Drugs Acting on the Sympathetic Nervous System I

Sympathetic Nervous System

In the course of phylogeny an efficient
control system evolved that enabled the
functions of individual organs to be orchestrated
in increasingly complex life
forms and permitted rapid adaptation
to changing environmental conditions.
This regulatory system consists of the
CNS (brain plus spinal cord) and two
separate pathways for two-way communication
with peripheral organs, viz.,
the somatic and the autonomic nervous
systems. The somatic nervous system
comprising extero- and interoceptive
afferents, special sense organs, and motor
efferents, serves to perceive external
states and to target appropriate body
movement (sensory perception: threat
response: flight or attack). The autonomic
(vegetative) nervous system
(ANS), together with the endocrine
system, controls the milieu interieur. It
adjusts internal organ functions to the
changing needs of the organism. Neural
control permits very quick adaptation,
whereas the endocrine system provides
for a long-term regulation of functional
states. The ANS operates largely beyond
voluntary control; it functions autonomously.
Its central components reside
in the hypothalamus, brain stem, and
spinal cord. The ANS also participates in
the regulation of endocrine functions.
The ANS has sympathetic and
parasympathetic branches. Both are
made up of centrifugal (efferent) and
centripetal (afferent) nerves. In many
organs innervated by both branches, respective
activation of the sympathetic
and parasympathetic input evokes opposing
responses.
In various disease states (organ
malfunctions), drugs are employed with
the intention of normalizing susceptible
organ functions. To understand the biological
effects of substances capable of
inhibiting or exciting sympathetic or
parasympathetic nerves, one must first
envisage the functions subserved by the
sympathetic and parasympathetic divisions
(A, Responses to sympathetic activation).
In simplistic terms, activation
of the sympathetic division can be considered
a means by which the body
achieves a state of maximal work capacity
as required in fight or flight situations.
In both cases, there is a need for
vigorous activity of skeletal musculature.
To ensure adequate supply of oxygen
and nutrients, blood flow in skeletal
muscle is increased; cardiac rate and
contractility are enhanced, resulting in a
larger blood volume being pumped into
the circulation. Narrowing of splanchnic
blood vessels diverts blood into vascular
beds in muscle.
Because digestion of food in the intestinal
tract is dispensable and only
counterproductive, the propulsion of intestinal
contents is slowed to the extent
that peristalsis diminishes and sphincteric
tonus increases. However, in order
to increase nutrient supply to heart and
musculature, glucose from the liver and
free fatty acid from adipose tissue must
be released into the blood. The bronchi
are dilated, enabling tidal volume and
alveolar oxygen uptake to be increased.
Sweat glands are also innervated by
sympathetic fibers (wet palms due to
excitement); however, these are exceptional
as regards their neurotransmitter
(ACh).
Although the life styles of modern
humans are different from those of
hominid ancestors, biological functions
have remained the same.

Structure of the Sympathetic Nervous
System


The sympathetic preganglionic neurons
(first neurons) project from the intermediolateral
column of the spinal gray
matter to the paired paravertebral ganglionic
chain lying alongside the vertebral
column and to unpaired prevertebral
ganglia. These ganglia represent
sites of synaptic contact between preganglionic
axons (1st neurons) and
nerve cells (2nd neurons or sympathocytes)
that emit postganglionic axons
terminating on cells in various end organs.
In addition, there are preganglionic
neurons that project either to peripheral
ganglia in end organs or to the adrenal
medulla.
Sympathetic Transmitter Substances
Whereas acetylcholine
serves as the chemical transmitter at
ganglionic synapses between first and
second neurons, norepinephrine
(= noradrenaline) is the mediator at
synapses of the second neuron (B). This
second neuron does not synapse with
only a single cell in the effector organ;
rather, it branches out, each branch
making en passant contacts with several
cells. At these junctions the nerve axons
form enlargements (varicosities) resembling
beads on a string. Thus, excitation
of the neuron leads to activation of
a larger aggregate of effector cells, although
the action of released norepinephrine
may be confined to the region
of each junction. Excitation of preganglionic
neurons innervating the adrenal
medulla causes a liberation of acetylcholine.
This, in turn, elicits a secretion
of epinephrine (= adrenaline) into the
blood, by which it is distributed to body
tissues as a hormone (A).
Adrenergic Synapse
Within the varicosities, norepinephrine
is stored in small membrane-enclosed
vesicles (granules, 0.05 to 0.2 μm in diameter).
In the axoplasm, L-tyrosine is
converted via two intermediate steps to
dopamine, which is taken up into the
vesicles and there converted to norepinephrine
by dopamine-!-hydroxylase.
When stimulated electrically, the sympathetic
nerve discharges the contents
of part of its vesicles, including norepinephrine,
into the extracellular space.
Liberated norepinephrine reacts with
adrenoceptors located postjunctionally
on the membrane of effector cells or
prejunctionally on the membrane of
varicosities. Activation of presynaptic
"2-receptors inhibits norepinephrine
release. By this negative feedback, release
can be regulated.
The effect of released norepinephrine
wanes quickly, because approx.
90 % is actively transported back into
the axoplasm, then into storage vesicles
(neuronal re-uptake). Small portions of
norepinephrine are inactivated by the
enzyme catechol-O-methyltransferase
(COMT, present in the cytoplasm of
postjunctional cells, to yield normetanephrine),
and monoamine oxidase
(MAO, present in mitochondria of nerve
cells and postjunctional cells, to yield
3,4-dihydroxymandelic acid).
The liver is richly endowed with
COMT and MAO; it therefore contributes
significantly to the degradation of
circulating norepinephrine and epinephrine.
The end product of the combined
actions of MAO and COMT is vanillylmandelic
acid.


Adrenoceptor Subtypes and Catecholamine Actions
Adrenoceptors fall into three major
groups, designated !1, !2, and ", within
each of which further subtypes can be
distinguished pharmacologically. The
different adrenoceptors are differentially
distributed according to region and
tissue. Agonists at adrenoceptors (direct
sympathomimetics) mimic the actions
of the naturally occurring catecholamines,
norepinephrine and epinephrine,
and are used for various therapeutic
effects.
Smooth muscle effects. The opposing
effects on smooth muscle (A) of
!-and "-adrenoceptor activation are
due to differences in signal transduction.
This is exemplified by vascular
smooth muscle (A). !1-Receptor stimulation
leads to intracellular release of
Ca2+ via activation of the inositol trisphosphate
(IP3) pathway. In concert
with the protein calmodulin, Ca2+ can
activate myosin kinase, leading to a rise
in tonus via phosphorylation of the contractile
protein myosin. cAMP inhibits
activation of myosin kinase. Via the former
effector pathway, stimulation of !-
receptors results in vasoconstriction;
via the latter, "2-receptors mediate vasodilation,
particularly in skeletal muscle
— an effect that has little therapeutic
use.
Vasoconstriction. Local application of
!-sympathomimetics can be employed
in infiltration anesthesia or for
nasal decongestion (naphazoline, tetrahydrozoline,
xylometazoline).
Systemically administered epinephrine
is important in the treatment
of anaphylactic shock for combating hypotension.
Bronchodilation. "2-Adrenoceptor-
mediated bronchodilation (e.g., with
terbutaline, fenoterol, or salbutamol)
plays an essential part in the treatment
of bronchial asthma.
Tocolysis. The uterine relaxant effect
of "2-adrenoceptor agonists, such as
terbutaline or fenoterol, can be used to
prevent premature labor. Vasodilation
with a resultant drop in systemic blood
pressure results in reflex tachycardia,
which is also due in part to the "1-stimulant
action of these drugs.
Cardiostimulation. By stimulating
"1-receptors, hence activation of adenylatcyclase
(Ad-cyclase) and cAMP
production, catecholamines augment all
heart functions, including systolic force
(positive inotropism), velocity of shortening
(p. clinotropism), sinoatrial rate
(p. chronotropism), conduction velocity
(p. dromotropism), and excitability (p.
bathmotropism). In pacemaker fibers,
diastolic depolarization is hastened, so
that the firing threshold for the action
potential is reached sooner (positive
chronotropic effect, B). The cardiostimulant
effect of "-sympathomimetics
such as epinephrine is exploited in the
treatment of cardiac arrest. Use of "-
sympathomimetics in heart failure carries
the risk of cardiac arrhythmias.
Metabolic effects. "-Receptors mediate
increased conversion of glycogen to
glucose (glycogenolysis) in both liver
and skeletal muscle. From the liver, glucose
is released into the blood, In adipose
tissue, triglycerides are hydrolyzed
to fatty acids (lipolysis, mediated by "3-
receptors), which then enter the blood
(C). The metabolic effects of catecholamines
are not amenable to therapeutic
use.


