You are on page 1of 105

T

C H

E
8 R
A

The minimum alveolar concentration


(MAC) is the alveolar concentration of an
inhaled anesthetic that prevents
movement in 50% of patients in response
to a standardized stimulus (eg, surgical
incision) .
Prolonged exposure to anesthetic
concentrations of nitrous oxide can result in
bone marrow depression (megaloblastic
anemia) and even neurological defi ciencies
(peripheral neuropathies) .
Halothane hepatitis is extremely rare (1 per
35,000 cases). Patients exposed to multiple
halothane anesthetics at short intervals,
middle-aged obese women, and persons
with a familial predisposition to halothane
toxicity or a personal history of toxicity are
considered to be at increased risk. Desfl
urane and isofl urane undergo much less
metabolism than halothane, resulting in
fewer of the metabolite protein adducts that
lead to immunologically mediated hepatic
injury .
Isofl urane dilates coronary arteries, but is
not nearly as potent a dilator as nitroglycerin
or adenosine. Dilation of normal coronary
arteries could theoretically divert blood away
from fi xed stenotic lesions .
154 SECTION II Clinical Pharmacology
The low solubility of desfl urane in blood
and body tissues causes a very rapid
induction of and emergence from
anesthesia.
—Continued next page

153

Inhalation
Anesthetics

KEYCONCEPTS

1 The greater the uptake of anesthetic


agent, 6 the greater the diff erence
between inspired and alveolar
concentrations, and the slower the rate
of induction.
2 Three factors aff ect anesthetic uptake:
solubility in the blood, alveolar blood fl
ow, and the diff erence in partial
pressure 7 between alveolar gas and
venous blood .
3 Low-output states predispose patients
to overdosage with soluble agents, as
the rate of rise in alveolar
concentrations will be
markedly increased .
4 Many of the factors that speed
induction also speed recovery:
elimination of rebreathing, high fresh
gas fl ows, low anesthetic-circuit
volume, low absorption by the
anesthetic circuit, decreased solubility,
high cerebral blood fl ow, and
increased ventilation .
CHAPTER 8 155
5 The unitary hypothesis proposes that
all inhalation agents share a common
mechanism of action at the molecular
level. This is supported by the
observation 9 that the
anesthetic potency of inhalation agents
correlates directly with their lipid
solubility (Meyer–Overton rule). There
is an ongoing debate as to the
mechanism of anesthetic action.
Anesthetic interactions at specifi c
protein 10 ion channels, as well as
more nonspecifi c membrane eff ects,
may combine to produce the
anesthetized state .
Continued— patients with
cardiovascular
11 Rapid increases disease .
in desfl urane
concentration 12 Nonpungency
lead to transient and rapid
but sometimes increases in
worrisome alveolar
elevations in anesthetic
heart rate, blood concentration
pressure, and make sevofl
catecholamine urane an
levels that are excellent choice
more for smooth and
pronounced rapid inhalation
than occur with inductions in
isofl urane, pediatric and
particularly in adult patients .

Nitrous oxide, Methoxyfl urane


chloroform, and was the most
ether were the fi rst potent inhalation
universally accepted agent, but its high
general anesthetics. solubility and low
Methoxyfl urane vapor pressure
and enfl urane, two yielded longer
potent halogenated inductions and
agents, were used emergences. Up to
for many years in 50% of it was
North American metabolized by
anesthesia practice. cytochrome P-450
156 SECTION II Clinical Pharmacology
(CYP) enzymes to Five inhalation
free fl uoride (F − ), agents continue to
oxalic acid, and be used in clinical
other nephrotoxic anesthesiology:
compounds. nitrous oxide,
Prolonged halothane, isofl
anesthesia with urane, desfl urane,
methoxyfl urane and sevofl urane.
was associated with Th e course of
a vasopressin- a general anesthetic
resistant, high- can be divided into
output, renal failure three phases: (1)
that was most induction, (2)
commonly seen maintenance, and
when F − levels (3) emergence.
increased to greater Inhalation
than 50 µmol/L. Enfl anesthetics, such as
urane has a halothane and
nonpungent odor sevofl urane, are
and is nonfl particularly useful in
ammable at clinical the induction of
concentrations. It pediatric patients in
depresses whom it may be diffi
myocardial cult to start an
contractility. It also intravenous line.
increases the Although adults are
secretion of usually induced with
cerebrospinal fl uid intravenous agents,
(CSF) and the the nonpungency
resistance to CSF and rapid onset of
outfl ow. During sevofl urane make
deep anesthesia inhalation induction
with hypocarbia practical for them as
electroencephalogr well. Regardless of
aphic changes can the patient’s age,
progress to a spike- anesthesia is oft en
and-wave pattern maintained with
producing tonic– inhalation agents.
clonic seizures. Emergence depends
Because of these primarily upon
concerns, methoxyfl redistribution from
urane and enfl the brain and
urane are no longer pulmonary
used.
CHAPTER 8 157
elimination of these depends on
agents. attainment of a
Because of therapeutic tissue
their unique route concentration in the
of administration, central nervous
inhalation system (CNS). Th
anesthetics have ere are many steps
useful in between the
pharmacological anesthetic vaporizer
properties not and the anesthetic’s
shared by other deposition in the
anesthetic agents. brain ( Figure 8–1 ).
For instance,
administration via
the pulmonary
circulation allows a
more rapid
appearance of the
drug in arterial
blood than
intravenous
administration.

Pharmacokin
etics of
Inhalation
Anesthetics

Although the
mechanism of
action of inhalation
anesthetics is
complex, likely
involving numerous
membrane proteins
and ion channels, it
is clear that
producing their
ultimate eff ect
158 SECTION II Clinical Pharmacology

FA the concentration
set on the
CT
OR
S
AF
FE
CTI
NG
INS
PIR
AT
OR
Y
CO
NC
EN
TR
ATI
ON

F I

Th e fresh gas
leaving the
anesthesia machine
mixes with gases in
the breathing circuit
before being
inspired by the
patient. Th erefore,
the patient is not
necessarily receiving
CHAPTER 8 159
Inhalation Anesthetics

Arterial
blood

FGF FI
FA
Breathing Fa Brain
circuit

FA

Venous
Lungs blood

FGF(fresh gas flow) is determined by the vaporizer and flowmeter settings.


Anesthesia machine
FI (inspired gas concentration) is determined by (1) FGF rate; (2) breathingcircuit
volume; and (3) circuit absorption.

FA (aveolar gas concentration) is determined by (1) uptake (uptake =


λb/g x C(A-V) x Q); (2) ventilation; and (3) the concentration effect
and second gas effect:
a) concentrating effect
b) augmented inflow effect

Fa (arterial gas concentration) is affected by ventilation/perfusion


mismatching.

FIGURE 81 Inhalation anesthetic agents must pass through many barriers between the anesthesia machine and
the brain. volume, and the
lower the circuit
absorption, the
vaporizer. Th e
closer the inspired
actual composition
gas concentration
of the inspired gas
will be to the fresh
mixture depends
gas concentration.
mainly on the fresh
Clinically, these
gas fl ow rate, the
attributes translate
volume of the
into faster induction
breathing system,
and recovery times.
and any absorption
by the machine or
breathing circuit. Th
e higher the fresh FACTORS
gas fl ow rate, the
smaller the AFFECTING
breathing system
160 SECTION II Clinical Pharmacology

ALVEOLAR anesthetic in the


blood and,
CONCENTRAT ultimately, in the
ION F A  brain. Similarly, the
partial pressure of
Uptake the anesthetic in the
If there were no brain is directly
uptake of anesthetic proportional to its
agent by the body, brain tissue
the alveolar gas concentration,
concentration (F a ) which determines
would rapidly clinical eff ect.
approach the
inspired gas 1 Th erefore, the
concentration (F i ). greater the uptake
Because anesthetic of anesthetic agent,
agents are taken up the greater the diff
by the pulmonary erence between
circulation during inspired and
induction, alveolar alveolar
concentrations lag concentrations, and
behind inspired the slower the rate
concentrations (F of induction.
a /F i <1.0). Th e TABLE 81
greater the uptake, Partition coeffi
the slower the rate cients of volatile
of rise of the anesthetics at 37°C.
alveolar 1

concentration and Blood/ Brain/


the lower the F a :F i Agent Gas Blood
ratio.
Nitrous oxide 0.47 1.1
Because the
concentration of a Halothane 2.4 2.9
gas is directly Isofl urane 1.4 2.6
proportional to its
partial pressure, the Desfl urane 0.42 1.3
alveolar partial Sevofl urane 0.65 1.7
pressure will also be 1 These values are
slow to rise. Th e averages derived from
alveolar partial multiple studies and should
be used for comparison
pressure is purposes, not as exact
important because numbers.
it determines the
partial pressure of
CHAPTER 8 161
2 Th ree coeffi cient (λ b/g ) of
factors aff ect nitrous oxide at
anesthetic uptake: 37°C is 0.47. In
solubility in the other words, at
blood, alveolar steady state, 1 mL of
blood fl ow, and the blood contains 0.47
diff erence in partial as much nitrous
pressure between oxide as does 1 mL
alveolar gas and of alveolar gas, even
venous blood. though the partial
Relatively pressures are the
soluble agents, such same. Stated
as nitrous oxide, are another way, blood
taken up by the has 47% of the
blood less avidly capacity for nitrous
than more soluble oxide as alveolar
agents, such as gas. Nitrous oxide is
halothane. As a much less soluble in
consequence, the blood than is
alveolar halothane, which
concentration of has a blood/gas
nitrous oxide rises partition coeffi cient
faster than that of at 37°C of 2.4. Th us,
halothane, and almost fi ve times
induction is faster. more halothane
Th e relative than nitrous oxide
solubilities of an must be dissolved to
anesthetic in air, raise the partial
blood, and tissues pressure of blood.
are expressed as Th e higher the
partition coeffi blood/gas coeffi
cients ( Table 8–1 ). cient, the greater
Each coeffi cient is the anesthetic’s
the ratio of the solubility and the
concentrations of greater its uptake by
the anesthetic gas in the pulmonary
each of two phases circulation. As a
at steady state. consequence of this
Steady state is defi increased solubility,
ned as equal partial alveolar partial
pressures in the two pressure rises more
phases. For slowly, and
instance, the induction is
blood/gas partition prolonged. Because
162 SECTION II Clinical Pharmacology
fat/blood partition concentrations will
coeffi cients are be markedly
greater than 1, increased.
blood/gas solubility Th e fi nal
is increased by factor aff ecting
postprandial uptake of anesthetic
lipidemia and is by the pulmonary
decreased by circulation is the
anemia. partial pressure
Th e second difference between
factor that aff ects alveolar gas and
uptake is alveolar venous blood. Th is
blood fl ow, which— gradient depends on
in the absence of tissue uptake. If
pulmonary shunting anesthetics did not
—is essentially pass into organs
equal to cardiac such as the brain,
output. If the venous and alveolar
cardiac output partial pressures
drops to zero, so will would become
anesthetic uptake. identical, and there
As cardiac output would be no
increases, pulmonary uptake.
anesthetic uptake Th e transfer of
increases, the rise in anesthetic from
alveolar partial blood to tissues is
pressure slows, and determined by three
induction is delayed. factors analogous to
Th e eff ect of systemic uptake:
changing cardiac tissue solubility of
output is less the agent
pronounced for (tissue/blood
insoluble partition coeffi
anesthetics, as so cient), tissue blood
little is taken up fl ow, and the diff
regardless of alve- erence in partial
3 olar blood pressure between
fl ow. L ow-output arterial blood and
states predispose the tissue.
patients to To better
overdosage with understand inhaled
soluble agents, as anesthetic uptake
the rate of rise in and distribution,
alveolar tissues have been
CHAPTER 8 163
Inhalation Anesthetics
Percentage of 10 50
body weight

1.0 Percentage of 75 19
Nitrous oxide cardiac output

Perfusion (mL/ 75 3
Desflurane
min/100 g)
0.8
Sevoflurane Relative solubility 1 1

0.6
Isoflurane
FA/FI

Halothane
0.4

0.2

0
10 20 30
Minutes

FIGURE 82 F A rises toward F I faster with nitrous oxide (an insoluble agent) than with halothane (a soluble agent). See
classifi ed into four Figure 8–1 for an
groups based on explanation of F A and F I
their solubility and .
blood fl ow (Table
8–2 ). Th e highly
perfused vessel-rich the fi rst to
group (brain, heart, encounter
liver, kidney, and appreciable
endocrine organs) is amounts of
anesthetic.
Moderate solubility
TABLE 82 Tissue and small volume
groups based on limit the capacity of
perfusion and this group, so it is
solubilities. also the fi rst to
reach steady state
Vessel
Characteristic Rich
(ie, arterial and
tissue partial
164 SECTION II Clinical Pharmacology
pressures are groups ( Figure 8–
equal). Th e muscle 3 ). Th e initial steep
group (skin and rate of uptake is due
muscle) is not as to unopposed fi lling
well perfused, so of the alveoli by
uptake is slower. In ventilation. Th e
addition, it has a rate of rise slows as
greater capacity due the vessel-rich
to a larger volume, group— and
and uptake will be eventually the
sustained for hours. muscle group—
Perfusion of the fat approach steady
group nearly equals state levels of
that of the muscle saturation.
group, but the
tremendous Ventilation
solubility of Th e lowering of
anesthetic in fat alveolar partial
leads to a total pressure by uptake
capacity can be countered by
(tissue/blood increasing alveolar
solubility × tissue ventilation. In other
volume) that would words, constantly
take days to replacing anesthetic
approach steady taken up by the
state. Th e minimal pulmonary
perfusion of the bloodstream results
vessel-poor group in better
(bones, ligaments, maintenance of
teeth, hair, and alveolar
cartilage) results in concentration. Th e
insignifi cant eff ect of increasing
uptake. ventilation will be
Anesthetic most obvious in
uptake produces a raising the F a /F i
characteristic curve for soluble
that relates the rise anesthetics, as they
in alveolar are more subject to
concentration to uptake. Because the
time ( Figure 8–2 ). F a /F i very rapidly
Th e shape of this approaches 1.0 for
graph is determined insoluble agents,
by the uptakes of increasing
individual tissue ventilation has
CHAPTER 8 165
minimal eff ect. In increasing the
contrast to the eff inspired
ect of anesthetics concentration not
on cardiac output, only increases the
anesthetics that alveolar
depress concentration, but
spontaneous also increases its
ventilation (eg, rate of rise (ie,
ether or increases F a /F i ),
Gas tension in various tissues

