Section I. Basic Principles
Chapter 1. Introduction
Definitions
Pharmacology can be defined as the study of substances that interact with living systems through
chemical processes, especially by binding to regulatory molecules and activating or inhibiting
normal body processes. These substances may be chemicals administered to achieve a beneficial
therapeutic effect on some process within the patient or for their toxic effects on regulatory
processes in parasites infecting the patient. Such deliberate therapeutic applications may be
considered the proper role of medical pharmacology, which is often defined as the science of
substances used to prevent, diagnose, and treat disease. Toxicology is that branch of pharmacology
which deals with the undesirable effects of chemicals on living systems, from individual cells to
complex ecosystems.
History of Pharmacology
Prehistoric people undoubtedly recognized the beneficial or toxic effects of many plant and animal
materials. The earliest written records from China and from Egypt list remedies of many types,
including a few still recognized today as useful drugs. Most, however, were worthless or actually
harmful. In the 2500 years or so preceding the modern era there were sporadic attempts to introduce
rational methods into medicine, but none were successful owing to the dominance of systems of
thought that purported to explain all of biology and disease without the need for experimentation
and observation. These schools promulgated bizarre notions such as the idea that disease was
caused by excesses of bile or blood in the body, that wounds could be healed by applying a salve to
the weapon that caused the wound, and so on.
Around the end of the 17th century, reliance on observation and experimentation began to replace
theorizing in medicine, following the example of the physical sciences. As the value of these
methods in the study of disease became clear, physicians in Great Britain and on the Continent
began to apply them to the effects of traditional drugs used in their own practices. Thus, materia
medica—the science of drug preparation and the medical use of drugs—began to develop as the
precursor to pharmacology. However, any understanding of the mechanisms of action of drugs was
prevented by the absence of methods for purifying active agents from the crude materials that were
available and—even more—by the lack of methods for testing hypotheses about the nature of drug
actions.
In the late 18th and early 19th centuries, François Magendie and later his student Claude Bernard
began to develop the methods of experimental animal physiology and pharmacology. Advances in
chemistry and the further development of physiology in the 18th, 19th, and early 20th centuries laid
the foundation needed for understanding how drugs work at the organ and tissue levels.
Paradoxically, real advances in basic pharmacology during this time were accompanied by an
outburst of unscientific promotion by manufacturers and marketers of worthless "patent medicines."
It was not until the concepts of rational therapeutics, especially that of the controlled clinical trial,
were reintroduced into medicine—about 50 years ago—that it became possible to accurately
evaluate therapeutic claims.
About 50 years ago, there also began a major expansion of research efforts in all areas of biology.
As new concepts and new techniques were introduced, information accumulated about drug action
and the biologic substrate of that action, the receptor. During the last half-century, many
fundamentally new drug groups and new members of old groups were introduced. The last 3
decades have seen an even more rapid growth of information and understanding of the molecular
basis for drug action. The molecular mechanisms of action of many drugs have now been identified,
and numerous receptors have been isolated, structurally characterized, and cloned. In fact, the use of
receptor identification methods (described in Chapter 2: Drug Receptors & Pharmacodynamics) has
led to the discovery of many orphan receptors—receptors for which no ligand has been discovered
and whose function can only be surmised. Studies of the local molecular environment of receptors
have shown that receptors and effectors do not function in isolation—they are strongly influenced
by companion regulatory proteins. Decoding of the genomes of many species—from bacteria to
humans—has led to the recognition of unsuspected relationships between receptor families.
Pharmacogenomics—the relation of the individual's genetic makeup to his or her response to
specific drugs—is close to becoming a practical area of therapy (see Pharmacology & Genetics).
Much of that progress is summarized in this resource.
The extension of scientific principles into everyday therapeutics is still going on, though the
medication-consuming public, unfortunately, is still exposed to vast amounts of inaccurate,
incomplete, or unscientific information regarding the pharmacologic effects of chemicals. This has
resulted in the faddish use of innumerable expensive, ineffective, and sometimes harmful remedies
and the growth of a huge "alternative health care" industry. Conversely, lack of understanding of
basic scientific principles in biology and statistics and the absence of critical thinking about public
health issues has led to rejection of medical science by a segment of the public and a common
tendency to assume that all adverse drug effects are the result of malpractice. Two general
principles that the student should always remember are, first, that all substances can under certain
circumstances be toxic; and second, that all therapies promoted as health-enhancing should meet the
same standards of evidence of efficacy and safety, ie, there should be no artificial separation
between scientific medicine and "alternative" or "complementary" medicine.
Pharmacology & Genetics
During the last 5 years, the genomes of humans, mice, and many other organisms have been
decoded in considerable detail. This has opened the door to a remarkable range of new approaches
to research and treatment. It has been known for centuries that certain diseases are inherited, and we
now understand that individuals with such diseases have a heritable abnormality in their DNA. It is
now possible in the case of some inherited diseases to define exactly which DNA base pairs are
anomalous and in which chromosome they appear. In a small number of animal models of such
diseases, it has been possible to correct the abnormality by "gene therapy," ie, insertion of an
appropriate "healthy" gene into somatic cells. Human somatic cell gene therapy has been attempted,
but the technical difficulties are great.
Studies of a newly discovered receptor or endogenous ligand are often confounded by incomplete
knowledge of the exact role of that receptor or ligand. One of the most powerful of the new genetic
techniques is the ability to breed animals (usually mice) in which the gene for the receptor or its
endogenous ligand has been "knocked out," ie, mutated so that the gene product is absent or
nonfunctional. Homozygous "knockout" mice will usually have complete suppression of that
function, while heterozygous animals will usually have partial suppression. Observation of the
behavior, biochemistry, and physiology of the knockout mice will often define the role of the
missing gene product very clearly. When the products of a particular gene are so essential that even
heterozygotes do not survive to birth, it is sometimes possible to breed "knockdown" versions with
only limited suppression of function. Conversely, "knockin" mice have been bred that overexpress
certain receptors of interest.
Some patients respond to certain drugs with greater than usual sensitivity. (Such variations are
discussed in Chapter 4: Drug Biotransformation.) It is now clear that such increased sensitivity is
often due to a very small genetic modification that results in decreased activity of a particular
enzyme responsible for eliminating that drug. Pharmacogenomics (or pharmacogenetics) is the
study of the genetic variations that cause individual differences in drug response. Future clinicians
may screen every patient for a variety of such differences before prescribing a drug.
The Nature of Drugs
In the most general sense, a drug may be defined as any substance that brings about a change in
biologic function through its chemical actions. In the great majority of cases, the drug molecule
interacts with a specific molecule in the biologic system that plays a regulatory role. This molecule
is called a receptor. The nature of receptors is discussed more fully in Chapter 2: Drug Receptors &
Pharmacodynamics. In a very small number of cases, drugs known as chemical antagonists may
interact directly with other drugs, while a few other drugs (eg, osmotic agents) interact almost
exclusively with water molecules. Drugs may be synthesized within the body (eg, hormones) or
may be chemicals not synthesized in the body, ie, xenobiotics (from Gr xenos "stranger"). Poisons
are drugs. Toxins are usually defined as poisons of biologic origin, ie, synthesized by plants or
animals, in contrast to inorganic poisons such as lead and arsenic.
In order to interact chemically with its receptor, a drug molecule must have the appropriate size,
electrical charge, shape, and atomic composition. Furthermore, a drug is often administered at a
location distant from its intended site of action, eg, a pill given orally to relieve a headache.
Therefore, a useful drug must have the necessary properties to be transported from its site of
administration to its site of action. Finally, a practical drug should be inactivated or excreted from
the body at a reasonable rate so that its actions will be of appropriate duration.
The Physical Nature of Drugs
Drugs may be solid at room temperature (eg, aspirin, atropine), liquid (eg, nicotine, ethanol), or
gaseous (eg, nitrous oxide). These factors often determine the best route of administration. For
example, some liquid drugs are easily vaporized and can be inhaled in that form, eg, halothane,
amyl nitrite. The most common routes of administration are listed in Table 3–3. The various classes
of organic compounds—carbohydrates, proteins, lipids, and their constituents—are all represented
in pharmacology. Many drugs are weak acids or bases. This fact has important implications for the
way they are handled by the body, because pH differences in the various compartments of the body
may alter the degree of ionization of such drugs (see below).
Drug Size
The molecular size of drugs varies from very small (lithium ion, MW 7) to very large (eg, alteplase
[t-PA], a protein of MW 59,050). However, the vast majority of drugs have molecular weights
between 100 and 1000. The lower limit of this narrow range is probably set by the requirements for
specificity of action. In order to have a good "fit" to only one type of receptor, a drug molecule
must be sufficiently unique in shape, charge, etc, to prevent its binding to other receptors. To
achieve such selective binding, it appears that a molecule should in most cases be at least 100 MW
units in size. The upper limit in molecular weight is determined primarily by the requirement that
drugs be able to move within the body (eg, from site of administration to site of action). Drugs
much larger than MW 1000 will not diffuse readily between compartments of the body (see
Permeation, below). Therefore, very large drugs (usually proteins) must be administered directly
into the compartment where they have their effect. In the case of alteplase, a clot-dissolving
enzyme, the drug is administered directly into the vascular compartment by intravenous infusion.
Drug Reactivity and Drug-Receptor Bonds
Drugs interact with receptors by means of chemical forces or bonds. These are of three major types:
covalent, electrostatic, and hydrophobic. Covalent bonds are very strong and in many cases not
reversible under biologic conditions. Thus, the covalent bond formed between the activated form of
phenoxybenzamine and the receptor for norepinephrine (which results in blockade of the receptor)
is not readily broken. The blocking effect of phenoxybenzamine lasts long after the free drug has
disappeared from the bloodstream and is reversed only by the synthesis of new receptors, a
process that takes about 48 hours. Other examples of highly reactive, covalent bond-forming drugs
are the DNA-alkylating agents used in cancer chemotherapy to disrupt cell division in the neoplastic
tissue.
Electrostatic bonding is much more common than covalent bonding in drug-receptor interactions.
Electrostatic bonds vary from relatively strong linkages between permanently charged ionic
molecules to weaker hydrogen bonds and very weak induced dipole interactions such as van der
Waals forces and similar phenomena. Electrostatic bonds are weaker than covalent bonds.
Hydrophobic bonds are usually quite weak and are probably important in the interactions of highly
lipid-soluble drugs with the lipids of cell membranes and perhaps in the interaction of drugs with
the internal walls of receptor "pockets."
The specific nature of a particular drug-receptor bond is of less practical importance than the fact
that drugs which bind through weak bonds to their receptors are generally more selective than drugs
which bind through very strong bonds. This is because weak bonds require a very precise fit of the
drug to its receptor if an interaction is to occur. Only a few receptor types are likely to provide such
a precise fit for a particular drug structure. Thus, if we wished to design a highly selective short-
acting drug for a particular receptor, we would avoid highly reactive molecules that form covalent
bonds and instead choose molecules that form weaker bonds.
A few substances that are almost completely inert in the chemical sense nevertheless have
significant pharmacologic effects. For example, xenon, an "inert gas," has anesthetic effects at
elevated pressures.
Drug Shape
The shape of a drug molecule must be such as to permit binding to its receptor site. Optimally, the
drug's shape is complementary to that of the receptor site in the same way that a key is
complementary to a lock. Furthermore, the phenomenon of chirality (stereoisomerism) is so
common in biology that more than half of all useful drugs are chiral molecules, ie, they exist as
enantiomeric pairs. Drugs with two asymmetric centers have four diastereomers, eg, ephedrine, a
sympathomimetic drug. In the great majority of cases, one of these enantiomers will be much more
potent than its mirror image enantiomer, reflecting a better fit to the receptor molecule. For
example, the (S)(+) enantiomer of methacholine, a parasympathomimetic drug, is over 250 times
more potent than the (R)(–) enantiomer. If one imagines the receptor site to be like a glove into
which the drug molecule must fit to bring about its effect, it is clear why a "left-oriented" drug will
be more effective in binding to a left-hand receptor than will its "right-oriented" enantiomer.
The more active enantiomer at one type of receptor site may not be more active at another type, eg,
a receptor type that may be responsible for some unwanted effect. For example, carvedilol, a drug
that interacts with adrenoceptors, has a single chiral center and thus two enantiomers (Table 1–1).
One of these enantiomers, the (S)(–) isomer, is a potent -receptor blocker. The (R)(+) isomer is
100-fold weaker at the receptor. However, the isomers are approximately equipotent as -receptor
blockers. Ketamine is an intravenous anesthetic. The (+) enantiomer is a more potent anesthetic and
is less toxic than the (–) enantiomer. Unfortunately, the drug is still used as the racemic mixture.
Table 1–1. Dissociation Constants (K
d
) of the Enantiomers and Racemate of Carvedilol.
1
Form of
Carvedilol
Inverse of Affinity for Receptors
(K
d
, nmol/L)
Inverse of Affinity for Receptors
(K
d
, nmol/L)
R(+) enantiomer 14 45
S(–) enantiomer 16 0.4
R,S(+/–)
enantiomers
11 0.9
Note: The K
d
is the concentration for 50% saturation of the receptors and is inversely proportionate
to the affinity of the drug for the receptors.
1
Data from Ruffolo RR et al: The pharmacology of carvedilol. Eur J Pharmacol 1990;38:S82.
Finally, because enzymes are usually stereoselective, one drug enantiomer is often more susceptible
than the other to drug-metabolizing enzymes. As a result, the duration of action of one enantiomer
may be quite different from that of the other.
Unfortunately, most studies of clinical efficacy and drug elimination in humans have been carried
out with racemic mixtures of drugs rather than with the separate enantiomers. At present, only about
45% of the chiral drugs used clinically are marketed as the active isomer—the rest are available
only as racemic mixtures. As a result, many patients are receiving drug doses of which 50% or more
is either inactive or actively toxic. However, there is increasing interest—at both the scientific and
the regulatory levels—in making more chiral drugs available as their active enantiomers.
Rational Drug Design
Rational design of drugs implies the ability to predict the appropriate molecular structure of a drug
on the basis of information about its biologic receptor. Until recently, no receptor was known in
sufficient detail to permit such drug design. Instead, drugs were developed through random testing
of chemicals or modification of drugs already known to have some effect (see Chapter 5: Basic &
Clinical Evaluation of New Drugs). However, during the past 3 decades, many receptors have been
isolated and characterized. A few drugs now in use were developed through molecular design based
on a knowledge of the three-dimensional structure of the receptor site. Computer programs are now
available that can iteratively optimize drug structures to fit known receptors. As more becomes
known about receptor structure, rational drug design will become more feasible.
Receptor Nomenclature
The spectacular success of newer, more efficient ways to identify and characterize receptors (see
Chapter 2: Drug Receptors & Pharmacodynamics, How Are New Receptors Discovered?) has
resulted in a variety of differing systems for naming them. This in turn has led to a number of
suggestions regarding more rational methods of naming them. The interested reader is referred for
details to the efforts of the International Union of Pharmacology (IUPHAR) Committee on Receptor
Nomenclature and Drug Classification (reported in various issues of Pharmacological Reviews)
and to the annual Receptor and Ion Channel Nomenclature Supplements published as special issues
by the journal Trends in Pharmacological Sciences (TIPS). The chapters in this book mainly use
these sources for naming receptors.
Drug-Body Interactions
The interactions between a drug and the body are conveniently divided into two classes. The actions
of the drug on the body are termed pharmacodynamic processes; the principles of
pharmacodynamics are presented in greater detail in Chapter 2: Drug Receptors &
Pharmacodynamics. These properties determine the group in which the drug is classified and often
play the major role in deciding whether that group is appropriate therapy for a particular symptom
or disease. The actions of the body on the drug are called pharmacokinetic processes and are
described in Chapters 3 and 4. Pharmacokinetic processes govern the absorption, distribution, and
elimination of drugs and are of great practical importance in the choice and administration of a
particular drug for a particular patient, eg, one with impaired renal function. The following
paragraphs provide a brief introduction to pharmacodynamics and pharmacokinetics.
Pharmacodynamic Principles
As noted above, most drugs must bind to a receptor to bring about an effect. However, at the
molecular level, drug binding is only the first in what is often a complex sequence of steps.
Types of Drug-Receptor Interactions
Agonist drugs bind to and activate the receptor in some fashion, which directly or indirectly brings
about the effect. Some receptors incorporate effector machinery in the same molecule, so that drug
binding brings about the effect directly, eg, opening of an ion channel or activation of enzyme
activity. Other receptors are linked through one or more intervening coupling molecules to a
separate effector molecule. The several types of drug-receptor-effector coupling systems are
discussed in Chapter 2: Drug Receptors & Pharmacodynamics. Pharmacologic antagonist drugs, by
binding to a receptor, prevent binding by other molecules. For example, acetylcholine receptor
blockers such as atropine are antagonists because they prevent access of acetylcholine and similar
agonist drugs to the acetylcholine receptor and they stabilize the receptor in its inactive state. These
agents reduce the effects of acetylcholine and similar drugs in the body.
"Agonists" That Inhibit Their Binding Molecules and Partial Agonists
Some drugs mimic agonist drugs by inhibiting the molecules responsible for terminating the action
of an endogenous agonist. For example, acetylcholinesterase inhibitors, by slowing the destruction
of endogenous acetylcholine, cause cholinomimetic effects that closely resemble the actions of
cholinoceptor agonist molecules even though cholinesterase inhibitors do not—or only incidentally
do—bind to cholinoceptors (see Chapter 7: Cholinoceptor-Activating & Cholinesterase-Inhibiting
Drugs). Other drugs bind to receptors and activate them but do not evoke as great a response as so-
called full agonists. Thus, pindolol, a adrenoceptor "partial agonist," may act as either an agonist
(if no full agonist is present) or as an antagonist (if a full agonist such as isoproterenol is present).
(See Chapter 2: Drug Receptors & Pharmacodynamics.)
Duration of Drug Action
Termination of drug action at the receptor level results from one of several processes. In some
cases, the effect lasts only as long as the drug occupies the receptor, so that dissociation of drug
from the receptor automatically terminates the effect. In many cases, however, the action may
persist after the drug has dissociated, because, for example, some coupling molecule is still present
in activated form. In the case of drugs that bind covalently to the receptor, the effect may persist
until the drug-receptor complex is destroyed and new receptors are synthesized, as described
previously for phenoxybenzamine. Finally, many receptor-effector systems incorporate
desensitization mechanisms for preventing excessive activation when agonist molecules continue to
be present for long periods. See Chapter 2: Drug Receptors & Pharmacodynamics for additional
details.
Receptors and Inert Binding Sites
To function as a receptor, an endogenous molecule must first be selective in choosing ligands (drug
molecules) to bind; and second, it must change its function upon binding in such a way that the
function of the biologic system (cell, tissue, etc) is altered. The first characteristic is required to
avoid constant activation of the receptor by promiscuous binding of many different ligands. The
second characteristic is clearly necessary if the ligand is to cause a pharmacologic effect. The body
contains many molecules that are capable of binding drugs, however, and not all of these
endogenous molecules are regulatory molecules. Binding of a drug to a nonregulatory molecule
such as plasma albumin will result in no detectable change in the function of the biologic system, so
this endogenous molecule can be called an inert binding site. Such binding is not completely
without significance, however, since it affects the distribution of drug within the body and will
determine the amount of free drug in the circulation. Both of these factors are of pharmacokinetic
importance (see below and Chapter 3: Pharmacokinetics & Pharmacodynamics: Rational Dosing &
the Time Course of Drug Action).
Pharmacokinetic Principles
In practical therapeutics, a drug should be able to reach its intended site of action after
administration by some convenient route. In some cases, a chemical that is readily absorbed and
distributed is administered and then converted to the active drug by biologic processes—inside the
body. Such a chemical is called a prodrug. In only a few situations is it possible to directly apply a
drug to its target tissue, eg, by topical application of an anti-inflammatory agent to inflamed skin or
mucous membrane. Most often, a drug is administered into one body compartment, eg, the gut, and
must move to its site of action in another compartment, eg, the brain. This requires that the drug be
absorbed into the blood from its site of administration and distributed to its site of action,
permeating through the various barriers that separate these compartments. For a drug given orally
to produce an effect in the central nervous system, these barriers include the tissues that comprise
the wall of the intestine, the walls of the capillaries that perfuse the gut, and the "blood-brain
barrier," the walls of the capillaries that perfuse the brain. Finally, after bringing about its effect, a
drug should be eliminated at a reasonable rate by metabolic inactivation, by excretion from the
body, or by a combination of these processes.
Permeation
Drug permeation proceeds by four primary mechanisms. Passive diffusion in an aqueous or lipid
medium is common, but active processes play a role in the movement of many drugs, especially
those whose molecules are too large to diffuse readily.
Aqueous Diffusion
Aqueous diffusion occurs within the larger aqueous compartments of the body (interstitial space,
cytosol, etc) and across epithelial membrane tight junctions and the endothelial lining of blood
vessels through aqueous pores that—in some tissues—permit the passage of molecules as large as
MW 20,000–30,000.
*
*
The capillaries of the brain, the testes, and some other tissues are characterized by absence of the
pores that permit aqueous diffusion of many drug molecules into the tissue. They may also contain
high concentrations of drug export pumps (MDR pumps; see text). These tissues are therefore
"protected" or "sanctuary" sites from many circulating drugs.
Aqueous diffusion of drug molecules is usually driven by the concentration gradient of the
permeating drug, a downhill movement described by Fick's law (see below). Drug molecules that
are bound to large plasma proteins (eg, albumin) will not permeate these aqueous pores. If the drug
is charged, its flux is also influenced by electrical fields (eg, the membrane potential and—in parts
of the nephron—the transtubular potential).
Lipid Diffusion
Lipid diffusion is the most important limiting factor for drug permeation because of the large
number of lipid barriers that separate the compartments of the body. Because these lipid barriers
separate aqueous compartments, the lipid:aqueous partition coefficient of a drug determines how
readily the molecule moves between aqueous and lipid media. In the case of weak acids and weak
bases (which gain or lose electrical charge-bearing protons, depending on the pH), the ability to
move from aqueous to lipid or vice versa varies with the pH of the medium, because charged
molecules attract water molecules. The ratio of lipid-soluble form to water-soluble form for a weak
acid or weak base is expressed by the Henderson-Hasselbalch equation (see below).
Special Carriers
Special carrier molecules exist for certain substances that are important for cell function and too
large or too insoluble in lipid to diffuse passively through membranes, eg, peptides, amino acids,
glucose. These carriers bring about movement by active transport or facilitated diffusion and, unlike
passive diffusion, are saturable and inhibitable. Because many drugs are or resemble such naturally
occurring peptides, amino acids, or sugars, they can use these carriers to cross membranes.
Many cells also contain less selective membrane carriers that are specialized for expelling foreign
molecules, eg, the P-glycoprotein or multidrug-resistance type 1 (MDR1) transporter found in
the brain, testes, and other tissues, and in some drug-resistant neoplastic cells. A similar transport
molecule, the multidrug resistance-associated protein-type 2 (MRP2) transporter, plays an
important role in excretion of some drugs or their metabolites into urine and bile.
Endocytosis and Exocytosis
A few substances are so large or impermeant that they can enter cells only by endocytosis, the
process by which the substance is engulfed by the cell membrane and carried into the cell by
pinching off of the newly formed vesicle inside the membrane. The substance can then be released
inside the cytosol by breakdown of the vesicle membrane. This process is responsible for the
transport of vitamin B
12
, complexed with a binding protein (intrinsic factor), across the wall of the
gut into the blood. Similarly, iron is transported into hemoglobin-synthesizing red blood cell
precursors in association with the protein transferrin. Specific receptors for the transport proteins
must be present for this process to work. The reverse process (exocytosis) is responsible for the
secretion of many substances from cells. For example, many neurotransmitter substances are stored
in membrane-bound vesicles in nerve endings to protect them from metabolic destruction in the
cytoplasm. Appropriate activation of the nerve ending causes fusion of the storage vesicle with the
cell membrane and expulsion of its contents into the extracellular space (see Chapter 6: Introduction
to Autonomic Pharmacology).
Fick's Law of Diffusion
The passive flux of molecules down a concentration gradient is given by Fick's law:
where C
1
is the higher concentration, C
2
is the lower concentration, area is the area across which
diffusion is occurring, permeability coefficient is a measure of the mobility of the drug molecules in
the medium of the diffusion path, and thickness is the thickness (length) of the diffusion path. In the
case of lipid diffusion, the lipid:aqueous partition coefficient is a major determinant of mobility of
the drug, since it determines how readily the drug enters the lipid membrane from the aqueous
medium.
Ionization of Weak Acids and Weak Bases; the Henderson-Hasselbalch Equation
The electrostatic charge of an ionized molecule attracts water dipoles and results in a polar,
relatively water-soluble and lipid-insoluble complex. Since lipid diffusion depends on relatively
high lipid solubility, ionization of drugs may markedly reduce their ability to permeate membranes.
A very large fraction of the drugs in use are weak acids or weak bases (Table 1–2). For drugs, a
weak acid is best defined as a neutral molecule that can reversibly dissociate into an anion (a
negatively charged molecule) and a proton (a hydrogen ion). For example, aspirin dissociates as
follows:
Table 1–2. Ionization Constants of Some Common Drugs.
