17,921
Views
36
CrossRef citations to date
0
Altmetric
Review

Pharmacokinetic considerations and recommendations in palliative care, with focus on morphine, midazolam and haloperidol

, , , , , , , & show all
Pages 669-680 | Received 25 Feb 2016, Accepted 13 Apr 2016, Published online: 29 Apr 2016

ABSTRACT

Introduction: A variety of medications are used for symptom control in palliative care, such as morphine, midazolam and haloperidol. The pharmacokinetics of these drugs may be altered in these patients as a result of physiological changes that occur at the end stage of life.

Areas covered: This review gives an overview of how the pharmacokinetics in terminally ill patients may differ from the average population and discusses the effect of terminal illness on each of the four pharmacokinetic processes absorption, distribution, metabolism, and elimination. Specific considerations are also given for three commonly prescribed drugs in palliative care: morphine, midazolam and haloperidol).

Expert opinion: The pharmacokinetics of drugs in terminally ill patients can be complex and limited evidence exists on guided drug use in this population. To improve the quality of life of these patients, more knowledge and more pharmacokinetic/pharmacodynamics studies in terminally ill patients are needed to develop individualised dosing guidelines. Until then knowledge of pharmacokinetics and the physiological changes that occur in the final days of life can provide a base for dosing adjustments that will improve the quality of life of terminally ill patients. As the interaction of drugs with the physiology of dying is complex, pharmacological treatment is probably best assessed in a multi-disciplinary setting and the advice of a pharmacist, or clinical pharmacologist, is highly recommended.

1. Introduction

In palliative care, when curation is no longer possible, the goal is to maintain or improve the quality of life. To achieve this, a variety of medications, such as morphine, midazolam, and haloperidol, are used for symptom control.[Citation1] Changes in the pharmacokinetics of these drugs may cause increased or decreased drug blood concentrations, which can result in altered efficacy or increased risk of adverse drug reactions. To optimize the use of these drugs, an understanding of pharmacokinetics in this specific patient population is therefore essential.

Pharmacokinetic (Pk) parameters (e.g. drug clearance and volume of distribution) are subject to interpatient variability and may be altered in the palliative population, as patients with cancer are known to differ from healthy volunteers with regards to, for example, age, body weight, and plasma protein levels.[Citation2] In addition, several physiological changes (e.g. decreased fluid intake, a catabolic state, inflammation, and cachexia) occur at the end of life, which can also influence pharmacokinetics.[Citation3Citation5]

So far there is limited knowledge on how these changes affect the different drugs used in palliative care, in particular in the terminal phase, i.e. the last days before death in which a patient is bedridden, semi-comatose, and are no longer able to take oral medication. The aim of this review is to give an overview of how the pharmacokinetics in terminally ill patient differ from the average population, and how changes in the palliative, and especially the terminal phase, can affect drug exposure (). We will discuss the effect of terminal illness on each of the four pharmacokinetic processes: absorption, distribution, metabolism, and elimination (ADME) and give some considerations for three drugs commonly prescribed in the terminal phase (i.e. morphine, midazolam, and haloperidol).[Citation6]

Figure 1. Physiological changes that can influence pharmacokinetics in the terminally ill adult patient.

Figure 1. Physiological changes that can influence pharmacokinetics in the terminally ill adult patient.

2. Absorption

Terminally ill patients frequently suffer from gastro intestinal (GI) problems, such as constipation, nausea, vomiting, and diarrhea. In the case of orally administered drugs, which are used in the palliative care setting when patients are still able to take oral medication, these symptoms can influence both the rate of absorption and bioavailability of a drug. Changes in the absorption rate will affect time-to-peak concentrations (Tmax), whereas changes in bioavailability will affect the initial peak concentration (Cmax) and total exposure, expressed as area under the curve (AUC). If and to what extent a drug is influenced by physiological changes will depend on the physicochemical properties of the drug and the pharmaceutical formulation (e.g. drug solubility and extended release formulations). An overview of the factors influencing absorption is given in . For this review, we will focus on the most commonly used routes of administration in palliative care, which are oral administration in the palliative phase and subcutaneous and transdermal administration in the terminal phase.

Table 1. Physiological changes affecting drug absorption.

2.1. Oral administration

The absorption of orally administered drugs is complex as a drug has to dissolve in the stomach, pass through either the stomach or gut wall, and pass the liver via the portal vein before they reach the systemic circulation. Whether the transportation of the dissolved drug into the bloodstream occurs in the stomach or gut is dependent on the drug’s physicochemical properties. Drugs that are weakly acidic are best absorbed within the acid environment of the stomach. Though most drugs are weak bases (e.g. morphine, haloperidol, and midazolam) and are therefore absorbed in the alkaline environment of the small intestine.

2.1.1. GI symptoms

Absorption of oral drugs can be altered in terminally ill (cancer) patients as this population often suffers from GI symptoms, such as constipation, vomiting, diarrhea, or a delayed gastric emptying due to cachexia. Constipation (i.e. decreases GI motility) occurs in around 50% of the terminal cancer patients and can be a result of dehydration, hypercalcaemia, a bedridden state, and medication use (e.g. opiates).[Citation7,Citation8] Decreased GI motility can result in a reduced absorption rate as it takes longer for the drug to reach the site of absorption.[Citation9Citation11] In the case of a sustained release formulation or drugs with an enterohepatic circulation, decreased GI motility can increase the absorption as there is more contact time with the GI mucosa.

Constipation can also cause nausea and vomiting. Vomiting can evidently decrease the bioavailability of oral medication. The same applies for unclamping the tube if medication is administered via this tube. To what extend the bioavailability is decreased will depend on the time between ingestion and vomiting or releasing the clamp of the tube. The time it takes for a drug to pass from the stomach to the intestine can range from 1 h, for healthy persons up to 4 h, for patients with delayed gastric emptying. As delayed gastric emptying is relevant in this patient population, it has to be taken into account that vomiting or unclamping the tube even several hours after intake of medication the bioavailability can be decreased.

Delayed gastric emptying by itself can also result in a decreased absorption rate for drugs that are absorbed through the small intestine.[Citation11,Citation12] In the case of a drug for which formulation dissolution in the stomach is the rate-limiting step in absorption, a decrease in gastric emptying time may increase the overall extent of absorption and, hence, systemic drug exposure.

Diarrhea can also influence the bioavailability of oral drugs. It can cause a decrease in bioavailability due to increased elimination from the gastro intestinal tract. On the other hand, if the intestinal mucosa is damaged (for instance in the case of an inflammatory process) it can also lead to increased bioavailability. These concepts cause drugs with low bioavailability generally have increased absorption in patients with diarrhea while drugs with good intestinal absorption are more affected by the increased GI motility and, therefore, will have lower absorption.[Citation13]

Furthermore, patients with a gastrointestinal malignancy may have some of their small intestine resected. Small intestine resections involving the loss of more than 100 cm of ileum frequently lead to malabsorption, which could also decrease drug absorption.[Citation14] Absorption might also be decreased by alterations in gut wall function, which is caused by body wasting or cachexia, or decreased splanchnic perfusion.[Citation13,Citation15]

2.1.2. First-pass metabolism

After absorption from the GI tract, the bioavailability of drugs may be altered in terminal patients due to changes in hepatic function or liver blood flow, which can occur in the case of hepatic cirrhosis or congestive heart failure. A decrease in hepatic blood flow can result in increased bioavailability of drugs with a high first-pass metabolism, as was shown for hydromorphone.[Citation16]

2.2. Subcutaneous/transdermal administration

Other common routes to administer drugs in palliative care are transdermal or by subcutaneous injection or infusion. These routes are advantageous in the case of GI problems as this route also bypasses the portal vein, first-pass metabolism does not occur. Factors that may influence absorption of subcutaneous or transdermal medication, however, are tissue blood perfusion and amount of subcutaneous fat. In terminally ill patients, reduced tissue blood perfusion, which can occur as a result of dehydration or old age, can result in a decrease in absorption rate or bioavailability after subcutaneous or transdermal administration.[Citation9,Citation17,Citation18] Alternatively a decrease in subcutaneous fat mass, which is also commonly seen in terminally ill patients, can in theory lead to increased absorption rate and possibly higher peak concentrations.[Citation19]

2.3. Clinical considerations

For clinical practice, we recommend that in the palliative phase GI problems should be closely monitored, and that medication and doses should be reassessed if changes in GI motility occur. As the effect of alterations in GI motility will differ per drug, depending on their chemical properties, this needs to be evaluated on a case by case basis. This assessment is preferably performed in a multi-disciplinary setting and the advice of a pharmacist, or clinical pharmacologist, is recommended. In the presence of a nasogastric tube that decompresses the gut in case of an intestinal obstruction, the administration of drugs through the oral route, or via the tube, is not rational. In the terminal phase, switching to subcutaneous administration, if possible, is preferred not only for the prescribing physician but also for patient’s comfort. In the case of subcutaneous or transdermal drug administration, changes will occur more gradually and monitoring of the clinical effect will probably suffice. If therapy is switched from oral to subcutaneous administration, one should correct for a difference in bioavailability, in addition, it is advisable to look for signs of diminished tissue perfusion (cool extremities, cyanosis, edema, and diminished or absent peripheral pulses) as this could result in a decreases absorption. Finally, in patients with an intestinal obstruction either anatomical or functional administering drugs via a tube followed by 1 or 2 h of clamping the tube will not likely lead to drug absorption, as most drugs are absorbed in the small intestine and in the case of delayed gastric emptying the drug may not have passed from the stomach yet. Therefore, in the case of intestinal obstruction drug administration via the subcutaneous route is preferred.