Structure – Activity Relationships of
Sympathomimetics


Due to its equally high affinity for all !-
and "-receptors, epinephrine does not
permit selective activation of a particular
receptor subtype. Like most catecholamines,
it is also unsuitable for oral
administration (catechol is a trivial
name for o-hydroxyphenol). Norepinephrine
differs from epinephrine by its
high affinity for !-receptors and low affinity
for "2-receptors. In contrast, isoproterenol
has high affinity for "-receptors,
but virtually none for !-receptors
(A).
norepinephrine ! !, "1
epinephrine ! !, "1, "2
isoproterenol ! "1, "2
Knowledge of structure–activity
relationships has permitted the synthesis
of sympathomimetics that display
a high degree of selectivity at
adrenoceptor subtypes.
Direct-acting sympathomimetics
(i.e., adrenoceptor agonists) typically
share a phenylethylamine structure. The
side chain "-hydroxyl group confers affinity
for !- and "-receptors. Substitution
on the amino group reduces affinity
for !-receptors, but increases it for "-receptors
(exception: !-agonist phenylephrine),
with optimal affinity being
seen after the introduction of only one
isopropyl group. Increasing the bulk of
the amino substituent favors affinity for
"2-receptors (e.g., fenoterol, salbutamol).
Both hydroxyl groups on the aromatic
nucleus contribute to affinity;
high activity at !-receptors is associated
with hydroxyl groups at the 3 and 4 positions.
Affinity for "-receptors is preserved
in congeners bearing hydroxyl
groups at positions 3 and 5 (orciprenaline,
terbutaline, fenoterol).
The hydroxyl groups of catecholamines
are responsible for the very low
lipophilicity of these substances. Polarity
is increased at physiological pH due
to protonation of the amino group. Deletion
of one or all hydroxyl groups improves
membrane penetrability at the
intestinal mucosa-blood and the bloodbrain
barriers. Accordingly, these noncatecholamine
congeners can be given
orally and can exert CNS actions; however,
this structural change entails a loss
in affinity.
Absence of one or both aromatic
hydroxyl groups is associated with an
increase in indirect sympathomimetic
activity, denoting the ability of a substance
to release norepinephrine from
its neuronal stores without exerting an
agonist action at the adrenoceptor.
An altered position of aromatic hydroxyl
groups (e.g., in orciprenaline, fenoterol,
or terbutaline) or their substitution
(e.g., salbutamol) protects
against inactivation by COMT. Indroduction
of a small alkyl residue at
the carbon atom adjacent to the amino
group (ephedrine, methamphetamine)
confers resistance to degradation by
MAO, as does replacement on the
amino groups of the methyl residue
with larger substituents (e.g., ethyl in
etilefrine). Accordingly, the congeners
are less subject to presystemic inactivation.
Since structural requirements for
high affinity, on the one hand, and oral
applicability, on the other, do not
match, choosing a sympathomimetic is
a matter of compromise. If the high affinity
of epinephrine is to be exploited,
absorbability from the intestine must be
foregone (epinephrine, isoprenaline). If
good bioavailability with oral administration
is desired, losses in receptor affinity
must be accepted (etilefrine).


Indirect Sympathomimetics
Apart from receptors, adrenergic neurotransmission
involves mechanisms
for the active re-uptake and re-storage
of released amine, as well as enzymatic
breakdown by monoamine oxidase
(MAO). Norepinephrine (NE) displays
affinity for receptors, transport systems,
and degradative enzymes. Chemical alterations
of the catecholamine differentially
affect these properties and result
in substances with selective actions.
Inhibitors of MAO (A). The enzyme
is located predominantly on mitochondria,
and serves to scavenge axoplasmic
free NE. Inhibition of the enzyme causes
free NE concentrations to rise. Likewise,
dopamine catabolism is impaired, making
more of it available for NE synthesis.
Consequently, the amount of NE stored
in granular vesicles will increase, and
with it the amount of amine released
per nerve impulse.
In the CNS, inhibition of MAO affects
neuronal storage not only of NE
but also of dopamine and serotonin.
These mediators probably play significant
roles in CNS functions consistent
with the stimulant effects of MAO inhibitors
on mood and psychomotor drive
and their use as antidepressants in the
treatment of depression (A). Tranylcypromine
is used to treat particular forms
of depressive illness; as a covalently
bound suicide substrate, it causes longlasting
inhibition of both MAO isozymes,
(MAOA, MAOB). Moclobemide reversibly
inhibits MAOA and is also used
as an antidepressant. The MAOB inhibitor
selegiline (deprenyl) retards the catobolism
of dopamine, an effect used in
the treatment of parkinsonism.
Indirect sympathomimetics (B)
are agents that elevate the concentration
of NE at neuroeffector junctions,
because they either inhibit re-uptake
(cocaine), facilitate release, or slow
breakdown by MAO, or exert all three of
these effects (amphetamine, methamphetamine).
The effectiveness of such
indirect sympathomimetics diminishes
or disappears (tachyphylaxis) when vesicular
stores of NE close to the axolemma
are depleted.
Indirect sympathomimetics can
penetrate the blood-brain barrier and
evoke such CNS effects as a feeling of
well-being, enhanced physical activity
and mood (euphoria), and decreased
sense of hunger or fatigue. Subsequently,
the user may feel tired and depressed.
These after effects are partly
responsible for the urge to re-administer
the drug (high abuse potential). To
prevent their misuse, these substances
are subject to governmental regulations
(e.g., Food and Drugs Act: Canada; Controlled
Drugs Act: USA) restricting their
prescription and distribution.
When amphetamine-like substances
are misused to enhance athletic performance
(doping), there is a risk of dangerous
physical overexertion. Because
of the absence of a sense of fatigue, a
drugged athlete may be able to mobilize
ultimate energy reserves. In extreme
situations, cardiovascular failure may
result (B).
Closely related chemically to amphetamine
are the so-called appetite
suppressants or anorexiants, such as
fenfluramine, mazindole, and sibutramine.
These may also cause dependence
and their therapeutic value and safety
are questionable.


!-Sympathomimetics,
!-Sympatholytics
!-Sympathomimetics can be used
systemically in certain types of hypotension
and locally for nasal or conjunctival
decongestion or
as adjuncts in infiltration anesthesia
for the purpose of delaying the removal
of local anesthetic. With local
use, underperfusion of the vasoconstricted
area results in a lack of oxygen
(A). In the extreme case, local hypoxia
can lead to tissue necrosis. The appendages
(e.g., digits, toes, ears) are particularly
vulnerable in this regard, thus precluding
vasoconstrictor adjuncts in infiltration
anesthesia at these sites.
Vasoconstriction induced by an !-
sympathomimetic is followed by a
phase of enhanced blood flow (reactive
hyperemia, A). This reaction can be observed
after the application of !-sympathomimetics
(naphazoline, tetrahydrozoline,
xylometazoline) to the nasal mucosa.
Initially, vasoconstriction reduces
mucosal blood flow and, hence, capillary
pressure. Fluid exuded into the
interstitial space is drained through the
veins, thus shrinking the nasal mucosa.
Due to the reduced supply of fluid, secretion
of nasal mucus decreases. In coryza,
nasal patency is restored. However,
after vasoconstriction subsides, reactive
hyperemia causes renewed exudation
of plasma fluid into the interstitial
space, the nose is “stuffy” again, and the
patient feels a need to reapply decongestant.
In this way, a vicious cycle
threatens. Besides rebound congestion,
persistent use of a decongestant entails
the risk of atrophic damage caused by
prolonged hypoxia of the nasal mucosa.
!-Sympatholytics (B). The interaction
of norepinephrine with !-adrenoceptors
can be inhibited by !-sympatholytics
( !-adrenoceptor antagonists, !-
blockers). This inhibition can be put to
therapeutic use in antihypertensive
treatment (vasodilation ! peripheral
resistance ", blood pressure ").
The first !-sympatholytics blocked the
action of norepinephrine at both postand
prejunctional !-adrenoceptors
(non-selective !-blockers, e.g., phenoxybenzamine,
phentolamine).
Presynaptic !2-adrenoceptors function
like sensors that enable norepinephrine
concentration outside the
axolemma to be monitored, thus regulating
its release via a local feedback
mechanism. When presynaptic !2-receptors
are stimulated, further release
of norepinephrine is inhibited. Conversely,
their blockade leads to uncontrolled
release of norepinephrine with
an overt enhancement of sympathetic
effects at #1-adrenoceptor-mediated
myocardial neuroeffector junctions, resulting
in tachycardia and tachyarrhythmia.
Selective !-Sympatholytics
!-Blockers, such as prazosin, or the
longer-acting terazosin and doxazosin,
lack affinity for prejunctional !2-adrenoceptors.
They suppress activation of
!1-receptors without a concomitant enhancement
of norepinephrine release.
!1-Blockers may be used in hypertension.
Because they prevent
reflex vasoconstriction, they are likely
to cause postural hypotension with
pooling of blood in lower limb capacitance
veins during change from the supine
to the erect position (orthostatic
collapse: " venous return, " cardiac output,
fall in systemic pressure, " blood
supply to CNS, syncope).
In benign hyperplasia of the prostate,
!-blockers (terazosin, alfuzosin)
may serve to lower tonus of smooth
musculature in the prostatic region and
thereby facilitate micturition.




!-Sympatholytics (!-Blockers)
!-Sympatholytics are antagonists of
norepiphephrine and epinephrine at !-
adrenoceptors; they lack affinity for "-
receptors.
Therapeutic effects. !-Blockers
protect the heart from the oxygenwasting
effect of sympathetic inotropism
(p. 306) by blocking cardiac !-receptors;
thus, cardiac work can no longer
be augmented above basal levels (the
heart is “coasting”). This effect is utilized
prophylactically in angina pectoris
to prevent myocardial stress that could
trigger an ischemic attack (p. 308, 310).
!-Blockers also serve to lower cardiac
rate (sinus tachycardia, p. 134) and elevated
blood pressure due to high cardiac
output (p. 312). The mechanism underlying
their antihypertensive action via
reduction of peripheral resistance is unclear.
Applied topically to the eye, !-
blockers are used in the management of
glaucoma; they lower production of
aqueous humor without affecting its
drainage.
Undesired effects. The hazards of
treatment with !-blockers become apparent
particularly when continuous
activation of !-receptors is needed in
order to maintain the function of an organ.
Congestive heart failure: In myocardial
insufficiency, the heart depends on
a tonic sympathetic drive to maintain
adequate cardiac output. Sympathetic
activation gives rise to an increase in
heart rate and systolic muscle tension,
enabling cardiac output to be restored
to a level comparable to that in a
healthy subject. When sympathetic
drive is eliminated during !-receptor
blockade, stroke volume and cardiac
rate decline, a latent myocardial insufficiency
is unmasked, and overt insufficiency
is exacerbated (A).
On the other hand, clinical evidence
suggests that !-blockers produce favorable
effects in certain forms of congestive
heart failure (idiopathic dilated cardiomyopathy).
Bradycardia, A-V block: Elimination
of sympathetic drive can lead to a
marked fall in cardiac rate as well as to
disorders of impulse conduction from
the atria to the ventricles.
Bronchial asthma: Increased sympathetic
activity prevents bronchospasm
in patients disposed to paroxysmal
constriction of the bronchial tree
(bronchial asthma, bronchitis in smokers).
In this condition, !2-receptor
blockade will precipitate acute respiratory
distress (B).
Hypoglycemia in diabetes mellitus:
When treatment with insulin or oral hypoglycemics
in the diabetic patient lowers
blood glucose below a critical level,
epinephrine is released, which then
stimulates hepatic glucose release via
activation of !2-receptors. !-Blockers
suppress this counter-regulation; in addition,
they mask other epinephrinemediated
warning signs of imminent
hypoglycemia, such as tachycardia and
anxiety, thereby enhancing the risk of
hypoglycemic shock.
Altered vascular responses: When
!2-receptors are blocked, the vasodilating
effect of epinephrine is abolished,
leaving the "-receptor-mediated vasoconstriction
unaffected: peripheral
blood flow # – “cold hands and feet”.
!-Blockers exert an “anxiolytic“
action that may be due to the suppression
of somatic responses (palpitations,
trembling) to epinephrine release that
is induced by emotional stress; in turn,
these would exacerbate “anxiety” or
“stage fright”. Because alertness is not
impaired by !-blockers, these agents are
occasionally taken by orators and musicians
before a major performance (C).
Stage fright, however, is not a disease
requiring drug therapy.



