Case Files
Pharmacology
Second Edition
EUGENE C. TOY, MD
The John S.Dunn Senior Academic Chief and Program
Director
Obstetrics and Gynecology Residency Program
The Methodist Hospital,Houston
Clerkship Director and Clinical Associate Professor
Department of Obstetrics and Gynecology
University of Texas Medical School at Houston
Houston,Texas
GARY C. ROSENFELD,PHD
Professor
Department of Integrative Biology and Pharmacology
Assistant Dean for Educational Programs
University of Texas Medical School at Houston
Houston,Texas
DAVID S.LOOSE, PHD
Associate Professor
Department of Integrative Biology and Pharmacology
University of Texas Medical School at Houston
Houston,Texas
DONALD BRISCOE, MD
Program Director
Family Medicine Residency
The Methodist Hospital,Houston
Medical Director
Houston Community Health Centers,Inc.
Denver Harbor Clinic
Houston,Texas
New York Chicago San Francisco
Lisbon London Madrid Mexico City
Milan New Delhi San Juan Seoul
Singapore Sydney Toronto
Copyright © 2008 by The McGraw-Hill Companies, Inc. All rights reserved. Manufactured in the
United States of America. Except as permitted under the United States Copyright Act of 1976, no
part of this publication may be reproduced or distributed in any form or by any means, or stored
in a database or retrieval system, without the prior written permission of the publisher.
0-07-164311-7
The material in this eBook also appears in the print version of this title: 0-07-148858-8.
All trademarks are trademarks of their respective owners. Rather than put a trademark
symbol after every occurrence of a trademarked name, we use names in an editorial fashion only,
and to the benefit of the trademark owner, with no intention of infringement of the trademark.
Where such designations appear in this book, they have been printed with initial caps.
McGraw-Hill eBooks are available at special quantity discounts to use as premiums and sales
promotions, or for use in corporate training programs. For more information, please contact
George Hoare, Special Sales, at george_hoare@mcgraw-hill.com or (212) 904-4069.
TERMS OF USE
This is a copyrighted work and The McGraw-Hill Companies, Inc. (“McGraw-Hill”) and its
licensors reserve all rights in and to the work. Use of this work is subject to these terms. Except as
permitted under the Copyright Act of 1976 and the right to store and retrieve one copy of the work,
you may not decompile, disassemble, reverse engineer, reproduce, modify, create derivative works
based upon, transmit, distribute, disseminate, sell, publish or sublicense the work or any part of it
without McGraw-Hill’s prior consent. You may use the work for your own noncommercial and
personal use; any other use of the work is strictly prohibited. Your right to use the work may be
terminated if you fail to comply with these terms.
THE WORK IS PROVIDED “AS IS.” McGRAW-HILL AND ITS LICENSORS MAKE NO
GUARANTEES OR WARRANTIES AS TO THE ACCURACY, ADEQUACY OR COMPLETE-
NESS OF OR RESULTS TO BE OBTAINED FROM USING THE WORK, INCLUDING ANY
INFORMATION THAT CAN BE ACCESSED THROUGH THE WORK VIA HYPERLINK OR
OTHERWISE, AND EXPRESSLY DISCLAIM ANY WARRANTY, EXPRESS OR IMPLIED,
INCLUDING BUT NOT LIMITED TO IMPLIED WARRANTIES OF MERCHANTABILITY
OR FITNESS FOR A PARTICULAR PURPOSE. McGraw-Hill and its licensors do not warrant
or guarantee that the functions contained in the work will meet your requirements or that its
operation will be uninterrupted or error free. Neither McGraw-Hill nor its licensors shall be liable
to you or anyone else for any inaccuracy, error or omission, regardless of cause, in the work or for
any damages resulting therefrom. McGraw-Hill has no responsibility for the content of any
information accessed through the work. Under no circumstances shall McGraw-Hill and/or its
licensors be liable for any indirect, incidental, special, punitive, consequential or similar damages
that result from the use of or inability to use the work, even if any of them has been advised of the
possibility of such damages. This limitation of liability shall apply to any claim or cause
whatsoever whether such claim or cause arises in contract, tort or otherwise.
DOI: 10.1036/0071488588
To Dr. Larry C. Gilstrap III, whose encouragement is largely responsible for
my writing this series of books. He has been a personal inspiration, mentor,
and role model of an outstanding physician, teacher, and leader;
and to Dr. Edward Yeomans who has been a dear friend and
gleaming light of brilliance in obstetrics.
ECT
To Dr. George Stancel who encouraged my passion for education;
to the medical students of the University of Texas, Medical School at Houston
who have consistently and constructively challenged me to be the
best teacher that I can be;
and to my special children and grandchildren,
Sydney, Stephanie, and Jacob.
GCR
To the medical and graduate students of the
University of Texas Health Science Center in Houston who continually challenge
and make both teachingand research far more interesting;
and to William and Jane
for their patience and encouragement during the writing
and editing of the manuscript.
DSL
To the dedicated staff, residents and faculty of the
Methodist Hospital Family Medicine Residency and Denver Harbor Clinic,
with whom I am privileged to work.
DB
DEDICATION
Copyright © 2008 by The McGraw-Hill Companies, Inc. Click here for terms of use.
This page intentionally left blank
CONTENTS
CONTRIBUTORS vii
ACKNOWLEDGMENTS ix
INTRODUCTION xi
SECTION I
Applying the Basic Sciences to Clinical Medicine 1
Part 1. Approach to Learning Pharmacology 3
Part 2. Approach to Disease 4
Part 3. Approach to Reading 5
SECTION II
Clinical Cases 11
Fifty-Three Case Scenarios 13
SECTION III
Listing of Cases 401
Listing by Case Number 403
Listing by Disorder (Alphabetical) 404
INDEX 407
For more information about this title, click here
This page intentionally left blank
CONTRIBUTORS
Jeané Simmons Holmes, MD, FACOG
Assistant Clinical Professor
Obstetrics and Gynecology Residency Program
The Methodist Hospital, Houston
Houston, Texas
Polycystic Ovarian Syndrome
Lacy Kessler
Medical Student
Class of 2008
The University of Texas Medical School at Houston
Houston, Texas
Ergot Akyloids
Eicosanoids
Priti P. Schachel, MD
Assistant Professor
Department of Obstetrics and Gynecology
Weill Medical College of Cornell University
The Methodist Hospital, Houston
Houston, Texas
Polycystic Ovarian Syndrome
vii
Copyright © 2008 by The McGraw-Hill Companies, Inc. Click here for terms of use.
This page intentionally left blank
ix
ACKNOWLEDGMENTS
The inspiration for this basic science series occurred at an educational retreat
led by Dr. L. Maximilian Buja, who at the time was the dean of the medical
school. It has been such a joy to work together with Drs. Gary Rosenfeld and
David Loose, who are both accomplished scientists and teachers. It has been
rewarding to collaborate with Dr. Donald Briscoe, a scholar and an excellent
teacher. I would like to thank McGraw-Hill for believing in the concept of
teaching by clinical cases. I owe a great debt to Catherine Johnson, who has
been a fantastically encouraging and enthusiastic editor.
At the University of Texas Medical School at Houston, we would like to
recognize the bright and enthusiastic medical students who have inspired us to
find better ways to teach. At The Methodist Hospital, I appreciate the support
from Dr. Mark Boom, Dr. Karin Larsen Pollock, Mr. Reggie Abraham, Mr.
John Lyle, and our fabulous Department Chair, Dr. Alan Kaplan. At St. Joseph
Medical Center, I would like to recognize some of the finest administrators I
have encountered: Phil Robinson, Pat Mathews, Laura Fortin, Dori Upton,
Cecile Reynolds, John Bertini, MD, and Thomas V. Taylor, MD. I appreciate
Marla Buffington’s excellent advice and assistance. Without the help from my
colleague and friend, Dr. John C. McBride, this book could not have been writ-
ten. Most importantly, I am humbled by the love, affection, and encouragement
from my lovely wife, Terri, and our four children, Andy, Michael, Allison, and
Christina.
Eugene C. Toy
Copyright © 2008 by The McGraw-Hill Companies, Inc. Click here for terms of use.
This page intentionally left blank
INTRODUCTION
Often, the medical student will cringe at the “drudgery” of the basic science
courses and see little connection between a field such as pharmacology and
clinical problems. Clinicians, however, often wish they knew more about the
basic sciences, because it is through the science that we can begin to under-
stand the complexities of the human body and thus have rational methods of
diagnosis and treatment.
Mastering the knowledge in a discipline such as pharmacology is a formi-
dable task. It is even more difficult to retain this information and to recall it
when the clinical setting is encountered. To accomplish this synthesis, phar-
macology is optimally taught in the context of medical situations, and this is
reinforced later during the clinical rotations. The gulf between the basic sci-
ences and the patient arena is wide. Perhaps one way to bridge this gulf is with
carefully constructed clinical cases that ask basic science-oriented questions.
In an attempt to achieve this goal, we have designed a collection of patient
cases to teach pharmacology-related points. More importantly, the explanations
for these cases emphasize the underlying mechanisms and relate the clinical
setting to the basic science data. The principles are explored rather than
overemphasizing rote memorization.
This book is organized for versatility: to allow the student “in a rush” to go
quickly through the scenarios and check the corresponding answers and to
provide more detailed information for the student who wants thought-
provoking explanations. The answers are arranged from simple to complex: a
summary of the pertinent points, the bare answers, a clinical correlation, an
approach to the pharmacology topic, a comprehension test at the end for rein-
forcement or emphasis, and a list of references for further reading. The clini-
cal cases are arranged by system to better reflect the organization within the
basic science. Finally, to encourage thinking about mechanisms and relation-
ships, we used open-ended questions in the clinical cases. Nevertheless, sev-
eral multiple-choice questions are included at the end of each scenario to
reinforce concepts or introduce related topics.
HOW TO GET THE MOST OUT OF THIS BOOK
Each case is designed to introduce a clinically related issue and includes open-
ended questions usually asking a basic science question, but at times, to break
up the monotony, there will be a clinical question. The answers are organized
into four different parts:
xi
Copyright © 2008 by The McGraw-Hill Companies, Inc. Click here for terms of use.
xii INTRODUCTION
PART I
1. Summary
2. A straightforward answer is given for each open-ended question.
3. Clinical Correlation—A discussion of the relevant points relating the
basic science to the clinical manifestations, and perhaps introducing
the student to issues such as diagnosis and treatment.
PART I I
An approach to the basic science concept consisting of three parts:
1. Objectives—A listing of the two to four main knowledge objectives
that are critical for understanding the underlying pharmacology to
answer the question and relate to the clinical situation.
2. Definitions of basic terminology.
3. Discussion of the specific class of agents.
PART III
Comprehension Questions—Each case includes several multiple-choice
questions that reinforce the material or introduces new and related concepts.
Questions about the material not found in the text are explained in the answers.
PART I V
Pharmacology Pearls—A listing of several important points, many clinically
relevant, reiterated as a summation of the text and to allow for easy review,
such as before an examination.
SECTION I
Applying the
Basic Sciences to
Clinical Medicine
Part 1. Approach to Learning Pharmacology
Part 2. Approach to Disease
Part 3. Approach to Reading
Copyright © 2008 by The McGraw-Hill Companies, Inc. Click here for terms of use.
This page intentionally left blank
PART 1. APPROACH TO LEARNING PHARMACOLOGY
Pharmacology is best learned by a systematic approach, understanding the
physiology of the body, recognizing that every medication has desirable and
undesirable effects, and being aware that the biochemical and pharmacologic
properties of a drug affects its characteristics such as duration of action, volume
of distribution, passage through the blood-brain barrier, mechanism of elimi-
nation, and route of administration. Rather than memorizing the characteris-
tics of a medication, the student should strive to learn the underlying rationale
such as, “Second-generation antihistamine agents are less lipid soluble than
first-generation antihistamines and therefore do not cross the blood-brain bar-
rier as readily; thus, second-generation antihistamines are not as sedating.
Because they both bind the histamine H
1
receptor, the efficacy is the same.”
KEY TERMS
Pharmacology: The study of substances that interact with living systems
through biochemical processes.
Drug:A substance used in the prevention, diagnosis, or treatment of disease.
Toxicology:A branch of pharmacology that studies the undesirable effects
of chemicals on living organisms.
Food and Drug Administration (FDA):The federal agency responsible
for the safety and efficacy of all drugs in the United States, as well as
food and cosmetics.
Adverse effect:Also known as side effect; all unintended actions of a drug
that result from the lack of specificity of drug action. All drugs are capa-
ble of producing adverse effects.
Pharmacodynamics: The actions of a drug on a living organism, includ-
ing mechanisms of action and receptor interaction.
Pharmacokinetics:The actions of the living organism on the drug, includ-
ing absorption, distribution, and elimination.
Volume of distribution (V
d
): The size of the “compartment” into which a
drug is distributed following absorption and is determined by the equation:
V
d
= Dose (mg) drug administered/Initial plasma concentration (mg/L)
Potency of drug: Relative amount of drug needed to produce a given
response, determined largely by the amount of drug that reaches the site
of action and by the affinity of the drug for the receptor.
Efficacy:Drug effect as the maximum response it is able to produce and is
determined by the number of drug-receptor complexes and the ability of
the receptor to be activated once bound. EC-50 refers to the drug con-
centration that produces 50 percent of the maximal response, whereas
ED-50 refers to the drug dose that is pharmacologically effective in
50 percent of the population.
APPLYING THE BASIC SCIENCES TO CLINICAL MEDICINE 3
Absorption:The movement of a drug from the administration site into the
blood stream usually requiring the crossing of one or more biologic
membranes. Important parameters include lipid solubility, ionization,
size of the molecule, and presence of a transport mechanism.
Elimination:Process by which a drug is removed from the body, generally
by either metabolism or excretion. Elimination follows various kinetic
models. For example, first-order kinetics describes most circum-
stances, and means that the rate of drug elimination depends on the con-
centration of the drug in the plasma as described by the equation:
Rate of elimination from body= Constant× Drug concentration
Zero-order kinetics:It is less common and means that the rate of elimina-
tion is constant and does not depend on the plasma drug concentration.
This may be a consequence of a circumstance such as saturation of liver
enzymes or saturation of the kidney transport mechanisms.
Bioavailability:The percentage of an ingested drug that is actually absorbed
into the bloodstream.
Route of administration:Drug may be delivered intravenously (IV or iv) for
delivery directly into the bloodstream, intramuscularly(IM), and subcu-
taneously (SC). The medication may be depot and slow release, inhalant
for rapid absorption and delivery to the bronchi and lungs, sublingual to
bypass the first-pass effect, intrathecal for agents that penetrate the
blood-brain barrier poorly, rectal to avoid hepatic first-pass effect and
for nausea, and topical administration when local effect is desired such
as dermatologic or ophthalmic agents.
PART 2. APPROACH TO DISEASE
Physicians usually tackle clinical situations by taking a history (asking
questions), performing a physical examination, obtaining selective labora-
tory and imaging tests, and then formulating a diagnosis. The synthesis of
the history, physical examination, and imaging or laboratory tests is called
the clinical database. After reaching a diagnosis, a treatment plan is usu-
ally initiated, and the patient is followed for a clinical response. Rational
understanding of disease and plans for treatment are best acquired by learn-
ing about the normal human processes on a basic science level; likewise,
being aware of how disease alters the normal physiologic processes is also
best understood on a basic science level. Pharmacology and therapeutics
require also the ability to tailor the correct medication to the patient’s situ-
ation and awareness of the medication’s adverse effect profile. Sometimes,
the patient has an adverse reaction to a medication as the chief complaint,
and the physician must be able to identify the medication as the culprit. An
understanding of the underlying basic science allows for more rational
analysis and medication choices.
4 CASE FILES: PHARMACOLOGY
PART 3. APPROACH TO READING
There are seven key questions that help to stimulate the application of basic
science information to the clinical setting. These are:
1. Which of the available medications is most likely to achieve the
desired therapeutic effect and/or is responsible for the described
symptoms or signs?
2. What is the likely mechanism for the clinical effect(s) and adverse
effect(s) of the medication?
3. What is the basic pharmacologic profile (e.g., absorption, elimina-
tion) for medications in a certain class, and what are the differ-
ences among the agents within the class?
4. Given basic pharmacologic definitions such as therapeutic index
(TI) or certain safety factor (TD
1
/ED
99
), or median lethal dose
(LD
50
), how do medications compare in their safety profile?
5. Given a particular clinical situation with described unique patient
characteristics, which medication is most appropriate?
6. What is the best treatment for the toxic effect of a medication?
7. What are the drug-drug interactions to be cautious about regard-
ing a particular medication?
1. Which of the following medications is most likely to be responsible
for the described symptoms or signs?
The student must be aware of the various effects, both desirable and
undesirable, produced by particular medications. Knowledge of desir-
able therapeutic effects is essential in selecting the appropriate drug for
the particular clinical application; likewise, an awareness of its adverse
effects is necessary, because patients may come into the physician’s
office with a complaint caused by a drug effect unaware that their
symptoms are because of a prescribed medication. It is only by being
aware of the common and dangerous effects that the clinician can
arrive at the correct diagnosis. The student is encouraged not to merely
memorize the comparative adverse effect profiles of the drugs, but
rather to understand the underlying mechanisms.
2. What is the likely mechanism for the clinical effect(s) and adverse
effect(s) of the medication?
As noted above, the student should strive to learn the underlying
physiologic, biochemical, or cellular explanation for the described
drug effect. This understanding allows for the rational choice of an
alternative agent or the reasonable choice of an agent to alleviate the
symptoms or explanatory advice to the patient regarding behavioral
changes to diminish any adverse affects. For example, if a 60-year-old
woman who takes medications for osteoporosis complains of severe
“heartburn,” one may be suspicious, knowing that the bisphosphonate
APPLYING THE BASIC SCIENCES TO CLINICAL MEDICINE 5
medication alendronate can cause esophagitis. Instruction to the
patient to take the medication while sitting upright and remaining
upright for at least 30 minutes would be the proper course of action,
because gravity will assist in keeping the alendronate in the stomach
rather than allowing regurgitation into the distal esophagus.
3. What is the basic pharmacologic profile (absorption, elimination,
volume of distribution) for medications in a certain class, and what
are differences among the agents within the class?
Understanding the pharmacologic profile of medications allows
for rational therapeutics. However, instead of memorizing the sepa-
rate profiles for every medication, grouping the drugs together into
classes allows for more efficient learning and better comprehension.
An excellent starting point for the student of pharmacology would be
to study how a prototype drug within a drug class organized by
structure or mechanism of action may be used to treat a condition
(such as hypertension). Then within each category of agents, the stu-
dent should try to identify important subclasses or drug differences.
For example, hypertensive agents can be categorized as diuretic
agents, β-adrenergic-blocking agents, calcium-channel-blocking
agents, and renin-angiotensin system inhibitors. Within the subclas-
sification of renin-angiotensin system inhibitors, the angiotensin-
converting enzyme inhibitors can cause the side effect of a dry cough
caused by the increase in bradykinin brought about by the enzyme
blockade; instead, the angiotensin-1 receptor blockers do not affect
the bradykinin levels and so do not cause the cough as often.
4. Given basic pharmacologic definitions such as therapeutic index
(TI) or certain safety factor (TD
1
/ED
99
), or median lethal dose
(LD
50
), how do medications compare in their safety profile?
Therapeutic index (TI): Defined as the TD
50
/ED
50
(the ratio of the
dose that produces a toxic effect in half the population to the dose that
produces the desired effect in half the population).
Certain safety factor (TD
1
/ED
99
): Defined as the ratio of the dose that
produces the toxic effect in 1 percent of the population to the dose that
produces the desired effect in 99 percent of the population; also known
as standard safety measure.
Median lethal dose (LD
50
): Defined as the median lethal dose, the
dose that will kill half the population.
Based on these definitions, a desirable medication would have a high
therapeutic index (toxic dose is many times that of the efficacious
dose), high certain safety factor, and high median lethal dose (much
higher than therapeutic dose). Likewise, medications such as digoxin
that have a low therapeutic index require careful monitoring of levels
and vigilance for side effects.
6 CASE FILES: PHARMACOLOGY
5. Given a particular clinical situation with described unique patient
characteristics, which medication is most appropriate?
The student must weigh various advantages and disadvantages, as
well as different patient attributes. Some of those may include compli-
ance with medications, allergies to medications, liver or renal insuffi-
ciency, age, coexisting medical disorders, and other medications. The
student must be able to sift through the medication profile and identify
the most dangerous adverse effects. For example, if a patient is already
taking a monoamine-oxidase-inhibiting agent for depression, then
adding a serotonin reuptake inhibitor would be potentially fatal, because
serotonin syndrome may ensue (hyperthermia, muscle rigidity, death).
6. What is the best treatment for the toxic effect of a medication?
If complications of drug therapy are present, the student should know
the proper treatment. This is best learned by understanding the drug
mechanism of action. For example, a patient who has taken excessive opi-
oids may develop respiratory depression, caused by either a heroin over-
dose or pain medication, which may be fatal. The treatment of an opioid
overdose includes the ABCs (airway, breathing, circulation) and the
administration of naloxone, which is a competitive antagonist of opioids.
7. What are the drug-drug interactions to be concerned with regard-
ing a particular medication?
Patients are often prescribed multiple medications, from either the
same practitioner or different clinicians. Patients may not be aware of
the drug-drug interactions; thus, the clinician must compile, as a com-
ponent of good clinical practice, a current list of all medications (pre-
scribed, over-the-counter, and herbal) taken by the patient. Thus, the
student should be aware of the most common and dangerous interac-
tions; once again, understanding the underlying mechanism allows for
lifelong learning rather than short-term rote memorization of facts that
are easily forgotten. For example, magnesium sulfate to stop preterm
labor should not be used if the patient is taking a calcium-channel-
blocking agent such as nifedipine. Magnesium sulfate acts as a com-
petitive inhibitor of calcium, and by decreasing its intracellular
availability it slows down smooth muscle contraction such as in the
uterus. Calcium-channel blockers potentiate the inhibition of calcium
influx and can lead to toxic effects, such as respiratory depression.
COMPREHENSION QUESTIONS
[I.1] Bioavailability of an agent is maximal when the drug has which of the
following qualities?
A. Highly lipid soluble
B. More than 100 Daltons in molecular weight
C. Highly bound to plasma proteins
D. Highly ionized
APPLYING THE BASIC SCIENCES TO CLINICAL MEDICINE 7
[I.2] An agent is noted to have a very low calculated volume of distribution
(V
d
). Which of the following is the best explanation?
A. The agent is eliminated by the kidneys, and the patient has renal
insufficiency.
B. The agent is extensively bound to plasma proteins.
C. The agent is extensively sequestered in tissue.
D. The agent is eliminated by zero-order kinetics.
[I.3] Which of the following describes the first-pass effect?
A. Inactivation of a drug as a result of the gastric acids.
B. Absorption of a drug through the duodenum.
C. Drug given orally is metabolized by the liver before entering the
circulation.
D. Drug given IV accumulates quickly in the central nervous system
(CNS).
[I.4] A laboratory experiment is being conducted in which a mammal is
injected with a noncompetitive antagonist to the histamine receptor.
Which of the following best describes this agent?
A. The drug binds to the histamine receptor and partially activates
it.
B. The drug binds to the histamine receptor but does not activate it.
C. The drug binds to the receptor, but not where histamine binds, and
prevents the receptor from being activated.
D. The drug irreversibly binds to the histamine receptor and renders it
ineffective.
[I.5] A 25-year-old medical student is given a prescription for asthma,
which the physician states has a very high therapeutic index. Which
of the statements best characterizes the drug as it relates to the ther-
apeutic index?
A. The drug’s serum levels will likely need to be carefully monitored.
B. The drug is likely to cross the blood-brain barrier.
C. The drug is likely to have extensive drug-drug interactions.
D. The drug is unlikely to have any serious adverse effects.
[I.6] A drug M is injected IV into a laboratory subject. It is noted to have
high serum protein binding. Which of the following is most likely to be
increased as a result?
A. Drug interaction
B. Distribution of the drug to tissue sites
C. Renal excretion
D. Liver metabolism
8 CASE FILES: PHARMACOLOGY
[I.7] A bolus of drug K is given IV. The drug is noted to follow first-order
kinetics. Which of the following describes the elimination of drug K?