Drug pK
a
1
Weak acids
Acetaminophen 9.5
Acetazolamide 7.2
Ampicillin 2.5
Aspirin 3.5
Chlorothiazide 6.8, 9.4
2
Chlorpropamide 5.0
Ciprofloxacin 6.09, 8.74
2
Cromolyn 2.0
Ethacrynic acid 2.5
Furosemide 3.9
Ibuprofen 4.4, 5.2
2
Levodopa 2.3
Methotrexate 4.8
Methyldopa 2.2, 9.2
2
Penicillamine 1.8
Pentobarbital 8.1
Phenobarbital 7.4
Phenytoin 8.3
Propylthiouracil 8.3
Salicylic acid 3.0
Sulfadiazine 6.5
Sulfapyridine 8.4
Theophylline 8.8
Tolbutamide 5.3
Warfarin 5.0
Weak bases
Albuterol (salbutamol) 9.3
Allopurinol 9.4, 12.3
Alprenolol 9.6
Amiloride 8.7
Amiodarone 6.56
Amphetamine 9.8
Atropine 9.7
Bupivacaine 8.1
Chlordiazepoxide 4.6
Chloroquine 10.8, 8.4
2
Chlorpheniramine 9.2
Chlorpromazine 9.3
Clonidine 8.3
Cocaine 8.5
Codeine 8.2
Cyclizine 8.2
Desipramine 10.2
Diazepam 3
Dihydrocodeine 3
Diphenhydramine 8.8
Diphenoxylate 7.1
Ephedrine 9.6
Epinephrine 8.7
Ergotamine 6.3
Fluphenazine 8.0, 3.9
2
Guanethidine 11.4, 8.3
2
Hydralazine 7.1
Imipramine 9.5
Isoproterenol 8.6
Kanamycin 7.2
Lidocaine 7.9
Metaraminol 8.6
Methadone 8.4
Methamphetamine 10.0
Methyldopa 10.6
Metoprolol 9.8
Morphine 7.9
Nicotine 7.9, 3.1
2
Norepinephrine 8.6
Pentazocine 7.9
Phenylephrine 9.8
Physostigmine 7.9, 1.8
2
Pilocarpine 6.9, 1.4
2
Pindolol 8.6
Procainamide 9.2
Procaine 9.0
Promazine 9.4
Promethazine 9.1
Propranolol 9.4
Pseudoephedrine 9.8
Pyrimethamine 7.0
Quinidine 8.5, 4.4
2
Scopolamine 8.1
Strychnine 8.0, 2.3
2
Terbutaline 10.1
Thioridazine 9.5
Tolazoline 10.6
A drug that is a weak base can be defined as a neutral molecule that can form a cation (a positively
charged molecule) by combining with a proton. For example, pyrimethamine, an antimalarial drug,
undergoes the following association-dissociation process:
Note that the protonated form of a weak acid is the neutral, more lipid-soluble form, whereas the
unprotonated form of a weak base is the neutral form. The law of mass action requires that these
reactions move to the left in an acid environment (low pH, excess protons available) and to the right
in an alkaline environment. The Henderson-Hasselbalch equation relates the ratio of protonated to
unprotonated weak acid or weak base to the molecule's pK
a
and the pH of the medium as follows:
This equation applies to both acidic and basic drugs. Inspection confirms that the lower the pH
relative to the pK
a
, the greater will be the fraction of drug in the protonated form. Because the
uncharged form is the more lipid-soluble, more of a weak acid will be in the lipid-soluble form at
acid pH, while more of a basic drug will be in the lipid-soluble form at alkaline pH.
An application of this principle is in the manipulation of drug excretion by the kidney. Almost all
drugs are filtered at the glomerulus. If a drug is in a lipid-soluble form during its passage down the
renal tubule, a significant fraction will be reabsorbed by simple passive diffusion. If the goal is to
accelerate excretion of the drug, it is important to prevent its reabsorption from the tubule. This can
often be accomplished by adjusting urine pH to make certain that most of the drug is in the ionized
state, as shown in Figure 1–1. As a result of this pH partitioning effect, the drug will be "trapped" in
the urine. Thus, weak acids are usually excreted faster in alkaline urine; weak bases are usually
excreted faster in acidic urine. Other body fluids in which pH differences from blood pH may cause
trapping or reabsorption are the contents of the stomach and small intestine; breast milk; aqueous
humor; and vaginal and prostatic secretions (Table 1–3).
Figure 1–1.
Trapping of a weak base (pyrimethamine) in the urine when the urine is more acidic than the
blood. In the hypothetical case illustrated, the diffusible uncharged form of the drug has
equilibrated across the membrane but the total concentration (charged plus uncharged) in the urine
is almost eight times higher than in the blood.
Table 1–3. Body Fluids with Potential for Drug "Trapping" Through the pH-Partitioning
Phenomenon.
Body Fluid Range
of pH
Total Fluid: Blood
Concentration Ratios for
Sulfadiazine (acid, pK
a
6.5)
1
Total Fluid: Blood Concentration
Ratios for Pyrimethamine (base,
pK
a
7.0)
1
Urine 5.0–8.0 0.12–4.65 72.24–0.79
Breast milk 6.4–7.6
2
0.2–1.77 3.56–0.89
Jejunum,
ileum
contents
7.5–8.0
3
1.23–3.54 0.94–0.79
Stomach
contents
1.92–
2.59
2
0.11
4
85,993–18,386
Prostatic
secretions
6.45–
7.4
2
0.21 3.25–1.0
Vaginal
secretions
3.4–4.2
3
0.11
4
2848–452
1
Body fluid protonated-to-unprotonated drug ratios were calculated using each of the pH extremes
cited; a blood pH of 7.4 was used for blood:drug ratio. For example, the steady-state urine:blood
ratio for sulfadiazine is 0.12 at a urine pH of 5.0; this ratio is 4.65 at a urine pH of 8.0. Thus,
sulfadiazine is much more effectively trapped and excreted in alkaline urine.
2
Lentner C (editor): Geigy Scientific Tables, vol 1, 8th ed. Ciba Geigy, 1981.
3
Bowman WC, Rand MJ: Textbook of Pharmacology, 2nd ed. Blackwell, 1980.
4
Insignificant change in ratios over the physiologic pH range.
As suggested by Table 1–2, a large number of drugs are weak bases. Most of these bases are amine-
containing molecules. The nitrogen of a neutral amine has three atoms associated with it plus a pair
of unshared electrons—see the display below. The three atoms may consist of one carbon
(designated "R") and two hydrogens (a primary amine), two carbons and one hydrogen (a
secondary amine), or three carbon atoms (a tertiary amine). Each of these three forms may
reversibly bind a proton with the unshared electrons. Some drugs have a fourth carbon-nitrogen
bond; these are quaternary amines. However, the quaternary amine is permanently charged and has
no unshared electrons with which to reversibly bind a proton. Therefore, primary, secondary, and
tertiary amines may undergo reversible protonation and vary their lipid solubility with pH, but
quaternary amines are always in the poorly lipid-soluble charged form.
Drug Groups
To learn each pertinent fact about each of the many hundreds of drugs mentioned in this book
would be an impractical goal and, fortunately, is in any case unnecessary. Almost all of the several
thousand drugs currently available can be arranged in about 70 groups. Many of the drugs within
each group are very similar in pharmacodynamic actions and often in their pharmacokinetic
properties as well. For most groups, one or more prototype drugs can be identified that typify the
most important characteristics of the group. This permits classification of other important drugs in
the group as variants of the prototype, so that only the prototype must be learned in detail and, for
the remaining drugs, only the differences from the prototype.
Sources of Information
Students who wish to review the field of pharmacology in preparation for an examination are
referred to Pharmacology: Examination and Board Review, by Trevor, Katzung, and Masters
(McGraw-Hill, 2002) or USMLE Road Map: Pharmacology, by Katzung and Trevor (McGraw-Hill,
2003).
The references at the end of each chapter in this book were selected to provide information specific
to those chapters.
Specific questions relating to basic or clinical research are best answered by resort to the general
pharmacology and clinical specialty serials. For the student and the physician, three periodicals can
be recommended as especially useful sources of current information about drugs: The New England
Journal of Medicine, which publishes much original drug-related clinical research as well as
frequent reviews of topics in pharmacology; The Medical Letter on Drugs and Therapeutics, which
publishes brief critical reviews of new and old therapies, mostly pharmacologic; and Drugs, which
publishes extensive reviews of drugs and drug groups.
Other sources of information pertinent to the USA should be mentioned as well. The "package
insert" is a summary of information the manufacturer is required to place in the prescription sales
package; Physicians' Desk Reference (PDR) is a compendium of package inserts published annually
with supplements twice a year; Facts and Comparisons is a more complete loose-leaf drug
information service with monthly updates; and the USP DI (vol 1, Drug Information for the Health
Care Professional) is a large drug compendium with monthly updates that is now published on the
Internet by the Micromedex Corporation. The package insert consists of a brief description of the
pharmacology of the product. While this brochure contains much practical information, it is also
used as a means of shifting liability for untoward drug reactions from the manufacturer onto the
practitioner. Therefore, the manufacturer typically lists every toxic effect ever reported, no matter
how rare. A useful and objective handbook that presents information on drug toxicity and
interactions is Drug Interactions. Finally, the FDA has an Internet World Wide Web site that carries
news regarding recent drug approvals, withdrawals, warnings, etc. It can be reached using a
personal computer equipped with Internet browser software at http://www.fda.gov.
The following addresses are provided for the convenience of readers wishing to obtain any of the
publications mentioned above:
Drug Interactions
Lea & Febiger
600 Washington Square
Philadelphia, PA 19106
Facts and Comparisons
111 West Port Plaza, Suite 300
St. Louis, MO 63146
Pharmacology: Examination & Board Review, 6th ed
McGraw-Hill Companies, Inc
2 Penn Plaza 12th Floor
New York, NY 10121-2298
USMLE Road Map: Pharmacology
McGraw-Hill Companies, Inc
2 Penn Plaza 12th Floor
New York, NY 10121-2298
The Medical Letter on Drugs and Therapeutics
56 Harrison Street
New Rochelle, NY 10801
The New England Journal of Medicine
10 Shattuck Street
Boston, MA 02115
Physicians' Desk Reference
Box 2017
Mahopac, NY 10541
United States Pharmacopeia Dispensing Information
Micromedex, Inc.
6200 S. Syracuse Way, Suite 300
Englewood, CO 80111
Chapter 2. Drug Receptors & Pharmacodynamics
Drug Receptors & Pharmacodynamics: Introduction
Therapeutic and toxic effects of drugs result from their interactions with molecules in the patient.
Most drugs act by associating with specific macromolecules in ways that alter the macromolecules'
biochemical or biophysical activities. This idea, more than a century old, is embodied in the term
receptor: the component of a cell or organism that interacts with a drug and initiates the chain of
biochemical events leading to the drug's observed effects.
Receptors have become the central focus of investigation of drug effects and their mechanisms of
action (pharmacodynamics). The receptor concept, extended to endocrinology, immunology, and
molecular biology, has proved essential for explaining many aspects of biologic regulation. Many
drug receptors have been isolated and characterized in detail, thus opening the way to precise
understanding of the molecular basis of drug action.
The receptor concept has important practical consequences for the development of drugs and for
arriving at therapeutic decisions in clinical practice. These consequences form the basis for
understanding the actions and clinical uses of drugs described in almost every chapter of this book.
They may be briefly summarized as follows:
(1) Receptors largely determine the quantitative relations between dose or concentration of
drug and pharmacologic effects. The receptor's affinity for binding a drug determines the
concentration of drug required to form a significant number of drug-receptor complexes, and the
total number of receptors may limit the maximal effect a drug may produce.
(2) Receptors are responsible for selectivity of drug action. The molecular size, shape, and
electrical charge of a drug determine whether—and with what affinity—it will bind to a particular
receptor among the vast array of chemically different binding sites available in a cell, tissue, or
patient. Accordingly, changes in the chemical structure of a drug can dramatically increase or
decrease a new drug's affinities for different classes of receptors, with resulting alterations in
therapeutic and toxic effects.
(3) Receptors mediate the actions of both pharmacologic agonists and antagonists. Some drugs
and many natural ligands, such as hormones and neurotransmitters, regulate the function of receptor
macromolecules as agonists; ie, they activate the receptor to signal as a direct result of binding to it.
Other drugs function as pharmacologic antagonists; ie, they bind to receptors but do not activate
generation of a signal; consequently, they interfere with the ability of an agonist to activate the
receptor. Thus, the effect of a so-called "pure" antagonist on a cell or in a patient depends entirely
on its preventing the binding of agonist molecules and blocking their biologic actions. Some of the
most useful drugs in clinical medicine are pharmacologic antagonists.
Macromolecular Nature of Drug Receptors
Most receptors are proteins, presumably because the structures of polypeptides provide both the
necessary diversity and the necessary specificity of shape and electrical charge. The section How
Are New Receptors Discovered? describes some of the methods by which receptors are discovered
and defined.
The best-characterized drug receptors are regulatory proteins, which mediate the actions of
endogenous chemical signals such as neurotransmitters, autacoids, and hormones. This class of
receptors mediates the effects of many of the most useful therapeutic agents. The molecular
structures and biochemical mechanisms of these regulatory receptors are described in a later section
entitled Signaling Mechanisms & Drug Action.
Other classes of proteins that have been clearly identified as drug receptors include enzymes, which
may be inhibited (or, less commonly, activated) by binding a drug (eg, dihydrofolate reductase, the
receptor for the antineoplastic drug methotrexate); transport proteins (eg, Na
+
/K
+
ATPase, the
membrane receptor for cardioactive digitalis glycosides); and structural pro-teins (eg, tubulin, the
receptor for colchicine, an anti-inflammatory agent).
This chapter deals with three aspects of drug receptor function, presented in increasing order of
complexity: (1) Receptors as determinants of the quantitative relation between the concentration of
a drug and the pharmacologic response. (2) Receptors as regulatory proteins and components of
chemical signaling mechanisms that provide targets for important drugs. (3) Receptors as key
determinants of the therapeutic and toxic effects of drugs in patients.
How Are New Receptors Discovered?
Because today's new receptor sets the stage for tomorrow's new drug, it is important to know how
new receptors are discovered. Receptor discovery often begins by studying the relations between
structures and activities of a group of drugs on some conveniently measured response. Binding of
radioactive ligands defines the molar abundance and binding affinities of the putative receptor and
provides an assay to aid in its biochemical purification.
Analysis of the pure receptor protein identifies the number of its subunits, its size, and (sometimes)
provides a clue to how it works (eg, agonist-stimulated autophosphorylation on tyrosine residues,
seen with receptors for insulin and many growth factors). These classic steps in receptor
identification serve as a warming-up exercise for molecular cloning of the segment of DNA that
encodes the receptor. Receptors within a specific class or subclass generally contain highly
conserved regions of similar or identical amino acid (and therefore DNA) sequence. This has led to
an entirely different approach toward identifying receptors by sequence homology to already known
(cloned) receptors.
Cloning of new receptors by sequence homology has identified a number of subtypes of known
receptor classes, such as -adrenoceptors and serotonin receptors, the diversity of which was only
partially anticipated from pharmacologic studies. This approach has also led to the identification of
receptors whose existence was not anticipated from pharmacologic studies. These putative
receptors, identified only by their similarity to other known receptors, are termed orphan receptors
until their native ligands are identified. Identifying such receptors and their ligands is of great
interest because this process may elucidate entirely new signaling pathways and therapeutic targets.
Relation between Drug Concentration & Response
The relation between dose of a drug and the clinically observed response may be complex. In
carefully controlled in vitro systems, however, the relation between concentration of a drug and its
effect is often simple and can be described with mathematical precision. This idealized relation
underlies the more complex relations between dose and effect that occur when drugs are given to
patients.
Concentration-Effect Curves & Receptor Binding of Agonists
Even in intact animals or patients, responses to low doses of a drug usually increase in direct
proportion to dose. As doses increase, however, the response increment diminishes; finally, doses
may be reached at which no further increase in response can be achieved. In idealized or in vitro
systems, the relation between drug concentration and effect is described by a hyperbolic curve
(Figure 2–1 A) according to the following equation:
where E is the effect observed at concentration C, E
max
is the maximal response that can be
produced by the drug, and EC
50
is the concentration of drug that produces 50% of maximal effect.
Figure 2–1.
Relations between drug concentration and drug effect (panel A) or receptor-bound drug (panel B).
The drug concentrations at which effect or receptor occupancy is half-maximal are denoted EC
50
and K
D
, respectively.
This hyperbolic relation resembles the mass action law, which predicts association between two
molecules of a given affinity. This resemblance suggests that drug agonists act by binding to
("occupying") a distinct class of biologic molecules with a characteristic affinity for the drug
receptor. Radioactive receptor ligands have been used to confirm this occupancy assumption in
many drug-receptor systems. In these systems, drug bound to receptors (B) relates to the
concentration of free (unbound) drug (C) as depicted in Figure 2–1 B and as described by an
analogous equation:
in which B
max
indicates the total concentration of receptor sites (ie, sites bound to the drug at
infinitely high concentrations of free drug). K
D
(the equilibrium dissociation constant) represents
the concentration of free drug at which half-maximal binding is observed. This constant
characterizes the receptor's affinity for binding the drug in a reciprocal fashion: If the K
D
is low,
binding affinity is high, and vice versa. The EC
50
and K
D
may be identical, but need not be, as
discussed below. Dose-response data is often presented as a plot of the drug effect (ordinate) against
the logarithm of the dose or concentration (abscissa). This mathematical maneuver transforms the
hyperbolic curve of Figure 2–1 into a sigmoid curve with a linear midportion (eg, Figure 2–2). This
expands the scale of the concentration axis at low concentrations (where the effect is changing
rapidly) and compresses it at high concentrations (where the effect is changing slowly), but has no
special biologic or pharmacologic significance.
Figure 2–2.
Logarithmic transformation of the dose axis and experimental demonstration of spare receptors,
using different concentrations of an irreversible antagonist. Curve A shows agonist response in the
absence of antagonist. After treatment with a low concentration of antagonist (curve B), the curve
is shifted to the right; maximal responsiveness is preserved, however, because the remaining
available receptors are still in excess of the number required. In curve C, produced after treatment
with a larger concentration of antagonist, the available receptors are no longer "spare"; instead,
they are just sufficient to mediate an undiminished maximal response. Still higher concentrations
of antagonist (curves D and E) reduce the number of available receptors to the point that maximal
response is diminished. The apparent EC
50
of the agonist in curves D and E may approximate the
K
D
that characterizes the binding affinity of the agonist for the receptor.
Receptor-Effector Coupling & Spare Receptors
When a receptor is occupied by an agonist, the resulting conformational change is only the first of
many steps usually required to produce a pharmacologic response. The transduction process
between occupancy of receptors and drug response is often termed coupling. The relative efficiency
of occupancy-response coupling is partially determined by the initial conformational change in the
receptor—thus, the effects of full agonists can be considered more efficiently coupled to receptor
occupancy than can the effects of partial agonists, as described below. Coupling efficiency is also
determined by the biochemical events that transduce receptor occupancy into cellular response.
High efficiency of receptor-effector interaction may also be envisioned as the result of spare
receptors. Receptors are said to be "spare" for a given pharmacologic response when the maximal
response can be elicited by an agonist at a concentration that does not result in occupancy of the full
complement of available receptors. Spare receptors are not qualitatively different from nonspare
receptors. They are not hidden or unavailable, and when they are occupied they can be coupled to
response. Experimentally, spare receptors may be demonstrated by using irreversible antagonists to
prevent binding of agonist to a proportion of available receptors and showing that high
concentrations of agonist can still produce an undiminished maximal response (Figure 2–2). Thus, a
maximal inotropic response of heart muscle to catecholamines can be elicited even under conditions
where 90% of the -adrenoceptors are occupied by a quasi-irreversible antagonist. Accordingly,
myocardial cells are said to contain a large proportion of spare -adrenoceptors.
How can we account for the phenomenon of spare receptors? In a few cases, the biochemical
mechanism is understood, such as for drugs that act on some regulatory receptors. In this situation,
the effect of receptor activation—eg, binding of guanosine triphosphate (GTP) by an intermediate—
may greatly outlast the agonist-receptor interaction (see the following section on G Proteins &
Second Messengers). In such a case, the "spareness" of receptors is temporal in that the response
initiated by an individual ligand-receptor binding event persists longer than the binding event itself.
In other cases, where the biochemical mechanism is not understood, we imagine that the receptors
might be spare in number. If the concentration or amount of a cellular component other than the
receptor limits the coupling of receptor occupancy to response, then a maximal response can occur
without occupancy of all receptors. This concept helps explain how the sensitivity of a cell or tissue
to a particular concentration of agonist depends not only on the affinity of the receptor for binding
the agonist (characterized by the K
D
) but also on the degree of spareness—the total number of
receptors present compared to the number actually needed to elicit a maximal biologic response.
The K
D
of the agonist-receptor interaction determines what fraction (B/B
max
) of total receptors will
be occupied at a given free concentration (C) of agonist regardless of the receptor concentration:
Imagine a responding cell with four receptors and four effectors. Here the number of effectors does
not limit the maximal response, and the receptors are not spare in number. Consequently, an agonist
present at a concentration equal to the K
D
will occupy 50% of the receptors, and half of the
effectors will be activated, producing a half-maximal response (ie, two receptors stimulate two
effectors). Now imagine that the number of receptors increases 10-fold to 40 receptors but that the
total number of effectors remains constant. Most of the receptors are now spare in number. As a
result, a much lower concentration of agonist suffices to occupy two of the 40 receptors (5% of the
receptors), and this same low concentration of agonist is able to elicit a half-maximal response (two
of four effectors activated). Thus, it is possible to change the sensitivity of tissues with spare
receptors by changing the receptor concentration.
Competitive & Irreversible Antagonists
Receptor antagonists bind to receptors but do not activate them. In general, the effects of these
antagonists result from preventing agonists (other drugs or endogenous regulatory molecules) from
binding to and activating receptors. Such antagonists are divided into two classes depending on
whether or not they reversibly compete with agonists for binding to receptors.
In the presence of a fixed concentration of agonist, increasing concentrations of a competitive
antagonist progressively inhibit the agonist response; high antagonist concentrations prevent
response completely. Conversely, sufficiently high concentrations of agonist can completely
surmount the effect of a given concentration of the antagonist; ie, the E
max
for the agonist remains
the same for any fixed concentration of antagonist (Figure 2–3 A). Because the antagonism is
competitive, the presence of antagonist increases the agonist concentration required for a given
degree of response, and so the agonist concentration-effect curve is shifted to the right.
Figure 2–3.
Changes in agonist concentration-effect curves produced by a competitive antagonist (panel A) or
by an irreversible antagonist (panel B). In the presence of a competitive antagonist, higher
concentrations of agonist are required to produce a given effect; thus the agonist concentration (C')
required for a given effect in the presence of concentration [I] of an antagonist is shifted to the
right, as shown. High agonist concentrations can overcome inhibition by a competitive antagonist.
This is not the case with an irreversible antagonist, which reduces the maximal effect the agonist
can achieve, although it may not change its EC
50
.
The concentration (C') of an agonist required to produce a given effect in the presence of a fixed
concentration ([I]) of competitive antagonist is greater than the agonist concentration (C) required
to produce the same effect in the absence of the antagonist. The ratio of these two agonist
concentrations (the "dose ratio") is related to the dissociation constant (K
I
) of the antagonist by the
Schild equation:
Pharmacologists often use this relation to determine the K
I
of a competitive antagonist. Even
without knowledge of the relationship between agonist occupancy of the receptor and response, the
K
I
can be determined simply and accurately. As shown in Figure 2–3, concentration response
curves are obtained in the presence and in the absence of a fixed concentration of competitive
antagonist; comparison of the agonist concentrations required to produce identical degrees of
pharmacologic effect in the two situations reveals the antagonist's K
I
. If C' is twice C, for example,
then [I] = K
I
.
For the clinician, this mathematical relation has two important therapeutic implications:
(1) The degree of inhibition produced by a competitive antagonist depends on the
concentration of antagonist. Different patients receiving a fixed dose of propranolol, for
example, exhibit a wide range of plasma concentrations, owing to differences in clearance of
the drug. As a result, the effects of a fixed dose of this competitive antagonist of
norepinephrine may vary widely in patients, and the dose must be adjusted accordingly.
(2) Clinical response to a competitive antagonist depends on the concentration of agonist
that is competing for binding to receptors. Here also propranolol provides a useful example:
When this competitive -adrenoceptor antagonist is administered in doses sufficient to block
the effect of basal levels of the neurotransmitter norepinephrine, resting heart rate is
decreased. However, the increase in release of norepinephrine and epinephrine that occurs
with exercise, postural changes, or emotional stress may suffice to overcome competitive
antagonism by propranolol and increase heart rate, and thereby can influence therapeutic
response.
Some receptor antagonists bind to the receptor in an irreversible or nearly irreversible fashion, ie,
not competitive. The antagonist's affinity for the receptor may be so high that for practical purposes,
the receptor is unavailable for binding of agonist. Other antagonists in this class produce
irreversible effects because after binding to the receptor they form covalent bonds with it. After
occupancy of some proportion of receptors by such an antagonist, the number of remaining
unoccupied receptors may be too low for the agonist (even at high concentrations) to elicit a
response comparable to the previous maximal response (Figure 2–3 B). If spare receptors are
present, however, a lower dose of an irreversible antagonist may leave enough receptors unoccupied
to allow achievement of maximum response to agonist, although a higher agonist concentration will
be required (Figures 2–2 B and C; see Receptor-Effector Coupling and Spare Receptors, above).