3. Distribution

The volume of distribution (Vd) of a drug is dependent on its chemical properties (e.g. its hydrophilicity and its affinity with plasma proteins). As a rule, hydrophilic drugs will diffuse into the intravascular, extracellular, and possibly intracellular water, and their Vd will not exceed the volume of total body water (around 42 L for an average adult of 70 kg). Whereas lipophilic drugs or drugs with high affinity to plasma proteins will have low free plasma concentrations and, therefore, a large volume of distribution. As the Vd is determined only by concentration and dose, the plasma concentrations of a drug can be influenced by body composition and amount of plasma protein. Both of these can be altered in terminally ill patients and can change over time, an overview of the factors influencing Vd is given in .

Table 2. Physiological changes affecting drug distribution.

3.1. Body composition

The main factors that influence body composition are loss of body weight and fluid deficit. Loss of body weight and cachexia are common in terminally ill patients, especially in cancer patients. The incidence of weight loss however differs between cancer types with the highest incidence (83–87%) for pancreatic or gastric cancers and the lowest frequency (31–40%) for favorable non-Hodgkin lymphoma, breast cancer, acute non-lymphocytic leukemia, and sarcomas.[Citation20] Fearon et al. showed that in cachectic patients the reduction in body weight is mainly caused by a reduction of adipose tissue (by 80%) and muscle protein (by 75%).[Citation21] A reduction of adipose tissue will result in a lower Vd for lipophilic drugs which will result in higher peak concentrations (Cmax).

Fluid deficit, which is also common among terminally ill patients, can also affect the body composition as it results in loss of total body water. The loss of water can be both intracellular, in the case of dehydration, and extracellular in the case of volume depletion.[Citation5,Citation17] A loss of water will result in a lower Vd for hydrophilic drugs and, therefore, initially lead to higher concentrations. Alternatively, the volume of distribution of hydrophilic drugs can also be increased as a result of ascites, pleural effusion, or generalized edema leading to a higher Vd and lower initial concentrations.[Citation13,Citation22Citation24]

3.2. Protein binding

Besides body composition, alterations in protein binding can also affect Vd. The two main drug binding proteins are albumin and α-1 acid glycoprotein (AAG). Albumin typically binds to weakly acidic drugs (e.g. temazepam and propofol), whereas AAG binds to weakly alkaline drugs (e.g. morphine and fentanyl).[Citation2] Changes in binding proteins can be caused by inflammatory responses. A long-lasting inflammatory response occurs in almost all types of solid tumors and can also be the result of cachexia, infection, and degenerative diseases.[Citation17,Citation25Citation27] As a result of the inflammatory response, albumin is downregulated and AAG is increased.[Citation27] Hypoalbuminemia is, therefore, often seen in various types of cancer, cachectic patients, and hospitalized or institutionalized elderly patients.[Citation14,Citation28Citation32] Increased plasma levels of AAG have also been shown in various types of cancer, acute illness, or chronic disease.[Citation33,Citation34] As a result, highly AAG bound drugs will have decreased unbound concentrations while highly albumin bound drugs will have increased unbound concentration. A decreased unbound concentration can result in decreased elimination and due to slow redistribution from the tissues, the effect can be prolonged. The clinical relevance of increased amounts of unbound drug on the other hand is limited as the elimination of a drug increases when the unbound concentration increases. Still if the elimination is otherwise inhibited, for example, in the case of renal failure, this might lead to accumulation.

3.3. Clinical considerations

As volume of distribution mainly affects the initial peak concentration (and also the time needed to reach steady-state concentrations), recommendations for clinical practice will primarily be relevant for drugs where an immediate response is desired. This is for instance the case in sedative or analgesic medication. For these drugs, a higher initial (loading) dose may be required if the volume of distribution in an individual is increased. For instance, to achieve adequate sedation, an obese patient will probably require a higher initial dose of midazolam (a lipophilic drug) than a cachectic patient. In addition, for pain management a patient with edema may probably need a higher initial dose of morphine (a hydrophilic drug) than a dehydrated patient.

4. Metabolism

Conversion of drugs into metabolites primarily takes place in the liver and largely determines the duration of a drug’s action, elimination, and toxicity. Hepatic clearance (ClH), the ability of the liver to remove drugs from the systemic circulation, is dependent on both liver blood flow and hepatic extraction ratio. The hepatic extraction ratio is the fraction of drug that is removed from the blood after a single passage through the liver. Drugs with a high extraction ratio will have a ClH that is primarily dependent on the liver blood flow. While for drugs with a low extraction ratio, this will be mainly dependent on intrinsic clearance (i.e. liver function). In patients with terminal illness, there are several factors that might influence drug metabolism, an overview is given in .

Table 3. Physiological changes affecting drug metabolism.

4.1. Liver blood flow

Liver blood flow reduces with age, and can also be decreased in dehydrated patients due to decreased cardiac output, in patients with liver cirrhosis due to intrahepatic and extrahepatic portal systemic shunting, or in patients with heart failure as a result of passive congestion or low cardiac output.[Citation10,Citation17,Citation35,Citation36] These patients can, therefore, have a decreased metabolism of drugs with a high extraction ratio, such as fentanyl, morphine, and propofol. As a result, the effect of these drugs can be increased and prolonged.

4.2. Intrinsic clearance

Intrinsic clearance is determined by the enzymatic capacity. There are two main enzymatic systems that are responsible for drug metabolism, i.e. phase I and phase II metabolism. Phase I metabolism includes oxidation, reduction, and hydrolysis and occurs predominantly by enzymes of the cytochrome P450 (CYP450) family. Phase II metabolism consists of conjugation with an endogenous substance (e.g. glucuronidation, acetylation, or sulfation). There are several factors that influence the metabolic capacity including genetic variability, enzyme induction, or inhibition (usually drug induced) and disease states including malignancies.[Citation14] Liver injury in terminally ill cancer patients can be due to primary liver tumors or more often due to the presence of liver metastases or as a result of chemotherapy. In non-malignant terminally ill patients liver function can also be affected, for instance in the case of alcoholic liver cirrhosis or in Chronic Obstructive Pulmonary Disease (COPD) patients, who have been also shown to be more at risk for hepatobiliary diseases.[Citation37]

The effect of liver pathology on metabolic capacity is, however, highly variable and difficult to predict. In fact, most liver functions can be maintained with some degree of liver injury, therefore liver pathology (including the presence of multiple liver metastases) can exist without affecting liver function. It is believed that unless liver cirrhosis is present, chronic liver diseases have little significant clinical impact on pharmacokinetics. In addition, phase II metabolism tends to be better preserved than phase I metabolism, only in advanced cirrhosis this pathway may also be impaired substantially.[Citation18,Citation38]

As the metabolic capacity depends on nutrients and co-factors, it is probable that malnutrition can result in altered metabolism. Indeed, some studies showed that deficiency of specific nutrients (e.g. proteins, lipids, vitamin C, vitamin B6, and vitamin E) can result in a decrease in metabolic rate. However, some deficiencies, such as riboflavin and iron have also shown to increase CYP450 metabolism by a still unknown mechanism.[Citation39] A reduction in the enzyme levels of some CYP450 enzymes (CYP2C8/10 and CYP2E1) have been shown, but this was not the case for some other CYP450 enzymes (CYP1A2 and CYP3A).[Citation40] Studies on the direct effect of malnutrition/cachexia on plasma drug levels are sparse and despite similar metabolic pathways, the influence of cachexia was divergent. Most of the drugs showed increased plasma levels after oral administration; however, with only plasma levels of the drug it is not possible to differentiate between changes in absorption, metabolism, or elimination. One study on oxycodone in cachectic cancer patients also measured the metabolite, noroxycodone, formed via the CYP3A enzyme and did show higher plasma levels of oxycodone and a lower noroxycodone/oxycodone ratio in patients with a higher GPS score (a measurement for cachexia) indicating that cachexia decreases the hepatic metabolism of oxycodone.[Citation41] This suggests a decrease in metabolic capacity, yet the overall effect of malnutrition and cachexia on metabolism is still unclear.