A. The rate of elimination of drug K is constant.
B. The rate of elimination of drug K is proportional to the patient’s
renal function.
C. The rate of elimination of drug K is proportional to its concentra-
tion in the patient’s plasma.
D. The rate of elimination of drug K is dependent on a nonlinear rela-
tionship to the plasma protein concentration.
Answers
[I.1] A. Transport across biologic membranes and thus bioavailability is
maximal with high lipid solubility.
[I.2] B. The volume of distribution is calculated by administering a known
dose of drug (mg) IV and then measuring an initial plasma concen-
tration (mg/L). The ratio of the mass of drug given (mg) divided by
the initial plasma concentration (mg/L) gives the V
d
. A very low V
d
may indicate extensive protein binding (drug is sequestered in the
bloodstream), whereas a high V
d
may indicate extensive tissue bind-
ing (drug is sequestered in the tissue).
[I.3] C. The first-pass effect refers to the process in which following oral
administration a drug is extensively metabolized as it initially passes
through the liver, before it enters the general circulation. Liver
enzymes may metabolize the agent to such an extent that the drug
cannot be administered orally.
[I.4] C. A noncompetitive antagonist binds to the receptor at a site other
than the agonist-binding site and renders it less effective by prevent-
ing agonist binding or preventing activation.
[I.5] D. An agent with a high therapeutic index means the toxic dose is
very much higher than the therapeutic dose, and it is less likely to
produce toxic effects at therapeutic levels.
[I.6] A. High protein binding means less drug to the tissue, the kidney, and
the liver. Drug interaction may occur if the agent binds to the same
protein site as other drugs, thus displacing drugs and increasing
serum levels.
[I.7] C. First-order kinetics means the rate of elimination of a drug is pro-
portional to the plasma concentration.
APPLYING THE BASIC SCIENCES TO CLINICAL MEDICINE 9
REFERENCES
Braunwald E, Fauci AS, Kasper KL, et al., eds. Harrison’s Principles of Internal
Medicine, 16th ed. New York: McGraw-Hill, 2004.
Rosenfeld GC, Loose-Mitchell DS. Pharmacology, 4th ed. Philadelphia, PA:
Lippincott, Williams & Wilkins, 2007:1.
10 CASE FILES: PHARMACOLOGY
PHARMACOLOGY PEARLS
Understanding the pharmacologic mechanisms of medications allows
for rational choices for therapy, fewer medication errors, and rapid
recognition and reversal of toxic effects.
The therapeutic index, certain safety factor (TD
1
/ED
99
), and median
lethal dose are various methods of describing the potential toxic-
ity of medications.
There are seven key questions to stimulate the application of basic
science information to the clinical arena.
SECTION II
Clinical Cases
Copyright © 2008 by The McGraw-Hill Companies, Inc. Click here for terms of use.
This page intentionally left blank
CASE 1
A 12-year-old girl presents to your office with a sore throat and fever. You
diagnose her with pharyngitis caused by group A β-hemolyticStreptococcus.
She is given an IM injection of penicillin. Approximately 5 minutes later, she
is found to be in respiratory distress and audibly wheezing. Her skin is mot-
tled and cool, she is tachycardic (rapid heart rate), and her blood pressure has
fallen to 70/20 mm Hg. You immediately diagnose her as having an anaphy-
lactic reaction to the penicillin and give an SC injection of epinephrine.
What effect will epinephrine have on this patient’s vascular system?
Which adrenoceptor primarily mediates the vascular response?
What effect will epinephrine have on her respiratory system?
Which adrenoceptor primarily mediates the respiratory system
response?
ANSWERS TO CASE 1: AUTONOMIC SYMPATHETIC
NERVOUS SYSTEM
Summary:A 12-year-old girl with “strep throat” is given an injection of peni-
cillin and develops an acute anaphylactic reaction.
Effect of epinephrine on vascular system:Vasoconstriction.
Adrenoceptor which primarily mediates the vascular response:
Alpha-1 (α
1
).
Effect of epinephrine on the pulmonary system: Bronchial muscle
relaxation.
Adrenoceptor which primarily mediates the pulmonary response:
Beta-2 (β
2
).
CLINICAL CORRELATION
Anaphylaxis is an acute, immune-mediated response to an allergen character-
ized by bronchospasm, wheezing, tachycardia, and hypotension. Epinephrine is
the drug of choice used to treat this condition because it appears, through the
activation of alpha (α)- and beta (β)-adrenoceptors, to counteract the patho-
physiologic processes underlying anaphylaxis. As with all emergencies, the
ABCs (airway, breathing, circulation) should be addressed first. Occasionally,
the anaphylaxis causes laryngeal edema to the extent that the airway is com-
promised, and intubation (placement of a tube in the trachea) is impossible. In
these circumstances, an emergency airway, such as a surgical cricothyroido-
tomy (creating an opening from the skin through the cricoid cartilage), is
required.
APPROACH TO THE AUTONOMIC SYMPATHETIC
NERVOUS SYSTEM
Objectives
1. List the neurotransmitters of the autonomic sympathetic nervous sys-
tem and describe their anatomical localization.
2. List the receptors and receptor-subtypes of the autonomic sympathetic
nervous system.
3. Predict the responses to activation and inhibition of autonomic sympa-
thetic nervous system receptors.
14
CASE FILES: PHARMACOLOGY
Definitions
Autonomic nervous system: Subdivision of the efferent peripheral nerv-
ous system that is largely under unconscious control (the somatic subdi-
vision of the peripheral nervous system is largely under conscious
control), shown in Figure 1-1.
Sympathetic nervous system:A division of the autonomic nervous system
(the other is the parasympathetic nervous system) that originates in nuclei
of the CNS. Preganglionic fibers exit through the thoracic and lumbar
spinal nerves to synapse on ganglia close to the spinal cord and also on
the adrenal medulla (considered modified ganglia). Postganglionic fibers
innervate a wide variety of effector organs and tissues, including arteriole
and bronchial smooth muscles.
Agonist: A drug that activates a receptor and results in a pharmacologic
response.
Antagonist:A drug that binds to receptors with little or no effect of its own,
but that can block the action of an agonist that binds to the same receptors.
DISCUSSION
Class
The neurotransmitter norepinephrine is released from the efferent nerves of
the sympathetic autonomic nervous system at postganglionic sympathetic
(also known as “adrenergic”) nerve endings, whereas epinephrine and some
norepinephrine are released from the adrenal medulla.
These endogenous catecholamine neurotransmitter agonists interact at
postjunctional specialized cell membrane components called adrenoceptors
(named after the adrenergic nerves that innervate them) that are classified as
either alpha (α) or beta (β).
There are two subtypes of the a-adrenoceptor, a
1
and a
2
, each of which
has variants of unclear pharmacologic importance. Activation of the autonomic
sympathetic nervous system α
1
-adrenoceptors by adrenergic agonists results in,
among other effects, contraction of most vascular smooth muscle (a
1
) caus-
ing increased peripheral resistance and blood pressure, contraction of the
pupillary dilator muscle resulting in mydriasis, an indirectly mediated
(through inhibition of acetylcholine [Ach] release) relaxation of gastrointesti-
nal smooth muscle and contraction of gastrointestinal sphincters (α
1
), and
activation of the seminal vesicles, prostate gland, and ductus deferens that
results in ejaculation. Activation of prejunctional adrenoceptor autoreceptors
(α
2
) by catecholamines results in (feedback) inhibition of the release of norep-
inephrine and other neurotransmitters from their respective nerve endings.
There are also three subtypes of the b-adrenoceptor, b
1
, b
2
, and b
3
.
Activation of the autonomic sympathetic nervous system α-adrenoceptors by
adrenergic agonists results in, among other effects, increased rate and force of
CLINICAL CASES 15
contraction of the heart (b
1
), smooth muscle relaxation of bronchi causing
bronchodilation (b
2
), and activation of fat cell lipolysis (b
3
).
Because the catecholamines epinephrine and norepinephrine have
important physiologic roles, drugs that block their actions, that is, adreno-
ceptor antagonists, can have important pharmacologic effects, many of
which are clinically useful. a-Adrenoceptor nonselective antagonists
(e.g., phentolamine) are used to treat the hypertension of pheochromocy-
toma (a tumor that secretes catecholamines) and male erectile dysfunc-
tion, whereas the more selective a
1
-adrenoceptor antagonists (e.g.,
prazosin, terazosin, doxazosin) are used to treat hypertension and benign
prostatic hyperplasia(Table 1-1).
16
CASE FILES: PHARMACOLOGY
Constricts
pupil of eye
Stimulates
salivary glands
Slows heart
Constricts bronchii
in lungs
Stimulates activity
of stomach
and intestines
Stimulates activity
of pancreas
Stimulates
gallbladder
Promotes voiding
from bladder
Promotes erection
of genitals
Dilates
pupil of eye
Sympathetic
Nervous System
Parasympathetic
Nervous System
Inhibits
salivary gland
secretion
Accelerates heart
Relaxes bronchii
in lungs
Inhibits activity
of stomach
and intestines
Inhibits activity
of pancreas
Stimulates
glucose release
from liver; inhibits
gallbladder
Inhibits voiding
from bladder
Stimulates
adrenal medulla
Promotes
ejaculation and
vaginal
contractions
Figure 1-1. Schematic of autonomic nervous system.
Structure
Epinephrine and norepinephrine are catecholamines, synthesized from
tyrosine, that possess a catechol nucleus with an ethylamine side chain (epi-
nephrine is the methylated side chain derivative of norepinephrine). The rate-
limiting enzyme in this process is tyrosine hydroxylase.
Mechanism of Action
Epinephrine binds to α
1
-adrenoceptors and, through a G-protein (Gq-type
GTP-binding protein [Gq])-mediated activation of phospholipase C and stimu-
lation of polyphosphoinositide hydrolysis, results in formation of inositol 1,4,5-
trisphosphate (IP
3
) that promotes the release of stored intracellular Ca
2+
.
Epinephrine interaction with α
2
-adrenoceptors results in activation of a Gi-type
GTP-binding protein (Gi) to inhibit adenylyl cyclase activity, thereby decreas-
ing cyclic adenosine monophosphate (cAMP) levels. Epinephrine perhaps also
increases β
1
-adrenoceptor-mediated influx of Ca
2+
across membrane channels.
In addition to the increased formation of the “second messenger” IP
3
, epi-
nephrine also increases the phospholipase-mediated formation of another sec-
ond messenger diacylglycerol (DAG) that activates protein kinase C that
influences the activity of a number of other signaling pathways. Epinephrine
also activates b
1
- and b
2
-adrenoceptors to increase a G-protein-mediated
stimulation of adenylyl cyclase activity, thereby increasing intracellular
cAMP levelsand the activity of cAMP-dependent protein kinases.
CLINICAL CASES 17
Table 1-1
SELECTED EFFECTS OF ADRENOCEPTOR ACTIVATION
ORGAN EFFECTS (ADRENOCEPTOR SUBTYPE)
Bronchial smooth muscle Dilates (β
2
)
Heart rate and contractile force Increases (β
1
)
Eye (pupil size) Dilates (α
1
)
*
Blood vessels Constrict (α
1
)
†,‡
Gastrointestinal tract Decrease (α
1
,β
2
)
(tone, motility, secretions)
Pancreas (insulin release) Decrease (α
2
)
Dilation (mydriasis)results from α
1
-adrenoceptor stimulation of the radial muscle.
Skeletal muscle blood vessels have β
2
-adrenoceptors that, when activated, result in vessel
constriction.
Coronary arteries also have β-adrenoceptors that, when activated, result in vessel dilation,
whichis the dominant effect.
Administration
Epinephrine is generally administered parenterally (IM) for treatment of
anaphylactic shock. For this and other conditions, it is also available as IV, SC,
ophthalmic, nasal, and aerosol preparations. Norepinephrine is only available
for parenteral, generally IV, administration.
Pharmacokinetics
Epinephrine released from the adrenal gland is metabolized primarily by
catechol-O-methyltransferase (COMT) and monoamine oxidase (MAO).
The action of norepinephrine released from nerve endings is terminated
primarily by reuptake into nerve terminals (uptake 1) and other cells
(uptake 2).
COMPREHENSION QUESTIONS
[1.1] A patient with septic shock is noted to have persistent hypotension
despite dopamine infusion. Epinephrine in an IV infusion is used.
With which adrenoceptor does epinephrine act to constrict vascular
smooth muscle?
A. α
1
-Adrenoceptors
B. α
2
-Adrenoceptors
C. β
1
-Adrenoceptors
D. β
2
-Adrenoceptors
[1.2] A 16-year-old male is having an acute asthmatic attack. Epinephrine is
given SC. With which of the following adrenoceptors does epinephrine
act to dilate bronchial smooth muscle?
A. α
1
-Adrenoceptors
B. α
2
-Adrenoceptors
C. β
1
-Adrenoceptors
D. β
2
-Adrenoceptors
[1.3] Which of the following best describes the cellular action of epinephrine?
A. Activation of adenylyl cyclase
B. Decreased activity of cAMP-dependent protein kinases
C. Increased intracellular stores of Ca
2+
D. Inhibition of the activity of phospholipase
[1.4] Epinephrine-mediated β
1
-adrenoceptor activation results in which of
the following?
A. Constriction of bronchial smooth muscle
B. Decreased gastrointestinal motility
C. Dilation of the pupils
D. Increased heart rate
18
CASE FILES: PHARMACOLOGY
Answers
[1.1] A.α
1
-Adrenoceptors mediate vasoconstriction in many vascular beds.
In skeletal muscle, epinephrine can act at β
2
-adrenoceptors to cause
vasodilation.
[1.2] D. Epinephrine acts on β
2
-adrenoceptors to cause smooth muscle
relaxation of bronchi resulting in bronchodilation. Because of
adverse cardiovascular effects of epinephrine (β
1
), more selective β
2
-
adrenoceptor agonists are now used (e.g., albuterol).
[1.3] A.Epinephrine activates α
1
-adrenoceptors to cause a release of intra-
cellular stored Ca
2+
andβ
1
- and β
2
-adrenoceptors to activate adenylyl
cyclase.
[1.4] D. Epinephrine activation of β
1
-adrenoceptors results in an
increase in heart rate. Activation of α
1
-adrenoceptors results in
dilation of the pupil. Activation of β
2
-adrenoceptors causes dila-
tion of bronchial smooth muscle and decreased gastrointestinal
(GI) motility.
CLINICAL CASES 19
PHARMACOLOGY PEARLS
Physiologically epinephrine acts as a hormone on distant cells after
its release from the adrenal medulla.
As exceptions, sympathetic postganglionic neurons that innervate
sweat glands and renal vascular smooth muscle release, respec-
tively, ACh and dopamine rather than norepinephrine.
Exogenously administered epinephrine increases blood pressure
through its action on β
1
-adrenoceptors in the heart, resulting in
increased heart rate and force of contraction and through its
action on α
1
-adrenoceptors in many vascular beds that results in
vasoconstriction.
In skeletal muscle, epinephrine injection can result in vasodilation
(β
2
) that in some cases may lead to a decreased total peripheral
resistance and a decrease in diastolic pressure.
Norepinephrine has little, if any, effect on β
2
-adrenoceptors (nor-
epinephrine and epinephrine have similar effects on α- and β
1
-
adrenoceptors), thus increasing both systolic and diastolic
blood pressure.
20 CASE FILES: PHARMACOLOGY
REFERENCES
Goldstein DS, Robertson D, Straus SE, et al. Dysautonomias: clinical disorders of
the autonomic nervous system. Ann Intern Med 2002;137(9):753–63.
Brown SG. Cardiovascular aspects of anaphylaxis: implications for treatment and
diagnosis. Curr Opin Allergy Clin Immunol 2005;5(4):359–64.
Clark AL, Cleland JG. The control of adrenergic function in heart failure: thera-
peutic intervention. Heart Fail Rev 2000;5(1):101–14.
August P. Initial treatment of hypertension. N Engl J Med 2003;348(7):610–7.
CASE 2
A 61-year-old man is noted to have increased intraocular pressure on a routine
eye examination. The visual acuity is normal in both eyes. The dilated eye
examination reveals no evidence of optic nerve damage. Visual field testing
shows mild loss of peripheral vision. He is diagnosed with primary open-angle
glaucoma and is started on pilocarpine ophthalmic drops.
What is the action of pilocarpine on the muscles of the iris and cilia?
What receptor mediates this action?
Is pilocarpine the appropriate first-line drug for treatment of
primary open-angle glaucoma?
ANSWERS TO CASE 2: MUSCARINIC
CHOLINOMIMETIC AGENTS
Summary: A 61-year-old man with open-angle glaucoma is prescribed pilo-
carpine ophthalmic drops.
Action of pilocarpine on muscles of the iris and cilia: Constriction of
the muscles
Receptor that mediates this action: Muscarinic cholinoreceptor
First-line drugs to treat primary open-angle glaucoma:
Prostaglandin analogs
CLINICAL CORRELATION
Open-angle glaucoma is a disease caused by obstruction of the outflow of
aqueous humor into the canal of Schlemm, causing an increase in intraocu-
lar pressure. The use of a direct-acting muscarinic agonist, such as pilo-
carpine, causes contraction of the muscles of the cilia and iris. Because
these are circular muscles, the pupil is constricted, which helps to relieve
the outflow obstruction and lower the intraocular pressure. Although not
common with the use of topical ophthalmic drops, bronchospasm and pul-
monary edema has been noted with the use of pilocarpine drops. More com-
monly, blurred vision and myopia (nearsightedness) occur as a result of the
impairment of accommodation caused by the contraction of the iris and cil-
iary muscles.
The use of a direct-acting muscarinic agonist such as pilocarpine to
treat open-angle glaucoma is now not common due to its numerous side
effects, the need to administer it up to four times per day, and the avail-
ability of other agents. Prostaglandin analogs such as latanoprost are now
considered first-line therapy for this condition followed by β-adrenoceptor
agonists.
APPROACH TO MUSCARINIC
CHOLINOMIMETIC AGENTS
Objectives
1. Be able to list the receptors of the parasympathetic nervous system.
2. Contrast the actions and effects of direct and indirect stimulation of
muscarinic cholinoreceptors.
3. List the therapeutic uses of parasympathomimetic agents.
4. List the adverse effects of parasympathomimetic agents.
22
CASE FILES: PHARMACOLOGY
Definitions
Parasympathetic nervous system:An anatomic division of the autonomic
nervous system (the other is the sympathetic nervous system) that orig-
inates in nuclei of the CNS. Preganglionic fibers exit through cranial
and sacral spinal nerves to synapse via short postganglionic nerve
fibers on ganglia, many of which are in the organs they innervate.
Cholinomimetic agents: Agents that mimic the action of ACh.These
act directly or indirectly to activate cholinoreceptors. Some directly
acting agents (pilocarpine, bethanechol, carbachol) are designed to act
selectively on either muscarinic or nicotinic cholinoreceptors, whereas
indirectly acting agents (such as neostigmine, physostigmine, edro-
phonium, demecarium), which inhibit the enzyme acetylcholinesterase
(AChE) that is responsible for the inactivation of ACh, can activate
both. Pilocarpine is a directly acting cholinomimetic agent that acts
chiefly at muscarinic cholinoreceptors. Additional selectivity of pilo-
carpine and other cholinomimetics in the treatment of glaucoma is
achieved by the use of an ophthalmic (topical) preparation.
DISCUSSION
Class
The efferent nerves of the parasympathetic autonomic nervous system
release the neurotransmitter AChat both preganglionic and postganglionic
(i.e., “cholinergic”) nerve endings, and also at somatic nerve endings. Nitric
oxide is a cotransmitter at many of the parasympathetic postganglionic sites.
TheAC h released from nerve endings of the parasympathetic nervous system
interacts at specialized cell membrane components called cholinoreceptors
that are classified as either nicotinic or muscarinicafter the alkaloids initially
used to distinguish them.
Nicotinic cholinoreceptors are localized at all postganglionic neurons
(the autonomic ganglia), including the adrenal medulla, and skeletal muscle
endplates innervated by somatic nerves. Muscarinic cholinoreceptors are
localized at organs innervated by parasympathetic postganglionic nerve end-
ings, for example, on cardiac atrial muscle, sinoatrial node cells, and atri-
oventricular node cells,where activation can cause a negative chronotropic
effect and delayed atrioventricular conduction. Cholinergic stimulation of
muscarinic receptorsin the smooth muscle, exocrine glands, and vascular
endothelium can cause, respectively, bronchoconstriction, increased acid
secretion, and vasodilation (Table 2-1).
There are two subtypes of the nicotinic cholinoreceptors: N
N
, localized to
postganglionic neurons, and N
M
, localized to the skeletal muscle endplates.
There are three pharmacologically important subtypes of the muscarinic choli-
noreceptors, M
1
, M
2
, and M
3
(two additional subtypes have been identified by
CLINICAL CASES 23
cloning), that alone or in combination are localized to sympathetic postganglionic
neurons (and the CNS), to the atrial muscle, sinoatrial (SA) cells, and atrioven-
tricular (AV) node of the heart, to smooth muscle, to exocrine glands, and to the
vascular endothelium that does not receive parasympathetic innervation.
Directly and indirectly acting parasympathetic cholinomimetic agents,
primarily pilocarpine and bethanechol, and neostigmine, are used most
often therapeutically to treat certain diseases of the eye (acute angle-closure
glaucoma), the urinary tract (urinary tract retention), the gastrointestinal
tract (postoperative ileus), salivary glands (xerostomia), and the neuromus-
cular junction (myasthenia gravis). The ACh is generally not used clinically
because of its numerous actions and very rapid hydrolysis by AChE and
pseudocholinesterase.
The adverse effects of direct- and indirect-acting cholinomimetics result
from cholinergic excess and may include diarrhea, salivation, sweating,
bronchial constriction, vasodilation, and bradycardia. Nausea and vom-
iting are also common. Adverse effects of cholinesterase inhibitors (most
often as a result of toxicity from pesticide exposure, e.g., organophos-
phates) also may include muscle weakness, convulsions, and respiratory
failure.
Structure
ACh is a choline ester that is not very lipid soluble because of its charged qua-
ternary ammonium group. It interacts with both muscarinic and nicotinic
cholinoreceptors. Choline esters similar in structure to ACh that are used thera-
peutically include methacholine, carbachol, and bethanechol. Unlike ACh and
24 CASE FILES: PHARMACOLOGY
Table 2-1
EFFECTS OF CHOLINORECEPTOR ACTIVATION
ORGAN EFFECTS
Bronchial smooth muscle Contracts
Heart rate Decreases
Eye smooth muscles Contracts
Pupil size Contracts
Accommodation
Blood vessels Dilate
*
Gastrointestinal tract (tone, motility, secretions) Increase
There is no parasympathetic innervation of blood vessels. However, they have cholinoreceptors
that when activated result in their dilation.
carbachol, methacholine and bethanechol are highly selective for muscarinic
cholinoreceptors.Pilocarpine is a tertiary amine alkaloid.
Mechanism of Action
Muscarinic cholinoreceptors activate inhibitory G-proteins (G
i
) to stimu-
late the activity of phospholipase C, which, through increased phospholipid
metabolism, results in production of inositol triphosphate (IP
3
) and DAG
that lead to the mobilization, respectively, of intracellular calciumfrom the
endoplasmic and sarcoplasmic reticulum and, through activation of protein
kinase C (PK-C), the opening of smooth muscle calcium channels with an
influx of extracellular calcium. Activation of muscarinic cholinoreceptors also
results in altered potassium flux that results in cell hyperpolarization, and in
inhibition of adenylyl cyclase activity and cAMP accumulation induced by
other hormones, including the catecholamines.
The nicotinic receptor functions as a cell membrane ligand-gated ion
channel pore. On interaction with ACh, the receptor undergoes a conforma-
tional change that results in influx of sodium with membrane depolarization
of the nerve cell or the skeletal muscle neuromuscular endplate.
Indirectly acting parasympathetic cholinomimetic agents inhibit AChE and
thereby increase ACh levels at both muscarinic and nicotinic cholinoreceptors.