100
as % off inspired tension

90 Alveolar
80 Vessel-rich group
70
60
)

50 Muscle group
40
30
20 Fat group
10
0
0 30 60 90
(

Minutes

FIGURE 83 The rise and fall in alveolar partial pressure precedes that of other tissues. (Modifi ed and reproduced, with
permission, from Cowles AL et al: Uptake and distribution of inhalation anesthetic agents in clinical practice. Anesth Analg 1968;4:404.)
halothane) will because of two
decrease the rate of phenomena (see
rise in alveolar Figure 8–1 ) that
concentration and produce a so-called
create a negative “concentrating eff
feedback loop. ect.” First, if 50% of
an anesthetic is
Concentration taken up by the
Th e slowing of pulmonary
induction due to circulation, an
uptake from inspired
alveolar gas can be concentration of
reduced by 20% (20 parts of
increasing the anesthetic per 100
inspired parts of gas) will
concentration. result in an alveolar
Interestingly, concentration of
166 SECTION II Clinical Pharmacology
11% (10 parts of anesthetic
anesthetic remaining in a total
remaining in a total volume of 50 parts
volume of 90 parts of gas).
of gas). On the Th e second
other hand, if the phenomenon
inspired responsible for the
concentration is concentration eff
raised to 80% (80 ect is the
parts of anesthetic augmented infl ow
per 100 parts of eff ect. Using the
gas), the alveolar example above, the
concentration will 10 parts of absorbed
be 67% (40 parts of gas must be
anesthetic replaced by an
remaining in a total equal volume of the
volume of 60 parts 20% mixture to
of gas). Th us, even prevent alveolar
though 50% of the collapse. Th us, the
anesthetic is taken alveolar
up in both concentration
examples, a higher becomes 12% (10
inspired plus 2 parts of
concentration anesthetic in a total
results in a of 100 parts of gas).
disproportionately In contrast, aft er
higher alveolar absorption of 50%
concentration. In of the anesthetic in
this example, the 80% gas
increasing the mixture, 40 parts of
inspired 80% gas must be
concentration 4-fold inspired. Th is
results in a 6-fold further increases
increase in alveolar the alveolar
concentration. Th e concentration from
extreme case is an 67% to 72% (40 plus
inspired 32 parts of
concentration of anesthetic in a
100% (100 parts of volume of 100 parts
100), which, despite of gas).
a 50% uptake, will Th e
result in an alveolar concentration eff
concentration of ect is more signifi
100% (50 parts of cant with nitrous
CHAPTER 8 167
oxide, than with the the arterial partial
volatile anesthetics, pressure is
as the former can be consistently less
used in much higher than endexpiratory
concentrations. gas would predict.
Nonetheless, a high Reasons for this
concentration of may include venous
nitrous oxide will admixture, alveolar
augment (by the dead space, and
same mechanism) nonuniform alveolar
not only its own gas distribution.
uptake, but Furthermore, the
theoretically that of existence of
a concurrently ventilation/perfusio
administered n mismatching will
volatile anesthetic. increase the
Th e concentration alveolar–arterial diff
eff ect of one gas erence. Mismatch
upon another is acts as a restriction
called the second to fl ow: It raises the
gas eff ect, which is pressure in front of
probably insignifi the restriction,
cant in the clinical lowers the pressure
practice of beyond the
anesthesiology. restriction, and
FACTORS reduces the fl ow
through the
AFFECTING restriction. Th e
overall eff ect is an
ARTERIAL
increase in the
CONCENTRAT alveolar partial
ION Fa pressure
(particularly for
highly soluble
Ventilation/Per agents) and a
decrease in the
fusion
arterial partial
Mismatch pressure
Normally, alveolar (particularly for
and arterial poorly soluble
anesthetic partial agents). Th us, a
pressures are bronchial intubation
assumed to be or a right-toleft
equal, but in fact, intracardiac shunt
168 SECTION II Clinical Pharmacology
will slow the rate of group of isozymes
induction with (specifi cally CYP
nitrous oxide more 2EI) seems to be
than with important in the
halothane. metabolism of some
volatile anesthetics.
Diff usion of
FACTORS anesthetic through
the skin is insignifi
AFFECTING cant.
ELIMINATION Th e most
important route for
Recovery from
elimination of
anesthesia depends
on lowering the
4 inhalation
concentration of
anesthetic in brain anesthetics is the
tissue. Anesthetics alveolus. Many of
can be eliminated the factors that
by speed induction also
biotransformation, speed recovery:
transcutaneous loss, elimination of
or exhalation. rebreathing, high
Biotransformation fresh gas
Inhalati
usually accounts for on
a minimal increase Anesth
etics
in the rate of
decline of alveolar
partial pressure. Its fl ows, low
greatest impact is anesthetic-circuit
on the elimination volume, low
of soluble absorption by the
anesthetics that anesthetic circuit,
undergo extensive decreased solubility,
metabolism (eg, high cerebral blood
methoxyfl urane). fl ow (CBF), and
Th e greater increased
biotransformation ventilation.
of halothane Elimination of
compared with isofl nitrous oxide is so
urane accounts for rapid that alveolar
halothane’s faster oxygen and CO 2 are
elimination, even diluted. Th e
though it is more resulting diff usion
soluble. Th e CYP
CHAPTER 8 169
hypoxia is
prevented by
administering 100%
oxygen for 5–10 min Pharmacodyn
aft er discontinuing amics of
nitrous oxide. Th e
rate of recovery is Inhalation
usually faster than
induction because
Anesthetics
tissues that have
not reached
equilibrium will THEORIE
continue to take up S OF
anesthetic until the
alveolar partial ANESTHE
pressure falls below TIC
the tissue partial
pressure. For ACTION
instance, fat will General anesthesia
continue to take up is an altered
anesthetic and physiological state
hasten recovery characterized by
until the partial reversible loss of
pressure exceeds consciousness,
the alveolar partial analgesia, amnesia,
pressure. Th is and some degree of
redistribution is not muscle relaxation.
as useful aft er Th e multitude of
prolonged substances capable
anesthesia (fat of producing
partial pressures of general anesthesia
anesthetic will have is remarkable: inert
come “closer” to elements (xenon),
arterial partial simple inorganic
pressures at the compounds (nitrous
time the anesthetic oxide), halogenated
was removed from hydrocarbons
fresh gas)—thus, (halothane), ethers
the speed of (isofl urane, sevofl
recovery also urane, desfl urane),
depends on the and complex
length of time the organic structures
anesthetic has been (propofol). A
administered. unifying theory
170 SECTION II Clinical Pharmacology
explaining membrane bilayer.
anesthetic action It is possible that
would have to inhalational
accommodate this anesthetics act on
diversity of multiple protein
structure. In fact, receptors that block
the various agents excitatory channels
probably produce and promote the
anesthesia by diff activity of inhibitory
ering sets of channels aff ecting
molecular neuronal activity, as
mechanisms. well as by some
Inhalational agents nonspecifi c
interact with membrane eff ects.
numerous ion Th ere does not
channels present in seem to be a single
the CNS and macroscopic site of
peripheral nervous action that is shared
system. Nitrous by all inhalation
oxide and xenon are agents. Specifi c
believed to inhibit brain areas aff ected
N -methyl D by various
-aspartate (NMDA) anesthetics include
receptors. NMDA the reticular
receptors are activating system,
excitatory receptors the cerebral cortex,
in the brain. Other the cuneate
inhalational agents nucleus, the
may interact at olfactory cortex, and
other receptors (eg, the hippocampus;
gamma- however, to be
aminobutyric acid clear, general
[GABA]-activated anesthetics bind
chloride channel throughout the CNS.
conductance) Anesthetics have
leading to also been shown to
anesthetic eff ects. depress excitatory
Additionally, some transmission in the
studies suggest that spinal cord,
inhalational agents particularly at the
continue to act in a level of the dorsal
nonspecifi c horn interneurons
manner, thereby aff that are involved in
ecting the pain transmission.
CHAPTER 8 171
Diff ering aspects of attempted to
anesthesia may be identify a unitary
related to diff erent hypothesis of
sites of anesthetic anesthetic eff ects.
action. For example, Th is hypothesis
unconsciousness proposes that all
and amnesia are inhalation agents
probably mediated share a common
by cortical mechanism of
anesthetic action, action at the
whereas the molecular level. Th
suppression of is was previously
purposeful supported by the
withdrawal from observation that the
pain may be related anesthetic potency
to subcortical of inhalation agents
structures, such as correlates directly
the spinal cord or with their lipid
brain stem. One solubility (Meyer–
study in rats Overton rule). Th e
revealed that implication is that
removal of the anesthesia results
cerebral cortex did from molecules
not alter the dissolving at specifi
potency of the c lipophilic sites. Of
anesthetic! Indeed, course, not all lipid-
measures of soluble molecules
minimal alveolar are anesthetics
concentration (some are actually
(MAC), the convulsants), and
anesthetic the correlation
concentration that between anesthetic
prevents movement potency and lipid
in 50% of subjects or solubility is only
animals, are approximate (
dependent upon Figure 8–4 ).
anesthetic eff ects Neuronal
at the spinal cord membranes contain
and not at the a multitude of
cortex. hydrophobic sites in
their phospholipid
5 Past bilayer. Anesthetic
understanding of binding to these
anesthetic action sites could expand
172 SECTION II Clinical Pharmacology
the bilayer beyond a critical volume
critical amount, hypothesis.
altering membrane General
function (critical anesthetic action
volume hypothesis). could be due to
Although this theory alterations in any
is almost certainly one (or a
an oversimplifi combination) of
cation, it explains an several cellular
interesting systems, including
phenomenon: the voltage-gated ion
reversal of channels, ligand-
anesthesia by gated ion channels,
increased pressure. second messenger
Laboratory animals functions, or
exposed to elevated neurotransmitter
hydrostatic pressure receptors. For
develop a resistance example, many
to anesthetic eff anesthetics enhance
ects. Perhaps the GABA inhibition of
pressure is the CNS.
displacing a number Furthermore, GABA
of molecules from receptor agonists
the membrane or seem to enhance
distorting the anesthesia, whereas
anesthetic binding GABA antagonists
sites in the reverse some
membrane, anesthetic eff ects.
increasing Th ere seems to be a
anesthetic strong correlation
requirements. between anesthetic
However, studies in potency and
the 1980s potentiation of
demonstrated the GABA receptor
ability of anesthetics activity. Th us,
to inhibit protein anesthetic action
actions, shift ing may relate to
attention to the binding in relatively
numerous ion hydrophobic
channels that might domains in channel
aff ect neuronal proteins (GABA
transmission and receptors).
away from the Modulation of GABA
function may prove
CHAPTER 8 173
to be a principal
mechanism of
action for many
anesthetic drugs.
Th e glycine
receptor α 1

-subunit, whose
function is
enhanced by
inhalation
anesthetics, is
another potential
anesthetic site of
action.
Th e tertiary
and quaternary
structure of amino
acids within an
anesthetic-binding
pocket could be
modifi ed by
inhalation agents,
perturbing the
receptor itself, or
indirectly producing
an eff ect at a
distant site.
Other ligand-
gated ion channels
whose modulation
may play a role in
anesthetic action
include nicotinic
acetylcholine
receptors and
NMDA receptors.
Investigations
into mechanisms of
anesthetic action
are likely to remain
ongoing for many
years,
174 SECTION II Clinical Pharmacology

100 Nitrous oxide

Ethylene

(
O
liv
e
)

oi
l

10
Cyclopropane
Log MAC

Fluroxene

Diethylether
Enflurane
Isoflurane
1.0

Halothane
(

Chloroform
W
hi
te
m
)

at
te
r

Trichloroethylene
Methoxyflurane

0.1
1 10 100 1000
Log partition coefficient

FIGURE 84 There is a good but not perfect correlation between anesthetic potency and lipid solubility. MAC,
minimum alveolar concentration. (Modifi ed and reproduced, with permission, from Lowe HJ, Hagler K: Gas Chromatography in Biology and
Medicine . Churchill, 1969.) actions will be the challenge in designing better
inhalational agents.

as many protein channels may be aff ected by ANESTHETIC NEUROTOXICITY


individual anesthetic agents, and no obligatory In recent years, there has been ongoing concern
site has yet been identifi ed. Selecting among so that general anesthetics damage the developing
many molecular targets for the one(s) that brain. It has been suggested that early exposure
provide optimum eff ects with minimal adverse to anesthetics can promote cognitive impairment
in later life. Concern has been raised that
CHAPTER 8 Inhalation Anesthetics 175

anesthetic exposure aff ects the development and cardiac protective eff ects against ischemia-
and the elimination of synapses in the infant reperfusion injury. Ischemic preconditioning
brain. For example, animal studies have implies that a brief ischemic episode protects a
demonstrated that isofl urane exposure cell from future, more pronounced ischemic
promotes neuronal apoptosis and subsequent events. Various molecular mechanisms have
learning disability. Volatile anesthetics have been been suggested to protect cells preconditioned
shown to promote apoptosis by altering cellular either through ischemic events or secondary to
calcium homeostatic mechanisms. pharmacologic mechanisms, such as through the
Human studies exploring whether use of inhalational anesthetics. In the heart,
anesthesia is harmful in children are diffi cult, as preconditioning in part arises from actions at
conducting a randomized controlled trial for that ATP-sensitive potassium (K ATP ) channels.
purpose only would be unethical. Studies that Th e exact mechanism of anesthetic
compare populations of children who have had preconditioning is likely to be multifocal and
anesthetics with those who have not are also includes the opening of K ATP channels, resulting in
complicated by the reality that the former less mitochondrial calcium ion concentration and
population is likewise having surgery and reduction of reactive oxygen species (ROS)
receiving the attention of the medical production. ROS are associated with cellular
community. Consequently, children receiving injury. For example, excitatory NMDA receptors
anesthetics may be more likely to be diagnosed are linked to the development of neuronal injury.
with learning diffi culties in the fi rst place. Data NMDA antagonists, such as the noble anesthetic
from one large study demonstrated that children gas Xenon, have been shown to be
who underwent surgery and anesthesia had a neuroprotective. Xenon has an anti-apoptotic eff
greater likelihood of carrying the diagnosis of a ect that may be secondary to its inhibition of
developmental disorder; however, the fi nding calcium ion infl ux following cell injury. Other
was not supported in twins (ie, the incidence of inhalational agents, such as sevofl urane, have
developmental disability was not greater in a been shown to reduce markers of myocardial cell
twin who was exposed to anesthesia and surgery injury (eg, troponin T), compared with
than in one who was not). intravenous anesthetic techniques.
Human, animal, and laboratory trials As with neurotoxicity, the role of
demonstrating or refuting that anesthetic inhalational anesthetics in tissue protection is the
neurotoxicity leads to developmental disability in subject of ongoing investigation.
children are underway. As of this writing, there is
insuffi cient and confl icting evidence to warrant
changes in anesthetic practice (see: MINIMUM ALVEOLAR
www.smarttots.org).
CONCENTRATION
6 Th e minimum alveolar concentration (MAC)
ANESTHETIC of an inhaled anesthetic is the alveolar
NEUROPROTECTION AND concen-
tration that prevents movement in 50% of
CARDIAC PRECONDITIONING patients in response to a standardized stimulus
Although inhalational agents have been (eg, surgical incision). MAC is a useful measure
suggested as contributing to neurotoxicity, they because it mirrors brain partial pressure, allows
have also been shown to provide both neurologic
176 SECTION II Clinical Pharmacology

comparisons of potency between agents, and the same MAC: 0.5 MAC of halothane causes
provides a standard for experimental evaluations more myocardial depression than 0.5 MAC of
( Table 8–3 ). Nonetheless, it should be nitrous oxide. MAC represents only one point on
remembered that this is a median value with the dose–response curve—it is the equivalent of
limited usefulness in managing individual a median eff ective dose (ED 50 ). MAC multiples
patients, particularly during times of rapidly are clinically useful if the concentration–response
changing alveolar concentrations (eg, induction). curves of the anesthetics being compared are
Th e MAC values for diff erent anesthetics parallel, nearly linear, and continuous for the eff
are roughly additive. For example, a mixture of ect being predicted. Roughly 1.3 MAC of any of
0.5 MAC of nitrous oxide (53%) and 0.5 MAC of the volatile anesthetics (eg, for halothane: 1.3 ×
halothane (0.37%) produces the same likelihood 0.74% = 0.96%) has been found to prevent
that movement in response to surgical incision movement in about 95% of patients (an
will be suppressed as 1.0 MAC of isofl urane approximation of the ED 95 ); 0.3–0.4 MAC is
(1.7%) or 1.0 MAC of any other single agent. In associated with awakening from anesthesia (MAC
contrast to CNS depression, the degree of awake) when the inhaled drug is the only agent
myocardial depression may not be equivalent at maintaining anesthetic (a rare circumstance).

