Pain and Cancer Mangement

23
3393 Cancer Pain Management Current Strategy Nathan I. Cherny, M.B.B.S., F.R.A.C.P., and Russell K. Porfenoy, M.D. Pain is among the most prevalent symptoms experienced by cancer patients. A strategy for the management of cancer pain is now widely accepted, and when well im- plemented, is usually effective. Unfortunately, many on- cologists are ill-prepared for the task of pain assessment and management, and the outcomes achieved in clinical practice are often suboptimal. The various elements in the pain management strat- egy are described. Patient assessment, the use of primary therapies and systemically administered nonopioid and opioid analgesics are pivotal to the strategy. Practical aspects of opioid pharmacotherapy encompass drug se- lection and dosing considerations including selection of an appropriate route of administration, dose titration, and the management of side effects. Specific approaches are described for the treatment of patients for whom an acceptable balance between relief and side effects of opioids is not achieved. These comprise noninvasive in- terventions, including the use of adjuvant analgesics, psy- chological therapies, and physiatric techniques, and in- vasive interventions, such as the use of intraspinal opioids, neural blockade, and neuroablative techniques. Finally, the use of sedation in the treatment of patients with pain that is refractory to other interventions is ad- dressed. The skilled application of this strategy can provide adequate relief to the vast majority of patients, most of whom will respond to systemic pharmacotherapy alone. Patients with refractory pain should see specialists in pain management or palliative medicine who can address these difficult problems. Cancer 1993; 72: 3393-415. Key words: cancer pain, pain assessment, pain manage- ment, analgesics, opioids, adjuvant analgesics, neural blockade, spinal opioids, sedation. Presented at the Second National Conference on New Oncolo- gic Agents: Practical Applications, San Diego, California, February From the Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York. Address for reprints: Russell K. Portenoy, M.D., Pain Service, Department of Neurology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021. 4-6, 1993. Accepted for publication July 30, 1993. Pain is among the most prevalent symptoms experi- enced by patients with cancer.'-3 Successful manage- ment depends on the ability of the clinician to assess initial problems, identify and evaluate pain syndromes, and formulate a plan for continuing care that is respon- sive to the evolving goals and needs of the ~ a t i e n t . ~ This process requires a familiarity with numerous therapeu- tic options. A general strategy for the management of cancer pain is now widely accepted (Table 1). When imple- mented well, this approach is usually eff e ~ t i v e . ~ , ~ Unfor- tunately, there is abundant evidence that the outcomes achieved in routine clinical practice are suboptimal7and that many oncologists are ill-prepared for the ongoing task of symptom assessment and management.7,8 Poor outcomes may contribute to physician b ~ r n o u t ~ , ~ and greatly increase patient morbidity. The American Soci- ety of Clinical Oncology recognized the need for im- proved clinician education and has recently published curriculum guidelines for cancer pain assessment and treatment." This publication represents a necessary first step, which must now be followed by widespread implementation in training programs and continuing education. An effective strategy for the management of cancer pain is built on a comprehensive assessment and the skillful administration of analgesic drugs. Survey data suggest that approximately 10-30% of patients fail to achieve satisfactory pain relief, and subsequently re- quire other approaches."-'6 Cancer Pain Assessment The formulation of an effective therapeutic strategy for the management of cancer pain is predicated on a com- prehensive assessment of the patient. A trusting rela- tionship with the patient is essential to this process, but does not negate the potential for under-reporting of symptom^.'^ A clinical posture should be adopted that affirms relief of pain and suffering as important goals of therapy, and encourages communication about symp- toms. To this end, it is useful to ask an open-ended

Transcript of Pain and Cancer Mangement

Page 1: Pain and Cancer Mangement

3393

Cancer Pain Management Current Strategy

Nathan I . Cherny, M.B.B.S., F.R.A.C.P., and Russell K. Porfenoy, M.D.

Pain is among the most prevalent symptoms experienced by cancer patients. A strategy for the management of cancer pain is now widely accepted, and when well im- plemented, is usually effective. Unfortunately, many on- cologists are ill-prepared for the task of pain assessment and management, and the outcomes achieved in clinical practice are often suboptimal.

The various elements in the pain management strat- egy are described. Patient assessment, the use of primary therapies and systemically administered nonopioid and opioid analgesics are pivotal to the strategy. Practical aspects of opioid pharmacotherapy encompass drug se- lection and dosing considerations including selection of an appropriate route of administration, dose titration, and the management of side effects. Specific approaches are described for the treatment of patients for whom an acceptable balance between relief and side effects of opioids is not achieved. These comprise noninvasive in- terventions, including the use of adjuvant analgesics, psy- chological therapies, and physiatric techniques, and in- vasive interventions, such as the use of intraspinal opioids, neural blockade, and neuroablative techniques. Finally, the use of sedation in the treatment of patients with pain that is refractory to other interventions is ad- dressed.

The skilled application of this strategy can provide adequate relief to the vast majority of patients, most of whom will respond to systemic pharmacotherapy alone. Patients with refractory pain should see specialists in pain management or palliative medicine who can address these difficult problems. Cancer 1993; 72: 3393-415.

Key words: cancer pain, pain assessment, pain manage- ment, analgesics, opioids, adjuvant analgesics, neural blockade, spinal opioids, sedation.

Presented at the Second National Conference on New Oncolo- gic Agents: Practical Applications, San Diego, California, February

From the Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, New York.

Address for reprints: Russell K. Portenoy, M.D., Pain Service, Department of Neurology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021.

4-6, 1993.

Accepted for publication July 30, 1993.

Pain is among the most prevalent symptoms experi- enced by patients with cancer.'-3 Successful manage- ment depends on the ability of the clinician to assess initial problems, identify and evaluate pain syndromes, and formulate a plan for continuing care that is respon- sive to the evolving goals and needs of the ~ a t i e n t . ~ This process requires a familiarity with numerous therapeu- tic options.

A general strategy for the management of cancer pain is now widely accepted (Table 1). When imple- mented well, this approach is usually eff e ~ t i v e . ~ , ~ Unfor- tunately, there is abundant evidence that the outcomes achieved in routine clinical practice are suboptimal7 and that many oncologists are ill-prepared for the ongoing task of symptom assessment and management.7,8 Poor outcomes may contribute to physician b ~ r n o u t ~ , ~ and greatly increase patient morbidity. The American Soci- ety of Clinical Oncology recognized the need for im- proved clinician education and has recently published curriculum guidelines for cancer pain assessment and treatment." This publication represents a necessary first step, which must now be followed by widespread implementation in training programs and continuing education.

An effective strategy for the management of cancer pain is built on a comprehensive assessment and the skillful administration of analgesic drugs. Survey data suggest that approximately 10-30% of patients fail to achieve satisfactory pain relief, and subsequently re- quire other approaches."-'6

Cancer Pain Assessment

The formulation of an effective therapeutic strategy for the management of cancer pain is predicated on a com- prehensive assessment of the patient. A trusting rela- tionship with the patient is essential to this process, but does not negate the potential for under-reporting of symptom^.'^ A clinical posture should be adopted that affirms relief of pain and suffering as important goals of therapy, and encourages communication about symp- toms. To this end, it is useful to ask an open-ended

Page 2: Pain and Cancer Mangement

3394 CANCER Supplement December 2, 2993, Volume 72, No. 11

Table 1. Clinical Strategies Essential in the Management of Cancer Pain

Comprehensive assessment Primary therapy and systemic nonopioid and opioid analgesic

therapy Role of primary therapies Selection of nonopioid and opioid analgesic agents Practical aspects of administration

Routes Dose selection and titration

Management of side effects Noninvasive interventions for patients unable to attain an

acceptable balance between relief and side effects of systemic opioid therapy

Reduce opioid requirement by: Appropriate primary therapy Addition of nonopioid analgesic Addition of an adjuvant analgesic Use of cognitive or behavioral techniques Use of an orthotic device or other physical medicine approach

Switch to another opioid Invasive interventions for patients unable to attain an acceptable

balance between relief and side effects during systemic pharmacotherapy

Regional analgesic techniques (spinal or intraventricular opioids) Neural blockade Neuroablative techniques

Consider increased sedation

question about the presence of pain at each patient visit in routine oncologic practice.

The assessment should clarify the pain characteris- tics and syndrome, infer the putative mechanisms that may underlie the pain, and determine the impact of the pain on function and psychological well-being. In addi- tion, the assessment should define the nature and ex- tent of the underlying disease; evaluate concurrent problems (physical, psychological, and social) that con- tribute, or may soon contribute, to patient distress; and clarify the goals of care. The goals of care are often complex, but can generally be grouped into three broad categories: (1) prolonging survival, (2) optimizing com- fort and (3) optimizing function. The relative priority of these goals provides an essential context for therapeutic decision-making.

A stepwise approach to cancer pain assessment be- gins with data collection and ends with a clinically rele- vant formulation (Table 2). The pain-related history should illuminate: characteristics of the pain; the re- sponses to previous disease-modifying and analgesic therapies; effects of the pain on activities of daily living, psychological state, familial and professional function; and associated symptom^.^^'^,'^ Numerous instruments, including symptom checklists and quality-of-life mea- sures, are available and may be useful in this evalua-

tion.’O,’l A careful review of medical history and the chronology of the cancer is important to place the pain complaint in context.

A physical examination, including a neurologic evaluation, is a necessary part of the initial pain assess- ment. The need for a thorough neurologic assessment is justified by the high prevalence of painful neurologic conditions in the cancer p~pulation.~’~’~ The physical examination should attempt to identify the underlying causes of the pain problem, clarify the extent of the underlying disease, and discern the relation of the pain complaint to the disease.

Careful review of previous laboratory and imaging studies can provide important information about the cause of the pain and the extent of the underlying dis- ease. The treating clinician is best able to correlate this information with the patient’s symptoms and signs, and should personally evaluate these studies.

The information derived from these data provides the basis for a provisional pain diagnosis that clarifies both the status of the disease and the nature of other concurrent concerns that may require therapeutic focus (Table 2). The provisional pain diagnosis includes infer- ences about the pathophysiology of the pain and an assessment of the pain syndrome. Evaluation of concur- rent concerns includes other symptoms and related psychosocial problems.

Additional investigations are often needed to clar- ify uncertainties in the provisional assessment.” The extent of the these investigations must be appropriate to the patient‘s general status and the overall goals of care. For some patients, multiple studies may be needed to evaluate the pain problem, clarify extent of disease, or assess other symptoms. Again, the physician ordering the diagnostic procedures should personally review them to correlate pathologic changes with the clinical findings. Expert assistance from physicians in other dis- ciplines, nurses, social workers, or others may be also be necessary to evaluate related physical or psychosocial problems identified during the initial assessment. Pain must be managed during this process to improve com- pliance and reduce the distress associated with proce- dures. No patient should be inadequately evaluated be- cause of poorly controlled pain.

The findings of this evaluation should be reviewed with the patient, and other appropriate persons, so that current problems can be listed in order of priority to reflect their importance to the patient. Potential out- comes that would benefit from contingency planning may also be identified, including the need for advanced medical directives, the evaluation home care resources, and prebereavement interventions with the family. A multimodality treatment approach targeted to specific problems can be developed from this assessment.

Page 3: Pain and Cancer Mangement

Current Strategy for Cancer Pain Management/Cherny and Portenoy 3395

Table 2. Stepwise Assessment of the Patient With Cancer Pain

Step 1. Data collection

Pain-related history Other relevant history and imaging data Physical examination

Chronology Disease-related Radiographs and scans Characteristics Other symptoms Tumor markers Impact on function Psychiatric history Hematologic parameters Prior treatment Social resources Biochemical parameters Other pain history

Available laboratory

Step 2. Provisional assessment

Provisional pain diagnosis

Syndrome identification Inferred pathophysiology

Global assessment Concurrent concerns

Extent of disease Other symptoms Goals of care Untreated concurrent diseases

Prolonging survival Psychosocial needs Optimizing function Rehabilitative needs Optimizing comfort Financial needs

Step 3. Diagnostic investigations and other assessments

Diagnostic investigations Other assessments

Symptom-specific Extent of disease

Psychological Social Financial Functional

Step 4. Initial formulation and problem list

Pain syndromes and pathophysiology Extent of disease Concurrent concerns Anticipated contigencies

Step 5. Patient review and formulation of prioritized problem lists

Current problems Anticipated contingencies

Step 6. Multimodality therapeutic plan

Primary anti-cancer treatment Chemotherapy Radiation therapy Surgery Immunotherapy Other

Treatment of concurrent disease processes Symptom-directed pharmacotherapy Rehabilitative approaches Psychological approaches Anesthetic approaches Neurostimulatory approaches From Chemy NI, Portenoy RKP. Cancer pain: principles of assessment and syndromes. In: Wall PD, Melzack R, eds. Textbook of pain. Edinburgh Churchill Livingstone, 1993. With permission.

Page 4: Pain and Cancer Mangement

3396 CANCER Supplement December 2, 2993, Volume 72, No. 13

The Role of Primary Therapy

The assessment process may reveal a cause for the pain that is amenable to primary therapy. Pain that is pro- duced by tumor infiltration may respond to antineoplas- tic treatment with surgery, radiotherapy, or chemother- apy, and pain caused by infections may be relieved with antibiotic therapy or drainage procedures. Specific an- algesic treatments are usually required as an adjunct to the primary therapy.

Radiation therapy. Radiation therapy has a pivotal role in the treatment of cancer pain caused by bone m e t a ~ t a s e s , ~ ~ , ~ ~ epidural ne~plasm,’~ and cerebral me- tastases.26 In other settings, conclusive data are lacking, but clinical observations support empirical radiation therapy. For example, the results with perineal pain caused by low sacral plexopathy appear to be encourag- ing,27,28 and hepatic radiation therapy (e.g., 2000-3000 cGy) can be well tolerated and effective for the pain of hepatic capsular distention in 50-9070 of patient^.'^-^'

Chemotherapy. There is a strong impression that tumor shrinkage is generally associated with relief of pain. Hence, analgesia can be anticipated after chemo- therapy in responsive tumors, such as lymphoma, small cell lung cancer, germ cell tumors, and previously un- treated breast cancer. Interestingly, there are isolated reports of analgesic effects from chemotherapy even in the absence of significant tumor shrinkage.33T34 In all situations, the decision to administer chemotherapy solely for the treatment of symptoms should be recon- sidered in the absence of a clearly favorable balance between relief and adverse effects.

Surgery. Surgery may have a role in the relief of symptoms caused by specific problems, such as ob- struction of a hollow viscus, unstable bony structures, and compression of neural t i s s ~ e s . ~ ~ - ~ ~ In all cases, the potential benefits must be weighed against the risks of surgery, the anticipated length of hospitalization and convalescence, and the predicted duration of benefit.35 For example, large volume paracentesis (up to 5-10 1) may provide prompt and prolonged relief from the pain and discomfort of tense as cite^,^^"^ with a small risk of h y p ~ t e n s i o n ~ ~ , ~ ’ or hyp~proteinemia.~~

Antibiotics. Antibiotics may have analgesic effects when the source of the pain involves infection, as illus- trated by the treatment of chronic sinus infection, pelvic abscess, pyonephrosis, and osteitis p ~ b i s . ~ ~ , ~ ~ Occasion- ally infection may be occult and confirmed only by the symptomatic relief provided by empirical treatment with these d r ~ g s . ~ ~ , ~ ~

Systemic Analgesic Pharmacotherapy

Systemically administered analgesic drug therapy, in- volving the use of nonopioid, opioid, and adjuvant an-

Figure 1. The three-step ”analgesic ladder,” proposed by an expert committee convened by the Cancer Unit of the Word Health Organization. From World Health Organization. Cancer pain relief and palliative care: report of a WHO expert committee. Geneva: World Health Organization, 1990. WHO Technical Report Series, no. 804. With permission.

algesics, is the cornerstone of cancer pain manage- ment.5,6T46 Nonopioid analgesics comprise acetamino- phen and the nonsteroidal anti-inflammatory drugs (NSAIDs) including aspirin. The term ”adjuvant anal- gesic” refers to a drug that has a primary indication other than pain, but is analgesic in selected circum- stances.

