toxicity (tremor, confusion, or seizures).
Opioid agonist-antagonists pentazocine (Talwin), butorphanol (Stadol), nalbuphine (Nubain) Risk of precipitating withdrawal in opioid-dependent patients.
Analgesic ceiling.
Possible production of unpleasant psychotomimetic effects (e.g., dysphoria, delusions, hallucinations).
Partial agonist buprenorphine (Buprenex) Analgesic ceiling.
May precipitate withdrawal.
Antagonists naloxone (Narcan), naltrexone (ReVia) May precipitate withdrawal.
Limit use to treatment of life-threatening respiratory depression. Give in diluted form to opioid-tolerant patients.
Combination preparations Brompton's cocktail No evidence of analgesic benefit to using Brompton's cocktail over single-opioid analgesics.
DPT (meperidine, promethazine, and chlorpromazine) Efficacy is poor compared with that of other analgesics.
High incidence of adverse effects.
Anxiolytics alone benzodiazepine (e.g., alprazolam, Xanax; diazepam, Valium; lorazepam, Ativan) Analgesic properties not demonstrated except for some instances of neuropathic pain.
Added sedation from anxiolytics may compromise neurologic assessment in patients receiving opioids.
Sedative/hypnotic drugs alone barbiturates, benzodiazepine Analgesic properties not demonstrated.
Added sedation from sedative/hypnotic drugs limits opioid dosing.

Routes of Administration To Be Avoided for Treatment of Cancer Pain
Routes of Administration  Rationale for Not Recommending 
Intramuscular Painful.
Absorption unreliable.
Should not be used in children or patients prone to develop dependent edema or patients with thrombocytopenia.
Transnasal The only drug approved by the FDA for transnasal administration is butorphanol, an agonist-antagonist drug that generally is not recommended. (See opioid agonist-antagonists above.)

Side effects of opioids

Clinicians should anticipate and monitor for side effects. The more common adverse effects include nausea, somnolence, and constipation. These should be discussed with patients before starting opioids. Somnolence and nausea are more often encountered with initiation of opioid treatment but tend to resolve within a few days. Clinicians who follow patients during long-term opioid treatment should watch for potential side effects and manage them as the need arises.

Constipation

Anticipate the constipating effects of analgesics. Opioids compromise gastrointestinal tract peristaltic function (a nearly universal side effect). Consequently, stool within the gut lumen becomes excessively dehydrated. The cornerstone of effective prophylaxis, therefore, is measurement aimed at keeping the patient well hydrated to maintain well-hydrated stool. All patients using opioid medications should be prescribed a scheduled regimen of stool-softening agents (e.g., docusate sodium) at the commencement of opioid treatment. Patients who do not adequately respond to an aggressive regimen with stool softeners may benefit from the addition of mild osmotic agents (e.g., 70% sorbitol solution, lactulose, milk of magnesia), lubricants (e.g., mineral oil), bulk-forming laxatives (e.g., psyllium) with appropriate orally administered hydration, or mild cathartic laxatives (e.g., senna). Stimulant cathartics (e.g., senna, bisacodyl) may be useful in severely constipated patients; however, they may be relatively ineffective in situations in which stool has become desiccated. Opioid-induced constipation is a frequent cause of chronic nausea and is observed in 40% to 70% of patients receiving opioids.[62] It appears to be dose-related, is characterized by large variability in individuals, and is opioid-receptor mediated via both central and peripheral mechanisms. Opioids extend the gastrointestinal transit time and desiccate the intraluminal content.[112] Unlike nausea, complete tolerance to this effect does not generally develop, and most patients require laxative/stool-softener therapy for as long as they take opioids. A plain x-ray of the abdomen may be helpful in assessing the extent of fecal load.[113]

Initiating a regular laxative regimen emphasizes prevention of opioid-induced constipation. Recommendations regarding laxative treatment have been largely based on clinical experiences and observations. Combinations of a sennoside and a stool softener such as docusate are generally suggested.[114] Reports that fentanyl causes less constipation than oral morphine are interesting but need to be confirmed in further prospective studies.[113,115,61] A recent study demonstrated decreased laxative use in patients on transdermal fentanyl as compared with patients receiving oral morphine treatment.[61] Whether this decrease in laxative usage is clinically significant, however, and whether the decrease relates to the route of administration instead of the opioid type need to be demonstrated. In a single small series, opioid switching of morphine to methadone resulted in a reduction in constipation.[116] Severe opioid-induced constipation may occur. At an extreme it may be present as a severe ileus and pseudo bowel obstruction.[117] As is the case with opioid-induced nausea and constipation, management relies on the use of gastrointestinal prokinetic agents. The use of orally administered opioid-antagonists such as naloxone is being studied.[118,119] Although the oral bioavailability of these medications is very limited, opioid withdrawal syndromes have been noted when higher doses have been used. Methylnaltrexone, a quaternary derivative of naltrexone, is an opioid antagonist that does not cross the blood-brain barrier. Preliminary studies suggest that it may be effective in the management of opioid-associated constipation without causing opioid withdrawal.[120] (Refer to the PDQ summaries on Gastrointestinal Complications; Nausea and Vomiting; and Nutrition in Cancer Care for more information.)

Nausea and vomiting

Nausea and vomiting occurs in approximately one third to two thirds of patients taking opioids.[121-123] It is a common complication of early exposure to opioids and usually disappears within the first week of treatment. Appropriate antiemetic coverage during the opioid-initiation phase is usually effective in limiting this adverse effect. Nausea alone does not represent an allergic reaction to the opioid. Occasionally, nausea may be experienced when an opioid dose is significantly increased. An antiemetic should be available on an as-needed basis to address this situation.

Three main mechanisms underlie this opioid-related adverse effect.[124] The predominant mechanism appears to be stimulation of the chemoreceptor trigger zone, where dopamine is the main neurotransmitter. Another is reduced gastrointestinal motility, including delayed gastric emptying. Nausea via increased vestibular sensitivity is uncommon.

Multiple antiemetic regimens have been proposed for the management of opioid-induced emesis, but prospective studies comparing one regimen over another are lacking.[124] Metoclopramide or domperidone are generally recommended as first-line agents because they improve gastrointestinal motility and are antidopaminergic.[124,125] Metoclopramide can be administered orally or subcutaneously at doses of 10 mg 4 times a day or every 4 hours, depending on the severity of the nausea. Rescue doses should also be ordered on an as-needed basis. Extrapyramidal-related adverse effects are a potential complication of these medications. The incidence of extrapyramidal reactions is low with domperidone, but this drug is not available in a parenteral formulation. The antihistamines act on the histamine receptors in the vomiting center and on vestibular afferents. They are generally reserved for cases in which vestibular sensitivity, often manifesting as motion-induced nausea, is suspected or for cases in which bowel obstruction precludes the use of gastrointestinal prokinetic agents. Haloperidol may also be used under the latter circumstances. The phenothiazines are an alternative group of antiemetics, but extrapyramidal and anticholinergic adverse effects may be dose-limiting. Chlorpromazine has modest antiemetic activity but a high incidence of sedation, postural hypotension, and anticholinergic adverse effects, whereas piperazine derivatives such as prochlorperazine are stronger antiemetics but cause more extrapyramidal side effects. Anticholinergic side effects also limit the use of anticholinergic agents such as hyoscine hydrobromide (scopolamine) in opioid-induced nausea, particularly in patients with advanced cancer. These patients seem to be more vulnerable to these adverse effects. The role of 5-HT3-receptor antagonists such as ondansetron in ameliorating opioid-induced nausea is not clear.[126]

There appear to be differences between individual patients in the extent to which different opioids cause nausea.[127] These differences form the basis for the strategy of switching from one opioid to another when a particular opioid produces persistent nausea.[128,129] Switching the route, specifically from the oral to the parenteral, has also been suggested, but the study supporting this strategy is small.[130]

Nausea and vomiting can sometimes persist beyond the opioid-initiation phase or occur de novo in patients on long-term opioid treatment. It may become chronic in nature. The multicausal nature of the problem needs to be recognized since management is directed at identifying and addressing the various causes.[131] Chronic nausea has been associated with the accumulation of active opioid metabolites.[90] A number of strategies are suggested to manage chronic nausea, including switching the opioid or decreasing the dose when pain is well controlled. (Refer to the PDQ summary on Nausea and Vomiting for more information.)

Cognitive and other neurotoxic side effects of opioids

Opioid-related neurotoxicity may manifest as cognitive impairment, hallucinations, delirium, generalized myoclonus, hyperalgesia and/or allodynia. Patients who have renal impairment or who are taking higher doses of opioids are at greater risk of developing these side effects. The mechanisms underlying these side effects are unclear, but the opioid metabolites are implicated. When patients present with generalized pain of an unknown source and the opioid dose has been recently increased, hyperalgesia should be considered as a possible diagnosis.[132,133] The etiological contribution of opioids to cognitive impairment and delirium in the cancer patient is often difficult to determine. This is the case particularly in patients with advanced disease in which the baseline vulnerability is associated with multisystem impairment, and the concurrent administration of other psychotropic agents can complicate the assessment of etiology. Nonetheless, opioid-induced cognitive problems have been reported with increasing frequency in the last decade.[71,134,135] In addition to cognitive impairment within the context of delirium, other effects include myoclonus, hyperalgesia, perceptual disturbance, and seizures.[136] Although the remarkable characteristics, potential severity, and impact of delirium contribute to its dominance in the spectrum of opioid-related cognitive dysfunction, more subtle psychomotor and cognitive opioid effects have been described. Neuropsychological testing has been used to study these more-subtle effects in less-advanced cancer disease,[137] chronic nonmalignant pain,[121,138] and in healthy volunteers.[139] Collectively, studies of neuropsychological testing have demonstrated somewhat mixed findings,[140] with some detecting opioid-associated impairment in certain aspects of psychomotor or cognitive function [138,139] and others detecting minimal or no impairment.[121,137] Clinical experience and some studies suggest that patients become tolerant of the sedating effects that accompany either the initiation of opioid therapy or dose increases,[141] thereby allowing patients who are otherwise physically able, and on stable opioid doses, to safely engage in activities such as driving.[137,142]

Decreased brain cholinergic activity is recognized as one of the potential underlying pathophysiological mechanisms of delirium.[143-145] In the case of meperidine, the anticholinergic activity associated with its active metabolite normeperidine is suspected to be the basis of the cognitive impairment and delirium occurring in association with this opioid.[146,147] Other opioid metabolites have been studied in relation to the generation of neuroexcitatory states in animal laboratory models and delirium in human subjects. A series of animal studies have demonstrated neuroexcitatory states in association with morphine metabolites, morphine-3-glucuronide (M-3-G) [148] and normorphine-3-glucuronide,[149] and the hydromorphone metabolite, hydromorphone-3-glucuronide.[150] In a hospice study of 36 patients with advanced cancer receiving morphine, both M-3-G and morphine-6-glucuronide (M-6-G) levels were studied in relation to the development of side effects, which included nausea and vomiting in 10 patients and cognitive impairment in 9 patients.[151] Creatinine levels, and plasma levels of M-3-G, M-6-G, and dose-corrected M-3-G and M-6-G, were higher in the 19 patients with side effects, suggesting that the elevation of morphine metabolites in association with renal impairment was associated with opioid toxicity, including cognitive impairment. Evidence is extensive demonstrating elevation of opioid-metabolite levels in the setting of renal impairment,[89,96,151-153] and some studies have noted an association with features of neurotoxicity, including cognitive impairment.[135,151] An accumulation of opioid metabolites possibly also occurs during dehydration, which was suggested as a contributory factor in a prospective study of predominantly opioid-related delirium.[154] Switching to another opioid is one strategy for abating the side effects in cases in which accumulation of active metabolites is considered responsible for side effects such as generalized myoclonus, sedation, confusion, or chronic nausea.[26]

