7. PHARMACOLOGY

Published on 09/04/2015 by admin

Filed under Hematology, Oncology and Palliative Medicine

Last modified 09/04/2015

Print this page

rate 1 star rate 2 star rate 3 star rate 4 star rate 5 star
Your rating: none, Average: 0 (0 votes)

This article have been viewed 1530 times

CHAPTER 7. PHARMACOLOGY
Phyllis A. Grauer
B9781416030799500154/f07-01-9781416030799.jpg is missing
Figure 7-1

(From Kuebler, K.K., Davis, M.P., & Moore, C.D. [2005]. Palliative practices: An interdisciplinary approach. St. Louis: Elsevier Mosby, Figure 4-1.)
Clinical pharmacokinetics examines the effects of the body on a drug, specifically absorption, distribution, metabolism, and excretion of drugs. Factors that influence these processes include the following:
▪ How quickly a drug is absorbed into the blood and how different dosages affect that absorption
▪ How the drug is distributed into organs or tissues of the body and to the site of action
▪ How the body metabolizes the drug and whether the drug is changed by the body into an active or inactive compound
▪ How long it takes the body to metabolize and eliminate half of the drug (the drug’s half-life)
▪ How long it takes the drug to be excreted from the body
Pharmacodynamics is the study of the body’s reaction to drugs. This area of pharmacology evaluates the body’s response to pharmacological, biochemical, physiological, and therapeutic effects of a drug. Basically, pharmacodynamics is the study of the activity of drugs on receptor sites within the body resulting in a clinical effect.
Of note, “pharmacogenomics” is an emerging area of study. The focus of pharmacogenomics is the identification of variations in the human genome (a total gene complement) that affect the response to medications. Advances in pharmacogenomics may permit drugs to be tailor-made for individuals and adapted to each person’s genetic makeup. Although environment, diet, age, lifestyle, and state of health all can influence a patient’s response to specific medications, understanding an individual’s genetic makeup is thought to be key in creating personalized drugs that would provide patients greater efficacy and safety. Because of the infancy and complexity of pharmacogenomics, it is beyond the scope of this chapter.
For the clinician, rational incorporation of the principles of pharmacology into therapeutic decision making will result in achieving the desired therapeutic outcome while preventing adverse drug events and promoting optimal symptom management.

UNDERSTANDING PHARMACOKINETIC PARAMETERS

Once a drug enters the circulatory system, it is distributed to tissues, reabsorbed into the bloodstream, and then eliminated from the body. Pharmacokinetic parameters define the factors that affect drug concentration within the human body over time.