Therapeutically, irreversible antagonists present distinctive advantages and disadvantages. Once the
irreversible antagonist has occupied the receptor, it need not be present in unbound form to inhibit
agonist responses. Consequently, the duration of action of such an irreversible antagonist is
relatively independent of its own rate of elimination and more dependent on the rate of turnover of
receptor molecules.
Phenoxybenzamine, an irreversible -adrenoceptor antagonist, is used to control the hypertension
caused by catecholamines released from pheochromocytoma, a tumor of the adrenal medulla. If
administration of phenoxybenzamine lowers blood pressure, blockade will be maintained even
when the tumor episodically releases very large amounts of catecholamine. In this case, the ability
to prevent responses to varying and high concentrations of agonist is a therapeutic advantage. If
overdose occurs, however, a real problem may arise. If the -adrenoceptor blockade cannot be
overcome, excess effects of the drug must be antagonized "physiologically," ie, by using a pressor
agent that does not act via receptors.
Partial Agonists
Based on the maximal pharmacologic response that occurs when all receptors are occupied, agonists
can be divided into two classes: partial agonists produce a lower response, at full receptor
occupancy, than do full agonists. Partial agonists produce concentration-effect curves that resemble
those observed with full agonists in the presence of an antagonist that irreversibly blocks some of
the receptor sites (compare Figures 2–2 [curve D] and 2–4 B). It is important to emphasize that the
failure of partial agonists to produce a maximal response is not due to decreased affinity for binding
to receptors. Indeed, a partial agonist's inability to cause a maximal pharmacologic response, even
when present at high concentrations that saturate binding to all receptors, is indicated by the fact
that partial agonists competitively inhibit the responses produced by full agonists (Figure 2–4 C).
Many drugs used clinically as antagonists are in fact weak partial agonists.
Figure 2–4.
Panel A: The percentage of receptor occupancy resulting from full agonist (present at a single
concentration) binding to receptors in the presence of increasing concentrations of a partial
agonist. Because the full agonist (filled squares) and the partial agonist (open squares) compete to
bind to the same receptor sites, when occupancy by the partial agonist increases, binding of the full
agonist decreases. Panel B: When each of the two drugs is used alone and response is measured,
occupancy of all the receptors by the partial agonist produces a lower maximal response than does
similar occupancy by the full agonist. Panel C: Simultaneous treatment with a single concentration
of full agonist and increasing concentrations of the partial agonist produces the response patterns
shown in the bottom panel. The fractional response caused by a single concentration of the full
agonist (filled squares) decreases as increasing concentrations of the partial agonist compete to
bind to the receptor with increasing success; at the same time the portion of the response caused by
the partial agonist (open squares) increases, while the total response—ie, the sum of responses to
the two drugs (filled triangles)—gradually decreases, eventually reaching the value produced by
partial agonist alone (compare panel B).
Other Mechanisms of Drug Antagonism
Not all of the mechanisms of antagonism involve interactions of drugs or endogenous ligands at a
single type of receptor. Indeed, chemical antagonists need not involve a receptor at all. Thus, one
drug may antagonize the actions of a second drug by binding to and inactivating the second drug.
For example, protamine, a protein that is positively charged at physiologic pH, can be used
clinically to counteract the effects of heparin, an anticoagulant that is negatively charged; in this
case, one drug antagonizes the other simply by binding it and making it unavailable for interactions
with proteins involved in formation of a blood clot.
The clinician often uses drugs that take advantage of physiologic antagonism between endogenous
regulatory pathways. For example, several catabolic actions of the glucocorticoid hormones lead to
increased blood sugar, an effect that is physiologically opposed by insulin. Although
glucocorticoids and insulin act on quite distinct receptor-effector systems, the clinician must
sometimes administer insulin to oppose the hyperglycemic effects of glucocorticoid hormone,
whether the latter is elevated by endogenous synthesis (eg, a tumor of the adrenal cortex) or as a
result of glucocorticoid therapy.
In general, use of a drug as a physiologic antagonist produces effects that are less specific and less
easy to control than are the effects of a receptor-specific antagonist. Thus, for example, to treat
bradycardia caused by increased release of acetylcholine from vagus nerve endings, the physician
could use isoproterenol, a -adrenoceptor agonist that increases heart rate by mimicking
sympathetic stimulation of the heart. However, use of this physiologic antagonist would be less
rational—and potentially more dangerous—than would use of a receptor-specific antagonist such as
atropine (a competitive antagonist at the receptors at which acetylcholine slows heart rate).
Signaling Mechanisms & Drug Action
Until now we have considered receptor interactions and drug effects in terms of equations and
concentration-effect curves. We must also understand the molecular mechanisms by which a drug
acts. Such understanding allows us to ask basic questions with important clinical implications:
• Why do some drugs produce effects that persist for minutes, hours, or even days after the
drug is no longer present?
• Why do responses to other drugs diminish rapidly with prolonged or repeated
administration?
• How do cellular mechanisms for amplifying external chemical signals explain the
phenomenon of spare receptors?
• Why do chemically similar drugs often exhibit extraordinary selectivity in their actions?
• Do these mechanisms provide targets for developing new drugs?
Most transmembrane signaling is accomplished by a small number of different molecular
mechanisms. Each type of mechanism has been adapted, through the evolution of distinctive protein
families, to transduce many different signals. These protein families include receptors on the cell
surface and within the cell, as well as enzymes and other components that generate, amplify,
coordinate, and terminate postreceptor signaling by chemical second messengers in the cytoplasm.
This section first discusses the mechanisms for carrying chemical information across the plasma
membrane and then outlines key features of cytoplasmic second messengers.
Five basic mechanisms of transmembrane signaling are well understood (Figure 2–5). Each uses a
different strategy to circumvent the barrier posed by the lipid bilayer of the plasma membrane.
These strategies use (1) a lipid-soluble ligand that crosses the membrane and acts on an intracellular
receptor; (2) a transmembrane receptor protein whose intracellular enzymatic activity is
allosterically regulated by a ligand that binds to a site on the protein's extracellular domain; (3) a
transmembrane receptor that binds and stimulates a protein tyrosine kinase; (4) a ligand-gated
transmembrane ion channel that can be induced to open or close by the binding of a ligand; or (5) a
transmembrane receptor protein that stimulates a GTP-binding signal transducer protein (G protein),
which in turn modulates production of an intracellular second messenger.
Figure 2–5.
Known transmembrane signaling mechanisms: 1: A lipid-soluble chemical signal crosses the
plasma membrane and acts on an intracellular receptor (which may be an enzyme or a regulator of
gene transcription); 2: the signal binds to the extracellular domain of a transmembrane protein,
thereby activating an enzymatic activity of its cytoplasmic domain; 3: the signal binds to the
extracellular domain of a transmembrane receptor bound to a protein tyrosine kinase, which it
activates; 4: the signal binds to and directly regulates the opening of an ion channel; 5: the signal
binds to a cell-surface receptor linked to an effector enzyme by a G protein. (A,C, substrates; B, D,
products; R, receptor; G, G protein; E, effector [enzyme or ion channel]; Y, tyrosine; P,
phosphate.)
While the five established mechanisms do not account for all the chemical signals conveyed across
cell membranes, they do transduce many of the most important signals exploited in
pharmacotherapy.
Intracellular Receptors for Lipid-Soluble Agents
Several biologic signals are sufficiently lipid-soluble to cross the plasma membrane and act on
intracellular receptors. One of these is a gas, nitric oxide (NO), that acts by stimulating an
intracellular enzyme, guanylyl cyclase, which produces cyclic guanosine monophosphate (cGMP).
Signaling via cGMP is described in more detail later in this chapter. Receptors for another class of
ligands—including corticosteroids, mineralocorticoids, sex steroids, vitamin D, and thyroid
hormone—stimulate the transcription of genes in the nucleus by binding to specific DNA sequences
near the gene whose expression is to be regulated. Many of the target DNA sequences (called
response elements) have been identified.
These "gene-active" receptors belong to a protein family that evolved from a common precursor.
Dissection of the receptors by recombinant DNA techniques has provided insights into their
molecular mechanism. For example, binding of glucocorticoid hormone to its normal receptor
protein relieves an inhibitory constraint on the transcription-stimulating activity of the protein.
Figure 2–6 schematically depicts the molecular mechanism of glucocorticoid action: In the absence
of hormone, the receptor is bound to hsp90, a protein that appears to prevent normal folding of
several structural domains of the receptor. Binding of hormone to the ligand-binding domain
triggers release of hsp90. This allows the DNA-binding and transcription-activating domains of the
receptor to fold into their functionally active conformations, so that the activated receptor can
initiate transcription of target genes.
Figure 2–6.
Mechanism of glucocorticoid action. The glucocorticoid receptor polypeptide is schematically
depicted as a protein with three distinct domains. A heat-shock protein, hsp90, binds to the
receptor in the absence of hormone and prevents folding into the active conformation of the
receptor. Binding of a hormone ligand (steroid) causes dissociation of the hsp90 stabilizer and
permits conversion to the active configuration.
The mechanism used by hormones that act by regulating gene expression has two therapeutically
important consequences:
(1) All of these hormones produce their effects after a characteristic lag period of 30 minutes to
several hours—the time required for the synthesis of new proteins. This means that the gene-
active hormones cannot be expected to alter a pathologic state within minutes (eg,
glucocorticoids will not immediately relieve the symptoms of acute bronchial asthma).
(2) The effects of these agents can persist for hours or days after the agonist concentration has
been reduced to zero. The persistence of effect is primarily due to the relatively slow turnover of
most enzymes and proteins, which can remain active in cells for hours or days after they have
been synthesized. Consequently, it means that the beneficial (or toxic) effects of a gene-active
hormone will usually decrease slowly when administration of the hormone is stopped.
Ligand-Regulated Transmembrane Enzymes Including Receptor Tyrosine Kinases
This class of receptor molecules mediates the first steps in signaling by insulin, epidermal growth
factor (EGF), platelet-derived growth factor (PDGF), atrial natriuretic peptide (ANP), transforming
growth factor- (TGF- ), and many other trophic hormones. These receptors are polypeptides
consisting of an extracellular hormone-binding domain and a cytoplasmic enzyme domain, which
may be a protein tyrosine kinase, a serine kinase, or a guanylyl cyclase (Figure 2–7). In all these
receptors, the two domains are connected by a hydrophobic segment of the polypeptide that crosses
the lipid bilayer of the plasma membrane.
Figure 2–7.
Mechanism of activation of the epidermal growth factor (EGF) receptor, a representative receptor
tyrosine kinase. The receptor polypeptide has extracellular and cytoplasmic domains, depicted
above and below the plasma membrane. Upon binding of EGF (circle), the receptor converts from
its inactive monomeric state (left) to an active dimeric state (right), in which two receptor
polypeptides bind noncovalently in the plane of the membrane. The cytoplasmic domains become
phosphorylated (P) on specific tyrosine residues (Y) and their enzymatic activities are activated,
catalyzing phosphorylation of substrate proteins (S).
The receptor tyrosine kinase signaling pathway begins with ligand binding to the receptor's
extracellular domain. The resulting change in receptor conformation causes receptor molecules to
bind to one another, which in turn brings together the tyrosine kinase domains, which become
enzymatically active, and phosphorylate one another as well as additional downstream signaling
proteins. Activated receptors catalyze phosphorylation of tyrosine residues on different target
signaling proteins, thereby allowing a single type of activated receptor to modulate a number of
biochemical processes. Insulin, for example, uses a single class of receptors to trigger increased
uptake of glucose and amino acids and to regulate metabolism of glycogen and triglycerides in the
cell. Similarly, each of the growth factors initiates in its specific target cells a complex program of
cellular events ranging from altered membrane transport of ions and metabolites to changes in the
expression of many genes. At present, a few compounds have been found to produce effects that
may be due to inhibition of tyrosine kinase activities. It is easy to imagine therapeutic uses for
specific inhibitors of growth factor receptors, especially in neoplastic disorders where excessive
growth factor signaling is often observed. For example, a monoclonal antibody (trastuzumab) that
acts as an antagonist of the HER2/neu receptor tyrosine kinase is effective in therapy of human
breast cancers associated with overexpression of this growth factor receptor.
The intensity and duration of action of EGF, PDGF, and other agents that act via receptor tyrosine
kinases are limited by receptor down-regulation. Ligand binding induces accelerated endocytosis of
receptors from the cell surface, followed by the degradation of those receptors (and their bound
ligands). When this process occurs at a rate faster than de novo synthesis of receptors, the total
number of cell-surface receptors is reduced (down-regulated) and the cell's responsiveness to ligand
is correspondingly diminished. A well-understood process by which many tyrosine kinases are
down-regulated is via ligand-induced internalization of receptors followed by trafficking to
lysosomes, where receptors are proteolyzed. EGF causes internalization and subsequent proteolytic
down-regulation after binding to the EGF receptor protein tyrosine kinase; genetic mutations that
interfere with this process of down-regulation cause excessive growth factor–induced cell
proliferation and are associated with an increased susceptibility to certain types of cancer.
Internalization of certain receptor tyrosine kinases, most notably receptors for nerve growth factor,
serves a very different function. Internalized nerve growth factor receptors are not rapidly degraded.
Instead, receptors remain intact and are translocated in endocytic vesicles from the distal axon
(where receptors are activated by nerve growth factor released from the innervated tissue) to the cell
body (where the signal is transduced to transcription factors regulating the expression of genes
controlling cell survival). This process effectively transports a critical survival signal released from
the target tissue over a remarkably long distance—more than 1 meter in certain sensory neurons. A
number of regulators of growth and differentiation, including TGF- , act on another class of
transmembrane receptor enzymes that phosphorylate serine and threonine residues. ANP, an
important regulator of blood volume and vascular tone, acts on a transmembrane receptor whose
intracellular domain, a guanylyl cyclase, generates cGMP (see below). Receptors in both groups,
like the receptor tyrosine kinases, are active in their dimeric forms.
Cytokine Receptors
Cytokine receptors respond to a heterogeneous group of peptide ligands that includes growth
hormone, erythropoietin, several kinds of interferon, and other regulators of growth and
differentiation. These receptors use a mechanism (Figure 2–8) closely resembling that of receptor
tyrosine kinases, except that in this case, the protein tyrosine kinase activity is not intrinsic to the
receptor molecule. Instead, a separate protein tyrosine kinase, from the Janus-kinase (JAK) family,
binds noncovalently to the receptor. As in the case of the EGF-receptor, cytokine receptors dimerize
after they bind the activating ligand, allowing the bound JAKs to become activated and to
phosphorylate tyrosine residues on the receptor. Tyrosine phosphates on the receptor then set in
motion a complex signaling dance by binding another set of proteins, called STATs (signal
transducers and activators of transcription). The bound STATs are themselves phosphorylated by
the JAKs, two STAT molecules dimerize (attaching to one another's tyrosine phosphates), and
finally the STAT/STAT dimer dissociates from the receptor and travels to the nucleus, where it
regulates transcription of specific genes.
Figure 2–8.
Cytokine receptors, like receptor tyrosine kinases, have extracellular and intracellular domains and
form dimers. However, after activation by an appropriate ligand, separate mobile protein tyrosine
kinase molecules (JAK) are activated, resulting in phosphorylation of signal transducers and
activation of transcription (STAT) molecules. STAT dimers then travel to the nucleus, where they
regulate transcription.
Ligand-Gated Channels
Many of the most useful drugs in clinical medicine act by mimicking or blocking the actions of
endogenous ligands that regulate the flow of ions through plasma membrane channels. The natural
ligands include acetylcholine, serotonin, -aminobutyric acid (GABA), and the excitatory amino
acids (eg, glycine, aspartate, and glutamate). All of these agents are synaptic transmitters.
Each of their receptors transmits its signal across the plasma membrane by increasing
transmembrane conductance of the relevant ion and thereby altering the electrical potential across
the membrane. For example, acetylcholine causes the opening of the ion channel in the nicotinic
acetylcholine receptor (AChR), which allows Na
+
to flow down its concentration gradient into cells,
producing a localized excitatory postsynaptic potential—a depolarization.
The AChR (Figure 2–9) is one of the best-characterized of all cell-surface receptors for hormones
or neurotransmitters. One form of this receptor is a pentamer made up of five polypeptide subunits
(eg, two chains plus one , one , and one chain, all with molecular weights ranging from 43,000
to 50,000). These polypeptides, each of which crosses the lipid bilayer four times, form a cylindric
structure 8 nm in diameter. When acetylcholine binds to sites on the subunits, a conformational
change occurs that results in the transient opening of a central aqueous channel through which
sodium ions penetrate from the extracellular fluid into the cell.
Figure 2–9.
The nicotinic acetylcholine receptor, a ligand-gated ion channel. The receptor molecule is depicted
as embedded in a rectangular piece of plasma membrane, with extracellular fluid above and
cytoplasm below. Composed of five subunits (two , one , one , and one ), the receptor opens a
central transmembrane ion channel when acetylcholine (ACh) binds to sites on the extracellular
domain of its subunits.
The time elapsed between the binding of the agonist to a ligand-gated channel and the cellular
response can often be measured in milliseconds. The rapidity of this signaling mechanism is
crucially important for moment-to-moment transfer of information across synapses. Ligand-gated
ion channels can be regulated by multiple mechanisms, including phosphorylation and
internalization. In the central nervous system, these mechanisms contribute to synaptic plasticity
involved in learning and memory.
G Proteins & Second Messengers
Many extracellular ligands act by increasing the intracellular concentrations of second messengers
such as cyclic adenosine-3',5'-monophosphate (cAMP), calcium ion, or the phosphoinositides
(described below). In most cases they use a transmembrane signaling system with three separate
components. First, the extracellular ligand is specifically detected by a cell-surface receptor. The
receptor in turn triggers the activation of a G protein located on the cytoplasmic face of the plasma
membrane. The activated G protein then changes the activity of an effector element, usually an
enzyme or ion channel. This element then changes the concentration of the intracellular second
messenger. For cAMP, the effector enzyme is adenylyl cyclase, a transmembrane protein that
converts intracellular adenosine triphosphate (ATP) to cAMP. The corresponding G protein, G
s
,
stimulates adenylyl cyclase after being activated by hormones and neurotransmitters that act via a
specific receptor (Table 2–1).
Table 2–1. A Partial List of Endogenous Ligands and Their Associated Second Messengers.
Ligand Second Messenger
Adrenocorticotropic hormone cAMP
Acetylcholine (muscarinic receptors) Ca
2+
, phosphoinositides
Angiotensin Ca
2+
, phosphoinositides
Catecholamines (
1
-adrenoceptors)
Ca
2+
, phosphoinositides
Catecholamines ( -adrenoceptors) cAMP
Chorionic gonadotropin cAMP
Follicle-stimulating hormone cAMP
Glucagon cAMP
Histamine (H
2
receptors)
cAMP
Luteinizing hormone cAMP
Melanocyte-stimulating hormone cAMP
Parathyroid hormone cAMP
Platelet-activating factor Ca
2+
, phosphoinositides
Prostacyclin, prostaglandin E2 cAMP
Serotonin (5-HT
4
receptors)
cAMP
Serotonin (5-HT
1C
and 5-HT
2
receptors)
Ca
2+
, phosphoinositides
Thyrotropin cAMP
Thyrotropin-releasing hormone Ca
2+
, phosphoinositides
Vasopressin (V
1
receptors)
Ca
2+
, phosphoinositides
Vasopressin (V
2
receptors) cAMP
Key: cAMP = cyclic adenosine monophosphate.
G
s
and other G proteins use a molecular mechanism that involves binding and hydrolysis of GTP
(Figure 2–10). This mechanism allows the transduced signal to be amplified. For example, a
neurotransmitter such as norepinephrine may encounter its membrane receptor for only a few
milliseconds. When the encounter generates a GTP-bound G
s
molecule, however, the duration of
activation of adenylyl cyclase depends on the longevity of GTP binding to G
s
rather than on the
receptor's affinity for norepinephrine. Indeed, like other G proteins, GTP-bound G
s
may remain
active for tens of seconds, enormously amplifying the original signal. This mechanism explains how
signaling by G proteins produces the phenomenon of spare receptors (described above). At low
concentrations of agonist the proportion of agonist-bound receptors may be much less than the
proportion of G proteins in the active (GTP-bound) state; if the proportion of active G proteins
correlates with pharmacologic response, receptors will appear to be spare (ie, a small fraction of
receptors occupied by agonist at any given time will appear to produce a proportionately larger
response).
Figure 2–10.
The guanine nucleotide-dependent activation-inactivation cycle of G proteins. The agonist
activates the receptor (R), which promotes release of GDP from the G protein (G), allowing entry
of GTP into the nucleotide binding site. In its GTP-bound state (G-GTP), the G protein regulates
activity of an effector enzyme or ion channel (E). The signal is terminated by hydrolysis of GTP,
followed by return of the system to the basal unstimulated state. Open arrows denote regulatory
effects. (P
i
, inorganic phosphate.)
The family of G proteins contains several functionally diverse subfamilies (Table 2–2), each of
which mediates effects of a particular set of receptors to a distinctive group of effectors. Receptors
coupled to G proteins comprise a family of "seven-transmembrane" or "serpentine" receptors, so
called because the receptor polypeptide chain "snakes" across the plasma membrane seven times
(Figure 2–11). Receptors for adrenergic amines, serotonin, acetylcholine (muscarinic but not
nicotinic), many peptide hormones, odorants, and even visual receptors (in retinal rod and cone
cells) all belong to the serpentine family. All were derived from a common evolutionary precursor.
Some serpentine receptors exist as dimers, but it is thought that dimerization is not usually required
for activation.
Table 2–2. G Proteins and Their Receptors and Effectors.
G
Protein
Receptors for: Effector/Signaling Pathway
G
s
-Adrenergic amines, glucagon, histamine,
serotonin, and many other hormones
Adenylyl cyclase cAMP
G
i1
, G
i2
,
G
i3
2
-Adrenergic amines, acetylcholine
(muscarinic), opioids, serotonin, and many others
Several, including:
Adenylyl cyclase cAMP
Open cardiac K
+
channels heart
rate
G
olf
Odorants (olfactory epithelium) Adenylyl cyclase cAMP
G
o
Neurotransmitters in brain (not yet specifically
identified)
Not yet clear
G
q
Acetylcholine (eg, muscarinic), bombesin,
serotonin (5-HT
1C
), and many others
Phospholipase C IP
3
,
diacylglycerol, cytoplasmic Ca
2+
G
t1
, G
t2
Photons (rhodopsin and color opsins in retinal
rod and cone cells)
cGMP phosphodiesterase cGMP
(phototransduction)
Key: cAMP = cyclic adenosine monophosphate; cGMP = cyclic guanosine monophosphate.
Serpentine receptors transduce signals across the plasma membrane in essentially the same way.
Often the agonist ligand—eg, a catecholamine, acetylcholine, or the photon-activated chromophore
of retinal photoreceptors—is bound in a pocket enclosed by the transmembrane regions of the
receptor (as in Figure 2–11). The resulting change in conformation of these regions is transmitted to
cytoplasmic loops of the receptor, which in turn activate the appropriate G protein by promoting
replacement of GDP by GTP, as described above. Considerable biochemical evidence indicates that
G proteins interact with amino acids in the third cytoplasmic loop of the serpentine receptor
polypeptide (shown by arrows in Figure 2–11). The carboxyl terminal tails of these receptors, also
located in the cytoplasm, can regulate the receptors' ability to interact with G proteins, as described
below.
Figure 2–11.
Transmembrane topology of a typical serpentine receptor. The receptor's amino (N) terminal is
extracellular (above the plane of the membrane), and its carboxyl (C) terminal intracellular. The
terminals are connected by a polypeptide chain that traverses the plane of the membrane seven
times. The hydrophobic transmembrane segments (light color) are designated by roman numerals
(I–VII). The agonist (Ag) approaches the receptor from the extracellular fluid and binds to a site
surrounded by the transmembrane regions of the receptor protein. G proteins (G) interact with
cytoplasmic regions of the receptor, especially with portions of the third cytoplasmic loop between
transmembrane regions V and VI. The receptor's cytoplasmic terminal tail contains numerous
serine and threonine residues whose hydroxyl (–OH) groups can be phosphorylated. This
phosphorylation may be associated with diminished receptor-G protein interaction.
Receptor Regulation
Receptor-mediated responses to drugs and hormonal agonists often desensitize with time (Figure 2–
12, top). After reaching an initial high level, the response (eg, cellular cAMP accumulation, Na
+
influx, contractility, etc) gradually diminishes over seconds or minutes, even in the continued
presence of the agonist. This desensitization is usually reversible; a second exposure to agonist, if
provided a few minutes after termination of the first exposure, results in a response similar to the
initial response.
Figure 2–12.
Possible mechanism for desensitization of the -adrenoceptor. The upper part of the figure depicts
the response to a -adrenoceptor agonist (ordinate) versus time (abscissa). The break in the time
axis indicates passage of time in the absence of agonist. Temporal duration of exposure to agonist
is indicated by the light-colored bar. The lower part of the figure schematically depicts agonist-
induced phosphorylation (P) by -adrenoceptor kinase ( -adrenergic receptor kinase, ARK) of
carboxyl terminal hydroxyl groups (–OH) of the -adrenoceptor. This phosphorylation induces
binding of a protein, -arrestin ( -arr), which prevents the receptor from interacting with G
s
.
