Translate

Sunday, November 16, 2014

PALLIATIVE CARE

PHYSICAL CARE

Among patients with life-limiting diseases, physical symptoms have been the cause of the most distress [212]. Patients usually have multiple symptoms, and a mean of nine to 11 symptoms per patient has been reported [204,205,206,213]. The presence of multiple symptoms can create challenges in identifying causes, as many symptoms are intricately linked with others, including symptoms in the psychosocial domain.
Several tools have been developed to assess factors in the end-of-life experience, including five tools to evaluate physical symptoms (three of which are used to assess pain), four to assess quality of life, and six to assess functional status [214]. However, a systematic review of 99 tools in these three domains plus six others (emotional and cognitive symptoms, advance care planning, continuity of care, spirituality, grief and bereavement, satisfaction and quality of care, and caregiver well-being) showed that data on the reliability and validity were lacking for most of the tools [215]. Assessment of symptoms should include comprehensive documentation of the patient's history and findings on physical examination and should be carried out at regular intervals [5]. To help ensure that patients' physical distress is alleviated, when clinicians ask patients about the presence and severity of symptoms, they should also ask which symptom is most troublesome, as patients do not often specifically state this [216].
Although asking open-ended questions about symptoms is helpful, systematic assessment of symptoms is also necessary. A study of patients in a palliative medicine program demonstrated that significantly more symptoms were identified on systematic assessment than through open-ended questioning (2,075 symptoms compared with 325) [217]. The missed symptoms were not inconsequential; 69% of symptoms that patients rated as "severe" and 79% of the symptoms described as "distressing" were not volunteered by the patients [217]. Studies have demonstrated that patients are often reluctant to report worsening symptoms because of fear that they indicate progressive disease. Clinicians should describe potential symptoms to help patients and their family understand which symptoms can be expected and when it is appropriate to notify a member of the healthcare team. It is important for the healthcare team to acknowledge the patient's symptoms as real and to take prompt actions to relieve them adequately. The patient's comfort should take precedence over the exact cause of the symptom. Diagnostic studies to determine the cause of symptoms should be undertaken only if the results will substantially help in directing effective treatment. The risks, benefits, costs, and options for treating an underlying cause should be discussed with the patient and family and considered within the context of the patient's culture, belief system, and expectations.
An important distinction of palliative care is the focus on dimensions other than the physical. Symptoms are accompanied by the patient's thoughts and feelings, and as such, nonpharmacologic strategies should be used to address the sensory, cognitive, affective, and functional components [66]. The healthcare team should talk to the patient and family about priorities for pharmacologic versus nonpharmacologic treatments. Although data are limited on some nonpharmacologic interventions, many patients have benefited from these approaches. As research expands in the field of palliative care, other innovative strategies are being scientifically evaluated, and results are sometimes conflicting. Nonpharmacologic measures should be carried out in conjunction with pharmacologic management before medications fail to provide relief, as interventions, especially cognitive/behavioral techniques, are more effective when symptoms are acute and/or mild.
Once the patient's needs have been assessed, the palliative care team should work with the patient (and family) to develop priorities and goals of care [110]. Continual reassessment of symptoms and periodic review and documentation of the patient's goals and care plan are necessary to ensure that his or her needs are met [110]. It may be helpful for patients or a family member to keep a pain or symptom diary to note which measures have or have not provided relief and the duration of relief. This information will help clinicians determine the efficacy of specific therapeutic options and modify the treatment plan as necessary.
The discussion of interventions that follows focuses on the care of adults. Palliative care for children is addressed later in this course.

PAIN

Unrelieved pain is the greatest fear among people with a life-limiting disease, and the need for an increased understanding of effective pain management is well-documented [218]. Although experts have noted that 75% to 90% of end-of-life pain can be managed effectively, rates of pain are high, even among people receiving palliative care [46,123,183,204,206,213,218,219,220,221].

Issues in Effective Pain Management

The inadequate management of pain is the result of several factors related to both patients and clinicians. In a survey of oncologists, patient reluctance to take opioids or to report pain were two of the most important barriers to effective pain relief [222]. This reluctance is related to a variety of attitudes and beliefs [218,222]:
  • Fear of addiction to opioids
  • Worry that if pain is treated early, there will be no options for treatment of future pain
  • Anxiety about unpleasant side effects from pain medications
  • Fear that increasing pain means that the disease is getting worse
  • Desire to be a "good" patient
  • Concern about the high cost of medications
Education and open communication are the keys to overcoming these barriers. Every member of the healthcare team should reinforce accurate information about pain management with patients and families. The clinician should initiate conversations about pain management, especially regarding the use of opioids, as few patients will raise the issue themselves or even express their concerns unless they are specifically asked [223]. It is important to acknowledge patients' fears individually and provide information to help them differentiate fact from fiction. For example, when discussing opioids with a patient who fears addiction, the clinician should explain that the risk of addiction is low [218]. It is also helpful to note the difference between addiction and physical dependence.
There are several other ways clinicians can allay patients' fears about pain medication:
  • Assure patients that the availability of pain relievers cannot be exhausted; there will always be medications if pain becomes more severe.
  • Acknowledge that side effects may occur but emphasize that they can be managed promptly and safely and that some side effects will abate over time.
  • Explain that pain and severity of disease are not necessarily related.
Encouraging patients to be honest about pain and other symptoms is also vital. Clinicians should ensure that patients understand that pain is multidimensional and emphasize the importance of talking to a member of the healthcare team about possible causes of pain, such as emotional or spiritual distress. The healthcare team and patient should explore psychosocial and cultural factors that may affect self-reporting of pain, such as concern about the cost of medication.
Clinicians' attitudes, beliefs, and experiences also influence pain management, with addiction, tolerance, side effects, and regulations being the most important concerns [218,219,222,224,225,226]. A lack of appropriate education and training in the assessment and management of pain has been noted to be a substantial contributor to ineffective pain management [222,224,226,227]. As a result, many clinicians, especially primary care physicians, do not feel confident about their ability to manage pain in their patients [222,224].
Clinicians require a clear understanding of available medications to relieve pain, including appropriate dosing, safety profiles, and side effects. If necessary, clinicians should consult with pain specialists to develop an effective approach.

Legal and Ethical Issues Related to the Treatment of Pain

Fear of license suspension for inappropriate prescribing of controlled substances is also prevalent, and a better understanding of pain medication will enable physicians to prescribe accurately, alleviating concern about regulatory oversight. Physicians must balance a fine line; on one side, strict federal regulations regarding the prescription of schedule II opioids (morphine, oxycodone, methadone, hydromorphone) raise fear of Drug Enforcement Agency investigation, criminal charges, and civil lawsuits [218,228]. Careful documentation on the patient's medical record regarding the rationale for opioid treatment is essential [228]. On the other side, clinicians must adhere to the American Medical Association's Code of Ethics, which states that failure to treat pain is unethical. The code states, in part: "Physicians have an obligation to relieve pain and suffering and to promote the dignity and autonomy of dying patients in their care. This includes providing effective palliative treatment even though it may foreseeably hasten death" [229]. In addition, the American Medical Association Statement on End-of-Life Care states that patients should have "trustworthy assurances that physical and mental suffering will be carefully attended to and comfort measures intently secured" [230].
Physicians should consider the legal ramifications of inadequate pain management and understand the liability risks associated with both inadequate treatment and treatment in excess. The undertreatment of pain carries a risk of malpractice liability, and this risk is set to increase as the general population becomes better educated about the availability of effective approaches to pain management at the end of life. Establishing malpractice requires evidence of breach of duty and proof of injury and damages. Before the development of various guidelines for pain management, it was difficult to establish a breach of duty, as this principle is defined by nonadherence to the standard of care in a designated specialty. With such standards now in existence, expert medical testimony can be used to demonstrate that a practitioner did not meet established standards of care for pain management. Another change in the analysis of malpractice liability involves injury and damages. Because pain management can be considered as separate from disease treatment and because untreated pain can lead to long-term physical and emotional damage, claims can be made for pain and suffering alone, without wrongful death or some other harm to the patient [231].
The proper storage and disposal of prescription pain medications should also be considered. Taking steps to ensure that medications are stored and destroyed securely and safely can help prevent unintended overdose and substance abuse. In 2010, the U.S. House of Representatives passed the Safe Disposal Drug Act, which amended the Controlled Substances Act to permit the take-back disposal of medications by authorized persons (rather than the patient with the prescription) [486]. As such, healthcare professionals may be required to dispose of drugs returned by patients in addition to drug samples that have expired or are not being dispensed. For best practice guidelines on the disposal of medications by patients or healthcare professionals, please visit the American Academy of Pain Management at http://www.aapainmanage.org/literature/Articles/DisposalOfDrugs.pdf [487].

Patients with History of Substance Abuse

This population of people with a history of substance abuse presents challenges to the effective use of pain medication, with issues related to trust, the appropriate use of pain medications, interactions between illicit drugs and treatment, and compliance with treatment. The issues differ depending on whether substance abuse is a current or past behavior.
With active substance abusers, it is difficult to know if patients' self-reports of pain are valid or are drug-seeking behaviors. It has been recommended that, as with other patients at the end of life, self-reports of pain should be believed [66,223]. A multidisciplinary approach, involving psychiatric professionals, addiction specialists, and, perhaps, a pain specialist, is necessary. To decrease the potential for the patient to seek illicit drugs for pain, an appropriate pain management plan should be implemented and the patient should be reassured that pain can be managed effectively [66,223]. When planning treatment, the patient's tolerance must be considered; higher doses may be needed initially, and doses can be reduced once acute pain is under control. Long-acting pain medications are preferred for active substance abusers, and the use of nonopioids and coanalgesics can help minimize the use of opioids. Setting limits as well as realistic goals is essential and requires establishing trust and rapport with the patient and caregivers.
Establishing trust is also essential for patients with former substance abuse behavior, who often must be encouraged to adhere to a pain management program because of their fears of addiction. Involving the patient's drug counselor is beneficial, and other psychologic clinicians may be helpful in assuring the patient that pain can be relieved without addiction. Recurrence of addiction is low, especially among people with cancer, but monitoring for signs of renewed abuse should be ongoing [223].
Patients who are following a methadone maintenance program may also fear effective pain management as a risk for recurrent abuse. Two approaches may be followed for these patients: they may receive an increased dose of methadone as the pain reliever or they may be given other opioids along with the same methadone dose, with the dose of the opioid titrated for effective pain relief [66,223]. Again, involvement of the drug counselor is important.

Prevalence

The prevalence of pain at the end of life has been reported to range from 8% to 96%, occurring at higher rates among people with cancer than among adults with other life-limiting diseases [208,232].

Etiology

Pain can be caused by a multitude of factors and is usually multidimensional, with pain frequently being exacerbated by other physical symptoms and by psychosocial factors, such as anxiety or depression [219].

Assessment

As the fifth vital sign, pain should be assessed routinely, and frequent assessment has become the standard of care [219]. Pain is a subjective experience, and as such, the patient's self-report of pain is the most reliable indicator. Research has shown that pain is underestimated by healthcare professionals and overestimated by family members [219,233]. Therefore, it is essential to obtain a pain history directly from the patient, when possible, as a first step toward determining the cause of the pain and selecting appropriate treatment strategies. When the patient is unable to orally communicate, other strategies must be used to determine the characteristics of the pain, as will be discussed.
Questions should be asked to elicit descriptions of the pain characteristics, including its location, distribution, quality, temporal aspect, and intensity. In addition, the patient should be asked about aggravating or alleviating factors. Pain is often felt in more than one area, and physicians should attempt to discern if the pain is focal, multifocal, or generalized. Focal or multifocal pain usually indicates an underlying tissue injury or lesion, whereas generalized pain could be associated with damage to the central nervous system. Pain can also be referred, usually an indicator of visceral pain.
The quality of the pain refers to the sensation experienced by the patient, and it often suggests the pathophysiology of the pain [219]. Pain that is well localized and described as aching, throbbing, sharp, or pressurelike is most likely somatic nociceptive pain. This type of pain is usually related to damage to bones and soft tissues. Diffuse pain that is described as squeezing, cramping, or gnawing is usually visceral nociceptive pain. Pain that is described as burning, tingling, shooting, or shocklike is neuropathic pain, which is generally a result of a lesion affecting the nervous system.
Temporal aspects of pain refer to its onset: acute, chronic, or "breakthrough." A recent onset characterizes acute pain, and there are accompanying signs of generalized hyperactivity of the sympathetic nervous system (diaphoresis and increased blood pressure and heart rate). Acute pain usually has an identifiable, precipitating cause, and appropriate treatment with analgesic agents will relieve the pain. When acute pain develops over several days with increasing intensity, it is said to be subacute. Episodic, or intermittent, pain occurs during defined periods of time, on a regular or irregular basis. Chronic pain is defined as pain that persists for at least 3 months beyond the usual course of an acute illness or injury. Such pain is not accompanied by overt pain behaviors (grimacing, moaning) or evidence of sympathetic hyperactivity.
"Breakthrough" is the term used to describe transitory exacerbations of severe pain over a baseline of moderate pain [234]. Breakthrough pain can be incident pain or pain that is precipitated by a voluntary act (such as movement or coughing), or can occur without a precipitating event. Breakthrough pain occurs in as many as 90% of people with cancer or in hospice settings and is often a consequence of inadequate pain management [218].
Documentation of pain intensity is key, as several treatment decisions depend on the intensity of the pain. For example, severe, intense pain requires urgent relief, which affects the choice of drug and the route of administration [3,219]. The numeric rating scale is the tool used most often to assess pain; with this tool, patients rate pain on a scale of 0 to 10 [219]. Visual analogue scales (patients rate pain on a line from 0 to 10) and verbal rating scales, which enable the patient to describe the pain as "mild," "moderate," or "severe," have also been found to be effective. Some patients, however, may have difficulty rating pain using even the simple scales. In an unpublished study involving 11 adults with cancer, the Wong-Baker FACES scale, developed for use in the pediatric setting, was found to be the easiest to use among three pain assessment tools that include faces to assess pain [235].
Functional assessment is important. The healthcare team should observe the patient to see how pain limits movements and should ask the patient or family how the pain interferes with normal activities. Determining functional limitations can help enhance patient compliance in reporting pain and adhering to pain-relieving measures, as clinicians can discuss compliance in terms of achieving established functional goals [223]. The Memorial Pain Assessment Card can be used to evaluate both the severity of pain and the effect of pain on function [219,236].
Physical examination can be valuable in determining an underlying cause of pain. Examination of painful areas can detect evidence of trauma, skin breakdown, or changes in osseous structures. Auscultation can detect abnormal breath or bowel sounds; percussion can detect fluid accumulation; and palpation can reveal tenderness. A neurologic examination should also be carried out to evaluate sensory and/or motor loss and changes in reflexes. During the examination, the clinician should watch closely for nonverbal cues that suggest pain, such as moaning, grimacing, and protective movements. These cues are especially important when examining patients who are unable to verbally communicate about pain.

Management

Strong evidence supports pain management approaches for people with cancer, but the evidence base for management of pain in people with other life-limiting diseases is weak [46,105,183,209,211,237]. Effective pain management involves a multidimensional approach involving pharmacologic and nonpharmacologic interventions that are individualized to the patient's specific situation [219].

Pharmacologic Interventions

The pharmacologic management of pain is best achieved with use of the WHO three-step analgesic ladder, which designates the type of analgesic agent based on the severity of pain (Figure 6) [238]. Step 1 of the WHO ladder involves the use of nonopioid analgesics, with or without an adjuvant (coanalgesic) agent, for mild pain (pain that is rated 1 to 3 on a 10-point scale). Step 2 treatment, recommended for moderate pain (score of 4 to 6), calls for a weak opioid, which may be used in combination with a step 1 nonopioid analgesic for unrelieved pain. Step 3 treatment is reserved for severe pain (score of 7 to 10) or pain that persists after Step 2 treatment. Strong opioids are the optimum choice of drug at Step 3. At any step, nonopioids and/or adjuvant drugs may be helpful.
THE WORLD HEALTH ORGANIZATION'S THREE-STEP LADDER OF ANALGESIA
The WHO ladder is also accompanied by five principles: prescribe analgesics according to the severity of pain (regardless of whether treatment at a previous step was carried out), use oral formulations (preferably), give analgesics at regular intervals around the clock (not on an as-needed basis), tailor the dose to the individual, and prescribe analgesics with attention to detail (document a treatment program) [238]. The pharmacologic treatment of pain involves the selecting the right drug(s) at the right dose, frequency, and route, and managing side effects [219]. A decision pathway was developed for use in the cancer setting and can be applied to other settings (Figure 7) [219].
DECISION PATHWAY FOR PAIN MANAGEMENT
Nonopioid analgesics, such as aspirin, acetaminophen (Tylenol), and nonsteroidal anti-inflammatory drugs (NSAIDs), are primarily used for mild pain (Step 1 of the WHO ladder) and may also be helpful as coanalgesics at Steps 2 and 3. Acetaminophen is among the safest of analgesic agents, but it has essentially no anti-inflammatory effect. Toxicity is a concern at high doses, and the maximum recommended dose is 3–4 g per day [219]. Acetaminophen should be avoided or given at lower doses in people with a history of alcohol abuse or renal or hepatic insufficiency [219].
NSAIDs are most effective for pain associated with inflammation. Among the commonly used NSAIDs are ibuprofen (Motrin, Advil), naproxen (Aleve, Naprosyn), and indomethacin (Indocin). There are several classes of NSAIDs, and the response differs among patients; trials of drugs for an individual patient may be necessary to determine which drug is most effective [66]. NSAIDs inhibit platelet aggregation, increasing the risk of bleeding, and also can damage the mucosal lining of the stomach, leading to gastrointestinal bleeding. There is a ceiling effect to the nonopioid analgesics; that is, there is a dose beyond which there is no further analgesic effect. In addition, many side effects of nonopioids can be severe and may limit their use or dosing.
Moderate pain (Step 2) has often been treated with analgesic agents that are combinations of acetaminophen and an opioid, such as codeine, oxycodone, or hydrocodone. However, it is now recommended that these combination drugs be avoided, as limits on the maximum dose of acetaminophen limits the use of a combination drug [202,219]. Individual drugs in combination is preferred, allowing for increases in the dose of the opioid without increasing the dose of the coanalgesic.
Strong opioids are used for severe pain (Step 3) [105,183,211,219]. Morphine, buprenorphine, oxycodone, hydromorphone, fentanyl, and methadone are the most widely used Step 3 opioids in the United States [239]. Unlike nonopioids, opioids do not have a ceiling effect, and the dose can be titrated until pain is relieved or side effects become unmanageable. For an opioid-naïve patient or a patient who has been receiving low doses of a weak opioid, the initial dose of a Step 3 opioid should be low, and, if pain persists, the dose may be titrated up daily until pain is controlled. Typical starting doses for patients who are opioid-naïve have been noted, but these doses should be used only as a guide, and the initial dose, as well as titrated dosing, should be done on an individual basis (Table 8). Guidelines suggest that the most appropriate dose is the one that relieves the patient's pain throughout the dosing interval without causing unmanageable side effects [183,202,243].
OPIOIDS FOR THE MANAGEMENT OF PAIN IN ADULTS*
DrugTypical Starting Dose†Onset of ActionDuration of Action
Codeine15–60 mg30 to 60 min4 to 6 hrs
Hydrocodone2.5–10 mg10 to 20 min4 to 8 hrs
Morphine, immediate release15–30 mg 15 to 30 min
5 to 10 min
3 to 6 hrs
Oxycodone, immediate release5–10 mg10 to 30 min3 to 4 hrs
Oxymorphone, sustained release10 mg5 to 10 min8 to 12 hrs
Hydromorphone2–4 mg15 to 30 min4 to 5 hrs
Methadone5–10 mg30 to 60 min4 to 6 hrs
Tapentadol50–100 mg<60 min4 to 6 hrs
Tapentadol, extended release50–100 mg
Fentanyl (buccal tablet)100–200 mcg5 to 15 min2 to 4 hrs
Fentanyl (transdermal patch)25 mcg/hr (worn for 3 days)12 to 18 hrs48 to 72 hrs
Buprenorphine (transdermal patch)5–10 mcg/hr (worn for 7 days)
*All information is given for oral formulations unless otherwise specified.
†Doses given are guidelines for opioid-naïve patients; actual doses should be determined on an individual basis.
More than one route of opioid administration will be needed by many patients during end-of-life care, but in general, opioids should be given orally, as this route is the most convenient and least expensive. The transdermal route is preferred to the parenteral route, although dosing with a transdermal patch is less flexible and so may not be appropriate for patients with unstable pain [219]. Intramuscular injections should be avoided because injections are painful, drug absorption is unreliable, and the time to peak concentration is long [219].
Morphine is considered to be the first-line treatment for a Step 3 opioid [202]. Morphine is available in both immediate-release and sustained-release forms, and the latter form can enhance patient compliance. The sustained-release tablets should not be cut, crushed, or chewed, as this counteracts the sustained-release properties. Morphine should be avoided in patients with severe renal failure [211].
Buprenorphine (Butrans) has the general structure of morphine but differs from it in several ways [239]. The transdermal formulation of the drug was approved in 2010 for moderate-to-severe chronic pain in patients requiring an around-the-clock opioid for an extended period [219]. It may be used for people with renal impairment but is contraindicated in patients who have substantial respiratory depression [239,240].
The sustained-release form of oxycodone (OxyContin) has been shown to be as safe and effective as morphine for cancer-related pain, and it may be associated with less common side effects, especially hallucinations and delirium [244]. Oxycodone is also available in an immediate-release form (Roxicodone). Oxycodone should be used in people with advanced chronic kidney disease only if alternative options are not available [211]. If the drug must be used, the intervals between doses should be increased, and the patient should be monitored closely [211].
Hydromorphone and fentanyl are the most potent opioids; neither drug should be given to an opioid-naïve patient. Hydromorphone (Dilaudid), which is four times as potent as morphine, is available in only an immediate-release form. Fentanyl is the strongest opioid (approximately 80 times the potency of morphine) and is available as a transdermal drug-delivery system (Duragesic), and a buccal tablet (Onsolis) was approved in 2009 [240,245]. Both the transdermal patch and buccal tablet have a more rapid onset than other opioids given nonparenterally [219]. Because of its potency, fentanyl must be used with extreme care, as deaths have been associated with its use. Physicians must emphasize to patients and their families the importance of following prescribing information closely, and members of the healthcare team should monitor the use of the drug. Fentanyl, administered subcutaneously, is the recommended choice for patients with advanced chronic kidney disease [211].
The use of methadone (Dolophine) to relieve pain has increased substantially over the past few years, moving from a second-line or third-line drug to a first-line medication for severe pain in people with life-limiting diseases [246]. A systematic review showed that methadone had efficacy similar to that of morphine [247]. Physicians must be well educated about the pharmacologic properties of methadone, as the risk for serious adverse events, including death, is high when the drug is not administered appropriately [247,248]. If the dose of methadone is increased too rapidly or administered too frequently, toxic accumulation of the drug can cause respiratory depression and death. Extreme care must be taken when titrating the drug, and close evaluation of the patient is necessary.
Propoxyphene (Darvon) is an opioid that is chemically similar to methadone. It is not recommended for use because of toxicity even at therapeutic doses and a lack of efficacy compared with placebo or acetaminophen [66,223]. Similarly, meperidine (Demerol) should not be used in the palliative care setting because of limited efficacy and potential for severe toxicity. Agonist-antagonist opioids (nalbuphine [Nubain], butorphanol [Stadol], and pentazocine [Talwin]) are not recommended for use with pure opioids, as they compete with them, leading to possible withdrawal symptoms.
Tapentadol (Nucynta) is a short-acting opioid approved for moderate to severe pain in adults; an extended release formulation (Nucynta ER) was approved in 2011 for moderate-to-severe chronic pain when an around-the-clock opioid is needed [240]. The drug is associated with a lower incidence of adverse effects than other opioids, and it has been shown to be highly effective for chronic pain conditions but has not been studied in cancer-related pain or the palliative care setting [249].
The most appropriate option for breakthrough pain is an immediate-release opioid taken in addition to the around-the-clock regimen [219]. The fentanyl buccal tablet has been shown to be effective and safe for relieving breakthrough pain in people who are opioid tolerant [183,250,251]. Between January 2011 and January 2012, three forms of fentanyl were approved for breakthrough pain in people with cancer: fentanyl sublingual tablet (Abstral), fentanyl nasal spray (Lazanda), and fentanyl sublingual spray (Subsys) [240]. For each formula, the initial dose may be repeated once if pain is not relieved adequately after 30 minutes. Patients must wait at least 2 hours before using the sublingual tablet or the nasal spray for another breakthrough pain episode; the interval is 4 hours for the sublingual spray [240].
When pain responds poorly to escalated doses of an opioid, other approaches should be considered, including alternative routes of administration, use of alternate opioids (termed opioid rotation or opioid switching), use of adjuvant analgesics, and nonpharmacologic approaches. A process for opioid switching has been established (Figure 8); the first step is to calculate the equianalgesic dose of the new drug (Table 9) [183,202,219]. Additional care is needed when switching to methadone, and conversion ratios have been established (Table 10) [183]. Evidence suggests that the traditionally recommended equianalgesic doses for the fentanyl transdermal patch are subtherapeutic for patients with chronic cancer-related pain, and more aggressive approaches may be warranted (Table 11) [183,219,252].
PROCESS FOR OPIOID SWITCHING
*Reduce by a greater percentage if the patient is older, frail, or has significant organ dysfunction. If changing to methadone, reduce the dose by 75%. If changing to transdermal fentanyl, do not reduce the dose and continue the current opioid for 12 to 48 hours.
OPIOID EQUIVALENT DOSES
DrugOral DoseParenteral Dose
Morphine30 mg10 mg
Codeine200 mgNA
Hydromorphone7.5 mg1.5 mg
Hydrocodone30–45 mgNA
Oxycodone20 mgNA
Oxymorphone10 mg1 mg
Methadone20 mg10 mg
Buprenorphine transdermal patch5–0 mcg/hrNA
DOSE CONVERSION RATIOS FOR METHADONE
Oral MorphineConversion Ratio (Morphine:Oral Methadone)
30–90 mg4:1
91–300 mg8:1
>300 mg12:1
100 mcg/hr800 mg
EQUIANALGESIC ORAL OPIOID DOSES FOR FENTANYL TRANSDERMAL PATCH
Transdermal FentanylMorphineHydromorphoneOxycodoneCodeine
25 mcg/hr60 mg7.5 mg30 mg200 mg
50 mcg/hr120 mg15 mg60 mg400 mg
75 mcg/hr180 mg22.5 mg90 mg600 mg
100 mcg/hr240 mg30 mg120 mg800 mg
Another approach that has been used for pain management in the cancer setting is combination opioid therapy, or the concurrent use of two strong opioids. The effectiveness of this approach has been evaluated in only two studies, and the combination was morphine and oxycodone or morphine with fentanyl or methadone [253]. The evidence to support a recommendation of combination opioid therapy is weak, and the side effects most likely outweigh the benefit [253].
Opioids are associated with many side effects, the most notable of which is constipation, occurring in nearly 100% of patients. The universality of this side effect mandates that once extended treatment with an opioid begins, prophylactic treatment with laxatives must also be initiated. Tolerance to other side effects, such as nausea and sedation, usually develops within 3 to 7 days. Some patients may state that they are "allergic" to an opioid. It is important for the physician to explore what the patient experienced when the drug was taken in the past, as many patients misinterpret side effects as an allergy. True allergy to an opioid is rare [219]. Opioid rotation may also be done to reduce adverse events.
When opioids are prescribed, careful documentation of the patient's history, examinations, treatments, progress, and plan of care are especially important from a legal perspective. This documentation must provide evidence that the patient is functionally better off with the medication than without [66]. In addition, physicians must note evidence of any dysfunction or abuse.
Adjuvant agents are often used in conjunction with opioids and are usually considered after the use of opioids has been optimized [66]. The primary indication for these drugs is adjunctive because they can provide relief in specific situations, especially neuropathic pain. Examples of adjuvant drugs are tricyclic antidepressants, anticonvulsants, muscle relaxants, and corticosteroids (Table 12) [183,219]. A systematic review found that there was limited evidence to support the use of selective serotonin reuptake inhibitors (SSRIs) for neuropathic pain, but one serotonin-norepinephrine reuptake inhibitor, venlafaxine (Effexor), was found to be effective [254].
ORAL ADJUVANT ANALGESICS
Drug ClassDrugTypical Starting DoseUsual Effective Dose
AnticonvulsantsGabapentin100–300 mg once daily300–1200 mg (2 or 3 divided doses)
Pregabalin25–75 mg twice daily75–200 mg (3 divided doses)
Carbamazepine100 mg twice daily300–800 mg twice daily
Topiramate25–50 mg daily50–200 mg twice daily
Oxcarbazepine150–300 mg twice daily150–600 mg twice daily
Tiagabine4 mg at bedtime4–12 mg twice daily
Tricyclic antidepressantsAmitriptyline Nortriptyline Desipramine10–25 mg at bedtime50–150 mg at bedtime
Serotonin-norepinephrine reuptake inhibitorsVenlafaxine37.5 mg daily150–350 mg daily
Skeletal muscle relaxantsBaclofen5 mg twice daily10–20 mg 2 or 3 times daily
Cyclobenzaprine5 mg 3 times daily10–20 mg 3 times daily
Metaxalone400 mg 3 times dailyNot defined
CorticosteroidsDexamethasone1–2 mgNot defined

