Comprehensive 22-32 Eng
Comprehensive 22-32 Eng
Agents for
Constipation, Diarrhea,
Hemorrhoids, and
Heartburn
JENNIFER D. SMITH
I. CONSTIPATION
A. General information
1. Definition. Constipation is defined as a decrease in the frequency of fecal elimination and is
characterized by the passage of hard, dry, and sometimes painful stools. Normal stool
frequency ranges from three times daily to three times per week. Patients may experience
abdominal bloating, headaches, low back pain, and/or a sense of rectal fullness from
incomplete evacuation of feces.
2. Epidemiology
a. Age. Constipation is common in all age groups, however; there is a higher prevalence in
people 65 years of age.
b. Gender. Women suffer from constipation more often than men.
3. Causes. Constipation can be caused by many factors, including the following:
a. Lifestyle. A diet insufficient in fiber and/or inadequate fluid intake, lack of exercise, and
poor bowel habits, such as failure to respond to the defecatory urge or hurried bowels (i.e.,
incomplete evacuation) can contribute to constipation.
b. Medications, such as narcotic analgesics, diuretics, or anticholinergics (e.g.,
antidepressants, antihypertensives, antihistamines, phenothiazines, antispasmodics). In
addition, nonpre-scription medications such as iron supplements, calcium- or aluminum-
containing antacids, nonprescription nonsteroidal anti-inflammatory drugs (NSAIDs), and
histamine-2 receptor antagonists (H2RAs; i.e., ranitidine) may contribute to constipation.
c. Pregnancy is a common contributor to constipation. The increased size of the uterus, hor-
monal changes, intake of calcium- and iron-containing prenatal vitamins, and a reduction in
physical activity are all considered contributing factors.
d. Systemic disorders, such as intestinal obstruction, tumor, inflammatory bowel disease,
diver-ticulitis, hypothyroidism, hyperglycemia, irritable bowel syndrome, cerebrovascular
disease, or Parkinson disease.
4. Assessment should include normal stool frequency, frequency and duration of the constipation,
exercise routine, daily dietary fiber and fluid intake, presence of other symptoms, and current
medications.
455
456 Chapter 23 I. B
B. Treatment
1. Nonpharmacologic
a. Increase intake of fluids (at least eight 8-oz servings of noncaffeinated fluids daily) and fiber
(e.g., whole-grain breads and cereals, beans, prunes, peas, carrots, corn). The recommended
adult intake of fiber is 20 to 35 g/day, but individuals who are consuming inadequate amounts of
fiber should increase the amount of fiber intake slowly to prevent gastrointestinal distress.
b. Increase exercise to increase and maintain bowel tone.
c. Bowel training to increase regularity (i.e., allowing regular and adequate time for defecation).
2. Pharmacologic recommendations for simple constipation should begin as a step-wise
approach with bulk-forming laxatives as first line, hyperosmotic laxatives as second line,
and then stimulant laxatives if the previous recommendations were ineffective or intolerable.
Self-care for the treatment of constipation should be limited to 7 days.
a. Bulk-forming laxatives are natural or synthetic polysaccharide or cellulose derivatives that
adsorb water to soften the stool and increase bulk, which stimulates peristalsis.
(1) Availability. Bulk-forming agents include psyllium (e.g., Metamucil, Konsyl,
Fiberall), methylcellulose (e.g., Citrucel), calcium polycarbophil (e.g., Konsyl Fiber
caplets, FiberCon), and wheat dextran (e.g., Benefiber). All bulk-forming agents must
be given with at least 8 oz of water to prevent choking.
(2) Onset of action is slow and ranges from 12 to 24 hrs up to 72 hrs.
(3) Adverse effects: Bloating, abdominal cramping, and flatulence
(4) Warnings and counseling points
(a) Bulk-forming agents should not be used if an obstructing bowel lesion, intestinal
strictures, or Crohn’s disease are present due to the potential to worsen the
situation or result in bowel perforation. Additionally, because of the administration
guide-lines, bulk-forming laxatives may be inappropriate for individuals with
severe fluid restrictions, such as those with heart failure or renal impairment.
(b) Sugar-free formulations should be considered for individuals with diabetes but
avoided in individuals with phenylketonuria.
(c) The use of bulk-forming laxatives for constipation relief should be limited to 1
week; however, they can be used on a long-term basis for prevention.
b. Hyperosmotic laxatives work by creating an osmotic gradient to pull water into the small
and large intestines, resulting in increased peristalsis and bowel motility.
(1) Availability
(a) Glycerin (e.g., Fleet Glycerin Suppositories, Fleet Pedia-Lax) is only safe and effective when
used rectally. Dosing for ages 6 years to adult is 2 g regular suppository or 5.4 g in liquid
suppository; ages 2 to 5 years is 1 g regular suppository or 2.8 g in liquid suppository.
(b) Polyethylene glycol 3350 (e.g., Miralax, Dulcolax Balance) is approved for self-care use
in ages 17 years and older. It is dosed as 17 g, stirred into 4 to 8 oz of fluid, once daily.
(2) Onset of action varies greatly between the two nonprescription items in this class.
Glycerin suppositories have an onset range of 15 mins to 1 hr and polyethylene
glycol has a longer onset of 1 to 3 days.
(3) Adverse effects. As expected, rectal burning may occur with glycerin products;
polyeth-ylene glycol causes adverse effects at excessive doses and can include diarrhea,
nausea, bloating, cramping, and flatulence.
(4) Warning. Avoid use in individuals with kidney disease, irritable bowel syndrome,
during pregnancy, or while breastfeeding.
c. Stimulant laxatives work by altering water and electrolyte transport by the intestines and
by stimulating bowel motility (i.e., propulsive peristaltic activity). They are recommended
for relief of constipation when an individual has failed or is intolerant to bulk-forming or
hyperosmotic agents. However, stimulant laxatives are being used more frequently as first-
line therapy for opiate-induced chronic constipation.
(1) Availability. The two main classes of stimulant laxatives are anthraquinones (e.g.,
senna, sennosides) and diphenylmethane (e.g., bisacodyl).
(a) Senna can be taken as single-entity products (e.g., Senokot, Ex-Lax, Perdiem,
Fletcher’s Laxative for Kids) or combined with a stool softener (e.g., Senokot-S, Peri-
Colace) for relief of constipation. Senna is available as a standardized concentrate or
Over-the-Counter Agents for Constipation, Diarrhea, Hemorrhoids, and Heartburn 457
as sennosides, which are derived from the senna leaves. Adult dosing in solid oral
formulations is 187 to 374 mg standardized concentrate and 8.6 to 17.2 mg sen-
nosides. Senna is appropriate for self-care for pediatrics (ages 2 and older) and is
available as a syrup, tablet, or chocolate chew.
(b) Bisacodyl (e.g., Dulcolax, Correctol) is available in rectal and oral formulations.
Adult doses are 5 to 15 mg by mouth or 10 mg rectally once daily. Pediatric dose
(ages 6 and older) is 5 mg orally or rectally once daily.
(2) Onset of action. The onset of action of the oral preparation is within 6 to 12 hrs, but
the rectal preparation onset of action is quicker, within 15 to 60 mins.
(3) Adverse effects. All stimulant laxatives can cause abdominal cramping. Electrolyte
and fluid deficiencies, enteric loss of protein, malabsorption, and hypokalemia are
additional possible adverse effects. The suppository form may cause rectal burning.
(4) Warnings and counseling points
(a) Individuals with undiagnosed rectal bleeding or signs of intestinal obstruction
should not use stimulant laxatives.
(b) Cathartic colon, which results in a poorly functioning colon, has been associated
with the chronic use of stimulant laxatives. However, there is no convincing
evidence that chronic use impairs the colon in structure or function. Caution should
still be taken with the chronic use of stimulant laxatives due to the other associated
adverse effects noted above.
(c) Sennosides may cause discoloration of the urine (pink/red, yellow, or brown), but
this is a harmless effect.
(d) Tablet formulations of bisacodyl should not be crushed or chewed due to the enteric
coating. Milk, H2RA (e.g., ranitidine, cimetidine, famotidine), and antacids may
erode this enteric coating and should be separated in dosing by at least 1 hr.
d. Emollient laxatives, also known as surfactants, work by allowing water to move more
easily into the stool. This creates a softer stool, which is easier to pass. Thus, these agents
are par-ticularly useful in those who must avoid straining to pass hard stools (e.g., recent
myocardial infarction, rectal surgery). Emollient laxatives have very few side effects, but
they are not as effective as other laxatives.
(1) Availability. Emollient laxatives are salts of the surfactant docusate. These products
con-tain insignificant amounts of calcium, sodium, or potassium, and there are no
specific guidelines for the selection of any one product. The products include docusate
sodium (e.g., Colace) and docusate calcium (e.g., Kaopectate Stool Softener). Adult
dosing is 50 to 300 mg/day and dosing for children 2 years of age is 50 to 150 mg/day.
Each dose should be taken with at least 8 oz of water.
(2) Onset of action. Slow onset of action (24 to 72 hrs).
(3) Adverse effects. Diarrhea and mild abdominal cramping are possible.
(4) Warnings. Use should be avoided in individuals with nausea and vomiting, symptoms
of appendicitis, or undetermined abdominal pain.
e. Saline laxatives, which include sodium and magnesium salts, work to draw water into
the colon. This class of laxatives includes magnesium citrate, magnesium hydroxide
(e.g., Milk of Magnesia, Phillips caplets), and sodium phosphate (e.g., Fleet’s Enema).
(1) Onset of action is 30 mins to 3 hrs when given orally and 2 to 5 mins when given rectally.
(2) Side effects include abdominal cramping, excessive diuresis, nausea and vomiting,
and dehydration. As much as 20% of magnesium may be absorbed from these
products, which may lead to hypermagnesemia in patients with preexisting renal
impairment. Excessive doses of magnesium result in diarrhea.
(3) Warnings and counseling points
(a) The adult dose of magnesium citrate is consumption of one full bottle. Refrigeration of
the product may increase palatability and help prevent crystallization.
(b) Sodium-containing salts should be avoided in those individuals with sodium
restrictions (e.g., heart failure, edema, renal failure) and magnesium-containing
products should be avoided in individuals with severe kidney disease.
(c) Oral sodium phosphate (OSP) products (e.g., Fleet’s Phospho-Soda) have been sold
for years as nonprescription products for constipation and for acute bowel evacuation.
458 Chapter 23 I. B
However, the U.S. Food and Drug Administration (FDA) received numerous reports of
acute phosphate nephropathy (acute kidney injury) associated with these prod-ucts
when used for acute bowel evacuation. FDA has allowed nonprescription OSP products
to remain available for the treatment of constipation because acute kidney injury has
not been reported at these doses. However, FDA has determined that non-prescription
OSP products are not appropriate for bowel cleansing. In response, Fleet Laboratories
voluntarily removed their OSP products from the market.
f. Lubricant laxative. Mineral oil is the only available agent in this class. It works at the
colon to increase water retention in the stool.
(1) Availability. Mineral oil can be given once daily orally or rectally. The dose for
adults (age 12 years) is 15 to 45 mL orally or 118 mL rectally and for children (age 6 to
11 years) is 10 to 15 mL orally or 59 mL rectally.
(2) Onset of action. 6 to 8 hrs (oral dosing) and 5 to 15 mins (rectal dosing)
(3) Adverse effects include anal seepage, which can result in itching and perianal discomfort.
(4) Warnings and counseling points
(a) Mineral oil can decrease absorption of fat-soluble vitamins (i.e., vitamins A, D,
E, and K), so it should not be used on a chronic basis.
(b) Mineral oil should be taken on an empty stomach. Because of possible aspiration
of mineral oil into the lungs (lipid pneumonitis), this agent should not be taken
at bedtime. Those who are elderly, debilitated, or have dysphagia are at the greatest
risk of lipid pneumonitis.
(c) Emollients (e.g., docusate) may increase the systemic absorption of mineral oil,
which can lead to hepatotoxicity. Thus emollient and lubricant laxatives should
not be given concomitantly.
(d) Mineral oil is contraindicated in persons with rectal bleeding, appendicitis, or age 6
years.
C. Special populations
1. Pediatrics. Constipation should be expected if there is a drastic change from baseline bowel
func-tion. Nonpharmacologic methods such as increasing the amount of fluid or sugar provided
or increasing the bulk content of the diet (fruit, fiber cereals, vegetables) should be tried before
medications are used. If nonpharmacologic methods do not work, those 2 years of age should
be referred to a medical provider. Glycerin suppositories may be used in those 2 years of age.
2. Pregnant. Constipation in pregnancy is common and can be the result of compression of the
colon by the enlarged uterus, ingestion of prenatal vitamins containing iron and calcium, and
the influence of progesterone can cause bowel hypomotility. Bulk-forming agents or stool
softeners are appropriate to recommend during pregnancy.
3. Geriatrics. The elderly tend to be at risk for constipation due to insufficient dietary (fiber) and
fluid ingestion, concurrent disease states (e.g., hypothyroidism), and/or medications (e.g.,
opiates, anticholinergics). A major concern with the geriatric population is the possible loss of
fluid that can be induced by aggressive laxative treatment (e.g., enemas, high-dose saline
laxatives). Hyper-osmotic or stimulant laxatives may be useful for initial treatment and bulk-
forming laxatives for prevention, depending on concomitant disease states.
II. DIARRHEA
A. General information
1. Definition. Diarrhea is defined as an increase in the frequency and looseness of stools compared to
one’s normal bowel pattern. The overall weight and volume of the stool is increased ( 200 g or
mL/day), and the water content is increased to 60% to 90%. In general, diarrhea results when the
intestine is unable to absorb water from the stool, which causes excess water in the stool.
2. Classification. Diarrhea can be classified based on mechanism or origin.
a. Classification by mechanism
(1) Osmotic diarrhea occurs when excess water is pulled into the intestinal tract. Osmotic
diarrhea ceases when the patient converts to a fasting state. This may be the result of
hypermagnesemia, undigested lactose or fructose, or celiac disease.
Over-the-Counter Agents for Constipation, Diarrhea, Hemorrhoids, and Heartburn 459
(2) Secretory diarrhea occurs when the intestinal wall is damaged, resulting in an increased
secretion rather than absorption of electrolytes into the intestinal tract. This can occur with
the ingestion of bacterial enteropathogens (e.g., Escherichia coli, Salmonella, Shigella).
(3) Motility-related diarrhea occurs when food moves through the intestines at such a
rapid pace (hypermotility) that insufficient time is allowed for water and nutrient
absorption. Those with diabetic neuropathy, gastric/intestinal resection, or a vagotomy
are suscep-tible to this type of diarrhea. Medications that can also cause hypermotility
include para-sympathomimetic agents (e.g., metoclopramide, bethanechol), digitalis,
quinidine, and antibiotics.
b. Classification by origin
(1) Viral gastroenteritis is typically caused by the noroviruses, which are transmitted by
contaminated water or food. Other attributable viruses include rotaviruses,
adenoviruses, and hepatitis A virus. Diarrhea associated with viral gastroenteritis is
usually self-limiting for 2 to 3 days but may last up to 2 weeks.
(2) Bacterial gastroenteritis typically results from consumption of contaminated water or
food. Common contributors include E. coli, Staphylococcus aureus, Vibrio cholerae,
Shigella, Salmonella, Campylobacter, and Clostridium difficile. Toxin-producing
bacteria affect the small intestines, resulting in a watery stool. Invasive bacteria affect
the large intestines, resulting in dysentery-like stools (e.g., extreme urgency to defecate,
abdominal cramping, tenesmus, fever, chills, and small-volume stools that contain
blood or pus). Onset of diarrhea may range between 1 and 72 hrs, depending on the
infecting bacteria. Symptoms typically subside over 3 to 5 days.
(3) Protozoal diarrhea, caused by Giardia lamblia, Entamoeba histolytica, or Cryptosporid-
ium, may be described as profuse watery diarrhea, which may be accompanied by flatu-
lence and/or abdominal pain. This type of diarrhea is self-limiting, but may persist for
several weeks. Due to the extent of fluid loss over an extended duration of time, individu-als
with protozoal-induced diarrhea are at risk for dehydration. Self-care is inappropriate for
this type of diarrhea; infected persons should be referred to a medical provider.
(4) Diet-induced diarrhea. Diarrhea induced by foods results from food allergies, high-
fiber diets, fatty or spicy foods, large amounts of caffeine, or lactose intolerance. The
best treat-ment is prevention by avoiding troublesome foods.
B. Evaluation
1. Assessment of the individual should include age, onset and duration of diarrhea, description of stool
(i.e., frequency, consistency, volume, and presence of blood or pus), other symptoms (i.e.,
abdominal pain, fever, chills), aggravating or remitting factors, recent travel, and medical history.
2. Exclusions to self-care include:
a. Younger than 3 years of age or older than 60 years of age (with multiple medical
problems), pregnant or breastfeeding patients, and patients with HIV.
b. Blood or mucus in the stools
c. High fever ( 101°F or 38°C)
d. Dehydration or weight loss 5% of total body weight; signs of dehydration—dry mouth,
sunken eyes, crying without tears, dry skin that is not elastic like normal skin
e. Duration of diarrhea 2 days
f. Vomiting
C. Treatment
1. Nonpharmacological. Normal dietary intake should be recommended during bouts of diarrhea.
However, fatty foods, caffeinated beverages, foods rich in simple sugars, and spicy foods
should be avoided. The most important recommendation for treating acute diarrhea is to keep
the indi-vidual hydrated.
a. Fluid and electrolyte replacement. If mild to moderate fluid loss is present, oral rehydration so-
lution (ORS) that contains water, salt, and sugar can be recommended. This solution can be made at
home by adding salt, baking soda, and sugar to water (see Table 23-1) or purchased ready-to-use (e.g.,
Pedialyte). Table 23-2 provides recommended doses for patient age and severity of diarrhea. If fluid
loss is severe ( 10% loss of body weight) and/or severe vomiting persists, then intravenous rehydration
is necessary and a referral to a medical provider is most appropriate.
460 Chapter 23 II. C
Ingredients Dose
b. Fluids to be avoided in the dehydrated person include hypertonic fruit juices and carbonated or
caffeinated beverages. These fluids may make the diarrhea worse and do not contain needed
electrolytes (i.e., Na , K ). Gatorade does not contain an equivalent electrolyte concentration to
ORS, but may be sufficient in an individual with diarrhea who is not dehydrated.
2. Pharmacologic. Although acute nonspecific diarrhea, including travelers’ diarrhea, is
typi-cally self-limiting, antidiarrheals may be used for symptom alleviation. Currently there
are only two available nonprescription agents that the FDA has deemed to be safe and effective
antidiarrheal agents: loperamide and bismuth subsalicylate. Self-treatment of diarrhea with
these products is limited to 2 days; individuals experiencing longer bouts of diarrhea should be
referred for medical care.
a. Loperamide. Loperamide, an antiperistaltic agent, is approved as a nonprescription
treatment for acute, nonspecific diarrhea, including traveler’s diarrhea. Loperamide (e.g.,
Imodium A–D) provides effective control of diarrhea as quickly as 1 hr after administration.
(1) Availability and dosing. Loperamide is available for ages 6 years in several oral
formulations, including a syrup, capsule, and chewable tablet. The adult dose is 4 mg
followed by 2 mg after each loose stool, not to exceed 16 mg/day. The dose for a child
(ages 6 to 11 years) is also 2 mg after each loose stool, but the maximum dosage for
ages 6 to 8 years is 4 mg/day, whereas the maximum dosage for ages 9 to 11 years is 6
mg/day.
(2) Mechanism of action. Loperamide stimulates microopioid receptors on the circular
and longitudinal musculature of the small and large intestines to normalize peristaltic
intesti-nal movements. They slow intestinal motility and affect water and electrolyte
movement through the bowel. Thus, the frequency of bowel movements is decreased,
and the consis-tency of stools is increased.
(3) Adverse effects. At recommended doses, loperamide is generally well tolerated. Side
ef-fects are infrequent and consist primarily of abdominal pain, distention, or
discomfort; drowsiness, dizziness, and dry mouth.
Dose
5 years of age 2 L/first 4 hr, then replace ongoing 2–4 L/first 4 hr, then replace
losses ongoing losses
5 years of age 50 mL/kg/first 4 hr, then 10 mL/kg 100 mL/kg/4 hr, then 10 mL/kg
or 1/2–1 cup per stool or 1/2–1 cup per stool
Over-the-Counter Agents for Constipation, Diarrhea, Hemorrhoids, and Heartburn 461
III. HEMORRHOIDS
A. General information
1. Definition. Hemorrhoids are defined as abnormally large, bulging, symptomatic clusters of
dilated blood vessels, supporting tissues, and overlying mucous membranes. Hemorrhoids can
present in the lower rectum (internal hemorrhoids) or anus (external hemorrhoids). Simply,
hemorrhoids represent downward displacement of anal cushions that contain arteriovenous
anastomoses.
2. Epidemiology. Hemorrhoids are common, with approximately 10% to 25% of the U.S. popula-
tion affected. The risk of developing hemorrhoids increases with advancing age and peaks in
indi-viduals 45 to 65 years of age. The incidence of hemorrhoids in pregnant women is higher
than that of nonpregnant women of similar age. Although they are considered a minor medical
problem, they may cause considerable discomfort and anxiety.
B. Types of hemorrhoids are determined by their anatomical position and vascular origin.
1. An internal hemorrhoid is an exaggerated vascular cushion with an engorged internal hemor-
rhoidal plexus located above the dentate line and covered with a mucous membrane.
2. An external hemorrhoid is a dilated vein of the inferior hemorrhoidal plexus located below the
dentate line and covered with squamous epithelium.
3. A mixed hemorrhoid appears as a baggy swelling and exhibits simultaneous characteristics of
internal and external hemorrhoids.
C. Origin. Although heredity may predispose a person to hemorrhoids, the exact cause is probably
related to acquired factors.
1. Situations that result in increased venous pressure in the hemorrhoidal plexus (e.g., chronic
straining during defecation; small, hard stools; prolonged sitting on the toilet; occupations that
routinely require heavy lifting; pelvic tumors; pregnancy) can transform an asymptomatic
hemor-rhoid into a problem.
2. The hemorrhoidal veins are pushed downward during defecation or straining; with increased
venous pressure, they dilate and become engorged. Over time, the fibers that anchor the
hemor-rhoidal veins to their underlying muscular coats stretch, which results in prolapse.
D. Signs/symptoms
1. The most common sign/symptom of hemorrhoids is painless bleeding occurring during a
bowel movement. The blood is usually bright red and may be visible on the stool, on the toilet
tissue, or coloring the water in the toilet.
2. Prolapse is the second most common sign/symptom of hemorrhoids. A temporary protrusion
may occur during defecation, and it may need to be replaced manually. A permanently
prolapsed hemorrhoid may give rise to chronic, moist soiling of the underwear. A dull, aching
feeling may be heard as a complaint.
3. Pain is unusual unless thrombosis involving external tissue is present.
4. Discomfort, soreness, pruritus, swelling, burning, and seepage may also occur with hemor-
rhoids.
E. Proper diagnosis is essential because other more serious conditions may produce symptoms
that mimic those of hemorrhoids and include anal fissure (small tear in the lining of the anus, anal
fistula (abnormal connection between the mucosa of the rectum and the skin adjacent to the anus),
polyps (tumor of the large intestine), or colorectal cancer.
F. Treatment
1. Nonpharmacologic
a. Avoid straining when defecating and avoid sitting on the toilet longer than necessary.
b. Increase dietary fiber, water intake, and physical activity.
c. Sitz baths for 15 mins, three to four times a day, can soothe the anal mucosa. Tepid water
should be used, and prolonged bathing should be avoided. Epsom salts (magnesium sulfate)
added to the bath or the application of an ice pack can help reduce the swelling of an
edema-tous or clotted hemorrhoid.
2. Pharmacologic treatment. Nonprescription agents are available for the treatment of burning,
discomfort, inflammation, irritation, itching, pain, and swelling associated with hemorrhoids in
individuals ages 12 years (Table 23-3). These products are simply palliative; they are not meant
to cure hemorrhoids or other anorectal disease. Medical care should be sought if symptoms do
Over-the-Counter Agents for Constipation, Diarrhea, Hemorrhoids, and Heartburn 463
not improve after 7 days of treatment or if any of the following occur: bleeding, prolapse,
seepage of feces or mucus, or severe pain. Available formulations include ointments, creams,
and suppos-itories. Generally, the ointment or cream dosage form is believed to be superior to
a suppository, which may bypass the affected area. Some formulations are available with rectal
pipes, which are most efficient when the pipe has holes. The pipe allows insertion of the
medication directly in the rectum and the openings allow the ointment to cover large areas of
the rectal mucosa unreachable with the finger.
a. Local anesthetics work by blocking nerve impulse transmission.
(1) Availability. FDA-approved agents include benzocaine 20% (e.g., Americaine), pramox-
ine 1% (e.g., Pramoxine Rectal Foam), and dibucaine 0.25% to 1.0% (e.g., Nupercainal).
(2) Adverse effects. These agents may produce a hypersensitivity reaction with burning
and itching similar to that of anorectal disease. Systemic absorption is minimal unless
the perianal skin is abraded. As a result of its unique chemical structure, pramoxine
exhibits little cross-sensitivity compared to the other local anesthetics.
(3) Warnings. Allergic reactions may occur in some patients.
b. Vasoconstrictors are chemicals that resemble endogenous catecholamines. These agents
cause arteriole constriction in the anorectal area to reduce swelling, but the mechanism by
which they relieve itching, discomfort, and irritation is unknown.
(1) Availability. Phenylephrine is the only FDA-approved vasoconstrictor found in non-
prescription hemorrhoidal relief agents. It is one of the active ingredients in the line of
Preparation H products available in ointment, gel, suppository, and cream.
(2) Adverse effects. At recommended doses, the risk of individuals receiving enough sys-
temic absorption from hemorrhoidal vasoconstrictors to develop cardiovascular or CNS
effects is minimal; however, in those with cardiovascular disease, anxiety disor-ders, or
thyroid disease, an agent that does not contain a vasoconstrictor is a prudent
recommendation.
(3) Warnings/contraindications. Avoid in individuals with cardiovascular disease,
hyper-tension, hyperthyroidism, diabetes, and prostate enlargement because of the
possibil-ity of systemic absorption.
c. Protectants provide a physical barrier between the irritated skin and stool or anal seepage,
thereby relieving the itching, irritation, discomfort, and burning. Protectants are often the
bases or vehicles for other agents used for anorectal disease. Products include aluminum
hydroxide gel, cocoa butter, kaolin, lanolin, hard fat, mineral oil, white petrolatum,
petrolatum, glycerin (external use only), topical starch, cod liver oil, shark liver oil, and
zinc oxide. When protectants are incorporated into the formulation of a nonprescription
product, they should make up at least 50% of the dosage unit. If two to four protectants are
used, their total concentration should represent at least 50% of the whole product. Lanolin, a
derivative of wool alcohol, may be allergenic to susceptible individuals.
464 Chapter 23 III. F
d. Astringents lessen mucus and other secretions and protect underlying tissue through a local
and limited protein coagulant effect. Action is limited to surface cells, but astringents
provide temporary relief of itching, discomfort, irritation, and burning. Products considered
to be safe and effective include calamine 5% to 25%, witch hazel 10% to 50% (external use
only), and zinc oxide 5% to 25%.
e. Keratolytics cause desquamation and debridement of the surface cells of the epidermis,
which in turn allows hemorrhoidal medications to penetrate deeper into the tissues, and thus
pro-vide temporary relief of discomfort and itching. Products considered to be safe and
effec-tive include aluminum chlorhydroxyallantoinate (0.2% to 2.0%) and resorcinol
(1% to 3%). Keratolytics are reserved for external use only and resorcinol should not be
used on an open wound due to the potential for a serious hypersensitivity reaction. Of note,
there are currently no marketed products for hemorrhoid relief that contain these agents.
f. Analgesics, anesthetics, and antipruritics provide temporary relief of burning, discomfort,
itching, pain, and soreness. Ingredients considered to be safe and effective for external use
in the anorectal area include menthol (0.1% to 1.0%), juniper tar (1% to 5%), and
camphor (0.1% to 3%). These agents should not be used to treat internal hemorrhoids.
g. Corticosteroids. Hydrocortisone (0.25% to 1.0%) is the only FDA-approved corticosteroid for
anorectal disorders. Hydrocortisone causes vasoconstriction, stabilization of lysosomal mem-
branes, and antimitotic activity, which in turn may reduce the itching, inflammation, and discom-
fort in the anorectal area. Hydrocortisone concentrations 1% are available by prescription only.
(c) Magnesium hydroxide rarely is used alone for heartburn because it frequently
causes diarrhea. If used in renal failure patients, hypermagnesemia can occur
rapidly.
(d) Combination products with aluminum and magnesium (e.g., Maalox, Mylanta)
provide the highest ANC per volume and are used most frequently. The predomi-
nant adverse effect of this combination is diarrhea.
(e) Patients with renal failure should avoid the use of all antacids. Potassium and
magnesium content of antacids should be considered for patients with cardiac
disease.
(f) Antacids can interfere with the absorption of many drugs. In general, antacids
should be spaced at least 2 hrs apart from the administration of interacting drugs.
This is of-ten quite difficult to accomplish. Important clinical interactions with
antacids may occur with the following drugs: tetracycline and quinolone
antibiotics, iron supple-ments, and digoxin.
b. H2-Receptor antagonists (H2RAs). These medications inhibit gastric acid secretion by
com-petitively blocking H2-receptors on the parietal cell. By decreasing gastric acid
secretion, the refluxed material is less damaging to the esophagus.
(1) Availability and dosing. Available nonprescription agents are all dosed as needed for
symptoms, up to twice daily and include cimetidine (Tagamet-HB) 200 mg;
famotidine (Pepcid-AC) 10 to 20 mg (maximum dose is 40 mg/day); ranitidine
(Zantac) 75 to 150 mg (maximum is 150 mg/day); and nizatidine (Axid-AR) 75 mg.
(2) Onset of action and duration. The onset of symptom relief is 1 to 2 hrs, which is con-
siderably longer than antacids; however, the duration of action can last up to 10 hrs.
(3) Counseling points
(a) All H2RAs are contraindicated in individuals 12 years of age.
(b) Nonprescription H2RAs can be used for relief or prevention of heartburn.
(c) Cimetidine may impair the hepatic metabolism and thus increase serum concen-
trations and the pharmacological effects of warfarin, phenytoin, and theophylline.
(4) Adverse effects. These agents are extremely well tolerated. The most common adverse
effects reported with nonprescription doses are headache, diarrhea, dizziness, and
nausea.
c. Proton pump inhibitors. These agents provide complete acid suppression by inhibiting the
hydrogen-potassium ATPase pump on the surface of the parietal cell. Proton pump
inhibitors (PPIs) are an appropriate recommendation for individuals who have failed other
“steps” in the treatment plan, or who are experiencing frequent heartburn (defined as
heartburn occurring 2 or more days a week).
(1) Availability. Nonprescription PPIs include omeprazole magnesium 20.6 mg (Prilosec
OTC) and lansoprazole 15 mg (Prevacid 24 hrs).
(2) Onset of action and duration. Onset of action is 2 to 3 hrs, but the duration of relief
is approximately 12 to 24 hrs. Thus, PPIs should not be used for immediate relief.
(3) Adverse effects include headache, constipation, abdominal pain, nausea, and diarrhea.
(4) Counseling points
(a) PPIs are activated by stomach acid and therefore should be dosed 30 mins prior to
a meal. Optimal symptom relief may not be achieved if taken after a meal.
(b) Drug interactions are attributable to effects on gastric pH and metabolism through
the CYP3A4 and CYP2C19 pathways and include clopidogrel, vitamin K
antagonists, and atazanavir.
(c) PPIs for self-care are approved for ages 18 years.
(d) As self-care, PPIs should only be recommended for the 14-day course of therapy
and repeated no more often than every 4 months, as necessary.
d. Bismuth subsalicylate (BSS) is FDA approved for the relief of upset stomach due to
overindul-gence of food and drink. Its mechanism of action for heartburn and dyspepsia is
unknown. A more thorough review of BSS can be found in section II.C.2.b.
e. Combination H2RAs and antacid (e.g., Pepcid Complete) may provide quick onset of relief
(within 30 mins) and an extended duration of relief (8 to 10 hrs).
Over-the-Counter Agents for Constipation, Diarrhea, Hemorrhoids, and Heartburn 467
Study
Questions 5. Which of the following is the most common
sign/ symptom of hemorrhoids?
Directions: Each of the questions, statements, or (A) bleeding
incomplete statements in this section can be correctly (B) pain
answered or completed by one of the suggested answers (C) seepage
or phrases. Choose the best answer. (D) pruritus
1. Which laxative should not be used to treat acute 6. Which of the following agents is designated as a safe
constipation because of its slow onset of action? and effective analgesic, anesthetic, and antipruritic
(A) glycerin by the FDA?
(B) bisacodyl suppository (A) witch hazel
(C) psyllium (B) juniper tar
(D) milk of magnesia (C) hydrocortisone
(D) phenylephrine
2. All of the following statements about emollient
stool softener laxatives are true except which one? 7. All of the following symptoms associated
(A) T hey are not first-line treatment for the with gastroesophageal reflux may be treated
typical individual with acute constipation. with nonprescription agents except
(B) T hey are appropriate for individuals who (A) burning sensation located in the lower chest.
should not strain by passing a hard stool. (B) pain that is worse after meals.
(C) T hey are known as surfactants and include (C) pain or difficulty when swallowing.
docusate calcium and docusate sodium. (D) pain that is worse in a recumbent position.
(D) T hey are highly associated with the 8. Which of the following is an appropriate
development of acute phosphate nephropathy. nonpharmacological recommendation for patients
3. Which of the following statements adequately with gastroesophageal reflux?
describes bulk-forming laxatives? (A) Eat larger but fewer meals.
(A) T hey can cause diarrhea if not taken with at (B) Avoid meals high in protein.
least 8 oz of water. (C) Eat evening meals at least 3 hrs before bed.
(B) T hey are derived from polysaccharides and (D) Prop the head up with two pillows at night.
resemble fiber in mechanism of action. 9. Which of the following would be the most appropriate
(C) T hey have a relatively fast onset of action to recommend for self-care to an individual wanting fast
of 4 to 8 hrs and duration of 12 to 24 hrs. but extended relief from heartburn?
(D) T hey produce a more complete evacuation of
(A) Proton pump inhibitor
the bowels than stimulant products.
(B) H2RA
4. Which local anesthetic should be used to treat (C) Aluminum ( ) Magnesium antacid
symptoms of pain, itching, burning, and discomfort (D) H2RA ( ) antacid
in patients with an established lidocaine allergy?
10. All of the following are acceptable uses of
(A) tetracaine
magnesium hydroxide except
(B) dibucaine
(A) heartburn.
(C) pramoxine
(D) benzocaine (B) dyspepsia.
(C) hemorrhoids.
(D) constipation.
468 Chapter 23
a great extent although it can be helpful for treating the headache and backache that
may accompany menstrual cramping. NSAID treatment provides relief for mild to
moderate symptoms but will probably not help patients with severe symptoms.
(a) Mechanism of action. NSAIDs inhibit the synthesis and action of prostaglandins,
which are responsible for the pain associated with dysmenorrhea.
(b) Administration guidelines. Begin therapy at the onset of pain; there is no proven value in
beginning therapy in anticipation of dysmenorrhea. Acceptable recommendations include
ibuprofen (e.g., Motrin, Midol Liquid Gels) 200 mg every 4 to 6 hrs (maximum 1200
mg/day) or naproxen sodium (e.g., Aleve, Pamprin All Day, Midol Extended Relief) 200 mg
of naproxen every 8 to 12 hrs (maximum naproxen dose is 600 mg/day).
(c) Adverse effects are limited because of the brief duration of need. However,
common adverse effects associated with NSAIDs include bleeding, upset stomach,
abdominal pain, diarrhea, heart failure exacerbation, cardiovascular thrombic
events, and diz-ziness. NSAIDs should be taken with food to decrease adverse
gastrointestinal (GI) effects.
(d) Warnings. The lowest effective dose for the shortest duration needed should be
rec-ommended to lessen the potential risk for cardiovascular events. These agents
are contraindicated in patients with an allergy to NSAIDs or active GI disease.
2. Premenstrual syndrome (PMS)
a. Symptoms (e.g., marked mood swings, fatigue, appetite changes, bloating, breast
tenderness, irritability, feelings of depression) begin 1 to 7 days before the onset of menses.
b. Nonpharmacological therapy includes regular exercise, dietary modifications, and reduction of
stress factors. Dietary modifications include avoiding alcohol and caffeine, which can in-crease
irritability, and consuming a balanced diet of fruits, vegetables, and complex carbohy-drates
while avoiding salty foods and simple sugars (can exacerbate fluid retention). Patients
experiencing symptoms abnormal to their cycle should be referred to a physician.
c. Pharmacological treatment
(1) Combination products are marketed for women (age 12 and older) with PMS
symptoms. These products contain analgesics (e.g., acetaminophen, aspirin, ibuprofen,
naproxen so-dium), diuretics (e.g., caffeine, pamabrom), and/or an antihistamine (e.g.,
pyrilamine, diphenhydramine).
(a) Diuretics are recommended by the U.S. Food and Drug Administration (FDA) for use
in eliminating water during premenstrual and menstrual periods. When admin-istered
approximately 5 days before menses, diuretics help relieve bloating, excess water,
cramps, and tension. Included in this category are caffeine and pamabrom.
(i) Caffeine (e.g., Pamprin Max, Midol Complete), a xanthine derivative, pro-
motes diuresis by inhibiting tubular reabsorption of sodium and chloride. The
recommended dosage is 100 to 200 mg every 3 to 4 hrs. Side effects
associated with caffeine use are GI disturbances and central nervous system
(CNS) stim-ulation. Patients should be counseled to limit the consumption
of caffeine-containing foods or beverages while taking this product.
(ii) Pamabrom (e.g., Aqua-ban, Midol Teen, Pamprin Cramp, Pamprin Multi-
Symptom) is a theophylline derivative often used in combination with analge-
sics and antihistamines. The recommended dosage is 50 mg four times daily
not to exceed 200 mg/day.
(b) Antihistamines, such as pyrilamine and diphenhydramine, are added to PMS prod-
ucts for their sedative effects although the need for sedation is questionable. At this
time, there is insufficient evidence to recommend agents containing antihistamines
(e.g., Pamprin Multi-Symptom, Midol Complete, Midol PM) for the alleviation of
PMS symptoms.
(2) Daily calcium supplementation (equivalent to the recommended daily calcium intake
for women of reproductive age) has been shown to reduce the emotional and physical
symptoms of PMS.
(3) Pyridoxine. Although clinical trials do not support the efficacy of pyridoxine (vitamin
B6), it is being used for the treatment of PMS. Vitamin B6 doses should be limited to
100 mg/day to prevent dose-associated neuropathy.
Over-the-Counter Menstrual, Vaginal, and Contraceptive Agents 471
C. Toxic shock syndrome (TSS) is a rare but sometimes fatal illness often associated with menstruation.
1. TSS can be categorized either as menstrual or nonmenstrual, with approximately two-thirds of
cases associated with menstruation. TSS is known to occur in both men and women.
2. The condition usually affects women 30 years of age who use tampons. Women between the
ages of 15 and 19 years are at the highest risk.
3. TSS is characterized by an abrupt onset (8 to 12 hrs) of flu-like symptoms (e.g., high fever, my-
algia, vomiting, diarrhea). Neurologic symptoms such as headache, agitation, lethargy,
seizures, and confusion can also occur.
4. TSS results from an exotoxin produced by Staphylococcus aureus.
5. The primary risk factor for TSS is the use of tampons, but the use of barrier contraceptives
(e.g., diaphragms, cervical sponges) also increases the risk.
6. When TSS is suspected, patients should be hospitalized immediately. To lower the risk of TSS, women
should use lower-absorbency tampons and alternate the use of tampons with feminine hygiene pads;
however, to lower the risk to nearly zero, women should use sanitary pads instead of tampons.
4. Pharmacologic treatment. Products proven safe and effective for the treatment of VVC are
imid-azole derivatives with antifungal activity and include tioconazole, miconazole, and
clotrimazole. Each agent is available in a variety of formulations, including intravaginal
creams, suppositories, ovules, and ointments.
a. The choice of product formulation is based on patient preference. One formulation is not
more effective than another.
b. External vaginal creams, mainly used to treat vulvar symptoms of pruritus, can be used
in combination with intravaginal products. Many products are available as a “combination
pack” including an internal cream, ovule, or suppository and an external cream.
c. For most available intravaginal products, application time should be recommended as
bedtime to increase vaginal mucosa contact time. However, external creams can be used
any time of day.
d. Products are available as 1-, 3-, or 7-day treatment plans. Available products include Moni-
stat (miconazole 1-, 3-, and 7-day), Vagistat (tioconazole 1-day; miconazole 3-day), and Gyne-
Lotrimin (clotrimazole 3- and 7-day). Efficacy rates for these products approach 80% to 90%.
(1) Each product must be used for the consecutive number of days outlined for the product
to be effective.
(2) The formulated products for 1- and 3-day treatments are for user convenience and
should not be misinterpreted for the time to symptom resolution.
(3) Symptoms of VVC typically do not resolve completely for 5 to 7 days. Thus when 1- and
3-day products are purchased, the woman should be aware that she may still experience
symptoms after she has completed the treatment but should resolve within several days.
If symptoms are still present after 7 days, a medical provider should be consulted.
(4) Products for internal use may come with prefilled applicators for single use or reusable
applicators. Reusable applicators should be washed with soap and water each time.
e. Patient counseling
(1) Complete the course of therapy even if symptoms improve. Symptom improvement
typi-cally will not be seen for 5 to 7 days after initiation of treatment. Seek medical
treatment if symptoms have not resolved after 7 days.
(2) Wash vaginal area with mild soap before application.
(3) Avoid sexual intercourse during therapy.
(4) Avoid condoms and diaphragm use for 72 hrs after therapy is completed.
(5) Continue use during menstrual period, but tampons should not be used during VVC
treatment.
(6) Sanitary pads can be used for leakage of intravaginal products.
(7) Side effects can include burning or irritation.
(8) Treatment of male partners is not necessary.
f. Lactobacillus acidophilus, taken by mouth or as a vaginal suppository, has inadequate data
at this time to make a sound recommendation for prevention or treatment of VVC.
5. Prevention
a. Dry vaginal area well with a towel after bathing.
b. Avoid tight or damp clothing.
c. Wear cotton underwear.
d. Use unscented soap to avoid irritation.
e. Avoid douching.
f. Decrease consumption of sucrose and simple sugars.
B. Feminine hygiene products. T here are a variety of feminine hygiene products available for
cleansing and controlling odor associated with normal vaginal discharge and products available for
vaginal dryness. These products are not used to treat vaginal infections.