TABLE 83 Properties of modern inhalation anesthetics .


Vapo
r
Press
ure
(mm
Hg at
Agent MAC% 1 20°C)
Nitrous oxide 105 2 —

2
Halothane (Fluothane)
4
0.75 3

Isoflurane (Forane)
1.2 2
4
0
Desflurane (Suprane)
6.0

6
Sevofl urane (Ultane) 8
1
2.0

1
6
0
1 These minimum alveolar concentration (MAC) values are for 30- to 55-year old human subjects and are expressed as a
percentage of 1 atmosphere. High altitude requires a higher inspired concentration of anesthetic to achieve the same partial
pressure.
2 A concentration greater than 100% means that hyperbaric conditions are required to achieve 1.0 MAC.
CHAPTER 8 Inhalation Anesthetics 177

MAC can be altered by several physiological NITROUS OXIDE


and pharmacological variables ( Table 8–4 ). One
of the most striking is the 6% decrease in MAC Physical Properties
per decade of age, regardless of volatile Nitrous oxide (N 2 O; laughing gas) is colorless
anesthetic . and essentially odorless. Although nonexplosive
MAC is relatively unaff ected by species, sex, or and nonfl ammable, nitrous oxide is as capable as
duration of anesthesia. Surprisingly, MAC is not oxygen of supporting combustion. Unlike the
altered aft er spinal cord transection in rats, potent volatile
leading to the hypothesis that the site of 1 These conclusions are based on human and animal
studies.
anesthetic inhibition of motor responses lies in
2 CSF, cerebrospinal fl uid.
the spinal cord.

Clinical Pharmacology of agents, nitrous oxide is a gas at room


temperature and ambient pressure. It can be
Inhalation Anesthetics kept as a liquid under pressure because its critical
temperature lies above room temperature.
Nitrous oxide is a relatively inexpensive
anesthetic; however, concerns regarding its
safety have led to continued interest in
alternatives such as xenon ( Table 8–5 ). As
TABLE 84 Factors aff ecting MAC. 1
Eff ect Eff ect
Variable on MAC Comments Variable on MAC Comments

Temperature Electrolytes
Hypothermia ↓ Hypercalcemia ↓
Hyperthermia ↓ Hypernatremia ↑ Caused by altered CSF2
↑ if > 42°C Hyponatremia ↓ Caused by altered CSF

Age Pregnancy ↓ MAC decreased by one-


Young ↑ third at 8 weeks’
Elderly ↓ gestation; normal by 72
h postpartum

Alcohol Drugs ↓ Except cocaine


Acute intoxication ↓ Local anesthetics ↓
Chronic abuse ↑ Opioids ↓
Ketamine ↓

Barbiturates
Anemia ↓
Benzodiazepines
Hematocrit < 10% ↓ ↓
Verapamil
Pa O2 ↓ Lithium ↓
<40 mm Hg Sympatholytics ↓
Methyldopa ↓
Pa CO 2 Clonidine
>95 mm Hg ↓ Caused by < pH in Dexmedetomidine
CSF Sympathomimetics ↓
Amphetamine ↑
Thyroid
Chronic ↑
Hyperthyroid No change Acute
Hypothyroid No change ↑
Cocaine
Blood pressure Ephedrine
Mean arterial pressure ↓
<40 mm Hg
178 SECTION II Clinical Pharmacology

noted earlier, nitrous oxide, like xenon, is an nitrous oxide directly depresses myocardial
NMDA receptor antagonist. contractility in TABLE 85 Advantages and
disadvantages of xenon (Xe) anesthesia.
Eff ects on Organ Systems 4
Percentage of absorbed anesthetic undergoing metabolism.

A. Cardiovascular
Nitrous oxide has a tendency to stimulate the
sympathetic nervous system. Th us, even though vitro, arterial blood pressure, cardiac output, and
heart rate are essentially unchanged or slightly
Advantages
Inert (probably nontoxic with no metabolism)
Minimal cardiovascular effects
Low blood solubility
Rapid induction and recovery
Does not trigger malignant hyperthermia
Environmentally friendly
Nonexplosive

Disadvantages
High cost
Low potency (MAC = 70%)

TABLE 86 Clinical pharmacology of inhalational anesthetics.


Nitrous Oxide Halothane Isofl urane Desfl urane Sevofl urane

Cardiovascular
Blood pressure N/C 1 ↓↓
Heart rate N/C ↓↓ ↓ ↓↓ ↑ N/C or ↑ ↓
Systemic vascular resistance N/C N/C ↓↓ ↓↓ N/C
Cardiac output 2 N/C ↓ N/C N/C or ↓ ↓↓

Respiratory
Tidal volume ↓ ↓
Respiratory rate ↑ ↓↓ ↑↑ ↓↓ ↑ ↑ ↓↑

Pa CO 2
Resting N/C ↑ ↑
Challenge ↑ ↑ ↑ ↑↑ ↑↑ ↑↑

Cerebral
Blood flow ↑ ↑↑
Intracranial pressure ↑ ↑↑ ↑ ↑
Cerebral metabolic rate ↑ ↓ ↑ ↑ ↑↑
Seizures ↓ ↓ ↓↓ ↓ ↓↓ ↓ ↓↓ ↓

Neuromuscular
Nondepolarizing blockade 3 ↑ ↑↑ ↑↑↑ ↑↑↑ ↑↑

Renal
Renal blood flow ↓
Glomerular filtration rate ↓↓ ↓↓ ↓↓ ↓↓ ↓↓ ↓↓ ↓ ↓↓
Urinary output ↓↓ ↓↓ ↓↓ ↓ ↓

Hepatic
Blood flow ↓ ↓↓ ↓ ↓ ↓

Metabolism 4
0.004% 15% to 20% 0.2% <0.1% 5%
1 N/C, no change.
2 Controlled ventilation.
3 Depolarizing blockage is probably also prolonged by these agents, but this is usually not clinically signifi cant.
CHAPTER 8 Inhalation Anesthetics 179

elevated in vivo because of its stimulation of leads to a drop in glomerular fi ltration rate and
catecholamines ( Table 8–6 ). Myocardial urinary output.
depression may be unmasked in patients with
coronary artery disease or severe hypovolemia. F. Hepatic
Constriction of pulmonary vascular smooth Hepatic blood fl ow probably falls during nitrous
muscle increases pulmonary vascular resistance, oxide anesthesia, but to a lesser extent than with
which results in a generally modest elevation of the volatile agents.
right ventricular end-diastolic pressure. Despite G. Gastrointestinal
vasoconstriction of cutaneous vessels, peripheral
Use of nitrous oxide in adults increases the risk
vascular resistance is not signifi cantly altered.
of postoperative nausea and vomiting,
B. Respiratory presumably as a result of activation of the
chemoreceptor trigger zone and the vomiting
Nitrous oxide increases respiratory rate
center in the medulla.
(tachypnea) and decreases tidal volume as a
result of CNS stimulation and, perhaps, activation
of pulmonary stretch receptors. Th e net eff ect is Biotransformation & Toxicity During
a minimal change in minute ventilation and emergence, almost all nitrous oxide is eliminated
resting arterial CO 2 levels. Hypoxic drive, the by exhalation. A small amount diff uses out
ventilatory response to arterial hypoxia that is through the skin. Biotransformation is limited to
mediated by peripheral chemoreceptors in the the less than 0.01% that undergoes reductive
carotid bodies, is markedly depressed by even metabolism in the gastrointestinal tract by
small amounts of nitrous oxide. Th is is a concern anaerobic bacteria.
in the recovery room. By irreversibly oxidizing the cobalt atom in
vitamin B 12 , nitrous oxide inhibits enzymes that
C. Cerebral are vitamin B 12 dependent. Th ese enzymes
By increasing CBF and cerebral blood volume, include methionine synthetase, which is
nitrous oxide produces a mild elevation of necessary for myelin formation, and thymidylate
intracranial pressure. Nitrous oxide also increases synthetase, which is
cerebral oxygen consumption (CMRO 2 ). Th ese
two eff ects make nitrous oxide theoretically less 7 necessary for DNA synthesis. Prolonged
attractive than other agents for neuroanesthesia. exposure to anesthetic concentrations of
Concentrations of nitrous oxide below MAC may
nitrous
provide analgesia in dental surgery, labor,
traumatic injury, and minor surgical procedures. oxide can result in bone marrow depression
(megaloblastic anemia) and even neurological
D. Neuromuscular defi ciencies (peripheral neuropathies). However,
In contrast to other inhalation agents, nitrous administration of nitrous oxide for bone marrow
oxide does not provide signifi cant muscle harvest does not seem to aff ect the viability of
relaxation. In fact, at high concentrations in bone marrow mononuclear cells. Because of
hyperbaric chambers, nitrous oxide causes possible teratogenic eff ects, nitrous oxide is oft
skeletal muscle rigidity. Nitrous oxide is not a en avoided in pregnant patients who are not yet
triggering agent of malignant hyperthermia. in the third trimester. Nitrous oxide may also
alter the immunological response to infection by
E. Renal aff ecting chemotaxis and motility of
Nitrous oxide seems to decrease renal blood fl polymorphonuclear leukocytes.
ow by increasing renal vascular resistance. Th is
180 SECTION II Clinical Pharmacology

Contraindications the concentration of volatile anesthetic


Although nitrous oxide is insoluble in comparison delivered. For example, decreasing nitrous oxide
with other inhalation agents, it is 35 times more concentration (ie, increasing oxygen
soluble than nitrogen in blood. Th us, it tends to concentration) increases the concentration of
diff use into air-containing cavities more rapidly volatile agent despite a constant vaporizer
than nitrogen is absorbed by the bloodstream. setting. Th is disparity is due to the relative
For instance, if a patient with a 100-mL solubilities of nitrous oxide and oxygen in liquid
pneumothorax inhales 50% nitrous oxide, the gas volatile anesthetics. Th e second gas eff ect was
content of the pneumothorax will tend to discussed earlier. Nitrous oxide is an ozone-
approach that of the bloodstream. Because depleting gas with greenhouse eff ects.
nitrous oxide will diff use into the cavity more
rapidly than the air (principally nitrogen) diff uses
out, the pneumothorax expands until it contains HALOTHANE
100 mL of air and 100 mL of nitrous oxide. If the
Physical Properties
walls surrounding the cavity are rigid, pressure
Halothane is a halogenated alkane (see Table 8–
rises instead of volume. Examples of conditions
3 ). Th e carbon–fl uoride bonds are responsible
in which nitrous oxide might be hazardous
for its nonfl ammable and nonexplosive nature.
include venous or arterial air embolism,
Th ymol preservative and amber-colored bottles
pneumothorax, acute intestinal obstruction
retard spontaneous oxidative decomposition. It is
with bowel distention, intracranial air
rarely used in the United States.
(pneumocephalus following dural closure or
pneumoencephalography), pulmonary air cysts,
intraocular air bubbles, and tympanic Eff ects on Organ Systems
membrane graft ing . Nitrous oxide will even diff A. Cardiovascular
use into tracheal tube cuff s, increasing the A dose-dependent reduction of arterial blood
pressure against the tracheal mucosa. Obviously, pressure is due to direct myocardial depression;
nitrous oxide is of limited value in patients 2.0 MAC of halothane in patients not
requiring high inspired oxygen concentrations. undergoing surgery results in a 50% decrease in
blood pressure and cardiac output. Cardiac
Drug Interactions depression— from interference with sodium–
Because the high MAC of nitrous oxide prevents calcium exchange and intracellular calcium
its use as a complete general anesthetic, it is utilization—causes an increase in right atrial
frequently used in combination with the more pressure. Although halothane is a coronary artery
potent volatile agents. Th e addition of nitrous vasodilator, coronary blood fl ow decreases, due
oxide decreases the requirements of these other to the drop in systemic arterial pressure.
agents (65% nitrous oxide decreases the MAC of Adequate myocardial perfusion is usually
the volatile anesthetics by approximately 50%). maintained, as oxygen demand also drops.
Although nitrous oxide should not be considered Normally, hypotension inhibits baroreceptors in
a benign carrier gas, it does attenuate the the aortic arch and carotid bifurcation, causing a
circulatory and respiratory eff ects of volatile decrease in vagal stimulation and a
anesthetics in adults. Nitrous oxide potentiates compensatory rise in heart rate. Halothane
neuromuscular blockade, but less so than the blunts this refl ex. Slowing of sinoatrial node
volatile agents. Th e concentration of nitrous conduction may result in a junctional rhythm or
oxide fl owing through a vaporizer can infl uence bradycardia. In infants, halothane decreases
cardiac output by a combination of decreased
CHAPTER 8 Inhalation Anesthetics 181