Nonpharmacologic Interventions

Several nonpharmacologic approaches are therapeutic complements to pain-relieving medication, lessening the need for higher doses and perhaps minimizing side effects. These interventions can help decrease pain or distress that may be contributing to the pain sensation. Approaches include palliative radiotherapy, complementary/alternative methods, manipulative and body-based methods, and cognitive/behavioral techniques. The choice of a specific nonpharmacologic intervention is based on the patient's preference, which, in turn, is usually based on a successful experience in the past.
Palliative radiotherapy is effective for managing cancer-related pain, especially bone metastases [46,255,256]. Bone metastases are the most frequent cause of cancer-related pain; 50% to 75% of patients with bone metastases will have pain and impaired mobility [255]. External-beam radiotherapy is the mainstay of treatment for pain related to bone metastases. At least some response occurs in 70% to 80% of patients, and the median duration of pain relief has been reported to be 11 to 24 weeks [255]. It takes 1 to 4 weeks for optimal therapeutic results [255,256].
However, palliative radiotherapy has become a controversial issue. Although the benefits of palliative radiotherapy are well documented and most hospice and oncology professionals believe that palliative radiotherapy is important, this treatment approach is offered at approximately 24% of Medicare-certified freestanding hospices, with less than 3% of hospice patients being treated [74,75,76]. As previously noted, reimbursement issues present a primary barrier to the use of palliative radiotherapy [74,75,76]. Among other barriers are short life expectancy, transportation issues, patient inconvenience, and lack of knowledge about the benefits of palliative radiotherapy in the primary care community [74,75,256,257].
More than half (54%) of people use complementary/alternative medicine therapies at the end of life [258]. The most commonly used therapies are massage, relaxation techniques, and acupuncture [258,259,260].
Massage, which can be broadly defined as stroking, compression, or percussion, has led to significant and immediate improvement in pain in the hospice setting [261]. Both massage and vibration are primarily effective for muscle spasms related to tension or nerve injury, and massage can be carried out with simultaneous application of heat or cold. Massage may be harmful for patients with coagulation abnormalities or thrombophlebitis [223].
Focused relaxation and breathing can help decrease pain by easing muscle tension. Progressive muscle relaxation, in which patients follow a sequence of tensing and relaxing muscle groups, has enabled patients to feel more in control and to experience less pain and can also help provide distraction from pain. [223]. This technique should be avoided if the muscle tensing will be too painful.
Acupuncture typically provides pain relief 15 to 40 minutes after stimulation. Relief seems to be related to the release of endorphins and a susceptibility to hypnosis [223]. The efficacy of acupuncture for relieving pain has not been proven, as study samples have been small. However, acupuncture has been found to be of some benefit for cancer-related pain when the therapy is given in conjunction with analgesic therapy [262].
Other nonpharmacologic interventions that have been helpful for some patients but lack a strong evidence base include manipulative and body-based methods (such as application of cold or heat, and positioning), yoga, distraction, and music or art therapy. The application of cold and heat are particularly useful for localized pain and have been found to be effective for cancer-related pain caused by bone metastases or nerve involvement, as well as for prevention of breakthrough incident pain [223]. Alternating application of heat and cold can be soothing for some patients, and it is often combined with other nonpharmacologic interventions.
Cold can be applied through wraps, gel packs, ice bags, and menthol. It provides relief for pain related to skeletal muscle spasms induced by nerve injury and inflamed joints. Cold application should not be used for patients with peripheral vascular disease. Heat can be applied as dry (heating pad) or moist (hot wrap, tub of water) and should be applied for no more than 20 minutes at a time, to avoid burning the skin. Heat should not be applied to areas of decreased sensation or with inadequate vascular supply, or for patients with bleeding disorders.
Changing the patient's position in the bed or chair may help relieve pain and also helps minimize complications such as decubitus ulcers, contractures, and frozen joints. Members of the healthcare team as well as family members and other informal caregivers can help reposition the patient for comfort and also perform range-of-motion exercises. Physical and occupational therapists can recommend materials, such as cushions, pillows, mattresses, splints, or support devices.
Hatha yoga is the branch of yoga most often used in the medical context, and it has been shown to provide pain relief for patients who have osteoarthritis and carpal tunnel syndrome but it has not been studied in patients at the end of life. Yoga may help relieve pain indirectly in some patients through its effects on reducing anxiety, increasing strength and flexibility, and enhancing breathing [263]. Yoga also helps patients feel a sense of control.
Methods to provide distraction from pain come in a wide variety of methods, including reciting poetry, meditating with a calm phrase, watching television or movies, playing cards, visiting with friends, or participating in crafts.
Music therapy and art therapy are also becoming more widely used as nonpharmacologic options for pain management. Listening to music has been shown to decrease the intensity of pain and reduce the amount of opioids needed, but the magnitude of the benefit was small [264]. Research suggests that art therapy contributes to a patient's sense of well-being [265]. Creating art helps patients and families to explore thoughts and fears during the end of life. An art therapist can help the creators reflect on the implications of the art work. Art therapy is especially helpful for patients who have difficulty expressing feelings with words, for physical or emotional reasons.

FATIGUE

Fatigue is a subjective feeling of tiredness, weariness, and lack of energy. Fatigue is often accompanied by a feeling of weakness (asthenia), which can be either generalized or localized. The National Comprehensive Cancer Network (NCCN) defines fatigue as "physical, emotional, and/or cognitive tiredness or exhaustion" [183]. Fatigue associated with life-limiting diseases is further defined by its disproportionate relation to recent activity and the lack of recovery with additional sleep [266]. Persistent fatigue has a significant impact on the quality of life by negatively affecting functional status, interfering with normal activities, and contributing to emotional distress [183]. Fatigue may also cause distress for a patient's family members, who may interpret this symptom as a sign of the patient "giving up." As is the case with pain, fatigue is underreported, underdiagnosed, and undertreated [266]. Studies have indicated that approximately half of patients do not report fatigue to their healthcare team, and the primary reasons were that they did not think effective treatments were available and their physicians did not offer interventions [267].
Fatigue is often part of a cluster of symptoms that may also include pain, depression, sleep disturbances, and anxiety/depression, especially at the end of life [183,268,269,270]. Analysis of 25 symptoms among 922 patients with advanced cancer demonstrated seven clusters. One of those clusters, referred to as the fatigue/anorexia-cachexia cluster, was composed of easy fatigue, weakness, lack of energy, anorexia, early satiety, weight loss, dry mouth, and taste changes [271]. Fatigue has often been reported to be the symptom that causes patients the most distress [272].

Prevalence

A sense of fatigue and weakness is one of the most common symptoms near the end of life, and patients often consider this symptom to be more troublesome than pain [273,274]. The prevalence of fatigue has been reported to range from 12% to 97% of patients with life-limiting diseases, and the prevalence is fairly consistent across disease settings [208,266].

Etiology

Among the most common contributors to fatigue in people with advanced life-limiting diseases are medications, anemia, dehydration, direct tumor effects on energy consumption and supply, infection, metabolic disturbances, fluid and electrolyte imbalance, dyspnea, sleep apnea, depression, and loss of skeletal muscle due to cachexia [105,183,266].

Prevention

Ensuring adequate management of symptoms related to fatigue may help in preventing the condition. Clinicians should advise the patient to conserve energy as much as possible, to follow a normal sleep cycle, and to engage in aerobic exercise [105,183,266,272].

Assessment

Assessing fatigue can be a challenge, but as with pain, the patient's report of how he or she is feeling is the gold standard in the assessment. For patients who speak a language other than English, questions about fatigue should include such words and phrases as "tired," "weak," and "lack of energy," as the word "fatigue" may translate differently in some languages [266]. Several tools are available to assess fatigue, but because it usually occurs in a cluster of symptoms, many of these tools are multidimensional instruments, often involving several questions, which can be impractical [266]. In assessing patients for fatigue, the clinician should ask such questions as "Do you feel unusually tired or weak?" or "How tired/weak are you?" [266].
An easy-to-use instrument is the Brief Fatigue Inventory, which includes four items that ask the patient to rate the severity of fatigue on a scale of 0 (no fatigue) to 10 ("as bad as you can imagine") [275]. The patient is asked to consider the current level of fatigue as well as fatigue experienced within the past 24 hours and to indicate the degree to which fatigue has interfered with activities, mood, relations with other people, and enjoyment of life.
Assessment should also include a physical examination to detect an underlying cause of fatigue, a focused history-taking, and laboratory tests, as appropriate, to rule out suspected causes (Figure 9) [266].
ALGORITHM FOR THE DIAGNOSIS OF FATIGUE IN PALLIATIVE CARE PATIENTS
ACTH = adrenocorticotropic hormone, Ca = calcium, CRP = C-reactive protein, Mg = magnesium, NRS = numerical rating scale, Phos = phosphate, TSH = thyroid-stimulating hormone.

Management

Little evidence is available to support guidelines for the management of fatigue during the end of life. Most of the research on nonpharmacologic and pharmacologic treatment options has been conducted with subjects receiving active cancer treatment or long-term follow-up care after cancer treatment. Fatigue in the palliative care setting is addressed specifically by the European Association for Palliative Care (EAPC) (all settings) and the NCCN (cancer setting) and is noted in guidelines for palliative care for advanced heart failure [105,183,266]. In addition, the AHRQ has addressed fatigue in the cancer setting, and systematic reviews have been done to help determine effective pharmacologic and nonpharmacologic interventions [268,276,277,278,279]. Management of fatigue should include treatment of an underlying cause, if one can be identified, but symptomatic relief should also be provided (Figure 10) [105,183,266].
ALGORITHM FOR THE MANAGEMENT OF FATIGUE IN PALLIATIVE CARE PATIENTS
When medications are the underlying cause of the fatigue, nonessential medications should be discontinued, and changing medications or the time of dosing may reduce tiredness during the day. Appropriate management of infection, cachexia, depression, and insomnia may also help reduce fatigue [266,273]. The patient's life expectancy and preferences should be considered before carrying out treatment of an underlying cause [266]. Fatigue may provide a protective effect for patients in the last days or hours of life [266]. As such, the patient may be more comfortable without aggressive treatment of fatigue during that period [266].
The treatment of anemia as an underlying cause of fatigue (and other symptoms) is a complex issue. Many studies have provided evidence to recommend the use of erythropoiesis-stimulating agents (erythropoietin [Epogen], darbepoetin [Procrit]) for anemia in people with cancer, HIV/AIDS, chronic kidney disease, and heart failure because of benefit in increasing the hemoglobin level, improving exercise tolerance, reducing symptoms, and decreasing the need for blood transfusions [266; 276; 280; 281; 282]. However, safety concerns led the U.S. Food and Drug Administration (FDA) to require a boxed warning on the label of erythropoiesis-stimulating agents regarding the increased risk of several adverse events (death, myocardial infarction, stroke, venous thromboembolism, thrombosis of vascular access, and tumor progression or recurrence) among people with chronic kidney disease or cancer [240]. The FDA recommends using the lowest dose sufficient to avoid red blood cell transfusion [240]. Recommendations for these agents in these populations have been withdrawn or revised [277; 283; 284]. A 2010 systematic review and meta-analysis (11 studies, 794 subjects) demonstrated benefit of erythropoiesis-stimulating agents among people with heart failure and mild anemia (>10 g/dL) with no increase in adverse events [282].
Most patients will try to manage fatigue by resting and/or sleeping more often, and many healthcare professionals will also recommend this strategy. However, additional rest and/or sleep usually does not restore energy in patients who have fatigue related to a life-limiting disease; continued lack of exercise may even promote fatigue [266]. Aerobic exercise has been found to alleviate fatigue, although much of the research in this area has been conducted with cancer survivors. For example, a meta-analysis (28 studies, 2,083 subjects) demonstrated a significant effect of exercise in the treatment of fatigue during and after cancer treatment [279]. Some small studies of fatigue have been done in the palliative care setting, and exercise was found to be beneficial [285,286,287].
Although an exercise program is recommended, decreasing activity to conserve energy is also encouraged [105,183,266]. Clinicians should talk to the patient and family about the importance of the patient conserving energy by adjusting daily activities to correspond to times of peak energy, setting priorities for activities, following a normal wake-sleep cycle, and using assistive devices, and delegating less important tasks [183,266]. Encouraging adequate nutrition, stress reduction through meditation or relaxation techniques, and engagement in enjoyable activities can help restore energy [183,266]. Counseling about setting realistic goals for activities and function may also help patients and family members adapt to new daily routines.
Pharmacologic treatment of fatigue should be undertaken only after potential causes of fatigue have been ruled out [183,266]. The EAPC notes that methylphenidate (Ritalin) and modafinil (Provigil) may reduce fatigue, and the NCCN recommends these two drugs as first-line options in the cancer palliative care setting [183,266]. These recommendations are based on systematic reviews showing a significant effect of methylphenidate for the treatment of fatigue in people with cancer or HIV/AIDS or for opioid-induced sedation [266,276,277,278,288]. An optimal dose of methylphenidate has not been defined, but an initial dose of 5–10 mg (given in the morning) has been used, with the dose titrated to 40–60 mg per day (given once in the morning and once at midday) [266]. Among the side effects are nervousness, jitteriness, agitation, arrhythmia, and tachycardia [266]. The initial recommended dose of modafinil is 200 mg per day [266]. Major side effects have included agitation, nervousness, sleep disturbances, nausea, and diarrhea. Since the publication of these recommendations, researchers conducting a systematic review concluded that the evidence was insufficient to recommend a specific drug for the treatment of fatigue in the palliative care setting [278].
Corticosteroids (prednisone and dexamethasone) have been used frequently to treat fatigue in the palliative care setting, but no research on their effectiveness is available [278]. These agents have provided short-term relief of fatigue and improved quality of life among people with cancer, but because of the toxicity associated with their long-term use, they should be considered only at the end life or to alleviate fatigue for a well-defined goal (such as allowing the patient to attend a special event) [183,266].

DYSPNEA

Dyspnea is a subjective sense of breathlessness (shortness of breath) and ranges from experiencing breathlessness on exertion to severe shortness of breath for longer periods of time. Patients may describe dyspnea as "smothering," "suffocating," or "drowning." Dyspnea can have a substantial impact on a patient's quality of life by restricting the patient's activities as well as causing distress for both patients and their families.

Prevalence

The prevalence of dyspnea among adults with life-limiting disease ranges from 10% to 95%, with the highest rates among people with COPD, lung cancer, and heart failure, especially in the last week of life [204,206,208,210,237,289,290].

Etiology

Pain and psychologic conditions such as anxiety and depression play a role in subjective symptoms of dyspnea [210,237]. Physical causes of dyspnea vary according to the life-limiting disease and/or comorbid conditions and include pleural effusion, airway obstruction, pulmonary embolism, pericardial effusion, and asthma [237].

Prevention

Measures to reduce anxiety can help to prevent dyspnea or reduce its severity. In addition, patients with heart failure or lung diseases should be advised to conserve energy.

Assessment

Practice guidelines recommend that clinicians regularly assess dyspnea in patients receiving end-of-life care [46,210,237]. Assessment should involve asking the patient to note the severity and/or distress related to dyspnea, as objective testing, such as respiratory rate, arterial blood gas levels, and pulse oximetry, do not always correlate with a patient's experience of shortness of breath [210]. Tools for patient-reported dyspnea include a modified Borg scale, a visual analog scale, or a numerical scale [210,291]. In addition to asking about the severity of breathlessness, the clinician should ask about other symptoms, especially concurrent chest pain, and about the activities that cause dyspnea. Patients with dyspnea often modify their activities to avoid dyspnea, so the clinician should ask the patient if he or she has changed or stopped any activities because of dyspnea [237]. Because of the link between psychologic factors and dyspnea, the clinician should also evaluate the patient's psychosocial status.
Physical assessment of the patient should include evaluation of breath sounds, heart rate, respiratory rate, jugular pressure, and functional status. Testing should be done to identify a suspected underlying cause of dyspnea [119,237].

Management

The American College of Physicians, the American College of Chest Physicians, the American Thoracic Society, and the NCCN have developed evidence-based guidelines for the management of dyspnea [46,119,183,210,237]. In addition, evidence-based recommendations for managing dyspnea in people with advanced heart failure are available [105]. A stepwise approach to managing dyspnea should be taken, with the first step being treatment of the underlying cause, if one can be identified [237]. Nonpharmacologic interventions should be used first; if the response is inadequate, pharmacologic interventions may be added.
Supplemental oxygen is commonly used to treat dyspnea. The ACCP notes that strong evidence supports the use of oxygen and pulmonary rehabilitation for dyspnea, and supplemental oxygen may provide relief of dyspnea for people with advanced lung or heart disease who have hypoxemia at rest or with minimal activity [46,183,209,210,237]. However, data suggest that oxygen offers no benefit to patients who do not have hypoxemia [105].
A variety of nonpharmacologic interventions have been suggested in several practice guidelines, although the evidence base varies (Table 13) [119,183,210,237,292]. In a systematic review of nonpharmacologic interventions for dyspnea in people with advanced malignant and nonmalignant diseases, there was strong evidence for chest wall vibration and neuroelectrical muscle stimulation and moderate evidence for walking aids and breathing training [293]. The data were insufficient to recommend the use of a fan, music, relaxation, counseling and support, and psychotherapy [293]. A subsequent small randomized controlled trial demonstrated that a handheld fan directed at the face reduced breathlessness [292].
NONPHARMACOLOGIC INTERVENTIONS FOR DYSPNEA RECOMMENDED IN PRACTICE GUIDELINES
Chest wall vibration
Neuroelectrical muscle stimulation
Walking aids
Breathing training
Handheld fan directed at the face
Pursed-lip breathing
Cool compress on the forehead
Cool room
Open windows
Activity pacing
Noninvasive positive pressure ventilation
Relaxation techniques
Psychosocial support
Patient and family education
Opioids represent the primary recommended pharmacologic intervention for severe dyspnea in people with advanced cancer and lung disease [46,210,237]. A systematic review and meta-analysis (18 randomized controlled trials) demonstrated a significant positive effect of opioids on breathlessness [294]. Guidelines recommend that oral or parenteral opioids be considered for all patients with severe and unrelieved dyspnea; nebulized opioids have not had an effect when compared with placebo [46,209,210,237]. Oral morphine is the most commonly prescribed opioid, but other opioids, such as diamorphine, dihydrocodeine, fentanyl, hydromorphone, and oxycodone, may be used [210]. The dose should be selected and titrated according to such factors as renal, hepatic, and pulmonary function and past use of opioids [210]. An oral dose of morphine of 2.5–10 mg every 4 hours as needed (1–5 mg intravenously) has been recommended for opioid-naïve patients [119,183]. Although respiratory depression is a side effect associated with opioids, especially morphine, this effect has not been found with doses used to relieve dyspnea [119,295]. Evidence-based recommendations for palliative care for people with heart failure note that diuretics represent the cornerstone of treatment of dyspnea [105]. Nitrates may also provide relief, and inotropes may be appropriate in select patients [105]. The recommendations also include the use of low-dose opioids [105].
Anxiolytics are often a recommended option for relief of breathlessness because of the association between anxiety and dyspnea. However, anxiolytic agents have not been found to be effective for the management of dyspnea. A systematic review published in 2010 (seven studies, 200 subjects) showed that benzodiazepines had no beneficial effect on breathlessness in people with advanced cancer or COPD [296]. Bronchodilators and systemic corticosteroids may be helpful in relieving dyspnea in people with lung cancer and underlying obstructive airway disease [237]. In addition, analgesics may help relieve dyspnea associated with pain [237].

CONSTIPATION

Constipation can be defined as a reduced frequency of bowel movements and an increased stool consistency. In defining constipation in people with life-limiting disease, measurable symptoms, as well as the person's perception of constipation and the level of discomfort, are factors [297,298]. The condition may be accompanied by cramps and a bloating feeling as well as discomfort while defecating, due to straining and rectal pressure. Constipation should be fundamentally defined by the patient [297].

Prevalence

The prevalence of constipation among adults with life-limiting disease ranges from 8% to 70%, and constipation occurs in almost all patients taking opioids [208,223,290].

Etiology

Opioids are the primary factor in constipation in the palliative care setting, and many other prescribed drugs can contribute to constipation, including tricyclic antidepressants, antacids, antiepileptic drugs, anticholinergic agents, and antihypertensives [297]. Additional factors that may contribute to constipation are diverticuli, inflammatory bowel disease, metabolic conditions (hypercalcemia, hypokalemia, hypothyroidism, uremia), cerebral tumors, dehydration, and radiation fibrosis [183,297]. For patients with cancer, constipation may be directly due to tumor involvement that causes intestinal obstruction. A diet low in fiber and decreased physical activity also increase the likelihood of constipation.

Prevention

Prevention of constipation is key, as prophylaxis is more effective than treatment after constipation has been identified. As such, all treatment guidelines strongly recommend that a prophylactic bowel regimen be initiated when treatment with opioids (or other constipation-causing drugs) begins [183,297,298]. The recommended prophylaxis is an osmotic and/or a stimulant laxative [297,298]. Many nonpharmacologic approaches are recommended, and patients should be encouraged to plan a diet with adequate fiber, to increase fluid intake, and to engage in physical activity, as appropriate [297,298]. Family members should be asked to help the patient comply with these measures. Ensuring that the patient has sufficient privacy and comfort with toileting is also recommended [297,298].

Assessment

Issues of personal privacy often lead to a reluctance of patients to discuss constipation, so clinicians and other healthcare professionals must initiate the discussion and talk honestly about what to expect and measures to prevent and manage the symptom. The assessment tools used most often are the Bristol Stool Form Scale and the Constipation Assessment Scale [297,298]. Assessment should include a review of the list of medications, a history of bowel habits, and abdominal and rectal examination. In addition to checking the list of prescribed medications to determine if constipation is a side effect, the physician should ask the patient about over-the-counter drugs and herbal remedies, as constipation can be a consequence of aluminum-containing antacids, ibuprofen, iron supplements, antidiarrhea drugs, antihistamines, mulberry, and flax. A detailed history of bowel habits helps to establish what is considered normal for the individual patient. The patient should be asked about frequency of stool, the appearance and consistency of stools, use of bowel medications, and previous occurrence of constipation. In general, physical examination of the abdomen for tenderness, distention, and bowel sounds can rule out intestinal obstruction as the cause of constipation. A rectal examination can identify the presence of stool, fecal impaction, or tumor. Imaging of the abdomen (by plain x-ray or computerized tomography) may be appropriate to confirm the presence of obstruction. Consideration of the patient's prognosis and preferences for care should be factored into a decision to carry out diagnostic testing. As with assessment of all symptoms, constipation should be reassessed frequently; assessment at least every 3 days is recommended [298].

Management

The goal of treatment should be relief of symptoms related to constipation and re-establishment of bowel habits to the patient's comfort and satisfaction; some recommend a goal of one nonforced bowel movement every 1 to 2 days [183,298]. Systematic reviews have demonstrated that data are insufficient to support one laxative or combination of laxatives over others [297,299,300].
Many laxatives are FDA approved for occasional constipation, and much of the evidence on their efficacy has come from studies of chronic constipation, not patients with life-limiting disease. In its guidelines for the management of chronic constipation, the American College of Gastroenterology notes the following [300]:
  • Polyethylene glycol (PEG) and lactulose (both osmotic) improve stool frequency and stool consistency.
  • Data are insufficient to make a recommendation about the efficacy of stool softeners (docusate [Colace or Surfak]); stimulant laxatives (senna [Senokot, Ex-lax] or bisacodyl [Dulcolax, Correctol]); milk of magnesia; herbal supplements (aloe); lubricants (mineral oil); or combination laxatives (psyllium plus senna).
The results of a systematic review of studies in the palliative care setting also demonstrated insufficient data for recommendations because of a lack of direct comparisons of laxatives [299]. Researchers have concluded that the choice of a laxative should be made on an individual basis, with considerations of patient preferences and the side-effect profile [183,297,299]. For all patients, oral formulations are recommended over rectal suppositories [297,298].
European and Canadian consensus groups and the NCCN have developed practice guidelines for constipation in the palliative care setting on the basis of the available data and expert opinion (Figure 11) [183,297,298]. First-line recommended treatment is a stimulant laxative plus a stool softener (PEG or lactulose) [183,297,299]. A small study of senna with and without docusate for hospitalized patients with cancer showed no significant benefit to the addition of docusate; docusate is specifically not recommended in the Canadian consensus recommendations [298,301]. If constipation persists, other options are bisacodyl, magnesium hydroxide, or sorbitol [183]. Methylnaltrexone (Relistor) was approved by the FDA in 2008 for the treatment of opioid-induced constipation. Although data are still limited, a systematic review indicated that the subcutaneous drug is effective in the palliative care setting, and is especially useful for patients with constipation refractory to conventional laxatives [302]. Practice recommendations note that methylnaltrexone should be considered for patients taking opioids after failure of other laxatives [183,297,298]. Withdrawal of opioids should never be a strategy to manage constipation.
ALGORITHM FOR THE MANAGEMENT OF CONSTIPATION IN PALLIATIVE CARE PATIENTS
Nonpharmacologic interventions are important adjuncts to laxatives, and the interventions used as prophylaxis are recommended for ongoing management [297,298].

NAUSEA AND VOMITING

Nausea may occur alone or with vomiting, a neuromuscular reflex. Nausea and vomiting can exacerbate pain and contribute to insomnia, fatigue and weakness, and anorexia. It can also limit activities and cause distress for the patient and family. Nausea is the result of stimulation of one of several pathways: the chemoreceptor trigger zone (located in the medulla), the cortex of the brain, the vestibulocochlear nerve, or the gastrointestinal tract [66].

Prevalence

Nausea alone affects approximately 6% to 68% of adults with life-limiting disease, and vomiting affects 40% [208]. The rate of nausea and vomiting is highest among patients with cancer [208].

Etiology

The potential causes of nausea and vomiting near the end of life vary according to life-limiting disease [202,211,243,303,304]:
  • Medications (chemotherapy agents, opioids, antidepressants, antibiotics)
  • Radiation therapy (especially to the abdomen or lumbosacral spine)
  • History of peptic ulcer disease or gastroesophageal reflux
  • Delayed gastric emptying
  • Primary or metastatic brain tumor
  • Gastrointestinal tract obstruction
  • Constipation
  • Renal failure
  • Hepatic failure
  • Pancreatitis
  • Hypercalcemia
  • High serum levels of dioxin or anticonvulsants
The causes also differ according to the pathway stimulated (Table 14) [303,304]. Most often the cause is multifactorial, but sometimes no cause can be determined.
CAUSES OF NAUSEA AND VOMITING ACCORDING TO PATHWAY STIMULATED AND CLASS OF ANTIEMETICS
Pathway StimulatedCausesClass of Antimetics
Chemoreceptor trigger zoneMetabolic disorders (hypercalcemia, hyponatremia, hepatic/renal failure)Dopamine antagonists
OpioidsProkinetic agent, dopamine antagonists
Malignant bowel obstructionProkinetic agent, dopamine antagonists, corticosteroids
Cortex of brainIncreased intracranial pressure, anxiety, five sensesCorticosteroids, anxiolytics
Peripheral pathways (gastrointestinal tract)GastroparesisProkinetic agent
Vestibular systemMotionMuscarinic acetylcholine receptor, antihistamine

Prevention

The prevention of nausea and vomiting has focused on prophylactic treatment for patients receiving chemotherapy or radiation therapy for cancer. Although most patients at the end of life do not receive anticancer treatment, chemotherapy may be given as part of palliative care. ASCO classifies chemotherapy drugs according to their emetigenic potential: high (>90% incidence of emesis without an antiemetic), moderate (30% to 90% incidence), low (10% to 30% incidence), and minimal (<10% incidence) [305]. According to ASCO guidelines, a 5-hydroxytryptamine type 3 (5-HT3) antagonist, dexamethasone (Decadron), and a neurokinin 1 (NK1) receptor antagonist (such as aprepitant [Emend]) should be used as prophylaxis for a highly emetic chemotherapy agent or combination (such as an anthracycline and cyclophosphamide) [305]. Palonosetron (Aloxi), in combination with dexamethasone, is recommended for chemotherapy agents with moderate emetic risk, and dexamethasone is recommended before the first dose of chemotherapy with a low emetic risk. For nausea and vomiting not related to chemotherapy, treatment with regular dosing of an antiemetic will help prevent subsequent episodes of the symptoms.