Managing cognitive and other neurotoxic effects of opioids

The general management approach to opioid-induced delirium requires a multidimensional assessment to determine the presence of other potentially treatable contributory factors such as dehydration, other centrally acting medications, sepsis, and hypercalcemia.[134,154,155] Clinical experience suggests that the presence of tactile hallucinations and myoclonus,[82] although not exclusively associated with opioid toxicity, raise the suspicion of this cause. A careful assessment can also identify prognostic factors associated with greater difficulty in achieving pain control, the need for higher opioid doses, and consequently greater risk of opioid-induced delirium. (Refer to the PDQ summary on Cognitive Disorders and Delirium for further information.) These factors include neuropathic pain, incidental pain, tolerance, somatization of psychological distress, and a positive history of drug or alcohol abuse.[156]

In addition to searching for underlying reversible causes of delirium, the symptomatic management of delirium requires the addition of a neuroleptic agent to control agitation and perceptual or delusional disturbance. Haloperidol is regarded as the drug of choice in this context,[157] and methotrimeprazine and chlorpromazine are considered useful alternatives,[158,159] especially when a greater level of sedation is required. Midazolam, a sedating and short-acting benzodiazepine given by continuous infusion, is sometimes necessary, especially in the case of nonreversible delirium.[160] Typical anxiolytics, including lorazepam, can be used to manage comorbid anxiety. Early data suggest that some atypical antipsychotics may be beneficial in improving pain control and decreasing opioid requirements in the cancer patient with mild cognitive impairment and/or anxiety. It is unclear whether this benefit is due to a primary effect or to its secondary impact on cognitive impairment and/or anxiety.[161]

The specific management approach to opioid-induced cognitive and other neurotoxic side effects involves either a dose reduction, a change in route, or an opioid switch.[162] If the pain is well controlled, and the cognitive and neurotoxic side effects are not severe, modest opioid dose reduction may be effective. The rationale for switching opioids, commonly referred to as opioid switching, is that a more favorable balance between analgesia and side effects can be achieved, often with a lower dose than that predicted by the conventional analgesic table.[83,134,163] This can reflect incomplete cross-tolerance among opioids in relation to analgesic and other effects.[164] It is also possible that switching to a new opioid could allow for the elimination of potentially toxic opioid metabolites.[165,166,134] Reduction in opioid dose in the context of an opioid-induced delirium has not been systematically evaluated but is also likely to have beneficial results. Although there is growing evidence to suggest a beneficial role for opioid switching,[165,167,129] controversy persists over the relative value of opioid switching versus dose reduction.[81]

Cognitive benefit has been reported with the use of methylphenidate in patients receiving a continuous infusion of opioids for cancer pain.[168] The psychostimulant benefit is likely to relate to mitigation of sedation associated with upward dose titration of opioid.[169] Although psychostimulants have been advocated for hypoactive delirium,[170]any evidence of perceptual or delusional disturbance is considered a contraindication. An open-label study of donepezil, a long-acting selective acetylcholinesterase inhibitor, suggests that it relieves opioid-associated fatigue and sedation in patients who are receiving opioids for cancer pain.[171]

Respiratory depression

Patients receiving long-term opioid therapy generally develop tolerance to the respiratory-depressant effects of these agents. However, concerns about respiratory depression with opioid use remain prevalent among clinicians and patients. Clinicians experienced in end-of-life care recognize that such concerns are generally exaggerated, though empirical research in the area is sparse. One observational study of 30 patients that evaluated the effect of parenteral opioid titration for the control of acute exacerbation of cancer pain showed no association between parenteral opioid titration and hypoventilation at pain control, as measured by change in end-tidal CO2 respiratory rate or oxygen saturation.[172]

When indicated for reversal of opioid-induced respiratory depression, naloxone titrated in small increments or as an infusion should be administered to improve respiratory function without reversing analgesia. The patient should be monitored carefully until the episode of respiratory depression resolves. The opioid antagonists have a short half-life and may have to be given repeatedly until the agonist drug is sufficiently cleared.[173]

Subacute overdose

Perhaps more common than acute respiratory depression, subacute overdose may manifest as slowly progressive (hours to days) somnolence and respiratory depression. Before analgesic doses are reduced, advancing disease must be considered, especially in the dying patient. Generally, withholding one or two doses of an opioid analgesic is adequate to assess whether mental and respiratory depression are opioid related. If symptoms resolve after temporary opioid withdrawal, reduce the scheduled opioid dosage by 25%. If symptoms do not abate, but the patient complains of or exhibits signs of increased pain, or if symptoms referable to opioid withdrawal occur, consider alternative causes for CNS depression and reinstate analgesic treatment. Ongoing assessment is essential to maintain adequate pain relief.

Effects of opioids on sexual function

Reduced libido is a well-known phenomenon for those using heroin or those in a methadone maintenance program; however, clinicians prescribing opioids for pain poorly understand this effect. Early case studies of persons using heroin or methadone described diminished libido, sexual dysfunction, reduced testosterone levels in men, and amenorrhea in women.[174-179] These effects resolve after the opioid has been discontinued. More recent case reports of patients receiving opioids for relief of chronic pain suggest these same findings.[180,181] The long-term effects of reduced testosterone and amenorrhea are not well known. Sexuality is an essential component of quality of life in many patients, including patients with advanced disease.[182] Patients should be assessed for changes in libido and sexual dysfunction. If these changes are distressing to the patient, serum testosterone levels may be obtained. Should the patient seek improvement in libido and performance, treatment is often empirical, keeping in mind that there are many potential causes of changes in sexual function. Treatment includes using nonopioids for pain, adding adjuvant analgesics in the hope the opioid dose may be reduced, or replacing testosterone through injections or a patch (if not contraindicated). More research is needed to understand the relationship between opioids and sexual function, as well as the most effective treatment strategies. (Refer to the PDQ summary on Sexuality and Reproductive Issues for more information.)

Other opioid side effects

Dry mouth, urinary retention, pruritus, dysphoria, euphoria, sleep disturbances, and inappropriate secretion of antidiuretic hormone are less common.

Adjuvant Drugs

Adjuvant drugs are valuable during all phases of pain management to enhance analgesic efficacy, treat concurrent symptoms, and provide independent analgesia for specific types of pain.[183] Adverse drug reactions are common, however, and there are wide interindividual and ethnic differences in drug metabolism.[184] A survey on symptom severity and management in 593 cancer patients treated for an average of 51 days reported that during this time, anticonvulsants were used in 11.8% of patients, antidepressants in 16%, corticosteroids in 28%, and bisphosphonates in 7.3%.[185] Patients with advanced cancer on palliative medicine services are reported to receive on average five medications for symptom relief, and as a result are at high risk of drug interactions.[184] A further note of caution appears in another study that questioned the concept of opioid-sparing effects of co-analgesics.[186] Nevertheless, adjuvant analgesics have been extensively studied and reviewed in noncancer settings and are generally endorsed as an important intervention in the provision of adequate pain management.[187-190] Few trials compare adjuvant analgesics in the cancer setting.

Adjuvant Medications With Possible Analgesic Activity
Class  Drug  Daily Dose Range*  Studies Conducted in: 
      Cancer Patients   Noncancer Patients  
Antidepressants amitriptyline (Elavil) 10–25 mg every day [191,192] [193]
desipramine (Norpramin) 10–150 mg every day [194] [195]
maprotiline (Ludiomil) 25 mg bid–50 mg tid [196]
duloxetine (Cymbalta) 20 mg bid–30 mg bid [197]
nortriptyline (Pamelor, Aventyl) 10–100 mg every day [198]
venlafaxine (Effexor) 37.5–225 mg every day [199,200] [201]
Anticonvulsants carbamazepine (Tegretol) 100 mg tid–400 mg tid [202]
valproate (Depacon) 500 mg tid–1,000 mg tid [203]
gabapentin (Neurontin) 100 mg tid–1,000 mg tid [204,205] [206]
clonazepam (Klonopin) 0.5 mg bid–4 mg bid [207]
lamotrigine (Lamictal) 25 mg bid–100 mg bid [208]
pregabalin (Lyrica) 150 mg divided into 2 or 3 doses; increase to 300 mg starting at day 3–7; if needed, increase to 600 mg 7 days later [209]
Local anesthetics mexiletine (Mexitil) 100 mg bid–300 mg tid [210]
lidocaine patch (Lidoderm) 5% patch contains 700 mg; one patch, 12 hours on, 12 hours off [211]
Corticosteroids dexamethasone (Decadron) See text
prednisone See text
Bisphosphonates clodronate See text
pamidronate (Aredia) See text
zoledronic acid (Zometa) See text [212]
NSAIDs See the Dosing Recommendations for Acetaminophen and NSAIDs table
Miscellaneous baclofen (Lioresal) 5 mg tid–20 mg tid [213]
calcitonin (Calcimar) 100–200 IU (subcutaneous or intranasal)
clonidine (Catapres) 0.1 mg bid–0.3 mg bid [214]
methylphenidate (Ritalin) 2.5 mg bid–20 mg bid [215] [216]
ketamine (Ketalar) See NMDA Receptor Antagonists section

bid = twice a day; tid = three times a day.
*Starting doses should incorporate the lowest possible dose.

Antidepressants

  • The analgesic benefits of tricyclic antidepressants have been well established and are generally considered first-line therapy for many neuropathic pain syndromes.[187-190,217] Supporting evidence is strong for amitriptyline and desipramine, and there is endorsement of other newer antidepressants such as maprotiline and paroxetine. Patients with neuropathic pain characterized by continuous dysesthesias are generally believed to be the most likely to benefit from antidepressant management; however, a randomized placebo-controlled study of amitriptyline for neuropathic pain in cancer patients found only slight analgesic benefit with significantly worse adverse effects.[192]


  • The most common side effects of tricyclic antidepressants are the following:
    • Constipation.
    • Dry mouth.
    • Blurred vision.
    • Cognitive changes.
    • Tachycardia.
    • Urinary retention.
    Caution has also been advised in treating patients with cardiac disease, and an electrocardiogram is sometimes recommended as a prudent measure. A slow upward titration is suggested as a good way to avoid side effects.[199]


Anticonvulsants

  • The group of commonly used anticonvulsants as adjuvant analgesics for neuropathic pain includes carbamazepine, valproate, phenytoin, and clonazepam.[187-190,217]

    Clinical experience with carbamazepine is extensive, but use of this drug is limited in the cancer population because of concern that it causes bone marrow suppression, in particular leukopenia. Other common adverse effects include nystagmus, dizziness, diplopia, cognitive impairment, and mood and sleep disturbance.

    Dosing guidelines for phenytoin are similar to those for the treatment for seizures.[187] This drug can be administered using a loading dose, which may be particularly useful in patients with severe pain.