Administration
Directly acting muscarinic cholinomimetic agents may be administered topically
as ophthalmic preparations (pilocarpine, carbachol), orally (bethanechol, pilo-
carpine), or parenterally (bethanechol). Depending on the agent, an indirectly act-
ing cholinesterase inhibitor may be administered topically, orally, or parenterally.
Pharmacokinetics
ACh is synthesized from choline and acetyl-coenzyme A (acetyl-CoA) by
the enzyme choline acetyltransferase and then transported into nerve end-
ing vesicles. Like ACh, methacholine, carbachol, and bethanechol are
poorly absorbed by the oral route and have limited penetration into the
CNS. Pilocarpine is more lipid soluble and can be absorbed and can
penetrate the CNS.
After release from nerve endings, ACh is rapidly metabolized into choline and
acetate, and its effects are terminated by the action of the enzymes AChE and
pseudocholinesterase. Methacholine and particularly carbachol and bethanechol
are resistant to the action of cholinesterases.
CLINICAL CASES 25
COMPREHENSION QUESTIONS
[2.1] A 62-year-old woman is noted to have open-angle glaucoma. She inad-
vertently applies excessive pilocarpine to her eyes. This may result in
which of the following?
A. Bronchial smooth muscle dilation
B. Decreased gastrointestinal motility
C. Dilation of blood vessels
D. Mydriasis
[2.2] Muscarinic cholinergic agonists
A. Activate inhibitory G-proteins (G
i
)
B. Decrease production of IP
3
C. Decrease release of intracellular calcium
D. Inhibit the activity of phospholipase C
[2.3] Choline esters like carbachol are most likely to cause which of the
following adverse effects?
A. Anhydrosis (dry skin)
B. Delirium
C. Salivation
D. Tachycardia (rapid heart rate)
Answers
[2.1] C. Excessive pilocarpine may initially result in dilation of blood vessels
with a drop in blood pressure and a compensatory reflex stimulation of
heart rate. Higher levels will directly inhibit the heart rate. In addition,
pilocarpine stimulation of muscarinic cholinoreceptors can result in
miosis, bronchial smooth muscle dilation, and increased GI motility.
[2.2] A. In addition to activating inhibitory G-proteins (G
i
), muscarinic
cholinergic agonists stimulate the activity of phospholipase C, increase
production of IP
3
, and increase release of intracellular calcium.
[2.3] C.Diarrhea, salivation, and lacrimation may be seen. The heart rate
is usually slowed. Choline esters do not cross the blood-brain barrier,
and therefore delirium is not an adverse effect.
26
CASE FILES: PHARMACOLOGY
REFERENCES
Felder C. Muscarinic acetylcholine receptors: signal transduction through multiple
effectors. FASEB J 1995;9(8):619–25.
Marquis RE, Whitson JT. Management of glaucoma: focus on pharmacological
therapy. Drugs Aging 2005;22(1):1–21.
Millard CB, Broomfield CA. Anticholinesterases: medical applications of neuro-
chemical principles. J Neurochem 1995;64(5):1909–18.
CLINICAL CASES
27
PHARMACOLOGY PEARLS
Cholinoreceptors are classified as either nicotinic or muscarinic.
Muscarinic cholinoreceptors are localized at organs such as the
heart, causing a negative chronotropic effect.
Stimulation of muscarinic receptors in the smooth muscle, exocrine
glands, and vascular endothelium cause bronchoconstriction, incr-
eased acid secretion, and vasodilation.
Methacholine and bethanechol are highly selective for muscarinic
cholinoreceptors.
Cholinomimetic agents, including anticholinesterase inhibitors, are
precluded for treatment of gastrointestinal or urinary tract disease
because of mechanical obstruction, where therapy can result in
increased pressure and possible perforation. They are also not
indicated for patients with asthma.
This page intentionally left blank
CASE 3
A 53-year-old woman comes to see you for a consultation. She is scheduled to
take a Caribbean cruise in 2 weeks but is concerned about sea sickness. She
has been on boats before and is very sensitive to motion sickness. A friend
mentioned to her that there is a patch that is effective for this problem. She is
in good health and takes no medications regularly. Her examination is normal.
You prescribe a scopolamine transdermal patch for her.
What is the mechanism of action of scopolamine?
What are the common side effects of this medication?
What are some relative contraindications to its use?
ANSWERS TO CASE 3: MUSCARINIC
CHOLINORECEPTOR ANTAGONISTS
Summary:A 53-year-old woman with motion sickness is prescribed transder-
mal scopolamine before she takes a sea cruise.
Mechanism of action of scopolamine: Competitive antagonist of
muscarinic cholinoreceptors in the vestibular system and the CNS
Common side effects: Mydriasis, dry mouth, tachycardia, urinary
retention, confusion, drowsiness
Relative contraindications: Glaucoma, urinary obstruction, heart
disease
CLINICAL CORRELATION
Scopolamine, like other antimuscarinic agents, including the prototype
atropine, is a selective competitive (surmountable) antagonist of ACh at mus-
carinic cholinoreceptors. Its actions can be overcome by increased concentra-
tions of ACh or other muscarinic cholinoreceptor agonists. Scopolamine
blocks muscarinic cholinoreceptors in the vestibular system and CNS to
prevent motion sickness.It has a relatively long duration of action and can be
given as a transdermal patch, making it well suited for the treatment of motion
sickness. Histamine H
1
-receptor antagonists, such as cyclizine, are also used
to treat motion sickness.
In addition to motion sickness, muscarinic cholinoreceptor antagonists
(e.g., benztropine) are used therapeutically to treat Parkinson disease. Short-
acting topical agents or ointments are used to facilitate ophthalmoscopic
examination (e.g., cyclopentolate, tropicamide). Ipratropium bromide, a qua-
ternary ammonium compound that does not cross the blood-brain barrier, is
used to treat asthma and has efficacy in chronic obstructive pulmonary disease
(COPD). They (e.g., trospium, tolterodine) are also used to treat certain blad-
der disorders. Because it penetrates the CNS, the tertiary amine atropine is
used to counter the muscarinic cholinoreceptor effects of cholinergic excess
resulting from organophosphate insecticide poisoning.
Theadverse effects of scopolamine and other muscarinic cholinoreceptor
antagonists are related to inhibition of muscarinic cholinoreceptors in organ
systems of the body. Drowsiness and sedation are caused by actions on the
CNS. Mydriasis is caused by blocking parasympathetic tone in the muscles of
the cilia and iris. This could increase intraocular pressure in a person with glau-
coma. Cholinoreceptor blockade at the sinoatrial node results in tachycardia.
This could cause arrhythmias,especially in someone with underlying heart dis-
ease. The urinary bladder is relaxed and the urinary sphincter constricted,
which may promote urinary retention.Blockade of muscarinic cholinoreceptors
30
CASE FILES: PHARMACOLOGY
in the salivary glands reduces salivation,causing dry mouth. Blockade of other
muscarinic cholinoreceptors in the CNS can lead to impairment of memory,
confusion, restlessness, drowsiness, or hallucinations.
Muscarinic cholinoreceptor antagonist drugs are used cautiously in
patients with angle-closure glaucoma (contraindicated), open-angle glau-
coma, urinary tract obstruction (e.g., prostatic hypertrophy), cardiac dis-
ease, and gastrointestinal infections, among other conditions. Elderly
patients are particularly sensitive to CNS effects.
APPROACH TO MUSCARINIC CHOLINORECEPTOR
ANTAGONISTS
Objectives
1. Describe the mechanism of action of muscarinic cholinoreceptor
antagonists.
2. Describe the physiologic effects of muscarinic cholinoreceptor antag-
onists.
3. List important therapeutic uses of muscarinic cholinoreceptor antagonists.
4. List the adverse effects and contraindications for muscarinic choli-
noreceptor antagonists.
Definitions
Chronic obstructive pulmonary disease [COPD]: Progressive, inflam-
matory lung conditions, including both chronic bronchitis and emphy-
sema, which result in airway obstruction that is not fully reversible.
Most COPD is due to smoking.
Asthma:An inflammatory lung condition characterized by reversible airway
obstruction that can be precipitated by irritants such as environmental
allergens, cigarette smoke, cold air or exercise.
Muscarinic Cholinoreceptor antagonists: Drugs that block the actions of
acetylcholine.
DISCUSSION
Class
Cholinoreceptor antagonists are distinguished by their specificity for mus-
carinic and nicotinic cholinoreceptors. Muscarinic cholinoreceptor antago-
nists block the effects of ACh at muscarinic cholinoreceptors in the
parasympathetic autonomic nervous system and in the CNS. Nicotinic
cholinoreceptor antagonists block the effects of ACh at ganglia of the
parasympathetic and sympathetic nervous system (and medulla), and at the
neuromuscular junction.
CLINICAL CASES 31
32 CASE FILES: PHARMACOLOGY
Structure
Like atropine, the prototype muscarinic cholinoreceptor antagonist scopo-
lamine is a tertiary amine. As such, it has ready access to the CNS when
administered parenterally, and it can be absorbed across the skin when com-
bined with a suitable vehicle in a transdermal patch. Quaternary amine
antimuscarinic agents,including tiotropium bromide, have limited access to
the CNS and thus are used therapeutically for their peripheral effects.
Mechanism of Action
Interaction of scopolamine, atropine, or other antimuscarinic agents with mus-
carinic cholinoreceptors prevents the typical actions of ACh, such as activation
of G-proteins and subsequent production of IP
3
, and DAG that results in mobi-
lization of calcium.
Administration
The patch formulation of scopolamine for motion sickness provides for up to
72 hours of pharmacologic activity. Scopolamine can also be administered IV,
IM, or PO. Ipratropium bromide and tiotropium are administered topically to
the airways as a metered-dose inhaler for COPD.
Pharmacokinetics
The duration of action of antimuscarinic agents ranges from less than a day
(tropicamide) to 3–10 days (scopolamine, atropine).
COMPREHENSION QUESTIONS
[3.1] Prescription of a muscarinic cholinoreceptor antagonist with a quater-
nary amine group is most appropriate for the patient with which of the
following conditions?
A. A 50-year-old woman with angle-closure glaucoma
B. A 34-year-old man with gastrointestinal infectious enteritis
C. A 66-year-old man with mild dementia
D. A 56-year-old diabetic woman with urinary tract obstruction
[3.2] A 16-year-old teenager is going on his first deep sea fishing trip and is
using a scopolamine patch to ward off sea sickness. Which of the fol-
lowing is the most likely adverse effect he will experience?
A. Bradycardia
B. Drowsiness
C. Miosis
D. Urinary urgency
[3.3] Cholinergic excess resulting from organophosphate insecticide poison-
ing can be treated with which of the following?
A. Atropine
B. Digoxin
C. Ipratropium bromide
D. Tropicamide
Answers
[3.1] C. Muscarinic cholinoreceptor antagonists with quaternary amine
groups do not penetrate the CNS and are therefore unlikely to impair
memory. By blocking gastrointestinal motility, these agents can cause
increased retention of infecting organisms.
[3.2] B.Scopolamine penetrates the CNS and can cause drowsiness and seda-
tion. It also can cause mydriasis, tachycardia, and urinary retention.
[3.3] A.Atropine is a tertiary amine that can penetrate the CNS. In addi-
tion to its peripheral blocking actions, it can also block the adverse
CNS effects as a result of cholinergic excess. Tropicamide is also a
tertiary amine. However, it has a very short duration of action and
would be an unsuitable antidote. Ipratropium bromide is a charged
quaternary ammonium compound that does not penetrate the CNS.
CLINICAL CASES 33
PHARMACOLOGY PEARLS
Many antihistaminic agents, antipsychotic agents, and antidepres-
sant agents have muscarinic cholinoreceptor antagonist (antimus-
carinic) activity.
Scopolamine is a tertiary amine and has ready access to the CNS
when administered parenterally, whereas quaternary amine
antimuscarinic agents, such as ipratropium bromide, have limited
access to the CNS.
Scopolamine can cause drowsiness and sedation, as well as mydria-
sis, tachycardia, and urinary retention.
Cholinoreceptor agonists cause symptoms of SLUD—salivation,
lacrimation,urination, diarrhea—whereas cholinoreceptor antag-
onists have the opposite effects—dry mouth, dry eyes, urinary
retention, constipation.
34 CASE FILES: PHARMACOLOGY
REFERENCES
Alhasso AA, McKinlay J, Patrick K et al. Anticholinergic drugs versus non-drug
active therapies for overactive bladder syndrome in adults. Cochrane Database Syst
Rev 2006;18(4):CD003193.
Eglen RM, Choppin A, Watson N. Therapeutic opportunities from muscarinic
receptor research. Trends Pharmacol Sci 2001;22(8):409–14.
Nachum Z, Shupak A, Gordon C. Transdermal scopolamine for prevention of
motion sickness: clinical pharmacokinetics and therapeutic applications. Clin
Pharmacokinet 2006;45(6):543–66.
CASE 4
A healthy 25-year-old man is undergoing a brief surgical procedure requiring
general anesthesia. He underwent an unremarkable intubation and induction of
anesthesia using IV succinylcholine and inhaled halothane. During the surgery
the patient develops muscle rigidity and tachycardia, and his temperature rap-
idly rises.
What is the mechanism of action of succinylcholine?
What reaction is occurring in the patient?
What drug should immediately be given to the patient, and what is
its mechanism of action?
ANSWERS TO CASE 4: SKELETAL MUSCLE
RELAXANTS
Summary:A 25-year-old man develops muscle rigidity, tachycardia, and high
fever during surgery.
Mechanism of action of succinylcholine: Nicotinic receptor agonist at
the motor endplate of the neuromuscular junction, which causes
persistent stimulation and depolarization of muscle cells.
Reaction that is occurring: Malignant hyperthermia.
Drug given for treatment and its mechanism of action:Dantrolene,
which acts by interfering with calcium release from the sarcoplasmic
reticulum.
CLINICAL CORRELATION
Succinylcholine is the only depolarizing neuromuscular agent in wide clini-
cal use. It is used for the rapid induction of a brief flaccid paralysis. It works
as an agonist of the nicotinic receptor at the motor endplate of the neuro-
muscular junction. This causes a persistent stimulation and depolarization of
the muscle, preventing stimulation of contraction by ACh. It has a rapid
onset and short duration of action because it is quickly hydrolyzed by plasma
and liver cholinesterase. Malignant hyperthermia, a rare but significant
cause of anesthetic morbidity and mortality, is an inherited autosomal dom-
inant disorder that results in tachycardia, muscle rigidity, and high body
temperatures in response to the use of certain inhaled anesthetics in com-
bination with muscle relaxants, usually succinylcholine. It is caused by a
release of calcium ions from the sarcoplasmic reticulum in muscle cells.
Dantrolene interferes with this release and is therefore the treatment of
choice for this condition.
APPROACH TO PHARMACOLOGY OF SKELETAL
MUSCLE RELAXANTS
Objectives
1. Contrast the mechanism of action of depolarizing and nondepolarizing
neuromuscular junction-blocking agents.
2. List the therapeutic uses and adverse effects of skeletal muscle
relaxants.
36
CASE FILES: PHARMACOLOGY
Definitions
Hyperkalemia: Elevated levels of the electrolyte potassium in the serum
Myalgia: Pain originating in skeletal muscle
Depolarizing neuromuscular agent:A drug that acts at the neuromuscular
junction to prevent the initiation of an action potential by ACh.
DISCUSSION
Class
Neuromuscular blocking agentsare classified as either depolarizing or non-
depolarizing (Table 4-1) and are used mostly as adjuncts with general anes-
thetics to block motor endplate activity of ACh at the neuromuscular junction.
Succinylcholine is the prototype for depolarizing agents and used for brief
paralysis for surgery and for intubation. Tubocurarine, the prototype, and
other nondepolarizing agents (e.g., cisatracurium, vecuronium, rocuronium)
are used for longer term paralysis for surgery.
In addition to malignant hyperthermia, succinylcholine administration may
result in hyperkalemia,particularly in patients with burn and trauma, which
could result in cardiac arrest. Myalgia is also commonly reported.
Certain nondepolarizing agents may produce hypotension, as a result of
histamine release and some ganglionic blocking activity, and tachycardia as a
result of vagolytic activity.
Numerous drug interactions between neuromuscular blocking agents and
other drugs have been reported that lead to increased neuromuscular blockade,
particularly with certain antibiotics and inhaled anesthetics.
CLINICAL CASES 37
Table 4-1
SELECTED SKELETAL MUSCLE RELAXANTS
MECHANISM SELECTED
TYPE OF AGENT OF ACTION ADVERSE EFFECTS
Depolarizing agents Persistent endplate Malignant hyperthermia,
(succinylcholine) depolarization and hyperkalemia, myalgia
desensitization
Nondepolarizing agents Reversible competitive Hypotension, tachycardia
(tubocurarine, cisatracurium antagonists that block the
vecuronium, rocuronium) action of ACh at nicotinic
cholinoreceptor
Structure
The neuromuscular blocking agents resemble ACh (succinylcholine con-
tains two linked ACh molecules) and contain one or two quaternary nitro-
gens that limit entry into the CNS.
Mechanism of Action
After a single dose, succinylcholine occupies the nicotinic receptor to pro-
duce a persistent endplate depolarization(phase I block) that results in flac-
cid paralysis because the muscles become unresponsive to endogenously
released ACh. The initial depolarization is accompanied by muscle fascicula-
tions. Continued exposure of endplates to succinylcholine results in their
repolarization. However, through an unclear mechanism, they become rela-
tively insensitive to subsequent depolarization (so-called desensitization, or
phase II block).
Nondepolarizing blocking agents act as reversible competitive antagonists
that block the action of ACh at nicotinic cholinoreceptors in muscle endplates
and autonomic ganglia.
Cholinesterase inhibitors (e.g., neostigmine, pyridostigmine) can effec-
tively antagonize and reverse the neuromuscular blocking action of non-
depolarizing agents and succinylcholine during phase II. However, they
will augment the action of succinylcholine during phase I.
Administration
The neuromuscular blocking agents are highly polar and therefore must be
administered parenterally. Most nondepolarizing agents are eliminated
through the kidney. Succinylcholine is eliminated by the hydrolytic action of
plasma butyrylcholinesterase (pseudocholinesterase).
Pharmacokinetics
Neuromuscular blocking agents are highly ionized and therefore have limited
volume of distribution and limited access to the CNS.
COMPREHENSION QUESTIONS
[4.1] The use of succinylcholine as an adjunct to general anesthetics during
surgery is based on its ability to:
A. Block the action of ACh at the motor endplate
B. Increase release of ACh from autonomic ganglia
C. Increase release of histamine from mast cells
D. Inhibit cholinesterase
38
CASE FILES: PHARMACOLOGY
[4.2] Continued exposure of muscle endplates to succinylcholine results
in their:
A. Conversion to ion channels
B. Enhanced sensitivity to ACh
C. Regeneration of ACh receptors
D. Repolarization
[4.3] Cholinesterase inhibitors can reverse the action of which of the following?
A. Cisatracurium
B. Succinylcholine
C. Both A and B
D. Neither A or B
[4.4] A 35-year-old man undergoes surgery for a hernia repair. After the surgery,
he complains of diffuse muscle aches, which the anesthesiologist states is
likely caused by the skeletal muscle relaxant. He has a temperature of
37.8°C (100°F). Which of the following is the most accurate statement?
A. The agent also commonly causes hypokalemia.
B. The agent blocks ACh at the nicotinic receptor.
C. The agent causes persistent endplate depolarization and desen-
sitization.
D. The patient likely has malignant hyperthermia.
Answers
[4.1] A.Succinylcholine acts like ACh to cause depolarization of the mus-
cle endplate. However, unlike ACh, succinylcholine is not metabo-
lized at the synapse. Therefore, the endplate remains depolarized and
unresponsive to endogenous ACh, resulting in muscle paralysis.
[4.2] D. Continued exposure of the muscle endplate to succinylcholine
results in desensitization (phase II block) where the endplate repolar-
izes but cannot readily be depolarized.
[4.3] C.Cholinesterase inhibitors like neostigmine can effectively antago-
nize and reverse the neuromuscular blocking action of nondepolariz-
ing agents and succinylcholine during phase II. However, they will
augment the action of succinylcholine during phase I.
[4.4] C.Myalgia (muscle aches) is a common adverse reaction of depolar-
izing agents such as succinylcholine; these agents also may induce
hyperkalemia and malignant hyperthermia.
CLINICAL CASES 39
REFERENCES
Bowman WC. Neuromuscular block. Br J Pharmacol 2006;147(Suppl 1):S277–86.
Lee C. Structure, conformation, and action of neuromuscular blocking drugs. Br J
Anaesth 2001;87(5):755–69.
Sparr HJ, Beaufort TM, Fuchs-Buder T. Newer neuromuscular blocking agents.
How do they compare with established drugs? Drugs 2001;61(7):919–42.
40 CASE FILES: PHARMACOLOGY
PHARMACOLOGY PEARLS
Malignant hyperthermia is a rare autosomal dominant disorder char-
acterized by tachycardia, muscle rigidity, and high body temper-
atures, which occurs when the patient is exposed to inhaled
anesthetics in combination with muscle relaxants, usually suc-
cinylcholine.
Dantrolene interferes with the release of intracellular calcium and is
therefore used to treat the muscle rigidity and hyperthermia asso-
ciated with malignant hyperthermia.
The neuromuscular blocking agents are highly polar and highly ion-
ized and therefore, must be administered parenterally and have
limited volume of distribution and limited access to the CNS.
A small number of patients (1:10,000) with atypical cholinesterase
experience long-lasting apnea of 1–4 hours following succinyl-
choline (or the nondepolarizing neuromuscular blocking drug
mivacurium that is also eliminated by the action of butyryl-
cholinesterase). Mechanical ventilation is used to manage the
apnea even though prescreening could detect this rare condition.
CASE 5
A 65-year-old woman is admitted to the intensive care unit (ICU) of the hos-
pital with sepsis caused by a urinary tract infection. She is hypotensive, with
a blood pressure of 80/40 mm Hg and has an elevated heart rate (tachycardia)
and decreased urine output (oliguria). Along with the institution of appropri-
ate antibiotic therapy and IV fluids, a decision is made to start her on an IV
infusion of dopamine to attempt to raise her blood pressure.
What effects can be expected with low-dose dopamine?
Which receptors mediate these effects?
What effects occur with higher dose dopamine and which receptors
mediate these?
ANSWERS TO CASE 5: SYMPATHOMIMETIC AGENTS
Summary: A 65-year-old woman in septic shock has persistent hypotension
and oliguria requiring IV dopamine.
Effects of low-dose dopamine: Reduces arterial resistance and
increases blood flow in renal, coronary, and splanchnic systems;
positive inotropic effect.
Receptors involved:β
1
-receptors and specific dopamine receptors.
Effects of higher dose dopamine and receptors involved:
Vasoconstriction mediated by α-receptors.
CLINICAL CORRELATION
Dopamine is frequently used to treat cardiogenic or septic shock. The β
1
-
adrenoceptor-mediated effects in the heart result in an increase in cardiac
output with minimal peripheral vasoconstriction. This contributes to
dopamine’s ability to raise systolic blood pressure with no effect or only a
slight effect on diastolic pressure. Specific dopamine receptors in the vascu-
lature of the renal, coronary, and splanchnic systems allow for reduced arte-
rial resistance and increased blood flow. At higher doses there is a peripheral
α-adrenoceptor effect that overrides dopamine receptor-mediated vasodila-
tion and results in vasoconstriction. The combination of renal blood-flow
preservation, while supporting the blood pressure, is desirable in conditions
of shock. This also contributes to increasing blood pressure. Prolonged high
doses of dopamine can result in peripheral tissue necrosis because of the
α-adrenoceptor-mediated vasoconstriction that reduces blood flow to the
extremities, particularly in the digits.
APPROACH TO PHARMACOLOGY OF AUTONOMIC
SYMPATHETIC AGENTS
Objectives
1. Outline the effects of sympathomimetic agents on peripheral organ
systems.
2. List the major sympathomimetic agonists and their routes of adminis-
tration.
3. Describe the therapeutic and adverse effects of the major sympath-
omimetic drugs.