Types of !-Blockers
The basic structure shared by most !-
sympatholytics is the side chain of !-
sympathomimetics (cf. isoproterenol
with the !-blockers propranolol, pindolol,
atenolol). As a rule, this basic structure
is linked to an aromatic nucleus by
a methylene and oxygen bridge. The
side chain C-atom bearing the hydroxyl
group forms the chiral center. With
some exceptions (e.g., timolol, penbutolol),
all !-sympatholytics are brought as
racemates into the market (p. 62).
Compared with the dextrorotatory
form, the levorotatory enantiomer possesses
a greater than 100-fold higher affinity
for the !-receptor and is, therefore,
practically alone in contributing to
the !-blocking effect of the racemate.
The side chain and substituents on the
amino group critically affect affinity for
!-receptors, whereas the aromatic nucleus
determines whether the compound
possess intrinsic sympathomimetic
activity (ISA), that is, acts as a
partial agonist (p. 60) or partial antagonist.
In the presence of a partial agonist
(e.g., pindolol), the ability of a full agonist
(e.g., isoprenaline) to elicit a maximal
effect would be attenuated, because
binding of the full agonist is impeded.
However, the !-receptor at which such
partial agonism can be shown appears
to be atypical (!3 or !4 subtype). Whether
ISA confers a therapeutic advantage
on a !-blocker remains an open question.
As cationic amphiphilic drugs, !-
blockers can exert a membrane-stabilizing
effect, as evidenced by the ability
of the more lipophilic congeners to inhibit
Na+-channel function and impulse
conduction in cardiac tissues. At the
usual therapeutic dosage, the high concentration
required for these effects will
not be reached.
Some !-sympatholytics possess
higher affinity for cardiac !1-receptors
than for !2-receptors and thus display
cardioselectivity (e.g., metoprolol, acebutolol,
bisoprolol). None of these
blockers is sufficiently selective to permit
its use in patients with bronchial
asthma or diabetes mellitus (p. 92).
The chemical structure of !-blockers
also determines their pharmacokinetic
properties. Except for hydrophilic
representatives (atenolol), !-sympatholytics
are completely absorbed from the
intestines and subsequently undergo
presystemic elimination to a major extent
(A).
All the above differences are of
little clinical importance. The abundance
of commercially available congeners
would thus appear all the more curious
(B). Propranolol was the first !-blocker
to be introduced into therapy in 1965.
Thirty-five years later, about 20 different
congeners are being marketed in different
countries. This questionable development
unfortunately is typical of any
drug group that has major therapeutic
relevance, in addition to a relatively
fixed active structure. Variation of the
molecule will create a new patentable
chemical, not necessarily a drug with a
novel action. Moreover, a drug no longer
protected by patent is offered as a generic
by different manufacturers under dozens
of different proprietary names.
Propranolol alone has been marketed by
13 manufacturers under 11 different
names.



Antiadrenergics
Antiadrenergics are drugs capable of
lowering transmitter output from sympathetic
neurons, i.e., “sympathetic
tone”. Their action is hypotensive (indication:
hypertension, p. 312); however,
being poorly tolerated, they enjoy only
limited therapeutic use.
Clonidine is an !2-agonist whose
high lipophilicity (dichlorophenyl ring)
permits rapid penetration through the
blood-brain barrier. The activation of
postsynaptic !2-receptors dampens the
activity of vasomotor neurons in the
medulla oblongata, resulting in a resetting
of systemic arterial pressure at a
lower level. In addition, activation of
presynaptic !2-receptors in the periphery
(pp. 82, 90) leads to a decreased release
of both norepinephrine (NE) and
acetylcholine.
Side effects. Lassitude, dry mouth;
rebound hypertension after abrupt cessation
of clonidine therapy.
Methyldopa (dopa = dihydroxyphenylalanine),
as an amino acid, is
transported across the blood-brain barrier,
decarboxylated in the brain to !-
methyldopamine, and then hydroxylated
to !-methyl-NE. The decarboxylation
of methyldopa competes for a portion of
the available enzymatic activity, so that
the rate of conversion of L-dopa to NE
(via dopamine) is decreased. The false
transmitter !-methyl-NE can be stored;
however, unlike the endogenous mediator,
it has a higher affinity for !2- than
for !1-receptors and therefore produces
effects similar to those of clonidine. The
same events take place in peripheral adrenergic
neurons.
Adverse effects. Fatigue, orthostatic
hypotension, extrapyramidal Parkinson-
like symptoms (p. 88), cutaneous
reactions, hepatic damage, immune-hemolytic
anemia.
Reserpine, an alkaloid from the
Rauwolfia plant, abolishes the vesicular
storage of biogenic amines (NE, dopamine
= DA, serotonin = 5-HT) by inhibiting
an ATPase required for the vesicular
amine pump. The amount of NE released
per nerve impulse is decreased.
To a lesser degree, release of epinephrine
from the adrenal medulla is also
impaired. At higher doses, there is irreversible
damage to storage vesicles
(“pharmacological sympathectomy”),
days to weeks being required for their
resynthesis. Reserpine readily enters
the brain, where it also impairs vesicular
storage of biogenic amines.
Adverse effects. Disorders of extrapyramidal
motor function with development
of pseudo-Parkinsonism (p. 88),
sedation, depression, stuffy nose, impaired
libido, and impotence; increased
appetite. These adverse effects have
rendered the drug practically obsolete.
Guanethidine possesses high affinity
for the axolemmal and vesicular
amine transporters. It is stored instead
of NE, but is unable to mimic the functions
of the latter. In addition, it stabilizes
the axonal membrane, thereby impeding
the propagation of impulses into
the sympathetic nerve terminals. Storage
and release of epinephrine from the
adrenal medulla are not affected, owing
to the absence of a re-uptake process.
The drug does not cross the blood-brain
barrier.
Adverse effects. Cardiovascular crises
are a possible risk: emotional stress
of the patient may cause sympathoadrenal
activation with epinephrine release.
The resulting rise in blood pressure
can be all the more marked because
persistent depression of sympathetic
nerve activity induces supersensitivity
of effector organs to circulating
catecholamines.

Drug-independent Effects II

Homeopathy is an alternative
method of therapy, developed in the
1800s by Samuel Hahnemann. His idea
was this: when given in normal (allopathic)
dosage, a drug (in the sense of
medicament) will produce a constellation
of symptoms; however, in a patient
whose disease symptoms resemble just
this mosaic of symptoms, the same drug
(simile principle) would effect a cure
when given in a very low dosage (“potentiation”).
The body’s self-healing
powers were to be properly activated
only by minimal doses of the medicinal
substance.
The homeopath’s task is not to diagnose
the causes of morbidity, but to
find the drug with a “symptom profile”
most closely resembling that of the
patient’s illness. This drug is then applied
in very high dilution.
A direct action or effect on body
functions cannot be demonstrated for
homeopathic medicines. Therapeutic
success is due to the suggestive powers
of the homeopath and the expectancy of
the patient. When an illness is strongly
influenced by emotional (psychic) factors
and cannot be treated well by allopathic
means, a case can be made in favor
of exploiting suggestion as a therapeutic
tool. Homeopathy is one of several
possible methods of doing so.

Drug-independent Effects I

Placebo
A placebo is a dosage form devoid of an
active ingredient, a dummy medication.
Administration of a placebo may elicit
the desired effect (relief of symptoms)
or undesired effects that reflect a
change in the patient’s psychological
situation brought about by the therapeutic
setting.
Physicians may consciously or unconsciously
communicate to the patient
whether or not they are concerned
about the patient’s problem, or certain
about the diagnosis and about the value
of prescribed therapeutic measures. In
the care of a physician who projects
personal warmth, competence, and confidence,
the patient in turn feels comfortable
and less anxious and optimistically
anticipates recovery.
The physical condition determines
the psychic disposition and vice versa.
Consider gravely wounded combatants
in war, oblivious to their injuries while
fighting to survive, only to experience
severe pain in the safety of the field hospital,
or the patient with a peptic ulcer
caused by emotional stress.
Clinical trials. In the individual
case, it may be impossible to decide
whether therapeutic success is attributable
to the drug or to the therapeutic
situation. What is therefore required is a
comparison of the effects of a drug and
of a placebo in matched groups of patients
by means of statistical procedures,
i.e., a placebo-controlled trial. A
prospective trial is planned in advance, a
retrospective (case-control) study follows
patients backwards in time. Patients
are randomly allotted to two
groups, namely, the placebo and the active
or test drug group. In a double-blind
trial, neither the patients nor the treating
physicians know which patient is
given drug and which placebo. Finally, a
switch from drug to placebo and vice
versa can be made in a successive phase
of treatment, the cross-over trial. In this
fashion, drug vs. placebo comparisons
can be made not only between two patient
groups, but also within either
group itself.