    Gabapentin is increasingly reported as useful for the management of neuropathic pain associated with cancer and its treatment.[218-221,204,205] Commonly reported side effects include somnolence, dizziness, ataxia, and fatigue.[190,221] One randomized open-label trial of gabapentin combined with an opioid (n = 38) versus an opioid alone (n = 37) for the management of neuropathic cancer pain suggests that the combination group achieved better relief than those receiving opioid monotherapy.[222]

    Clonazepam is an anticonvulsant from the benzodiazepine class and is commonly used for treating lancinating or paroxysmal neuropathic pain.[187] The patient must be monitored carefully for drowsiness and cognitive impairment.



Local anesthetics

  • The use of mexiletine has been described for chronic neuropathic pain.[187,188,190] Side effects are reported as common and include gastrointestinal toxicity, in particular nausea, and CNS side effects such as dizziness. Patients with a history of cardiac disease and those on higher doses are at increased risk of adverse effects, and it is recommended that they receive appropriate cardiac evaluation, including an electrocardiogram.

Corticosteroids

  • These drugs have achieved wide acceptance in the management of patients with cancer pain. They are indicated as adjuvant analgesics for cancer pain of bone, visceral, and neuropathic origin. Adverse effects include neuropsychiatric syndromes, gastrointestinal disturbances, proximal myopathy, hyperglycemia, aseptic necrosis, capillary fragility, and immunosuppression. The risk of adverse effects increases with the duration of use. As a result, use is often restricted to patients with a limited life expectancy; in addition, once effective pain control is obtained, it is commonly recommended that the dose be tapered as much as possible. Dosage recommendations vary from a trial of low-dose therapy such as dexamethasone 1 to 2 mg or prednisone 5 to 10 mg once or twice daily,[187] to a starting dose of dexamethasone 10 mg twice daily with subsequent tapering to the minimal effective dose.[223]

    Another suggested use of corticosteroids is in high doses for short periods in patients with severe pain. This empirical approach recommends a regime of a single bolus of dexamethasone 100 mg IV followed by a small amount given 4 times per day and then tapered over the next few weeks.[187]

    Although there is widespread acceptance of steroid therapy, mostly via the oral route but also subcutaneously and intravenously, data remain inadequate for definitive conclusions regarding efficacy and dosing guidelines.[187,224,189,190,223,225-228]



Bisphosphonates

  • These drugs have been recommended for the management of bone pain as well as the prevention of skeletal complications in patients with metastatic bone disease.[187-190,229-232] Their use in a study of breast cancer patients resulted in improved quality of life compared with that of patients not using bisphosphonates.[233] The bisphosphonates most frequently used are clodronate, pamidronate,[234] and zoledronic acid.[235]


  • Clodronate can be given orally or intravenously. Dosage recommendations are oral clodronate, 1,600 mg per day; or IV clodronate, 600 to 1,500 mg every 2 to 3 weeks. Clodronate is not available in the United States.


  • Pamidronate has been recommended in the dose range of 60 to 90 mg IV over 2 hours every 3 to 4 weeks; however, pooled results from two multicenter, double-blind, randomized, placebo-controlled trials (n = 350) using pamidronate (90 mg every 3 weeks) failed to demonstrate a benefit for bone pain.[236]


  • Zoledronic acid is a potent bisphosphonate that can be given as an IV bolus over 15 to 30 minutes in the dose range of 4 to 8 mg; however, the 8-mg dose has been associated with deterioration of renal function.[237-240] The few studies to date suggest administration at 3- to 4-week intervals.[241]


  • Ibandronate can be given orally or intravenously. Dosing recommendations are 50 mg orally daily or 6 mg intravenously every 3 to 4 weeks.[242]


  • The FDA has highlighted the possibility of severe and sometimes incapacitating bone, joint, and/or muscle pain in patients who are taking bisphosphonates. The musculoskeletal pain may occur within days, months, or years after starting treatment with a bisphosphonate. This pain contrasts with the acute-phase response characterized by fever, chills, bone pain, myalgias, and arthralgias that may sometimes accompany initial administration of intravenous bisphosphonates. The FDA recommends that bisphosphonates be considered a possible cause of severe musculoskeletal pain in patients who present with these symptoms, and health care professionals should consider temporary or permanent discontinuation of the drug. The risk factors and the incidence of the association of this musculoskeletal pain with bisphosphonates remain unknown.[243]


  • The use of bisphosphonates carries a risk of developing bisphosphonate-associated osteonecrosis (BON). (Refer to the Oral Toxicities Not Related to Chemotherapy or Radiation Therapy section in the PDQ summary on Oral Complications of Chemotherapy and Head/Neck Radiation for more information.)


Miscellaneous

  • Baclofen
    • This drug is generally used for spasticity but may also be used for the treatment of neuropathic pain.[187,188,190] Side effects include drowsiness, dizziness, ataxia, confusion, and nausea and vomiting.


  • Calcitonin
    • Although the mechanism by which calcitonin produces analgesia is unknown, historically it has been recommended for the treatment of both bone and neuropathic pain.[187,188,190,229,230] However, a systematic review of randomized double-blind clinical trials assessing the efficacy of calcitonin for control of metastatic bone pain does not support its use.[244] Because only two of these studies were evaluated as well designed, further research is necessary. The utility of calcitonin for bone pain is unclear.


  • Clonidine
    • This traditional antihypertensive can be given via the oral, epidural, or transdermal route and has been recommended as a trial for the management of neuropathic pain. Reported side effects include dry mouth, dizziness and hypotension, sedation, and constipation.[187,188,190] The maximum recommended dose is 2.4 mg per day.


  • Psychostimulants
    • Psychostimulants such as dextroamphetamine, methylphenidate, and modafinil may enhance the analgesic effects of opioids.[168,169,189] They may also be used to diminish opioid-induced sedation when reducing the dose is not possible.


  • NMDA Receptor Antagonists
    • There is increasing evidence for the importance of NMDA receptors and the possibility that NMDA antagonists may have a role in refractory cancer pain management.[187,245] Ketamine in subanesthetic doses has been used in this setting.[246] The severe psychomimetic adverse effects associated with this treatment, including vivid hallucinations, limit widespread use of ketamine. Coadministration of a neuroleptic or benzodiazepine is recommended to limit the emergence of these effects. Ketamine is generally given subcutaneously at a low starting dose such as 0.1 mg per kg of body weight per hour, with a gradual escalation. Oral ketamine may be a more potent analgesic and have a more favorable side-effect profile than parenteral ketamine.[231] One study suggests short-duration therapy of a continuous subcutaneous infusion of ketamine over 3 to 5 days. The initial dose is 100 mg per day, and if pain control is inadequate, the dose is escalated to 300 mg per day and then to a maximum dose of 500 mg per day. Treatment is continued for 3 days at either the lowest effective dose or 500 mg per day and then discontinued.[245] A systematic review of the benefits and harms of ketamine in managing cancer pain revealed a general lack of studies and small subject numbers,[247,248] precluding a definitive conclusion on benefits and harms.


    • Methadone, particularly the racemic mixture, appears to have significant NMDA-antagonist properties.[249] The d-isomer blocks the NMDA receptor and as a result may yield independent analgesic effects and perhaps reverse some analgesic tolerance to the opioid.[250] This may explain the often-unanticipated high potency of methadone.


    • Dextromethorphan (DM), a commonly prescribed antitussive, may also have NMDA-blocking properties.[250] The clinical significance of this effect, however, is unclear and studies have not been able to determine at what dose these effects may manifest. Oral DM in doses of 60 or 90 mg given preoperatively and postoperatively has been shown to reduce pain intensity and opioid use after orthopedic oncology surgery.[251,252] A randomized, double-blind, placebo-controlled study of 65 patients evaluated the efficacy and safety of DM or placebo with slow-release morphine. The dose of DM was 60 mg 4 times daily, increased after 7 days to 120 mg 4 times daily if tolerated. While the DM group showed somewhat more improvement than the placebo group, the differences were not significant; furthermore, the DM group experienced more toxic effects, particularly dizziness.[253] The authors concluded that DM does not enhance opioid analgesia or modulate opioid tolerance enough in cancer patients to warrant continued use.




  • Octreotide
    • Data from a case series of 16 patients with symptomatic hepatic metastases from a variety of nonneuroendocrine primary sites suggest that octreotide palliates pain and improves a variety of quality-of-life indices as measured by the EORTC QLQ-C30 questionnaire.[254]


References

  1. World Health Organization.: Cancer Pain Relief. 2nd ed. Geneva, Switzerland: World Health Organization, 1996. 

  2. Stockler M, Vardy J, Pillai A, et al.: Acetaminophen (paracetamol) improves pain and well-being in people with advanced cancer already receiving a strong opioid regimen: a randomized, double-blind, placebo-controlled cross-over trial. J Clin Oncol 22 (16): 3389-94, 2004.  [PUBMED Abstract]

  3. Oneschuk D, Bruera E: The "dark side" of adjuvant analgesic drugs. Progress in Palliative Care 5(1): 5-13, 1997. 

  4. Caraceni A, Gorni G, Zecca E, et al.: More on the use of nonsteroidal anti-inflammatories in the management of cancer pain. J Pain Symptom Manage 21 (2): 89-91, 2001.  [PUBMED Abstract]

  5. Jenkins CA, Bruera E: Nonsteroidal anti-inflammatory drugs as adjuvant analgesics in cancer patients. Palliat Med 13 (3): 183-96, 1999.  [PUBMED Abstract]

  6. McNicol E, Strassels S, Goudas L, et al.: Nonsteroidal anti-inflammatory drugs, alone or combined with opioids, for cancer pain: a systematic review. J Clin Oncol 22 (10): 1975-92, 2004.  [PUBMED Abstract]

  7. Mercadante S, Fulfaro F, Casuccio A: A randomised controlled study on the use of anti-inflammatory drugs in patients with cancer pain on morphine therapy: effects on dose-escalation and a pharmacoeconomic analysis. Eur J Cancer 38 (10): 1358-63, 2002.  [PUBMED Abstract]

  8. Fitzgerald GA: Coxibs and cardiovascular disease. N Engl J Med 351 (17): 1709-11, 2004.  [PUBMED Abstract]

  9. Rømsing J, Møiniche S: A systematic review of COX-2 inhibitors compared with traditional NSAIDs, or different COX-2 inhibitors for post-operative pain. Acta Anaesthesiol Scand 48 (5): 525-46, 2004.  [PUBMED Abstract]

  10. Sibbald B: Rofecoxib (Vioxx) voluntarily withdrawn from market. CMAJ 171 (9): 1027-8, 2004.  [PUBMED Abstract]

  11. Whitcomb LA, Kirsh KL, Passik SD: Substance abuse issues in cancer pain. Curr Pain Headache Rep 6 (3): 183-90, 2002.  [PUBMED Abstract]

  12. Hanks GW, Conno F, Cherny N, et al.: Morphine and alternative opioids in cancer pain: the EAPC recommendations. Br J Cancer 84 (5): 587-93, 2001.  [PUBMED Abstract]

  13. Cherny N, Ripamonti C, Pereira J, et al.: Strategies to manage the adverse effects of oral morphine: an evidence-based report. J Clin Oncol 19 (9): 2542-54, 2001.  [PUBMED Abstract]