heart rate and depressed myocardial C. Cerebral


contractility. Halothane sensitizes the heart to By dilating cerebral vessels, halothane lowers
the arrhythmogenic eff ects of epinephrine, so cerebral vascular resistance and increases CBF.
that doses of epinephrine above 1.5 mcg/kg Autoregulation , the maintenance of constant
should be avoided. Although organ blood fl ow is CBF during changes in arterial blood pressure, is
redistributed, systemic vascular resistance is blunted. Concomitant rises in intracranial
unchanged. pressure can be prevented by establishing
hyperventilation prior to administration of
B. Respiratory
halothane. Cerebral activity is decreased, leading
Halothane typically causes rapid, shallow
to electroencephalographic slowing and modest
breathing. Th e increased respiratory rate is not
reductions in metabolic oxygen requirements.
enough to counter the decreased tidal volume,
D. Neuromuscular
so alveolar ventilation drops, and resting Paco 2 is
Halothane relaxes skeletal muscle and
elevated. Apneic threshold , the highest Pa co2
at which a patient remains apneic, also rises potentiates nondepolarizing neuromuscular-
blocking agents (NMBA). Like the other potent
because the difference between it and resting Pa
co 2 is not altered by general anesthesia. volatile anesthetics, it is a triggering agent of
malignant hyperthermia.
Similarly, halothane limits the increase in minute
ventilation that normally accompanies a rise in
E. Renal
Pa co2 . Halothane’s ventilatory eff ects are
Halothane reduces renal blood fl ow, glomerular
probably due to central (medullary depression)
fi ltration rate, and urinary output. Part of this
and peripheral (intercostal muscle dysfunction)
decrease can be explained by a fall in arterial
mechanisms. Th ese changes are exaggerated by
blood pressure and cardiac output. Because the
preexisting lung disease and attenuated by
reduction in renal blood fl ow is greater than the
surgical stimulation. Th e increase in Pa co 2 and
reduction in glomerular fi ltration rate, the fi
the decrease in intrathoracic pressure that
ltration fraction is increased. Preoperative
accompany spontaneous ventilation with
hydration limits these changes.
halothane partially reverse the depression in
cardiac output, arterial blood pressure, and heart F. Hepatic
rate described above. Hypoxic drive is severely Halothane causes hepatic blood fl ow to
depressed by even low concentrations of decrease in proportion to the depression of
halothane (0.1 MAC). cardiac output. Hepatic artery vasospasm has
Halothane is considered a potent been reported during halothane anesthesia. Th e
bronchodilator, as it oft en reverses asthma- metabolism and clearance of some drugs (eg,
induced bronchospasm. Th is action is not fentanyl, phenytoin, verapamil) seem to be
inhibited by β-adrenergic blocking agents. impaired by halothane. Other evidence of
Halothane attenuates airway refl exes and hepatic cellular dysfunction includes
relaxes bronchial smooth muscle by inhibiting sulfobromophthalein (BSP) dye retention and
intracellular calcium mobilization. Halothane also minor liver transaminase elevations.
depresses clearance of mucus from the
respiratory tract (mucociliary function),
promoting postoperative hypoxia and atelectasis.
Biotransformation & Toxicity
Halothane is oxidized in the liver by a particular
isozyme of CYP (2EI) to its principal metabolite,
trifl uoroacetic acid. Th is metabolism can be
182 SECTION II Clinical Pharmacology

inhibited by pretreatment with disulfi ram. ed by trifl uoroacetic acid as the triggering
Bromide, another oxidative metabolite, has been antigens (trifl uoroacetylated liver proteins such
incriminated in (but is an improbable cause of) as microsomal carboxylesterase). As with
postanesthetic changes in mental status. In the halothane, other inhalational agents that
absence of oxygen, reductive metabolism may undergo oxidative metabolism can likewise lead
result in a small amount of hepatotoxic end to hepatitis. However, newer agents undergo
products that covalently bind to tissue little to no metabolism, and therefore do not
macromolecules. Th is is more apt to occur form trifl uroacetic acid protein adducts or
following enzyme induction by phenobarbital. produce the immune response leading to
Elevated fl uoride levels signal signifi cant hepatitis.
anaerobic metabolism.
Postoperative hepatic dysfunction has Contraindications
several causes: viral hepatitis, impaired hepatic
It is prudent to withhold halothane from patients
perfusion, preexisting liver disease, hepatocyte
with unexplained liver dysfunction following
hypoxia, sepsis, hemolysis, benign postoperative
previous anesthetic exposure.
intrahepatic cho-
Halothane, like all inhalational anesthetics,
should be used with care in patients with
8 lestasis, and drug-induced hepatitis. “
intracranial mass lesions because of the
Halothane hepatitis” is extremely rare (1 per
possibility of intracranial hypertension secondary
35,000 cases). Patients exposed to multiple
to increased cerebral blood volume and blood fl
halothane anesthetics at short intervals, middle-
ow.
aged obese women, and persons with a familial
Hypovolemic patients and some patients
predisposition to halothane toxicity or a personal
with severe reductions in left ventricular
history of toxicity are considered to be at
function may not tolerate halothane’s negative
increased risk. Signs are mostly related to hepatic
inotropic eff ects. Sensitization of the heart to
injury, such as increased serum alanine and
catecholamines limits the usefulness of
aspartate transferase, elevated bilirubin (leading
halothane when exogenous epinephrine is
to jaundice), and encephalopathy.
administered or in patients with
Th e hepatic lesion seen in humans—
pheochromocytoma.
centrilobular necrosis—also occurs in rats
pretreated with an enzyme inducer
(phenobarbital) and exposed to halothane under
hypoxic conditions (F io 2 < 14%). Th is halothane
hypoxic model implies hepatic damage from
reductive metabolites or hypoxia.
More likely evidence points to an immune
mechanism. For instance, some signs of the
disease indicate an allergic reaction (eg,
eosinophilia, rash, fever) and do not appear until
a few days aft er exposure. Furthermore, an
antibody that binds to hepatocytes previously
exposed to halothane has been isolated from
patients with halothane-induced hepatic
dysfunction. Th is antibody response may involve
liver microsomal proteins that have been modifi
CHAPTER 8 183
Drug Interactions anesthetics, except that tachypnea is less
Th e myocardial depression seen with halothane pronounced. Th e net eff ect is a more
pronounced fall in minute ventilation. Even low
is exacerbated by β-adrenergic-blocking agents
levels of isofl urane (0.1 MAC) blunt the normal
and calcium channel-blocking agents. Tricyclic
ventilatory response to hypoxia and hypercapnia.
antidepressants and monoamine oxidase Inhalation Anesthetics
inhibitors have been associated with fl uctuations
in blood pressure and arrhythmias, although
neither represents an absolute contraindication. Despite a tendency to irritate upper airway refl
Th e combination of halothane and aminophylline exes, isofl urane is considered a good
has resulted in serious ventricular arrhythmias. bronchodilator, but may not be as potent a
bronchodilator as halothane.

C. Cerebral
ISOFLURANE At concentrations greater than 1 MAC, isofl urane
Physical Properties increases CBF and intracranial pressure. Th ese eff
Isofl urane is a nonfl ammable volatile anesthetic ects are thought to be less pronounced than with
with a pungent ethereal odor. Although it is a halothane and are reversed by hyperventilation. In
chemical isomer with the same molecular weight contrast to halothane, the hyperventilation does
as enfl urane, it has diff erent physicochemical not have to be instituted prior to the use of isofl
properties (see urane to prevent intracranial hypertension. Isofl
Table 8–3 ). urane reduces cerebral metabolic oxygen
requirements, and at 2 MAC, it produces an
Eff ects on Organ Systems electrically silent electroencephalogram (EEG).
A. Cardiovascular D. Neuromuscular
Isofl urane causes minimal left ventricular Isofl urane relaxes skeletal muscle.
depression in vivo. Cardiac output is maintained
by a rise in heart rate due to partial preservation E. Renal
of carotid barorefl exes. Mild β-adrenergic Isofl urane decreases renal blood fl ow,
stimulation increases skeletal muscle blood fl ow, glomerular fi ltration rate, and urinary output.
decreases systemic vascular resistance, and lowers
arterial blood pressure. Rapid increases in isofl
F. Hepatic
urane concentration lead to transient increases in Total hepatic blood fl ow (hepatic artery and
heart rate, arterial blood pres- portal vein fl ow) may be reduced during isofl
urane anesthesia. Hepatic oxygen supply is better
maintained with isofl urane than with halothane,
9 sure, and plasma levels of norepinephrine.
however, because hepatic artery perfusion is
Isofl urane dilates coronary arteries, but not
preserved. Liver function tests are usually not aff
nearly as potently as nitroglycerin or adenosine.
ected.
Dilation of normal coronary arteries could
theoretically divert blood away from fi xed
stenotic lesions, which was the basis for concern Biotransformation & Toxicity
about coronary “steal” with this agent, a concern Isofl urane is metabolized to trifl uoroacetic acid.
that has largely been forgotten. Although serum fl uoride fl uid levels may rise,
nephrotoxicity is extremely unlikely, even in the
B. Respiratory presence of enzyme inducers. Prolonged sedation
Respiratory depression during isofl urane (>24 h at 0.1–0.6% isofl urane) of critically ill
anesthesia resembles that of other volatile patients has resulted in elevated plasma fl uoride
184 SECTION II Clinical Pharmacology
levels (15–50 µmol/L) without evidence of renal nitrous oxide (0.47). Although desfl urane is
impairment. Similarly, up to 20 MAC-hours of isofl roughly one-fourth as potent as the other volatile
urane may lead to fl uoride levels exceeding 50 agents, it is 17 times more potent than nitrous
µmol/L without detectable postoperative renal oxide. A high vapor pressure, an ultrashort
dysfunction. Its limited oxidative metabolism also duration of action, and moderate potency are the
minimizes any possible risk of signifi cant hepatic most characteristic features of desfl urane.
dysfunction.
Eff ects on Organ Systems
Contraindications A. Cardiovascular
Isofl urane presents no unique contraindications. Th e cardiovascular eff ects of desfl urane seem to
Patients with severe hypovolemia may not be similar to those of isofl urane. Increasing the
tolerate its vasodilating eff ects. It can trigger dose is associated with a decline in systemic
malignant hyperthermia. vascular resistance that leads to a fall in arterial
blood pressure. Cardiac output remains relatively
Drug Interactions unchanged or slightly depressed at 1–2 MAC. Th
Epinephrine can be safely administered in doses ere is a moderate rise in heart rate, central venous
up to 4.5 mcg/kg. Nondepolarizing NMBAs are pressure, and pulmonary artery pressure that oft
potentiated by isofl urane. en does not become

11 apparent at low doses. Rapid increases in


DESFLURANE desfl urane concentration lead to transient but
sometimes worrisome elevations in heart rate,
Physical Properties blood pressure, and catecholamine levels that are
Th e structure of desfl urane is very similar to that more pronounced than occur with isofl urane,
of isofl urane. In fact, the only diff erence is the particularly in patients with cardiovascular
substitution of a fl uorine atom for isofl urane’s disease. Th ese cardiovascular responses to rapidly
chlorine atom. Th at “minor” change has profound increasing desfl urane concentration can be
eff ects on the physical properties of the drug, attenuated by fentanyl, esmolol, or clonidine.
however. For instance, because the vapor
pressure of desfl urane at 20°C is 681 mm Hg, at B. Respiratory
high altitudes (eg, Denver, Colorado) it boils at Desfl urane causes a decrease in tidal volume and
room temperature. Th is problem necessitated the an increase in respiratory rate. Th ere is an overall
development of a special decrease in alveolar ventilation that causes a rise
in resting Pa co 2 . Like other modern volatile
10 desfl urane vaporizer. F urthermore, the low anesthetic agents, desfl urane depresses the
solubility of desfl urane in blood and body tis- ventilatory response to increasing Pa co 2 .
Pungency and airway irritation during desfl urane
sues causes a very rapid induction and emergence
induction can be manifested by salivation, breath-
of anesthesia. Th erefore, the alveolar
holding, coughing, and laryngospasm. Airway
concentration of desfl urane approaches the
resistance may increase in children with reactive
inspired concentration much more rapidly than
airway susceptibility. Th ese problems make desfl
the other volatile agents, giving the
urane a poor choice for inhalation induction.
anesthesiologist tighter control over anesthetic
levels. Wakeup times are approximately 50% less C. Cerebral
than those observed following isofl urane . Th is Like the other volatile anesthetics, desfl urane
is principally attributable to a blood/gas partition directly vasodilates the cerebral vasculature,
coeffi cient (0.42) that is even lower than that of increasing CBF, cerebral blood volume, and
CHAPTER 8 185
intracranial pressure at normotension and percutaneous loss. Desfl urane, more than other
normocapnia. Countering the decrease in cerebral volatile anesthetics, is degraded by desiccated CO
vascular resistance is a marked decline in the 2 absorbent (particularly barium hydroxide lime,