1. Vaginal douches (Summer’s Eve) irrigate the vagina and can be used for cleansing, for
soothing as an astringent, or to produce a mucolytic effect.
2. Vaginal lubricants are used for immediate relief of vaginal dryness, which can cause pain
dur-ing intercourse. Vaginal lubricants are available as oil-, water-, or silicone-based.
a. Oil-based lubricants typically contain baby oil or petroleum jelly and should not be
recom-mended for use with latex condoms because they can deteriorate the latex.
Additionally, oil-based lubricants can harbor bacteria in the vagina and lead to infections.
Over-the-Counter Menstrual, Vaginal, and Contraceptive Agents 473
b. Water-based vaginal lubricants (e.g., Astroglide, K-Y Jelly) are known to be compatible
with latex condoms.
c. Silicone-based vaginal lubricants (e.g., K-Y INTRIGUE) are longer lasting than water-
based lubricants and are compatible only with latex condoms.
3. Vaginal moisturizers. Unlike lubricants, which provide immediate relief, vaginal moisturizers
(e.g., Replens, K-Y Silk-E) are for chronic vaginal dryness. Thus if used for intercourse,
vaginal moisturizers need to be applied 2 hrs prior to allow for adequate lubrication.
Oral contraceptives
Mechanical/chemical
d
Cervical cap
Multiparous 40 26.0
Nulliparous 20 9.0
Male condom without spermicide 12–14 3.0
Male condom with spermicide 4–6 1.8
d
Diaphragm 20 6.0
Female condom 21 5.0
Intrauterine device 1–2 1.0–1.5
Levonorgestrel implants 1.0 1.0
Medroxyprogesterone injection 1.0 1.0
Spermicide alone 20–22 6.0
Other
estimated to average 2 to 3 days (up to 5 days), and the fertile period of the ovum, which is
es-timated to be 24 hr. Recent studies indicate that conception is most likely to occur when
couples have intercourse during a 6-day period ending on the day of ovulation. Conception is
highly un-likely if sexual intercourse occurs 6 or more days before ovulation or the day
after ovulation. These methods are based on reproductive anatomy and physiology and are
applied according to the signs and symptoms naturally occurring in the menstrual cycle.
a. Calendar method. This method estimates the possible day of ovulation, based on
documen-tation of prior menstrual cycle events. Abstinence should be practiced during the
period around ovulation when there may be a fertilizable egg present. The calendar method
is not as well-used as it once was and is not accurate for women with irregular cycles,
women who are breastfeeding, or women with postponed ovulation.
b. Temperature method. Documentation of basal body temperature (BBT) can be made
using a basal thermometer, which is available without a prescription.
(1) Approximately 24 hrs prior to ovulation, there is a moderate drop in basal temperature
followed by a rise in temperature 24 hrs after ovulation. The rise in temperature is thought
to be the result of progesterone release, which indicates the occurrence of ovulation.
(2) For this method to be successful, abstinence should be practiced 5 days after the
onset of menses until 3 days after the drop in basal temperature.
(3) Because the basal temperature reflects the amount of heat radiation when the body is at
its metabolic low, the temperature should be taken first thing in the morning (i.e.,
before any activity) after at least 5 hrs of restful sleep.
(4) The BBT thermometer may be used orally, vaginally, or rectally, depending on the
model selected.
(5) False changes in BBT may be caused by infection, tension, a restless night, or any type
of excessive movement.
2. Spermicidal agents are composed of an active spermicidal chemical, which immobilizes or
kills sperm, and an inert base (e.g., foam, cream, jelly, gel, tablet, or suppository), which
localizes the spermicidal chemical in proximity to the cervical os. The only FDA-approved
spermicidal agent is nonoxynol-9. However, although it is spermicidal, it is not a microbicide
and therefore cannot be used alone for protection against sexually transmitted infections.
a. Dosage forms. Contraceptive spermicides, which are available in several forms, offer the
greatest variety within one specific method of contraception (Table 24-2).
(1) Creams, jellies, and gels are used with a diaphragm. The concentration of spermicide
is less than the necessary 8% to be employed as a single contraceptive method.
Film Inserted by the female directly over the cervix; insert not less than
15 mins and not more than 3 hrs before intercourse; contraceptive
protection begins 5–15 mins after insertion and remains effective no
more than 3 hrs
Foam Contraceptive protection is immediate; remains effective no more
than 1 hr, additional dose is needed before any subsequent
intercourse
Jellies, creams, gels Contraceptive protection is immediate; used alone remains
effective no more than 1 hr; when used with diaphragm or cap,
keep diaphragm or cap in place for at least 6 hrs after last
intercourse
Suppositories and tablets Contraceptive protection begins 10–15 mins after insertion; remains
effective no more than 1 hr
(2) Foams disperse better into the vagina and over the cervical opening but provide less
lubrication than creams, jellies, and gels. They usually contain a higher concentration
of spermicide (i.e., the optimal concentration of 8% or higher). Because of volume
differ-ences among brands, the dosage amounts vary. If vaginal or penile irritation
develops, another brand should be tried.
(a) The can should be shaken vigorously 20 times before use.
(b) The foam should be inserted intravaginally about two-thirds the length of the appli-
cator, and the contents should be discharged.
(c) Foam should be reapplied during prolonged intercourse (i.e., lasting 1 hr) and
before every subsequent act of intercourse.
(d) To ensure efficacy, the patient should wait at least 8 hrs before douching to avoid
diluting the spermicide effect or “forcing” sperm into the cervix.
(3) Suppositories and foaming tablets. T hese agents are both small and convenient. Al-
though solid at room temperature, suppositories melt at body temperature, whereas
foaming tablets effervesce.
(a) The tablets should be wetted and inserted high into the vagina 10 to 15 mins before
intercourse. Intercourse must occur within 1 hr or the dose must be repeated. Each
repeated act of intercourse requires insertion of another tablet/suppository.
(b) To ensure efficacy, the patient should wait 6 to 8 hrs after the last act of
intercourse before douching.
(4) Film comes as small paper-thin sheets (e.g., vaginal contraceptive film [VCF]). It is
inserted on the tip of the finger into the vagina, and placed at the cervical opening 5 to
15 mins before intercourse.
(5) Sponge. The Today Sponge is a doughnut-shaped polyurethane device containing the
spermicide nonoxynol-9. However, it may also provide contraceptive effects by serving
as a mechanical barrier to the cervical entrance.
(a) The sponge should remain in place for at least 6 hrs and up to 30 hrs. Contraceptive
benefits are for 24 hrs, regardless of the frequency of intercourse during this period.
(b) Increased risk of TSS with use (rare).
(c) Not recommended for use during menstruation.
(d) This method may have a higher pregnancy rate for women who have vaginally
deliv-ered a baby.
b. Side effects. Side effects are minimal, but may include sensation of warmth and rare
allergic reactions (contact dermatitis with rash, stinging, itching, and swelling). If a
suspected reac-tion occurs, one should be instructed to use another product because the issue
might be the concentration of the spermicide or an additive specific to a given brand. There
are no signifi-cant differences in birth defect rates between users and nonusers.
3. Male condoms are used to prevent transmission of sperm into the vagina.
a. Types. There are four different types of materials for male condoms, which include latex
rub-ber, processed collagenous lamb cecum sheaths (lambskin), polyurethane, or
polyisoprene. Condoms are labeled that they are intended to prevent HIV and other sexually
transmitted infections, but caution that they do not completely eliminate the risk,
particularly human papillomavirus (HPV) and herpes simplex virus (HSV).
(1) Latex condoms afford greater elasticity than lambskin and are more likely to remain in
place on the penis.
(a) Availability. Various types are available (e.g., lubricated, ribbed, colored),
including some with spermicide (concentration much less than that of a vaginal
spermicide product). It is doubtful that spermicide-lubricated condoms offer any
better protec-tion than plain latex condoms, and they have a shorter shelf life.
There is a standard size, but smaller and larger versions are available.
(b) Concerns. Latex rubber may cause an allergic reaction. An estimated 1% to 2% of the
population is sensitized to natural rubber latex, and higher percentages are likely for those
frequently exposed to latex (e.g., health care workers). The most common symptoms are
genital inflammation with redness, itching, and burning. Sometimes, antioxidants or ac-
celerators used during the manufacturing process may be the cause of the allergy.
476 Chapter 24 III. B
(2) Lambskin condoms (e.g., Trojan NaturaLamb) are not considered as effective as
latex condoms (and cannot be labeled as such) in preventing the transmission of STDs,
includ-ing HIV. The lambskin condoms are structured to consist of membranes that
reveal layers of fibers crisscrossing in various patterns. This gives the lambskin
strength but also allows for an occasional pore. Therefore, lambskin may allow HIV
and hepatitis B virus, which are smaller than sperm, to pass through.
(a) Lambskin has less elasticity than latex, and lambskin condoms may slip off the penis.
(b) Lambskin affords greater sensitivity than latex.
(c) Lambskin condoms are more expensive than latex condoms.
(3) A polyurethane condom (e.g., Avanti, Trojan Supra) is marketed for individuals
who are allergic to latex. Some evidence exists that slippage and breakage rates may
be higher than for latex condoms. In contrast to the latex condom, petroleum-based
products will not degrade the polyurethane.
(4) A polyisoprene condom (e.g., LifeStyles Skyn) is the newest material to be marketed
for individuals who are allergic to latex. The condom is promoted as superior to
poly-urethane in malleability and comfort and does protect against most sexually
transmitted infections.
b. Advantages. The relative accessibility, ease of transport, and low cost make condoms an at-
tractive method of contraception.
c. Disadvantages. Condoms are not 100% effective against preventing pregnancy or the
spread of sexually transmitted infections. Although accessible to purchase, male condoms
are, for most, not easily transportable. Male condoms should not be stored in wal-lets,
glove compartments, etc. because of the potential for deterioration of the condom.
However, a new product, Trojan2GO, provides condoms packaged as “condom cards”—
packaging that is resistant to breakdown and which can be easily carried in the wallet or
pocket.
d. Use. Condoms are now packaged with detailed instructions for proper application
tech-nique and disposal. A new condom should be used correctly each time.
e. Counseling points
(1) Proper lubrication can minimize the possibility of tearing and can be ensured by using
either a lubricated condom or by applying a water-based lubricant to either the condom
or the woman’s genitalia. (Note: Petroleum jelly [Vaseline] should never be used
because it causes deterioration of the rubber [latex] and is a poor lubricant.)
(2) Condoms should never be reused.
(3) Condoms should not be stored near excessive heat. Be sure to store condoms in a cool,
dry place, out of direct sunlight. The glove compartment of a car is not a good place to
store condoms. Do not store condoms in pockets, purses, or wallets for more than a few
hours, unless packaging has been specifically designed for such purpose.
(4) If the condom should break or leak, spermicide foam should be immediately inserted
vaginally.
(5) Do not buy or use condoms that have passed their expiration date.
(6) Condoms are not 100% effective against preventing pregnancy or the spread of
sexually transmitted infections. If a sexually transmitted infection is suspected, contact
a health care provider or public health agency.
4. The female condom is a disposable nitrile sheath that fits into the vagina, and provides
protec-tion from pregnancy and some sexually transmitted infections. The original female
condom (FC1) was made of polyurethane, an expensive material. In 2009, FDA approved a
second-generation product, the nitrile female condom (FC2), which is made of synthetic latex
and thus less expensive to manufacture than the FC1. Production of FC1 has ceased.
a. The sheath resembles a plastic vaginal pouch and consists of an inner ring, which is
inserted into the vagina near the cervix much like a diaphragm, whereas the outer ring
remains outside the vagina, covering the labia. The condom is prelubricated, and additional
lubricant (oil- or water-based) may be used if needed. It may be inserted up to 8 hrs before
intercourse, and can be removed at any time after ejaculation. However, the FC should be
removed prior to the woman standing up to prevent semen spillage.
Over-the-Counter Menstrual, Vaginal, and Contraceptive Agents 477
b. Female condoms should not be used concurrently with a male condom because the
friction between the two condoms may contribute to condom breakage.
c. The FC has a higher pregnancy failure rate than male condoms, but does appear to
protect against some sexually transmitted diseases, including HIV and cytomegalovirus.
However, these data are limited, and further research is necessary to determine the extent of
protection these condoms afford against sexually transmitted infections.
d. Female condoms should not be reused.
e. A main complaint with the FC1 was a “squeaking” noise with use. The FC2 has less noise
associated with it, but it still can make noise during intercourse.
5. The diaphragm is a contraceptive device that is self-inserted into the vagina to block access of
sperm to the cervix. It requires a prescription and must be used in conjunction with a nonpre-
scription spermicide to seal off crevices between the vaginal wall and the device.
6. The cervical cap is a prescription rubber device smaller than a diaphragm that fits over the
cervix like a thimble. It is more difficult to fit than the diaphragm.
7. Emergency contraception (EC) is the use of a medication or device after coitus to prevent
pregnancy. Currently, the only available nonprescription agent for emergency contraception is
levonorgestrel, but it is only available for self-care in females ages 17 and older.
a. Mechanism of action. The exact mechanism for EC is unknown but thought to be at least
two or more of the following:
(1) inhibit or delay ovulation
(2) prevent implantation
(3) inhibit fertilization
b. Availability and dose
(1) Next Choice: contains two tablets of levonorgestrel 0.75 mg to be taken 12 hrs apart
(2) Plan B One-Step: contains one tablet of levonorgestrel 1.5 mg to be taken as a single dose
c. Timing. Levonorgestrel for EC is most efficacious when taken within 24 hrs of
intercourse but can be taken up to 72 hrs after intercourse. Some data demonstrate
efficacy up to 120 hrs (5 days) after intercourse, but if used during this time frame, women
should be aware that efficacy may be diminished.
d. Safety. When used as emergency contraception, there are no contraindications, given the
short duration of use. Adverse effects may include nausea, vomiting, cramping, or irregular
bleeding. If vomiting occurs within 2 hrs of dosing, the dose may need to be repeated.
e. Counseling points
(1) Levonorgestrel EC should not be used for ongoing contraception.
(2) EC is not effective against the prevention of sexually transmitted infections.
(3) Levonorgestrel EC is most efficacious if taken within 24 hrs of unprotected intercourse.
(4) Levonorgestrel EC is ineffective once implantation has occurred.
(D) anaerobic bacteria.
Study
Questions
Directions for questions 1–8: Each of the questions,
statements, or incomplete statements in this section can
be correctly answered or completed by one of the
suggested answers or phrases. Choose the best answer.
1. T he most common cause of vaginal yeast infections is
(A) Candida albicans.
(B) Candida glabrata.
(C) Trichomonas.
2. A female complains of vaginal burning and itching
with a distinct “fishy” odor. Which of the following
would be the most appropriate recommendation?
(A) Purchase the Vagisil pH monitor.
(B) Referral to a medical provider.
(C) Treatment with a 1-day VVC product.
(D) Treatment with a 7-day VVC product.
478 Chapter 24 7. Which of the following agents can be used alone for
protection against sexually transmitted infections?
(A) Contraceptive foam
3. Which of the following would be appropriate to
(B) Contraceptive sponge
counsel on for a woman who is purchasing a
(C) Female condom
female condom? (D) Diaphragm
(A) T he FC2 is superior to the male condom
8. A 17-year-old female reports breakage of a condom
for pregnancy prevention.
last night during intercourse. She has not been using
(B) T he FC2 can protect against all
sexually transmitted infections. oral contraception because she smokes. Which of
(C) T he FC2 should be used concomitantly with the following would be appropriate to counsel for
a male condom. this patient on Plan-B One-Step?
(D) T he FC2 can be inserted several hours prior (A) She needs a prescription to purchase Plan-
to intercourse. B One-Step.
(B) Take one tablet now and the second tablet 12
4. When assisting a female with the purchase of a
hrs later.
vaginal pH monitor, which of the following (C) Take Plan B One-Step within 72 hrs of intercourse.
should be discussed? (D) She is not a candidate for EC because of
(A) T he pH monitor is not recommended her tobacco use.
in postmenopausal women. (E) She can use this continuously for back-
(B) An elevated pH indicates the presence of a up contraception.
yeast infection.
Directions for questions 9–10: Each statement in this
(C) T he pH of a clean-catch morning urine
sample should be used. section is most closely related to one of the following
(D) T he pH monitor can detect the presence drug types. The drug types may be used more than once
of sexually transmitted infections. or not at all. Choose the best answer, A–D.
I. INTRODUCTION. Many of the drugs available on the market are derived from plants. Some of
those include aspirin, atropine, belladonna, capsaicin, cascara, colchicine, digoxin, ephedrine, ergotamine,
ipecac, opium, physostigmine, pilocarpine, podophyllum, psyllium, quinidine, reserpine, scopolamine,
senna, paclitaxel, tubocurarine, vinblastine, and vincristine. Herb products are also derived from plants;
however, these products are not considered drugs by the U.S. Food and Drug Administration (FDA).
A. Regulations
1. The Federal Food, Drug, and Cosmetic Act of 1938 mandated pharmaceutical companies to
test drugs for safety before marketing.
2. The Kefauver–Harris Drug Amendments of 1962 mandated pharmaceutical companies to
test drugs for efficacy before marketing.
3. The Dietary Supplement Health and Education Act of 1994 mandated the following:
a. Dietary supplements are not drugs or food. They are intended to supplement the diet.
b. Herbs are considered dietary supplements.
c. Dietary supplements do not have to be standardized.
d. The secretary of Health and Human Services may remove a supplement from the market
only when it has been shown to be hazardous to health.
e. Dietary supplements may make claims only regarding the effects on structure or function of
the body. No claims regarding a particular disease or condition may be made.
f. The following statement is required on the product label: “This product has not been
evaluated by the FDA. It is not intended to diagnose, treat, cure, or prevent.”
4. Final rule for Current Good Manufacturing Practices (CGMPs) for dietary supplements (2007)
a. Manufacturers are required to evaluate for identity, purity, strength, composition
5. German Federal Health Agency
a. In 1978, the German Federal Health Agency established Commission E.
b. Commission E evaluates the safety and efficacy of herbs through clinical trials and cases
pub-lished in scientific literature.
c. There are . 380 published monographs on herbs.
B. Herbs considered unsafe for human consumption
1. Carcinogenic herbs include borage, calamus, coltsfoot, comfrey, liferoot, and sassafras.
2. Hepatotoxic herbs include chaparral, germander, kava, and liferoot.
3. High doses of licorice for long periods may cause pseudoaldosteronism, a condition that may
include headache, lethargy, sodium and water retention, hypokalemia, high blood pressure,
heart failure, and cardiac arrest.
4. Ma huang may cause myocardial infarction, strokes, or seizures.
5. Pokeroot may be fatal in children.
6. Unsafe herbs according to the FDA. In the 1990s, the FDA’s Center for Food Safety and Applied
Nutrition created the Special Nutritional Adverse Event Monitoring System Web site for dietary
supplements. Unfortunately, by 1999 the site was no longer being updated and thus was eventually
480
Herbal Medicines and Nutritional Supplements 481
deleted. According to the last update from that Web site, the following dietary supplements
were considered unsafe by the FDA:
a. Arnica: muscle paralysis, death
b. American and European mistletoe: seizures, coma
c. Bittersweet and deadly nightshade: cardiac toxicity
d. Bloodroot: hypotension, coma
e. Broom: dehydration
f. Comfrey: cancer
g. Dutch and English tonka bean: hepatotoxicity
h. Heliotrope: hepatotoxicity
i. Horse chestnut: bleeding
j. Jimson weed: anticholinergic, hallucinations
k. Kava: hepatotoxicity
l. Lily of the valley: cardiac toxicity
m. Lobelia (nicotine): coma, death
n. Mandrake/mayapple: severe gastrointestinal symptoms
o. Morning glory: psychosis
p. Periwinkle: renal and hepatotoxicity
q. Snakeroot: reserpine derivative
r. Spindle tree: seizures
s. St. John’s wort: drug interactions
t. Sweet flag: hallucinations, liver cancer
u. True jalap: purgative cathartic
v. Wahoo: seizures
w. Wormwood: seizures, paralysis
x. Yohimbe: renal failure, hypertension
5. Drug interactions
a. Cisplatin (Platinol) efficacy may be reduced.
b. Theoretically, black cohosh may interact with hepatotoxic drugs such as acetaminophen
(Tylenol), carbamazepine (Tegretol), and isoniazid (Nydrazid) because it is an inhibitor of
cytochrome P450 3A4 (CYP3A4) and CYP2D6 isoenzymes.
c. A case reports a patient taking atorvastatin (Lipitor) developing significantly elevated liver
function enzymes after black cohosh was initiated.3
6. Side effects
a. Occasional intestinal problems may occur, such as nausea and vomiting; weight gain is possible.
b. Liver toxicity may occur; liver function tests should be monitored periodically.
c. Large doses of black cohosh may cause dizziness, nausea, severe headaches, stiffness, and
trembling limbs.
d. Does not seem to increase risk of endometrial hyperplasia.
7. Dosage. Remifemin is a standardized product that contains 20 mg black cohosh and is taken
twice daily. It is standardized to 1 mg of 27-deoxyactein per tablet.
1. Commission E indications
a. Internal use: supportive therapy for infections of the upper respiratory tract (cold) and lower
urinary tract
b. External use: local application for the treatment of hard-to-heal superficial wounds and ulcers
2. Mechanism of action. Echinacea increases the body’s resistance to bacteria by the following:
a. Caffeic acid derivatives, which include cichoric acid, chlorogenic acid, and cynarin,
increase phagocytosis and stimulate the production of immunopotentiating substances such
as inter-feron, interleukins, and tumor necrosis factor.
b. Polysaccharides, such as inulin, stimulate macrophages and inhibit hyaluronidase activity to
decrease inflammation.
c. Alkylamides, such as echinacein, have a local anesthetic effect and inhibit hyaluronidase
activity to decrease inflammation.
d. Echinacea has little or no direct bactericidal or bacteriostatic properties.
3. Efficacy
a. Treatment of common cold. In a review of 26 controlled clinical trials evaluating
echinacea’s ability to strengthen the body’s own defense mechanisms, it was found that 30
of 34 echinacea therapies were more effective compared to controls. 8
b. Prevention of common cold. Studies have shown that taking echinacea prophylactically to
prevent the development of a cold does not seem to be effective.
4. Contraindications/precautions
a. Echinacea is contraindicated in infectious and autoimmune diseases such as tuberculosis,
leukosis, collagenosis, multiple sclerosis, AIDS, HIV, and lupus.
b. Caution should be used in patients who are allergic to members of the ragweed or chrysan-
themum family.
c. The effects of echinacea in pregnancy, lactation, and children are unknown. Comparison
with a control group suggested no increased risk of major malformations in 206 pregnant
women.9
d. Therapy should not exceed 8 weeks. Theoretically, prolonged use of echinacea may depress
the immune system, possibly through overstimulation.
5. Drug interactions
a. Unknown if echinacea interacts with immunosuppressants
b. Echinacea inhibits cytochrome P450 1A2. Some drugs metabolized by CYP1A2 are
caffeine (Cafcit) and theophylline.
c. Echinacea induces and inhibits CYP3A4. Some drugs metabolized by CYP3A4 are
midazolam (Versed), itraconazole (Sporanox), and fexofenadine (Allegra).
6. Side effects
a. Nausea, vomiting, dizziness, tiredness
b. Allergic reactions, acute asthma, leukopenia, and anaphylaxis
c. May interfere with male fertility
7. Dosage. T here are a variety of doses recommended. The most common dose is as the dried
powder, 1 g or two 500-mg capsules orally three times daily. Recommended to use for 2 weeks
only during a cold.
3. Efficacy. An evaluation of five trials for the efficacy of feverfew in the prevention of mi-
graines compared to placebo was conducted.10 A variety of doses and durations were used.
Some trials showed the number and severity of migraine attacks and the degree of vomiting
was reduced with feverfew. The duration of attacks was unaltered. Other trials showed no
benefit.
4. Contraindications/precautions
a. Feverfew should be avoided in pregnancy, lactation, and children , 2 years of age.
b. Contraindicated in individuals with allergies to chrysanthemums or ragweed.
c. Contraindicated in patients with bleeding disorders. Discontinue 2 weeks before surgery.
5. Drug interactions
a. Feverfew may interact with anticoagulants, increasing the risk of bleeding.
b. Feverfew may inhibit the following cytochrome P450 isoenzymes: 1A2, 2C19, 2C9, and
3A4.
6. Side effects
a. Gastric discomfort on oral consumption
b. Contact dermatitis
c. Minor ulcerations of oral mucosa, irritation of tongue, and swelling of lips may occur when
fresh leaves are chewed.
d. Palpitations
e. Post-feverfew syndrome: Discontinuation of feverfew may produce muscle and joint
stiffness and a cluster of nervous system reactions (rebound of migraines, anxiety, and
insomnia).
7. Dosage. The usual dose of feverfew is 50 to 100 mg daily. A product containing at least 0.2%
par-thenolide is recommended.
2. Mechanism of action
a. The seed coat gum of flaxseed is soluble fiber.
b. Flaxseed contains fatty acids; of which, 55% are alpha-linolenic acid. The human body
converts alpha-linolenic acid into omega-3 fatty acids, such as eicosapentaenoic (EPA) and
docosahexaenoic acids (DHA).
c. Lignans are phytoestrogens with weak estrogenic and anti-estrogenic effects.
3. Efficacy
a. Diabetes. Flaxseed decreased A1c but did not decrease fasting blood glucose or insulin
levels (Pan).
b. Hypercholesterolemia. Flaxseed 40 to 50 g/day decreases LDL by 8% to 18% and
decreases total cholesterol by 5% to 9%, but flaxseed does not affect HDL.
c. Menopause. Flaxseed 40 g/day has been shown to significantly reduce hot flashes by about
35% and night sweats by about 44%.
4. Contraindications/precautions. Bleeding disorders, diabetes, gastrointestinal obstruction, and
hormone-sensitive cancers
5. Drug interactions
a. Anticoagulants/antiplatelets. Theoretically may have additive effects.
b. Antidiabetic agents. Theoretically may have additive effects.
c. Estrogens. Theoretically may have competitive effects.
d. Flaxseed may decrease the absorption of oral drugs. It is recommended to administer
medica-tions an hour before or 2 hrs after taking flaxseed.
6. Side effects
a. Soluble fiber causes bloating, flatulence, abdominal pain, diarrhea, constipation, dyspepsia,
nausea.
b. Allergic reactions
7. Dosage
a. For hypercholesterolemia, baked goods (muffins or bread) containing flaxseed and ground
flaxseed with 40 to 50 g of flaxseed per day
b. For menopausal symptoms, 40 g daily of crushed flaxseed
c. Adequate amount of fluid to prevent potential bowel obstruction
d. Antihypertensives (hypotension)
e. Antidiabetic agents (hypoglycemia)
f. May induce the following cytochrome P450 isoenzymes: 2C9, 2C19, 3A4, 2D6, and 2E1.
Caution should be used with contraceptive medications, cyclosporine, diltiazem, and vera-
pamil (decreased efficacy).
g. Isoniazid. Decreased efficacy.
6. Side effects. Gastrointestinal discomfort (heartburn, flatulence), sweating, light-headedness,
allergic reactions, and menorrhagia
7. Dosage. Between 0.6 and 1.2 g dried powder (2 to 5 mg of allicin) daily or 2 to 4 g fresh garlic
8. Comments
a. Alliinase (the enzyme that converts alliin to allicin) is inactivated by acids. Enteric-coated
tablets or capsules allow more absorption because they pass through the stomach and
release their contents in the alkaline medium of the small intestine.
b. Effective preparations include freeze-dried garlic power and aged garlic extract.
c. Odorless garlic preparations may not contain the active compounds.
2. Mechanism of action
a. Ginkgo contains flavonoids (quercetin, kaempferol, and isorhamnetin) and terpenoids
(ginkgolides A, B, and C and bilobalide).
b. Flavonoids provide the antioxidant activity, reduce capillary fragility, and increase the
thresh-old of blood loss from capillaries.
c. Ginkgolides antagonize platelet-activating factor (PAF), which induces platelet aggregation,
the degranulation of neutrophils, and the production of oxygen radicals.
3. Efficacy
a. In a review of the clinical and pharmacological studies on ginkgo and cerebral insuf-
ficiency, eight were found to be of good quality. 14 Seven of the trials showed positive ef-
fects of ginkgo compared to placebo. Symptoms of cerebral insufficiency evaluated were
difficulties of concentration and memory, absentmindedness, confusion, lack of energy,
tiredness, decreased physical performance, depression, anxiety, dizziness, tinnitus, and
headaches.
b. A randomized, double-blind, placebo-controlled study included 202 patients with either
Alzheimer disease or multi-infarct dementia. These patients were given ginkgo 40 mg three
times daily or placebo for 1 year. Ginkgo had a statistically significant improvement by at
least two points or better on the Alzheimer Disease Assessment Scale—Cognitive (a 70-
point subscale) compared to placebo (50% vs. 29%). Ginkgo showed statistically significant
im-provement on the Geriatric Evaluation by Relative’s Rating Instrument (37% vs. 23%).
There was no difference between ginkgo and placebo on the Clinical Global Impression of
Change Scale.15
4. Contraindications/precautions
a. Epilepsy. Ginkgotoxin may cause neurotoxicity and seizures.
b. Bleeding disorders. Discontinue 2 weeks before surgery
c. Diabetes (hypoglycemia)
d. Infertility. Caution in difficulty conceiving.
5. Drug interactions
a. Ginkgo may potentiate the bleeding properties of antiplatelets/anticoagulants.
b. Aminoglycosides (increased ototoxicity)
c. Thiazide (increases blood pressure)
d. Trazodone (Desyrel) (coma)
e. Seizure threshold lowering drugs
f. Anticonvulsants (decreased efficacy)
g. Antidiabetic drugs (hypoglycemia)
h. Ginkgo may mildly affect the cytochrome P450 isoenzymes 1A2, 2C19, 2C9, 2D6, and
3A4.
6. Side effects
a. Gastric disturbances, headache, dizziness, and vertigo
b. Toxic ingestion may produce tonic–clonic seizures and loss of consciousness
c. Spontaneous bleeding; mild to severe (intracerebral hemorrhage)
7. Dosage. Recommended dose is 40 mg three times daily with meals for at least 4 to 6 weeks.
Standardized preparations that contain 6% terpene lactones and 24% ginkgo flavone glycosides
are recommended.
1. Commission E indications. Tonic to combat feelings of lassitude and debility, lack of energy,
and ability to concentrate, and during convalescence
2. Mechanism of action
a. At least 28 active ingredients, known as ginsenosides, have been identified.
b. Ginseng effects vary with extract derivative, drying method, dose, duration of treatment,
and animal species that were studied. Each ginsenoside produces different pharmacological
effects on the CNS, cardiovascular system, and other body systems. Different ginsenosides
are capable of producing biological effects in direct opposition with those produced by
others. For example, the ginsenoside Rb1 has been shown to have a suppressive effect on
the CNS, whereas Rg1 produces a stimulatory effect.
Herbal Medicines and Nutritional Supplements 489
3. Efficacy
a. A randomized, double-blind, placebo-controlled, crossover study included 50 male sports
teachers who performed a treadmill exercise test. Volunteers received two ginseng capsules
(Geriatric Pharmaton) daily for 6 weeks or placebo. Volunteers used energy more
efficiently and had greater endurance while taking ginseng.16 However, other studies have
not shown Asian ginseng to be effective.
4. Contraindications/precautions
a. Pregnancy and lactation
b. Children
c. Avoid in patients with hypertension, emotional/psychological imbalances, headaches, heart
palpitations, insomnia, asthma, inflammation, or infections with high fever.
d. Caution should be used in patients with a history of bleeding. Discontinue 2 weeks before surgery.
e. Diabetes (hypoglycemia)
f. Schizophrenia
g. Caution should be used in patients with a history of breast cancer. Ginseng may stimulate
breast cancer cells.
5. Drug interactions
a. Ginseng may interact with phenelzine (Nardil), producing hallucinations and psychosis.
b. Ginseng may decrease the INR of warfarin (Coumadin).
c. Ginseng may interact with stimulants, including caffeine (Cafcit).
d. Ginseng may interact with oral hypoglycemic and insulin, causing hypoglycemia.
e. It is unknown whether ginseng interacts with hormonal therapy, antihypertensives, or
cardiac medications.
f. It may inhibit cytochrome P450 2D6. Caution should be used with drugs that are metabolized
via cytochrome P450 2D6, such as amitriptyline (Elavil) and fluoxetine (Prozac).
g. It may interfere with immunosuppressants such as cyclosporine (Sandimmune) or
tacrolimus (Prograf).
6. Side effects
a. Nervousness, excitation, insomnia
b. Inability to concentrate with long-term use
c. Diffuse mammary nodularity and vaginal bleeding may be caused by ginseng’s estrogen-
like effect in women.
d. Hypertension, euphoria, restlessness, nervousness, insomnia, skin eruptions, edema, and diarrhea
have been reported with long-term ginseng use with an average dose of 3 g ginseng root daily.
e. Tachyarrhythmia due to increased QT interval
7. Dosage. 1 to 2 g crude herb daily or 100 to 300 mg ginseng extract three times daily.
Standardized products that contain at least 4% to 5% ginsenosides are recommended.
6. Side effects include diarrhea and other gastrointestinal reactions (nausea, dyspepsia, flatulence)
and allergic reactions.
7. Dosage. Recommended dose of milk thistle is 200 to 400 mg/day divided into three doses using
a standardized product that includes 70% to 80% silymarin.
5. Drug interactions
a. Antidepressants such as paroxetine (Paxil), sertraline (Zoloft), and nefazodone have been
reported to cause serotonin syndrome when taken with St. John’s wort.
b. Antiretroviral (protease inhibitors and nonnucleoside reverse transcriptase inhibitors) levels
may decrease.
c. St. John’s wort may decrease the efficacy of barbiturates.
d. St. John’s wort may increase the efficacy of clopidogrel (Plavix).
e. Cyclosporine (Sandimmune) levels may decrease.
f. St. John’s wort may interact with other drugs metabolized through the cytochrome P450
isoenzymes 1A2, 2C9, 2C19, and 3A4.
g. Digoxin (Lanoxin) levels may decrease.
h. Irinotecan (Camptosar) and imatinib (Gleevec) levels may decrease.
i. Methadone (Dolophine) levels may decrease.
j. St. John’s wort may decrease the efficacy of omeprazole (Prilosec).
k. Oral contraceptives may have a decreased effect.
l. St. John’s wort may decrease the efficacy of HMG coenzyme reductase inhibitors (simvastatin).
m. Tacrolimus (Prograf) levels may decrease.
n. Theophylline levels may decrease.
o. Triptans. Theoretically, St. John’s wort may interact with the triptans.
p. Verapamil (Calan, Covera-HS, Isoptin, Verelan) levels may decrease.
q. St. John’s wort may decrease the INR of warfarin (Coumadin).
r. Serotonergic agents such as dextromethorphan, fenfluramine, narcotics.
s. Anticonvulsants. Phenytoin, phenobarbital, mephenytoin.
6. Food interactions
a. Older studies suggested that St. John’s wort was an MAOI.20
b. Newer studies suggest St. John’s wort is a weak MAOI.21
c. One case report published of MAOI-type food interactions such as tyramine-containing
foods: cheeses, beer, wine, herring, and yeast.
7. Side effects
a. Photodermatitis, allergic reactions
b. Gastrointestinal irritations
c. Tiredness, restlessness, sleep disturbances
d. Elevated thyroid-stimulating hormone
e. Elevated blood pressure
f. Mania or hypomania
g. May cause infertility
8. Dosage. Recommended 2 to 4 g daily in two to three divided doses. Standardized products con-
taining 0.4 to 2.7 mg hypericin/day or 0.3% hypericin are recommended.
4. Contraindications/precautions
a. Caution while driving or performing other tasks requiring alertness and coordination is
recommended.
b. Pregnancy and lactation
c. Surgery. Valerian may have an additive effect to anesthesia.
5. Drug interactions
a. CNS depressants. Valerian may potentiate the sedative effect of barbiturates,
benzodiazepines, opiates, alcohol, or other sedatives.
b. Valerian inhibits cytochrome P450 3A4.
6. Side effects
a. Headaches, hangover, excitability, insomnia, uneasiness, and cardiac disturbances
b. Hepatotoxicity
c. Toxicity includes ataxia, decreased sensibility, hypothermia, hallucinations, and increased
muscle relaxation
d. Patients may experience a benzodiazepine-like withdrawal, so doses should be tapered down slowly.
7. Dosage. Dried root: 2 to 3 g/cup, one to three times daily. Standardized to contain 0.8% to 1.0%
valeremic acids/dose extract: 400 to 900 mg 30 to 60 mins before bedtime.
1. Nonapproved indications. Heart failure (HF), hypertension, stable angina, ventricular arrhyth-
mias, cancer, heart surgery, and periodontal disease
2. Mechanism of action
a. It is a naturally occurring coenzyme that has a predominant role in oxidative phosphorylation and
synthesis of adenosine triphosphate (ATP), which is needed for muscle contraction and relaxation.
Herbal Medicines and Nutritional Supplements 493
b. Statin-induced myalgias. Studies suggest that there is a 20% to 50% reduction in serum
lev-els of coenzyme Q10 in hypercholesterolemic patients after a statin has been initiated.
The reduction in coenzyme Q10 concentration is believed to be dose- related.25 Several
studies have evaluated the effect of coenzyme Q10 supplementation in patients taking
statins. When coenzyme Q10 is given 30 to 300 mg/day, coenzyme Q10 serum
concentrations significantly elevate.25 However, a systematic review of studies showed that
exogenous coenzyme Q10 did not improve statin-induced myalgias.26
4. Contraindications/precautions
a. Hyper and hypotension
b. Pregnancy and lactation
5. Drug interactions
a. HMG reductase inhibitors may lower plasma concentrations of coenzyme Q10.
b. Doxorubicin toxicity may be decreased.
c. Antihypertensives (additive effect)
d. Warfarin. Coenzyme Q10 is structurally related to vitamin K 2 so may have coagulant
effects.
6. Side effects
a. Rash and gastrointestinal disturbances, such as nausea, anorexia, epigastric pain, and
diarrhea
b. Elevations of serum aminotransferases have occurred with relatively high oral doses
7. Dosage. Depends on indication: 100 mg daily, up to 3000 mg daily in two to three divided
doses.
5. Drug interactions
a. Glucosamine may interact with antidiabetic agents.
b. Glucosamine may induce resistance to antimitotic chemotherapy (etoposide [VePesid],
doxorubicin).
c. Theoretically, glucosamine may interact with warfarin.
6. Side effects
a. Gastrointestinal side effects such as epigastric pain and tenderness, heartburn, diarrhea, and
nausea
b. CNS side effects such as drowsiness, headache, and insomnia
c. Long-term side effects are unknown.
d. Elevated blood glucose
7. Dosage. Recommended: 500 mg three times daily
1. Orphan drug status. Treatment of circadian rhythm sleep disorders in blind people who have
no light perception
2. Nonapproved indications. Jet lag, insomnia, depression, and cancer
3. Mechanism of action
a. It is a hormone made from serotonin and secreted by the pineal gland. Melatonin controls
the periods of sleepiness and wakefulness. It increases GABA receptor binding.
4. Efficacy
a. Jet lag. A randomized, placebo-controlled trial evaluated the effect of melatonin in 52
aircraft personnel. Melatonin was given either 5 mg daily 3 days before departure until 5
days after arrival (early group) or 5 mg daily upon arrival and for 3 additional days (late
group). The late group had significantly less jet lag, fewer overall sleep disturbances, and a
faster recovery of energy compared to the placebo group and the early group. 27
b. Insomnia. A meta-analysis on 17 randomized, double-blind, placebo-controlled trials that
evaluated the sleep effect of melatonin in subjects showed that melatonin significantly
decreased time to sleep onset, increased sleep efficiency, and increased total sleep duration
compared to placebo.28 Unfortunately, melatonin preparations were varied and study
designs differed.
c. Children. Melatonin may be effective in decreasing the time to sleep onset in children with
neurodevelopmental disabilities. Melatonin does not improve total sleep time or nighttime
awakenings.29
5. Contraindications/precautions
a. Avoid in pregnancy and lactation
b. Melatonin may aggravate depressive symptoms.
c. Melatonin may increase the incidence of seizures.
d. Diabetes (hyperglycemia)
e. Hypertension (exacerbated)
f. Caution while driving or performing other tasks requiring alertness and coordination
6. Drug interactions
a. Selective serotonin reuptake inhibitors may increase melatonin serum concentrations.
b. Other sedatives such as alcohol and benzodiazepines may exacerbate the sedative effects of
melatonin.
c. Melatonin may interfere with immunosuppressants.
d. Antidiabetic agents may be less effective.
e. Anticoagulants and antiplatelets (increased effect)
f. Caffeine (theoretically, efficacy of melatonin may be decreased)
g. Contraceptives (theoretically, efficacy of melatonin may be increased)
h. Verapamil (increased melatonin excretion)
7. Side effects
a. Side effects include drowsiness, daytime fatigue, headache, and transient depression.
b. Long-term side effects are unknown.
8. Dosage. 0.3 to 5.0 mg at bedtime
Herbal Medicines and Nutritional Supplements 495
IV. REFERENCES
1. Mahady GB. Black cohosh (Actaea/Cimicifuga racemosa): review of the clinical data for safety
and efficacy in menopausal symptoms. Treat Endocrinol. 2005;4(3):177–184.
2. Jacobson JS, Troxel AB, Evans J, et al. Randomized trial of black cohosh for the treatment of hot
flashes among women with a history of breast cancer. J Clin Oncol. 2001;19(10):2739–2745.
3. Patel NM, Derkits RM. Possible increase in liver enzymes secondary to atorvastatin and black
cohosh administration. J Pharm Prac. 2007;20(4):341–346.
4. Schellenberg R. Treatment for the premenstrual syndrome with agnus castus fruit extract:
prospec-tive, randomised, placebo controlled study. BMJ. 2001;322(7279):134–137.
5. Avorn J, Monane M, Gurwitz JH, et al. Reduction of bacteriuria and pyuria after ingestion of
cran-berry juice. JAMA. 1994;271(10):751–754.
6. Murray M, Pizzorno J. Encyclopedia of Natural Medicine. 2nd ed. Roseville, CA: Prima Publishing;
1998.
7. Hirata JD, Swiersz LM, Zell B, et al. Does dong quai have estrogenic effects in postmenopausal
women? A double-blind, placebo-controlled trial. Fertil Steril. 1997;68(6):981–986.
8. Melchart D, Linde K, Worku F, et al. Immunomodulation with echinacea: a systematic review of
controlled clinical trials. Phytomedicine. 1994;1:245–254.
9. Gallo M, Sarkar M, Au W, et al. Pregnancy outcome following gestational exposure to echinacea:
a prospective controlled study. Arch Intern Med. 2000;160(20):3141–3143.