  14. Bruera E, Schoeller T, Fainsinger RL, et al.: Custom-made suppositories of methadone for severe cancer pain. J Pain Symptom Manage 7 (6): 372-4, 1992.  [PUBMED Abstract]

  15. Hunt G, Bruera E: Respiratory depression in a patient receiving oral methadone for cancer pain. J Pain Symptom Manage 10 (5): 401-4, 1995.  [PUBMED Abstract]

  16. Crews JC, Sweeney NJ, Denson DD: Clinical efficacy of methadone in patients refractory to other mu-opioid receptor agonist analgesics for management of terminal cancer pain. Case presentations and discussion of incomplete cross-tolerance among opioid agonist analgesics. Cancer 72 (7): 2266-72, 1993.  [PUBMED Abstract]

  17. Sjøgren P, Jensen NH, Jensen TS: Disappearance of morphine-induced hyperalgesia after discontinuing or substituting morphine with other opioid agonists. Pain 59 (2): 313-6, 1994.  [PUBMED Abstract]

  18. Leng G, Finnegan MJ: Successful use of methadone in nociceptive cancer pain unresponsive to morphine. Palliat Med 8 (2): 153-5, 1994.  [PUBMED Abstract]

  19. Thomas Z, Bruera E: Use of methadone in a highly tolerant patient receiving parenteral hydromorphone. J Pain Symptom Manage 10 (4): 315-7, 1995.  [PUBMED Abstract]

  20. Manfredi PL, Borsook D, Chandler SW, et al.: Intravenous methadone for cancer pain unrelieved by morphine and hydromorphone: clinical observations. Pain 70 (1): 99-101, 1997.  [PUBMED Abstract]

  21. Bruera E, Watanabe S, Fainsinger RL, et al.: Custom-made capsules and suppositories of methadone for patients on high-dose opioids for cancer pain. Pain 62 (2): 141-6, 1995.  [PUBMED Abstract]

  22. Bruera E, Pereira J, Watanabe S, et al.: Opioid rotation in patients with cancer pain. A retrospective comparison of dose ratios between methadone, hydromorphone, and morphine. Cancer 78 (4): 852-7, 1996.  [PUBMED Abstract]

  23. Vigano A, Fan D, Bruera E: Individualized use of methadone and opioid rotation in the comprehensive management of cancer pain associated with poor prognostic indicators. Pain 67 (1): 115-9, 1996.  [PUBMED Abstract]

  24. Mercadante S, Casuccio A, Fulfaro F, et al.: Switching from morphine to methadone to improve analgesia and tolerability in cancer patients: a prospective study. J Clin Oncol 19 (11): 2898-904, 2001.  [PUBMED Abstract]

  25. Watanabe S, Belzile M, Kuehn N, et al.: Capsules and suppositories of methadone for patients on high-dose opioids for cancer pain: clinical and economic considerations. Cancer Treat Rev 22 (Suppl A): 131-6, 1996.  [PUBMED Abstract]

  26. Fainsinger R, Schoeller T, Bruera E: Methadone in the management of cancer pain: a review. Pain 52 (2): 137-47, 1993.  [PUBMED Abstract]

  27. Ripamonti C, Zecca E, Bruera E: An update on the clinical use of methadone for cancer pain. Pain 70 (2-3): 109-15, 1997.  [PUBMED Abstract]

  28. Ripamonti C, Bianchi M: The use of methadone for cancer pain. Hematol Oncol Clin North Am 16 (3): 543-55, 2002.  [PUBMED Abstract]

  29. Mathew P, Storey P: Subcutaneous methadone in terminally ill patients: manageable local toxicity. J Pain Symptom Manage 18 (1): 49-52, 1999.  [PUBMED Abstract]

  30. Gutstein A: Opioid analgesics. In: Goodman LS, Hardman JG, Limbird LE, et al.: Goodman & Gilman's The Pharmacological Basis of Therapeutics. 10th ed. New York, NY: McGraw-Hill, 2001, pp 596-7. 

  31. Bruera E, Palmer JL, Bosnjak S, et al.: Methadone versus morphine as a first-line strong opioid for cancer pain: a randomized, double-blind study. J Clin Oncol 22 (1): 185-92, 2004.  [PUBMED Abstract]

  32. Gagnon B, Almahrezi A, Schreier G: Methadone in the treatment of neuropathic pain. Pain Res Manag 8 (3): 149-54, 2003 Fall.  [PUBMED Abstract]

  33. Krantz MJ, Lewkowiez L, Hays H, et al.: Torsade de pointes associated with very-high-dose methadone. Ann Intern Med 137 (6): 501-4, 2002.  [PUBMED Abstract]

  34. Krantz MJ, Kutinsky IB, Robertson AD, et al.: Dose-related effects of methadone on QT prolongation in a series of patients with torsade de pointes. Pharmacotherapy 23 (6): 802-5, 2003.  [PUBMED Abstract]

  35. Gil M, Sala M, Anguera I, et al.: QT prolongation and Torsades de Pointes in patients infected with human immunodeficiency virus and treated with methadone. Am J Cardiol 92 (8): 995-7, 2003.  [PUBMED Abstract]

  36. Walker PW, Klein D, Kasza L: High dose methadone and ventricular arrhythmias: a report of three cases. Pain 103 (3): 321-4, 2003.  [PUBMED Abstract]

  37. Kornick CA, Kilborn MJ, Santiago-Palma J, et al.: QTc interval prolongation associated with intravenous methadone. Pain 105 (3): 499-506, 2003.  [PUBMED Abstract]

  38. Reddy S, Fisch M, Bruera E: Oral methadone for cancer pain: no indication of Q-T interval prolongation or torsades de pointes. J Pain Symptom Manage 28 (4): 301-3, 2004.  [PUBMED Abstract]

  39. Martell BA, Arnsten JH, Ray B, et al.: The impact of methadone induction on cardiac conduction in opiate users. Ann Intern Med 139 (2): 154-5, 2003.  [PUBMED Abstract]

  40. Ripamonti C, Groff L, Brunelli C, et al.: Switching from morphine to oral methadone in treating cancer pain: what is the equianalgesic dose ratio? J Clin Oncol 16 (10): 3216-21, 1998.  [PUBMED Abstract]

  41. Fisch MJ, Cleeland CS: Managing cancer pain. In: Skeel RT, ed.: Handbook of Cancer Chemotherapy. 6th ed. Philadelphia, Pa: Lippincott Williams & Wilkins, 2003, pp 663. 

  42. Morley JS, Watt JW, Wells JC, et al.: Methadone in pain uncontrolled by morphine. Lancet 342 (8881): 1243, 1993.  [PUBMED Abstract]

  43. Mercadante S, Sapio M, Serretta R, et al.: Patient-controlled analgesia with oral methadone in cancer pain: preliminary report. Ann Oncol 7 (6): 613-7, 1996.  [PUBMED Abstract]

  44. Mercadante S, Casuccio A, Calderone L: Rapid switching from morphine to methadone in cancer patients with poor response to morphine. J Clin Oncol 17 (10): 3307-12, 1999.  [PUBMED Abstract]

  45. Nauck F, Ostgathe C, Dickerson ED: A German model for methadone conversion. Am J Hosp Palliat Care 18 (3): 200-2, 2001 May-Jun.  [PUBMED Abstract]

  46. Derby S, Chin J, Portenoy RK: Systemic opioid therapy for chronic cancer pain: practical guidelines for converting drugs and routes of administration. CNS Drugs 9 (2): 99-109, 1998. 

  47. O'Rourke MA, Shalabi A, Webb S: Methadone for treatment of cancer pain. JAMA 275 (7): 519, 1996.  [PUBMED Abstract]

  48. Dworkin RH, O'Connor AB, Backonja M, et al.: Pharmacologic management of neuropathic pain: evidence-based recommendations. Pain 132 (3): 237-51, 2007.  [PUBMED Abstract]

  49. Arbaiza D, Vidal O: Tramadol in the treatment of neuropathic cancer pain: a double-blind, placebo-controlled study. Clin Drug Investig 27 (1): 75-83, 2007.  [PUBMED Abstract]

  50. Petzke F, Radbruch L, Sabatowski R, et al.: Slow-release tramadol for treatment of chronic malignant pain--an open multicenter trial. Support Care Cancer 9 (1): 48-54, 2001.  [PUBMED Abstract]

  51. Mercadante S, Arcuri E, Fusco F, et al.: Randomized double-blind, double-dummy crossover clinical trial of oral tramadol versus rectal tramadol administration in opioid-naive cancer patients with pain. Support Care Cancer 13 (9): 702-7, 2005.  [PUBMED Abstract]

  52. Negro S, Martín A, Azuara ML, et al.: Stability of tramadol and haloperidol for continuous subcutaneous infusion at home. J Pain Symptom Manage 30 (2): 192-9, 2005.  [PUBMED Abstract]

  53. Paice JA, Noskin GA, Vanagunas A, et al.: Efficacy and safety of scheduled dosing of opioid analgesics: a quality improvement study. J Pain 6 (10): 639-43, 2005.  [PUBMED Abstract]

  54. Mercadante S, Villari P, Ferrera P, et al.: Safety and effectiveness of intravenous morphine for episodic (breakthrough) pain using a fixed ratio with the oral daily morphine dose. J Pain Symptom Manage 27 (4): 352-9, 2004.  [PUBMED Abstract]

  55. Ryan M, Moynihan TJ, Loprinzi CL: As-needed morphine: yes, but at what dose and at what interval? J Clin Oncol 23 (16): 3849-52, 2005.  [PUBMED Abstract]

  56. Miaskowski C, Dodd MJ, West C, et al.: Lack of adherence with the analgesic regimen: a significant barrier to effective cancer pain management. J Clin Oncol 19 (23): 4275-9, 2001.  [PUBMED Abstract]

  57. Mercadante S, Villari P, Ferrera P, et al.: Optimization of opioid therapy for preventing incident pain associated with bone metastases. J Pain Symptom Manage 28 (5): 505-10, 2004.  [PUBMED Abstract]

  58. Kornick CA, Santiago-Palma J, Schulman G, et al.: A safe and effective method for converting patients from transdermal to intravenous fentanyl for the treatment of acute cancer-related pain. Cancer 97 (12): 3121-4, 2003.  [PUBMED Abstract]

  59. Quigley C, Joel S, Patel N, et al.: Plasma concentrations of morphine, morphine-6-glucuronide and morphine-3-glucuronide and their relationship with analgesia and side effects in patients with cancer-related pain. Palliat Med 17 (2): 185-90, 2003.  [PUBMED Abstract]

  60. Klepstad P, Kaasa S, Jystad A, et al.: Immediate- or sustained-release morphine for dose finding during start of morphine to cancer patients: a randomized, double-blind trial. Pain 101 (1-2): 193-8, 2003.  [PUBMED Abstract]

  61. Radbruch L, Sabatowski R, Loick G, et al.: Constipation and the use of laxatives: a comparison between transdermal fentanyl and oral morphine. Palliat Med 14 (2): 111-9, 2000.  [PUBMED Abstract]

  62. Ahmedzai S, Brooks D: Transdermal fentanyl versus sustained-release oral morphine in cancer pain: preference, efficacy, and quality of life. The TTS-Fentanyl Comparative Trial Group. J Pain Symptom Manage 13 (5): 254-61, 1997.  [PUBMED Abstract]