cerebral metabolic rate of oxygen (CMRO 2 ) that but also sodium and potassium hydroxide) into
tends to cause cerebral vasoconstriction and potentially clinically signifi cant levels of carbon
moderate any increase in CBF. Th e cerebral monoxide. Carbon monoxide poisoning is diffi cult
vasculature remains responsive to changes in Pa to diagnose under general anesthesia, but the
co2 , however, so that intracranial pressure can be presence of carboxyhemoglobin may be
lowered by hyperventilation. Cerebral oxygen detectable by arterial blood gas analysis or lower
consumption is decreased during desfl urane than expected pulse oximetry readings (although
anesthesia. Th us, during periods of desfl urane- still falsely high). Disposing of dried out absorbent
induced hypotension (mean arterial pressure = 60 or use of calcium hydroxide can minimize the risk
mm Hg), CBF is adequate to maintain aerobic of carbon monoxide poisoning.
metabolism despite a low cerebral perfusion
pressure. Th e eff ect on the EEG is similar to that Contraindications
of isofl urane. Initially, EEG frequency is increased, Desfl urane shares many of the contraindications
but as anesthetic depth is increased, EEG slowing of other modern volatile anesthetics: severe
becomes manifest, leading to burst suppression at hypovolemia, malignant hyperthermia, and
higher inhaled concentrations. intracranial hypertension.
Inhalation Anesthetics
D. Neuromuscular
Desfl urane is associated with a dose-dependent
decrease in the response to train-of-four and Drug Interactions
tetanic peripheral nerve stimulation.
Desfl urane potentiates nondepolarizing
E. Renal neuromuscular blocking agents to the same extent
Th ere is no evidence of any signifi cant as isofl urane. Epinephrine can be safely
nephrotoxic eff ects caused by exposure to desfl administered in doses up to 4.5 mcg/kg as desfl
urane. However, as cardiac output declines, urane does not sensitize the myocardium to the
decreases in urine output and glomerular fi arrhythmogenic eff ects of epinephrine. Although
ltration should be expected with desfl urane and emergence is more rapid following desfl urane
all other anesthetics. anesthesia than aft er isofl urane anesthesia,
switching from isofl urane to desfl urane toward
F. Hepatic the end of anesthesia does not signifi cantly
Hepatic function tests are generally unaff ected by accelerate recovery, nor does faster emergence
desfl urane, assuming that organ perfusion is translate into faster discharge times from the
maintained perioperatively. Desfl urane postanesthesia care unit. Desfl urane emergence
undergoes minimal metabolism, therefore the risk has been associated with delirium in some
of anesthetic-induced hepatitis is likewise pediatric patients.
minimal. As with isofl urane and sevofl urane,
hepatic oxygen delivery is generally maintained.
SEVOFLURANE
Biotransformation & Toxicity Desfl urane
Physical Properties
undergoes minimal metabolism in humans. Serum
and urine inorganic fl uoride levels following desfl Like desfl urane, sevofl urane is halogenated with
urane anesthesia are essentially unchanged from fl uorine. Sevofl urane’s solubility in blood is
preanesthetic levels. Th ere is insignifi cant slightly greater than des fl urane (λ b/g 0.65 versus
0.42)
186 SECTION II Clinical Pharmacology
D. Neuromuscular
12 (see T able 8–3) . N onpungency and rapid Sevofl urane produces adequate muscle
increases in alveolar anesthetic concentration relaxation for intubation of children following an
make sevofl urane an excellent choice for smooth inhalation induction.
and rapid inhalation inductions in pediatric and
adult patients. In fact, inhalation induction with E. Renal
4% to 8% sevofl urane in a 50% mixture of nitrous Sevofl urane slightly decreases renal blood fl ow.
oxide and oxygen can be achieved within 1 min. Its metabolism to substances associated with
Likewise, its low blood solubility results in a rapid impaired renal tubule function (eg, decreased
fall in alveolar anesthetic concentration upon concentrating ability) is discussed below.
discontinuation and a more rapid emergence
compared with isofl urane (although not an earlier F. Hepatic
discharge from the post-anesthesia care unit). Sevofl urane decreases portal vein blood fl ow,
Sevofl urane’s modest vapor pressure permits the but increases hepatic artery blood fl ow, thereby
use of a conventional variable bypass vaporizer. maintaining total hepatic blood fl ow and oxygen
delivery. It is generally not associated with
Eff ects on Organ Systems immune-mediated anesthetic hepatotoxicity
A. Cardiovascular
Sevofl urane mildly depresses myocardial Biotransformation & Toxicity
contractility. Systemic vascular resistance and Th e liver microsomal enzyme P-450 (specifi cally
arterial blood pressure decline slightly less than the 2E1 isoform) metabolizes sevofl urane at a
with isofl urane or desfl urane. Because sevofl rate onefourth that of halothane (5% versus 20%),
urane causes little, if any, rise in heart rate, but 10 to 25 times that of isofl urane or desfl
cardiac output is not maintained as well as with urane and may be induced with ethanol or
isofl urane or desfl urane. Sevofl urane may phenobarbital pretreatment. Th e potential
prolong the QT interval, the clinical signifi cance of nephrotoxicity of the resulting rise in inorganic fl
which is unknown. QT prolongation may be uoride (F − ) was discussed earlier. Serum fl uoride
manifest 60 min following anesthetic emergence concentrations exceed 50 µmol/L in approximately
in infants. 7% of patients who receive sevofl urane, yet
clinically signifi cant renal dysfunction has not
B. Respiratory been associated with sevofl urane anesthesia. Th e
Sevofl urane depresses respiration and reverses overall rate of sevofl urane metabolism is 5%, or
bronchospasm to an extent similar to that of isofl 10 times that of isofl urane. Nonetheless, there
urane. has been no association with peak fl uoride levels
following sevofl urane and any renal concentrating
C. Cerebral abnormality.
Similar to isofl urane and desfl urane, sevofl urane Alkali such as barium hydroxide lime or soda
causes slight increases in CBF and intracranial lime (but not calcium hydroxide) can degrade
pressure at normocarbia, although some studies sevofl urane, producing another proven (at least in
show a decrease in cerebral blood fl ow. High rats) nephrotoxic end product ( compound A , fl
concentrations of sevofl urane (>1.5 MAC) may uoromethyl-2,2-difluoro-1-[trifluoromethyl]vinyl
impair autoregulation of CBF, thus allowing a drop ether). Accumulation of compound A increases
in CBF during hemorrhagic hypotension. Th is eff with increased respiratory gas temperature, lowfl
ect on CBF autoregulation seems to be less ow anesthesia, dry barium hydroxide absorbent
pronounced than with isofl urane. Cerebral (Baralyme), high sevofl urane concentrations, and
metabolic oxygen requirements decrease, and anesthetics of long duration.
seizure activity has not been reported.
CHAPTER 8 187
Most studies have not associated sevofl competing with glycine at the glycine binding site.
urane with any detectable postoperative Xenon seems to have little eff ect on
impairment of renal function that would indicate cardiovascular, hepatic, or renal systems and has
toxicity or injury. Nonetheless, some clinicians been found to be protective against neuronal
recommend that fresh gas fl ows be at least 2 ischemia. As a natural element, it has no eff ect
L/min for anesthetics lasting more than a few upon the ozone layer compared with another
hours and that sevofl urane not be used in NMDA antagonist, nitrous oxide. Cost and limited
patients with preexisting renal dysfunction. availability have prevented its widespread use.
Sevofl urane can also be degraded into
hydrogen fl uoride by metal and environmental
impurities present in manufacturing equipment,
glass bottle packaging, and anesthesia equipment.
SUGGESTED READING
Banks P, Franks N, Dickinson R: Competitive inhibition at
Hydrogen fl uoride can produce an acid burn on
the glycine site of the N-methyl-d-aspartate receptor
contact with respiratory mucosa. Th e risk of
mediates xenon neuroprotection against hypoxia
patient injury has been substantially reduced by ischemia. Anesthesiology 2010;112:614.
inhibition of the degradation process by adding Bantel C, Maze M, Trapp S: Neuronal preconditioning by
water to sevofl urane during the manufacturing inhalational anesthetics. Anesthesiology
process and packaging it in a special plastic 2009;11:986.
container. Th e manufacturer has also distributed Cittanova M-L, Lelongt B, Verpont M-C: Fluoride ion
a “Dear Provider” letter warning of isolated toxicity in human kidney collecting duct cells.
incidents of fi re in the respiratory circuits of Anesthesiology 1996;84:428.
anesthesia machines with desiccated CO 2 Coburn M, Maze M, Franks N: Th e neuroprotective
absorbent when sevofl urane was used. eff ects of xenon and helium in an in vitro model
of traumatic brain injury. Crit Care Med
2008;36:588.
Contraindications De Hert S, Preckel B, Schlack W: Update on inhalational
Contraindications include severe hypovolemia, anaesthetics. Curr Opin Anaesthesiol 2009;22:491.
susceptibility to malignant hyperthermia, and DiMaggio C, Sun L, Li G: Early childhood exposure to
intracranial hypertension. anesthesia and risk of developmental and behavioral
disorders in a sibling birth cohort. Anesth Analg
Drug Interactions 2011; 113:1143.
Like other volatile anesthetics, sevofl urane Ebert TJ: Myocardial ischemia and adverse cardiac
potentiates NMBAs. It does not sensitize the heart outcomes in cardiac patients undergoing noncardiac
to catecholamine-induced arrhythmias. Inhalation Anesthetics

surgery with sevofl urane and isofl urane. Anesth


XENON Analg 1997;85:993.
Xenon is a “noble” gas that has long been known Eger EI 2nd, Bowland T, Ionescu P, et al: Recovery and
to have anesthetic properties. It is an inert kinetic characteristics of desfl urane and sevofl urane
in volunteers aft er 8-h exposure, including kinetics
element that does not form chemical bonds.
of degradation products. Anesthesiology
Xenon is scavenged from the atmosphere through
1997;87:517.
a costly distillation process. It is an odorless, Eger EI 2nd, Raines DE, Shafer SL, Hemmings HC Jr,
nonexplosive, naturally occurring gas with a MAC Sonner JM: Is a new paradigm needed to explain how
of .71 and a blood/gas coeffi cient of 0.115, giving inhaled anesthetics produce immobility? Anesth
it very fast onset and emergence parameters. As Analg 2008;107: 832.
previously mentioned, xenon’s anesthetic eff ects
seem to be mediated by NMDA inhibition by
188 SECTION II Clinical Pharmacology
Ghatge S, Lee J, Smith I: Sevofl urane: an ideal agent for
adult day-case anesthesia? Acta Anaesthesiol Scand
2003;47:917.
Ishizawa Y: General anesthetic gases and the global
environment. Anesth Analg 2011; 112:213.
Jevtovic-Todorovic V: Pediatric anesthesia
neurotoxicity: an overview of the 2011 Smart Tots
panel. Anesth Analg 2011;113:965.
Jordan BD, Wright EL: Xenon as an anesthetic agent.
AANA J 2010;78:387.
Loeckinger A, Kleinsasser A, Maier S, et.al: Sustained
prolongation of the QTc interval aft er anesthesia
with sevofl urane in infants during the fi rst 6 months
of life.
Anesthesiology 2003;98:639.
Njoku D, Laster MJ, Gong DH: Biotransformation of
halothane, enfl urane, isofl urane, and desfl urane to
trifl uoroacetylated liver proteins: association
between protein acylation and hepatic injury. Anesth
Analg 1997;84:173.
Preckel B, Weber N, Sanders R, et al: Molecular
mechanisms transducing the anesthetic analgesic
and organ protective actions of Xenon.
Anesthesiology 2006;105:187.
Stratmann G: Neurotoxicity of anesthetic drugs in the
developing brain. Anesth Analg 2011;113:1170.
Summors AC, Gupta AK, Matta BF: Dynamic cerebral
autoregulation during sevofl urane anesthesia: a
comparison with isofl urane. Anesth Analg
1999;88:341.
Sun X, Su F, Shi Y, Lee C: Th e “second gas eff ect” is not
a valid concept. Anesth Analg 1999;88:188.
Th omas J, Crosby G, Drummond J, et.al: Anesthetic
neurotoxicity: a diffi cult dragon to slay. Anesth
Analg 2011;113;969.
Torri G: Inhalational anesthetics: a review. Minerva
Anestesiol 2010;76:215.
Wang L, Traystman R, Murphy S: Inhalational agents in
ischemic brain. Curr Opin Pharmacol 2008;8:104.
Wei H: Th e role of calcium dysregulation in anesthetic
mediated neurotoxicity. Anesth Analg 2011;113:972.
Intravenous Anesthetics 9 C H

P
E
A

T
R
KEYCONCEPTS anesthesia with EMLA (eutectic mixture of local
anesthetic) cream, LMX (plain lidocaine cream 4%
and 5%), or 2% lidocaine jelly has increased the
1 Repetitive administration of barbiturates ease of intravenous inductions in children.
(eg, infusion of thiopental for “barbiturate Maintenance of general anesthesia is
coma” and brain protection) saturates the 4 In contrast to other anesthetic agents,
peripheral compartments, minimizing any ketamine increases arterial blood pressure,
eff ect of redistribution, and rendering the heart rate, and cardiac output, particularly
duration of action more dependent on after rapid bolus injections.
elimination. This is an example of context
5 Induction doses of etomidate transiently
sensitivity.
inhibit enzymes involved in cortisol and
2 Barbiturates constrict the cerebral aldosterone synthesis. Etomidate was
vasculature, causing a decrease in cerebral often used in the past for ICU sedation
blood fl ow, cerebral blood volume, and before reports of its consistent ability to
intracranial pressure. produce adrenocortical suppression in that
3 Although apnea may be relatively circumstance appeared.
uncommon after benzodiazepine 6 P ropofol formulations can support the
induction, even small intravenous doses of growth of bacteria, so sterile technique
diazepam and midazolam have resulted in must be observed in preparation and
respiratory arrest. handling. Propofol should be administered
within 6 h of opening the ampule.

General anesthesia began with inhaled agents


but now can be induced and maintained with feasible with a total intravenous anesthesia
drugs that enter the patient through a wide range (TIVA) technique. Th is chapter focuses on the
of routes. Drug administration can be oral, rectal, intravenous agents used to produce hypnosis,
transdermal, transmucosal, intramuscular, or including barbiturates, benzodiazepines,
intravenous for the purpose of producing or ketamine, etomidate, and propofol.
enhancing an anesthetic state. Preoperative
sedation of adults is usually accomplished by way
of oral or intravenous routes. Induction of
general anesthesia in adults usually includes
intravenous drug administration. Eff ective topical
176 SECTION II Clinical Pharmacology

BARBITURATES potency than does a short straight chain.


Likewise, the phenyl group in pheno barbital is
Mechanisms of Action anticonvulsive, whereas the methyl group in
Barbiturates depress the reticular activating metho hexital is not. Replacing the oxygen at C 2 (
system in the brainstem, which controls multiple oxy barbiturates) with a sulfur atom ( thio
vital functions, including consciousness. In clinical barbiturates) increases lipid solubility. As a result,
concentrations, barbiturates more potently aff thiopental and thiamylal have a greater potency,
ect the function of nerve synapses than axons. Th more rapid onset of action, and shorter durations
eir primary of action (aft er a single “sleep dose”) than
175
pentobarbital. Th e sodium salts of the
barbiturates are water soluble but markedly

FIGURE 91 Barbiturates share the structure of barbituric acid and diff er in the C 2 , C 3 , and N 1 substitutions.
alkaline (pH of 2.5% thiopental >10) and
mechanism of action is believed to be through relatively unstable (2-week shelf-life for
2.5% thiopental solution). Concentrations greater
binding to the γ-aminobutyric acid type A (GABA A
than recommended cause an unacceptable
) receptor. Barbiturates potentiate the action of
incidence of pain on injection and venous
GABA in increasing the duration of openings of a
thrombosis.
chloridespecifi c ion channel.

Structure–Activity Relationships Pharmacokinetics


Barbiturates are derived from barbituric acid ( A. Absorption
Figure 9–1 ). Substitution at carbon C 5 In clinical anesthesiology, thiopental, thiamylal,
determines hypnotic potency and anticonvulsant and methohexital were frequently administered
activity. A longbranched chain conveys more intravenously for induction of general anesthesia
CHAPTER 9 177
in adults and children (prior to the introduction of typically lose consciousness within 30 s and
propofol). Rectal thiopental or, more oft en, awaken within 20 min.
methohexital has been used for induction in Th e minimal induction dose of thiopental
children, and intramuscular (or oral) will depend on body weight and age. Reduced
pentobarbital was oft en used in the past for induction doses are required for elderly patients
premedication of all age groups. primarily due to slower redistribution. In contrast
to the rapid initial distribution half-life of a few
B. Distribution minutes, elimination of thiopental is prolonged
Th e duration of sleep doses of the highly lipid- (elimination half-life ranges of 10–12 h). Th
soluble barbiturates (thiopental, thiamylal, and iamylal and methohexital have similar distribution
methohexital) is determined by redistribution, patterns, whereas less lipid-soluble barbiturates
not by metabolism or elimination. For example, have much longer distribution half-lives and
although thiopental is highly protein bound durations of action aft er a sleep dose. Repetitive
(80%), its great lipid solubility and high
nonionized fraction (60%)
1 administration of barbiturates (eg, infusion of
100 thiopental for “barbiturate coma” and brain
Plasma protection) saturates the peripheral
MG
75 compartments,
Intravenous Anesthetics
% of dose

VRG
50

FG
minimizing any eff ect of redistribution, and
25 rendering the duration of action more dependent
on elimination. Th is is an example of context
sensitivity.
0.1 1.0 10 100
Time (min) C. Biotransformation
Barbiturates are principally biotransformed via
FIGURE 92 Distribution of thiopental from hepatic oxidation to inactive water-soluble
plasma to the vessel-rich group (VRG; brain, heart, metabolites. Because of greater hepatic
liver, kidney, endocrine glands), to the muscle group extraction, methohexital is cleared by the liver
(MG), and fi nally to the fat group (FG). (Modifi ed and more rapidly than thiopental. Although
reproduced, with permission, from Price HL et al: The uptake of redistribution is responsible for the awakening
thiopental by body tissues and its relation to the duration of
narcosis. Clin Pharmacol Ther 1960;1:16.) from a single sleep dose of any of these lipid-
soluble barbiturates, full recovery of
psychomotor function is more rapid following
account for rapid brain uptake (within 30 s). If the methohexital due to its enhanced metabolism.
central compartment is contracted (eg,
D. Excretion
hypovolemic shock), if the serum albumin is low
Increased protein binding decreases barbiturate
(eg, severe liver disease or malnutrition), or if the
glomerular fi ltration, whereas increased lipid
nonionized fraction is increased (eg, acidosis),
solubility tends to increase renal tubular
larger brain and heart concentrations will be
reabsorption. Except for the less protein-bound
achieved for a given dose. Redistribution to the
and less lipid-soluble agents such as
peripheral compartment— specifi cally, the
phenobarbital, renal excretion is limited to water-
muscle group—lowers plasma and brain
soluble end products of hepatic
concentration to 10% of peak levels within 20–30
biotransformation. Methohexital is excreted in
min ( Figure 9–2 ). Th is pharmacokinetic profi le
the feces.
correlates with clinical experience—patients
178 SECTION II Clinical Pharmacology