Adverse Drug Effects V

Drug Toxicity in Pregnancy and
Lactation
Drugs taken by the mother can be
passed on transplacentally or via breast
milk and adversely affect the unborn or
the neonate.
Pregnancy
Limb malformations induced by the
hypnotic, thalidomide, first focused attention
on the potential of drugs to
cause malformations (teratogenicity).
Drug effects on the unborn fall into two
basic categories:
1. Predictable effects that derive from
the known pharmacological drug
properties. Examples are: masculinization
of the female fetus by androgenic
hormones; brain hemorrhage
due to oral anticoagulants; bradycardia
due to !-blockers.
2. Effects that specifically affect the developing
organism and that cannot
be predicted on the basis of the
known pharmacological activity profile.
In assessing the risks attending
drug use during pregnancy, the following
points have to be considered:
a) Time of drug use. The possible sequelae
of exposure to a drug depend on
the stage of fetal development, as
shown in A. Thus, the hazard posed
by a drug with a specific action is limited
in time, as illustrated by the tetracyclines,
which produce effects on
teeth and bones only after the third
month of gestation, when mineralization
begins.
b) Transplacental passage. Most drugs
can pass in the placenta from the maternal
into the fetal circulation. The
fused cells of the syncytiotrophoblast
form the major diffusion barrier.
They possess a higher permeability to
drugs than is suggested by the term
“placental barrier”.
c) Teratogenicity. Statistical risk estimates
are available for familiar, frequently
used drugs. For many drugs,
teratogenic potency cannot be demonstrated;
however, in the case of
novel drugs it is usually not yet possible
to define their teratogenic hazard.
Drugs with established human teratogenicity
include derivatives of vitamin
A (etretinate, isotretinoin [used
internally in skin diseases]), and oral
anticoagulants. A peculiar type of damage
results from the synthetic estrogenic
agent, diethylstilbestrol, following its
use during pregnancy; daughters of
treated mothers have an increased incidence
of cervical and vaginal carcinoma
at the age of approx. 20.
In assessing the risk: benefit ratio, it is
also necessary to consider the benefit
for the child resulting from adequate
therapeutic treatment of its mother. For
instance, therapy with antiepileptic
drugs is indispensable, because untreated
epilepsy endangers the infant at least
as much as does administration of anticonvulsants.
Lactation
Drugs present in the maternal organism
can be secreted in breast milk and thus
be ingested by the infant. Evaluation of
risk should be based on factors listed in
B. In case of doubt, potential danger to
the infant can be averted only by weaning.

Adverse Drug Effects IV

Type 2, cytotoxic reaction. Drugantibody
(IgG) complexes adhere to the
surface of blood cells, where either circulating
drug molecules or complexes already
formed in blood accumulate.
These complexes mediate the activation
of complement, a family of proteins that
circulate in the blood in an inactive
form, but can be activated in a cascadelike
succession by an appropriate stimulus.
“Activated complement” normally
directed against microorganisms, can
destroy the cell membranes and thereby
cause cell death; it also promotes phagocytosis,
attracts neutrophil granulocytes
(chemotaxis), and stimulates other
inflammatory responses. Activation
of complement on blood cells results in
their destruction, evidenced by hemolytic
anemia, agranulocytosis, and
thrombocytopenia.
Type 3, immune complex vasculitis
(serum sickness, Arthus reaction).
Drug-antibody complexes precipitate on
vascular walls, complement is activated,
and an inflammatory reaction is triggered.
Attracted neutrophils, in a futile
attempt to phagocytose the complexes,
liberate lysosomal enzymes that damage
the vascular walls
. Symptoms may include fever,
exanthema, swelling of lymph
nodes, arthritis, nephritis, and neuropathy.
Type 4, contact dermatitis. A cutaneously
applied drug is bound to the
surface of T-lymphocytes directed specifically
against it. The lymphocytes release
signal molecules (lymphokines)
into their vicinity that activate macrophages
and provoke an inflammatory
reaction.

Adverse Drug Effects III

Drug Allergy
The immune system normally functions
to rid the organism of invading foreign
particles, such as bacteria. Immune responses
can occur without appropriate
cause or with exaggerated intensity and
may harm the organism, for instance,
when allergic reactions are caused by
drugs (active ingredient or pharmaceutical
excipients). Only a few drugs, e.g.
(heterologous) proteins, have a molecular
mass (> 10,000) large enough to act
as effective antigens or immunogens,
capable by themselves of initiating an
immune response. Most drugs or their
metabolites (so-called haptens) must
first be converted to an antigen by linkage
to a body protein. In the case of penicillin
G, a cleavage product (penicilloyl
residue) probably undergoes covalent
binding to protein. During initial contact
with the drug, the immune system
is sensitized: antigen-specific lymphocytes
of the T-type and B-type (antibody
formation) proliferate in lymphatic tissue
and some of them remain as socalled
memory cells. Usually, these processes
remain clinically silent. During
the second contact, antibodies are already
present and memory cells proliferate
rapidly. A detectable immune response,
the allergic reaction, occurs.
This can be of severe intensity, even at a
low dose of the antigen. Four types of
reactions can be distinguished:
Type 1, anaphylactic reaction.
Drug-specific antibodies of the IgE type
combine via their Fc moiety with receptors
on the surface of mast cells. Binding
of the drug provides the stimulus for the
release of histamine and other mediators.
In the most severe form, a lifethreatening
anaphylactic shock develops,
accompanied by hypotension,
bronchospasm (asthma attack), laryngeal
edema, urticaria, stimulation of gut
musculature, and spontaneous bowel
movements .

Adverse Drug Effects II

Increased Sensitivity . If certain
body functions develop hyperreactivity,
unwanted effects can occur even at normal
dose levels. Increased sensitivity of
the respiratory center to morphine is
found in patients with chronic lung disease,
in neonates, or during concurrent
exposure to other respiratory depressant
agents. The DRC is shifted to the left
and a smaller dose of morphine is sufficient
to paralyze respiration. Genetic
anomalies of metabolism may also lead
to hypersensitivity. Thus, several drugs
(aspirin, antimalarials, etc.) can provoke
premature breakdown of red blood cells
(hemolysis) in subjects with a glucose-
6-phosphate dehydrogenase deficiency.
The discipline of pharmacogenetics deals
with the importance of the genotype for
reactions to drugs.
The above forms of hypersensitivity
must be distinguished from allergies involving
the immune system .
Lack of selectivity . Despite appropriate
dosing and normal sensitivity,
undesired effects can occur because the
drug does not specifically act on the targeted
(diseased) tissue or organ. For instance,
the anticholinergic, atropine, is
bound only to acetylcholine receptors of
the muscarinic type; however, these are
present in many different organs.
Moreover, the neuroleptic, chlorpromazine,
formerly used as a neuroleptic,
is able to interact with several
different receptor types. Thus, its action
is neither organ-specific nor receptorspecific.
The consequences of lack of selectivity
can often be avoided if the drug
does not require the blood route to
reach the target organ, but is, instead,
applied locally, as in the administration
of parasympatholytics in the form of eye
drops or in an aerosol for inhalation.
With every drug use, unwanted effects
must be taken into account. Before
prescribing a drug, the physician should
therefore assess the risk: benefit ratio.

Adverse Drug Effects I

Adverse Drug Effects
The desired (or intended) principal effect
of any drug is to modify body function
in such a manner as to alleviate
symptoms caused by the patient’s illness.
In addition, a drug may also cause
unwanted effects that can be grouped
into minor or “side” effects and major or
adverse effects. These, in turn, may give
rise to complaints or illness, or may
even cause death.
Causes of adverse effects: overdosage
. The drug is administered in
a higher dose than is required for the
principal effect; this directly or indirectly
affects other body functions. For instances,
morphine , given in the
appropriate dose, affords excellent pain
relief by influencing nociceptive pathways
in the CNS. In excessive doses, it
inhibits the respiratory center and
makes apnea imminent. The dose dependence
of both effects can be graphed
in the form of dose-response curves
(DRC). The distance between both DRCs
indicates the difference between the
therapeutic and toxic doses. This margin
of safety indicates the risk of toxicity
when standard doses are exceeded.
“The dose alone makes the poison”
(Paracelsus). This holds true for both
medicines and environmental poisons.
No substance as such is toxic! In order to
assess the risk of toxicity, knowledge is
required of) the effective dose during
exposure) the dose level at which
damage is likely to occur) the duration
of exposure.

Drug-Receptor Interaction IX

Time Course of Plasma Concentration
and Effect
After the administration of a drug, its
concentration in plasma rises, reaches a
peak, and then declines gradually to the
starting level, due to the processes of
distribution and elimination.
Plasma concentration at a given point in
time depends on the dose administered.
Many drugs exhibit a linear relationship
between plasma concentration and
dose within the therapeutic range
(dose-linear kinetic ; note different
scales on ordinate). However, the
same does not apply to drugs whose
elimination processes are already sufficiently
activated at therapeutic plasma
levels so as to preclude further proportional
increases in the rate of elimination
when the concentration is increased
further. Under these conditions,
a smaller proportion of the dose administered
is eliminated per unit of time.
The time course of the effect and of
the concentration in plasma are not
identical, because the concentrationeffect
relationships obeys a hyperbolic
function. This means
that the time course of the effect exhibits
dose dependence also in the presence
of dose-linear kinetics .
In the lower dose range (example
1), the plasma level passes through a
concentration range (0 ! 0.9) in which
the concentration effect relationship is
quasi-linear. The respective time courses
of plasma concentration and effect
are very similar.
However, if a high dose (100) is applied,
there is an extended period of time during
which the plasma level will remain
in a concentration range (between 90
and 20) in which a change in concentration
does not cause a change in the size
of the effect. Thus, at high doses (100),
the time-effect curve exhibits a kind of
plateau. The effect declines only when
the plasma level has returned (below
20) into the range where a change in
plasma level causes a change in the intensity
of the effect.
The dose dependence of the time
course of the drug effect is exploited
when the duration of the effect is to be
prolonged by administration of a dose
in excess of that required for the effect.
This is done in the case of penicillin G
, when a dosing interval of 8 h is
being recommended, although the drug
is eliminated with a half-life of 30 min.
This procedure is, of course, feasible only
if supramaximal dosing is not associated
with toxic effects.
Futhermore it follows that a nearly
constant effect can be achieved, although
the plasma level may fluctuate
greatly during the interval between
doses.
The hyperbolic relationship be
tween plasma concentration and effect
explains why the time course of the effect,
unlike that of the plasma concentration,
cannot be described in terms of
a simple exponential function. A halflife
can be given for the processes of
drug absorption and elimination, hence
for the change in plasma levels, but generally
not for the onset or decline of
the effect.