  63. Heiskanen T, Kalso E: Controlled-release oxycodone and morphine in cancer related pain. Pain 73 (1): 37-45, 1997.  [PUBMED Abstract]

  64. Curtis GB, Johnson GH, Clark P, et al.: Relative potency of controlled-release oxycodone and controlled-release morphine in a postoperative pain model. Eur J Clin Pharmacol 55 (6): 425-9, 1999.  [PUBMED Abstract]

  65. Bruera E, Belzile M, Pituskin E, et al.: Randomized, double-blind, cross-over trial comparing safety and efficacy of oral controlled-release oxycodone with controlled-release morphine in patients with cancer pain. J Clin Oncol 16 (10): 3222-9, 1998.  [PUBMED Abstract]

  66. Hagen NA, Babul N: Comparative clinical efficacy and safety of a novel controlled-release oxycodone formulation and controlled-release hydromorphone in the treatment of cancer pain. Cancer 79 (7): 1428-37, 1997.  [PUBMED Abstract]

  67. van Seventer R, Smit JM, Schipper RM, et al.: Comparison of TTS-fentanyl with sustained-release oral morphine in the treatment of patients not using opioids for mild-to-moderate pain. Curr Med Res Opin 19 (6): 457-69, 2003.  [PUBMED Abstract]

  68. Galer BS, Coyle N, Pasternak GW, et al.: Individual variability in the response to different opioids: report of five cases. Pain 49 (1): 87-91, 1992.  [PUBMED Abstract]

  69. Sjøgren P, Jonsson T, Jensen NH, et al.: Hyperalgesia and myoclonus in terminal cancer patients treated with continuous intravenous morphine. Pain 55 (1): 93-7, 1993.  [PUBMED Abstract]

  70. Fainsinger RL, Bruera E: Is this opioid analgesic tolerance? J Pain Symptom Manage 10 (7): 573-7, 1995.  [PUBMED Abstract]

  71. MacDonald N, Der L, Allan S, et al.: Opioid hyperexcitability: the application of alternate opioid therapy. Pain 53 (3): 353-5, 1993.  [PUBMED Abstract]

  72. Fainsinger R, Schoeller T, Boiskin M, et al.: Palliative care round: cognitive failure and coma after renal failure in a patient receiving captopril and hydromorphone. J Palliat Care 9 (1): 53-5, 1993 Spring.  [PUBMED Abstract]

  73. Bruera E, Schoeller T, Montejo G: Organic hallucinosis in patients receiving high doses of opiates for cancer pain. Pain 48 (3): 397-9, 1992.  [PUBMED Abstract]

  74. Mercadante S, Villari P, Ferrera P, et al.: Opioid plasma concentrations during a switch from transdermal fentanyl to methadone. J Palliat Med 10 (2): 338-44, 2007.  [PUBMED Abstract]

  75. Drake R, Longworth J, Collins JJ: Opioid rotation in children with cancer. J Palliat Med 7 (3): 419-22, 2004.  [PUBMED Abstract]

  76. Pereira J, Lawlor P, Vigano A, et al.: Equianalgesic dose ratios for opioids. a critical review and proposals for long-term dosing. J Pain Symptom Manage 22 (2): 672-87, 2001.  [PUBMED Abstract]

  77. Indelicato RA, Portenoy RK: Opioid rotation in the management of refractory cancer pain. J Clin Oncol 21 (Suppl 9): 87-91, 2003. 

  78. Rauck RL, Cherry D, Boyer MF, et al.: Long-term intrathecal opioid therapy with a patient-activated, implanted delivery system for the treatment of refractory cancer pain. J Pain 4 (8): 441-7, 2003.  [PUBMED Abstract]

  79. Morita T, Takigawa C, Onishi H, et al.: Opioid rotation from morphine to fentanyl in delirious cancer patients: an open-label trial. J Pain Symptom Manage 30 (1): 96-103, 2005.  [PUBMED Abstract]

  80. Riley J, Ross JR, Rutter D, et al.: No pain relief from morphine? Individual variation in sensitivity to morphine and the need to switch to an alternative opioid in cancer patients. Support Care Cancer 14 (1): 56-64, 2006.  [PUBMED Abstract]

  81. Fallon M: Opioid rotation: does it have a role? Palliat Med 11 (3): 177-8, 1997.  [PUBMED Abstract]

  82. Mercadante S: Pathophysiology and treatment of opioid-related myoclonus in cancer patients. Pain 74 (1): 5-9, 1998.  [PUBMED Abstract]

  83. Mercadante S: Opioid rotation for cancer pain: rationale and clinical aspects. Cancer 86 (9): 1856-66, 1999.  [PUBMED Abstract]

  84. Hanks GW, Hoskin PJ, Aherne GW, et al.: Explanation for potency of repeated oral doses of morphine? Lancet 2 (8561): 723-5, 1987.  [PUBMED Abstract]

  85. Pasternak GW, Bodnar RJ, Clark JA, et al.: Morphine-6-glucuronide, a potent mu agonist. Life Sci 41 (26): 2845-9, 1987.  [PUBMED Abstract]

  86. Potter JM, Reid DB, Shaw RJ, et al.: Myoclonus associated with treatment with high doses of morphine: the role of supplemental drugs. BMJ 299 (6692): 150-3, 1989.  [PUBMED Abstract]

  87. Sear JW, Hand CW, Moore RA, et al.: Studies on morphine disposition: influence of general anaesthesia on plasma concentrations of morphine and its metabolites. Br J Anaesth 62 (1): 22-7, 1989.  [PUBMED Abstract]

  88. Glare PA, Walsh TD, Pippenger CE: Normorphine, a neurotoxic metabolite? Lancet 335 (8691): 725-6, 1990.  [PUBMED Abstract]

  89. McQuay HJ, Carroll D, Faura CC, et al.: Oral morphine in cancer pain: influences on morphine and metabolite concentration. Clin Pharmacol Ther 48 (3): 236-44, 1990.  [PUBMED Abstract]

  90. Hagen NA, Foley KM, Cerbone DJ, et al.: Chronic nausea and morphine-6-glucuronide. J Pain Symptom Manage 6 (3): 125-8, 1991.  [PUBMED Abstract]

  91. Sjøgren P, Dragsted L, Christensen CB: Myoclonic spasms during treatment with high doses of intravenous morphine in renal failure. Acta Anaesthesiol Scand 37 (8): 780-2, 1993.  [PUBMED Abstract]

  92. Bowsher D: Paradoxical pain. BMJ 306 (6876): 473-4, 1993.  [PUBMED Abstract]

  93. Goucke CR, Hackett LP, Ilett KF: Concentrations of morphine, morphine-6-glucuronide and morphine-3-glucuronide in serum and cerebrospinal fluid following morphine administration to patients with morphine-resistant pain. Pain 56 (2): 145-9, 1994.  [PUBMED Abstract]

  94. Smith M, Cramond T: Comments on Goucke et al., PAIN, 56 (1994) 145-149. Pain 59 (1): 155-6, 1994.  [PUBMED Abstract]

  95. Smith GD, Smith MT: Morphine-3-glucuronide: evidence to support its putative role in the development of tolerance to the antinociceptive effects of morphine in the rat. Pain 62 (1): 51-60, 1995.  [PUBMED Abstract]

  96. Tiseo PJ, Thaler HT, Lapin J, et al.: Morphine-6-glucuronide concentrations and opioid-related side effects: a survey in cancer patients. Pain 61 (1): 47-54, 1995.  [PUBMED Abstract]

  97. Nugent M, Davis C, Brooks D, et al.: Long-term observations of patients receiving transdermal fentanyl after a randomized trial. J Pain Symptom Manage 21 (5): 385-91, 2001.  [PUBMED Abstract]

  98. Muijsers RB, Wagstaff AJ: Transdermal fentanyl: an updated review of its pharmacological properties and therapeutic efficacy in chronic cancer pain control. Drugs 61 (15): 2289-307, 2001.  [PUBMED Abstract]

  99. Solassol I, Caumette L, Bressolle F, et al.: Inter- and intra-individual variability in transdermal fentanyl absorption in cancer pain patients. Oncol Rep 14 (4): 1029-36, 2005.  [PUBMED Abstract]

  100. Payne R, Coluzzi P, Hart L, et al.: Long-term safety of oral transmucosal fentanyl citrate for breakthrough cancer pain. J Pain Symptom Manage 22 (1): 575-83, 2001.  [PUBMED Abstract]

  101. Hanks GW, Nugent M, Higgs CM, et al.: Oral transmucosal fentanyl citrate in the management of breakthrough pain in cancer: an open, multicentre, dose-titration and long-term use study. Palliat Med 18 (8): 698-704, 2004.  [PUBMED Abstract]

  102. Christie JM, Simmonds M, Patt R, et al.: Dose-titration, multicenter study of oral transmucosal fentanyl citrate for the treatment of breakthrough pain in cancer patients using transdermal fentanyl for persistent pain. J Clin Oncol 16 (10): 3238-45, 1998.  [PUBMED Abstract]

  103. Portenoy RK, Payne R, Coluzzi P, et al.: Oral transmucosal fentanyl citrate (OTFC) for the treatment of breakthrough pain in cancer patients: a controlled dose titration study. Pain 79 (2-3): 303-12, 1999.  [PUBMED Abstract]

  104. Portenoy RK, Taylor D, Messina J, et al.: A randomized, placebo-controlled study of fentanyl buccal tablet for breakthrough pain in opioid-treated patients with cancer. Clin J Pain 22 (9): 805-11, 2006 Nov-Dec.  [PUBMED Abstract]

  105. Elsner F, Radbruch L, Loick G, et al.: Intravenous versus subcutaneous morphine titration in patients with persisting exacerbation of cancer pain. J Palliat Med 8 (4): 743-50, 2005.  [PUBMED Abstract]

  106. Dudgeon DJ, Lertzman M: Dyspnea in the advanced cancer patient. J Pain Symptom Manage 16 (4): 212-9, 1998.  [PUBMED Abstract]

  107. Davis C: The role of nebulised drugs in palliating respiratory symptoms of malignant disease. European Journal of Palliative Care 2(1): 9-15, 1995. 

  108. Smith TJ, Coyne PJ, Staats PS, et al.: An implantable drug delivery system (IDDS) for refractory cancer pain provides sustained pain control, less drug-related toxicity, and possibly better survival compared with comprehensive medical management (CMM). Ann Oncol 16 (5): 825-33, 2005.  [PUBMED Abstract]

  109. Smith TJ, Coyne PJ: Implantable drug delivery systems (IDDS) after failure of comprehensive medical management (CMM) can palliate symptoms in the most refractory cancer pain patients. J Palliat Med 8 (4): 736-42, 2005.  [PUBMED Abstract]

  110. Smith TJ, Staats PS, Deer T, et al.: Randomized clinical trial of an implantable drug delivery system compared with comprehensive medical management for refractory cancer pain: impact on pain, drug-related toxicity, and survival. J Clin Oncol 20 (19): 4040-9, 2002.  [PUBMED Abstract]

  111. Kalso E, Heiskanen T, Rantio M, et al.: Epidural and subcutaneous morphine in the management of cancer pain: a double-blind cross-over study. Pain 67 (2-3): 443-9, 1996.  [PUBMED Abstract]

  112. Derby S, Portenoy RK: Assessment and management of opioid-induced constipation. In: Portenoy RK, Bruera E, eds.: Topics in Palliative Care. Volume 1. New York, NY: Oxford University Press, 1997, pp 95-112. 