Another possible method by which CYP450 metabolism can be reduced in cancer patients is by inflammatory response. This is mediated largely through downregulation of gene transcription caused by pro-inflammatory cytokines.[Citation27] This effect has not been studied extensively but it has been shown in some studies for the metabolism of CYP3A4 and CYP2C19.[Citation42Citation44] In addition, there are also implications that inflammation may reduce the metabolic capacity of CYP1A2.[Citation45Citation47] The clinical relevance of these reduction in metabolism, however, remains to be further investigated.

4.3. Clinical considerations

For clinical practice, one should be aware that drug metabolism can be altered in patients with heart failure or those that suffer from decreased cardiac output due to dehydration (resulting in decreased hepatic blood flow) or patients with liver disease. In addition, drugs that are metabolized via the CYP450 enzyme system are likely to be affected more than drugs which are metabolized via phase II metabolism. As the effect of liver disease, dehydration, inflammation, and cachexia on liver metabolic capacity, is difficult to predict no specific recommendations can be made. Instead, care givers should be aware of the fact that patients with liver diseases can have a different reaction to medication, and they should look out for signs of altered efficacy and side effects in these patients, especially in the case of drugs with active metabolites.

5. Elimination

The elimination of drugs and metabolites can occur through a number of different routes (e.g. bile, sweat, and saliva); however, the main route of elimination is via the kidneys through glomerular filtration. Renal function, including glomerular filtration rate, decreases with increasing age. This alone means that most terminally ill patients will have a reduced elimination, as they are usually older (on average 63 years) than the healthy volunteers in which most pharmacokinetic studies are performed (on average 25–29 years).[Citation2,Citation48] Renal elimination can also be decreased in terminally ill patients as a result of renal insufficiency, which occurs in a large portion (50–60%) of the cancer patients.[Citation49] Most terminally ill patients have a diminished fluid intake, which will cause prerenal kidney failure. Co-administration of non-steroidal anti-inflammatory drugs (NSAIDs) in this situation will severely compromise renal function.[Citation24]

It is important to note that although renal insufficiency is common in this population, it might not be recognized using the standard blood chemistry tests. This is because glomular filtration is estimated using serum creatinine levels. In the case of terminally ill patients, this can be misleading as the production of creatinine is reduced as muscle mass is decreased. Therefore, glomular filtration rate can decrease without a change in serum creatinine concentrations. It is therefore important to realize that the eGFR provided by modification of diet in renal disease (MDRD) formula gives an overestimation of the renal function in patients with low muscle mass. For drugs that are not eliminated via kidneys but undergo hepatic elimination, accumulation can occur if the liver decompensates in the terminal phase. This can also happen if the bile is the primary route of elimination and the patient becomes icteric.[Citation24] An overview of the factors affecting elimination is given in .

Table 4. Physiological changes affecting drug elimination.

5.1. Clinical considerations

In clinical practice, renal-eliminated drugs (or metabolites) will accumulate in the final days of life, if fluid intake is limited. Measuring renal function based on serum creatinine will not be very helpful in these patients. It is therefore recommended to either determine renal function using other parameters that correct for the loss of muscle for instance albumin or weight besides creatinine clearance or to measure drug concentrations. As both these interventions require blood sampling, it is probably of more practical value, to be aware of the fact that accumulation of certain drugs can occur and to monitor fluid intake and urinary output and look out for (increased) side effects in patients where these functions are diminished.

6. Conclusion

In conclusion, there are numerous ways by which comorbidities and other physiological changes can alter pharmacokinetics in patients with terminal illness. The net effect of these alterations and the clinical relevance will be dependent on both the status of the individual patient and the properties of the drug in question. For clinical practice, we will discuss three commonly prescribed drugs in the terminal phase, i.e. morphine, midazolam, and haloperidol.

7. Morphine

Morphine is widely used to treat pain and dyspnea in terminally ill patients.[Citation50] In a palliative setting, it is usually administered either orally (as normal release liquid or modified release tablets) or subcutaneously (as bolus injection or continuous infusion). Morphine is a relatively hydrophilic drug and is only partially bound (34–37.5%) to plasma proteins, predominantly albumin.[Citation51] The metabolism of morphine takes place primarily in the liver. Morphine has a high extraction ratio and is metabolized mainly by Uridine 5ʹ-diphospho-glucuronosyltransferase (UGT) enzymes into morphine-3-glucuronide (M3G) for 60%, and morphine-6-glucuorinide (M6G) for 10%.[Citation52Citation54] The M6G metabolite is pharmacologically active and is 10–60 times as potent as morphine.[Citation53Citation60] Its ability to cross the blood–brain barrier is, however, far less (1/57th) than that of morphine.[Citation61] Nonetheless after chronic morphine administration, the gradual accumulation of M6G in the brain can account for increased potency compared to single administration.[Citation53,Citation60,Citation62,Citation63] The M3G metabolite does not bind to the opioid receptors and, therefore, does not possess analgesic properties.[Citation56,Citation64Citation67] Conversely, it has been suggested that M3G may be responsible for the side effects of morphine.[Citation54,Citation68] Both glucuronide metabolites are eliminated through renal excretion. Overall, this pharmacokinetic profile of morphine means that its concentrations and effect may be influenced by changes in total body water (by influencing Vd), liver blood flow (by influencing metabolism and also via first-pass absorption), and renal function (by influencing elimination of the metabolites).

The effect of total body water on the Vd of morphine have been shown by Baillie et al. [Citation69]. Their results showed a decreased volume of distribution in elderly patients when compared to younger adults, which is in line with the fact that total body water declines with age. The clinical relevance of this will, however, be limited for terminally ill patients as the volume of distribution only determines the initial peak concentration and most patients will receive multiple doses of morphine.

An increased interpatient variability in morphine metabolism in terminally ill patients has been shown. This has been suggested to be due to reduced hepatic blood flow and subsequent reduction in morphine clearance in these patients.[Citation69] As a result of variability in metabolism, interpatient variability in oral bioavailability (between 15% and 49%) has also been shown.[Citation70,Citation71] The fact that this is caused by liver metabolism instead of absorption in the GI tract is supported by the fact that patients with icterus had an even higher oral bioavailability of 64%.[Citation70] In addition, the fact that first-pass metabolism determines its bioavailability also means that the ratio of morphine to its metabolites will differ for different routes of administration.[Citation72Citation74] This can be relevant as the metabolites of morphine can influence both its efficacy and side effects.

As the kidneys are responsible for the elimination of the glucuronide metabolites, renal function is an important aspect in morphine pharmacokinetics. This is especially relevant in terminally ill patients as renal insufficiency is common in this population. Accumulation of M3G and M6G in patients with renal insufficiency has been shown in several studies.[Citation72,Citation73,Citation75Citation77] This can be advantageous due to the increased levels of the active M6G metabolite. It has indeed been shown that patients with renal insufficiency had an increased response to morphine and that they required lower doses.[Citation77Citation80] Another advantage is that M6G has a lower risk of respiratory depression or hypoxia compared to morphine itself.[Citation67,Citation81Citation83] However, other side effects, such as delirium, myoclonus, and hyperalgesia/allodynia have been related to higher metabolite levels in terminally ill patients and are probably caused by accumulation of the M3G metabolite.[Citation84Citation91]

In clinical practice, this means that physicians and nurses should be aware that if renal function declines (for instance if fluid intake ceases) delirium and myoclonus can occur. At the same time, the pain symptoms can both increase (hyperalgesia due to M3G accumulation) or decrease (due to M6G accumulation). If the pain is not increased, a reduction in morphine dose can be considered, otherwise switching to an analgesic without active metabolites (for instance fentanyl) may be an option. Furthermore, dosing forms that bypass the portal vein and, therefore, do not undergo first-pass metabolism (e.g. intravenous or subcutaneous injections) will probably have less side effects as the morphine–metabolites ratio is higher. This might therefore also be beneficial in patients with renal insufficiency.