Routes of Administration

A myriad of factors affect the dosage and matrix used in medications that influence the drug’s delivery to the specific site of action. These factors include the drug’s ability to penetrate barriers (i.e., the wall of the gastrointestinal tract and skin), the stability of the drug in acid environments such as the stomach (pH 2), the degree of tissue irritation when the drug is administered intramuscularly or subcutaneously, and the fraction of drug that is inactivated by the first pass through the liver. Table 7-1 describes the most common routes of administration.
TABLE 7-1 Characteristics of Various Routes of Administration
Data from Olson, J (2003). Clinical pharmacology made ridiculously simple (2nd ed.). Miami, Fla: MedMaster. © MedMaster2003
Route Characteristics
Oral (PO) Drug must be dispersed in solid dosage form to permeate the gastrointestinal lining and enter circulatory system.
Most is absorbed in small intestine, where there is less acidic environment.
Rate of absorption is dependent on gastric emptying and intestinal motility.
Extent of absorption is dependent on drug’s ability to permeate gastrointestinal lining and enter circulatory system.
Drug enters portal circulation, passes through liver, and therefore is subject to hepatic extraction and metabolism.
Drugs inactivated by acidic environment of stomach are typically enteric coated to prevent contact with stomach acid. Once the drug enters the less acidic environment of small intestine, the enteric coating dissolves, allowing drug to be dispersed and then absorbed.
Extended-release drugs use various forms of pharmaceutically prepared release mechanisms so drug is released from oral dosage form over time.
Sublingual (SL) This route avoids contact with acidic stomach environment.
Drug is absorbed through mucosa under the tongue and enters bloodstream through numerous capillary beds.
Much drug that is absorbed sublingually bypasses the liver.
There is greater lipophilicity of drug and more is completely absorbed sublingually.
Rectal (PR) Rectal mucosa is fed by blood vessels that pass through liver and by blood vessels that avoid portal circulation.
Percent of drug absorbed through each system depends on where drug is placed in rectum.
Many drugs administered rectally have erratic and often unpredictable absorption.
Do not administer drugs dependent on constant serum concentration within a narrow therapeutic range (e.g., phenytoin, digoxin, warfarin).
Transdermal (TD) Skin is the body’s strongest barrier against absorption of toxins from environment into systemic circulation.
Few drugs will penetrate skin and be absorbed into subcutaneous capillary beds.
Extent of absorption is dependent on lipophilicity and drug’s molecular structure.
Amount absorbed is determined by surface area to which it is applied.
Drugs administered topically for systemic absorption are best formulated in predetermined patch sizes (e.g., fentanyl TM patch).
Intravenous (IV) Drug has rapid onset of action.
Rate-limiting step is time it takes to reach site of action and produce therapeutic effect.
Only soluble drugs are able to be administered by IV injection.
Drugs administered IV are not affected by first-pass liver extraction and inactivation.
Intramuscular (IM) Rate at which a drug is absorbed from muscle into bloodstream is dependent on type of diluent used to prepare drug formulation.
Oil-based drugs are typically absorbed more slowly that those in aqueous solution.
Drug is not affected by first-pass liver extraction and inactivation.
Subcutaneous (SC) Route is used for drugs that are not irritating to surrounding tissue and where volume of drug product administered does not typically exceed 2 ml of fluid.
Type of formulation used should determine how rapidly drug is absorbed into capillary walls and enters circulatory system.
Drug is not affected by first-pass liver extraction and inactivation.
Intraspinal Some drugs that act on the central nervous system can be administered epidurally and intrathecally.
Route often allows for decreased dosage requirements and localized action, reducing intensity of adverse effects.
Route can be used for opioids and other adjuvant pain medications such as local anesthetics.
Inhalation (INH) Drug is generally absorbed rapidly if particles are small.
Multidose inhalers require good administration technique in order to deliver drug through bronchial tree to alveolar bed for absorption.
Nebulizer administration of drug is less dependent on technique and is more efficacious in patients who are weak and debilitated (although absorption is less).
Topical (TOP) Route is typically intended to exert action locally and considered to avoid systemic absorption.
Sites of action include skin, eyes, nose, ears, and vaginal and rectal tissues.
It is important to note that there are several determinants associated with oral medication use that may interfere with the drug’s absorption from the gastric mucosa. These determinants include dissolution, gastric emptying time, intestinal motility, drug interactions in the gut lumen, and passage through the gut wall. Box 7-1 outlines these in further detail.
Box 7-1

McGraw-Hill Australia Pty. Ltd.

DISSOLUTION

• Physical/chemical properties of the drug
• Crystal size and form
• Excipients (e.g., tablet fillers such as lactose)
• Dosage forms (enteric coated, sustained-release formulations)
• pH of the stomach and intestines

GASTRIC EMPTYING RATE

• Stability of the drug in an acid pH
• Solution or solid dosage forms (liquids and small particles empty more quickly)
• Presence of food or antacids
• Drugs (opioids and anticholinergics slow emptying time, metoclopramide increases emptying time)
• Disease (autonomic nervous system abnormalities such as Parkinson’s disease)
• Intestinal interactions in the gut
• Formation of complexes (tetracyclines and divalent metal compounds, e.g., Al 2+)
• Absorption (ion exchange resins, cholestyramine)
• Food (i.e., dairy products, proteins) (many antibiotics)