10. Pittler MH, Ernst E. Feverfew for preventing migraine. Cochrane Database Syst Rev. 2004;(1):
CD002286.
11. Silagy CA, Neil HA. A meta-analysis of the effect of garlic on blood pressure. J Hypertension.
1994;12(4):463–468.
12. Warshafsky S, Kamer RS, Sivak SL. Effect of garlic on total serum cholesterol. A meta-analysis.
Ann Intern Med. 1993;119(7 pt 1):599–605.
13. Mowrey DB, Clayson DE. Motion sickness, ginger, and psychophysics. Lancet. 1982;1(8273):
655–657.
14. Kleijnen J, Knipschild P. Ginkgo biloba. Lancet. 1992;340:1136–1139.
15. Le Bars PL, Katz MM, Berman N, et al. A placebo-controlled, double-blind, randomized trial of
an extract of Ginkgo biloba for dementia. JAMA. 1997;278(16):1327–1332.
16. Pieralisi G, Ripari P, Vecchiet L. Effects of a standardized ginseng extract combined with dimeth-
ylaminoethanol bitartrate, vitamins, minerals, and trace elements on physical performance during
exercise. Clin Ther. 1991;13(3):373–382.
17. Rambaldi A, Jacobs BP, Iaquinto G, et al. Milk thistle for alcoholic and/or hepatitis B or C virus
liver diseases. Cochrane Database Syst Rev. 2005;(2):CD003620.
18. Tacklind J, MacDonald R, Rutks I, et al. Serenoa repens for benign prostatic hyperplasia.
Cochrane Database Syst Rev. 2009;(2):CD001423. doi:10.1002/14651858.CD001423.pub2
19. Linde K, Mulrow CD, Berner M, et al. St. John’s wort for depression. Cochrane Database Syst Rev.
2005;(2):CD000448.
20. Suzuki O, Katsumata Y, Oya M, et al. Inhibition of monoamine oxidase by hypericin. Planta Med.
1984;50(3):272–274.
21. Müller WE, Rolli M, Schäfer C, et al. Effects of hypericum extract (LI 160) in biochemical
models of antidepressant activity. Pharmacopsychiatry. 1997;30(suppl 2):102–107.
22. Leathwood PD, Chauffard F. Aqueous extract of valerian reduces latency to fall asleep in man.
Planta Med. 1985;(2):144–148.
23. Clegg DO, Reda DJ, Harris CL, et al. Glucosamine, chondroitin sulfate, and the two in
combination for painful knee osteoarthritis. N Engl J Med. 2006;354(8):795–808.
24. Morisco C, Trimarco B, Condorelli M. Effect of coenzyme Q10 therapy in patients with congestive
heart failure: a long-term multicenter randomized study. Clin Investig. 1993;71(8 suppl):S134–S136.
25. Nawarskas JJ. HMG-CoA reductase inhibitors and coenzyme Q10. Cardiol Rev. 2005;13(2):
76–79.
26. Marcoff L, Thompson PD. The role of coenzyme Q10 in statin-associated myopathy: a systematic
review. J Am Coll Cardiol. 2007;49(23):2231–2237.
496 Chapter 25 IV
27. Petrie K, Dawson AG, Thompson L, et al. A double-blind trial of melatonin as a treatment for jet
lag in international cabin crew. Biol Psychiatry. 1993;33(7):526–530.
28. Brzezinski A, Vangel MG, Wurtman RJ, et al. Effects of exogenous melatonin on sleep: a meta-
analysis. Sleep Med Rev. 2005;9(1):41–50.
29. Phillips L, Appleton RE. Systematic review of melatonin treatment in children with neurodevelop-
mental disabilities and sleep impairment. Dev Med Child Neurol. 2004;46(11):771–775.
Study Questions
Directions for questions: Each of the questions, 4. Mary has a family history of heart disease and
statements, or incomplete statements can be correctly wonders if garlic would be beneficial to her. Which
answered or completed by one of the suggested answers of the following statements is correct about garlic?
or phrases. Choose the best answer. (A) Enteric-coated tablets release their contents in
1. Which of the following herbs is known to the stomach.
cause cancer? (B) Side effects include heartburn, flatulence,
(A) Chaparral and sweating.
(C) T he safety of garlic in pregnancy is unknown.
(B) Comfrey
(D) Garlic does not interact with warfarin.
(C) Ma huang
(D) Licorice 5. An 80-year-old man takes warfarin for his mechanical
(E) St. John’s wort heart valve. He would also like to take the following
herbs: Asian ginseng, feverfew, garlic, and dong quai.
2. Which of the following is a correct statement?
Which of these herbs may decrease the effectiveness
(A) Dietary supplements must be proven safe and
of warfarin?
effective before marketing in the United States.
(A) Asian ginseng
(B) T he following statement is optional for
(B) Feverfew
labeling of herbal products: “This product has
(C) Fish oil
not been evaluated by the FDA. It is not
(D) Garlic
intended to diagnose, treat, cure, or prevent.”
(E) Dong quai
(C) Herbs must be standardized to be considered
dietary supplements. 6. A 30-year-old female is 10 weeks pregnant with her
(D) Dietary supplement manufacturers may claim second child. During her first pregnancy, she became
that their products affect the structure and depressed and was started on Prozac 20 mg every day.
function of the human body. She is already beginning to notice early symptoms of
(E) Congress determines what is considered a depression during her second pregnancy. She would
supplement and what is considered a drug. like to try St. John’s wort for her depression. Which of
the following statements is correct regarding
3. Tom would like to try echinacea to prevent cold and
St. John’s wort?
flu during the winter months. Which of the
following statements is true about echinacea? (A) T he safety of St. John’s wort in
pregnancy is unknown.
(A) It is contraindicated in patients allergic to parsley.
(B) St. John’s wort is not helpful in treating
(B) It should be taken continuously only
mild depression.
for 3 months.
(C) St. John’s wort may interact with
(C) It is contraindicated in patients with lupus and
serotonin reuptake inhibitors.
leukosis.
(D) St. John’s wort may interact with dairy
(D) Prolonged use of echinacea will up regulate
products like milk and eggs.
the immune system.
(E) St. John’s wort decreases the effects of
(E) Side effects include headache, rash, and dizziness.
clopidogrel (Plavix).
7. A 65-year-old is interested in taking ginkgo. Which of 497
Herbal Medicines and Nutritional Supplements
the following statements is correct regarding ginkgo?
(A) T here are no contraindications with ginkgo.
(B) T here is a drug–herb interaction between 8. A 20-year-old athletic man would like to take Asian
ginkgo and aspirin. ginseng to increase his physical stamina. His
(C) Toxic effects include hypertension and girlfriend suggested that he ask a pharmacist about
cardiac arrest. the safety of Asian ginseng. Which of the following
(D) T here is a drug–herb interaction between statements is not correct?
ginkgo and phenelzine. (A) Asian ginseng may interact with
(E) Ginkgo is contraindicated in patients phenelzine, warfarin, and digoxin.
with gallstone pain. (B) Asian ginseng should be used with caution
in patients with a history of breast cancer.
(C) Asian ginseng may interact with
stimulants, including caffeine (Cafcit).
(D) Asian ginseng should be avoided in patients
with hypertension.
(E) Asian ginseng may cause bradycardia due
to increasing the QT interval.
I. INTRODUCTION
A. Objectives
1. Therapeutic drug monitoring (TDM) in a general sense is about using serum drug concentra-
tions (SDCs), pharmacokinetics, and pharmacodynamics to individualize and optimize patient
responses to drug therapy.
2. TDM aims to promote optimum drug treatment by maintaining SDC within a therapeutic range—
above, when drug-induced toxicity occurs too often; and below, when the drug is too often ineffective.
B. Definitions
1. Specifically, TDM is a practice applied to a small group of drugs in which there is a direct
relation between SDCs and pharmacological response as well as a narrow range of
concentrations that are effective and safe and for which SDCs are used in conjunction with
other measures of clinical observation to assess patient status.
2. Clinical pharmacokinetics, a term often used interchangeably with TDM, is more generally the ap-
plication of pharmacokinetic principles for the rational design of an individualized dosage regimen.
3. For definitions of the terms used and the concepts applicable in basic and clinical
pharmacokinet-ics, see Chapter 5 on pharmacokinetics.
C. Rationale and reasons
1. The rationale for TDM makes three assumptions.
a. Measuring patient SDC provides an opportunity to adjust for variations in patient
pharmaco-kinetics by individualizing drug dosage.
b. The SDC is a better predictor of patient response than dose.
c. There is a good relation between SDCs and pharmacological response.
2. Reasons for measuring SDC
a. Drug levels are used in conjunction with other clinical data to assist practitioners in
determin-ing how a patient is responding.
b. Drug levels provide a basis for individualizing patient dosage regimens.
c. Drug levels assist in determining if a change in patient-specific pharmacokinetics has
occurred during a course of treatment, whether as a result of a change in physiological state,
a change in diet, or addition of other drugs.
d. Assuring drug compliance is often cited as a reason for measuring SDC, but it is unreliable for
this purpose. In truth, a noncompliant patient may outwit practitioners by manipulating preap-
pointment behavior to induce an SDC that is nonreflective of the patient’s drug-taking behavior.
499
500 Chapter 26 II. A
Therapeutic
range
D f
3 Cl 5 CSS
t
Figure 26-1. Pharmacokinetic factors infl uencing serum
drug concentration and pharmacological response.
f, bioavailability; Cl, clearance; Css, steady-state
Practitioner Patient Pharmacological serum drug concentration; D, dose; , frequency of
variables variables response administration of dose.
Clinical Pharmacokinetics and Therapeutic Drug Monitoring 501
a
Amikacin Cmax,ss 20–30 g/mL; Cmin,ss 10 g/mL
Cyclosporine Varies with transplanted organ, time after transplant, time of sampling during
dosage interval, and method of analysis
b
Digoxin 0.8–2.0 ng/mL
a
Gentamicin Cmax,ss 5–10 g/mL; Cmin,ss 2 g/mL
Phenytoin 10–20 g/mL
a
Tobramycin Cmax,ss 5–50 g/mL; Cmin,ss 2 g/mL
a
Vancomycin Cmax,ss 30–50 g/mL; Cmin,ss 5–10 g/mL
aEnd of 30- to 60-min infusion.
b
Levels for atrial fi brillation often exceed 2 ng/mL.
502 Chapter 26 III. B
Carbamazepine
Indinavin
Lithium
Methotrexate
Mycophenolate
Serolimus
Tacrolimus
where S is the fraction of the dosage form that is the active moiety and f is the bioavailability.
3. Minimum concentration (Cmin,ss) during ss, when absorption is assumed to be much faster
than elimination.
(S)( f )(dose/tinf)
Cinf,ss (5)
Cl
B. Linear pharmacokinetic drug clearance—impaired renal function
1. Estimating creatinine clearance from serum creatinine when serum creatinine is assumed to be stable,
not changing daily, and weight is expressed by the patient’s total weight, unless total weight is equal to or
more than 20% of ideal (lean) body weight, in which case ideal weight should be used in the calculation.
(140 age)(weight)
__
Clcr (mL/min, males) (6)
(Crs)(72)
where Crs denotes serum creatinine in mg/dL and weight expressed in kg. The female value is
85% of the estimated male value in (6).
Clinical Pharmacokinetics and Therapeutic Drug Monitoring 503
3. In the renal impaired, what should be done to modify the preceding regimen for a patient who
is 60 years old, 70 kg, male, with a Crs of 2.5 mg/dL? In this patient, if the preceding regimen
were used, the prolonged t½ would yield Cmax,ss 15.2 mcg/mL and Cmin,ss 8.4 mcg/mL, values
clearly above the target concentration ranges.
a. Using equation (6) for estimating Clcr in this patient.
Cl (140 60 yrs)(70 kg) 31 mL/min
__
cr
(2.5 mg/dL)(72)
b. Then using equation (7) for estimating t½ in this patient, assuming F 0.98 and (Clcr)n 120
mL/min.
(2.5 hrs)
_ 1 0.98 0.98 (31/120) 0.27(t½)ri 9.3 hrs
(t½)ri
c. Then, using equation (9), first determine a loading dose (DL) to achieve the same Cmax,ss of
approximately 8 mcg/mL as estimated in IV.D.2 for a patient with normal renal function:
DL (Cmax,ss)(V)/(S)(f)
DL (8 mcg/mL)(0.25 L/kg)/(1)(1) 2 mg/kg
Next, determine fraction of drug lost during , assuming a ri of 24 hrs, and using the (t½)ri of
9.3 hrs estimated in IV.D.3.b.
F. From a clinical viewpoint, serum protein binding of drugs becomes an important issue in TDM
for drugs bound more than 80% to serum proteins. Because hepatic and renal clearances, volume
of distribution, and pharmacological response are mediated by the free (unbound) form of the drug
in serum, interpatient variations in protein binding not only result in variations in pharmacokinetics
in normals, but loss of serum proteins during renal and hepatic impairments may also result in
modi-fied drug clearance and pharmacological response.
5. Illustrating the use of PPV, NPV, PLR, and NLR. For digoxin, using a test upper cutoff of 2
ng/mL, PPV is 0.59, NPV is 0.95, PLR is 7, and NLR is 0.2.
a. For PPV, this means that the proportion of patients with a positive test result (SDC upper
cutoff ) who truly have digoxin-induced toxicity is 59%. For an individual patient with SDC
cutoff, the probability of toxicity is 0.59.
506 Chapter 26 VI. B
b. For NPV, this means that the proportion of patients with a negative test result (SDC upper
cutoff ) who truly are nontoxic is 95%. For an individual patient with SDC cutoff, the prob-
ability of nontoxicity is 0.95.
c. For PLR, digoxin-induced toxic patients will have a positive test result seven times more
often than do nontoxic patients.
d. For NLR, digoxin-induced toxic patients will have a negative test result 20% as often as
will nontoxic patients.
e. These results suggest that a negative digoxin test result rules out toxicity (0.95) about 1.6
times (0.95/0.59) as effectively as a positive test rules in digoxin-induced toxicity (0.59). A
positive test appears to be unreliable as an indicator of digoxin-induced toxicity, but a
negative test appears to be highly predictive of nontoxicity. Furthermore, the PLR suggests
that a posi-tive SDC test is six times more likely to come from a digoxin-induced toxic than
a nontoxic patient. On the other hand, the NLR implies that it is one-fifth as likely (0.2) that
a negative test comes from a digoxin-induced toxic compared to a nontoxic patient.
f. Knowledge of test performance characteristics of SDC measures provides the practitioner
with an index of the usefulness of the SDC in categorizing patients.
C. Using a Bayesian approach. Using SDC in conjunction with likelihood ratio information enhances the
application of the SDC in decision making. A Bayesian approach to probability revision allows the
practitioner to make a pretest assessment of patient status, order a diagnostic test, and use the probability
information contained in the test result to revise the assessment of status.
1. The relation between pretest, test, and posttest assessment is shown in the following equation:
(pretest odds)(likelihood ratio) (posttest odds) (13)
where pretest refers to the pretest odds of the condition being present prior to obtaining the patient’s
SDC and posttest refers to the posttest odds of the condition being present after learning the SDC.
2. Odds are defined as results divided by results ( / ). Probability is defined as results divided by
total results ( /total, where total is the sum of and results).
3. Odds are converted to probability as follows:
Probability odds/(1 odds) (14)
4. Probability is converted to odds as follows:
Odds probability/(1 probability) (15)
5. An example of applying a Bayesian approach to modifying the probability of patient status.
Using the digoxin test performance characteristics noted in VI.B.5:
a. Assume that a practitioner assesses by visual inspection that the probability of digoxin-
induced toxicity in a patient is 0.25 (the pretest probability). She orders a serum digoxin
concentration (SDC), and uses the measurement to revise her assessment of digoxin-
induced toxicity in the patient (the posttest probability).
b. Further assume that the test performance characteristics for the SDC are: PLR 7 and NLR 0.2.
c. Using equations (13), (14), and (15), a pretest probability of 0.25 is the same as pretest odds of
toxicity of ⅓ [0.25/(1 0.25)], using equation (15).
d. If the SDC test for the patient comes back positive (SDC 2 ng/mL), then PLR 7 is used to
revise the odds of toxicity. If the SDC test is negative (SDC 2 ng/mL), then NLR 0.2 is used.
e. Assume the patient’s SDC 2.4 ng/mL, then using equation (13) yields (⅓)(7) 2.3. So, the
posttest odds of toxicity are 2.3/1. Converting these odds to probability using equation (14):
[2.3/(1 2.3) ] 0.71. The posttest probability of toxicity is 0.71. The pretest probability of
0.25 has nearly tripled (0.71/0.25) using the SDC as feedback.
f. Assume instead that the patient’s SDC 1.4 ng/mL; then using equation (13) yields (⅓)
(0.2) 0.07. So, the posttest odds of toxicity are 0.07/1. Converting these odds to probability
using equation (14): [0.07/(1 0.07) ] 0.07. The posttest probability of toxicity is 0.07.
g. Although a positive SDC test nearly tripled the probability of toxicity earlier, a negative test
cuts the probability to less than ⅓ of its pretest value. This demonstrates the usefulness of
us-ing SDC in combination with practitioner assessment as a guide to quantifying the
probability of patient status.
Clinical Pharmacokinetics and Therapeutic Drug Monitoring 507
VII. SUMMARY
A. TDM applies to a small number of drugs with a narrow range of effective and safe SDC wherein
optimum drug treatment is promoted by maintaining SDC within a population or patient-specific
therapeutic range, above which drug-induced toxicity occurs too often and below which the drug is
too often ineffective.
B. A practitioner initiates drug treatment using a dose rate that assumes that the patient shows mean
population values for the pharmacokinetic variables, even though it is expected that few patients
will ever possess the mean value being used. If the resulting Css and/or pharmacological response
is other than expected and the patient becomes at risk for subtherapeutic or drug-induced toxicity,
it is likely due to the interpatient variability in pharmacokinetic values and pharmacodynamic
response that characterizes the need for TDM; therefore, the dose rate is modified to produce a
patient-specific Css that represents the best tradeoff of effectiveness and toxicity.
C. Timing of sampling of SDC is critical to reduce errors in interpretation of the measurement. SDC
should be sampled at steady state, after postabsorption and postdistribution equilibrium is achieved, and
a time during ss that matches the time at which the therapeutic range was established.
D. T he choice of Cmax,ss, Cmin,ss, Cavg,ss, or Cinf,ss to estimate dosage regimens depends on the
therapeutic range objective and, in the latter case, intravenous rather than intermittent administration.
E. For the renal-impaired patient, the dosage reduction factor is calculated to achieve, depending on
the therapeutic range objective, a Cmax,ss or Cavg,ss in the renal-impaired patient that is similar to
that desired if the patient had normal renal function.
F. T ehSDC measure is more than a number used to relate the patient’s value to a population
therapeutic range. The SDC is a form of diagnostic test used: (1) to assist in classifying patient
status and (2) as feedback to revise practitioner estimates of patient status. Therefore, it is
important to know the pre-dictive values and likelihood ratios of SDC tests.
Study
Questions 2. T he therapeutic range for digoxin is often stated as
0.8 to 2.0 ng/mL. What does this mean?
Directions: Each of the numbered items or incomplete (A) Fifty percent of people taking digoxin show a safe
statements in this section is followed by answers or by and effective response when the serum drug
completions of the statement. Select the one lettered concentration is between 0.8 and 2.0 ng/mL.
answer or completion that is best in each case. (B) Most people achieve the desired response to
digoxin with minimum adverse effects when the
1. Define therapeutic drug monitoring. What is meant by
serum digoxin concentration is maintained between
the term TDM? 0.8 and 2.0 ng/mL. Fewer patients are managed
(A) T he use of drug serum concentration effectively at 0.8 ng/mL, but some may respond
measurements for drugs in which there is (1) a quite appropriately at lower levels. The frequency
correlation between serum concentration and of adverse effects increases as the level increases
response and (2) a narrow range of effective above the upper limit of the therapeutic range, but a
and safe concentrations, to assess patient status few patients are managed effectively without
as an adjunct to clinical observation adversity above the range.
(B) T he use of drug serum concentration (C) Twenty-five percent of people show an effective
measurements to determine population values for response to digoxin at 0.8 ng/mL, and 75% show
a drug’s half-life value an effective response at 2.0 ng/mL.
(C) T he use of drug serum concentration (D) Twice daily administration of digoxin, but not
measurements to assess the accuracy of the three times daily, requires that serum drug
drug concentration assay concentrations stay within 0.8 to 2.0 ng/mL.
(D) Observing the effects of drugs in human (E) Digoxin serum drug concentrations outside of the
(E) Using drug serum concentration measurements 0.8 to 2.0 ng/mL range are ineffective and/or unsafe.
to differentiate effective from ineffective drugs
508 Chapter 26 5. What is the positive predictive value of
a diagnostic test?
(A) T he fraction of patients with a positive
3. Assume that for digoxin, the therapeutic range is
outcome who have a positive test result.
cited as Cavg,ss 0.8 to 2.0 ng/mL. If the patient is (B) Being more than 50% correct in predicting
assumed to have an estimated digoxin t½ of 48 hrs, success or failure upon using a drug regimen.
how long would you wait to take a serum digoxin (C) T he fraction of patients who achieve a
concentration measurement, and when during would successful response in using a drug.
you schedule it? (D) T he fraction of patients with a positive test
(A) 28 days, then 3 to 4 hrs after the dose result who turn out to have a positive outcome.
is administered (E) T he probability that knowledge of a drug
(B) 14 days, then 6 to 8 hrs after the dose serum concentration results in a successful
is administered response to treatment.
(C) 7 days, then 10 to 14 hrs after the dose
6. A 70-year-old, 80-kg male with serum creatinine of
is administered
3 mg/dL is scheduled to start tobramycin therapy.
(D) 3 days, then 1 to 2 hrs after the dose
is administered What regimen is recommended to achieve Cmax,ss
within 5 to 10 mcg/mL (use the midpoint of 7.5 mcg/
(E) 1 day, then 18 to 22 hrs after the dose
is administered mL for the calculation) and Cmin,ss 2 mcg/mL. Try
an every 24-hr regimen to start and, if unsuccessful
4. Differentiate linear from nonlinear drug in achieving the target concentration goals, alter and
clearance. What is the effect on TDM? recalculate. Assume in normals the following values:
(A) Linear drug clearance is first order, the Cl and t½ 2.5 hrs, V 0.25 L/kg, F 0.98, S 1, f 1, Clcr 120
t½ are independent of drug dosage, and mL/min
proportional changes in dose result in the same (A) A loading dose of 1.8 to 2.0 mg/kg followed
proportional changes in Css. Nonlinear drug by 1.0 mg/kg every day
clearance is zero order, Cl and t½ change as (B) A loading dose of 1.8 to 2.0 mg/kg followed
dose changes (or as the amount of drug in the by 1.5 mg/kg every day
body changes), and proportional changes in (C) 2.0 mg/kg every day
dose yield disproportionate changes in Css. (D) 1.0 mg/kg every day
(B) Linear drug clearance presents fewer (E) 0.5 mg/kg every day
serum concentration peaks and troughs
during the dosage interval than does
nonlinear drug clearance.
(C) Linear drug clearance is zero order, the Cl and
t½ are dependent on drug dosage, and
proportional changes in dose do not result in the
same proportional changes in Css. Nonlinear
drug clearance is first order, and equations are
not available to predict drug serum
concentration from the dose rate.
(D) Drugs with linear clearance have shorter t½
values than drugs with nonlinear clearance.
(E) Drugs with linear clearance are administered
less often than drugs with nonlinear clearance.
Clinical Pharmacokinetics and Therapeutic Drug Monitoring 509
I. PEDIATRIC PHARMACOTHERAPY
A. General considerations
1. Most pharmacists in community and hospital settings provide care for children. In the United
States, children make up nearly one-third of the total population. Although children typically
require fewer medications than adults because of their relative good health, approximately 30%
of all prescriptions filled in community pharmacies are for pediatric patients. As a result,
pharmacists need to have a basic knowledge of pediatric pharmacotherapy to
appropriately assess and monitor drug therapy.
2. Providing care for children is often a challenge. There is limited information on the selection,
dosing, and monitoring of drugs in this population. It is estimated that only 25% of the drugs
available on the market in the United States carry a U.S. Food and Drug Administration (FDA)-
approved indication for use in pediatric patients, although nearly 75% have been used to treat
children. As a result, most pediatric dosing is considered off-label. Dosing information
typically comes directly from case reports and small clinical trials published in the medical
literature. In addition to being able to obtain pediatric-specific drug information, pharmacists
must also be familiar with differences in pediatric pharmacokinetics and phar-macodynamics as
well as the unique aspects of medication monitoring and compliance in this population.
B. Pharmacokinetic considerations
1. Unlike the relatively stable pharmacokinetic profile of most drugs in adults, children’s
pharmacokinetic parameters change during maturation from neonates into adolescents
(Table 27-1). As a result, the pediatric population is a diverse and dynamic group. Each aspect
of drug disposition is affected, including absorption, distribution, metabolism, and elimination.
None of these processes is fully mature at birth, and they develop at different rates over the first
years of life. The study of these changes is known as developmental pharmacology.
Newborn
a
Preterm or premature 36 weeks gestation
a
Term 36 weeks gestation
Neonate 1 month
Infant 1 month–1 year
Child 1–11 years
Adolescent 12–16 years
510
Pediatrics 511
2. Drug absorption. For most routes of drug administration, the greatest change in absorption
takes place during infancy. Differences in drug absorption during this period may affect the
choice of delivery method, dose, or monitoring.
a. Gastrointestinal absorption. Oral drug absorption is most greatly altered during the first
year of life. Several aspects of absorption are age dependent, including gastric pH, gastric
emptying time, intestinal motility, bile salt production, and pancreatic enzyme function.
(1) Elevated gastric pH. At birth, gastric pH is elevated to nearly neutral. This results
from a relative decrease in gastric acid production and an overall decrease in the
volume of gastric secretions. As a result, acid-labile drugs such as penicillin G may
have a greater bioavailability in neonates than in older infants. Conversely, drugs that
are weak acids such as phenobarbital (Luminal) and phenytoin (Dilantin) may not be as
well absorbed. To compensate for this effect, neonates may require larger oral doses of
these drugs than older infants to produce the desired therapeutic effect. Acid production
rises steadily after birth but do not reach adult levels until 2 to 3 years of age.
(2) Prolonged gastric emptying and intestinal motility. T he rate of passage of drugs
through the gastrointestinal tract is also important in determining the rate and extent of
their absorption. Coordination of contractions within the stomach begins to improve
shortly after birth, whereas intestinal peristalsis increases more slowly, over the first 4
to 8 months of life. As a result of this prolonged transit time, absorption of drugs may
be significantly slowed.
(a) Preterm neonates typically have more delayed gastric emptying time than term
neonates, so that changes in oral absorption may be most pronounced in these patients.
Clinical studies have demonstrated that peak acetaminophen (Tylenol) serum concen-
trations occur up to 2 hrs later in premature neonates compared to older infants.
(b) Breastfed infants empty their stomachs approximately twice as fast as formula-fed
infants. The increased caloric density of formula feedings delays gastric emptying.
(3) Bile salt and pancreatic enzyme production. The rate of bile salt synthesis in preterm
and term infants is approximately 50% of adult values. Decreased fat absorption from
enteral feedings, as well as decreased drug absorption, can occur. For example, when
lipid-soluble vitamin D is administered to infants, bioavailability is only 30% as
compared with 70% in adults. The absorption of lipid-soluble drugs is further reduced
due to lower levels of pancreatic enzymes.
(4) Other factors affecting oral drug absorption include reduced splanchnic blood flow in
the first month of life and reduced activity of intestinal metabolic enzymes, which
alters the first-pass effect. In addition, neonates lack normal gut microflora. Although
bacterial colonization normally occurs shortly after birth, it may be significantly
delayed in preterm neonates who are being cared for in the sterile environment of an
intensive care unit.
b. Percutaneous absorption
(1) Absorption of drugs through the skin is enhanced in infants and young children owing
to better hydration of the epidermis, greater perfusion of the subcutaneous layer, and
the larger ratio of total body surface area to body mass compared to adults.
(2) In preterm neonates, the stratum corneum is also thinner, further increasing the
potential for the absorption of topical products.
(3) This route of administration should be used with caution in infants and young children to
avoid overdosage. There are numerous accounts of toxicity resulting from percutaneous
absorption. Repeated use of the skin cleanser hexachlorophene (Phisohex) has resulted in
seizures in preterm neonates and application of povidone–iodine (Betadine) to prepare the
skin for surgery or sterile procedures has caused iodine toxicity in infants. Repeated
applications of topical hydrocortisone cream for diaper rash or eczema can produce adrenal
axis suppression after as little as 2 weeks of use in an infant.
c. Intramuscular absorption
(1) The absorption of drugs administered by this route may be reduced in infants as a result of
reduced blood flow to skeletal muscles. In addition, weak or erratic muscle contractions in
neonates may result in reduced drug distribution. These factors may be partially offset
512 Chapter 27 I. B
by the higher density of capillaries in skeletal muscle during infancy, which increases
blood circulation.
(2) Intramuscular administration of drugs is generally discouraged in the pediatric
population because of the pain associated with the injection and the risk of nerve
damage from inadvertent injection into nerve tissue.
(3) This route is generally reserved for the administration of vitamin K where it produces a
longer lasting depot-like effect to prevent hemorrhagic disease of the newborn,
vaccines, and occasionally antibiotics when intravenous access is not available. If it is
used, the volume should not exceed 0.5 mL for infants and younger children and 1.0
mL for older children.
d. Rectal administration. Absorption by this route is fairly reliable, even for preterm
neonates. Administration may be complicated in infants; however, by the increased number
of pulsa-tile contractions in the rectum compared to adults, making expulsion of a
suppository more likely.
e. Pulmonary administration. Inhalation of medications is increasingly being used in infants
and older children to avoid systemic exposure. Although developmental changes in the
pulmonary vasculature and respiratory mechanics likely alter the pharmacokinetics of drugs
given by inhalation, little is known about the effects of growth and maturation on this route
of drug administration.
3. Drug distribution. Growth and maturation affect many of the factors that determine drug
distri-bution. Body water content, fat stores, plasma protein concentrations, organ size and
perfusion, hemodynamic stability, tissue perfusion, acid–base balance, and cell membrane
permeability all undergo significant changes from infancy to adolescence.
a. Body water and fat content. Total body water content decreases with increasing age. This
is primarily the result of a larger extracellular body water content in neonates and young
infants that decreases with age. Approximately 80% of a newborn’s body weight is water.
By 1 year of age, this value declines to 60%, similar to that of an adult. Highly water-
soluble compounds, such as gentamicin (Garamycin), have a larger volume of distribution
in neonates than in older children. As a result, larger milligram per kilogram doses are often
needed to achieve desired therapeutic concentrations. Conversely, body fat increases with
age from 1% to 2% in a preterm neonate to 10% to 15% in a term neonate and 20% to 25%
in a 1-year-old. Lipophilic drugs, such as diazepam (Valium), have a smaller volume of
distribution in infants than in older children and adults.
b. Protein binding. Acidic drugs bind to albumin, whereas basic substances bind primarily to
1-acid glycoprotein (AGP). The quantity of total plasma proteins, including both of these
substances, is reduced in neonates and young infants. In addition, the serum albumin of new-
borns may have a reduced binding affinity. These two factors result in an increase in the free
fraction of many drugs (Table 27-2). The increase in the free fraction may result in enhanced
pharmacological activity for a given dose. The relative decrease in serum proteins may also
produce increased competition by drugs and endogenous substances, such as bilirubin, for
binding sites. Drugs that are highly bound to albumin, such as the sulfonamides, may displace
bilirubin from its binding sites and allow deposition in the brain, referred to as kernicterus. As a
result, these drugs are contraindicated in the first 2 months of life.
c. Although most of the recent research into pediatric drug metabolism has focused on the
development of enzyme function in neonates, several new studies highlight additional
changes in metabolic function during adolescence. For example, lopinavir (marketed with
ritonavir as Kaletra) pharmacokinetics undergoes significant age and gender-related
changes at the time of puberty. Lopinavir clearance increases by more than 30% in boys
after age 12, compared to minimal changes in clearance in girls after the onset of puberty.
5. Elimination. Development of renal function is a complex and dynamic process. Nephrons begin
forming as early as the ninth week of gestation and are complete by 36 weeks. Although the
functional units of the kidneys are present, their capacity is significantly reduced at birth.
Glomerular filtration rate in neonates is approximately half that of adults. Values are further reduced
in preterm neonates. Glomerular filtration rate increases rapidly during the first 2 weeks of life and
typically reaches adult values by 8 to 12 months of age. Tubular secretion rate is also reduced at
birth to approximately 20% of adult capacity but matures by 12 months of age.
a. Immature renal function results in significant alterations in the elimination of many
drugs. Pharmacokinetic studies for several drugs, including the aminoglycosides, digoxin
(Lanoxin), and vancomycin (Vancocin), have shown a direct correlation between clearance
and patient age. Prolongation of the half-life should be anticipated for all renally eliminated
drugs administered to neonates and is most pronounced in preterm neonates.
b. To account for the reduced ability of a neonate to eliminate these drugs, longer dosing
intervals are often required. Failure to account for the reduction in renal function may
result in drug accumulation and toxicity. Dosage adjustment is typically based on urine
output, with values greater than 1 mL/kg/hr indicating adequate renal function. Although a
trend towards increasing serum creatinine over a period of time may indicate worsening
renal function, calculation of creatinine clearance is not routinely used to determine drug
dosing in pediatrics. Creatinine values in neonates may be falsely elevated during the
perinatal period due to transfer of maternal creatinine across the placenta, whereas values in
older infants and young children may be low due to limited muscle mass and provide
misleading assurance that renal function is adequate. As a result, the equations used for
determining creatinine clear-ance, such as Cockcroft–Gault or Modification of Diet in Renal
Disease (MDRD), are not considered appropriate for use in infants and children.
C. Pharmacodynamic considerations. Unlike the rapidly accelerating knowledge of the pharma-
cokinetic changes associated with development, less is known of the pharmacodynamic changes
associated with maturation. Preliminary investigations of warfarin (Coumadin) pharmacodynamics
have demonstrated a relationship between greater anticoagulant response and increasing patient
age. Studies have suggested that neonates may have lower numbers of -adrenergic receptors,
reduced binding affinity, or conformational changes of the receptor binding site, which might
explain their decreased responsiveness to dopamine and epinephrine for the treatment of
hypotension. Future research in age-related pharmacodynamic changes is needed to optimize the
safe and effective use of drugs in infants and children.
D. Pediatric drug administration and monitoring. Drug administration, including the selection of agent
and dose as well as the preparation of the dose and therapeutic drug monitoring, is complicated in the
pediatric population. Pharmacists caring for children must not only be knowledgeable about the changes
in pharmacokinetics and pharmacodynamics described previously, but must also be able to use this
information in the assessment of the appropriateness of a medication order or prescription. In addition,
they should understand the need to carefully check all dosage calculations and be able to alter available
dosage formulations when necessary to tailor them for infants and young children.
1. Accurate dosage calculations are critical in the care of infants and children. Pharmacists should
use pediatric dosing information available in general drug references or pediatric-specific references
such as The Pediatric Dosage Handbook (Lexi-Comp), the Harriet Lane Handbook (Mosby), or
Neofax (Thomson Reuters). To account for differences in pharmacokinetic parameters, most pe-
diatric doses are based on body weight. In the case of some drugs, such as chemotherapy, doses are
based on body surface area (BSA). This value can be determined from the patient’s height and
weight, using either a nomogram or the following equation:
height (cm) weight (kg)
2
BSA (m ) ___
3600
Pediatrics 515
There is no absolute rule for when adolescent patients should start to be dosed as adults. In
general, adult dosing guidelines may be used in patients weighing more than 40 to 50 kg or
when the calculated weight-based pediatric dose exceeds the standard adult dose.
2. The need to calculate pediatric doses introduces a greater risk for dosage errors. Dosing errors
have been found to be more common in medication orders for children than in any other patient
population. Studies suggest a medication error rate of 5% to 15% in pediatric inpatients, with the
greatest number of errors occurring during the ordering process. The need to calcu-late individual
doses, along with potential decimal errors and transcription errors, increases the potential for
mistakes. In addition, there is often a wide range of patient ages and weights within a single hospital
or clinic. It is not uncommon to have patient doses vary by 10-fold, as an infant weighing 5 kg and
an adolescent weighing 50 kg may be cared for by the same health care pro-viders. Pharmacists
practicing in a neonatal intensive care unit may have extremely low birth weight patients weighing
as little as 0.5 kg alongside large infants with congenital or birth-related complications weighing 5
kg. All calculations should be double-checked, and orders outside of the normal pediatric dosage
range verified with the prescriber. Computerized physician order entry (CPOE) with embedded
clinical decision support and dose-checking software reduces the potential for errors but does not
replace the need for order review by a trained pharmacist.
3. Dosage formulations may need to be altered to make them useful for infants and children.
Because young children cannot typically swallow tablets and capsules, these solid dosage
formulations must often be converted to oral solutions or suspensions. Several compounding
resources, including Extemporaneous Formulations for Pediatric, Geriatric and Special Needs
Patients (American Society of Health-System Pharmacists) and Pediatric Drug Formulations
(Harvey Whitney Books), are available to provide pharmacists with formulations that have
been tested to ensure drug stability. With coaching, most children can learn to swallow solid
dosage forms between 6 and 8 years of age.
4. Medications must be stored appropriately and doses measured accurately. Families should be
given complete instructions for the storage, use, and disposal of all medications for children. This
should include instructions on the accurate measurement of oral liquids. Oral syringes are preferred
for infants, but oral dosing spoons and cups may be used for older children. Written information,
ideally targeted to the pediatric patient population as in The Pediatric Medication Education Text
(American College of Clinical Pharmacy), is a useful adjunct to oral communication and allows
families to provide detailed instructions to extended family members, preschool and school
personnel, babysitters, and other care providers. Safe storage of all household medications should be
stressed with all families as a part of good poison prevention practices.
5. Intravenous (IV) medications may be prepared as more concentrated solutions because of
the limits on fluid administration to infants and young children. Although a typical adult
may receive up to 4 to 5 L of IV fluids per day, the total daily fluid requirements for a preterm
neonate may be as little as 20 to 50 mL. As a result of these limitations, medication volumes
are typically minimized in order to allow more of the daily total to be used to provide either
enteral or parenteral nutrition. Special equipment, such as syringe pumps and microbore tubing,
is used to ensure accurate delivery of drugs in small fluid volumes. Currently available syringe
pumps can deliver volumes as small as 0.1 mL over 1 hr. Smart pump technology, which
incorporates dose checking and the ability to set dosing limits, has significantly reduced the
frequency of IV medication errors in hospitalized pediatric patients. Microbore IV tubing is
used to minimize the amount of dead space between the delivery device and the patient, further
improving the accuracy of drug delivery. Pharmacists must also be capable of assessing
literature on the compatibility with drugs and IV fluids because pediatric patients often have
limited IV access, so that multiple solutions may need to be infused through the same IV site.
E. Therapeutic drug monitoring is often essential to optimizing drug therapy in infants and children. For
most drugs, including aminoglycosides, antiepileptics, cyclosporine, methotrexate, tacrolimus, and
vancomycin, the therapeutic ranges developed for and used in adults are also appropriate for monitoring
pediatric patients. A potential complication in interpreting serum drug concentrations is the presence of
endogenous substances, which may cross-react with analytical drug assays. This has been demonstrated
for digoxin (Lanoxin) in neonates and infants, for whom endogenous digoxin-like reactive substances
(EDLRS) may produce falsely elevated serum digoxin concentrations. Standard assay techniques can be
modified to exclude EDLRS as a complicating factor.
516 Chapter 27 I. F
F. Pharmacists should also be aware of differences in adverse drug reactions between children
and adults. Most of the adverse reaction information available in drug product labeling or cited in
pharmacy references has been obtained from clinical trials in adults. As several studies have
demonstrated, the adverse reaction profile in children may be significantly different from that
observed in older subjects. For example, severe dermatologic reactions to lamotrigine (Lamictal),
including Stevens–Johnson syndrome, were infrequent during premarketing phase III clinical
trials. When the drug was introduced into the market in the United States and began to be used in
children off-label, a higher rate of dermatologic reactions was reported in children. Subsequent
research revealed the incidence of severe dermatologic reactions to be 0.8% in children compared
to 0.3% in adults. A reduction in the pediatric starting dose, along with recommendations for a
more gradual dose escalation, has reduced the number of severe reactions. There are also adverse
effects known to occur primarily in children due to pharmacokinetic differences. Valproic acid-
induced hepatotoxicity has been reported most frequently in children less than 2 years of age,
likely related to their reduced ability to eliminate the toxic 4-en-valproate metabolite. Other
examples of drug reactions that occur more often in children than adults include the development
of Reye syndrome, a derangement of cellular mitochondrial function resulting in liver failure
following aspirin use, and the higher incidence of renal dysfunction with nonsteroidal anti-
inflammatory drugs used during the first 6 months of life. Differences such as these further
highlight the need for more clinical research in children.
G. In addition, pharmacists should take an active role in promoting medication adherence (compli-
ance) in children. Several studies have shown adherence rates in pediatric patients to be 30% to 70%. It
is surprising that some of the poorest adherence rates have been associated with chronic diseases such as
asthma, epilepsy, and diabetes and in children requiring immunosuppressive therapy after organ
transplantation. As in adults, counseling about medication adherence should include efforts to identify
and overcome barriers to therapy, education about the importance of medical management, and
programs to incorporate medication regimens into normal daily tasks. When working with fami-lies of
younger children, pharmacists should also explore problems with dosage formulation (such as taste or
texture aversion) and dosing frequency. Medications with once or twice daily dosing avoid the need for
administration at school or day care and have been shown to increase adherence. In older children and
adolescents, pharmacists should be aware of their growing need for autonomy and work with the patient
and his or her family to foster the goal of self-care. Most children can begin to take an active role in
their medication preparation and administration by the age of 12, although the age at which they can
assume responsibility for their treatment varies considerably. As part of an interdisciplinary health care
team, pharmacists can serve a vital role in improving medication adherence, reducing the likelihood of
treatment failure and supporting a lifelong commitment to a healthy lifestyle among their pediatric
patients.
Study Questions
Directions for questions 1–14: Each of the questions, 1. Which of the following variables will most likely
statements, or incomplete statements in this section can be used to calculate doses for AH’s antibiotics?
be correctly answered or completed by one of the (A) Height
suggested answers or phrases. Choose the best answer. (B) Hepatic function
For questions 1–4: AH is a 1.2-kg female born prematurely (C) Weight
at 30 weeks of gestational age. Her mother had an infection (D) Age
with a fever at the time of delivery. AH was admitted to the (E) Serum creatinine
neonatal intensive care unit for presumed sepsis and placed
on empiric antibiotic therapy with ampicillin (50 mg/kg IV
every 12 hrs) and gentamicin (2.5 mg/kg IV every 8 hrs).