Assessment

A detailed history, physical examination, and review of the medication list are essential for planning effective management of nausea and vomiting. In talking with the patient, the clinician should ensure that the patient is actually experiencing nausea, as patients have used the term nausea to describe other feelings, such as pain, distention, abdominal discomfort, and early satiety [66]. The clinician should ask about the onset of the nausea, how frequently it occurs, if there are precipitating factors, and if there is a relationship to food intake. It may be helpful to ask the patient to rate the intensity of nausea on a scale similar to a pain scale (a 10-point numerical scale). Because the cause of nausea and vomiting is often multifactorial, a multidimensional assessment is beneficial, with particular attention paid to such other symptoms as pain, appetite, fatigue, depression, and anxiety. The physical examination should include evaluation for signs of cachexia or malnutrition, assessment of the abdomen for evidence of bowel obstruction, increased bowel sounds, and abdominal distention. In addition, a neurologic examination should be done to determine if there are signs of increased intracranial pressure, papilledema, or autonomic insufficiency [66]. Diagnostic testing may include laboratory studies to rule out metabolic disorders, renal impairment, or liver failure, or radiographs of the abdomen to determine if there is obstruction.
Nausea is often not reported; patients should be asked if they have experienced nausea even if they have not vomited [243].

Management

Evidence-based guidelines for the management of nausea and vomiting unrelated to chemotherapy and radiation are lacking [306]. In addition, most studies of these symptoms and recommendations are related to the cancer setting. In general, experts have recommended that antiemetics be selected on the basis of the emetic pathway and the etiology of the nausea and/or vomiting, but systematic reviews have found that the evidence for recommendations is weak to moderate at best [202,303,304,306,307,308]. One systematic review found no evidence that the choice of antiemetic according to etiology or multiple antiemetics was better than a single antiemetic [306].
Several classes of pharmacologic agents can be used to manage nausea and vomiting; the main classes used in the end-of-life setting are prokinetic agents, dopamine receptor antagonists, antihistamines, anticholinergics, 5-HT3 receptors, and corticosteroids (Table 15) [303,304,308]. The prokinetic agent metoclopramide (Reglan) has been recommended as a first-line treatment because of its central and peripheral actions and its effectiveness for many chemical causes of nausea [202,243,303]. The drug should be used with caution in patients with heart failure, diabetes, and kidney or liver disease, and the dose should be reduced by half for older patients and those with moderate-to-severe renal impairment [304]. Octreotide (Sandostatin), dexamethasone, and hyoscine hydrobromide (Scopolamine) are recommended for bowel obstruction [89,303,304,306]. Ondansetron (Zofran) has been suggested for chronic nausea, but in September 2011, the FDA issued a safety announcement about the drug, noting that it may increase the risk of QT prolongation on electrocardiogram; more research is being done [240,304]. Haloperidol (Haldol) is recommended for uremia-induced nausea in people with end-stage chronic kidney disease [211]. Dexamethasone is used for nausea and vomiting related to increased intracranial pressure and, although the evidence is limited, it is also used as second-line treatment for intractable nausea and vomiting and as an adjuvant antiemetic [243,303,304]. Olanzapine (Zyprexa), an atypical antipsychotic, has also been effective for nausea that has been resistant to other traditional antiemetics, as well as for opioid-induced nausea [309]. A benzodiazepine (such as lorazepam [Ativan]) may be of benefit if anxiety is thought to be contributing to nausea or vomiting [304].
PHARMACOLOGIC MANAGEMENT OF NAUSEA AND VOMITING
Drug ClassDrugTypical Starting Dose and Frequency
Prokinetic agentsMetoclopramide10–20 mg PO/IV/SC, every 6 to 8 hrs
Dopamine antagonistsHaloperidol1.5–5 mg PO/IV/SC 2 or 3 times daily
Prochlorperazine5–10 mg PO 3 or 4 times daily
Chlorpromazine10–25 mg PO/IV every 4 to 6 hrs
Olanzapine5–10 mg PO daily
Levomepromazine6.25–25 mg SC twice daily
AntihistaminesPromethazine25 PO 4 to 6 hrs
AnticholinergicsHyoscine hydrobromide0.1–0.4 mcg PO/IV/SC every 4 hrs
5-hydroxytryptamine type 3 receptor antagonistsOndansetron4–8 mg PO/IV 1 or 2 times daily
Granisetron1 mg twice daily
Dolasetron200 mg daily
Palonosetron0.25 mg IV daily
Mirtazapine15–45 mg PO, every night
CorticosteroidsDexamethasone4–8 mg daily
PO = orally, IV = intravenously, SC = subcutaneously.
In addition to pharmacologic management of nausea and vomiting, other supportive approaches include maintenance of oral hygiene, regular baths to reduce unpleasant odors, and small meals at regular intervals [202,243]. Cold foods may be better tolerated than hot foods because of decreased smells.

ANOREXIA AND CACHEXIA

The symptoms of anorexia and cachexia often occur in tandem. While anorexia encompasses decreased appetite and food intake, cachexia involves loss of skeletal mass, with accompanying asthenia and autonomic failure. The two conditions are often linked by the term "anorexia-cachexia syndrome," but the exact relationship between the two conditions is unclear [310]. For example, decreased food intake may lead to weight loss, but the body wasting of cachexia is not solely the result of decreased intake [311]. "Wasting" is often used as a synonym for cachexia, but wasting indicates weight loss due to inadequate nutritional intake, whereas cachexia refers to a loss of lean body mass resulting metabolic derangement rather than nutritional deficiency [312].
Cachexia is associated with a poor prognosis in many life-limiting diseases. In fact, unintentional, progressive weight loss of more than 10% of body weight over the past 6 months, with an albumin level less than 2.5 mg/dL is a prognostic indicator for hospice referral [73]. Despite this relationship between cachexia and poor prognosis, the condition is underrecognized and underdiagnosed [313].
Cachexia has also been challenging to define. The lack of an operational definition led to a consensus conference at which a definition was crafted [311]. This definition joins others for disease-specific cachexia (Table 16). The diagnosis and management of anorexia/cachexia has been studied the most in the settings of cancer and HIV infection.
DEFINITIONS AND DIAGNOSTIC CRITERIA FOR CACHEXIA
ConditionDefinition and/or Diagnostic Criteria
All patients with chronic disease
Cachexia is a complex metabolic syndrome associated with underlying illness and characterized by loss of muscle with or without loss of fat mass.
Chronic disease AND
Loss of body weight of 5% or more within the past 3 to 12 months AND
Presence of at least three of the following:
Reduced muscle strength
Fatigue
Anorexia
Low fat-free mass index
Abnormal inflammatory marker levels, anemia, or low albumin level
Cancer cachexiaA multifactorial syndrome defined by an ongoing loss of skeletal muscle mass (with or without loss of fat mass) that cannot be fully reversed by conventional nutritional support and leads to progressive functional impairment. Its pathophysiology is characterized by a negative protein and energy balance driven by a variable combination of reduced food intake and abnormal metabolism.
Cardiac cachexia6% non-edematous, nonvoluntary weight loss over 6 months
HIV-associated wasting
At least one of the following:
10% unintentional weight loss over 12 months
7.5% unintentional weight loss over 6 months
5% body cell mass (BCM) loss within 6 months
Body mass index (BMI) <20 kg/m2
BCM <35% body weight AND BMI <27 kg/m2 (men)
BCM <23% body weight AND BMI <27 kg/m2 (women)

Prevalence

Anorexia occurs in 21% to 92% of adults with life-limiting disease, with the highest rates found among patients with cancer [204,208,290]. Cachexia has been reported in 16% to 57%, again with the highest rates found among people with cancer [313].

Etiology

Across life-limiting diseases, anorexia can occur as a result of several other symptoms, such as fatigue, constipation, xerostomia, dysphagia, mucositis, and nausea. Endocrine abnormalities may also be the cause, and psychologic, social, and spiritual distress can affect the desire to eat [183,317]. Changes in taste sensations (leading to food aversions), altered sense of smell, and early satiety have been common among people with cancer and anorexia [317,318].
Studies have shown that multiple factors contribute to cachexia. Abnormal metabolism is thought to lead to a negative protein and energy balance, with subsequent loss of muscle mass [197,311,314,315]. Inflammation, increased neurohormonal activity, insulin resistance, and increased muscle protein breakdown are often associated with cachexia [311,315,319]. The role these factors play in the development of cachexia may differ according to the underlying chronic condition.

Prevention

Preventive measures for anorexia include effective management of symptoms that are known to have a potential impact on the desire and/or ability to eat. No appropriate measures to prevent cachexia are available.

Assessment

Guidelines for assessing anorexia and cachexia have been developed for the cancer and HIV settings [183,320]. According to NCCN guidelines, assessment of anorexia and cachexia in patients with cancer include the following [183]:
  • Determination of the rate and severity of weight loss
  • Examination of the oral cavity (the mucous membranes, teeth, gingiva, and lips)
  • Review of the medications list for drugs that interfere with intake
  • Evaluation of symptoms that have the potential to interfere with eating and drinking
  • Evaluation for endocrine abnormalities that may be an underlying cause
  • Assessment of social and economic factors
The guidelines for the assessment of HIV-related wasting recommend the following [320]:
  • Thorough and complete history and physical examination, with specific questions related to the patient's nutritional status, caloric intake, appetite, and gastrointestinal and physiologic functioning
  • Measurements of body composition (considering the following factors: age, height, weight, ideal body weight, body cell mass (by BIA), and body mass index
  • Laboratory tests (plasma HIV RNA, CD4 cell count, free and total serum testosterone, and serum albumin and thyroid function (if clinically warranted)
  • Psychosocial evaluation
  • Dietary assessment

Management

Few evidence-based guidelines for the treatment of anorexia and cachexia are available, primarily because of the lack of studies on these underrecognized conditions and the still-emerging understanding of the causes of cachexia. The first step in managing anorexia is to treat symptoms that interfere with appetite and/or the ability to eat. In addition, nonpharmacologic interventions should be directed at improving enjoyment of food, increasing the sense of well-being, and enhancing a sense of normalcy in daily activities. The patient should be encouraged to try favorite foods, to eat small frequent meals, and to drink high-calorie nutritional supplements [183,319,321,322]. Other interventions include an exercise program and consultation with a nutritionist [183]. For people with end-stage liver disease and an inadequate caloric intake, protein restriction (to prevent hepatic encephalopathy) should be avoided [197].
Two drugs are FDA approved as appetite stimulants for anorexia associated with life-limiting disease (Table 17). Megestrol acetate is FDA approved for the treatment of anorexia, cachexia, or unexplained weight loss in patients with AIDS [323]. It has become the most widely used drug for these indications for people with other life-limiting diseases, and a meta-analysis of data from studies (involving people with a variety of life-limiting illnesses) demonstrated that megestrol acetate was beneficial, especially with respect to improving appetite and weight gain in people with cancer [323]. The data were insufficient to recommend megestrol acetate for conditions other than cancer or to recommend an optimal dose [323]. Dronabinol (Marinol), an oral cannabinoid, is FDA approved for anorexia associated with weight loss in people with AIDS [240]. Because of its effects, dronabinol should be used with caution for people with cardiac disorders, depression, or a history of substance abuse; people taking concomitant sedatives or hypnotics; and older individuals [240].
PHARMACOLOGIC MANAGEMENT OF ANOREXIA AND CACHEXIA
DrugDose RangeFindingsFDA Approval
Megestrol acetate400–800 mg/dayIncreased appetite, food intake, and weightFor the treatment of anorexia, cachexia, or unexplained weight loss in patients with AIDS
Dronabinol2.5–20 mg, twice daily (before lunch and dinner)Stimulated appetite and improved body weightFor anorexia associated with weight loss in people with AIDS
Metoclopramide10 mg, 3 times dailyEnhanced appetite in people with early satietyFor nausea and vomiting
Recombinant human growth hormone0.1 mg/kg SC at bedtime (max: 6 mg)Increased lean body mass and improved physical endurance and quality of life among people with HIV-related cachexiaFor HIV-related wasting or cachexia (with concomitant antiretroviral therapy)
Oxandrolone (anabolic steroid)5–20 mg/dayIncreased body weight and lean body mass in cachexia related to HIV and COPDAdjunctive therapy to promote weight gain after weight loss following extensive surgery, chronic infection, or severe trauma and for some patients without a definitive pathophysiologic cause of weight loss
GhrelinNot definedIncreased lean body mass in people with end-stage renal disease, COPD, and heart failureNot approved
AIDS = acquired immune deficiency syndrome, COPD = chronic obstructive pulmonary disease, HIV = human immunodeficiency virus, SC = subcutaneous.
In addition to appetite stimulants, metoclopramide, a drug approved for treatment of nausea and vomiting, is recommended for anorexia related to early satiety in people with cancer [183,321].
The treatment of cachexia is more challenging because its pathophysiology is poorly understood and because treatments may differ according to the life-limiting disease. According to the guidelines for cachexia related to cancer and HIV infection, management includes improving nutritional intake, treating disease-related causes of cachexia, treating anorexia, and addressing psychosocial or lifestyle issues [183,320].
Currently, there is no one treatment or combination of treatments that is effective for all patients with cachexia [322]. Increasing oral intake alone is not sufficient, and reversal of wasting may not always be possible; the goal should be to prevent or delay further wasting and functional decline [183,320,322]. As noted, the use of megestrol acetate is effective in increasing weight, but increased nutrition and weight are not sufficient to effectively manage cachexia, and more research is needed to identify agents to increase body mass and to define a multimodal strategy to stop and/or reverse wasting. These strategies may differ according to the underlying chronic disease.
Studies have indicated that recombinant human growth hormone (rhGH) significantly increases lean body mass and improved physical endurance and quality of life in people with HIV [324,325]. In addition, rhGH has shown benefit in cachexia related to pulmonary and cardiac disease [326]. Recombinant somatropin (Serostim) is approved for the treatment of people with HIV with wasting or cachexia; concomitant antiretroviral therapy is necessary [240]. The drug is contraindicated in active neoplasia [240].
The anabolic steroid oxandrolone (Oxandrin) is FDA approved as adjunctive therapy to promote weight gain after weight loss following extensive surgery, chronic infection, or severe trauma and for some patients without a definitive pathophysiologic cause of weight loss [240]. The drug has shown benefit in increasing body weight and lean body mass in cachexia related to HIV and COPD [327,328]. The drug is safe and well tolerated, but more studies are needed to determine its risk-benefit ratio before it can be used more widely [329].
Ghrelin has been evaluated for the treatment of cachexia, and its anti-inflammatory properties may address the proposed role of inflammation in the development of cachexia [330]. The results of small studies have demonstrated that ghrelin increases lean body mass in people with end-stage renal disease, COPD, and heart failure [326]. Again, more research is needed before this agent can become part of clinical practice.
For people with a limited life expectancy (weeks to days), the clinician should assess the importance of anorexia and cachexia to the patient and the family before prescribing interventions [183]. The clinician should also talk to the patient and family about the risks of artificial nutrition and should consider consulting a spiritual counselor or bioethicist about discontinuing nutrition [183].

DIARRHEA

Diarrhea is characterized by the frequent passage of loose, watery stools, usually defined as more than three unformed stools within a 24-hour period [331]. Diarrhea is most often acute, lasting for a few days; diarrhea is chronic when it persists for more than 3 weeks [331]. Left unchecked, diarrhea can result in dehydration, electrolyte imbalance, and fatigue.

Prevalence

The prevalence of diarrhea among adults with life-limiting disease varies widely, ranging from 3% to 90%, with the highest rates reported among people with HIV infection or AIDS [208].

Etiology

The most frequent cause of diarrhea in patients receiving palliative care is overuse of laxatives and leakage around a fecal impaction [331]. Other causes are infectious disease or underlying disease in people with HIV/AIDS or metastatic colorectal cancer. Diarrhea is also a side effect of many drugs, including antihypertensives, antacids containing magnesium, some NSAIDs, potassium supplements, quinidine, thiazide diuretics, retroviral agents, prokinetic agents (metoclopramide), and antibiotics [331].

Prevention

No appropriate measures to prevent diarrhea are available.

Assessment

A detailed history is the cornerstone of assessing patients for diarrhea. The condition is distressful, yet embarrassing, and direct questions should be asked because the patient may not be forthcoming about the symptom. The clinician should ask the patient about the onset of diarrhea, dietary habits and food intolerances, timing of diarrhea in relation to eating, and medications [331,332]. The patient should also describe bowel movements in terms of frequency, color, and consistency. If possible, a stool specimen should be evaluated.
If infectious diarrhea is suspected, a stool sample should be evaluated to identify the causative organism [332,333].

Management

The American Gastroenterological Association developed guidelines for the treatment of chronic disease in the general clinical setting, but no guidelines are available for the management of diarrhea in palliative care [333].
Treatment of an underlying condition is the optimal approach to managing diarrhea. The clinician should review the medication list and discontinue or reduce the dose of any medication that may be the cause [331,332].
Nonpharmacologic approaches to managing diarrhea include avoiding gas-forming and bulky foods, hot spices, fats, alcohol, and milk until diarrhea is controlled. The patient should be encouraged to drink plenty of fluids to avoid dehydration; beverages with added electrolytes, such as sports drinks, can help maintain proper electrolyte balance.
Pharmacologic management includes the use of bulk-forming agents, adsorbents, and opioids [332]. Kaolin and pectin (Kaopectate), available over the counter, is a combination of adsorbent and bulk-forming agents. However, it provides modest relief and it may take up to 48 hours to be effective [332]. Loperamide (Imodium) is the drug of choice for diarrhea because its side effect profile is better than that for codeine or diphenoxylate (Lomotil) [332]. The initial dose of loperamide is 4 mg, with an additional 2 mg after each loose stool [332]. The package insert for loperamide notes that the maximum daily dose in a 24-hour period is 16 mg, but doses of up to 54 mg a day have been used as part of palliative care with few adverse events [332]. Octreotide has been effective for profuse secretory diarrhea associated with HIV infection and can be used to treat refractory diarrhea [332]. The use of octreotide for diarrhea in the palliative setting is usually off-label, as the drug is FDA approved for the treatment of diarrhea and flushing associated with metastatic carcinoid tumors [240]. Octreotide is administered as a continuous subcutaneous infusion at a rate of 10–80 mcg/hr until improvement of symptoms [332]. Infectious diarrhea should be treated with an appropriate antibiotic. A systematic review found probiotic agents to be of benefit in the management of acute infectious diarrhea [334].

INSOMNIA

As defined, insomnia refers to a variety of sleep disturbances, including difficulty falling asleep and difficulty staying asleep (insufficient amount of sleep or frequent awakenings), that results in impaired function during the day [335]. The most frequent type of insomnia among people at the end of life is difficulty staying asleep, primarily because of pain [336]. A lack of sufficient sleep affects the quality of life by contributing to daytime fatigue and weakness, exacerbating pain, and increasing the potential for depression. Family members also become distressed when the patient is unable to sleep, which, in turn, may increase the burden on caregivers.

Prevalence

Insomnia is common among the general population, and rates reported for adults with life-limiting disease are even higher, ranging from 9% to 83% [197,204,208,337]. The highest rates have been found among patients with end-stage renal disease [208].

Etiology

The primary difference between insomnia in the general population and in people with life-limiting diseases is that insomnia in the latter group is usually secondary to the life-limiting disease or its symptoms [336]. Overall, uncontrolled pain is the most common contributor to the inability to sleep well [336,337]. Other common physical symptoms such as dyspnea, nocturnal hypoxia, nausea and vomiting, pruritus, and hot flashes are also causes of insomnia. Restless legs syndrome may be a substantial contributor to the disruption of sleep in persons with end-stage renal disease [207,338,339].
In addition, many psychologic conditions associated with a life-limiting disease can cause insomnia; depression, anxiety, delirium, spiritual distress, and grief can make it difficult to fall or remain asleep [336]. Insomnia is a side effect of many drugs, most notably corticosteroids, antidepressants, decongestants, opioids, and some antiemetics [335,340]. Patients also may have difficulty sleeping because of disruptions in the normal sleep-wake cycle that result from inactivity and napping during the day. Lastly, stimulants, such as caffeine, and alcohol may keep patients from falling asleep easily.

Prevention

Adequate relief of pain and other symptoms is the mainstay of preventing insomnia. The most effective preventive measure is limiting the amount of time in bed during the day and restricting the amount of daytime sleep [336]. Encouraging patients to increase activity during the day, as tolerated; to adhere to a regular schedule with limited naps; and to avoid caffeine and alcohol in the afternoon and evening can help lead to more healthy sleep patterns.

Assessment

Few patients with life-limiting diseases report insomnia, and few clinicians pursue sleep symptoms in their patients [336]. Clinicians should obtain a sleep history from all patients, following guidelines developed by the American Academy of Sleep Table 18 [335]. The Epworth Sleepiness Scale has been recommended as an assessment tool [183,341].
QUESTIONS TO OBTAIN A SLEEP HISTORY
What is your primary problem with sleep: difficulty falling asleep, waking up frequently during the night, and/or poor quality of sleep?
When did your sleep problems begin?
How often do you have trouble sleeping (every night, most nights)?
Have you ever taken any medication for sleep problems in the past? If so, what did and did not help?
What do you do before you go to bed?
What is your bedroom environment like?
How do you feel (physically and emotionally) in the evening?
What is your average sleep-wake schedule?
How long does it typically take you to fall asleep?
What factors make it longer for you to fall asleep?
What factors shorten your sleep?
How often do you awaken during the night?
When you awaken during the night, how long are you awake?
Do you have symptoms that cause you to awaken during the night?
What do you do to try to fall back asleep after awakening during the night?
How many hours do you sleep each night (on average)?
Do you nap during the day? If so, how often and for how long?
Do you feel sleepy during the day?
How do your sleep problems affect you during the day? Do you have mood disturbances? Feel confused? Feel like your symptoms are worse?
Clinicians should evaluate patients physically as well as psychologically for signs and symptoms that have been identified as contributors to sleep disturbances.

Management

The American Academy of Sleep has developed an evidence-based guideline for the evaluation and management of chronic insomnia in adults and a practice parameter for the psychologic and behavioral treatment of insomnia, but neither offers specific guidelines for managing insomnia at the end of life [335,342]. Nonpharmacologic interventions should be implemented first, with pharmacologic therapy added to the treatment plan if these interventions are not effective [336]. Optimizing sleep habits can be useful, especially if they are begun early in the course of the disease.
The nonpharmacologic approaches used to prevent insomnia are also the primary management strategies. Among the recommended behavioral strategies are the following [335,342]:
  • Stimulus control therapy: Training the patient to reassociate the bed and bedroom with sleep and to re-establish a consistent sleep-wake cycle
  • Relaxation training: Progressive muscle relaxation and reducing thoughts that interfere with sleep
  • Sleep restriction: Limiting the time spent in bed to time spent sleeping
Cognitive behavioral therapy has also been reported to be effective when used in combination with behavioral interventions [335,342]. No nonpharmacologic strategy has been found to be superior to another [342]. These interventions are effective and recommended for older individuals and can also be effective for people with life-limiting disease when strategies are individualized according to the patient [336,342].
Several drugs have been approved by the FDA for the treatment of insomnia; the classes of these drugs are sedative-hypnotics and benzodiazepines Table 19. In addition, antidepressants and antihistamines are often used for insomnia, but this use is off-label.
PHARMACOLOGIC MANAGEMENT OF INSOMNIA
DrugTypical Dose (mg)*Comments
Sedative-Hypnotics (FDA approved for insomnia)
Zolpidem5–20 mgUseful for sleep-onset insomnia; lower dose should be used for older or debilitated individuals or those with impaired hepatic function
Zaleplon5–20 mgUseful for sleep-onset insomnia; lower dose should be used for older or debilitated individuals, patients with impaired hepatic function, and patients taking cimetidine
Eszopiclone1–3 mgHas favorable side-effect profile in older individuals; FDA approved for long-term use
Benzodiazepines (FDA approved for insomnia)
Flurazepam15–30 mgLower dose should be used for older or debilitated individuals; long-acting effect increases risk of daytime drowsiness
Estazolam0.5–2 mgLower dose should be used for older or debilitated individuals
Temazepam7.5–30 mgLower dose should be used for older or debilitated individuals
Triazolam0.125–0.25 mgLower dose should be used for older or debilitated individuals
Quazepam7.5–15 mg
Melatonin Receptor Agonists (FDA approved for insomnia)
Ramelteon8 mgUseful for sleep-onset insomnia; FDA approved for long-term use
Antidepressants (Not FDA approved for insomnia)
Trazodone50–150 mg
Amitriptyline10–100 mg
Doxepin75–150 mg
Trimipramine25–100 mg
Nonprescription (Not FDA approved for insomnia)
Diphenhydramine25–50 mg
*Doses are given as guidelines; actual doses should be determined on an individual basis.
Among sedative-hypnotics, zolpidem (Ambien) is a short- to intermediate-acting drug used primarily for sleep-onset insomnia [335,343]. Zolpidem is recommended by the NCCN for insomnia as part of palliative care for people with cancer [183]. Another sedative-hypnotic, eszopiclone (Lunesta), is intermediate-acting and is one of only two insomnia medications approved by the FDA for long-term use [343].
A systematic search of the literature found no evidence from randomized controlled trials regarding the use of benzodiazepines in palliative care [344]. However drugs in this class are the most commonly used drugs for the treatment of short-term insomnia in people with life-limiting disease [336]. Benzodiazepines are effective in decreasing the time needed to fall asleep as well as the likelihood of waking up during the night [336,343]. Their use should be short term, as their long-term efficacy has not been clearly defined, although this issue is not as important for patients with a limited life expectancy [336]. Lorazepam (Ativan) is a recommended drug for insomnia in people with cancer [183]. The long-acting effect of flurazepam (Dalmane) may be of benefit for some patients [336].
The antidepressant trazodone (Desyrel) is the preferred antidepressant for insomnia (although it is not FDA approved for this indication) [336]. It is the drug of choice among tricyclic antidepressants because of its shorter half-life and its milder anticholinergic side effects [336]. Antidepressants are especially useful for people who have anxiety or depression.
The most recently FDA-approved drug for insomnia is ramelteon (Rozerem), a melatonin receptor agonist. This drug is short acting and used primarily for sleep-onset insomnia [343]. Ramelteon is FDA approved for long-term use [335].
For insomnia related to restless legs, a systematic review showed that dopamine agonists are effective, with cabergoline (Dostinex) and pramipexole (Mirapex) often having a greater efficacy than levodopa (L-Dopa) [345].
Barbiturates are not recommended for insomnia because of the rapid development of tolerance [336]. Two supplements promoted for sleep enhancement—melatonin and valerian—have not been shown to be effective for managing insomnia [336,346].
Several factors must be considered when treating older patients with insomnia. For example, it has been recommended that benzodiazepines be avoided in older individuals because of side effects such as increased risk for falls, confusion, and "hangover" [343]. However, these side effects must be considered in light of an individual's particular situation and weighed against the benefits [343,336]. Eszopiclone and ramelteon have been studied in older individuals and have a favorable side-effect profile for that population [343]. Lower doses are often recommended for older individuals [335].

DELIRIUM

Delirium is a disturbance of consciousness with reduced ability to focus, sustain, or shift attention, as well as changes in cognition (disorientation, memory deficit, language impairment) [347]. Patients may seem confused or be restless, agitated, or combative. Delirium is often difficult to recognize because it shares diagnostic features with other symptoms, especially dementia and depression. As a result, delirium is often unrecognized or misdiagnosed and consequently inappropriately treated or not treated [348]. Delirium is classified into three clinical subtypes: hypoactive, hyperactive, and mixed [349]. Hypoactive delirium is characterized by lethargy, reduced awareness of surroundings, sedation, and psychomotor retardation, whereas hyperactive delirium is characterized by agitation, restlessness, hallucinations, hypervigilance, and delusions [349]. In the palliative care setting, about half of patients with delirium will have the hypoactive subtype [349,350].
Delirium can be extremely distressful for the patient and even more so for family members. The healthcare team can help alleviate family members' distress by educating them about the nature and cause of the syndrome and the potential for reversal. Encouraging them to participate in nonpharmacologic interventions may also help to provide a positive experience.

Prevalence

The prevalence of delirium among adults with life-limiting disease ranges from 6% to 93%, occurring most frequently among patients with cancer [208]. Terminal delirium is a distinct entity that occurs within the last days or hours of life, and it is estimated to occur in 80% of dying patients [351].