42
CASE FILES: PHARMACOLOGY
Definitions
Sympathomimetic agents: Drugs that either directly or indirectly mimic
all or some of the effects of epinephrine or norepinephrine.
Receptor selectivity: Preferential binding (greater affinity) of a drug to a
specific receptor group or receptor subtype at concentrations below which
there is little, if any, interaction with another receptor group or subtype.
DISCUSSION
Class
Sympathomimetic agents act directly (e.g., epinephrine, norepinephrine,
dopamine, dobutamine, phenylephrine, metaraminol, methoxamine,
albuterol, terbutaline) or indirectly (amphetamine, ephedrine) to activate
a- and b-adrenoceptors (Table 5-1). Sympathomimetic agent adrenoceptor
CLINICAL CASES 43
Table 5-1
AUTONOMIC NERVOUS SYSTEM EFFECTS*
ADRENERGIC MUSCARINIC CHOLINERGIC
ORGAN RECEPTOR/ACTION RECEPTOR/ACTION
Heart β
1
—increased heart rate and Decreased heart rate and
contractility contractility
Blood vessels
α
1
—constriction Dilation
β
2
—dilation
Bronchi β
2
—bronchial smooth muscle Bronchial smooth muscle
relaxation contraction
GI tract α
1
—sphincter contraction Overall contraction relaxation
β
2
—relaxation of sphincter
Kidney β
1
—renin release No effect
Urinary bladder α
1
—sphincter contraction Wall contraction sphincter
β
2
—wall relaxation relaxation
Adipose tissue β
1
—increased lipolysis No effect
Eye α
1
—radial muscle contraction Sphincter muscle contraction
with pupil dilation with pupil constriction ciliary
muscle contraction
*
See also Figure 1-1.
No direct parasympathetic innervation.
selectivity varies. Some are nonselective (e.g., ephedrine), whereas some
have greater affinity for α-adrenoceptors (e.g., phenylephrine, metaraminol,
methoxamine) or β
1
-adrenoceptor (e.g., dobutamine) or β
2
-adrenoceptor
(e.g., terbutaline, albuterol) subgroups. However, selectivity is often lost as
the dose of a sympathomimetic agent is increased. Compared to nonselective
β-receptor agonists (isoproterenol), b
1
-selective sympathomimetic agents
may increase cardiac output with minimal reflex tachycardia. a
2
-Selective
agents, which decrease blood pressure by a prejunctional action in the CNS
(clonidine, methyldopa), are used to treat hypertension. Fenoldopam is a
potent D
1
agonist with a short half-life, useful in severe hypertension; its
effects include decreasing systemic vascular resistance.
Dopamine interacts with specific subtypes of dopamine receptors in the
periphery (D
1
and D
2
). Stimulation of the D
1
receptor on the vasculature is
principally vasodilation, and on the renal proximal tubules leads to natriuresis
and diuresis; stimulation of the D
2
receptor on the presynaptic sympathetic
nerve endings inhibits norepinephrine release. It also has direct and indirect
sympathomimetic activity where, at lower doses, it has greater affinity for β-
adrenoceptors than it does for α-adrenoceptors.
The clinical utility of a particular sympathomimetic agent depends on,
among other factors, the specific organ system and receptor subtypes that are
involved. In the cardiovascular system, a reduction in blood flow by rela-
tively selective α-adrenoceptor sympathomimetic agents is used to achieve
surgical hemostasis, reduced diffusion of local anesthetics, and a reduction of
mucous membrane congestion in hay fever and for the common cold. An
increase in blood flow or blood pressure by α-adrenoceptor sympathomimetic
agents is beneficial for the management of hypotensive emergencies (e.g.,
phenylephrine, methoxamine, norepinephrine) and chronic orthostatic
hypotension (oral ephedrine). Sympathomimetic agents such as isoproterenol
(and epinephrine) are also used for emergency short-term treatment of com-
plete heart block and cardiac arrest.
Treatment of bronchial asthmarepresents a major use of b
2
-selective sym-
pathomimetic agents (e.g., terbutaline, albuterol). Its effect is bronchodilation
and relaxation of the smooth muscles of the bronchioles.
Ophthalmic examination is facilitated with the use of the directly acting
a-adrenoceptor sympathomimetic agonist, phenylephrine. Phenylephrine
(and the indirectly acting sympathomimetic agent, cocaine) is also used to
localize the lesion in Horner syndrome. In addition to β-adrenoceptor-blocking
agents, α
2
-selective agents (e.g., apraclonidine) are used to lower intraocular
pressure in glaucoma.
The peripheral adverse effects of the sympathomimetic agents are gen-
erally an extension of their pharmacologic effects. These are most often
cardiovascular in nature, particularly when they are administered parenter-
ally, and may include increased blood pressure, arrhythmias, and cardiac
failure.
44 CASE FILES: PHARMACOLOGY
Structure
Sympathomimetic agents, as well as norepinephrine and epinephrine, are
derived from phenylethylamine.Substitutions on the amino group, the ben-
zene ring or the α- or β-carbon, markedly alter the selectivity, activity, and
metabolism of the sympathomimetic agents. For example, alkyl substitutions
on the amino group tend to markedly increase β-adrenoceptor selectivity.
Mechanism of Action
Directly acting sympathomimetic agents bind to and activate adrenoceptors to
mimic the actions of epinephrine or norepinephrine. Indirectly acting sympa-
thomimetic agents mimic the actions of norepinephrine by either displacing it
or inhibiting its reuptake from adrenergic nerve endings.
Administration
Sympathomimetic agents are available for administration by topical, nasal,
oral, ophthalmic, and parenteral routes depending on the drug and condi-
tion being treated.
Pharmacokinetics
Like the catecholamines norepinephrine and epinephrine, direct and indirect
sympathomimetic agent may be subject to metabolism and inactivation by
COMT and MAO. Phenylephrine is not metabolized by COMT, whereas
metaraminol and methoxamine are not substrates for either COMT or MAO.
Their durations of action, therefore, are relatively long (20–60 minutes).
COMPREHENSION QUESTIONS
[5.1] A 25-year-old man is noted to be in septic shock. A low-dose dopamine
infusion is administered, and will likely result in which of the following?
A. Decrease cardiac output
B. Decrease systolic blood pressure
C. Increase renal blood flow
D. Produce significant peripheral vasoconstriction
[5.2] In contrast to norepinephrine, metaraminol, and methoxamine are
metabolized by which of the following?
A. COMT
B. MAO
C. Both
D. Neither
CLINICAL CASES 45
[5.3] Which of the following is the most accurate statement?
A. α-Adrenoceptor sympathomimetic agonists are used to reduce
mucous membrane congestion.
B. α-Adrenoceptor agonists are used to treat bronchospasm.
C. β-Adrenoceptor agonists are used to reduce surgical bleeding.
D. β
2
-Adrenoceptor agonist agents are used to prolong local anesthesia.
Answers
[5.1] C.Dopamine binding to specific dopamine receptors in the vascula-
ture in the kidney increases renal blood flow. Cardiac output is
increased by dopamine action on β
1
-adrenoceptors. Dopamine causes
minimal peripheral vasoconstriction.
[5.2] D. Metaraminol and methoxamine are not substrates for either
COMT or MAO and have a longer duration of action than norepi-
nephrine, which is a substrate for both.
[5.3] A.α-Adrenoceptor sympathomimetic agents will cause vasoconstric-
tion and thereby reduce mucous membrane congestion.
46 CASE FILES: PHARMACOLOGY
PHARMACOLOGY PEARLS
β
1
-Selective sympathomimetic agents may increase cardiac output
with minimal reflex tachycardia.
α
2
-Selective agents decrease blood pressure by a prejunctional
action in the CNS.
Terbutaline and albuterol are preferred over ephedrine for relieving
the bronchoconstriction of asthma, and other bronchial condi-
tions, because of their greater bronchiolar selectivity.
Dopamine binding to specific dopamine receptors in the vasculature in
the kidney increases renal blood flow. Cardiac output is increased
by dopamine action on β
1
-adrenoceptors. Low-dose dopamine
causes minimal peripheral vasoconstriction.
REFERENCES
Cleland JG. Beta-blockers for heart failure: why, which, when, and where. Med Clin
North Am 2003;87(2):339–71.
Graham RM, Perez DM, Hwa J, et al. Alpha
1
-adrenergic receptor subtypes.
Molecular structure, function and signaling. Circ Res 1996;78(5):737–49.
Mann HJ, Nolan PE. Update on the management of cardiogenic shock. Curr Opin
Crit Care 2006;12(5):431–6.
CASE 6
A 70-year-old man is seen in follow-up at your office after he has been hospi-
talized for a myocardial infarction (MI). He underwent successful angioplasty
and is currently asymptomatic. Prior to his MI, he was not on medications. He
is not a smoker and is not diabetic. During his hospitalization, he was noted
to have persistently elevated blood pressure readings. He had asthma as a
child, but has not had any recent wheezing episodes. While in the hospital, he
was started on oral metoprolol.
Metoprolol is selective for which adrenoceptor?
What effects do agents such as metoprolol have on the
cardiovascular system?
In which organ is metoprolol primarily metabolized?
Why must b-adrenergic antagonists be used with caution in
asthmatics?
ANSWERS TO CASE 6: ADRENOCEPTOR
ANTAGONISTS
Summary:A 70-year-old hypertensive man with a childhood history of asthma
had a recent myocardial infarction and is prescribed metoprolol.
Adrenoceptor selectively antagonized by metoprolol:β
1
.
Effect of b-adrenoceptor antagonists on the cardiovascular system:
Reduction of sympathetic-stimulated increases in heart rate,
contractility, and cardiac output; lower blood pressure as a result of
effects on the heart, renin-angiotensin system, and CNS; increased
atrioventricular (AV) conduction time and refractoriness.
Organ in which metoprolol is metabolized:Liver.
Reason for caution in use in asthmatics: Blockade of β
2
-adrenoceptor
in bronchial smooth muscle may cause increased airway resistance and
bronchospasm.
CLINICAL CORRELATION
β-Adrenergic receptor antagonists are widely used in medicine, primarily for
their beneficial effects on the cardiovascular system and for lowering intraoc-
ular pressure in patients with glaucoma. Both the nonselective β-adrenorecep-
tor antagonists and the relatively β
1
-adrenoceptor selective antagonists are
used to treat hypertension. The mechanism of their action is multifactorial,
probably including reduction in cardiac output, reduction in renin release, and
some CNS effect. They are also beneficial for treating coronary artery disease
and postmyocardial infarction patients as they reduce sympathetic-stimulated
increases in heart rate and contractility. This helps to reduce myocardial oxy-
gen demand, providing prophylaxis for angina. b-Adrenoceptor antagonists
have a proven benefit in prolonging survival after heart attacks. They
lengthen AV conduction time and refractoriness and suppress automaticity.
This helps to prevent both supraventricular and ventricular arrhythmias.
Caution must be used when giving β-blockers to patients with asthma, COPD,
and diabetes. All β-adrenoceptor blockers, including those that are β
1
-adreno-
ceptor selective, have some β
2
-adrenoceptor antagonist activity and may
cause bronchospasm by their effects on bronchial smooth muscle. They can
also mask the symptoms of hypoglycemia in a diabetic by blocking the
adrenergic stimulated symptoms of tremor, tachycardia, and nervousness that
would normally occur.
48
CASE FILES: PHARMACOLOGY
APPROACH TO PHARMACOLOGY
OF ADRENOCEPTOR AGONISTS
Objectives
1. Describe the therapeutic uses and adverse effects of α-adrenoceptor
antagonists.
2. Describe the therapeutic uses and adverse effects of β-adrenoceptor
antagonists.
3. Contrast the differences between the nonselective and relatively
β
1
-selective adrenoceptor antagonists.
Definitions
Pheochromocytoma:A tumor of the adrenal medulla that releases excess
levels of epinephrine and norepinephrine that can result in hypertension,
cardiac anomalies, and severe headache.
Myocardial infarction:Death of cardiac muscle as a result of ischemia.
DISCUSSION
Class
There are two classes of clinically important α-adrenoceptor antagonists: non-
selective antagonists and selective α
1
-antagonists. Phentolamine, a nonselec-
tive, competitive a-adrenoceptor antagonist, and phenoxybenzamine, a
nonselective, noncompetitive a-adrenoceptor antagonist are used for the
preoperative management of the marked catecholamine-induced vasoconstric-
tionassociated with pheochromocytoma. Prazosin and other a
1
-adrenoceptor
selective antagonists(doxazosin, terazosin) are used to manage chronic mild-
to-moderate hypertension and benign prostatic hypertrophy.
In addition to the nonselective β-adrenoceptor antagonists, there are two
classes of clinically important selective β-adrenoceptor antagonists, β
1
and β
2
(Table 6-1). The major clinical uses for β-adrenoceptor antagonists include
ischemic heart disease, cardiac arrhythmias, hypertension, hyperthyroidism, and
glaucoma. Ischemic heart disease is managed with nonselective β-adrenoceptor
antagonists, propranolol, timolol, and nadolol, as well as β
1
-adrenoceptor selec-
tive antagonists, metoprolol, atenolol, and esmolol. Cardiac arrhythmias are
managed, depending on the arrhythmia, with propranolol and esmolol.
Hypertension is managed with a wide variety of nonselective and b
1
-adreno-
ceptor selective antagonists, except esmolol. Timololand other β-adrenocep-
tor antagonists are used to manage glaucoma by decreasing aqueous humor
production and thereby reducing intraocular pressure.
CLINICAL CASES 49
Labetalol (and several other agents, including carvedilol), in formulations
used clinically, blocks both b- and a
1
-adrenoceptors in a 3:1 ratio. It also
has some β
2
-adrenoceptor agonist activity. Labetalol lowers blood pressure
bydecreasing systemic vascular resistance without any major effect on heart
rate or cardiac output. It is used to treat hypertensive emergencies and hyper-
tension from pheochromocytoma. Table 6-2 has a listing of selected sympa-
thomimetic agents.
The major adverse effects of nonselective α-adrenoceptor antagonists
arecardiac stimulation, primarily tachycardia because of baroreflex-mediated
sympathetic discharge, and postural hypotension.Additional cardiac stimu-
lation by phentolamine may be caused by antagonist activity at prejunctional
α
2
-adrenoceptors that result in increased norepinephrine release. (Prazosin and
other selective α
1
-adrenoceptor selective antagonists are less likely to cause
reflex tachycardia.) A β-adrenoceptor antagonist may be required to counter
the cardiac effects. α-Antagonists are rarely used as first-line agents for hyper-
tension, as they are associated with a higher rate of congestive heart failure
than other agents.
The major adverse effects of nonselective b-adrenoceptor antagonists
are related to their effects on bronchial smooth muscle and on carbohydrate
metabolism. Blockade of b
2
-receptors in smooth muscle may increase air-
way resistance in patients with asthma or other airway obstruction dis-
eases.Although the clinical use of a selective β
1
-adrenoceptor antagonist may
offer some protection in patients with asthma, the selectivity of these agents is
not great, and therefore they should be used judiciously, if at all. In patients
with insulin-dependent diabetes, nonselective b-adrenoceptor antagonists
increase the incidence and severity of hypoglycemic episodes. The use of
selective β
1
-adrenoceptor antagonists in patients with this condition offers
some potential benefit.
50 CASE FILES: PHARMACOLOGY
Table 6-1
β-ADRENOCEPTOR ANTAGONIST SELECTIVITY
Nonselectiveb-Adrenoceptor Antagonists
Propranol
Nadolol
Timolol
Selectiveb
1
-Adrenoceptor Antagonists
Atenolol
Metoprolol
Esmolol
Nonselectiveb- and a
1
-Adrenoceptor Antagonists
Labetalol
Carvedilol
Mechanism of Action
α-Adrenoceptor antagonists and β-adrenoceptor antagonists interact directly,
and either competitively or irreversibly with, respectively, α-adrenoceptors
andβ-adrenoceptors to block actions of the endogenous catecholamines (nor-
epinephrine and epinephrine), and exogenously administered sympath-
omimetic agents.
Administration
α- and β-adrenoceptor antagonists are administered orally or parenterally.
β-Adrenoceptor antagonists are also available for ophthalmic application.
CLINICAL CASES 51
Table 6-2
SELECTED DRUGS AND THEIR EFFECTS ON THE AUTONOMIC
NERVOUS SYSTEM
ADRENOCEPTOR MECHANISM OF
DRUG ACTIVITY ACTION CLINICAL USE
Epinephrine Nonselective α- Bronchial smooth Asthma and other
andβ-adrenoceptor muscle dilation allergic diseases to
agonist relax airways and
reduce swelling
Phenylephrine α
1
-Adrenoreceptor Vasoconstriction Rhinitis and colds as
stimulation decongestant
Propranolol Nonselective β- Decreases heart Hypertension, coronary
adrenoceptor rate, cardiac heart disease
agonist contractility
Albuterol β
2
-Adrenoceptor Bronchial smooth Asthma
agonist muscle dilation
Phentolamine Nonselective Vasodilation Preoperative manage-
competitive α- ment of the marked
adrenoceptor catecholamine-induced
antagonist vasoconstriction
associated with
pheochromocytoma
Prazosin α
1
-Adrenoreceptor Vasodilation Chronic mild to
selective moderate hypertension
antagonists and benign prostatic
hypertrophy
Pharmacokinetics
Metoprolol and propranolol undergo extensive and variable interindividual
first-pass hepatic metabolism resulting in relatively low bioavailability. Oral
sustained-release preparations of these agents are available. Drugs that inhibit
cytochrome P
450
2D6 may decrease the metabolism of carvedilol. Esmolol is
ultra-short-acting as a result of its ester linkage that is rapidly metabolized by
plasma esterases.
COMPREHENSION QUESTIONS
[6.1] Which of the following actions of epinephrine are blocked by prazosin?
A. Bronchial dilation
B. Increased cardiac stroke volume
C. Increased heart rate
D. Mydriasis
[6.2] A 34-year-old man is prescribed labetalol for hypertension. The effect
on the cardiovascular system is a result of its action as an antagonist at
which of the following?
A. α-Adrenoceptors
B. β-Adrenoceptors
C. Both α- and β-adrenoceptors
D. Muscarinic cholinoreceptors
[6.3] Which of the following is the least likely clinical use for β-adrenoceptor
antagonists?
A. Benign prostatic hypertrophy
B. Cardiac arrhythmias
C. Hypertension
D. Ischemic heart disease
Answers
[6.1] D.Prazosin is an α-adrenoceptor antagonist that will block epinephrine-
mediated contraction of the radial smooth muscle of the eye that
results in mydriasis. All the other actions listed are mediated by β-
adrenoceptors, which would be blocked by β-adrenoceptor antago-
nists like propranolol.
[6.2] C. Labetalol blocks both β- and α-adrenoceptors. It lowers blood
pressure by decreasing systemic vascular resistance (α-adrenoceptor
antagonist activity), without any major effect on heart rate or cardiac
output (β-adrenoceptor antagonist activity).
52
CASE FILES: PHARMACOLOGY
[6.3] A. β-Adrenoceptor antagonists are used therapeutically to manage
ischemic heart disease, cardiac arrhythmias, and hypertension. α
1
-
Adrenoceptor selective antagonists are used to manage benign pro-
static hypertrophy.
CLINICAL CASES 53
PHARMACOLOGY PEARLS
α
1
-Adrenoceptor selective antagonists, such as doxazosin and tera-
zosin, are used for mild chronic hypertension and benign prosta-
tic hypertrophy.
The major clinical uses for β-adrenoceptor antagonists include
ischemic heart disease, cardiac arrhythmias, hypertension, hyper-
thyroidism, and glaucoma.
The major adverse effects of nonselective b-adrenoceptor antag-
onists are related to their effects on bronchial smooth muscle
(increased airway resistance in asthmatics) and on carbohydrate
metabolism (hypoglycemia in insulin-dependent diabetics).
REFERENCES
Shin J, Johnson JA. Pharmacogenetics of beta-blockers. Pharmacotherapy
2007;27(6):874–87.
Piascik MT, Perez DM. Alpha 1-adrenergic receptors: new insights and directions.
J Pharmacol Exp Ther 2001;293(2):403–10.
This page intentionally left blank
CASE 7
A 64-year-old woman with a history of two previous myocardial infarctions
(MIs) comes to the emergency room with shortness of breath. In the previous
2 weeks, she has developed dyspnea with exertion and swelling of her legs.
She sleeps on three pillows because she coughs and gets short of breath if she
tries to lie flat. In the emergency department, she is sitting upright, appears to
be in moderate respiratory distress, and is tachycardic and hypertensive. She
has jugular venous distension to the angle of her jaw. On auscultation of her
lungs, wet rales are heard bilaterally. She has pitting edema of both lower legs
up to her knees. A chest x-ray confirms the diagnosis of pulmonary edema.
She is placed on oxygen and immediately given an IV injection of furosemide.
What is the mechanism of action of furosemide?
What electrolyte abnormalities can be caused by furosemide?
ANSWERS TO CASE 7: DIURETICS
Summary: A 64-year-old woman with pulmonary edema is prescribed
furosemide.
Mechanism of action of furosemide: Inhibit active NaCl reabsorption
in the ascending limb of the loop of Henle, increasing water and
electrolyte excretion.
Potential electrolyte abnormalities:Hypokalemia, hypomagnesemia,
and metabolic alkalosis because of enhanced H
+
excretion.
CLINICAL CORRELATION
Loop diuretics given intravenously promote diuresis within minutes, mak-
ing them ideal for the treatment of acute pulmonary edema. Furosemide
is the prototype and most widely used drug in this class. Loop diuretics inhibit
NaCl reabsorption in the ascending limb of the loop of Henle. This causes a
marked increasein the excretion of both water and electrolytes. The excretion
of potassium, magnesium, and calcium ions are all increased, which may
cause clinically significant adverse effects. A metabolic alkalosis may also
occur as a result of the excretion of hydrogen ions. However, the ability to
cause excretion of these electrolytes may also provide a clinical benefit in cer-
tain situations. Forced diuresis by giving IV saline and furosemide is a pri-
mary method of treatment of hypercalcemia.
APPROACH TO PHARMACOLOGY OF THE
LOOP DIURETICS
Objectives
1. Know the site and mechanism of action of diuretic agents.
2. Know the electrolyte effects of the various diuretic agents.
3. Know the therapeutic uses, adverse effects, and contraindications to
diuretic use.
Definitions
Diuretic:An agent that increases the production of urine. The most com-
mon are natriuretic diuretics, agents that increase urine production by
interfering with sodium reabsorption in the kidney.
Edema: Accumulation of water in interstitial spaces. Causes include ele-
vated blood pressure, a decrease in plasma oncotic pressure caused by a
reduction in hepatic protein synthesis, or an increase in the oncotic pres-
sure within the interstitial space.
56
CASE FILES: PHARMACOLOGY
DISCUSSION
Class
Natriuretic diuretics all act within the kidney to reduce the reabsorption
of Na
and Cl
.There are four sites within the kidney where various diuret-
ics act; these correspond to four anatomic regions of the nephron. The proxi-
mal tubule (site 1)is the site of approximately 60 percent Na
+
reabsorption, but
diuretics acting here are relatively ineffective because of the sodium-reabsorbing
capacity in more distal regions of the nephron. The ascending loop of Henle
(site 2)has active reabsorption of approximately 35 percent of the filtered Na
+
.
This is mediated by a cotransporter termed NKCC2 that transports 1 Na
+
, 1 K
+
,
and 2 Cl
, and this is the molecular target of furosemide and other loop or
“high-ceiling” diuretics. The distal convoluted tubule (site 3)is responsible
for transport of approximately 15 percent of filtered sodium. Thiazidediuret-
ics act in this segment of the nephron by interfering with another cotransporter,
NCC, which cotransports Na
+
and Cl
. Site 4 diuretics act in the collecting
tubule by interfering with Na
+
reabsorption through a specific channel, the
epithelial sodium channel (ENaC), also called the amiloride-sensitive sodium
channel (Figure 7-1).