PASSAGE THROUGH THE GUT WALL

• Physical/chemical characteristics of the drug (quaternary ammonium compound, vancomycin)
• Metabolism by enzymes in the intestinal endothelium
Modified from Birkett, D.J. (2003). Pharmacokinetics made easy. North Ryde, Australia: McGraw-Hill Australia Pty. Ltd., Table 5-1, p. 36.
At the end of life when patients are unable to swallow and/or a parenteral route is unavailable, many drugs used for the treatment of symptoms in palliative care can be given via alternative routes of administration. Although there is a lack of published information and controlled studies regarding these alternate routes of administration, knowledge of the characteristics of the drug, including lipophilicity, molecular weight, and p Ka, can help the pharmacist predict whether a drug is likely to be absorbed via a particular route. Literature supports both benzodiazepines such as lorazepam and diazepam and certain opioids such as methadone and fentanyl as being well absorbed into the sublingual capillaries, whereas morphine is absorbed to a much lesser extent through the sublingual mucosa (Akinbi & Welty, 1999; Weinberg, Inturrisi, Reidenberg et al., 1988). The majority of the effect of morphine, as well as that of oxycodone and hydromorphone, occurs when the sublingually administered drug trickles down the esophagus and is absorbed by the gastrointestinal tract (Akinbi & Welty, 1999; Weinberg et al., 1988). Methadone administered rectally has a more rapid onset of action than does oral methadone, making it a feasible alternative when the oral and parenteral routes are not options (Dale, Sheffels, & Kharasch, 2004). Knowledge of the extent of absorption through each route of administration is imperative in order to appropriately guage the dose of medication (Katzung, 2003). The quantitative measure of the absorption of a drug into the circulation is known as the drug’s bioavailability.

Bioavailability

The bioavailability of a drug is the fraction of the administered dose that reaches the systemic circulation. For example, the bioavailability of an intravenous injection is 100% (Birkett, 2003), whereas bioavailability will vary for other routes of administration depending on factors that affect the extent of absorption into the circulatory system and first-pass hepatic metabolism. The measurement that determines absolute bioavailability is called the area under the curve (AUC). This measurement is determined by calculating the AUC of the plasma concentration plotted over time (Birkett, 2003) (Figure 7-2).
B9781416030799500154/f07-02-9781416030799.jpg is missing
Figure 7-2

(From Birkett, D.J. [2003]. Pharmacokinetics made easy. North Ryde, Australia: McGraw-Hill Australia Pty. Ltd., Figure 1-2.)
McGraw-Hill Australia
The bioavailability of a specific drug is the determinant of the dosage and is equivalent to the drug administered via various routes of administration. For example, chronic use of morphine administered orally has a bioavailability of around 20% to 30% of the parenteral dose (100% bioavailability) (Doyle, Hanks, Cherny et al., 2005). This means that a patient who has been receiving 10 mg of morphine intravenously will require approximately 30 mg of morphine orally to achieve the same analgesic effect as experienced from the intravenous dose (American Pain Society, 2003).
Drugs are considered bioequivalent when the extent and rate of absorption are similar and there is no difference between the therapeutic and adverse effects. A generic drug company, for example, that manufactures a drug in the same dosage form as a brand-name product will often use the AUC bioavailability data when comparing the two drugs. For a generic drug to be considered equivalent to the brand drug, the bioavailability must be similar to that of the brand product (USDHHS-FDA, 2006). The Electronic Orange Book is an online publication of the U.S. Department of Health and Human Services Food and Drug Administration that lists approved drug products and their therapeutic equivalence evaluations.