Removal of agonist for a short period of time allows dissociation of -arr, removal of phosphate
(Pi) from the receptor by phosphatases (P'ase), and restoration of the receptor's normal
responsiveness to agonist.
Although many kinds of receptors undergo desensitization, the mechanism is in many cases
obscure. A molecular mechanism of desensitization has been worked out in some detail, however,
in the case of the -adrenoceptor (Figure 2–12, bottom). The agonist-induced change in
conformation of the receptor causes it to bind, activate, and serve as a substrate for a specific
kinase, -adrenoceptor kinase (also called ARK). ARK then phosphorylates serine or threonine
residues in the receptor's carboxyl terminal tail. The presence of phosphoserines increases the
receptor's affinity for binding a third protein, -arrestin. Binding of -arrestin to cytoplasmic loops
of the receptor diminishes the receptor's ability to interact with G
s
, thereby reducing the agonist
response (ie, stimulation of adenylyl cyclase). Upon removal of agonist, however, cellular
phosphatases remove phosphates from the receptor and ARK stops putting them back on, so that
the receptor—and consequently the agonist response—return to normal. This mechanism of
desensitization, which rapidly and reversibly modulates the ability of the receptor to signal to G
protein, turns out to regulate many G protein–coupled receptors. Another important regulatory
process is down-regulation. Down-regulation, which decreases the actual number of receptors
present in the cell or tissue, occurs more slowly than rapid desensitization and is less readily
reversible. This is because down-regulation involves a net degradation of receptors present in the
cell, requiring new receptor biosynthesis for recovery, in contrast to rapid desensitization which
involves reversible phosphorylation of existing receptors. Many G protein–coupled receptors are
down-regulated by undergoing ligand-induced endocytosis and delivery to lysosomes, similar to
down-regulation of protein tyrosine kinases such as the EGF receptor. Down-regulation generally
occurs only after prolonged or repeated exposure of cells to agonist (over hours to days). Brief
periods of agonist exposure (several minutes) can also induce internalization of receptors. In this
case, many receptors, including the -adrenoceptor, do not down-regulate but instead recycle intact
to the plasma membrane. This rapid cycling through endocytic vesicles promotes dephosphorylation
of receptors, increasing the rate at which fully functional receptors are replenished in the plasma
membrane. Thus, depending on the particular receptor and duration of activation, internalization
can mediate quite different effects on receptor signaling and regulation.
Well-Established Second Messengers
Cyclic Adenosine Monophosphate (cAMP)
Acting as an intracellular second messenger, cAMP mediates such hormonal responses as the
mobilization of stored energy (the breakdown of carbohydrates in liver or triglycerides in fat cells
stimulated by -adrenomimetic catecholamines), conservation of water by the kidney (mediated by
vasopressin), Ca
2+
homeostasis (regulated by parathyroid hormone), and increased rate and
contraction force of heart muscle (
-adrenomimetic catecholamines). It also regulates the
production of adrenal and sex steroids (in response to corticotropin or follicle-stimulating
hormone), relaxation of smooth muscle, and many other endocrine and neural processes.
cAMP exerts most of its effects by stimulating cAMP-dependent protein kinases (Figure 2–13).
These kinases are composed of a cAMP-binding regulatory (R) dimer and two catalytic (C) chains.
When cAMP binds to the R dimer, active C chains are released to diffuse through the cytoplasm
and nucleus, where they transfer phosphate from ATP to appropriate substrate proteins, often
enzymes. The specificity of cAMP's regulatory effects resides in the distinct protein substrates of
the kinases that are expressed in different cells. For example, liver is rich in phosphorylase kinase
and glycogen synthase, enzymes whose reciprocal regulation by cAMP-dependent phosphorylation
governs carbohydrate storage and release.
Figure 2–13.
The cAMP second messenger pathway. Key proteins include hormone receptors (Rec), a
stimulatory G protein (G
s
), catalytic adenylyl cyclase (AC), phosphodiesterases (PDE) that
hydrolyze cAMP, cAMP-dependent kinases, with regulatory (R) and catalytic (C) subunits, protein
substrates (S) of the kinases, and phosphatases (P'ase), which remove phosphates from substrate
proteins. Open arrows denote regulatory effects.
When the hormonal stimulus stops, the intracellular actions of cAMP are terminated by an elaborate
series of enzymes. cAMP-stimulated phosphorylation of enzyme substrates is rapidly reversed by a
diverse group of specific and nonspecific phosphatases. cAMP itself is degraded to 5'-AMP by
several cyclic nucleotide phosphodiesterases (PDE, Figure 2–13). Competitive inhibition of cAMP
degradation is one way caffeine, theophylline, and other methylxanthines produce their effects (see
Chapter 20: Drugs Used in Asthma).
Calcium and Phosphoinositides
Another well-studied second messenger system involves hormonal stimulation of phosphoinositide
hydrolysis (Figure 2–14). Some of the hormones, neurotransmitters, and growth factors that trigger
this pathway (see Table 2–1) bind to receptors linked to G proteins, while others bind to receptor
tyrosine kinases. In all cases, the crucial step is stimulation of a membrane enzyme, phospholipase
C (PLC), which splits a minor phospholipid component of the plasma membrane,
phosphatidylinositol-4,5-bisphosphate (PIP
2
), into two second messengers, diacylglycerol and
inositol-1,4,5-trisphosphate (IP
3
or InsP
3
). Diacylglycerol is confined to the membrane where it
activates a phospholipid- and calcium-sensitive protein kinase called protein kinase C. IP
3
is water-
soluble and diffuses through the cytoplasm to trigger release of Ca
2+
from internal storage vesicles.
Elevated cytoplasmic Ca
2+
concentration promotes the binding of Ca
2+
to the calcium-binding
protein calmodulin, which regulates activities of other enzymes, including calcium-dependent
protein kinases.
Figure 2–14.
The Ca
2+
-phosphoinositide signaling pathway. Key proteins include hormone receptors (R), a G
protein (G), a phosphoinositide-specific phospholipase C (PLC), protein kinase C substrates of the
kinase (S), calmodulin (CaM), and calmodulin-binding enzymes (E), including kinases,
phosphodiesterases, etc. (PIP
2
, phosphatidylinositol-4,5-bisphosphate; DAG, diacylglycerol.
Asterisk denotes activated state. Open arrows denote regulatory effects.)
With its multiple second messengers and protein kinases, the phosphoinositide signaling pathway is
much more complex than the cAMP pathway. For example, different cell types may contain one or
more specialized calcium- and calmodulin-dependent kinases with limited substrate specificity (eg,
myosin light chain kinase) in addition to a general calcium- and calmodulin-dependent kinase that
can phosphorylate a wide variety of protein substrates. Furthermore, at least nine structurally
distinct types of protein kinase C have been identified.
As in the cAMP system, multiple mechanisms damp or terminate signaling by this pathway. IP
3
is
inactivated by dephosphorylation; diacylglycerol is either phosphorylated to yield phosphatidic
acid, which is then converted back into phospholipids, or it is deacylated to yield arachidonic acid;
Ca
2+
is actively removed from the cytoplasm by Ca
2+
pumps.
These and other nonreceptor elements of the calcium-phosphoinositide signaling pathway are now
becoming targets for drug development. For example, the therapeutic effects of lithium ion, an
established agent for treating manic-depressive illness, may be mediated by effects on the
metabolism of phosphoinositides (see Chapter 29: Antipsychotic Agents & Lithium).
Cyclic Guanosine Monophosphate (cGMP)
Unlike cAMP, the ubiquitous and versatile carrier of diverse messages, cGMP has established
signaling roles in only a few cell types. In intestinal mucosa and vascular smooth muscle, the
cGMP-based signal transduction mechanism closely parallels the cAMP-mediated signaling
mechanism. Ligands detected by cell surface receptors stimulate membrane-bound guanylyl cyclase
to produce cGMP, and cGMP acts by stimulating a cGMP-dependent protein kinase. The actions of
cGMP in these cells are terminated by enzymatic degradation of the cyclic nucleotide and by
dephosphorylation of kinase substrates.
Increased cGMP concentration causes relaxation of vascular smooth muscle by a kinase-mediated
mechanism that results in dephosphorylation of myosin light chains (see Figure 12–2). In these
smooth muscle cells, cGMP synthesis can be elevated by two different transmembrane signaling
mechanisms utilizing two different guanylyl cyclases. ANP, a blood-borne peptide hormone,
stimulates a transmembrane receptor by binding to its extracellular domain, thereby activating the
guanylyl cyclase activity that resides in the receptor's intracellular domain. The other mechanism
mediates responses to NO (see Chapter 19: Nitric Oxide, Donors, & Inhibitors), which is generated
in vascular endothelial cells in response to natural vasodilator agents such as acetylcholine and
histamine (NO is also called endothelium-derived relaxing factor [EDRF]). After entering the target
cell, NO binds to and activates a cytoplasmic guanylyl cyclase. A number of useful vasodilating
drugs act by generating or mimicking NO, or by interfering with the metabolic breakdown of cGMP
by phosphodiesterase (see Chapter 11: Antihypertensive Agents and Chapter 12: Vasodilators & the
Treatment of Angina Pectoris).
Interplay among Signaling Mechanisms
The calcium-phosphoinositide and cAMP signaling pathways oppose one another in some cells and
are complementary in others. For example, vasopressor agents that contract smooth muscle act by
IP
3
-mediated mobilization of Ca
2+
, whereas agents that relax smooth muscle often act by elevation
of cAMP. In contrast, cAMP and phosphoinositide second messengers act together to stimulate
glucose release from the liver.
Phosphorylation: A Common Theme
Almost all second messenger signaling involves reversible phosphorylation, which performs two
principal functions in signaling: amplification and flexible regulation. In amplification, rather like
GTP bound to a G protein, the attachment of a phosphoryl group to a serine, threonine, or tyrosine
residue powerfully amplifies the initial regulatory signal by recording a molecular memory that the
pathway has been activated; dephosphorylation erases the memory, taking a longer time to do so
than is required for dissociation of an allosteric ligand. In flexible regulation, differing substrate
specificities of the multiple protein kinases regulated by second messengers provide branch points
in signaling pathways that may be independently regulated. In this way, cAMP, Ca
2+
, or other
second messengers can use the presence or absence of particular kinases or kinase substrates to
produce quite different effects in different cell types. Inhibitors of protein kinases have great
potential as therapeutic agents, particularly in neoplastic diseases. Trastuzumab, an antibody that
antagonizes growth factor receptor signaling, was discussed earlier as a therapeutic agent for breast
cancer. Another example of this general approach is imatinib (Gleevec, STI571), a small molecule
inhibitor of the cytoplasmic tyrosine kinase Bcr/Abl, which is activated by growth factor signaling
pathways and is overexpressed in chronic myelogenous leukemia (CML). This compound, a
promising agent for treating CML, was recently approved by the US Food and Drug Administration
(FDA) for clinical use.
Receptor Classes & Drug Development
The existence of a specific drug receptor is usually inferred from studying the structure-activity
relationship of a group of structurally similar congeners of the drug that mimic or antagonize its
effects. Thus, if a series of related agonists exhibits identical relative potencies in producing two
distinct effects, it is likely that the two effects are mediated by similar or identical receptor
molecules. In addition, if identical receptors mediate both effects, a competitive antagonist will
inhibit both responses with the same K
I
; a second competitive antagonist will inhibit both responses
with its own characteristic K
I
. Thus, studies of the relation between structure and activity of a series
of agonists and antagonists can identify a species of receptor that mediates a set of pharmacologic
responses.
Exactly the same experimental procedure can show that observed effects of a drug are mediated by
different receptors. In this case, effects mediated by different receptors may exhibit different orders
of potency among agonists and different K
I
values for each competitive antagonist.
Wherever we look, evolution has created many different receptors that function to mediate
responses to any individual chemical signal. In some cases, the same chemical acts on completely
different structural receptor classes. For example, acetylcholine uses ligand-gated ion channels
(nicotinic AChRs) to initiate a fast excitatory postsynaptic potential (EPSP) in postganglionic
neurons. Acetylcholine also activates a separate class of G protein–coupled receptors (muscarinic
AChRs), which modulate responsiveness of the same neurons to the fast EPSP. In addition, each
structural class usually includes multiple subtypes of receptor, often with significantly different
signaling or regulatory properties. For example, norepinephrine activates many structurally related
receptors, including -adrenergic (stimulation of G
s
, increased heart rate),
1
-adrenergic
(stimulation of G
q
, vasoconstriction), and
2
-adrenergic (stimulation of G
i
, opening of K
+
channels)
(see Table 2–2). The existence of multiple receptor classes and subtypes for the same endogenous
ligand has created important opportunities for drug development. For example, propranolol, a
selective antagonist of -adrenergic receptors, can reduce an accelerated heart rate without
preventing the sympathetic nervous system from causing vasoconstriction, an effect mediated by
1
receptors.
The principle of drug selectivity may even apply to structurally identical receptors expressed in
different cells, eg, receptors for steroids such as estrogen (Figure 2–6). Different cell types express
different accessory proteins, which interact with steroid receptors and change the functional effects
of drug-receptor interaction. For example, tamoxifen acts as an antagonist on estrogen receptors
expressed in mammary tissue but as an agonist on estrogen receptors in bone. Consequently,
tamoxifen may be useful not only in the treatment and prophylaxis of breast cancer but also in the
prevention of osteoporosis by increasing bone density (see Chapter 40: The Gonadal Hormones &
Inhibitors and Chapter 42: Agents That Affect Bone Mineral Homeostasis). Tamoxifen may also
create complications in postmenopausal women, however, by exerting an agonist action in the
uterus, stimulating endometrial cell proliferation.
New drug development is not confined to agents that act on receptors for extracellular chemical
signals. Pharmaceutical chemists are now determining whether elements of signaling pathways
distal to the receptors may also serve as targets of selective and useful drugs. For example,
clinically useful agents might be developed that act selectively on specific G proteins, kinases,
phosphatases, or the enzymes that degrade second messengers.
Relation between Drug Dose & Clinical Response
We have dealt with receptors as molecules and shown how receptors can quantitatively account for
the relation between dose or concentration of a drug and pharmacologic responses, at least in an
idealized system. When faced with a patient who needs treatment, the prescriber must make a
choice among a variety of possible drugs and devise a dosage regimen that is likely to produce
maximal benefit and minimal toxicity. In order to make rational therapeutic decisions, the
prescriber must understand how drug-receptor interactions underlie the relations between dose and
response in patients, the nature and causes of variation in pharmacologic responsiveness, and the
clinical implications of selectivity of drug action.
Dose & Response in Patients
Graded Dose-Response Relations
To choose among drugs and to determine appropriate doses of a drug, the prescriber must know the
relative pharmacologic potency and maximal efficacy of the drugs in relation to the desired
therapeutic effect. These two important terms, often confusing to students and clinicians, can be
explained by refering to Figure 2–15, which depicts graded dose-response curves that relate dose of
four different drugs to the magnitude of a particular therapeutic effect.
Figure 2–15.
Graded dose-response curves for four drugs, illustrating different pharmacologic potencies and
different maximal efficacies. (See text.)
Potency
Drugs A and B are said to be more potent than drugs C and D because of the relative positions of
their dose-response curves along the dose axis of Figure 2–15. Potency refers to the concentration
(EC
50
) or dose (ED
50
) of a drug required to produce 50% of that drug's maximal effect. Thus, the
pharmacologic potency of drug A in Figure 2–15 is less than that of drug B, a partial agonist,
because the EC
50
of A is greater than the EC
50
of B. Potency of a drug depends in part on the
affinity (K
D
) of receptors for binding the drug and in part on the efficiency with which drug-
receptor interaction is coupled to response. Note that some doses of drug A can produce larger
effects than any dose of drug B, despite the fact that we describe drug B as pharmacologically more
potent. The reason for this is that drug A has a larger maximal efficacy, as described below.
For clinical use, it is important to distinguish between a drug's potency and its efficacy. The clinical
effectiveness of a drug depends not on its potency (EC
50
), but on its maximal efficacy (see below)
and its ability to reach the relevant receptors. This ability can depend on its route of administration,
absorption, distribution through the body, and clearance from the blood or site of action. In deciding
which of two drugs to administer to a patient, the prescriber must usually consider their relative
effectiveness rather than their relative potency. Pharmacologic potency can largely determine the
administered dose of the chosen drug.
For therapeutic purposes, the potency of a drug should be stated in dosage units, usually in terms of
a particular therapeutic end point (eg, 50 mg for mild sedation, 1
g/kg/min for an increase in heart
rate of 25 beats/min). Relative potency, the ratio of equi-effective doses (0.2, 10, etc), may be used
in comparing one drug with another.
Maximal Efficacy
This parameter reflects the limit of the dose-response relation on the response axis. Drugs A, C,
and D in Figure 2–15 have equal maximal efficacy, while all have greater maximal efficacy than
drug B. The maximal efficacy (sometimes referred to simply as efficacy) of a drug is obviously
crucial for making clinical decisions when a large response is needed. It may be determined by the
drug's mode of interactions with receptors (as with partial agonists, described above)
*
or by
characteristics of the receptor-effector system involved.
*
Note that "maximal efficacy," used in a therapeutic context, does not have exactly the same
meaning the term denotes in the more specialized context of drug-receptor interactions described
earlier in this chapter. In an idealized in vitro system, efficacy denotes the relative maximal efficacy
of agonists and partial agonists that act via the same receptor. In therapeutics, efficacy denotes the
extent or degree of an effect that can be achieved in the intact patient. Thus, therapeutic efficacy
may be affected by the characteristics of a particular drug-receptor interaction, but it also depends
on a host of other factors as noted in the text.
Thus, diuretics that act on one portion of the nephron may produce much greater excretion of fluid
and electrolytes than diuretics that act elsewhere. In addition, the practical efficacy of a drug for
achieving a therapeutic end point (eg, increased cardiac contractility) may be limited by the drug's
propensity to cause a toxic effect (eg, fatal cardiac arrhythmia) even if the drug could otherwise
produce a greater therapeutic effect.
Shape of Dose-Response Curves
While the responses depicted in curves A, B, and C of Figure 2–15 approximate the shape of a
simple Michaelis-Menten relation (transformed to a logarithmic plot), some clinical responses do
not. Extremely steep dose-response curves (eg, curve D) may have important clinical consequences
if the upper portion of the curve represents an undesirable extent of response (eg, coma caused by a
sedative-hypnotic). Steep dose-response curves in patients could result from cooperative
interactions of several different actions of a drug (eg, effects on brain, heart, and peripheral vessels,
all contributing to lowering of blood pressure).
Quantal Dose-Effect Curves
Graded dose-response curves of the sort described above have certain limitations in their
application to clinical decision making. For example, such curves may be impossible to construct if
the pharmacologic response is an either-or (quantal) event, such as prevention of convulsions,
arrhythmia, or death. Furthermore, the clinical relevance of a quantitative dose-response
relationship in a single patient, no matter how precisely defined, may be limited in application to
other patients, owing to the great potential variability among patients in severity of disease and
responsiveness to drugs.
Some of these difficulties may be avoided by determining the dose of drug required to produce a
specified magnitude of effect in a large number of individual patients or experimental animals and
plotting the cumulative frequency distribution of responders versus the log dose (Figure 2–16). The
specified quantal effect may be chosen on the basis of clinical relevance (eg, relief of headache) or
for preservation of safety of experimental subjects (eg, using low doses of a cardiac stimulant and
specifying an increase in heart rate of 20 beats/min as the quantal effect), or it may be an inherently
quantal event (eg, death of an experimental animal). For most drugs, the doses required to produce a
specified quantal effect in individuals are lognormally distributed; ie, a frequency distribution of
such responses plotted against the log of the dose produces a gaussian normal curve of variation
(colored area, Figure 2–16). When these responses are summated, the resulting cumulative
frequency distribution constitutes a quantal dose-effect curve (or dose-percent curve) of the
proportion or percentage of individuals who exhibit the effect plotted as a function of log dose
(Figure 2–16).
Figure 2–16.
Quantal dose-effect plots. Shaded boxes (and the accompanying curves) indicate the frequency
distribution of doses of drug required to produce a specified effect; ie, the percentage of animals
that required a particular dose to exhibit the effect. The open boxes (and the corresponding curves)
indicate the cumulative frequency distribution of responses, which are lognormally distributed.
The quantal dose-effect curve is often characterized by stating the median effective dose (ED
50
),
the dose at which 50% of individuals exhibit the specified quantal effect. (Note that the abbreviation
ED
50
has a different meaning in this context from its meaning in relation to graded dose-effect
curves, described above.) Similarly, the dose required to produce a particular toxic effect in 50% of
animals is called the median toxic dose (TD
50
) If the toxic effect is death of the animal, a median
lethal dose (LD
50
) may be experimentally defined. Such values provide a convenient way of
comparing the potencies of drugs in experimental and clinical settings: Thus, if the ED
50
s of two
drugs for producing a specified quantal effect are 5 and 500 mg, respectively, then the first drug can
be said to be 100 times more potent than the second for that particular effect. Similarly, one can
obtain a valuable index of the selectivity of a drug's action by comparing its ED
50
s for two different
quantal effects in a population (eg, cough suppression versus sedation for opioid drugs).
Quantal dose-effect curves may also be used to generate information regarding the margin of safety
to be expected from a particular drug used to produce a specified effect. One measure, which relates
the dose of a drug required to produce a desired effect to that which produces an undesired effect, is
the therapeutic index. In animal studies, the therapeutic index is usually defined as the ratio of the
TD
50
to the ED
50
for some therapeutically relevant effect. The precision possible in animal
experiments may make it useful to use such a therapeutic index to estimate the potential benefit of a
drug in humans. Of course, the therapeutic index of a drug in humans is almost never known with
real precision; instead, drug trials and accumulated clinical experience often reveal a range of
usually effective doses and a different (but sometimes overlapping) range of possibly toxic doses.
The clinically acceptable risk of toxicity depends critically on the severity of the disease being
treated. For example, the dose range that provides relief from an ordinary headache in the great
majority of patients should be very much lower than the dose range that produces serious toxicity,
even if the toxicity occurs in a small minority of patients. However, for treatment of a lethal disease
such as Hodgkin's lymphoma, the acceptable difference between therapeutic and toxic doses may be
smaller.
Finally, note that the quantal dose-effect curve and the graded dose-response curve summarize
somewhat different sets of information, although both appear sigmoid in shape on a
semilogarithmic plot (compare Figures 2–15 and 2–16). Critical information required for making
rational therapeutic decisions can be obtained from each type of curve. Both curves provide
information regarding the potency and selectivity of drugs; the graded dose-response curve
indicates the maximal efficacy of a drug, and the quantal dose-effect curve indicates the potential
variability of responsiveness among individuals.
Variation in Drug Responsiveness
Individuals may vary considerably in their responsiveness to a drug; indeed, a single individual may
respond differently to the same drug at different times during the course of treatment. Occasionally,
individuals exhibit an unusual or idiosyncratic drug response, one that is infrequently observed in
most patients. The idiosyncratic responses are usually caused by genetic differences in metabolism
of the drug or by immunologic mechanisms, including allergic reactions.
Quantitative variations in drug response are in general more common and more clinically important.
An individual patient is hyporeactive or hyperreactive to a drug in that the intensity of effect of a
given dose of drug is diminished or increased in comparison to the effect seen in most individuals.
(Note: The term hypersensitivity usually refers to allergic or other immunologic responses to
drugs.) With some drugs, the intensity of response to a given dose may change during the course of
therapy; in these cases, responsiveness usually decreases as a consequence of continued drug
administration, producing a state of relative tolerance to the drug's effects. When responsiveness
diminishes rapidly after administration of a drug, the response is said to be subject to
tachyphylaxis.
Even before administering the first dose of a drug, the prescriber should consider factors that may
help in predicting the direction and extent of possible variations in responsiveness. These include
the propensity of a particular drug to produce tolerance or tachyphylaxis as well as the effects of
age, sex, body size, disease state, genetic factors, and simultaneous administration of other drugs.
Four general mechanisms may contribute to variation in drug responsiveness among patients or
within an individual patient at different times.
Alteration in Concentration of Drug That Reaches the Receptor
Patients may differ in the rate of absorption of a drug, in distributing it through body compartments,
or in clearing the drug from the blood (see Chapter 3: Pharmacokinetics & Pharmacodynamics:
Rational Dosing & the Time Course of Drug Action). By altering the concentration of drug that
reaches relevant receptors, such pharmacokinetic differences may alter the clinical response. Some
differences can be predicted on the basis of age, weight, sex, disease state, liver and kidney
function, and by testing specifically for genetic differences that may result from inheritance of a
functionally distinctive complement of drug-metabolizing enzymes (see Chapter 3:
Pharmacokinetics & Pharmacodynamics: Rational Dosing & the Time Course of Drug Action and
Chapter 4: Drug Biotransformation).
Variation in Concentration of an Endogenous Receptor Ligand
This mechanism contributes greatly to variability in responses to pharmacologic antagonists. Thus,
propranolol, a -adrenoceptor antagonist, will markedly slow the heart rate of a patient whose
endogenous catecholamines are elevated (as in pheochromocytoma) but will not affect the resting
heart rate of a well-trained marathon runner. A partial agonist may exhibit even more dramatically
different responses: Saralasin, a weak partial agonist at angiotensin II receptors, lowers blood
pressure in patients with hypertension caused by increased angiotensin II production and raises
blood pressure in patients who produce small amounts of angiotensin.