Loop diuretics—furosemide, ethacrynic acid, bumetanide, and
torsemide—are highly acidic drugs that act on the luminal side of the
tubule. They reach this site by being secreted into the tubule by anion secre-
tion in the proximal tubule. Compared with other diuretics, loop diuretics
cause the greatest diuresis because the Na
+
K
2Cl
transporter is responsible
for a large fraction of Na
+
reabsorption, and regions distal to the ascending
limb have more limited capacity for sodium transport. Loop diuretics are use-
ful for the treatment of peripheral and pulmonary edema, which may occur
secondarily as a consequence of cardiac failure, liver failure, or renal failure.
Loop diuretics increase the excretion of Na
+
, Cl
, K
+
, Mg
2+
, Ca
2+
and decrease
the excretion of Li
+
. The increased excretion of Ca
2+
is clinically relevant, and
loop diuretics can be used to treat hypercalcemia. Some of the diuretic actions
of furosemide are mediated via prostaglandins because inhibitors of
prostaglandin biosynthesis diminish the increase in diuresis produced by the
drug. In addition, furosemide has actions on the vascular system that occur
prior to diuresis and this action may be mediated by prostaglandins. Other
effects include changes in renal blood flow and a reduction in left-ventricular-
filling pressure. Loop diuretics increase urine production and decrease plasma
K
+
in patients with acute renal failure.
Major adverse effects of loop diuretics are electrolyte imbalances.
Increased delivery of Na
+
to the collecting duct increases K
+
and H
+
excretion.
Loop diuretics therefore cause hypokalemia, hypochloridemia, and meta-
bolic alkalosis. Hyperuricemiamay be caused by the volume contraction and
enhanced uric acid reabsorption by the proximal tubule. Loop diuretics can
produce dose-dependent ototoxicity.
CLINICAL CASES 57
Figure 7-1. Sites of action of the nephron and diuretic agents.
Distal convoluted tubule
Proximal convoluted tubule
Proximal straight tubule
Descending limb
of loop of Henle
Ascending limb
of loop of Henle
Collecting
tubule
2Cl
Na
+
Na
+
Na
+
H
+
Na
+
Cl
K
+
Site 1
Carbonic anhydrase
inhibitors
Site 3
Thiazide
diurectics
Site 4
Aldosterone
antagonists
Site 2
Loop
diurectics
Structure
Most loop diuretics are sulfonamide derivatives;the exceptions are ethacrynic
acid, which is a phenoxyacetic acid derivative, and torsemide, which is a
sulfonylurea.
Mechanism of Action
The molecular target of furosemide and other loop or high-ceiling diuretics is the
sodium-potassium-2 chloride cotransporter (NKCC2), which transports 1 Na
+
,
1 K
+
, and 2 Cl
. The activity of this transporter is blocked by loop diuretics.
58
CASE FILES: PHARMACOLOGY
Administration
All loop diuretics can be administered orally, and their onset of action is
approximately 1 hour (torsemide) to 2 hours (furosemide). Loop diuretics can
also be administered IV, and for furosemide, this produces vasodilation in as
little as 5 minutes and diuresis in 20 minutes.
Pharmacokinetics
All loop diuretics are extensively bound to plasma proteins. Half-lives vary
from 45 minutes (bumetanide) to 3.5 hours (torsemide). Approximately
65 percent of a dose of furosemide is eliminated by the kidney, and the
remainder is metabolized. Only 20 percent of torsemide is eliminated by the
kidney, and 80 percent is metabolized.
COMPREHENSION QUESTIONS
[7.1] Furosemide acts to inhibit Na
+
reabsorption in which of the following
locations?
A. Ascending limb of the loop of Henle
B. Collecting duct
C. Descending limb of the loop of Henle
D. Distal convoluted tubule
[7.2] A patient arrives in the emergency room in a coma and has a serum Ca
2+
of 4.5 mM. You start a saline infusion of which of the following drugs?
A. Calcitonin
B. Ethacrynic acid
C. Hydrochlorothiazide
D. Spironolactone
[7.3] A 55-year-old man with congestive heart failure is noted to be taking
furosemide each day. Which of the following is most likely to be found
in the serum?
A. Decreased potassium level
B. Decreased uric acid level
C. Elevated magnesium level
D. Low bicarbonate level
Answers
[7.1] A.Furosemide acts specifically on a Na
+
K
+
2Cl
transporter in the
ascending limb of the loop of Henle.
[7.2] B.Loop diuretics such as ethacrynic acid increase the excretion of Ca
2+
.
[7.3] A. Furosemide leads to hypokalemia, hypomagnesemia, and meta-
bolic alkylosis (elevated bicarbonate level).
CLINICAL CASES 59
REFERENCES
Paul RV. Rational diuretic management in congestive heart failure: a case-based
review. Crit Care Nurs Clin North Am 2003;15(4):453–60.
Padilla MC, Armas-Hernandez MJ, Hernandez RJ, et al. Update of diuretics in the
treatment of hypertension. Am J Ther 2007;14(4): 331–5.
60 CASE FILES: PHARMACOLOGY
PHARMACOLOGY PEARLS
Furosemide, which acts on the loop of Henle, is the most efficacious
diuretic.
Hypokalemia is a frequent adverse effect encountered with loop
diuretics, and this can be managed with the concomitant use of
potassium-sparing diuretics such as triamterene or spironolactone.
Loop diuretics can produce dose-dependent ototoxicity.
CASE 8
Following his third episode of gouty arthritis, a 50-year-old man sees you in
the clinic. Each case was successfully treated acutely; however, your patient is
interested in trying to prevent future episodes. He is not on regular medications
and has a normal physical examination today. Blood work reveals an elevated
serum uric acid level and otherwise normal renal function and electrolytes. A
24-hour urine collection for uric acid reveals that he is under-excreting uric
acid. Suspecting that this is the cause of his recurrent gout, you place him on
probenecid.
What is the mechanism of action of probenecid?
Which drugs could have their excretion inhibited by probenecid?
ANSWERS TO CASE 8: NONDIURETIC INHIBITORS
OF TUBULAR TRANSPORT
Summary: A 50-year-old man with recurrent gout is prescribed probenecid.
Mechanism of action of probenecid: Inhibits secretion of organic
acids and decreases reabsorption of uric acid, causing a net increase in
secretion.
Other drugs whose secretion could be inhibited: Penicillin,
indomethacin, and methotrexate.
CLINICAL CORRELATION
Gout is a disease in which uric acid crystals deposit in joints, causing an
extremely painful acute inflammatory arthritis. Persons with recurrent gout
often have chronically elevated levels of uric acid in their blood. This hyper-
uricemia is frequently caused by either overproduction of uric acid or under-
excretion of uric acid by the kidneys. Probenecid (and other uricosuric drugs)
promotes the excretion of uric acid. It works by inhibiting the secretion of
organic acids from the plasma into the tubular lumen and blocking the reup-
take of uric acid. The net result of this is an increase in the secretion of uric
acid. The benefit of this is the prevention of recurrent gout attacks in chronic
underexcreters of uric acid. In those individuals who overproduce uric acid,
allopurinol is used. This inhibits xanthine oxidase, a key enzyme in the pro-
duction of uric acid.
APPROACH TO PHARMACOLOGY
OF URICOSURIC AGENTS
Objectives
1. Understand the mechanism of action of uricosuric agents.
2. Know the therapeutic uses, adverse effects, and contraindications to
uricosurics.
3. Know the mechanism of action and use of allopurinol.
Definitions
Uricosuric agents: Increase the mass of uric acid that is excreted in the
urine.
Renal secretion: Moves solutes such as urate from the plasma into the
urine.
Renal reabsorption:Moves solutes from the urine back into the plasma.
62 CASE FILES: PHARMACOLOGY
DISCUSSION
Class
Urate is both secreted and reabsorbed by at least three independent molecular
transporters located in the proximal tubule. Urate is nearly completely secreted
into the lumen of the nephron against an electrochemical gradient by the
action of organic acid transporter-1 (OAT-1) and organic acid transporter-3
(OAT-3).These cotransporters exchange α-ketoglutarate and urate (or other
organic anions) and move urate from the plasma into the tubular cell. The pro-
tein UAT is an electrically neutral channel that permits uric acid to leave tubu-
lar cells and enter either the tubular lumen or the plasma. URAT1, located on
the apical membrane of tubular cells, is thought to be responsible for most of
the reabsorption of urate from the filtrate. URAT1 is a transporter that is capa-
ble of exchanging a variety of anions with urate in an electrically neutral manner.
Interaction of uricosuric agents such as probenecid with URAT1 diminishes the
reabsorption of urate and increases urate excretion. All of these transporters or
channels are relatively nonselective with respect to the organic acid transported.
OAT-1 and OAT-3 are capable of secreting most organic acids including
probenecid, penicillin, aspirin, furosemide, and hydrochlorothiazide.
In patients with gout, probenecid can be used prophylactically; urico-
suric drugs will not diminish the severity of an acute attack. An acute
gouty attackmay be precipitated by the initiation of probenecid treatment
as uric acid is mobilized out of joints. Adequate hydrationshould be ensured,
because probenecid predisposes patients to the formation of uric acid kidney
stones.
The alternate therapeutic approach to the treatment of gout is to reduce the
production of uric acid with allopurinol. The enzyme xanthine oxidase
produces uric acid in a two-step reaction from the purine hypoxanthine.
Allopurinol is metabolized to alloxanthine by xanthine oxidase, and this
metabolite is a long-lasting inhibitor of the enzyme.
Probenecid is also useful for decreasing the excretion of penicillin,
because penicillin is eliminated primarily by renal secretion mediated by
OAT-1 and OAT-3. Probenecid competes for this secretion and thereby
reduces the rate of elimination and increases both the biological half-life of
penicillin and the plasma concentrationof the antibiotic more than twofold.
This adjunct use of probenecid is particularly useful in single-dose regimens
for the treatment of gonococcal infections with long-acting penicillins such as
penicillin G.
Secretion of organic acids is quite nonspecific, and most acidic drugs are
secreted by the same transporters OAT-1 and OAT-3. This implies that nearly
any combination of acidic drugs will compete for elimination at the level of
the transporters, and the effects on elimination of each individual drug must be
considered. For example, the half-life of diuretics such as furosemide will be
increased by probenecid, and this may require dosage adjustment. Aspirin,
CLINICAL CASES 63
another acidic drug, will compete with probenecid for secretion. This
reduces the action of probenecid to increase uric acid excretion and thus
increases plasma urate. Therefore aspirin is contraindicated in patients with
gout who are taking probenecid.
The most common adverse effect of probenecid is gastrointestinal (GI)
upset, and approximately 2 percent of patients experience a hypersensitivity
reaction usually manifest as a skin rash. The incidence of hypersensitivity is
lower with sulfinpyrazone, but the incidence of GI upset is higher.
Structure
Probenecid is a lipid-soluble benzoic acid derivative with a pKa of 3.4.
Another agent in this class is sulfinpyrazone, a pyrazolone derivative similar
to the anti-inflammatory agent phenylbutazone. It has a pKa of 2.8 but is no
longer marketed in the United States.
Mechanism of Action
Both probenecid and sulfinpyrazone are secreted into the lumen of the nephron
via OAT-1 and OAT-3 where the drugs can diminish the ability of URAT1 to
reabsorb urate.
Administration
Both drugs are active orally, and both are nearly completely absorbed.
Pharmacokinetics
The half-life of probenecid is 5–8 hours; sulfinpyrazone is approximately 3 hours,
but its uricosuric actions can last as long as 10 hours. Increased excretion of
uric acid occurs promptly after oral administration. Both agents are eliminated
in the urine.
COMPREHENSION QUESTIONS
[8.1] Probenecid is effective in treating gout because it decreases which of
the following?
A. Inflammation in affected joints
B. Production of uric acid
C. Reabsorption of uric acid
D. Secretion of uric acid
64 CASE FILES: PHARMACOLOGY
[8.2] Which of the following describes the action of allopurinol?
A. Inhibits metabolism of purines to uric acid
B. Inhibits prostaglandin biosynthesis
C. Inhibits uric acid reabsorption
D. Interferes with cytokine production
[8.3] An 18-year-old man who is known to have non-penicillinase-producing
gonococcal urethritis is given an injection of penicillin and probenecid.
What is the mechanism used by probenecid that makes penicillin more
efficacious?
A. Decreases the bacterial resistance by inhibiting penicillinase
production
B Increases the half-life and serum level by decreasing the renal
excretion of penicillin
C. Prolongs the duration of action by affecting the liver metabolism of
penicillin
D. Promotes entry of the penicillin into the bacteria
Answers
[8.1] C. Probenecid does inhibit renal tubular secretion of urate, but at
therapeutic doses it inhibits reabsorption to a greater degree, thereby
increasing net excretion urate.
[8.2] A.Allopurinol interferes with the metabolism of purines by inhibit-
ing the enzyme xanthine oxidase.
[8.3] B. Probenecid decreases the renal excretion of penicillin, thereby
increasing both the half-life and the serum level.
PHARMACOLOGY PEARLS
At low doses, probenecid inhibition of urate secretion predominates,
and this paradoxically increases plasma urate.
At higher doses, inhibition of reabsorption predominates, leading to
the therapeutically useful increased excretion of urate.
An acute gouty attack may be precipitated by the initiation of
probenecid treatment as uric acid is mobilized out of joints.
Probenecid is also useful for decreasing the excretion of penicillin
and cephalosporins.
Patients are typically begun on a high loading dose to ensure the
action on reabsorption is achieved.
CLINICAL CASES 65
REFERENCES
Dantzler WH. Regulation of renal proximal and distal tubule transport: sodium,
chloride and organic ions. Comp Biochem Physiol Part A 2003;136:453–78.
Stamp LK, O’Donnell JL, Chapman PT. Emerging therapies in the long-term man-
agement of hyperuricemia and gout. Intern Med J 2007;37:258–66.
66 CASE FILES: PHARMACOLOGY
CASE 9
A 72-year-old man presents to the office for routine follow-up. He is under
treatment for hypertension and congestive heart failure with enalapril and a
diuretic. His blood pressure is under acceptable control and he has no symp-
toms of heart failure at present. He does complain that he has been coughing
frequently in the past few months. History and examination reveal no other
cause of a chronic cough, so you decide to discontinue his enalapril and start
him on losartan.
What is the mechanism of action of enalapril?
By what mechanism does enalapril convert to its active form
enalaprilat?
What is the likely cause of the cough?
What is the mechanism of action of losartan?
ANSWERS TO CASE 9: DRUGS ACTIVE ON THE
RENIN-ANGIOTENSIN SYSTEM
Summary: A 72-year-old man with hypertension and congestive heart failure
presents with an ACE inhibitor-induced cough, and is switched to losartan.
Mechanism of action of enalapril: Inhibits the conversion of
angiotensin I to angiotensin II, this also inhibits the angiotensin
II-stimulated release of aldosterone. Angiotensin-converting enzyme
(ACE) inhibitors also impair the inactivation of bradykinin.
Mechanism of converting enalapril to enalaprilat:Deesterification in
the liver.
Mechanism of ACE inhibitor-induced cough:Secondary to the
increased bradykinin levels, which is caused by reduction in the
inactivation of bradykinin.
Mechanism of action of angiotensin receptor blockers (ARBs):
Antagonists of angiotensin-1 (AT-1) receptors which mediate the
pressor effects of angiotensin II.
CLINICAL CORRELATION
ACE inhibitors have gained wide-scale use in medicine for their effectiveness
in hypertension, congestive heart failure, coronary artery disease, and renal
protection in diabetics. They inhibit the conversion of angiotensin I to
angiotensin II. Angiotensin II is a potent vasoconstrictor and stimulates the
release of aldosterone, which promotes sodium retention. Angiotensin II also
increases catecholamine release by the adrenal medulla and at sympathetic
nerves. Inhibition of the production of angiotensin II reduces vascular resist-
ance and sodium and water retention. Another effect of ACE inhibitors is to
reduce the inactivation of bradykinin. Active bradykinin is a vasodilator, pro-
viding an additive effect in lowering blood pressure. However, raising
bradykinin levels contributes to one of the ACE inhibitors’most bothersome
side effects, chronic dry cough. ACE inhibitors in general are well tolerated,
but along with cough, can cause hyperkalemia and should be used with cau-
tion with potassium-sparing diuretics or in persons with impaired renal func-
tion. ARBs are antagonists of the angiotensin I receptor, which mediates the
direct vasoconstrictor effect of angiotensin II. This also blocks the release of
aldosterone. ARBs do not affect the bradykinin system and therefore do not
cause a cough. They are also well tolerated but, like ACE inhibitors, can cause
hyperkalemia. Aliskiren (Tekturna), a renin inhibitor has recently been intro-
duced in the United States. It appears to be as efficacious as ACE inhibitors or
ARBs, but clinical experience is limited.
68
CASE FILES: PHARMACOLOGY
APPROACH TO PHARMACOLOGY OF THE
RENIN-ANGIOTENSIN SYSTEM
Objectives
1. Know the mechanism of action of ACE inhibitors.
2. Know the therapeutic uses, side effects, and contraindications to ACE
inhibitor use.
3. Know the mechanism of action of ARBs.
4. Know the therapeutic uses, side effects, and contraindications to ARB
use.
Definitions
Hypertension: From the Seventh Report, Joint National Committee on
Detection, Evaluation, and Treatment of High Blood Pressure, normal
blood pressure is 120/80 mm Hg. Progressive disease may be staged as
prehypertensive (120–139/80–89), Stage 1 (140–159/90–99), and Stage
2 (>160/> 100).
Bradykinin:A member of a class of peptides, the kinins, that have a vari-
ety of effects on the cardiovascular system, including vasodilatation and
inflammation.
ARB:Angiotensin receptor blocker, more precisely angiotensin AT-1 receptor
blockers.
DISCUSSION
Class
The renin-angiotensin-aldosterone system provides a humoral system for
controlling blood pressure and electrolyte levels.The “sensors” in this sys-
tem monitor Na
+
, K
+
, vascular volume, and blood pressure.A reduction in
blood pressure, detected by intrarenal stretch receptors, or a fall in the deliv-
ery of Na
+
to the distal portions of the nephron results in release of renin from
the juxtaglomerular apparatus (JGA). Renin secretion can also be increased
through the baroreceptor reflex mediated by increased central nervous system
(CNS) outflow and β
1
-adrenergic receptors on the JGA. Renin is an aspartyl
protease that cleaves angiotensinogen, a 56-kD polypeptide produced in the
liver, to the decapeptide angiotensin I (Figure 9-1, “classic” pathway).
Angiotensin I is biologically inactive and is rapidly converted to the
octapeptide angiotensin II by the action of ACE, a dipeptidyl peptidase.
Angiotensin II is further metabolized within the brain and in the plasma by
aminopeptidase A, which removes the N-terminal aspartic acid to produce
angiotensin III, which may itself be further metabolized by aminopeptidase
N, which removes the N-terminal arginine yielding angiotensin IV. The latter
CLINICAL CASES 69
Figure 9-1. Schematic of angiotensin pathway.
+
“CLASSIC” INTRAVASCULAR
PRODUCTION OF AGII
LOCAL TISSUE
PRODUCTION OF AGII
Angiotensin
(56 Kd, liver)
Angiotensin I
(10 amino acids)
Angiotensin II
(8 amino acids)
Angiotensin III
(AgII 2–8)
Angiotensin IV
(AgII 3–8)
Renin
Local
Renin
ACE (dipeptidyl-peptidase)
Aminopeptidase A
Aminopeptidase N
Aminopeptidase A
Aminopeptidase N
Local ACE
Cathepsin G
Chymase
t-PA
Cythepsin G
70 CASE FILES: PHARMACOLOGY
two metabolites may play a critical role in regulating blood pressure in the
brain. Distinct from this classic intravascular pathway for the formation of
angiotensins, evidence has accumulated indicating that angiotensins can also
be produced within various tissues by a local conversion to angiotensins II, III,
and IV (see Figure 9-1).
Angiotensin II has multiple actions that act in concert to increase blood
pressure and alter electrolyte levels. Angiotensin II is a potent vasoconstric-
tor,10–40 times more potent than epinephrine, an effect mediated by recep-
tor-coupled Ca
2
channels in vascular smooth muscle cells, as described
below. Angiotensin II enhances the release of catecholaminesfrom both the
adrenal medulla and at peripheral nerve endings. Within the adrenal cortex,
angiotensin II increases the biosynthesis of aldosterone, which leads to an
increase in Na
+
and water reabsorption in the kidneys and volume expansion.
Angiotensin II has several actions within the CNS including altering vagal
tone to increase blood pressure, increasing thirst, and increasing the release of
antidiuretic hormone.
Angiotensin II also has effects on the heart and the vasculature that do not
directly affect blood pressure. Angiotensin II induces cardiac hypertrophy,
is proproliferative, and enhances matrix remodeling and the deposition of
matrix proteins, which leads to increased myocardial stiffness.Within vessel
walls, angiotensin II is proinflammatory and can stimulate the release of sev-
eral chemokines.
Three angiotensin receptors mediate these actions. The AT-1 and
angiotensin-2 (AT-2) receptors have been described in various tissues. Both
are seven-transmembrane receptors that appear to couple to various signaling
pathways. AT-1 receptors bind angiotensin II, angiotensin III, and angiotensin
IV. This receptor mediates most of the cardiovascular and central responses to
angiotensin II, including vasoconstriction of vascular smooth muscle and
aldosterone biosynthesis in the adrenal medulla. AT-1 receptors also mediate
the cardiac hypertrophic and proproliferative responses to angiotensin II. AT-2
receptors also bind angiotensin II and play a role in the development of the car-
diovascular system. In general, activation of AT-2 receptors is physiologically
antagonistic to the action of AT-1 receptors. Activation of AT-2 receptors is
hypotensive and antiproliferative and is coupled to distinctly different signal-
ing pathways compared to AT-1 receptors. Angiotensin-4 (AT-4) receptors
appear to be identical to transmembrane aminopeptidase insulin-regulated
aminopeptidase (IRAP) and have a single transmembrane domain. AT-4 recep-
tors are expressed in the numerous tissues and bind angiotensin IV. Activation
of these receptors has been reported to regulate cerebral blood flow, and to
stimulate endothelial cell expression of plasminogen activator inhibitor, and
has effects on both memory and learning.
Inhibition of the renin-angiotension system (RAS) is accomplished phar-
macologically in three ways: inhibition of the production of angiotensin II,
blockade of AT-1 receptors, or inhibition of renin activity. ACE inhibitors, or
peptidyl dipeptidase (PDP) inhibitors, include enalapril, lisinopril, fosinopril,
CLINICAL CASES 71
captopril, and seven others. These drugs differ in their chemistry and phar-
macokinetic properties, but all are orally active, have the same range of activ-
ities, and are equally effective clinically. ACE is the enzyme responsible for
both activation of angiotensin I (metabolism to angiotensin II) and inacti-
vation of bradykinin. The decreased metabolism of bradykinin is partly
responsible for the hypotensive action of ACE inhibitors, and is also responsi-
ble for enhancing the irritability of airways that leads to the dry cough asso-
ciated with ACE inhibitors.
ARBs block the action of angiotensin II by acting as antagonists at AT-1
receptors. These nonpeptide antagonists include losartan, valsartan, can-
desartan, and others. ARBs bind with high affinity to AT-1 receptors without
interfering with AT-2 or AT-4 receptors.
The ACE inhibitors and ARBs are equally effective in reducing blood
pressure. More clinical experience exists with the ACE inhibitors and it has
been well established that this class of drugs reduces the risk of second events
in patients who have had an MI and in reducing renal damage in patients with
diabetic nephropathy. Hypotension and hyperkalemia are adverse effects
seen with both classes of RAS inhibitors. Cough and angioedema, caused
by increased bradykinin levels, are more frequently seen with the ACE
inhibitors.
A newly approved agent, aliskiren, has been approved for use in the treatment
of hypertension. Aliskiren is a small molecule inhibitor of renin. In clinical trials
of more than 2000 patients, aliskiren was effective in 24-hour blood pressure
control. The effect was maintained for at least a year. Aliskiren was about as
effective as ACE inhibitors or ARBs but may cause a greater rebound in renin
production when discontinued than the other agents.
Structure
Although the various ACE inhibitors have different chemical structures, they
are mostly based on extensive modifications of L-proline. The ARBs are also
quite distinct chemically: Valsartan is an L-valine derivative, and losartan is an
imidazole derivative. Aliskiren was designed based on the crystal structure of
renin and is a nonpeptide, small molecule, transition-state mimetic that binds
to the active site of the enzyme.