2. After the administration of four doses of gentamicin, a Pediatrics 517
serum concentration obtained 5 mins before the next
dose was 2.5 mcg/mL. Which of the following answers
best describes the pharmacokinetic differences observed 6. Administration of a sulfonamide antibiotic may
in premature neonates (compared to older children and displace bilirubin from binding sites on which of the
adults) that might explain this value? following substances, leading to passage of bilirubin
(A) Larger volume of distribution, longer half-life into the brain and the development of kernicterus?
(B) Larger volume of distribution, shorter half-life (A) 1-acid glycoprotein
(C) Smaller volume of distribution, longer half-life (B) albumin
(D) Smaller volume of distribution, shorter half-life (C) -adrenergic receptors
(E) Similar volume of distribution, shorter half-life (D) uridine 5 -diphosphate glucuronosyltransferase
3. Ampicillin may exhibit pharmacokinetic differences (E) sulfotransferase
in AH because of its protein binding characteristics. 7. Caffeine administered to neonates for the treatment
Which of the following answers best describes the of apnea of prematurity undergoes metabolism
effect of AH’s age on ampicillin protein binding? through which of the following cytochrome P450
(A) Increased protein binding, resulting in a greater enzymes, also the last major drug-metabolizing
free fraction enzyme to develop after birth?
(B) Increased protein binding, resulting in a reduced (A) CYP1A2
free fraction (B) CYP2C19
(C) Decreased protein binding, resulting in a greater (C) CYP2D6
free fraction (D) CYP3A4
(D) Decreased protein binding, resulting in a reduced (E) CYP3A7
free fraction
8. Both glomerular filtration and tubular secretion rates
(E) Decreased protein binding, resulting in no
are reduced in infants compared to adults. Which list
significant change in free fraction
of drugs is the most likely to have prolonged
4. As the pharmacist providing services for the neonatal clearance during infancy because of immature renal
intensive care unit, you evaluate medication orders function alone?
and make recommendations to the medical team. (A) Amikacin, caffeine, and retinoic acid
Which of the following would be the most (B) Acetaminophen, atomoxetine, and retinoic acid
appropriate recommendation for AH’s care? (C) Acetaminophen, benzyl alcohol, and vancomycin
(A) Change to oral antibiotics for better absorption. (D) Amikacin, gentamicin, and vancomycin
(B) Double-check the calculations to avoid (E) Atomoxetine, lopinavir, and vancomycin
decimal errors.
9. As a pharmacy manager in a children’s hospital, you
(C) Dilute the gentamicin with a larger volume of
are responsible for implementing new technologies
IV fluids to make it easier to measure.
that have the potential to reduce medication errors in
(D) Use a pediatric-specific therapeutic range
children. Which of the following has the most
for monitoring gentamicin.
(E) Change to sulfamethoxazole and trimethoprim potential to reduce the largest number of errors?
(Bactrim) for single-agent antibacterial coverage. (A) Bar-code scanning
(B) CPOE with dose-checking software
5. Although the total body water content of an adult
(C) Inventory management systems
accounts for approximately 60% of body weight, the
(D) Smart pump technology
total body water content of a healthy newborn is (E) Standard IV concentrations
(A) 40%.
10. You are counseling the grandmother of a 3-year-old boy
(B) 50%.
who has a prescription for amoxicillin/clavulanate
(C) 70%.
(Augmentin) to treat uncomplicated otitis media. Which
(D) 80%.
of the following issues would be least likely to affect
(E) 90%.
medication adherence (compliance)?
(A) Lack of education about the medication
(B) Cost
(C) Dosing interval (frequency)
(D) Taste
(E) Autonomy
518 Chapter 27
I. DRUG USE IN GERIATRIC PATIENTS. In 2000, more than 12% of the American
population was 65 years of age, representing more than 35 million Americans, while consuming
approximately 30% of all health care costs. It is estimated that three out of every four elderly people are
taking prescription medications. Anticipated total drug usage, including nonprescription medications,
reveals that 50% of all drugs used in the United States are used by the geriatric population.
A. Adverse drug reactions. Geriatric patients are at increased risk for drug-induced adverse effects.
Incidence of adverse drug reactions (ADRs) in patients older than 65 years is two to three times
greater compared to younger patients. The risk is five times higher in people approaching age 90.
One in five of all elderly patients experience an ADR. In some patients, these are overlooked
because they mimic the characteristics of other diseases.
1. Factors that are responsible for the higher prevalence of ADRs in the geriatric population
include polypharmacy, multiple disease states, altered pharmacokinetic disposition among the
elderly (reduced drug elimination, increased sensitivity to drug effects, etc.), poor relation-ship
with health care providers, increasing severity of illness, and poor patient compliance and
supervision.
a. Studies have shown that 35% of geriatric patients living in the community use six or more
medications; approximately one-half of patients residing in long-term care facilities use five
or more medications. People 65 years and older use approximately 40% of the drugs
prescribed and 50% of the over-the-counter (OTC) medications taken.
b. Patients taking multiple medications have a greater chance of experiencing ADRs owing to
drug–drug interactions and the potential for overlap or synergy between adverse effect
profiles.
c. Patients with multiple disease states are at higher risk for having a drug–disease state interaction.
d. In addition to the aforementioned risk factors for developing an ADR, it is difficult to
predict how geriatric patients will respond to any given medication owing to altered
pharmacokinetic and pharmacodynamic profiles.
e. Another issue complicating geriatric drug therapy is adherence. Factors that have been
shown to increase nonadherence include poor relationships with health care providers,
lower socioeconomic status, living alone, polypharmacy, complicated drug regimens, and
multiple comorbidities. As many as 60% of geriatric patients do not take their medications
as prescribed and may self-medicate as often as once a week. If patients are hospitalized,
their prescribed drug doses may represent a significant overdosage or underdosage, which
could cause unintended effects.
f. Elderly patients can have diseases that make adhering to drug therapy difficult. Conditions
that affect vision, such as macular degeneration or cataract formation, can make reading
prescription labels and medication instructions troublesome. Hearing loss can prevent
patients from understanding and health care professionals from effectively communicating
medication information and patient instructions. Arthritis can add to the difficulty of
opening medication bottles. In these instances, providing patients with medication or “pill”
boxes and written medication lists may limit potential barriers to patient adherence.
Recognizing these factors, pharmacists can increase adherence in elderly patients.
519
520 Chapter 28 I. A
2. Pharmacists can provide recommendations to eliminate unnecessary drug therapy and monitor
medication profiles to avoid potential drug–drug or drug–disease state interactions that may
prove harmful. Efforts to optimize drug therapy, including simplifying and employing more
cost-effective regimens, may ultimately afford better patient adherence.
B. Pharmacokinetics. Pharmacokinetic parameters may be altered in the elderly owing to age-related
physiological changes. Specific age-related physiological changes affecting drug therapy are given
in Table 28-1.
1. Absorption. Physiological changes that can alter absorption in the geriatric population include
delayed gastric emptying, decreased splanchnic blood flow, elevated gastric pH, and impaired
intestinal motility. Although the rate of drug absorption may be altered in some patients, the ex-tent
of absorption is rarely affected, and this represents the least affected parameter in the elderly.
2. Distribution. Several age-related physiological changes may affect drug distribution.
a. Elderly patients have a decrease in total body water with a proportionate increase in body
fat, causing water-soluble drugs (e.g., digoxin [Lanoxin®]) to have a smaller volume of
distribu-tion. Peak concentrations of water-soluble agents will be higher, and loading doses
may need to be adjusted to incorporate this change.
b. The volume of distribution of lipid-soluble drugs (e.g., diazepam [Valium®]; propranolol [Inderal®])
is increased due to the proportionate increase in body fat, which will result in their accumulation in fat
stores and increasing their duration of action. For some patients, this will increase the risk of adverse
effects due to prolonged sedation as well as greater predisposition to falls.
c. The aging process has also been shown to affect the pharmacokinetics of drugs that are
highly protein bound. Drugs that are highly bound to albumin (e.g., warfarin [Coumadin®];
phenytoin [Dilantin®]) may have a greater free concentration due to decreased albumin
levels in the elderly who are chronically ill.
d. Additionally, there are other substances, some medications as well as other substances
within the body, that may alter binding affinity in the elderly to albumin, or albumin may
even experience a change in configuration, which can also lower binding capacity.
Competition for acidic binding sites could occur with accumulated endogenous substances,
altering the free fraction of drugs.
Pharmacokinetic
Gastrointestinal motility ↓ May affect the rate but not the extent of drug
absorption
Gastric pH ↑ No signifi cant change in drug absorption
Renal function ↓ Reduced elimination of renally excreted drugs
Serum albumin ↓ Decreased protein binding leading to an increased
free fraction of drug
Phase I hepatic metabolism ↓ Potential accumulation of drugs metabolized by
oxidation, reduction, or hydrolysis reactions
Body fat to lean muscle mass ↑ Increased volume of distribution of fat-soluble drugs
ratio Total body water ↓ Decreased volume of distribution of water-soluble
drugs
Pharmacodynamic
e. Although 1-acid glycoprotein (AGP) concentrations tend to increase with age, the increase
in the concentrations of basic drugs (e.g., lidocaine [Xylocaine®]; propranolol [Inderal®])
that bind to AGP are usually clinically meaningful only in acutely ill patients.
f. Based on available clinical literature, the elderly have a higher predisposition to the acute effects
of medications when combinations of highly protein bound agents are given concomitantly
without taking into account the drug–drug interaction taking place at protein binding sites. Drugs
such as phenytoin (Dilantin®), diazepam (Valium®), propranolol (Inderal®), valproic acid
(Depakene®), warfarin (Coumadin®), and salicylates require extra monitoring in the elderly in
order to avoid the consequences associated with altered protein binding.
3. Renal excretion. Perhaps the best documented and most predictable age-related physiologi-cal
change is the decline in renal function, specifically glomerular filtration rate and the tubular
secretion rate.
a. In patients without renal dysfunction, it is estimated that there is a 50% decline in renal
function by age 80. Renal blood flow is reduced by approximately 10% each decade of life.
Estimates of the serum creatinine level are the closest predictor of renal function that is
currently available. As serum creatinine increases, there is a reciprocal reduction in
creatinine clearance and glomerular filtration rate.
b. Although serum creatinine may be the best predictor of renal function, it is not always
accurate in the elderly because creatinine production also declines with age and with lower
activity levels (such as being bedridden or debilitated).
c. Medications primarily eliminated by the kidneys may have increased concentrations of both
the active drug and its metabolites possibly leading to subsequent adverse effects. All
renally eliminated drugs used in geriatric patients should be monitored for the need for dose
reductions and potential toxicity.
d. Cockcroft and Gault equation is a useful “estimate” of creatinine clearance, but its
limitations in the elderly must be appreciated, due to reduced creatinine synthesis and
consequential reductions in levels due to reduced muscle mass.
(140 age) weight kg
Males: Creatinine clearance (mL/min) ___
72 serum creatinine (mg/dL)
Females:
85% of male value (not applicable if serum creatinine 5 mg/dL)
4. Hepatic metabolism. Age-related changes affecting the liver include a reduction in hepatic
blood flow and a decline in hepatic metabolism.
a. A reduction in phase I reactions (oxidation, reduction, and hydrolysis) occurs in the elderly
and may prolong the activities of drugs metabolized by these enzymes. The long-acting
benzodiazepine diazepam (Valium®), phenytoin (Dilantin®), cimetidine (Tagamet®), and
cer-tain analgesics, which depend on phase I reactions for metabolism, represent situations
in which changes in hepatic metabolism may be important. The elimination half-lives of
these agents would be prolonged and may result in drug accumulation and the
predisposition to adverse drug effects.
b. Phase II reactions (glucuronidation, acetylation, and sulfation) are relatively unchanged in
the elderly. Several short acting benzodiazepines such as oxazepam (Serax®) and
lorazepam (Ativan®) undergo phase II reactions and represent relatively safe agents to
administer to the elderly for that reason.
c. Liver blood flow has been demonstrated to fall approximately 50% between the ages of 20
and 80 years. This becomes a critical parameter for those drugs, which have high extraction
rates by the liver such as propranolol (Inderal®), lidocaine (Xylocaine®), and the earlier
tricyclic antidepressants. Such alterations in the rate of liver extraction would require
careful titration to pharmacologic effect in order to avoid an excess response and the
potential for toxicity.
d. Liver volume and cell mass decrease in the elderly. It appears that this change in size
enhances the reduced activity of oxidative and demethylation metabolism. Conjugation
reactions do not appear to be affected by liver mass.
522 Chapter 28 I. C
C. Pharmacodynamics
1. Geriatric patients can be more or less responsive to certain drugs compared to younger patients.
Reasons for this may include altered receptor sensitivity, receptor number, or receptor response.
Studies have shown that elderly patients may show a diminished response to -blockers and -agonists.
2. In contrast, geriatric patients have an exaggerated response to anticholinergic drugs—drugs
affecting the CNS such as narcotics, analgesics, benzodiazepines, and warfarin. Elderly patients
should be monitored carefully when taking these medications. Initiation of any of these
therapies should be at lower doses than recommended for younger patients. Avoidance of these
drugs may not be possible, but if better alternatives exist, they should be used first.
D. Drug therapy considerations
1. Drug therapy in geriatric patients is an involved process and can be very complex because of
age-related changes in pharmacokinetics and pharmacodynamics.
2. Additionally, a lack of clinical trials designed to evaluate the safety and efficacy of drug
therapy in the elderly population increases the problem.
3. The higher incidence of adverse effects in geriatric patients may be in part the result of the
complexity of drug therapy and the relative lack of clinical trials in this population. Table 28-2
lists several drugs and doses that should be avoided in the elderly owing to higher risks of
adverse effects and/or lack of efficacy.
4. Owing to alterations in gait, balance, and mobility, falls and consequent adverse events are
frequent occurrences in geriatric patients.
a. The high prevalence of osteoporosis in the elderly results in an increased incidence of
fractures. Complications associated with fractures, particularly hip fractures, are significant
causes of morbidity and mortality.
b. Medications causing orthostatic hypotension, drowsiness, dizziness, blurred vision, or confu-sion
have the potential to cause or worsen postural instability and increase falls in the elderly.
c. It is well established that many psychoactive agents, especially long-acting benzodiazepines, are
associated with an increased risk of falls in the elderly. If a benzodiazepine must be prescribed,
low-dose lorazepam (Ativan®) or oxazepam (Serax®) are better choices because of the lack of
active metabolites and their metabolism involves phase II hepatic reactions only.
5. Geriatric patients tend to be sensitive to medications that possess anticholinergic effects. Dry
mouth, urinary retention, blurred vision, constipation, tachycardia, memory impairment, and
confusion are typical anticholinergic adverse effects associated with several classes of drugs
(Table 28-3). These agents can also induce delirium in some people.
a. When possible, drugs with anticholinergic effects should be avoided in the elderly. In those
instances when this is not an option, the least anticholinergic agent should be chosen and
initiated at the lowest effective dose. For example, if a tricyclic antidepressant is needed,
desipramine (Norpramin®) and nortriptyline (Pamelor®) possess less anticholinergic
activity than amitriptyline (Elavil®) and imipramine (Tofranil®) and therefore would be
better initial therapeutic options.
b. Frequent monitoring for and patient and family education on signs and symptoms of possible
anticholinergic adverse effects is always warranted when these drugs are prescribed in the elderly.
E. General principles. To aid clinicians in providing appropriate geriatric drug therapy, some general
principles have been developed.
1. Start with a low dose, and titrate the medication dose slowly.
2. Owing to reduced renal and hepatic function, the half-lives of many drugs are prolonged in the
elderly. Selection of agents should involve consideration of the specific pharmacokinetics of
each drug in the geriatric population.
3. Rapid dose escalations prevent attainment of the optimal therapeutic response because a steady-state
concentration of the drug is not reached and increases the risks of developing an ADR.
4. The fewest number of drugs should always be used to treat patients.
5. Always evaluate possible drug toxicity. Geriatric patients can have atypical presentations of
ADRs, which may manifest as CNS changes (e.g., altered mental status).
6. Review concomitant medications and diseases to evaluate the possible interactions with new
drugs.
7. Reassess the need for each medication on a regular basis.
Geriatrics 523
Drug Comments
Analgesics
Antidepressants
Antiemetics
Antihistamines
Antihypertensives
Antipsychotics
Haloperidol (Haldol) Avoid unless indicated for psychotic disorder; use in small
doses (1 mg); risk of sudden death in higher doses
Thioridazine (Mellaril) Avoid unless indicated for psychotic disorder
Antispasmodics
Drug Comments
Decongestants
H2 antagonists
Hypoglycemic agents
Muscle relaxants
Carisoprodol (Soma) Risk of adverse events greater than potential benefi ts; all use
should be avoided
Cyclobenzaprine (Flexeril) Risks of adverse events greater than potential benefits
Methocarbamol (Robaxin) Risks of adverse events greater than potential benefits
Orphenadrine (Norflex) Risks of adverse events greater than potential benefits
NSAIDs
Platelet inhibitors
Dipyridamole (Persantine) Avoid; lack of effi cacy and adverse effects (orthostatic
hypotension) at high doses; aspirin is safer and more
effective
Sedative hypnotics
CNS, central nervous system; NSAIDs, nonsteroidal anti-inflammatory drugs; OTC, over-the-counter; SIADH, syndrome of inappropriate
antidiuretic hormone secretion. Modified with permission from Fick DM, Cooper JW, Wade WE, et al. Updating the Beers criteria for potentially
inappropriate medication use in older adults. Arch Intern Med. 2003;163:2716–2724.
Geriatrics 525
Study
Questions 3. Which of the following antihypertensive agents
should be avoided in elderly patients?
Directions for questions 1–14: Each of the questions, (A) amlodipine (Norvasc®) 5 mg every day
statements, or incomplete statements in this section can (B) atenolol (Tenormin®) 25 mg every day
be correctly answered or completed by one of the (C) benazepril (Lotensin®) 10 mg every day
suggested answers or phrases. Choose the best answer. (D) hydrochlorothiazide (HydroDIURIL®) 25
mg every day
1. Which of the following medications due to its lack of
(E) methyldopa (Aldomet®) 250 mg three times a day
active metabolites and dependency on phase II
hepatic reactions is least likely to cause a fall in a 4. Which of the following benzodiazepines is expected to
geriatric patient? cause the least amount of adverse effects in the elderly?
(A) amitriptyline (Elavil®) (A) chlordiazepoxide (Librium®)
(B) haloperidol (Haldol®) (B) diazepam (Valium®)
(C) benztropine (Cogentin®) (C) f urazepaml (Dalmane®)
(D) diazepam (Valium®) (D) lorazepam (Ativan®)
(E) oxazepam (Serax®) (E) temazepam (Restoril®)
2. Which of the following drugs would be expected 5. Which of the following factors is associated with
to cause anticholinergic adverse effects in the an increased risk of noncompliance in the elderly?
elderly? (A) Polypharmacy
(A) diazepam (Valium®) (B) Hypertension
(B) ciprofloxacin (Cipro®) (C) Living with a spouse in an isolated environment
(C) tolterodine (Detrol®) (D) Expensive medications
(D) propranolol (Inderal®) (E) Good relationship with physician
(E) cimetidine (Tagamet®)
526 Chapter 28 7. Which of the following statements regarding
renal excretion in the geriatric patient is correct?
I. All renally eliminated drugs should be
Directions for questions 6-7: The questions and
monitored for the need for dose reductions in
incomplete statements in this section can be correctly
order to reduce potential toxicity.
answered or completed by one or more of the
II. Cockcroft–Gault formula provides a
suggested answers. Choose the answer, A–E. good estimation of creatinine clearance
A if I only is correct in most patient populations.
B if III only is correct III. Serum creatinine is a very sensitive indicator
C if I and II are correct of renal function in the elderly.
D if II and III are correct 8. Patient AM is a 60-year-old obese Caucasian male
E if I, II, and III are correct who is about to be started on several renally
6. Which of the following pharmacokinetic parameters eliminated drugs for various diseases he has been
is most likely to affect the manner in which a drug diagnosed with. Patient is 5 9 , weighs 220 lb, and
will affect a geriatric patient? has a serum creatinine of 2.0 mg/dL. What is Mr.
I. Drug absorption AM’s calculated creatinine clearance?
II. Phase I reactions within the (A) 56 mL/min
liver III. Drug distribution (B) 128 mL/min
(C) 47 mL/min
(D) 109 mL/min
(E) 100 mL/min
I. PREMENSTRUAL SYNDROME/PREMENSTRUAL
DYSPHORIC DISORDER
A. Definition. Premenstrual syndrome (PMS) occurs in many women from 2 to 14 days before the
onset of menstruation. Women with PMS can experience abdominal bloating, weight gain, appe-
tite changes, fatigue, tension, mastalgia, headaches, irritability, depressed or labile mood, anxiety,
or be oversensitive to environmental stimuli. Women with premenstrual dysphoric disorder
(PMDD) often have major depression, frequently have persistent anger, irritability, anhedonia, and
feel over-whelmed or out of control.
B. Epidemiology. PMS is quite common (up to 75%) and usually mild; however, 20% to 30% of
wom-en have clinically significant PMS that interferes with their normal routines. The more
debilitating PMDD is seen only in 3% to 8% of menstruating women. Symptoms typically cease at
or soon after the onset of menstruation.
C. Cause. T he exact cause of the conditions is unknown, but hormones likely play a role.
D. Treatment is multimodal, including lifestyle and dietary changes, cognitive behavioral therapy,
and pharmacological treatment.
1. Lifestyle/dietary changes. Regular exercise, good sleep hygiene, and reduced sodium intake
are often helpful.
2. Vitamin supplementation. Some recommendations include 600 mg calcium twice daily, 200
to 400 mg magnesium daily, pyridoxine 50 to 100 mg daily, or vitamin E 400IU daily to
decrease symptoms.
3. Analgesics. Agents such as nonsteroidal anti-inflammatory drugs, including mefenamic acid
(Ponstel) or acetaminophen for headaches, menstrual cramping, and breast tenderness.
4. Diuretics. Spironolactone (Aldactone) may be helpful for fluid weight gain.
5. Oral contraceptives. These may be helpful to reduce symptoms. Drospirenone-containing
con-traceptives (e.g., Yaz, Yasmin) may be most beneficial because of the similarity to
spironolactone. A shortened hormone-free period also should be considered.
6. Antidepressants (Table 29-1). Agents may be taken throughout the cycle or only during the lu-
teal phase (days 14 to 28). Relief of symptoms should occur in the first cycle. Meta-analyses
have demonstrated that intermittent dosing is less effective than continuous dosing.
528
Women’s Health 529
III. DRUG USE IN PREGNANT PATIENTS. Most women would like to avoid
pharmacologic therapy during pregnancy if at all possible. However, . 80% of women are exposed to
substances such as medication during their pregnancies. Preexisting conditions and other problems
occurring during the pregnancy may require continuation or initiation of drug therapy. In rare
circumstances, fetal therapy can be administered through the mother. It is important to understand
maternal pharmacokinetic changes, placental drug transfer, eventual disposition of the drug, and
limitations of the FDA classification system to safely treat pregnant women.
A. Fetal development. T he effects of drug therapy in pregnancy depend largely on the stage of fetal
development during which the exposure occurs. Pregnancies are normally dated from the first day of the
last menstrual cycle; however, when discussing fetal development, fertilization occurs on day 1.
1. Weeks 1 to 2. During the first days after fertilization, the zygote forms in the fallopian tube. Over
the next few days, division of the zygote eventually results in the formation of the blastocyst, which
530 Chapter 29 III. A
travels through the tube into the uterus. The blastocyst contains numerous types of relatively un-
differentiated tissues that will ultimately become the fetus, the placenta, and the fetal membranes.
Superficial implantation in the endometrium occurs within the first 5 days. During the second week,
differentiation begins and the placenta has started to form. During these weeks, there is an “all or
none” phenomenon. With no placenta to transfer substances to the blastocyst, there is no
susceptibility to teratogens. Exposure to environmental agents during this time will have either little
or no effect on the embryo or will destroy most cells, leading to pregnancy termination.
2. Weeks 3 to 8. It is during this time that the placenta becomes fully functional and organogenesis oc-curs.
This is the most critical period of development, when the embryo is most susceptible to terato-gens. All
major organ systems develop structurally during these weeks. All are completely formed by the end of the
ninth week, with the exception of the central nervous system (CNS). Major congenital anomalies, such as
cardiac abnormalities, spina bifida, and limb defects occur during this time.
3. Weeks 9 to 38 (the fetal period). At the ninth week, the embryo is referred to as a fetus.
Development during this time is primarily functional, with overall growth occurring
throughout. The fetus may be at risk during exposure to potentially fetotoxic drugs or viruses.
Exposure to a drug is generally not associated with major congenital malformations; however,
minor congenital anomalies and functional defects may occur during this time.
B. Placental transfer of drugs. T he placenta is the functional unit between the fetal and the maternal
blood supply. There is no mixing of the two systems, but exchange of nutrients, oxygen, and waste
products occurs primarily via passive diffusion. This process is driven by the concentration
gradient between the two systems. There are a few substances that are actively transported across
the placenta (e.g., amino acids); drugs that are structurally similar to these compounds will also be
transported by this mechanism.
1. Placental metabolism. T he placenta produces several pregnancy-related hormones that are
mainly secreted into the maternal circulation. Some of the other substances produced by the
pla-centa are enzymes that metabolize drugs. A common example of this is prednisone
metabolism, so that very little steroid reaches the fetus.
2. Factors affecting placental drug transfer. For a drug to cause a teratogenic or pharmacological
effect in the embryo or fetus, it must cross from the maternal circulation to the fetal circulation or
tissues. Generally, the principles that apply to drug transfer across any lipid membrane can be
applied to placental transfer of a drug. Most substances administered for therapeutic purposes have,
by design, the ability to cross the placenta to the fetus. The critical factor is whether the rate and
extent of transfer are sufficient to cause significant drug concentrations in the fetus. There are many
factors that affect the rate and extent of placental drug transfer.
a. Molecular weight. Low-molecular-weight drugs (, 500 Da) diffuse freely across the pla-
centa. Drugs of a higher molecular weight (500 to 1000 Da) cross less easily. Drugs
composed of very large molecules (e.g., heparin) do not cross the placental membranes.
b. Drug pKa. Weakly acidic and weakly basic drugs tend to rapidly diffuse across the
placental membranes. Ionized compounds do not cross the placenta.
c. Lipid solubility. Moderately lipid-soluble drugs easily diffuse across the placental mem-
branes. It is important to note that many drugs have been formulated for oral administration
and are designed for optimal lipid membrane transfer.
d. Drug absorption. During pregnancy, gastric tone and motility are decreased, which results
in delayed gastrointestinal emptying time. This typically does not affect drug absorption.
How-ever, nausea and vomiting, which are most common in the first trimester but may
continue throughout pregnancy, may affect absorption.
e. Drug distribution. T he volume of distribution increases significantly during pregnancy and
increases with advancing gestational age. The alteration in volume of distribution is the result of
an increased plasma volume. Total body fluid (intravascular and extravascular volume)
increases, as does adipose tissue. The placenta itself may also be a site for distribu-tion.
Hydrophilic drugs will have a higher volume of distribution leading to lower peak levels. Plasma
concentrations of drugs that are widely distributed are usually lower than those with a small
volume of distribution. Therefore, less drug is available to cross the placenta.
f. Plasma protein binding. Placental transfer of a highly plasma-protein-bound drug is less likely
because only the free drug crosses the placenta. During pregnancy, a reduction in the levels of
Women’s Health 531
two major drug-binding proteins—albumin and AGP—is observed. A dilutional effect occurs
with albumin and other protein concentrations, which can increase the free fraction of drugs.
g. Physical characteristics of the placenta. As a pregnancy progresses, the placental
membranes become progressively thinner, resulting in a decrease in diffusion distance. The
placenta also expands, causing a greater surface area for the transfer of substances.
h. Maternal pharmacokinetic changes. The dramatic increase in blood volume that occurs
pri-marily during the first 30 weeks of pregnancy enhances blood flow through the kidneys
and liver. Drugs that are excreted renally will experience a more rapid clearance, which
could decrease the overall exposure time of the drug to the placenta. Metabolism of some
drugs is increased, whereas others decrease; cytochrome P450 system enzymes 2D6, 2C9,
and 3A4 increase in activity, whereas 1A2 and 2C19 are inhibited.
i. Drug half-life. Drugs that remain in circulation longer have a higher likelihood of placental
passage due to the prolonged exposure.
3. Teratogenic drugs
a. Teratogens are defined as agents that increase the risk of or cause a congenital anomaly to
oc-cur. These defects can be structural, functional, or behavioral in nature. Women may
blame a specific exposure during their pregnancy as the cause of a fetal anomaly; however,
the defect may have no known cause, as is the case in 3% of all births in the United States.
b. It may take years of exposures to actually link a specific drug to certain defects. Animal studies
can only suggest potential problems in humans but are often the only source of information
regarding safety of agents during early pregnancy. The dose that animals often receive exceeds
the normal human dose, which diminishes the applicability of these data to humans.
c. The fetus is unable to metabolize or eliminate drugs as quickly as the mother. Some
substances may be excreted into the amniotic fluid and then resorbed in the fetal intestines
after the fluid is swallowed. Therefore, some drugs may have a longer exposure time in the
fetus, whereas others are eliminated more rapidly.
d. Because fetal organ systems develop at different times, specific teratogenic effects depend
mainly on the point of gestation when the drug was ingested.
e. The teratogenic rate of substances indicates how frequently anomalies occur and over what
exposure period. For example, one of the most potent known teratogens, thalidomide, had a
teratogenic rate of 20% with a single exposure, yet only one-third of women who ingested
the drug gave birth to affected infants. Other agents may increase the rate of specific defects
over the general population, but the absolute incidence may be extremely low.
f. The FDA developed a classification system that groups drugs according to the degree of
their potential risk during pregnancy.1
(1) Category A. Adequate, well-controlled studies in pregnant women have not shown an
increased risk of fetal abnormalities.
(2) Category B. Animal studies have revealed no evidence of harm to the fetus; however,
there are no adequate and well-controlled studies in pregnant women. Or animal stud-
ies have shown an adverse effect, but adequate and well-controlled studies in pregnant
women have failed to demonstrate a risk to the fetus.
(3) Category C. Animal studies have shown an adverse effect, and there are no adequate
and well-controlled studies in pregnant women. Or no animal studies have been
conducted, and there are no adequate and well-controlled studies in pregnant women.
(4) Category D. Studies—adequate, well-controlled, or observational—in pregnant
women have demonstrated a risk to the fetus. However, the benefits of therapy may
outweigh the potential risk.
(5) Category X. Studies—adequate, well-controlled, or observational—in animals or preg-
nant women have demonstrated positive evidence of fetal abnormalities. The use of the
product is contraindicated in women who are or may become pregnant.
g. Problems with the current system. The current system was created in 1979 and has not been
revised since its inception. With many agents there is a paucity of human data, despite the
1
Federal Register, June 26, 1979;44(124):37434–37467.
532 Chapter 29 III. B
fact the drug may carry a category B rating. Most newly marketed agents will be placed in
category C, amid other agents with little data in humans or animals. This is the most
difficult category to assess. Although there may be case reports of drug exposures, these
tend to bias information toward fetal risk; most publish only outcomes with potential drug-
related effects. Few drugs will ever be assigned a category A status because large,
randomized, well-controlled trials are rarely conducted in pregnant women.
h. Examples of teratogenic agents
(1) Vitamin A derivatives. Drugs such as isotretinoin (Accutane) and etretinate (Tegison)
are potent teratogens in humans. These agents should be discontinued several months
before pregnancy.
(2) Warfarin (Coumadin) is most teratogenic in the first trimester (weeks 6 to 9), but can
also cause malformations during the second and third trimesters as well. Early exposure
is associated with a pattern of defects known as fetal warfarin syndrome. These defects
can include hypoplasia of the nose and extremities, congenital heart disease, and
seizures. CNS abnormalities are increased with later use. Heparins may be an
appropriate substi-tute when anticoagulation is necessary; however, they are not as
effective for preventing thrombosis in women with artificial heart valves.
(3) Androgenic agents can cause virilization of female fetuses, creating ambiguous genitalia.
Finasteride (Propecia) can cause genital abnormalities in male offspring. Estrogen and
progestins, fortunately, do not have this effect. Many women continue to take birth con-trol
pills during the first month or two after conception until the pregnancy is discovered.
(4) Ethanol. Alcohol consumed in large amounts for prolonged periods during pregnancy
(. 4 to 5 drinks per day) is known to cause fetal alcohol syndrome (FAS). Features of
FAS include growth restriction, craniofacial dysmorphology, and CNS malfunctions,
along with various other abnormalities. At least 30% of women who abuse alcohol will
deliver an infant affected by FAS. Moderate alcohol consumption (2 drinks per day)
can also lead to similar defects, although usually not the complete syndrome. Even
though the most problematic time is during the first 2 months of pregnancy, moderate
drink-ing during the second trimester is associated with an increased rate of
spontaneous abortions.
(5) Antineoplastics. Many agents in this class are associated with fetal anomalies after
first trimester chemotherapy administration. Growth restriction often occurs regardless
of the timing of exposure. Owing to the mechanism of action of these agents, many are
embryocidal.
(6) Anticonvulsants. Drugs such as phenytoin (Dilantin), valproic acid (Depakene), and
carbamazepine (Tegretol) have all been associated with fetal anomalies. However, ma-
ternal benefit from these agents often outweighs the risk to the fetus. Anticonvulsants
should not be stopped during pregnancy, but if appropriate, they should be discontinued
several months before fertilization. Valproic acid and carbamazepine can increase the
risk of neural tube defects; women taking these agents should receive folic acid
supplementa-tion starting before conception. Toxic epoxide radicals are thought to be
the mechanism of teratogenicity with several of these agents. Genetic alterations in the
epoxide hydrolase enzyme activity can reduce or increase the severity of abnormalities.
(7) Infections. Viral infections, such as rubella, cytomegalovirus, parvovirus, coxsackie,
and varicella can be associated with growth restriction, congenital anomalies,
premature de-livery, and potential embryo toxicity or fetal demise. Nearly all maternal
infections have been thought to cause growth restriction.
(8) Cigarette smoking. Cigarettes contain many toxic and carcinogenic compounds in addi-
tion to nicotine. Nicotine is a potent vasoconstricting agent capable of reducing uterine
blood flow and increasing uterine vascular resistance. Smoking not only increases the risk
of a growth-restricted fetus but also increases the risk of spontaneous abortions, prema-ture
delivery, placental abruption, and premature rupture of the membranes. Small in-creases in
defects of the heart, limbs and feet, skull, urinary system, abdomen, intestines, and muscles
have also been associated with cigarettes. Smoking may also alter the effects of other
substances, perhaps enhancing toxicity of both agents.
Women’s Health 533
4. Other problematic therapies. Some agents given during pregnancy may result in
pharmacologi-cal effects that are not necessarily toxic, yet need to be considered when
medications are given during the later weeks of pregnancy.
a. CNS depression may occur with barbiturates, tranquilizers, antidepressants, and nar-cotics.
Also, anesthetics and other agents commonly given during labor may cause sig-nificant
CNS and respiratory depression in newborns (e.g., magnesium sulfate or opioid analgesics).
IV. DRUG EXCRETION IN BREAST MILK. Today, more than 60% of women choose to
breastfeed their infants. Of these women, 90% to 95% receive a medication during the first postpartum
week, most commonly for pain control after delivery. It is important to understand the principles of
drug excretion in breast milk and specific information on the various medications to minimize risks
from drug effects in the nursing infant.
A. Transfer of drugs from plasma to breast milk. Drug transfer into breast milk is governed by
many of the same principles that influence human placental drug transfer.
1. Most drugs cross into breast milk via passive diffusion along a concentration gradient formed
by the un-ionized drug content on each side of the membrane.
2. Breast milk contents change throughout a feeding. Colostrum, the very first milk produced, is
much higher in protein than mature milk and the fat content is minimal. Mature milk consists of
foremilk at the beginning of a feeding and hindmilk at the end. The protein and fat content
increase throughout the nursing session. Therefore drugs that partition into more lipid solutions
will have the highest concentration in hindmilk.
3. A milk to plasma ratio can be determined for specific agents when both blood and milk concen-
trations are known. Most drugs have a ratio , 1; lower numbers indicate that less drug crosses
into breast milk. Because the milk to plasma ratio may change within a feeding, the average
breast milk concentration is usually used, if available.
4. It is possible to calculate the dose an infant receives if the breast milk concentration is known. A typ-ical
infant drinks 150 mL/kg/day. Multiplying the average concentration by the breast milk volume consumed
will give the total daily exposure. It is important to remember that this drug must now be ingested by the
infant, so the bioavailability of the drug is critical to calculate the actual daily dose. Doses , 10% of the
maternal dose on a milligram per kilogram per day basis are preferable.
5. Some medications are not absorbed orally but may pass into breast milk when administered in-
travenously to the mother. Although the drug may not enter the infant’s blood, it may have
effects on the gastrointestinal tract. For example, the bioavailability of gentamicin is negligible;
however, it may cause diarrhea or sterilize the bowel.
B. Drug factors. T he drug and its environment influence the rate and extent of drug passage into the
breast milk.
1. Molecular weight. Drugs weighing , 200 Da cross into milk easily. Larger molecules can dis-
solve in the lipid membrane or pass through small pores. Large molecules, such as insulin, do
not cross into breast milk.
2. pH gradient. Human milk is more acidic than plasma.
a. Weak acids may diffuse across the membrane and remain un-ionized, allowing for passage back
into the plasma. Lower amounts of these drugs will cross than those that are weak bases.
b. Weak bases may diffuse into the breast milk and ionize, which causes drug trapping. This
cre-ates higher levels of drug in the breast milk; these drugs will have a milk to plasma ratio
. 1. This effect though is not usually clinically significant, especially when the maternal
serum concentration is very low.
534 Chapter 29 IV. B
3. Drug pKa. Only the un-ionized form of a drug is able to pass through the lipid membrane.
4. Plasma protein binding. The free fraction of a drug is available to pass into the breast milk. In
general, drugs with high plasma-protein-binding properties tend to remain in the plasma and
pass into the milk in low concentrations. Milk proteins and lipids also may bind drugs when
they are created in the mammary glands; this may represent another route of entry, rather than
passive diffusion.
5. Lipid solubility. Lipid solubility is necessary for a drug to pass into the breast milk. Highly
lipid-soluble drugs (e.g., diazepam [Valium]) may pass into the breast milk in relatively high
amounts and, therefore, may present a significant dose of drug to the nursing infant.
6. Equilibration. Some drugs may rapidly equilibrate between maternal plasma and breast milk. These
agents will diffuse across the membrane as the drug concentration changes in the maternal system.
Other agents may never reach equilibrium between milk and plasma. These drugs tend to slowly
diffuse into breast milk and will respond gradually to changes in maternal concentrations.
C. Maternal factors. Maternal pharmacology plays a significant role in the rate and extent of drug
passage into breast milk. The extent of plasma protein binding and changes in the mother’s ability
to metabolize or eliminate the drug influence the amount of drug that is available to pass into the
breast milk. Equally important are the maternal dose of the drug, the dosing schedule or frequency,
and the route of administration.
D. Drugs affecting hormonal influence of breast milk production. T he primary hormone
responsible for controlling breast milk production is prolactin. A decrease in milk production may
result in di-minished weight gain in the nursing infant, the need for supplementation, or premature
cessation of breastfeeding.
1. Drugs that decrease serum prolactin levels. Drugs such as bromocriptine have been used to
suppress lactation in women who choose not to breastfeed. This practice has long been aban-
doned because myocardial infarctions, seizures, and stroke were attributed to its use. Other
drugs include
a. ergot alkaloids
b. l-dopa
2. Drugs that increase serum prolactin levels. Metoclopramide (Reglan) has been useful
therapeu-tically to enhance milk production. The following drugs are known to increase serum
prolactin levels, but they are not used for this purpose. These drugs include
a. methyldopa (Aldomet)
b. haloperidol (Haldol)
c. phenothiazines
E. Factors to assess. In assessing the safety of an agent during breastfeeding, several considerations
should be addressed.
1. Inherent toxicity of the drug
2. Drug safety data in infants
3. Amount of drug ingested
4. Duration of therapy
5. Age of the infant or degree of prematurity
6. Drug pharmacokinetics in the mother and child
F. Factors to minimize drug exposure to the infant. One of the goals when using medications in
the breastfeeding mother is to maintain a natural, uninterrupted pattern of nursing. In many
instances, it may be possible to withhold a drug when it is not essential or delay therapy until after
weaning. Other factors include
1. Medication selection. When a specific product is being selected, it is important to choose the
agent that is distributed into the milk the least, if possible.
a. Other desirable characteristics include a short half-life, inactive metabolites, and high
protein binding.
b. In addition, it is desirable to select agents with lower plasma concentrations, which may in-
volve an alternative route of administration.
c. Single doses may be preferable to a longer therapy course if the agent is contraindicated in
breastfeeding. This can allow for the mother to pump and discard her milk for a defined
time, often 12 to 24 hrs, rather than discontinue breastfeeding altogether.
Women’s Health 535
2. Maternal dose relative to infant feeding. One of the goals of drug dosing in lactating women
is minimal infant exposure to the drug. It is desirable to adjust the dosing and nursing schedules
so that a drug dose is administered at the time of or immediately after the infant’s feeding.
Medica-tions should be dosed before the infant’s longest sleep.
G. Examples of drugs that readily enter breast milk. T hese agents should be used with caution in
nursing mothers.
1. Narcotics, barbiturates, and benzodiazepines. CNS active agents, such as diazepam, may
have a hypnotic effect on the nursing infant. These effects are related to the maternal dose.
Alcohol consumption may have a similar effect.
2. Antidepressants and antipsychotics. These classes of drugs appear to pass into the breast
milk; however, no serious adverse effects have been reported. The long-term behavioral effects
of chronic exposure to these drugs on developing newborns are unknown.
3. Anticholinergic compounds. These drugs may result in adverse CNS effects in the infant and
may reduce milk volume in the mother. Dicyclomine (Bentyl) is contraindicated in nursing
mothers because it may result in neonatal apnea.
V. INCONTINENCE
A. Definition. Incontinence is defined as an inability to control urine or feces elimination. There are
four major types of urinary incontinence: stress, urge (overactive bladder), overflow, and mixed
(stress and urge incontinence).
B. Epidemiology. T he reported incidence of urinary incontinence varies widely, ranging from 10%
to 35% in women. Incontinence becomes more common as women age but is not a normal part of
aging.