Drug-Receptor Interaction VIII

Mode of Operation of G-Protein-
Coupled Receptors
Signal transduction at G-protein-coupled
receptors uses essentially the same
basic mechanisms . Agonist binding
to the receptor leads to a change in receptor
protein conformation. This
change propagates to the G-protein: the
!-subunit exchanges GDP for GTP, then
dissociates from the two other subunits,
associates with an effector protein, and
alters its functional state. The !-subunit
slowly hydrolyzes bound GTP to GDP.
G!-GDP has no affinity for the effector
protein and reassociates with the " and
subunits . G-proteins can undergo
lateral diffusion in the membrane; they
are not assigned to individual receptor
proteins. However, a relation exists
between receptor types and G-protein
types . Furthermore, the !-subunits
of individual G-proteins are distinct in
terms of their affinity for different effector
proteins, as well as the kind of influence
exerted on the effector protein. G-
GTP of the GS-protein stimulates adenylate
cyclase, whereas G-GTP of the Giprotein
is inhibitory. The G-proteincoupled
receptor family includes muscarinic
cholinoceptors, adrenoceptors
for norepinephrine and epinephrine, receptors
for dopamine, histamine, serotonin,
glutamate, GABA, morphine, prostaglandins,
leukotrienes, and many other
mediators and hormones.
Major effector proteins for G-protein-
coupled receptors include adenylate
cyclase (ATP ! intracellular messenger
cAMP), phospholipase C (phosphatidylinositol
intracellular messengers
inositol trisphosphate and diacylglycerol),
as well as ion channel
proteins. Numerous cell functions are
regulated by cellular cAMP concentration,
because cAMP enhances activity of
protein kinase A, which catalyzes the
transfer of phosphate groups onto functional
proteins. Elevation of cAMP levels
inter alia leads to relaxation of smooth
muscle tonus and enhanced contractility
of cardiac muscle, as well as increased
glycogenolysis and lipolysis .
Phosphorylation of cardiac calcium-
channel proteins increases the
probability of channel opening during
membrane depolarization. It should be
noted that cAMP is inactivated by phosphodiesterase.
Inhibitors of this enzyme
elevate intracellular cAMP concentration
and elicit effects resembling those
of epinephrine.
The receptor protein itself may
undergo phosphorylation, with a resultant
loss of its ability to activate the associated
G-protein. This is one of the
mechanisms that contributes to a decrease
in sensitivity of a cell during prolonged
receptor stimulation by an agonist
(desensitization).
Activation of phospholipase C leads
to cleavage of the membrane phospholipid
phosphatidylinositol-4,5 bisphosphate
into inositol trisphosphate (IP3)
and diacylglycerol (DAG). IP3 promotes
release of Ca2+ from storage organelles,
whereby contraction of smooth muscle
cells, breakdown of glycogen, or exocytosis
may be initiated. Diacylglycerol
stimulates protein kinase C, which
phosphorylates certain serine- or threonine-
containing enzymes.
The subunit of some G-proteins
may induce opening of a channel protein.
In this manner, K+ channels can be
activated (e.g., ACh effect on sinus node,
; opioid action on neural impulse
transmission).

Drug-Receptor Interaction VII

The insulin receptor protein represents
a ligand-operated enzyme , a
catalytic receptor. When insulin binds
to the extracellular attachment site, a
tyrosine kinase activity is “switched on”
at the intracellular portion. Protein
phosphorylation leads to altered cell
function via the assembly of other signal
proteins. Receptors for growth hormones
also belong to the catalytic receptor
class.
Protein synthesis-regulating receptors
for steroids, thyroid hormone,
and retinoic acid are found in the
cytosol and in the cell nucleus, respectively.
Binding of hormone exposes a normally
hidden domain of the receptor
protein, thereby permitting the latter to
bind to a particular nucleotide sequence
on a gene and to regulate its transcription.
Transcription is usually initiated or
enhanced, rarely blocked.

Drug-Receptor Interaction VI

Receptor Types
Receptors are macromolecules that bind
mediator substances and transduce this
binding into an effect, i.e., a change in
cell function. Receptors differ in terms
of their structure and the manner in
which they translate occupancy by a ligand
into a cellular response (signal
transduction).
G-protein-coupled receptors
consist of an amino acid chain that
weaves in and out of the membrane in
serpentine fashion. The extramembranal
loop regions of the molecule may
possess sugar residues at different Nglycosylation
sites. The seven !-helical
membrane-spanning domains probably
form a circle around a central pocket
that carries the attachment sites for the
mediator substance. Binding of the mediator
molecule or of a structurally related
agonist molecule induces a change
in the conformation of the receptor protein,
enabling the latter to interact with
a G-protein (= guanyl nucleotide-binding
protein). G-proteins lie at the inner
leaf of the plasmalemma and consist of
three subunits .
There are various G-proteins that differ
mainly with regard to their !-unit. Association
with the receptor activates the
G-protein, leading in turn to activation
of another protein (enzyme, ion channel).
A large number of mediator substances
act via G-protein-coupled receptors.
An example of a ligand-gated ion
channel is the nicotinic cholinoceptor
of the motor endplate. The receptor
complex consists of five subunits, each
of which contains four transmembrane
domains. Simultaneous binding of two
acetylcholine (ACh) molecules to the
two !-subunits results in opening of the
ion channel, with entry of Na+ (and exit
of some K+), membrane depolarization,
and triggering of an action potential .
The ganglionic N-cholinoceptors
apparently consist only of ! and " subunits
. Some of the receptors for
the transmitter #-aminobutyric acid
(GABA) belong to this receptor family:
the GABAA subtype is linked to a chloride
channel (and also to a benzodiazepine-
binding site). Glutamate
and glycine both act via ligandgated
ion channels.

Drug-Receptor Interaction V

Enantioselectivity of affinity. If a
receptor has sites for three of the substituents
(symbolized in B by a cone, a
sphere, and a cube) on the asymmetric
carbon to attach to, only one of the
enantiomers will have optimal fit. Its affinity
will then be higher. Thus, dexetimide
displays an affinity at the muscarinic
ACh receptors almost 10000 times
that of levetimide; and at !-
adrenoceptors, S(-)-propranolol has an
affinity 100 times that of the R(+)-form.
Enantioselectivity of intrinsic activity.
The mode of attachment at the
receptor also determines whether an effect
is elicited and whether or not a substance
has intrinsic activity, i.e., acts as
an agonist or antagonist. For instance,
(-) dobutamine is an agonist at "-adrenoceptors
whereas the (+)-enantiomer is
an antagonist.
Inverse enantioselectivity at another
receptor. An enantiomer may
possess an unfavorable configuration at
one receptor that may, however, be optimal
for interaction with another receptor.
In the case of dobutamine, the
(+)-enantiomer has affinity at !-adrenoceptors
10 times higher than that of the
(-)-enantiomer, both having agonist activity.
However, the "-adrenoceptor
stimulant action is due to the (-)-form
(see above).
As described for receptor interactions,
enantioselectivity may also be
manifested in drug interactions with
enzymes and transport proteins. Enantiomers
may display different affinities
and reaction velocities.
Conclusion: The enantiomers of a
racemate can differ sufficiently in their
pharmacodynamic and pharmacokinetic
properties to constitute two distinct
drugs.

Drug-Receptor Interaction IV

Enantioselectivity of Drug Action
Many drugs are racemates, including !-
blockers, nonsteroidal anti-inflammatory
agents, and anticholinergics (e.g.,
benzetimide A). A racemate consists of
a molecule and its corresponding mirror
image which, like the left and right
hand, cannot be superimposed. Such
chiral (“handed”) pairs of molecules are
referred to as enantiomers. Usually,
chirality is due to a carbon atom
linked to four different substituents
(“asymmetric center”). Enantiomerism is
a special case of stereoisomerism. Nonchiral
stereoisomers are called diastereomers
(e.g., quinidine/quinine).
Bond lengths in enantiomers, but
not in diastereomers, are the same.
Therefore, enantiomers possess similar
physicochemical properties (e.g., solubility,
melting point) and both forms are
usually obtained in equal amounts by
chemical synthesis. As a result of enzymatic
activity, however, only one of the
enantiomers is usually found in nature.
In solution, enantiomers rotate the
wave plane of linearly polarized light
in opposite directions; hence they are
refered to as “dextro”- or “levo-rotatory”,
designated by the prefixes d or (+) and l
or (-), respectively. The direction of rotation
gives no clue concerning the spatial
structure of enantiomers. The absolute
configuration, as determined by
certain rules, is described by the prefixes
S and R. In some compounds, designation
as the D- and L-form is possible
by reference to the structure of D- and
L-glyceraldehyde.
For drugs to exert biological actions,
contact with reaction partners in
the body is required. When the reaction
favors one of the enantiomers, enantioselectivity
is observed.

Drug-Receptor Interaction III

Agonist stabilizes spontaneously
occurring active conformation. The
receptor can spontaneously “flip” into
the active conformation. However, the
statistical probability of this event is
usually so small that the cells do not reveal
signs of spontaneous receptor activation.
Selective binding of the agonist
requires the receptor to be in the active
conformation, thus promoting its existence.
The “antagonist” displays affinity
only for the inactive state and stabilizes
the latter. When the system shows minimal
spontaneous activity, application
of an antagonist will not produce a measurable
effect. When the system has
high spontaneous activity, the antagonist
may cause an effect that is the opposite
of that of the agonist: inverse agonist.
A “true” antagonist lacking intrinsic
activity (“neutral antagonist”) displays
equal affinity for both the active and inactive
states of the receptor and does
not alter basal activity of the cell.
According to this model, a partial agonist
shows lower selectivity for the active
state and, to some extent, also binds
to the receptor in its inactive state.
Other Forms of Antagonism
Allosteric antagonism. The antagonist
is bound outside the receptor agonist
binding site proper and induces a decrease
in affinity of the agonist. It is also
possible that the allosteric deformation
of the receptor increases affinity for an
agonist, resulting in an allosteric synergism.
Functional antagonism. Two agonists
affect the same parameter (e.g.,
bronchial diameter) via different receptors
in the opposite direction (epinephrine
dilation; histamine ! constriction).