Mechanism of Action
ACE inhibitors are all competitive inhibitors of angiotensin-converting enzyme.
ARBs are competitive antagonists of the angiotensin II type 1 receptor (AT-1)
Administration
All ACE inhibitors are available for oral administration. Enalaprilat, the active
metabolite of enalapril, is available for intravenous infusion. Aliskiren is an
oral agent.
72 CASE FILES: PHARMACOLOGY
Pharmacokinetics
Most of the current ACE inhibitors are prodrugs and require conversion to the
active metabolite in the liver. For example, enalapril is converted to enalapri-
lat, fosinopril is converted into fosinoprilat. Captopril and lisinopril are active
drugs that do not require metabolism. The onset of action of ACE inhibitors is
0.5–2 hours, and the duration of action is typically 24 hours (captopril is 6 hours).
Most are eliminated in the urine.
COMPREHENSION QUESTIONS
[9.1] Losartan acts to decrease which of the following?
A. AT-1 receptor activity
B. Bradykinin production
C. Production of angiotensin II
D. Renin production
[9.2] Which of the following is a limiting adverse effect of ACE inhibitors?
A. Acidosis
B. Hyperkalemia
C. Hypernatremia
D. Hypokalemia
E. Hyponatremia
[9.3] Which of the following is an advantage of losartan over enalapril?
A. Better efficacy in lower blood pressure
B. Better prevention of secondary myocardial events
C. Less cost
D. Less incidence of angioedema
Answers
[9.1] A.Losartan is a prototypical angiotensin AT-1 receptor antagonist.
[9.2] B.By reducing aldosterone levels, ACE inhibitors decrease K
+
excre-
tion in the distal nephron.
[9.3] D.Losartan does not lead to elevated bradykinin levels; thus, there is
less of an incidence of angioedema and dry cough. The effects on
blood pressure are equal. The track record for prevention of secondary
cardiovascular events is well established for ACE inhibitors, although
the same is speculated for ARBs.
CLINICAL CASES 73
74 CASE FILES: PHARMACOLOGY
PHARMACOLOGY PEARLS
Elevation of the bradykinin levels is thought to be the etiology of the
dry cough and angioedema of ACE inhibitors.
ACE inhibitors improve outcome in patients with cardiovascular dis-
ease and have been recommended as therapy in several guide-
lines.
Clinical experience suggests that inhibitors of the renin-angiotensin
system are somewhat less effective in African Americans.
ARBs block the action of angiotensin II by acting as antagonists at
AT-1 receptors.
REFERENCES
Yusuf S, Sleight P, Pogue J, et al. Effects of an angiotensin-converting-enzyme
inhibitor, ramipril, on cardiovascular events in high-risk patients: the heart out-
comes prevention evaluation study investigators. N Engl J Med 2000;342:
145–53.
Stojiljkovic L, Behnia R. Role of angiotensin system inhibitors in cardiovascular
and renal protection: a lesson from clinical trials. Curr Pharm Des 2007;13:
1335–45.
CASE 10
A 69-year-old man sees you in the office for follow-up of his chronic conges-
tive heart failure. He has a marked reduction in his ejection fraction following
a series of MIs. He also has hypertension and type II diabetes mellitus. His
symptoms include dyspnea on exertion, orthopnea, paroxysmal nocturnal dys-
pnea, and peripheral edema. He has normal renal function. He is on appropriate
treatment of his diabetes, along with an ACE inhibitor and a loop diuretic. You
decide to add digoxin to his regimen.
What is the effect of digoxin on the normal heart?
What is the effect of digoxin on the failing heart?
What neural effects does digoxin have?
What are the side effects and toxicities of digoxin?
ANSWERS TO CASE 10: AGENTS USED TO TREAT
CONGESTIVE HEART FAILURE
Summary:A 69-year-old man with congestive heart failure, hypertension, and
diabetes mellitus has a markedly low ejection fraction and is prescribed
digoxin.
Effect on a normal heart: Increased systemic vascular resistance and
constriction of smooth muscle in veins, which may decrease cardiac
output.
Effect on a failing heart: Increased stroke volume and increased
cardiac output.
Neural effects: Decreased sympathetic tone and increased vagal
activity, resulting in inhibition of sinoatrial (SA) node and delayed
conduction through atrioventricular (AV) node.
Side effects and toxicities: Induction of arrhythmias, anorexia, nausea,
vomiting, diarrhea, disorientation, and visual disturbance.
CLINICAL CORRELATION
Digoxin can be useful in improving some of the symptoms of congestive heart
failure, but its use must be closely monitored. Digoxin works by inhibiting the
sodium-potassium adenosine triphosphatase (ATPase), primarily in cardiac
muscle cells. This causes increased intracellular sodium and decreased intra-
cellular potassium. The increased sodium reduces the exchange of intracellu-
lar calcium for extracellular sodium, causing an increased intracellular
calcium level. The overall effect of this is to allow for a greater release of cal-
cium with each action potential. This has a positive inotropic effect. In a fail-
ing heart, stroke volume and cardiac output are increased. End-diastolic
volume, venous pressure, and blood volume are decreased. These circulatory
improvements also result in a reduction of sympathetic tone. This further
improves circulation by lowering systemic vascular resistance. Digoxin also
has the effect of increasing vagal activity,which inhibits the SA node and
slows conduction through the AV node. This is beneficial in patients with
atrial tachyarrhythmias such as atrial fibrillation, atrial flutter, and atrial tachy-
cardias. Digoxin has a narrow therapeutic index, and its level in the blood
must be closely monitored.The dose must be adjusted for renal impairment,
because it is cleared by the kidney. Toxic digoxin levels may produce many
types of arrhythmias, with AV blocks and bradycardia being common.
Mental status changes and gastrointestinal symptoms are common as well.
Asymptomatic elevations in digoxin levels are usually treated by discontinu-
ing or reducing the drug’s dosage. Symptomatic toxicity, particularly arrhyth-
mias, is most often treated by the IV infusion of digoxin-binding
antibodies.
76
CASE FILES: PHARMACOLOGY
APPROACH TO PHARMACOLOGY
OF THE CARDIAC GLYCOSIDES
Objectives
1. Know the mechanism of action of the cardiac glycosides.
2. Know the therapeutic uses, adverse effects, and toxicities of cardiac
glycosides.
3. Know the other agents used frequently in the treatment of congestive
heart failure.
Definitions
Cardiac glycosides:The cardenolides include digitalis, digoxin, digitoxin,
and ouabain.
Inotropic:Affecting myocardial contractility.
Chronotropic:Affecting heart rate.
Congestive heart failure:A syndrome with multiple causes that may affect
either systole or diastole. Left heart failure leads to pulmonary conges-
tion and reduced cardiac output and appears in patients with MI, aortic
and mitral valve disease, and hypertension. Right heart failure leads to
peripheral edema and ascites and appears in patients with tricuspid valve
disease, cor pulmonale, and prolonged left heart failure. The New York
Heart Association classification of congestive heart failure includes class I
(mild disease) to class IV (severe disease).
DISCUSSION
Class
The medicinal actions of the cardiac glycosides, digitalis, have been used suc-
cessfully for over 200 years, and they have both positive inotropic and antiar-
rhythmic properties. Digoxin is the most commonly used cardiac
glycoside. Cardiac glycosides act to indirectly increase intracellular cal-
cium (Figure 10-1). Digitalis binds to a specific site on the outside of the
Na
-K
-ATPaseand this reduces the activity of the enzyme. All cells express
Na
+
/K
+
-ATPase but there are several different isoforms of the enzyme; the iso-
forms expressed by cardiac myocytes and vagal neurons are the most sus-
ceptible to digitalis. Inhibition of the enzyme by digitalis causes an increase
in intracellular Na
+
and decreases the Na
+
concentration gradient across the
plasma membrane. It is this Na
+
concentration that provides the driving force
for the Na
+
-Ca
2+
antiporter. The rate of transport of Ca
2+
out of the cell is
reduced, and this leads to an increase in intracellular Ca
2+
, greater activation
of contractile elements, and an increase in the force of contraction of the
heart. The electrical characteristics of myocardial cells are also alteredby
CLINICAL CASES 77
Figure 10-1. Digoxin acts to indirectly increase intracellular calcium levels
by binding to the Na
+
-K
+
-ATPase.
K
+
K
+
K
+
Na
+
Na
+
Na
+
Ca
2
+
Ca
2
+
Na
+
Na
+
Digoxin
Outside
Inside
Na
+
Na
+
Outside
Inside
the cardiac glycosides. The most important effect is a shortening of the
action potential that produces a shortening of both atrial and ventricular
refractoriness. There is also an increase in the automaticity of the heart,
both within the AV node and in the cardiac myocytes.
Within the nervous system cardiac glycosides affect both the sympathetic
and parasympathetic systems, and parasympatheticomimetic effects predomi-
nate at therapeutic doses. Increased vagal activity inhibits the SA node and
delays conduction through the AV node.
Inacute heart failure, digitalis clearly improves contractility. Ejection frac-
tion and cardiac output is increased and symptoms are decreased. In congestive
78 CASE FILES: PHARMACOLOGY
heart failure, digitalis is used primarily in patients who are symptomatic
after optimal therapy with diuretics, ACE inhibitors, and beta blockers.
In this setting, digitalis decreases symptoms and increases exercise tolerance.
However, in patients with normal sinus rhythm,there is no decline in over-
all mortality because of deaths associated with digitalis toxicity.
Because of its action in increasing vagal tone, cardiac glycosidesare use-
ful in the treatment of several supraventricular arrhythmias including
atrial flutterand atrial fibrillation. Digitalis can control paroxysmal atrial
and AV nodal tachycardia.Its use is contraindicated in Wolff-Parkinson-
White syndrome, where it can induce arrhythmias in the alternate pathway.
Cardiac glycosides have a narrow therapeutic index. Toxic levels of car-
diac glycosides lead to depletion of intracellular K
and accumulation of
Na
(because of inhibition of Na
+
/K
+
-ATPase). This leads to partial depolar-
ization of the cell and increased excitability, both of which can lead to arrhyth-
mias including supraventricular and ventricular tachyarrhythmias. Bradycardia
and heart block are also manifestations of digitalis toxicity in the heart.
Adverse effects of digitalis on the gastrointestinal (GI) tract are common
including anorexia, vomiting, pain, and diarrhea. Central nervous system
effects include yellowed and blurred vision, dizziness, fatigue, and delir-
ium. At very high toxic ranges digitalis inhibits Na
+
/K
+
-ATPase in skeletal
muscle, resulting in hyperkalemia.
K
+
competes with digitalis for binding to the Na
+
/K
+
-ATPase;
hypokalemia increases the effectiveness of digitalis and increases toxicity.
Hypercalcemia can also increase the action of digitalis and increase toxicity.
Dopamine and dobutamine are positive inotropic agents that can be
used on a short-term basis in congestive heart failure. Dobutamine stimu-
lates D
1
- and D
2
-adrenergic receptors.The action on b
1
-adrenoreceptorsis
responsible for most of the beneficial actions of dobutamine.It is useful in
patients with acute left ventricular failure or to prevent pulmonary edema in
heart failure. At sufficient doses, dopamineinteracts with b
1
receptors and
increases myocardial contractility. It is useful in the treatment of cardio-
genic and septic shock.
Structure
These compounds share two structural features: an aglycone steroid nucleus
with a lactone at carbon 17 in the D ring, which confers the cardiotonic prop-
erties, and polymeric sugar moieties attached to carbon 3 of the A ring. Both
features are necessary for pharmacologic activity; the sugar groups are largely
responsible for the pharmacokinetic properties of these drugs.
Mechanism of Action
Inhibition of the activity of the Na
-K
-ATPase; this indirectly increases
intracellular Ca
2
.
CLINICAL CASES 79
Administration
Digoxin can be administered IV or orally. Oral bioavailability is approxi-
mately 75 percent. Digitoxin is available only as an oral agent and its bioavail-
ability is greater than 90 percent. Ouabain has limited bioavailability and is not
used clinically.
Pharmacokinetics
Digoxin is excreted by the kidney and is not metabolized. Patients with com-
promised renal function must be monitored carefully for digoxin toxicity.
Digitoxin is metabolized in the liver and renal impairment does not affect the
half-life of the drug.
COMPREHENSION QUESTIONS
[10.1] Digoxin increases cardiac contractility by directly engaging in which
of the following?
A. Activating L-type Ca
2+
channels
B. Inhibiting cardiac phosphodiesterase
C. Inhibiting myocardial Na
+
/Ca
2+
-ATPase
D. Inhibiting myocardial Na
+
/K
+
-ATPase
[10.2] Which of the following drugs may be used to increase cardiac output
in a patient with pulmonary edema secondary to MI?
A. Captopril
B. Dobutamine
C. Metoprolol
D. Verapamil
[10.3] Which of the following is the most accurate statement regarding
digoxin?
A. Decreased mortality in patients with congestive heart failure with
normal sinus rhythm
B. Increases vagal tone and decreases AV node conduction
C. Lengthens the action potential and increases the refractoriness of
the heart
D. Useful in the treatment of Wolff-Parkinson-White syndrome
Answers
[10.1] D. While digoxin reduces the amount of Na
+
-Ca
2+
exchange, this
effect is indirect and mediated by the inhibition of the Na
+
/K
+
-ATPase.
[10.2] B. Dobutamine is useful in this setting; the other choices would not
increase cardiac output.
80
CASE FILES: PHARMACOLOGY
[10.3] B. Cardiac glycosides increase vagal tone and decrease AV node con-
duction. The action potential is decreased and the refractoriness of
the heart is decreased. Mortality is not decreased in patients with nor-
mal sinus rhythm because of digoxin toxicity. Digoxin is contraindi-
cated in Wolff-Parkinson-White syndrome.
PHARMACOLOGY PEARLS
Cardiac glycosides inhibit the activity the Na
+
-K
+
-ATPase; this
indirectly increases intracellular Ca
2+
.
While several studies have found that digitalis does not improve
mortality, it is still useful in reducing symptoms in congestive
heart failure.
The increased effectiveness of digitalis as serum K
+
falls is signifi-
cant because most patients with congestive heart failure are also
frequently treated with diuretics that cause potassium loss.
Hypokalemia exacerbates digoxin toxicity.
REFERENCE
Hood W, Jr, Dans A, Guyatt G, et al. Digitalis for treatment of congestive heart failure
in patients in sinus rhythm. Cochrane Database Syst Rev 2004;2:CD002901.
CLINICAL CASES
81
This page intentionally left blank
CASE 11
A 62-year-old man is being managed in the intensive care unit following a
large anterior wall MI. He has been appropriately managed with oxygen,
aspirin, nitrates, and β-adrenergic receptor blockers but has developed recur-
rent episodes of ventricular tachycardia. During these episodes he remains
conscious but feels dizzy, and he becomes diaphoretic and hypotensive. He is
given an IV bolus of lidocaine and started on an IV lidocaine infusion.
To what class of antiarrhythmic does lidocaine belong?
What is lidocaine’s mechanism of action?
ANSWERS TO CASE 11: ANTIARRHYTHMIC DRUGS
Summary: A 62-year-old man develops symptomatic ventricular tachycardia
after an MI. He is begun on IV lidocaine.
Class of antiarrhythmic to which lidocaine belongs: Ib.
Mechanism of action: Specific Na
+
channel blocker, reduces the rate of
phase 0 depolarization, primarily in damaged tissue.
CLINICAL CORRELATION
Lidocaine is a common treatment for ventricular tachycardia in a patient who
is symptomatic and remains conscious. It works by blocking Na
+
channels and
is highly selective for damaged tissue. This makes it useful for the treatment
of ventricular ectopy associated with an MI. It is administered as an IV bolus
followed by a continuous drip infusion. It is metabolized in the liver and
undergoes a large first-pass effect. It has many neurological side effects,
including agitation, confusion, and tremors, and can precipitate seizures.
APPROACH TO PHARMACOLOGY
OF THE ANTIARRHYTHMICS
Objectives
1. Know the classes of antiarrhythmic agents and their mechanisms of action.
2. Know the indications for the use of antiarrhythmic agents.
3. Know the adverse effects and toxicities of the antiarrhythmic agents.
Definitions
Paroxysmal atrial tachycardias (PAT):Arrhythmia caused by reentry
through the AV node.
Heart block: Failure of normal conduction from atria to ventricles.
WPW:Wolff-Parkinson-White syndrome.
DISCUSSION
Class
Arrhythmias arise as a result of improper impulse generation or improper
impulse conduction. The abnormal action potentials cause disturbances in the
rate of contraction or in the coordination of myocardial contraction. The
molecular targets of antiarrhythmics are ion channels in the myocardium or
conduction pathways; these may be direct or indirect effects.
84
CASE FILES: PHARMACOLOGY
There are four ion channels of pharmacologic importance in the heart:
Voltage-activated Na
+
channel—SCN5A
Voltage-activated Ca
2+
channel—L-type
Voltage-activated K
+
channel—IKr
Voltage-activated K
+
channel—IKs
Most antiarrhythmic drugs either bind directly to sites within the pore of a
channel or indirectly alter channel activity. There are approximately 20 antiar-
rhythmics approved for use today. They are classified according to which
of the ion channels they affect and their mechanism of action (Table 11-1).
CLINICAL CASES 85
Table 11-1
SELECTED ANTIARRHYTHMIC AGENTS
PROTOTYPE
CLASS DRUG Na
+
K
+
Ca
2+
EFFECT
Ia Quinidine X X Increases refractory period,
slows conduction
Ib Lidocaine X Shortens duration of
refractory period
Ic Flecainide X X Slows conduction
II Propranolol X
*
Blocksβ
1
-adrenergic receptors
III Amiodarone X X Increases refractory period
IV Verapamil X Increases refractory period
AV node
Other
Adenosine X X
*
Decreases AV node conduction
Moricizine X
Atropine Decreases vagal tone
Digoxin Increases vagal tone
Sotalol X
Also nonselective beta blocker
*
Indirect effect mediated by decreasing cAMP.
Moricizine blocks Na
+
channels and is usually considered a class 1 antiarrhythmic, but it has
properties of Ia, Ib, and Ic drugs.
Solotol has α- and β-adrenergic antagonist properties and also inhibits K
+
channels.
The major arrhythmias of clinical concern are ventricular arrhythmias, atrial
arrhythmias, bradycardias, and heart blocks. There is also the pharmacologic
need to convert an abnormal rhythm to normal sinus rhythm (cardioconver-
sion). The class of antiarrhythmics used for any particular arrhythmia depends
on the clinical circumstances. The treatment of acute, life-threatening disease,
in contrast with the long-term management of chronic disease, requires a dif-
ferent selection of antiarrhythmics.
Class I Antiarrhythmics
Class I antiarrhythmics bind to Na
+
channels and prevent their activation.
This increases their effective refractory period and decreases conduction
velocity. Class I antiarrhythmics have a greater effect on damaged tissue com-
pared to normal tissue. This may be because of several factors:
Depolarization. Damaged tissues tend to be depolarized because of K
+
leakage—many class I antiarrhythmics preferentially bind to depolar-
ized tissues.
pH. Ischemic tissues are more acidic, and many class I antiarrhythmics
preferentially bind to membranes at low pH.
Inactivation frequency. During arrhythmias, Na
+
channels undergo more
rapid cycles of activation/inactivation. At any given time there will be an
increase in the number of inactive channels compared to normal tissues
in a normal rhythm. Class I antiarrhythmics generally bind preferentially
to Na
+
channels in the inactive state.
The subclasses a, b, and c of class I antiarrhythmics are distinguished based on
their ability to inhibit K
+
channels.
Class Ia. Procainamide is a prototype class Ia antiarrhythmic that sup-
presses the activity of Na
+
and also suppresses K
+
-channel activity.
Administered IV, it is used for the acute suppression of supraventricu-
lar and ventricular arrhythmias and for suppressing episodes of atrial
flutter and atrial fibrillation. It may be administered orally for the
long-term suppression of both supraventricular and ventricular
arrhythmia, but toxicity limits this application. Procainamide can
suppress sinoatrial (SA) and AV nodal activity, especially in patients
with nodal disease, and cause heart block. Prolonged use of pro-
cainamide is associated with increased risk of ventricular tachycar-
dias. Procainamide has some ganglionic blocking activity and can
cause hypotension and decreased myocardial contractility.A limit-
ing adverse effect of procainamide is the development of lupus-like syn-
drome characterized by skin rash, arthritis, and serositis. All patients on
procainamide will develop antinuclear antibodies within 2 years.
Procainamide is metabolized to N-acetyl procainamide (NAPA), which
has K
-channel-blocking effects. NAPA is excreted by the kidney,
and plasma levels of procainamide and NAPA should both be moni-
tored especially in patients with renal disease.
86 CASE FILES: PHARMACOLOGY
Class Ib. Lidocaine is very specific for the Na
+
channel and it blocks both
activated and inactivated states of the channel. It must be administered
parenterally. Lidocaine has been used extensively to suppress ven-
tricular arrhythmias associated with acute MI or cardiac damage
(surgery). It has been used prophylactically to prevent arrhythmias
in patients with MI, but there is controversy as to the overall bene-
fit in decreasing mortality. Lidocaine is metabolized in the liver and
has relatively short half-life (60 minutes). This limits its adverse
effects which generally are mild and rapidly reversible.Overdose can
produce sedation, hallucinations, and convulsions.
Class Ic. Flecainide inhibits both Na
+
and K
+
channels but shows no pref-
erence for inactivated Na
+
channels. It delays conduction and increases
refractoriness.It is effective for the control of atrial arrhythmias and
it is very effective in suppressing supraventricular arrhythmias.A
recent large clinical trial with patients with ischemic heart disease
demonstrated that flecainide is associated with increased mortality.
Currently its use is restricted to patients with atrial arrhythmias
without underlying ischemic heart disease.
Class II Agents
Endogenous catecholamines increase myocardial excitability and can
trigger ventricular arrhythmias. β-Adrenergic receptor blockade indirectly
suppresses L-type Ca
2+
-channel activity. This slows phase 3 repolarization and
lengthens the refractory period. Reduction in sympathetic tone depresses auto-
maticity, decreases AV conduction, and decreases heart rate and contractility.
Beta blockers are useful for the long-term suppression of ventricular
arrhythmias particularly in patients at risk for sudden cardiac arrest.
Beta blockers are most effective in patients with increased adrenergic activity:
Surgical or anesthetic stress.
Anginal pain and MI.
Congestive heart failure and ischemic heart disease.
Hyperthyroidism.
Beta blockers have been shown to reduce mortality and second cardio-
vascular events by 25–40% in patients with post-MI.
There are a large number of beta blockers approved for use as antiarrhyth-
mics. Two of particular interest are
1. d,l-sotalol, which is particularly effective as an antiarrhythmic agent
because it combines inhibition of K
+
channels with beta-blocker activity
2. Metoprolol, a specific β
1
antagonist, which reduces the risk of pul-
monary complications
d,l-sotalol is a racemic mixture; l-sotalol is an effective, nonselective β-
adrenergic antagonist;and d-sotalol is a class III antiarrhythmic that inhibits K
+
channels. It is an oral agent with a long half-life (20 hours) that can maintain
CLINICAL CASES 87
therapeutic blood levels with once a day dosing. d,l-sotalol is useful for the
long-term suppression of ventricular arrhythmias, especially in patients at risk
of sudden death. It is also used to suppress atrial flutter and fibrillation and
paroxysmal atrial tachycardia. It is a valuable adjunct in the use of implantable
cardiac defibrillators, decreasing the number of events that require defibrilla-
tion. At low doses, the β-adrenergic-blocking activity, and associated adverse
effects, predominates. At higher doses, the K
+
-channel inhibitory effects pre-
dominate with the risk of developing ventricular tachycardia.
Class III Antiarrhythmics
Drugs in this class include bretylium, dofetilide, and amiodarone.These
agents act predominantly to inhibit cardiac K
channels (IKr). This length-
ens the time to repolarize and prolongs the refractory period. Amiodarone is
also a potent inhibitor of Na
+
channels and has α- and β-adrenergic antagonist
activity.