Alterations in Number or Function of Receptors
Experimental studies have documented changes in drug responsiveness caused by increases or
decreases in the number of receptor sites or by alterations in the efficiency of coupling of receptors
to distal effector mechanisms. In some cases, the change in receptor number is caused by other
hormones; for example, thyroid hormones increase both the number of receptors in rat heart
muscle and cardiac sensitivity to catecholamines. Similar changes probably contribute to the
tachycardia of thyrotoxicosis in patients and may account for the usefulness of propranolol, a -
adrenoceptor antagonist, in ameliorating symptoms of this disease.
In other cases, the agonist ligand itself induces a decrease in the number (eg, down-regulation) or
coupling efficiency (eg, desensitization) of its receptors. These mechanisms (discussed above, under
Signaling Mechanisms & Drug Actions) may contribute to two clinically important phenomena:
first, tachyphylaxis or tolerance to the effects of some drugs (eg, biogenic amines and their
congeners), and second, the "overshoot" phenomena that follow withdrawal of certain drugs. These
phenomena can occur with either agonists or antagonists. An antagonist may increase the number of
receptors in a critical cell or tissue by preventing down-regulation caused by an endogenous
agonist. When the antagonist is withdrawn, the elevated number of receptors can produce an
exaggerated response to physiologic concentrations of agonist. Potentially disastrous withdrawal
symptoms can result for the opposite reason when administration of an agonist drug is discontinued.
In this situation, the number of receptors, which has been decreased by drug-induced down-
regulation, is too low for endogenous agonist to produce effective stimulation. For example, the
withdrawal of clonidine (a drug whose
2
-adrenoceptor agonist activity reduces blood pressure) can
produce hypertensive crisis, probably because the drug down-regulates
2
-adrenoceptors (see
Chapter 11: Antihypertensive Agents).
Genetic factors also can play an important role in altering the number or function of specific
receptors. For example, a specific genetic variant of the
2C
-adrenoceptor—when inherited together
with a specific variant of the
1
-adrenoceptor—confers a greatly increased risk for developing
congestive heart failure which may be reduced by early intervention using antagonist drugs. The
identification of such genetic factors, part of the rapidly developing field of pharmacogenetics,
holds exciting promise for clinical diagnosis and may help physicians design the most appropriate
pharmacologic therapy for individual patients.
Changes in Components of Response Distal to the Receptor
Although a drug initiates its actions by binding to receptors, the response observed in a patient
depends on the functional integrity of biochemical processes in the responding cell and physiologic
regulation by interacting organ systems. Clinically, changes in these postreceptor processes
represent the largest and most important class of mechanisms that cause variation in responsiveness
to drug therapy.
Before initiating therapy with a drug, the prescriber should be aware of patient characteristics that
may limit the clinical response. These characteristics include the age and general health of the
patient and—most importantly—the severity and pathophysiologic mechanism of the disease. The
most important potential cause of failure to achieve a satisfactory response is that the diagnosis is
wrong or physiologically incomplete. Drug therapy will always be most successful when it is
accurately directed at the pathophysiologic mechanism responsible for the disease.
When the diagnosis is correct and the drug is appropriate, an unsatisfactory therapeutic response
can often be traced to compensatory mechanisms in the patient that respond to and oppose the
beneficial effects of the drug. Compensatory increases in sympathetic nervous tone and fluid
retention by the kidney, for example, can contribute to tolerance to antihypertensive effects of a
vasodilator drug. In such cases, additional drugs may be required to achieve a useful therapeutic
response.
Clinical Selectivity: Beneficial Versus Toxic Effects of Drugs
Although we classify drugs according to their principal actions, it is clear that no drug causes only a
single, specific effect. Why is this so? It is exceedingly unlikely that any kind of drug molecule will
bind to only a single type of receptor molecule, if only because the number of potential receptors in
every patient is astronomically large. Even if the chemical structure of a drug allowed it to bind to
only one kind of receptor, the biochemical processes controlled by such receptors would take place
in multiple cell types and would be coupled to many other biochemical functions; as a result, the
patient and the prescriber would probably perceive more than one drug effect. Accordingly, drugs
are only selective—rather than specific—in their actions, because they bind to one or a few types of
receptor more tightly than to others and because these receptors control discrete processes that
result in distinct effects.
It is only because of their selectivity that drugs are useful in clinical medicine. Selectivity can be
measured by comparing binding affinities of a drug to different receptors or by comparing ED
50
s for
different effects of a drug in vivo. In drug development and in clinical medicine, selectivity is
usually considered by separating effects into two categories: beneficial or therapeutic effects
versus toxic effects. Pharmaceutical advertisements and prescribers occasionally use the term side
effect, implying that the effect in question is insignificant or occurs via a pathway that is to one side
of the principal action of the drug; such implications are frequently erroneous.
Beneficial and Toxic Effects Mediated by the Same Receptor-Effector Mechanism
Much of the serious drug toxicity in clinical practice represents a direct pharmacologic extension of
the therapeutic actions of the drug. In some of these cases (bleeding caused by anticoagulant
therapy; hypoglycemic coma due to insulin), toxicity may be avoided by judicious management of
the dose of drug administered, guided by careful monitoring of effect (measurements of blood
coagulation or serum glucose) and aided by ancillary measures (avoiding tissue trauma that may
lead to hemorrhage; regulation of carbohydrate intake). In still other cases, the toxicity may be
avoided by not administering the drug at all, if the therapeutic indication is weak or if other therapy
is available.
In certain situations, a drug is clearly necessary and beneficial but produces unacceptable toxicity
when given in doses that produce optimal benefit. In such situations, it may be necessary to add
another drug to the treatment regimen. In treating hypertension, for example, administration of a
second drug often allows the prescriber to reduce the dose and toxicity of the first drug (see Chapter
11: Antihypertensive Agents).
Beneficial and Toxic Effects Mediated by Identical Receptors But in Different Tissues or by
Different Effector Pathways
Many drugs produce both their desired effects and adverse effects by acting on a single receptor
type in different tissues. Examples discussed in this book include: digitalis glycosides, which act by
inhibiting Na
+
/K
+
ATPase in cell membranes; methotrexate, which inhibits the enzyme
dihydrofolate reductase; and glucocorticoid hormones.
Three therapeutic strategies are used to avoid or mitigate this sort of toxicity. First, the drug should
always be administered at the lowest dose that produces acceptable benefit. Second, adjunctive
drugs that act through different receptor mechanisms and produce different toxicities may allow
lowering the dose of the first drug, thus limiting its toxicity (eg, use of other immunosuppressive
agents added to glucocorticoids in treating inflammatory disorders). Third, selectivity of the drug's
actions may be increased by manipulating the concentrations of drug available to receptors in
different parts of the body, for example, by aerosol administration of a glucocorticoid to the bronchi
in asthma.
Beneficial and Toxic Effects Mediated by Different Types of Receptors
Therapeutic advantages resulting from new chemical entities with improved receptor selectivity
were mentioned earlier in this chapter and are described in detail in later chapters. Such drugs
include the
- and -selective adrenoceptor agonists and antagonists, the H
1
and H
2
antihistamines,
nicotinic and muscarinic blocking agents, and receptor-selective steroid hormones. All of these
receptors are grouped in functional families, each responsive to a small class of endogenous
agonists. The receptors and their associated therapeutic uses were discovered by analyzing effects
of the physiologic chemical signals—catecholamines, histamine, acetylcholine, and corticosteroids.
A number of other drugs were discovered by exploiting therapeutic or toxic effects of chemically
similar agents observed in a clinical context. Examples include quinidine, the sulfonylureas,
thiazide diuretics, tricyclic antidepressants, opioid drugs, and phenothiazine antipsychotics. Often
the new agents turn out to interact with receptors for endogenous substances (eg, opioids and
phenothiazines for endogenous opioid and dopamine receptors, respectively). It is likely that other
new drugs will be found to do so in the future, perhaps leading to the discovery of new classes of
receptors and endogenous ligands for future drug development.
Thus, the propensity of drugs to bind to different classes of receptor sites is not only a potentially
vexing problem in treating patients, it also presents a continuing challenge to pharmacology and an
opportunity for developing new and more useful drugs.
Chapter 3. Pharmacokinetics & Pharmacodynamics: Rational
Dosing & the Time Course of Drug Action
Pharmacokinetics & Pharmacodynamics: Rational Dosing & the Time Course of Drug Action:
Introduction
The goal of therapeutics is to achieve a desired beneficial effect with minimal adverse effects.
When a medicine has been selected for a patient, the clinician must determine the dose that most
closely achieves this goal. A rational approach to this objective combines the principles of
pharmacokinetics with pharmacodynamics to clarify the dose-effect relationship (Figure 3–1).
Pharmacodynamics governs the concentration-effect part of the interaction, whereas
pharmacokinetics deals with the dose-concentration part (Holford & Sheiner, 1981). The
pharmacokinetic processes of absorption, distribution, and elimination determine how rapidly and
for how long the drug will appear at the target organ. The pharmacodynamic concepts of maximum
response and sensitivity determine the magnitude of the effect at a particular concentration (see E
max
and EC
50
, Chapter 2: Drug Receptors & Pharmacodynamics).
Figure 3–1.
The relationship between dose and effect can be separated into pharmacokinetic (dose-
concentration) and pharmacodynamic (concentration-effect) components. Concentration provides
the link between pharmacokinetics and pharmacodynamics and is the focus of the target
concentration approach to rational dosing. The three primary processes of pharmacokinetics are
absorption, distribution, and elimination.
Figure 3–1 illustrates a fundamental hypothesis of pharmacology, namely, that a relationship exists
between a beneficial or toxic effect of a drug and the concentration of the drug. This hypothesis has
been documented for many drugs, as indicated by the Target Concentrations and Toxic
Concentrations columns in Table 3–1. The apparent lack of such a relationship for some drugs does
not weaken the basic hypothesis but points to the need to consider the time course of concentration
at the actual site of pharmacologic effect (see below).
Table 3–1. Pharmacokinetic and Pharmacodynamic Parameters for Selected Drugs. (See Speight &
Holford, 1997, for a More Comprehensive Listing.)
Drug Oral
Availabi
lity (F)
(%)
Urinar
y
Excreti
on (%)
Boun
d in
Plas
ma
(%)
Cleara
nce
(L/h/70
kg)
1
Volume
of
Distribut
ion (L/70
kg)
Half-
Life (h)
Target
Concentrat
ions
Toxic
Concentrat
ions
Acetaminoph
en
88 3 0 21 67 2 15 mg/L >300 mg/L
Acyclovir 23 75 15 19.8 48 2.4 . . . . . .
Amikacin . . . 98 4 5.46 19 2.3 . . . . . .
Amoxicillin 93 86 18 10.8 15 1.7 . . . . . .
Amphoterici
n
. . . 4 90 1.92 53 18 . . . . . .
Ampicillin 62 82 18 16.2 20 1.3 . . . . . .
Aspirin 68 1 49 39 11 0.25 . . . . . .
Atenolol 56 94 5 10.2 67 6.1 1 mg/L . . .
Atropine 50 57 18 24.6 120 4.3 . . . . . .
Captopril 65 38 30 50.4 57 2.2 50 ng/mL . . .
Carbamazepi
ne
70 1 74 5.34 98 15 6 mg/L >9 mg/L
Cephalexin 90 91 14 18 18 0.9 . . . . . .
Cephalothin . . . 52 71 28.2 18 0.57 . . . . . .
Chloramphen
icol
80 25 53 10.2 66 2.7 . . . . . .
Chlordiazepo
xide
100 1 97 2.28 21 10 1 mg/L . . .
Chloroquine 89 61 61 45 13000 214 20 ng/mL 250 ng/mL
Chlorpropam
ide
90 20 96 0.126 6.8 33 . . . . . .
Cimetidine 62 62 19 32.4 70 1.9 0.8 mg/L . . .
Ciprofloxaci
n
60 65 40 25.2 130 4.1 . . . . . .
Clonidine 95 62 20 12.6 150 12 1 ng/mL . . .
Cyclosporine 23 1 93 24.6 85 5.6 200 ng/mL >400 ng/mL
Diazepam 100 1 99 1.62 77 43 300 ng/mL . . .
Digitoxin 90 32 97 0.234 38 161 10 ng/mL >35 ng/mL
Digoxin 70 60 25 7 500 50 1 ng/mL >2 ng/mL
Diltiazem 44 4 78 50.4 220 3.7 . . . . . .
Disopyramid
e
83 55
2
5.04 41 6 3 mg/L >8 mg/L
Enalapril 95 90 55 9 40 3 > 0.5 ng/mL . . .
Erythromyci
n
35 12 84 38.4 55 1.6 . . . . . .
Ethambutol 77 79 5 36 110 3.1 . . . >10 mg/L
Fluoxetine 60 3 94 40.2 2500 53 . . . . . .
Furosemide 61 66 99 8.4 7.7 1.5 . . . >25 mg/L
Gentamicin . . . 90 10 5.4 18 2.5 . . . . . .
Hydralazine 40 10 87 234 105 1 100 ng/mL . . .
Imipramine 40 2 90 63 1600 18 200 ng/mL >1 mg/L
Indomethacin 98 15 90 8.4 18 2.4 1 mg/L >5 mg/L
Labetalol 18 5 50 105 660 4.9 0.1 mg/L . . .
Lidocaine 35 2 70 38.4 77 1.8 3 mg/L >6 mg/L
Lithium 100 95 0 1.5 55 22 0.7 mEq/L >2 mEq/L
Meperidine 52 12 58 72 310 3.2 0.5 mg/L . . .
Methotrexate 70 48 34 9 39 7.2 750 M-h
3
>950 M-h
Metoprolol 38 10 11 63 290 3.2 25 ng/mL . . .
Metronidazol
e
99 10 10 5.4 52 8.5 4 mg/L . . .
Midazolam 44 56 95 27.6 77 1.9 . . . . . .
Morphine 24 8 35 60 230 1.9 60 ng/mL . . .
Nifedipine 50 0 96 29.4 55 1.8 50 ng/mL . . .
Nortriptyline 51 2 92 30 1300 31 100 ng/mL >500 ng/mL
Phenobarbita
l
100 24 51 0.258 38 98 15 mg/L >30 mg/L
Phenytoin 90 2 89 Conc-
depende
nt
4
45 Conc-
depende
nt
5
10 mg/L >20 mg/L
Prazosin 68 1 95 12.6 42 2.9 . . . . . .
Procainamide 83 67 16 36 130 3 5 mg/L >14 mg/L
Propranolol 26 1 87 50.4 270 3.9 20 ng/mL . . .
Pyridostigmi
ne
14 85 . . . 36 77 1.9 75 ng/mL . . .
Quinidine 80 18 87 19.8 190 6.2 3 mg/L >8 mg/L
Ranitidine 52 69 15 43.8 91 2.1 100 ng/mL . . .
Rifampin ? 7 89 14.4 68 3.5 . . . . . .
Salicylic acid 100 15 85 0.84 12 13 200 mg/L >200 mg/L
Sulfamethox
azole
100 14 62 1.32 15 10 . . . . . .
Terbutaline 14 56 20 14.4 125 14 2 ng/mL . . .
Tetracycline 77 58 65 7.2 105 11 . . . . . .
Theophylline 96 18 56 2.8 35 8.1 10 mg/L >20 mg/L
Tobramycin . . . 90 10 4.62 18 2.2 . . . . . .
Tocainide 89 38 10 10.8 210 14 10 mg/L . . .
Tolbutamide 93 0 96 1.02 7 5.9 100 mg/L . . .
Trimethopri
m
100 69 44 9 130 11 . . . . . .
Tubocurarine . . . 63 50 8.1 27 2 0.6 mg/L . . .
Valproic acid 100 2 93 0.462 9.1 14 75 mg/L >150 mg/L
Vancomycin . . . 79 30 5.88 27 5.6 . . . . . .
Verapamil 22 3 90 63 350 4 . . . . . .
Warfarin 93 3 99 0.192 9.8 37 . . . . . .
Zidovudine 63 18 25 61.8 98 1.1 . . . . . .
1
Convert to mL/min by multiplying the number given by 16.6.
2
Varies with concentration.
3
Target area under the concentration time curve after a single dose.
4
Can be estimated from measured C
p
using CL = V
max
/(K
m
+ C
p
); V
max
= 415 mg/d, K
m
= 5 mg/L.
See text.
5
Varies because of concentration-dependent clearance.
Knowing the relationship between dose, drug concentration and effects allows the clinician to take
into account the various pathologic and physiologic features of a particular patient that make him or
her different from the average individual in responding to a drug. The importance of
pharmacokinetics and pharmacodynamics in patient care thus rests upon the improvement in
therapeutic benefit and reduction in toxicity that can be achieved by application of these principles.
Pharmacokinetics
The "standard" dose of a drug is based on trials in healthy volunteers and patients with average
ability to absorb, distribute, and eliminate the drug (see Clinical Trials: The IND and NDA in
Chapter 5: Basic & Clinical Evaluation of New Drugs). This dose will not be suitable for every
patient. Several physiologic processes (eg, maturation of organ function in infants) and pathologic
processes (eg, heart failure, renal failure) dictate dosage adjustment in individual patients. These
processes modify specific pharmacokinetic parameters. The two basic parameters are clearance, the
measure of the ability of the body to eliminate the drug; and volume of distribution, the measure of
the apparent space in the body available to contain the drug. These parameters are illustrated
schematically in Figure 3–2, where the volume of the compartments into which the drugs diffuse
represents the volume of distribution and the size of the outflow "drain" in Figures 3–2 B and D
represents the clearance.
Figure 3–2.
Models of drug distribution and elimination. The effect of adding drug to the blood by rapid
intravenous injection is represented by expelling a known amount of the agent into a beaker. The
time course of the amount of drug in the beaker is shown in the graphs at the right. In the first
example (A), there is no movement of drug out of the beaker, so the graph shows only a steep rise
to maximum followed by a plateau. In the second example (B), a route of elimination is present,
and the graph shows a slow decay after a sharp rise to a maximum. Because the level of material in
the beaker falls, the "pressure" driving the elimination process also falls, and the slope of the curve
decreases. This is an exponential decay curve. In the third model (C), drug placed in the first
compartment ("blood") equilibrates rapidly with the second compartment ("extravascular volume")
and the amount of drug in "blood" declines exponentially to a new steady state. The fourth model
(D) illustrates a more realistic combination of elimination mechanism and extravascular
equilibration. The resulting graph shows an early distribution phase followed by the slower
elimination phase.
Volume of Distribution
Volume of distribution (V
d
) relates the amount of drug in the body to the concentration of drug (C)
in blood or plasma:
The volume of distribution may be defined with respect to blood, plasma, or water (unbound drug),
depending on the concentration used in equation (1) (C = C
b
, C
p
, or C
u
).
That the V
d
calculated from equation (1) is an apparent volume may be appreciated by comparing
the volumes of distribution of drugs such as digoxin or chloroquine (Table 3–1) with some of the
physical volumes of the body (Table 3–2). Volume of distribution can vastly exceed any physical
volume in the body because it is the volume apparently necessary to contain the amount of drug
homogeneously at the concentration found in the blood, plasma, or water. Drugs with very high
volumes of distribution have much higher concentrations in extravascular tissue than in the vascular
compartment, ie, they are not homogeneously distributed. Drugs that are completely retained within
the vascular compartment, on the other hand, have a minimum possible volume of distribution equal
to the blood component in which they are distributed, eg, 0.04 L/kg body weight or 2.8 L/70 kg
(Table 3–2) for a drug that is restricted to the plasma compartment.
Table 3–2. Physical Volumes (in L/Kg Body Weight) of Some Body Compartments into Which
Drugs May Be Distributed.
Compartment and Volume Examples of Drugs
Water
Total body water (0.6 L/kg
1
)
Small water-soluble molecules: eg, ethanol.
Extracellular water (0.2 L/kg) Larger water-soluble molecules: eg, gentamicin.
Blood (0.08 L/kg); plasma (0.04
L/kg)
Strongly plasma protein-bound molecules and very large
molecules: eg, heparin.
Fat (0.2–0.35 L/kg) Highly lipid-soluble molecules: eg, DDT.
Bone (0.07 L/kg) Certain ions: eg, lead, fluoride.
1
An average figure. Total body water in a young lean man might be 0.7 L/kg; in an obese woman,
0.5 L/kg.
Clearance
Drug clearance principles are similar to the clearance concepts of renal physiology. Clearance of a
drug is the factor that predicts the rate of elimination in relation to the drug concentration:
Clearance, like volume of distribution, may be defined with respect to blood (CL
b
), plasma (CL
p
),
or unbound in water (CL
u
), depending on the concentration measured.
It is important to note the additive character of clearance. Elimination of drug from the body may
involve processes occurring in the kidney, the lung, the liver, and other organs. Dividing the rate of
elimination at each organ by the concentration of drug presented to it yields the respective clearance
at that organ. Added together, these separate clearances equal total systemic clearance:
"Other" tissues of elimination could include the lungs and additional sites of metabolism, eg, blood
or muscle.
The two major sites of drug elimination are the kidneys and the liver. Clearance of unchanged drug
in the urine represents renal clearance. Within the liver, drug elimination occurs via
biotransformation of parent drug to one or more metabolites, or excretion of unchanged drug into
the bile, or both. The pathways of biotransformation are discussed in Chapter 4: Drug
Biotransformation. For most drugs, clearance is constant over the concentration range encountered
in clinical settings, ie, elimination is not saturable, and the rate of drug elimination is directly
proportional to concentration (rearranging equation [2]):
This is usually referred to as first-order elimination. When clearance is first-order, it can be
estimated by calculating the area under the curve (AUC) of the time-concentration profile after a
dose. Clearance is calculated from the dose divided by the AUC.
Capacity-Limited Elimination
For drugs that exhibit capacity-limited elimination (eg, phenytoin, ethanol), clearance will vary
depending on the concentration of drug that is achieved (Table 3–1). Capacity-limited elimination is
also known as saturable, dose- or concentration-dependent, nonlinear, and Michaelis-Menten
elimination.
Most drug elimination pathways will become saturated if the dose is high enough. When blood flow
to an organ does not limit elimination (see below), the relation between elimination rate and
concentration (C) is expressed mathematically in equation (5):
The maximum elimination capacity is V
max
, and K
m
is the drug concentration at which the rate of
elimination is 50% of V
max
. At concentrations that are high relative to the K
m
, the elimination rate is
almost independent of concentration—a state of "pseudo-zero order" elimination. If dosing rate
exceeds elimination capacity, steady state cannot be achieved: The concentration will keep on rising
as long as dosing continues. This pattern of capacity-limited elimination is important for three drugs
in common use: ethanol, phenytoin, and aspirin. Clearance has no real meaning for drugs with
capacity-limited elimination, and AUC cannot be used to describe the elimination of such drugs.
Flow-Dependent Elimination
In contrast to capacity-limited drug elimination, some drugs are cleared very readily by the organ of
elimination, so that at any clinically realistic concentration of the drug, most of the drug in the
blood perfusing the organ is eliminated on the first pass of the drug through it. The elimination of
these drugs will thus depend primarily on the rate of drug delivery to the organ of elimination. Such
drugs (see Table 4–7) can be called "high-extraction" drugs since they are almost completely
extracted from the blood by the organ. Blood flow to the organ is the main determinant of drug
delivery, but plasma protein binding and blood cell partitioning may also be important for
extensively bound drugs that are highly extracted.
Half-Life
Half-life (t
1/2
) is the time required to change the amount of drug in the body by one-half during
elimination (or during a constant infusion). In the simplest case—and the most useful in designing
drug dosage regimens—the body may be considered as a single compartment (as illustrated in
Figure 3–2 B) of a size equal to the volume of distribution (V
d
). The time course of drug in the body
will depend on both the volume of distribution and the clearance:
*
The constant 0.7 in equation (6) is an approximation to the natural logarithm of 2. Because drug
elimination can be described by an exponential process, the time taken for a twofold decrease can
be shown to be proportional to ln(2).
Half-life is useful because it indicates the time required to attain 50% of steady state—or to decay
50% from steady-state conditions—after a change in the rate of drug administration. Figure 3–3
shows the time course of drug accumulation during a constant-rate drug infusion and the time
course of drug elimination after stopping an infusion that has reached steady state.
Figure 3–3.
The time course of drug accumulation and elimination. Solid line: Plasma concentrations
reflecting drug accumulation during a constant rate infusion of a drug. Fifty percent of the steady-
state concentration is reached after one half-life, 75% after two half-lives, and over 90% after four
half-lives. Dashed line: Plasma concentrations reflecting drug elimination after a constant rate
infusion of a drug had reached steady state. Fifty percent of the drug is lost after one half-life, 75%
after two half-lives, etc. The "rule of thumb" that four half-lives must elapse after starting a drug-
dosing regimen before full effects will be seen is based on the approach of the accumulation curve
to over 90% of the final steady-state concentration.
Disease states can affect both of the physiologically related primary pharmacokinetic parameters:
volume of distribution and clearance. A change in half-life will not necessarily reflect a change in
drug elimination. For example, patients with chronic renal failure have decreased renal clearance of
digoxin but also a decreased volume of distribution; the increase in digoxin half-life is not as great
as might be expected based on the change in renal function. The decrease in volume of distribution
is due to the decreased renal and skeletal muscle mass and consequent decreased tissue binding of
digoxin to Na
+
/K
+
ATPase.
Many drugs will exhibit multicompartment pharmacokinetics (as illustrated in Figures 3–2 C and
D). Under these conditions, the "true" terminal half-life, as given in Table 3–1, will be greater than
that calculated from equation (6).