Drug-Receptor Interaction II

Agonists – Antagonists
An agonist has affinity (binding avidity)
for its receptor and alters the receptor
protein in such a manner as to generate
a stimulus that elicits a change in cell
function: “intrinsic activity“. The biological
effect of the agonist, i.e., the
change in cell function, depends on the
efficiency of signal transduction steps
initiated by the activated receptor.
Some agonists attain a maximal
effect even when they occupy only a
small fraction of receptors
. Other ligands , possessing
equal affinity for the receptor but lower
activating capacity (lower intrinsic activity),
are unable to produce a full maximal
response even when all receptors
are occupied: lower efficacy. Ligand B is
a partial agonist. The potency of an agonist
can be expressed in terms of the
concentration (EC50) at which the effect
reaches one-half of its respective maximum.
Antagonists attenuate the effect
of agonists, that is, their action is
“anti-agonistic”.
Competitive antagonists possess
affinity for receptors, but binding to the
receptor does not lead to a change in
cell function (zero intrinsic activity).
When an agonist and a competitive
antagonist are present simultaneously,
affinity and concentration of the two rivals
will determine the relative amount
of each that is bound. Thus, although the
antagonist is present, increasing the
concentration of the agonist can restore
the full effect . However, in the presence
of the antagonist, the concentration-
response curve of the agonist is
shifted to higher concentrations (“rightward
shift”).
Molecular Models of Agonist/Antagonist
Action
Agonist induces active conformation.
The agonist binds to the inactive receptor
and thereby causes a change from
the resting conformation to the active
state. The antagonist binds to the inactive
receptor without causing a conformational
change.

Drug-Receptor Interaction I

Types of Binding Forces
Unless a drug comes into contact with
intrinsic structures of the body, it cannot
affect body function.
Covalent bond. Two atoms enter a
covalent bond if each donates an electron
to a shared electron pair (cloud).
This state is depicted in structural formulas
by a dash. The covalent bond is
“firm”, that is, not reversible or only
poorly so. Few drugs are covalently
bound to biological structures. The
bond, and possibly the effect, persist for
a long time after intake of a drug has
been discontinued, making therapy difficult
to control. Examples include alkylating
cytostatics or organophosphates
. Conjugation reactions
occurring in biotransformation also
represent a covalent linkage (e.g., to
glucuronic acid).
Noncovalent bond. There is no formation
of a shared electron pair. The
bond is reversible and typical of most
drug-receptor interactions. Since a drug
usually attaches to its site of action by
multiple contacts, several of the types of
bonds described below may participate.
Electrostatic attraction. A positive
and negative charge attract each
other.
Ionic interaction: An ion is a particle
charged either positively (cation) or
negatively (anion), i.e., the atom lacks or
has surplus electrons, respectively. Attraction
between ions of opposite
charge is inversely proportional to the
square of the distance between them; it
is the initial force drawing a charged
drug to its binding site. Ionic bonds have
a relatively high stability.
Dipole-ion interaction: When bond
electrons are asymmetrically distributed
over both atomic nuclei, one atom
will bear a negative, and its partner
a positive partial charge. The molecule
thus presents a positive and a negative
pole, i.e., has polarity or a dipole. A
partial charge can interact electrostatically
with an ion of opposite charge.
Dipole-dipole interaction is the electrostatic
attraction between opposite
partial charges. When a hydrogen atom
bearing a partial positive charge bridges
two atoms bearing a partial negative
charge, a hydrogen bond is created.
A van der Waals’ bond is
formed between apolar molecular
groups that have come into close proximity.
Spontaneous transient distortion
of electron clouds (momentary faint dipole)
may induce an opposite dipole
in the neighboring molecule. The van
der Waals’ bond, therefore, is a form of
electrostatic attraction, albeit of very
low strength (inversely proportional to
the seventh power of the distance).
Hydrophobic interaction. The
attraction between the dipoles of water
is strong enough to hinder intercalation
of any apolar (uncharged) molecules. By
tending towards each other, H2O molecules
squeeze apolar particles from
their midst. Accordingly, in the organism,
apolar particles have an increased
probability of staying in nonaqueous,
apolar surroundings, such as fatty acid
chains of cell membranes or apolar regions
of a receptor.

Quantification of Drug Action V

Concentration-Binding Curves
In order to elicit their effect, drug molecules
must be bound to the cells of the
effector organ. Binding commonly occurs
at specific cell structures, namely,
the receptors. The analysis of drug binding
to receptors aims to determine the
affinity of ligands, the kinetics of interaction,
and the characteristics of the
binding site itself.
In studying the affinity and number
of such binding sites, use is made of
membrane suspensions of different tissues.
This approach is based on the expectation
that binding sites will retain
their characteristic properties during
cell homogenization. Provided that
binding sites are freely accessible in the
medium in which membrane fragments
are suspended, drug concentration at
the “site of action” would equal that in
the medium. The drug under study is radiolabeled
(enabling low concentrations
to be measured quantitatively),
added to the membrane suspension,
and allowed to bind to receptors. Membrane
fragments and medium are then
separated, e.g., by filtration, and the
amount of bound drug is measured.
Binding increases in proportion to concentration
as long as there is a negligible
reduction in the number of free binding
sites (c = 1 and B ! 10% of maximum
binding; c = 2 and B ! 20 %). As binding
approaches saturation, the number of
free sites decreases and the increment
in binding is no longer proportional to
the increase in concentration (in the example
illustrated, an increase in concentration
by 1 is needed to increase
binding from 10 to 20 %; however, an increase
by 20 is needed to raise it from 70
to 80 %).

The differing affinity of different ligands
for a binding site can be demonstrated
elegantly by binding assays. Although
simple to perform, these binding
assays pose the difficulty of correlating
unequivocally the binding sites concerned
with the pharmacological effect;
this is particularly difficult when more
than one population of binding sites is
present. Therefore, receptor binding
must not be implied until it can be
shown that
• binding is saturable (saturability);
• the only substances bound are those
possessing the same pharmacological
mechanism of action (specificity);
• binding affinity of different substances
is correlated with their pharmacological
potency.
Binding assays provide information
about the affinity of ligands, but they do
not give any clue as to whether a ligand
is an agonist or antagonist . Use of
radiolabeled drugs bound to their receptors
may be of help in purifying and
analyzing further the receptor protein.

Quantification of Drug Action IV

Disadvantages are:
1. Unavoidable tissue injury during dissection.
2. Loss of physiological regulation of
function in the isolated tissue.
3. The artificial milieu imposed on the
tissue.
Concentration-Effect Curves (B)
As the concentration is raised by a constant
factor, the increment in effect diminishes
steadily and tends asymptotically
towards zero the closer one comes
to the maximally effective concentration.
The concentration at which a maximal
effect occurs cannot be measured
accurately; however, that eliciting a
half-maximal effect (EC50) is readily determined.
It typically corresponds to the
inflection point of the concentration–
response curve in a semilogarithmic
plot (log concentration on abscissa).
Full characterization of a concentration–
effect relationship requires determination
of the EC50, the maximally
possible effect (Emax), and the slope at
the point of inflection.

Quantification of Drug Action III

Concentration-Effect Relationship (A)
The relationship between the concentration
of a drug and its effect is determined
in order to define the range of active
drug concentrations (potency) and
the maximum possible effect (efficacy).
On the basis of these parameters, differences
between drugs can be quantified.
As a rule, the therapeutic effect or toxic
action depends critically on the response
of a single organ or a limited
number of organs, e.g., blood flow is affected
by a change in vascular luminal
width. By isolating critical organs or tissues
from a larger functional system,
these actions can be studied with more
accuracy; for instance, vasoconstrictor
agents can be examined in isolated
preparations from different regions of
the vascular tree, e.g., the portal or
saphenous vein, or the mesenteric, coronary,
or basilar artery. In many cases,
isolated organs or organ parts can be
kept viable for hours in an appropriate
nutrient medium sufficiently supplied
with oxygen and held at a suitable temperature.
Responses of the preparation to a
physiological or pharmacological stimulus
can be determined by a suitable recording
apparatus. Thus, narrowing of a
blood vessel is recorded with the help of
two clamps by which the vessel is suspended
under tension.
Experimentation on isolated organs
offers several advantages:
1. The drug concentration in the tissue
is usually known.
2. Reduced complexity and ease of relating
stimulus and effect.
3. It is possible to circumvent compensatory
responses that may partially
cancel the primary effect in the intact
organism — e.g., the heart rate increasing
action of norepinephrine
cannot be demonstrated in the intact
organism, because a simultaneous
rise in blood pressure elicits a counter-
regulatory reflex that slows cardiac
rate.
4. The ability to examine a drug effect
over its full rage of intensities — e.g.,
it would be impossible in the intact
organism to follow negative chronotropic
effects to the point of cardiac
arrest.