C. Cause. Gender, age, hormonal status, birthing trauma, and genetic differences in connective tissue
all contribute to the development of incontinence.
D. Diagnosis is made with a voiding diary (frequency of urination, leakage of urine, symptoms during
urination such as pain or discomfort, or the need to strain or splint to urinate), a physical examina-
tion, surgical and obstetric history, medication history, mental status examination in the elderly,
and a urinalysis. Special diagnostic procedures, such as a cystourethrogram, can be performed if
the ini-tial diagnosis and treatment is ineffective.
E. Stress urinary incontinence (SUI) involves the involuntary loss of urine during physical activity that
increases the intra-abdominal pressure (e.g., coughing, sneezing). This is usually caused by urethral
underactivity, decreased tone of the urethral sphincter, or hypersensitivity of the bladder neck.
1. Treatment of SUI usually involves
a. Behavioral therapy. Bladder retraining involves voiding on a schedule, usually hourly to
start. Once this is managed without leakage, the time between voids is increased by 15-min
inter-vals, until the elapsed time between voids is between 2 and 4 hrs. Pelvic floor muscle
strength-ening, known as Kegel exercises, can reduce incontinence by up to 50%.
b. Weight loss of 5% to 10% can dramatically improve symptoms especially in the
postpartum period when stress incontinence is a common problem.
c. Fluid restriction to less than 2L daily.
d. Drug therapy
(1) Vaginal estrogen improves tone and blood supply of the urethral sphincter muscles
increasing urethral closure pressure and increasing mucosal thickness, thus improving
functioning.
(2) Alpha-adrenergic agents induce muscle contraction in the bladder neck and urethra,
increasing outlet resistance. Pseudoephedrine (Sudafed) is the remaining agent in this
class.
(3) Duloxetine (Cymbalta) which has a direct effect on the urethra (enhancing closure) via
peripheral adrenergic activation (40 to 80 mg daily).
(4) Imipramine (Tofranil), a tricyclic antidepressant, decreases bladder contractility and
in-creases outlet resistance. Doses of 75 mg once daily and 25 mg three times daily
have been successful in studies.
e. Surgical options. Colposuspension elevates the bladder neck and the urethra, restoring the
urethrovesical junction; sling procedures elevate the urethra and increase urethral
536 Chapter 29 V. E
compression; or tension-free tape reduces urethral mobility or produces a kink in the urethra
during increases in intra-abdominal pressure.
F. Urge incontinence is described by the sudden sense of needing to urinate followed by the loss of
urine. This involuntary contraction of the bladder or bladder overactivity is known as detrusor in-
stability. The detrusor is the muscle located in the bladder wall; relaxation allows bladder to
expand. Another common symptom is frequent urination during the day and night.
1. Treatment of urge incontinence involves
a. Behavioral therapy. Bladder retraining entails initially voiding every 1 to 1.5 hrs even if
leak-age or the urge to void occurs before this time, eventually increasing the interval by 30
mins until 3 to 4 hrs elapses between voids. Also, avoiding foods that may irritate the
bladder such as caffeine, carbonation, acidic foods, and spicy foods is helpful.
b. Drug therapy
(1) Anticholinergic agents act on muscarinic receptors to decrease contractility. However,
these agents have the potential to cause significant side effects, especially in the
elderly. Extended-release formulations usually have lower anticholinergic effects.
(a) oxybutynin (Ditropan, Ditropan XL, Oxytrol), 2.5 mg to 5.0 mg two to four times
daily, 5 mg to 30 mg daily extended-release, 3.9 mg patch twice weekly.
(b) tolterodine (Detrol, Detrol LA), 2 mg twice daily or 4 mg daily extended-release.
The dose should be decreased by 50% in women taking CYP3A4 inhibitors.
(c) trospium (Sanctura), 20 mg twice daily, adjusted to 20 mg daily in renal
impairment or elderly women (. 75 years).
(d) darifenacin (Enablex), 7.5 to 15 mg daily, women with moderate hepatic impair-
ment or taking CYP3A4 inhibitors should not receive more than 7.5 mg daily.
(e) solifenacin (VESIcare), 5 to 10 mg daily with 5 mg daily recommended in women
with renal impairment, moderate hepatic impairment, or in those taking CYP3A4
inhibitors.
(2) Flavoxate (Urispas) is a urinary antispasmodic that can be helpful. Doses range from
100 to 200 mg three to four times daily.
c. Surgery. Augmentation cystoplasty uses intestinal segments to improve the storage
function of the bladder.
G. Overflow incontinence is a constant loss of urine or inadequate bladder emptying. This type of in-
continence is not common in women. Women with neurological impairment who may not feel the
urge to void are predisposed to this type of incontinence because of bladder overdistention. A post-
void residual, measuring the urine volume remaining in the bladder after urination, can make this
diagnosis. A blockage in the urethra can also cause overflow incontinence.
1. Treatment
a. Behavioral therapy. Voiding or self-catheterization should occur at specific intervals to
pre-vent overdistention. In severe cases of neurological impairment (such as significant
demen-tia), the patient may require incontinence pads or long-term catheterization.
b. Drug therapy. Tamsulosin (Flomax) may be helpful by relaxing striated and smooth
muscle, decreasing urethral resistance, and relieving symptoms. Initial doses should be 0.4
mg daily with a maximum of 0.8 mg daily. It is important to note that some women
receiving alpha-adrenergic blocking agents for hypertension develop incontinence.
c. Surgery. Surgery may be required if a urethral blockage is the cause of incontinence.
VI. MENOPAUSE
A. Definition. Menopause is defined as the irreversible cessation of the reproductive cycle and
menses after the ovaries permanently fail to respond to gonadotropins. Removal of the ovaries
(oophorectomy) in premenopausal women and premature ovarian failure also will lead to
menopausal symptoms. Menopause occurs naturally around age 51, but often occurs earlier in
smokers due to increased estrogen metabolism in these women.
B. Symptoms. Symptoms of menopause are due to the lack of estrogen. They include vasomotor
insta-bility (hot flushes), atrophic changes of the vagina and urethra/bladder (symptoms of burning,
itch-ing, bleeding, dyspareunia/painful intercourse, or incontinence), and a menopausal syndrome
(sleep disturbances and mood swings).
Women’s Health 537
PO, by mouth; TID, three times daily; BID, twice daily; TD, transdermal; XL, extended release.
C. Treatment
1. Lifestyle changes. Recommendations include lowering the core body temperature by dressing
in layers, consuming cold food and beverages, and using cooling techniques, such as a fan or
cold towels placed on the back of the neck.
2. Hormone therapy. Hormone therapy has become controversial in recent years but can be helpful in
very symptomatic women. The lowest dose for the shortest duration is the current recommendation.
Estrogen decreases the risk of colon cancer and decreases the risk of noninsulin dependent diabetes.
However, combined hormone therapy can increase the risk for breast cancer, whereas estrogen alone
does not appear to have this effect. Estrogen also increases cardiovascular risks and thromboses.
3. Nonhormonal therapy. Many other therapies have been studied for the treatment of
vasomotor symptoms (Table 29-2).
4. Herbal therapies/phytoestrogens. Many therapies have been studied, but few agents have
been shown to be more effective than placebo.
a. Black cohosh possibly decreases hot flashes, but therapy should be limited to 6 months or
less. Black cohosh can increase the density of breast tissue, making mammograms more
difficult to interpret.
b. Red clover has a small decrease in frequency of hot flushes over placebo.
c. Soy has not been shown to be beneficial in many studies.
d. Other herbs. Other herbs studied or recommended by some for hot flashes include ginseng,
dong quai, damiana, licorice, motherwort, fennel, and evening primrose oil.
VII. OSTEOPOROSIS
A. Definition. Osteoporosis can be defined as an imbalance of osteoblast and osteoclast activity.
Osteo-blasts form bone, whereas osteoclasts break down bone (resorption). Receptor activator of
nuclear factor kappa-B (RANK) and osteoprotegerin (OPG) are proteins that are intimately
involved with bone remodeling. RANK is key for the differentiation and activation of osteoclasts.
OPG has the opposite effect, blocking the effects of RANK by binding to the RANK ligand. The
ultimate result of OPG is to decrease the activity of osteoclasts, thus preventing bone resorption.
B. Epidemiology. In 2009, approximately 13% of the American population was . 65 years of age,
rep-resenting nearly 38 million Americans. The risk of a fracture doubles every 7 to 8 years in
women older than the age of 50. Nonmodifiable risk factors for osteoporosis include female sex,
age, low body mass index, small frame, family history of osteoporosis, menopausal status, and
ethnicity/race. In an observational study, osteoporosis was diagnosed in different ethnicities.
Women with highest risk to lowest rate of osteoporosis were Native Americans . Asians .
Hispanics . Caucasians . African Americans. Risk factors that can be modified include alcohol
intake . 3 servings per day, low activity level, smoking, and estrogen deficiency.
C. Diagnosis. Routine health care should include measuring height; vertebral fractures will shorten stature
and having one vertebral fracture increases the risk for more. Screening for osteoporosis should be done
in women 60 to 64 years with one major risk factor and all women . 65 years, women with a low trauma
fracture, women who have lost . 0.5 inch of height within 1 year or . 1.5 inches
538 Chapter 29 VII. C
overall, and to monitor therapy. The World Health Organization developed the Fracture Risk As-
sessment tool (FRAX) to evaluate a woman’s risk of having a hip or any major osteoporotic fracture
over the next 10 years. It takes into account family history and many risk factors. It is only indicated in
women . 50 years who have not started any drug therapy for osteoporosis. Peripheral bone min-eral
density scans can be done at community health screenings using heel ultrasonography on the
nondominant foot. These screenings can help identify women who need further screening with a dual
energy x-ray absorptiometry (DXA) scan. The DXA scan is the gold standard and bone mineral density
(BMD) is measured at the hip and spine. The results generate a T score; a score of 21 to 22.5 indicates
low bone mass/osteopenia and a score # 22.5 makes the diagnosis of osteoporosis. A DXA scan should
not be performed more frequently than every 1 to 2 years.
D. Therapy
1. Lifestyle changes are important to increase bone mass. Adolescent and young women need to build
bone early in life. A healthy lifestyle incorporates weight bearing exercises such as walking and
isometric exercises such as weight lifting, not smoking, and limiting alcohol and caffeine intake.
Sunlight converts vitamin D into an active form in the skin, but adequate exposure is dif-ficult for
most women to achieve. Using suntan lotion negates the effect of sunlight and only more equatorial
areas of the world have enough direct sunlight during the winter months.
2. Calcium and vitamin D. Adequate calcium ingestion may not occur with dietary intake and
supplementation is often recommended. Vitamin D has been shown in studies to decrease the
number of falls, thus reducing the number of fractures. The recommended intake of calcium
according to the National Osteoporosis Foundation (NOF) is 1200 mg with 800 to 1000
interna-tional units of vitamin D for women older than age 50. Calcium therapy taken without
vitamin D might increase the risk of myocardial infarctions.
3. Bisphosphonates are selected as first line in many women (Table 29-3). They are effective in re-
ducing fractures in the hip and spine, working to decrease osteoclast activity. Oral dosing is com-
plicated by their poor absorption when taken within 1 hr of a meal or beverage. They also require the
woman to remain upright after dosing to prevent esophageal irritation and erosion. They can cause
unusual side effects, such as jaw necrosis and atypical fractures of the femur. They are gener-ally
contraindicated in women with a creatinine clearance less than 30 mL/min. The duration of therapy
is not completely established, although 5 years is recommended by some experts.
4. Hormone therapy. Estrogens inhibit bone resorption and promote bone formation. They
reduce fractures in the hip, spine, and nonvertebral locations. Bone mass is maintained with
conjugated equine estrogen doses of 0.3 mg daily. The higher the estrogen dose, the higher the
increase in BMD; however, the lowest dose of estrogen should be used in naturally
postmenopausal women to control vasomotor symptoms.
5. Selective estrogen receptor modulators (SERMs). Raloxifene (Evista) increases BMD spine and
hip and reduces vertebral fractures. Raloxifene is helpful in women also needing protection against
breast cancer, but the agent can cause menopausal symptoms in women due to its estrogen antago-
nism in selective tissues. The dose for prevention and treatment of osteoporosis is 60 mg daily.
H, hip fracture; V, vertebral (spine) fracture; NV, nonvertebral fracture; PO, by mouth; IV, intravenous.
Women’s Health 539
a
Estrogen 34% 35% 5%
Raloxifene 40%
Bisphosphonates 40% 60% 3.3%–4.8%
Calcitonin 33% 1%
Teriparatide 35% 65% 35% 5%
Denosumab 40% 68% 20% 5%
Study
Questions
Directions for questions 1–8: Each of the questions,
statements, or incomplete statements in these sections can
be correctly answered or completed by one of the
suggested answers or phrases. Choose the best answer.
1. A 23-year-old woman was diagnosed with
significant PMS in her teens, now presents with
persistent irritation with her significant other
resulting in frequent arguments. Possible therapies
for her include all of the following except
(A) sertraline 100 mg days 14 to 28.
(B) paroxetine 30 mg daily.
(C) venlafaxine XR 75 mg daily.
(D) clonazepam 1 mg twice daily.
2. T he best therapy for an obese woman with PCOS to
improve menstrual regularity will be
(A) spironolactone.
(B) metformin.
(C) weight loss.
(D) pioglitazone.
3. Which of the following therapies enhances osteoblast
activity?
(A) Calcium and vitamin D
(B) Calcitonin
(C) Denosumab
(D) Teriparatide
(E) Risedronate
540 Chapter 29 Directions for questions 9–12: The questions and
incomplete statements in this section can be correctly
answered or completed by one or more of the
4. Which of the following medications is safe to use suggested answers. Choose the answer, A–E.
in the third trimester of pregnancy? (A) if I only is correct
(A) Acetaminophen (B) if III only is correct
(B) Nonsteroidal anti-inflammatory drugs (C) if I and II are correct
(C) Warfarin (D) if II and III are correct
(D) OxyContin (E) if I, II, and III are correct
(E) Aspirin 9. T he definition of polycystic ovarian syndrome includes
5. Placental transfer of a drug is affected by all of I. hyperandrogenism
the following characteristics except II. diabetes
(A) molecular weight. III. obesity
(B) fetal gender. 10. T herapy for stress urinary incontinence can include
(C) gestational age.
I. oxybutynin
(D) lipid solubility of the drug.
(E) plasma protein binding. II. tamsulosin
III. imipramine
6. When selecting a benzodiazepine product for a
woman who has chronic panic disorder, all of the 11. Behavioral therapy and bladder retraining is
following drug properties are desirable for most helpful in which types of incontinence?
breastfeeding her 8-month-old infant who was born at I. stress incontinence
term except II. urge incontinence
(A) hepatic metabolism to inactive metabolites. III. overflow incontinence
(B) a short half-life. 12. According to the principles of drug excretion into
(C) a rapid onset of action. the breast milk, which combination of the
(D) high lipid solubility. following properties would result in the highest
7. Drug safety in pregnancy of a specific agent can drug concentration in breast milk?
be assessed best by I. low molecular weight, moderately lipophilic
(A) the FDA classification system, II. low plasma protein bound, weakly basic
especially category C drugs. III. highly plasma protein bound, weakly acidic
(B) case reports. 13. Following principles of teratogenicity, drug
(C) physician knowledge. exposure during which of the following times could
(D) databases such as REPROTOX. cause fetal abnormalities?
8. T he primary difference between PMS and PMDD is I. f rsti 2 weeks of gestation
(A) PMS has an earlier onset of symptoms. II. weeks 3 to 8 of
(B) PMS is more debilitating. gestation III. the fetal period
(C) PMDD can be treated with antidepressants.
(D) PMDD is more common.
I. GENERAL PRINCIPLES
A. Laboratory tests are performed for multiple purposes, including to discover a disease, confirm or
differentiate a diagnosis, stage or classify a disease, and monitor effectiveness of therapy.
B. Laboratory tests are classified as screening or diagnostic. Screening tests are used in patients with no
signs or symptoms of a disease (e.g., serum cholesterol for assessing cardiovascular disease risk). Diag-
nostic tests are done in patients with signs and symptoms of disease or with an abnormal screening test.
C. Monitoring drug therapy
1. Laboratory test results are used to investigate potential problems with a patient’s anatomy or
physiology. Pharmacists usually monitor laboratory tests to
a. Assess the therapeutic and adverse effects of a drug (e.g., monitoring the serum uric acid
level after allopurinol is administered, checking for increased liver function test values after
administration of isoniazid)
b. Determine the proper drug dose (e.g., assessment of the serum creatinine or creatinine
clearance value before use of a renally excreted drug)
c. Assess the need for additional or alternate drug therapy (e.g., assessment of white blood
cell count after an antibiotic is administered)
d. Prevent test misinterpretation resulting from drug interference (e.g., determination of a
false-positive result for a urine glucose test after cephalosporin administration)
2. These tests can be expensive, and requests for them must be balanced against potential benefits for
patients and how the laboratory test will affect your decision regarding therapy. Generally, lab tests
should be ordered only if the results will affect the decisions about the management of the patient.
D. Definition of normal values
1. Normal laboratory test results fall within a predetermined range of values, and abnormal
values fall outside that range. The normal range of a laboratory test is usually determined by
applying statistical methods to results from a representative sample of the general population.
Usually, the mean 2 standard deviations is taken as the normal range.
a. Normal limits may be defined somewhat arbitrarily; thus, values outside the normal
range may not necessarily indicate disease or the need for treatment (e.g., asymptomatic
hyperuricemia).
b. Many factors (e.g., age, sex, time since last meal) must be taken into account when
evaluating test results.
c. Normal values also vary among institutions and may depend on the method used to
perform the test.
d. The goal is not to make all laboratory values normal; resist urges to do something in a
clinically stable patient.
e. Attempts have been made in recent years to standardize the presentation of laboratory data by
using the International System of Units (SI units). Controversy surrounds this issue in the United
States, and resistance to adopt this system continues. The SI unit of measure is a method of
reporting clinical laboratory data in a standard metric format. The basic unit of mass for the SI is
the mole. The mole is not influenced by the addition of excess weight of salt or ester
formulations. Technically and pharmacologically, the mole is more meaningful than
the gram because each physiological reaction occurs on a molecular level.
542
Clinical Laboratory Tests 543
Efforts to implement the SI system began in the 1970s, resulting in the adoption of full SI-
transition policies by a few major medical and pharmaceutical journals in the 1980s.
Reluctance to use this system by many clinicians in the United States has forced changes in
the policies by some journals to report both conventional and SI units or to report the
conver-sion factor between the two systems. It is still controversial which method should be
used to report clinical laboratory values. There are arguments for and against the universal
conversion to the SI system. Readers should be aware that some journals report SI and/or
conventional units in their text. Particular attention should be paid to the units associated
with a reported laboratory value, and access to a conversion table may be necessary to avoid
confusion in the interpretation of the data. When appropriate, both conventional and SI units
will be reported in this chapter.
2. Laboratory error must always be considered when test results do not correlate with
expected results for a given patient. If necessary, the test should be repeated. Common
sources of laboratory error include spoiled specimens, incomplete specimens, specimens taken
at the wrong time, faulty reagents, technical errors, incorrect procedures, and failure to take diet
or medication into account.
3. During hospital admission or routine physical examination, a battery of tests is usually given
to augment the history and physical examination. Basic tests may include an electrocardio-
gram (ECG), a chest x-ray, a sequential multiple analyzer (SMA) profile, electrolyte tests
(e.g., Chemistry or Basic Metabolic Panel [BMP]), a complete blood count (CBC), and
urinalysis.
E. Quantitative tests, qualitative tests, and analytical performance
1. Tests with normal values reported in ranges (i.e., 3.5 to 5.0 mEq/L) are called quantitative.
2. Tests with positive ( ) or negative ( ) outcomes are called qualitative.
3. Those with varying degrees of positivity (e.g., 1 , 2 , 3 glucose in the urine) are termed
semiquantitative.
4. The quality of a quantitative assay is measured in terms of accuracy (accuracy is defined as the
extent to which the mean measurement is close to the true value). Precision refers to the repro-
ducibility of the assay.
II. HEMATOLOGICAL TESTS. Blood contains three types of formed elements: red blood
cells (RBCs), white blood cells (WBCs), and platelets (Figure 30-1). A CBC typically includes RBC
count, total WBC count, hemoglobin (Hb), hematocrit (Hct), RBC indices (mean cell volume [MCV],
mean cell Hb [MCH], mean cell Hb concentration [MCHC]), reticulocyte count, and platelet count.
Figure 30-1. Derivation of blood elements from stem cells. Cells located below the horizontal line are found in
normal peripheral blood, with the exception of the late normoblasts.
544 Chapter 30 II. A
A. RBCs (erythrocytes)
1. The RBC count, which reports the number of RBCs found in a given volume of blood, provides an
indirect estimate of the blood’s Hb content. Values are often reported in cells/microliter ( L) or
cells/liter and less commonly as cells/cubic millimeter (mm3). Normal values are
a. 4.3 to 5.9 1012 cells/L of blood for men
b. 3.5 to 5.0 1012 cells/L of blood for women
2. The Hct or packed cell volume (PCV) measures the percentage by volume of packed RBCs in
a whole blood sample after centrifugation. The Hct value is usually three times the Hb value
(see II.A.3) and is given as a percent or fraction of 1 (42% to 52% or 0.42 to 0.52 for men; 37%
to 47% or 0.37 to 0.47 for women).
a. Low Hct values indicate such conditions as anemia, overhydration, or blood loss.
b. High Hct values indicate such conditions as polycythemia vera or dehydration.
3. The Hb test measures the grams of Hb contained in 100 mL (1 dL) or 1 L of whole blood and
provides an estimate of the oxygen-carrying capacity of the RBCs. The Hb value depends on
the number of RBCs and the amount of Hb in each RBC.
a. Normal values are 14 to 18 g/dL for men and 12 to 16 g/dL for women.
b. Low Hb values indicate anemia.
4. RBC indices provide important information regarding RBC size, Hb concentration, and Hb
weight. They are used primarily to categorize anemias, although they may be affected by
average cell measurements. A peripheral blood smear can provide most of the information
obtained through RBC indices. Observations of a smear may show variation in RBC shape
(poikilocytosis), as might occur in sickle-cell anemia, or it may show a variation in RBC size (
anisocytosis), as might occur in a mixed anemia (folic acid and iron deficiency).
a. MCV is the ratio of the Hct to the RBC count. It essentially assesses average RBC size and
reflects any anisocytosis.
Hct (%) 10
MCV __
RBC (millions)
(1) Low MCV indicates microcytic (undersize) RBCs, as occurs in iron deficiency.
(2) High MCV indicates macrocytic (oversize) RBCs, as occurs in a vitamin B12 or folic
acid deficiency.
(3) Normal range for MCV is 90 10.
b. Mean cell hemoglobin (MCH) assesses the amount of Hb in an average RBC.
(1) MCH is defined as:
MCH __Hb10
RBC (millions)
(2) Normal range for MCH is 30 4.
c. Mean cell hemoglobin concentration (MCHC) represents the average concentration of Hb
in an average RBC, defined as:
MCHC _Hb100
Hct
(1) Normal range for MCHC is 34 3.
(2) Low MCHC indicates hypochromia (pale RBCs resulting from decreased Hb
content), as occurs in iron deficiency.
d. Red blood cell distribution width (RDW) is a relatively new index of RBCs. Normally,
most RBCs are approximately equal in size, so that only one bell-shaped histogram peak is
generated. Disease may change the size of some RBCs—for example, the gradual change in
size of newly produced RBCs in folic acid or iron deficiency. The difference in size
between the abnormal and the less abnormal RBCs produces either more than one
histogram peak or a broadening of the normal peak. This value is used primarily with other
tests to diagnose iron-deficiency anemia.
(1) An increased RDW is found in factor deficiency anemia (e.g., iron, folate, vitamin B 12).
(2) A normal RDW is found in such conditions as anemia or chronic disease.
(3) The RDW index is never decreased.
Clinical Laboratory Tests 545
5. The reticulocyte count provides a measure of immature RBCs (reticulocytes), which contain
remnants of nuclear material (reticulum). Normal RBCs circulate in the blood for about 1 to 2
days in this form. Hence, this test provides an index of bone marrow production of
matureRBCs.
a. Reticulocytes normally make up 0.1% to 2.4% of the total RBC count.
b. Increased reticulocyte count occurs with such conditions as hemolytic anemia, acute
blood loss, and response to the treatment of a factor deficiency (e.g., an iron, vitamin B 12,
or folate deficiency). Polychromasia (the tendency to stain with acidic or basic dyes) noted
on a peripheral smear laboratory report usually indicates increased reticulocytes.
c. Decreased reticulocyte count occurs with such conditions as drug-induced aplastic anemia.
6. The erythrocyte sedimentation rate (ESR) measures the rate of RBC settling of whole,
uncoagu-lated blood over time, and it primarily reflects plasma composition. Most of the
sedimentation effect results from alterations in plasma proteins.
a. Normal ESR rates range from 0 to 20 mm/hr for males and from 0 to 30 mm/hr for females.
b. ESR values increase with acute or chronic infection, tissue necrosis or infarction, well-
established malignancy, and rheumatoid collagen diseases.
c. ESR values are used to
(1) Follow the clinical course of a disease
(2) Demonstrate the presence of occult organic disease
(3) Differentiate conditions with similar symptomatology—for example, angina pectoris (no
change in ESR value) as opposed to a myocardial infarction (increase in ESR value)
B. WBCs (leukocytes)
1. The WBC count reports the number of leukocytes in a given volume of whole blood.
a. Normal values range from 4,000 to 11,000 103 cells/mm3 (or 109 cells/L)
b. Increased WBC count (leukocytosis) usually signals infection; it may also result from
leukemia, tissue necrosis, or administration of corticosteroids. It is most often found with
bacterial infection.
c. Decreased WBC count (leukopenia) indicates bone marrow depression, which may result
from metastatic carcinoma, lymphoma, or toxic reactions to substances such as
antineoplastic agents.
2. The WBC differential evaluates the distribution and morphology of the five major types of
WBCs: the granulocytes (neutrophils, basophils, eosinophils) and the nongranulocytes
(lymphocytes, monocytes). A certain percentage of each type makes up the total WBC count
(Table 30-1).
a. Neutrophils may be mature or immature. Mature neutrophils are polymorphonuclear
leukocytes (PMNs), also referred to as polys; segmented neutrophils, or segs; immature
neu-trophils are referred to as bands or stabs.
(1) Chemotaxis. Neutrophils that phagocytize and degrade many types of particles
serve as the body’s first line of defense when tissue is damaged or foreign material
gains entry. They congregate at sites in response to a specific stimulus, through a
process known as chemotaxis.
Table 30-2 EXAMPLES OF CHANGES IN TOTAL WHITE BLOOD CELL (WBC) COUNT
AND WBC DIFFERENTIAL IN RESPONSE TO BACTERIAL INFECTION
WBC Count
(2) Lymphopenia, a decreased number of lymphocytes, may result from severe debili-
tating illness, immunodeficiency, or from AIDS, which has a propensity to attack TH4
cells.
(3) Atypical lymphocytes (i.e., T lymphocytes in a state of immune activation) are
classically associated with infectious mononucleosis.
e. Monocytes are phagocytic cells. Monocytosis, an increased number of monocytes, may
occur with tuberculosis (TB), subacute bacterial endocarditis, and during the recovery
phase of some acute infections.
C. Platelets (thrombocytes). T hese are the smallest formed elements in the blood, and they are
involved in blood clotting and vital to the formation of a hemostatic plug after vascular injury.
1. Normal values for a platelet count are 150,000 to 300,000/mm3 (1.5 to 3.0 1011/L).
2. Thrombocytopenia, a decreased platelet count, can occur with a variety of conditions, such as
idiopathic thrombocytopenic purpura or, occasionally, from such drugs as quinidine and
sulfonamides.
a. Thrombocytopenia is moderate when the platelet count is 100,000/mm3.
b. Thrombocytopenia is severe when the platelet count is 50,000/mm3.
III. COMMON SERUM ENZYME TESTS. Small amounts of enzymes (catalysts) circulate
in the blood at all times and are released into the blood in larger quantities when tissue damage occurs.
Thus, serum enzyme levels can be used to aid in the diagnosis of certain diseases.
A. Creatine kinase (CK)
1. Creatine kinase—formerly known as creatine phosphokinase (CPK)—is found primarily in
heart muscle, skeletal muscle, and brain tissue.
2. CK levels are used primarily to aid in the diagnosis of acute myocardial (Figure 30-2) or
skeletal muscle damage. However, vigorous exercise, a fall, or deep intramuscular injections
can cause significant increases in CK levels.
3. The isoenzymes of CK—CK-MM, found in skeletal muscle; CK-BB, found in brain tissue;
and CK-MB, found in heart muscle—can be used to differentiate the source of damage.
a. Normally, serum CK levels are virtually all the CK-MM isoenzyme.
b. Increase in CK-MB levels provides a sensitive indicator of myocardial necrosis.
B. Lactate dehydrogenase (LDH)
1. LDH catalyzes the interconversion of lactate and pyruvate and represents a group of enzymes
present in almost all metabolizing cells.
4. An increase in serum bilirubin results in jaundice from bilirubin deposition in the tissues.
There are three major causes of increased serum bilirubin.
a. Hemolysis increases total bilirubin; direct bilirubin (conjugated) is usually normal or
slightly increased. Urine color is normal, and no bilirubin is found in the urine.
b. Biliary obstruction, which may be intrahepatic (as with a chlorpromazine reaction) or
extrahepatic (as with a biliary stone), increases total bilirubin and direct bilirubin;
intrahepatic cholestasis (e.g., from chlorpromazine) may increase direct bilirubin as well.
Urine color is dark, and bilirubin is present in the urine.
c. Liver cell necrosis, as occurs in viral hepatitis, may cause an increase in both direct
bilirubin (because inflammation causes some bile sinusoid blockage) and indirect bilirubin
(because the liver’s ability to conjugate is altered). Urine color is dark, and bilirubin is
present in the urine.
C. Serum proteins
1. Primary serum proteins measured are albumin and the globulins (i.e., , , ).
a. Albumin (4 to 6 g/dL) maintains serum oncotic pressure and serves as a transport agent.
Because it is primarily manufactured by the liver, liver disease can decrease albumin levels.
Albumin can also be used to assess nutritional status.
b. Globulin (23 to 35 g/L) relates to the total measurement of immunoglobins (antibodies)
found in the serum and function as transport agents and play a role in certain immunologi-
cal mechanisms. A decrease in albumin levels usually results in a compensatory increase in
globulin production.
2. Normal values for total serum protein levels are 6 to 8 g/dL (60 to 80 g/L).
V. URINALYSIS. Composed of chemical and microscopic tests of the urine used to provide basic
information regarding renal function, urinary tract disease, and the presence of certain systemic
diseases. Components of a standard urinalysis include physical (color, turbidity, odor, specific gravity,
and osmolality), chemical (pH, Hb, glucose, protein, glucose, ketone, leukocyte esterase, nitrites,
bilirubin) and microscopic examination (RBC, WBC, epithelial cells, casts, bacteria).
A. Appearance. Normal urine is clear and ranges in color from pale yellow to deep gold. Changes
in color can result from drugs, diet, or disease.
1. A red color may indicate, among other things, the presence of blood or phenolphthalein (a
laxative).
550 Chapter 30 V. A
b. In many renal diseases, urea and creatinine accumulate in the blood because they are not
excreted properly.
c. These tests also aid in determining drug dosage for drugs excreted through the kidneys.
2. Azotemia describes excessive retention of nitrogenous waste products (BUN and creatinine) in the
blood. The clinical syndrome resulting from decreased renal function and azotemia is called uremia.
a. Renal azotemia results from renal disease, such as glomerulonephritis and chronic
pyelonephritis.
b. Prerenal azotemia results from such conditions as severe dehydration, hemorrhagic shock,
and excessive protein intake.
c. Postrenal azotemia results from such conditions as ureteral or urethral stones or tumors
and prostatic obstructions.
3. Clearance—a theoretical concept defined as the volume of plasma from which a measured
amount of substance can be completely eliminated, or cleared, into the urine per unit time—can
be used to estimate glomerular function.
B. BUN
1. Urea, an end product of protein metabolism, is produced in the liver. From there, it travels
through the blood and is excreted by the kidneys. Urea is filtered at the glomerulus, where the
tubules reabsorb approximately 40%. Thus, under normal conditions, urea clearance is about
60% of the true GFR.
2. Normal values for BUN range from 8 mg/dL to 18 mg/dL (3.0 to 6.5 mmol/L).
a. Decreased BUN levels occur with significant liver disease.
b. Increased BUN levels may indicate renal disease. However, factors other than glomerular
function (e.g., protein intake, reduced renal blood flow, blood in the gastrointestinal tract)
readily affect BUN levels, sometimes making interpretation of results difficult.
C. Serum creatinine
1. Creatinine (CR), the metabolic breakdown product of muscle creatine phosphate, has a
relatively constant level of daily production. Blood levels vary little in a given individual.
2. Creatinine is excreted by glomerular fi ltration and tubular secretion. Creatinine clearance parallels the
GFR within a range of 10% and is a more sensitive indicator of renal damage than BUN levels because
renal impairment is almost the only cause of an increase in the serum creatinine level.
3. Normal values for serum creatinine range from 0.6 to 1.2 mg/dL (50 to 110 mmol/L).
a. Values vary with the amount of muscle mass—a value of 1.2 mg/dL in a muscular athlete
may represent normal renal function, whereas the same value in a small, sedentary person
with little muscle mass may indicate significant renal impairment.
b. Generally, the serum creatinine value doubles with each 50% decrease in GFR. For
example, if a patient’s normal serum creatinine is 1 mg/dL, 1 mg/dL represents 100% renal
function, 2 mg/dL represents 50% function, and 4 mg/dL represents 25% function.
D. Creatinine clearance
1. Creatinine clearance, which represents the rate at which creatinine is removed from the
blood by the kidneys, roughly approximates the GFR.
a. The value is given in units of milliliters per minute, representing the volume of blood
cleared of creatinine by the kidney per minute.
b. Normal values for men range from 75 to 125 mL/min.
2. Calculation requires knowledge of urinary creatinine excretion (usually over 24 hrs) and con-
current serum creatinine levels. Creatinine clearance is calculated as follows:
CuV
ClCR
CCR
where ClCR is the creatinine clearance in milliliters per minute, CU is the concentration of
creatinine in the urine, V is the volume of urine (in milliliters per minute of urine formed over
the collection period), and CCR is the serum creatinine concentration.
3. Suppose the serum creatinine concentration is 1 mg/dL, and 1440 mL of urine was collected in
24 hrs (1440 mins) for a urine volume of 1 mL/min. The urine contains 100 mg/dL of
creatinine. Creatinine clearance is calculated as:
100 mg/mL 1 mL/min
___
100 mL/min
1 mg/dL
552 Chapter 30 VII. D
4. Incomplete bladder emptying and other problems may interfere with obtaining an accurate
timed urine specimen. Thus, estimations of creatinine clearance may be necessary. These
estimations require only a serum creatinine value. One estimation uses the method of
Cockcroft and Gault, which is based on body weight, age, and gender.
a. This formula provides an estimated value, calculated for males as:
Clcr [140 age (in years)] body weight (in kg)
72 Ccr (in mg/dL)
b. For females, use 0.85 of the value calculated for males.
c. Example: A 20-year-old man weighing 72 kg has a CCR of 1.0 mg/dL; thus
Ccr
(140 20 years) 72 kg 120 mL/min
72 1 mg/dL
5. Determination of GFR. T he modified diet in renal disease (MDRD) equation is considered a
more accurate measurement of GFR than other equations used to estimate renal function (e.g.,
Cockcroft–Gault) in patients with reduced GFR and is used in staging renal disease. Patients
must have a serum creatinine concentration.
a. The MDRD equation for males is as follows:
GFR 186 (Pcr) 1.154 age 0.203
where Pcr is serum creatinine. For females, multiply the result by 0.742; for African
Americans, multiply by 1.210.
b. The MDRD has been validated in Caucasians, patients with diabetic kidney disease, kid-ney
transplant recipients, and African Americans and Asians with nondiabetic kidney disease.
c. The MDRD equation has not been validated in patients 18 years of age, pregnant women,
patients 70 years of age, other ethnic groups, patients with normal kidney function who are
at an increased risk for chronic kidney disease, and patients with normal renal function.
d. Many institutions are routinely reporting an MDRD-derived GFR estimation for patients as
a routine component of a blood chemistry study. This value should be used to assist the
clini-cian in staging a patient’s degree of renal dysfunction and is not a substitute for
creatinine clearance as estimated by the Cockcroft and Gault equation, which should be
used for drug dosing in renal impairment. The MDRD estimate has not been evaluated for
the purpose of drug dosing.
VII. ELECTROLYTES
A. Sodium (Na)
1. Sodium is the major cation of the extracellular fl uid. Sodium, along with chloride (Cl), potassium
(K), and water, is important in the regulation of osmotic pressure and water balance between
intracellular and extracellular fluids. Normal values are 135 to 147 mEq/L or mmol/L.
2. The sodium concentration is defined as the ratio of sodium to water, not the absolute amounts
of either. Laboratory tests for sodium are used mainly to detect disturbances in water balance
and body osmolality. The kidneys are the major organs of sodium and water balance.
3. An increase in sodium concentration (hypernatremia) may indicate impaired sodium excretion
or dehydration. A decrease in sodium concentration (hyponatremia) may reflect overhydration,
abnormal sodium loss, or decreased sodium intake.
4. Patients with kidney, heart, or pulmonary disease may have difficulty with sodium and water
balance. In adults, changes in sodium concentrations most often reflect changes in water
balance, not salt imbalances. Therefore, sodium concentration is often used as an indicator of
fluid status, rather than salt imbalance.
5. Control of sodium by the body is accomplished mainly through the hormones aldosterone and
antidiuretic hormone (ADH).
a. ADH is released from the pituitary gland in response to signals from the hypothalamus.
ADH’s presence in the distal tubules and collecting ducts of the kidney causes them to
become more permeable to the reabsorption of water; therefore, concentrating urine.
Clinical Laboratory Tests 553
C. Chloride (Cl)
1. Chloride is the major anion of the extracellular fluid and is important in the maintenance of
acid–base balance. Alterations in the serum chloride concentration are rarely a primary
indicator of major medical problems. Chloride itself is not of primary diagnostic significance. It
is usually measured to confirm the serum sodium concentration. The relationship among
sodium, chloride, and HCO 3 is described by the following:
Cl HCO 3 R Na
where R is the anion gap. The normal value for Cl is 95 to 105 mEq/L or mmol/L.
2. Hypochloremia is a decreased chloride concentration, and it is often accompanied by metabolic
alkalosis or acidosis caused by organic or other acids. Other causes include chronic renal failure,
adrenal insufficiency, fasting, prolonged diarrhea, severe vomiting, and diuretic therapy.
3. Hyperchloremia is an increased chloride concentration that may indicate hyperchloremic
metabolic acidosis. Hyperchloremia in the absence of metabolic acidosis is unusual because
chloride retention is often accompanied by sodium and water retention. Other causes include
acute renal failure, dehydration, and excess chloride administration.
D. Bicarbonate (HCO 3)/carbon dioxide (CO2) content
1. The carbon dioxide (CO2) content represents the sum of the bicarbonate (HCO 3) concentration and
the concentration of CO2 dissolved in the serum. The HCO 3/CO2 system is the most impor-tant
buffering system to maintain pH within physiological limits. Most disturbances of acid–base balance
can be considered in terms of this system. Normal values are 22 to 28 mEq/L or mmol/L.
2. The relationship among this system is defined as follows:
HCO 3 H H2CO3 H2O CO2
(bicarbonate ions bind hydrogen ions to form carbonic acid). Clinically, the serum HCO 3
concentration is measured because acid–base balance can be inferred if the patient has normal
pulmonary function.
3. Hypobicarbonatemia is usually caused by metabolic acidosis, renal failure, hyperventilation,
severe diarrhea, drainage of intestinal fluid, and by drugs such as acetazolamide. Toxicity
caused by salicylates, methanol, and ethylene glycol can also decrease the HCO 3 level.
4. Hyperbicarbonatemia is usually caused by alkalosis, hypoventilation, pulmonary disease,
persis-tent vomiting, excess HCO 3 intake with poor renal function, and diuretics.
VIII. MINERALS
A. Calcium (Ca)
1. Calcium plays an important role in nerve impulse transmission, muscle contraction, pancreatic
insulin release, and hydrogen ion release from the stomach, as a cofactor for some enzyme
reac-tions and blood coagulation and, most important, bone and tooth structural integrity.
Normal total calcium values are 8.8 to 10.3 mg/dL or 2.20 to 2.56 mmol/L.
2. The total calcium content of normal adults is 20 to 25 g/kg of fat-free tissue, and about 44% of
this calcium is in the body skeleton. Approximately 1% of skeletal calcium is freely
exchangeable with that of the extracellular fluid. The reservoir of calcium in bones maintains
the concentra-tion of calcium in the plasma constant. About 40% of the calcium in the
extracellular fluid is bound to plasma proteins (especially albumin), 5% to 15% is complexed
with phosphate and citrate, and 45% to 55% is in the unbound, ionized form. Most laboratories
measure the total calcium concentration; however, it is the free, ionized calcium that is
important physiologically. Ionized calcium levels may be obtained from the laboratory.
Clinically, the most important de-terminant of ionized calcium is the amount of serum protein
(albumin) available for binding. The normal serum calcium range is for a serum albumin of 4.0
g/dL. A good approximation is that for every 1.0 g/dL decrease in albumin, 0.8 g/dL should be
added to the calcium laboratory result. Doing this corrects the total plasma concentration to
reflect the additional amount of free (active) calcium.
3. Hypocalcemia usually implies a deficiency in either the production or response to parathyroid
hormone (PTH) or vitamin D. PTH abnormalities include hypoparathyroidism, pseudohypo-
parathyroidism, or hypomagnesemia. Vitamin D abnormalities can be caused by decreased
Clinical Laboratory Tests 555
Study
Questions 2. Hematological studies are most likely to show a
low reticulocyte count in a patient who has which
Directions for questions 1–16: Each of the questions, of the following abnormalities?
statements, or incomplete statements in this section can (A) aplastic anemia secondary to cancer chemotherapy
be correctly answered or completed by one of the (B) acute hemolytic anemia secondary to quinidine
suggested answers or phrases. Choose the best answer. treatment
(C) severe bleeding secondary to an
1. Hematological testing of a patient with AIDS is most
automobile accident
likely to show which of the following abnormalities? (D) iron-deficiency anemia 1 week after
(A) basophilia treatment with ferrous sulfate
(B) eosinophilia (E) megaloblastic anemia owing to folate deficiency
(C) lymphopenia 1 week after treatment with folic acid
(D) reticulocytosis
(E) agranulocytosis
556 Chapter 30 7. A 52-year-old male construction worker who drinks
“fairly heavily” when he gets off work is seen in the
emergency room with, among other abnormal
3. All of the following findings on a routine urinalysis laboratory results, an increased creatine kinase (CK)
would be considered normal except which one? level. All of the following circumstances could
(A) pH: 6.5 explain this increase except which one?