Drug Accumulation
Whenever drug doses are repeated, the drug will accumulate in the body until dosing stops. This is
because it takes an infinite time (in theory) to eliminate all of a given dose. In practical terms, this
means that if the dosing interval is shorter than four half-lives, accumulation will be detectable.
Accumulation is inversely proportional to the fraction of the dose lost in each dosing interval. The
fraction lost is 1 minus the fraction remaining just before the next dose. The fraction remaining can
be predicted from the dosing interval and the half-life. A convenient index of accumulation is the
accumulation factor.
For a drug given once every half-life, the accumulation factor is 1/0.5, or 2. The accumulation
factor predicts the ratio of the steady-state concentration to that seen at the same time following the
first dose. Thus, the peak concentrations after intermittent doses at steady state will be equal to the
peak concentration after the first dose multiplied by the accumulation factor.
Bioavailability
Bioavailability is defined as the fraction of unchanged drug reaching the systemic circulation
following administration by any route (Table 3–3). The area under the blood concentration-time
curve (area under the curve, AUC) is a common measure of the extent of bioavailability for a drug
given by a particular route (Figure 3–4). For an intravenous dose of the drug, bioavailability is
assumed to be equal to unity. For a drug administered orally, bioavailability may be less than 100%
for two main reasons—incomplete extent of absorption and first-pass elimination.
Table 3–3. Routes of Administration, Bioavailability, and General Characteristics.
Route Bioavailability
(%)
Characteristics
Intravenous (IV) 100 (by
definition)
Most rapid onset
Intramuscular
(IM)
75 to 100 Large volumes often feasible; may be painful
Subcutaneous
(SC)
75 to 100 Smaller volumes than IM; may be painful
Oral (PO) 5 to <100 Most convenient; first-pass effect may be significant
Rectal (PR) 30 to <100 Less first-pass effect than oral
Inhalation 5 to <100 Often very rapid onset
Transdermal 80 to 100 Usually very slow absorption; used for lack of first-pass
effect; prolonged duration of action
Figure 3–4.
Blood concentration-time curves, illustrating how changes in the rate of absorption and extent of
bioavailability can influence both the duration of action and the effectiveness of the same total
dose of a drug administered in three different formulations. The dashed line indicates the target
concentration (TC) of the drug in the blood.
Extent of Absorption
After oral administration, a drug may be incompletely absorbed, eg, only 70% of a dose of digoxin
reaches the systemic circulation. This is mainly due to lack of absorption from the gut. Other drugs
are either too hydrophilic (eg, atenolol) or too lipophilic (eg, acyclovir) to be absorbed easily, and
their low bioavailability is also due to incomplete absorption. If too hydrophilic, the drug cannot
cross the lipid cell membrane; if too lipophilic, the drug is not soluble enough to cross the water
layer adjacent to the cell. Drugs may not be absorbed because of a reverse transporter associated
with P-glycoprotein. This process actively pumps drug out of gut wall cells back into the gut lumen.
Inhibition of P-glycoprotein and gut wall metabolism, eg, by grapefruit juice, may be associated
with substantially increased drug absorption.
First-Pass Elimination
Following absorption across the gut wall, the portal blood delivers the drug to the liver prior to
entry into the systemic circulation. A drug can be metabolized in the gut wall (eg, by the CYP3A4
enzyme system) or even in the portal blood, but most commonly it is the liver that is responsible for
metabolism before the drug reaches the systemic circulation. In addition, the liver can excrete the
drug into the bile. Any of these sites can contribute to this reduction in bioavailability, and the
overall process is known as first-pass elimination. The effect of first-pass hepatic elimination on
bioavailability is expressed as the extraction ratio (ER):
where Q is hepatic blood flow, normally about 90 L/h in a person weighing 70 kg.
The systemic bioavailability of the drug (F) can be predicted from the extent of absorption (f) and
the extraction ratio (ER):
A drug such as morphine is almost completely absorbed (f = 1), so that loss in the gut is negligible.
However, the hepatic extraction ratio for morphine is 0.67, so (1 – ER) is 0.33. The bioavailability
of morphine is therefore expected to be about 33%, which is close to the observed value (Table 3–
1).
Rate of Absorption
The distinction between rate and extent of absorption is shown in Figure 3–4. The rate of absorption
is determined by the site of administration and the drug formulation. Both the rate of absorption and
the extent of input can influence the clinical effectiveness of a drug. For the three different dosage
forms depicted in Figure 3–4, there would be significant differences in the intensity of clinical
effect. Dosage form B would require twice the dose to attain blood concentrations equivalent to
those of dosage form A. Differences in rate of availability may become important for drugs given as
a single dose, such as a hypnotic used to induce sleep. In this case, drug from dosage form A would
reach its target concentration earlier than drug from dosage form C; concentrations from A would
also reach a higher level and remain above the target concentration for a longer period. In a multiple
dosing regimen, dosage forms A and C would yield the same average blood level concentrations,
although dosage form A would show somewhat greater maximum and lower minimum
concentrations.
The mechanism of drug absorption is said to be zero-order when the rate is independent of the
amount of drug remaining in the gut, eg, when it is determined by the rate of gastric emptying or by
a controlled-release drug formulation. In contrast, when the full dose is dissolved in gastrointestinal
fluids, the rate of absorption is usually proportional to the gastrointestinal concentration and is said
to be first-order.
Extraction Ratio & the First-Pass Effect
Systemic clearance is not affected by bioavailability. However, clearance can markedly affect the
extent of availability because it determines the extraction ratio (equation [8a]). Of course,
therapeutic blood concentrations may still be reached by the oral route of administration if larger
doses are given. However, in this case, the concentrations of the drug metabolites will be increased
significantly over those that would occur following intravenous administration. Lidocaine and
verapamil are both used to treat cardiac arrhythmias and have bioavailability less than 40%, but
lidocaine is never given orally because its metabolites are believed to contribute to central nervous
system toxicity. Other drugs that are highly extracted by the liver include isoniazid, morphine,
propranolol, verapamil, and several tricyclic antidepressants (Table 3–1).
Drugs with high extraction ratios will show marked variations in bioavailability between subjects
because of differences in hepatic function and blood flow. These differences can explain the marked
variation in drug concentrations that occurs among individuals given similar doses of highly
extracted drugs. For drugs that are highly extracted by the liver, shunting of blood past hepatic sites
of elimination will result in substantial increases in drug availability, whereas for drugs that are
poorly extracted by the liver (for which the difference between entering and exiting drug
concentration is small), shunting of blood past the liver will cause little change in availability.
Drugs in Table 3–1 that are poorly extracted by the liver include chlorpropamide, diazepam,
phenytoin, theophylline, tolbutamide, and warfarin.
Alternative Routes of Administration & the First-Pass Effect
There are several reasons for different routes of administration used in clinical medicine (Table 3–
3)—for convenience (eg, oral), to maximize concentration at the site of action and minimize it
elsewhere (eg, topical), to prolong the duration of drug absorption (eg, transdermal), or to avoid the
first-pass effect.
The hepatic first-pass effect can be avoided to a great extent by use of sublingual tablets and
transdermal preparations and to a lesser extent by use of rectal suppositories. Sublingual absorption
provides direct access to systemic—not portal—veins. The transdermal route offers the same
advantage. Drugs absorbed from suppositories in the lower rectum enter vessels that drain into the
inferior vena cava, thus bypassing the liver. However, suppositories tend to move upward in the
rectum into a region where veins that lead to the liver predominate. Thus, only about 50% of a
rectal dose can be assumed to bypass the liver.
Although drugs administered by inhalation bypass the hepatic first-pass effect, the lung may also
serve as a site of first-pass loss by excretion and possibly metabolism for drugs administered by
nongastrointestinal ("parenteral") routes.
The Time Course of Drug Effect
The principles of pharmacokinetics (discussed in this chapter) and those of pharmacodynamics
(discussed in Chapter 2: Drug Receptors & Pharmacodynamics; Holford & Sheiner, 1981) provide a
framework for understanding the time course of drug effect.
Immediate Effects
In the simplest case, drug effects are directly related to plasma concentrations, but this does not
necessarily mean that effects simply parallel the time course of concentrations. Because the
relationship between drug concentration and effect is not linear (recall the E
max
model described in
Chapter 2: Drug Receptors & Pharmacodynamics), the effect will not usually be linearly
proportional to the concentration.
Consider the effect of an angiotensin-converting enzyme (ACE) inhibitor, such as enalapril, on
plasma ACE. The half-life of enalapril is about 3 hours. After an oral dose of 10 mg, the peak
plasma concentration at 3 hours is about 64 ng/mL. Enalapril is usually given once a day, so seven
half-lives will elapse from the time of peak concentration to the end of the dosing interval. The
concentration of enalapril after each half-life and the corresponding extent of ACE inhibition are
shown in Figure 3–5. The extent of inhibition of ACE is calculated using the E
max
model, where
E
max
, the maximum extent of inhibition, is 100% and the EC
50
is about 1 ng/mL.
Figure 3–5.
Time course of angiotensin-converting enzyme (ACE) inhibitor concentrations and effects. The
black line shows the plasma enalapril concentrations in nanograms per milliliter after a single oral
dose. The colored line indicates the percentage inhibition of its target, ACE. Note the different
shapes of the concentration-time course (exponentially decreasing) and the effect-time course
(linearly decreasing in its central portion).
Note that plasma concentrations of enalapril change by a factor of 16 over the first 12 hours (four
half-lives) after the peak, but ACE inhibition has only decreased by 20%. Because the
concentrations over this time are so high in relation to the EC
50
, the effect on ACE is almost
constant. After 24 hours, ACE is still 33% inhibited. This explains why a drug with a short half-life
can be given once a day and still maintain its effect throughout the day. The key factor is a high
initial concentration in relation to the EC
50
. Even though the plasma concentration at 24 hours is
less than 1% of its peak, this low concentration is still half the EC
50
. This is very common for drugs
that act on enzymes (eg, ACE inhibitors) or compete at receptors (eg, propranolol).
When concentrations are in the range between one fourth and four times the EC
50
, the time course
of effect is essentially a linear function of time—13% of the effect is lost every half-life over this
concentration range. At concentrations below one fourth the EC
50
, the effect becomes almost
directly proportional to concentration and the time course of drug effect will follow the exponential
decline of concentration. It is only when the concentration is low in relation to the EC
50
that the
concept of a "half-life of drug effect" has any meaning.
Delayed Effects
Changes in drug effects are often delayed in relation to changes in plasma concentration. This delay
may reflect the time required for the drug to distribute from plasma to the site of action. This will be
the case for almost all drugs. The delay due to distribution is a pharmacokinetic phenomenon that
can account for delays of a few minutes. This distributional delay can account for the lag of effects
after rapid intravenous injection of central nervous system (CNS)–active agents such as thiopental.
A common reason for more delayed drug effects—especially those that take many hours or even
days to occur—is the slow turnover of a physiologic substance that is involved in the expression of
the drug effect (Jusko & Ko, 1994). For example, warfarin works as an anticoagulant by inhibiting
vitamin K epoxidase in the liver. This action of warfarin occurs rapidly, and inhibition of the
enzyme is closely related to plasma concentrations of warfarin. The clinical effect of warfarin, eg,
on the prothrombin time, reflects a decrease in the concentration of the prothrombin complex of
clotting factors (see Figure 34–7). Inhibition of vitamin K epoxidase decreases the synthesis of
these clotting factors, but the complex has a long half-life (about 14 hours), and it is this half-life
that determines how long it takes for the concentration of clotting factors to reach a new steady state
and for a drug effect to become manifest that reflects the warfarin plasma concentration.
Cumulative Effects
Some drug effects are more obviously related to a cumulative action than to a rapidly reversible
one. The renal toxicity of aminoglycoside antibiotics (eg, gentamicin) is greater when administered
as a constant infusion than with intermittent dosing. It is the accumulation of aminoglycoside in the
renal cortex that is thought to cause renal damage. Even though both dosing schemes produce the
same average steady-state concentration, the intermittent dosing scheme produces much higher peak
concentrations, which saturate an uptake mechanism into the cortex; thus, total aminoglycoside
accumulation is less. The difference in toxicity is a predictable consequence of the different patterns
of concentration and the saturable uptake mechanism.
The effect of many drugs used to treat cancer also reflects a cumulative action—eg, the extent of
binding of a drug to DNA is proportional to drug concentration and is usually irreversible. The
effect on tumor growth is therefore a consequence of cumulative exposure to the drug. Measures of
cumulative exposure, such as AUC, provide a means to individualize treatment (Evans et al, 1998).
The Target Concentration Approach to Designing a Rational Dosage Regimen
A rational dosage regimen is based on the assumption that there is a target concentration that will
produce the desired therapeutic effect. By considering the pharmacokinetic factors that determine
the dose-concentration relationship, it is possible to individualize the dose regimen to achieve the
target concentration. The effective concentration ranges shown in Table 3–1 are a guide to the
concentrations measured when patients are being effectively treated. The initial target concentration
should usually be chosen from the lower end of this range. In some cases, the target concentration
will also depend on the specific therapeutic objective—eg, the control of atrial fibrillation by
digoxin often requires a target concentration of 2 ng/mL, while heart failure is usually adequately
managed with a target concentration of 1 ng/mL.
Maintenance Dose
In most clinical situations, drugs are administered in such a way as to maintain a steady state of
drug in the body, ie, just enough drug is given in each dose to replace the drug eliminated since the
preceding dose. Thus, calculation of the appropriate maintenance dose is a primary goal. Clearance
is the most important pharmacokinetic term to be considered in defining a rational steady state drug
dosage regimen. At steady state, the dosing rate ("rate in") must equal the rate of elimination ("rate
out"). Substitution of the target concentration (TC) for concentration (C) in equation (4) predicts the
maintenance dosing rate:
Thus, if the desired target concentration is known, the clearance in that patient will determine the
dosing rate. If the drug is given by a route that has a bioavailability less than 100%, then the dosing
rate predicted by equation (9) must be modified. For oral dosing:
If intermittent doses are given, the maintenance dose is calculated from:
(See Example: Maintenance Dose Calculation.)
Note that the steady-state concentration achieved by continuous infusion or the average
concentration following intermittent dosing depends only on clearance. The volume of distribution
and the half-life need not be known in order to determine the average plasma concentration
expected from a given dosing rate or to predict the dosing rate for a desired target concentration.
Figure 3–6 shows that at different dosing intervals, the concentration time curves will have different
maximum and minimum values even though the average level will always be 10 mg/L.
Estimates of dosing rate and average steady-state concentrations, which may be calculated using
clearance, are independent of any specific pharmacokinetic model. In contrast, the determination of
maximum and minimum steady-state concentrations requires further assumptions about the
pharmacokinetic model. The accumulation factor (equation [7]) assumes that the drug follows a
one-compartment body model (Figure 3–2 B), and the peak concentration prediction assumes that
the absorption rate is much faster than the elimination rate. For the calculation of estimated
maximum and minimum concentrations in a clinical situation, these assumptions are usually
reasonable.
Example: Maintenance Dose Calculation
A target plasma theophylline concentration of 10 mg/L is desired to relieve acute bronchial asthma
in a patient. If the patient is a nonsmoker and otherwise normal except for asthma, we may use the
mean clearance given in Table 3–1, ie, 2.8 L/h/70 kg. Since the drug will be given as an intravenous
infusion, F = 1.
Therefore, in this patient, the proper infusion rate would be 28 mg/h/70 kg.
If the asthma attack is relieved, the clinician might want to maintain this plasma level using oral
theophylline, which might be given every 12 hours using an extended-release formulation to
approximate a continuous intravenous infusion. According to Table 3–1, F
oral
is 0.96. When the
dosing interval is 12 hours, the size of each maintenance dose would be:
A tablet or capsule size close to the ideal dose of 350 mg would then be prescribed at 12-hourly
intervals. If an 8-hour dosing interval was used, the ideal dose would be 233 mg; and if the drug
was given once a day, the dose would be 700 mg. In practice, F could be omitted from the
calculation since it is so close to 1.
Loading Dose
When the time to reach steady state is appreciable, as it is for drugs with long half-lives, it may be
desirable to administer a loading dose that promptly raises the concentration of drug in plasma to
the target concentration. In theory, only the amount of the loading dose need be computed—not the
rate of its administration—and, to a first approximation, this is so. The volume of distribution is the
proportionality factor that relates the total amount of drug in the body to the concentration in the
plasma (C
p
); if a loading dose is to achieve the target concentration, then from equation (1):
For the theophylline example given in Example: Maintenance Dose Calculation, the loading dose
would be 350 mg (35 L x 10 mg/L) for a 70 kg person. For most drugs, the loading dose can be
given as a single dose by the chosen route of administration.
Up to this point, we have ignored the fact that some drugs follow more complex multicompartment
pharmacokinetics, eg, the distribution process illustrated by the two-compartment model in Figure
3–2. This is justified in the great majority of cases. However, in some cases the distribution phase
may not be ignored, particularly in connection with the calculation of loading doses. If the rate of
absorption is rapid relative to distribution (this is always true for intravenous bolus administration),
the concentration of drug in plasma that results from an appropriate loading dose—calculated using
the apparent volume of distribution—can initially be considerably higher than desired. Severe
toxicity may occur, albeit transiently. This may be particularly important, for example, in the
administration of antiarrhythmic drugs such as lidocaine, where an almost immediate toxic response
may occur. Thus, while the estimation of the amount of a loading dose may be quite correct, the
rate of administration can sometimes be crucial in preventing excessive drug concentrations, and
slow administration of an intravenous drug (over minutes rather than seconds) is almost always
prudent practice. For intravenous doses of theophylline, initial injections should be given over a 20-
minute period to reduce the possibility of high plasma concentrations during the distribution phase.
When intermittent doses are given, the loading dose calculated from equation (12) will only reach
the average steady-state concentration and will not match the peak steady-state concentration (see
Figure 3–6). To match the peak steady-state concentration, the loading dose can be calculated from
equation (13):
Figure 3–6.
Relationship between frequency of dosing and maximum and minimum plasma concentrations
when a steady-state theophylline plasma level of 10 mg/L is desired. The smoothly rising line
(solid black) shows the plasma concentration achieved with an intravenous infusion of 28 mg/h.
The doses for 8-hourly administration (light color) are 224 mg; for 24-hourly administration (dark
color), 672 mg. In each of the 3 cases, the mean steady-state plasma concentration is 10 mg/L.
Therapeutic Drug Monitoring: Relating Pharmacokinetics & Pharmacodynamics
The basic principles outlined above can be applied to the interpretation of clinical drug
concentration measurements on the basis of three major pharmacokinetic variables: absorption,
clearance, and volume of distribution (and the derived variable, half-life); and two
pharmacodynamic variables: maximum effect attainable in the target tissue and the sensitivity of the
tissue to the drug. Diseases may modify all of these parameters, and the ability to predict the effect
of disease states on pharmacokinetic parameters is important in properly adjusting dosage in such
cases. (See The Target Concentration Strategy.)
The Target Concentration Strategy
Recognition of the essential role of concentration in linking pharmacokinetics and
pharmacodynamics leads naturally to the target concentration strategy. Pharmacodynamic principles
can be used to predict the concentration required to achieve a particular degree of therapeutic effect.
This target concentration can then be achieved by using pharmacokinetic principles to arrive at a
suitable dosing regimen (Holford, 1999). The target concentration strategy is a process for
optimizing the dose in an individual on the basis of a measured surrogate response such as drug
concentration:
1. Choose the target concentration, TC.
2. Predict volume of distribution (V
d
) and clearance (CL) based on standard population
values (eg, Table 3–1) with adjustments for factors such as weight and renal function.
3. Give a loading dose or maintenance dose calculated from TC, V
d
, and CL.
4. Measure the patient's response and drug concentration.
5. Revise V
d
and/or CL based on the measured concentration.
6. Repeat steps 3–5, adjusting the predicted dose to achieve TC.
Pharmacokinetic Variables
Absorption
The amount of drug that enters the body depends on the patient's compliance with the prescribed
regimen and on the rate and extent of transfer from the site of administration to the blood.
Overdosage and underdosage relative to the prescribed dosage—both aspects of failure of
compliance—can frequently be detected by concentration measurements when gross deviations
from expected values are obtained. If compliance is found to be adequate, absorption abnormalities
in the small bowel may be the cause of abnormally low concentrations. Variations in the extent of
bioavailability are rarely caused by irregularities in the manufacture of the particular drug
formulation. More commonly, variations in bioavailability are due to metabolism during absorption.
Clearance
Abnormal clearance may be anticipated when there is major impairment of the function of the
kidney, liver, or heart. Creatinine clearance is a useful quantitative indicator of renal function.
Conversely, drug clearance may be a useful indicator of the functional consequences of heart,
kidney, or liver failure, often with greater precision than clinical findings or other laboratory tests.
For example, when renal function is changing rapidly, estimation of the clearance of
aminoglycoside antibiotics may be a more accurate indicator of glomerular filtration than serum
creatinine.
Hepatic disease has been shown to reduce the clearance and prolong the half-life of many drugs.
However, for many other drugs known to be eliminated by hepatic processes, no changes in
clearance or half-life have been noted with similar hepatic disease. This reflects the fact that hepatic
disease does not always affect the hepatic intrinsic clearance. At present, there is no reliable marker
of hepatic drug-metabolizing function that can be used to predict changes in liver clearance in a
manner analogous to the use of creatinine clearance as a marker of renal drug clearance.
Volume of Distribution
The apparent volume of distribution reflects a balance between binding to tissues, which decreases
plasma concentration and makes the apparent volume larger, and binding to plasma proteins, which
increases plasma concentration and makes the apparent volume smaller. Changes in either tissue or
plasma binding can change the apparent volume of distribution determined from plasma
concentration measurements. Older people have a relative decrease in skeletal muscle mass and
tend to have a smaller apparent volume of distribution of digoxin (which binds to muscle proteins).
The volume of distribution may be overestimated in obese patients if based on body weight and the
drug does not enter fatty tissues well, as is the case with digoxin. In contrast, theophylline has a
volume of distribution similar to that of total body water. Adipose tissue has almost as much water
in it as other tissues, so that the apparent total volume of distribution of theophylline is proportional
to body weight even in obese patients.
Abnormal accumulation of fluid—edema, ascites, pleural effusion—can markedly increase the
volume of distribution of drugs such as gentamicin that are hydrophilic and have small volumes of
distribution.
Half-Life
The differences between clearance and half-life are important in defining the underlying
mechanisms for the effect of a disease state on drug disposition. For example, the half-life of
diazepam increases with age. When clearance is related to age, it is found that clearance of this drug
does not change with age. The increasing half-life for diazepam actually results from changes in the
volume of distribution with age; the metabolic processes responsible for eliminating the drug are
fairly constant.
Pharmacodynamic Variables
Maximum Effect
All pharmacologic responses must have a maximum effect (E
max
). No matter how high the drug
concentration goes, a point will be reached beyond which no further increment in response is
achieved.
If increasing the dose in a particular patient does not lead to a further clinical response, it is possible
that the maximum effect has been reached. Recognition of maximum effect is helpful in avoiding
ineffectual increases of dose with the attendant risk of toxicity.
Sensitivity
The sensitivity of the target organ to drug concentration is reflected by the concentration required to
produce 50% of maximum effect, the EC
50
. Failure of response due to diminished sensitivity to the
drug can be detected by measuring—in a patient who is not getting better—drug concentrations that
are usually associated with therapeutic response. This may be a result of abnormal physiology—eg,
hyperkalemia diminishes responsiveness to digoxin—or drug antagonism—eg, calcium channel
blockers impair the inotropic response to digoxin.
Increased sensitivity to a drug is usually signaled by exaggerated responses to small or moderate
doses. The pharmacodynamic nature of this sensitivity can be confirmed by measuring drug
concentrations that are low in relation to the observed effect.
Interpretation of Drug Concentration Measurements
Clearance
Clearance is the single most important factor determining drug concentrations. The interpretation of
measurements of drug concentrations depends on a clear understanding of three factors that may
influence clearance: the dose, the organ blood flow, and the intrinsic function of the liver or
kidneys. Each of these factors should be considered when interpreting clearance estimated from a
drug concentration measurement. It must also be recognized that changes in protein binding may
lead the unwary to believe there is a change in clearance when in fact drug elimination is not altered
(see Plasma Protein Binding: Is It Important?). Factors affecting protein binding include the
following:
1. Albumin concentration: Drugs such as phenytoin, salicylates, and disopyramide are
extensively bound to plasma albumin. Albumin levels are low in many disease states, resulting in
lower total drug concentrations.
2. Alpha
1
-acid glycoprotein concentration:
1
-Acid glycoprotein is an important binding protein
with binding sites for drugs such as quinidine, lidocaine, and propranolol. It is increased in acute
inflammatory disorders and causes major changes in total plasma concentration of these drugs
even though drug elimination is unchanged.
3. Capacity-limited protein binding: The binding of drugs to plasma proteins is capacity-limited.
Therapeutic concentrations of salicylates and prednisolone show concentration-dependent protein
binding. Because unbound drug concentration is determined by dosing rate and clearance—
which is not altered, in the case of these low-extraction-ratio drugs, by protein binding—
increases in dosing rate will cause corresponding changes in the pharmacodynamically important
unbound concentration. Total drug concentration will increase less rapidly than the dosing rate
would suggest as protein binding approaches saturation at higher concentrations.
Plasma Protein Binding: Is It Important?