8. Midazolam

Midazolam can be used intermittently for the night times and is the drug of choice for palliative sedation in terminally ill patients.[Citation6,Citation92Citation94] It is commonly administered via subcutaneous infusion but can also be administered orally to treat anxiety or insomnia. Midazolam is a highly permeable drug and is, therefore, believed to be completely absorbed through the GI tract, if given orally.[Citation95] However, midazolam has limited bioavailability due to first-pass metabolism via CYP3A enzymes in the liver and gut wall. As midazolam is a highly permeable drug, the extent of first-pass metabolism can be influenced by variability in intestinal blood flow.[Citation95] In addition, it has also been proposed that midazolam bioavailability can be influenced by CYP3A metabolizing activity in the intestine.[Citation96] Midazolam is highly lipophilic at physiological pH and is also highly bound to albumin (96–97%), resulting in a large volume of distribution.[Citation97,Citation98] It is metabolized in the liver, mainly by CYP3A into 1-hydroxymidazolam, which is then glucuronidated and excreted via the kidneys. 1-Hydroxymidazolam is pharmacologically active, although to a lesser extent than midazolam.[Citation97] Midazolam has an intermediate extraction ratio its metabolism is, therefore, dependent on both liver blood flow and enzymatic activity.[Citation99Citation101] Overall, this pharmacokinetic profile of midazolam means that its concentrations and effect may be influenced by changes in total body fat and albumin levels (by influencing Vd), liver blood flow, intestinal blood flow and CYP3A activity (by influencing metabolism and also via first-pass absorption) and renal function (by influencing elimination of the metabolites).

The effect of total body fat on the volume of distribution of midazolam has been studied primarily in obese patients. As expected, obese patient had a larger volume of distribution for midazolam.[Citation96,Citation102Citation104] We would therefore expect the opposite in terminally ill patients, and a study on cancer cachexia in rats did indeed show a decrease in Vd after the animals became cachectic. Increased plasma concentrations as a result of a decrease in Vd can be further enhanced as a result of hypoalbuminemia. Increased plasma concentrations as result of decreased Vd or hypoalbuminemia can have an impact on the onset of sedation after first administration. Halliday et al. showed that hypoalbuminemia was associated with shorter time to induction suggesting that higher levels of free midazolam will give a more rapid response.[Citation105] On the other hand, if midazolam is given continuously over a longer period of time the higher free plasma levels will also result in a higher elimination.

Midazolam metabolism can be reduced in terminally ill patients as a result of reduced liver blood flow. This has been shown in elderly patients who compared to younger adults had a decreased midazolam clearance.[Citation102] As midazolam is primarily metabolized by CYP3A, a reduction of CYP3A activity can also lead to decreased midazolam metabolism. Reduced CYP3A activity as a result of cachexia has been suggested to occur in cachectic patients and decreased midazolam clearance has also been shown in an animal model of cancer cachexia.[Citation41,Citation106] Reduced CYP3A activity can also occur as a result of liver disease and a correlation between midazolam clearance and liver failure has been shown in intensive care unit (ICU) patients.[Citation107] In palliative patients, no correlation was found between midazolam concentrations and liver disease, probably because liver diseases in this population are not as severe as in ICU patients.[Citation108] Finally, CYP3A metabolism can also be affected by the use of other drugs. In the palliative setting, there might be a relevant interaction with dexamethasone. Dexamethasone is used for a variety of symptoms in the terminal phase, and there are suggestions that it may induce CYP3A.[Citation109,Citation110] However, the extent by which dexamethasone induces CYP3A has not been completely clarified.

Finally, the elimination of the glucuronidated metabolites by the liver is reduced in patients with renal insufficiency, resulting in accumulation. Although glucuronidated 1-hydroxymidazol has only 1/10th of the potency of midazolam itself, this can result in prolonged sedation.[Citation111]

In clinical practice, the onset of sedation can be different between patients due to changes in Vd. Patients with higher body weight may, therefore, require a higher initial dose, whereas hypoalbumineamic patients may require a lower initial dose. Patients who have used a CYP3A inducer, such as carbamazepine, in the past week may need higher midazolam doses to achieve accurate sedation. Finally, in patients with renal insufficiency, the sedative effect may be prolonged. This will probably be of little clinical relevance in the case of palliative sedation as most patients will only require sedation for less than 48 h. Nevertheless, it is something to keep in mind if midazolam is given for anxiety or insomnia.

9. Haloperidol

Haloperidol is a typical antipsychotic drug that is used in palliative care to treat delirium and might also be prescribed to treat nausea and vomiting.[Citation1,Citation112] In terminally ill patients, it is administered either orally or as a subcutaneous injection.[Citation113] If given orally, it has a bioavailability of 60–70% due to first-pass metabolism.[Citation112,Citation114,Citation115] For the subcutaneous route, there is no information available but bioavailability is probably around 100% as it diffuses from the subcutaneous tissue directly to the systemic circulation. Haloperidol is a lipophilic drug, and it is bound to albumin for more than 90%. Therefore, haloperidol has a large volume of distribution.[Citation116,Citation117] The hepatic metabolism of haloperidol is extensive (<1% is excreted unchanged) and includes both irreversible and reversible metabolic biotransformation. The main metabolic pathway is glucuronidation by UGT, which accounts for 50–60% of the total metabolism.[Citation118] An estimated 20–30% of haloperidol is metabolized via CYP3A4 and CYP2D6.[Citation119] Both these pathways are irreversible. The reversible part of the haloperidol metabolism is its conversion into reduced haloperidol by carbonyl reductase, which accounts for approximately 23% of the total metabolism.[Citation120Citation122] The reduction of haloperidol is reversible as reduced haloperidol can be converted back into haloperidol through oxidation by CYP3A4.[Citation119,Citation123,Citation124] Haloperidol has an intermediate extraction ratio therefore its metabolism is dependent on both enzymatic activity and liver blood flow.[Citation114] Haloperidol metabolites are eliminated both with the urine and via the bile.[Citation125,Citation126] Overall, this pharmacokinetic profile of haloperidol means that its concentrations and effect may be influenced by changes in body fat and albumin levels (by influencing Vd), liver blood flow and metabolic activity (by influencing metabolism and also via first-pass absorption).

In terminally ill patients, a reduction in body fat, and consequently Vd, can result in higher initial plasma concentrations. Furthermore, hypoalbuminemia can also result in higher free haloperidol concentrations and thereby possibly shorter the time-to-peak plasma concentrations. These changes can be clinically relevant as a rapid onset of action is desired in treating delirium. A large interpatient variability in time-to-peak plasma concentrations, between 2 and 6 h, has been shown in patients taking oral haloperidol.[Citation114,Citation127] It is, however, not known if this is due to changes in plasma albumin if there are other explanations, for instance delayed gastric emptying.

Haloperidol metabolism might be reduced in terminally ill patients as a result of reduced liver blood flow. It has been shown that elderly patients had higher steady-state plasma concentrations than younger patients.[Citation127] As steady-state concentrations are only influenced by changes in clearance (not in Vd) a decrease in liver blood flow, which is common in elderly, might explain this.

Finally, differences in metabolic capacity might also influence haloperidol metabolism and thereby plasma concentrations. Interpatient variability in metabolism is unlikely to be caused by changes in UGT activity, as its capacity is relatively large compared to the other metabolic pathways.[Citation114] The conversion of haloperidol into reduced haloperidol is also unlikely to cause much interpatient variability as little variation in enzyme activity has been shown for carbonyl reductase.[Citation114] Changes in CYP3A4 or CYP2D6 activity on the other hand may lead to altered plasma concentrations. In the case of CYP3A4, it has been shown that co-administration of haloperidol with carbamazepine, a CYP3A4 inducer, resulted in significantly lower haloperidol concentrations.[Citation128Citation131] The combination of carbamazepine and haloperidol might be relevant in patients with brain tumors or metastases. Another drug that might induce CYP3A is dexamethasone, this is commonly used in palliative care but the relevance of this combination remains to be determined.[Citation109,Citation110] A decrease in haloperidol metabolism in terminally ill patients is also possible as result of reduced CYP3A activity due to cachexia.[Citation41] Variability in CYP2D6 metabolic capacity may also influence haloperidol concentrations. This has been shown by Mihara et al. for patients with a genetic variation in CYP2D6 enzyme.[Citation132] In terminally ill patients, this could be relevant in the case of co-administration of haloperidol with CYP2D6 inhibitors, like fluoxetine or paroxetine. Although these drugs are not commonly given in the terminal phase. There have been some studies on the effect of fluoxetine on haloperidol levels and this showed a 20–35% increase in plasma levels. However, this was not associated with clinical effects.[Citation133Citation136] So far, the effect of alteration in haloperidol metabolism due to cachexia, dexamethasone use or fluoxetine, or paroxetine use are merely theoretical and more research on its clinical relevance is needed.