Etiology

Many factors may cause delirium, and although the cause is usually multifactorial, often no cause is found [352]. In one comprehensive review, the primary contributor to delirium was unrelieved pain [353]. Delirium is also often caused by medications, including several that are used in the end-of-life setting, such as opioids, corticosteroids, benzodiazepines, and NSAIDs [351]. In addition, age, cognitive deficits, impaired vision/hearing, emotional stress, depression, and comorbidities are predisposing factors of delirium [350,351].

Prevention

Because of the substantial influence of unrelieved pain, adequate pain management can help prevent delirium. Prevention strategies are directed at minimizing precipitating factors, which include a high number of medications (more than six), dehydration, decreased sensory input, psychotropic medications, and a change in environment.

Assessment

The diagnosis of delirium relies on identifying its two features: cognitive impairment and deficits in attention; these features can be assessed with the Mini-Mental State Examination [351]. The Confusion Assessment Method (CAM) is considered to be the gold standard for distinguishing between delirium from other causes of altered mental status, and other tools to evaluate delirium include the Delirium Rating Scale, the Delirium Symptom Interview, and the Memorial Delirium Assessment Scale [351]. Communication with the healthcare team and family is vital in assessing the patient to help determine the onset and course of delirium as well as signs indicative of the syndrome. Some specific ways to help determine if a patient has delirium include [349]:
  • Ask the patient "Do you feel 100% awake?" If they do not, ask "How awake do you feel?"
  • Evaluate whether the patient is easily distracted.
  • Test registration and immediate recall.
  • Assess psychomotor disturbances by noting whether the patient is restless and agitated or slow and hypoactive.
  • Ask the patient if he or she is seeing or hearing strange things.
  • Ask the patient to state the days of the week or months backward, or to give a span of numbers frontward and backward.
  • Ask the patient open-ended questions, and listen for incoherent speech or tangential thought processes.
Clinical assessment and physical examination should also be directed at ruling out underlying causes, such as infection or metabolic abnormalities, and the medication list should be reviewed carefully [183,351].

Management

The treatment of an underlying cause, if identified, is a key step in managing delirium. Whether delirium can be reversed depends on the cause. Delirium caused by psychotropic medications, dehydration, or hypercalcemia is more likely to be reversible than delirium caused by hypoxia, metabolic abnormalities, or nonrespiratory infections [349,354].
Several nonpharmacologic interventions have been successful in preventing and managing delirium Table 20 [183,349,351]. If delirium is refractory to nonpharmacologic measures, medications may be prescribed. The American Psychiatric Association developed guidelines in 1999 for the treatment of delirium with antipyschotics [355]. Level 1 evidence supports the use of haloperidol and chlorpromazine (Thorazine) (typical antipsychotics), and these drugs have the advantage of being available in formulations that allow for multiple routes of administration and of being the most cost-effective [349]. Several systematic reviews have been done to determine the efficacy of antipsychotics for delirium, and although each review has identified only a few well-designed trials, the results have supported the continued use of these drugs (Table 21) [183,349,351,356,357,358]. One of these reviews focused on patients with terminal illness; the review identified only one small study (30 subjects) eligible for analysis; haloperidol and chlorpromazine were equally effective, but the risk for cognitive impairment was slightly greater with chlorpromazine [356]. In the other reviews, the efficacy of haloperidol was found to be similar to that of olanzapine, risperidone (Risperdal), and quetiapine (Seroquel) (atypical antipsychotics) [357,358]. In two small nonrandomized studies—one involving hospitalized patients with cancer—aripiprazole (Abilify) was safe and effective for the treatment of delirium, especially the hypoactive subtype [359,360]. Mild-to-moderate delirium can be managed with low oral doses of antipsychotics, titrating the dose to optimum relief; higher doses can be used for severe delirium [183,351]. For older patients and those with multiple comorbidities, treatment should begin with lower doses and titration should be slow [349]. Factors to consider when selecting a drug include the side-effect profile, the patient's age and baseline mental status, the time to response, and the subtype of delirium [349].
NONPHARMACOLOGIC TREATMENT OPTIONS FOR DELIRIUM
Review all medications; discontinue any unnecessary ones and replace those with a high likelihood of delirium as a side effect.
Rotate opioids or lower the opioid dose.
Provide orienting cues (e.g., calendar, clock, familiar objects) in the patient's room.
Encourage family to sit with the patient.
Encourage activities that are cognitively stimulating (e.g., word puzzles).
Ensure good sleep hygiene.
Minimize noise and interventions at bedtime.
Encourage patient to get out of bed as much as possible.
Provide visual and hearing aids, if appropriate.
Monitor for dehydration.
Minimize use of devices/equipment that are immobilizing (e.g., catheter, intravenous lines).
PHARMACOLOGIC OPTIONS FOR DELIRIUM
DrugDose RangeRoutes of AdministrationComments
Haloperidol0.5–2 mg every 2 to 12 hrsPO, IV, IM, SCConsidered to be first-line treatment.
Chlorpromazine12.5–50 mg every 4 to 6 hrsPO, IV, IM, SC, PRHas more sedative effect than haloperidol, thus is preferred for patients with agitation.
Olanzapine2.5–5 mg every 12 to 24 hrsPOSedation has been a dose-limiting effect; poorer response has been associated with older age, pre-existing dementia, and hypoactive subtype.
Risperidone0.25–1 mg every 12 to 24 hrsPOResponse may be better with hypoactive subtype; orthostatic hypotension is possible adverse effect.
Quetiapine12.5–100 mg every 12 to 24 hrsPOSedation and orthostatic hypotension are possible adverse effects.
Aripiprazole5–30 mg every 24 hrsPOResponse may be better with hypoactive subtype.
Lorazepam0.5–2 mg every 2 to 4 hrsIV, SCMay be added to treatment with haloperidol if agitation is refractory to high doses.
PO = orally, IV = intravenously, IM = intramuscularly, SC = subcutaneously, PR = rectally.
The goal of treatment is to reach patients' baseline mental state, not to sedate them, and patients should be reassessed frequently until this goal is met [351]. If agitation is refractory to high doses of haloperidol, the antipsychotic lorazepam may be helpful [183,351]. The management of delirium also includes providing support to family, to help them cope with the condition [183,351]. The management of terminal delirium will be discussed later in this course.

PSYCHOSOCIAL CARE

The natural initial reaction to a limited life expectancy is emotional, and patients as well as their families experience a wide range of emotions, including disbelief, anger, fear, and sadness. Over time, these emotions broaden; patients may feel isolated and lonely, anxious about the burden on their family, or hopeless. Family members may have guilt about their own well-being, anxiety about the future, and grief about the loss of their loved one. Practical issues such as the cost of care and loss of income from the patient and/or caregiver can add substantially to the feelings of stress.
The prevalence of psychologic suffering is high during the last year of life, and addressing this aspect of care is integral to the patient's overall comfort and quality of life. Anxiety and depression are the most common psychologic symptoms at the end of life, yet they are among the most underdiagnosed and untreated symptoms [66,361]. Psychologic suffering exacerbates pain and other symptoms, limits the patient's capacity for pleasurable activities, and causes distress for both the patient and the family [202,362].
The word "distress" has become standard to describe the psychologic suffering experienced by patients with life-limiting disease. The NCCN notes that the word "distress" is more acceptable and is associated with less stigma than words such as "psychosocial" or "emotional" [183]. In its guidelines on distress management, the NCCN defines distress as existing "along a continuum, ranging from common normal feelings of vulnerability, sadness, and fears to problems that can become disabling, such as depression, anxiety, panic, social isolation, and existential and spiritual crisis" [183]. According to a study of patients in a palliative care program, the answers to the question "What bothers you most?" included [212]:
  • Emotional, spiritual, existential, or nonspecific distress (16%)
  • Relationships (15%)
  • Concerns about the dying process and death (15%)
  • Loss of function and normalcy (12%)
Patients at increased risk of distress include individuals with a history of psychiatric disorder, substance abuse, or depression/suicide attempt; with cognitive impairment or communication barriers; with severe comorbid conditions; and with spiritual/religious concerns. Other factors that predispose a patient to distress include rapidly progressing disease, unrelieved pain, and uncontrolled symptoms [202]. Women, younger individuals, and individuals with young children are also at increased risk [183]. Gay and lesbian patients with life-limiting diseases often have distinct sources of suffering [66]. These patients may be disenfranchised from their families or have been subjected to social stigma, leading to fears of abandonment and isolation. In some instances, spiritual crises may be the result of guilt and shame from past behaviors. Many patients with HIV/AIDS have suffered through the loss of loved ones to the same disease, some of whom may have been part of the individual's defined family and network of social support.
As with physical symptoms, assessment of distress and the psychosocial and spiritual well-being of the patient must be ongoing, as changes occur over time [5,66]. In addition, worsening symptoms and disease progression can affect patients' coping mechanisms [223]. One study found significant correlations between the will to live and existential, psychologic, and social sources of distress. In that study, hopelessness, burden to others, and dignity were the variables with the most influence [363]. Other studies have consistently shown that psychosocial suffering has a stronger association than pain with a desire to hasten death [364,365,366,367,368,369].
How a patient responds to his or her disease and care is strongly influenced by attitudes and values learned through family interactions, and social workers should evaluate the patient and family to assess psychosocial as well as practical problems and recommend and/or carry out interventions [5,183]. For many patients, the primary concern about their illness is its impact on the family. The need for palliative care raises issues regarding power, structure, and roles among the patient and his or her family [66]. The impact of a life-limiting disease and the ensuing care threatens the structure and integrity of the family, as family roles are reassigned, the rules of daily living are altered, and methods of problem-solving are revised. Families vary in their ability to adapt to such restructuring, and dysfunction can result from either limited or excessive adaptation. At one end of this spectrum, family members have difficulty breaking away from coping mechanisms, even though they are ineffective. At the other end of the spectrum, family members continually try new coping strategies to meet each crisis, resulting in chaos [66]. Both types of dysfunction can lead to increased demands on the healthcare team and can interfere with the delivery of appropriate care.

ANXIETY

Anxiety is a feeling of fear, apprehension, and dread. The patient feels uneasy, insecure, and uncertain about the future. Often, the patient is not able to identify the source of anxiety, but it can be related to any number of physical, psychologic, social, spiritual, or practical issues common during the end of life.

Prevalence

Severe anxiety varies widely among adults with life-limiting disease, ranging from 8% to 79%, with the highest rate among patients with cancer [208].

Etiology

One of the primary causes of anxiety is inadequate pain relief. Anxiety may also be the result of a patient's overwhelming concern about his or her illness, the burden of the illness on the family, and the prospect of death. In addition, anxiety is a potential side effect of many medications, including corticosteroids, metoclopramide, theophylline, albuterol, antihypertensives, neuroleptics, psychostimulants, antiparkinsonian medications, and anticholinergics. Lastly, withdrawal from opiates, alcohol, caffeine, and sedatives can result in anxiety.

Prevention

Effective pain management is the best way to prevent anxiety. Also, educating the patient and the family about what to expect over the course of the illness and providing adequate psychologic and spiritual support can help comfort the patient, thereby preventing anxiety.

Assessment

Family members and friends may be able to provide information about the level of anxiety experienced by the patient currently and in past situations. All members of the healthcare team should evaluate the patient and the clinical record for reversible causes of anxiety, such as those caused by medications or withdrawal syndromes, and should try to distinguish anxiety from delirium, depression, or bipolar disorder [370,371].
Anxiety manifests itself through physical as well as psychologic and cognitive signs and symptoms. These signs and symptoms include dyspnea, paresthesias, tachycardia, chest pain, urinary frequency, pallor, restlessness, agitation, hyperventilation, insomnia, tremors, excessive worrying, and difficulty concentrating.

Management

Nonpharmacologic approaches are essential for managing anxiety, and the addition of pharmacologic treatment depends on the severity of the anxiety [66,372]. Effective management of pain and other distressing symptoms, such as constipation, dyspnea, and nausea, will also help to relieve anxiety. If the anxiety is thought to be caused by medications, they should be replaced by alternate drugs. Other strategies include psychologic support that allows the patient to explore fears and concerns and to discuss practical issues with appropriate healthcare team members. Relaxation and guided imagery may also be of benefit [373]. A consult for psychologic therapy may be needed for patients with severe anxiety.
When pharmacologic management is deemed necessary, benzodiazepines are generally preferred, and administration on an as-needed basis is usually sufficient [66]. Neuroleptics and tricyclic antidepressants may also be effective (Table 22). For all medications, the initial dose should be low and subsequently titrated to produce the desired effect within the level of tolerance. Benzodiazepines should be given with caution in older patients, as these drugs may harm memory or cause confusion and agitation in patients who have cognitive impairment [375].
PHARMACOLOGIC MANAGEMENT OF ANXIETY AND DEPRESSION
ConditionDrug Class, DrugsTypical Starting Oral Dose*Titration RecommendedMaximum Daily DoseComments
AnxietyBenzodiazepines
Lorazepam0.5–2 mg, every 1 to 6 hrsMay titrate upwardFirst choice
Diazepam2.5–10 mg, every 3 to 6 hrsMay titrate upward
Midazolam2–10 mg/day (SC)May titrate upward
Clonazepam0.5–1.0 mg, 3 times per dayMay titrate upward 4 mg
Neuroleptics
Haloperidol0.5–4.0 mg, every 4 to 6 hrsMay titrate upward
Thioridazine10 mg, 3 times per dayMay titrate upward
Tricyclic Antidepressant
Imipramine10–25 mg, 3 times per dayMay titrate upward
DepressionSSRIs
Fluoxetine20 mg/dayIncrease by 10 mg every 1 to 2 wks20–60 mgFirst choice when immediate onset not needed (onset at 4 to 6 wks)
Paroxetine10 mg/dayIncrease by 10 mg every 1 wk10–50 mg
Sertraline50 mg/dayIncrease by 25 mg every 1 wk50–150 mg
Escitalopram10 mg/day20 mg
Venlafaxine18.75 mg/dayIncrease by 75 mg every 1 wk75–225 mg
Tricyclic Antidepressants
Amitriptyline25 mg/dayIncrease by 25 mg every 1 to 2 days50–150 mgLess useful because of side effects; slow onset of action (3 to 6 wks)
Nortriptyline25 mg/day50–150 mg
Desipramine25 mg/day50–150 mg
Doxepin25 mg/day50–200 mg
*Doses are given as guidelines; actual doses should be determined on an individual basis.

DEPRESSION

Depression is linked to many other symptoms, especially pain, and is a primary source of suffering. Depression in patients with life-limiting disease is a challenge to identify, as feelings of sadness, helplessness, and hopelessness are a typical reaction to the situation [202,373]. Depression is more likely when sadness and/or hopelessness is overwhelming or pervasive and is accompanied by a sense of despair [373,376]. Early diagnosis is essential for effective treatment and relief of other symptoms.

Prevalence

The prevalence of depression varies widely among adults with life-limiting diseases, ranging from 3% to 82%, with the highest rate among patients with HIV/AIDS and end-stage liver disease [197,208].

Etiology

Unrelieved pain is one of the primary risk factors for depression. Other causes within the physical domain include metabolic disorders (hyponatremia or hypercalcemia), lesions in the brain, insomnia, or side effects of medications (corticosteroids or opioids). Many patients with heart failure have comorbidities and polypharmacy, both of which can increase the risk of depression [377,378]. Psychosocial causes include despair about progressive physical impairment and loss of independence, financial stress, family concerns, lack of social support, and spiritual distress.

Prevention

Adequate management of pain, attention to psychosocial and spiritual well-being, and early referral for mental health or pastoral counseling are the best strategies to prevent depression.

Assessment

The diagnosis of depression is complicated, as the usual somatic signs of depression—anorexia, sleep disturbances, weight loss, and fatigue—are often symptoms related to the underlying disease or part of the constellation of symptoms experienced by patients with life-limiting disease [223]. Because of this, assessment should focus on psychologic and cognitive symptoms, such as:
  • Persistent dysphoria
  • Loss of pleasure in activities
  • Frequent crying
  • Loss of self-esteem
  • Sense of worthlessness
  • Excessive guilt
  • Pervasive despair
  • Thoughts of suicide
A diagnosis of depression requires the presence of at least five depression-related symptoms within the same 2-week period, and the symptoms must represent a change from a previous level of functioning [347]. A simple screening tool that has been found to be effective is to ask the patient, "Are you depressed?" or, "Do you feel depressed most of the time?" [223,379,380]. The physician should also discuss the patient's mood and behavior with other members of the healthcare team and family to help determine a diagnosis. Patients who have thoughts of suicide must be assessed carefully. The physician should differentiate between depression and a desire to hasten death because of uncontrolled symptoms [66]. Psychologic counseling should be sought, as well as measures to enhance the management of symptoms.

Management

The effective management of depression requires a multimodal approach, incorporating supportive psychotherapy, cognitive strategies, behavioral techniques, and antidepressant medications [46]. Patients with depression should be referred to mental health services for evaluation, and resultant approaches may include formal therapy sessions with psychiatrists or psychologists or counseling from social workers or pastoral advisors. In addition, physicians can help by having discussions with the patient to enhance his or her understanding of the disease, treatments, and outcomes, and to explore expectations, fears, and goals. Behavioral interventions, such as relaxation techniques, distraction therapy, and pleasant imagery have been effective for patients with mild-to-moderate depression [46].
Strong evidence supports the use of tricyclic antidepressants or SSRIs, along with psychosocial interventions, for the management of depression in patients with cancer [46,209]. Evidence to support the use of specific pharmacologic agents to treat depression in patients with noncancer diagnoses is not as strong, but psychostimulants may also be helpful [46,66,223,381]. The choice of medication depends on the time available for treatment. The most immediate effect (within days) is achieved with a rapid-acting psychostimulant; longer times to therapeutic effect are associated with SSRIs (2 to 4 weeks) and tricyclic antidepressants (3 to 6 weeks).

SPIRITUAL NEEDS

Spirituality is unique to each person. It is founded in cultural, religious, and family traditions and is modified by life experiences. Spirituality is considered to be separate from religious faith, and many surveys have shown that spirituality or religion is an integral component of people's lives [66,382]. Spirituality also plays a significant role in health and illness. Studies have shown spirituality to be the greatest factor in protecting against end-of-life distress and to have a positive effect on a patient's sense of meaning [376,383]. Thus, a spiritual assessment and spiritual care to address individual needs are essential components of the multidimensional evaluation of the patient and family [202,384].
A life-limiting disease will lead patients to ask questions that may give way to spiritual conflicts, such as "Why would God let me suffer this way?" Patients may also carry out life review in search of meaning for their illness; some may view their illness as punishment for past "sins." Left unanswered, spiritual questions and concerns lead to spiritual distress and suffering, which can cause or exacerbate pain and other physical and psychosocial symptoms. It then becomes critical for the healthcare team to facilitate pastoral services to address patients' spiritual concerns [5]. In general, the spiritual and existential concerns of patients at the end of life relate to four areas: the past, the present, the future, and religion (Table 23) [202].
SPIRITUAL AND EXISTENTIAL CONCERNS OF PATIENTS AT THE END OF LIFE
Relation of ConcernConcerns
Past
Value and meaning of the person's life
Worth of relationships
Value of previous achievements
Painful memories or shame
Guilt about failures, unfulfilled aspirations
Present
Disruption of personal integrity
Physical, psychologic, and social changes
Increased dependency
Meaning of the person's life
Meaning of suffering
Future
Impending separation
Hopelessness
Meaninglessness
Death
Religion
Strength of faith
A life lived without disgrace to the faith
Existence of afterlife
The need for spirituality at the end of life is heightened, and patients will search for meaning as a way to cope with emotional and existential suffering [385]. Spirituality helps patients cope with dying through hope. At the time of diagnosis, patients hope for cure, but over time, the object of hope changes and the patient may hope for enough time to achieve important goals, personal growth, reconciliation with loved ones, and a peaceful death [13,66]. Spirituality can also help a patient gain a sense of control, acceptance, and strength. As a result, greater spiritual well-being has been associated with decreased rates of anxiety and depression among people with advanced disease [204,386].
There has been a growing emphasis on the need for physicians to discuss spirituality with their patients [384,387]. A spiritual history should be obtained to elicit answers to such questions as:
  • Do you consider yourself spiritual or religious?
  • Do you have spiritual beliefs that help you cope with stress?
  • What importance does your faith or belief have in your life?
  • Are you part of a spiritual or religious community?
One recommended mnemonic for the components of a spiritual history is SPIRIT: spiritual belief system; personal spirituality; integration with a spiritual community; ritualized practices and restrictions; implications for medical care; and terminal events planning [388].
Although spiritual care is an essential component of palliative care, what patients and families perceive to be spiritual care and how it should be delivered have not been well-defined [382]. Patients and families have found spiritual comfort with friends and family, clergy and other pastoral care providers, and healthcare professionals [382]. Among healthcare professionals, barriers to providing spiritual care are time; social, religious, or cultural discordance; and lack of privacy and care continuity [382].

FAMILY-CENTERED PSYCHOSOCIAL NEEDS

Adequate psychosocial support is also needed for the patient's family. The structure of families varies widely, and it is important to note that what constitutes a family is defined by the patient. It is essential for the healthcare team to talk to patients during the initial assessment about who provides support, with whom they wish to share information, and who should be involved in planning care and decision making [5,66]. For some patients, friends provide the support network when families are not near or the patient is disenfranchised from his or her family. Social workers have a prominent role in helping these patients overcome such barriers as discrimination and legal and financial issues, as well as ensuring appropriate support for grieving partners who may be disenfranchised [389].
Family caregivers can become overwhelmed with added responsibilities. Often, the caregiver is a spouse who is older and may also have illnesses. In addition, children and teenagers are frequently forgotten, but addressing their concerns and needs is essential for their psychologic well-being and appropriate grieving [66]. Young children will realize that the family structure has been disrupted. They should be encouraged to ask questions, and they usually need time to interpret answers. Adolescence is a challenging time in itself, and dealing with the illness and loss of a parent or close family member may result in aggressive behavior, isolation, or sexuality. Frequent evaluation of family members' coping strategies, moods, and behaviors can help to determine if early referral for individual counseling or family therapy is necessary. Support should be provided to ensure that the patient and family has access to resources to help with finances, that the home environment is safe, that caregivers are available, and that adequate transportation is available [183].
Family roles are also important to understand, and these roles are strongly influenced by culture. Many cultures highly value family, with strong family ties across many generations. Patients from such cultures will often have many visitors at one time. The palliative care team should accommodate such visits when possible. In addition, family hierarchy may dictate behavior of family caregivers. For example, in traditional Vietnamese families, a female member of the family is expected to stay at the bedside of the patient for comfort and support [390]. In Asian families, elders are revered and a young person cannot tell an older person what to do [391]. This may make it difficult for a healthcare professional who is younger than the patient. Patients and families who adhere to Native American cultures have unique traditions and rituals that should be respected [392].
All members of the healthcare team should become familiar with the cultural context of their patients and provide resources from within the cultural community if possible. A bilingual healthcare worker can provide an important link to a community [393].

IMMINENT DEATH AND LOSS

In the last days, the goals of the healthcare team are to ensure a peaceful death for the patient and to support the family during the dying process and throughout grief and mourning. The focus for the patient is management of symptoms and emotional and spiritual ease, and the focus for the family is education to prepare them for the dying process.

THE PATIENT'S NEEDS

During the last days, all care should be directed at comfort, and the NCCN has listed several interventions for imminently dying patients (Table 24) [183]. The physician should minimize the number of medications by reassessing the need for each one. The symptoms that occur most commonly during the last days are pain, noisy breathing, dyspnea, and delirium, and medications to manage these symptoms should be maintained or initiated [66]. In addition, medication may be required to reduce the risk of seizures. Medications should be prescribed for the least invasive route of administration (oral or buccal mucosa), but patients may lose the ability to swallow, making a subcutaneous, transdermal, or intravenous route necessary.
INTERVENTIONS FOR PATIENTS WHO ARE IMMINENTLY DYING
Intensify ongoing care.
Try to ensure privacy (if not at home, arrange for private room if possible).
Discontinue diagnostic tests.
Reposition for comfort as appropriate.
Avoid unnecessary needle sticks.
Provide mouth care (e.g., hydrogen peroxide/water solution).
Treat for urinary retention and fecal impaction.
Ensure access to medication even when oral route is not available.
Prepare to meet request for organ donation and autopsy.
Allow patient and family uninterrupted time together.
Ensure the patient and family understand the signs and symptoms of imminent death and are supported through the dying process.
Offer anticipatory bereavement support.
Provide support to children and grandchildren.
Encourage visits by children if consistent with family values.
Support culturally meaningful rituals.
Facilitate around-the-clock family presence.
Ensure that caregivers understand and will honor advance directives.
Provide respectful space for families.
Facilitate closure.
Treatment of pain should continue, and knowledge of opioid pharmacology becomes critical during the last hours of life [66,394]. The metabolites of morphine and some other opioids remain active until they are cleared through the kidneys. If urine output stops, alternative opioids, such as fentanyl or methadone, should be considered, as they have inactive metabolites [211,395].
Anticholinergic medications can eliminate the so-called "death rattle" brought on by the build-up of secretions when the gag reflex is lost or swallowing is difficult. Specific drugs recommended include scopolamine, glycopyrrolate, hyoscyamine, and atropine (Table 25) [66,183,394,396]. For patients with advanced kidney disease, the dose of glycopyrrolate should be reduced 50% (because evidence indicates that the drug accumulates in renal impairment) and hyoscine butylbromide should not be used (because of a risk of excessive drowsiness or paradoxical agitation) [211]. Some evidence suggests that treatment is more effective when given earlier; however, if the patient is alert, the dryness of the mouth and throat caused by these medications can be distressful. Repositioning the patient to one side or the other or in the semiprone position may reduce the sound. Oropharyngeal suctioning is not only often ineffective but also may disturb the patient or cause further distress for the family. Therefore, it is not recommended.
TREATMENT OF EXCESSIVE RESPIRATORY SECRETIONS CAUSING "DEATH RATTLE"
DrugDose
Scopolamine (transdermal patch)1 or 2 (1.5-mg) patches applied behind the ear and changed every 48 to 72 hours; if ineffective, switch to 50 mcg/hr continuous IV or SC infusion and double the dose every hour, up to 200 mcg/hr
Glycopyrrolate (Robinul)1–2 mg PO or 0.1–0.2 mg SC/IV, every 4 to 8 hrs, as needed; or 0.4–1.2 mg/day continuous infusion
Hyoscyamine (Levsin)0.125–0.5 mg PO/SL/SC/IV every 4 hrs as needed
Atropine (1%) eye drops1 or 2 drops PO/SL, titrate every 8 hrs; 0.4 mg SL every 15 min, as needed
PO = orally, IV = intravenously, SC = subcutaneously, SL = sublingually, PR = rectally.
Terminal delirium should be treated aggressively at its first signs (restlessness, moaning, increasing confusion, and drowsiness). Haloperidol is frequently the first choice for its relatively quick action [202,394]. Other drugs may include olanzapine, chlorpromazine, levomepromazine, and benzodiazepines [202,394]. For terminal delirium associated with agitation, benzodiazepines, including clonazepam, midazolam, diazepam, and lorazepam may be helpful [202,223,394]. Depending on which drug is used, administration may intravenous, subcutaneous, or rectal, and the dose can be titrated until effective.
Seizures at the end of life may be managed with high doses of benzodiazepines. Other antiepileptics such as phenytoin (administered intravenously), fosphenytoin (administered subcutaneously), or phenobarbital (60–120 mg rectally, intravenously, or intramuscularly every 10 to 20 minutes as needed) may become necessary until control is established.
A calm and peaceful environment should be maintained for the patient. Family and spiritual leaders should be allowed to carry out traditional rites and rituals associated with death.

Palliative Sedation

Palliative sedation may be considered when an imminently dying patient is experiencing suffering (physical, psychologic, and/or spiritual) that is refractory to the best palliative care efforts. Terminal restlessness and dyspnea have been the most common indications for palliative sedation, and thiopental and midazolam are the typical sedatives used [183,397,398]. For patients who have advanced kidney disease, midazolam is recommended, but the dose should be reduced because more unbound drug becomes available [211]. Before beginning palliative sedation, the clinician should consult with a psychiatrist and pastoral services (if appropriate) and talk to the patient, family members, and other members of the healthcare team about the medical, emotional, and ethical issues surrounding the decision [66,183,223,399,400]. Formal informed consent should be obtained from the patient or from the healthcare proxy.