Quantification of Drug Action II

To illustrate this point, we consider
an experiment in which the subjects individually
respond in all-or-none fashion,
as in the Straub tail phenomenon
. Mice react to morphine with excitation,
evident in the form of an abnormal
posture of the tail and limbs. The dose
dependence of this phenomenon is observed
in groups of animals (e.g., 10
mice per group) injected with increasing
doses of morphine. At the low dose,
only the most sensitive, at increasing
doses a growing proportion, at the highest
dose all of the animals are affected
. There is a relationship between the
frequency of responding animals and
the dose given. At 2 mg/kg, one out of 10
animals reacts; at 10 mg/kg, 5 out of 10
respond. The dose-frequency relationship
results from the different sensitivity
of individuals, which as a rule exhibits
a log-normal distribution (C, graph at
right, linear scale). If the cumulative frequency
(total number of animals responding
at a given dose) is plotted
against the logarithm of the dose (abscissa),
a sigmoidal curve results .
The inflection point of the curve lies at
the dose at which one-half of the group
has responded. The dose range encompassing
the dose-frequency relationship
reflects the variation in individual sensitivity
to the drug. Although similar in
shape, a dose-frequency relationship
has, thus, a different meaning than does
a dose-effect relationship. The latter can
be evaluated in one individual and results
from an intraindividual dependency
of the effect on drug concentration.
The evaluation of a dose-effect relationship
within a group of human subjects
is compounded by interindividual
differences in sensitivity. To account for
the biological variation, measurements
have to be carried out on a representative
sample and the results averaged.
Thus, recommended therapeutic doses
will be appropriate for the majority of
patients, but not necessarily for each individual.
The variation in sensitivity may be
based on pharmacokinetic differences
(same dose ! different plasma levels)
or on differences in target organ sensitivity
(same plasma level !different effects).

Quantification of Drug Action I

Dose–Response Relationship
The effect of a substance depends on the
amount administered, i.e., the dose. If
the dose chosen is below the critical
threshold (subliminal dosing), an effect
will be absent. Depending on the nature
of the effect to be measured, ascending
doses may cause the effect to increase in
intensity. Thus, the effect of an antipyretic
or hypotensive drug can be quantified
in a graded fashion, in that the extent
of fall in body temperature or blood
pressure is being measured. A dose-effect
relationship is then encountered.
The dose-effect relationship may
vary depending on the sensitivity of the
individual person receiving the drug,
i.e., for the same effect, different doses
may be required in different individuals.
Interindividual variation in sensitivity is
especially obvious with effects of the
“all-or-none” kind.

Pharmacokinetics VIII

Change in Elimination Characteristics
During Drug Therapy
With any drug taken regularly and accumulating
to the desired plasma level, it
is important to consider that conditions
for biotransformation and excretion do
not necessarily remain constant. Elimination
may be hastened due to enzyme
induction (p. 32) or to a change in urinary
pH (p. 40). Consequently, the
steady-state plasma level declines to a
new value corresponding to the new
rate of elimination. The drug effect may
diminish or disappear. Conversely,
when elimination is impaired (e.g., in
progressive renal insufficiency), the
mean plasma level of renally eliminated
drugs rises and may enter a toxic concentration
range.

Pharmacokinetics VII

Accumulation: Dose, Dose Interval, and
Plasma Level Fluctuation
Successful drug therapy in many illnesses
is accomplished only if drug concentration
is maintained at a steady high
level. This requirement necessitates
regular drug intake and a dosage schedule
that ensures that the plasma concentration
neither falls below the therapeutically
effective range nor exceeds
the minimal toxic concentration. A constant
plasma level would, however, be
undesirable if it accelerated a loss of effectiveness
(development of tolerance),
or if the drug were required to be
present at specified times only.
A steady plasma level can be
achieved by giving the drug in a constant
intravenous infusion, the steadystate
plasma level being determined by
the infusion rate, dose D per unit of time
and the clearance
This procedure is routinely used in
intensive care hospital settings, but is
otherwise impracticable. With oral administration,
dividing the total daily
dose into several individual ones, e.g.,
four, three, or two, offers a practical
compromise.
When the daily dose is given in several
divided doses, the mean plasma
level shows little fluctuation. In practice,
it is found that a regimen of frequent
regular drug ingestion is not well
adhered to by patients. The degree of
fluctuation in plasma level over a given
dosing interval can be reduced by use of
a dosage form permitting slow (sustained)
release .
The time required to reach steadystate
accumulation during multiple
constant dosing depends on the rate of
elimination. As a rule of thumb, a plateau
is reached after approximately
three elimination half-lives (t1/2).
For slowly eliminated drugs, which
tend to accumulate extensively (phenprocoumon,
digitoxin, methadone), the
optimal plasma level is attained only after
a long period. Here, increasing the
initial doses (loading dose) will speed
up the attainment of equilibrium, which
is subsequently maintained with a lower
dose (maintenance dose).

Pharmacokinetics VI

Time Course of Drug Plasma Levels
During Irregular Intake
In practice, it proves difficult to achieve
a plasma level that undulates evenly
around the desired effective concentration.
For instance, if two successive doses
are omitted, the plasma level will
drop below the therapeutic range and a
longer period will be required to regain
the desired plasma level. In everyday
life, patients will be apt to neglect drug
intake at the scheduled time. Patient
compliance means strict adherence to
the prescribed regimen. Apart from
poor compliance, the same problem
may occur when the total daily dose is
divided into three individual doses (tid)
and the first dose is taken at breakfast,
the second at lunch, and the third at
supper. Under this condition, the nocturnal
dosing interval will be twice the
diurnal one. Consequently, plasma levels
during the early morning hours may
have fallen far below the desired or,
possibly, urgently needed range.

Pharmacokinetics V

Time Course of Drug Plasma Levels
During Repeated Dosing
When a drug is administered at regular
intervals over a prolonged period, the
rise and fall of drug concentration in
blood will be determined by the relationship
between the half-life of elimination
and the time interval between
doses. If the drug amount administered
in each dose has been eliminated before
the next dose is applied, repeated intake
at constant intervals will result in similar
plasma levels. If intake occurs before
the preceding dose has been eliminated
completely, the next dose will add on to
the residual amount still present in the
body, i.e., the drug accumulates. The
shorter the dosing interval relative to
the elimination half-life, the larger will
be the residual amount of drug to which
the next dose is added and the more extensively
will the drug accumulate in
the body. However, at a given dosing
frequency, the drug does not accumulate
infinitely and a steady state (Css) or
accumulation equilibrium is eventually
reached. This is so because the activity
of elimination processes is concentration-
dependent. The higher the drug
concentration rises, the greater is the
amount eliminated per unit of time. After
several doses, the concentration will
have climbed to a level at which the
amounts eliminated and taken in per
unit of time become equal, i.e., a steady
state is reached. Within this concentration
range, the plasma level will continue
to rise (peak) and fall (trough) as dosing
is continued at a regular interval.
The height of the steady state (Css) depends
upon the amount administered
per dosing interval and the
clearance (Cltot)

Pharmacokinetics IV

Drug entry into hepatic and renal
tissue constitutes movement into the
organs of elimination. The characteristic
phasic time course of drug concentration
in plasma represents the sum of
the constituent processes of absorption,
distribution, and elimination,
which overlap in time. When distribution
takes place significantly faster than
elimination, there is an initial rapid and
then a greatly retarded fall in the plasma
level, the former being designated
the !-phase (distribution phase), the
latter the "-phase (elimination phase).
When the drug is distributed faster than
it is absorbed, the time course of the
plasma level can be described in mathematically
simplified form by the Bateman
function (k1 and k2 represent the
rate constants for absorption and elimination,
respectively).
B. The velocity of absorption depends
on the route of administration.
The more rapid the administration, the
shorter will be the time (tmax) required
to reach the peak plasma level (cmax),
the higher will be the cmax, and the earlier
the plasma level will begin to fall
again.
The area under the plasma level time
curve (AUC) is independent of the route
of administration, provided the doses
and bioavailability are the same (Dost’s
law of corresponding areas). The AUC
can thus be used to determine the bioavailability
F of a drug. The ratio of AUC
values determined after oral or intravenous
administration of a given dose of a
particular drug corresponds to the proportion
of drug entering the systemic
circulation after oral administration.
The determination of plasma levels affords
a comparison of different proprietary
preparations containing the same
drug in the same dosage. Identical plasma
level time-curves of different
manufacturers’ products with reference
to a standard preparation indicate bioequivalence
of the preparation under
investigation with the standard.

Pharmacokinetics III

Time Course of Drug Concentration in
Plasma
A. Drugs are taken up into and eliminated
from the body by various routes. The
body thus represents an open system
wherein the actual drug concentration
reflects the interplay of intake (ingestion)
and egress (elimination). When an
orally administered drug is absorbed
from the stomach and intestine, speed
of uptake depends on many factors, including
the speed of drug dissolution (in
the case of solid dosage forms) and of
gastrointestinal transit; the membrane
penetrability of the drug; its concentration
gradient across the mucosa-blood
barrier; and mucosal blood flow. Absorption
from the intestine causes the
drug concentration in blood to increase.
Transport in blood conveys the drug to
different organs (distribution), into
which it is taken up to a degree compatible
with its chemical properties and
rate of blood flow through the organ.
For instance, well-perfused organs such
as the brain receive a greater proportion
than do less well-perfused ones. Uptake
into tissue causes the blood concentration
to fall. Absorption from the gut diminishes
as the mucosa-blood gradient
decreases. Plasma concentration reaches
a peak when the drug amount leaving
the blood per unit of time equals that
being absorbed.

Pharmacokinetics II

The constancy of the process permits
calculation of the plasma volume
that would be cleared of drug, if the remaining
drug were not to assume a homogeneous
distribution in the total volume
(a condition not met in reality).
This notional plasma volume freed of
drug per unit of time is termed the
clearance. Depending on whether plasma
concentration falls as a result of urinary
excretion or metabolic alteration,
clearance is considered to be renal or
hepatic. Renal and hepatic clearances
add up to total clearance (Cltot) in the
case of drugs that are eliminated unchanged
via the kidney and biotransformed
in the liver. Cltot represents the
sum of all processes contributing to
elimination; it is related to the half-life
(t1/2) and the apparent volume of distribution
Vapp (p. 28) by the equation:
Vapp t1/2 = In 2 x ––––
Cltot
The smaller the volume of distribution
or the larger the total clearance, the
shorter is the half-life.
In the case of drugs renally eliminated
in unchanged form, the half-life of
elimination can be calculated from the
cumulative excretion in urine; the final
total amount eliminated corresponds to
the amount absorbed.
Hepatic elimination obeys exponential
kinetics because metabolizing
enzymes operate in the quasilinear region
of their concentration-activity
curve; hence the amount of drug metabolized
per unit of time diminishes
with decreasing blood concentration.
The best-known exception to exponential
kinetics is the elimination of alcohol
(ethanol), which obeys a linear
time course (zero-order kinetics), at
least at blood concentrations > 0.02 %. It
does so because the rate-limiting enzyme,
alcohol dehydrogenase, achieves
half-saturation at very low substrate
concentrations, i.e., at about 80 mg/L
(0.008 %). Thus, reaction velocity reaches
a plateau at blood ethanol concentrations
of about 0.02 %, and the amount of
drug eliminated per unit of time remains
constant at concentrations above
this level.