  113. Bruera E, Suarez-Almazor M, Velasco A, et al.: The assessment of constipation in terminal cancer patients admitted to a palliative care unit: a retrospective review. J Pain Symptom Manage 9 (8): 515-9, 1994.  [PUBMED Abstract]

  114. Mancini I, Bruera E: Constipation in advanced cancer patients. Support Care Cancer 6 (4): 356-64, 1998.  [PUBMED Abstract]

  115. Payne R, Mathias SD, Pasta DJ, et al.: Quality of life and cancer pain: satisfaction and side effects with transdermal fentanyl versus oral morphine. J Clin Oncol 16 (4): 1588-93, 1998.  [PUBMED Abstract]

  116. Daeninck PJ, Bruera E: Reduction in constipation and laxative requirements following opioid rotation to methadone: a report of four cases. J Pain Symptom Manage 18 (4): 303-9, 1999.  [PUBMED Abstract]

  117. Bruera E, Brenneis C, Michaud M, et al.: Continuous Sc infusion of metoclopramide for treatment of narcotic bowel syndrome. Cancer Treat Rep 71 (11): 1121-2, 1987.  [PUBMED Abstract]

  118. Meissner W, Schmidt U, Hartmann M, et al.: Oral naloxone reverses opioid-associated constipation. Pain 84 (1): 105-9, 2000.  [PUBMED Abstract]

  119. Sykes NP: An investigation of the ability of oral naloxone to correct opioid-related constipation in patients with advanced cancer. Palliat Med 10 (2): 135-44, 1996.  [PUBMED Abstract]

  120. Foss JF: A review of the potential role of methylnaltrexone in opioid bowel dysfunction. Am J Surg 182 (5A Suppl): 19S-26S, 2001.  [PUBMED Abstract]

  121. Moulin DE, Iezzi A, Amireh R, et al.: Randomised trial of oral morphine for chronic non-cancer pain. Lancet 347 (8995): 143-7, 1996.  [PUBMED Abstract]

  122. Aparasu R, McCoy RA, Weber C, et al.: Opioid-induced emesis among hospitalized nonsurgical patients: effect on pain and quality of life. J Pain Symptom Manage 18 (4): 280-8, 1999.  [PUBMED Abstract]

  123. Hanks G, Cherny N: Opioid analgesic therapy. In: Doyle D, Hanks GW, MacDonald N, eds.: Oxford Textbook of Palliative Medicine. 2nd ed. New York, NY: Oxford University Press, 1998, pp 331-355. 

  124. Mannix KA: Palliation of nausea and vomiting. In: Doyle D, Hanks GW, MacDonald N, eds.: Oxford Textbook of Palliative Medicine. 2nd ed. New York, NY: Oxford University Press, 1998, pp 489-499. 

  125. Bruera E, Seifert L, Watanabe S, et al.: Chronic nausea in advanced cancer patients: a retrospective assessment of a metoclopramide-based antiemetic regimen. J Pain Symptom Manage 11 (3): 147-53, 1996.  [PUBMED Abstract]

  126. Pereira J, Bruera E: Successful management of intractable nausea with ondansetron: a case study. J Palliat Care 12 (2): 47-50, 1996 Summer.  [PUBMED Abstract]

  127. Kalso E, Vainio A: Morphine and oxycodone hydrochloride in the management of cancer pain. Clin Pharmacol Ther 47 (5): 639-46, 1990.  [PUBMED Abstract]

  128. Cherny NJ, Chang V, Frager G, et al.: Opioid pharmacotherapy in the management of cancer pain: a survey of strategies used by pain physicians for the selection of analgesic drugs and routes of administration. Cancer 76 (7): 1283-93, 1995.  [PUBMED Abstract]

  129. Maddocks I, Somogyi A, Abbott F, et al.: Attenuation of morphine-induced delirium in palliative care by substitution with infusion of oxycodone. J Pain Symptom Manage 12 (3): 182-9, 1996.  [PUBMED Abstract]

  130. Drexel H, Dzien A, Spiegel RW, et al.: Treatment of severe cancer pain by low-dose continuous subcutaneous morphine. Pain 36 (2): 169-76, 1989.  [PUBMED Abstract]

  131. Pereira J, Bruera E: Chronic nausea. In: Bruera E, Higginson I, eds.: Cachexia-Anorexia in Cancer Patients. New York, NY: Oxford University Press, 1996, pp 23-37. 

  132. Mao J: Opioid-induced abnormal pain sensitivity: implications in clinical opioid therapy. Pain 100 (3): 213-7, 2002.  [PUBMED Abstract]

  133. Mercadante S, Ferrera P, Villari P, et al.: Hyperalgesia: an emerging iatrogenic syndrome. J Pain Symptom Manage 26 (2): 769-75, 2003.  [PUBMED Abstract]

  134. Lawlor PG, Bruera E: Side-effects of opioids in chronic pain treatment. Current Opinion in Anaesthesiology 11 (5): 539-45, 1998. 

  135. Hagen N, Swanson R: Strychnine-like multifocal myoclonus and seizures in extremely high-dose opioid administration: treatment strategies. J Pain Symptom Manage 14 (1): 51-8, 1997.  [PUBMED Abstract]

  136. Daeninck PJ, Bruera E: Opioid use in cancer pain. Is a more liberal approach enhancing toxicity? Acta Anaesthesiol Scand 43 (9): 924-38, 1999.  [PUBMED Abstract]

  137. Vainio A, Ollila J, Matikainen E, et al.: Driving ability in cancer patients receiving long-term morphine analgesia. Lancet 346 (8976): 667-70, 1995.  [PUBMED Abstract]

  138. Sjogren P, Thomsen AB, Olsen AK: Impaired neuropsychological performance in chronic nonmalignant pain patients receiving long-term oral opioid therapy. J Pain Symptom Manage 19 (2): 100-8, 2000.  [PUBMED Abstract]

  139. Zacny JP, Lichtor JL, Flemming D, et al.: A dose-response analysis of the subjective, psychomotor and physiological effects of intravenous morphine in healthy volunteers. J Pharmacol Exp Ther 268 (1): 1-9, 1994.  [PUBMED Abstract]

  140. Ersek M, Cherrier MM, Overman SS, et al.: The cognitive effects of opioids. Pain Manag Nurs 5 (2): 75-93, 2004.  [PUBMED Abstract]

  141. Bruera E, Macmillan K, Hanson J, et al.: The cognitive effects of the administration of narcotic analgesics in patients with cancer pain. Pain 39 (1): 13-6, 1989.  [PUBMED Abstract]

  142. Galski T, Williams JB, Ehle HT: Effects of opioids on driving ability. J Pain Symptom Manage 19 (3): 200-8, 2000.  [PUBMED Abstract]

  143. Flacker JM, Cummings V, Mach JR Jr, et al.: The association of serum anticholinergic activity with delirium in elderly medical patients. Am J Geriatr Psychiatry 6 (1): 31-41, 1998 Winter.  [PUBMED Abstract]

  144. Mach JR Jr, Dysken MW, Kuskowski M, et al.: Serum anticholinergic activity in hospitalized older persons with delirium: a preliminary study. J Am Geriatr Soc 43 (5): 491-5, 1995.  [PUBMED Abstract]

  145. Mussi C, Ferrari R, Ascari S, et al.: Importance of serum anticholinergic activity in the assessment of elderly patients with delirium. J Geriatr Psychiatry Neurol 12 (2): 82-6, 1999 Summer.  [PUBMED Abstract]

  146. Eisendrath SJ, Goldman B, Douglas J, et al.: Meperidine-induced delirium. Am J Psychiatry 144 (8): 1062-5, 1987.  [PUBMED Abstract]

  147. Marcantonio ER, Juarez G, Goldman L, et al.: The relationship of postoperative delirium with psychoactive medications. JAMA 272 (19): 1518-22, 1994.  [PUBMED Abstract]

  148. Bartlett SE, Cramond T, Smith MT: The excitatory effects of morphine-3-glucuronide are attenuated by LY274614, a competitive NMDA receptor antagonist, and by midazolam, an agonist at the benzodiazepine site on the GABAA receptor complex. Life Sci 54 (10): 687-94, 1994.  [PUBMED Abstract]

  149. Smith GD, Smith MT: The excitatory behavioral and antianalgesic pharmacology of normorphine-3-glucuronide after intracerebroventricular administration to rats. J Pharmacol Exp Ther 285 (3): 1157-62, 1998.  [PUBMED Abstract]

  150. Wright AW, Nocente ML, Smith MT: Hydromorphone-3-glucuronide: biochemical synthesis and preliminary pharmacological evaluation. Life Sci 63 (5): 401-11, 1998.  [PUBMED Abstract]

  151. Ashby M, Fleming B, Wood M, et al.: Plasma morphine and glucuronide (M3G and M6G) concentrations in hospice inpatients. J Pain Symptom Manage 14 (3): 157-67, 1997.  [PUBMED Abstract]

  152. Faura CC, Collins SL, Moore RA, et al.: Systematic review of factors affecting the ratios of morphine and its major metabolites. Pain 74 (1): 43-53, 1998.  [PUBMED Abstract]

  153. Osborne R, Joel S, Grebenik K, et al.: The pharmacokinetics of morphine and morphine glucuronides in kidney failure. Clin Pharmacol Ther 54 (2): 158-67, 1993.  [PUBMED Abstract]

  154. Lawlor PG, Gagnon B, Mancini IL, et al.: Occurrence, causes, and outcome of delirium in patients with advanced cancer: a prospective study. Arch Intern Med 160 (6): 786-94, 2000.  [PUBMED Abstract]

  155. Fainsinger RL, Tapper M, Bruera E: A perspective on the management of delirium in terminally ill patients on a palliative care unit. J Palliat Care 9 (3): 4-8, 1993 Autumn.  [PUBMED Abstract]

  156. Bruera E, Schoeller T, Wenk R, et al.: A prospective multicenter assessment of the Edmonton staging system for cancer pain. J Pain Symptom Manage 10 (5): 348-55, 1995.  [PUBMED Abstract]

  157. Lipowski ZJ: Delirium (acute confusional states). JAMA 258 (13): 1789-92, 1987.  [PUBMED Abstract]

  158. Breitbart W, Marotta R, Platt MM, et al.: A double-blind trial of haloperidol, chlorpromazine, and lorazepam in the treatment of delirium in hospitalized AIDS patients. Am J Psychiatry 153 (2): 231-7, 1996.  [PUBMED Abstract]

  159. Breitbart W, Bruera E, Chochinov H, et al.: Neuropsychiatric syndromes and psychological symptoms in patients with advanced cancer. J Pain Symptom Manage 10 (2): 131-41, 1995.  [PUBMED Abstract]

  160. Burke AL, Diamond PL, Hulbert J, et al.: Terminal restlessness--its management and the role of midazolam. Med J Aust 155 (7): 485-7, 1991.  [PUBMED Abstract]

  161. Khojainova N, Santiago-Palma J, Kornick C, et al.: Olanzapine in the management of cancer pain. J Pain Symptom Manage 23 (4): 346-50, 2002.  [PUBMED Abstract]