Physician-Assisted Suicide

Physician-assisted suicide, or hastened death, is defined as active euthanasia (direct administration of a lethal agent with a merciful intent) or assisted suicide (aiding a patient in ending his or her life at the request of the patient) [66]. The following are not considered to be physician-assisted suicide: carrying out a patient's wishes to refuse treatment, withdrawal of treatment, and the use of high-dose opioids with the intent to relieve pain. The American Medical Association Code of Ethics explicitly states, "Physician-assisted suicide is fundamentally incompatible with the physician's role as healer, would be difficult or impossible to control, and would pose serious societal risks" [401]. Position statements against the use of physician-assisted suicide have been issued by many other professional organizations, including the NHPCO, the AAHPM, and the NCCN [183,402,403]. The basis for these declarations is that appropriate hospice care is an effective choice for providing comfort to dying patients.
In 2010, in a first-of-its-kind comprehensive consensus statement, the Heart Rhythm Society in collaboration with the major cardiology, geriatrics, and palliative care societies, emphasized that deactivation of implantable cardioverter-defibrillators is neither euthanasia nor physician-assisted suicide [404]. The organizations urged clinicians to respect the right of patients to request deactivation.
The NCCN guidelines recommend that physicians explore requests for assisted suicide and explain to the patient the distinctions among assisted suicide, treatment withdrawal, and aggressive symptom management [183]. Some states have enacted assisted suicide statutes. State laws vary, and knowledge of your local statutes is necessary.

THE FAMILY'S NEEDS

Ongoing communication with family members is essential to ensure their well-being as their loved one dies. The healthcare team should discuss what will happen over the course of dying so the family can be better prepared for symptoms such as altered breathing patterns and sounds, terminal delirium, and unconsciousness [5,183,394]. The family should be reassured that what they may think the patient is experiencing is not the patient's actual reality.
The altered breathing patterns that are present as death is imminent are distressful for family members, as they believe that the patient is experiencing a sense of suffocation. Also distressful to family is the sound of the death rattle. The healthcare team should assure family that these signs do not indicate that the patient is suffering and explain that additional therapy will not be of benefit.
Families often misinterpret the early signs of terminal delirium as signs of uncontrollable pain. However, if pain has been adequately managed throughout the delivery of palliative care, such pain will not begin during the last hours. As the patient slips in and out of consciousness, family members may become increasingly distressed about not being able to communicate anymore with their loved one. Although it is unknown what a dying patient can hear, other experiences in medicine suggest that awareness may be greater than the ability to respond. Family members should be encouraged to continue talking with their loved one to help them attain a sense of closure.
Despite the best efforts to prepare the family, reactions are unpredictable when death occurs. The clinician should take time to answer questions from family members, including children, and perhaps provide information on the physiologic events associated with death [66]. For family members who were not present during the death, the clinician should describe the event, while reassuring them that the patient died peacefully.
Many experts believe that people can handle grief better if they spend time with a loved one immediately after death. Family members should be allowed to touch, hold, and kiss their loved one as they feel comfortable. The healthcare team should respect the needs of the family to conduct personal, cultural, or religious traditions, rites, and rituals.

GRIEF, MOURNING, AND BEREAVEMENT

Palliative care extends beyond the patient's death, with the focus shifting to support of the family during bereavement and mourning. Although the terms "grief," "mourning," and "bereavement" are often used interchangeably, their definitions are different. Grief is a normal reaction to a loss; mourning is the process by which individuals adjust to the loss; and bereavement is the period of time during which grief and mourning occur [66,405]. Psychosocial support of the family is essential throughout the duration of palliative care and can help to decrease the risks of morbidity, substance abuse, and mortality that have been found among spouses and other loved ones of patients who have died [5].

Grief

Grief comprises a range of feelings, thoughts, and behaviors that fall in the realm of the physical, emotional, and social domains [66]. Individuals may have trouble sleeping, changes in appetite, or other physical symptoms or illness. Emotions can include sadness, anxiety, guilt, and anger. Return to work, activities with friends, and taking care of family can be beneficial.
Grief counseling for the family and patient should begin when the patient is alive, with a focus on life meaning and the contributions from the patient's family. An understanding of the mediators of the grief response can help physicians and other members of the healthcare team recognize the family members who may be at increased risk for adapting poorly to the loss [406]. These mediators are:
  • Nature of attachment (how close and/ or dependent the individual was with regard to the patient)
  • Mode of death (the suddenness of the death)
  • Historical antecedents (how the individual has handled loss in the past)
  • Personality variables (factors related to age, gender, ability to express feelings)
  • Social factors (availability of social support, involvement in ethnic and religious groups)
  • Changes and concurrent stressors (number of other stressors in the individual's life, coping styles)
Clinical assessment should be carried out for individuals at risk of complicated grief. Distinguishing between grief and depression can be challenging, as many signs and symptoms are similar. However, the hallmarks of depression are constant and unremitting feelings of worthlessness, hopelessness, helplessness, anhedonia, and suicidal ideation [202].

Mourning

Satisfactory adaptation to loss depends on "tasks" of mourning [406]. Previous research referred to "stages" of mourning, but the term "task" is now used because the stages were not clear-cut and were not always followed in the same order. The tasks include:
  • Accepting the reality of the loss
  • Experiencing the pain of the loss
  • Adjusting to the environment in which the deceased is missing (external, internal, and spiritual adjustments)
  • Finding a way to remember the deceased while moving forward with life
After the patient's death, members of the palliative care team should encourage the family to talk about the patient, as this promotes acceptance of the death. Explaining that a wide range of emotions is normal during the mourning process can help family members understand that experiencing these emotions is a necessary aspect of grieving. Frequent contact with family members after the loved one's death can ensure that the family is adjusting to the loss. Referrals for psychosocial and spiritual interventions should be made as early as possible to optimize their efficacy.

Bereavement

Bereavement support should begin immediately with a handwritten condolence note from the clinician. Such notes have been found to provide comfort to the family [407,408]. The physician should emphasize the personal strengths of the family that will help them cope with the loss and should offer help with specific issues. Attendance at the patient's funeral, if possible, is also appropriate.
How bereavement services are provided through a hospice/palliative care program vary. Programs usually involve contacting the family at regular intervals to provide resources on grieving, coping strategies, professional services, and support groups [183,223]. When notes are sent, family members should be invited to contact the physician or other members of the healthcare team with questions. Notes are especially beneficial at the time of the first holidays without the patient, significant days for the family (patient's birthday, spouse's birthday), and the anniversary of the patient's death. Bereavement services should extend for at least 1 year after the patient's death, but a longer period may be necessary [5,223].

PALLIATIVE CARE FOR SPECIFIC POPULATIONS

Some patient populations present with unique needs that create challenges to the delivery of high-quality palliative care. Among these populations are older patients, including those with dementia and/or debility; children and adolescents; and patients in the critical care setting.

OLDER PATIENTS AND NURSING FACILITY RESIDENTS

Older patients comprise the largest percentage of patients who receive palliative care through hospice. In 2010, patients 75 years and older represented two-thirds of the individuals who received such care [1]. The number of hospices providing care in nursing homes has increased, growing from 1,850 in 1999 to 2,768 in 2006 [409]. The mean length of hospice stay also increased during that time [409].
The primary issues for this population at the end of life are the variation in care settings, a high level of comorbidities, inadequate management of pain and suffering, and a high prevalence of dementia [101].
The majority of older individuals receive hospice care at home, but up to 25% are residents at a nursing facility at the time of death [1,410]. Each setting presents different issues. For patients at home, caregiver burden is high, as the long disease trajectory requires an extended need for family caregivers. In addition, the primary caregiver may be a spouse who is older than 75 years of age and may have multiple health issues. For patients in nursing facilities, care may be fragmented and staff often lack an appropriate understanding of pharmacology, drug addiction and dependence, management of side effects, and effective nonpharmacologic therapies [411,412,413]. Also, family members often have grief symptoms before the death of the patient; the most frequent grief symptom is yearning (separation distress) [414]. Thus, early psychosocial support and bereavement services for family are important.
Older patients, especially those with end-stage organ disease, often have substantial comorbidities and take multiple medications, both of which add to the complexity of care [101,377,415]. One study of patients with heart failure found that approximately 33% had COPD, 40% had diabetes, and more than 50% had coronary heart disease or hypertension [416]. With respect to multiple medications, a study found that older patients took an average of 6.5 medications and that 29% of the patients were taking a medication that was considered to be "never appropriate" [417]. Polypharmacy increases the likelihood of drug interactions, and clinicians should review the medication list and eliminate those drugs that are not providing clear benefit [415]. Knowledge of pharmacokinetics, pharmacodynamics, and pathophysiology are needed in making decisions to stop or adjust drugs [418]. Consulting with a pharmacist can be valuable.
As with the overall population of patients at the end of life, pain management is inadequate for older patients, with pain experienced by more than 50% of patients at home and as many as 80% of patients in nursing facilities [419,420,421]. Studies have confirmed that older patients receive less pain medication at the end of life than younger patients and that pain management is inadequate for residents of nursing facilities [422,423,424,425]. The American Geriatrics Society has issued guidelines for the management of chronic pain for older patients, and physicians and nursing facility staff should become familiar with this resource and other guidelines for pain [421]. Improvement is also needed in the treatment of patients who have psychosocial symptoms, such as depression, agitation, anxiety, and loneliness [426].
Perhaps the greatest issue is the need for better palliative care for patients with dementia [100,411,427]. The prevalence of dementia has been reported to be 40% to 50% among persons older than 80 years of age [409]. Underlying dementia makes it difficult to identify symptoms, especially pain and psychosocial disorders. As a result, suffering is prevalent among patients with dementia. In fact, one study showed that 93% of patients with dementia died with an intermediate or high level of suffering [428]. The assessment of pain can be particularly challenging when the patient is unable to communicate. This situation calls for a multipronged approach consisting of observation, discussion with family and caregivers, and evaluation of the response to pain medication or nonpharmacologic measures. Recommendations for assessing pain in nonverbal patients have been developed by the American Society for Pain Management Nursing [429].
As dementia progresses, behavioral disturbances become more frequent, and symptoms include hallucinations, sleep disorders, agitation, paranoia, delusions, anxiety, and combativeness. Care should be taken to differentiate these symptoms from those associated with the underlying disease or as an adverse effect of drugs. In addition, dementia can affect the prognosis of other chronic diseases, and health events or complications such as hip fracture, pneumonia, febrile episodes, or eating problems can substantially reduce the life span for patients with advanced dementia [170,422].
The understanding of advanced dementia is limited, and as noted, the uncertainty of the disease course makes it difficult for advance care planning and referral to hospice care [170,411,430,431,432]. The progressive nature of dementia adds importance to the need for advance directives, and involvement of the family in decision making is crucial [170,433]. Educational resources about palliative care and hospice can help family and patients better understand the language needed in advance directives and the benefit of hospice services [410,433].
In an effort to enhance the quality of care at the end of life for older patients, the CAPC published the report Improving Palliative Care in Nursing Homes [21]. Based on their research, the authors of this report identified four different models for integrating preferred practices for palliative and hospice care for patients in nursing homes [21,434]:
  • Palliative Care Consult Service: Palliative care services are provided by healthcare professionals as requested by the nursing home Medical Director or Director of Nursing or the patient's attending physician.
  • Hospice-based Palliative Care Consult Service: Palliative care services are provided by healthcare professionals employed at a local hospice as requested by the nursing home Medical Director or Director of Nursing or the patient's attending physician.
  • Nursing Home Services Integrated Palliative Care: Palliative care services are provided by staff employed directly by a nursing home that incorporates one or more of the NQF's domains of care.
  • Hospice Care: Specialized end-of-life palliative care services are provided by contracted hospice providers to hospice-eligible residents.

CHILDREN/ADOLESCENTS

Although the majority of physicians involved in the care of children/adolescents with life-limiting diseases are likely to make referrals for palliative or hospice care, the rate of hospice use among eligible children/adolescents is lower than that among adults, ranging from 5% to 25% in the United States and Canada [435,436,437]. The cause of most deaths among children is cancer, and it has been proposed that referral to hospice at the time of disease relapse would enhance the quality of care for children and their families; yet, only 2.5% of referrals are made at that time [438,439]. Instead, most hospice referrals are made at the time of disease progression (44%), at the end of therapeutic options (26%), or at the time of imminent death (20%) [438]. Similarly, most palliative care referrals are made late, with 30% to 44% of pediatricians preferring a palliative care consultation when curative therapy is no longer the goal [439].
One factor contributing to inadequate palliative/hospice care referral may be availability of appropriate services. A survey of institutions participating in Children's Oncology Group clinical trials found that a palliative care team was available in 58% of institutions and hospice care in 60% [440]. Furthermore, even when available, most services were not well used by patients [440]. In addition, many healthcare professionals are inexperienced with pediatric palliative care, and the availability of sufficiently trained pediatric hospice professionals is limited [87,435,438].
Research has identified several additional barriers to palliative care at the end of life for children/adolescents, many of which differ from those in the adult setting (Table 26) [14,87,438,439]. The sense of failure or of "giving up" may be heightened among both pediatric healthcare professionals and family members because the potential death of a child goes against the natural order. Compared with pediatric oncology professionals, parents are more likely to favor the use of aggressive treatment near the end of the child's life and consider hope a more important factor in treatment decision making [441]. As with adults, integrating palliative care early in the disease continuum can help overcome conflicts in treatment goals related to uncertainty of the prognosis [87]. Although aggressive treatment should be discontinued when it is of no benefit, the Patient Protection and Affordable Care Act of 2010 now allows for disease-directed treatment to be given concurrently with hospice [442]. (A life expectancy of 6 months is still a criterion for eligibility.) Clinicians usually recognize the lack of a realistic chance for cure before parents do and should talk openly with parents about discontinuing aggressive treatment and directing attention to enhancing the quality of life that remains for the child [443]. Members of the palliative care team should discuss treatment goals with the family, outline choices for interventions as the end of life draws near, and establish limits of care as the health status changes [435,444,445].
BARRIERS TO EFFECTIVE PALLIATIVE CARE FOR CHILDREN
Rarity of death among children
Uncertain prognosis
Unrealistic expectations or denial of parents
Association of palliative care with "giving up" or hopelessness
Immeasurable parental distress at loss of child
Lack of pediatricians' knowledge about distinction between palliative care and hospice
Provider sense of failure when a child dies
Lack of symptom assessment tools
Lack of knowledge regarding pediatric dosing of symptom-relief medications
Fragmentation of medial and psychosocial/spiritual services for children
Lack of adequately trained pediatric hospice professionals
Inadequate education for providers and families about palliative care
Lack of adequate reimbursement
The involvement of the young patient in discussions about diagnosis, prognosis, and treatment goals is another important issue in the pediatric population. Members of the healthcare team should collaborate with parents to determine how much information should be shared with the child and how involved the child should be with decision making; these determinations should be based on the child's intellectual and emotional maturity [14,446]. Many parents wish to protect their child by withholding information, but studies have shown that children often recognize the seriousness of their illness and prefer open communication about their disease and prognosis [447,448]. Such open exchange of information can help to avoid the fear of the unknown and preserve the child's trust in his or her parents and/or family and caregivers [448]. Thus, as much as possible and appropriate, the child should be allowed to participate in discussions about the direction of care [446].
When parents and clinicians involve the child in discussions, the language used should be developmentally appropriate for the child and the clinician should check often to make sure the child understands. Having the child repeat the information in his or her own words is one way to assess comprehension. When the child demonstrates an understanding of the illness and the prognosis, the emphasis should be on his or her preferences for care, and the child's preferences should be given equal weight in the decision making [434,449,450]. The physician should be an advocate for the child's preferences and decision [451].
Symptom management is a key issue in the pediatric setting. One study indicated that 89% of dying children suffered "a lot" or "a great deal" from at least one symptom in their last month of life, and other end-of-life symptoms have often been intractable [443,452]. These problems are compounded by the fact that many clinicians who provide components of pediatric palliative care do not have confidence in their ability to manage end-of-life symptoms [437]. Inadequate training and the paucity of data on symptoms in children/adolescents contribute to this lack of confidence. Few studies have been done to determine the prevalence of symptoms in children/adolescents with life-limiting diseases, the studies that do exist are in the cancer setting, and evidence-based recommendations for interventions are not available.
According to reports of parents, the most common symptoms during the last month of life are similar to those among adults; fatigue (weakness) and pain have been the most frequently reported Table 27 [443,452,453]. When evaluating fatigue in children, age is a consideration in how fatigue is discussed. Children think about fatigue as a physical sensation, and adolescents think about fatigue as either physical and/or mental tiredness [454]. Parents or other caregivers tend to report fatigue in terms of how it interferes with the child's activities [454].
PREVALENCE OF SYMPTOMS AMONG CHILDREN IN THE LAST MONTH OF LIFE
SymptomRange in Prevalence
Pain73% to 92%
Fatigue/weakness86% to 91%
Anorexia68% to 81%
Reduced mobility61% to 76%
Nausea/vomiting57% to 63%
Constipation55% to 59%
Anxiety/depression45% to 48%
Dyspnea41% to 81%
As with adults, the patient's self-report of pain is the most reliable indicator [66,455], which makes assessment particularly challenging in young children. Pain assessment must be appropriate for each child's age, developmental level, and cultural context, and assessment tools have been developed for four age groups, from infants to 18 years of age, and for nonverbal or cognitively impaired children [456,457,458]. These tools include lists of behaviors for the parents or caregivers to rate, as well as areas for parents to provide their own rating of the child's pain and to note what has previously helped to alleviate pain (Table 28). The tools for children who are 5 years of age and older include age-appropriate items such as drawings of a child's body on which the child is asked to mark with a crayon or pencil the area that hurts and different sized circles to indicate pain intensity. The tools are available on the Promoting Excellence in End-of-Life Care website (http://www.promotingexcellence.org). The Wong-Baker FACES scale is recommended for children who are at least 3 years of age [235]. This scale has been found to be valid and reliable for Japanese, Thai, Chinese, and black children and has been modified for use with Alaska native children [460,461,462].
BEHAVIORS TO EVALUATE IN ASSESSING PAIN IN CHILDREN AND ADOLESCENTS
Age of ChildBehaviors
Infants (<1 year)
Sleep during the previous hour
Facial expressions (frown, furrowed brow, quivering chin)
Consolability
Crying
Sucking
Flexing of fingers and toes
Motor activity
Breath-holding
Children (1 year and older)
Energy level
Eating behavior
Interest in usual activities
Whining, crying, groaning, complaining
Holding or protecting part of body
Seeking comfort, closeness
For children who are too young to verbally express pain, clinicians and parents must rely on behavioral cues, such as frowning, a furrowed brow, a quivering chin, crying, sucking, flexing of fingers and toes, and breath-holding in infants. Behavioral indicators in older children include decreased energy level, eating, and interest in usual activities; holding or protecting part of the body; seeking comfort or closeness; and whining or groaning [456,457,458].
Pain management according to the WHO ladder has been found to be effective for children/adolescents, and the NCCN has developed guidelines for pediatric pain management [183,463,464]. Acetaminophen or NSAIDs, codeine, or oxycodone is recommended for pain rated as 0-3 on a scale of 0 to 10; an acetaminophen/opioid combination, NSAIDs, oxycodone, or morphine is recommended for pain rated as 4-6; and morphine or oxycodone is recommended for pain rated as 7-10 [183]. It is important to note that codeine may not be metabolized in 35% of children, and analgesia will be ineffective in those children [183]. Pharmacokinetic data for pediatric medications are lacking, and physicians should consult pediatric specialists for appropriate dosing of medications for symptom relief. Pain medication should be complemented by age-appropriate nonpharmacologic interventions; touch, massage, stroking, and rocking are effective for infants, toddlers, and young children, and guided imagery, music and art therapy, play therapy, controlled breathing, and relaxation techniques are beneficial for older children [455,465,466].
Attention to psychosocial support for the patient, parents, and other family members is crucial in the pediatric setting. Although most parents think that psychosocial issues should be discussed with the child's physician and would find that discussion to be valuable, fewer than half of parents raise such topics [467]. Furthermore, parents report that only 15% to 20% of physicians assess the family's psychosocial issues [467]. Among the psychosocial issues common in children/adolescents and their families are ineffective family coping strategies, the patient's relationships with peers, psychologic adjustment of healthy siblings, and long-term psychologic adjustment for parents [446,465,468,469,470,471,472]. The palliative care team must carefully evaluate the patient and family and provide resources and appropriate referrals.

CRITICAL CARE SETTING

Nearly 50% of patients who die in the hospital are in the ICU for some period of time during the last 3 days of life [473,474]. In addition, 13% of patients admitted to the ICU with traumatic injury will die [473]. The abruptness of a traumatic injury is vastly different from the illness trajectories of life-limiting diseases, and palliative care seems incongruous in the ICU, a high-technology environment of the most aggressive life-prolonging treatments. The effective delivery of palliative care is challenged by many factors inherent in the ICU setting, including inadequate training of healthcare professionals, unrealistic expectations of patients and families, misunderstanding of lifesaving measures, and a greater need for surrogate decision making [473,475,476]. As these factors gain greater recognition, there is a growing emphasis on integrating palliative care elements into the care of patients with traumatic injury and/or patients in an ICU [119,473,475,476,477,478].
The focus on complex, lifesaving care in the ICU creates a gap in providing relief of patients' symptoms. As in all settings, symptom assessment and management must be a priority for ICU patients. It has been suggested that an interdisciplinary palliative care assessment be carried out early in an ICU stay, preferably within 24 hours after admission, with documentation of a comprehensive care plan within 72 hours after admission [476,479].
ICU patients are often young, and families expect lifesaving procedures to be effective [476]. Misunderstanding of lifesaving measures has been reported to be an obstacle to high-quality palliative care [480]. Clinicians and other members of the team should maintain open, ongoing communication about the patient's prognosis, the feasibility of recovery, and the burden of treatment. The sudden, often catastrophic events that bring patients to the ICU compound stress and grief in family members, whose psychosocial needs peak earlier than in other palliative care settings [476]. As a result, psychosocial and bereavement support for families must begin early in the course of the patient's stay in ICU, preferable within 24 hours after the patient's admission to the ICU [476].
The abruptness of traumatic injury or catastrophic illness is also associated with the lack of preparation of advance directives for many patients. There is often no time for planning during the short end-of-life process, and approximately 95% of patients are unable to participate in their care [476]. As a consequence, surrogates must make decisions, and such decisions have been shown to correlate poorly with the preferences of patients [481,482].
The most critical decision in the ICU setting is the withdrawal of life-support technologies. Withdrawal of mechanical ventilator support should be discussed with the family or surrogate when they (or the patient) raise the issue or when the clinician believes that the ventilator is no longer meeting the patient's goals or is more burdensome than beneficial [119]. To ease the discussion for families, the clinician should review the patient's status and care goals before discussing withdrawal of support [119]. Once the decision has been made to withdraw life support, the physician should review the process with family members, clarify the decision, ensure that the patient's spiritual and cultural context are considered, and reassure the family that comfort measures will be carried out [119,476]. Withdrawal of life support should then be immediate, not carried out over hours or days, and established protocols for withdrawal of mechanical ventilation should be followed [476,483].
Recognizing the importance of palliative care in critical care settings, the Society of Critical Care Medicine developed recommendations calling for, among other improvements, [473]:
  • Increased competency in all aspects of palliative care, including the use of sedatives, analgesics, and nonpharmacologic approaches to manage symptoms
  • Improved communication with family
  • Better understanding of the practical and ethical aspects of withdrawing life-sustaining treatment
  • Development of comprehensive bereavement programs to support both families and the needs of the clinical staff
To specifically address the needs of patients' families, the American College of Critical Care Medicine developed guidelines with 43 recommendations that included endorsement of a shared decision-making model, early and repeated care conferencing to reduce family stress and improve consistency in communication, honoring culturally appropriate requests for truth-telling and informed refusal, spiritual support, family presence at rounds and resuscitation, open flexible visitation, and family support before, during, and after a death [484].
Many other initiatives have focused on improving palliative care in the ICU, primarily by having a palliative care team screen patients for potential consultation and increasing communication between the team and attending physicians. This approach increased the use palliative care consultation 113% in one study and from 5% to 21% in another study [24,485]. Another model that integrates palliative care into the ICU improved the quality of care and led to a higher rate of formalization of advance directives, better utilization of hospice, and a decreased use of nonbeneficial life-prolonging treatments [474].

CONCLUSION

As a result of ongoing advances in medicine, the trajectory of illness for many diseases has shifted, yielding an increasing number of patients needing palliative care throughout the continuum of care and, especially, at the end of life. High-quality palliative care focuses on the physical, psychosocial, and spiritual well-being of the patient as well as the family. Care is provided by a palliative healthcare team composed of members who have expertise in communication, pharmacologic principles of pain management, and identification of psychosocial and spiritual symptoms. Palliative care enables patients to die without suffering and provides for grief counseling and bereavement services for a family adjusting to loss. Physicians and other healthcare professionals should strive to enhance their knowledge of key strategies to achieve high-quality palliative care, as detailed in this course.
Complete for credit