Pharmacokinetics I

Drug Concentration in the Body
as a Function of Time. First-Order
(Exponential) Rate Processes
Processes such as drug absorption and
elimination display exponential characteristics.
As regards the former, this follows
from the simple fact that the
amount of drug being moved per unit of
time depends on the concentration difference
(gradient) between two body
compartments (Fick’s Law). In drug absorption
from the alimentary tract, the
intestinal contents and blood would
represent the compartments containing
an initially high and low concentration,
respectively. In drug elimination via the
kidney, excretion often depends on glomerular
filtration, i.e., the filtered
amount of drug present in primary
urine. As the blood concentration falls,
the amount of drug filtered per unit of
time diminishes. The resulting exponential
decline is illustrated in . The
exponential time course implies constancy
of the interval during which the
concentration decreases by one-half.
This interval represents the half-life
(t1/2) and is related to the elimination
rate constant k by the equation t1/2 = ln
2/k. The two parameters, together with
the initial concentration co, describe a
first-order (exponential) rate process.

Drug Elimination X

Lipophilic drugs that are converted
in the liver to hydrophilic metabolites
permit better control, because the
lipophilic agent can be eliminated in
this manner. The speed of formation of
hydrophilic metabolite determines the
drug’s length of stay in the body.
If hepatic conversion to a polar metabolite
is rapid, only a portion of the
absorbed drug enters the systemic circulation
in unchanged form, the remainder
having undergone presystemic
(first-pass) elimination. When biotransformation
is rapid, oral administration
of the drug is impossible (e.g.,
glyceryl trinitate). Parenteral or,
alternatively, sublingual, intranasal, or
transdermal administration is then required
in order to bypass the liver. Irrespective
of the route of administration,
a portion of administered drug may be
taken up into and transiently stored in
lung tissue before entering the general
circulation. This also constitutes presystemic
elimination.
Presystemic elimination refers to
the fraction of drug absorbed that is
excluded from the general circulation
by biotransformation or by first-pass
binding.
Presystemic elimination diminishes
the bioavailability of a drug after its
oral administration. Absolute bioavailability
= systemically available amount/
dose administered; relative bioavailability
= availability of a drug contained
in a test preparation with reference to a
standard preparation.

Drug Elimination IX

Elimination of Lipophilic and
Hydrophilic Substances
The terms lipophilic and hydrophilic
(or hydro- and lipophobic) refer to the
solubility of substances in media of low
and high polarity, respectively. Blood
plasma, interstitial fluid, and cytosol are
highly polar aqueous media, whereas
lipids — at least in the interior of the lipid
bilayer membrane — and fat constitute
apolar media. Most polar substances
are readily dissolved in aqueous media
(i.e., are hydrophilic) and lipophilic
ones in apolar media. A hydrophilic
drug, on reaching the bloodstream,
probably after a partial, slow absorption
(not illustrated), passes through the liver
unchanged, because it either cannot,
or will only slowly, permeate the lipid
barrier of the hepatocyte membrane
and thus will fail to gain access to hepatic
biotransforming enzymes. The unchanged
drug reaches the arterial blood
and the kidneys, where it is filtered.
With hydrophilic drugs, there is little
binding to plasma proteins (protein
binding increases as a function of lipophilicity),
hence the entire amount
present in plasma is available for glomerular
filtration. A hydrophilic drug is
not subject to tubular reabsorption and
appears in the urine. Hydrophilic drugs
undergo rapid elimination.
If a lipophilic drug, because of its
chemical nature, cannot be converted
into a polar product, despite having access
to all cells, including metabolically
active liver cells, it is likely to be retained
in the organism. The portion filtered
during glomerular passage will be
reabsorbed from the tubules. Reabsorption
will be nearly complete, because
the free concentration of a lipophilic
drug in plasma is low (lipophilic substances
are usually largely proteinbound).
The situation portrayed for a
lipophilic non-metabolizable drug
would seem undesirable because pharmacotherapeutic
measures once initiated
would be virtually irreversible (poor
control over blood concentration).

Drug Elimination VIII

During passage down the renal tubule,
urinary volume shrinks more than
100-fold; accordingly, there is a corresponding
concentration of filtered drug
or drug metabolites . The resulting
concentration gradient between urine
and interstitial fluid is preserved in the
case of drugs incapable of permeating
the tubular epithelium. However, with
lipophilic drugs the concentration gradient
will favor reabsorption of the filtered
molecules. In this case, reabsorption
is not based on an active process
but results instead from passive diffusion.
Accordingly, for protonated substances,
the extent of reabsorption is
dependent upon urinary pH or the degree
of dissociation. The degree of dissociation
varies as a function of the urinary
pH and the pKa, which represents
the pH value at which half of the substance
exists in protonated (or unprotonated)
form. This relationship is graphically
illustrated with the example of
a protonated amine having a pKa of 7.0.
In this case, at urinary pH 7.0, 50 % of the
amine will be present in the protonated,
hydrophilic, membrane-impermeant
form (blue dots), whereas the other half,
representing the uncharged amine
(orange dots), can leave the tubular lumen
in accordance with the resulting
concentration gradient. If the pKa of an
amine is higher (pKa = 7.5) or lower (pKa
= 6.5), a correspondingly smaller or
larger proportion of the amine will be
present in the uncharged, reabsorbable
form. Lowering or raising urinary pH by
half a pH unit would result in analogous
changes for an amine having a pKa of
7.0.
The same considerations hold for
acidic molecules, with the important
difference that alkalinization of the
urine (increased pH) will promote the
deprotonization of -COOH groups and
thus impede reabsorption. Intentional
alteration in urinary pH can be used in
intoxications with proton-acceptor substances
in order to hasten elimination of
the toxin (alkalinization ! phenobarbital;
acidification !amphetamine).

Drug Elimination VII

The Kidney as Excretory Organ
Most drugs are eliminated in urine either
chemically unchanged or as metabolites.
The kidney permits elimination
because the vascular wall structure in
the region of the glomerular capillaries
allows unimpeded passage of blood
solutes having molecular weights (MW)
<> 70000. With
few exceptions, therapeutically used
drugs and their metabolites have much
smaller molecular weights and can,
therefore, undergo glomerular filtration,
i.e., pass from blood into primary
urine. Separating the capillary endothelium
from the tubular epithelium, the
basal membrane consists of charged
glycoproteins and acts as a filtration
barrier for high-molecular-weight substances.
The relative density of this barrier
depends on the electrical charge of
molecules that attempt to permeate it.
Apart from glomerular filtration
, drugs present in blood may pass
into urine by active secretion. Certain
cations and anions are secreted by the
epithelium of the proximal tubules into
the tubular fluid via special, energyconsuming
transport systems. These
transport systems have a limited capacity.
When several substrates are present
simultaneously, competition for the
carrier may occur .

Drug Elimination VI

Conjugations
The most important of phase II conjugation
reactions is glucuronidation. This
reaction does not proceed spontaneously,
but requires the activated form of
glucuronic acid, namely glucuronic acid
uridine diphosphate. Microsomal glucuronyl
transferases link the activated
glucuronic acid with an acceptor molecule.
When the latter is a phenol or alcohol,
an ether glucuronide will be
formed. In the case of carboxyl-bearing
molecules, an ester glucuronide is the
result. All of these are O-glucuronides.
Amines may form N-glucuronides that,
unlike O-glucuronides, are resistant to
bacterial !-glucuronidases.
Soluble cytoplasmic sulfotransferases
conjugate activated sulfate (3’-
phosphoadenine-5’-phosphosulfate)
with alcohols and phenols. The conjugates
are acids, as in the case of glucuronides.
In this respect, they differ from
conjugates formed by acetyltransferases
from activated acetate (acetylcoenzyme
A) and an alcohol or a phenol.
Acyltransferases are involved in the
conjugation of the amino acids glycine
or glutamine with carboxylic acids. In
these cases, an amide bond is formed
between the carboxyl groups of the acceptor
and the amino group of the donor
molecule (e.g., formation of salicyluric
acid from salicylic acid and glycine).
The acidic group of glycine or glutamine
remains free.

Drug Elimination V

Enterohepatic Cycle
After an orally ingested drug has been
absorbed from the gut, it is transported
via the portal blood to the liver, where it
can be conjugated to glucuronic or sulfuric
acid (shown in B for salicylic acid
and deacetylated bisacodyl, respectively)
or to other organic acids. At the pH of
body fluids, these acids are predominantly
ionized; the negative charge confers
high polarity upon the conjugated
drug molecule and, hence, low membrane
penetrability. The conjugated
products may pass from hepatocyte into
biliary fluid and from there back into
the intestine. O-glucuronides can be
cleaved by bacterial !-glucuronidases in
the colon, enabling the liberated drug
molecule to be reabsorbed. The enterohepatic
cycle acts to trap drugs in the
body. However, conjugated products
enter not only the bile but also the
blood. Glucuronides with a molecular
weight (MW) > 300 preferentially pass
into the blood, while those with MW >
300 enter the bile to a larger extent.
Glucuronides circulating in the blood
undergo glomerular filtration in the kidney
and are excreted in urine because
their decreased lipophilicity prevents
tubular reabsorption.
Drugs that are subject to enterohepatic
cycling are, therefore, excreted
slowly. Pertinent examples include digitoxin
and acidic nonsteroidal anti-inflammatory
agents .