The three-step ”analgesic ladder,” proposed by an expert committee convened by the Cancer Unit of the World Health Organization, has emphasized that pain intensity should be the prime consideration in the se- lection of these (Fig. 1). Patients with mild to moderate cancer-related pain should be treated with a nonopioid analgesic, which should be combined with an adjuvant analgesic if a specific indication for one exists. Patients who are relatively nontolerant and have moderate to severe pain, or who fail to obtain adequate relief after a trial of a nonopioid analgesic, are usually treated with an opioid. This treatment is typically ac- complished using a combination product containing a nonopioid (e.g., aspirin or acetaminophen) and an opioid conventionally used for this indication, such as

Page 5: Pain and Cancer Mangement

Current Strategy for Cancer Pain Management/Cherny and Portenoy 3397

Table 3. Nonoplioid Analgesics

Maximal recommended

Half-life Starting dose dose Chemical class Generic name (hr) (mg) (mg/day) Comments

Nonacidic p-Aminophenol derivatives Acetaminophen Naphthylalkanones Nabumetone

Salicylates Aspirin Acidic

Diflunisal

Choline magnesium trisalicylate

Salsalate

Proprionic acids

Acetic acids

Ibuprofen Naproxen Fenoprofen Ketoprofen Flurbiprofen lndomethacin

Sulindac Diclofenac Ketorolac

Tolmen tin Oxicams Piroxicam Fenamates Mefenamic acid I'yranocarboxylic acids Etodolac

3-4 22-30

3-12 8-12

8-12

8-12

3-4 1-3 2-3 2-3 5-6 4-5

14 2

4-7

1 45

2 7

750 every 4 hr 500 every 12 hr

650 every 4-6 hr 500 every 12 hr

1000 every 12 hr

1000 every 12 hr

400 every 6 hr 250 every 12 hr 200 every 6 hr

25 every 8 hr 100 every 12 hr 25 every 12 hr

150 every 12 hr 25 every 8 hr 30 every 6 hr

200 every 8 hr

250 every 6 hr 20 every 24 hr

1000 every 24

6000 2000

6000 1500

4000

4000

4200 1000 3200

200 300 200

400 200 240

2000 40

1000 2000

Available over the counter

Avaiiable over the counter Less GI toxicity than

Minimal GI toxicity No effect on platelet

Minimal GI toxicity No effect on platelet

function at usual doses Available over the counter

aspirin

function at usual doses

Sustained release and rectal preparations

Oral or parenteral preparation

GI: gastrointestinal.

codeine, oxycodone, or propoxyphene. This drug should be coadministered with an adjuvant analgesic if needed. Finally, patients who have severe pain, or fail to achieve adequate relief after appropriate administra- tion of drugs on the second rung of the analgesic ladder, should receive an opioid agonist conventionally used for severe pain. This drug may also be combined with a nonopioid or adjuvant analgesic.

The Use of Nonopioid Analgesics

The nonopioid analgesics are useful alone for mild to moderate pain and provide additive analgesia when combined with opioid drugs in the treatment of more severe pain. These drugs constitute a heterogeneous group of compounds that differ in chemical structure but share many pharmacologic actions (Table 3). Unlike opioid analgesics, all have a "ceiling" effect for analge- sia and produce neither tolerance nor physical depen- dence. The major site of action is presumably periph- eral. Aspirin and the NSAIDs inhibit the enzyme cy- clooxygenase and consequently block the biosynthesis

of prostaglandins, inflammatory mediators known to sensitize peripheral noci~eptors .~~ A central mechanism of action is also likely to c ~ n t r i b u t e , ~ ~ ' ~ ~ however, and probably predominates in acetaminophen analge~ia.~'

The safe administration of nonopioid analgesics re- quires familiarity with their potential adverse effects.51 Aspirin and the other NSAIDs have a broad spectrum of potential toxicity. Bleeding diathesis due to inhibition of platelet aggregation, gastroduodenopathy (including peptic ulcer disease), and renal impairment are the most common. The less common adverse effects include con- fusion, precipitation of cardiac failure, and exacerba- tion of hypertension. Particular caution is required in the administration of these agents to patients at in- creased risk of adverse effects, including the elderly and those with blood clotting disorders, predilection to pep- tic ulceration, impaired renal function, and concurrent corticosteroid therapy. Of the NSAIDs, the nonacety- lated salicylates ( e g , choline magnesium trisalicylate and salsalate) are preferred for patients who have a bleeding ulcer or ulcer diathesis; these drugs have rela-

Page 6: Pain and Cancer Mangement

3398 CANCER Supplement December 1, 1993, Volume 72. No. 11

Table 4. Classification of Opioid Analgesics Based on Receptor Interactions

Partial Agonists Agonists/antagonists agonists

Codeine Butorphanol Buprenorphine Oxycodone Nalbuphine Dezocine I'ropoxyphene I'entazocine Morphine Hydromorphone Methadone Meperidine Oxymorphone Heroin Levorphanol Fentanyl

tively less effect on gastric mucosa and platelet aggre- gation, and no effect on bleeding time at routine clinical doses5* Acetaminophen rarely produces gastrointesti- nal toxicity, and has no adverse effects on platelet func- tion. Hepatic toxicity is possible, however, and patients with liver disease can develop severe hepatopathy at the usual therapeutic dose.53

Standard recommended doses for nonopioid drugs are derived from studies performed in populations (gen- erally healthy patients with an inflammatory disease) that potentially have little in common with cancer pa- tients, who often have numerous medical problems and may be receiving multiple other drugs. Consequently, there is no certain knowledge of the minimal effective analgesic dose, ceiling dose, or toxic dose for any pa- tient with cancer pain, and doses may be higher or lower than the usual dose ranges recommended for the drug involved. These considerations and the observa- tion that the effects of these drugs are (at least partially) dose-dependent, support an approach to the adminis- tration of NSAIDs that incorporates both low initial doses and dose titration. With gradual dose escalation, it may be possible to identify the ceiling dose and re- duce the risk of significant toxicity. Clinical experience suggests that 1 week is usually adequate to evaluate the efficacy of a dose. The potential for dose-dependent toxicity requires the use of an empirical upper limit for dose titration, which is usually in the range of 1.5-2 times the standard recommended dose of the drug in question. Because failure with one NSAID can be fol- lowed by success with another, sequential trials of sev- eral NSAIDs may be needed to identify a drug with a favorable balance between analgesia and side effects.

Systemically Administered Opioid Therapy

Based on their interactions with the various receptor subtypes, opioid compounds can be divided into ago- nist, agonist-antagonist, and antagonist classes (Table

4). The pure agonist drugs (Table 5) are most commonly used in cancer pain management. The mixed agonist- antagonist opioids (pentazocine, nalbuphine, and bu- torphanol) and the partial agonist opioids (buprenor- phine and probably dezocine) have limited usefulness in this setting because of a ceiling effect for analgesia, precipitation of withdrawal reactions in patients physi- cally dependent to opioid agonists, and a high preva- lence of dose-dependent psychotomimetic side ef- fects. 4,5

Opioid selection. A trial of opioid therapy should be administered to all patients with pain of moderate or greater severity, regardless of the pathophysiological mechanism underlying the pain.5,54-57 In accordance with the analgesic ladder approach,46 patients with moderate pain are commonly treated with a combina- tion product, which in the United States typically con- tains codeine, oxycodone or propoxyphene plus acet- aminophen, or aspirin. The dose of these combination products can be increased until the maximum dose of the nonopioid coanalgesic is attained (e.g., 4000-6000 mg acetaminophen). If pain relief is inadequate at this dose, the opioid contained in the combination product can be increased as a single agent ( e g , oxycodone tab- lets or syrup can be used to supplement an oxycodone- aspirin combination product), or the patient can be switched to an opioid customarily used to treat severe pain.

Most patients with cancer pain require long-term treatment with an opioid conventionally used to man- age severe pain (third rung of the analgesic ladder). Po- tentially any of these opioids could be used. Ease of titration and convenience of administration are impor- tant considerations, and morphine sulphate is usually preferred because it has a short half-life, is easy to ti- trate in its immediate release form, and is also available as a controlled-release preparation that allows an 8-12- hour dosing interval. The long half-life drugs, metha- done and levorphanol, are not preferred for the elderly or those with major organ failure, because they can be difficult to titrate and present challenging management problems should delayed toxicity develop as plasma concentrations gradually rise after dose increments.

The use of meperidine is generally not recom- mended for the management of cancer pair^.^,^,^' Me- peridine is demethylated to normeperidine, an active metabolite that is twice as potent as a convulsant and has a half-life 4-5 times as longer than the parent com- pound.59 Repetitive dosing of meperidine can result in normeperidine accumulation, which may cause central nervous system toxicity characterized by subtle adverse mood effects, tremulousness, multifocal myoclonus and, occasionally, seizure^.^^-^' Naloxone does not re- verse normeperidine-induced seizures, and indeed,

Page 7: Pain and Cancer Mangement

Current Strategy for Cancer Pain Management/Cherny and Portenoy 3399

Table 5. Opioid Agonist Drugs Dose (mg) equianalgesic to 10 mg im morphine Duration

Half-life of action Drug IM PO (hr) (hr) Comments

Opioid agonists customarily used to treat moderate pain

Codeine 130 200

Oxycodone*

Propoxyphene

15 30

50 100

Opioid agonists customarily used to treat severe pain

Morphine

Oxy codone Hydromorphone Methadone

Meperidine

Oxymorphone

Heroin Levorphanol

Fentanyl transdermal system

10 30 (repeated dose) 60 (single dose)

15 30

10 20 1.5 7.5

75 300

1 10 (PR)

5 60 2 4

2-3 2-4

2-3 2-4

2-3 2-4

2-3 3-4

2-3 2-4 2-3 2-4

15-190 4-8

2-3

2-3

0.5 12-15

2-4

3-4

3-4 4-8

48-72

Usually combined with a nonopioid Available over the counter Formulated as single agent or

combined with a nonopioid Usually combined with a nonopioid Norpropoxyphene toxicity may

cause seizures

M6G accumulation in renal failure may predispose to additional toxicity

Formulated as single agent No known active metabolites Plasma accumulation may lead to

Dosing should be initiated on a

Not recommended for cancer pain Normeperidine toxicity limits utility Contraindicated in patients with

renal failure and those receiving MA0 inhibitors

delayed toxicity

PRN basis

No oral formulation available Less histamine release High solubility morphine prodrug Plasma accumulation may lead to

Patches available to deliver 25, 50, delayed toxicity

75, and 100 g d h r IM: intramuscular; PO: per 0s; PR per rectum; PRN: as needed; MAO: monoamine oxidase. * When combined with a nonoDioid.

could theoretically precipitate seizures in patients re- ceiving meperidine by blocking the depressant action of the parent compound and allowing the convulsant ac- tivity of normeperidine to become manifest.62 Although normeperidine toxicity is most likely to affect the el- derly and patients with overt renal disease (who are unable to renally clear the metabolite), it is sometimes observed in younger patients with normal renal func- tion.59,63 In the unusual patient who is administered meperidine, concurrent therapy with a monoamine oxi- dase inhibitor is specifically contraindicated because of the risk of a severe drug reaction characterized by exci- tation, hyperpyrexia, and convulsions.

Renal impairment may reduce the clearance of the active metabolites of propoxyphene (norpropoxy- phene), meperidine (normeperidine), and morphine (morphine-6-glucuronide). Particular caution is re- quired in the titration of these agents in this ~ e t t i n g , ~ ~ - ~ ~

and alternative opioids are often recommended. Similar care is required when prescribing meperidine, pentazo- cine, or propoxyphene to patients with hepatic dys- function. The clearance of these drugs is diminished with liver disease and plasma concentrations may be- come higher than normal. 70,71 Morphine clearance is only minimally affected by mild or moderate hepatic i m ~ a i r m e n t , ~ ~ , ~ ~ but may be significantly reduced in ad- vanced disease.74

Patients who have experienced dose-limiting side effects with one opioid may benefit from sequential trials of other opioid For each patient, the bal- ance between side effects and analgesic efficacy varies among drugs in an unpredictable manner. Sequential trials may therefore identify a more acceptable drug. An explanation for the variability in response is lacking, but hypotheses have been proposed based on the exis- tence of receptor binding difference^^^ and the occur-

Page 8: Pain and Cancer Mangement

3400 CANCER Supplement December 2, 2993, Volume 72. No. 11

rence of incomplete cross-tolerance to the analgesic and adverse effects of different ~ p i o i d . ~ ~

A successful switch from one opioid to another re- quires understanding of relative analgesic potency, which can be defined as the ratio of the dose of two analgesics required to produce the same analgesic effect (equianalgesia). Reference tables conventionally ex- press these relative potencies in terms of a dose equian- algesic to 10 mg of parenteral morphine (Table 5).77 Equianalgesic doses should not be viewed as standard starting doses when the drug or route of administration is changed, but rather as a useful guide for dose selec- tion. Numerous other variables may influence the ap- propriate dose for the individual patient, including pain severity, prior opioid exposure (and the degree of cross- tolerance this confers), age, route of administration, level of consciousness, and metabolic abnormalities (see later).

Selecting a route of systemic opioid administra- tion, Opioids should be administered by the least inva- sive and most convenient route capable of providing adequate analgesia for the patient. In routine practice, the oral route is usually the most appropriate.5r6r58 Al- ternative noninvasive routes, including the rectal, sub- lingual, and transdermal routes, should be used for pa- tients who have impaired swallowing or gastrointesti- nal obstruction.

The potency of opioids administered rectally is be- lieved to approximate oral d ~ s i n g . ~ ~ , ~ ~ Rectal supposito- ries containing oxymorphone, hydromorphone, and morphine are available, and controlled-release mor- phine tablets can also be administered rectally."

The bioavailability of the sublingual route is very poor with opioid drugs that are not highly lipophilic," and the likelihood of an adequate analgesic response is low. Sublingual buprenorphine, a relatively lipophilic partial agonist, has been proven eff ective,82 but is not available in the United States. Sublingual morphine has also been reported anecdotally to be effe~tive,'~ but this drug has poor sublingual absorption,'l and efficacy may be related to swallowing of the dose.84 Both fen- tanyl and methadone are relatively well absorbed through the buccal mucosa,81 and sublingual adminis- tration of an injectable formulation might be a reason- able approach in the patient who transiently loses the option of oral dosing.

A transdermal formulation of fentanyl is available in systems that deliver 25, 50, 75, and 100 ~ g / h . ' ~ The transdermal system consists of a drug reservoir sepa- rated from the skin by a rate-limiting membrane that controls the rate of drug delivery such that the drug is released into the skin at a nearly constant amount per unit time. The dosing interval for each system is usually 72 hours, but interindividual pharmacokinetic variabil-

ity is and some patients require dosing inter- vals as brief as 48 hours. Limitations of the transdermal delivery system include its expense and the require- ment for an alternative short-acting opioid for break- through pain.