(B) glucose: negative (A) He fell against the bumper of his car in a
(C) ketones: negative drunken stupor and bruised his right side.
(D) white blood cells (WBCs): 3 per high-power (B) He is showing evidence of some liver
field (HPF), no casts damage owing to the heavy alcohol intake.
(E) red blood cells (RBCs): 5 per HPF (C) He has experienced a heart attack.
4. A 12-year-old boy is treated for otitis media with (D) He received an intramuscular (IM) injection a
cefaclor (Ceclor). On the seventh day of therapy, few hours before the blood sample was drawn.
he spikes a fever and develops an urticarial rash (E) He pulled a muscle that day when lifting a
on his trunk. Which of the following laboratory heavy concrete slab.
tests could best confirm the physician’s suspicion 8. A 45-year-old man with jaundice has spillage of
of a hypersensitivity (allergic) reaction? bilirubin into his urine. All of the following state-
(A) complete blood count (CBC) and differential ments could apply to this patient except which one?
(B) serum hemoglobin (Hb) and reticulocyte count (A) His total bilirubin is increased.
(C) liver function test profile (B) His direct bilirubin is increased.
(D) lactate dehydrogenase (LDH) isoenzyme profile (C) He may have viral hepatitis.
(E) red blood cell (RBC) count and serum bilirubin (D) He may have hemolytic anemia.
5. An increased hematocrit (Hct) is a likely finding in (E) He may have cholestatic hepatitis.
all of the following individuals except which one? For questions 9–11: A 70-year-old black man weighing
(A) a man who has just returned from a 3- 154 lbs complains of chronic fatigue. Several laboratory
week skiing trip in the Colorado Rockies tests were performed with the following results:
(B) a woman who has polycythemia vera blood urea nitrogen (BUN) 15 mg/dL
(C) a hospitalized patient who mistakenly aspartate aminotransferase (AST) within normal
received 5 L of intravenous (IV) dextrose 5%
limits white blood cell (WBC) count 7500/mm3
in water (D5W) over the last 24 hrs red blood cell (RBC) count 4
(D) a man who has been rescued from the Arizona
million/mm3 hematocrit (Hct) 29%
desert after spending 4 days without water
hemoglobin (Hb) 9 g/dL
(E) a woman who has chronic obstructive pulmonary
disease 9. T his patient’s mean cell hemoglobin
concentration (MCHC) is
6. A 29-year-old white man is seen in the emergency
room. His white blood cell (WBC) count is 14,200 (A) 27.5.
with 80% polys. All of the following conditions (B) 28.9.
could normally produce these laboratory findings (C) 31.0.
except which one? (D) 33.5.
(E) 35.4.
(A) a localized bacterial infection on the tip of
the index finger 10. His mean cell volume (MCV) is
(B) acute bacterial pneumonia caused (A) 61.3.
by Streptococcus pneumoniae (B) 72.5.
(C) a heart attack (C) 77.5.
(D) a gunshot wound to the abdomen with a loss (D) 90.2.
of 2 pints of blood (E) 93.5.
(E) an attack of gout
11. From the data provided and from the calculations in Clinical Laboratory Tests 557
questions 9 and 10, this patient is best described as
(A) normal except for a slightly increased blood
urea nitrogen (BUN). 15. All of the following statements about calcium (Ca)
(B) having normochromic, microcytic anemia. and phosphorus (PO4) are true except which one?
(C) having sickle-cell anemia. (A) An alcoholic with a serum albumin of 2.0
(D) having hypochromic, normocytic anemia. g/dL and a serum total calcium of 8.0 mg/dL
(E) having folic acid deficiency. has a corrected total calcium of 9.6 mg/dL.
12. All of the following statements about sodium (Na) (B) Calcium and PO4 levels should be
are true except which one? interpreted together because many of the
same factors influence both minerals.
(A) T he normal range for sodium is 135
(C) Metastatic cancer often induces a decrease
to 147 mEq/L. in serum calcium levels.
(B) Sodium is the major cation of the extracellular (D) A patient with renal failure may present
fluid, and the laboratory test is used mainly to
with hypocalcemia and hyperphosphatemia.
detect disturbances in water balance.
(C) Hyponatremia usually results from the total 16. All of the following are important functions
body depletion of sodium or through a dilutional of magnesium (Mg) except
effect. (A) nerve conduction.
(D) Control of the sodium concentration is mainly (B) phospholipid synthesis.
through regulation of arterial pH. (C) muscle contractility.
13. A 53-year-old woman with diabetes mellitus is seen in (D) carbohydrate, fat, and electrolyte metabolism.
the emergency room. Her blood glucose is 673 mg/dL Directions for questions 17–19: The questions and
and ketones are present in her blood. A diagnosis of incomplete statements in this section can be correctly
diabetic ketoacidosis (DKA) is made. Other important answered or completed by one or more of the
laboratory values are potassium of 4.8 mEq/L, 4 glucose suggested answers. Choose the answer, A–E.
in urine, and an arterial pH of 7.1. All of
A if I only is correct B if
the following statements apply to this patient
III only is correct C if I
except which one?
and II are correct D if II
(A) Her potassium value is normal; and III are correct
therefore, no potassium supplementation E if I, II, and III are correct
is likely to be necessary.
(B) Her potassium value should be corrected 17. Factors likely to cause an increase in the blood
owing to her acidosis; a corrected potassium urea nitrogen (BUN) level include
would be 3.0 mEq/L. I. intramuscular (IM) injection of diazepam
(C) Potassium supplementation should be instituted (Valium).
because her total body potassium is depleted. II. severe liver disease.
(D) Factors affecting potassium in this patient III. chronic kidney disease.
include glycosuria and arterial pH.
18. A patient who undergoes serum enzyme testing is
14. A 50-year-old man presents with bicarbonate of found to have an increased aspartate
18 mEq/L. All of the following could be a cause aminotransferase (AST) level. Possible underlying
of his low bicarbonate level except causes of this abnormality include
(A) metabolic acidosis. I. methyldopa-induced hepatitis.
(B) salicylate toxicity. II. congestive heart failure (CHF).
(C) diuretic therapy. III. pneumonia.
(D) diarrhea.
19. Serum enzyme tests that may aid in the diagnosis
of myocardial infarction include
I. alkaline phosphatase.
II. creatine kinase (CK).
III. lactate dehydrogenase (LDH).
558 Chapter 30
I. INTRODUCTION
A. Definition. Coronary artery disease (CAD) is a general term that refers to several diseases other
than atherosclerosis, which causes a narrowing of the major epicardial coronary arteries. Ischemic
heart disease (IHD) is a form of heart disease with primary manifestations that result from myo-
cardial ischemia owing to atherosclerotic CAD. This term encompasses a spectrum of conditions,
ranging from the asymptomatic preclinical phase to acute myocardial infarction and sudden cardiac
death and is used throughout this chapter.
B. Incidence
1. IHD continues to be the most common chronic life-threatening illness in the United States
(231.1 to 297.9 deaths per 100,000); cancer is the second leading cause of death (159.1 to 228.1
deaths per 100,000). It currently affects 11 million citizens in the United States, and it has been
estimated that if all forms of major CAD were eliminated, life expectancy would increase by
almost 7 years to our current population.
2. Each year, more than 5 million patients present to emergency rooms with chest discomfort and
related symptoms, and approximately 1.5 million are hospitalized for acute coronary syn-
dromes. Each year in the United States, more than 1 million patients suffer an acute myocardial
infarction (MI).
C. Economics. Based on models evaluating the costs associated with the treatment of Medicare pa-tients
with common IHD-related diagnosis, it has been estimated that the direct costs of hospitaliza-tion are
$15 billion yearly, with an additional $4.5 billion yearly in diagnostic procedures.
D. Clinical guidelines. Owing to the clinical, humanistic, and economic effect that IHD has in the
United States, evidence-based practice guidelines have evolved based on the differences in the
diagnosis and management of IHD. The author has relied heavily on the use of those guidelines to
ensure the most up-to-date recommendations based on the clinical literature. However, the author
has chosen to limit the treatment options to those within a class I benefit; (Benefit Risk) with the
associated level of evidence rather than providing each recommendation for all four classes (I,
IIa, IIb, III). Guidelines that are pertinent to daily pharmacy practice include the following:
1. Fraker TD Jr, Fihn SD, 2002 Chronic Stable Angina Writing Committee, et al. 2007 chronic
angi-na focused update of the ACC/AHA 2002 guidelines for the management of patients with
chronic stable angina: a report of the American College of Cardiology/American Heart
Association Task Force on Practice Guidelines Writing Group to develop the focused update of
the 2002 guide-lines for the management of patients with chronic stable angina. J Am Coll
Cardiol. 2007;50(23): 2264–2274. http://content.onlinejacc.org/cgi/reprint/50/23/2264.pdf
2. Anderson JL, Adams CD, Antman EM, et al. ACC/AHA 2007 guidelines for the management of
patients with unstable angina/non–ST-elevation myocardial infarction: a report of the American
College of Cardiology/American Heart Association Task Force on Practice Guidelines (Writing
Committee to Revise the 2002 Guidelines for the Management of Patients With Unstable Angina/
Non–ST-Elevation Myocardial Infarction) developed in collaboration with the American College
560
Coronary Artery Disease 561
of Emergency Physicians, the Society for Cardiovascular Angiography and Interventions, and
the Society of Thoracic Surgeons endorsed by the American Association of Cardiovascular and
Pulmonary Rehabilitation and the Society for Academic Emergency Medicine. J Am Coll
Cardiol. 2007;50(7):e1–e157. http://content.onlinejacc.org/cgi/reprint/50/7/e1.pdf
3. Wright RS, Anderson JL, Adams CD, et al. 2011 ACCF/AHA focused update incorporated into
the ACC/AHA 2007 of the guidelines for the management of patients with unstable
angina/non– ST-elevation myocardial infarction: a report of the American College of
Cardiology Foundation/ American Heart Association Task Force on Practice Guidelines
developed in collaboration with the American Academy of Family Physicians, Society for
Cardiovascular Angiography and Interventions, and the Society of Thoracic Surgeons. J Am
Coll Cardiol. 2011;57(19):e215–e367. http://content.onlinejacc.org/cgi/reprint/57/19/e215.pdf
4. Antman EM, Anbe DT, Armstrong PW, et al. ACC/AHA guidelines for the management of
patients with ST-elevation myocardial infarction—executive summary. A report of the Ameri-
can College of Cardiology/American Heart Association Task Force on Practice Guidelines
(Writing Committee to revise the 1999 guidelines for the management of patients with acute
myocardial infarction). J Am Coll Cardiol. 2004;44(3):671–719.
http://content.onlinejacc.org/cgi/ reprint/44/3/671.pdf
5. Canadian Cardiovascular Society, American Academy of Family Physicians, American College
of Cardiology, et al. 2007 focused update of the ACC/AHA 2004 guidelines for the manage-
ment of patients with ST-elevation myocardial infarction: a report of the American College of
Cardiology/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol.
2008;51(2):210–247. http://content.onlinejacc.org/cgi/reprint/51/2/210.pdf
6. Kushner FG, Hand M, Smith SC Jr, et al. 2009 focused updates: ACC/AHA guidelines for the
management of patients with ST-elevation myocardial infarction (updating the 2004 guideline and
2007 focused update) and ACC/AHA/SCAI guidelines on percutaneous coronary interven-tion
(updating the 2005 guideline and 2007 focused update): a report of the American College of
Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J Am Coll
Cardiol. 2009;54(23):2205–2241. http://content.onlinejacc.org/cgi/reprint/54/23/2205.pdf
7. National Cholesterol Education Program (NCEP). Third report of the National Cholesterol Education
Program (NCEP) Expert Panel: Detection, evaluation, and treatment of high blood cholesterol in adults
(Adult Treatment Panel III). Final Report. NIH Publication No. 02-5215. Bethesda, MD: National In-
stitutes of Health; 2002. Available at http://www.nhlbi.nih.gov/guidelines/cholesterol/atp3_rpt.htm
E. Manifestations
1. Angina pectoris, an episodic, reversible oxygen insufficiency, is the most common form of
IHD (see II).
2. The term acute ischemic (coronary) syndromes describes a group of clinical symptoms repre-
senting acute myocardial ischemia. The clinical symptoms include acute myocardial infarction,
which includes either ST-segment elevation (STEMI) or non–ST-segment elevation
(NSTEMI), and unstable angina (UA) (see III).
F. Etiology. T he processes, singly or in combination, that produce IHD include decreased blood flow
to the myocardium, increased oxygen demand, and decreased oxygenation of the blood. Generally,
significant IHD is defined via angiography as a stenosis that is 70% of the diameter of at least one
major coronary artery segment or 50% of the diameter of the left main coronary artery.
1. Decreased blood flow (Figure 31-1)
a. Atherosclerosis, with or without coronary thrombosis, is the most common cause of IHD.
In this condition, the coronary arteries are progressively narrowed by smooth muscle cell
prolif-eration and the accumulation of lipid deposits (plaque) along the inner lining (intima)
of the arteries.
b. Coronary artery spasm, a sustained contraction of one or more coronary arteries, can occur
spontaneously or be induced by irritation (e.g., by coronary catheter or intimal hemorrhage),
exposure to the cold, and ergot-derivative drugs. One long-term study demonstrated that
coronary spasm was most often associated with an atypical chest pain syndrome and cigarette
smoking. These spasms can cause Prinzmetal angina and even MI. Variant angina (Prinzmetal
angina) is a form of unstable angina that usually occurs spontaneously, is characterized by
transient ST-segment elevation, and most commonly resolves without progression to MI.
562 Chapter 31 I. F
Figure 31-1. Oxygen and other nutrients are borne to the myocardium through the two major coronary
arteries (the left and right) and their tributaries. The hemodynamic consequences of ischemic heart disease
depend on which of the coronary vessels are involved and what part of the myocardium those vessels supply.
c. Traumatic injury, whether blunt or penetrating, can interfere with myocardial blood
supply (e.g., the impact of a steering wheel on the chest causing a myocardial contusion in
which the capillaries hemorrhage).
d. Embolic events, even in otherwise normal coronary vessels, can abruptly restrict the
oxygen supply to the myocardium.
2. Increased oxygen demand usually in the presence of a fixed restricted oxygen supply can
occur with exertion (e.g., exercise, shoveling snow) and emotional stress as well as under
circumstances external to the coronary arterial bed, which increases sympathetic stimulation
and heart rate. Some factors affecting cardiac workload and therefore myocardial oxygen
supply and demand are listed in Table 31-1.
a. Diastole. Under normal circumstances, almost all of the oxygen is removed (during
diastole) from the arterial blood as it passes through the heart. Thus, little remains to be
extracted if oxygen demand increases. To increase the coronary oxygen supply, blood flow
has to increase. The normal response mechanism is for the blood vessels, particularly the
coronary arteries, to dilate, thereby increasing blood flow.
b. Systole. The two phases of systole—contraction and ejection—strongly influence oxygen demand.
(1) The contractile (inotropic) state of the heart influences the amount of oxygen it
requires to perform.
Factor Heart Rate Blood Pressure Ejection Time Ventricular Volume Inotropic Effect
(2) Increases in systolic wall tension, influenced by left ventricular volume and systolic
pressure, increase oxygen demand.
(3) Lengthening of ejection time (i.e., the duration of systolic wall tension per cardiac
cycle) also increases oxygen demand.
(4) Changes in heart rate influence oxygen consumption by changing the ejection time.
3. Reduced blood oxygenation. T he oxygen-carrying capacity of the blood may be reduced, as
occurs in various forms of anemia or hypoxemia.
G. Risk factors for IHD and goals for secondary prevention have been updated with the recently
released Guidelines for the Management of Patients with ST-Elevation Myocardial Infarction and
are provided in Table 31-2.
H. Therapeutic considerations. Because most IHD occurs secondary to atherosclerosis, which is a
long-term, cumulative process, medical efforts focus on reducing risk factors through individual
pa-tient education and media campaigns. Once manifestations occur, treatment addresses their
specific variables.
Table 31-2 RISK FACTORS FOR ISCHEMIC HEART DISEASE AND GUIDELINES FOR
THEIR MODIFICATION, WHEN APPLICABLE
The addition of other blood pressure lowering drugs such as thiazides as needed to achieve the goal blood
pressure.
Blood lipid management: Low density lipoprotein-cholesterol (LDL-C) levels should be less than 100 mg/dL. (If
triglycerides are greater than or equal to 200 mg/dL, total cholesterol minus high density lipoprotein
cholesterol (HDL-C) should be less than 130 mg/dL.) *Class I recommendation
Initiate dietary therapy in all patients; which includes reducing intake of saturated fats ( 7% of total calories),
trans fatty acids, and cholesterol ( 200 mg/day).
The addition of plant stanol/sterols (2 g/day) and/or viscous fiber ( 10 g/day) to further lower LDL-C.
Promotion of daily physical activity and weight management.
It may be reasonable to encourage the increase in the consumption of omega-3 fatty acids in the form of fish or
in capsule form (1 g/d) for risk reduction.
A fasting lipid profile should be assessed in all patients and within 24 hrs of hospitalization for those with an
acute cardiovascular or coronary event.
Physical activity: Participate in 30 mins of physical activity 7 days a week. *Class 1 recommendation
It is recommended that all patients have a risk assessment with a physical activity history and/or an exercise
test to guide prescription.
All patients should be encouraged to participate in 30 to 60 mins of moderate-intensity aerobic activity, (i.e.,
brisk walking) on most, preferably all, days of the week, supplemented by an increase in daily lifestyle
activities (e.g., walking breaks at work, gardening, household work).
Advise medically supervised programs for high-risk patients (e.g., recent acute coronary syndrome
or revascularization, heart failure).
Weight management: Body mass index (BMI) of 18.5 to 24.9 kg/m 2 with waist circumference of less than 35
inches (women) and less than 40 inches (men). *Class 1 recommendation
Assessment of body mass index and/or waist circumference on each visit and consistently.
Encourage weight maintenance/reduction through an appropriate balance of physical activity, caloric intake, and formal
behavioral programs when indicated to maintain/achieve a body mass index between 18.5 and 24.9 kg/m 2.
The initial goal of weight loss therapy should be to reduce body weight by approximately 10% from baseline.
With success, further weight loss can be attempted if indicated.
If waist circumference is greater than or equal to 35 inches in women and greater than or equal to 40 inches in men, it is
useful to initiate lifestyle changes and consider treatment strategies for metabolic syndrome as indicated.
Diabetes management: Achieve a glycosylated hemoglobin (HbA1c) level of less than 7% *Class 1
recommendation
It is recommended to initiate lifestyle and pharmacotherapy to achieve near-normal levels of HbA1c.
Aggressive modification of other risk factors such as physical activity, weight management, blood
pressure control, and cholesterol management, as recommended above is benefi cial.
Coordination of diabetic care with patient’s primary care physician or endocrinologist is beneficial.
*Class I recommendation: The benefits of the intervention/treatment are much greater than the risk, and the procedure/treatment should be
performed.
ACE, angiotensin-converting enzyme.
exercise. Exercise stress testing is preferable to other variations of the stress test (pharmacolo-
gical) in patients who are able to exercise.
3. Pharmacological stress testing is performed in suspected IHD patients when they are not able
to perform more than moderate exercise due to various reasons (i.e., severe arthritis, prior
injury, reduced exercise tolerance as a result of debilitating illnesses, etc.), or in patients who
are unable to increase the heart rate.
a. Intravenous dipyridamole (Persantine®; coronary vasodilation), adenosine (Adenocard®;
coronary vasodilation) by inhibiting cellular uptake and degradation of adenosine increase
coronary blood flow, and high-dose dobutamine (Dobutrex®; 20 to 40 mcg/kg/min)
increase oxygen demand through increased heart rate, systolic blood pressure, and
myocardial con-tractility causing an increase in myocardial blood flow are all able to induce
detectable cardiac ischemia in conjunction with EKG/ECG testing.
b. Side effects occur for each of the agents and include dipyridamole (angina, 18% to 42%;
arrhythmias, 2%; headache, 5% to 23%; dizziness, 5% to 21%; nausea, 8% to 12%; and flush-
ing, 3%), adenosine (chest pain, 57%; headache, 35%; flushing, 25%; shortness of breath, 15%;
and first-degree atrioventricular [AV] heart block, 18%), dobutamine (premature ventricular
beats, 15%; premature atrial beats, 8%; supraventricular tachycardia and nonsustained ven-
tricular tachycardia, 3% to 4%; nausea, 8%; anxiety, 6%; headache, 4%; and tremor, 4%).
4. Stress perfusion imaging with thallium-201 or more recently, technetium-99m (99mTc)
(sestamibi [Cardiolite®] or tetrafosmin [Myoview®]), can diagnose multivessel disease,
localized ischemia, and may be able to determine myocardial viability. Coronary arteriography
and cardiac catheter-ization are very specific and sensitive but are also invasive, expensive, and
risky (the mortality rate is 1% to 2%); therefore, they must be used judiciously when trying to
confirm suspected angina and to differentiate its origin.
5. Various drugs can have an effect on the EKG/ECG and should be considered before, during,
and after an exercise test is carried out. Examples include
a. Digoxin (Lanoxin®) produces abnormal exercise-induced ST depression in 25% to 40% of
apparently healthy, normal subjects without ischemia.
b. -Adrenergic blockers may delay the development of an abnormal EKG/ECG if patients
receive them before or during a stress test. If possible, therapy should be slowly withheld
from the patient at least four to five half-lives before the exercise testing. If it is not possible
to with-draw therapy, the clinician needs to recognize that the test might be less reliable.
c. Antihypertensives such as vasodilators can alter the stress test by altering the normal
hemody-namic response of blood pressure. In addition, short-term use of nitrates can
attenuate angina and ST-segment changes associated with myocardial ischemia.
F. Treatment goals
1. To prevent MI and death, thereby increasing a patient’s quality of life
2. To reduce symptoms of angina and occurrence of ischemia, which should improve a patient’s
quality of life
3. To remove or reduce risk factors
4. The management of angina pectoris includes therapies aimed at reversing cardiac risk factors.
a. Hyperlipidemia, if present, should be treated. Reducing cholesterol and low-density lipo-
protein (LDL) is associated with a reduced risk of cardiovascular disease and incidence of
ischemic cardiac events, as demonstrated by several recent studies using -hydroxy- -
methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors. The NCEP has published
guidelines for treatment of high blood cholesterol (Adult Treatment Panel III), and a fourth
set of guidelines is currently being developed with a publication date in 2012.
(1) Total cholesterol is no longer the primary target of treatment; LDL cholesterol is now
the primary target.
(2) Current recommendations include the completion of a lipoprotein profile—total choles-
terol, LDL cholesterol, high-density lipoprotein (HDL) cholesterol, and triglycerides—
as the preferred initial test, rather than screening for total cholesterol and HDL alone.
Additionally, the ATP-3 guidelines identify LDL cholesterol 100 mg/dL as optimal and
raises categorical low HDL cholesterol from 35 mg/dL to 40 mg/dL because the latter
is a better measure of a depressed HDL.
Coronary Artery Disease 567
Finally, the ATP-3 lowered the triglyceride classification cutpoints to give more
attention to moderate elevations.
(3) Persons are categorized into one of four levels of risk to identify group-specific
treatment modalities: (1) high-risk, established IHD or IHD risk equivalents (diabetes,
noncoronary forms of atherosclerotic disease); (2) moderately high-risk, multiple (more
than two) risk factors and a calculated 10-year risk of 10% to 20%; (3) moderate risk,
multiple (more than two) risk factors and a calculated 10-year risk of 10%; and (4)
lower risk, zero to one risk factor.
(4) All individuals with IHD or IHD risk equivalents have been called “high risk,” and the
treatment goals is to have LDL-cholesterol (LDL-C) levels 100 mg/dL. Other recom-
mendations are as follows:
(a) If baseline LDL-C is 100 mg/dL (goal of treatment), patients with IHD should be given
instructions on diet and exercise and have levels monitored annually; although the
guidelines suggest an optional goal of obtaining an LDL-C of 70 mg/dL.
(b) If baseline LDL-C is 100 mg/dL, an LDL-lowering drug should be started along
with therapeutic lifestyle changes (TLCs). Lifestyle-related risk factors include
things such as obesity, physical inactivity, elevated triglyceride, low-HDL-C, or
metabolic syndrome.
(c) Adult Treatment Panel III did not mandate LDL-lowering drugs for patients with
baseline LDL-C levels between 100 and 129 mg/dL, but suggested intensifying
life-style and the optional use of drug therapies to lower LDL to 100 mg/dL.
However, if the patient had elevated triglycerides or low high-density lipoprotein
cholesterol (HDL-C), a drug should be started that targets these abnormalities—for
example, nicotinic acid or fibric acid if the patient has elevated triglycerides ( 200
mg/dL) or low HDL levels ( 40 mg/dL). If triglycerides are 500 mg/dL, consider
fibrate or niacin before LDL-lowering therapy.
(5) For moderately high-risk patients, the recommended LDL-C goal is 130 mg/dL, but a
goal of 100 mg/dL is an optional goal, based on recent clinical trial evidence. In addi-
tion, a recommendation to initiate TLCs when the patient has an LDL-C 130 mg/dL.
(6) For moderate risk patients, the recommended LDL-C goal is 130 mg/dL. In addition, a
recommendation to initiate TLC when the patient has an LDL-C 160 mg/dL.
(7) For lower risk patients, the recommended LDL-C goal is 160 mg/dL, but again, a
recommendation to initiate TLC for those patients with an LDL 190 mg/dL.
(8) The metabolic syndrome is closely linked to insulin resistance, when the normal actions of
insulin are impaired. Excess body fat and physical inactivity promote the development of
the syndrome; however, some individuals may be predisposed genetically. Patients with
three or more of the following characteristics are referred to as having the metabolic syn-
drome and should be treated accordingly: abdominal obesity, triglycerides 150 mg/dL, HDL
levels of 40 mg/dL for men and 50 mg/dL for women, blood pressure readings 130/85 mm
Hg, and a fasting serum glucose level 110 mg/dL.
(9) Increased interest in triglycerides due to a long-standing association between elevated
triglyceride levels and cardiovascular disease. The continuing rise since 1976 of triglycer-
ide levels in concert with the growing incidence of obesity, type 2 diabetes mellitus, and
insulin resistance has occurred at a time when LDL-C levels have continued to decrease.
This ongoing concern has resulted in a recent scientific statement from the American Heart
Association with the intent to update clinicians on the continued role that triglycerides play
in cardiovascular disease. (Available through the Internet at: http://circ.ahajournals
.org/cgi/reprint/123/20/2292?maxtoshow &hits 10&RESULTFORMAT &fulltext
triglycerides&searchid 1&FIRSTINDEX 0&volume 123&issue 20&resource type
HWCIT)
G. Individual drug classes
1. Bile acid-binding resins
a. Mechanism of action. These insoluble, nonabsorbable, anion-exchange resins bind bile
acids within the intestines and prevent them from being reabsorbed. Bile acids are
synthesized from cholesterol.
568 Chapter 31 II. G
b. Indications. These agents have been shown to be safe and effective in lowering LDL-C,
espe-cially in patients with modestly elevated levels, in primary prevention, in young adult
men, and in postmenopausal women. They are effective in combination with other agents.
c. Currently available agents include the following:
(1) Cholestyramine (Questran®): 4 to 24 g by mouth in two daily doses
(2) Colestipol (Colestid®): 2 to 16 g by mouth in one to four daily doses
(3) Colesevelam (Welchol®): six to seven tablets (625 mg/tablet) by mouth in one daily dose
d. Precautions and monitoring effects
(1) These resins are taken just before meals and present palatability problems in patients.
(2) Gastrointestinal (GI) intolerance, especially constipation, flatulence, and dyspepsia are
frequent.
(3) Absorption of many other drugs can be affected. Other drugs should be taken 1 hr
before or 4 to 6 hrs after resins.
2. Statins or HMG-CoA reductase inhibitors
a. Mechanism of action. These agents inhibit the enzyme HMG-CoA, resulting in a reduction
in cholesterol production.
b. Indications. These agents are effective in lowering LDL levels while increasing HDL
levels and lowering triglyceride levels. They are primarily used to lower LDL cholesterol
levels and are generally considered to be the most effective of the lipid lowering agents.
c. Currently available agents include the following:
(1) Atorvastatin (Lipitor®): 10 to 80 mg by mouth daily
(2) Fluvastatin (Lescol®): 20 to 80 mg by mouth in one to two doses
(3) Lovastatin (Mevacor®, various): 10 to 80 mg by mouth in the evening
(4) Pitavastatin (Livalo®): 1 to 4 mg by mouth daily
(5) Pravastatin (Pravachol®): 10 to 80 mg by mouth daily
(6) Rosuvastatin (Crestor®): 5 to 40 mg by mouth daily
(7) Simvastatin (Zocor®, various): 5 to 80 mg by mouth in the evening
(a) Recent FDA alert on dosing limitations for products containing simvastatin:
Simvastatin 80 mg is limited to patients that have been taking this dose for 12 con-
secutive months without evidence of myopathy and are not currently taking or begin-
ning to take a simvastatin dose-limiting or contraindicated interacting medication.
d. Precautions and monitoring effects
(1) GI adverse effects are less frequently seen than with other classes of agents. Headache
and dyspepsia frequently occur and should be evaluated, then followed up in 6 to 8
weeks, and then at each follow-up visit thereafter.
(2) These agents can elevate liver function tests—alanine aminotransferase (ALT) and
aspar-tate aminotransferase (AST), which requires initial evaluation, then after
approximately 12 weeks of therapy, then annually thereafter.
(3) Although the incidence of myopathy is believed to be low (0.08%) for lovastatin and
simvastatin, elevations of creatine kinase (CK) 10 times the upper limit of normal have
been reported with pravastatin, with similar potential for the other members of the
group. Cerivastatin (Baycol) was voluntarily removed from the market owing to the
reported deaths of 31 patients from rhabdomyolysis while receiving the drug alone or in
combination with gemfibrozil (Lopid). Consequently, routine monitoring is necessary
in all patients as follows:
(a) Evaluate muscle symptoms and check CK before starting therapy, evaluate in 6 to
12 weeks after starting therapy, and then at each follow-up visit.
(b) Patients presenting with muscle soreness, tenderness, or pain should have a CK
mea-surement on presentation to minimize the development of myopathies.
(c) Concurrent therapy with other agents, including cyclosporine, macrolide
antibiotics, azole antifungals, niacin, fibrates, or nefazodone, may increase the risk.
3. Fibric acid derivatives
a. Mechanism of action. These agents are presumed to inhibit cholesterol synthesis and lower
LDL-C and bile acids. They are most effective in lowering triglyceride levels by reducing
the concentration of very-low-density lipoproteins (VLDL). In some patients, they modestly
lower LDL-C and raise HDL-C.
Coronary Artery Disease 569
HMG-CoA LDL: 18%–55% reduction Lovastatin: 10–80 mg Myopathy, increased liver enzymes
reductase HDL: 5%–15% increase Pravastatin: 20–40 mg
inhibitors TG: 7%–30% reduction Simvastatin: 5–80 mg
(statins) Fluvastatin: 20–80 mg
Atorvastatin: 10–80 mg
Rosuvastatin: 5–40 mg
Bile acid LDL: 15%–30% reduction Cholestyramine: 4–16 g GI distress, constipation, decreased
sequestrants HDL: 3%–5% increase Colestipol: 5–20 g absorption of other drugs
TG: No change or increase Colesevelam: 2.6–3.8 g
Nicotinic acid LDL: 5%–25% reduction Immediate-release: 1.5–3.0 g Flushing, hyperglycemia,
HDL: 15%–35% increase Extended-release: 1–2 g hyperuricemia (or gout),
TG: 20%–50% reduction Sustained-release: 1–2 g upper GI distress, hepatotoxicity
Fibric acids LDL: 5%–20% reduction Gemfi brozil: 600 mg Dyspepsia, gallstones,
HDL: 10%–20% increase twice daily myopathy, unexplained non-CHD
TG: 20%–50% reduction Fenofibrate: 145–160 mg deaths in WHO study
Clofibrate: 1000 mg
twice daily
CHD, coronary heart disease; GI, gastrointestinal; HDL, high-density lipoprotein; HMG-CoA, -hydroxy- -methylglutaryl-coenzyme A; LDL,
low-density lipoprotein; TG, triglycerides; WHO, World Health Organization. Adapted from Grundy SM, Becher D, Clark LT, et al. Third
Report of the National Cholesterol Education Program (NCEP): detection, evaluation, and treatment of high blood cholesterol in adults (Adult
Treatment Panel III) [NIH Publication No. 01-3670]. Washington, DC: U.S. Department of Health and Human Services; May 2001. Available
at http://www.nhlbi.nih.gov/guidelines/cholesterol/atp3xsum.pdf.
570 Chapter 31 II. G
5. Ezetimibe (Zetia®)
a. Mechanism of action. Works by selectively inhibiting the intestinal absorption of choles-
terol and related phytosterols, with a resultant decrease in intestinal cholesterol delivered to
the liver, decreased hepatic cholesterol stores, and an increase in the clearance of cho-
lesterol from the blood. Ezetimibe has demonstrated the ability to reduce total cholesterol,
LDL-C, apolipoprotein B, and triglyceride levels while increasing HDL levels in patients
with hypercholesterolemia.
b. Indications. Ezetimibe as adjunctive therapy along with dietary measures, alone in patients with
primary heterozygous familial and nonfamilial hypercholesterolemia, or in combination with the
HMG-CoA reductase inhibitors in homozygous familial hypercholesterolemia.
c. Dose. Normal dosing recommendations are a 10-mg dose given once daily.
d. Precautions. As monotherapy, studies to date have not revealed significant side effects
above those seen with placebo administration. However, when used in combination with
HMG-CoA reductase inhibitors, reports have described an increased incidence
(approximately 1.4%) in the elevation of liver transaminase levels (three times the upper
limit of normal) as compared to the incidence of 0.4% with HMG-CoA agents used alone.
6. Combination products
a. Ezetimibe/simvastatin (Vytorin®) uses the individual class properties of the HMG-CoA
reduc-tase inhibitors (simvastatin) to reduce cholesterol production with the absorption-
inhibiting properties of ezetimibe to target cholesterol with two differing mechanisms,
which might aid in improving patient compliance.
(1) Available product is supplied as 10/10, 10/20, 10/40, 10/80 combinations of ezetimibe and sim-
vastatin, respectively, and the daily recommended dose is one tablet by mouth every evening.
(2) Side effects reflect the additive side effect properties of ezetimibe and simvastatin and
warrant consideration when occurring during therapy. Recent FDA alert on dosing
limitations for products containing simvastatin: Simvastatin 80 mg is limited to pa-
tients that have been taking this dose for 12 consecutive months without evidence of
myopathy and are not currently taking or beginning to take a simvastatin dose-limiting
or contraindicated interacting medication.
b. Lovastatin/niacin (Advicor®) is a product that incorporates the actions of the HMG-CoA
re-ductase inhibitor lovastatin with an extended-release niacin component into a single
product, using differing mechanisms, which might aid in improving patient compliance.
(1) Available product is supplied as 20/500, 20/750, 20/1000, 40/1000 immediate-release
tab-let combinations of lovastatin and niacin, respectively. The recommended daily
dose is one to two tablets by mouth at bedtime.
(2) Side effects reflect the additive side effect properties for both lovastatin and niacin and
warrant consideration during therapy.
c. Amlodipine/atorvastatin (Caduet®) is a combination product that incorporates the actions of
the dihydropyridine calcium-channel blocker amlodipine with the lipid-lowering properties
of the HMG-CoA reductase inhibitor atorvastatin. The calcium-channel blocker offers no
benefit for lipid lowering but represents a potentially beneficial product for a patient with
coexisting disease states, such as IHD and hypertension, for which the use of a calcium-
channel blocker would be warranted.
(1) Available product is supplied as 2.5/10, 2.5/20, 2.5/40, 5/10, 5/20, 5/40, 5/80, 10/10,
10/20, /10/40, and 10/80; which consist of amlodipine and atorvastatin, respectively, in
an immediate-release dosage form. The recommended daily dose is one to two tablets
by mouth at bedtime.
(2) Side effects reflect the additive side effect properties for both amlodipine and
atorvastatin and warrant consideration during therapy.
d. Simvastatin/niacin (Simcor®) is a product that incorporates the actions of the HMG-CoA
reductase inhibitor simvastatin with an extended-release niacin component into a single
product, using differing mechanisms, which might aid in improving patient compliance.
(1) Available product is supplied as 500/20, 500/40, 750/20, 1000/20, 1000/40 which consist of
extended-release niacin and simvastatin, respectively, in an immediate-release dosage form.
The recommended daily dose is one to two tablets by mouth at bedtime.
Coronary Artery Disease 571
(2) Side effects reflect the additive side effect properties for both niacin and simvastatin
and warrant consideration during therapy. Recent FDA alert on dosing limitations for
products containing simvastatin: Simvastatin 80 mg is limited to patients that have
been taking this dose for 12 consecutive months without evidence of myopathy and are
not currently taking or beginning to take a simvastatin dose-limiting or contraindicated
in-teracting medication.
e. Omega-3-acid ethyl esters (Lovaza®) is the first FDA-approved omega-3 fatty acid, which
is indicated for patients with elevated levels of triglycerides ( 500 mg/dL).
(1) Mechanism of action. Although not entirely worked out yet, may work by one of
several mechanisms, including an increased hepatic beta-oxidation, a reduction in the
hepatic synthesis of triglycerides, or an increase in plasma lipoprotein lipase activity.
(2) Available product is supplied as a 1-g capsule; the recommended daily dose is four
capsules by mouth daily in one to two doses.
(3) Side effects that have been reported include burping, infection, flu-like symptoms,
upset stomach, change in one’s sense of taste, back pain, and rash.
f. Hypertension. Treatment of hypertension according to the Joint National Conference VII
guidelines has received a class I recommendation based on data from multiple randomized
clinical trials with large numbers of patients (A, high) and should be controlled. Class I rec-
ommendations are based on evidence or general agreement that a given procedure or treat-
ment is useful and effective (see Chapter 33).
g. Smoking should be stopped if at all possible and has received a class I recommendation
based on data derived from a limited number of randomized trials with small numbers of
patients (B, intermediate).
(1) Transdermal use of nicotine-containing patches has become one strategy for aiding the
cessation of smoking. Products such as Nicotrol®, NicoDerm®, and others are
available in varying strengths both as prescription and over-the-counter treatments, in
order to wean patients off the use of cigarettes over an 8- to 12-week period, using
descending doses.
(2) Nicotine gum (oral nicotine polacrilex chewing pieces) is available in 2- or 4-mg pieces
as an over-the-counter product. Nicorette® is usually used for 3 months to aid in
cessation of smoking.
(3) Bupropion is a prescription antidepressant, which is also marketed under the brand
name of Zyban® as an aid to smoking cessation.
(4) Varenicline (Chantix®) is an approved product, which works as a partial neuronal 4- 2
nicotinic receptor agonist in the brain to reduce the craving for nicotine. Therapy is
started with a dose of 0.5 mg by mouth daily for 3 days, followed by 0.5 mg tablets
twice daily for 4 days, then 1 mg twice daily thereafter for 12 weeks, which can be
given for an additional 12 weeks if successful.
h. Obesity should be reduced through diet and an appropriate exercise program in patients
with hypertension, hyperlipidemia, or diabetes mellitus and has received a class I
recommendation based on expert consensus as the primary basis (C, low).
H. Therapeutic agents
1. Recent evidence-based guidelines have provided recommendations for the treatment of patients
with chronic stable angina. (See I.D.1; Fraker et al.). Recommendations use the following
levels of rankings:
Estimate of Certainty
for Treatment Effect Extent of Populations Studied Types of Trials Used
sulfhydryl radicals. However, practical considerations suggest that less frequent administra-
tion (8 to 12 hrs of nitrate-free intervals) is effective without introducing additional agents.
3. -Adrenergic blockers. Based on the 2007 Focused Guidelines for Patients With Chronic
Stable Angina, a class 1a recommendation states that it is beneficial to start and continue -
blocker therapy indefinitely in all patients who have had MI, acute coronary syndrome, or left
ventricular dysfunction with or without heart failure symptoms, unless contraindicated.
a. Mechanism of action. -Blockers reduce oxygen demand, both at rest and during exertion, by de-
creasing the heart rate and myocardial contractility, which also decreases arterial blood pressure.
b. Indications
(1) These agents reduce the frequency and severity of exertional angina that is not
controlled by nitrates.
(2) Nitrates have been combined with calcium antagonists, when slow-release dihydropyr-
idines (e.g., felodipine [Plendil®], amlodipine [Norvasc®]) are preferred over
diltiazem (Cardizem®) or verapamil (Calan®). If patients need to receive a -adrenergic
blocker along with verapamil or diltiazem owing to the added effects, they have the
potential to induce bradycardia, AV heart block, and fatigue.
c. Precautions and monitoring effects
(1) Doses should be increased until the anginal episodes have been reduced or until unac-
ceptable side effects occur.
(2) -Blockers should be avoided in Prinzmetal angina (caused by coronary vasospasm)
because they increase coronary resistance and may induce vasospasm.
(3) Asthma is a relative contraindication because all -blockers increase airway resistance
and have the potential to induce bronchospasm in susceptible patients.
(4) Patients with diabetes and others predisposed to hypoglycemia should be warned that -
blockers mask tachycardia, which is a key sign of developing hypoglycemia.
(5) Patients should be monitored for excessive negative inotropic effects. Findings such as
fatigue, shortness of breath, edema, and paroxysmal nocturnal dyspnea may signal devel-
oping cardiac decompensation, which also increases the metabolic demands of the heart.
(6) Sudden cessation of -blocker therapy may trigger a withdrawal syndrome that can
exacerbate anginal attacks (especially in patients with IHD) or cause MI.
d. Choice of preparations. All -blockers are likely to be equally effective for stable
(exertional) angina. For further review of -adrenergic blockers, see Chapter 33-
Hypertension. For a list of -adrenergic blockers, see Table 33-4, and for complete
descriptions for each -adrenergic blocker, see Chapter 33: III.B.3.a.
4. Calcium-channel blockers
a. Mechanism of action. Two actions are most pertinent in the treatment of angina.
(1) These agents prevent and reverse coronary spasm by inhibiting calcium influx into
vascular smooth muscle and myocardial muscle. This results in increased blood flow,
which enhances myocardial oxygen supply.
(2) Calcium-channel blockers decrease coronary vascular resistance and increase coronary
blood flow, resulting in increased oxygen supply.