Works Cited

1. National Hospice and Palliative Care Organization. NHPCO Facts and Figures: Hospice Care in America. Alexandria, VA: National Hospice and Palliative Care Organization; 2011.
2. Mount BM. The Royal Victoria Hospital Palliative Care Service: a Canadian experience. In: Saunders C, Kastenbaum R (eds). Hospice Care on the International Scene. New York, NY: Springer; 1997: 73-85.
3. World Health Organization. Cancer Pain Relief and Palliative Care. Technical Report Series 804. Geneva: World Health Organization; 1990.
4. Doyle D, Hanks GWC, McDonald N. Introduction. In: Doyle D, Hanks GWC, MacDonald D (eds). Oxford Textbook of Palliative Medicine. Oxford: Oxford University Press; 1993: 3.
5. National Consensus Project for Quality Palliative Care Consortium Organizations. Clinical Practice Guidelines for Quality Palliative Care. Pittsburgh, PA: National Consensus Project; 2004.
6. World Health Organization. WHO Definition of Palliative Care. Geneva: World Health Organization; 2007.
7. Ferris F, Bruera E, Cherny N, et al. ASCO Special Article. Palliative cancer care a decade later: accomplishments, the need, next steps—from the American Society of Clinical Oncology. J Clin Oncol. 2009;27(18):3052-3058.
8. Lynn J, Adamson DM. Living Well at the End of Life. Adapting Health Care to Serious Chronic Illness in Old Age. Santa Monica: Rand; 2003.
9. Pastrana T, Junger S, Ostgathe C, Elsner F, Radbruch L. A matter of definition—key elements identified in a discourse analysis of definitions of palliative care. Palliat Med. 2008;22(3):222-232.
10. National Institutes of Health. State-of-the Science Conference Statement on Improving End-of-Life Care. Available at http://consensus.nih.gov/2004/2004EndOfLifeCareSOS024html.htm. Last accessed April 27, 2012.
11. The SUPPORT Investigators. A controlled trial to improve care for seriously ill hospitalized patients in the Study to Understand Prognoses and Preferences for Outcomes and Risk of Treatments. JAMA. 1995;274:1591-1598.
12. Freeborne N, Lynn J, Desbiens NA. Insights about dying from the SUPPORT project. The Study to Understand Prognoses and Preferences for Outcomes and Risks of Treatments. J Am Geriatr Soc. 2000;48(5 Suppl):S199-S205.
13. Field MJ, Cassel CK (eds). Approaching Death: Improving Care at the End of Life. Washington, DC: National Academies Press, 1997.
14. Field MJ, Behrman RE (eds). When Children Die: Improving Palliative and End-of-Life Care for Children and Their Families. Report of the Institute of Medicine. Washington, DC: National Academies Press, 2003.
15. National Quality Forum. A National Framework and Preferred Practices for Palliative and Hospice Care Quality. A Consensus Report. Available at http://www.qualityforum.org/Projects/n-r/Palliative_and_Hospice_Care_Framework/Palliative___Hospice_Care__Framework_and_Practices.aspx. Last accessed April 27, 2012.
16. Singer PA, Martin DK, Kelner M. Quality end-of-life care: patients' perspectives. JAMA. 1999;281:163-168.
17. Steinhauser K, Clipp EC, McNeilly M, et al. In search of a good death: observations of patients, families, and providers.Ann Intern Med. 2000;132:825-832.
18. Center to Advance Palliative Care. The Joint Commission Advanced Certification for Palliative Care. Available at http://www.capc.org/palliative-care-professional-development/Licensing/joint-commission. Last accessed April 27, 2012.
19. National Rural Health Association. Providing Hospice and Palliative Care in Rural and Frontier Areas. Kansas City, MO: National Rural Health Association; 2005.
20. Linder JF, Meyers FJ. Palliative care for prison inmates. "Don't let me die in prison." JAMA. 2007;298(8):894-901.
21. Center to Advance Palliative Care. Improving Palliative Care in Nursing Homes. Available at http://www.capc.org/support-from-capc/capc_publications/nursing_home_report.pdf. Last accessed April 27, 2012.
22. National Hospice and Palliative Care Organization. NHPCO Facts and Figures: Pediatric Palliative and Hospice Care in America. Alexandria, VA: National Hospice and Palliative Care Organization; 2009.
23. Mosenthal AC, Weissman DE, Curtis JR, et al. Integrating palliative care in the surgical and trauma intensive care unit: a report from the Improving Palliative Care in the Intensive Care Unit (IPAL-ICU) Project Advisory Board and the Center to Advance Palliative Care. Crit Care Med. 2011; [Epub ahead of print].
24. Villarreal D, Restrepo MI, Healy J, et al. A model of increasing palliative care in the intensive care unit: enhancing interprofessional consultation rates and communication. J Pain Symptom Manage. 2011;42(5):676-679.
25. American Society of Health-System Pharmacists. ASHP statement on the pharmacist's role in hospice and palliative care.Am J Health Syst Pharm. 2002;59:1770-1773.
26. Fine PG. The evolving and important role of anesthesiology in palliative care. Anesth Analg. 2005;100:183-188.
27. Elkington H, White P, Addington-Hall J, Higgs R, Pettinari C. The last year of life of COPD: a qualitative study of symptoms and services. Respir Med. 2004;98(5):439-445.
28. Higginson IJ, Evans CJ. What is the evidence that palliative care teams improve outcomes for cancer patients and their families? Cancer J. 2010;16(5):423-435.
29. Zimmermann C, Riechelmann R, Krzyzanowska M, Rodin G, Tannock I. Effectiveness of specialized palliative care. A systematic review. JAMA. 2008;299(14):1698-1709.
30. Caffrey C, Sengupta M, Moss A, Harris-Kojetin L, Valverde R. Home health care and discharged hospice care patients: United States, 2000 and 2007. Natl Health Stat Report. April 27, 2011;(38):1-27.
31. Lynn J. Serving patients who may die soon and their families: the role of hospice and other services. JAMA. 2001;285(7):925-932.
32. Weckmann M. The role of the family physician in the referral and management of hospice patients. Am Fam Physician. 2008;77(6):807-812, 817-818.
33. Centers for Medicare and Medicaid Services. Medicare Hospice Benefits. CMS Pub No. 02154. Baltimore, MD: U.S. Department of Health and Human Services, 2011.
34. Epstein R, Street RL Jr. Patient-Centered Communication in Cancer Care: Promoting Healing and Reducing Suffering. NIH Publication No. 07-6225. Bethesda, MD: National Cancer Institute; 2007.
35. Tattersall M, Butow PN, Clayton JM. Insights from cancer patient communication research. Hematol Oncol Clin North Am. 2002;16:731-743.
36. de Haes H, Teunissen S. Communication in palliative care: a review of recent literature. Curr Opin Oncol. 2005;17:345-350.
37. Connor S, Elwert F, Spence C, Christakis NA. Racial disparity in hospice use in the United States in 2002. Palliat Med. 2008;22(3):205-213.
38. Cohen LL. Racial/ethnic disparities in hospice care: a systematic review. J Palliat Med. 2008;11(5):763-768.
39. Ngo-Metzger Q, Phillips RS, McCarthy EP. Ethnic disparities in hospice use among Asian-American and Pacific Islander patients dying with cancer. J Am Geriatr Soc. 2008;56(1):139-144.
40. Lepore MJ, Miller SC, Gozalo P. Hospice use among urban Black and White U.S. nursing home decedents in 2006. Gerontologist. 2011;51(2):251-260.
41. Washington KT, Bickel-Swenson D, Stephens N. Barriers to hospice use among African Americans: a systematic review.Health Soc Work. 2008;33(4):267-274.
42. Mitchell BL, Mitchell LC. Review of the literature on cultural competence and end-of-life treatment decisions: the role of the hospitalist. J Natl Med Assoc. 2009;101(9):920-926.
43. Colon M. Hospice and Latinos: a review of the literature. J Soc Work End Life Palliat Care. 2005;1(2):27-43.
44. El-Jawahri A, Greer JA, Temel JS. Does palliative care improve outcomes for patients with incurable illness? A review of the evidence. J Support Oncol. 2011;9(3):87-94.
45. Brumley R, Enquidanos S, Jamison P, et al. Increased satisfaction with care and lower costs: results of a randomized trial of in-home palliative care. J Am Geriatr Soc. 2007;55(7):993-1000.
46. Qaseem A, Snow V, Shekelle P, et al. Evidence-based interventions to improve the palliative care of pain, dyspnea, and depression at the end of life: a clinical practice guideline from the American College of Physicians. Ann Intern Med. 2008;148(2):141-146.
47. Casarett D, Pickard A, Bailey FA, et al. A nationwide VA palliative care quality measure: the family assessment of treatment at the end of life. J Palliat Med. 2008;11(1):68-75.
48. Gelfman LP, Meier DE, Morrison RS. Does palliative care improve quality? A survey of bereaved family members. J Pain Symptom Manage. 2008;36(1):22-28.
49. Abernethy AP, Currow DC, Fazekas BS, Luszcz MA, Wheeler JL, Kuchibhatla M. Specialized palliative care services are associated with improved short- and long-term caregiver outcomes. Support Care Cancer. 2008;16(6):585-597.
50. Connor S, Pyenson B, Fitch K, Spence C, Iwasaki K. Comparing hospice and nonhospice patient survival among patients who die within a three-year window. J Pain Symptom Manage. 2007;33(3):238-246.
51. Ternel JS, Greer JA, Muzikansky A, et al. Early palliative care for patients with non-small-cell lung cancer. N Engl J Med. 2010;363(8):733-742.
52. Saito AM, Landrum MB, Neville BA, Ayanian JZ, Weeks JC, Earle CC. Hospice care and survival among elderly patients with lung cancer. J Palliat Med. 2011;14(8):929-939.
53. Smith TJ, Temin S, Alesi ER, et al. American Society of Clinical Oncology provisional clinical opinion: the integration of palliative care into standard oncology care. J Clin Oncol. 2012;30(8):880-887.
54. Goodman DC, Esty AR, Fisher ES, Chang C-H. Trends and Variation in End-of-Life Care for Medicare Beneficiaries with Severe Chronic Illness: A Report of the Dartmouth Atlas Project. Available at http://tdi.dartmouth.edu/documents/EOL_Trend_Report_0411%20%283%29.pdf. Last accessed May 16, 2012.
55. Zhang B, Wright AA, Huskamp HA, et al. Health care costs in the last week of life. Arch Intern Med. 2009;169(5):480-488.
56. Emanuel EJ. Cost savings at the end of life: What do the data show? JAMA. 1996;275:1907-1914.
57. Emanuel E, Ash A, Yu W, et al. Managed care, hospice use, site of death, and medical expenditures in the last year of life.Arch Intern Med. 2002;162(15):1722-1728.
58. Taylor D, Ostermann J, Van Houtven CH, Tulsky JA, Steinhauser K. What length of hospice use maximizes reduction in medical expenditures near death in the US Medicare program? Soc Sci Med. 2007;65:1466-1478.
59. Morrison RS, Dietrich J, Ladwig S, et al. Palliative care consultation teams cut hospital costs for Medicaid beneficiaries.Health Aff. 2011;30(3):454-463.
60. Bekelman DB, Havranek EP. Palliative care for patients with acute decompensated heart failure: an underused service?Nat Clin Pract Cardiovasc Med. 2008;5(5):250-251.
61. Curtis JR. Palliative and end-of-life care for patients with severe COPD. Eur Respir J. 2008;32(3):796-803.
62. Roberts C, Seiger A, Buckingham R, Stone R. Clinician perceived good practice in end-of-life care for patients with COPD.Palliat Med. 2008;22(7):855-858.
63. Berzoff J, Swantkowski J, Cohen LM. Developing a renal supportive care team from the voices of patients, families, and palliative care staff. Palliat Support Care. 2008;6(2):133-139.
64. Jablonski A. Palliative care: misconceptions that limit access for patients with chronic renal disease. Semin Dial. 2008;21(3): 206-209.
65. Hinds PS, Oakes LL, Hicks J, Anghelescu DL. End-of-life care for children and adolescents. Semin Oncol Nurs. 2005;21(1):53-62.
66. Berger AM, Portenoy RK, Weissman DE (eds). Principles & Practice of Palliative Care & Supportive Oncology. 2nd ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2002.
67. Ross DD, Shpritz D, Hull MM, Goloubeva O. Long-term evaluation of required coursework in palliative and end-of-life care for medical students. J Palliat Med. 2005;8:962-974.
68. Scharfenberger J, Furman CD, Rotella J, Pfeifer M. Meeting American Council of Graduate Medical Education guidelines for a palliative medicine fellowship through diverse community partnerships. J Palliat Med. 2008;11(3):428-430.
69. American Medical Association. FREIDA Online. Available at http://www.ama-assn.org/ama/pub/education-careers/graduate-medical-education/freida-online.page. Last accessed April 27, 2012.
70. American Board of Medical Specialties. 2011 Certificate Statistics. Chicago, IL: American Board of Medical Specialties, 2011.
71. Wilkinson S, Perry R, Blanchard K, Linsell L. Effectiveness of a three-day communication skills course in changing nurses' communication skills with cancer/palliative care patients: a randomised controlled trial. Palliat Med. 2008;22(4):365-375.
72. National Board for Certification in Hospice and Palliative Care Nurses. March 2012 CBT Testing Results. NBCHPN Certification Newsletter. April 2012. Available at http://www.nbchpn.org/DisplayPage.aspx?Title=Certification%20Newsletter%20-%20April%202012. Last accessed May 5, 2012.
73. National Hospice Organization. Medical guidelines for determining prognosis in selected non-cancer diseases. Hosp J. 1996;11(2):47-63.
74. Lutz S, Spence C, Chow E, et al. Survey on use of palliative radiotherapy in hospice care. J Clin Oncol. 2004;22:3581-3586.
75. McCloskey S, Tao ML, Rose CM, Fink A, Amadeo AM. National survey of perspectives of palliative radiation therapy: role, barriers, and needs. Cancer J. 2007;13(2):130-137.
76. Jarosek SL, Virnig BA, Feldman R. Palliative radiotherapy in Medicare-certified freestanding hospices. J Pain Symptom Manage. 2009;37(5):780-787.
77. Wright A, Katz IT. Letting go of the rope—aggressive treatment, hospice care, and open access. N Engl J Med. 2007;357:324-327.
78. Finlay E, Casarett D. Making difficult decisions easier. Using prognosis to facilitate transitions to hospice. Ca Cancer J Clin. 2009;59:250-263.
79. Goodlin SJ. Palliative care in congestive heart failure. J Am Coll Cardiol. 2009;54(5):386-396.
80. Salz T, Brewer NT. Offering chemotherapy and hospice jointly: one solution to hospice underuse. Med Decis Making. 2009;29(4):521-531.
81. Casarett DJ. Rethinking hospice eligibility criteria. JAMA. 2011;305(10):1031-1032.
82. Donnell RW. The Provision on Reimbursement for End of Life Counseling. Available at http://doctorrw.blogspot.com/2011/01/provision-on-reimbursement-for-end-of.html. Last accessed May 16, 2012.
83. Sophocles A. Time is of the essence: coding on the basis of time for physician services. Fam Pract Manage. 2003;10(6):27-31.
84. University of Minnesota Physicians. E&M Coding Based on Time. Available at http://www.med.umn.edu/gim/prod/groups/med/@pub/@med/documents/asset/med_96209.pdf. Last accessed May 16, 2012.
85. Brickner L, Scannell K, Marquet S, Ackerson L. Barriers to hospice care and referrals: survey of physicians' knowledge, attitudes, and perceptions in a health maintenance organization. J Palliat Med. 2004;7(3):411-418.
86. Hauptman P, Swindle J, Hussain Z, Biener L, Burroughs TE. Physician attitudes toward end-stage heart failure: a national survey. Am J Med. 2008;121:127-135.
87. Davies B, Sehring SA, Partridge JC, et al. Barriers to palliative care for children: perceptions of pediatric health care providers. Pediatrics. 2008;121(2):282-288.
88. Twomey F, O'Leary N, O'Brien T. Prediction of patient survival by healthcare professionals in a specialist palliative care inpatient unit: a prospective study. Am J Hosp Palliat Care. 2008;25(2):139-145.
89. Glare PA, Sinclair CT. Palliative medicine review: prognostication. J Palliat Med. 2008;11:84-103.
90. Christakis NA, Lamont EB. Extent and determinants of error in doctors' prognoses in terminally ill patients: Prospective cohort study. BMJ. 2000;320:469-472.
91. Joshi K, Guthmann R, Kishman C. How do we decide when a patient with nonmalignant disease is eligible for hospice care?Family Pract. 2006;55(6).
92. Boyd K, Murray SA. Recognising and managing key transitions in end of life care. BMJ. 2010;341:c4863.
93. Glare P, Virik K, Jones M, et al. A systematic review of physicians' survival predictions in terminally ill cancer patients. BMJ. 2003;327(7408):195-198.
94. Alexander M, Christakis NA. Bias and asymmetric loss in expert forecasts: a study of physician prognostic behavior with respect to patient survival. J Health Econ. 2008;27:1095-1108.
95. Casarett DJ, Quill TE. "I'm not ready for hospice:" strategies for timely and effective hospice discussions. Ann Intern Med. 2007;146(6):443-449.
96. Coventry PA, Grande GE, Richards DA, Todd CJ. Prediction of appropriate timing of palliative care for older adults with non-malignant life-threatening disease: a systematic review. Age Aging. 2005;34:218-227.
97. Lunney JR, Lynn J, Hogan C. Profiles of older Medicare decedents. J Am Geriatr Soc. 2002;50(6):1108-1112.
98. Spence A, Hasson F, Waldron M, et al. Professionals delivering palliative care to people with COPD: qualitative study. Palliat Med. 2009;23(2):126-131.
99. McClung J. End-of-life care in the treatment of heart failure in the elderly. Clin Geriatr Med. 2007;23:235-248.
100. Birch D, Draper J. A critical literature review exploring the challenges of delivering effective palliative care to older people with dementia. J Clin Nurs. 2008;17(9):1144-1163.
101. Kapo J, Morrison LJ, Liao S. Palliative care for the older adult. J Palliat Med. 2007;10(1):185-209.
102. Knaus WA, Harrerll FE, Lynn J et al. The SUPPORT prognostic model. Objective estimates of survival for seriously ill hospitalised adults. Ann Intern Med. 1995;122:191-203.
103. Pirovano M, Maltoni M, Nanni O, et al. A new palliative prognostic score: a first step for the staging of terminally ill cancer patients. J Pain Symptom Manage. 1999;17(4):231-239.
104. Morita T, Tsunoda J, Inoue S, et al. The Palliative Prognostic Index: a scoring system for survival prediction of terminally ill cancer patients. Support Care Cancer. 1999;7(3):128-133.
105. Adler ED, Goldfinger JZ, Park ME, Meier DE. Palliative care in the treatment of advanced heart failure. Circulation. 2009;120: 2597-2606.
106. Levy W, Mozaffarian D, Linker DT, et al. The Seattle Heart Failure model: prediction of survival in heart failure. Circulation. 2006;113:1424-1433.
107. Feliu J, Jimenez-Gordo AM, Madero R, et al. Development and validation of a prognostic nomogram for terminally ill cancer patients. J Natl Cancer Inst. 2011;103(21):1613-1620.
108. Mitchell SL, Miller SC, Teno JM, Kiely DK, Davis RB, Shaffer ML. Prediction of 6-month survival of nursing home residents with advanced dementia using ADEPT vs hospice eligibility guidelines. JAMA. 2010;304(17):1929-1935.
109. Han PK, Lee M, Reeve BB, et al. Development of a prognostic model for six-month mortality in older adults with declining health. J Pain Symptom Manage. 2012;43(3):527-539.
110. Institute for Clinical Systems Improvement. Health Care Guideline: Palliative Care. 3rd ed. Available at http://www.icsi.org/guidelines_and_more/gl_os_prot/other_health_care_conditions/palliative_care/palliative_care_11875.html. Last accessed May 16, 2012.
111. Bergman J, Saigal CS, Lorenz KA, et al. Hospice use and high-intensity care in men dying of prostate cancer. Arch Intern Med. 2011;171(3):204-210.
112. Haupt BJ. Characteristics of hospice care discharges and their length of service: United States, 2000. Vital Health Stat. 2003;(154): 1-36.
113. Teno JM, Shu JE, Casarett D, Spence C, Rhodes R, Connor S. Timing of referral to hospice and quality of care: length of stay and bereaved family members' perceptions of the timing of hospice referral. J Pain Symptom Manage. 2007;34(2):120-125.
114. Clayton JM, Hancock KM, Butow PN, et al. Clinical practice guidelines for communicating prognosis and end-of-life issues with adults in the advanced stages of a life-limiting illness, and their caregivers. Med J Aust. 2007;188(12 Suppl):S77, S79, S83-S108.
115. Weissman DE, Meier DE. Identifying patients in need of a palliative care assessment in the hospital setting: a consensus report from the Center to Advance Palliative Care. J Palliat Med. 2011;14(1):17-23.
116. Swetz KM, Freeman MR, AbouEzzeddine OF, et al. Palliative medicine consultation for preparedness planning in patients receiving left ventricular assist devices as destination therapy. Mayo Clin Proc. 2011;86(6):493-500.
117. Swetz KM, Ottenberg AL, Freeman MR, Mueller PS. Palliative care and end-of-life issues in patients treated with left ventricular assist devices as destination therapy. Curr Heart Fail Rep. 2011;8(3):212-218.
118. Renal Physicians Association. Shared Decision-Making in the Appropriate Initiation and Withdrawal from Dialysis. Clinical Practice Guideline. 2nd ed. Rockville, MD: Renal Physicians Association; 2010.
119. Lanken PN, Terry PB, Delisser HM, et al. An official American Thoracic Society clinical policy statement: palliative care for patients with respiratory diseases and critical illnesses. Am J Respir Crit Care Med. 2008;177(8):912-927.
120. Larson AM, Curtis JR. Integrating palliative care for liver transplant candidates: "too well for transplant, too sick for life."JAMA. 2006;295(108):2168-2176.
121. Center to Advance Palliative Care. 2011 Public Opinion Research on Palliative Care: A Report Based on Research by Public Opinion Strategies. New York, NY: Center to Advance Palliative Care; 2011.
122. Cambia Health Foundation. Living Well at the End of Life: A National Conversation: Results of 2011 Poll. Available at http://www.cambiahealthfoundation.org/nationalJournal.html. Last accessed May 16, 2012.
123. Davison SN, Jhangri GS. Impact of pain and symptom burden on the health-related quality of life of hemodialysis patients.J Pain Symptom Manage. 2010;39(3):477-485.
124. Naierman N. Debunking the Myths of Hospice. Available at http://www.americanhospice.org/articles-mainmenu-8/about-hospice-mainmenu-7/36-debunking-the-myths-of-hospice. Last accessed May 16, 2012.
125. McGorty E, Bornstein BH. Barriers to physicians' decisions to discuss hospice: insights gained from the United States hospice model. J Eval Clin Pract. 2003;9:363-372.
126. Balaban RB. A physician's guide to talking about end-of-life care. J Gen Intern Med. 2000;15:195-200.
127. Ngo-Metzger Q, August KJ, Srinivasan M, Liao S, Meyskens FL Jr. End-of-life care: guidelines for patient-centered communication. Am Fam Physician. 2008;77(2):167-174.
128. Larochelle MR, Rodriguez KL, Arnold RM, Barnato AE. Hospital staff attributions of the causes of physician variation in end-of-life treatment intensity. Palliat Med. 2009;23(5):460-470.
129. Office of Minority Health. Cultural Competency. Available at http://www.minorityhealth.hhs.gov/templates/browse.aspx?lvl=1&lvlID=3. Last accessed May 15, 2012.
130. Paez K, Allen JK, Beach MC, Carson KA, Cooper LA. Physician cultural competence and patient ratings of the patient-physician relationship. J Gen Intern Med. 2009;24(4):495-498.
131. Powers BJ, Trinh JV, Bosworth HB. Can this patient read and understand written health information? JAMA. 2010;304(1):76-84.
132. U.S. Census Bureau. Selected Social Characteristics in the United States: 2010. Available at http://factfinder2.census.gov/faces/tableservices/jsf/pages/productview.xhtml?pid=ACS_10_1YR_DP02&prodType=table. Last accessed May 16, 2012.
133. Karliner L, Napoles-Springer AM, Schillinger D, Bibbins-Domingo K, Pérez-Stable EJ. Identification of limited English proficient patients in clinical care. J Gen Intern Med. 2008;23(10):1555-1560.
134. Flores G. The impact of medical interpreter services on the quality of health care: a systematic review. Med Care Res Rev. 2005;62(3):255-299.
135. Karliner L, Jacobs EA, Chen AH, Mutha S. Do professional interpreters improve clinical care for patients with limited English proficiency? A systematic review of the literature. Health Serv Res. 2007;42(2):727-754.
136. Ngo-Metzger Q, Massagli MP, Clarridge BR, et al. Linguistic and cultural barriers to care: perspectives of Chinese and Vietnamese immigrants. J Gen Intern Med. 2003;18(1):44-52.
137. Norris WM, Wenrich MD, Nielsen EL, Treece PD, Jackson JC, Curtis JR. Communication about end-of-life care between language-discordant patients and clinicians: insights from medical interpreters. J Palliat Med. 2005;8(5):1016-1024.
138. Committee on Health Literacy, Board on Neuroscience and Behavioral Health. Health Literacy: A Prescription to End Confusion. Washington, DC: The National Academies Press; 2004.
139. Paasche-Orlow MK, Parker RM, Gazmararian JA, Nielsen-Bohlman LT, Rudd RR. The prevalence of limited health literacy.J Gen Intern Med. 2005;20(2):175-184.
140. Kutner M, Greenberg E, Jin,Y, Paulsen C, White S. The Health Literacy of America's Adults: Results from the 2003 National Assessment of Adult Literacy. Washington, DC: National Center for Education Statistics; 2006.
141. Shah LC, West P, Bremmeyr K, Savoy-Moore RT. Health literacy instrument in family medicine: the "newest vital sign" ease of use and correlates. J Am Board Fam Med. 2010;23(2):195-203.
142. Weiss B, Mays MZ, Martz W, et al. Quick assessment of literacy in primary care: the newest vital sign. Ann Fam Med. 2005;3: 514-522.
143. Crawley LM, Marshall PA, Lo B, Koenig BA, for the End-of-Life Care Consensus Panel. Strategies for culturally effective end-of-life care. Ann Intern Med. 2002;136(9):673-679.
144. Shanawani H, Wenrich MD, Tonelli MR, Curtis JR. Meeting physicians' responsibilities in providing end-of-life care. Chest. 2008;133(3):775-786.
145. Quill TE. Initiating end-of-life discussions with seriously ill patients: addressing the "elephant in the room." JAMA. 2000;284(19):2502-2507.
146. Parker SM, Clayton JM, Hancock K, et al. A systematic review of prognostic/end-of-life communication with adults in the advanced stages of a life-limiting illness: patient/caregiver preferences for the content, style, and timing of information n.J Pain Symptom Manage. 2007;34(1):81-93.
147. Werb R. Palliative care in the treatment of end-stage renal failure. Prim Care Clin Office Pract. 2011;38:299-309.
148. Mack JW, Weeks JC, Wright AA, Block SD, Prigerson HG. End-of-life discussions, goal attainment, and distress at the end of life: predictors and outcomes of receipt of care consistent with preferences. J Clin Oncol. 2010;28(7):1203-1208.
149. Huskamp HA, Keating NL, Malin JL, et al. Discussions with physicians about hospice among patients with metastatic lung cancer. Arch Intern Med. 2009;169(10):954-962.
150. Curtis J, Engelberg RA, Nielsen EL, et al. Patient-physician communication about end-of-life care for patients with severe COPD. Eur Respir J. 2004;24(2):200-205.
151. Reinke LF, Slatore CG, Uman J, et al. Patient-clinician communication about end-of-life care topics: is anyone talking to patients with chronic obstructive pulmonary disease? J Palliat Med. 2011;14(8):923-928.
152. Halliwell J, Mulcahy P, Buetow S, Bray Y, Coster G, Osman LM. GP discussion of prognosis with patients with severe chronic obstructive pulmonary disease: a qualitative study. Br J Gen Pract. 2004;54(509):904-908.
153. Keating NL, Landrum MB, Rogers SO Jr, et al. Physician factors associated with discussions about end-of-life care. Cancer. 2010;116(4):998-1006.
154. Jessup M, Abraham WT, Casey DE, et al. 2009 focused update: ACCF/AHA guidelines for the diagnosis and management of heart failure in adults. Circulation. 2009;119:1977-2016.
155. Hancock K, Clayton JM, Parker SM, et al. Truth-telling in discussing prognosis in advanced life-limiting illnesses: a systematic review. Palliat Med. 2007;21(6):507-517.
156. Wailing A, Lorenz KA, Dy SM, et al. Evidence-based recommendations for information and care planning in cancer care.J Clin Oncol. 2008;26(23):3896-3902.
157. Buckman R. Breaking bad news: why is it still so difficult? Br Med J. 1984;288:1597-1599.
158. Baile WF, Buckman R, Lenzi R, Glober G, Beale EA, Kudelka AP. SPIKES—A six-step protocol for delivering bad news: application to the patient with cancer. Oncologist. 2000;5:302-311.