In clinical practice, it may be the case that patients with hypoalbuminemia or loss of body fat will have a more rapid onset of action, and a lower initial dose might be sufficient. In addition, patients with reduced liver blood flow, or co-administration of dexamethasone might also need a lower dose. While patients with cachexia or fluoxetine or paroxetine use might need higher doses, it is not yet possible to make any real recommendations as there has been very little research on haloperidol pharmacokinetics in terminally ill patients, especially about the use of the subcutaneous injections.

10. Expert opinion

The pharmacokinetics of drugs in terminally ill patients can be complex due to the pathophysiological changes that occur near the end of life. Although there are several guidelines for symptom management in terminally ill patients, limited evidence exists on guided drug use in these patients. Even for the most commonly used medications in this population (i.e. morphine, midazolam, and haloperidol) much is still unknown. The medication dose is therefore usually guided by experience and clinical effect, resulting in adaptation of a universal starting dose rather than defining a personalized dose beforehand based on solid PK characteristics.

Besides comorbidities, co-medication can also influence the action of drugs (both on the level of pharmacokinetics as pharmacodynamics). If a new drug, which could potentially interfere with the current medication, is added to the regimen caution is essential and short acting formulations are preferred when treatment is initiated and polypharmacy should be avoided. This may be more relevant in the pre-terminal phase as medication is reassessed in the terminal phase and most medication (besides analgesic and anxiolytics) is usually discontinued.

Such personalized treatment may significantly improve the quality of life for these patients and their family members, especially in the final days of life. To achieve this not only more knowledge but also more studies on the pharmacokinetics in terminally ill patients are necessary. A growing number of pharmacokinetic studies are being performed in special patient populations (e.g. ICU patients), yet these studies in terminally ill patients are still lacking to a large extent. In addition, there is also a need for pharmacodynamic (Pd) studies in this population as pharmacokinetics will give information on the achieved drug concentrations but not on the preferred clinical effect. Pd studies that measure the effect on for instance pain, sedation, or delirium would be of great clinical importance. The fact that so little studies are being performed in terminally ill patients might be because terminally ill patients are considered a vulnerable population, and it has been argued that including them in clinical research is inappropriate or even unethical. These ethical concerns are, however, often unjustified and studies in this population, if carefully designed and executed, can be very valuable.[Citation137] A crucial aspect is to minimize the burden for patients and their families. Population Pk/Pd studies using limited sampling strategies may therefore provide a solution and may eventually lead to individualized dosing guidelines.

While Pk/Pd studies are lacking, there are several studies on factors predicting death in terminally ill patients.[Citation19,Citation25,Citation26,Citation138] These studies give valuable insight in the changes in body functions that occur in the final days of life. Together with the knowledge of pharmacokinetics mentioned in this review this should provide a base on which pharmacological interventions can be made which will improve the quality of life of terminally ill patients. The difficulty in this is, however, that although a common final pathway is hypothesized, the terminally ill population can be very heterogeneous, they require different types of medication and will have different comorbidities. As the net result of drug concentrations is dependent on both physiological changes as well as chemical drug properties, these are probably best assessed by a multi-disciplinary team with a specialist pharmacist or clinical pharmacologist with specific knowledge of the last phase of life.

Article highlights

  • In terminally ill patients, pharmacokinetics may be altered as a result of co-morbidities and physiological changes that occur at the end stage of life.

  • The absorption of orally administered drug, can be altered significantly as a results of GI symptoms. In the palliative phase GI problems should be closely monitored, and medication (both dose and route of administration) should be reassessed if changes in GI motility occur.

  • The volume of distribution of drugs can vary in terminally ill patients due to changes in body composition and plasma proteins. This is particularly relevant for drugs for which rapid response is desired.

  • Drug metabolism can be diminished in case of liver disease, dehydration, inflammation or cachexia. Care givers should be aware of different reaction in patients with these symptoms and should look out for signs of altered efficacy and side effects in these patients, especially in the case of drugs with active metabolites.

  • Renal eliminated drugs (or metabolites) can accumulate in the final days of life if fluid intake is limited, which can cause side effects due to accumulation of drugs or metabolites

  • As the interaction of drugs with the physiology of dying is complex pharmacological treatment is probably best assessed in a multi-disciplinary setting and the advice of a pharmacist, or clinical pharmacologist, is highly recommended.

This box summarizes key points contained in the article

Acknowledgments

The authors would like to thank W Bramer (Medical Library, Erasmus Medical, Centre) for assistance with the literature search.