(3) Calcium-channel blockers decrease systemic vascular resistance and arterial pressure; in
addition, they decrease inotropic effects, resulting in decreased myocardial oxygen demand.
b. Indications
(1) Calcium-channel blockers are used in stable (exertional) angina that is not controlled
by nitrates and -blockers and in patients for whom -blocker therapy is inadvisable.
Combination therapy—with nitrates, -blockers, or both—may be most effective.
(2) These agents, alone or with a nitrate, are particularly valuable in the treatment of Prinzmetal
angina. They are considered the drug of choice in treatment of angina at rest.
c. Individual agents
(1) Diltiazem (Cardizem®) and verapamil (Calan®)
(a) These drugs produce negative inotropic effects, and patients must be monitored closely for
signs of developing cardiac decompensation (i.e., fatigue, shortness of breath, edema,
paroxysmal nocturnal dyspnea). When coadministered with -blockers or other agents that
produce negative inotropic effects (e.g., disopyramide [Norpace®], quinidine Vari-ous,
procainamide [Pronestyl®], flecainide [Tambocor®]), the negative effects are additive.
574 Chapter 31 II. H
Nitrates
-Adrenergic blockers
Propranolol (Inderal®) 20–80 mg twice daily Possesses both 1- and 2-blocker effects
Metoprolol (Lopressor®) 50–200 mg twice daily Possesses 1-blocker effects
Atenolol (Tenormin®) 50–200 mg/day Possesses 1-blocker effects
Nadolol (Corgard®) 40–80 mg/day Possesses both 1- and 2-blocker effects
Timolol 10 mg twice daily Possesses both 1- and 2-blocker effects
Acebutolol (Sectral®) 200–600 mg twice daily Possesses 1-blocker effects
Betaxolol (Kerlone®) 10–20 mg/day Possesses 1-blocker effects
Bisoprolol (Zebeta®) 10 mg/day Possesses 1-blocker effects
Esmolol (intravenous) 50–300 mcg/kg/min Possesses 1-blocker effects
(Brevibloc ®)
Labetalol (Trandate®, Various) 200–600 mg twice daily Possesses both 1-, 1-, and 2-blocker
effects
Pindolol (Various) 2.5–7.5 mg three times daily Possesses both 1- and 2-blocker effects
Carvedilol (Coreg®) 25 mg twice daily Possesses both 1- 1-, and 2-blocker
effects
Penbutolol (Levatol® ) Possesses both 1- and 2-blocker effects
Calcium-channel blockers
Dihydropyridine derivatives
Nifedipine (Procardia®) Immediate-release; Short duration of action of 4–6 hrs
30–90 mg daily
Amlodipine (Norvasc®, Various) 5–10 mg once daily Long duration of action
Felodipine (Plendil®) 5–10 mg once daily Long duration of action
Isradipine (Dynacirc®) 2.5–10 mg twice daily Intermediate duration of action
Nicardipine (Cardene®) 20–40 mg three times daily Short duration of action
Nisoldipine (Sular®) 20–40 mg once daily Short duration of action
Coronary Artery Disease 575
Miscellaneous
(b) Patients should be monitored for signs of developing bradyarrhythmias and heart
block because these agents have negative chronotropic effects.
(c) Verapamil (Calan®) frequently causes constipation that must be treated as needed
to prevent straining at stool, which could cause an increased oxygen demand
(Valsalva maneuver). Verapamil is not recommended in patients with sick sinus
syndromes, AV nodal disease, or heart failure (HF).
(2) Nifedipine (Procardia®)
(a) This calcium-channel blocker is believed to possess the greatest degree of negative
inotropic effects compared to the newer second-generation members of this group,
amlodipine (Norvasc®) and felodipine (Plendil®). Nifedipine 10 mg (chewed or
swal-lowed) has been used to treat Prinzmetal angina or refractory spasm in
patients who are not hypotensive. Controversy still exists about the use of short-
acting, rapid-release agents such as nifedipine in patients with IHD.
(b) Because nifedipine increases the heart rate somewhat, it can produce tachycardia,
which would increase oxygen demand. Coadministration of a -blocker should pre-
vent reflex tachycardia.
(c) Its potent peripheral dilatory effects can decrease coronary perfusion and produce
excessive hypotension, which can aggravate myocardial ischemia.
(d) Dizziness, light-headedness, and lower extremity edema are the most common
adverse effects, but these tend to disappear with time or dose adjustment.
(3) Amlodipine (Norvasc®), clevidipine (Cleviprex®), felodipine (Plendil®),
isradipine (Dynacirc®), nicardipine (Cardene®), and nisoldipine (Sular®) are
second-generation dihydropyridine derivative, calcium-channel blockers. They have
been used effectively as once- or twice-a-day agents owing to their long activity.
Because of the potent negative inotropic effects of these agents, they are not
recommended in patients with HF (amlo-dipine has been shown to have less negative
potential in HF than other members of the class). Clevidipine is available in the
injectable form only and is administered as a con-tinuous slow IV infusion.
5. Antiplatelet agents
a. Aspirin. Based on the 2007 Focused Guidelines for Patients With Chronic Stable Angina, a
class Ia recommendation states that aspirin should be started at 75 to 162 mg/day and
contin-ued indefinitely in all patients unless contraindicated.
b. Ticlopidine (Ticlid®) is a thienopyridine derivative that inhibits platelet aggregation induced by
adenosine diphosphate. However, unlike aspirin, it has not been shown to decrease adverse
576 Chapter 31 II. H
cardiovascular events in patients with stable angina and has been associated with thrombotic
thrombocytopenic purpura on an infrequent basis.
c. Clopidogrel (Plavix®) is also a thienopyridine derivative related to ticlopidine, but it possesses
antithrombotic effects that are greater than those of ticlopidine. Clopidogrel is a therapeutic
option in those angina patients who cannot take aspirin because of contraindications. Doses of 75
mg daily are recommended to prevent the development of acute coronary syndromes.
(1) Recently there has been added concern regarding the interaction which takes place when
patients receiving clopidogrel are also given one of the proton pump inhibitors (PPIs), such
as omeprazole (Nexium®). The American College of Cardiology Foundation, in conjunc-
tion with the American College of Gastroenterology and the American Heart Association,
has issued a consensus document regarding the concomitant use of PPIs and thienopyri-
dines, specifically clopidogrel. The following highlighted recommendations are discussed
within the consensus statement as they relate to the combinations of clopidogrel and PPIs:
(a) PPIs are appropriate in patients with multiple risk factors for GI bleeding who are
also receiving antiplatelet therapy (e.g., clopidogrel).
(b) Although pharmacokinetic and pharmacodynamic studies have demonstrated vary-ing
effects of PPIs on the extent of clopidogrel metabolic conversion to the active
metabolite, no evidence has established clinically meaningful differences in outcomes.
(c) A clinically significant interaction cannot be excluded in subgroups who are poor
metabolizers of clopidogrel.
(d) Until solid evidence exists to support staggering PPIs with clopidogrel, the dosing
of PPIs should not be altered.
d. Based on the 2007 Focused Guidelines for Patients With Chronic Stable Angina, a class Ib
recommendation states that the use of warfarin in conjunction with aspirin and/or clopido-
grel is associated with an increased risk of bleeding and should be monitored closely.
6. ACE inhibitors. Based on the 2007 Focused Guidelines for Patients With Chronic Stable
Angina, the following recommendations have been made for ACE inhibitors.
a. Class 1a recommendation states that ACE inhibitors should be started and continued indefi-
nitely in all patients with left ventricular ejection fraction 40% and in those with hyperten-
sion, diabetes, or chronic kidney disease unless contraindicated.
b. Class Ib recommendation that ACE inhibitors should be started and continued indefinitely
in patients who are not lower risk (lower risk defined as those with normal left ventricular
ejec-tion fraction in whom cardiovascular risk factors are well controlled and
revascularization has been performed), unless contraindicated.
c. Class IIa recommendation that it is reasonable to use ACE inhibitors among lower risk pa-
tients with mildly reduced or normal left ventricular ejection fraction in whom
cardiovascular risk factors are well controlled and revascularization has been performed.
d. Current guidelines do not suggest which agent to use, and it is anticipated that ongoing trials
with additional agents will provide additional information regarding dosing regimens and
potential differences that might exist among the class of drugs.
7. Angiotensin receptor blockers (ARBs). Based on the 2007 Focused Guidelines for Patients
With Chronic Stable Angina, three new recommendations have been made for the use of ARBs
in pa-tients with chronic stable angina.
a. Class Ia recommendation that ARBs are recommended for patients who have hypertension,
have indications for but are intolerant of ACE inhibitors, have heart failure, or have had a
myocardial infarction with left ventricular ejection fraction 40%.
b. Class IIb recommendation that ARBs may be considered in combination with ACE
inhibitors for heart failure due to left ventricular systolic dysfunction.
c. Class Ia recommendation that aldosterone blockade is recommended for use in post-MI
patients without significant renal dysfunction or hyperkalemia who are already receiving
therapeutic doses of an ACE inhibitor and a -blocker, have a left ventricular ejection
fraction 40%, and have either diabetes or heart failure.
8. Chelation therapy. Based on the 2007 Focused Guidelines for Patients With Chronic Stable Angina, a
class IIIc recommendation stated that chelation therapy (intravenous infusions of ethylenediami-
netetraacetic acid or EDTA) is not recommended for the treatment of chronic angina or arterioscle-rotic
cardiovascular disease and may be harmful because of its potential to cause hypocalcemia.
Coronary Artery Disease 577
Figure 31-2. Evolutionary progression of ACSs. As atherosclerosis (most common cause of ischemic heart
disease) advances, the reduction in myocardial perfusion results in the development of the ACS owing to
unstable angina, NSTEMI, or STEMI. The thrombi, which form in unstable angina, NSTEMI, and STEMI, are
rich in both fibrin and platelets. Adapted from Vanscoy G. Integrating new fi brinolytic fi ndings into AMI
reperfusion and combination therapy: 2002 and beyond. Paper presented at the meeting of the Delaware
Valley Chapter of the Pennsylvania Society of Health-Systems Pharmacists; March 7, 2002. STEMI, ST-
segment elevation; NSTEMI, non–ST-segment elevation.
578 Chapter 31 III. D
are considered to have either UA or NSTEMI; the final diagnosis is made later, after the presence
or absence of serial cardiac markers is determined.
1. Diagnostic test results. The development of an ACS is a life-threatening emergency; diagnosis
is presumed—and treatment is instituted—based on the patient’s complaints and the results of
an immediate 12-lead EKG/ECG. Laboratory tests and further diagnostic tests can rule out or
provide confirmation and help identify the locale and extent of myocardial damage.
2. Serial 12-lead EKG. Abnormalities may be absent or inconclusive during the first few hours
after presentation of the ACS and may not aid the diagnosis in about 15% of the cases. When
present, characteristic findings show progressive changes.
a. First, ST-segment elevation (injury current) appears in the leads, reflecting the injured area.
Peaked upright or inverted T waves usually indicate acute myocardial injury, the early stages of
a transmural Q-wave MI. Persistent ST depression may also indicate a non–Q-wave MI.
b. Q waves developing (indicating necrosis) is generally diagnostic of an MI but can be seen
in other conditions.
c. Unequivocal diagnosis can be made only in the presence of all three abnormalities.
However, the manifestations depend on the area of injury. For example, in non–Q-wave
infarction, only ST-segment depression may appear.
d. The most serious arrhythmic complication of an AMI is ventricular fibrillation, which may
occur without warning.
e. Ventricular premature beats (VPBs) are the most commonly encountered arrhythmias and
may require treatment.
3. Cardiac enzymes
a. Creatine kinase–heart muscle (CK-MB) is first elevated 3 to 12 hrs after the onset of pain,
peaks in 24 hrs, and returns to baseline in 48 to 72 hrs. Other conditions elevate the CK-MB
enzyme but do not demonstrate the typical pattern of rise and fall as seen in an MI. Until
recently, CK-MB had been the principal serum cardiac marker used in the evaluation of ACS.
b. Cardiac troponin I (cTnI) and cardiac troponin T (cTnT) are even more sensitive than
CK-MB. They represent a powerful tool for risk stratification and have greater sensitivity
and specificity than CK-MB. However, they do provide a low sensitivity in the early phases
of an MI ( 6 hrs after symptom onset) and require repeat measurements at 12 to 16 hrs, if
negative. Levels increase 3 to 12 hrs after the onset of pain, peak at 24 to 48 hrs, and return
to baseline over 5 to 14 days.
c. Lactate dehydrogenase (LDH) is followed for its characteristic patterns of rise and fall.
The ratio of LDH1:LDH2 is helpful in diagnosing an MI. LDH assays are being replaced by
cTnT assays.
4. Cardiac imaging. As cardiac enzyme assays improve, the use of noninvasive cardiac imaging
techniques are not indicated for initial diagnosis of an MI. Tests include 99mTc-pyrophosphate
scintigraphy, myocardial perfusion imaging, radionucleotide ventriculography, two-
dimensional echocardiography, and coronary angiography.
E. Signs and symptoms
1. Recent evidence-based clinical guidelines indicate a class I recommendation for patients with
suspected ACS with chest discomfort at rest for longer than 20 mins; hemodynamic instability or
recent syncope or presyncope should be strongly considered for immediate referral to an emer-gency
department or specialized chest pain unit. The foremost characteristic of ACS is persistent, severe
chest pain or pressure, commonly described as crushing, squeezing, or heavy (likened to having an
elephant sitting on the chest). The pain generally begins in the chest and, like angina, may radiate to
the left arm, the abdomen, back, neck, jaw, or teeth. The onset of pain generally occurs at rest or
with normal daily activities; it is not commonly associated with exertion.
2. Other common complaints include a sense of impending doom, sweating, nausea, vomiting,
and difficulty breathing. In some patients, fainting and sudden death may be the initial
presentation of ACS.
3. Observable findings include extreme anxiety, restless, agitated behavior, and ashen pallor.
4. Some patients, particularly those with diabetes or the elderly, may experience only mild or
indigestion-like pain or a clinically silent MI, which may only manifest in worsening heart fail-
ure, loss of consciousness, acute confusion, dyspnea, a sudden drop in blood pressure, or a
lethal arrhythmia.
Coronary Artery Disease 579
Oral antiplatelets/anticoagulants
Aspirin* For acute STEMI patients:
162 mg should be chewed by patients who have not taken aspirin before presentation with STEMI. Class I
For all post-PCI STEMI stented patients:
1. 162–325 mg daily for at least 1 month after bare-metal stent implantation, 3 months after Class IIa
sirolimus-eluting stent, and 6 months after paclitaxel-eluting stent, and then indefi nitely at
doses of 75–132 mg daily.
2. In patients where there is concern of bleeding, 75–162 mg (lower dose) aspirin is Class IIa
reasonable during the initial period after stent implantation.
Clopidogrel (Plavix) 1. 75 mg orally daily added to ASA in STEMI patients. Class I-Post-STEMI patients
2. Treatment should be at least 14 days. Class I-Post-STEMI patients
3. Loading dose of 300 mg orally (patients 75 years who receive fi brinolytic therapy or who do Class IIa-Post-STEMI patients
not receive reperfusion therapy)
4. 75 mg orally per day, long-term maintenance therapy (e.g., 1 year) (regardless of whether Class IIa-Post-STEMI patients
they undergo reperfusion with fi brinolytic therapy).
For all post-PCI STEMI stented patients:
5. Receive a drug-eluting stent (DES), clopidogrel 75 mg daily should be given for at least 12 Class I
months if patients are not at high risk of bleeding.
6. Receive a bare metal stent (BMS), clopidogrel should be given for a minimum of 1 month Class I
and ideally up to 12 months (unless the patient is at increased risk of bleeding; then it should
be given for a minimum of 2 weeks).
7. For patients taking clopidogrel for whom CABG is planned, if possible, the drug should be Class I
withheld for at least 5 days, and preferably for 7, unless the urgency for revascularization
outweighs the risks of bleeding.
Warfarin 1. Managing warfarin to INR 2.0 to 3.0 in post-STEMI patients. Use of warfarin in conjunction Class I-Post-STEMI patients
with aspirin and/or clopidogrel is associated with increased risk of bleeding and should be
monitored closely.
2. In patients requiring warfarin, clopidogrel, and aspirin therapy, an INR of 2.0 to 2.5 is recommended with low-dose
aspirin (75–81 mg) and a 75 mg dose of clopidogrel.
Coronary
Parenteral antiplatelets/anticoagulants
Artery Disease
Heparin (UFH) 1. Bolus of 60 U/kg, maximum 4000 U IV followed by an initial infusion 12 U/kg/hr, Class I-Post-STEMI
(maximum of 1000 U/hr) in patient at high risk for systemic emboli (large or anterior MI,
atrial fibrillation, previous embolus, known LV thrombus, or cardiogenic shock.
2. IV or SQ UFH or with subcutaneous LMWH for at least 48 hrs. In patients whose clinical Class IIa-Post-STEMI and not undergoing
condition necessitates prolonged bed rest and/or minimized activities, it is reasonable that reperfusion, and no contraindications.
treatment be continued until the patient is ambulatory.
581
582
Table 31-5 Continued.
Chapter 31
3. SQ UFH, 7,500 units–12,500 units twice daily for prophylaxis for deep venous thrombosis Class IIb
(DVT) until completely ambulatory, may be useful, but the effectiveness of such a strategy is
not well established in the contemporary era of routine aspirin use and early mobilization.
For STEMI patients receiving PCI:
4. Bolus of 70–100 U/kg, and maintenance to target 1.5–2.0 times aPTT, if no GP IIb/IIIa Class I
previously given. However, if a GP IIb/ IIIa has been given previously, 50–70 U/kg bolus, and
maintenance to target, as above.
III. H
Enoxaparin (Lovenox) Patients undergoing reperfusion with fibrinolytics:
For patients younger than 75 years of age:
1. 30 mg intravenous bolus, followed in 15 mins by 1.0 mg/kg SQ every 12 hrs (serum Class I-Post-STEMI
creatinine 2.5 mg/dL in men and 2.0 mg/dL in women).
For patients 75 years of age,
2. 0.75 mg/kg SQ every 12 hrs. (No loading dose). Maintenance doses with enoxaparin should Class I-Post-STEMI
continue for the duration of the index hospitalization, up to 8 days. Regardless of age, if the
creatinine clearance (using the Cockcroft-Gault formula) during the course of treatment is
estimated to be less than 30 mL/min, the subcutaneous regzimen is 1.0 mg/kg every 24 hrs.
Fondaparinux (Arixtra) Patients undergoing reperfusion with fibrinolytics:
1. 2.5 mg IV, followed by 2.5 mg SQ once daily. (serum creatinine is less than 3.0 mg/dL) Class I-Post-STEMI
2. Maintenance doses with fondaparinux should be continued for the duration of the index Class I-Post-STEMI
hospitalization, up to 8 days.
Bivalirudin (Angiomax) Patients undergoing PCI:
1. Initial IV bolus dose of 0.75 mg/kg, followed by 1.75 mg/kg/hr infusion for the duration of the Class I-Post-STEMI
procedure.
Abciximab (ReoPro) 0.25 mg/kg IV bolus followed by infusion of 0.125 mcg/kg/min for 12–24 hrs Class IIa, for use in addition to aspirin and heparin
in STEMI patients for whom catheterization and
PCI are planned just before PCI.
Class III, in STEMI patients for whom PCI is
not planned.
Eptifi batide (Integrilin) 180 mcg/kg IV bolus followed by infusion of 2.0 mcg/kg/min for 72–96 hrs. Class I, for use in addition to aspirin and heparin
Tirofiban (Aggrestat) 0.4 mcg/kg/min for 30 mins followed by infusion of 0.1 mcg/kg/min for 48–96 hrs. in STEMI patients for whom catheterization and
PCI are planned, and in patients just before PCI.
Class IIa, in addition to aspirin and a LMWH or
UFH in STEMI patients without continuing
ischemia who have no other high-risk features
and for whom PCI is not planned.
Class IIb, in addition to aspirin and a LMWH or
UFH in STEMI patients without continuing
ischemia who have no other high-risk features
and for whom PCI is not planned.
2. When the lesion ruptures, it triggers the release of adenosine diphosphate (ADP), serotonins,
and thromboxane A2, which leads to platelet aggregation and the formation of the primary clot.
Thromboplastin, released from the injured vessel initiates the clotting cascade, and the result-
ing fibrin traps red blood cells (RBCs), platelets, and plasma protein to form an intraluminal
thrombus. The subsequent clot dissolution is caused by the conversion of plasminogen to plas-
min, which is mediated by plasminogen activators.
3. According to updated management guidelines (see I.D.6.), all patients with STEMI should
receive either primary PCI within 90 mins of first medical contact (class Ia recommendation) or
fibrino-lytic therapy within 30 mins of hospital presentation if they cannot be transferred to a
PCI center and undergo PCI within 90 mins of first medical contact, unless fibrinolytic therapy
is contrain-dicated (class Ib recommendation).
4. It is beyond the realm of this text to expand on the clinical implications for primary PCI, facilitat-ed
PCI, and rescue PCI, as described in the 2009 Focused Updates: ACC/AHA Guidelines for the
Management of Patients With ST-Elevation Myocardial Infarction (Updating the 2004 Guideline
and 2007 Focused Update) and ACC/AHA/SCAI Guidelines on Percutaneous Coronary Inter-
vention (Updating the 2005 Guideline and 2007 Focused Update). Consequently, the review will
focus strictly on the pharmacologic agents recommended for use in the STEMI patient popula-tion,
which include fibrinolytics, analgesics, anticoagulants, thienopyridines, antiplatelets, ACE
inhibitors, ARBs, and -adrenergic receptor blockers as either primary treatment or secondary
preventive and long-term management, as discussed in the guidelines.
a. Fibrinolytics. Administration of thrombolytic agents causes the thrombus clot to be lysed
when administered early after symptom onset ( 6 to 12 hrs) and to restore blood flow. The
conversion of plasminogen to plasmin promotes fibrinolysis and breakdown of the clot.
(1) Indications
(a) Thrombolytic agents were used in patients with STEMI with chest pain 6 to 12 hrs. Suc-
cessful early reperfusion has been shown to reduce infarct size, improve ventricular func-
tion, and improve mortality. However, benefits may be seen in patients using thrombolytic
therapy as late as 12 hrs after pain starts. Therapy of choice when PCI is not available.
(b) Intravenous administration of a recombinant tissue-plasminogen activator (t-PA)
such as alteplase (Activase®), a recombinant plasminogen activator (r-PA) such as
re-teplase (Retavase®), or tenecteplase (Tnkase®), may restore blood flow in an
occluded artery if administered within 12 hrs of an AMI, although 6 hrs is optimal.
The goal of treatment of STEMI patients is to initiate thrombolytic therapy within
30 to 60 mins of arrival in an emergency room.
(i) t-PA is relatively fibrin specific and is able to lyse clots without depleting fi
brinogen, and Tnkase has a greater fibrin specificity.
(ii) Most studies have shown that each agent, when used early, can reopen
(reperfuse) occluded coronary arteries and reduce mortality by up to 30%
from STEMI. However, considerations such as ease of use, market
availability, onset of action, incidence of bleed, and cost are important factors
in determining which agent to use for a given hospital and patient.
(2) Individual agents
(a) Alteplase (Activase®)
(i) Absolute contraindications to t-PA include active internal bleeding; recent
cerebrovascular accident (CVA); intracranial neoplasm; aneurysm;
pregnancy; arteriovenous malformations; recent (within 2 months)
intracranial surgery, spinal surgery, or trauma; and severe uncontrolled
hypertension, bleeding dia-thesis, or hemorrhagic ophthalmic conditions.
(ii) A front-loaded regimen—an accelerated infusion that consists of a total dose of
100 mg or less that is given over 1.5 hr—may be more beneficial. The initial dose
of 15 mg is given as an IV bolus, 1 to 2 mins, while an infusion is begun to
(a) Infuse t-PA at the rate of 0.75 mg/kg over 30 mins (not to exceed 50 mg)
(b) Followed by t-PA infused at 0.5 mg/kg over 60 mins (not to exceed 35 mg)
(iii) An alternate dosing regimen is based on the patient’s weight.
(a) Dosage for patients 65 kg. A total of 100 mg of t-PA is generally admin-
istered to all patients who weigh 65 kg over a 3-hr period. Although many
584 Chapter 31 III. H
(ii) Patients who receive anticoagulants for more than 48 hrs should receive an
agent other than unfractionated heparin (UFH) due to the increased risk of
heparin-induced thrombocytopenia with its prolonged use (class Ia).
(iii) Anticoagulant regimens with established efficacy in STEMI include:
— UFH. Initial IV bolus of 60 units/kg (maximum 4000 units) followed by
an IV infusion of 12 units/kg/hr (maximum of 1000 units/hr) initially.
The dose should be adjusted to maintain an activated partial
thromboplastin time (aPTT) of 1.5 to 2 times control (class Ic).
— Enoxaparin (Lovenox®). An initial 30 mg IV bolus, followed in 15 mins by
a subcutaneous injection of 1 mg/kg every 12 hrs (assuming serum cre-
atinine is less than 2.5 mg/dL in men, 2.0 mg/dL in women in patients
younger than 75 years of age). For patients 75 years of age and older, the IV
bolus dose is eliminated, and the patient is given a subcutaneous dose of 0.75
mg/kg every 12 hrs. CAUTION: In all patients, if the creatinine clear-ance is
estimated to be less than 30 mL/min, the dosing regimen should be changed
to 1 mg/kg every 24 hrs. The current guidelines recommend the use of
maintenance dosing of enoxaparin for the duration of hospitaliza-tion, up to a
maximum of 8 days (class Ia).
— Fondaparinux (Arixtra®). An initial dose of 2.5 mg intravenously and
subsequent subcutaneous injections of 2.5 mg given once daily
(assuming serum creatinine is less than 3.0 mg/dL). The current
guidelines, as in the case of enoxaparin, recommend the use of
maintenance dosing of fondaparinux for the duration of hospitalization,
up to a maximum of 8 days (class Ib).
(iv) Anticoagulants have also been shown to be effective in STEMI patients prior
to undergoing PCI, with the following dosing recommendations:
— UFH. For prior treatment, administer additional boluses of UFH as
needed to support the procedure, but take into account whether other
agents such as GP IIb/IIIa receptor antagonists (class Ic). Bivalirudin
(Angiomax®) can also be used in patients previously treated with UFH
(class Ic).
— Enoxaparin (Lovenox®). For prior treatment, if the last subcutaneous
dose of enoxaparin was given within the previous 8 hrs, no additional
drug is needed; however, if the last dose was administered 8 to 12 hrs
earlier, an intravenous dose of 0.3 mg/kg should be given (class Ib).
— Bivalirudin (Angiomax®). With the most recent national guidelines up-
dated for STEMI/PCI, bivalirudin received a class I, level b
recommenda-tion as useful supportive measure for primary PCI with or
without prior treatment with UFH. Bivalirudin has also received a class
IIa, level b rec-ommendation in STEMI patients undergoing PCI who are
at a high risk of bleeding due to its relatively low risk of bleeding
compared to other anticoagulants.
(v) Anticoagulants (non-UFH regimens) have also been recommended for
patients with STEMI who do not undergo reperfusion therapy for the duration
of the initial hospitalization (class IIa). Dosing regimens that can be used
include enoxaparin or fondaparinux in the same dosing regimens as in those
patients receiving fibrinolytic (see iv previously).
(e) Thienopyridines
(i) Clopidogrel (Plavix®) should be added to aspirin in STEMI patients
regardless of whether they undergo reperfusion with fibrinolytics or do not
receive re-perfusion (class Ib). Doses of 75 mg by mouth daily should be
administered. Treatment should continue for at least 14 days (class Ib).
(ii) Prasugrel (Effient®) 60 mg should be given as soon as possible prior to PCI,
if clopidogrel is not being used (class Ib). Prasugrel should be given in those
patients not receiving clopidogrel within 1 hr of PCI (class Ib).
586 Chapter 31 III. H
Study
Questions
Directions for questions 1–16: Each of the questions,
statements, or incomplete statements in this section can
be correctly answered or completed by one of the
suggested answers or phrases. Choose the best answer.
1. Exertion-induced angina, which is relieved by
rest, nitroglycerin, or both, is referred to as
(A) Prinzmetal angina.
(B) unstable angina.
(C) stable angina.
(D) variant angina.
2. Myocardial oxygen demand is increased by all of
the following factors except
(A) exercise.
(B) smoking.
(C) cold temperatures.
(D) isoproterenol.
(E) metoprolol.
588 Chapter 31 6. Which of the following is considered a component
of acute coronary syndrome (ACS)?
I. unstable angina
3. Which of the following agents used in
II. non–ST-segment elevated myocardial
Prinzmetal angina has spasmolytic actions,
infarction (NSTEMI)
which increase coronary blood supply? III. ST-segment elevated myocardial
(A) nitroglycerin infarction (STEMI)
(B) diltiazem
Directions for questions 7–11: Each of the following
(C) timolol
descriptions is most closely related to one of the following
(D) isosorbide mononitrate
(E) propranolol drugs. The descriptions may be used more than once or
not at all. Choose the best answer, A–E.
4. T he development of ischemic pain occurs when the
(A) Inhibition of intestinal absorption of cholesterol
demand for oxygen exceeds the supply.
Determinants of oxygen demand include all of the (B) Lowering of low-density lipoproteins
following choices except which one? (LDLs), triglycerides, and increased high-
density lipoprotein (HDL) along with
(A) contractile state of the heart
potential anti-inflammatory effects
(B) myocardial ejection time (C) Recommendations for this agent have been
(C) left ventricular volume substantially expanded beyond an alternative for
(D) right atrial pressure aspirin-intolerant patients due to recent trials
(E) systolic pressure demonstrating its benefit in select ACS patients.
5. Myopathy is an adverse effect of all the (D) Recommended for ACS patients who
following agents except cannot tolerate aspirin
(A) lovastatin. (E) Recommended over unfractionated heparin
(B) simvastatin. (UFH) as an anticoagulant in patients with
(C) pravastatin. unstable angina (UA) or NSTEMI
(D) gemfibrozil. 7. tirofiban (Aggrastat®)
(E) colestipol.
8. enoxaparin (Lovenox®)
Directions for question 6 : The question can be correctly
answered by one or more of the suggested answers. 9. simvastatin (Various)
Choose the answer, A–E.
10. clopidogrel (Plavix®)
(A) if I only is correct
(B) if III only is correct 11. ezetimibe (Zetia®)
(C) if I and II are correct
(D) if II and III are correct
(E) if I, II, and III are correct
I. INTRODUCTION. Sudden death from cardiac causes in the United States has been estimated to
occur in the range of 300,000 to 350,000 cases annually. Depending on the definition, sudden cardiac
death could be classified as low as 13% of all natural deaths up to approximately 50% of all deaths
from cardiovascular causes occurring shortly after onset (instantaneous to 1 hr), with the majority of
sudden deaths being caused by acute ventricular tachyarrhythmias. These incidence reports might
appear to create greater need for the knowledge necessary to appropriately use antiarrhythmics for this
high-risk patient population. However, previously conducted studies have cast doubt on the true place
of antiarrhythmics in the treatment and prevention of cardiac arrhythmias. Studies such as the Cardiac
Arrhythmia Suppression Trial (CAST) have demonstrated that certain classes of antiarrhythmics
increased mortality in patients treated with antiarrhythmics as compared to placebo. Since the release
of the data from the CAST trial, subsequent studies have confirmed the finding that certain
antiarrhythmics do possess “proarrhythmic” effects when used injudiciously. Consequently, the use of
trial and error to determine antiarrhythmic therapy has given way to an era of outcome-based
antiarrhythmic drug decision making. By understanding the causes of arrhythmias and being aware of
drug–drug and drug– target interactions, we are more likely to understand the key considerations to
maximize therapeutic strategies while minimizing drug-induced toxicities.
A. Definition. Cardiac arrhythmias are deviations from the normal heartbeat pattern. They include
abnormalities of impulse formation, such as heart rate, rhythm, or site of impulse ori-gin and
conduction disturbances, which disrupt the normal sequence of atrial and ventricular activation.
B. Electrophysiology
1. Conduction system
a. Two electrical sequences that cause the heart chambers to fill with blood and contract are
initiated by the conduction system of the heart.
(1) Impulse formation, the first sequence, takes place when an electrical impulse is gener-
ated automatically.
(2) Impulse transmission, the second sequence, occurs once the impulse has been gener-
ated, signaling the heart to contract.
b. Four main structures composed of tissue that can generate or conduct electrical impulses
make up the conduction system of the heart.
(1) The sinoatrial (SA) node in the wall of the right atrium contains cells that
spontaneously initiate an action potential. Serving as the main pacemaker of the heart,
the SA node initi-ates 60 to 100 beats/min.
(a) Impulses generated by the SA node trigger atrial contraction.
(b) Impulses travel through internodal tracts—the anterior tract, middle tract, posterior
tract, and anterior interatrial tract (Figure 32-1).
(2) At the atrioventricular (AV) node situated in the lower interatrial septum, the
impulses are delayed briefly to permit completion of atrial contraction before
ventricular contrac-tion begins.
(3) At the bundle of His—muscle fibers arising from the AV junction—impulses travel
along the left and right bundle branches, located on either side of the intraventricular
septum.
590
Cardiac Arrhythmias 591
Figure 32-1. Electrical pathways of the heart. AV, atrioventricular; SA, sinoatrial.
(4) The impulses reach the Purkinje fibers, a diffuse network extending from the bundle
branches and ending in the ventricular endocardial surfaces. Ventricular contraction
then occurs.
c. Latent pacemakers. The AV junction, bundle of His, and Purkinje fibers are latent
pacemakers; they contain cells capable of generating impulses. However, these regions have
a slower firing rate than the SA node. Consequently, the SA node predominates except
when it is depressed or injured, which is known as overdrive suppression.
2. Myocardial action potential. Before cardiac contraction can take place, cardiac cells must
depo-larize and repolarize.
a. Depolarization and repolarization result from changes in the electrical potential across the
cell membrane, caused by the exchange of sodium and potassium ions.
b. Action potential, which reflects this electrical activity, has five phases (Figure 32-2).
(1) Phase 0 (rapid depolarization) takes place as sodium ions enter the cell through fast
channels; the cell membrane’s electrical charge changes from negative to positive.
(2) Phase 1 (early rapid repolarization). As fast sodium channels close and potassium
ions leave the cell, the cell rapidly repolarizes (i.e., returns to resting potential).
160 1
2
Transmembrane potential (mV)
140
120
0
3
220
240 0
(3) Phase 2 (plateau). Calcium ions enter the cell through slow channels while potassium
ions exit. As the cell membrane’s electrical activity temporarily stabilizes, the action
potential reaches a plateau (represented by the notch at the beginning of this phase in
Figure 32-2).
(4) Phase 3 (final rapid repolarization). Potassium ions are pumped out of the cell as the
cell rapidly completes repolarization and resumes its initial negativity.
(5) Phase 4 (slow depolarization). The cell returns to its resting state with potassium ions
inside the cell and sodium and calcium ions outside.
c. During both depolarization and repolarization, a cell’s ability to initiate an action potential
varies.
(1) The cell cannot respond to any stimulus during the absolute refractory period (begin-
ning during phase 1 and ending at the start of phase 3).
(2) A cell’s ability to respond to stimuli increases as repolarization continues. During the
relative refractory period, which occurs during phase 3, the cell can respond to a
strong stimulus.
(3) When the cell has been completely repolarized, it can again respond fully to stimuli.
d. Cells in different cardiac regions depolarize at different speeds, depending on whether fast
or slow channels predominate.
(1) Sodium flows through fast channels; calcium flows through slow channels.
(2) Where fast channels dominate (e.g., in cardiac muscle cells), depolarization occurs
quickly. Where slow channels dominate (e.g., in the electrical cells of the SA node and
AV junction), depolarization occurs slowly.
3. Electrocardiography. The electrical activity occurring during depolarization–repolarization
can be transmitted through electrodes attached to the body and transformed by an
electrocardio-graph (EKG/ECG) machine into a series of waveforms (EKG/ECG
waveform). Figure 32-3 shows a normal EKG/ECG waveform.
a. The P wave reflects atrial depolarization.
b. The PR interval represents the spread of the impulse from the atria through the Purkinje
fibers.
c. The QRS complex reflects ventricular depolarization (phase 0).
d. The ST segment represents phase 2 of the action potential—the absolute refractory period
(part of ventricular repolarization).
e. The T wave shows phase 3 of the action potential—ventricular repolarization.
C. Classification. Arrhythmias generally are classified by origin (i.e., supraventricular or ventricular).
1. Supraventricular arrhythmias stem from enhanced automaticity of the SA node (or another
pacemaker region, above the bundle of His or ventricular tissue) or from reentry conduction.
2. Ventricular arrhythmias occur below the bundle of His, when an ectopic (abnormal)
pacemaker triggers a ventricular contraction before the SA node fires (e.g., from a conduction
disturbance or ventricular irritability).
3. Special note
a. Torsades de pointes (TdP; French for “twisting of the points”) has received increased atten-
tion during recent years as a major proarrhythmic event, which has been reported with antiar-
rhythmic drug therapy. It is defined as a polymorphic ventricular tachycardia with a twisting
QRS complex morphology, which sometimes occurs with drugs that prolong ventricular re-
polarization (QT interval widening). Although initial reports of TdP centered around antiar-
rhythmic drugs (quinidine), today, more than 50 drugs, both antiarrhythmic agents and other
classes of drugs, such as antibiotics, have been shown to affect the duration of the QT
interval and have been associated with this arrhythmia.
b. A Web site devoted to providing education and research on drug-induced arrhythmias,
especially those due to prolongation of the QT interval on the electrocardiogram
(EKG/ECG), is available. The site, http://www.torsades.org/medical-pros/drug-lists/drug-
lists.htm, is cur-rently maintained by Dr. Raymond Woosley.
c. Dr. Woosley and colleagues have established the International Registry for Drug-Induced
Arrhythmias, to which one can submit a suspected “drug-induced arrhythmia event.” The
registry also provides a list of drugs reported to prolong the QT interval or cause TdP
(http:// www.Torsades.org). Four drug lists have been created based on the relative risk of
inducing TdP or a prolonged QT interval.
d. It is beyond the intention of this chapter to provide a comprehensive listing, as done on Dr.
Woosley’s Web site. However, the four categories of drug lists include the following with
select examples:
(1) List 1: Drugs that are generally accepted by authorities to carry a risk of causing
TdP; that is, amiodarone (Cordarone®), chlorpromazine (Thorazine®), clarithromycin
(Biaxin®), disopyramide (Norpace®), dofetilide (Tikosyn®), erythromycin
(Erythrocin®), haloperidol (Haldol®), ibutilide (Corvert®), methadone (Dolophine®),
moxifloxacin (Avelox®), pentam-idine (NebuPent®), pimozide (Orap®), procainamide
(Various), quinidine (Quinaglute®), sotalol (Betapace®), and thioridazine (Mellaril®).
(2) List 2: Drugs that in some reports have been associated with TdP and/or QT pro-
longation but at this time lack substantial evidence for causing TdP; that is, aman-
tadine (Symmetrel®), azithromycin (Zithromax®), clozapine (Clozaril®), dolasetron
(Anzemet®), dronedarone (Multaq®), escitalopram (Lexapro®), felbamate
(Felbatol®), flecainide (Tambocor®), foscarnet (Foscavir®), fosphenytoin
(Cerebyx®), granisetron (Kytril®), isradipine (DynaCirc®), levofloxacin (Levaquin®),
nicardipine (Cardene®), ondansetron ( Zofran®), quetiapine (Seroquel®), risperidone
(Risperdal®), sunitinib (Sutent®), tacrolimus (Prograf®), venlafaxine (Effexor®),
voriconazole (Vfend®), and ziprasidone (Geodon®).
(3) List 3: Drugs with a conditional risk of TdP—drugs that carry a risk of TdP and/or
QT prolongation under certain conditions, such as patients with long QT syndrome,
drug overdose, or coadministration of interacting drugs; that is, amitriptyline (Elavil®),
ciprofloxacin (Cipro®), citalopram (Celexa®), clomipramine (Anafranil®), desipramine
(Norpramin®), diphenhydramine (Benadryl®), doxepin (Silenor®), fluconazole
(Diflucan®), fluoxetine (Prozac®), galantamine (Razadyne®), imipramine (Tofranil®),
itraconazole (Sporanox®), ketoconazole (Nizoral®), nortriptyline (Pamelor®), paroxetine
(Paxil®), rito-navir (Norvir®), sertraline (Zoloft®), solifenacin (VESIcare®), trazodone
(Desyrel®), trim-ethoprim-sulfamethoxazole (Bactrim), trimipramine (Surmontil).
(4) List 4: Drugs to be avoided for use in patients with diagnoses or suspected
congeni-tal long QT syndrome. (Drugs on Lists 1, 2, and 3 would also be included
here.) That is, albuterol (Ventolin®), amphetamine (Adderall®), dobutamine
(Dobutrex®), dopamine (Intropin®), isoproterenol (Isuprel®), lapatinib (Tykerb®),
levalbuterol (Xopenex®), and vardenafil (Levitra®).
D. Causes
1. Precipitating causes. Arrhythmias result from various conditions, including
a. Heart disease—infection, coronary artery disease (CAD), valvular heart disease, rheumatic
heart disease, ischemic heart disease
b. Myocardial infarction (MI)
c. Systemic hypertension
d. Hyperkalemia/hypokalemia
e. Chronic obstructive pulmonary disease (COPD)—emphysema, bronchitis
f. Thyroid disorders
g. Drug therapy (both antiarrhythmic and nonantiarrhythmic drugs)
h. Toxic doses of cardioactive drugs (e.g., digitalis preparations)
i. Increased sympathetic tone
594 Chapter 32 I. D
Figure 32-4. Reentry arrhythmias. A. Two waves of excitation going in opposite directions. B. A
unidirectional wave of excitation. C. Reexcitation of tissue in a slow conduction area.
E. Pathophysiology. Arrhythmias may decrease cardiac output, reduce blood pressure, and disrupt per-
fusion of vital organs. Specific pathophysiological consequences depend on the arrhythmia present.
F. Clinical evaluation
1. Physical findings. Although some arrhythmias are silent, most produce signs and symptoms.
Only an EKG/ECG can definitively identify an arrhythmia. However, physical findings may
sug-gest which arrhythmia is present; they also yield information about the patient’s clinical
status and may help identify associated complications. Signs and symptoms that typically
accompany arrhythmias include
a. Chest pain
b. Anxiety and confusion (reduced brain perfusion)
c. Dyspnea
d. Skin pallor or cyanosis
e. Abnormal pulse rate, rhythm, or amplitude
f. Reduce blood pressure
g. Palpitations
h. Syncope
i. Weakness
j. Convulsions
k. Hypotension
l. Decreased urinary output
2. Diagnostic test results
a. An EKG/ECG can identify a specific arrhythmia; usually, a 12-lead EKG/ECG is used.
b. Electrophysiological (EP) testing. T his intracardiac procedure determines the location of
ectopic foci and bypass tracts and may help assess therapeutic response to antiarrhythmic
drug therapy. It also can determine the need for a pacemaker or surgical intervention.
c. His bundle study, a type of EP testing, can locate the origin of a heart block or reentry pattern.
d. Laboratory findings. Some arrhythmias result from electrolyte abnormalities—most
commonly, hyperkalemia and hypocalcemia.