159. Buckman R. Communication skills in palliative care: a practical guide. Neurol Clin. 2001;19(4):989-1004.
160. Back AL, Arnold RM, Baile WF, Tulsky JA, Fryer-Edwards K. Approaching difficult communication tasks in oncology.CA Cancer J Clin. 2005;55:164-177.
161. Hancock K, Clayton JM, Parker SM, et al. Discrepant perceptions about end-of-life communication: a systematic review.J Pain Symptom Manage. 2007;34(2):190-200.
162. Fellowes D, Wilkinson S, Moore P. Communication skills training for health care professionals working with cancer patients, their families and/or carers. Cochrane Database Syst Rev. 2004;(2):CD003751.
163. Back AL, Arnold RM, Baile WF, et al. Efficacy of communication skills training for giving bad news and discussing transitions to palliative care. Arch Intern Med. 2007;167(5):453-460.
164. Clayton JM, Butow PN, Tattersall MNH, et al. Randomized controlled trial of a prompt list to help advanced cancer patients and their caregivers to ask questions about prognosis and end-of-life care. J Clin Oncol. 2007;25(6):715-723.
165. Au DH, Udris EM, Engelberg RA, et al. A randomized trial to improve communication about end-of-life care among patients with COPD. Chest. 2012;141(3):726-735.
166. Brink P, Smith TF, Kitson M. Determinants of do-not-resuscitate orders in palliative home care. J Palliat Med. 2008;11(2):226-232.
167. Swetz KM, Ottenberg AL, Freeman MR, Mueller PS. Palliative care and end-of-life issues in patients treated with left ventricular assist devices as destination therapy. Curr Heart Fail Rep. 2011;8(3):212-218.
168. Swetz KM, Freeman MR, AbouEzzeddine OF, et al. Palliative medicine consultation for preparedness planning in patients receiving left ventricular assist devices as destination therapy. Mayo Clin Proc. 2011;86(6):493-500.
169. O'Connor NR, Kumar P. Conservative management of end-stage renal disease without dialysis: a systematic review. J Palliat Med. 2012;15(2):228-235.
170. Stapleton R, Curtis JR. End-of-life considerations in older patients who have lung disease. Clin Chest Med. 2007;28:801-811.
171. Audrey S, Abel J, Blazeby JM, Falk S, Campbell R. What oncologists tell patients about survival benefits of palliative chemotherapy and implications for informed consent: qualitative study. BMJ. 2008;337:a752.
172. Goldstein N, Lampert R, Bradley E, et al. Management of implantable cardioverter defibrillators in end-of-life care. Ann Intern Med. 2004;141:835-838.
173. Smith TJ, Hillner BE. Bending the cost curve in cancer care. N Engl J Med. 2011;364(21):2060-2065.
174. Weeks JC, Cook EF, O'Day SJ, et al. Relationship between cancer patients' predictions of prognosis and their treatment preferences. JAMA. 1998;279(21):1709-1714.
175. Sheffield KM, Boyd CA, Benarroch-Gampel J, Kuo YF, Cooksley CD, Riall TS. End-of-life care in Medicare beneficiaries dying with pancreatic cancer. Cancer. 2011;117(21):5003-5012.
176. Unroe KT, Greiner MA, Hernandez AF, et al. Resource use in the last 6 months of life among Medicare beneficiaries with heart failure, 2000–2007. Arch Intern Med. 2011;171(3):196-203.
177. Earle C, Neville BA, Landrum MB, et al. Trends in the aggressiveness of cancer care near the end of life. J Clin Oncol. 2004;22: 315-321.
178. Kao S, Shafiq J, Vardy J, Adams D. Use of chemotherapy at end of life in oncology patients. Ann Oncol. 2009;20(9):1555-1559.
179. Saito AM, Landrum MB, Neville BA, Zyanian JZ, Earle CC. The effect on survival of continuing chemotherapy to near death. BMC Palliat Care. 2011;10:14.
180. Curtis JR, Engelberg RA. What is the "right" intensity of care at the end of life and how do we get there? [editorial]. Ann Intern Med. 2011;154(4):283-284.
181. Wright AA, Zhang B, Ray A, et al. Associations between end-of-life discussions, patient mental health, medical care near death, and caregiver bereavement adjustment. JAMA. 2008;300(14):1665-1673.
182. Smith L. Guidelines for delivering quality palliative care. Am Fam Physician. 2006;73(6):1105-1111.
183. National Comprehensive Cancer Network. NCCN Clinical Practice Guidelines in Oncology. Available at http://www.nccn.org/professionals/physician_gls/f_guidelines.asp. Last accessed May 16, 2012.
184. McCabe M, Storm C. When doctors and patients disagree about medical futility. J Oncol Pract. 2008;4(4):207-209.
185. Smith CB, Bunch O'Neill L. Do not resuscitate does not mean do not treat: how palliative care and other modalities can help facilitate communication about goals of care in advanced illness. Mt Sinai J Med. 2008;75(5):460-465.
186. Lee D, Austin PC, Stukel TA, et al. "Dose-dependent" impact of recurrent cardiac events on mortality in patients with heart failure. Am J Med. 2009;122(2):162-169.
187. Holley J. Palliative care in end-stage renal disease: illness trajectories, communication, and hospice use. Adv Chronic Kidney Dis. 2007;14(4):402-408.
188. Peppercorn JM, Smith TJ, Helft PR, et al. American Society of Clinical Oncology statement: toward individualized care for patients with advanced cancer. J Clin Oncol. 2011;29(6):755-760.
189. Torke AM, Siegler M, Abalos A, Moloney RM, Alexander GC. Physicians' experience with surrogate decision making for hospitalized adults. J Gen Intern Med. 2009;24(9):1023-1028.
190. Silveira MJ, Kim SY, Langa KM. Advance directives and outcomes of surrogate decision making before death. N Engl J Med. 2010;362(13):1211-1218.
191. Nicholas LN, Langa KM, Iwashyna TJ, Weir DR. Regional variation in the association between advance directives and end-of-life Medicare expenditures. JAMA. 2011;306(13):1447-1453.
192. Teno JM, Licks S, Lynn J, et al. Do advance directives provide instructions that direct care? SUPPORT Investigators. Study to Understand Prognoses and Preferences for Outcomes and Risks of Treatment. J Am Geriatr Soc. 1997;45(4):508-512.
193. Olick RS. Defining features of advance directives in law and clinical practice. Chest. 2012;14(1):232-238.
194. Dula A, Williams S. When race matters. Clin Geriatr Med. 2005;21:239-253.
195. Reynolds K, Hanson LC, Henderson M, Steinhauser KE. End-of-life care in nursing home settings: do race or age matter?Palliat Support Care. 2008;6(1):21-27.
196. Kwak J, Haley WE. Current research findings on end-of-life decision making among racially or ethnically diverse groups. Gerontologist. 2005;45(5):634-641.
197. Sanchez W, Walwalkar JA. Palliative care for patients with end-stage liver disease ineligible for liver transplanation. Gastroenterol Clin North Am. 2006;35(1):201-219.
198. Stein G, Bonuck KA. Attitudes on end-of-life care and advance care planning in the lesbian and gay community. J Palliat Med. 2001;4(2):173-190.
199. Center for Ethics in Health Care, Oregon Health and Science University. Physician Orders for Life-Sustaining Treatment. Available at http://www.ohsu.edu/polst/programs/state+programs.htm. Last accessed May 16, 2012.
200. Hickman SE, Nelson CA, Moss AH, et al. Use of the Physicians Orders for Life-Sustaining Treatment (POLST) paradigm program in the hospice setting. J Palliat Med. 2009;12(2):133-141.
201. Fromme EK, Zive D, Schmidt TA, Olszewski E, Tolle SW. POLST registry do-not-resuscitate orders and other patient treatment preferences [research letter]. JAMA. 2012;307(1):34-35.
202. Doyle D, Woodruff R. The IAHPC Manual of Palliative Care. 2nd ed. Houston, TX: International Association for Hospice and Palliative Care Press; 2008.
203. Mast K, Salama M, Silverman GK, Arnold RM. End-of-life content in treatment guidelines for life-limiting diseases. J Palliat Med. 2004;7(6):754-763.
204. Blinderman CD, Homel P, Billings JA, Portenoy RK, Tennstedt SL. Symptom distress and quality of life in patients with advanced congestive heart failure. J Pain Symptom Manage. 2008;35(6):594-603.
205. Bekelman DB, Rumsfeld JS, Havranek EP, et al. Symptom burden, depression, and spiritual well-being: a comparison of heart failure and advanced cancer patients. J Gen Intern Med. 2009;24(5):592-598.
206. Blinderman CD, Homel P, Billings JA, Tennstedt S, Portenoy RK. Symptom distress and quality of life in patients with advanced chronic obstructive pulmonary disease. J Pain Symptom Manage. 2009;38(1):115-123.
207. Murtagh F, Addington-Hall JM, Higginson IJ. The prevalence of symptoms in end-stage renal disease: a systematic review.Adv Chronic Kidney Dis. 2007;14(1):82-99.
208. Solano J, Gomes B, Higginson IJ. A comparison of symptom prevalence in far advanced cancer, AIDS, heart disease, chronic obstructive pulmonary disease and renal disease. J Pain Symptom Manage. 2006;31:58-69.
209. Lorenz KA, Lynn J, Dy SM, et al. Evidence for improving palliative care at the end of life: a systematic review. Ann Intern Med. 2008;148(2):147-159.
210. Mahler DA, Selecky PA, Harrod CG, et al. American College of Chest Physicians consensus statement on the management of dyspnea in patients with advanced lung or heart disease. Chest. 2010;137(3):674-691.
211. Douglas C, Murtagh FE, Chambers EJ, Howse M, Ellershaw J. Symptom management for the adult patient dying with advanced chronic kidney disease: a review of the literature and development of evidence-based guidelines by a United Kingdom Expert Consensus Group. Palliat Med. 2009;23(2):103-110.
212. Shah M, Quill T, Norton S, Sada Y, Buckley M, Fridd C. "What bothers you the most?" Initial responses from patients receiving palliative care consultation. Am J Hosp Palliat Care. 2008;25(2):88-92.
213. Esther Kim J-E, Dodd MJ, Aouizerat BE, Jahan T, Miaskowski C. A review of the prevalence and impact of multiple symptoms in oncology patients. J Pain Symptom Manage. 2009;37(4):715-736.
214. Teno J. TIME: Toolkit of Instruments to Measure End-of-Life Care. Available at http://www.chcr.brown.edu/pcoc/toolkit.htm. Last accessed May 16, 2012.
215. Mularski RA, Dy SM, Shugarman LR, et al. A systematic review of measures of end-of-life care and its outcomes. Health Serv Res. 2007;42(5):1848-1870.
216. Hoekstra J, Vernooij-Dassen MJ, de Vos R, Bindels PJ. The added value of assessing the "most troublesome" symptom among patients with cancer in the palliative phase. Patient Educ Couns. 2007;65(2):223-229.
217. Homsi J, Walsh D, Rivera N, et al. Symptom evaluation in palliative medicine: patient report vs. systematic assessment.Support Care Cancer. 2006;14(5):444-453.
218. American Pain Foundation. Breakthrough cancer pain: mending the break in the continuum of care. J Pain and Palliat Care Pharmacother. 2011;25(3):252-264.
219. Dalal S, Bruera E. Assessment and management of pain in the terminally ill. Prim Care Clin Office Pract. 2011;38:195-223.
220. Gao W, Gulliford M, Higginson IJ. Prescription patterns of analgesics in the last 3 months of life: a retrospective analysis of 10,202 lung cancer patients. Br J Cancer. 2011;104(11):1704-1710.
221. Godfrey C, Harrison MB, Medves J, Tranmer JE. The symptom of pain with heart failure: a systematic review. J Card Fail. 2006;12(4):307-313.
222. Breuer B, Fleishman SB, Cruciani RA, Portenoy RK. Medical oncologists' attitudes and practice in cancer pain management: a national survey. J Clin Oncol. 2011;[Epub ahead of print].
223. Abrahm JL. A Physician's Guide to Pain and Symptom Management in Cancer Patients. 2nd ed. Baltimore, MD: Johns Hopkins University Press; 2005.
224. D'Arcy Y. Nursing2008 pain management survey report. Nursing 2008. 2008;38(6):42-49.
225. Zerzan J, Lee CA, Haverhals LM, Nowels CT. Exploring physician decisions about end-of-life opiate prescribing: a qualitative study. J Palliat Med. 2011;14(5):567-572.
226. Fineberg IC, Wenger NS, Brown-Saltzman K. Unrestricted opiate administration for pain and suffering at the end of life: knowledge and attitudes as barriers to care. J Palliat Med. 2006;9(4):873-883.
227. Mezei L, Murinson BB, Johns Hopkins Pain Curriculum Development Team. Pain education in North American medical schools.J Pain. 2011;12(12):1199-1208.
228. Schmidt C. Experts worry about chilling effect of federal regulations on treating pain. J Natl Cancer Inst. 2005;97(8):554-555.
229. American Medical Association. AMA Code of Medical Ethics. Opinion 2.20: Withholding or Withdrawing Life-Sustaining Medical Treatment. Available at http://www.ama-assn.org/ama/pub/physician-resources/medical-ethics/code-medical-ethics/opinion220.page. Last accessed May 2, 2012.
230. American Medical Association. AMA Statement on End-of-Life Care. Available at http://www.ama-assn.org/ama/pub/physician-resources/medical-ethics/about-ethics-group/ethics-resource-center/end-of-life-care/ama-statement-end-of-life-care.page. Last accessed May 2, 2012.
231. Bagdasarian N. A prescription for mental distress: the principles of psychosomatic medicine with the physical manifestation requirement in NIED cases. Am J Law Med. 2000;26:401-438.
232. Goudas LC, Bloch R, Gialeli-Goudas M, Lau J, Carr DB. The epidemiology of cancer pain. Cancer Invest. 2005;23:182-190.
233. Desbiens NA, Mueller-Rizner N. How well do surrogates assess the pain of seriously ill patients? Crit Care Med. 2000;28(5): 1347-1352.
234. Portenoy RK, Hagen NA. Breakthrough pain: definition, prevalence and characteristics. Pain. 1990;41:273-281.
235. Hockenberry M, Wilson D. Wong's Essentials of Pediatric Nursing. 8th ed. St. Louis, MO: Mosby; 2008.
236. Fishman B, Pasteranak S, Wallenstien SL, Houde RW, Holland JC, Foley KM. The Memorial Pain Assessment Card: a valid instrument for the evaluation of cancer pain. Cancer. 1987;60:1151-1158.
237. Kvale PA, Selecky PA, Prakash UBS. Palliative care in lung cancer: ACCP evidence-based clinical practice guidelines (2nd edition). Chest. 2007;132(3 suppl):368S-403S.
238. World Health Organization. Cancer Pain Relief. 2nd ed. Geneva: World Health Organization, 1996.
239. Pergolizzi JV Jr, Mercadante S, Echaburu AV, et al. The role of transdermal buprenorphine in the treatment of cancer pain: an expert panel consensus. Curr Med Res Opin. 2009;25(6):1517-1528.
240. U.S. Food and Drug Administration. Drug Approvals and Database. Available at http://www.fda.gov/Drugs/InformationOnDrugs/default.htm. Last accessed May 16, 2012.
241. Swetz KM, Kamal AH. Palliative care. Ann Intern Med. 2012;156(3):ITC21.
242. Argoff CE, Silvershein DI. A comparison of long- and short-acting opioids for the treatment of chronic noncancer pain: tailoring therapy to meet patient needs. Mayo Clin Proc. 2009;84(7):602-612.
243. Scottish Partnership for Palliative Care. NHS Lothian Palliative Care Guidelines. 3rd ed. Scotland: National Health Service Scotland; 2009.
244. Lauretti GR, Oliveira GM, Pereira NL. Comparison of sustained-release morphine with sustained-release oxycodone in advanced cancer patients. Br J Cancer. 2003;89:2027-2030.
245. McCleane G, Smith HS. Opioids for persistent noncancer pain. Anesthesiol Clin. 2007;25:787-807.
246. Shaiova L, Berger A, Blinderman CD, et al. Consensus guideline on parenteral methadone use in pain and palliative care.Palliat Support Care. 2008;6(2):165-176.
247. Nicholson AB. Methadone for cancer pain. Cochrane Database Syst Rev. 2007;(4):CD003971.
248. Dart RC, Woody GE, Kleber HD. Prescribing methadone as an analgesic [letter]. Ann Intern Med. 2005;143:620.
249. Vadivelu N, Timchenko A, Huang Y, Sinatra R. Tapentadol extended-release for treatment of chronic pain: a review. J Pain Res. 2011;4:211-218.
250. Blick SK, Wagstaff AJ. Fentanyl buccal tablet: in breakthrough pain in opioid-tolerant patients with cancer. Drugs. 2006;66(18):2387-2395.
251. Messina J, Darwish M, Fine PG. Fentanyl buccal tablet. Drugs Today (Barc). 2008;44(1):41-54.
252. Skaer T. Transdermal opioids for cancer pain. Health Qual Life Outcomes. 2006;4:24.
253. Fallon MT, Laird BJA. A systematic review of combination step III opioid therapy in cancer pain: an EPCRC opioid guideline project. Palliat Med. 2011;25(5):597-603.
254. Saarto T, Wiffen PJ. Antidepressants for neuropathic pain. Cochrane Database Syst Rev. 2007;(4):CD005454.
255. Dolinsky C, Metz JM. Palliative radiation therapy in oncology. Anesthesiol Clin North Am. 2006;24:113-128.
256. Fine PG. Palliative radiation therapy in end-of-life care: evidence-based utilization. Am J Hospice Palliat Med. 2002;19(3):166-170.
257. Berrang T, Samant R. Palliative radiotherapy knowledge among community family physicians and nurses. J Cancer Educ. 2008;23(3):156-160.
258. Tilden VP, Drach LL, Tolle SW. Complementary and alternative therapy use at end-of-life in community settings. J Altern Complement Med. 2004;10(5):811-817.
259. Demmer C. A survey of complementary therapy services provided by hospices. J Palliat Med. 2004;7(4):510-516.
260. Kozak LE, Kayes L, McCarty R, et al. Use of complementary and alternative medicine (CAM) by Washington State hospices.Am J Hosp Palliat Care. 2008;25(6):463-468.
261. Polubinski JP, West L. Implementation of a massage therapy program in the home hospice setting. J Pain Symptom Manage. 2005;30:104-106.
262. Choi TY, Lee MS, Kim TH, Zaslawski C, Ernst E. Acupuncture for the treatment of cancer pain: a systematic review of randomised clinical trials. Support Care Cancer. 2012;20(6):1147-1158.
263. Raub JA. Psychophysiologic effects of hatha yoga on musculoskeletal and cardiopulmonary function: a literature review.J Altern Complement Med. 2002;8:797-812.
264. Cepeda MS, Carr DB, Lau J, Alvarez H. Music for pain relief. Cochrane Database System Rev. 2006;(2):CD004843.
265. Reynolds MW, Nabors L, Quinlan A. The effectiveness of art therapy: does it work? J Am Art Ther Assoc. 2000;17:207-213.
266. Radbruch L, Strasser F, Elsner F, et al. Fatigue in palliative care patients—an EAPC approach. Palliat Med. 2008;22(1):13-32.
267. Passik SD, Kirsh KL, Donaghy K et al. Patient-related barriers to fatigue communication: initial validation of the fatigue management barriers questionnaire. J Pain Symptom Manage. 2002;24: 481-493.
268. Agency for Healthcare Research and Quality. Management of Cancer Symptoms: Pain, Depression, and Fatigue. Evidence Report/Technology Assessment No. 61. Rockville, MD: U.S. Department of Health and Human Services, 2002.
269. Fleishman SB. Treatment of symptom clusters: pain, depression, and fatigue. J Natl Cancer Inst Monogr. 2004;(32):119-23.
270. Barsevick A. The elusive concept of the symptom cluster. Oncol Nurse Forum. 2007;34(5):971-980.
271. Walsh D, Rybicki L. Symptom clustering in advanced cancer. Support Care Cancer. 2006;14(8):831-836.
272. Ross D, Alexander CS. Management of common symptoms in terminally ill patients: part I. Fatigue, anorexia, cachexia, nausea and vomiting. Am Fam Physician. 2001;64:807-814.
273. Harris J. Fatigue in chronically ill patients. Curr Opin Support Palliat Care. 2008;2(3):180-186.
274. Curt G, Breitbart W, Cella D, et al. Impact of cancer-related fatigue on the lives of patients: new findings from the Fatigue Coalition. Oncologist. 2000;5(5):353-360.
275. Mendoza T, Wang XS, Cleeland CS, et al. The rapid assessment of fatigue severity in cancer patients: use of the Brief Fatigue Inventory. Cancer. 1999;85:1186-1196.
276. Minton O, Stone P, Richardson A, Sharpe M, Hotopf M. Drug therapy for the management of cancer related fatigue.Cochrane Database Syst Rev. 2008;(1):CD006704.
277. Minton O, Richardson A, Sharpe M, Hotopf M, Stone P. Drug therapy for the management of cancer-related fatigue.Cochrane Database Syst Rev. 2010;(7):CD006704.
278. Peuckmann-Post V, Elsner F, Krumm N, Trottenberg P, Radbruch L. Pharmacological treatments for fatigue associated with palliative care. Cochrane Database System Rev. 2010;(11):CD006788.
279. Cramp F, Daniel J. Exercise for the management of cancer-related fatigue in adults. Cochrane Database of Syst Rev. 2008;(2):CD006145.
280. Volberding PA, Levine AM, Dieterich D, et al. Anemia in HIV infection: clinical impact and evidence-based management strategies. Clin Infect Dis. 2004;38:1454-1463.
281. Schmidt RJ, Dalton CL. Treating anemia of chronic kidney disease in the primary care setting: cardiovascular outcomes and management recommendations. Osteopath Med Primary Care. 2007;1:14.
282. Ngo K, Kotecha D, Walters JA, et al. Erythropoiesis-stimulating agents for anaemia in chronic heart failure patients.Cochrane Database Syst Rev. 2010;(1):CD007613.
283. National Kidney Foundation, Kidney Disease Outcomes Quality Initiative. KDOQI Clinical Practice Guideline and Clinical Practice Recommendations for Anemia in Chronic Kidney Disease: 2007 Update of Hemoglobin Target. Available at http://www.kidney.org/professionals/kdoqi/guidelines_anemiaUP/guide1.htm. Last accessed May 16, 2012.
284. Rizzo JD, Brouwers M, Hurley P, et al. American Society of Clinical Oncology/American Society of Hematology clinical practice guideline update on the use of epoetin and darbepoetin in adult patients with cancer. J Clin Oncol. 2010;28(33):4996-5010.
285. Porock K, Kristjanson LJ, Tinnelly K, et al. An exercise intervention for advanced cancer patients experiencing fatigue: a pilot study. J Palliat Care. 2000;16:30-36.
286. Segal R, Reid RD, Courneya KS, et al. Resistance exercise in men receiving androgen deprivation therapy for prostate cancer.J Clin Oncol. 2003;21:1653-1659.
287. Oldervoll L, Loge JH, Paltiel H, et al. The effect of a physical exercise program in palliative care: a phase II study. J Pain Symptom Manage. 2006;31:421-430.
288. Rabkin JG, McElihiney MC, Rabkin R, McGrath PJ. Modafinil treatment for fatigue in HIV/AIDS: a randomized placebo-controlled study. J Clin Psychiatry. 2010;71(6):707-715.
289. Bausewein C, Farquhar M, Booth S, Gysels M, Higginson IJ. Measurement of breathlessness in advanced disease: a systematic review. Respir Med. 2007;101(3):399-410.
290. Edmonds P, Karlsen S, Khan S, Addington-Hall J. A comparison of the palliative care needs of patients dying from chronic respiratory diseases and lung cancer. Palliat Med. 2001;15(4):287-295.
291. Borg G. Borg's Perceived Exertion and Pain Scales. Champaign, IL: Human Kinetics; 1998.
292. Galbraith S, Fagan P, Perkins P, Lynch A, Booth S. Does the use of a handheld fan improve chronic dyspnea? A randomized, controlled, crossover trial. J Pain Symptom Manage. 2010;39(5):831-838.
293. Bausewein C, Booth S, Gysels M, Higginson I. Non-pharmacological interventions for breathlessness in advanced stages of malignant and non-malignant diseases. Cochrane Database Syst Rev. 2008;(2):CD005623.
294. Jennings AL, Davies AN, Higgins JP, et al. A systematic review of the use of opioids in the management of dyspnoea. Thorax. 2002;57:939-944.
295. Clemens KE, Klaschik E. Effect of hydromorphone on ventilation in palliative care patients with dyspnea. Support Care Cancer. 2008;16(1):93-99.
296. Simon ST, Higginson IJ, Booth S, H arding R, Bausewein C. Benzodiazepines for the relief of breathlessness in advanced malignant and non-malignant diseases in adults. Cochrane Database Syst Rev. 2010;(1):CD007354.
297. Larkin PJ, Sykes NP, Centeno C, et al. The management of constipation in palliative care: clinical practice recommendations. Palliat Med. 2008;22(7):796-807.
298. Librach SL, Bouvette M, De Angelis C, et al. Consensus recommendations for the management of constipation in patients with advanced, progressive illness. J Pain Symptom Manage. 2010;40(5):761-773.
299. Miles C, Fellowes, D, Goodman ML, Wilkinson S. Laxatives for the management of constipation in palliative care patients. Cochrane Database Syst Rev. 2006;(4):CD003448.
300. American College of Gastroenterology Chronic Constipation Task Force. An evidence-based approach to the management of chronic constipation in North America. Am J Gastroenterol. 2005;100:S1-S22.
301. Hawley PH, Byeon JJ. A comparison of sennosides-based bowel protocols with and without docusate in hospitalized patients with cancer. J Palliat Med. 2008;11(4):575-581.
302. Candy B, Jones L, Goodman ML, Drake R, Tookman A. Laxatives or methylnaltrexone for the management of constipation in palliative care patients. Cochrane Database System Rev. 2011;(1):CD003448.
303. Wood GJ, Shega JW, Lynch B, Von Roenn JH. Management of intractable nausea and vomiting in patients at the end of life. JAMA. 2007;298(10):1196-1207.
304. Glare P, Miller J, Nikolova T, Tickoo R. Treating nausea and vomiting in palliative care: a review. Clin Intervent Aging. 2011;6: 243-259.
305. Basch E, Prestrud AA, Hesketh PJ, et al. Antiemetics: American Society of Clinical Oncology clinical practice guideline update.J Clin Oncol. 2011;29(31):4189-4198.
306. Davis MP, Hallerberg G, Palliative Medicine Study Group of the Multinational Association of Supportive Care in Cancer. A systematic review of the treatment of nausea and/or vomiting in cancer unrelated to chemotherapy or radiation.J Pain Symptom Manage. 2010;39(4):756-767.
307. Glare P, Pereira G, Kristjanson LJ, Stockler M, Tattersall M. Systematic review of the efficacy of antiemetics in the treatment of nausea in patients with far-advanced cancer. Support Care Cancer. 2004;12(6):432-440.
308. Glare P, Dunwoodie D, Clark K, et al. Treatment of nausea and vomiting in terminally ill cancer patients. Drugs. 2008;68(18): 2575-2590.
309. Pommer E. Olanzapine: palliative medicine update. Am J Hosp Palliat Care. 2012; [Epub ahead of print].
310. Donohoe CL, Ryan AM, Reynolds JV. Cancer cachexia: mechanisms and clinical implications. Gastroenterol Res Pract. 2011:601434.
311. Evans WJ, Morley JE, Argiles J, et al. Cachexia: a new definition. Clin Nutr. 2008;27(6):793-799.
312. Mangili A, Murman DH, Zampini AM, Wanke CA, Mayer KH. Nutrition and HIV infection: review of weight loss and wasting in the era of highly active antiretroviral therapy from the Nutrition for Healthy Living Cohort. Clin Infect Dis. 2006;42(6): 836-842.
313. von Haehling S, Anker SD. Cachexia as a major underestimated and unmet medical need: facts and numbers. J Cachexia Sarcopenia Muscle. 2010;1:1-5.
314. Fearon K, Strasser F, Anker SD, et al. Definition and classification of cancer cachexia: an international consensus. Lancet Oncol. 2011;12(5):489-495.
315. Springer J, Filippatos G, Akashi YJ, Anker SD. Prognosis and therapy approaches of cardiac cachexia. Curr Opin Cardiol. 2006;21(3):229-233.
316. Consensus Development Panel Meeting. Treatment Guidelines for HIV-Associated Wasting. New York, NY: 2000.
317. Lasheen W, Walsh D. The cancer anorexia-cachexia syndrome: myth or reality? Support Care Cancer. 2010;18(2):265-272.
318. Yavuzsen T, Walsh D, Davis MP, et al. Components of the anorexia-cachexia syndrome: gastrointestinal symptom correlates of cancer anorexia. Support Care Cancer. 2009;17(12):1531-1541.
319. Kalantar-Zadeh K, Anker SD, Horwich TB, Fonarow GC. Nutritional and anti-inflammatory interventions in chronic heart failure. Am J Cardiol. 2008;101(11A):89E-103E.
320. Polsky B, Kotler D, Steinhart C. Treatment guidelines for HIV-associated wasting. HIV Clin Trials. 2004;5(1):50-61.
321. Sarhill N, Mahmoud F. Hiccups and other gastrointestinal symptoms. In: Berger AM, Shuster JL, Von Roenn JH (eds). Principles and Practice of Palliative Care and Supportive Oncology. 3rd ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2007: 193-204.
322. Bosaeus I. Nutritional support in multimodal therapy for cancer cachexia. Support Care Cancer. 2008;16(5):447-451.
323. Berenstein EG, Ortiz Z. Megestrol acetate for the treatment of anorexia-cachexia syndrome. Cochrane Database Syst Rev. 2005;(2):CD004310.
324. Moyle GJ, Schoelles K, Fahrback K, et al. Efficacy of selected treatments of HIV wasting: a systematic review and meta-analysis.J Acquir Immune Defic Syndr. 2004;37(Suppl 5):S262-S276.
325. Gelato M, McNurlan M, Freedland E. Role of recombinant human growth hormone in HIV-associated wasting and cachexia: pathophysiology and rationale for treatment. Clin Ther. 2007;29(11):2269-2288.