Eff ects on Organ Systems Barbiturates incompletely depress airway refl ex


A. Cardiovascular responses to laryngoscopy and intubation, and
airway instrumentation may lead to
Intravenous bolus induction doses of
bronchospasm (in asthmatic patients) or
barbiturates cause a decrease in blood pressure
laryngospasm in lightly anesthetized patients.
and an increase in heart rate. Hemodynamic
responses to barbiturates are reduced by slower
C. Cerebral
rates of induction. Depression of the medullary
vasomotor center produces vasodilation of Barbiturates constrict the cerebral
2
peripheral capacitance vessels, which increases
vasculature, causing a decrease in cerebral
peripheral pooling of blood, mimicking a reduced
blood
blood volume. Tachycardia following
administration is probably due to a central fl ow, cerebral blood volume, and intracranial
vagolytic eff ect and refl ex responses to pressure. Intracranial pressure decreases to a
decreases in blood pressure. Cardiac output is oft greater extent than arterial blood pressure, so
en maintained by an increased heart rate and cerebral perfusion pressure (CPP) usually
increased myocardial contractility from increases. (CPP equals cerebral artery pressure
compensatory baroreceptor refl exes. minus the greater of jugular venous pressure or
Sympathetically induced vasoconstriction of intracranial pressure.) Barbiturates induce a
resistance vessels (particularly with intubation greater decline in cerebral oxygen consumption
under light planes of general anesthesia) may (up to 50% of normal) than in cerebral blood fl
actually increase peripheral vascular resistance. ow; therefore the decline in cerebral blood fl ow
However, in situations where the baroreceptor is not detrimental. Barbiturate-induced
response will be blunted or absent (eg, reductions in oxygen requirements and cerebral
metabolic activity are mirrored by changes in the
hypovolemia, congestive heart failure, β-
electroencephalogram (EEG), which progress
adrenergic blockade), cardiac output and
from low-voltage fast activity with small doses to
arterial blood pressure may fall dramatically due
highvoltage slow activity, burst suppression, and
to uncompensated peripheral pooling of blood
electrical silence with larger doses. Barbiturates
and direct myocardial depression . Patients with
may protect the brain from transient episodes of
poorly controlled hypertension are particularly
focal ischemia (eg, cerebral embolism) but
prone to wide swings in blood pressure during
probably do not protect from global ischemia (eg,
anesthesia induction. Th e cardiovascular eff ects
cardiac arrest). Abundant animal data document
of barbiturates therefore vary markedly,
these eff ects but the clinical data are sparse and
depending on rate of administration, dose,
inconsistent. Furthermore, thiopental doses
volume status, baseline autonomic tone, and
required to maintain EEG suppression (most oft
preexisting cardiovascular disease. A slow rate of
en burst suppression or fl at line) are associated
injection and adequate preoperative hydration
with prolonged awakening, delayed extubation,
attenuates or eliminates these changes in most
and the need for inotropic support.
patients.
Th e degree of central nervous system
B. Respiratory depression induced by barbiturates ranges from
mild sedation to unconsciousness, depending on
Barbiturates depress the medullary ventilatory
center, decreasing the ventilatory response to the dose administered ( Table 9–1 ). Some
patients relate a taste sensation of garlic, onions,
hypercapnia and hypoxia. Deep barbiturate
sedation oft en leads to upper airway obstruction; or pizza during induction with thiopental.
Barbiturates do not impair the perception of pain.
apnea oft en follows an induction dose. During
awakening, tidal volume and respiratory rate are In fact, they sometimes appear to lower the pain
threshold. Small doses occasionally cause a state
decreased following barbiturate induction.
of excitement and disorientation that can be
CHAPTER 9 179
disconcerting when sedation is the objective. potentiating the organ system eff ects of a given
Barbiturates do not produce muscle relaxation, dose.
and some induce involuntary skeletal muscle Ethanol, opioids, antihistamines, and other
contractions (eg, methohexital). Relatively small central nervous system depressants potentiate
doses of thiopental (50–100 mg intravenously) the sedative eff ects of barbiturates. Th e
rapidly (but temporarily) control most grand mal common clinical
seizures. Unfortunately, acute tolerance and Intravenous Anesthetics

physiological dependence on the sedative eff ect


of barbiturates develop quickly.
impression that chronic alcohol abuse is
D. Renal associated with increased thiopental
Barbiturates reduce renal blood fl ow and requirements during induction lacks scientifi c
glomerular fi ltration rate in proportion to the fall proof.
in blood pressure.

E. Hepatic
Hepatic blood fl ow is decreased. Chronic BENZODIAZEPINES
exposure to barbiturates has opposing eff ects on Mechanisms of Action
drug biotransformation. Induction of hepatic Benzodiazepines bind the same set of receptors
enzymes increases the rate of metabolism of in the central nervous system as barbiturates but
some drugs, whereas binding of barbiturates to bind to a diff erent site on the receptors.
the cytochrome P-450 enzyme system interferes Benzodiazepine binding to the GABA A receptor
with the biotransformation of other drugs (eg, increases the frequency of openings of the
tricyclic antidepressants). Barbiturates promote associated chloride ion channel. For example,
aminolevulinic acid synthetase, which stimulates benzodiazepine-receptor binding facilitates
the formation of porphyrin (an intermediary in binding of GABA to its receptor. Flumazenil (an
heme synthesis). Th is may precipitate acute imidazobenzodiazepine) is a specifi c
intermittent porphyria or variegate porphyria in benzodiazepine–receptor antagonist that eff
susceptible individuals. ectively reverses most of the central nervous
system eff ects of benzodiazepines (see Chapter
F. Immunological
17).
Anaphylactic or anaphylactoid allergic reactions
are rare. Sulfur-containing thiobarbiturates evoke
mast cell histamine release in vitro, whereas Structure–Activity Relationships
oxybarbiturates do not. For this reason, some Th e chemical structure of benzodiazepines
anesthesiologists prefer induction agents other includes a benzene ring and a seven-member
than thiopental or thiamylal in asthmatic or diazepine ring ( Figure 9–3 ). Substitutions at
atopic patients, but the evidence for this choice is various positions on these rings aff ect potency
sparse. Th ere is no question that airway and biotransformation. Th e imidazole ring of
instrumentation with light anesthesia is midazolam contributes to its water solubility at
troublesome in patients with reactive airways. low pH. Diazepam and lorazepam are insoluble in
water so parenteral preparations contain
Drug Interactions propylene glycol, which can produce venous
Contrast media, sulfonamides, and other drugs irritation.
that occupy the same protein-binding sites as
thiopental may displace the barbiturate,
increasing the amount of free drug available and
Diazepam Lorazepam Flumazenil
CH3 N
O O COOC2H5
180 SECTION II Clinical
N Pharmacology N N
OH
N N N
Cl Cl F
CH3
Cl O

Midazolam

CH3 CH3 N
N

N N
pH < 6.0 CH2NH2
N O
Cl pH > 6.0 Cl C
F
F

(lipid-soluble) (water-soluble)

Pharmacokinetics TABLE 92 Uses and doses of commonly used


A. Absorption benzodiazepines.
Benzodiazepines are commonly administered Agent Use Route 1 Dose (mg/kg)
orally, intramuscularly, and intravenously to Diazepam Premedication Oral 0.2–0.52
provide sedation or, less commonly, to induce Sedation IV 0.04–0.2
general anesthesia ( Table 9–2 ). Diazepam and Midazolam Premedication IM 0.07–0.15
lorazepam are well absorbed from the Sedation IV 0.01–0.1
gastrointestinal tract, with peak plasma levels Induction IV 0.1–0.4
usually achieved in 1 and 2 h, respectively. Oral Lorazepam Premedication Oral 0.05
midazolam has not been approved by the U.S.
with permission, from White PF: Pharmacologic and clinical aspects of
Food and Drug Administration, nevertheless this preoperative medication. Anesth Analg 1986;65:963. With permission
route of administration has been popular for from the International Anesthesia Research Society.)

pediatric premedication. Likewise, intranasal


(0.2– 0.3 mg/kg), buccal (0.07 mg/kg), and
sublingual (0.1 mg/kg) midazolam provide eff B. Distribution
ective preoperative sedation. Diazepam is relatively lipid soluble and readily
FIGURE 93 The structures of commonly used penetrates the blood–brain barrier. Although
benzodiazepines and their antagonist, fl umazenil, share midazolam is water soluble at reduced pH, its
a seven-member diazepine ring. (Modifi ed and reproduced, imidazole ring closes at physiological pH,
increasing its lipid solubility (see Figure 9–3 ). Th
e moderate lipid solubility of lorazepam accounts
Intramuscular injections of diazepam are for its slower brain uptake and onset of action.
painful and unreliably absorbed. In contrast, Redistribution is fairly rapid for the
midazolam and lorazepam are well absorbed aft benzodiazepines (the initial distribution half-life is
er intramuscular injection, with peak levels 3–10 min) and, like the barbiturates, is
achieved in 30 and 90 min, respectively. Induction responsible for awakening. Although midazolam
of general anesthesia with midazolam is has been used as an induction agent, neither
convenient only with intravenous administration. midazolam nor any other of the benzodiazepines
can match the rapid onset and short duration of
1 IV, intravenous; IM,
intramuscular. 2 Maximum dose
is 15 mg.
CHAPTER 9 181
action of propofol or even thiopental. All three
benzodiazepines are highly protein bound (90–
98%).

C. Biotransformation
Th e benzodiazepines rely on the liver for
biotransformation into water-soluble
glucuronidated end products. Th e phase I
metabolites of diazepam are pharmacologically
active.
182 SECTION II Clinical Pharmacology

Slow hepatic extraction and a large volume of B. Respiratory


distribution ( Vd ) result in a long elimination half- Benzodiazepines depress the ventilatory response
life for diazepam (30 h). Although lorazepam also to CO 2 . Th is depression is usually insignifi cant
has a low hepatic extraction ratio, its lower lipid unless the drugs are administered intravenously
solubility limits its V d , resulting in a shorter or in association with other respiratory
elimination half-life (15 h). Nonetheless, the depressants. Although
clinical duration of lorazepam is oft en quite
prolonged due to increased receptor affi nity. Th 3 apnea may be relatively uncommon aft er
ese diff erences between lorazepam and diazepam benzodiazepine induction, even small
illustrate the low utility of individual intravenous doses of diazepam and
pharmacokinetic halflives in guiding clinical midazolam have resulted in respiratory
practice. Midazolam shares diazepam’s Vd , but arrest. Th e steep dose–response curve,
its elimination half-life (2 h) is the shortest of the slightly prolonged onset (compared with
group because of its increased hepatic extraction propofol or thiopental), and potency of
ratio. midazolam necessitate careful titration to
avoid overdosage and apnea. Ventilation
D. Excretion
must be monitored in all patients receiving
Th e metabolites of benzodiazepine intravenous benzodiazepines, and
biotransformation are excreted chiefl y in the resuscitation equipment must be
urine. Enterohepatic circulation produces a immediately available.
secondary peak in diazepam plasma concentration
6–12 h following administration. Kidney failure C. Cerebral
may lead to prolonged sedation in patients Benzodiazepines reduce cerebral oxygen
receiving larger doses of midazolam due to the consumption, cerebral blood fl ow, and
accumulation of a conjugated metabolite (α- intracranial pressure but not to the extent the
hydroxymidazolam). barbiturates do. Th ey are eff ective in preventing
and controlling grand mal seizures. Oral sedative
Eff ects on Organ Systems doses oft en produce antegrade amnesia, a useful
A. Cardiovascular premedication property. Th e mild muscle-relaxing
Th e benzodiazepines display minimal property of these drugs is mediated at the spinal
cardiovascular depressant eff ects even at general cord level, not at the neuromuscular junction. Th e
anesthetic doses, except when they are antianxiety, amnestic, and sedative eff ects seen
coadministered with opioids (these agents at lower doses progress to stupor and
interact to produce myocardial depression and unconsciousness at induction doses. Compared
arterial hypotension). Benzodiazepines given with propofol or thiopental, induction with
alone decrease arterial blood pressure, cardiac benzodiazepines is associated with a slower rate
output, and peripheral vascular resistance slightly, of loss of consciousness and a longer recovery.
and sometimes increase heart rate. Intravenous Benzodiazepines have no direct analgesic
midazolam tends to reduce blood pressure and properties.
peripheral vascular resistance more than
diazepam. Changes in heart rate variability during Drug Interactions
midazolam sedation suggest decreased vagal tone Cimetidine binds to cytochrome P-450 and
(ie, drug-induced vagolysis). reduces the metabolism of diazepam.
Erythromycin inhibits metabolism of midazolam
and causes a two- to threefold prolongation and
CHAPTER 9 Intravenous Anesthetics 183

intensifi cation of its eff ects. Heparin displaces Ketamine Phencyclidine


diazepam from protein-binding sites and increases
the free drug concentration.
O
As previously mentioned, the combination of
opioids and benzodiazepines markedly reduces Cl N
NHCH3
arterial blood pressure and peripheral vascular
resistance. Th is synergistic interaction has oft en
been observed in patients with ischemic or Etomidate Propofol
valvular heart disease who oft en receive O N
OH
benzodiazepines for premedication and during CH3CH2OC
N (CH3)2HC CH(CH3)2
induction of anesthesia with opioids.
Benzodiazepines reduce the minimum CH3CH
alveolar concentration of volatile anesthetics as
much as 30%.
Ethanol, barbiturates, and other central
nervous system depressants potentiate the FIGURE 94 The structures of ketamine, etomidate,
sedative eff ects of the benzodiazepines. and propofol. Note the similarities between ketamine and
phencyclidine.
KETAMINE of phencyclidine’s psychotomimetic eff ects.
Mechanisms of Action Ketamine is used for intravenous induction of
Ketamine has multiple eff ects throughout the anesthesia, particularly in settings where its
central nervous system, inhibiting polysynaptic tendency to produce sympathetic stimulation are
refl exes in the spinal cord as well as excitatory useful (hypovolemia, trauma). When intravenous
neurotransmitter eff ects in selected areas of the access is lacking, ketamine is useful for
brain. In contrast to the depression of the reticular intramuscular induction of general anesthesia in
activating system induced by the barbiturates, children and uncooperative adults. Ketamine can
ketamine functionally “dissociates” the thalamus be combined with other agents (eg, propofol or
(which relays sensory impulses from the reticular midazolam) in small bolus doses or infusions for
activating system to the cerebral cortex) from the deep conscious sedation during nerve blocks,
limbic cortex (which is involved with the endoscopy, etc. Even subanesthetic doses of
awareness of sensation). Clinically, this state of ketamine may cause hallucinogenic eff ects but
dissociative anesthesia may cause the patient to usually do not do so in clinical practice, where
appear conscious (eg, eye opening, swallowing, many patients will have received at least a small
muscle contracture) but unable to process or dose of midazolam (or a related agent) for
respond to sensory input. Ketamine has been amnesia and sedation. Th e increased anesthetic
demonstrated to be an N -methyl- d -aspartate potency and decreased psychotomimetic side eff
(NMDA) receptor (a subtype of the glutamate ects of one isomer (S[+] versus R[–]) are the result
receptor) antagonist. of stereospecifi c receptors. Th e single S(+)
stereoisomer preparation is not available in the
United States (but widely available throughout the
Structure–Activity Relationships world), and it has considerably greater affi nity
Ketamine ( Figure 9–4 ) is a structural analogue of than the racemic mixture for the NMDA receptor
phencyclidine (an anesthetic that has been used in as well as several-fold greater potency as a
veterinary medicine, and a drug of abuse). It is general anesthetic.
one-tenth as potent, yet retains many
184 SECTION II Clinical Pharmacology