Parenteral routes of administration should be con- sidered for patients who have impaired swallowing or gastrointestinal obstruction, those who require the rapid onset of analgesia, and highly tolerant patients who require doses that cannot otherwise be conve- niently administered. Repeated parenteral bolus injec- tions, which may be administered by the intravenous (IV), intramuscular, or subcutaneous (SC) routes, may be useful in some patients, but are often complicated by the occurrence of prominent "bolus" effects (toxicity at peak concentration and/or pain breakthrough at the trough). Although repetitive intramuscular injections are a common practice, they are painful and offer no pharmacokinetic advantage; their use is not recom- mended.58 Repeated bolus doses, if required, can be ac- complished without frequent skin punctures through the use of an indwelling IV or SC infusion device. To deliver repeated SC injections, a 27-gauge infusion de- vice (known as a "butterfly") can be left under the skin for up to 1 week.

Continuous infusions avoid the problems asso- ciated with the bolus effect and may be administered intravenously or subcutaneously.s9-93 Ambulatory in- fusion devices vary in complexity, cost, and ability to provide patient-controlled "rescue doses" as an adjunct to a continuous basal infusion (see later). Opioids suit- able for continuous SC infusion must be soluble, well absorbed, and nonirritant. Extensive experience has been reported with heroin, hydromorphone, oxymor- phone, and Methadone appears to be relatively irritating and is not preferred for SC infu- ~ i o n . ~ ~ The bioavailability of SC hydromorphone is 7 8 Y 0 , ~ ~ and is assumed to be similarly high with other opioids. This bioavailability supports the recommenda- tion that dosing with SC administration proceed in a manner identical to continuous IV infusion. To main- tain the comfort of an infusion site, the SC infusion rate should not exceed 5 ml/hr. Patients who need high doses may benefit from the use of concentrated solu- tions, which in selected cases can be compounded spe- cifically for continuous SC infusion. Continuous IV in- fusion may be the most appropriate way of delivering an opioid when there is a need for infusion of large volume of solution, or when using methadone. If con- tinuous IV infusion is to be continued on a long-term basis, a permanent central venous port is recom- mended.

Continuous infusions of drug combinations may be indicated when pain is accompanied by nausea, anxi-

Page 9: Pain and Cancer Mangement

Current Strategy for Cancer Pain Management/Cherny and Portenoy 3401

ety, or agitation. An antiemetic, neuroleptic, or anxioly- tic agent may be combined with an opioid provided it is nonirritant, miscible, and stable in combined solution. Experience has been reported with infusions of an opioid combined with metoclopromide, haloperidol, scopolamine, cyclizine, methotrimeprazine, chlorprom- azine, and midazolam.'00-'02

The switch between the oral and parenteral routes requires careful attention to relative potency (see Table 5). To avoid inadvertent overdosing or underdosing, it is useful to perform the change in steps if possible. Slowly reducing the parenteral dose while increasing the oral dose during a 2-3-day period can minimize the potential problems caused by an abrupt change of routes associated with different potencies.

Scheduling of opioid administration. Scheduled, around-the-clock dosing can provide the patient with continuous relief and is generally preferred when pain is constant or frequently recurring. The initiation of opioid therapy with an around-the-clock regimen, re- quires particular vigilance when patients have had no previous opioid exposure and when drugs with long half-lives are selected. Drugs with long half-lives may produce the delayed onset of adverse effects as plasma concentration rises toward steady-state levels.

Breakthrough pain is highly prevalent among pa- tients with controlled baseline pain.'03 To help manage these episodes, all patients who receive an around-the- clock opioid regimen should also have access to a sup- plemental rescue dose on an as-needed basis. The "res- cue" drug is typically identical to that administered on a continuous basis with the exception of transdermal fen- tanyl and methadone, for which the use of an alterna- tive short half-life opioid is recommended. Clinical ex- perience suggests that the size of the rescue dose should be equivalent to approximately 5-15% of the 24-hour baseline dose. The frequency with which the rescue dose can be offered depends on the time to peak effect for the drug and the route of administration. Oral res- cue doses are offered up to every 1-2 hours and paren- teral doses are offered up to every 15-30 minutes. The integration of scheduled dosing with rescue doses pro- vides a method for safe and rational stepwise dose escalation, which is applicable to all routes of opioid administration (Table 6).

Controlled-release preparations can lessen the in- convenience associated around-the-clock administra- tion of drugs with a short duration of action.'04 In the United States, controlled-release morphine sulphate is available in 15, 30, 60, and 100 mg tablets, and formu- lations of controlled-release oxycodone and hydromor- phone are under development. Controlled-release prep- arations of morphine sulphate typically achieve peak levels 3-5 hours after administration and have a dura-

tion of effect of 8-12 hours.'05 Immediate-release mor- phine is generally used as the rescue medication for patients receiving these formulations'06 (Table 6). Con- trolled-release morphine should not be used to rapidly titrate the dose in patients with severe pain'06; repeated dose adjustments at short intervals are performed more efficiently with an immediate-release preparation, which may be converted on a milligram to milligram basis to a controlled-release preparation when the ef- fective around-the-clock dose is identified.

In some situations, an as-needed dosing regimen alone can be recommended, This type of dosing pro- vides additional safety during the initiation of opioid therapy in the opioid-naive patient, particularly when rapid dose escalation is needed. This technique is strongly recommended when starting methadone ther- apy. As-needed dosing may also be appropriate for pa- tients who have rapidly decreasing analgesic require- ment or intermittent pains separated by pain-free inter- vals.

Patient-controlled analgesia (PCA) is a technique of parenteral drug administration (see above) in which the patient controls a pump that delivers bolus doses of an analgesic according to parameters set by the physi- ~ i a n . ~ ~ , ' ~ ' Use of a patient-controlled analgesia device allows the patient to carefully titrate the opioid dose to his or her individual analgesic needs. The technique is most commonly applied postoperatively. For patients with cancer pain patient-controlled analgesia is typi- cally used in conjunction with continuous opioid infu- sion. Long-term patient-controlled analgesia in cancer patients is most commonly accomplished via the subcu- taneous route using an ambulatory infusion device (see earlier).

Dose selection and adjustment. Patients who are opioid-naive or who have had limited exposure to an opioid conventionally used for moderate pain should generally begin one of the opioids used for more severe pain at a dose equivalent to 5-10 mg intramuscular morphine every 3-4 hours. If morphine is used, an in- tramuscu1ar:oral relative potency ratio of 1 :3 is gener- ally recommended.

When patients receiving higher doses of opioids are switched to an alternative opioid drug, the equianalge- sic dose table (Table 5) is used as a guide to the starting dose. For patients with good pain control but unaccept- able side effects, the starting dose of the new drug should be reduced to 50-75% of the equianalgesic dose (and to 33-50% when the switch is to methadone) to account for incomplete cross-tolerance. For patients with poor pain control and unacceptable side effects, the starting dose of the new drug can usually be 75- 100% of the equianalgesic dose (and 50-75% when the switch is to methadone). Patients must be monitored to

Page 10: Pain and Cancer Mangement

3402 CANCER Supplement December 1, 1993, Volume 72, No. 11

Table 6. Examples of Stepwise Dose Escalation of Morphine Sulfate Administered as Oral Immediate Release Preparation and Oral Controlled Release and Continuous Infusion

Oral controlled release (with SC infusion with SC rescue Oral immediate release immediate release rescue dosing) doses

mg every 4 hr Rescue dose PRN Rescue dose PRN Rescue dose PRN "around the every 1 hr mg "around every 1 hr q15-30 min

Step* clock" (mg) the c l o c k (mg) m g l h r (mg)

1 10 5 30 every 12 hr 7.5 3 2.0

3 30 15.0 60 every 12 hr 15.0 7 3.5

5 60 30.0 100 every 8 hr 45.0 15 7.5

2 15 7.5 30 every 8 hr 15.0 5 2.5

4 45 22.5 100 every 12 hr 30.0 10 5.0

* Indications for progression from one step to the next: requirement of z 2 rescue doses in any 4-hour interval; requirement of > 6 rescue doses in 24 hours.

assess the adequacy of analgesia and to detect the devel- opment of side effects. Subsequent dose adjustments are usually necessary.

At any point during the course of therapy, the de- velopment of inadequate pain relief should be ad- dressed through a stepwise escalation of the opioid dose. The opioid dose should be increased until ade- quate analgesia is reported or unmanageable side ef- fects supervene. Most patients reach plateaus that re- main constant for a prolonged p e r i ~ d . ' ~ ~ , ~ ~ ~ Patients in whom develop dose-limiting side effects during dose titration require the use of another analgesic approach or a technique to reduce toxicity (see later).

Because analgesia increases linearly with the log of the opioid dose, necessary dose escalations should usually be in the range of 30-50°% of the previous dose. Smaller dose increments are not likely to significantly improve analgesia. The absolute dose, which may be- come very large, is immaterial as long as the balance between analgesia and side effects remains favorable. For example, in a retrospective study of 100 patients with advanced cancer, the average daily opioid require- ment was equivalent to 400-600 mg of intramuscular morphine, but approximately 10% of patients needed more than 2000 mg and one patient needed more than 30,000 mg every 24 hours3

The severity of the pain should determine the rate of dose titration. Patients with very severe pain can be treated by repeated parenteral dosing every 15-30 min- utes until pain is partially relieved. Guidelines have been proposed for the calculation of hourly mainte- nance dosing after parenteral loading with a short half- life opioid.l10 These guidelines recommend that the starting hourly maintenance dose be approximated by dividing the total loading dose by twice the elimination half-life of the drug. For example, the starting mainte- nance dose for a patient who requires an intravenous loading dose of morphine sulphate 120 mg (half-life approximately 3 hours) would be 20 mg/h. Patients

with less severe pain may not require a loading dose of the opioid. In this situation, patients who are treated with short half-life opioids can undergo dose incre- ments as often as twice daily. The dose of controlled-re- lease preparations of oral morphine or transdermal fen- tanyl can be increased every 24-48 hours.

The need for escalating doses is a complex phenom- enon that may be precipitated by any of a variety of distinct processes. Extensive clinical experience sug- gests that most patients who require an escalation in dose to manage increasing pain have demonstrable pro- gression of d i s e a ~ e . ' ~ ~ ~ " ' - ~ ~ ~ True pharmacologic toler- ance to the analgesic effect of an opioid, which could potentially compromise the usefulness of treatment, can only be said to occur if a patient manifests the need for increasing opioid doses in the absence of other fac- tors (e.g., progressive disease, psychological factors, or pharmacokinetic factors) that would be capable of ex- plaining the increase in opioid requirement. This phe- nomenon is distinctly uncommon in the clinical setting. This observation suggests, first, that the concern about tolerance should not impede the use of opioids early in the course of the disease, and second, that worsening pain in a patient receiving a stable dose of opioids should generally be assessed as presumptive evidence of disease progression or, less commonly, increasing psychological distress.

Managing side effects. Management of the poten- tial adverse effects of opioids is necessary to optimize the therapeutic index of these drugs. The most common adverse effects are constipation, nausea, and vomiting. Other important dose-limiting adverse effects include sedation, delirium, myoclonus, and respiratory depres- sion.

The likelihood of opioid-induced constipation is so great that laxative medications should be prescribed prophylactically to most patients. Recommendations for laxative therapy are empirical. A combination of a softening agent (docusate) and a cathartic agent (e.g.,

Page 11: Pain and Cancer Mangement

Current Strategy for Cancer Pain Management/Cherny and Portenoy 3403

Table 7. Putative Mechanisms of Opioid-Induced Nausea and Vomiting and Their Management Mechanism Suggestive clinical features Antiemetic drugs

Stimulation of the medullary Nausea and or vomiting shortly after Metoclopramide, prochlorperazine, chlorpromazine, chemoreceptor trigger opioid administration haloperidol, corticosteroid, or lorazepam zone

sensitivity and vomiting, or vertigo Enhanced vestibular Prominent movement-induced nausea Scopolamine, meclizine, or lorazepam

Increased gastric antral tone Early satiety, postprandial bloating, or Metoclopramide vomi tine.

senna, bisocodyl, or phenophthalein) is frequently used. The doses of these drugs should be increased as necessary, and an osmotic laxative (e.g., lactulose or milk of magnesia) can be added if needed. Occasionally patients are managed with intermittent colonic lavage using an oral bowel preparation such as Golytely (Braintree Laboratories, Braintree, MA), and rarely pa- tients with refractory constipation can undergo a trial of oral naloxone, which has a bioavailability less than 3% and presumably acts selectively on opioid receptors in the gut.'14 Because there is a small risk of systemic with- drawal from oral nal~xone,"~ the initial dose should be conservative (0.8-1.2 mg once or twice daily); this dose can be escalated slowly until either favorable effects occur or abdominal cramps, diarrhea, or any other ad- verse effect develop^."^

The incidence of opioid-induced nausea and vomit- ing has been estimated to be 10-40% and 15-40%, re- spectively."6 Three putative mechanisms may produce these symptoms, and their respective recommended therapies are summarized in Table 7. Tolerance to these effects usually develops within weeks, and routine pro- phylactic administration of an antiemetic is not usually indicated except in patients with a history of severe opioid-induced nausea and vomiting.

Initiation of opioid therapy or significant dose esca- lation is often associated with sedation that may persist for days to weeks. Although tolerance to this effect usually develops, some patients have a persistent prob- lem, particularly if other contributing factors exist. A stepwise strategy for the management of persistent se- dation is most useful (Table 8).

Opioid-induced cognitive impairment also appears to be transient in most patient^."^ Although persistent cognitive impairment attributable to opioid alone oc- curs, most patients with persistent delirium have sev- eral contributing factors, including electrolyte dis- orders, neoplastic involvement of the central nervous system, sepsis, vital organ failure, or hypoxemia."* Again a stepwise approach to management is appro- priate (Table 9). Myoclonus is also a common dose-re- lated adverse effect of opioids, and may similarly re- solve spontaneously. If the myoclonus is symptomatic

and distressing, it can be treated empirically with a ben- zodiazepine (particularly clonazepam"'), barbiturate, or valproate.

Respiratory depression, the most serious adverse effect of opioid therapy, is uncommon in the cancer patient for whom opioid doses are carefully titrated. Continued opioid administration rapidly induces toler- ance to this effect.12' Clinically significant respiratory depression is always accompanied by other signs of central nervous system depression, including sedation and mental clouding. Respiratory distress associated with tachypnea and anxiety is never a primary opioid event and alternative explanations (e.g., pneumonia or pulmonary embolism) should be sought.