  162. Enting RH, Oldenmenger WH, van der Rijt CC, et al.: A prospective study evaluating the response of patients with unrelieved cancer pain to parenteral opioids. Cancer 94 (11): 3049-56, 2002.  [PUBMED Abstract]

  163. Portenoy RK, Lesage P: Management of cancer pain. Lancet 353 (9165): 1695-700, 1999.  [PUBMED Abstract]

  164. Portenoy RK: Tolerance to opioid analgesics: clinical aspects. Cancer Surv 21: 49-65, 1994.  [PUBMED Abstract]

  165. de Stoutz ND, Bruera E, Suarez-Almazor M: Opioid rotation for toxicity reduction in terminal cancer patients. J Pain Symptom Manage 10 (5): 378-84, 1995.  [PUBMED Abstract]

  166. Lawlor P, Turner K, Hanson J, et al.: Dose ratio between morphine and hydromorphone in patients with cancer pain: a retrospective study. Pain 72 (1-2): 79-85, 1997.  [PUBMED Abstract]

  167. Bruera E, Franco JJ, Maltoni M, et al.: Changing pattern of agitated impaired mental status in patients with advanced cancer: association with cognitive monitoring, hydration, and opioid rotation. J Pain Symptom Manage 10 (4): 287-91, 1995.  [PUBMED Abstract]

  168. Bruera E, Miller MJ, Macmillan K, et al.: Neuropsychological effects of methylphenidate in patients receiving a continuous infusion of narcotics for cancer pain. Pain 48 (2): 163-6, 1992.  [PUBMED Abstract]

  169. Bruera E, Fainsinger R, MacEachern T, et al.: The use of methylphenidate in patients with incident cancer pain receiving regular opiates. A preliminary report. Pain 50 (1): 75-7, 1992.  [PUBMED Abstract]

  170. Stiefel F, Bruera E: Psychostimulants for hypoactive-hypoalert delirium? J Palliat Care 7 (3): 25-6, 1991 Autumn.  [PUBMED Abstract]

  171. Bruera E, Strasser F, Shen L, et al.: The effect of donepezil on sedation and other symptoms in patients receiving opioids for cancer pain: a pilot study. J Pain Symptom Manage 26 (5): 1049-54, 2003.  [PUBMED Abstract]

  172. Estfan B, Mahmoud F, Shaheen P, et al.: Respiratory function during parenteral opioid titration for cancer pain. Palliat Med 21 (2): 81-6, 2007.  [PUBMED Abstract]

  173. O'Mahony S, Coyle N, Payne R: Current management of opioid-related side effects. Oncology (Huntingt) 15 (1): 61-73, 77; discussion 77-8, 80-2, 2001.  [PUBMED Abstract]

  174. De Leon G, Wexler HK: Heroin addiction: its relation to sexual behavior and sexual experience. J Abnorm Psychol 81 (1): 36-8, 1973.  [PUBMED Abstract]

  175. Pelosi MA, Sama JC, Caterini H, et al.: Galactorrhea-amenorrhea syndrome associated with heroin addiction. Am J Obstet Gynecol 118 (7): 966-70, 1974.  [PUBMED Abstract]

  176. Cicero TJ, Bell RD, Wiest WG, et al.: Function of the male sex organs in heroin and methadone users. N Engl J Med 292 (17): 882-7, 1975.  [PUBMED Abstract]

  177. Wang C, Chan V, Yeung RT: The effect of heroin addiction on pituitary-testicular function. Clin Endocrinol (Oxf) 9 (5): 455-61, 1978.  [PUBMED Abstract]

  178. Chan V, Wang C, Yeung RT: Effects of heroin addiction on thyrotrophin, thyroid hormones and porlactin secretion in men. Clin Endocrinol (Oxf) 10 (6): 557-65, 1979.  [PUBMED Abstract]

  179. Spagnolli W, Torboli P, Mattarei M, et al.: Calcitonin and prolactin serum levels in heroin addicts: study on a methadone treated group. Drug Alcohol Depend 20 (2): 143-8, 1987.  [PUBMED Abstract]

  180. Paice JA, Penn RD, Ryan WG: Altered sexual function and decreased testosterone in patients receiving intraspinal opioids. J Pain Symptom Manage 9 (2): 126-31, 1994.  [PUBMED Abstract]

  181. Paice JA, Penn RD: Amenorrhea associated with intraspinal morphine. J Pain Symptom Manage 10 (8): 582-3, 1995.  [PUBMED Abstract]

  182. Lemieux L, Kaiser S, Pereira J, et al.: Sexuality in palliative care: patient perspectives. Palliat Med 18 (7): 630-7, 2004.  [PUBMED Abstract]

  183. Mercadante S, Portenoy RK: Opioid poorly-responsive cancer pain. Part 3. Clinical strategies to improve opioid responsiveness. J Pain Symptom Manage 21 (4): 338-54, 2001.  [PUBMED Abstract]

  184. Davis MP, Homsi J: The importance of cytochrome P450 monooxygenase CYP2D6 in palliative medicine. Support Care Cancer 9 (6): 442-51, 2001.  [PUBMED Abstract]

  185. Meuser T, Pietruck C, Radbruch L, et al.: Symptoms during cancer pain treatment following WHO-guidelines: a longitudinal follow-up study of symptom prevalence, severity and etiology. Pain 93 (3): 247-57, 2001.  [PUBMED Abstract]

  186. Kloke M, Rapp M, Bosse B, et al.: Toxicity and/or insufficient analgesia by opioid therapy: risk factors and the impact of changing the opioid. A retrospective analysis of 273 patients observed at a single center. Support Care Cancer 8 (6): 479-86, 2000.  [PUBMED Abstract]

  187. Portenoy RK, Frager G: Pain management: pharmacological approaches. In: von Gunten CF, ed.: Palliative Care and Rehabilitation of Cancer Patients. Boston, Mass: Kluwer Academic Publishers, 1999, pp 1-29. 

  188. Portenoy RK: Adjuvant analgesic agents. Hematol Oncol Clin North Am 10 (1): 103-19, 1996.  [PUBMED Abstract]

  189. Breitbart W: Psychotropic adjuvant analgesics for pain in cancer and AIDS. Psychooncology 7 (4): 333-45, 1998 Jul-Aug.  [PUBMED Abstract]

  190. Guay DR: Adjunctive agents in the management of chronic pain. Pharmacotherapy 21 (9): 1070-81, 2001.  [PUBMED Abstract]

  191. Kalso E, Tasmuth T, Neuvonen PJ: Amitriptyline effectively relieves neuropathic pain following treatment of breast cancer. Pain 64 (2): 293-302, 1996.  [PUBMED Abstract]

  192. Mercadante S, Arcuri E, Tirelli W, et al.: Amitriptyline in neuropathic cancer pain in patients on morphine therapy: a randomized placebo-controlled, double-blind crossover study. Tumori 88 (3): 239-42, 2002 May-Jun.  [PUBMED Abstract]

  193. Leijon G, Boivie J: Central post-stroke pain--a controlled trial of amitriptyline and carbamazepine. Pain 36 (1): 27-36, 1989.  [PUBMED Abstract]

  194. Holland JC, Romano SJ, Heiligenstein JH, et al.: A controlled trial of fluoxetine and desipramine in depressed women with advanced cancer. Psychooncology 7 (4): 291-300, 1998 Jul-Aug.  [PUBMED Abstract]

  195. Max MB, Kishore-Kumar R, Schafer SC, et al.: Efficacy of desipramine in painful diabetic neuropathy: a placebo-controlled trial. Pain 45 (1): 3-9; discussion 1-2, 1991.  [PUBMED Abstract]

  196. Vrethem M, Boivie J, Arnqvist H, et al.: A comparison a amitriptyline and maprotiline in the treatment of painful polyneuropathy in diabetics and nondiabetics. Clin J Pain 13 (4): 313-23, 1997.  [PUBMED Abstract]

  197. Raskin J, Pritchett YL, Wang F, et al.: A double-blind, randomized multicenter trial comparing duloxetine with placebo in the management of diabetic peripheral neuropathic pain. Pain Med 6 (5): 346-56, 2005 Sep-Oct.  [PUBMED Abstract]

  198. Raja SN, Haythornthwaite JA, Pappagallo M, et al.: Opioids versus antidepressants in postherpetic neuralgia: a randomized, placebo-controlled trial. Neurology 59 (7): 1015-21, 2002.  [PUBMED Abstract]

  199. Tasmuth T, Härtel B, Kalso E: Venlafaxine in neuropathic pain following treatment of breast cancer. Eur J Pain 6 (1): 17-24, 2002.  [PUBMED Abstract]

  200. Reuben SS, Makari-Judson G, Lurie SD: Evaluation of efficacy of the perioperative administration of venlafaxine XR in the prevention of postmastectomy pain syndrome. J Pain Symptom Manage 27 (2): 133-9, 2004.  [PUBMED Abstract]

  201. Rowbotham MC, Goli V, Kunz NR, et al.: Venlafaxine extended release in the treatment of painful diabetic neuropathy: a double-blind, placebo-controlled study. Pain 110 (3): 697-706, 2004.  [PUBMED Abstract]

  202. Harke H, Gretenkort P, Ladleif HU, et al.: The response of neuropathic pain and pain in complex regional pain syndrome I to carbamazepine and sustained-release morphine in patients pretreated with spinal cord stimulation: a double-blinded randomized study. Anesth Analg 92 (2): 488-95, 2001.  [PUBMED Abstract]

  203. Kochar DK, Garg P, Bumb RA, et al.: Divalproex sodium in the management of post-herpetic neuralgia: a randomized double-blind placebo-controlled study. QJM 98 (1): 29-34, 2005.  [PUBMED Abstract]

  204. Caraceni A, Zecca E, Bonezzi C, et al.: Gabapentin for neuropathic cancer pain: a randomized controlled trial from the Gabapentin Cancer Pain Study Group. J Clin Oncol 22 (14): 2909-17, 2004.  [PUBMED Abstract]

  205. Ross JR, Goller K, Hardy J, et al.: Gabapentin is effective in the treatment of cancer-related neuropathic pain: a prospective, open-label study. J Palliat Med 8 (6): 1118-26, 2005.  [PUBMED Abstract]

  206. Levendoglu F, Ogün CO, Ozerbil O, et al.: Gabapentin is a first line drug for the treatment of neuropathic pain in spinal cord injury. Spine 29 (7): 743-51, 2004.  [PUBMED Abstract]

  207. Hugel H, Ellershaw JE, Dickman A: Clonazepam as an adjuvant analgesic in patients with cancer-related neuropathic pain. J Pain Symptom Manage 26 (6): 1073-4, 2003.  [PUBMED Abstract]

  208. Simpson DM, McArthur JC, Olney R, et al.: Lamotrigine for HIV-associated painful sensory neuropathies: a placebo-controlled trial. Neurology 60 (9): 1508-14, 2003.  [PUBMED Abstract]

  209. Lesser H, Sharma U, LaMoreaux L, et al.: Pregabalin relieves symptoms of painful diabetic neuropathy: a randomized controlled trial. Neurology 63 (11): 2104-10, 2004.  [PUBMED Abstract]