1
IV, intravenous; IM, intramuscular.
Pharmacokinetics 2
Almost always in combination with propofol.
A. Absorption
Ketamine has been administered orally, nasally,
rectally, subcutaneously, and epidurally, but in
usual clinical practice it is given intravenously or anesthetic activity. Induction of hepatic enzymes
intramuscularly ( Table 9–3 ). Peak plasma levels only partially explains the tolerance that patients
are usually achieved within 10–15 min aft er who receive multiple doses of ketamine will
intramuscular injection. develop. Extensive hepatic uptake (hepatic
extraction ratio of 0.9) explains ketamine’s
B. Distribution relatively short elimination half-life (2 h).
Ketamine is more lipid soluble and less protein
bound than thiopental. Th ese characteristics, D. Excretion
along with ketamine-induced increase in cerebral End products of ketamine biotransformation are
blood fl ow and cardiac output, lead to rapid brain excreted renally.
uptake and subsequent redistribution (the Eff ects on Organ Systems
distribution half-life is 10–15 min). Awakening is A. Cardiovascular
due to redistribution from brain to peripheral
compartments. 4 In contrast to other anesthetic agents,
C. Biotransformation ketamine increases arterial blood pressure, heart
rate, and cardiac output ( Table 9–4 ), particularly
Ketamine is biotransformed in the liver to several
aft er rapid bolus injections. Th ese indirect
metabolites, one of which (norketamine) retains
cardiovascular eff ects are due to central
TABLE 93 Uses and doses of ketamine,
stimulation of the sympathetic nervous system
etomidate, and propofol.
and inhibition of the reuptake of norepinephrine
Agent Use Route 1 Dose
aft er release at nerve terminals. Accompanying
Ketamine Induction IV 1–2 mg/kg these changes are increases in pulmonary artery
IM 3–5 mg/kg
Sedation 2 IV 2.5–15 mcg/kg/min
pressure and myocardial work. For these reasons,
large bolus injections of ketamine should be
Etomidate Induction IV 0.2–0.5 mg/kg
administered cautiously in patients with coronary
Propofol Induction IV 1–2.5 mg/kg artery disease, uncontrolled hypertension,
Maintenance IV 50–200 mcg/kg/min congestive heart failure, or arterial aneurysms. Th
infusion e direct myocardial depressant eff ects of large
Sedation IV 25–100 mcg/kg/min doses of ketamine, probably due to inhibition of
infusion
calcium transients, are unmasked by sympathetic
blockade (eg, spinal cord transection) or
exhaustion of catecholamine stores (eg, severe
end-stage shock). On the other hand, ketamine’s
indirect stimulatory eff ects may be benefi cial to
patients with acute shock.

1 HR, heart rate; MAP, mean arterial pressure; Vent, ventilatory drive;
B’dil, bronchodilation; CBF, cerebral blood fl ow; CMRO 2 , cerebral
oxygen consumption; ICP, intracranial pressure; 0, no eff ect; 0/↑, no
change or mild increase; ↓, decrease (mild, moderate, marked); ↑,
increase (mild, moderate, marked).
2 Minimal change in CBF and ICP when coadministered with other agents
(see text).
CHAPTER 9 Intravenous Anesthetics 185

B. Respiratory and controlled ventilation, but not with nitrous


Ventilatory drive is minimally aff ected by oxide, ketamine is not associated with increased
induction doses of ketamine, although rapid intracranial pressure. Myoclonic activity is
intravenous bolus associated with increased subcortical electrical

TABLE 94 Summary of nonvolatile anesthetic eff ects on organ systems. 1


Cardiovascular Respiratory Cerebral

Agent HR MAP Vent B’dil CBF CMRO 2 ICP

Barbiturates
Thiopental ↓
Thiamylal ↑↑ ↑↑ ↓↓ ↓↓ ↓↓↓ ↓↓↓ ↓ ↓↓↓ ↓↓↓ ↓↓↓ ↓↓↓ ↓↓↓
Methohexital ↑↑ ↓↓ ↓↓↓ 0 ↓↓↓ ↓↓↓ ↓↓↓ ↓↓↓

Benzodiazepines
Diazepam 0/↑ ↓ 0
Lorazepam 0/↑ ↓ ↓↓ ↓↓ 0 ↓↓ ↓↓ ↓↓ ↓↓ ↓↓
Midazolam ↑ ↓↓ ↓↓ 0 ↓↓ ↓↓ ↓↓ ↓↓

Ketamine ↑↑ ↑↑ ↓ ↑↑↑ ↑↑ 2 ↑ ↑↑ 2

Etomidate 0 ↓ ↓ 0 ↓↓↓ ↓↓↓ ↓↓↓

Propofol 0 ↓↓ ↓↓↓ 0 ↓↓↓ ↓↓↓ ↓↓↓

administration or combinations of ketamine with activity, which is not apparent on surface EEG.
opioids occasionally produce apnea. Racemic Undesirable psychotomimetic side eff ects (eg,
ketamine is a potent bronchodilator, making it a disturbing dreams and delirium) during
good induction agent for asthmatic patients; emergence and recovery are less common in
however, S(+) ketamine produces minimal children and in patients premedicated with
bronchodilation. Upper airway refl exes remain benzodiazepines or those in whom ketamine is
largely intact, but partial airway obstruction may combined with propofol in a TIVA technique. Of
occur, and patients at increased risk for aspiration the nonvolatile agents, ketamine comes closest to
pneumonia (“full stomachs”) should be intubated being a “complete” anesthetic as it induces
during ketamine general anesthesia (see Case analgesia, amnesia, and unconsciousness.
Discussion, Chapter 17). Th e increased salivation
associated with ketamine can be attenuated by Drug Interactions
premedication with an anticholinergic agent such Ketamine interacts synergistically (more than
as glycopyrrolate additive) with volatile anesthetics but in an
additive way with propofol, benzodiazepines, and
C. Cerebral other GABA-receptor–mediated agents. In animal
Th e received dogma about ketamine is that it experiments nondepolarizing neuromuscular
increases cerebral oxygen consumption, cerebral blocking agents are minimally potentiated by
blood fl ow, and intracranial pressure. Th ese eff ketamine (see Chapter 11). Diazepam and
ects would seem to preclude its use in patients midazolam attenuate ketamine’s
with space-occupying intracranial lesions such as cardiostimulatory eff ects and diazepam prolongs
occur with head trauma; however, recent ketamine’s elimination half-life.
publications off er convincing evidence that when α -Adrenergic and β-adrenergic antagonists
combined with a benzodiazepine (or another (and other agents and techniques that diminish
agent acting on the same GABA receptor system)
186 SECTION II Clinical Pharmacology

sympathetic stimulation) unmask the direct B. Distribution


myocardial depressant eff ects of ketamine, which Although it is highly protein bound, etomidate is
are normally overwhelmed by sympathetic characterized by a very rapid onset of action due
stimulation. Concurrent infusion of ketamine and to its great lipid solubility and large nonionized
propofol, oft en in a fi xed infusion rate ratio of fraction at physiological pH. Redistribution is
1:10, has achieved great popularity for sedation responsible for decreasing the plasma
with local and regional anesthesia, particularly in concentration to awakening levels. Etomidate
offi ce-based settings. plasma kinetics are well explained by a two-
compartment model.

ETOMIDATE C. Biotransformation
Mechanisms of Action Hepatic microsomal enzymes and plasma
esterases rapidly hydrolyze etomidate to an
Etomidate depresses the reticular activating
inactive metabolite.
system and mimics the inhibitory eff ects of GABA.
Specifi cally, etomidate—particularly the R(+) D. Excretion
isomer— appears to bind to a subunit of the GABA Th e end products of etomidate hydrolysis are
A receptor, increasing the receptor’s affi nity for primarily excreted in the urine.
GABA. Unlike barbiturates, etomidate may have
disinhibitory eff ects on the parts of the nervous
Eff ects on Organ Systems
system that control extrapyramidal motor activity.
Th is disinhibition off ers a potential explanation A. Cardiovascular
for the 30–60% incidence of myoclonus with Etomidate has minimal eff ects on the
etomidate induction of anesthesia. cardiovascular system. A mild reduction in
peripheral vascular resistance is responsible for a
Structure–Activity Relationships slight decline in arterial blood pressure.
Myocardial contractility and cardiac output are
Etomidate contains a carboxylated imidazole and
usually unchanged. Etomidate does not release
is structurally unrelated to other anesthetic agents
histamine. However, etomidate by itself, even in
(see Figure 9–4 ). Th e imidazole ring provides
large doses, produces relatively light anesthesia
water solubility in acidic solutions and lipid
for laryngoscopy, and marked increases in heart
solubility at physiological pH. Th erefore
rate and blood pressure may be recorded when
etomidate is dissolved in propylene glycol for
etomidate provides the only anesthetic depth for
injection. Th is solution oft en causes pain on
intubation.
injection that can be lessened by a prior
intravenous injection of lidocaine. B. Respiratory
Ventilation is aff ected less with etomidate than
Pharmacokinetics with barbiturates or benzodiazepines. Even
A. Absorption induction doses usually do not result in apnea
Etomidate is available only for intravenous unless opioids have also been administered.
administration and is used primarily for induction
C. Cerebral
of general anesthesia (see Table 9–3). It is
Etomidate decreases cerebral metabolic rate,
sometimes used for brief production of deep
cerebral blood fl ow, and intracranial pressure.
(unconscious) sedation such as prior to placement
Because of minimal cardiovascular eff ects, CPP is
of retrobulbar blocks.
well maintained. Although changes on EEG
resemble those associated with barbiturates,
CHAPTER 9 Intravenous Anesthetics 187

etomidate increases the amplitude of mg/mL) is available for intravenous administration


somatosensory evoked potentials. Postoperative as an oil-in-water emulsion containing soybean oil,
nausea and vomiting are more common following glycerol, and egg lecithin. A history of egg allergy
etomidate than following propofol or barbiturate does not necessarily contraindicate the use of
induction. Etomidate lacks analgesic properties. propofol because most egg allergies involve a
reaction to egg white (egg albumin), whereas egg
D. Endocrine lecithin is extracted from egg yolk. Th is
formulation will oft en cause pain during injection
5 Induction doses of etomidate transiently inhibit that can be decreased by prior injection of
enzymes involved in cortisol and aldosterone lidocaine or less eff ectively by mixing lidocaine
synthesis. It was used in the past for sedation in with propofol prior to injection (2 mL of 1%
the intensive care unit (ICU) before reports of its lidocaine in 18 mL propo-
consistent ability to produce adrenocortical
suppression in that circumstance appeared. 6 fol). P ropofol formulations can support the
Longterm infusion and adrenocortical growth of bacteria, so sterile technique must be
suppression were associated with an increased observed in preparation and handling. Propofol
mortality rate in critically ill (particularly septic) should be administered within 6 h of opening the
patients. ampule. Sepsis and death have been linked to
contaminated propofol preparations. Current
Drug Interactions formulations of propofol contain 0.005% disodium
Fentanyl increases the plasma level and prolongs edetate or 0.025% sodium metabisulfi te to help
the elimination half-life of etomidate. Opioids retard the rate of growth of microorganisms;
decrease the myoclonus characteristic of an however, these additives do not render the
etomidate induction. product “antimicrobially preserved” under United
States Pharmacopeia standards.

PROPOFOL
Pharmacokinetics
Mechanisms of Action
A. Absorption
Propofol induction of general anesthesia may
Propofol is available only for intravenous
involve facilitation of inhibitory neurotransmission
administration for the induction of general
mediated by GABA A receptor binding. Propofol
anesthesia and for moderate to deep sedation
allosterically increases binding affi nity of GABA
(see Table 9–3).
for the GABA A receptor. Th is receptor, as
previously noted, is coupled to a chloride channel, B. Distribution
and activation of the receptor leads to Propofol has a rapid onset of action. Awakening
hyperpolarization of the nerve membrane. from a single bolus dose is also rapid due to a very
Propofol (like most general anesthetics) binds short initial distribution half-life (2–8 min). Most
multiple ion channels and receptors. Propofol investigators believe that recovery from propofol
actions are not reversed by the specifi c is more rapid and is accompanied by less
benzodiazepine antagonist fl umazenil. “hangover” than recovery from methohexital,
thiopental, ketamine, or etomidate. Th is makes it
Structure–Activity Relationships a good agent for outpatient anesthesia. A smaller
Propofol consists of a phenol ring substituted with induction dose is recommended in elderly patients
two isopropyl groups (see Figure 9–4 ). Propofol is because of their smaller V d . Age is also a key
not water soluble, but a 1% aqueous solution (10 factor determining required propofol infusion
188 SECTION II Clinical Pharmacology

rates for TIVA. In countries other than the United bradycardia. Changes in heart rate and cardiac
States, a device called the Diprifusor is oft en used output are usually transient and insignifi cant in
to provide target (concentration) controlled healthy patients but may be severe in patients at
infusion of propofol. Th e user must enter the the extremes of age, those receiving β-adrenergic
patient’s age and weight and the desired target blockers, or those with impaired ventricular
concentration. Th e device uses these data, a function. Although myocardial oxygen
microcomputer, and standard pharmacokinetic consumption and coronary blood fl ow usually
parameters to continuously adjust the infusion decrease comparably, coronary sinus lactate
rate. production increases in some patients, indicating
some mismatch between myocardial oxygen
C. Biotransformation supply and demand.
Th e clearance of propofol exceeds hepatic blood
fl ow, implying the existence of extrahepatic B. Respiratory
metabolism. Th is exceptionally high clearance Propofol is a profound respiratory depressant that
rate probably contributes to relatively rapid usually causes apnea following an induction dose.
recovery aft er continuous infusions. Conjugation Even when used for conscious sedation in
in the liver results in inactive metabolites that are subanesthetic doses, propofol inhibits hypoxic
eliminated by renal clearance. Th e ventilatory drive and depresses the normal
pharmacokinetics of propofol do not appear to be response to hypercarbia. As a result, only properly
aff ected by obesity, cirrhosis, or kidney failure. educated and qualifi ed personnel should
Use of propofol infusion for long-term sedation of administer propofol for sedation. Propofol-
children who are critically ill or young adult induced depression of upper airway refl exes
neurosurgical patients has been associated with exceeds that of thiopental, allowing intubation,
sporadic cases of lipemia, metabolic acidosis, and endoscopy, or laryngeal mask placement in the
death, the so-termed propofol infusion syndrome. absence of neuromuscular blockade. Although
propofol can cause histamine release, induction
D. Excretion with propofol is accompanied by a lower incidence
Although metabolites of propofol are primarily of
excreted in the urine, chronic kidney failure does
not aff ect clearance of the parent drug.