Plasma protein binding is often mentioned as a factor playing a role in pharmacokinetics,
pharmacodynamics, and drug interactions. However, there are no clinically relevant examples of
changes in drug disposition or effects that can be clearly ascribed to changes in plasma protein
binding (Benet & Hoener 2002). The idea that if a drug is displaced from plasma proteins it would
increase the unbound drug concentration and increase the drug effect and, perhaps, produce toxicity
seems a simple and obvious mechanism. Unfortunately, this simple theory, which is appropriate for
a test tube, does not work in the body, which is an open system capable of eliminating unbound
drug.
First, a seemingly dramatic change in the unbound fraction from 1% to 10% releases less than 5%
of the total amount of drug in the body into the unbound pool because less than one third of the
drug in the body is bound to plasma proteins even in the most extreme cases, eg, warfarin. Drug
displaced from plasma protein will of course distribute throughout the volume of distribution, so
that a 5% increase in the amount of unbound drug in the body produces at most a 5% increase in
pharmacologically active unbound drug at the site of action.
Second, when the amount of unbound drug in plasma increases, the rate of elimination will increase
(if unbound clearance is unchanged), and after four half-lives the unbound concentration will return
to its previous steady state value. When drug interactions associated with protein binding
displacement and clinically important effects have been studied, it has been found that the
displacing drug is also an inhibitor of clearance, and it is the change in clearance of the unbound
drug that is the relevant mechanism explaining the interaction.
The clinical importance of plasma protein binding is only to help interpretation of measured drug
concentrations. When plasma proteins are lower than normal, then total drug concentrations will be
lower but unbound concentrations will not be affected.
Dosing History
An accurate dosing history is essential if one is to obtain maximum value from a drug concentration
measurement. In fact, if the dosing history is unknown or incomplete, a drug concentration
measurement loses all predictive value.
Timing of Samples for Concentration Measurement
Information about the rate and extent of drug absorption in a particular patient is rarely of great
clinical importance. However, absorption usually occurs during the first 2 hours after a drug dose
and varies according to food intake, posture, and activity. Therefore, it is important to avoid
drawing blood until absorption is complete (about 2 hours after an oral dose). Attempts to measure
peak concentrations early after oral dosing are usually unsuccessful and compromise the validity of
the measurement, because one cannot be certain that absorption is complete.
Some drugs such as digoxin and lithium take several hours to distribute to tissues. Digoxin samples
should be taken at least 6 hours after the last dose and lithium just before the next dose (usually 24
hours after the last dose). Aminoglycosides distribute quite rapidly, but it is still prudent to wait 1
hour after giving the dose before taking a sample.
Clearance is readily estimated from the dosing rate and mean steady-state concentration. Blood
samples should be appropriately timed to estimate steady-state concentration. Provided steady state
has been approached (at least three half-lives of constant dosing), a sample obtained near the
midpoint of the dosing interval will usually be close to the mean steady-state concentration.
Initial Predictions of Volume of Distribution & Clearance
Volume of Distribution
Volume of distribution is commonly calculated for a particular patient using body weight (70 kg
body weight is assumed for the values in Table 3–1). If a patient is obese, drugs that do not readily
penetrate fat (eg, gentamicin and digoxin) should have their volumes calculated from ideal body
weight as shown below:
Patients with edema, ascites, or pleural effusions offer a larger volume of distribution to the
aminoglycoside antibiotics (eg, gentamicin) than is predicted by body weight. In such patients, the
weight should be corrected as follows: Subtract an estimate of the weight of the excess fluid
accumulation from the measured weight. Use the resultant "normal" body weight to calculate the
normal volume of distribution. Finally, this normal volume should be increased by 1 L for each
estimated kilogram of excess fluid. This correction is important because of the relatively small
volumes of distribution of these water-soluble drugs.
Clearance
Drugs cleared by the renal route often require adjustment of clearance in proportion to renal
function. This can be conveniently estimated from the creatinine clearance, calculated from a single
serum creatinine measurement and the predicted creatinine production rate.
The predicted creatinine production rate in women is 85% of the calculated value, because they
have a smaller muscle mass per kilogram and it is muscle mass that determines creatinine
production. Muscle mass as a fraction of body weight decreases with age, which is why age appears
in the Cockcroft-Gault equation, given in Chapter 61: Special Aspects of Geriatric Pharmacology.
The decrease of renal function with age is independent of the decrease in creatinine production.
Because of the difficulty of obtaining complete urine collections, creatinine clearance calculated in
this way is at least as reliable as estimates based on urine collections. Ideal body weight should be
used for obese patients, and correction should be made for muscle wasting in severely ill patients.
Revising Individual Estimates of Volume of Distribution & Clearance
The commonsense approach to the interpretation of drug concentrations compares predictions of
pharmacokinetic parameters and expected concentrations to measured values. If measured
concentrations differ by more than 20% from predicted values, revised estimates of V
d
or CL for
that patient should be calculated using equation (1) or equation (2). If the change calculated is more
than a 100% increase or 50% decrease in either V
d
or CL, the assumptions made about the timing of
the sample and the dosing history should be critically examined.
For example, if a patient is taking 0.25 mg of digoxin a day, a clinician may expect the digoxin
concentration to be about 1 ng/mL. This is based on typical values for bioavailability of 70% and
total clearance of about 7 L/h (CL
renal
4 L/h, CL
nonrenal
3 L/h). If the patient has heart failure, the
nonrenal (hepatic) clearance might be halved because of hepatic congestion and hypoxia, so the
expected clearance would become 5.5 L/h. The concentration is then expected to be about 1.3
ng/mL. Suppose that the concentration actually measured is 2 ng/mL. Common sense would
suggest halving the daily dose to achieve a target concentration of 1 ng/mL. This approach implies a
revised clearance of 3.5 L/h. The smaller clearance compared with the expected value of 5.5 L/h
may reflect additional renal functional impairment due to heart failure.
This technique will often be misleading if steady state has not been reached. At least a week of
regular dosing (three to four half-lives) must elapse before the implicit method will be reliable.
Chapter 4. Drug Biotransformation
Drug Biotransformation: Introduction
Humans are exposed daily to a wide variety of foreign compounds called xenobiotics—substances
absorbed across the lungs or skin or, more commonly, ingested either unintentionally as compounds
present in food and drink or deliberately as drugs for therapeutic or "recreational" purposes.
Exposure to environmental xenobiotics may be inadvertent and accidental or—when they are
present as components of air, water, and food—inescapable. Some xenobiotics are innocuous, but
many can provoke biologic responses. Such biologic responses often depend on conversion of the
absorbed substance into an active metabolite. The discussion that follows is applicable to
xenobiotics in general (including drugs) and to some extent to endogenous compounds.
Why Is Drug Biotransformation Necessary?
Renal excretion plays a pivotal role in terminating the biologic activity of some drugs, particularly
those that have small molecular volumes or possess polar characteristics such as functional groups
that are fully ionized at physiologic pH. However, many drugs do not possess such physicochemical
properties. Pharmacologically active organic molecules tend to be lipophilic and remain un-ionized
or only partially ionized at physiologic pH. They are often strongly bound to plasma proteins. Such
substances are not readily filtered at the glomerulus. The lipophilic nature of renal tubular
membranes also facilitates the reabsorption of hydrophobic compounds following their glomerular
filtration. Consequently, most drugs would have a prolonged duration of action if termination of
their action depended solely on renal excretion.
An alternative process that may lead to the termination or alteration of biologic activity is
metabolism. In general, lipophilic xenobiotics are transformed to more polar and hence more
readily excretable products. The role metabolism may play in the inactivation of lipid-soluble drugs
can be quite dramatic. For example, lipophilic barbiturates such as thiopental and pentobarbital
would have extremely long half-lives if it were not for their metabolic conversion to more water-
soluble compounds.
Metabolic products are often less pharmacodynamically active than the parent drug and may even
be inactive. However, some biotransformation products have enhanced activity or toxic properties.
It is noteworthy that the synthesis of endogenous substrates such as steroid hormones, cholesterol,
active vitamin D congeners and bile acids involves many pathways catalyzed by enzymes
associated with the metabolism of xenobiotics. Finally, drug-metabolizing enzymes have been
exploited in the design of pharmacologically inactive prodrugs that are converted to active
molecules in the body.
Where Do Drug Biotransformations Occur?
Although every tissue has some ability to metabolize drugs, the liver is the principal organ of drug
metabolism. Other tissues that display considerable activity include the gastrointestinal tract, the
lungs, the skin, and the kidneys. Following oral administration, many drugs (eg, isoproterenol,
meperidine, pentazocine, morphine) are absorbed intact from the small intestine and transported
first via the portal system to the liver, where they undergo extensive metabolism. This process has
been called a first-pass effect (see Chapter 3: Pharmacokinetics & Pharmacodynamics: Rational
Dosing & the Time Course of Drug Action). Some orally administered drugs (eg, clonazepam,
chlorpromazine) are more extensively metabolized in the intestine than in the liver. Thus, intestinal
metabolism may contribute to the overall first-pass effect. First-pass effects may so greatly limit the
bioavailability of orally administered drugs that alternative routes of administration must be used to
achieve therapeutically effective blood levels. The lower gut harbors intestinal microorganisms that
are capable of many biotransformation reactions. In addition, drugs may be metabolized by gastric
acid (eg, penicillin), by digestive enzymes (eg, polypeptides such as insulin), or by enzymes in the
wall of the intestine (eg, sympathomimetic catecholamines).
Although drug biotransformation in vivo can occur by spontaneous, noncatalyzed chemical
reactions, the vast majority of transformations are catalyzed by specific cellular enzymes. At the
subcellular level, these enzymes may be located in the endoplasmic reticulum, mitochondria,
cytosol, lysosomes, or even the nuclear envelope or plasma membrane.
Microsomal Mixed Function Oxidase System & Phase I Reactions
Many drug-metabolizing enzymes are located in the lipophilic membranes of the endoplasmic
reticulum of the liver and other tissues. When these lamellar membranes are isolated by
homogenization and fractionation of the cell, they re-form into vesicles called microsomes.
Microsomes retain most of the morphologic and functional characteristics of the intact membranes,
including the rough and smooth surface features of the rough (ribosome-studded) and smooth (no
ribosomes) endoplasmic reticulum. Whereas the rough microsomes tend to be dedicated to protein
synthesis, the smooth microsomes are relatively rich in enzymes responsible for oxidative drug
metabolism. In particular, they contain the important class of enzymes known as the mixed
function oxidases (MFOs), or monooxygenases. The activity of these enzymes requires both a
reducing agent (NADPH) and molecular oxygen; in a typical reaction, one molecule of oxygen is
consumed (reduced) per substrate molecule, with one oxygen atom appearing in the product and the
other in the form of water.
In this oxidation-reduction process, two microsomal enzymes play a key role. The first of these is a
flavoprotein, NADPH-cytochrome P450 reductase. One mole of this enzyme contains 1 mol each
of flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD). The second microsomal
enzyme is a hemoprotein called cytochrome P450 that serves as the terminal oxidase. In fact, the
microsomal membrane harbors multiple forms of this hemoprotein, and this multiplicity is
increased by repeated administration of exogenous chemicals (see below). The name cytochrome
P450 (abbreviated as CYP or P450) is derived from the spectral properties of this hemoprotein. In
its reduced (ferrous) form, it binds carbon monoxide to give a complex that absorbs light maximally
at 450 nm. The relative abundance of P450, compared with that of the reductase in the liver,
contributes to making P450 heme reduction a rate-limiting step in hepatic drug oxidations.
Microsomal drug oxidations require P450, P450 reductase, NADPH, and molecular oxygen. A
simplified scheme of the oxidative cycle is presented in Figure 4–3. Briefly, oxidized (Fe
3+
) P450
combines with a drug substrate to form a binary complex (step 1). NADPH donates an electron to
the flavoprotein reductase, which in turn reduces the oxidized P450-drug complex (step 2). A
second electron is introduced from NADPH via the same flavoprotein reductase, which serves to
reduce molecular oxygen and to form an "activated oxygen"- P450-substrate complex (step 3). This
complex in turn transfers activated oxygen to the drug substrate to form the oxidized product (step
4).
Figure 4–3.
Cytochrome P450 cycle in drug oxidations. (R-H, parent drug; R-OH, oxidized metabolite; e
-
,
electron.)
The potent oxidizing properties of this activated oxygen permit oxidation of a large number of
substrates. Substrate specificity is very low for this enzyme complex. High solubility in lipids is the
only common structural feature of the wide variety of structurally unrelated drugs and chemicals
that serve as substrates in this system (Table 4–1).
Table 4–1. Phase I Reactions.
Enzyme Induction
Some of these chemically dissimilar drug substrates, on repeated administration, "induce" P450 by
enhancing the rate of its synthesis or reducing its rate of degradation. Induction results in an
acceleration of substrate metabolism and usually in a decrease in the pharmacologic action of the
inducer and also of coadministered drugs. However, in the case of drugs metabolically transformed
to reactive metabolites, enzyme induction may exacerbate metabolite-mediated toxicity.
Various substrates appear to induce P450 isoforms having different molecular masses and
exhibiting different substrate specificities and immunochemical and spectral characteristics. The
isoforms that have been most extensively studied include CYP2B1 (formerly P450b), induced by
phenobarbital treatment; CYP1A1 (P
1
450 or P448), induced by polycyclic aromatic hydrocarbons
("PAHs" such as benzo[a]pyrene and 3-methylcholanthrene); CYPs3A (including CYPs 3A4 and
3A5, the major human liver isoforms) induced by glucocorticoids, macrolide antibiotics,
anticonvulsants, and some steroids. Chronic administration of isoniazid or ethanol induces a
different isoform, CYP2E1, that oxidizes ethanol and activates carcinogenic nitrosamines. The
VLDL-lowering drug clofibrate induces other distinct enzymes of the CYP4A class that are
responsible for -hydroxylation of several fatty acids, leukotrienes, and prostaglandins.
Environmental pollutants are also capable of inducing P450 enzymes. As noted above, exposure to
benzo[a]pyrene and other polycyclic aromatic hydrocarbons, which are present in tobacco smoke,
charcoal-broiled meat, and other organic pyrolysis products, is known to induce CYP1A enzymes
and to alter the rates of drug metabolism. Other environmental chemicals known to induce specific
P450s include the polychlorinated biphenyls (PCBs), which were used widely in industry as
insulating materials and plasticizers, and 2,3,7,8-tetrachlorodibenzo-p-dioxin (dioxin, TCDD), a
trace byproduct of the chemical synthesis of the defoliant 2,4,5-T (see Chapter 57: Introduction to
Toxicology: Occupational & Environmental).
Increased P450 synthesis requires enhanced transcription and translation. A cytoplasmic receptor
(termed AhR) for polycyclic aromatic hydrocarbons (eg, benzo[a]pyrene, dioxin) has been
identified, and the translocation of the inducer-receptor complex into the nucleus and subsequent
activation of regulatory elements of genes have been documented. A pregnane X receptor (PXR), a
member of the steroid-retinoid-thyroid hormone receptor family, has recently been shown to
mediate CYP3A induction by various chemicals (dexamethasone, rifampin) in the liver and
intestinal mucosa. A similar receptor, the constitutive androstane receptor (CAR) has been
identified for the phenobarbital class of inducers (Sueyoshi, 2001; Willson, 2002).
P450 enzymes may also be induced by "substrate stabilization," ie, decreased degradation, as is the
case with troleandomycin- or clotrimazole-mediated induction of CYP3A enzymes and the ethanol-
mediated induction of CYP2E1.
Enzyme Inhibition
Certain drug substrates may inhibit cytochrome P450 enzyme activity. Imidazole-containing drugs
such as cimetidine and ketoconazole bind tightly to the P450 heme iron and effectively reduce the
metabolism of endogenous substrates (testosterone) or other coadministered drugs through
competitive inhibition. However, macrolide antibiotics such as troleandomycin, erythromycin, and
other erythromycin derivatives are metabolized, apparently by CYP3A, to metabolites that complex
the cytochrome heme-iron and render it catalytically inactive. Another compound that acts through
this mechanism is the well-known inhibitor proadifen (SKF-525-A), which binds tightly to the
heme-iron and quasi-irreversibly inactivates the enzyme, thereby inhibiting the metabolism of
potential substrates.
Some substrates irreversibly inhibit P450s via covalent interaction of a metabolically generated
reactive intermediate that may react with the P450 apoprotein or heme moiety or even cause the
heme to fragment and irreversibly modify the apoprotein. The antibiotic chloramphenicol is
metabolized by CYP2B1 to a species that modifies its protein and thus also inactivates the enzyme.
A growing list of "suicide inhibitors"—inactivators that attack the heme or the protein moiety—
includes the steroids ethinyl estradiol, norethindrone, and spironolactone; the anesthetic agent
fluroxene; the barbiturate allobarbital; the analgesic sedatives allylisopropylacetylurea,
diethylpentenamide, and ethchlorvynol; the solvent carbon disulfide; and propylthiouracil. On the
other hand, the barbiturate secobarbital is found to inactivate CYP2B1 by modification of both its
heme and protein moieties.
Human Liver P450 Enzymes
Immunoblotting analyses—coupled with the use of relatively selective functional markers and
selective P450 inhibitors—have identified numerous P450 isoforms (CYPs 1A2, 2A6, 2B6, 2C9,
2C19, 2D6, 2E1, 3A4, 3A5, 4A11 and 7) in human liver microsomal preparations. Of these, CYPs
1A2, 2A6, 2C9, 2D6, 2E1, and 3A4 appear to be the major forms, accounting for approximately,
12, 4, 20, 4, 6, and 28 percent, respectively, of the total human liver P450 content. Together they are
responsible for catalyzing the bulk of the hepatic drug and xenobiotic metabolism (Table 4–2). It is
noteworthy that CYP3A4 alone is responsible for metabolism of more than 50% of the clinically
prescribed drugs metabolized by the liver. The involvement of individual P450s in the metabolism
of a given drug may be screened in vitro by means of selective functional markers, selective
chemical P450 inhibitors, and anti-P450 antibodies. In vivo, such screening may be accomplished
by means of relatively selective noninvasive markers, which include breath tests or urinary analyses
of specific metabolites after administration of a P450-selective substrate probe.
Table 4–2. Human Liver P450s (CYPs), and Some of the Drugs Metabolized (Substrates),
Inducers, and Drugs Used for Screening (Noninvasive Markers).
CYP Substrates Inducers Noninvasive Markers
1A2 Acetaminophen, antipyrine,
caffeine, clomipramine, phenacetin,
tamoxifen, theophylline, warfarin
Smoking, charcoal-
broiled foods,
cruciferous
vegetables,
omeprazole
Caffeine
2A6 Coumarin Coumarin
2B6 Artemisinin, bupropion,
cyclophosphamide, S-
mephobarbital, S-mephenytoin (N-
demethylation to nirvanol),
propofol, selegiline, sertraline
Phenobarbital,
cyclophosphamide
S-Mephenytoin
2C9 Hexobarbital, ibuprofen, phenytoin,
tolbutamide, trimethadione,
sulfaphenazole, S-warfarin,
Barbiturates, rifampin Tolbutamide,warfarin
ticrynafen
2C19 Diazepam, S-mephenytoin,
naproxen, nirvanol, omeprazole,
propranolol
Barbiturates, rifampin S-Mephenytoin
2D6 Bufuralol, bupranolol,
clomipramine, clozapine, codeine,
debrisoquin, dextromethorphan,
encainide, flecainide, fluoxetine,
guanoxan, haloperidol,
hydrocodone, 4-methoxy-
amphetamine, metoprolol,
mexiletine, oxycodone, paroxetine,
phenformin, propafenone,
propoxyphene, risperidone,
selegiline (deprenyl), sparteine,
thioridazine, timolol, tricyclic
antidepressants
None known Debrisoquin,dextromethorphan
2E1 Acetaminophen, chlorzoxazone,
enflurane, halothane, ethanol (a
minor pathway)
Ethanol, isoniazid Chlorzoxazone
3A4 Acetaminophen, alfentanil,
amiodarone, astemizole, cocaine,
cortisol, cyclosporine, dapsone,
diazepam, dihydroergotamine,
dihydropyridines, diltiazem, ethinyl
estradiol, gestodene, indinavir,
lidocaine, lovastatin, macrolides,
methadone, miconazole,
midazolam, mifepristone (RU 486),
paclitaxel, progesterone, quinidine,
rapamycin, ritonavir, saquinavir,
spironolactone, sulfamethoxazole,
sufentanil, tacrolimus, tamoxifen,
terfenadine, testosterone,
tetrahydro-cannabinol, triazolam,
troleandomycin, verapamil
Barbiturates,
carbamazepine,
macrolides,
glucocorticoids,
pioglitazone,
phenytoin, rifampin
Erythromycin, 6 -
hydroxycortisol
Phase II Reactions
Parent drugs or their phase I metabolites that contain suitable chemical groups often undergo
coupling or conjugation reactions with an endogenous substance to yield drug conjugates (Table 4–
3). In general, conjugates are polar molecules that are readily excreted and often inactive. Conjugate
formation involves high-energy intermediates and specific transfer enzymes. Such enzymes
(transferases) may be located in microsomes or in the cytosol. They catalyze the coupling of an
activated endogenous substance (such as the uridine 5'-diphosphate [UDP] derivative of glucuronic
acid) with a drug (or endogenous compound), or of an activated drug (such as the S-CoA derivative
of benzoic acid) with an endogenous substrate. Because the endogenous substrates originate in the
diet, nutrition plays a critical role in the regulation of drug conjugations.
Table 4–3. Phase II Reactions.
Type of
Conjugation
Endogenous
Reactant
Transferase
(Location)
Types of
Substrates
Examples
Glucuronidation UDP glucuronic
acid
UDP
glucuronosyl-
transferase
(microsomes)
Phenols, alcohols,
carboxylic acids,
hydroxylamines,
sulfonamides
Nitrophenol,
morphine,
acetaminophen,
diazepam, N-
hydroxydapsone,
sulfathiazole,
meprobamate,
digitoxin, digoxin
Acetylation Acetyl-CoA N-
Acetyltransferase
(cytosol)
Amines Sulfonamides,
isoniazid,
clonazepam,
dapsone, mescaline
Glutathione
conjugation
Glutathione GSH-S-transferase
(cytosol,
microsomes)
Epoxides, arene
oxides, nitro
groups,
hydroxylamines
Ethacrynic acid,
bromobenzene
Glycine
conjugation
Glycine Acyl-CoA
glycinetransferase
(mitochondria)
Acyl-CoA
derivatives of
carboxylic acids
Salicylic acid,
benzoic acid,
nicotinic acid,
cinnamic acid, cholic
acid, deoxycholic
acid
Sulfate
conjugation
Phosphoadenosyl
phosphosulfate
Sulfotransferase
(cytosol)
Phenols, alcohols,
aromatic amines
Estrone, aniline,
phenol, 3-hydroxy-
coumarin,
acetaminophen,
methyldopa
Methylation S-adenosyl-
methionine
Transmethylases
(cytosol)
Catecholamines,
phenols, amines
Dopamine,
epinephrine,
pyridine, histamine,
thiouracil
Epoxide hydrolase
(microsomes)
Arene oxides, cis-
disubstituted and
monosubstituted
oxiranes
Benzopyrene 7,8-
epoxide, styrene 1,2-
oxide,
carbamazepine
epoxide
Water
conjugation
Water
(cytosol) Alkene oxides,
fatty acid epoxides
Leukotriene A
4
Drug conjugations were once believed to represent terminal inactivation events and as such have
been viewed as "true detoxification" reactions. However, this concept must be modified, since it is
now known that certain conjugation reactions (acyl glucuronidation of nonsteroidal anti-
inflammatory drugs, O-sulfation of N-hydroxyacetylaminofluorene, and N-acetylation of isoniazid)
may lead to the formation of reactive species responsible for the hepatotoxicity of the drugs.
Furthermore, sulfation is known to activate the orally active prodrug minoxidil into a very
efficacious vasodilator.
Metabolism of Drugs to Toxic Products
It has become evident that metabolism of drugs and other foreign chemicals may not always be an
innocuous biochemical event leading to detoxification and elimination of the compound. Indeed,
several compounds have been shown to be metabolically transformed to reactive intermediates that
are toxic to various organs. Such toxic reactions may not be apparent at low levels of exposure to
parent compounds when alternative detoxification mechanisms are not yet overwhelmed or
compromised and the availability of endogenous detoxifying cosubstrates (glutathione [GSH],
glucuronic acid, sulfate) is not limited. However, when these resources are exhausted, the toxic
pathway may prevail, resulting in overt organ toxicity or carcinogenesis. The number of specific
examples of such drug-induced toxicity is expanding rapidly. An example is acetaminophen
(paracetamol)-induced hepatotoxicity (Figure 4–4). This analgesic antipyretic drug is quite safe in
therapeutic doses (1.2 g/d for an adult). It normally undergoes glucuronidation and sulfation to the
corresponding conjugates, which together comprise 95% of the total excreted metabolites. The
alternative P450-dependent glutathione conjugation pathway accounts for the remaining 5%. When
acetaminophen intake far exceeds therapeutic doses, the glucuronidation and sulfation pathways are
saturated, and the P450-dependent pathway becomes increasingly important. Little or no
hepatotoxicity results as long as glutathione is available for conjugation. However, with time,
hepatic glutathione is depleted faster than it can be regenerated, and a reactive and toxic metabolite
accumulates. In the absence of intracellular nucleophiles such as glutathione, this reactive
metabolite (N-acetylbenzoiminoquinone) reacts with nucleophilic groups of cellular proteins,
resulting in hepatotoxicity (Figure 4–4).