  210. Oskarsson P, Ljunggren JG, Lins PE: Efficacy and safety of mexiletine in the treatment of painful diabetic neuropathy. The Mexiletine Study Group. Diabetes Care 20 (10): 1594-7, 1997.  [PUBMED Abstract]

  211. Meier T, Wasner G, Faust M, et al.: Efficacy of lidocaine patch 5% in the treatment of focal peripheral neuropathic pain syndromes: a randomized, double-blind, placebo-controlled study. Pain 106 (1-2): 151-8, 2003.  [PUBMED Abstract]

  212. Polascik TJ, Given RW, Metzger C, et al.: Open-label trial evaluating the safety and efficacy of zoledronic acid in preventing bone loss in patients with hormone-sensitive prostate cancer and bone metastases. Urology 66 (5): 1054-9, 2005.  [PUBMED Abstract]

  213. Dapas F, Hartman SF, Martinez L, et al.: Baclofen for the treatment of acute low-back syndrome. A double-blind comparison with placebo. Spine 10 (4): 345-9, 1985.  [PUBMED Abstract]

  214. Lavand'homme PM, Eisenach JC: Perioperative administration of the alpha2-adrenoceptor agonist clonidine at the site of nerve injury reduces the development of mechanical hypersensitivity and modulates local cytokine expression. Pain 105 (1-2): 247-54, 2003.  [PUBMED Abstract]

  215. Bruera E, Chadwick S, Brenneis C, et al.: Methylphenidate associated with narcotics for the treatment of cancer pain. Cancer Treat Rep 71 (1): 67-70, 1987.  [PUBMED Abstract]

  216. Cantello R, Aguggia M, Gilli M, et al.: Analgesic action of methylphenidate on parkinsonian sensory symptoms. Mechanisms and pathophysiological implications. Arch Neurol 45 (9): 973-6, 1988.  [PUBMED Abstract]

  217. George RM, Ahmedzai SH: The management of neuropathic pain in cancer: clinical guidelines for the use of adjuvant analgesics. Indian J Cancer 37 (1): 4-9, 2000.  [PUBMED Abstract]

  218. Caraceni A, Zecca E, Martini C, et al.: Differences in gabapentin efficacy for cancer pain more apparent than real? J Pain Symptom Manage 21 (2): 93-4, 2001.  [PUBMED Abstract]

  219. Chandler A, Williams JE: Gabapentin, an adjuvant treatment for neuropathic pain in a cancer hospital. J Pain Symptom Manage 20 (2): 82-6, 2000.  [PUBMED Abstract]

  220. Caraceni A, Zecca E, Martini C, et al.: Gabapentin as an adjuvant to opioid analgesia for neuropathic cancer pain. J Pain Symptom Manage 17 (6): 441-5, 1999.  [PUBMED Abstract]

  221. Oneschuk D, al-Shahri MZ: The pattern of gabapentin use in a tertiary palliative care unit. J Palliat Care 19 (3): 185-7, 2003 Fall.  [PUBMED Abstract]

  222. Keskinbora K, Pekel AF, Aydinli I: Gabapentin and an opioid combination versus opioid alone for the management of neuropathic cancer pain: a randomized open trial. J Pain Symptom Manage 34 (2): 183-9, 2007.  [PUBMED Abstract]

  223. Watanabe S, Bruera E: Corticosteroids as adjuvant analgesics. J Pain Symptom Manage 9 (7): 442-5, 1994.  [PUBMED Abstract]

  224. Lussier D, Huskey AG, Portenoy RK: Adjuvant analgesics in cancer pain management. Oncologist 9 (5): 571-91, 2004.  [PUBMED Abstract]

  225. Hardy J, Ling J, Mansi J, et al.: Pitfalls in placebo-controlled trials in palliative care: dexamethasone for the palliation of malignant bowel obstruction. Palliat Med 12 (6): 437-42, 1998.  [PUBMED Abstract]

  226. Hardy J: Corticosteroids in palliative care. European Journal of Palliative Care 5(2): 46-50, 1998. 

  227. Feuer DJ, Broadley KE: Corticosteroids for the resolution of malignant bowel obstruction in advanced gynaecological and gastrointestinal cancer. Cochrane Database Syst Rev (2): CD001219, 2000.  [PUBMED Abstract]

  228. Wooldridge JE, Anderson CM, Perry MC: Corticosteroids in advanced cancer. Oncology (Huntingt) 15 (2): 225-34; discussion 234-6, 2001.  [PUBMED Abstract]

  229. Ripamonti C, Fulfaro F: Malignant bone pain: pathophysiology and treatments. Curr Rev Pain 4 (3): 187-96, 2000.  [PUBMED Abstract]

  230. Ripamonti C, Fulfaro F: Pathogenesis and pharmacological treatment of bone pain in skeletal metastases. Q J Nucl Med 45 (1): 65-77, 2001.  [PUBMED Abstract]

  231. McDonnell FJ, Sloan JW, Hamann SR: Advances in cancer pain management. Curr Pain Headache Rep 5 (3): 265-71, 2001.  [PUBMED Abstract]

  232. Rodrigues P, Hering F, Campagnari JC: Use of bisphosphonates can dramatically improve pain in advanced hormone-refractory prostate cancer patients. Prostate Cancer Prostatic Dis 7 (4): 350-4, 2004.  [PUBMED Abstract]

  233. Diel IJ, Body JJ, Lichinitser MR, et al.: Improved quality of life after long-term treatment with the bisphosphonate ibandronate in patients with metastatic bone disease due to breast cancer. Eur J Cancer 40 (11): 1704-12, 2004.  [PUBMED Abstract]

  234. Lucas LK, Lipman AG: Recent advances in pharmacotherapy for cancer pain management. Cancer Pract 10 (Suppl 1): S14-20, 2002 May-Jun.  [PUBMED Abstract]

  235. Weinfurt KP, Anstrom KJ, Castel LD, et al.: Effect of zoledronic acid on pain associated with bone metastasis in patients with prostate cancer. Ann Oncol 17 (6): 986-9, 2006.  [PUBMED Abstract]

  236. Small EJ, Smith MR, Seaman JJ, et al.: Combined analysis of two multicenter, randomized, placebo-controlled studies of pamidronate disodium for the palliation of bone pain in men with metastatic prostate cancer. J Clin Oncol 21 (23): 4277-84, 2003.  [PUBMED Abstract]

  237. Berenson JR, Rosen LS, Howell A, et al.: Zoledronic acid reduces skeletal-related events in patients with osteolytic metastases. Cancer 91 (7): 1191-200, 2001.  [PUBMED Abstract]

  238. Rosen LS, Gordon D, Antonio BS, et al.: Zoledronic acid versus pamidronate in the treatment of skeletal metastases in patients with breast cancer or osteolytic lesions of multiple myeloma: a phase III, double-blind, comparative trial. Cancer J 7 (5): 377-87, 2001 Sep-Oct.  [PUBMED Abstract]

  239. Rosen LS, Gordon D, Tchekmedyian S, et al.: Zoledronic acid versus placebo in the treatment of skeletal metastases in patients with lung cancer and other solid tumors: a phase III, double-blind, randomized trial--the Zoledronic Acid Lung Cancer and Other Solid Tumors Study Group. J Clin Oncol 21 (16): 3150-7, 2003.  [PUBMED Abstract]

  240. Saad F, Gleason DM, Murray R, et al.: A randomized, placebo-controlled trial of zoledronic acid in patients with hormone-refractory metastatic prostate carcinoma. J Natl Cancer Inst 94 (19): 1458-68, 2002.  [PUBMED Abstract]

  241. Neville-Webbe H, Coleman RE: The use of zoledronic acid in the management of metastatic bone disease and hypercalcaemia. Palliat Med 17 (6): 539-53, 2003.  [PUBMED Abstract]

  242. Body JJ, Diel IJ, Bell R, et al.: Oral ibandronate improves bone pain and preserves quality of life in patients with skeletal metastases due to breast cancer. Pain 111 (3): 306-12, 2004.  [PUBMED Abstract]

  243. US Food and Drug Administration.: FDA Alert (January 7, 2008): Information on Bisphosphonates (marketed as Actonel, Actonel+Ca, Aredia, Boniva, Didronel, Fosamax, Fosamax+D, Reclast, Skelid, and Zometa). Rockville, Md: Food and Drug Administration, Center for Drug Evaluation and Research, 2008. Available online. Last accessed July 7, 2008. 

  244. Martinez MJ, Roqué M, Alonso-Coello P, et al.: Calcitonin for metastatic bone pain. Cochrane Database Syst Rev (3): CD003223, 2003.  [PUBMED Abstract]

  245. Jackson K, Ashby M, Martin P, et al.: "Burst" ketamine for refractory cancer pain: an open-label audit of 39 patients. J Pain Symptom Manage 22 (4): 834-42, 2001.  [PUBMED Abstract]

  246. Lossignol DA, Obiols-Portis M, Body JJ: Successful use of ketamine for intractable cancer pain. Support Care Cancer 13 (3): 188-93, 2005.  [PUBMED Abstract]

  247. Bell R, Eccleston C, Kalso E: Ketamine as an adjuvant to opioids for cancer pain. Cochrane Database Syst Rev (1): CD003351, 2003.  [PUBMED Abstract]

  248. Bell RF, Eccleston C, Kalso E: Ketamine as adjuvant to opioids for cancer pain. A qualitative systematic review. J Pain Symptom Manage 26 (3): 867-75, 2003.  [PUBMED Abstract]

  249. Shimoyama N, Shimoyama M, Elliott KJ, et al.: d-Methadone is antinociceptive in the rat formalin test. J Pharmacol Exp Ther 283 (2): 648-52, 1997.  [PUBMED Abstract]

  250. Elliott K, Hynansky A, Inturrisi CE: Dextromethorphan attenuates and reverses analgesic tolerance to morphine. Pain 59 (3): 361-8, 1994.  [PUBMED Abstract]

  251. Weinbroum AA, Gorodetzky A, Nirkin A, et al.: Dextromethorphan for the reduction of immediate and late postoperative pain and morphine consumption in orthopedic oncology patients: a randomized, placebo-controlled, double-blind study. Cancer 95 (5): 1164-70, 2002.  [PUBMED Abstract]

  252. Weinbroum AA, Bender B, Bickels J, et al.: Preoperative and postoperative dextromethorphan provides sustained reduction in postoperative pain and patient-controlled epidural analgesia requirement: a randomized, placebo-controlled, double-blind study in lower-body bone malignancy-operated patients. Cancer 97 (9): 2334-40, 2003.  [PUBMED Abstract]

  253. Dudgeon DJ, Bruera E, Gagnon B, et al.: A phase III randomized, double-blind, placebo-controlled study evaluating dextromethorphan plus slow-release morphine for chronic cancer pain relief in terminally ill patients. J Pain Symptom Manage 33 (4): 365-71, 2007.  [PUBMED Abstract]

  254. Pistevou-Gombaki K, Eleftheriadis N, Plataniotis GA, et al.: Octreotide for palliative treatment of hepatic metastases from non-neuroendocrine primary tumours: evaluation of quality of life using the EORTC QLQ-C30 questionnaire. Palliat Med 17 (3): 257-62, 2003.  [PUBMED Abstract]

Back to Top

< Previous Section  |  Next Section >


A Service of the National Cancer Institute
Department of Health and Human Services National Institutes of Health USA.gov