Therapeutic Range

Therapeutic range is defined as the plasma concentration that occurs between the concentration of drug needed to achieve the desired pharmacological effect and the concentration where adverse effects are observed (Figure 7-3). For some drugs, this range is narrow, and for other drugs, it is wide. The narrower the range, the more monitoring is needed to prevent adverse drug effects or clinical misadventures. Some monitoring is done based on plasma drug concentrations (e.g., gentamicin, theophylline, and digoxin). Other drug monitoring is done by measuring physiological changes that are caused by the drug (e.g., measuring international normalized ratio for Coumadin [warfarin] and thyroid-stimulating hormone for levothyroxine).
B9781416030799500154/f07-03-9781416030799.jpg is missing
Figure 7-3

(From Adams, M.P., Josephson, D.L., & Holland, L.N. [2005]. Pharmacology in nursing: A pathophysiologic approach. © 2005, pp. 53, 54. Reprinted by permission of Pearson Education, Inc., Upper Saddle River, N.J.)
Pearson Education, Inc.

Volume of Distribution

Once the drug is absorbed into the systemic circulation, it is distributed throughout the body. The apparent volume of distribution (V D) is a measure of where the drug goes once it is completely distributed throughout the body (Table 7-2). It is the ratio of the fraction of drug unbound to protein in the plasma to the fraction of unbound drug in the tissue, and it is expressed as liters per kilogram (L/kg). The volume of distribution is also determined by the strength of binding of the drug to plasma proteins in relationship to the strength of binding to tissue components. If a drug is highly bound by tissue and not by blood, it will allow most of the drug to be held in the tissues of the body and little will be held within the blood. In this case, the drug will have a large volume of distribution. The V D is measured by plotting the logarithm of the plasma concentration against time. The result is a straight line that can be extrapolated back to zero (Birkett, 2003). The plasma concentration at zero time (C 0) divided by the dose equals the volume of distribution:
TABLE 7-2 Examples of Volumes of Distribution (V D) and Half-life ( t½)
Data from Lexi-Comp Online. (2006). Retrieved March 30, 2006, from www.lexi.com. © 2006
*Biphasic elimination: initial (terminal).
Data from Lexi-Comp Online. (2006). Retrieved March 30, 2006, from www.lexi.com.
Drug V D (L/kg) V D/70 kg (L) t½ (hr)
Morphine 3.3 (3-4) 230 2-4 (Immediate release)
Lorazepam 1.3 91 12-16
Diazepam 1.1 77 20-50
Chlorpromazine 20 1400 2 (30) *
Haloperidol 20 1400 20
Fentanyl 6 420 2–4 (IV), 17 (TD)
Methadone 4 (1-8) 280 7-59
Warfarin 7 (6-7) 490 20-60
Digoxin 7 (6-7) 490 37-48
B9781416030799500154/si1.gif is missing

Loading Doses

A loading dose (LD) can be used when attempting to achieve a rapid concentration of a specific drug. The volume of distribution is the pharmacokinetic parameter used when calculating an LD of a drug. For example, if a drug has a V D of 40 liters (L) and the desired concentration (c) is 10 mg/L, then a loading dose to achieve that concentration is 400 mg:
B9781416030799500154/si2.gif is missing

Half-Life

The half-life ( t1/2) is identified by the time it takes for the plasma concentration and the amount of drug within the body to fall by half after it has undergone absorption and distribution. Half-life is the reciprocal function of the elimination rate constant. The half-life and elimination rate constant are determined by both clearance (CL) and V D (Urso, Blardi, & Giorgi, 2002) (Figure 7-4). Half-life determines the duration of action after a single dose of a drug and the amount of time required to reach steady state with constant dosing.
B9781416030799500154/f07-04-9781416030799.jpg is missing
Figure 7-4

(From Adams, M.P., Josephson, D.L., & Holland, L.N. [2005]. Pharmacology in nursing: A pathophysiologic approach. © 2005, pp. 53, 54. Reprinted by permission of Pearson Education, Inc., Upper Saddle River, NJ.)
Pearson Education, Inc.
The therapeutic concentration range, also known as the therapeutic window, is the drug concentration range where most patients will have a therapeutic effect with the least amount of adverse effects. In general, the usual therapeutic range and drug effect are proportional to the logarithm of drug concentration. Therefore, drug effect after a single dose usually declines in a linear relationship over time. Notwithstanding, a number of mechanisms result in a dissociation of the usual relationship between drug concentration and effect. Changes in volume of distribution due to dehydration of overhydration and changes in renal or liver function are examples of factors that will affect the drug concentration–and–effect relationship.