Because of the risk of systemic withdrawal and the return of pain, naloxone should only be administered for symptomatic respiratory depression. If the patient is arousable, and the peak plasma levels of the opioid have already been reached, the opioid dose should be withheld and the patient monitored until their condi- tion is improved. If the patient is unarousable or respira- tory depression is severe, naloxone should be used to improve ventilation using small bolus injections of di- lute solution (0.4 mg in 10 ml saline), which are titrated against respiratory rate.'21*'22 An intercurrent cardiac or pulmonary process can precipitate respiratory depres- sion in patients receiving chronic opioid therapy that is

Table 8. Stepwise Management of Opioid-Induced Sedation Eliminate nonessential central nervous system depressant

If analgesia is satisfactory, reduce opioid dose by 25% If analgesia is unsatisfactory, try addition of a psychostimulant

(starting dose: methylphenidate 5 mg twice daily, dextroamphetamine 5 mg twice daily, or pemoline 18.75 mg twice daily)

medications

If somnolence persists, consider: Addition of a nonopioid or adjuvant that will allow reduction in

Change to an alternative opioid drug Change to the intraspinal opioid (+local anesthetic) Consider invasive anesthetic or neurosurtzical techniques

opioid dose

Page 12: Pain and Cancer Mangement

3404 CANCER Supplement December 1, 2993, Volume 72, No. 11

Table 9. Management of Cognitive Impairment in Cancer Patients Receiving Opioid Therapy

Eliminate nonessential centrally acting medications If analgesia is satisfactory, reduce opioid dose by 25% Evaluate patient for concurrent causes (e.g., sepsis, metabolic

derangement, intracerebral or leptomeningeal metastases) and treat if possible

If delirium persists, consider Trial of neuroleptic (e.g., haloperidol) Change to an alternative opioid drug Change to the intraspinal opioid (flocal anesthetic) Consider invasive anesthetic or neurosurgical techniques

similarly responsive to naloxone. Hence, a naloxone re- sponse does not obviate the need for subsequent careful patient evaluation for a concurrent cardiopulmonary process.

Dependence and Addiction

Confusion about physical dependence and addiction augment the fear of opioid drugs and contribute to the undertreatment of pain.'7,'23-'27 This confusion derives, in part, from misunderstanding of the nomenclature used to describe drug use. For example, the term de- pendence is commonly used but is often misappre- hended. Patients who require a specific pharmacother- apy to control a symptom or disease process are depen- dent on the therapeutic efficacy of the drugs in question. Examples of this therapeutic dependence in- clude the requirements of patients with congestive car- diac failure for cardiotonic and diuretic medications, or the reliance of insulin-dependent diabetics on insulin therapy. Undermedication or withdrawal of treatment from these patients can result in serious untoward con- sequences, the fear of which can induce aberrant psy- chological responses and drug-seeking behaviors. In this sense, most patients with chronic cancer pain are therapeutically dependent on an opioid; this response may or may not be associated with the development of physical dependence, but is virtually never associated with addiction.

Physical dependence. Physical dependence is a pharmacologic property of opioid drugs solely defined by the development of an abstinence (withdrawal) syn- drome after either abrupt dose reduction or administra- tion of an antagonist. Although physical dependence is most commonly observed in patients taking large doses for a prolonged period of time, withdrawal can also occur in patients after low doses or short duration of treatment. Physical dependence is not a clinical prob- lem if patients are warned to avoid abrupt discontinua- tion of the drug, a tapering schedule is used if treatment cessation is indicated, and opioid antagonist drugs (in- cluding agonist-antagonist analgesics) are avoided.

Addiction. The term addiction should never be used when physical dependence is meant. Addiction refers to a psychological and behavioral syndrome characterized by a continued craving for an opioid drug to achieve a psychic effect (psychological dependence) and associated aberrant drug-related behaviors (e.g., compulsive use, or continued use despite harm to self or others). Addiction should be suspected if patients dem- onstrate compulsive use, loss of control over drug use, and continuing use despite harm.

The medical use of opioids is very rarely associated with the development of addiction."2~'24~'28 In the larg- est prospective study, only four cases of iatrogenic ad- diction could be identified among 11,882 patients with no history of addiction who received at least one opioid preparation in the hospital ~ett ing."~ Although there are no prospective studies in patients with chronic cancer pain, there is an extensive clinical experience that affirms the extremely low risk of addiction in this popU~ation~5,6,46,58,94,11Z,lZ3,1Z5,1Z8,130-13Z H ealth care pro- viders, patients, and families often require vigorous and repeated reassurance that the risk of addiction is ex- tremely small.

Pseudoaddiction. The distress engendered in pa- tients who are therapeutically dependent on analgesic pharmacotherapy but who continue to experience unrelieved pain is occasionally expressed in behaviors that are reminiscent of addiction, such as intense con- cern about opioid availability and unsanctioned dose escalation. Pain relief, usually produced by dose escala- tion, eliminates these aberrant behaviors and distin- guishes the patient from the true addict. This syndrome has been termed pseud~addiction. '~~ Misunderstand- ing these phenomena may lead the clinician to inappro- priately stigmatize the patient with the label "addict," which may compromise care and erode the doctor-pa- tient relationship. In the setting of unrelieved pain, aberrant drug-related behaviors require careful assess- ment, renewed efforts to manage pain, and avoidance of stigmatizing labels.

Noninvasive Interventions for Patients Unable to Attain an Acceptable Balance Between Relief and Side Effects of Systemic Opioids

Even with optimal management of adverse effects, some patients fail to attain an acceptable balance be- tween pain relief and the side effects of an opioid. Nu- merous noninvasive interventions (Table 1) can im- prove this balance by reducing the opioid requirement. These include the concurrent use of an appropriate pri- mary therapy, alternative pharmacologic approaches (nonopioid analgesic or an adjuvant analgesic) and the use of psychological, rehabilitative, or neurostimula- tory techniques (e.g., transcutaneous electrical nerve

Page 13: Pain and Cancer Mangement

Current Strategy for Cancer Pain ManagementlCherny and Portenoy 3405

stimulation). A switch to another opioid, as described previously, may also improve the therapeutic out- come.75

Adjuvant Analgesics

Adjuvant analgesics can be broadly divided into gen- eral purpose analgesics, and drugs with specific utility for either neuropathic or bone pain. There is a large interindividual and intraindividual variability in the re- sponses to these agents (including those within the same class), and sequential trials are frequently needed to identify optimal therapy.

General-purpose adjuvant analgesics. Corticoste- roids are the most widely used general-purpose adju- vant These drugs may ameliorate pain and produce beneficial effects on appetite, nausea, mood, and malai~e. '~~- '~ ' The painful conditions that commonly respond to corticosteroids include raised in- tracranial pressure, acute spinal cord compression, supe- rior vena cava syndrome, metastatic bone pain, neuro- pathic pain due to infiltration or compression by tumor, symptomatic lymphedema, and hepatic capsular dis- t e n t i ~ n . ' ~ ~ , ' ~ ~ Patients with advanced cancer who expe- rience pain and other symptoms that may respond to steroids are usually given a small dose (e.g., dexametha- sone 1-2 mg twice daily). An acute episode of very se- vere pain that is related to a neuropathic lesion ( e g , plexopathy or epidural spinal cord compression) or bony metastasis and cannot be promptly reduced with opioids may respond dramatically to a short course of relatively high doses ( e g , dexamethasone 100 mg IV followed initially by 96 mg/day in divided doses). In all cases, the dose should be gradually lowered after pain reduction to the minimum needed to sustain relief.

Adjuvant analgesics used for neuropathic pain. Neuropathic pain such as brachial or lumbosacral plex- opathy can be a major therapeutic challenge, and adju- vant drugs can play an important role in the manage- ment of these problems. For purpose of drug selection, it is useful to distinguish between continuous, lancinat- ing, and sympathetically-maintained neuropathic pain based on the patient's history and physical examination (Table

The tricyclic antidepressants are truly multipurpose analgesics, but are generally prescribed for neuropathic pain in cancer patients. They are typically used to man- age continuous dysesthesias that have not responded adequately to an opioid, and lancinating neuropathic pains refractory to other specific adjuvant d r ~ g s . ' ~ ~ , ' ~ ~ These compounds are also useful in patients with pain complicated by depression and i n s ~ m n i a . ' ~ ~ , ' ~ ~ Al- though the evidence for analgesic efficacy is greatest for the tertiary amine tricyclic drugs, such as amitriptyline, doxepin, and i m i ~ r a r n i n e , ' ~ ~ - ' ~ ~ the secondary amine

Table 10. Selection of Adjuvant Analgesics for Neuropathic Pain Based on Clinical Characteristics of the Pain

Sympathetically Continuous pain Lancinating pain maintained pain

Antidepressants Anticonvulsant drugs Phenoxybenzamine Amitript yline Carbamazepine Prazosin Doxepin Phen ytoin Corticosteroid Imipramine Clonazepam Nifedipine Desipramine Valproate Propranolol Notriptyline Baclofen Calcitonin Trazodone Local anesthetics Maprotiline Mexiletine

Local anesthetics Lidocaine Mexiletine Pimozide Lidocaine Antidepressants

Clonidine Capsaicin Calcitonin

drugs, such as desipramine and nortryptyline may be preferred when concern about sedation, anticholinergic effects, or cardiovascular toxicity is high. There is lim- ited evidence that the newer antidepressants, trazo- done and maprotiline, are analgesic and virtually no data in support of f l~oxet ine . '~~ The starting dose of a tricyclic antidepressant should be low (e.g., amitripty- line 10 mg in the elderly and 25 mg in younger pa- tients). Doses can be increased every few days by incre- ments the same size as the starting dose. The usual ef- fective dose for the most widely used of these drugs, amitriptyline, is 50-150 mg per day. It is reasonable to continue upward dose titration beyond this range, how- ever, when patients fail to achieve benefit and have no limiting side effects. Plasma drug concentration, if avail- able, may provide useful information and should be followed during the course of therapy. Very low levels in nonresponders suggest either poor compliance or an unusually rapid metabolism. In the latter case, doses can be increased while repeatedly monitoring the plasma drug level. Likewise, nonresponders whose plasma concentration is not very low, but is lower than the antidepressant range, should be considered for a trial of higher doses if side effects are not a problem.

Anticonvulsant drugs appear to be analgesic for di- verse types of lancinating (shooting) neuropathic pain.'42,'49 Clinical experience also supports the use of these drugs for patients with paroxysmal (but not lan- cinating) pains, and for those with continuous neuro- pathic pains that have not responded to other agents. Although carbamazepine is often preferred because of the high response rate observed in trigeminal neural- gia,150-153 caution is required in cancer patients with thrombocytopenia, those at risk for marrow failure

Page 14: Pain and Cancer Mangement

3406 CANCER Supplement December 2 , 2993, Volume 7 2 , No. 11

(e.g., after chemotherapy), and those whose blood counts must be monitored to determine disease status. If carbamazepine is used, a complete blood count should be obtained before the start of therapy, after 2 and 4 weeks, and then every 3-4 months thereafter. A leukocyte count below 4000 is usually considered to be a contraindication to treatment, and a decline to less than 3000, or an absolute neutrophil count of less than 1500 during therapy, should prompt discontinuation of the drug. Published reports and clinical experience also support trials with other anticonvulsant drugs, includ- ing p h e n y t ~ i n , ' ~ ~ , ' ~ ~ - ' ~ ~ ~ l o n a z e p a m , ' ~ ~ - ' ~ ~ and val- p r ~ a t e . ' ~ ~ , ' ~ ' Dosing guidelines for the use of these drugs as adjuvant analgesics are customarily identical to those employed in the treatment of seizures. Low initial doses are appropriate for carbamazepine, val- proate, and clonazepam, and the administration of phe- nytoin often begins with the presumed therapeutic dose (e.g., 300 mg/day) or a prudent oral loading regimen ( e g , 500 mg twice, separated by 4 hours). When low initial doses are used, dose escalation should ensue un- til a favorable effect occurs, intolerable side effects su- pervene, or the plasma drug concentration has reached a predetermined level, which is customarily at the up- per end of the therapeutic range for seizure manage- ment.

Baclofen, a gamma-aminobutyric acid-agonist ef- fective for trigeminal n e ~ r a l g i a , ' ~ ~ , ' ~ ~ is often employed in the management of lancinating pains due to neural injury of any type. A starting dose of 5 mg two to three times per day is gradually escalated until analgesia is achieved or adverse effects of sedation or confusion emerge.

Oral local anesthetic drugs may be considered in the management of neuropathic pains characterized by either continuous or lancinating dyse~thesias. '~~ The supporting evidence for this therapy is less abundant than that available for other drug classes; it is, there- fore, reasonable to undertake a trial with an oral local anesthetic in patients with continuous dysesthesias who fail to respond to tricyclic antidepressants, and in patients with lancinating pains refractory to trials of anticonvulsant drugs and baclofen. Mexiletine is the safest of these drugs and is preferred over flecainide and t ~ c a i n i d e . ' ~ ~ , ' ~ ~ Dosing with mexiletine should usually start at 100-150 mg/day. If intolerable side ef- fects do not occur, the dose can be increased by a like amount every few days, until the usual maximum dose of 300 mg three times per day is reached. Plasma drug concentrations, if available, can be helpful in monitor- ing the progress of the patient.

Topical administration of capsaicin cream depletes peptides in small primary afferent neurons, including compounds that are putative mediators of nociceptive

transmission (e.g., substance P).176,177 Analgesic effects have been observed in postherpetic neuralgia, painful peripheral neuropathies, and postmastectomy ~ a i n . ' ~ ~ - ' ~ ~ Tw o concentrations (0.025% and 0.075%) are commercially available. Although the dose-re- sponse relationship has not been evaluated in con- trolled studies, it is reasonable to use the higher concen- tration for either the initial trial or a subsequent trial after failure of the lower concentration product. A burning sensation can follow topical application of cap- saicin. This wanes spontaneously in some patients and others can reduce it through the prior use of an oral analgesic or cutaneous application of lidocaine 5% ointment. Some patients report intolerable burning and cannot continue therapy. In those who can tolerate the drug, an adequate trial should be considered at least four applications every day for 4 weeks.

Experience with other drugs in the treatment of cancer-related neuropathic pain is very limited. Cloni- dine, an alpha-2 adrenergic agonist available in oral or transdermal formulations, has antinociceptive effects in the management of diverse pains, but like the tricyclic antidepressants, is conventionally used for continuous neuropathic pain in the cancer p ~ p u l a t i o n . ' ~ ~ Calci- tonin (200 IU/day) has been shown to be an active analgesic in the management of some neuropathic pains,'83 and pimozide, a phenothiazine neuroleptic, has activity against lancinating neuropathic pain. The latter drug is not preferred because of a high incidence of adverse effects, including physical and mental slow- ing, tremor, and parkinsonian symptoms.