326. Gullett NP, Hebbar G, Ziegler TR. Update on clinical trials of growth factors and anabolic steroids in cachexia and wasting.Am J Clin Nutr. 2010;91(4):1143S-1147S.
327. Grunfeld C, Kotler DP, Dobs A, Glesby M, Bhasin S. Oxandrolone in the treatment of HIV-associated weight loss in men: a randomized, double-blind, placebo-controlled study. J Acquir Immune Defic Syndr. 2006;41(3):304-314.
328. Yeh SS, DeGuzman B, Kramer T, M012 Study Group. Reversal of COPD-associated weight loss using the anabolic agent oxandrolone. Chest. 2002;122(2):421-428.
329. Orr R, Fiatarone Singh M. The anabolic androgenic steroid oxandrolone in the treatment of wasting and catabolic disorders: review of efficacy and safety. Drugs. 2004;64(7):725-750.
330. DeBoer MD. Emergence of ghrelin as a treatment for cachexia syndromes. Nutrition. 2008;24(9):806-814.
331. Beckwith MC. Diarrhea in palliative care patients. In: Lipman AG, Jackson KC II, Tyler LS (eds). Evidence-Based Symptom Control in Palliative Care: Systematic Reviews and Validated Clinical Practice Guidelines for 15 Common Problems in Patients with Life-Limiting Disease. Binghamton, NY: Haworth Press; 2000: 99-101.
332. Alderman J. Fast Facts and Concepts #96: Diarrhea in Palliative Care, 2nd edition. Available at http://www.eperc.mcw.edu/fastfact/ff_096.htm. Last accessed May 2, 2012.
333. American Gastroenterological Association. American Gastroenterological Association medical position statement: guidelines for the evaluation and management of chronic diarrhea. Gastroenterology. 1999;116:1461-1463.
334. Allen SJ, Martinez EG, Gregorio GV, Dans LF. Probiotics for treating acute infectious diarrhea. Cochrane Database Syst Rev. 2010;(11):CD003048.
335. Schutte-Rodin S, Broch L, Buysse D, Dorsey C, Sateia M. Clinical guideline for the evaluation and management of chronic insomnia in adults. J Clin Sleep Med. 2008;4(5):487-504.
336. Hajjar RR. Sleep disturbance in palliative care. Clin Geriatr Med. 2008;24(1):83-91.
337. Hugel H, Ellershaw JE, Cook L, Skinner J, Irvine C. The prevalence, key causes and management of insomnia in palliative care patients. J Pain Symptom Manage. 2004;27(4):316-321.
338. Shayamsunder AK, Patel SS, Jain V, Peterson RA, Kimmel PL. Sleepiness, sleeplessness, and pain in end-stage renal disease: distressing symptoms for patients. Semin Dial. 2005;18:109-118.
339. Earley CJ. Restless legs syndrome. N Engl J Med. 2003; 348:2103-2109.
340. Savard J, Marin CM. Insomnia in the context of cancer: a review of a neglected problem. J Clin Oncol. 2001;19:895-908.
341. Johns MW. A new method for measuring daytime sleepiness: the Epworth sleepiness scale. Sleep. 1991;14:54-545.
342. Morgenthaler T, Kramer M, Alessi C, et al. Practice parameters for the psychological and behavioral treatment of insomnia: an update. An American Academy of Sleep Medicine report. Sleep. 2006;29(11):1415-1419.
343. Tariq SH, Pulisetty S. Pharmacotherapy for insomnia. Clin Geriatr Med. 2008;24(1):93-105.
344. Hirst A, Sloan R. Benzodiazepines and related drugs for insomnia in palliative care. Cochrane Database Syst Rev. 2002;(4):CD003346.
345. Scholz H, Trenkwalder C, Kohnen R, Riemann D, Kriston L, Hornyak M. Dopamine agonists for restless legs syndrome.Cochrane Database Syst Rev. 2011;(3):CD006009.
346. Barton DL, Atherton PJ, Bauer BA, et al. The use of Valeriana officinalis (valerian) in improving sleep in patients who are undergoing treatment for cancer: a phase III randomized, placebo-controlled, double-blind study (NCCTG Trial, N01C5).J Support Oncol. 2011;9(1):24-31.
347. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 4th ed. Washington, DC: American Psychiatric Association; 1994.
348. Friedlander M, Brayman Y, Breitbart WS. Delirium in palliative care. Oncology (Williston Park). 2004;18(12):1541-1550.
349. Breitbart W, Alici Y. Agitation and delirium at the end of life. JAMA. 2008;300(24):2898-2910.
350. Rudolph JL, Marcantonio ER. Diagnosis and prevention of delirium. Geriat Aging. 2003;6(10):14-19.
351. Casarett D, Inouye S. Diagnosis and management of delirium near the end of life. Ann Intern Med. 2001;135:32-40.
352. Leonard M, Agar M, Mason C, Lawlor P. Delirium issues in palliative care settings. J Psychosom Res. 2008;65(3):289-298.
353. Cobb JL, Glantz MJ, Martin WE, et al. Delirium in patients with cancer at the end of life. Cancer Pract. 2000;8:172-177.
354. Lawlor PG, Fainsinger RL, Bruera ED. Delirium at the end of life. Critical issues in clinical practice and research. JAMA. 2000;284:2427-2429.
355. American Psychiatric Association. Practice guidelines for the treatment of patients with delirium. Am J Psychiatry. 1999;156 (5 suppl):1-20.
356. Jackson KC, Lipman AG. Drug therapy for delirium in terminally ill adult patients. Cochrane Database System Rev. 2004;(2):CD004770.
357. Boettger S, Breitbart W. Atypical antipsychotics in the management of delirium: a review of the empirical literature.Palliat Support Care. 2005;3(3):227-237.
358. Lonergan E, Britton AM, Luxenberg J, Wyller T. Antipsychotics for delirium. Cochrane Database Syst Rev. 2007;(2):CD005594.
359. Straker DA, Shapiro PA, Muskin PR. Aripiprazole in the treatment of delirium. Psychosomatics. 2006;47(5):385-391.
360. Boettger S, Breitbart W. An open trial of aripiprazole for the treatment of delirium in hospitalized cancer patients.Palliat Support Care. 2011; 9(4):351-357.
361. Noorani NH, Montagnini M. Recognizing depression in palliative care patients. J Palliat Med. 2007;10(2):458-464.
362. von Roenn JH, Paice JA. Control of common, non-pain cancer symptoms. Semin Oncol. 2005;32:200-210.
363. Chochinov HM, Hack T, Hassard T, Kristjanson LJ, McClement S, Harlos M. Understanding the will to live in patients nearing death. Psychosomatics. 2005;46:7-10.
364. Breitbart W, Rosenbeld B, Pessin H, et al. Depression, hopelessness, and desire for hastened death in terminally ill patients with cancer. JAMA. 2000;284:2907-2911.
365. Emanuel EJ, Fairclough DL, Emanuel LL. Attitudes and desires related to euthanasia and physician-assisted suicide among terminally ill patients and their caregivers. JAMA. 2000;284:2460-2468.
366. Mystakidou K, Rosenfeld B, Parpa E, et al. Desire for death near the end of life: the role of depression, anxiety and pain.Gen Hosp Psychiatry. 2005;27(4):258-262.
367. Dees MK, Vernooij-Dassen MJ, Dekkers WJ, et al. "Unbearable suffering:" a qualitative study on the perspectives of patients who request assistance in dying. J Med Ethics. 2011;37:727-723.
368. Hudson P, Kristianson L, Ashby M, et al. Desire for hastened death in patients with advanced disease and the evidence base of clinical guidelines: a systematic review. Palliat Med. 2006;20(7):693-701.
369. Suarez-Almazor M, Newman C, Hanson J, et al. Attitudes of terminally ill cancer patients about euthanasia and assisted suicide: predominance of psychosocial determinants and beliefs over symptom distress and subsequent survival. J Clin Oncol. 2002;20(8):2134-2141.
370. Dein S. Psychiatric liaison in palliative care. Adv Psychiatr Treat. 2003;9:241-248.
371. Stoklosa J, Patterson K, Rosielle D, Arnold R. #186 Fast Facts: Anxiety in Palliative Care—Causes and Diagnosis. Available at http://www.eperc.mcw.edu/EPERC/FastFactsIndex/ff_186.htm. Last accessed May 16, 2012.
372. Roth A, Massie M. Anxiety in palliative care. In: Chochinov M, Breitbart W (eds). Handbook of Psychiatry in Palliative Medicine. 2nd ed. New York, NY: Oxford University Press; 2009: 69-80.
373. Gibson C, Lichtenthal W, Berg A, Breitbart W. Psychologic issues in palliative care. Anesthesiol Clin North Am. 2006;24:61-80.
374. National Cancer Institute. Depression (PDQ). Available at http://www.cancer.gov/cancertopics/pdq/supportivecare/depression/HealthProfessional. Last accessed May 16, 2012.
375. Longo L, Johnson B. Addiction: Part 1. Benzodiazepines: side effects, abuse risk and alternatives. Am Fam Physician. 2000;61(7):2121-2128.
376. McClain C, Rosenfeld B, Breitbart W. Effect of spiritual well-being on end-of-life despair in terminally ill cancer patients.Lancet. 2003;361(9369):1603-1607.
377. Bekelman DB, Hutt E, Masoudi FA, Kutner JS, Rumsfeld JS. Defining the role of palliative care in older adults with heart failure. Int J Cardiol. 2008;125(2):183-190.
378. Lang C, Mancini D. Non-cardiac comorbidities in chronic heart failure. Heart. 2007;93(6):665-671.
379. Lloyd-Williams M, Spiller J, Ward J. Which depression screening tools should be used in palliative care? Palliat Med. 2003;17(1): 40-43.
380. Chochinov HM, Wilson KG. "Are you depressed?" Screening for depression in the terminally ill. Am J Psychiatry. 1997;154: 674-676.
381. Huffman J, Stern T. Using psychostimulants to treat depression in the medically ill. Prim Care Companion J Clin Psychiatry. 2004;6(1):44-46.
382. Daaleman T, Usher BM, Williams SW, Rawlings J, Hanson LC. An exploratory study of spiritual care at the end of life.Am Fam Med. 2008;6:406-411.
383. Sinclair S, Pereira J, Raffin S. A thematic review of the spirituality literature within palliative care. J Palliat Med. 2006;9(2): 464-479.
384. Puchalski C, Ferrell B, Virani R, et al. Improving the quality of spiritual care as a dimension of palliative care: the report of the consensus conference. J Palliat Med. 2009;12(10):885-904.
385. Breitbart W, Gibson C, Poppito SR, Berg A. Psychotherapeutic interventions at the end of life: a focus on meaning and spirituality. Can J Psychiatry. 2004;49:366-372.
386. Johnson KS, Tulsky JA, Hays JC, et al. Which domains of spirituality are associated with anxiety and depression in patients with advanced illness? J Gen Intern Med. 2011;26(7):751-758.
387. Murray SA, Kendall M, Boyd K, Worth A, Benton TF. Exploring the spiritual needs of people dying of lung cancer or heart failure: a prospective qualitative interview study of patients and their carers. Palliat Med. 2004;18:39-45.
388. Maugans TA. The SPIRITual history. Arch Fam Med. 1996;5(1):11-16.
389. Smolinski K, Colon Y. Silent voices and invisible walls: exploring end of life care with lesbians and gay men. J Psychosoc Oncol. 2006;24(1):51-64.
390. Galanti G-A. Vietnamese family relationships. West J Med. 2000;172:415-416.
391. Pennachio D. Cultural competence: caring for Chinese, Japanese, and Korean patients. Med Econ. 2004;81:54.
392. Dayer-Berenson, L. Native Americans: traditional health beliefs. In: Cultural Competencies for Nurses. Impact on Health and Illness. Sudbury, MA: Jones and Bartlett Publishers, LLC; 2011: 252-255.
393. Ackroyd R. Audit of referrals to a hospital palliative care team: role of the bilingual health-care worker. Int J Palliat Nurs. 2003;9(8):352-357.
394. National Cancer Institute. Last Days of Life (PDQ). Available at http://www.cancer.gov/cancertopics/pdq/supportivecare/lasthours/healthprofessional. Last accessed May 16, 2012.
395. Manfredonia J. Prescribing methadone for pain management in end-of-life care. JAOA. 2005;105(3 Suppl): 18S-21S.
396. Wee B, Hillier R. Interventions for noisy breathing in patients near to death. Cochrane Database Syst Rev. 2008;(1):CD005177.
397. Elsayem A, Curry E III, Boohene J, et al. Use of palliative sedation for intractable symptoms in the palliative care unit of a comprehensive cancer center. Support Care Cancer. 2009;17(1):53-59.
398. Rietjens JA, van Zuylen L, van Veluw H, van der Wijk L, van der Heide A, van der Rijt CC. Palliative sedation in a specialized unit for acute palliative care in a cancer hospital: comparing patients dying with and without palliative sedation.J Pain Symptom Manage. 2008;36(3):228-234.
399. Lo B, Rubenfeld G. Palliative sedation in dying patients. JAMA. 2005; 294:1810-1816.
400. Kirk T, Mahon M. National Hospice and Palliative Care Organization (NHPCO) position statement and commentary on the use of palliative sedation in imminently dying terminally ill patients. J Pain Symptom Manage. 2010;39(5):914-923.
401. American Medical Association. AMA Opinion 2.211: Physician-Assisted Suicide. Available at http://www.ama-assn.org/ama/pub/physician-resources/medical-ethics/code-medical-ethics/opinion2211.page. Last accessed May 17, 2012.
402. American Academy of Hospice and Palliative Medicine. Position Statements. Physician-Assisted Suicide. Available at http://www.aahpm.org/positions/default/suicide.html. Last accessed May 15, 2012.
403. National Hospice and Palliative Care Organization. Commentary and Resolution on Physician-Assisted Suicide. Available at http://www.nhpco.org/files/public/pas_resolution_commentary.pdf. Last accessed May 15, 2012.
404. Lampert R, Hayes DL, Annas GJ, et al. HRS expert consensus statement on the management of cardiovascular implantable electronic devices (CIEDs) in patients nearing end of life or requesting withdrawal of therapy. Heart Rhythm. 2010;7(7): 1008-1026.
405. Stroebe MS, Hansson RO, Schut H, et al. (eds). Handbook of Bereavement Research and Practice: Advances in Theory and Intervention. Washington, DC: American Psychological Association; 2008.
406. Worden JW. Grief Counseling and Grief Therapy. 3rd ed. New York, NY: Springer; 2002.
407. Main J. Improving management of bereavement in general practice based on a survey of recently bereaved subjects in a single general practice. Br J Gen Pract. 2000;50:863-866.
408. Bedell S, Cadenhead K, Braboys T. The doctor's letter of condolence: national vital statistics reports final data 1998. N Engl J Med. 2001;344:1161-1162.
409. Miller SC, Lima J, Gozalo PL, Mor V. The growth of hospice care in U.S. nursing homes. J Am Geriatr Soc. 2010;58(8):1481-1488.
410. Casarett D, Karlawish J, Morales K, Crowley R, Mirsch T, Asch DA. Improving the use of hospice services in nursing homes: a randomized controlled trial. JAMA. 2005;294:211-217.
411. Powers BA, Watson NM. Meaning and practice of palliative care for nursing home residents with dementia at end of life.Am J Alzheimers Dis Other Demen. 2008;23(4):319-325.
412. Brazil K, Vohra JU. Identifying educational needs in end-of-life care for staff and families of residents in care facilities. Int J Palliat Nurs. 2005;11(9):475-480.
413. Jones KR, Fink R, Pepper G, et al. Improving nursing home staff knowledge and attitudes about pain. Gerontologist. 2004;44: 469-478.
414. Kiely D, Prigerson H, Mitchell SL. Health care proxy grief symptoms before the death of nursing home residents with advanced dementia. Am J Geriatr Psychiatry. 2008;16(8):664-673.
415. Masoudi FA, Krumholz HM. Polypharmacy and comorbidity in heart failure. BMJ. 2003;327(7414):513-514.
416. Havranek E, Masoudi FA, Westfall KA, Wolfe P, Ordin DL, Krumholz HM. Spectrum of heart failure in older patients: results from the National Heart Failure project. Am Heart J. 2002;143(3):412-417.
417. Holmes HM, Sachs GA, Shega JW, Hougham GW, Cox Hayley D, Dale W. Integrating palliative medicine into the care of persons with advanced dementia: identifying appropriate medication use. J Am Geriatr Soc. 2008;56(7):1306-1311.
418. Stevenson J, Abernethy AP, Miller C, Currow DC. Managing comorbidities in patients at the end of life. BMJ. 2004;329(7471):909-912.
419. Helme RD, Gibson SJ. The epidemiology of pain in elderly people. Clin Geriatr Med. 2001;17:417-431.
420. Ferrell BA, Ferrell BR, Rivera L. Pain in cognitively impaired nursing home patients. J Pain Sympt Manage. 1995;10:591-598.
421. American Geriatrics Society Panel on Pharmacological Management of Persistent Pain in Older Persons. Pharmacological management of persistent pain in older persons. J Am Geriatr Soc. 2009;57: 1331-1346.
422. Mitchell SL, Teno JM, Kiely DK, et al. The clinical course of advanced dementia. N Engl J Med. 2009;361:1529-1538.
423. Hanson LC, Eckert KJ, Dobbs D, et al. Symptom experience of dying long-term care residents. J Am Geriatr Soc. 2008;56:91-98.
424. Teno JM, Clarridge BR, Casey V, et al. Family perspectives on end-of-life care at the last place of care. JAMA. 2004;291:88-93.
425. Ferrell BA. Assessing pain in the elderly. Consult Pharm. 2010;25(Suppl A):5-10.
426. Vandenberg EV, Tvrdik A, Keller BK. Use of the quality improvement process in assessing end-of-life care in the nursing home.J Am Med Dir Assoc. 2005;6(5):334-339.
427. Mahon MM, Sorrell JM. Palliative care for people with Alzheimer's disease. Nurs Philos. 2008;9(2):110-120.
428. Aminoff BZ. Dying dementia patients: too much suffering, too little palliation. Am J Hosp Palliat Care. 2005;22(5):344-348.
429. Herr K, Coyne PJ, Key T, et al. Pain assessment in the nonverbal patient: position statement with clinical practice recommendations. Pain Manage Nursing. 2006;7(2):44-52.
430. Mitchell SL, Morris JN, Park PS, Fries BE. Terminal care for persons with advanced dementia in the nursing home and home care settings. J Palliat Med. 2004;7:808-816.
431. Sachs GA, Shega JW, Cox-Hayley D. Barriers to excellent end-of-life care for patients with dementia. J Gen Intern Med. 2004;19:1057-1063.
432. Sampson EL, Thune-Boyle I, Kukkastenvehmas R, et al. Palliative care in advanced dementia: a mixed methods approach for the development of a complex intervention. BMC Palliat Care. 2008;7:8.
433. Triplett P, Black BS, Phillips H, et al. Content of advance directives for individuals with advanced dementia. J Aging Health. 2008;20(5):583-596.
434. Center to Advance Palliative Care. Improving Palliative Care in Nursing Homes. Available at http://www.capc.org/support-from-capc/capc_publications/nursing_home_report.pdf. Last accessed May 16, 2012.
435. Himelstein B. Palliative care in pediatrics. Anesthesiol Clin North Am. 2005;23:837-856.
436. Widger K, Davies D, Drouin DJ, et al. Pediatric patients receiving palliative care in Canada: results of a multicenter review.Arch Pediatr Adolesc Med. 2007;161(6):597-602.
437. Sheetz MJ, Bowman MA. Pediatric palliative care: an assessment of physicians' confidence in skills, desire for training, and willingness to refer for end-of-life care. Am J Hosp Palliat Care. 2008;25(2):100-105.
438. Fowler K, Poehling K, Billheimer D, et al. Hospice referral practices for children with cancer: a survey of pediatric oncologists.J Clin Oncol. 2006;24(7):1099-1104.
439. Thompson L, Knapp C, Madden V, et al. Pediatricians' perceptions of and preferred timing for pediatric palliative care. Pediatrics. 2009;123;e777.
440. Johnston DL, Nagel K, Friedman DL, Meza JL, Hurwitz CA, Friebert S. Availability and use of palliative care and end-of-life services for pediatric oncology patients. J Clin Oncol. 2008;26(28):4646-4650.
441. Tomlinson D, Bartels U, Gammon J, et al. Chemotherapy versus supportive care alone in pediatric palliative care for cancer: comparing the preferences of parents and health care professionals. CMAJ. 2011;183(17):E1252-E1258.
442. Crane K. Pediatric palliative care gains recognition. J Natl Cancer Inst. 2011;103(119):1432-1433.
443. Wolfe J, Grier HE, Klar N, et al. Symptoms and suffering at the end of life in children with cancer. N Engl J Med. 2000;342(5): 326-333.
444. Kang T, Hoehn KS, Licht DJ, et al. Pediatric palliative, end-of-life, and bereavement care. Pediatr Clin North Am. 2005;52(4): 1029-1046.
445. Himelstein B, Hilden JM, Boldt AM, Weissman D. Pediatric palliative care. N Engl J Med. 2004;350(17):1752-1762.
446. American Academy of Pediatrics. Committee on Bioethics and Committee on Hospital Care. Palliative care for children.Pediatrics. 2000;106:351-357.
447. Hilden J, Watterson J, Chrastek J. Tell the children. J Clin Oncol. 2003;21(9S):37s-39s.
448. Beale E, Baile WF, Aaron J. Silence is not golden: communicating with children dying from cancer. J Clin Oncol. 2005;23(15): 3629-3631.
449. Donnelly JP, Huff SM, Lindsey ML, McMahon KA, Schumacher JD. The needs of children with life-limiting conditions: a healthcare-provider-based model. Am J Hosp Palliat Care. 2005;22:259-267.
450. Graham R, Levetown M, Comeau M. Decision making. In: Carter B, Levetown M, Freibert S (eds). Palliative Care for Infants, Children, and Adolescents: A Practical Handbook. 2nd ed. Baltimore, MD: The Johns Hopkins University Press; 2011.
451. Whitney S, Ethier AM, Fruge E, Berg S, McCullough LB, Hockenberry M. Decision making in pediatric oncology: who should take the lead? The decisional priority in pediatric oncology model. J Clin Oncol. 2006;24(1):160-165.
452. Goldman A, Hewitt M, Collins GS, Childs M, Hain R, United Kingdom Children's Cancer Study Group/Paediatric Oncology Nurses' Forum Palliative Care Working Group. Symptoms in children/young people with progressive malignant disease: United Kingdom Children's Cancer Study Group/Paediatric Oncology Nurses Forum Survey. Pediatrics. 2006;117(6):e1179-1186.
453. Jalmsell L, Kreicbergs U, Onelov E, Steineck G, Henter J-I. Symptoms affecting children with malignancies during the last month of life: a nationwide follow-up. Pediatrics. 2006;117(4):1314-1320.
454. Ullrich C, Mayer OH. Assessment and management of fatigue and dyspnea in pediatric palliative care. Pediatr Clin North Am. 2007;54:735-756.
455. Armendi A, Anghelescu DL. Pain management. In: Pui C (ed). Childhood Leukemias. 2nd ed. New York, NY: Cambridge University Press; 2006.
456. Savedra MC, Holzemer WL, Tesler MD, Wilkie DJ. Assessment of postoperation pain in children and adolescents using the adolescent pediatric pain tool. Nurs Res. 1993;42:5-9.
457. McGrath K, Rosmus C, Canfield C, Campbell MA, Hennigar A. Behaviours caregivers use to determine pain in nonverbal, cognitively impaired children. Develop Med Child Neurol. 1998;40:340-343.
458. Chambers C, Reid GJ, McGrath PJ, Finley GA. Development and preliminary validation of a postoperative pain measure for parents. Pain. 1996;68:307-313.
459. Buccholz M, Karl HW, Pomietto M, Lynn A. Pain scores in infants: a modified infant pain scale vs. visual analog scale.Pain Symp Manag. 1998;15(2):117-124.
460. Newman CJ, Lolekha R, Limkittikul K, Luangxay K, Chotpitayasunondh T, Chanthavanich P. A comparison of pain scales in Thai children. Arch Dis Child. 2005;90:269-270.
461. Luffy R, Grove SK. Examining the validity, reliability, and preference of three pediatric pain measurement tools in African-American children. Pediatr Nurs. 2003;29:54-59.
462. Garra G, Singer AJ, Taira BR, et al. Validation of the Wong-Baker FACES pain rating scale in pediatric emergency department patients. Acad Emerg Med. 2010;17:50-54.
463. Monteiro Caran EM, Dias CG, Seber A, Petrilli AS. Clinical aspects and treatment of pain in children and adolescents with cancer. Pediatr Blood Cancer. 2005;45:925-932.
464. Zernikow B, Smale H, Michel E, Hasan C, Jorch N, Andler W. Paediatric cancer pain management using the WHO analgesic ladder: results of a prospective analysis from 2265 treatment days during a quality improvement study. Eur J Pain. 2006;10(7):587-595.
465. Tournay A. An introduction to pediatric palliative care. Clin Fam Pract. 2004;6(2):499-516.
466. Anghelescu D, Oakes L, Hinds PS. Palliative care and pediatrics. Anesthesiol Clin North Am. 2006;24(1):145-161.
467. American Academy of Pediatrics. Family pediatrics: report of the Task Force on the Family. Pediatrics. 2003;111(6):1541-1571.
468. Kazak A, Alderfer M, Rourke MT, Simms S, Streisand R, Grossman JR. Posttraumatic stress disorder (PTSD) and posttraumatic stress symptoms (PTSS) in families of adolescent childhood cancer survivors. J Pediatr Psychol. 2004;29(3):211-219.
469. Murray J. Attachment theory and adjustment difficulties in siblings of children with cancer. Issues Ment Health Nurs. 2000;21(2):149-169.
470. Sharpe D, Rossiter L. Siblings of children with a chronic illness: a meta-analysis. J Pediatr Psychol. 2002;27(8):699-710.
471. Grootenhuis M, Last BF. Children with cancer with different survival perspectives: defensiveness, control strategies, and psychological adjustment. Psycho-oncology. 2001;10(4):305-314.
472. Mularski R, Puntillo K, Varkey B, et al. Pain management within the palliative and end-of-life care experience in the ICU.Chest. 2009;135:1360-1369.
473. Truog R, Campbell ML, Curtis JR, et al. Recommendations for end-of-life care in the intensive care unit: a consensus statement by the American College of Critical Care Medicine. Crit Care Med. 2008;36(3):953-963.
474. O'Mahony S, McHenry J, Blank AE, et al. Preliminary report of the integration of a palliative care team into an intensive care unit. Palliat Med. 2010;24(2):154-165.
475. Nelson J. Identifying and overcoming the barriers to high-quality palliative care in the intensive care unit. Crit Care Med. 2006;34(Suppl):S324-S331.
476. Mosenthal A, Murphy PA. Interdisciplinary model for palliative care in the trauma and surgical intensive care unit: Robert Wood Johnson Foundation Demonstration Project for Improving Palliative Care in the Intensive Care Unit. Crit Care Med. 2006;34(Suppl):S399-S403.
477. Thompson BT, Cox PN, Antonelli M, et al. Challenges in end-of-life care in the ICU: statement of the 5th International Consensus Conference in Critical Care: Brussels, Belgium, April 2003: executive summary. Crit Care Med. 2004;32:1781-1784.
478. Mosenthal AC, Murphy PA. Trauma care and palliative care: time to integrate the two? J Am Coll Surg. 2003;197:509-516.
479. Norton S, Hogan LA, Holloway RG, et al. Proactive palliative care in the medical intensive care unit: effects on length of stay for selected high-risk patients. Crit Care Med. 2007;35:1530-1535.
480. Beckstrand RL, Kirchhoff KT. Providing end-of-life care to patients: critical care nurses' perceived obstacles and supportive behaviors. Am J Crit Care. 2005;14:395-403.
481. Sulmasy DP, Terry PB, Weisman CS, et al. The accuracy of substituted judgments in patients with terminal diagnoses.Ann Intern Med. 1998;128:621-629.
482. Covinsky KE, Fuller JD, Yaffe K, et al. Communication and decision-making in seriously ill patients: findings of the SUPPORT project. The Study to Understand Prognoses and Preferences for Outcomes and Risks of Treatments. J Am Geriatr Soc. 2000;48:S187-S193.
483. Campbell M. How to withdraw mechanical ventilation: a systematic review of the literature. AACN. 2007;18:397-403.
484. Davidson J, Powers K, Hedayat KM, et al. Clinical practice guidelines for the support of the family in the patient-centered intensive care unit: American College of Critical Care Medicine Task Force 2004–2005. Crit Care Med. 2007;35(2):605-622.
485. Sihra L, Harris M, O'Reardon C. Using the Improving Palliative Care in the Intensive Care Unit (IPAL-ICU) project to promote palliative care consultation. J Pain Symptom Manage. 2011;42(5):672-675.
486. Library of Congress. House Bill HR 5809: Safe Drug Disposal Act of 2010. Available at http://thomas.loc.gov/cgi-bin/query/z?c111:H.R.5809:&utm_source=BenchmarkEmail&utm_campaign=e_News_9_29_10_to_staff&utm_medium=email. Last accessed September 17, 2012.
487. American Academy of Pain Management. Prescribing Issue: Proper Disposal of Medications. Available at http://www.aapainmanage.org/literature/Articles/DisposalOfDrugs.pdf. Last accessed September 17, 2012.

Evidence-Based Practice Recommendations Citations

1. National Consensus Project for Quality Palliative Care. Clinical Practice Guidelines for Quality Palliative Care. 2nd ed. Pittsburgh, PA: National Consensus Project for Quality Palliative Care; 2009. Summary retrieved from National Guideline Clearinghouse at http://www.guideline.gov/content.aspx?id=14423. Last accessed May 24, 2012.
2. Qaseem A, Snow V, Shekelle P, et al. Evidence-based interventions to improve the palliative care of pain, dyspnea, and depression at the end of life: a clinical practice guideline from the American College of Physicians. Ann Intern Med. 2008;148(2):141-146. Summary retrieved from National Guideline Clearinghouse at http://www.guideline.gov/content.aspx?id=12149. Last accessed May 24, 2012.


Copyright © 2012 NetCE, P.O. Box 997571, Sacramento, CA 95899-7571
Mention of commercial products does not indicate endorsement.

No comments:

Post a Comment