Declaration of interests

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

References

  • De Lima L. International Association for Hospice and Palliative Care list of essential medicines for palliative care. Ann Oncol. 2007 Feb;18(2):395–399.
  • Cheeti S, Budha NR, Rajan S, et al. A physiologically based pharmacokinetic (PBPK) approach to evaluate pharmacokinetics in patients with cancer. Biopharm Drug Dispos. 2013 Apr;34(3):141–154.
  • Stevenson J, Abernethy AP, Miller C, et al. Managing comorbidities in patients at the end of life. BMJ. 2004 Oct 16;329(7471):909–912.
  • Bruera E, Hui D. Palliative care research: lessons learned by our team over the last 25 years. Palliat Med. 2013 Dec;27(10):939–951.
  • Sarhill N, Walsh D, Nelson K, et al. Evaluation and treatment of cancer-related fluid deficits: volume depletion and dehydration. Support Care Cancer. 2001 Sep;9(6):408–419.
  • Masman AD, van Dijk M, Tibboel D, et al. Medication use during end-of-life care in a palliative care centre. Int J Clin Pharm. 2015 Oct;37(5):767–775.
  • Reuben DB, Mor V, Hiris J. Clinical symptoms and length of survival in patients with terminal cancer. Arch Intern Med. 1988 Jul;148(7):1586–1591.
  • Van Lancker A, Velghe A, Van Hecke A, et al. Prevalence of symptoms in older cancer patients receiving palliative care: a systematic review and meta-analysis. J Pain Symptom Manage. 2014 Jan;47(1):90–104.
  • Turnheim K. When drug therapy gets old: pharmacokinetics and pharmacodynamics in the elderly. Exp Gerontol. 2003 Aug;38(8):843–853.
  • Lichtman SM. Guidelines for the treatment of elderly cancer patients. Cancer Control. 2003;10(6):445–453.
  • Smith BS, Yogaratnam D, Levasseur-Franklin KE, et al. Introduction to drug pharmacokinetics in the critically ill patient. Chest. 2012 May;141(5):1327–1336.
  • Morley JE, Thomas DR, Wilson MM. Cachexia: pathophysiology and clinical relevance. Am J Clin Nutr. 2006 Apr;83(4):735–743.
  • Trobec K, Kerec Kos M, von Haehling S, et al. Pharmacokinetics of drugs in cachectic patients: a systematic review. PLoS One. 2013;8(11):e79603.
  • Mihelic RA. Pharmacology of palliative medicine. Semin Oncol Nurs. 2005 Feb;21(1):29–35.
  • Smucker D, Wang R. The effect of aging on digestive processes. In: Masoro EJ, editor. CRC handbook of physiology in aging. Boca Raton (FL): CRC Press; 1981. p. 235–285.
  • Chang SF, Moore L, Chien YW. Pharmacokinetics and bioavailability of hydromorphone: effect of various routes of administration. Pharm Res. 1988 Nov;5(11):718–721.
  • Sarhill N, Mahmoud FA, Christie R, et al. Assessment of nutritional status and fluid deficits in advanced cancer. Am J Hosp Palliat Care. 2003 Nov–Dec;20(6):465–473.
  • Krishnan V, Murray P. Pharmacologic issues in the critically ill. Clin Chest Med. 2003 Dec;24(4):671–688.
  • Viganò A, Dorgan M, Buckingham J, et al. Survival prediction in terminal cancer patients: a systematic review of the medical literature. Palliat Med. 2000 Sep;14(5):363–374.
  • Tisdale MJ. Molecular pathways leading to cancer cachexia. Physiology. 2005 Oct;20:340–348.
  • Fearon KC, Preston T. Body composition in cancer cachexia. Infusionstherapie. 1990 Apr;17(Suppl 3):63–66.
  • Pichard C, Kyle UG. Body composition measurements during wasting diseases. Curr Opin Clin Nutr Metab Care. 1998 Jul;1(4):357–361.
  • Muscaritoli M, Anker SD, Argilés J, et al. Consensus definition of sarcopenia, cachexia and pre-cachexia: joint document elaborated by Special Interest Groups (SIG) ‘cachexia-anorexia in chronic wasting diseases’ and ‘nutrition in geriatrics’. Clin Nut. 2010 Apr;29(2):154–159.
  • Verhagen CAHHVM. Pathofysiologie, symptomen en problematiek rond de terminale patiënt. In: van Bommel EMG, Tukker J, editors. 26e Anselmus Colloquium De laatste fase. Stichting Organisatie Anselmus Colloquium, Houten, 2014 Sept 18, Utrecht; 2014.
  • Mahmoud FA, Rivera NI. The role of C-reactive protein as a prognostic indicator in advanced cancer. Curr Oncol Rep. 2002 May;4(3):250–255.
  • Lindenmann J, Fink-Neuboeck N, Koesslbacher M, et al. The influence of elevated levels of C-reactive protein and hypoalbuminemia on survival in patients with advanced inoperable esophageal cancer undergoing palliative treatment. J Surg Oncol. 2014 Nov;110(6):645–650.
  • Slaviero KA, Clarke SJ, Rivory LP. Inflammatory response: an unrecognised source of variability in the pharmacokinetics and pharmacodynamics of cancer chemotherapy. Lancet Oncol. 2003;4:224–232.
  • Evans WJ, Morley JE, Argilés J, et al. Cachexia: a new definition. Clin Nut. 2008 Dec;27(6):793–799.
  • Kemik O, Sumer A, Kemik AS, et al. The relationship among acute-phase response proteins, cytokines and hormones in cachectic patients with colon cancer. World J Surg Oncol. 2010;8:85.
  • Araújo JP, Lourenço P, Rocha-Gonçalves F, et al. Nutritional markers and prognosis in cardiac cachexia. Int J Cardiol. 2011 Feb 3;146(3):359–363.
  • Woo J, Chan HS, Or KH, et al. Effect of age and disease on two drug binding proteins: albumin and alpha-1- acid glycoprotein. Clin Biochem. 1994 Aug;27(4):289–292.
  • Piafsky KM. Disease-induced changes in the plasma binding of basic drugs. Clin Pharmacokinet. 1980 May–Jun;5(3):246–262.
  • Duché JC, Urien S, Simon N, et al. Expression of the genetic variants of human alpha-1-acid glycoprotein in cancer. Clin Biochem. 2000 Apr;33(3):197–202.
  • Israili ZH, Dayton PG. Human alpha-1-glycoprotein and its interactions with drugs. Drug Metab Rev. 2001 May;33(2):161–235.
  • Delcò F, Tchambaz L, Schlienger R, et al. Dose adjustment in patients with liver disease. Drug Saf. 2005;28(6):529–545.
  • Alvarez AM, Mukherjee D. Liver abnormalities in cardiac diseases and heart failure. Int J Angiol. 2011 Sep;20(3):135–142.
  • Mapel D. Renal and hepatobiliary dysfunction in chronic obstructive pulmonary disease. Curr Opin Pulm Med. 2014 Mar;20(2):186–193.
  • Morgan DJ, McLean AJ. Clinical pharmacokinetic and pharmacodynamic considerations in patients with liver disease. An update. Clin Pharmacokinet. 1995 Nov;29(5):370–391.
  • Raiten DJ. Nutrition, pharmacology, and toxicology: a dialectic. In: Massaro E, editor. Handbook of human toxicology. Boca Raton (FL): CRC Press; 1997.
  • George J, Byth K, Farrell GC. Influence of clinicopathological variables on CYP protein expression in human liver. J Gastroenterol Hepatol. 1996 Jan;11(1):33–39.
  • Naito T, Tashiro M, Yamamoto K, et al. Impact of cachexia on pharmacokinetic disposition of and clinical responses to oxycodone in cancer patients. Eur J Clin Pharmacol. 2012 Oct;68(10):1411–1418.
  • Chen YL, Le Vraux V, Leneveu A, et al. Acute-phase response, interleukin-6, and alteration of cyclosporine pharmacokinetics. Clin Pharmacol Ther. 1994 Jun;55(6):649–660.
  • Williams ML, Bhargava P, Cherrouk I, et al. A discordance of the cytochrome P450 2C19 genotype and phenotype in patients with advanced cancer. Br J Clin Pharmacol. 2000 May;49(5):485–488.
  • Rivory LP, Slaviero KA, Clarke SJ. Hepatic cytochrome P450 3A drug metabolism is reduced in cancer patients who have an acute-phase response. Br J Cancer. 2002 Jul 29;87(3):277–280.
  • Yamaguchi A, Tateishi T, Okano Y, et al. Higher incidence of elevated body temperature or increased C-reactive protein level in asthmatic children showing transient reduction of theophylline metabolism. J Clin Pharmacol. 2000 Mar;40(3):284–289.
  • Frye RF, Schneider VM, Frye CS, et al. Plasma levels of TNF-alpha and IL-6 are inversely related to cytochrome P450-dependent drug metabolism in patients with congestive heart failure. J Card Fail. 2002 Oct;8(5):315–319.
  • Islam M, Frye RF, Richards TJ, et al. Differential effect of IFNalpha-2b on the cytochrome P450 enzyme system: a potential basis of IFN toxicity and its modulation by other drugs. Clin Cancer Res. 2002 Aug;8(8):2480–2487.
  • Rowe JW, Andres R, Tobin JD, et al. The effect of age on creatinine clearance in men: a cross-sectional and longitudinal study. J Gerontol. 1976 Mar;31(2):155–163.