Amiodarone has an unusual structure related to thyroxine. It can be
administered IV or orally, but its actions differ depending on route of admin-
istration. IV-administered amiodarone has acute effects to inhibit K
+
-channel
activity, slowing repolarization, and increasing the refractory period of all
myocardial cell types. Administered orally in a more chronic setting, it leads
to long-term alterations in membrane properties with a reduction in both Na
+
-
and K
+
-channel activity and decrease in adrenergic receptor activity.
Amiodarone is used extensively for ventricular and atrial arrhythmias
and has little myocardial depressant activity, allowing it to be used in
patients with diminished cardiac function. Administered IV, amiodarone is
effective in treating ventricular tachycardia and to prevent recurrent ventricu-
lar tachycardia, and to suppress atrial fibrillation. Oral amiodarone is useful
for arrhythmias that have not responded to other drugs (such as adenosine) and
for long-term suppression of arrhythmias in patients at risk of sudden cardiac
death.
Amiodarone has little myocardial toxicity, does not impair contractility,
and rarely induces arrhythmias. Most of the adverse effects of amiodarone
result from its long half-life (13–103 days) and poor solubility. Amiodarone
deposits in the lung and can cause irreversible pulmonary damage.
Similarly, amiodarone can be deposited in the cornea causing visual dis-
turbances or in the skin where it can cause a bluish tinge.
Class IV Antiarrhythmics
The class IV antiarrhythmics act by directly blocking the activity of L-
type Ca
2
channels. Verapamiland diltiazem are the major members of this
class, and they have a similar pharmacology. Verapamil blocks both active
and inactive Ca
2
channels and has effects that are equipotent in cardiac and
peripheral tissues. The dihydropyridinessuch as nifedipine have little effect
on Ca
2
channels in the myocardium, but are effective in blocking Ca
2
88 CASE FILES: PHARMACOLOGY
channels in the vasculature. Verapamilhas marked effects on both SA and
AV nodesbecause these tissues are highly dependent on Ca
2+
currents. AV
node conduction and refractory period are prolonged and the SA node is
slowed. Verapamil and diltiazem are useful for reentrant supraventricu-
lar tachycardias and can also be used to reduce the ventricular rate in atrial
flutter or fibrillation. The major adverse effect of verapamilis related to its
inhibition of myocardial contractility.It can cause heart block at high doses.
Other Antiarrhythmics
Adenosine is a very short-acting drug (approximately 10 seconds) used
specifically to block PAT. Adenosine binds to purinergic A1 receptors.
Activation of these receptors leads to increased potassium conductance and
decreased in calcium influx. This results in hyperpolarization and a decrease
in Ca
2+
-dependent action potentials. The effect in the AV node is marked with
a decrease in conduction and an increase in nodal refractory period. Effects on
the SA node are smaller. Adenosine is nearly 100 percent effective in convert-
ing PAT to sinus rhythm. Adenosine must be given IV, and because of its short
half-life, it has few adverse effects. Flushing and chest pain are frequent but
typically resolve quickly.
Digoxin (see Case 10) blocks Na
+
-K
+
-ATPase and indirectly increases
intracellular Ca
2+
. In the myocardium this causes an increase in contractility;
in nerve tissue the predominant effect is to increase neurotransmitter release;
and the parasympathetic system (vagus) is affected more than the sympathetic
system. The increased vagal tone results in increased stimulation of mus-
carinic acetylcholine receptors that slow conduction in the AV node. Digoxin
is very effective controlling the ventricular response rate in patients with
atrial fibrillation or flutter.Digoxin can be administered IV to acutely treat
atrial arrhythmias or orally for long-term suppression of abnormal atrial
rhythms. Digitalis is less effective than adenosine in PAT and should not be
used in Wolff-Parkinson-White syndrome.
Atropine is a muscarinic antagonist that can be used in some brady-
cardias and heart blocks. It can be administered to reverse heart block
caused by increased vagal tone such as an MI or digitalis toxicity.
Atropine is administered IV, and it exerts its effect within minutes.
COMPREHENSION QUESTIONS
[11.1] Which of the following is the most effective agent for converting
paroxysmal atrial tachycardia to normal sinus rhythm?
A. Adenosine
B. Atropine
C. Digoxin
D. Lidocaine
CLINICAL CASES 89
[11.2] Which of the following best describes a pharmacologic property of
amiodarone?
A. α-Adrenergic agonist
B. β-Adrenergic agonist
C. Activation of Ca
2+
channels
D. Inhibition of K
+
channels
[11.3] A 45-year-old man is noted to have dilated cardiomyopathy with atrial
fibrillation and a rapid ventricular rate. An agent is used to control the
ventricular rate, but the cardiac contractility is also affected, placing
him in pulmonary edema. Which of the following agents was most
likely used?
A. Amiodarone
B. Digoxin
C. Nifedipine
D. Verapamil
Answers
[11.1] A. Adenosine is nearly 100 percent effective in converting PAT.
Digoxin could be used but is less effective.
[11.2] D. Amiodarone blocks both Na
+
and K
+
channels and has α- and β-
adrenoreceptor antagonist activities. The latter would indirectly
decrease Ca
2+
-channel activity.
[11.3] D. Verapamil is a calcium-channel-blocking agent that slows conduc-
tion in the AV node, but it also has a negative inotropic effect on the
heart.
PHARMACOLOGY PEARLS
Amiodarone is typically the first choice in acute ventricular arrhyth-
mias.
Adenosine is the best choice to convert PAT to sinus rhythm.
Long-term benefit of using class I antiarrhythmics is uncertain, but
mortality is not decreased.
Beta blockers have been shown to reduce mortality and second
cardiovascular events by 25–40% in patients post-MI.
90
CASE FILES: PHARMACOLOGY
REFERENCES
Cooper HA, Bloomfield DA, Bush DE, et al. Relation between achieved heart rate
and outcomes in patients with atrial fibrillation (from the atrial fibrillation follow-
up investigation of rhythm management [AFFIRM] study): AFFIRM investiga-
tors. Am J Cardiol 2004;93(10):1247–53.
Boriani G, Diemberger I, Biffi M et al. Pharmacological cardioversion of atrial fib-
rillation: current management and treatment options. Drugs 2007;64:2641–62.
CLINICAL CASES
91
This page intentionally left blank
CASE 12
A 50-year-old man presents for follow-up of his hypertension. He is main-
taining a low-sodium diet, exercising regularly, and taking metoprolol at max-
imum dosage. He is on no other medications. His blood pressure remains
elevated at 150/100 mm Hg. His examination is otherwise unremarkable. You
decide to add a thiazide diuretic to his regimen.
What is the mechanism of action of metoprolol?
What is the mechanism of action of thiazide diuretics?
What electrolyte abnormalities commonly occur with thiazide
diuretics?
ANSWERS TO CASE 12: ANTIHYPERTENSIVE
AGENTS
Summary: A 50-year-old man with inadequately controlled hypertension is
prescribed a thiazide diuretic.
Mechanism of action of metoprolol:β
1
-Selective adrenoreceptor antagonist.
Mechanism of action of thiazide diuretics: Inhibit active reabsorption
of NaCl in the distal convoluted tubule by interfering with a specific
Na
+
/Cl
cotransporter.
Electrolyte abnormalities seen with thiazide diuretics:Hypokalemia,
hyponatremia, hypochloremia.
CLINICAL CORRELATION
Thiazide diuretics are the recommended first-line agents for most people
with hypertension. They are frequently used in combination with other
classes of antihypertensives. Thiazides inhibit the active reabsorption of Na
+
.
This causes an increase in the excretion of Na
+
, Cl
, and K
+
. They also reduce
the excretion of Ca
2+
by increasing its absorption. The excretion of sodium and
water reduces intravascular volume and contributes to their antihypertensive
effect. Thiazides are used as single agents primarily in mild to moderate hyper-
tension. They are often added as second agents when other drugs alone cannot
control a patient’s hypertension. The electrolyte abnormalities caused by thi-
azides can be clinically important. Hypokalemia occurs frequently, especially
when higher doses of thiazides are used. Patients need to be instructed to fol-
low a high potassium diet and frequently require potassium supplementation.
Thiazides can also elevate serum uric acid levels, which can precipitate gout
in susceptible individuals.
APPROACH TO PHARMACOLOGY
OF ANTIHYPERTENSIVE AGENTS
Objectives
1. Know the classes of antihypertensive medications and their mecha-
nisms of action.
2. Know the common side effects of the antihypertensive agents.
Definitions
Hypertension: Blood pressure continuously elevated to levels greater than
120/80 mm Hg. Pressures of 130/90 mm Hg are considered prehyper-
tensive.
94
CASE FILES: PHARMACOLOGY
Essential hypertension: Hypertension of unknown etiology makes up
approximately 90 percent of hypertensive patients.
DISCUSSION
Class
There are 12 major classes of drugs that are used as oral antihypertensive
drugs, and these include drugs that act centrally and those that work in the
periphery. Antihypertensive drugs may cause vascular smooth-muscle relax-
ation, vascular volume reduction, or a decrease in cardiac output. This is
accomplished by decreasing Ca
2+
in vascular smooth muscle cells or by reduc-
ing Na
+
reabsorption on the kidney. Table 12-1 lists these major classes.
Lifestyle modifications include smoking cessation, weight management,
and commencement of an exercise program.
The Joint National Commission (JNC) also emphasized the need to recog-
nize and treat systolic hypertension,which is associated with a higher degree
of risk of MI in patients older than 45 years. Systolic hypertension is more
difficult to treat than diastolic hypertension and frequently requires multiple
drugs acting via different mechanisms.
The JNC-7 report and other recent studies recommend thiazide diuretics as
the first-line agent for the treatment of hypertension in most cases(Table 12-2).
This conservative approach is based on data supporting the fact that these
agents decrease morbidity and mortality in clinical trials. The other
agents that should be considered for initial monotherapy include the beta
blockers, the renin-angiotensin system inhibitors (either ACE inhibitors or
ARBs), α-adrenoreceptor antagonists, calcium-channel antagonists, and
arterial vasodilators. All have been shown to reduce blood pressure by
10–15 mm Hg.
Diuretics
Diuretics cause an initial reduction in blood pressure by facilitating loss of
Na
and water.This leads to a decrease in cardiac output and blood pressure.
However, after 8 weeks, cardiac output returns to normal while blood pressure
remains reduced. This is thought to be caused by a reduction in the vasocon-
strictive activities of Na
on vascular smooth musclesthat include elevation
of intracellular Ca
2
via the Ca
2
/Na
antiporter. Thiazidediuretics, which
reduce the activity of a specific Na
+
Cl
cotransporter (NCC2) in the distal
convoluted tubule,are the class of diuretics most often used for hypertension.
In refractory cases or in patients with concomitant edema, loop diuretics can
be used with caution. Loop diuretics reduce Na
+
reabsorption in the ascending
limb of the loop of Henle by reducing the activity of another Na
+
-K
+
-2Cl
cotransporter (NKCC) and can produce a profound loss of Na
+
and K
+
. Both
thiazides and loop diuretics can cause hypokalemia and hyponatremia. A
common complaint associated with diuretic use is the increased frequency of
CLINICAL CASES 95
96 CASE FILES: PHARMACOLOGY
Table 12-1
SELECTIVE CLASSES OF ANTIHYPERTENSIVE AGENTS
COMMON
PROTOTYPE ADVERSE
CLASS DRUG MOA EFFECT
Beta blocker Propranolol Adrenergic Fatigue, reduction
β-receptor antagonist on libido
α
1
-Antagonist Prazosin Adrenergic receptor Orthostatic
antagonist hypotension
ACE inhibitor Enalapril Reduces production Hyperkalemia
of angiotensin II
ARB (angiotenson Losartan AT-1 receptor Hyperkalemia
receptor blocker) antagonist
Renin Aliskiren Inhibits renin Angioedema,
inhibitor activity headache, dizziness,
gastrointestinal
events
Specific Eplerenone Aldosterone-receptor Hyperkalemia
aldosterone- antagonists
receptor antagonist
Diuretic—loop Furosemide Reduces Na
+
Hypokalemia
reabsorption in
loop of Henle
Diuretic—distal Hydrochlorothiazide Reduces Na
+
Hypokalemia
tubule reabsorption at site 3
Ca
2+
channel Nifedipine Blocks Ca
2+
entry Hypotension
blocker in vascular smooth arrhythmias
muscle cells
Arterial Minoxidil Hyperpolarizes Orthostatic
vasodilators VSMC hypotension
Central acting Clonidine α
2
-Adrenergic Sedation,
vasodilator agonist, I
2
-receptor depression
agonist
Adrenergic Guanethidine Inhibits release of Postural
neuron blockers norepinephrine hypotension
Neuronal Reserpine Depletes neurons of Sedation
uptake inhibitor neurotransmitters
urination. Spironolactone and eplerenone are antagonists of the aldosterone
receptor and are weakly diuretic. Eplerenone is much more specific for
the alososterone receptor compared to spironolactone.
Beta () Blockers
Use of β-adrenoreceptor blockers for hypertension relies on decreasing
cardiac output and decreasing peripheral vascular resistance. The vari-
ous drugs in this class vary in their potency on β
1
receptors; metoprolol is
more than 1000 times more potent in blocking β
1
compared to β
2
receptors,
giving this drug a relative cardioselectivity.Blockade of β
1
-adrenoreceptors
in the JGA of the kidney reduces renin secretion, and this reduces the pro-
duction of angiotensin II. Nonselective beta blockers such as propranolol
cause a number of predictable adverse effects including bronchoconstriction
(contraindicating use in asthmatics); a decrease in the production of insulin
(contraindicating use in diabetics); and central nervous system (CNS) effects
including depression, insomnia, and a decline in male potency. In addition,
the nonselective agents increase both triglycerides and low-density lipopro-
tein (LDL). These effects are reduced but not eliminated with the more β
1
-
selective agents.
Alpha
1
(
1
) Blockers
Prazosin, doxazosin, and terazosin reduce blood pressure by antagoniz-
ing α
1
-adrenoreceptors in vascular smooth muscle.Blockade of this recep-
tor reduces intracellular cyclic adenosine monophosphate (cAMP) and leads
to a reduction in intracellular Ca
2+
. Orthostatic hypotension is common on
initiation of therapy but diminishes. Dizziness and headache are also adverse
effects. α
1
-Blockers appear to reduce LDL cholesterol. Alphablockers are used
CLINICAL CASES 97
Table 12-2
THE JOINT NATIONAL COMMITTEE ON HYPERTENSION HAS
DEFINED FOUR CATEGORIES OF HYPERTENSION
BLOOD BLOOD
PRESSURE PRESSURE
SYSTOLIC DIASTOLIC RECOMMENDED
STAGE (mm Hg) (mm Hg) TREATMENT
Normal <120 and <80
Prehypertensive 120–139 or 80–89 Lifestyle
modification
Hypertensive stage 1 140–159 or 90–99 Lifestyle modification, R
x
Hypertensive stage 2 160 or 100 Lifestyle modification, R
x
primarily for hypertension in patients who also have symptomatic prostatic
hyperplasia. Because of excess cases of congestiveheart failurein users of
alpha blockers, these agents should not be used as first-line therapy in hyper-
tension.
Calcium Channel Blockers
Calcium channel (Ca
2
-channel) blockers are useful antihypertensives
and can reduce blood pressure by 10–15 mm Hg. These agents exert their anti-
hypertensive effect by blocking L-type (voltage-sensitive) Ca
2
channels.
By blocking the entry of Ca
2+
into the cell, less is available to activate the con-
tractile apparatus, and within vascular smooth muscle, this produces a reduc-
tion in vascular tone. Three distinct chemical classes comprise the
Ca
2+
-channel antagonists, dihydropyridinesinclude nifedipine, diphenylalky-
lamines include verapamil, and benzothiazepines include diltiazem. All are
approved for treating hypertension. Nifedipine and the other dihydropyridines
have less of an effect on the heart than verapamil and diltiazem. Verapamil
has the greatest effect on the heart and can significantly reduce contrac-
tility. Because of its effect on the heart, verapamil can be used to treat
supraventricular arrhythmias as well as variant angina. Depression of cardiac
function is the greatest adverse effect of the Ca
2+
-channel blockers, and this is
markedly diminished with the dihydropyridines. Dihydropyridines can induce
a reflex tachycardia in response to their blood pressure–lowering effect.
However, clinical trials with short-acting nifedipine suggested that there was
an increase in the risk of MI in patients treated for hypertension, and these
agents should not be used to treat the disease.
Renin-Angiotensin System Inhibitors
Inhibitors of the renin-angiotensin system, both ACE inhibitors and ARBs
are effective for hypertension monotherapy. ACE inhibitors block the conver-
sion of the inactive angiotensin I to the potent angiotensin II. Angiotensin II
acts to increase blood pressure in several ways. In vascular smooth muscle it
increases intracellular Ca
2+
and produces pronounced vasoconstriction. At
peripheral nerve endings and in the adrenal medulla it increases the amount of
catecholamines released on stimulation. In the zona glomerulosa of the adre-
nal cortex it acts to stimulate the biosynthesis of aldosterone, which increases
renal Na
+
and water retention. Adverse effects include hypotension, dizziness,
and fatigue; rarely, hyperkalemia may occur. A dry cough and angioedema
may occur as a result of the reduction in degradation of bradykinin that is
brought about by these drugs.
Aliskiren (Tekturna) reduces the activity of renin; this in turn causes a
reduction in the production of angiotensin II. Clinical experience is lacking for
aliskiren, but it appears about as effective as ACE inhibitors and has fewer side
effects. During clinical trials, headache, dizziness, and some gastrointestinal
98
CASE FILES: PHARMACOLOGY
events were the most common side effects, and angioedema was observed in a
few patients.
Angiotensin II acts through AT-1 and AT-2 receptors, which in turn couple to
numerous signal transduction pathways. The hypertensive actions of angiotensin
II are mediated by AT-1 receptors. Losartan, valsartan, and other AT-1 receptor
blockers are also effective in reducing blood pressure by 10–15 mm Hg. The
adverse-effect profile is similar to the ACE inhibitors but without the cough or
angioedema.
Direct Arterial Vasodilators
Arterial vasodilators act by increasing the efflux of potassium from the
cell. This causes hyperpolarization across the plasma membrane that dimin-
ishes the activity of the voltage-regulated L-type calcium channel. In vascular
smooth muscle cells this produces a reduction in vascular tone. Minoxidil and
hydralazine are the two most commonly used oral vasodilators used to treat
hypertension. Both have pronounced effects on the resistance vessels and little
effect on veins. Because of their predominant effect on arterioles, these agents
provoke the baroreceptor reflex that includes tachycardia, vasoconstric-
tion, and the release of renin.For this reason, these agents are usually com-
bined with a beta blocker and a diuretic.
Centrally Acting Agents
Centrally acting vasodilators such as clonidine and methyldopa act as
2
-adrenergic receptor agonists in the vasomotor center within the
medulla. These agents decrease sympathetic outflow and thereby decrease
vascular tone and cardiac output. The use of these agents as antihypertensives
has been overshadowed by the introduction of ACE inhibitors and ARBs and
Ca
2+
-channel blockers. This is largely a result of the adverse effects, which are
mostly in the CNS and include sedation, depression, and dry mouth. However,
they are still used in cases of refractory hypertension.
Peripheral Sympathetic Inhibitors
Peripheral sympatholytic agents used for hypertension include guanethi-
dine and reserpine. Guanethidine enters sympathetic nerve terminals by trans-
port and replaces norepinephrine in transmitter vesicles. Release of
norepinephrine is thereby diminished. Reserpine blocks the uptake and storage
of biogenic amines, and this diminishes the amount of transmitter released on
stimulation. Because of much higher rates of adverse effects, these agents are
rarely used to treat simple hypertension but may be combined in the treatment
of refractory hypertension.
CLINICAL CASES 99
COMPREHENSION QUESTIONS
[12.1] The inclusion of spironolactone with a thiazide diuretic in a regimen to
treat hypertension is done to achieve which of the following?
A. Reduce hyperuricemia
B. Reduce Mg
+
loss
C. Decrease the loss of Na
+
D. Reduce K
+
loss
[12.2] Which of the following drugs would be the best to treat moderate
hypertension in a diabetic patient with mild proteinuria?
A. Enalapril
B. Propranolol
C. Hydrochlorothiazide
D Nifedipine
[12.3] A 33-year-old man is diagnosed with essential hypertension. He is
started on a blood pressure medication, and after 6 weeks, he notes
fatigue, rash over his face, joint aches, and effusions. A serum antinu-
clear antibody (ANA) test is positive. Which of the following is the
most likely agent?
A. Hydralazine
B. Propranolol
C. Thiazide diuretic
D. Nifedipine
E. Enalapril
Answers
[12.1] D. Spironolactone is a “potassium-sparing” diuretic that reduces K
+
excretion in the collecting duct. It diminishes the K
+
-wasting effects
of thiazide diuretics.
[12.2] A. ACE inhibitors, such as enalapril, have been shown to reduce the
progressive loss of renal function that is often seen in diabetic
patients. The nonselective beta blocker, propranolol, would worsen
the diabetes.
[12.3] A. Hydralazine is associated with a lupus-like presentation, with pho-
tosensitivity, malar rash, joint pain, and sometimes pericardial effu-
sion or pleural effusion.
100
CASE FILES: PHARMACOLOGY
PHARMACOLOGY PEARLS
The ALLHAT clinical trial (Antihypertensive and lipid-lowering
treatment to prevent heart attack) compared amlodipine, a dihy-
dropyridine Ca
2+
-channel blocker, lisinopril, an ACE inhibitor,
doxazosin, an α
1
-adrenergic antagonist with chlorthalidone, a
thiazide diuretic.
Thiazide diuretics are the preferred initial therapy for hypertension
in most cases.
Beta-blocking agents can cause depression, insomnia, male impo-
tency, bronchoconstriction, and decreased production of insulin.
REFERENCES
Kostis JB. The importance of managing hypertension and dyslipemia to decrease
cardiovascular disease. Cardiovasc Drugs Ther Epub; 21(4):297–309:2007.
ALLHAT Officers and Coordinators for the ALLHAT Collaborative Research
Group. Major outcomes in moderately hypercholesterolemic, hypertensive
patients randomized to pravastatin vs usual care: the antihypertensive and lipid-
lowering treatment to prevent heart attack trial (ALLHAT-LLT). The antihyper-
tensive and lipid-lowering treatment to prevent heart attack trial. JAMA
2002;288(23):2998–3007.
CLINICAL CASES
101
This page intentionally left blank
CASE 13
A 60-year-old man with hypertension and type II diabetes comes in for a
follow-up visit. Along with making appropriate diet and lifestyle changes, he
is taking an ACE inhibitor-thiazide diuretic combination for his hypertension
and metformin for his diabetes. His blood pressure and diabetes are under
acceptable control. Routine blood work revealed normal electrolytes, renal
function, and liver enzymes. He is noted to have elevated total cholesterol and
low-density lipoprotein (LDL) levels, which have remained high in spite of
his lifestyle changes. In an effort to reduce his risk of developing coronary
artery disease, you start him on a 3-hydroxy-3-methylglutaryl-coenzyme A
(HMG-CoA) reductase inhibitor.
What is the mechanism of action of HMG-CoA reductase
inhibitors?
What effect do they have on total and LDL cholesterol levels?
What are the common adverse effects of HMG-CoA reductase
inhibitors?
ANSWERS TO CASE 13: LIPID-LOWERING AGENTS
Summary:A 60-year-old man has hypertension, diabetes, and hyperlipidemia
and is started on an HMG-CoA reductase inhibitor.
Mechanism of action of HMG-CoA reductase inhibitors:
Competitive inhibition of the rate-limiting enzyme in cholesterol
biosynthesis results in compensatory increase in plasma cholesterol
uptake in the liver mediated by an increase in the number of LDL
receptors.
Effect on total cholesterol: Up to 30 percent reduction.
Effect on LDL cholesterol: Up to 50 percent reduction.
Common adverse events:Elevated liver enzymes and hepatotoxicity,
myalgia and myositis, irritability, sleep disturbance, anxiety.