Some drugs are used specifically in the manage- ment of sympathetically-maintained pain (pain asso- ciated with local signs of autonomic dysregulation, such as edema, vasomotor instability, or sweating abnormali- ties). Although the preferred approach for this type of pain includes sympathetic nerve blocks, blocks may oc- casionally be contraindicated or fail. In this situation, several specific adjuvant analgesics have been advo- cated. Phenoxybenzamine, 60-1 20 mg per day, has been reported to be effective in a survey of patients with ~ausalgia , '~~ and there is similar limited evidence for the efficacy of p r a z o ~ i n , ' ~ ~ p r ~ p r a n o l o l , ' ~ ~ * ' ~ ~ and nifedipine. A recent controlled trial suggests that cal- citonin may also be useful in some patients with sym- pathetically maintained pain.'89

Adjuvant analgesics used for bone pain. Manage- ment of bone pain frequently requires the integration of opioid therapy with multiple ancillary approaches (Ta- ble 11). Anecdotally, NSAIDs appear to be particularly efficacious in pain of this type, and corticosteroids are often advocated in difficult cases.'90 The bisphospho- nate drugs, pamidronate and clodronate, inhibit osteo- clast activity and have been demonstrated to relieve

Page 15: Pain and Cancer Mangement

Current Strategy for Cancer Pain ManagementlCherny and Portenoy 3407

Table 11. Nonopioid Therapies for Bone Fain Primary therapy Psychiatric Drug therapy

Radiation therapy Orthotics Anti-inflammatory drugs Chemotherapy Assistive devices Nonstimulatory anti-inflammatory drugs Hormonal therapy Corticosteroids Surgical stabilization Bisphosphonates

Pamidronate Clodronate

Calcitonin Radiopharmaceuticals

Strontium-89 Samarium-153-ethylenediaminetetramethylene

Rhenium-1 86-hydroxyethylidene diphosphonate phosphonic acid

malignant bone pain in several recent surveys and con- trolled trial^.'^'-'^^ A tr ial of one of these agents can be recommended for patients with refractory bone pain. Serum calcium, phosphate, magnesium, and potassium should be monitored regularly during bisphosphonate therapy. Clinical experience also suggests that calci- tonin may occasionally provide significant relief of re- fractory bone pain, and a trial should be considered in patients who do not respond to a bisphosphonate. Fi- nally, newly developed radiopharmaceuticals that are absorbed at areas of high bone turnover may provide an important means of treating metastatic bone pain. These compounds, which include ~ t r o n t i u m - 8 9 , ~ ~ ~ * ~ ~ ' samarium-153-ethylenediaminetetramethylene phos- phonic a ~ i d , l ~ ~ - ~ " and rhenium-186-hydroxyethyli- dene diphosphonate,200~20z~zo3 are likely to become avail- able in the near future. Further studies are needed to identify the risks and benefits of each agent and the durability of the effects produced.

Other Noninvasive Analgesic Techniques

Psychological therapies. While all cancer patients benefit from psychological assessment and support, some will benefit from specific psychological interven- tions used in the management of pain. Cognitive-beha- vioral interventions can help reduce the perception of distress caused by the pain through the development of coping skills and the modification of thoughts, feelings, and behavior^.^^^,^^^ Some patients may be able to use relaxation techniques to reduce muscular tension and emotional arousal, or enhance pain Other approaches reduce anticipatory anxiety that may lead to avoidant behavior^."^ Successful application of these therapies requires a cognitively intact patient and a dedicated, well-trained clinician.204

Physiatric techniques. Physiatric techniques can be used to enhance analgesia and optimize the function of the patient with chronic cancer pain. Therapeutic

modalities such as electrical stimulation (including transcutaneous electrical nerve stimulation), heat, or cryotherapy can be useful adjuncts to standard analge- sic therapy. The treatment of lymphedema by use of wraps, pressure stockings, or pneumatic pump devices can both improve function and relieve pain and heavi- ness.208 Orthotic devices can immobilize and support painful or weakened structures, and assistive devices can enhance comfort for patients with pain precipitated by weight bearing or ambulation.

lnvasive Interventions for Patients Unable to Attain an Acceptable Balance Between Relief and Side Effects of Systemic Pharmacotherapy

Patients who are unable to achieve a satisfactory bal- ance between analgesia and side effects from systemic analgesic therapies may be candidates for the use of invasive analgesic techniques. Anesthetic and neurosur- gical approaches (Table 12) may reduce or eliminate the requirement for systemically administered opioids. Techniques such as intraspinal opioid and local anes- thetic administration were developed to achieve this end without need for compromising neurologic integ- rity. The use of neurodestructive procedures should be based on a careful evaluation of the likelihood and dura- tion of analgesic benefit, the immediate and long-term risks, the anticipated duration of survival of the patient, and the anticipated length of hospitalization.

Regional Opioid Analgesia

Epidural and intrathecal opioids. The delivery of low opioid doses near the sites of action in the spinal cord may decrease supraspinally mediated adverse ef- fects. Compared to neuroablative therapies, spinal opioids have the advantage of preserving sensation, strength, and sympathetic function. Contraindications include bleeding diathesis, profound leukopenia, and sepsis. A temporary trial of spinal opioid therapy

Page 16: Pain and Cancer Mangement

3408 CANCER Supplement December 1, 2993, Volume 72, No. 11

Table 12. Commonly Performed Anesthetic and Neurosurgical Analgesic Techniques for Pain Refractory to Systemic Pharmacotherapy

Class Technique Clinical situation

Regional analgesia

Sympathetic blockade and neurolysis

Somatic neurolysis or pathway ablation

Other

Spinal opioids and/or local anesthetics

Intraventricular

Celiac plexus block

Lumbar sympathetic

Stellate ganglion blockade Chemical or surgical

blockade

rhizotomy

Trigeminal neurolysis Transacral neurolysis Cordotomy Cingulotomy

Pituitarv ablation

Systemic opioid analgesia complicated by unmanageable supraspinally

Little reported experience, no well-defined indications Reported efficacy with both upper body and generalized pain Refractory malignant pain involving the upper abdominal viscera

including the upper retroperitoneum, liver, small bowel, and proximal colon

mediated adverse effects

Sympathetically maintained pain involving the legs

Sympathetically maintained pain involving the head, neck, or arms Refractory brachial plexopathy or arm pain Intercostal nerve pain, chest wall pain Refractory bilateral pelvic or lumbosacral plexus pain in a bedridden

patient with urinary diversion Refractory unilateral facial pain Refractory pain limited to the perineum Refractory unilateral pain arising in the torso or lower extremity Little reported experience, no well-defined indications Has been used for refractory multifocal pain Refractorv multifocal pain

should be performed to assess the potential benefits of this approach before implantation of a permanent cath- eter.

Opioid selection for intraspinal delivery is in- fluenced by several factors. Hydrophilic drugs, such as morphine and hydromorphone, have a prolonged half- life in cerebrospinal fluid and significant rostral redis- t r i b ~ t i o n . ~ ' ~ - ~ ~ ' Lipophilic opioids, such as fentanyl and sufentanil, have less rostral redistribution212 and may be preferable for segmental analgesia at the level of spinal infusion. In some patients, the addition of a low concentration of a local anesthetic, such as 0.125- 0.25% bupivacaine, to an epidural opioid has been dem- onstrated to increase analgesic effect without increasing t o x i ~ i t y . ~ ~ ~ - ~ ' ~ The potential morbidity for these proce- dures indicates the need for a well-trained clinician and long-term monitoring.

Intraventricular opioids. Limited experience exists with the administration of an opioid into the cerebral ventricles via an Ommaya re~ervoi r .~~ ' -~ '~ This tech- nique has been used for patients with upper body or head pain, or severe diffuse pain. In relatively nontoler- ant patients, the analgesia produced by a dose of mor- phine has a rapid onset and a long duration of

Anesthetic Techniques for Sympathetically- maintained Pain and Visceral Pain

Celiac plexus block. Neurolytic celiac plexus blockade can be considered in the management of pain caused by neoplastic infiltration of the upper abdomi- nal viscera, including the pancreas, upper retroperiton-

e m , liver, gall bladder, and proximal small b o ~ e 1 . ~ ~ ~ - ~ ~ ~ Reported analgesic response rates in patients with pan- creatic cancer are 50-90%, with the duration of effect lasting from 1 to 12 month^.'*^-^^^ Given the generally favorable response to this approach, most clinicians will recommend it as the next intervention for patients with an appropriate pain syndrome who fail to obtain an adequate balance between analgesia and side effects from an oral opioid. Common transient complications include postural hypotension and diarrhea.224-226 Poste- rior spread of neurolytic solution can lead to involve- ment of lower thoracic and lumbar somatic nerves and potentially result in neuropathic pain in the region of the lower rib cage or upper thigh. Uncommon compli- cations include pneumothorax, retroperitoneal hema- toma, and paraparesis.

Sympathetic blockade of somatic structures. Sympathetically maintained pain syndromes may be relieved by interruption of sympathetic outflow to the affected region of the body. Lumbar sympathetic block- ade should be considered for sympathetically main- tained pain involving the legs, and stellate ganglion blockade may be useful for sympathetically maintained pain involving the face or arms.

Neuroablative Techniques for Somatic and Neuropathic Pain

Chemical rhizotomy. Chemical rhizotomy, which may be produced by the instillation of a neurolytic so- lution into either the epidural or intrathecal space, can be an effective method of pain control for patients with

Page 17: Pain and Cancer Mangement

Current Strategy for Cancer Pain Management/Cherny and Portenoy 3409

otherwise refractory localized pain syndrome^.^^^,^^^ The technique is most commonly used in the manage- ment of chest wall pain due to tumor invasion of so- matic and neural structures. Other indications include refractory upper limb, lower limb, pelvic, or perineal pain.

Satisfactory analgesia is achieved in about 50% of patients who undergo epidural or intrathecal neuroly- is."^ Adverse effects can be related to the injection technique (e.g., spinal headache, infection, and arach- noiditis) or to the destruction of non-nociceptive nerve fibers. Specific complications of the procedure depend on the site of neurolysis. Complications of lumbosacral neurolysis include paresis (5-20%), sphincter dysfunc- tion (5-60%), impairment of touch and proprioception, and dysesthesias. Fortunately, neurologic deficits are usually transient, although fatal meningitis, paraplegia, and permanent impairment of sphincter function have been re~orded.”~ Because of the risk of increased dis- ability through weakness, sphincter incompetence, and loss of positional sense, chemical rhizotomy of lumbo- sacral nerve roots is best reserved for patients with lim- ited function and preexistent urinary diversion. It is es- sential that patients be counseled about the risks in- volved with these techniques.

Other chemical neurolyses. Neurolysis of primary afferent nerves may provide significant relief for se- lected patients with localized pain. Refractory unilateral facial or pharyngeal pain may be amenable to trigemi- nal neurolysis, Gasserian gangliolysis, or glossopha- ryngeal neurolysis. Intercostal or paravertebral neuro- lysis are alternatives to rhizotomy for patients with chest wall pain. Severe pain limited to the perineum may be treated by neurolysis of the 54 nerve root via the ipsilateral posterior sacral foramen, a procedure that carries a minimal risk of motor or sphincter impair- men^"^

Cordotomy. During cordotomy, the anterolateral spinothalamic tract is ablated to produce contralateral loss of pain and temperature s e n ~ i b i l i t y . ~ ~ ~ , ~ ~ ~ The pa- tient with severe unilateral pain arising in the torso or lower extremity is most likely to benefit from this pro- cedure.228 The percutaneous technique is generally pre- ferredZ3’; open cordotomy is usually reserved for pa- tients who are unable to lie in the supine position or are not cooperative enough to undergo a percutaneous pro- cedure, 230

Significant pain relief is achieved in more than 90% of patients during the period immediately following ord do to my.^^^^^^^-^^^ F ifty percent of surviving patients have recurrent pain after 1 year, and repeat cordotomy can sometimes be effective. The neurologic complica- tions of cordotomy include paresis, ataxia, and bladder dysfunction.232 The complications are usually transient,

but are protracted and disabling in approximately 5% of cases. Rarely, patients with a long duration of sur- vival (> 12 months) develop a delayed-onset dysesthe- tic pain. The most serious potential complication is re- spiratory dysfunction, which may result from phrenic nerve paralysis or occur as sleep-induced apnea (in pa- tients who undergo bilateral high c~rdotomy).’~~ The potential for this complication relatively contraindi- cates bilateral high cervical cordotomies or a unilateral cervical cordotomy ipsilateral to the site of the only functioning lung.

Other Techniques

Pituitary ablation. Pituitary ablation, by chemical or surgical hypophysectomy, has been reported to re- lieve diffuse and multifocal pain syndromes that have been refractory to opioid therapy and are unsuitable for any regional neuroablative p r o c e d ~ r e . ~ ~ ~ , ~ ~ ~ Pain relief has been observed from pain due to both hormone-de- pendent and hormone-independent t ~ m o r s . ~ ~ ~ , ~ ~ ~

Cingulotomy. Anecdotal reports also support the efficacy of c i n g u l ~ t o m y ~ ~ ~ in the management of dif- fuse pain syndromes that have been refractory to opioid therapy. The mode of action is unknown and the proce- dure is considered rarely.

Therapy for Patients with Refractory Pain: The Role of Sedation

The use of sedation therapy to manage intractable pain and suffering in the small population that fails to bene- fit from optimal therapy has recently received increas- ing at ten ti or^.^^^*^^' In one study, 52% of terminally ill patients developed otherwise unendurable symptoms that required deep sedation for adequate relief; in just under half of these patients, pain was the predominant symptom.”

The patient with advanced cancer and uncontrolled symptoms may choose transitory use of sedating ther- apy while continuing trials of analgesic approaches. Al- ternatively, a strategy can be designed to provide ade- quate sedation until death. The ethical basis of the latter approach is predicated on informed consent and an ac- knowledgment of the ”principle of double effect,” which distinguishes between the compelling primary therapeutic intent (to relieve suffering) and unavoid- able untoward consequences (the potential for accelerat- ing death).238 The use of this approach must recognize the right of dying patients to adequate relief of pain and the right of all patients to choose among appropriate therapeutic option^.'^^-'^^ No patient should have to ask to be killed because of persistently unrelieved pain, and on the contrary, no patient should be sedated with- out appropriate informed consent of the patient or

Page 18: Pain and Cancer Mangement

3410 CANCER Supplement December 1, 1993, Volume 72, No. 11

proxy. The process of informed decision making re- quires candid discussion that clarifies the prevailing clinical predicament and presents the alternative anal- gesic options (including sedation). The potentially con- flicting goals of comfort versus function may need to be made explicit. Other relevant considerations, including existential, ethical, religious, and familial concerns, may benefit from the participation of a religious counselor, social worker, or clinical ethics specialist.

Sedation can potentially be accomplished through the use of systemic opioids, with either a benzodiaze- pine (e.g., lorazepam or midazolam), a neuroleptic (e.g., chlorpromazine or methotrimeprazine), or a barbiturate (e.g., thiopental). Experience has been reported in the use of an opioid in conjunction with a continuous infu- sion of m i d a ~ o l a m ~ ~ ~ or t h i ~ p e n t a l . ~ ~ ~ , * ~ ~ It is the respon- sibility of the physician to ensure that the patient, fam- ily, and staff have a comprehensive understanding of this intervention.

Conclusion

Currently available techniques can provide adequate pain relief to the vast majority of cancer patients, most of whom will respond to systemic pharmacotherapy alone. Successful long-term management requires com- prehensive assessment and a continuity of care that provides an appropriate level of monitoring and re- sponds quickly, flexibly, and expertly to the changing needs of the patient. Patients with refractory pain, or unremitting suffering related to other losses or distress- ing symptoms should have access to specialists in pain management, palliative medicine and psychooncology, who can provide an approach capable of addressing these complex problems.

References

1. Johanson GA. Symptom character and prevalence during cancer patients’ last days of life. A m ] Hosp Palliat Care 1991; 8:6-8.

2. Bonica JJ, Ventafridda V, Twycross RG. Cancer Pain. In: Bonica JJ, editor. The management of pain. 2nd ed. Philadelphia: Lea & Febiger, 1990: 400-60.

3. Coyle N, Adelhardt J, Foley KM, Portenoy RK. Character of terminal illness in the advanced cancer patient: pain and other symptoms during last four weeks of life. ]Pain Symptom Manage

4. Foley KM, Inturrisi CE. Analgesic drug therapy in cancer pain: principles and practice. Med Clin North A m 1987; 71207-32.