Figure 4–4.
Metabolism of acetaminophen (Ac) to hepatotoxic metabolites. (GSH, glutathione; GS,
glutathione moiety; Ac*, reactive intermediate.)
The chemical and toxicologic characterization of the electrophilic nature of the reactive
acetaminophen metabolite has led to the development of effective antidotes—cysteamine and N-
acetylcysteine. Administration of N-acetylcysteine (the safer of the two) within 8–16 hours
following acetaminophen overdosage has been shown to protect victims from fulminant
hepatotoxicity and death (see Chapter 59: Management of the Poisoned Patient).
Clinical Relevance of Drug Metabolism
The dose and the frequency of administration required to achieve effective therapeutic blood and
tissue levels vary in different patients because of individual differences in drug distribution and
rates of drug metabolism and elimination. These differences are determined by genetic factors and
nongenetic variables such as age, sex, liver size, liver function, circadian rhythm, body temperature,
and nutritional and environmental factors such as concomitant exposure to inducers or inhibitors of
drug metabolism. The discussion that follows summarizes the most important of these variables.
Individual Differences
Individual differences in metabolic rate depend on the nature of the drug itself. Thus, within the
same population, steady-state plasma levels may reflect a 30-fold variation in the metabolism of one
drug and only a twofold variation in the metabolism of another.
Genetic Factors
Genetic factors that influence enzyme levels account for some of these differences. Succinylcholine,
for example, is metabolized only half as rapidly in persons with genetically determined defects in
pseudocholinesterase as in persons with normally functioning pseudocholinesterase. Analogous
pharmacogenetic differences are seen in the acetylation of isoniazid (Figure 4–5) and the
hydroxylation of warfarin. The defect in slow acetylators (of isoniazid and similar amines) appears
to be caused by the synthesis of less of the enzyme rather than of an abnormal form of it. Inherited
as an autosomal recessive trait, the slow acetylator phenotype occurs in about 50% of blacks and
whites in the USA, more frequently in Europeans living in high northern latitudes, and much less
commonly in Asians and Inuits (Eskimos). Similarly, genetically determined defects in the
oxidative metabolism of debrisoquin, phenacetin, guanoxan, sparteine, phenformin, warfarin and
others have been reported (Table 4–4). The defects are apparently transmitted as autosomal
recessive traits and may be expressed at any one of the multiple metabolic transformations that a
chemical might undergo.
Figure 4–5.
Genetic polymorphism in drug metabolism. The graph shows the distribution of plasma
concentrations of isoniazid in 267 individuals 6 hours after an oral dose of 9.8 mg/kg. This
distribution is clearly bimodal. Individuals with a plasma concentration greater than 2.5 mg/mL at
6 hours are considered slow acetylators. (Redrawn, with permission, from Evans DAP, Manley
KA, McKusick VA: Genetic control of isoniazid metabolism in man. Br Med J 1960;2:485.)
Table 4–4. Some Examples of Genetic Polymorphisms in Drug Metabolism.
Defect Drug and Therapeutic Use Clinical Consequences
1
Oxidation Bufuralol ( -adrenoceptor blocker) Exacerbation of -blockade,
nausea
Oxidation Codeine (analgesic)
2
Reduced analgesia
Oxidation Debrisoquin (antihypertensive) Orthostatic hypotension
N-Demethylation Ethanol Facial flushing, cardiovascular
symptoms
Oxidation Ethanol Facial flushing, cardiovascular
symptoms
N-Acetylation Hydralazine (antihypertensive) Lupus erythematosus-like
syndrome
N-Acetylation Isoniazid (antitubercular) Peripheral neuropathy
Oxidation Mephenytoin (antiepileptic) Overdose toxicity
Thiopurine methyl-
transferase
Mercaptopurines (antileukemic) Myelotoxicity
Oxidation Nicotine (stimulant) Lesser addiction
Oxidation Nortriptyline (antidepressant) Toxicity
O-Demethylation Omeprazole (antiulcer) Increased therapeutic efficacy
Oxidation Sparteine Oxytocic symptoms
Ester hydrolysis Succinylcholine (neuromuscular
blocker)
Prolonged apnea
Oxidation S-warfarin (anticoagulant) Bleeding
Oxidation Tolbutamide (hypoglycemic) Cardiotoxicity
1
Observed or predictable.
2
Prodrug
Of the several recognized genetic variations of drug metabolism polymorphisms, three have been
particularly well characterized and afford some insight into possible underlying mechanisms. First
is the debrisoquin-sparteine oxidation type of polymorphism, which apparently occurs in 3–10% of
whites and is inherited as an autosomal recessive trait. In affected individuals, the CYP2D6-
dependent oxidations of debrisoquin and other drugs (see Table 4–2) are impaired. These defects in
oxidative drug metabolism are probably coinherited. The precise molecular basis for the defect
appears to be faulty expression of the P450 protein, resulting in little or no isoform-catalyzed drug
metabolism. More recently, however, another polymorphic genotype has been reported that results
in ultrarapid metabolism of relevant drugs due to the presence of 2D6 allelic variants with up to 13
gene copies in tandem. This genotype is most common in Ethiopians and Saudi Arabians,
populations that display it in up to one third of individuals. As a result, these subjects require
twofold to threefold higher daily doses of nortriptyline (a 2D6 substrate) to achieve therapeutic
plasma levels. Conversely, in these ultrarapidly metabolizing populations, the prodrug codeine
(another 2D6 substrate) is metabolized much faster to morphine, often resulting in undesirable side
effects of morphine such as severe abdominal pain.
A second well-studied genetic drug polymorphism involves the stereoselective aromatic (4)-
hydroxylation of the anticonvulsant mephenytoin, catalyzed by CYP2C19. This polymorphism,
which is also inherited as an autosomal recessive trait, occurs in 3–5% of Caucasians and 18–23%
of Japanese populations. It is genetically independent of the debrisoquin-sparteine polymorphism.
In normal "extensive metabolizers," (S)-mephenytoin is extensively hydroxylated by CYP2C19 at
the 4 position of the phenyl ring before its glucuronidation and rapid excretion in the urine, whereas
(R)-mephenytoin is slowly N-demethylated to nirvanol, an active metabolite. "Poor metabolizers,"
however, appear to totally lack the stereospecific (S)-mephenytoin hydroxylase activity, so both
(S)- and (R)-mephenytoin enantiomers are N-demethylated to nirvanol, which accumulates in much
higher concentrations. Thus, poor metabolizers of mephenytoin show signs of profound sedation
and ataxia after doses of the drug that are well tolerated by normal metabolizers. The molecular
basis for this defect is a single base pair mutation in exon 5 of the CYP2C19 gene that creates an
aberrant splice site, a correspondingly altered reading frame of the mRNA, and, finally, a truncated
nonfunctional protein. It is clinically important to recognize that the safety of a drug may be
severely reduced in individuals who are poor metabolizers.
The third genetic polymorphism recently characterized is that of CYP2C9. Two well-characterized
variants of this enzyme exist, each with amino acid mutations that result in altered metabolism:
CYP2C9*2 allele encodes an Arg144Cys mutation, exhibiting impaired functional interactions with
P450 reductase. The other allelic variant, CYP2C9*3, encodes an enzyme with an Ile359Leu
mutation that has lowered affinity for many substrates. Consequently, individuals displaying the
CYP2C9*3 phenotype have greatly reduced tolerance for the anticoagulant warfarin. The warfarin
clearance in CYP2C9*3-homozygous individuals is only 10% of normal values, and these people
can tolerate much smaller daily doses of the drug than those who are homozygous for the normal
wild type allele. These individuals also have a much higher risk of adverse effects with warfarin
(eg, bleeding) and with other CYP2C9 substrates such as phenytoin, losartan, tolbutamide, and
some NSAIDs.
Allelic variants of CYP3A4 have also been reported but their contribution to its well-known
interindividual variability in drug metabolism apparently is limited. On the other hand, the
expression of CYP3A5, another human liver isoform, is markedly polymorphic, ranging from 0% to
100% of the total hepatic CYP3A content. This CYP3A5 protein polymorphism is now known to
result from a single nucleotide polymorphism (SNIP) within intron 3 which enables normally
spliced CYP3A5 transcripts in 5% of Caucasians, 29% of Japanese, 27% of Chinese, 30% of
Koreans, and 73% of African Americans. Thus, it can significantly contribute to interindividual
differences in the metabolism of preferential CYP3A5 substrates such as midazolam.
Additional genetic polymorphisms in drug metabolism that are inherited independently from those
already described are being discovered. Studies of theophylline metabolism in monozygotic and
dizygotic twins that included pedigree analysis of various families have revealed that a distinct
polymorphism may exist for this drug and may be inherited as a recessive genetic trait. Genetic
drug metabolism polymorphisms also appear to occur for aminopyrine and carbocysteine
oxidations. Regularly updated information on human P450-polymorphisms is available at
http://www.imm.ki.se/CYPalleles/.
Although genetic polymorphisms in drug oxidations often involve specific P450 enzymes, such
genetic variations can occur at other sites. The recent descriptions of a polymorphism in the
oxidation of trimethylamine, believed to be metabolized largely by the flavin monooxygenase
(Ziegler's enzyme), suggest that genetic variants of other non-P450-dependent oxidative enzymes
may also contribute to such polymorphisms.
Diet & Environmental Factors
Diet and environmental factors also contribute to individual variations in drug metabolism.
Charcoal-broiled foods and cruciferous vegetables are known to induce CYP1A enzymes, whereas
grapefruit juice is known to inhibit the CYP3A metabolism of coadministered drug substrates.
Cigarette smokers metabolize some drugs more rapidly than nonsmokers because of enzyme
induction (see above). Industrial workers exposed to some pesticides metabolize certain drugs more
rapidly than nonexposed individuals. Such differences make it difficult to determine effective and
safe doses of drugs that have narrow therapeutic indices.
Age & Sex
Increased susceptibility to the pharmacologic or toxic activity of drugs has been reported in very
young and old patients compared with young adults (see Chapters 60 and 61). Although this may
reflect differences in absorption, distribution, and elimination, differences in drug metabolism also
plays a role. Slower metabolism could be due to reduced activity of metabolic enzymes or reduced
availability of essential endogenous cofactors.
Sex-dependent variations in drug metabolism have been well documented in rats but not in other
rodents. Young adult male rats metabolize drugs much faster than mature female rats or prepubertal
male rats. These differences in drug metabolism have been clearly associated with androgenic
hormones. Clinical reports suggest that similar sex-dependent differences in drug metabolism also
exist in humans for ethanol, propranolol, some benzodiazepines, estrogens, and salicylates.
Drug-Drug Interactions during Metabolism
Many substrates, by virtue of their relatively high lipophilicity, are retained not only at the active
site of the enzyme but remain nonspecifically bound to the lipid membrane of the endoplasmic
reticulum. In this state, they may induce microsomal enzymes; depending on the residual drug
levels at the active site, they also may competitively inhibit metabolism of a simultaneously
administered drug.
Enzyme-inducing drugs include various sedative-hypnotics, tranquilizers, anticonvulsants, and
insecticides (Table 4–5). Patients who routinely ingest barbiturates, other sedative-hypnotics, or
tranquilizers may require considerably higher doses of warfarin (an oral anticoagulant) to maintain
a prolonged prothrombin time. On the other hand, discontinuance of the sedative may result in
reduced metabolism of the anticoagulant and bleeding—a toxic effect of the ensuing enhanced
plasma levels of the anticoagulant. Similar interactions have been observed in individuals receiving
various combination drug regimens such as antipsychotics or sedatives with contraceptive agents,
sedatives with anticonvulsant drugs, and even alcohol with hypoglycemic drugs (tolbutamide).
Table 4–5. Partial List of Drugs That Enhance Drug Metabolism in Humans.
Inducer Drug Whose Metabolism Is Enhanced
Benzo[a]pyrene Theophylline
Chlorcyclizine Steroid hormones
Ethchlorvynol Warfarin
Glutethimide Antipyrine, glutethimide, warfarin
Griseofulvin Warfarin
Phenobarbital and
other barbiturates
1
Barbiturates, chloramphenicol, chlorpromazine, cortisol, coumarin
anticoagulants, desmethylimipramine, digitoxin, doxorubicin, estradiol,
phenylbutazone, phenytoin, quinine, testosterone
Phenylbutazone Aminopyrine, cortisol, digitoxin
Phenytoin Cortisol, dexamethasone, digitoxin, theophylline
Rifampin Coumarin anticoagulants, digitoxin, glucocorticoids,methadone,
metoprolol, oral contraceptives, prednisone, propranolol, quinidine
1
Secobarbital is an exception. See Table 4–6 and text.
It must also be noted that an inducer may enhance not only the metabolism of other drugs but also
its own metabolism. Thus, continued use of some drugs may result in a pharmacokinetic type of
tolerance—progressively reduced effectiveness due to enhancement of their own metabolism.
Conversely, simultaneous administration of two or more drugs may result in impaired elimination
of the more slowly metabolized drug and prolongation or potentiation of its pharmacologic effects
(Table 4–6). Both competitive substrate inhibition and irreversible substrate-mediated enzyme
inactivation may augment plasma drug levels and lead to toxic effects from drugs with narrow
therapeutic indices. Similarly, allopurinol both prolongs the duration and enhances the
chemotherapeutic action of mercaptopurine by competitive inhibition of xanthine oxidase.
Consequently, to avoid bone marrow toxicity, the dose of mercaptopurine is usually reduced in
patients receiving allopurinol. Cimetidine, a drug used in the treatment of peptic ulcer, has been
shown to potentiate the pharmacologic actions of anticoagulants and sedatives. The metabolism of
the sedative chlordiazepoxide has been shown to be inhibited by 63% after a single dose of
cimetidine; such effects are reversed within 48 hours after withdrawal of cimetidine.
Table 4–6. Partial List of Drugs That Inhibit Drug Metabolism in Humans.
Inhibitor Drug Whose Metabolism Is Inhibited
Allopurinol, chloramphenicol,
isoniazid
Antipyrine, dicumarol,probenecid, tolbutamide
Cimetidine Chlordiazepoxide, diazepam, warfarin, others
Dicumarol Phenytoin
Diethylpentenamide Diethylpentenamide
Disulfiram Antipyrine, ethanol, phenytoin, warfarin
Ethanol Chlordiazepoxide (?), diazepam (?), methanol
Grapefruit juice
1
Alprazolam, atorvastatin,cisapride, cyclosporine, midazolam,
triazolam
Ketoconazole Cyclosporine, astemizole, terfenadine
Nortriptyline Antipyrine
Oral contraceptives Antipyrine
Phenylbutazone Phenytoin, tolbutamide
Secobarbital Secobarbital
Troleandomycin Theophylline, methylprednisolone
1
Active components in grapefruit juice include furanocou-marins such as 6´, 7´-
dihydroxybergamottin (which is known to inactivate both intestinal and liver CYP3A4) as well as
other unknown components that inhibit P-glycoprotein-mediated intestinal drug efflux and
consequently further enhance the bioavailability of certain drugs such as cyclosporine.
Impaired metabolism may also result if a simultaneously administered drug irreversibly inactivates
a common metabolizing enzyme. These inhibitors, in the course of their metabolism by cytochrome
P450, inactivate the enzyme and result in impairment of their own metabolism and that of other
cosubstrates.
Interactions between Drugs & Endogenous Compounds
Various drugs require conjugation with endogenous substrates such as glutathione, glucuronic acid,
and sulfate for their inactivation. Consequently, different drugs may compete for the same
endogenous substrates, and the faster-reacting drug may effectively deplete endogenous substrate
levels and impair the metabolism of the slower-reacting drug. If the latter has a steep dose-response
curve or a narrow margin of safety, potentiation of its pharmacologic and toxic effects may result.
Diseases Affecting Drug Metabolism
Acute or chronic diseases that affect liver architecture or function markedly affect hepatic
metabolism of some drugs. Such conditions include alcoholic hepatitis, active or inactive alcoholic
cirrhosis, hemochromatosis, chronic active hepatitis, biliary cirrhosis, and acute viral or drug-
induced hepatitis. Depending on their severity, these conditions may significantly impair hepatic
drug-metabolizing enzymes, particularly microsomal oxidases, and thereby markedly affect drug
elimination. For example, the half-lives of chlordiazepoxide and diazepam in patients with liver
cirrhosis or acute viral hepatitis are greatly increased, with a corresponding prolongation of their
effects. Consequently, these drugs may cause coma in patients with liver disease when given in
ordinary doses.
Some drugs are metabolized so readily that even marked reduction in liver function does not
significantly prolong their action. However, cardiac disease, by limiting blood flow to the liver, may
impair disposition of those drugs whose metabolism is flow-limited (Table 4–7). These drugs are so
readily metabolized by the liver that hepatic clearance is essentially equal to liver blood flow.
Pulmonary disease may also affect drug metabolism as indicated by the impaired hydrolysis of
procainamide and procaine in patients with chronic respiratory insufficiency and the increased half-
life of antipyrine in patients with lung cancer. Impairment of enzyme activity or defective formation
of enzymes associated with heavy metal poisoning or porphyria also results in reduction of hepatic
drug metabolism.
Table 4–7. Rapidly Metabolized Drugs Whose Hepatic Clearance Is Blood Flow-Limited.
Alprenolol
Amitriptyline
Clomethiazole
Desipramine
Imipramine
Isoniazid
Labetalol
Lidocaine
Meperidine
Morphine
Pentazocine
Propoxyphene
Propranolol
Verapamil
Although the effects of endocrine dysfunction on drug metabolism have been well-explored in
experimental animal models, corresponding data for humans with endocrine disorders are scanty.
Thyroid dysfunction has been associated with altered metabolism of some drugs and of some
endogenous compounds as well. Hypothyroidism increases the half-life of antipyrine, digoxin,
methimazole, and practolol, whereas hyperthyroidism has the opposite effect. A few clinical studies
in diabetic patients indicate no apparent impairment of drug metabolism although impairment has
been noted in diabetic rats. Malfunctions of the pituitary, adrenal cortex, and gonads markedly
impair hepatic drug metabolism in rats. On the basis of these findings, it may be supposed that such
disorders could significantly affect drug metabolism in humans. However, until sufficient evidence
is obtained from clinical studies in patients, such extrapolations must be considered tentative.
Chapter 5. Basic & Clinical Evaluation of New Drugs
Basic & Clinical Evaluation of New Drugs: Introduction
*
Acknowledgment: I thank Wallace Dairman for comments and Kevin Abbey and Sema Ashkouri
for their research and administrative efforts.
New drug developments have revolutionized the practice of medicine, converting many once fatal
or debilitating diseases into almost routine therapeutic exercises. For example, deaths from
cardiovascular disease and stroke have decreased by more than 50% in the USA over the past 30
years. This decline is due—in part—to the discovery and increased use of antihypertensives,
cholesterol synthesis inhibitors, and drugs that prevent or dissolve blood clots. The process of drug
discovery and development has been greatly affected by investment in new technology and by
governmental support of medical research.
In most countries, the testing of therapeutic agents is now regulated by legislation and closely
monitored by governmental agencies. This chapter summarizes the process by which new drugs are
discovered, developed, and regulated. While the examples used reflect the experience in the USA,
the pathway of new drug development is generally the same worldwide.
One of the first steps in the development of a new drug is the discovery or synthesis of a potential
new drug molecule and correlating this molecule with an appropriate biologic target. Repeated
application of this approach leads to compounds with increased potency and selectivity (Figure 5–
1). By law, the safety and efficacy of drugs must be defined before they can be marketed. In
addition to in vitro studies, most of the biologic effects of the molecule must be characterized in
animals before human drug trials can be started. Human testing must then go forward in three
conventional phases before the drug can be considered for approval for general use. A fourth phase
of data gathering and safety monitoring follows after approval for general use.
Figure 5–1.
The development and testing process required to bring a drug to market in the USA. Some of the
requirements may be different for drugs used in life-threatening diseases.
Enormous costs, from $150 million to over $800 million, are involved in the research and
development of a single successful new drug. Thousands of compounds may be synthesized and
hundreds of thousands tested from existing libraries of compounds for each successful new drug
that reaches the market. It is primarily because of the economic investment and risks involved as
well as the need for multiple interdisciplinary technologies that most new drugs are developed in
pharmaceutical companies. At the same time, the incentives to succeed in drug development are
equally enormous. The worldwide market for ethical (prescription) pharmaceuticals in 2001 was
$364 billion. Moreover, it has been estimated that during the second half of the 20th century,
medications produced by the pharmaceutical industry saved more than 1.5 million lives and $140
billion in the costs of treatment for tuberculosis, poliomyelitis, coronary artery disease, and
cerebrovascular disease alone. In the USA, approximately 10% of the health care dollar is presently
spent on prescription drugs.
Drug Discovery
Most new drug candidates are launched through one or more of five approaches:
1. Identification or elucidation of a new drug target
2. Rational drug design based on an understanding of biologic mechanisms, drug receptor
structure, and drug structure
3. Chemical modification of a known molecule
4. Screening for biologic activity of large numbers of natural products; banks of previously
discovered chemical entities; and large libraries of peptides, nucleic acids, and other organic
molecules
5. Biotechnology and cloning using genes to produce larger peptides and proteins.
Moreover, automation, miniaturization and informatics have facilitated the process known
as "high through-put screening," which permits millions of assays per month.
Major attention is now being given to the discovery of entirely new targets for drug therapy. These
targets are emerging from studies with genomics, proteomics, and molecular pharmacology and are
expected to increase the number of useful biologic or disease targets ten-fold and thus be a positive
driver for new and improved drugs.
Drug Screening
Regardless of the source or the key idea leading to a drug candidate molecule, testing it involves a
sequence of experimentation and characterization called drug screening. A variety of biologic
assays at the molecular, cellular, organ system, and whole animal levels are used to define the
activity and selectivity of the drug. The type and number of initial screening tests depend on the
pharmacologic goal. Anti-infective drugs will generally be tested first against a variety of infectious
organisms, hypoglycemic drugs for their ability to lower blood sugar, etc. In addition, the molecule
will also be studied for a broad array of other actions to establish the mechanism of action and
selectivity of the drug. This has the advantage of demonstrating unsuspected toxic effects and
occasionally discloses a previously unsuspected therapeutic action. The selection of molecules for
further study is most efficiently conducted in animal models of human disease. Where good
predictive models exist (eg, hypertension or thrombotic disease), we generally have adequate drugs.
Good drugs are conspicuously lacking for diseases for which models are poor or not yet available,
eg, Alzheimer's disease.
Some of the studies performed during drug screening are listed in Table 5–1 and define the
pharmacologic profile of the drug. For example, a broad range of tests would be performed on a
drug designed to act as an antagonist at vascular -adrenoceptors for the treatment of hypertension.
At the molecular level, the compound would be screened for receptor binding affinity to cell
membranes containing receptors, possibly cloned human receptors, other receptors, and binding
sites on enzymes. Early studies would be done on liver cytochrome P450 enzymes to determine
whether the molecule of interest is likely to be a substrate or inhibitor of these enzymes.
Table 5–1. Pharmacologic Profile Tests.
Experimental Method or
Target Organ
Species or Tissue Route of
Administration
Measurement
Molecular
Receptor binding
(example: -
adrenoceptors)
Cell membrane
fractions from
organs or cultured
cells; cloned
receptors
In vitro Receptor affinity and
selectivity
Enzyme activity
(examples: tyrosine
Sympathetic nerves;
adrenal glands;
In vitro Enzyme inhibition and
selectivity
hydroxylase, dopamine-3-
hydroxylase, monoamine
oxidase)
purified enzymes
Cytochrome P450 Liver In vitro Enzyme inhibition; effects on
drug metabolism
Cellular
Cell function Cultured cells In vitro Evidence for receptor
activity—agonism or
antagonism (example: effects
on cyclic nucleotides)
Isolated tissue Blood vessels, heart,
lung, ileum (rat or
guinea pig)
In vitro Effects on vascular contraction
and relaxation; selectivity for
vascular receptors; effects on
other smooth muscles
Systems/disease models
Dog, cat
(anesthetized)
Parenteral Systolic-diastolic changes Blood pressure
Rat, hypertensive
(conscious)
Oral Antihypertensive effects
Dog (conscious) Oral Electrocardiography Cardiac effects
Dog (anesthetized) Parenteral Inotropic, chronotropic effects,
cardiac output, total peripheral
resistance
Peripheral autonomic
nervous system
Dog (anesthetized) Parenteral Effects on response to known
drugs and electrical
stimulation of central and
peripheral autonomic nerves
Respiratory effects Dog, guinea pig Parenteral Effects on respiratory rate and
amplitude, bronchial tone
Diuretic activity Dog Oral, parenteral Natriuresis, kaliuresis, water
diuresis, renal blood flow,
glomerular filtration rate
Gastrointestinal effects Rat Oral Gastrointestinal motility and
secretions
Circulating hormones,
cholesterol, blood sugar
Rat, dog Parenteral, oral Serum concentration
Blood coagulation Rabbit Oral Coagulation time, clot
retraction, prothrombin time
Central nervous system Mouse, rat Parenteral, oral Degree of sedation, muscle
relaxation, locomotor activity,
stimulation
Effects on cell function would be studied to determine the efficacy of the compound. Evidence
would be obtained about whether the drug is an agonist, partial agonist, or antagonist at receptors.