CLINICAL CORRELATION
HMG-CoA reductase inhibitors are in wide clinical use with proven benefit in
lowering cholesterol levels and reducing the risk of coronary artery disease in
susceptible individuals. They competitively antagonize the rate-limiting
enzyme in cholesterol biosynthesis. Reduced cholesterol synthesis spurs a
compensatory increase in hepatic uptake of plasma cholesterol mediated by an
increase in the number of LDL receptors. The net effect of this is to lower the
plasma levels of lipoproteins, especially LDL cholesterol. The effect on high-
density lipoprotein (HDL) cholesterol is less pronounced. Although generally
very well tolerated, severe hepatotoxicity has occurred, and monitoring of
liver enzymes is mandatory while taking these medications. Myalgiais a com-
mon side effect, but rarely severe, myositis and rhabdomyolysis have occurred.
Hepatotoxicity and myositis can occur while using an HMG-CoA reductase
inhibitor alone, but they become more likely when combinations of medica-
tions are used.
APPROACH TO PHARMACOLOGY
OF LIPID-LOWERING DRUGS
Objectives
1. Know the drugs used to treat hyperlipoproteinemias.
2. Know the adverse effects and toxicities of the drugs.
3. Know the therapeutic uses of each of the lipid-lowering agents.
104
CASE FILES: PHARMACOLOGY
Definitions
Hyperlipidemia: An elevation in either plasma cholesterol or plasma
triglycerides or both.
Myopathy:General term for any disease of muscle.
Myositis: Muscle pain with increased creatinine kinase levels.
Rhabdomyolysis: Muscle pain accompanied by a greater than tenfold
increase in creatinine kinase above upper limits of normal, indicating
serious muscle damage.
LDL cholesterol: Low-density lipoprotein. Atherogenic lipoprotein parti-
cle. Several subfractions have been identified, and the smallest are the
most atherogenic. It contains apolipoproteins B
100
(apo B
100
; interacts
with LDL receptor), Apo E (interacts with LDL receptor and Apo E
receptor), and Apo C (activates lipoprotein lipase).
HDL cholesterol: High-density lipoprotein particle involved in transport-
ing cholesterol from the periphery back to the liver. Has antiatheroscle-
rotic activity. Contains Apo A, C, and D.
VLDL:Very-low-density lipoprotein, a triglyceride-rich lipoprotein particle
synthesized in the liver.
DISCUSSSION
Class
Drugs that decrease plasma lipids are among the most commonly prescribed
today. Some of these affect primarily cholesterol (e.g., the statins) and are use-
ful in the treatment of hypercholesterolemia while other agents affect primarily
triglycerides (e.g., gemfibrozil).
The National Cholesterol Education Program (NCEP) has classified
levels of plasma cholesterol (Table 13-1). The LDL cholesterol treatment
goal is determined by assessing the risk of cardiovascular disease of indi-
vidual patients. The major risk factors that modify LDL goals are listed in
Table 13-2.
Known CHD include patients who have had an infarction or angina or a
surgical procedure for cardiovascular disease. In addition, patients with
peripheral arterial disease, abdominal aortic aneurism, or symptomatic
carotid artery disease or diabetes are considered to have known CHD or a
high risk for CHD. The NCEP classification and the risk assessment are
combined and used to modify the LDL cholesterol goals as illustrated in
Table 13-3.
CLINICAL CASES 105
106 CASE FILES: PHARMACOLOGY
Table 13-2
RISK FACTORS FOR CARDIOVASCULAR DISEASE
Clinical CVD
Cigarette Smoking
Hypertension (BP >140/90 mm Hg) or on an antihypertensive drug
Low HDL cholesterol (<40 mg/dL)
Family history of premature coronary heart disease
Age (men >45 years, women > 55 years)
Poor nutrition
Table 13-1
NATIONAL CHOLESTEROL EDUCATION PROGRAM (NCEP) LEVELS
OF PLASMA CHOLESTEROL
LDL CHOLESTEROL (mg/dL) CATEGORIZATION
<100 Optimal
100–129 Near/above optimal
130–139 Borderline high
160–189 High
>190 Very high
TOTAL CHOLESTEROL (mg/dL)
<200 Desirable
200–239 Borderline high
>240 High
HDL CHOLESTEROL (mg/dL)
<40 Low
>60 High
Agents Used for Hypercholesterolemia
Statins
Of the drugs that decrease plasma cholesterol, the statins have gained the
widest use. The statins are structural analogs of the substrate HMG-CoA
that inhibit the activity of the enzyme HMG-CoA reductase at nanomolar
concentrations. This enzyme is required for the synthesis of isoprenoids and
cholesterol. By inhibiting de novo biosynthesis of cholesterol, cellular uptake
of cholesterol from plasma via the LDL receptor is increased, reducing plasma
cholesterol levels. Because statins have additional actions to inhibit the
production of the triglyceride-rich VLDL, this makes them useful in the
management of patients with hypertriglyceridemia; atorvastatin and rosuvas-
tatin are particularly effective in this regard. There is evidence that statins also
have anti-inflammatory activity, and this may contribute to their reduction in
cardiovascular events. Statins may also reduce the rate of bone resorption
and thereby lessen osteoporosis. This effect is thought to be caused by the
inhibition of isoprenoid biosynthesis in osteoclast precursors, which inhibits
their differentiation into mature osteoclasts. Six statins are approved in the
United States: lovastatin, rosuvastatin, fluvastatin, atorvastatin, pravas-
tatin, and simvastatin. They differ in efficacy: Rosuvastatin has been
reported to reduce LDL cholesterol by more than 60 percent; atorvastatin,
approximately 50 percent; and pravastatin and fluvastatin, approximately
35 percent. All of the statins are active orally. Lovastatin and simvastatin are
prodrugs that are converted to their active metabolite by the liver.
The two major adverse effects associated with statin use are hepato-
toxicity and myopathy.Hepatotoxicity was initially thought to be as high as
1 percent with elevations in hepatic transaminases as high as three times the
upper limits. Subsequent clinical trials indicate that the actual incidence of
hepatotoxicity is much lower. Hepatic transaminase levels should be moni-
tored on initiation of therapy and at least yearly thereafter. The myopathy asso-
ciated with statin use occurs in less than 0.1 percent of patients. However,
severe rhabdomyolysis has occurred rarely, and one statin, cerivastatin, was
removed from the market after several rhabdomyolysis-associated deaths.
CLINICAL CASES 107
Table 13-3
CARDIOVASCULAR RISK AND LDL GOAL
LDL GOAL
RISK LEVEL (mg/dL)
Known CHD <100
2 risk factors <130
0–1 risk factors <160
Bile-Acid-Binding Resins
The bile acid sequestrants are also useful in reducing plasma cholesterol.
Cholestyramine, colestipol, and colesevelam are ion-exchange resins that
nonspecifically bind bile acids within the intestine and thereby reduce their
enterohepatic circulation. This increases de novo hepatic bile acid synthesis
and the cholesterol for this synthesis comes, in part, from the plasma via the
LDL receptor. Bile acid sequestrants typically reduce plasma cholesterol
by 15–20 percent with no effect on triglycerides. Because they are not
absorbed, the bile acid sequestrants are quite safe, and adverse effects are typ-
ically gastrointestinal and include bloating and constipation. In the intestine,
these agents bind many molecules other than bile acids and they impair the
absorption of lipid-soluble vitamins and many drugs including digoxin,
furosemide, thiazides, coumarin, and some statins. Patient adherence with
these drugs is poor.
Inhibitors of Cholesterol Absorption
Ezetimibe is a new class of cholesterol-lowering drug that acts within
the intestine to reduce cholesterol absorption.Cholesterol is absorbed from
the small intestine by a process that includes specific transporters that have not
been completely characterized. Ezetimibe appears to block one or more of
these cholesterol transporters, thereby reducing cholesterol absorption.
Ezetimibe used alone produces a reduction in plasma cholesterol of approxi-
mately 19 percent and an approximate 10 percent decline in triglyceride
levels. When combined with a statin, reductions in plasma cholesterol as
high as 72 percent have been reported in clinical trials. The complementary
mechanisms—inhibition of cholesterol biosynthesis by statins and inhibition
of cholesterol absorption by ezetimibe—may be useful in treating patients
with refractory hypercholesterolemia. Few adverse effects have been reported
with ezetimibe, but clinical experience is limited. The most frequently
reported adverse effects are back and joint pain.
Nicotinic Acid
Niacin, at doses well beyond those used as a vitamin, has effects on all
plasma lipids. It reduces LDL cholesterol by 20–30 percent and reduces
triglycerides by 35–45 percent. It is the best agent available for increasing
HDL. Niacin inhibits VLDL production in the liver by inhibiting both the syn-
thesis and esterification of fatty acids. LDL levels are reduced as a consequence
of the decline in VLDL synthesis. Niacin inhibits lipolysis in adipose tissue
which reduces the supply of fatty acids to the liver, further decreasing VLDL
synthesis. HDL levels are increased because niacin decreases the catabolism of
Apo A
1
. Niacin is useful in treating hypertriglyceridemia as well as hypercho-
lesterolemia especially in the presence of low HDL. The limiting adverse effect
of niacin is cutaneous flushing and itching, and dyspepsia is common at the
doses (1 g/day) necessary to affect lipids. More medically serious adverse
108
CASE FILES: PHARMACOLOGY
effects include hepatotoxicity and hyperglycemia. Niacin induces an insulin-
resistant state causing hyperglycemia. For this reason niacin should not be
used in diabetic patients.
Agents Used for Hypertriglyceridemia—Fibrates
The fibrates include clofibrate, fenofibrate, ciprofibrate, bezafibrate,
and gemfibrozil.These agents predominantly cause a decline plasma triglyc-
eridesand a small decrease in LDL cholesterol. HDL levels are increased. The
fibrates bind to a nuclear receptor peroxisomal proliferator-activator receptor γ
(PPAR-γ) mostly in liver and skeletal muscle. Agonist-bound PPAR-γ induces
lipoprotein lipase (LPL), which increases the lipolysis of triglyceride-rich
VLDL and chylomicrons. Fibrates reduce triglycerides by 35–50 percent and
LDL cholesterol by 10–20 percent. HDL levels are increased by 10–15 percent.
All of the fibrates are orally active, but their absorption is decreased by food.
The major adverse effect is gastrointestinal upset, cutaneous rash, and itching.
Fibrates should not be used in patients with compromised renal function.
COMPREHENSION QUESTIONS
[13.1] Lovastatin reduces plasma cholesterol by which of the following
processes?
A. Inhibiting Apo B
100
biosynthesis
B. Inhibiting cholesterol absorption
C. Inhibiting cholesterol biosynthesis
D. Interfering with bile acid reabsorption
[13.2] Which of the following is a usual effect of niacin?
A. Increases HDL
B. Increases LDL
C. Increases total cholesterol
D. Increases triglycerides
[13.3] A 33-year-old man has been prescribed medication for hyperlipidemia.
He has been noted to have bleeding from his gums and easy bruisability.
His prothrombin time is elevated. Which of the following agents is
most likely to be involved?
A. Atorvastatin
B. Cholestyramine
C. Gemfibrozil
D. Niacin
CLINICAL CASES 109
Answers
[13.1] C. The statins are competitive inhibitors of HMG-CoA reductase and
thereby inhibit de novo cholesterol biosynthesis.
[13.2] A. Niacin increases HDL, decreases total and LDL cholesterol, and
decreases triglycerides.
[13.3] B. Cholestyramine interferes with the absorption of lipid-soluble
vitamins such as vitamin K, leading to decreased levels of vitamin
K–dependent coagulation factors.
PHARMACOLOGY PEARLS
The HMG-CoA reductase inhibitors, the statins, are the initial
choice of drug for the treatment of hypercholesterolemia.
The statins are structural analogs of the substrate HMG-CoA
(3-hydroxy-3-methylglutaryl-coenzyme A) that inhibit the activ-
ity of the enzyme HMG-CoA reductase.
The two major adverse effects associated with statin use are hepato-
toxicity and myopathy.
Bile acid sequestrants impair the absorption of lipid-soluble vita-
mins and many drugs including digoxin, furosemide, thiazides,
coumarin, and some statins.
The fibrates including clofibrate, fenofibrate, ciprofibrate, bezafi-
brate, and gemfibrozil predominantly cause a decline in plasma
triglycerides.
Niacin has effects on all plasma lipids and has side effects of flush-
ing and itching.
REFERENCES
NCEP Report. Implications of recent clinical trials for the National Cholesterol
Education Program Adult Treatment Panel III guidelines. Circulation
2004;110:227–39.
Wierzbicki AS, Mikhailidis DP, Wray R. Drug treatment of combined hyperlipi-
demia. Am J Cardiovasc Drugs 2001;1(5):327–36.
110 CASE FILES: PHARMACOLOGY
CASE 14
A 19-year-old man is brought to the physician’s office by his very concerned
mother. He has been kicked out of the dormitory at college for his “bizarre”
behavior. He has accused several fellow students and professors of spying on
him for the CIA. He stopped attending his classes and spends all of his time
watching TV because the announcers are sending him secret messages on how
to save the world. He has stopped bathing and will only change his clothes
once a week. In your office you find him to be disheveled, quiet, and unemo-
tional. The only spontaneous statement he makes is when he asks why his
mother brought him to the office of “another government spy.” His physical
examination and blood tests are normal. A drug screen is negative. You diag-
nose him with acute psychosis secondary to schizophrenia, admit him to the
psychiatric unit of the hospital, and start him on haloperidol.
What is the mechanism of therapeutic action of haloperidol?
What mediates the extrapyramidal side effects (EPSs) of the
antipsychotic agents?
Which autonomic nervous system receptors are antagonized by
antipsychotic agents?
112 CASE FILES: PHARMACOLOGY
ANSWERS TO CASE 14: ANTIPSYCHOTIC DRUGS
Summary:A 19-year-old man with acute psychosis from schizophrenia is pre-
scribed haloperidol.
Mechanism of therapeutic action of haloperidol:Antagonist activity at
postjunctional dopamine D
2
-receptors in the mesolimbic and mesocortical
areas of the brain.
Mechanism of EPSs:Antagonist activity at dopamine receptors in the
basal ganglia and other dopamine receptor sites in the central nervous
system (CNS).
Autonomic nervous system receptors blocked by antipsychotic
agents: α-Adrenoceptors and muscarinic cholinoreceptors.
CLINICAL CORRELATION
Schizophrenia is a chronic thought disorder that often presents in adolescence
or early adulthood. It is characterized by the presence of “positive symptoms,”
which include delusions, hallucinations, and paranoia, and “negative symp-
toms,” which include blunt affect, withdrawal, and apathy. The therapeutic
effects of the antipsychotic agents result from their antagonist actions on
postjunctional dopamine D
2
receptors in the mesolimbic and mesocortical
areas of the brain, although their benefits may also be related to their antago-
nist activity at dopamine receptors in other areas of the CNS; additionally,
atypical antipsychotic agents have efficacy at serotonin receptors. The
dopamine receptor antagonist activity of antipsychotic agents at multiple sites
in the CNS, and their antagonist activity at various other receptors in the CNS
and throughout the body, contributes to the presence of numerous adverse
effects. The presence of so many, and frequently severe, side effects makes
patient compliance with long-term antipsychotic therapy an important clinical
issue. However, newer, “atypical” agents are now available with greater speci-
ficity for the receptors that mediate antipsychotic actions than for the receptors
that mediate adverse effects.
APPROACH TO PHARMACOLOGY
OF ANTIPSYCHOTIC DRUGS
Objectives
1. List the classes and specific drugs that have antipsychotic activity.
2. Describe the mechanism of therapeutic action of antipsychotic agents.
3. Describe the common side effects of antipsychotic agents and indicate
the receptors that mediate them.
112
CASE FILES: PHARMACOLOGY
CLINICAL CASES 113
Definitions
Acute dystonia: Sustained painful muscle spasms producing twisting
abnormal posture usually occurring shortly after taking an antipsychotic
medication.
Akathisia: Characterized by feelings of intense muscle restlessness or
strong desire to move about, usually during the first 2 weeks of treatment
with an antipsychotic medication.
Parkinson syndrome: Characterized by flat affect, shuffling gait, joint
rigidity, and tremor that occurs weeks to months after treatment.
Neuroleptic malignant syndrome: Characterized by the acute onset of
hyperthermia, muscle rigidity, tremor, tachycardia, mental status changes,
diaphoresis, labile blood pressure, and exposure to a neuroleptic. This
syndrome is associated with a significant mortality rate and usually
occurs within the first few weeks of therapy.
DISCUSSION
Class
Antipsychotic drugscan be classified according to chemical structure as phe-
nothiazines, butyrophenones, and an important group with diverse atypical
structures. The phenothiazines are further subdivided according to side-chain
constituents: aliphatic, piperidine, and piperazine (Table 14-1).
Although very similar in their therapeutic efficacy, the “low- (oral-)
potency” aliphatic and piperidine phenothiazines have a somewhat different
Table 14-1
REPRESENTATIVE ANTIPSYCHOTIC DRUGS (SIDE CHAINS)
Phenothiazines
Chlorpromazine, triflupromazine (aliphatic)
Thioridazine, mesoridazine (piperidine)
Fluphenazine, trifluoperazine (piperazine)
Butyrophenone
Haloperidol
Atypical
Clozapine
Risperidone
Olanzapine
Quetiapine
Aripiprazole
Ziprasidone
adverse effect profile than the “high-potency” agents that include the piper-
azine phenothiazines, and also thiothixene, and haloperidol.
The newer, atypical agents have generally unique structures; some studies
have suggested that they may have greater therapeutic efficacy with regard to
the negative symptoms of schizophrenia. They also have been documented to
have superior adverse effect profiles. Recent clinical trials have called into
question the safety of several of the newer agents. In summary, individual
patient response to antipsychotic agents varies widely and often dictates drug
selection.
Administration of the low-potency antipsychotic agents is more likely to
result in autonomic adverse effects that include orthostatic hypotension
caused by
a
-adrenoceptor blockade, and dry mouth, urinary retention,
and tachycardia resulting from blockade of muscarinic cholinoreceptors.
Their blockade of histamine H
1
receptors in the CNS results in sedation.
The still widely used high-potency agents, for example, haloperidol,are more
likely to result in adverse neurologic effects. Among these are the EPSs, acute
dystonia, akathisia, and Parkinson syndrome, which occur relatively early
in therapy and are thought to be primarily mediated by blockade of dopamine
D
2
receptors in the nigrostriatal dopamine pathway of the basal ganglia. A
late-occurring tardive dyskinesiathat is often irreversible and that may be a
result of the slow development of dopamine receptor supersensitivity also in
the basal ganglia is more or less likely to occur with all antipsychotic agents
except clozapine. A potentially fatal neuroleptic malignant syndrome is
another serious adverse effect of antipsychotic agents in sensitive patients
(1%).Also, hyperprolactinemia in women may occur as a result of enhanced
prolactin release from the posterior pituitary, because of antipsychotic drug
blockade (phenothiazines, butyrophenones, risperidone) of dopamine D
2
receptors of the tuberoinfundibular dopaminergic pathway, which may lead to
amenorrhea, galactorrhea, gynecomastia, decreased libido, and impotence.
Weight gain is also a likely effect of many of these antipsychotic agents.
Theatypical agents are less likely than the conventional agents to result in
adverse EPSs. However, weight gain (clozapine, olanzapine, quetiapine),
hypotension, and sedation are not uncommon events. Seizures (2–5%) and
agranulocytosis (2% risk, 10% fatality) limit the use of clozapine to patients
unresponsive to other agents.
Mechanism of Action
All clinically useful antipsychotic drugs block postjunctional dopamine D
2
recep-
tors,although the degree of blockade among the drugs varies greatly in relation to
their action on other neuroreceptors, particularly serotonin 5-hydroxytryptamine
2A (5-HT
2A
) receptors and certain other dopamine receptor subtypes.
Antipsychotic drugs appear to exert their therapeutic effect, at least in part, by inhi-
bition of dopamine’s action in the mesocortical and mesolimbic dopaminer-
gic pathways of the CNS.
114 CASE FILES: PHARMACOLOGY114 CASE FILES: PHARMACOLOGY
CLINICAL CASES 115
Administration
All antipsychotic agents can be administered by either the oral or parenteral
route or both. Fluphenazine decanoate and haloperidol decanoate are available
as parenteral depot preparations.
Pharmacokinetics
Most antipsychotic agents are readily but incompletely absorbed. They are
highly lipid soluble and have longer clinical duration of action than would be
expected from their plasma half-life, probably as a consequence of their dep-
osition in fat tissue.
Thioridazine, which is metabolized to mesoridazine, is the exception to the
rule that hepatic metabolism of the antipsychotic agents results in less active
metabolites.
Concurrent use of certain antipsychotic agents with other drugs that also
block cholinoreceptors may result in additive peripheral and CNS dysfunction.
COMPREHENSION QUESTIONS
[14.1] Haloperidol-induced Parkinson syndrome is a result of haloperidol’s
action in which of the following tracts?
A. Mesocortical tract
B. Mesolimbic tract
C. Nigrostriatal tract
D. Tuberoinfundibular tract
[14.2] The therapeutic effect of haloperidol is mediated, at least in part, by its
blockade of which of the following receptors?
A. α-Adrenoceptors
B. Dopamine D
2
receptors
C. Histamine H
1
receptors
D. Muscarinic receptors
[14.3] Compared to the low-potency phenothiazine antipsychotic agents,
haloperidol is more like to cause which of the following adverse effects?
A. Akathisia
B. Orthostatic hypotension
C. Sedation
D. Urinary retention
Answers
[14.1] C. Haloperidol-induced Parkinson syndrome is a result of inhibition
of dopamine D
2
receptors in the nigrostriatal tract of the CNS.
[14.2] B. Antipsychotic drugs like haloperidol exert their therapeutic effect,
at least in part, by inhibition of dopamine’s action at dopamine D
2
116 CASE FILES: PHARMACOLOGY
receptors in the mesocortical and mesolimbic dopaminergic path-
ways of the CNS. A number of adverse effects of these drugs are
caused by inhibition of dopamine action in the nigrostriatal and
tuberoinfundibular dopaminergic pathways of the CNS; blockade of
histamine, muscarinic, cholinergic, and α-adrenergic receptors in the
CNS and the peripheral nervous system are also contributory.
[14.3] A. Haloperidol is most likely to cause dystonia, akathisia, and
Parkinson syndrome, whereas the low-potency phenothiazines are
more likely to cause autonomic adverse effects that include orthosta-
tic hypotension, sedation, and urinary retention.
116 CASE FILES: PHARMACOLOGY
PHARMACOLOGY PEARLS
The low-potency antipsychotic agents are more likely to result in
autonomic adverse effects that include orthostatic hypotension as
a consequence of α-adrenoceptor blockade, dry mouth, urinary
retention, and tachycardia resulting from blockade of muscarinic
cholinoreceptors, and sedation (histamine H
1
-receptor blockade).
High-potency agents, for example, haloperidol, are more likely to
result in EPSs, acute dystonia, akathisia, and Parkinson syn-
drome, mediated by blockade of dopamine D
2
receptors in the
nigrostriatal pathway of the basal ganglia.
A late-occurring tardive dyskinesia is often irreversible and is a seri-
ous effect of many antipsychotic agents.
A potentially fatal neuroleptic malignant syndrome is another seri-
ous adverse effect of antipsychotic agents in sensitive patients.
Hyperprolactinemia may occur as a result of enhanced prolactin
release from the posterior pituitary, as a result of antipsychotic
drug blockade of dopamine D
2
receptors in the tuberoinfundibu-
lar tract.
Agranulocytosis may occur in patients treated with clozapine.
REFERENCES
Ananth J, Burgoyne KS, Gadasalli R, et al. How do atypical antipsychotics work? J
Psychiatry Neurosci 2001;26(5):385–94.
Freedman R. Schizophrenia. N Engl J Med 2003;349(13):1738–49.
Lieberman JA, Stroup TS, McEvoy JP, et al. Effectiveness of antipsychotic drugs in
patients with chronic schizophrenia. N Engl J Med 2005;353:1209–23.
Thacker GK, Carpenter WT. Advances in schizophrenia. Nature Med 2001;7(6):
667–71.