5. Foley KM. The treatment of cancer pain. N Engl ] Med 1985;

6. World Health Organization. Cancer pain relief and palliative care. Geneva: World Health Organization, 1990.

7. Von Roenn JH, Cleeland CS, Gonin R, Pandya KJ. Results of a physician attitude toward cancer pain management survey by ECOG. Proc A m Soc Clin Oncol 1991; 10:326.

8. Whippen DA, Canellos GP. Burnout syndrome in the practice of medical oncology: result of a random survey of 1000 oncolo- gists. ] Clin Oncol 1991; 9:1916-20.

1990; 5~83-9.

313:84-95.

9. Abeloff MD. Burnout in oncology-physician heal thyself. ] Clin Oncol 1991; 9:1721-2.

10. Ad Hoc Committee on Cancer Pain of the American Cancer Society Cancer pain assessment and treatment curriculum guidelines. ] Clin Oncol 1992; 10:1976-82.

11. Walker VA, Hoskin PJ, Hanks GW, et al. Evaluation of WHO analgesic guidelines for cancer pain in a hospital-based pallia- tive care unit. J Pain Symptom Manage 1988; 3:145-50.

12. Ventafridda V, Tamburini M, Caraceni A, DeConno F, Naldi F. A validation study of the WHO method for cancer pain relief. Cancer 1987; 59351-6.

13. Takeda F. Results of field-testing in Japan of WHO Draft Interim Guidelines on Relief of Cancer Pain. Pain Clinic 1986; 1:83-9.

14. Schug SA, Zech D, Dorr U. Cancer pain management according to WHO analgesic guidelines. J Pain Symptom Manage 1990;

15. Grond S, Zech D, Schug SA, Lynch J, Lehman KA. Validation of the World Health Organization guidelines for cancer pain relief during the last days and hours of life. ] Pain Symptom Manage

16. Jorgensen L, Mortensen MB, Jensen NH, Eriksen J. Treatment of cancer pain patients in a multidisciplinary pain clinic. Pain Clinic

17. Cleeland CS. Pain control: public and physician’s attitudes. In: Hill CS, Fields WS, editors. Drug treatment of cancer pain in a drug-oriented society: advances in pain research and therapy. Vol 11. New York: Raven Press, 1989: 81-9.

18. Ventafridda V, Ripamonti C, De Conno F, Tamburini M, Cassi- leth BR. Symptom prevalence and control during cancer pa- tients’ last days of life. ] Palliative Care 1990; 6:7-11.

19. Reuben DB, Mor V, Hiris J . Clinical symptoms and length of survival in patients with terminal cancer. Arch Intern Med 1988;

20. Moinpour CM, Feigl P, Metch B, Hayden KA, Meyskens FJ, Crowley J. Quality of life endpoints in cancer clinical trials. ] Natl Cancer Inst 1989; 81:485-95.

21. Moinpour CM, Hayden KA, Thomson IM, Feigl P, Metch B. Quality of life assessment in Southwest Oncology Group trials. In: Tchekmedyian NS, Cella D, editors. Quality of life in oncol- ogy practice and research. Williston Park: Dominus Publishing

22. Gonzales GR, Elliot KJ, Portenoy RK, Foley KM. The impact of a comprehensive evaluation in the management of cancer pain. Pain 1991; 47:141-4.

23. Clouston P, De Angelis L, Posner JB. The spectrum of neurologic disease in patients with systemic cancer. Ann Neurol 1992;

24. Bates T. A review of local radiotherapy in the treatment of bone metastases and cord compression. Int ] Radiat Oncol Biol Phys

25. Bates T, Yarnold JR, Blitzer P, Nelson OS, Rubin P, Maher J. Bone metastasis consensus statement. Int ] Radiat Oncol Biol

26. Coia LR. The role of radiotherapy in the treatment of brain me- tastases. Int ] Radiat Oncol Biol Phys 1992; 23239-44.

27. Dobrowsky W, Schmidt AP. Radiotherapy for presacral recur- rence following radical surgery for rectal carcinoma. Dis Colon Rectum 1985; 28:917-9.

5: 2 7-32.

1991; 6:411-22.

1990; 3:83-9.

148:1586-91.

CO., 1991: 43-50.

31 ~268-73.

1992; 23:217-22.

Phys 1992; 23:215-6.

28. Bosch A. Radiotherapy. Clin Oncology 1984; 3:47-53. 29. Borgelt BB, Gelber R, Brady LW, Griffin T, Hendrickson FR. The

palliation of hepatic metastases. Results of a radiation therapy oncology group pilot study. Int ] Radiat Oncol Biol Phys 1981; 7:587-9 1.

Page 19: Pain and Cancer Mangement

Current Strategy for Cancer Pain Management/Cherny and Portenoy 3411

30. Prassad B, Lee M, Hendrickson FR. Irradiation of hepatic metas- tases. Int J Radiat Oncol Biol Phys 1977; 2:129-32.

31. Sherman DM, Weichselbaum R, Order SE, et al. Palliation of hepatic metastases. Cancer 1978; 41:2013-7.

32. Minsky BD, Leibel S. The treatment of hepatic metastases from colorectal cancer with radiation therapy alone or combined with chemotherapy or misonidazole. Cancer Treat Rev 1989; 16:213-9.

33. Patt YZ, Peters RE, Chuang VP, Wallace S, Claghorn L, Mavligit G. Palliation of pelvic recurrence of colorectal cancer with intra- arterial 5-fluorouracil and mitomycin. Cancer 1985; 56:2175-80.

34. MacDonald N. The role of medical and surgical oncology in the management of cancer pain. In: Foley KM, Bonica JJ, Venta- fridda V, editors. Second International Congress on Cancer Pain: advances in pain research and therapy. Vol 16. New York: Raven Press, 1990: 27-39.

35. Boraas M. Palliative surgery. Semin Oncol 1985; 12:368-74. 36. Sundaresan N, DiGiacinto GV. Antitumor and anti nociceptive

approaches to control cancer pain. Med Clin North A m 1987;

37. Williams MR. The place of surgery in terminal care. In: Saunders C, editor. The management of terminal disease. London: Ed- ward Arnold, 1984: 148-53.

38. Petrelli NJ, Velez A, Herrera L, Walsh D, Mittelman A. Ileal conduits for recurrent unresectable colorectal adenocarcinoma. A m J Surg 1985; 150:239-42.

39. Ross GJ, Kessler HB, Clair MR, Gatenby RA, Hartz WH, Ross LV. Sonographically guided paracentesis for palliation of symp- tomatic malignant ascites. A m J Roentgenol 1989; 153:1309-11.

40. Cruikshank DP, Buchsbaum HJ. Effects of rapid paracentesis. Cardiovascular dynamics and body fluid composition. J A M

41. Lifshitz S , Buchsbaum HJ. The effect of paracentesis on serum proteins. Gynecol Oncol 1976; 4:347-51.

42. Burney RE, Faulkner DJ. Recurrent abdominal abscess caused by retained colorectum after proctocolectomy. Surgery 1988;

43. Butch RJ, Wittenberg J, Mueller PR, Simeone JF, Meyer JE, Ferrucci JJ. Presacral masses after abdominoperineal resection for colorectal carcinoma: the need for needle biopsy. Am J Roent- genol 1985; 144:309-12.

44. Bruera E, McDonald N. Intractable pain in patients with ad- vanced head and neck tumors: a possible role of local infection. Cancer Treat Rep 1986; 70:691-692.

45. Coyle N, Portenoy RK. Infection as a cause of rapidly increasing pain in cancer patients. J Pain Symptom Manage 1991; 6:266-9.

46. World Health Organization. Cancer pain relief. Geneva: World Health Organization, 1986.

47. Vane JR. Inhibition of prostaglandin synthesis as a mechanism of action for aspirin-like drugs. Nature New Biol 1971; 234:231-8.

48. Malmberg AB, Yaksh TL. Hyperalgesia mediated by spinal glu- tamate and substance P receptors blocked by spinal cyclooxy- genase inhibition. Science 1992; 92:1276-9.

49. Willer J, DeBroucker T, Bussel B, Robi-Brami A, Harrewyn J. Central analgesic effect of ketoprofen in humans: electrophysio- logic evidence for a supraspinal mechanism in a double-blind and cross-over study. Pain 1989; 38:l-8.

50. Piletta P, Porchet HC, Dayer P. Central analgesic effect of acet- aminophen but not of aspirin. Clin Pharm Ther 1991; 49:350-4.

51. Brooks I'M, Wood AJJ. Nonsteroidal antiinflammatory drugs- differences and similarities. N EngI J Med 1991; 324:1716-25.

52. Insel PA. Analgesic-antipyretics and antiinflammatory agents: drugs employed in the treatment of rheumatoid arthritis and gout. In: Gilman AG, Rall TW, Nies AS, Taylor P, editors. The pharmacological basis of therapeutics. 8th ed. New York: Perga- mon Press, 1990: 638-81.

71~329-48.

1973; 225:1361-5.

104:580-2.

53. Seeff LB, Cuccherini BA, Zimmerman H1, Adler E, Benjamin S. Acetaminophen hepatotoxicity in alcoholics. Ann Intern Med

54. McQuay HJ, Jadad AR, Carroll D, Faura C, Glynn CJ, Moore RA, et al. Opioid sensitivity of chronic pain: a patient controlled an- algesia method. Anaesthesia 1992; 47757-67.

55. Jadad AR, Carroll D, Glynn CJ, Moore RA, McQuay HJ. Mor- phine responsiveness of chronic pain: double blind randomised crossover study with patient controlled analgesia. Lancet ii 1992;

56. Portenoy RK, Foley KM, Inturrisi CE. The nature of opioid re- sponsiveness and its implications for neuropathic pain: new hy- potheses derived from studies of opioid infusions. Pain 1990;

57. Cherny NI, Thaler HT, Friedlander-Klar H, Lapin J, Portenoy RK. Opioid responsiveness of neuropathic cancer pain: com- bined analysis of single-dose analgesic trials. Proc A m Soc Clin Oncol 1992; 11:1330.

58. American Pain Society. Principles of analgesic use in the treat- ment of acute pain and chronic cancer pain. A concise guide to medical practice. 3rd ed. Skokie, Illinois: American Pain Society, 1992.

59. Szeto HH, Inturrisi CE, Houde R, Saal R, Cheigh J, Reidengerg MM. Accumulation of normeperidine an active metabolite of meperidine, in patients with renal failure or cancer. Ann Intern Med 1977; 85:738-41.

60. Eisendrath SJ, Goldman B, Douglas J, Dimatteo L, Van DC. Me- peridine-induced delirium. A m ] Psychiatry 1987; 144:1062-5.

61. Intumsi CE, Umans JG. Meperidine biotransformation and cen- tral nervous system toxicity in animals and humans. In: Foley KM, Inturrisi CE, editors. Opioid analgesics in the management of clinical pain: advances in pain research and therapy. Vol. 8. New York: Raven Press, 1986: 143-54.

62. Umans JG, Inturrisi CE. Antinociceptive activity and toxicity of meperidine and normeperidine in mice. J Pharmacol Exp Ther

63. Kaiko RF, Foley KM, Grabinski PV, Heinrich G, Rogers AG, Inturrisi CE, et al. Central nervous system excitatory effects of meperidine in cancer patients. Ann Neurol 1983; 13:180-5.

64. Chan GL, Matzke GR. Effects of renal insufficiency on the phar- macokinetics and pharmacodynamics of opioid analgesics. Drug Intel1 Clin Pharm 1987; 21:773-83.

65. Hagen N, Foley KM, Cerbones DJ, Portenoy RK, Inturrisi CE. Chronic nausea and morphine-6-glucuronide. ] Pain Symptom Manage 1991; 6:125-8.

66. Osborne RJ, Joel SP, Slevin ML. Morphine intoxication in renal failure: the role of morphine-6-glucuronide. BMJ 1986; 292:

67. Sawe J, Svensson JO, Odar-Cederlof I. Kinetics of morphine in patients with renal failure. Lancet 1985; ii:211.

68. Portenoy RK, Foley KM, Stulman J, Khan E, Adelhardt J, Lay- man M, et al. Plasma morphine and morphine-6-glucuronide during chronic morphine therapy for cancer pain: plasma pro- files, steady state concentrations and the consequences of renal failure. Pain 1991; 47:13-9.

69. McQuay HJ, Carroll D, Faura CC, Gavaghan DJ, Hand CW, Moore RA. Oral morphine in cancer pain: influences on mor- phine and metabolite concentration. Clin Pharmacol Ther 1990;

70. Giacomini KM, Giacomini JC, Gibson TP, Levy G. Propoxy- phene and norpropoxyphene plasma concentrations after oral propoxyphene in cirrhotic patients with and without surgically constructed portacaval shunt. Clin Pharmacol Ther 1980;

1986; 104:399-404.

339~1367-71.

43~273-86.

1982; 2231203-6.

1548-9.

48~236-44.

28:417-24.

Page 20: Pain and Cancer Mangement

3412 CANCER Supplement December 1, 2993, Volume 72, No. 11

71. Neal EA, Meffin PJ, Gregory PB, Blaschke TF. Enhanced bio- availability and decreased clearance of analgesics in patients with cirrhosis. Gastroenterology 1979; 77:96-102.

72. Crotty B, Watson KJR, Desmond PV, Marshford ML, Wood LJ, Coleman J, et al. Hepatic extraction of morphine is impaired in cirrhosis. Eur Clin Pharmacol 1989; 36:501-6.

73. Patwardhan R, Johnson RF, Hoyumpa A, Sheehan JJ, Desmond PV, Wilkinson GR, et al. Normal metabolism of morphine in cirrhosis. Gastroenterology 1981; 81:1006-11.

74. Hasselstrom J, Eriksson LS, Persson A, Rane A, Svensson J, Sawe J. The metabolism and bioavailability of morphine in pa- tients with severe liver cirrhosis patients. Br ] Clin Pharmacol 1990; 29:289-97.

75. Galer BS, Coyle N, Pasternak GW, Portenoy RK. Individual vari- ability in the response to different opioids: report of five cases. Pain 1992; 49237-91.

76. Moulin DE, Ling GS, Pasternak GW. Unidirectional cross toler- ance between morphine and levorphanol in the rat. Pain 1988;

77. Houde RW, Wallenstein SL, Beaver WT. Evaluation of analge- 331233-9.

78

79

80

81

83

84

85

86

sics in patients with cancer pain. In: Lasagna L, editor. Interna- tional encylcopedia of pharmacology and therapeutics. Vol l. New York: Pergamon Press, 1966: 59-67. Beaver WT, Feise G. Comparison of the analgesic effect oxymor- phone by rectal suppository and intramuscular injection in pa- tients with postoperative pain. ] Clin Pharmacol 1977; 17:276. Hanning CD. The rectal absorption of opioids. In: Benedetti C, Chapman CR, Giron G, editors. Opioid analgesia, advances in pain research and therapy. Vol 14 New York: Raven Press, 1990: 259-69. Kaiko RF, Cronin C, Healy N, Pav J, Thomas G, Goldenheim PD. Bioavailability of rectal and oral MS-Contin. Proc A m Soc Clin Oncol 1989; 8:1307. Weinberg DS, Inturrisi CE, Reidenberg B, Moulin D, Nip TJ, Wallenstein SL, et al. Sublingual absorption of selected opioid analgesics. Cl fn Pharm Ther 1988; 44:335-42. 82. Bullingham R, McQuay H, Porter E, Allen M, Moore R. Sub- lingual buprenorphine used postoperatively: ten hour plasma drug concentration analysis. Br 1 Clin Pharmacol 1982; 13:665- 73. Whitman HH. Sublingual morphine: a novel route of narcotic administration. A m Nursing 1 1984; 84:939-44. Hirsch JD. Sublingual morphine sulphate in chronic pain man- agement. Clin Pharmacol 1984; 3:585. Calk KA, Kohler DR, Corso DM. Transdermally administered fentanyl for pain management. Clin Pharmacol 1992; 11:22-36. Portenoy PK, Southam M, Gupta SK, Lapin J, Layman M, In- turrisi CE, et al. Transdermal fentanyl for cancer pain: repeated dose pharmacokinetics. Anesthesiology 1993; 78:36-43.