  • Launay-Vacher V, Oudard S, Janus N, et al. Prevalence of renal insufficiency in cancer patients and implications for anticancer drug management: the renal insufficiency and anticancer medications (IRMA) study. Cancer. 2007 Sep 15;110(6):1376–1384.
  • WHO. Essential medicines in palliative care: executive summary. WHO Press, Geneva. 2013.
  • Olsen GD. Morphine binding to human plasma proteins. Clin Pharmacol Ther. 1975 Jan;17(1):31–35.
  • Le Couteur DG, McLean AJ. The aging liver. Drug clearance and an oxygen diffusion barrier hypothesis. Clin Pharmacokinet. 1998 May;34(5):359–373.
  • Lötsch J, Stockmann A, Kobal G, et al. Pharmacokinetics of morphine and its glucuronides after intravenous infusion of morphine and morphine-6-glucuronide in healthy volunteers. Clin Pharmacol Ther. 1996 Sep;60(3):316–325.
  • Christrup LL. Morphine metabolites. Acta Anaesthesiol Scand. 1997 Jan;41(1 Pt 2):116–122.
  • Donnelly S, Davis MP, Walsh D, et al. Morphine in cancer pain management: a practical guide. Supportive Care Cancer. 2002;10(1):13–35.
  • Glare PA, Walsh TD. Clinical pharmacokinetics of morphine. Ther Drug Monit. 1991 Jan;13(1):1–23.
  • Francés B, Gout R, Campistron G, et al. Morphine-6-glucuronide is more mu-selective and potent in analgesic tests than morphine. Prog Clin Biol Res. 1990;328:477–480.
  • Pasternak GW, Bodnar RJ, Clark JA, et al. Morphine-6-glucuronide, a potent mu agonist. Life Sci. 1987 Dec 28;41(26):2845–2849.
  • Paul D, Standifer KM, Inturrisi CE, et al. Pharmacological characterization of morphine-6 beta-glucuronide, a very potent morphine metabolite. J Pharmacol Exp Ther. 1989 Nov;251(2):477–483.
  • Klimas R, Mikus G. Morphine-6-glucuronide is responsible for the analgesic effect after morphine administration: a quantitative review of morphine, morphine-6-glucuronide, and morphine-3-glucuronide. Br J Anaesth. 2014 Jul 1;113:935–944.
  • Wu D, Kang YS, Bickel U, et al. Blood-brain barrier permeability to morphine-6-glucuronide is markedly reduced compared with morphine. Drug Metab Dispos. 1997 Jun;25(6):768–771.
  • Janicki PK, Erskine WA, James MF. The route of prolonged morphine administration affects the pattern of its metabolites in the urine of chronically treated patients. Eur J Clin Chem Clin Biochem. 1991 Jun;29(6):391–393.
  • Portenoy RK, Thaler HT, Inturrisi CE, et al. The metabolite morphine-6-glucuronide contributes to the analgesia produced by morphine infusion in patients with pain and normal renal function. Clin Pharmacol Ther. 1992 Apr;51(4):422–431.
  • Bartlett SE, Dodd PR, Smith MT. Pharmacology of morphine and morphine-3-glucuronide at opioid, excitatory amino acid, GABA and glycine binding sites. Pharmacol Toxicol. 1994 Aug;75(2):73–81.
  • Löser SV, Meyer J, Freudenthaler S, et al. Morphine-6-O-beta-D-glucuronide but not morphine-3-O-beta-D-glucuronide binds to mu-, delta- and kappa- specific opioid binding sites in cerebral membranes. Naunyn Schmiedebergs Arch Pharmacol. 1996 Jul;354(2):192–197.
  • Chen ZR, Irvine RJ, Somogyi AA, et al. Mu receptor binding of some commonly used opioids and their metabolites. Life Sci. 1991;48(22):2165–2171.
  • Penson RT, Joel SP, Bakhshi K, et al. Randomized placebo-controlled trial of the activity of the morphine glucuronides. Clin Pharmacol Ther. 2000 Dec;68(6):667–676.
  • Lewis SS, Hutchinson MR, Rezvani N, et al. Evidence that intrathecal morphine-3-glucuronide may cause pain enhancement via toll-like receptor 4/MD-2 and interleukin-1beta. Neuroscience. 2010 Jan 20;165(2):569–583.
  • Baillie SP, Bateman DN, Coates PE, et al. Age and the pharmacokinetics of morphine. Age Ageing. 1989;18:258–262.
  • Säwe J, Dahlström B, Paalzow L, et al. Morphine kinetics in cancer patients. Clin Pharmacol Ther. 1981 Nov;30(5):629–635.
  • Säwe J, Kager L, Svensson Eng JO, et al. Oral morphine in cancer patients: in vivo kinetics and in vitro hepatic glucuronidation. Br J Clin Pharmacol. 1985 Apr;19(4):495–501.
  • Faura CC, Collins SL, Moore RA, et al. Systematic review of factors affecting the ratios of morphine and its major metabolites. Pain. 1998 Jan;74(1):43–53.
  • Klepstad P, Dale O, Kaasa S, et al. Influences on serum concentrations of morphine, M6G and M3G during routine clinical drug monitoring: a prospective survey in 300 adult cancer patients. Acta Anaesthesiol Scand. 2003 Jul;47(6):725–731.
  • Osborne R, Joel S, Trew D, et al. Morphine and metabolite behavior after different routes of morphine administration: demonstration of the importance of the active metabolite morphine-6-glucuronide. Clin Pharmacol Ther. 1990 Jan;47(1):12–19.
  • Peterson GM, Randall CT, Paterson J. Plasma levels of morphine and morphine glucuronides in the treatment of cancer pain: relationship to renal function and route of administration. Eur J Clin Pharmacol. 1990;38(2):121–124.
  • Osborne R, Joel S, Grebenik K, et al. The pharmacokinetics of morphine and morphine glucuronides in kidney failure. Clin Pharmacol Ther. 1993 Aug;54(2):158–167.
  • D’Honneur G, Gilton A, Sandouk P, et al. Plasma and cerebrospinal fluid concentrations of morphine and morphine glucuronides after oral morphine. The influence of renal failure. Anesthesiology. 1994 Jul;81(1):87–93.
  • Chan GL, Matzke GR. Effects of renal insufficiency on the pharmacokinetics and pharmacodynamics of opioid analgesics. Drug Intell Clin Pharm. 1987 Oct;21(10):773–783.
  • Regnard CF, Twycross RG. Metabolism of narcotics. BMJ. 1984 Mar 17;288(6420):860.
  • Hoskin PJ, Hanks GW. The management of symptoms in advanced cancer: experience in a hospital-based continuing care unit. J R Soc Med. 1988 Jun;81(6):341–344.
  • Thompson PI, Joel SP, John L, et al. Respiratory depression following morphine and morphine-6-glucuronide in normal subjects. Br J Clin Pharmacol. 1995 Aug;40(2):145–152.
  • Peat SJ, Hanna MH, Woodham M, et al. Morphine-6-glucuronide: effects on ventilation in normal volunteers. Pain. 1991 Apr;45(1):101–104.
  • Romberg R, Olofsen E, Sarton E, et al. Pharmacodynamic effect of morphine-6-glucuronide versus morphine on hypoxic and hypercapnic breathing in healthy volunteers. Anesthesiology. 2003;99(4):788–798.
  • Morita T, Tei Y, Tsunoda J, et al. Increased plasma morphine metabolites in terminally ill cancer patients with delirium: an intra-individual comparison. J Pain Symptom Manage. 2002;23(2):107–113.
  • Sawe J. Morphine and its 3- and 6-glucuronides in plasma and urine during chronic oral administration in cancer patients. In: Foley KM, Lnturrisi CE, editors. Advances in pain research and therapy. New York (NY): Raven Press; 1986. p. 45–55.
  • Sjøgren P, Jonsson T, Jensen NH, et al. Hyperalgesia and myoclonus in terminal cancer patients treated with continuous intravenous morphine. Pain. 1993;55(1):93–97.
  • Gretton SK, Ross JR, Rutter D, et al. Plasma morphine and metabolite concentrations are associated with clinical effects of morphine in cancer patients. J Pain Symptom Manage. 2013;45(4):670–680.
  • Ferris DJ. Controlling myoclonus after high-dosage morphine infusions. Am J Health Syst Pharm. 1999 May 15;56(10):1009–1010.
  • Smith MT, Wright AW, Williams BE, et al. Cerebrospinal fluid and plasma concentrations of morphine, morphine-3-glucuronide, and morphine-6-glucuronide in patients before and after initiation of intracerebroventricular morphine for cancer pain management. Anesth Analg. 1999 Jan;88(1):109–116.
  • McCann S, Yaksh TL, Von Gunten CF. Correlation between myoclonus and the 3-glucuronide metabolites in patients treated with morphine or hydromorphone: a pilot study. J Opioid Manage. 2010;6(2):87–94.
  • Oosten AW, Oldenmenger WH, van Zuylen C, et al. Higher doses of opioids in patients who need palliative sedation prior to death: cause or consequence? Eur J Cancer. 2011 Oct;47(15):2341–2346.
  • Cherny NI. Sedation for the care of patients with advanced cancer. Nat Clin Pract Oncol. 2006;3(9):492–500.
  • Farriols C, Ferrández O, Planas J, et al. Changes in the prescription of psychotropic drugs in the palliative care of advanced cancer patients over a seven-year period. J Pain Symptom Manage. 2012 May;43(5):945–952.