Steady State

Steady state occurs when the rate of drug administered is equal to the rate of elimination from the body. It generally takes three to five drug half-lives to reach steady state. For example, when immediate-release morphine ( t1/2 ≈4 hours) is administered routinely every 4 hours, steady state is achieved within 24 hours. For a drug with a longer half-life, such as methadone ( t1/2 ≈20 to 35 hours), steady state is achieved in approximately 6 to 10 days (Lugo, Satterfield, & Kern, 2006).
Drug concentrations are often measured as unbound plasma drug concentrations rather than as whole blood concentrations. With oral and intermittent dosing, once the steady state has been achieved, despite a fluctuation in drug doses, the amount of drug administered will equal the amount of drug eliminated. This results in an average drug concentration that is equal to an intravenous infusion (Olson, 2003).
With intermittent dosing, when the dosing interval is equal to the half-life of the drug, the result is about a two-fold fluctuation in drug concentration over the dosing interval. In cases where the drug is administered orally and at an interval that is not equal to the drug’s half-life, the degree of plasma concentration fluctuation over the dosing interval is determined by both the absorption rate and the relationship of the dosing interval to the half-life. Although a drug may be at steady state, if the dose is changed, it again will take three to five half-lives to reach a new steady state. Conversely, upon discontinuation of a dose, it takes four half-lives to eliminate 94% of the drug from the body (Birkett, 2003; Olson, 2003).

Dosing Intervals

To achieve a sustained systemic blood level of medication that has a short half-life and a narrow therapeutic window, drugs are often formulated in an oral sustained-release matrix. Morphine is an example of a drug that is available in an immediate-release form (i.e., on administration, the entire dose is available to be absorbed and distributed to the site of action) and in a sustained-release format where systemic blood levels are available for 8, 12, and 24 hours after ingestion of a single-unit dose. When morphine is administered as an immediate-release dose, it is typically dosed at an interval close to its half-life (i.e., every 4 hours) to maintain a constant therapeutic systemic level. Sustained-release morphine has the same half-life and clearance as immediate-release morphine. The sustained-release matrix formulation is designed to gradually release drug, resulting in a fraction of morphine available for absorption and distribution. Consequently, altering the sustained-release matrix through crushing, for example, will destroy the delayed absorption property and result in a bolus of the entire dosage, leading to loss of a sustained serum concentration and potentially toxicity (Olson, 2003).

Clearance

Medication clearance (CL) is the most important pharmacokinetic property of a drug. It is the measure of the efficiency of irreversible elimination of the drug from the systemic circulation and is expressed as the volume of blood cleared of unchanged drug per unit of time. Furthermore, it is the sum of drug clearance from systemic circulation, which includes the body’s vital organs. CL determines the steady-state concentration of a drug for a given dose rate.
B9781416030799500154/si3.gif is missing
However, if a drug is administered intermittently, as in the case of oral dosing, C ss is determined by
B9781416030799500154/si4.gif is missing

where F is the fraction of the dose absorbed into the systemic circulation, D is the dose of the drug, and τ is the dosage interval.

It is important for the clinician to note that the clearance of a drug can be altered by several factors, including liver and kidney insufficiency, changes in protein and tissue binding, and the concomitant administration of other medications.

Linear Pharmacokinetics

Once a drug is in the body, the process of elimination begins. The majority of drugs are eliminated by “first-order,” or linear, pharmacokinetics. This process of elimination is exponential or logarithmic (Figure 7-5). As an example of linear pharmacokinetics, when a dose is doubled from 200 mg/day to 400 mg/day, the patient’s serum drug concentration also doubles (Birkett, 2003).
Buy Membership for Hematology, Oncology and Palliative Medicine Category to continue reading. Learn more here