87. Holley FO, Van-Steennis C. Postoperative analgesia with fen- tanyl: pharmacokinetics and pharmacodynamics of constant- rate I.V. and transdermal delivery. Br JAnaes th 1988; 60:608-13.

88. Gourlay GK, Kowalski SR, Plummer JL, Cherry DA, Szekely SM, Mather LE, et al. The efficacy of transdermal fentanyl in the treatment of postoperative pain: a double blind comparison of fentanyl and placebo systems. Pain 1990; 40:21-8.

89. Portenoy RK. Continuous intravenous infusions of opioid drugs. Med Clin North A m 1987; 71:233-41.

90. Bruera E, Brenneis C, Michaud M, Bacovsky R, Chadwick S, Emeno A, et al. Use of the subcutaneous route for the administra- tion of narcotics in patients with cancer pain. Cancer 1988;

91. Coyle N, Mauskop A, Maggard J . Continuous subcutaneous in- fusions of opiates in cancer patients with pain. Oncol Nursing Forum 1986; 13:53-7.

62:407-11.

92. Swanson G, Smith J, Bulich R, New P, Shiffman R. Patient-con- trolled analgesia for chronic cancer pain in the ambulatory set- ting: a report of 117 patients. ] Clin Oncol 1989; 7:1903-8.

93. Storey P, Hill HH, St. Louis R, Tarver EE. Subcutaneous infu- sions for control of cancer symptoms. 1 Pain Symptom Manage

94. Twycross RG, Lack SA. Symptom control in far-advanced cancer: pain relief. London: Pitman, 1984.

95. Bruera E, Brenneis C, Michaud M, MacMillan K, Hanson J, Mac- Donald RN. Patient-controlled subcutaneous hydromorphone versus continuous subcutaneous infusion for the treatment of cancer pain. 1 Nat l Cancer lnst 1988; 80:1152-4.

96. Bruera E, Macmillan K, Selmser P, MacDonald RN. Decreased local toxicity with subcutaneous diamorphine (heroin): a prelimi- nary report. Pain 1990; 43:91-4.

97. Moulin DE, Kreeft JH, Murray PN, Bouquillon AI. Comparison of continuous subcutaneous and intravenous hydromorphone infusions for management of cancer pain. Lancet i 1991;

98. Moulin DE, Johnson NG, Murray PN, Geoghegan MF, Good- win VA, Chester MA. Subcutaneous narcotic infusions for cancer pain: treatment outcome and guidelines for use. Can Med

99. Bruera E, Fainsinger R, Moore M, Thibault R, Spoldi E, Venta- fridda V. Local toxicity with subcutaneous methadone. Experi- ence of two centers. Pain 1991; 45:141-5.

100. Dover SB. Syringe driver in terminal care. BM] 1987; 294:553-5. 101. Amesbury BDW. Use of SC Midazolam in the home care setting.

Palliative Med 1989; 3:299-301. 102. Oliver DJ. Syringe drivers in palliative care: a review. Palliative

Med 1988; 2 2 - 6 . 103. Portenoy RK, Hagen NA. Breakthrough pain: definition, preva-

lence and characteristics. Pain 1990; 41:273-81. 104. Arkinstall WW, Goughnour BR, White JA, Stewart JH. Control

of severe pain with sustained-release morphine tablets v. oral morphine solution. Can Med Assoc 1 1989; 140:653-7.

105. Shepherd K. Review of a controlled-release morphine prepara- tion. In: Foley KM, Bonica JJ, Ventafridda V. Second Interna- tional Congress on Cancer Pain: advances in pain research and therapy. Vol 16. New York: Raven Press, 1990: 191-202.

106. Warfield CA. Guidelines for the use of MS Contin tablets in the management of cancer pain. Postgrad Med 1 1991; 67:12-6.

107. Citron M, Johnston-Early A, Boyer M, Brasnow S, Hood M, Co- hen M. Patient-controlled analgesia for severe cancer pain. Arch lntern Med 1986; 146:734-6.

108. Brescia FJ, Portenoy RK, Ryan M, Krasnoff L, Gray G. Pain, opioid use, and survival in hospitalized patients with advanced cancer. 1 Clin Oncol 1992; 10:149-55.

109. Kanner RM, Foley KM. Patterns of narcotic drug use in a cancer pain clinic. A n n NY Acad Sci 1981; 362161-72.

110. Edwards WT, Breed RJ. Treatment of postoperative pain in the post anesthesia care unit. Anes th Clin North A m 1990; 8935-65.

11 1. Foley KM. Clinical tolerance to opioids. In: Basbaum AI, Bessom JM, editors. Towards a new pharmacotherapy of pain, Dahlem Konfrenzen. Chichester: John Wiley & Sons, 1991: 181-204.

112. Foley KM. Controversies in cancer pain: medical perspective. Cancer 1989; 63:2257-65.

113. Coyle N, Adelhardt J, Foley KM. Disease progression and toler- ance in the cancer pain patient. 2nd International Congress on Cancer Pain. 1 Pain Symptom Manage 1988; 3:S25.

114. Sykes NP. Oral naloxone in opioid associated constipation. Lan- cet i 1991; 337:1475.

115. Culpepper-Morgan JA, Inturrisi CE, Portenoy RK, Foley KM, Houde RW, Marsh F, et al. Treatment of opioid induced consti-

1990; 5133-41.

337:465-8.

ASSOC 1 1992; 146:891-7.

Page 21: Pain and Cancer Mangement

Current Strategy for Cancer Pain ManagementlCherny and Portenoy 3413

pation with oral naloxone: a pilot study. Clin Pharmacol Ther

116. Campora E, Merlini L, Pace M, Bruzzone M, Luzzani M, Gott- leib A, et al. The incidence of narcotic induced emesis. ] Pain Symptom Manage 1991; 6:428-430.

117. Bruera E, Macmillan K, Hanson J, MacDonald RN. The cognitive effects of the administration of narcotic analgesics in patients with cancer pain. Pain 1989; 39:13-6.

118. Breitbart W, Holland JC. Psychiatric complications of cancer. Curr Therap Hemat Oncol 1988; 3:268-75.

119. Eisele JH, Grigsby EJ, Dea G. Clonazepam treatment of myo- clonic contractions associated with high dose opioids: a case re- port. Pain 1992; 49:231-2.

120. Foley KM. Clinical tolerance to opioids. In: Basbaum AI, Besson JM, editors. Towards a new pharmacotherapy of pain, Dahlem Konferenzen. Chichester: John Wiley & Sons, 1991: 181-204.

121. Goldfrank L, Weisman RS, Errick JK, Lo MW. A dosing nomo- gram for continuous infusion of intravenous naloxone. A n n Emergency Medicine 1986; 15:91-5.

122. Bradberry JC, Reabel MA. Continuous infusion of naloxone in the treatment of narcotic overdose. Drug Intell Clin Pharm 1981;

123. Hill CS, Fields WS, editors. Drug treatment of cancer pain in a drug oriented society: advances in pain research and therapy. Vol 11. New York: Raven Press, 1989.

124. Schuster CR. Does treatment of cancer pain with narcotics pro- duce junkies? In: Hill CS, Fields WS. Drug treatment of cancer pain in a drug oriented society: advances in pain research and therapy. Vol 11. New York: Raven Press, 1989: 1-3.

125. Foley KM. The decriminalization of cancer pain. In: Hill CS, Field H. Drug treatment of cancer pain in a drug-oriented soci- ety: advances in pain research and therapy. Vol 11. New York: Raven Press, 1989: 5-18.

126. Jaffe JH. Misinformation: euphoria and addiction. In: Hill CS, Fields WS. Drug treatment of cancer pain in a drug oriented society: advances in pain research and therapy. Vol 11. New York: Raven Press, 1989: 163-74.

127. Morgan JP. American opiophobia: customary underutilization of opioid analgesics. In: Hill CS, Fields WS, editors. Drug treat- ment of cancer pain in a drug oriented society: advances in pain research and therapy. Vol 11. New York Raven Press, 1989:

128. Chapman CR, Hill HF. Prolonged morphine self-administration and addiction liability. Cancer 1989; 63:1636-44.

129. Porter J, Jick H. Addiction rare in patients treated with narcotics. N Engl Med 1980; 302:123.

130. American College of Physicians Health and Public Policy Com- mittee. Drug therapy for severe chronic pain in terminal illness. A n n Intern Med 1983; 99:870-80.

131. Inturrisi CE. Management of cancer pain. Cancer 1989; 63:2308- 20.

132. Walsh TD. Prevention of opioid side effects. ] Pain Symptom Manage 1990; 5:363-7.

133. Weissman DE, Haddox JD. Opioid pseudoaddiction-an iatro- genic syndrome. Pain 1989; 36:363-6.

134. Ettinger AB, Portenoy RK. The use of corticosteroids in the treat- ment of symptoms associated with cancer. J P a i n Symptom Man- age 1988; 3:99-103.

135. Walsh TD. Adjuvant analgesic therapy in cancer pain. In: Foley KM, Bonica JJ, Ventafrida V, editors. The Second International Conference on Cancer Pain: advances in pain research and ther- apy. Vol 16. New York: Raven Press, 1990: 155-68

136. Della Cuna GR, Pellegrini A, Piazzi M. Effect of methylpredni- solone sodium succinate on quality of life in preterminal cancer

1992; 52:90-5.

15185-90.

181-9.

patients. A placebo-control multicenter study. Eur J Cancer Clin

137. Tannock I, Gospodarowicz M, Meakin W, Panzarella T, Stewart L, Rider W. Treatment of metastatic prostatic cancer with low- dose prednisone: evaluation of pain and quality of life as prag- matic indices of response. J CIin Oncol 1989; 7:590-7.

138. Popiela T, Lucchi R, Giongo F. Methylprednisolone as palliative therapy for female terminal cancer patients. The Methylpredni- solone Female Preterminal Cancer Study Group. Eur J Cancer Clin Oncol 1989; 25:1823-9.

139. Moertel CG, Shutte A, Reitemeier R, et al. Corticosteroid ther- apy of preterminal gastrointestinal cancer. Cancer 1974;

140. Wilcox JC, Corr J, Shaw J, Richardson M, Calman KC. Predniso- lone as appetite stimulant in patients with cancer. BMJ 1984; 288:27.

141. Bruera E, Roca E, Cedaro L, Carraro S, Chacon R. Action of oral methylprednisolone in terminal cancer patients: a prospective randomized double-blind study. Cancer Treat Rep 1985; 69:751-4.

142. Portenoy RK. Adjuvant analgesics in pain management. In: Doyle D, Hanks GW, MacDonald N, editors. Oxford textbook of palliative medicine. Oxford: Oxford University Press, 1993:

143. Walsh TD. Antidepressants in chronic pain. Clin Neuropharma-

144. Breivik H, Rennemo F. Clinical evaluation of combined treat- ment with methadone and psychotropic drugs in cancer pa- tients. Acta Anaesth Scand 1982; 74:135-40.

145. Magni G, Arsie D, DeLeo D. Antidepressants in the treatment of cancer pain. A survey in Italy. Pain 1987; 29:347-53.

146. Hugues A, Chauvergne J, Lissilour T, Lagarde C. L'imipramine utilisee comme antalgique majeur en carcinologie: etude de 118 cas. Presse Medicale 1963; 71:1073-4.

147. Walsh TD. Controlled study of imipramine and morphine in chronic pain due to cancer. Proc A m Soc Clin Oncol 1986; 5:237.

148. Panerai AE, Bianchi M, Sacerdote P, Ripamonti C, Ventafridda V, De Conno F. Antidepresessants in cancer pain. ] Palliat Care

149. Swerdlow M. Anticonvulsant drugs and chronic pain. Clin Neuropharmacol 1984; 7:51-82.

150. Lloyd-Smith DL, Sachdev KK. A long-term, low dosage study of carbamazepine in trigeminal neuralgia. Headache 1969; 9:64-72.

151. Rockliff BW, Davis EH. Controlled sequential trials of carba- mazepine in trigeminal neuralgia. Arch Neural 1966; 15:129- 136.

152. Taylor JC, Brauer S, Espir MLE. Long-term treatment of trigemi- nal neuralgia with carbamazepine. Postgrad Med J 1980; 5716-8.

153. Tomson T, Bertilsson L. Potent therapeutic effects of carbamaz- epine 10,ll-epoxide in trigeminal neuralgia. Arch Neurol 1984;

154. Lockman LA, Hunninghake DB, Drivit W, Desnick RJ. Relief of pain of Fabry's disease by diphenylhydantoin. Neurology (Min- neapj 1973; 232371-5.

155. Chadda VS, Mathur MS. Double-blind study of the effects of diphenylhydantoin sodium in diabetic neuropathy. J Assoc Phy- sicians India 1978; 26:403-6.

156. Braham J, Saia A. Phenytoin in the treatment of trigeminal and other neuralgias. Lancet 1960; ii:892-4.

157. Ellenberg M. Treatment of diabetic neuropathy with diphenyl- hydantoin. Ny State ] Med 1968; 68:2633-55.

158. Ianmore A, Baker AR, Morrell F. Dilantin in the treatment of trigeminal neuralgia. Neurology 1958; 8:126-8.

159. Taylor PH, Gray K, Bicknell RG, Rees JR. Glossopharyngeal neu- ralgia with syncope. Laryngol Otol 1977; 91:859-68.

Oncol 1989; 29:1817-21.

33:1607-9.

187-203.

C O ~ 1983; 6:271-95.

1991; 7:42-4.

411598-601.

Page 22: Pain and Cancer Mangement

3414 CANCER Supplement December 2,2993, Volume 72, No. 11

160. Ekbom K. Carbamazepine in the treatment of tabetic lightning pains. Arch Neurol 1972; 26:374-8.

161. Hatangdi VS, Boas RA, Richards EG. Postherpetic neuralgia: management with antiepileptic and tricyclic drugs. In: Bonica JJ, Albe-Fessard D, editors. Advances in pain research and therapy. Vol 1. New York: Raven Press, 1976: 583-7.

162. Hallag IY, Harris JD. The syndrome of postherpetic neuralgia: complications and an approach to therapy. J A m Osteopathic

163. Cantor FK. Phenytoin treatment of thalamic pain. BMJ 1972; 2590.

164. Mladinich EK. Diphenylhydantoin in the Wallenberg syn- drome. JAMA 1974; 230:372-3.

165. Raskin NH, Levinson SA, Hoffman PM, Pickett JBE, Fields HL. Postsympathectomy neuralgia: amelioration with diphenylhy- dantoin and carbamazepine. A m J Surg 1974; 128:75-8.