  • Cherny NI, Grp EGW. ESMO Clinical Practice Guidelines for the management of refractory symptoms at the end of life and the use of palliative sedation. Ann Oncol. 2014 Sep;25:143–152.
  • Jamei M, Turner D, Yang J, et al. Population-based mechanistic prediction of oral drug absorption. AAPS J. 2009 Jun;11(2):225–237.
  • Brill MJ, van Rongen A, Houwink AP, et al. Midazolam pharmacokinetics in morbidly obese patients following semi-simultaneous oral and intravenous administration: a comparison with healthy volunteers. Clin Pharmacokinet. 2014 Oct;53(10):931–941.
  • Reves JG, Fragen RJ, Vinik HR, et al. Midazolam: pharmacology and uses. Anesthesiology. 1985 Mar;62(3):310–324.
  • Moschitto LJ, Greenblatt DJ. Concentration-independent plasma protein binding of benzodiazepines. J Pharm Pharmacol. 1983 Mar;35(3):179–180.
  • Kanto JH. Midazolam: the first water-soluble benzodiazepine. Pharmacology, pharmacokinetics and efficacy in insomnia and anesthesia. Pharmacotherapy. 1985 May–Jun;5(3):138–155.
  • Maitre PO, Funk B, Crevoisier C, et al. Pharmacokinetics of midazolam in patients recovering from cardiac surgery. Eur J Clin Pharmacol. 1989;37(2):161–166.
  • Malacrida R, Fritz ME, Suter PM, et al. Pharmacokinetics of midazolam administered by continuous intravenous infusion to intensive care patients. Crit Care Med. 1992 Aug;20(8):1123–1126.
  • Greenblatt DJ, Abernethy DR, Locniskar A, et al. Effect of age, gender, and obesity on midazolam kinetics. Anesthesiology. 1984 Jul;61(1):27–35.
  • Brill MJ, van Rongen A, van Dongen EP, et al. The pharmacokinetics of the CYP3A substrate midazolam in morbidly obese patients before and one year after bariatric surgery. Pharm Res. 2015 Jul 23;32:3927–3936.
  • van Rongen A, Vaughns JD, Moorthy G, et al. Population pharmacokinetics of midazolam and its metabolites in overweight and obese adolescents. Br J Clin Pharmacol. 2015 Jun 4;80:1185–1196.
  • Halliday NJ, Dundee JW, Collier PS, et al. Influence of plasma proteins on the onset of hypnotic action of intravenous midazolam. Anaesthesia. 1985 Aug;40(8):763–766.
  • Cvan Trobec K, Kerec Kos M, Trontelj J, et al. Influence of cancer cachexia on drug liver metabolism and renal elimination in rats. J Cachexia Sarcopenia Muscle. 2015 Mar;6(1):45–52.
  • Albarmawi A, Czock D, Gauss A, et al. CYP3A activity in severe liver cirrhosis correlates with Child-Pugh and model for end-stage liver disease (MELD) scores. Br J Clin Pharmacol. 2014;77(1):160–169.
  • Bleasel MD, Peterson GM, Dunne PF. Plasma concentrations of midazolam during continuous subcutaneous administration in palliative care. Palliat Med. 1994;8(3):231–236.
  • Matsunaga T, Maruyama M, Matsubara T, et al. Mechanisms of CYP3A induction by glucocorticoids in human fetal liver cells. Drug Metab Pharmacokinet. 2012;27(6):653–657.
  • McCune JS, Hawke RL, LeCluyse EL, et al. In vivo and in vitro induction of human cytochrome P4503A4 by dexamethasone. Clin Pharmacol Ther. 2000 Oct;68(4):356–366.
  • Bauer TM, Ritz R, Haberthür C, et al. Prolonged sedation due to accumulation of conjugated metabolites of midazolam. Lancet. 1995 Jul 15;346(8968):145–147.
  • Prommer E. Role of haloperidol in palliative medicine: an update. Am J Hosp Palliat Care. 2012 Jun;29(4):295–301.
  • Bartz L, Klein C, Seifert A, et al. Subcutaneous administration of drugs in palliative care: results of a systematic observational study. J Pain Symptom Manage. 2014 Apr 21;48:540–547.
  • Kudo S, Ishizaki T. Pharmacokinetics of haloperidol: an update. Clin Pharmacokinet. 1999 Dec;37(6):435–456.
  • Nishimuta H, Nakagawa T, Nomura N, et al. Significance of reductive metabolism in human intestine and quantitative prediction of intestinal first-pass metabolism by cytosolic reductive enzymes. Drug Metab Dispos. 2013 May;41(5):1104–1111.
  • Forsman A, Ohman R. Studies on serum protein binding of haloperidol. Curr Ther Res Clin Exp. 1977 Feb;21(2):245–255.
  • Rowell FJ, Hui SM, Fairbairn AF, et al. Total and free serum haloperidol levels in schizophrenic patients and the effect of age thioridazine and fatty acid on haloperidol-serum protein binding in vitro. Br J Clin Pharmacol. 1981 Apr;11(4):377–382.
  • Someya T, Shibasaki M, Noguchi T, et al. Haloperidol metabolism in psychiatric patients: importance of glucuronidation and carbonyl reduction. J Clin Psychopharmacol. 1992 Jun;12(3):169–174.
  • Fang J, Baker GB, Silverstone PH, et al. Involvement of CYP3A4 and CYP2D6 in the metabolism of haloperidol. Cell Mol Neurobiol. 1997 Apr;17(2):227–233.
  • Midha KK, Hawes EM, Hubbard JW, et al. Interconversion between haloperidol and reduced haloperidol in humans. J Clin Psychopharmacol. 1987 Oct;7(5):362–364.
  • Chakraborty BS, Hubbard JW, Hawes EM, et al. Interconversion between haloperidol and reduced haloperidol in healthy volunteers. Eur J Clin Pharmacol. 1989;37(1):45–48.
  • Jann MW, Lam YW, Chang WH. Reversible metabolism of haloperidol and reduced haloperidol in Chinese schizophrenic patients. Psychopharmacology (Berl). 1990;101(1):107–111.
  • Kudo S, Odomi M. Involvement of human cytochrome P450 3A4 in reduced haloperidol oxidation. Eur J Clin Pharmacol. 1998 May;54(3):253–259.
  • Pan LP, De Vriendt C, Belpaire FM. In-vitro characterization of the cytochrome P450 isoenzymes involved in the back oxidation and N-dealkylation of reduced haloperidol. Pharmacogenetics. 1998 Oct;8(5):383–389.
  • Johnson PC Jr, Charalampous KD, Braun GA. Absorption and excretion of tritiated haloperidol in man. (A preliminary report). Int J Neuropsychiatry. 1967 Aug;3(Suppl 1):24–25.
  • Anderson RJ, Gambertoglio JG, Schrier RW. Clinical use of drugs in renal failure. Springfield (IL): Thomas; 1976.
  • Forsman AO. Individual variability in response to haloperidol. Proc R Soc Med. 1976;69(suppl 1):9–12.
  • Jann MW, Ereshefsky L, Saklad SR, et al. Effects of carbamazepine on plasma haloperidol levels. J Clin Psychopharmacol. 1985 Apr;5(2):106–109.
  • Kidron R, Averbuch I, Klein E, et al. Carbamazepine-induced reduction of blood levels of haloperidol in chronic schizophrenia. Biol Psychiatry. 1985 Feb;20(2):219–222.
  • Arana GW, Goff DC, Friedman H, et al. Does carbamazepine-induced reduction of plasma haloperidol levels worsen psychotic symptoms? Am J Psychiatry. 1986 May;143(5):650–651.
  • Hesslinger B, Normann C, Langosch JM, et al. Effects of carbamazepine and valproate on haloperidol plasma levels and on psychopathologic outcome in schizophrenic patients. J Clin Psychopharmacol. 1999 Aug;19(4):310–315.
  • Mihara K, Suzuki A, Kondo T, et al. Effects of the CYP2D6*10 allele on the steady-state plasma concentrations of haloperidol and reduced haloperidol in Japanese patients with schizophrenia. Clin Pharmacol Ther. 1999 Mar;65(3):291–294.
  • Goff DC, Midha KK, Brotman AW, et al. Elevation of plasma concentrations of haloperidol after the addition of fluoxetine. Am J Psychiatry. 1991 Jun;148(6):790–792.
  • Viala A, Aymard N, Leyris A, et al. [Pharmaco-clinical correlations during fluoxetine administration in patients with depressive schizophrenia treated with haloperidol decanoate]. Therapie. 1996 Jan–Feb;51(1):19–25. French.
  • Goff DC, Midha KK, Sarid-Segal O, et al. A placebo-controlled trial of fluoxetine added to neuroleptic in patients with schizophrenia. Psychopharmacology (Berl). 1995 Feb;117(4):417–423.
  • Avenoso A, Spinà E, Campo G, et al. Interaction between fluoxetine and haloperidol: pharmacokinetic and clinical implications. Pharmacol Res. 1997 Apr;35(4):335–339.
  • Gysels MH, Evans C, Higginson IJ. Patient, caregiver, health professional and researcher views and experiences of participating in research at the end of life: a critical interpretive synthesis of the literature. BMC Med Res Methodol. 2012;12:123.
  • Ohde S, Hayashi A, Takahasi O, et al. A 2-week prognostic prediction model for terminal cancer patients in a palliative care unit at a Japanese general hospital. Palliat Med. 2011 Mar;25(2):170–176.