(1) A serum potassium level . 5 mEq/L reflects hyperkalemia, a serum calcium level , 4.5 mEq/L
signifies hypocalcemia, and serum magnesium levels , 2.5 mEq/L signify hypomagnesemia.
(2) An EKG/ECG tracing may suggest an electrolyte abnormality. For example, prolonged
QRS complexes, tented T waves, and lengthened PR intervals may signal hyperkalemia;
prolonged QT intervals and flattened or inverted T waves suggest hypocalcemia.
596 Chapter 32 I. G
G. Treatment objectives
1. Terminate or suppress the arrhythmia if it causes hemodynamic compromise or disturbing
symptoms.
2. Maintain adequate cardiac output and tissue perfusion.
3. Correct or maintain fluid balance (some arrhythmias cause hypervolemia).
H. National guidelines. During recent years, several prominent national organizations have
formalized a structure, which encourages the dissemination of “clinical practice guidelines,” which
focus on “evidence-based practice.” The American College of Cardiology, in collaboration with
the American Heart Association and European Society of Cardiology, has established a routine
standard for the evaluation, development, approval, and subsequent distribution of such practice
guidelines for a wide array of cardiac situations. We have used guidelines published in the
following text to serve as primary reference for this chapter:
• Fuster V, Rydén LE, Cannom DS, et al. ACC/AHA/ESC 2006 guidelines for the
management of patients with atrial fibrillation-executive summary: a report of the American
College of Cardiology/American Heart Association Task Force on Practice Guidelines and
the European Society of Cardiology Committee for Practice Guidelines (Writing Committee
to revise the 2001 guidelines for the management of patients with atrial fibrillation). Eur
Heart J. 2006;27(16):1979–2030.
• Fuster V, Rydén LE, Cannom DS, et al. 2011 ACCF/AHA/HRS focused updates
incorporated into the ACC/AHA/ESC 2006 guidelines for the management of patients with
atrial fibrilla-tion: a report of the American College of Cardiology Foundation/American
Heart Associa-tion Task Force on Practice Guidelines developed in partnership with the
European Society of Cardiology and in collaboration with the European Heart Rhythm
Association and the Heart Rhythm Society. J Am Coll Cardiol. 2011;57(11):e101–e198.
• Zipes DP, Camm AJ, Borggrefe M, et al. ACC/AHA/ESC 2006 guidelines for management of
patients with ventricular arrhythmias and the prevention of sudden cardiac death—executive
summary: a report of the American College of Cardiology/American Heart Association Task
Force and the European Society of Cardiology Committee for Practice Guidelines (Writing
Committee to develop guidelines for management of patients with ventricular arrhythmias and
the prevention of sudden cardiac death). Developed in collaboration with the European Heart
Rhythm Association and the Heart Rhythm Society. Eur Heart J. 2006;27(17):2099–2140.
• Wann LS, Curtis AB, January CT, et al. 2011 ACCF/AHA/HRS focused update on the
management of patients with atrial fibrillation (updating the 2006 guideline): a report of the
American College of Cardiology Foundation/American Heart Association Task Force on
Practice Guidelines. Circulation. 2011;123(1):104–123.
2. Ranking Recommendations
2. Each recommendation is given two scores: one referring to the relationship between benefit of
effect/treatment and one being the level of evidence, based on populations studied. Best
ranking would be a class I recommendation with level A evidence versus the lowest ranking,
which would be a class III recommendation with level C evidence, where it would be more
risky then beneficial and perhaps even harmful to use a treatment, which has been studied in
very limited populations and only supported by opinions or case reports.
II. THERAPY. Antiarrhythmic agents, directly or indirectly, alter the duration of the myocardial action
potential. Most antiarrhythmics fall into one of four classes, depending on their specific effects on the
heart’s electrical activity (Table 32-1) and on what portion of the action potential they directly affect.
A. Class I antiarrhythmics interfere with the sodium ion channel
1. Indications
a. Class Ia drugs-Agents in this class include quinidine, procainamide, and disopyramide.
(1) Quinidine (Various) is used to conversion and prevention of relapse into atrial fibrillation and/or
flutter and the suppression of ventricular arrhythmia. Due to a substantial number of concerns
with its administration, its use today has been reduced due to other therapeutic options currently
available. Quinidine poses added concern when used intravenously because of increased
cardiovascular effects (i.e., hypotension, syncope, myocardial depression).
T e latesth set of guidelines provides a class IIb recommendation with a “C” level of
evidence for its use in conversion of atrial fibrillation.
a
Singh BN, Vaughan Williams EM. Classification of antiarrhythmic drugs. In: Sandoe E, Flensted-Jensen E, Olesen KH, eds. Symposium on
Cardiac Arrhythmias. Sodertalige, Sweden: AB Astra; 1970.
bSotalol is a b-adrenergic blocker that prolongs the action potential of phase 3 and is classifi ed as type III.
598 Chapter 32 II. A
(2) Procainamide (Various) is used for the same arrhythmias for which quinidine is given. It is
used more frequently than quinidine because it can be administered intravenously and in
sustained-release oral preparations. The most recent guidelines give procainamide a class IIa
recommendation for patients where cardioversion from atrial fibrillation is not necessary
with a “C” level of evidence. It is also rated as class IIb for patients in atrial fibril-lation
involving conduction over an accessory pathway, and similar to quinidine, has been given a
class IIb ranking for conversion from atrial fibrillation with a “C” level of evidence.
(3) Disopyramide (Norpace®) may be used for the suppression and prevention of atrial and
premature ventricular complexes. Also effective in the conversion of atrial fibrillation, atrial
flutter, and paroxysmal atrial tachycardia to normal sinus rhythm and prevention of their
recurrences. Similar to procainamide, disopyramide is rated as class IIb for patients in atrial
fibrillation involving conduction over an accessory pathway with a “B” level of evidence,
and similar to quinidine and procainamide, has been given a class IIb ranking for conversion
from atrial fibrillation with a “C” level of evidence.
b. Class Ib drugs-Agents in this class include lidocaine, phenytoin, and flecainide, although
phe-nytoin has begun to be removed from the antiarrhythmic categories.
(1) Lidocaine (Xylocaine®) is used therapeutically for ventricular arrhythmias (especially pre-
mature ventricular contractions [PVCs] and ventricular tachycardia) that result from acute
MI and open-heart surgery. Controversy still exists as to the benefits of lidocaine when used
prophylactically in patients with acute MI to prevent ventricular fibrillation. A recent
analysis showed an increase in the number of deaths in patients receiving lidocaine during
an acute MI as compared to placebo. Many feel that its current use prophylactically post-MI
is no longer justified. Lidocaine has been given a rating of class IIb in the initial treat-ment
of patients with MI or myocardial ischemia with a “C” level of evidence. It has also been
given a class IIb rating in patients with ventricular tachycardia with MI or myocardial
ischemia and in patients presenting with long QT syndrome and TdP.
(2) Phenytoin (Dilantin®) in the past has been primarily indicated in the treatment of digitalis-
induced ventricular arrhythmias and heart block. However, most recent guide-lines provide
it with a class III recommendation and a “C” level of evidence. It will no longer be included
in this chapter until updated guidelines reintroduce it as an antiarrhythmic.
(3) Mexiletine (Various) is closely related to lidocaine structurally, with modifications
that reduce first-pass liver metabolism, making oral therapy possible. It is most
commonly used to treat patients in with serious ventricular arrhythmias or PVCs.
Similar to lidocaine, mexiletine is rated as a class IIb in patients who have a long QT
syndrome and TdP.
c. Class Ic drugs-Agents in this class include flecainide and propafenone.
(1) Flecainide (Tambocor®) is indicated in the prevention and treatment of life-threatening
ventricular arrhythmias, such as sustained ventricular tachycardia, controlling symptomatic,
and disabling supraventricular tachycardias, in patients without structural heart disease
when other agents fail. Flecainide is reserved for patients with refractory life-threatening
ventricular arrhythmias who do not have CAD. Flecainide has received a class Ia
recommen-dation in the conversion from atrial fibrillation with an “A” class of evidence.
(2) Propafenone (Rythmol®) is indicated in the treatment of life-threatening ventricular
arrhythmias and in the maintenance of normal sinus rhythm in patients with symptomatic
atrial fibrillation. Similar to flecainide, propafenone has also received a class Ia recom-
mendation in the conversion from atrial fibrillation with an “A” class of evidence.
2. Mechanism of action. As a class, all of the agents work by blocking the rapid inward sodium
current and thereby slow down the rate of rise of the cardiac tissue’s action potential. However,
differences in EP effects has led to a subclassification of the class I agents into three subsets
(Ia, Ib, and Ic), based on these EP effects.
a. Class Ia drugs moderately reduce the depolarization rate and prolong repolarization
(refractory period).
b. Class Ib drugs shorten repolarization (refractory period); they also weakly affect the
repolarization rate.
c. Class Ic drugs strongly depress depolarization but have a negligible effect on the duration
of repolarization or refractoriness.
Cardiac Arrhythmias 599
when decision makers choose between outpatient and inpatient initiation of antiarrhythmic
therapy.
a. Quinidine (Various)
(1) This drug is contraindicated in patients with
(a) Complete AV block unless a ventricular pacemaker is in place.
(b) Marked prolongation of the QT interval or prolonged QT syndrome because
ventricular tachyarrhythmia (TdP) may arise, resulting in quinidine syncope (i.e.,
syncope or sudden death).
(2) An increase of 50% or more in the duration of the QRS complex necessitates dosage
reduction.
(3) Quinidine has a narrow therapeutic index. Therapeutic serum levels are in the range of
2 to 6 mg/mL, depending on the specificity of the assay. Toxicity may cause acute
cardiac effects, such as pronounced slowing of conduction in all heart regions; this, in
turn, may lead to SA block or arrest, ventricular tachycardia, or asystole.
(4) The EKG/ECG should be monitored during quinidine therapy to detect signs of car-
diotoxicity. To counteract quinidine-induced ventricular tachyarrhythmias, catechol-
amines, glucagon, or sodium lactate may be given.
(5) In patients receiving quinidine for atrial tachyarrhythmias, vagolytic effects may in-
crease impulse conduction at the AV node, resulting in an accelerated ventricular re-
sponse. To prevent this, agents that slow AV nodal conduction (e.g., verapamil,
digoxin) may be administered.
(6) The dosage should be reduced in elderly patients (. 60 years old) and in patients with
hepatic dysfunction or congestive heart failure (CHF).
(7) Embolism may occur on restoration of normal sinus rhythm after prolonged atrial
fibril-lation. To prevent or minimize this complication, anticoagulants may be
administered before quinidine therapy begins.
(8) Quinidine may cause cinchonism at high serum concentrations, manifested by
tinnitus, hearing loss, blurred vision, and gastrointestinal (GI) disturbances. In severe
cases, nausea, vomiting, diarrhea, headache, confusion, delirium, photophobia,
diplopia, and psychosis may occur.
(9) GI reactions are the most common adverse reactions to quinidine. About 30% of
patients experience diarrhea; nausea and vomiting may also occur. Arising almost
immediately after the first dose, these symptoms sometimes warrant discontinuing the
drug. However, aluminum hydroxide or use of the polygalacturonate salt may reverse
this.
(10) Hypersensitivity reactions include anaphylaxis, thrombocytopenia, respiratory
distress, and vascular collapse.
b. Procainamide (Various)
(1) This drug is contraindicated in patients with hypersensitivity to procaine and related
drugs, myasthenia gravis, second- or third-degree AV block with no pacemaker, a
history of procainamide-induced systemic lupus erythematosus (SLE), prolonged QT
syndrome, or TdP.
(2) An increase of 50% or more in the duration of the QRS complex necessitates dosage
reduction.
(3) Procainamide has a narrow therapeutic index. Therapeutic serum levels are report-ed
in the range of 4 to 10 mg/mL. N-acetyl procainamide (NAPA) levels of 15 to 25
mg/mL are considered therapeutic. The active metabolite, NAPA, possesses differing
pharmacological cardiovascular effects, and serum levels need to be evaluated inde-
pendently of procainamide. Toxicity may cause acute cardiac effects (e.g.,
pronounced slowing of conduction in all heart regions), which, in turn, may lead to
SA block or ar-rest, ventricular tachycardia, or asystole.
(4) High serum procainamide levels may induce ventricular arrhythmias (e.g., PVCs, ven-
tricular tachycardia, or fibrillation). The EKG/ECG should be monitored continuously
to detect these problems. Catecholamines, glucagon, or sodium lactate may be
adminis-tered to counteract these arrhythmias.
Cardiac Arrhythmias 601
(2) The EKG/ECG should be monitored during flecainide therapy because this drug may
exacerbate existing arrhythmias or precipitate new ones. Flecainide was shown in the
CAST study to increase mortality in patients with asymptomatic ventricular
arrhythmias and, therefore, should be reserved for patients with life-threatening
ventricular arrhyth-mias that are refractory to other drugs.
(3) This drug has a significant negative inotropic effect and may bring on or worsen CHF
and cardiomyopathy.
(4) Adverse CNS effects (e.g., dizziness, headache, tremor) and GI effects (e.g., nausea,
abdominal pain) may occur.
(5) Blurred vision and dyspnea have been reported.
(6) Therapeutic serum levels recommended for flecainide are between 0.2 and 1.0 mg/mL.
g. Propafenone (Rythmol®)
(1) This drug, such as other antiarrhythmic agents, may cause new or worsened arrhythmias.
Such proarrhythmic properties range from an increased frequency of PVCs to the
development of severe ventricular tachycardia, ventricular fibrillation, and TdP. This pro-
arrhythmic effect has been under discussion for the class Ic agents; thus, when used, these
agents should be monitored closely. The findings from the CAST study must be weighed
against the benefits of using these agents for treating significant ventricular arrhythmias.
(2) Dizziness is a side effect that has been reported in as many as 10% to 15% of patients
taking the drug.
(3) Other associated side effects include vomiting, a metallic bitter taste in the mouth,
constipation, headache, and new or worsening CHF and asthma.
(4) Therapeutic serum levels recommended for propafenone are between 0.06 and 1.0 mg/mL.
5. Significant interactions
a. Quinidine
(1) Quinidine may increase serum levels of digoxin and increase the effects of digitalis on
the heart, with a resultant increase in toxicity.
(2) Severe orthostatic hypotension may occur with concomitant administration of vasodila-
tors (e.g., nitroglycerin).
(3) Phenytoin, rifampin (Rifadin®), and barbiturates may antagonize quinidine activity
and reduce its therapeutic efficacy.
(4) Nifedipine (Procardia®) may reduce plasma quinidine levels.
(5) Antacids, sodium bicarbonate, and sodium acetazolamide (Diamox®) may increase
plasma quinidine levels, possibly resulting in toxicity.
(6) Quinidine may produce additive hypoprothrombinemic effects with coumarin anti-
coagulants.
b. Amiodarone (Cordarone®) and cimetidine (Tagamet®) may increase plasma
procainamide levels, possibly leading to drug toxicity.
c. Phenytoin accelerates disopyramide metabolism, possibly reducing its therapeutic efficacy.
d. Lidocaine
(1) Phenytoin may increase the cardiodepressant effects of lidocaine.
(2) -Blockers (class II antiarrhythmics) may reduce lidocaine metabolism, possibly
leading to drug toxicity.
e. Mexiletine (Various). Phenobarbital, rifampin (Rifadin®), and phenytoin (Dilantin®)
reduce plasma mexiletine levels and may decrease therapeutic efficacy.
B. Class II antiarrhythmics
1. Indications. These drugs— -adrenergic blockers—among the drugs in this class indicated for
the treatment of select arrhythmias are propranolol (Inderal®), esmolol (Brevibloc®), and
acebuto-lol (Sectral®) are approved for antiarrhythmic use.
a. Propranolol (Inderal®) may be given to
(1) Control supraventricular arrhythmias (e.g., atrial fibrillation or flutter, paroxysmal
supra-ventricular tachycardia [PSVTs])
(2) Treat tachyarrhythmias caused by catecholamine stimulation (e.g., in hyperthyroidism,
during anesthesia)
(3) Suppress severe ventricular arrhythmias in prolonged QT syndrome
(4) Treat digitalis-induced ventricular arrhythmias
Cardiac Arrhythmias 603
(5) Terminate certain ventricular arrhythmias (e.g., PVCs in patients without structural
heart disease)
(6) In the most recently completed guidelines, the intravenous administration of b-
adrenergic blockers was given a class I recommendation with a “B” level of evidence
to slow the ventricular response to atrial fibrillation in acute setting, with cautious
considerations to those with hypotension or HF.
b. Esmolol (Brevibloc®) is used to treat supraventricular tachycardias; it possesses a very
short (9 mins) half-life, and has been used to control the ventricular response to atrial
fibrillation or flutter during or after surgery.
c. Acebutolol (Sectral®) has been used in the management of ventricular arrhythmias; although not
specifically listed within the recent guidelines, like esmolol and propranolol, the b-adrenergic blockers
were given a class I ranking for ventricular control in patients with atrial fibrillation.
2. Mechanism of action. Class II antiarrhythmics reduce sympathetic stimulation of the heart,
decreasing impulse conduction through the AV node and lengthening the refractory period.
Additionally, this class of antiarrhythmics slows the sinus rhythm without significantly changing the
QT or QRS intervals, resulting in a reduced heart rate and a decrease in myocardial oxygen demand.
3. Administration and dosage
a. Propranolol (Inderal®) may be given intravenously or orally when used as an antiarrhythmic.
(1) Emergency therapy calls for slow intravenous administration of 1 to 3 mg diluted in 50
mL dextrose 5% in water or normal saline solution. This dose is infused slowly (no
faster than 1 mg/min). A second dose of 1 to 3 mg may be given 2 mins later.
(2) For oral therapy, 10 to 80 mg/day is given in three to four doses. (However, 1000 mg or
more may be required for resistant ventricular arrhythmias.)
b. Esmolol (Brevibloc®) is given intravenously. A loading dose of 500 mcg/kg/min is infused
over 1 min, followed by a 4-min maintenance infusion of 50 mcg/kg/min. If a satisfactory
response is not achieved within 5 mins, the loading dose is repeated and followed by a
main-tenance infusion of 100 mcg/kg/min.
c. Acebutolol (Sectral®) is given orally. An oral dose of 400 mg is given by mouth daily up
to a maximum of 1200 mg/day for the treatment of ventricular arrhythmias.
4. Precautions and monitoring effects
a. Propranolol (Inderal®)
(1) This drug is contraindicated in patients with sinus bradycardia, second- or third-degree
AV block, cardiogenic shock, severe HF, or asthma.
(2) The b-blocking effects of this drug may lead to marked hypotension, exacerbation of
HF and left ventricular failure, or cardiac arrest.
(3) Blood pressure, heart rate, and the EKG/ECG should be monitored during intravenous
infusion.
(4) Embolism may occur upon restoration of normal sinus rhythm after sustained atrial fi
brillation. An anticoagulant may be given before propranolol therapy begins to prevent
this complication.
(5) Propranolol may depress AV node conduction and ventricular pacemaker activity,
resulting in AV block or asystole.
(6) This drug may mask the signs and symptoms of hypoglycemia. It also may mask signs
of shock.
(7) Fatigue, lethargy, increased airway resistance, and skin rash have been reported.
(8) Nausea, vomiting, and diarrhea may occur.
(9) Sudden withdrawal of propranolol may lead to acute MI, arrhythmias, or angina in
cardiac patients. Drug therapy is discontinued by tapering the dose over 4 to 7 days.
b. Esmolol (Brevibloc®)
(1) This drug is contraindicated in patients with severe CHF or sinus bradycardia.
(2) Hypotension occurs in approximately 30% of patients receiving esmolol. This effect
can be reversed by reducing the dosage or stopping the infusion.
(3) This drug is for short-term use only and should be replaced by a long-acting
antiarrhythmic once the patient’s heart stabilizes.
(4) Dizziness, headache, fatigue, and agitation may occur.
(5) Other adverse effects include nausea, vomiting, and bronchospasm.
604 Chapter 32 II. B
c. Acebutolol (Sectral®)
(1) Similar contraindications as other b-blockers; heart block, uncompensated HF, sick
sinus syndrome.
(2) Major monitoring parameters for patients include serum creatinine level, heart rate, and
blood pressure.
(3) Therapy should not be rapidly discontinued but rather gradually over 10 to 14 days.
Abrupt discontinuation in cardiac patients may exacerbate an angina attack or a more
significant acute coronary syndrome.
5. Significant interactions
a. Propranolol (Inderal®)
(1) Severe vasoconstriction may occur with concomitant epinephrine administration.
(2) Digitalis preparations can cause excessive bradycardia.
(3) Calcium-channel blockers—for example, diltiazem (Cardizem®) and verapamil
(Calan®)—and other negative inotropic and chronotropic drugs—such as disopyramide
(Norpace®) and quinidine—add to the myocardial depressant effects of propranolol.
b. Esmolol (Brevibloc®). Morphine (MS Contin®) may raise plasma esmolol levels.
C. Class III antiarrhythmics
1. Indications
a. Amiodarone (Cordarone®) is given to control malignant ventricular arrhythmias and is
recommended within the advance cardiac life support (ACLS) guidelines, has been given a
class I recommendation for victims with ventricular tachyarrhythmia during a cardiac ar-
rest, for attempting a stable rhythm after further defibrillations. A “B” level of evidence has
been documented. Unlike most other antiarrhythmics, with the exception of the b-
adrenergic blockers, amiodarone has been shown to reduce arrhythmic deaths in patients
after an MI. Additionally, amiodarone received a class IIa recommendation for patients with
sustained ventricular tachycardia that is hemodynamically unstable and refractory to
conversion with countershock or procainamide or other agents. Amiodarone has also
received a class I rating for intravenous administration to control the heart rate in atrial
fibrillation and HF patients who do not have an accessory pathway.
b. Sotalol (Betapace®) is used to treat supraventricular and ventricular tachyarrhythmias. Sotalol
antagonizes both b1- and b2-adrenergic receptors but also prolongs the phase 3 action potential.
It is this property that distinguishes it from other b-adrenergic blockers and is the reason why it
is classified as a class III antiarrhythmic agent rather than a class II agent (b-adrenergic blocker).
c. Ibutilide (Corvert®) is used in the conversion of atrial fibrillation and flutter of recent
onset (duration , 30 days). In the most recent guidelines provided for the treatment of atrial
fi brillation, ibutilide received along with dofetilide, a class I recommendation with “A”
level of evidence for its use in the pharmacologic cardioversion of atrial fibrillation.
d. Dofetilide (Tikosyn®) is available in the United States under restricted access in the
treatment of atrial fibrillation/flutter. As mentioned earlier, within the most recent
guidelines provided for the treatment of atrial fibrillation, dofetilide received, along with
ibutilide, a class I recom-mendation with “A” level of evidence for its use in the
pharmacological cardioversion of atrial fibrillation.
2. Mechanism of action. Class III antiarrhythmic drugs primarily work on the potassium
channels of the action potential and prolong the refractory period and action potential; they
have no effect on myocardial contractility or conduction time.
3. Administration and dosage
a. Amiodarone (Cordarone®). Available for both oral and intravenous use and should only
be initiated in the hospital setting. Amiodarone has been incorporated into the ACLS
guidelines and recommended by the expert panel members as the first-choice
antiarrhythmic for shock-refractory ventricular fibrillation/ventricular tachycardia.
(1) It is available for oral use; 800 to 1600 mg every 12 hrs is given for 7 to 14 days, then
200 to 400 mg daily thereafter.
(2) Oral treatment is used to suppress ventricular and supraventricular arrhythmias but can
take days or weeks to take effect. Oral doses of 100 to 600 mg/day (usually 300 to 400
mg/day) for maintenance therapy in ventricular tachycardia and 100 to 200 mg/day for
maintenance therapy for supraventricular tachycardias are given.
Cardiac Arrhythmias 605
(3) Intravenous formulation is available for treatment and prophylaxis of recurrent ventricular
fibrillation or hemodynamically unstable ventricular tachycardia in refractory patients.
(4) The intravenous form is rapidly distributed throughout the body. Recommended doses include a
rapid loading infusion of 150 mg over 10 mins, followed by a slow infusion of 1.0 mg/min for 6
hrs (360 mg), and then a maintenance infusion of 0.5 mg/min for the remainder of the 24-hr
period. Patients usually receive 2 to 4 days of infusions before conversion to oral form. However,
a maintenance infusion can be continued for 2 to 3 weeks.
b. Sotalol (Betapace®) is available commercially as an oral tablet, and therapy should be
initi-ated within the hospital setting. Normal dosing of 80 mg twice daily initially and
increasing doses at 2- to 3-day intervals to a maximum dose of 640 mg/day, given in two to
three doses throughout the day.
c. Ibutilide (Corvert®) is available only for injection in a 0.1-mg/mL, 10-mL vial. Normal
doses for the conversion of recent-onset atrial fibrillation to normal sinus rhythm is a dose
of 1 mg (0.01 mg/kg for those , 60 kg) over 10 mins, with a repeat dose in 10 mins if the
arrhythmia does not end.
d. Dofetilide (Tikosyn®) is available only for oral administration in 0.125-, 0.25-, and 0.5-mg
capsules under the trade name of Tikosyn and should only be initiated in a hospital setting
with trained personnel and the equipment necessary to provide continuous cardiac monitor-
ing during initiation of therapy.
(1) A normal dose for the conversion of recent-onset atrial fibrillation to normal sinus
rhythm is 0.5 mg twice daily for patients with creatinine clearance values . 60 mL/min;
doses are reduced 50% (0.25 mg) for those with creatinine clearance values of 40 to 60
mL/min, and doses are reduced an additional 50% (0.125 mg) for those with creatinine
clearance values of 20 to 40 mL/min.
(2) Maintenance therapy is based on the EKG/ECG, with doses being reduced with QTc
pro-longation exceeding 15% of the baseline value. Any patient developing a QTc
interval exceeding 500 msec should have therapy discontinued immediately.
(3) Dofetilide should not be given to those with creatinine clearance values , 20 mL/min.
4. Precautions and monitoring effects
a. Amiodarone (Cordarone®)
(1) Life-threatening pulmonary toxicity may occur during amiodarone therapy, especially
in patients receiving . 400 mg/day. Baseline as well as routine pulmonary function tests
reveal relevant pulmonary changes.
(2) Most patients develop corneal microdeposits 1 to 4 months after amiodarone therapy
begins. However, this reaction rarely causes visual disturbance, but the patient should
be monitored with routine ophthalmological examinations.
(3) Blood pressure and heart rate and rhythm should be monitored for hypotension and
bradyarrhythmias.
(4) Patients should be monitored routinely for the possible development of hepatic dysfunc-
tion, thyroid disorders (e.g., hyperthyroidism, hypothyroidism), and photosensitivity.
(5) CNS reactions include fatigue, malaise, peripheral neuropathy, and extrapyramidal effects.
(6) Nausea and vomiting have been reported.
(7) This drug has an extremely long half-life (up to 60 days). Therapeutic response may be
delayed for weeks after oral therapy begins; adverse reactions may persist up to 4
months after therapy ends.
b. Sotalol (Betapace®)
(1) Side effects of this drug are directly related to b-blockade and prolongation of repolarization.
(2) Transient hypotension, bradycardia, myocardial depression (negative inotropic effect),
and bronchospasm have all been associated with this drug.
(3) This drug carries all the contraindications associated with other b-blockers along with
those owing to its electrophysiologic properties.
c. Ibutilide (Corvert®)
(1) Infusion should be discontinued as soon as the atrial arrhythmia is terminated or if sus-
tained or nonsustained ventricular arrhythmia or marked QT prolongation is documented.
(2) Continuous EKG/ECG monitoring is required for at least 4 hrs after discontinuing the
infusion or until the QT interval returns to baseline.
606 Chapter 32 II. C
d. Dofetilide (Tikosyn®)
(1) Patients need to be monitored closely for the subsequent development of ventricular
arrhythmias with increasing doses of dofetilide or with declining renal status. In clinical
trials, ventricular tachycardias, including TdP, are the most frequently occurring
arrhyth-mias due to dofetilide.
(2) Hypokalemia and those situations that might cause hypotension will predispose a
patient to prolongation of the QT interval, which could put a dofetilide patient at risk
for toxic arrhythmias.
5. Significant interactions
a. Amiodarone (Cordarone®)
(1) Amiodarone may increase the plasma levels of quinidine, procainamide, diltiazem,
digitalis, and flecainide.
(2) It may increase the pharmacological effect of -blockers, calcium-channel blockers,
and warfarin.
(3) Special note: Amiodarone has been reported to have numerous drug–drug interactions
among all categories of drugs. To avoid the development of a significant drug–drug in-
teraction, a thorough patient medication profile should be carried out for each patient
having amiodarone therapy initiated, as well as each time a patient currently receiving
amiodarone is given an additional drug.
b. Sotalol (Betapace®)
(1) Sotalol must be used cautiously in those patients receiving agents with cardiac-
depressant properties.
(2) Agents such as sotalol that prolong the QT interval may induce malignant arrhythmias
when used in combination with other class Ia antiarrhythmics, especially in the
presence of low potassium levels.
c. Ibutilide (Corvert®) should be avoided with other agents that prolong repolarization or
with-in 4 hrs of administration.
d. Dofetilide (Tikosyn®) should be avoided in patients who have hypokalemia or preexisting
QT prolongation.
D. Class IV antiarrhythmics
1. Indications
a. Calcium-channel blockers (e.g., verapamil [Calan®]; diltiazem [Cardizem®]) are used
mainly to treat and prevent supraventricular arrhythmias.
(1) They are first-line agents for the suppression of PSVTs stemming from AV nodal reentry.
(2) They can rapidly control the ventricular response to atrial flutter and fibrillation.
b. Other calcium-channel blockers available include amlodipine (Norvasc®), clevidipine
(Cleviprex®), felodipine (Plendil®), isradipine (DynaCirc®), nicardipine (Cardene®),
nifedipine (Procardia®), nimodipine (Nimotop®), and nisoldipine (Sular®), but these
agents have primar-ily been used in the treatment of angina pectoris and hypertension and
will not be discussed further in this section.
2. Mechanism of action. Class IV antiarrhythmics are calcium-channel blockers. They decrease
conduction through those areas of the heart dependent on slow calcium channels, which include
the SA and AV nodes.
3. Administration and dosage
a. To control atrial arrhythmias, verapamil usually is administered intravenously. A dose of
2.5 to 10 mg is given over at least 2 mins and may be repeated in 30 mins, if necessary. A 5
to 10 mg/ hr continuous intravenous infusion has also been used in treating arrhythmias.
b. To prevent PSVTs, verapamil may be given orally in four daily doses of 80 to 120 mg each.
c. To control atrial arrhythmias, diltiazem usually is administered intravenously. A dose of 20
mg (0.25 mg/kg) is given over 2 mins. If an adequate response is not obtained, a second
dose of 25 mg (0.35 mg/kg) is administered after 15 mins. A 5 to 15 mg/hr intravenous
continuous infusion has also been used in treating arrhythmias.
4. Precautions and monitoring effects
a. Verapamil and diltiazem are contraindicated in patients with AV block; left ventricular dys-
function; severe hypotension; concomitant, intravenous b-blockers; and atrial fibrillation
with an accessory AV pathway.
Cardiac Arrhythmias 607
b. These drugs must be used cautiously in patients with CHF, sick sinus syndrome, MI, and
hepatic or renal impairment.
c. Because of the negative chronotropic effect, verapamil and diltiazem must be used
cautiously in patients who have slow heart rates or who are receiving digitalis glycosides.
d. The EKG/ECG (especially the respiratory rate (RR) interval) should be monitored during
therapy.
e. Patients . 60 years old should receive reduced dosages and slower injection rates.
f. Constipation and nausea have been reported with verapamil.
5. Significant interactions
a. Concomitant administration of -blockers or disopyramide may precipitate HF.
b. Quinidine may increase the risk of calcium-channel blocker–induced hypotension.
c. Verapamil may increase serum digoxin concentrations, and diltiazem may do the same to a
lesser extent.
d. Rifampin may enhance the metabolism of calcium-channel blockers, with a resultant
decrease in pharmacological effect.
e. Verapamil and diltiazem may inhibit theophylline metabolism and may require reductions
in theophylline dosage.
f. Diltiazem and verapamil inhibit the metabolism of cyclosporine (Gengraf®) and may
require reductions in cyclosporine dosages.
E. Unclassified antiarrhythmics
1. Atropine (Various)
a. Indications. Atropine is therapeutic for symptomatic sinus bradycardia and junctional
rhythm.
b. Mechanism of action. An anticholinergic, atropine blocks vagal effects on the SA node,
promoting conduction through the AV node and increasing the heart rate.
c. Administration and dosage. For antiarrhythmic use, atropine is administered in a dose of
0.4 to 1.0 mg by intravenous push; the dose is given every 5 mins to a maximum of 2.0 mg.
d. Precautions and monitoring effects
(1) Thirst and dry mouth are the most common adverse effects of atropine.
(2) CNS reactions (e.g., restlessness, headache, disorientation, dizziness) may occur with
doses over 5 mg.
(3) Tachycardia and ophthalmic disturbances (e.g., mydriasis, blurred vision, photophobia)
may occur with doses of 1 mg or more.
(4) Initial doses may induce a reflex bradycardia owing to incomplete suppression of vagal
impulses.
2. Adenosine (Adenocard®)
a. Indications. Adenosine is indicated for the conversion of acute supraventricular tachycardia
(SVT) to normal sinus rhythm.
b. Mechanism of action. Adenosine is a naturally occurring nucleoside, which is normally
present in all cells of the body. It has been shown to
(1) slow conduction through the AV node,
(2) interrupt reentry pathways through the AV node, and
(3) restore normal sinus rhythm in patients with PSVTs.
c. Administration and dosage. For antiarrhythmic effects, adenosine is given as a rapid bolus
intravenous injection in a 6-mg dose over 1 to 2 secs. If the first dose does not eliminate the
arrhythmia within 1 to 2 mins, the dose should be increased to 12 mg and again given as a
rapid intravenous dose. An additional 12-mg dose may be repeated if necessary.
d. Precautions and monitoring effects
(1) The effects of adenosine are antagonized by methylxanthines, such as caffeine and theophylline.
Theophylline has been successfully used for treating adenosine-induced side effects, such as
hypotension, sweating, and palpitations. If side effects are encountered, ag-gressive therapy is not
required because of the ultrashort half-life of the drug (10 secs or less).
(2) The main side effect associated with adenosine use in up to 18% of patients is facial fl
ushing, but this effect is normally very short-lived.
(3) Other side effects associated with adenosine use include shortness of breath, chest
pressure, nausea, headache, and a metallic taste.
608 Chapter 32 II. E
e. Additional use. Adenosine has been used as an adjunctive agent in patients undergoing
various types of pharmacological stress testing (e.g., thallium). In this situation, adenosine
is given as a continuous infusion over a period of 4 to 6 mins and is able to provide a form
of exercise tolerance test in patients not able to exert themselves owing to age, fatigue, and
various other physical handicaps.
3. Magnesium sulfate (Various)
a. Indications. Previous national guidelines have recommended magnesium for the treatment
of drug-induced long QT syndrome. In the most recent ACLS guidelines, published in 2010,
magnesium received a class IIb rating with a “class B” level of evidence, for its use strictly
in patients presenting with TdP associated with a long QT interval. Additionally,
magnesium has been used in the treatment of arrhythmias (ventricular
tachycardia/fibrillation, due to hypo-magnesemia).
b. Mechanism of action. Acts on the myocardium by slowing the rate of impulse formation at
the SA node and therefore slows down conduction. Magnesium is also necessary in the
exchange of calcium, sodium, and potassium in and out of cells, which in the case of TdP
might lower the amplitude of the early after depolarizations.
c. Administration and dosage. For the treatment of TdP, 1 to 6 g over several minutes,
occasion-ally followed by approximately 3 to 20 mg/min by IV infusion for 5 to 48 hrs,
depending on response and serum magnesium concentrations.
(1) Alternatively, for TdP associated with cardiac (pulseless) arrest, 1 to 2 g in 10 mL 5%
dex-trose injection over 5 to 20 mins.
(2) Alternatively, for TdP in a patient with pulses, give a loading dose of 1 to 2 g (8 to 16
mEq) in 50 to 100 mL 5% dextrose injection over 5 to 60 mins.
(3) Intraosseous TdP associated with cardiac (pulseless) arrest, 1 to 2 g in 10 mL 5%
dextrose injection over 5 to 20 mins.
d. Precautions and monitoring effects
(1) Routine monitoring of magnesium levels, in order to prevent hypermagnesemia, while
also monitoring calcium levels and phosphorus levels, which can be reduced when
administering IV magnesium.
4. Digoxin (Lanoxin®)
a. Indications. Recent guidelines recommend that digoxin is effective in stable, narrow-
complex regular tachycardias if rhythm remains uncontrolled or unconverted by adenosine
or vagal maneuvers or if SVT is recurrent. Additionally, digoxin has been shown effective
to control the ventricular rate in patients with atrial fibrillation or atrial flutter.
b. Mechanism of action. As a cardiac glycoside, digoxin has positive inotropic effects; however,
for rhythm control digoxin relies on its “parasympathomimetic properties,” which slow
conduction through the AV node and decrease impulses going through to the ventricles.
c. Administration and dosage. Digoxin can be given as 8 to 12 mcg/kg with half of that
being given over 5 mins, and the remaining 50% given in two doses at 4 and 8 hrs later.
d. Precautions and monitoring effects
(1) The onset of action is slow and depending on the circumstances might not be
acceptable for acute arrhythmias.
(2) Long-term use of digoxin will expose the patient to numerous monitoring requirements,
including renal function, drug–drug interactions, and fluid and electrolyte monitoring,
in order to minimize likelihood of adverse drug reactions.
5. Dronedarone (Multaq®)
a. The newest antiarrhythmic, which was a major focal point for recently updated guidelines
for the treatment of atrial fibrillation, and which has not yet been formally added into the
current Vaughan Williams classification.
b. Within the updated guidelines, dronedarone (Multaq®) received a class IIa rating with a
“class B” level of evidence, to decrease the need for hospitalization for cardiovascular
events in patients with paroxysmal atrial fibrillation or after conversion of persistent atrial
fibrilla-tion. Additionally, it was felt that dronedarone could be initiated during outpatient
therapy. However, within the same guidelines, dronedarone was also given a class III for
use in class IV HF or patients who have had an episode of decompensated HF in the past 4
weeks, especially with depressed left ventricular function (ejection fraction , 35%).
Cardiac Arrhythmias 609
c. Dronedarone (Multaq®) is available and is doses as 400 mg by mouth twice daily in the
prevention of atrial fibrillation and/or flutter.
d. Dronedarone (Multaq®), similar to amiodarone, there are numerous drug–drug interactions
with dronedarone; consequently, each newly added therapy should be evaluated in order to
identify the potential impact when starting, as well as when ending each therapy.
Study
Questions 5. Class III antiarrhythmics have which of the following
effects to the cardiac cell’s action potential?
Directions: Each of the questions, statements, or (A) Slow the rate of rise for phase 0 of depolarization.
incomplete statements can be correctly answered or (B) Delay the fast-channel conductance of
completed by one of the suggested answers or sodium ions.
phrases. Choose the best answer. (C) Prolong phases 2 and 3 of repolarization.
(D) Inhibit the slow-channel conductance
1. Strong anticholinergic effects limit the
of calcium ions.
antiarrhythmic use of (E) Prolong the refractory period of the action
(A) quinidine (Various). potential.
(B) procainamide (Various).
6. Which of the following drugs is a class IV
(C) mexiletine (Various).
antiarrhythmic that is primarily indicated for the
(D) f lecainide (Tambocor®).
(E) disopyramide (Norpace®). treatment of supraventricular tachyarrhythmias?
(A) ibutilide (Corvert®)
2. A pronounced slowing of phase 0 of the myocardial
(B) mexiletine (Various)
action potential results in a prolongation of either
(C) diltiazem (Cardizem®)
atrial depolarization, causing a prolonged P wave on
(D) procainamide (Various)
the electrocardiogram (EKG/ECG), or ventricular
(E) propranolol (Inderal®)
depolarization, causing a prolonged QRS complex
characterized by which class of antiarrhythmics? 7. Relatively new antiarrhythmic agent, which has not
(A) Class I yet been formally added into the Vaughan Williams
classification table but received a class IIa rating
(B) Class II
with a class B level of evidence to decrease the need
(C) Class III
for hospitalization in patients with paroxysmal atrial
(D) Class IV
(E) Class V fibrillation or after conversion of persistent atrial
fibrillation. Which of the following agents is best
3. Which of the following class III antiarrhythmics has described by the above statements?
been reported as “carrying a risk for” causing (A) aliskiren (Tekturna®)
torsades de pointes? (B) inamrinone (Various)
(A) lidocaine (Xylocaine®) (C) dronedarone (Multaq®)
(B) amiodarone (Cordarone®) (D) amiodarone (Cordarone®)
(C) quinidine (Various) (E) dofetilide (Tikosyn®)
(D) f lecainide (Tambocor®)
8. Which of the following drugs is a class III
(E) diltiazem (Cardizem®)
antiarrhythmic agent that is effective in the acute
4. A patient receiving a class I antiarrhythmic agent management of atrial fibrillation or atrial flutter
complains of GI symptoms, including nausea, of recent onset?
vomiting, and occasional diarrhea after taking a (A) propranolol (Inderal®)
dose. The patient is most likely receiving (B) ibutilide (Corvert®)
(A) lidocaine (Xylocaine®). (C) metoprolol (Lopressor®)
(B) procainamide (Various). (D) disopyramide (Norpace®)
(C) quinidine (Various). (E) diltiazem (Cardizem®)
(D) f lecainide (Tambocor®).
(E) propranolol (Inderal®).
610 Chapter 32 10. Based on the criteria used for recent national
guidelines used within the cardiology arena, which
of the following recommendations would most
9. All of the following problems represent concerns likely result in the use of a selected antiarrhythmic
when patients are started on amiodarone except therapy for a patient with atrial fibrillation?
(A) extremely long t½. (A) Class I; level C
(B) need for multiple daily doses. (B) Class I; level A
(C) development of hyperthyroidism or (C) Class IIa; level B
hypothyroidism. (D) Class III; level A
(D) development of liver toxicity. (E) Class IIb; level A
(E) interactions with numerous other drugs.