166. Green JB. Dilantin in the treatment of lightning pains. Neurology (Minneap) 1961; 11257-8.

167. Caccia MR. Clonazepam in facial neuralgia and cluster head- ache: clinical and electrophysiological study. Eur Neurol 1975;

168. Martin G. The management of pain following laminectomy for lumbar disc lesions. Ann R Coll Surg Engl 1981; 63:244-52.

169. Swerdlow M, Cundill JG. Anticonvulsant drugs used in the treatment of lancinating pains. A comparison. Anesthesia 1981;

170. Peiris JB, Perera GLS, Davendra SV, Lionel NDW. Sodium val- proate in trigeminal neuralgia. Med J Aust 1980; 2:278.

171. Raftery H. The management of postherpetic pain using sodium valproate and amitriptyline. J Irish Med Assoc 1979; 72399-401

172. Fromm GH, Terrence CF, Chattha AS. Baclofen in the treatment of trigeminal neuralgia: double-blind study and long-term fol- low-up. Ann Neurol 1984; 15:240-4.

173. Fromm GH. Trigeminal neuralgia and related disorders. In: Por- tenoy RK, editor. Pain: mechanisms and syndromes, neurologic clinics. Vol 7. Philadelphia: WB Saunders Co, 1989: 305-19.

174. CAST (Cardiac Arrhythmia Suppression Trial) Investigators. Preliminary report: effect of encainide and flecainide on mortal- ity in a randomized trial of arrhythmia suppression after acute myocardial infarction. N Engl J Med 1989; 321:406-12.

175. Horn HR, Hadidian Z, Johnson JL, Vasallo HG, Williams JH, Young MD. Safety evaluation of tocainide in an American emer- gency use program. A m Hearf 11980; 100:1037-40.

176. Jesse1 TM, Dodd J. Functional chemistry of primary afferent neu- rons. In: Wall PD, Melzack R, editors. Textbook of pain. Edin- burgh: Churchill Livingston, 1989: 82-99.

177. Dubner R. Topical capsaicin therapy for neuropathic pain. Pain

178. Watson CPN, Evans RJ, Watt VR. Postherpetic neuralgia and topical capsaicin. Pain 1988; 33:333-40.

179. The Capsaicin Study Group. Treatment of painful diabetic neu- ropathy with topical capsaicin. A multicenter, double-blind, ve- hicle-controlled study. Arch Intern Med 1991; 151:2225-9.

180. Scheffler NM, Sheitel PL, Lipton MN. Treatment of painful dia- betic neuropathy with capsaicin 0.075%. J A m Podiatr Med Assoc

181. Tandan R, Lewis GA, Krusinski PB, Badger GB, Fries TJ. Topical capsaicin in painful diabetic neuropathy. Controlled study with long-term follow-up. Diabefes Care 1992; 15:8-14.

182. Watson CPN, Evans RJ, Watt VR. The post-mastectomy pain syndrome and the effect of topical capsaicin. Pain 1989; 38:177- 86.

183. Jaeger H, Maier C. Calcitonin in phantom limb pain: a double- blind study. Pain 1992; 4821-7.

ASSOC 1968; 681~1255-8.

13:560-3.

36:1129-32.

1991; 47~247-8.

1991; 81 :288-93.

184. Ghostine SY, Comair YG, Turner DM, Kassell NF, Azar CG. Phenoxybenzamine in the treatment of causalgia. 1 Neurosurg

185. Abram SE, Lightfoot RW. Treatment of longstanding causalgia with prazosin. Reg Anesth 1981; 6:79-81.

186. Simson G. Propranolol for causalgia and Sudeks atrophy. JAMA 1974; 227:32?.

187. Scadding JW. Clinical trial of propranolol in post-traumatic neu- ralgia. Pain 1982; 14983-92.

188. Prough DS, McLeskey CH, Poehling GG, Koman LA, Weeks DB, Whitworth T, et al. Efficacy of oral nifedipine in the treat- ment of reflex sympathetic dystrophy. Anesthesiology 1985;

189. Gobelet C, Waldburger M, Meier JL. The effect of adding calci- tonin to physical treatment on reflex sympathetic dystrophy.

190. Nielsen OS, Munro AJ, Tannock IF. Bone metastases: pathophys- iology and management policy. J Clin Oncol 1991; 9:509-24.

191. Coleman RE, Woll PJ, Miles M, Scrivener W, Rubens RD. Treat- ment of bone metastases with (3 amino-1-hydroxyprpy1idene)- 1, 1-bisphosphonate (APD). Br J Cancer 1988; 58:621-5.

192. Thiebaud D, Leyvraz S, von Fliedner V, Perey L, Cornu P, Thie- baud S, et al. Treatment of bone metastases from breast cancer and myeloma with pamidronate. Eur J Cancer 1991; 27:37-41.

193. Morton AR, Cantrill JA, Pillai GV, McMahon A, Anderson DC, Howell A. Sclerosis of lytic bony metastases after aminohy- droxyprpylidene bisphosphonate (APD) in patients with breast cancer. BMI 1988; 297:772-3.

194. Van Holten-Verzantvoort ATM, Zwinderman AH, Aaronson NK, Hermans J, van Emmerik B, van Dam FSAM, et al. The effect of supportive pamidronate treatment on aspects of quality of life of patients with advanced breast cancer. Eur J Cancer 1991; 27:544-9.

195. Clarke NW, Holbrook IB, McClure J, George N]. Osteoclastinhi- bition by pamidronate in metastatic prostate cancer: a prelimi- nary study. Br J Cancer 1991; 63:420-3.

196. Ernst DS, MacDonald RN, Paterson AHG, Jensen J, Brasher P, Bruera E. A double blind, crossover trial of I.V. clodronate in metastatic bone pain. J Pain Symptom Manage 1992; 7:4-11.

197. Silberstein EB, Williams C. Strontium-89 therapy for the pain of osseous metastases. J Nucl Med 1985; 26:345-8.

198. Robinson RG, Preston DF, Spicer JA, Baxter KG. Radionuclide therapy of intractable bone pain: emphasis on strontium-89. Se- min Nucl Med 1992; 22:28-32.

199. Turner JH, Claringbold BG, Heatherington EL, Sorby P, Martin- dal AA. A phase 1 study of samarium-153 ethylenedi-amene- tetramethylene phosphonate therapy for disseminated skeletal metastases. J Clin Oncol 1989; 7:1926-31.

200. Holmes RA. [153Sm]EDTMP: a potential therapy for bone cancer pain. Semin Nucl Med 1992; 2241-5.

201. Turner JH, Claringbold PG. A phase I1 study of treatment of painful multifocal skeletal metastases with single and repeated dose samarium-153 ethylenediaminetetramethylene phospho- nate. EurJ Cancer 1991; 27:1084-6.

202. Maxon HR, Deutsch EA, Thomas SR, Libson K, Lukes SJ, Wil- liams CC, et al. Initial experience with 186-Re(Sn)-HEDP in the treatment of painful skeletal metastases. J Nucl Med 1988; 29:776.

203. Maxon H, Thomas SR, Hertzberg VS, Schroder LE, Englaro EE, Samaratunga R, et al. Rhenium-186 hydroxyethylidene diphos- phonate for the treatment of painful osseous metastases. Semin Nucl Med 1992; 2233-40.

204. Fishman B. The treatment of suffering in patients with cancer pain: cognitive behavioral approaches. In: Foley KM, Bonica JJ,

1984; 60:1263-8.

62:796-9.

Pait1 1992; 48~171-5.

Page 23: Pain and Cancer Mangement

Current Strategy for Cancer Pain Management/Cherny and Portenoy 3415

Ventafridda V, editors. Second International Congress on Cancer Pain: advances in pain research and therapy. Vol 16. New York: Raven Press, 1990: 301-16.

205. Fordyce WE. Behavioral methods for chronic pain and illness. St. Louis: C.V. Mosby, 1976.

206. Linton SL, Melin L. Applied relaxation in the management of cancer pain. Behav Psychother 1983; 11:337-50.

207. Turk D, Meichenbaum D, Genest M. Pain and behavioral medi- cine. New York: Guilford Press, 1983.

208. Brennan MJ. Management of lymphedema: review of patho-

209

210

211

212

213.

214

215.

216.

217.

218.

219.

physiology and treatment. ] Pain Symptom Manage 1992; 7:

Moulin DE, lnturrisi CE, Foley KM. Epidural and intrathecal opioids: cerebrospinal fluid and plasma pharmacokinetics in cancer pain patients. In: Foley KM, Inturrisi CE, editors. Opioid analgesics in the management of clinical pain: advances in pain research and therapy. Vol8. New York: Raven Press, 1986: 369- 84. Max MB, Inturrisi CE, Kaiko RF, Grabinski PY, Li CH, Foley KM. Epidural and intrathecal opiates: cerebrospinal fluid and plasma profiles in patients with chronic cancer pain. Clin Pharmacol Ther 1985; 38:631-41. Brose WG, Tanalian DL, Brodsky JB, Mark JBD, Cousins MJ. C.S.F. and blood pharmacokinetics of hydromorphone and morphine following lumbar epidural administration. Pain 1991;

Chrubasik J, Wust H, Schulte MJ, Thon K, Zindler M. Relative analgesic potency of epidural fentanyl, alfentanil, and morphine in treatment of postoperative pain. Anesthesiology 1988; 68:929- 33. Du Pen SL, Ramsey DH. Compounding local anaesthetics and narcotics for epidural analgesia in cancer out patients. Anaesthe- siology 1988; 69(3A):A404. Nitescu P, Appelgren L, Linder LE, Sjoberg M, Hultman E, Cur- elaru I. Epidural versus intrathecal morphine-bupivicaine: as- sessment of consecutive treatment is advanced cancer pain. ] Pain Symptom Manage 1990; 5:18-26. Hogan Q, Haddox JD, Abram S, Weissman D, Taylor ML, Janjan N. Epidural opiates and local anesthetics for the management of cancer pain. Pain 1991; 46:271-9. Sjoberg M, Appelgren L, Einarsson S, Hultman E, Linder LE, Nitescu P, et al. Long-term intrathecal morphine and bupiva- caine in “refractory” cancer pain. Results from the first series of 52 patients. Acta Anaesfhesiol Scand 1991; 35:30-43. Obbens EAMT, Hill CS, Leavens ME, Ruthenbeck SS, Otis F. Intraventricular morphine administration for control of chronic cancer pain. Pain 1987; 28:61-8. Lobato RD, Madrid JL, Fatela LV, Sarabia R, Rivas JJ, Gozalo A. Intraventricular morphine for intractable cancer pain: rationale, methods, clinical results. Acta Anaesth Scand Suppl 1987; 31:68- 74. Leavens ES, Hill CS, Cech DA, Weyland JB, Weston JS. lntrathe-

110-6.

45:ll-7.

cal and intraventricular morphine for pain in cancer patients. ] Neurosurg 1982; 56:241-5.

220. Lee TL, Kumar A, Baratham G. Intraventricular morphine for intractable craniofacial pain. Singapore Med ] 1990; 31:273-6.

221. Nurchi G. Use of intraventricular and intrathecal morphine in intractable pain associated with cancer. Neurosurgery 1984;

222. Roquefeil B, Benezich J, Blanchet P, Baiter C, Frerebeau P, Gros C. Intraventricular administration of morphine in patients with neoplastic intractable pain. Surg Neurol 1984; 21:155-8.

223. Sandouk P, Serrie A, Urtizberea M, Debray M, Got P, Scherr- mann MJ. Morphine pharmacokinetics and pain assessment

15:801-4.

after intracerebroventricular administration in patients with ter- minal cancer. Clin Pharmacol Ther 1991; 49:442-8.

224. Brown DL, Bulley CK, Quiel EC. Neurolytic celiac plexus block- ade for pancreatic cancer pain. Anesth Analg 1987; 66:869-73.

225. Cousins MJ, Dwyer B, Gibb D. Chronic pain and neurolytic blockade. In: Cousins MJ, Bridenbaugh PO, editors. Neural blockade in clinical anesthesia and management of pain. 2nd ed. Philadelphia: JB Lippincott, 1988: 1053-84.

226. Bonica JJ, Buckley FP, Moricca G, Murphey TM. Neurolytic blockade and hypophysectomy. In: Bonica JJ, editor. The man- agement of pain. 2nd ed. Vol 2. Philadelphia: Lea & Febiger,

227. Hellendoorn SM, Overweg van Kints J. Perineal pain: a case report. Eur ] Surg Oncol 1988; 14:197-8.

228. Ischia S, Luzzani A, Ischia A, Magon F, Toscano D. Subarach- noid neurolytic block (L5-S1) and unilateral percutaneous cervi- cal cordotomy in the treatment of pain secondary to pelvic malig- nant disease. Pain 1984; 20:139-49.

229. Robertson DH. Transsacral neurolytic nerve block. An alterna- tive approach to intractable perineal pain. Br J Anaesth 1983;

230. Arbit E. Neurosurgical management of cancer pain. In: Foley KM, Bonica JJ, Ventafrida V. Second International Congress on Cancer Pain: advances in pain research and therapy. Vol 16. New York Raven Press, 1990: 289-300.

231. Rosomoff HL, Carroll F, Brown J. Percutaneous radiofrequency cervical cordotomy: technique. ] Neurosurg 1965; 23:639-644.

232. Tasker RR, Tsuda T, Howrylyshn P. Percutaneous cordotomy- the lateral high cervical technique. In: Schmidek HH, Sweet WH, editors. Operative neurosurgical technique indications, methods and results. New York: Grune and Stratton, 1988:

233. Polatty RC, Cooper KR. Respiratory failure after percutaneous cordotomy. South Med ] 1986; 79:367-79.

234. Levin AB, Ramirez LL. Treatment of cancer pain with hypophy- sectomy: surgical and chemical. In: Benedetti C, Chapman CR, Moricca G, editors. Recent advances in the management of pain, advances in pain research and therapy. Vol 7. New York: Raven Press, 1984: 63 1-45.

235. Hassenbusch SJ, Pillay PK, Barnett GM. Radiofrequency cingu- lotomy for intractable cancer pain using stereotaxis guided by magnetic resonance imaging. Neurosurgery 1990; 27:220-3.

236. Greene WR, Davis WH. Titrated intravenous barbiturates in the control of symptoms in patients with terminal cancer. South Med

237. Troug RD, Berde CB, Mitchell C, Greir HE. Barbiturates in the care of the terminally ill. N Engl ] Med 1992; 327:1678-82.

238. Latimer EJ. Ethical decision-making in the care of the dying and its application to clinical practice. ]Pain Symptom Manage 1991;

239. Edwards RB. Pain management and the values of health care providers. In: Hill CS, Fields WS, editors. Drug treatment of cancer pain in a drug oriented society: advances in pain research and therapy. Vol 11. New York: Raven Press, 1989: 101-12.

240. Wanzer SH, Federman DD, Adelstein SJ, Cassel CK, Cassem EH, Cranford RE, et al. The physician’s responsibility toward hopelessly ill patients-a second look. N Engl ] Med 1989;

241. Martin RS. Mortal values: healing, pain and suffering. In: Hill CS, Fields WS. Drug treatment of cancer pain in a drug oriented society: advances in pain research and therapy. Vol 11. New York Raven Press, 1989: 19-26.

242. Burke AL, Diamond PL, Hulbert J, Yeatman J, Farr EA. Terminal restlessness-its management and the role of midazolam. Med ]

1990: 1980-2039.

55:873-5.

1191-205.

1991; 84~332-7.

6:329-36.

120:844-9.

Aust 1991; 155:485-7.