Eff ects on Organ Systems


A. Cardiovascular
Th e major cardiovascular eff ect of propofol is a
decrease in arterial blood pressure due to a drop
in systemic vascular resistance (inhibition of
sympathetic vasoconstrictor activity), preload, and
cardiac contractility. Hypotension following
induction is usually reversed by the stimulation
accompanying laryngoscopy and intubation.
Factors associated with propofol-induced
hypotension include large doses, rapid injection,
and old age. Propofol markedly impairs the
normal arterial barorefl ex response to
hypotension. Rarely, a marked drop in preload
may lead to a vagally mediated refl ex
CHAPTER 9 189
wheezing in asthmatic and nonasthmatic patients showing that it produces more complete amnesia
compared with barbiturates or etomidate. and better conscious sedation for endoscopy than
midazolam plus fentanyl. It has a slower onset and
C. Cerebral Intravenous Anesthetics
Propofol decreases cerebral blood fl ow and
intracranial pressure. In patients with elevated
intracranial pressure, propofol can cause a critical slower recovery than propofol, off ering little
reason for anesthesiologists to use fospropofol in
reduction in CPP (<50 mm Hg) unless steps are
place of propofol. Th e place (if any) of fospropofol
taken to support mean arterial blood pressure.
relative to other competing agents has not yet
Propofol and thiopental probably provide a similar
been established in clinical practice.
degree of cerebral protection during experimental
focal ischemia. Unique to propofol are its
antipruritic properties. Its antiemetic eff ects CASE DISCUSSION
(requiring a blood propofol concentration of 200
ng/mL) provide yet another reason for it to be a Premedication of the Surgical Patient
preferred drug for outpatient anesthesia. An extremely anxious 17-year-old woman
Induction is occasionally accompanied by presents for dilation and curettage. She demands
excitatory phenomena such as muscle twitching, to be asleep before going to the operating room
spontaneous movement, opisthotonus, or and does not want to remember anything .
hiccupping. Although these reactions may
occasionally mimic tonic–clonic seizures, propofol What are the goals of administering preoperative
medication?
has anticonvulsant properties and has been used
successfully to terminate status epilepticus. Anxiety is a normal response to impending
Propofol may be safely administered to epileptic surgery. Diminishing anxiety is usually the major
patients. Propofol decreases intraocular pressure. goal of preoperative medication. For many
Tolerance does not develop aft er long-term patients, the preoperative interview with the
propofol infusions. Propofol is an uncommon anesthesiologist allays fears more eff ectively than
agent of physical dependence or addiction; sedative drugs. Preoperative medication may also
however, both anesthesia personnel and provide relief of preoperative pain or perioperative
medically untrained individuals have died while amnesia .
using propofol inappropriately to induce sleep in There may also be specifi c medical indications
for preoperative medication: prophylaxis against
nonsurgical settings.
postoperative nausea and vomiting (5-HT 3 s) and
against aspiration pneumonia (eg, antacids),
Drug Interactions prevention of allergic reactions (eg,
Fentanyl and alfentanil concentrations may be antihistamines), or decreasing upper airway
increased with concomitant administration of secretions (eg, anticholinergics). The goals of
propofol. Many clinicians administer a small preoperative medication depend on many factors,
amount of midazolam (eg, 30 mcg/kg) prior to including the health and emotional status of the
induction with propofol; midazolam can reduce patient, the proposed surgical procedure, and the
the required propofol dose by more than 10%. anesthetic plan. For this reason, the choice of
anesthetic premedication must be individualized
and must follow a thorough preoperative
FOSPROPOFOL evaluation .
Fospropofol is a water-soluble prodrug that is Do all patients require preoperative medication?
metabolized in vivo to propofol, phosphate, and No—customary levels of preoperative anxiety
formaldehyde. It has been released in the United do not harm most patients. Some patients dread
States and other countries based on studies intramuscular injections, and others fi nd altered
190 SECTION II Clinical Pharmacology
states of consciousness more unpleasant than duration (90 min), but intravenous midazolam has an
nervousness. If the surgical procedure is brief, the even better pharmacokinetic profi le .
eff ects of some sedatives may extend into the Which factors must be considered in selecting the
postoperative period and prolong recovery time. anesthetic premedication for this patient?
This is particularly troublesome for patients
First, it must be made clear to the patient that
undergoing ambulatory surgery. Specifi c
in most centers, lack of necessary equipment and
contraindications for sedative premedication
concern for patient safety preclude anesthesia
include severe lung disease, hypovolemia,
being induced in the preoperative holding room.
impending airway obstruction, increased
Long-acting agents such as morphine or lorazepam
intracranial pressure, and depressed baseline
are poor choices for an outpatient procedure.
mental status. Premedication with sedative drugs
Diazepam can also aff ect mental function for
should never be given before informed consent has
several hours. One alternative is to establish an
been obtained .
intravenous line in the preoperative holding area
Which patients are most likely to benefi t from and titrate small doses of midazolam using slurred
preoperative medication?
speech as an end point. At that time, the patient
Some patients are quite anxious despite the can be taken to the operating room. Vital signs—
preoperative interview. Separation of young children particularly respiratory rate—must be continuously
from their parents is often a traumatic ordeal, monitored .
particularly if they have endured multiple prior
surgeries. Medical conditions such as coronary artery
disease or hypertension may be aggravated by
psychological stress . SUGGESTED READING
How does preoperative medication infl uence the Domino EF: Taming the ketamine tiger. Anesthesiology
induction of general anesthesia? 2010;113:678.
Some medications often given preoperatively (eg, Garnock-Jones KP, Scott LJ: Fospropofol. Drugs
opioids) decrease anesthetic requirements and can 2010;70:469.
smooth induction. However, intravenous Jensen LS, Merry AF, Webster CS, et al: Evidence-based
administration of these medications just prior to strategies for preventing drug administration errors
induction is a more reliable method of achieving the during anaesthesia. Anaesthesia 2004;59:493.
Leslie K, Clavisi O, Hargrove J: Target-controlled infusion
same benefi ts .
versus manually-controlled infusion of propofol for
What governs the choice among the preoperative general anaesthesia or sedation in adults. Cochrane
medications commonly administered? Database Syst Rev 2008;(3):CD006059.
After the goals of premedication have been Raeder J: Ketamine, revival of a versatile intravenous
determined, the clinical eff ects of the agents dictate anaesthetic. Adv Exp Med Biol 2003;523:269.
choice. For instance, in a patient experiencing Short TG, Young R: Toxicity of intravenous anaesthetics.
preoperative pain from a femoral fracture, the Best Pract Res Clin Anaesthesiol 2003;17:77.
Vanlersberghe C, Camu F: Etomidate and other
analgesic eff ects of an opioid (eg, fentanyl, morphine,
nonbarbiturates. Handb Exp Pharmacol 2008;
hydromorphone) will decrease the discomfort
(182):267.
associated with transportation to the operating room
Vanlersberghe C, Camu F: Propofol. Handb Exp
and positioning on the operating room table. On the
Pharmacol 2008;(182):227.
other hand, respiratory depression, orthostatic
hypotension, and nausea and vomiting may result
from opioid premedication . Analgesic Agents
Benzodiazepines relieve anxiety, often provide
amnesia, and are relatively free of side eff ects;
however, they are not analgesics. Diazepam and
lorazepam are available orally. Intramuscular KEYCONCEPTS
midazolam has a rapid onset (30 min) and short
1 The accumulation of morphine of opioids are required for postoperative
metabolites analgesia.
(morphine 3-glucuronide and morphine 6- 4 The neuroendocrine stress response to
glucuronide) in patients with kidney failure surgical stimulation is measured in terms
has been associated with narcosis and of the secretion of specifi c hormones,
ventilatory depression lasting several days. including catecholamines, antidiuretic
2 Rapid administration of larger doses of hormone, and cortisol. Large doses of
opioids (particularly fentanyl, sufentanil, opioids block the release of these
remifentanil, and alfentanil) can induce hormones in response to surgery more
chest wall rigidity severe enough to completely than volatile anesthetics.
prevent adequate bag-and-mask 5 Aspirin is unique in that it irreversibly
ventilation. inhibits COX-1 by acetylating a serine
3 Prolonged dosing of opioids can produce residue in the enzyme. The irreversible
“opioid-induced hyperalgesia,” in which nature of its inhibition underlies the nearly
patients become more sensitive to painful 1-week duration of its clinical eff ects (eg,
stimuli. Infusion of large doses of (in return of platelet aggregation to normal)
particular) remifentanil during general after drug discontinuation.
anesthesia can produce acute tolerance, in
which much larger than usual doses

OPIOIDS
Regardless of how expertly surgical and Mechanisms of Action
anesthetic procedures are performed, appropriate Opioids bind to specifi c receptors located
prescription of analgesic drugs, especially opioids throughout the central nervous system and other
and cyclooxygenase (COX) inhibitors, can make tissues. Four major opioid receptor types have
the diff erence between a satisfi ed and an been identifi ed ( Table 10–1 ): mu (µ, with
unsatisfi ed postoperative patient. Studies have subtypes µ 1 and µ 2 ), kappa (κ), delta (δ), and
shown that outcomes can be improved when
sigma (σ). All opioid receptors couple to G
analgesia is provided in a “multimodal” format
proteins; binding of an agonist to an opioid
(typically emphasizing COX inhibitors and local
receptor causes membrane hyperpolarization.
anesthetic techniques while minimizing opioid
Acute opioid eff ects are mediated by inhibition of
use) as one part of a welldefi ned and well-
adenylyl cyclase (reductions in intracellular cyclic
organized plan for postoperative care (see Chapter
48).

189

A
10 P T E
C
192 SECTION II Clinical Pharmacology

TABLE 101 Classifi cation of opioid receptors. butorphanol, and pentazocine) have less effi cacy
1
than so-called full agonists (eg, fentanyl) and
Receptor Clinical Eff ect Agonists under some circumstances will antagonize the
actions of full agonists. Th e pure opioid
µ Supraspinal Morphine
antagonists are discussed in Chapter 17.
analgesia (µ 1 ) Met-enkephalin 2

Respiratory depression β-Endorphin 2 Th e opioid drugs mimic endogenous


(µ2 ) Fentanyl compounds. Endorphins, enkephalins, and
Physical dependence dynorphins are endogenous peptides that bind to
Muscle rigidity opioid receptors. Th ese three families of opioid
κ Sedation Morphine peptides diff er in their amino acid sequences,
Spinal analgesia Nalbuphine anatomic distributions, and receptor affi nities.
Butorphanol
Opioid receptor activation inhibits the
Dynorphin 2
Oxycodone
presynaptic release and postsynaptic response to
excitatory neurotransmitters (eg, acetylcholine,
δ Analgesia Leu-enkephalin 2
Behavioral β-Endorphin 2
substance P) from nociceptive neurons . Th e
Epileptogenic cellular mechanism for this action was described
at the beginning of this chapter. Transmission of
σ Dysphoria Pentazocine
Hallucinations Nalorphine pain impulses can be selectively modifi ed at the
Respiratory stimulation Ketamine level of the dorsal horn of the spinal cord with
intrathecal or epidural administration of opioids.
1
Note: The relationships among receptor, clinical eff ect, and agonist Opioid receptors also respond to systemically
are more complex than indicated in this table. For example,
pentazocine is an antagonist at µ receptors, a partial agonist at κ administered opioids. Modulation through a
receptors, and an agonist at σ receptors. 2 Endogenous opioid. descending inhibitory pathway from the
periaqueductal gray matter to the dorsal horn of
the spinal cord may also play a role in opioid
adenosine monophosphate concentrations) and analgesia. Although opioids exert their greatest eff
activation of phospholipase C. Opioids inhibit ect within the central nervous system, opiate
voltage-gated calcium channels and activate receptors have also been identifi ed on somatic
inwardly rectifying potassium channels. Opioid eff and sympathetic peripheral nerves. Certain opioid
ects vary based on the duration of exposure, and side eff ects (eg, depression of gastrointestinal
opioid tolerance leads to changes in opioid motility) are the result of opioid binding to
responses. receptors in peripheral tissues (eg, the wall of the
Although opioids provide some degree of gastrointestinal tract), and there are now selective
sedation and (in many species) can produce antagonists for opioid actions outside the central
general anesthesia when given in large doses, they nervous system (alvimopan and oral naltrexone).
are principally used to provide analgesia. Th e Th e distribution of opioid receptors on axons of
properties of specifi c opioids depend on which primary sensory nerves and the clinical
receptor is bound (and in the case of spinal and importance of these receptors (if present) remains
epidural administration of opioids, the location in speculative, despite the persisting practice of
the neuraxis where the receptor is located) and compounding of opioids in local anesthetic
the binding affi nity of the drug. Agonist– solutions applied to peripheral nerves.
antagonists (eg, nalbuphine, nalorphine,
CHAPTER 10 193

Structure–Activity
Relationships
Opioid receptor binding is a property shared by a
chemically diverse group of compounds.
Nonetheless, there are common structural
characteristics, which are shown in Figure 10–1 .
As is true for most classes of drugs, small
molecular changes can convert an agonist into an
antagonist. Th e levorotatory isomers are
generally more potent than the dextrorotatory
opioid isomers.
194 SECTION II Clinical Pharmacology

FIGURE 101 Opioid agonists and antagonists share hydrocodone (most oft Nonionized Pr
part of their chemical structure, which is outlined in cyan. en in combination with Agent Fraction Bi
reservoir of drug in the acetaminophen),
Morphine ++
upper dermis delays by codeine, tramadol,
Pharmacokinetics several hours the Meperidine +
morphine,
A. Absorption achievement of eff hydromorphone, and Fentanyl +
Rapid and complete ective blood methadone. Th ese
absorption follows the concentrations. Serum agents are much used
intramuscular injection Sufentanil ++ +
concentrations of for outpatient pain
of hydromorphone, fentanyl reach a management.
morphine, or plateau within 14–24 h Fentanyl is oft en Alfentanil ++++ +
meperidine, with peak of application (with administered in small Remifentanil +++
plasma levels usually peak levels occurring doses (10–25 mcg)
reached aft er 20–60
1
+ , very low; + +, low; + + +,
aft er a longer delay in with local anesthetics high; + + + +, very high.
min. Oral elderly than in younger for spinal anesthesia,
transmucosal fentanyl patients) and remain and adds to the
citrate absorption constant for up to 72 analgesia when
(fentanyl “lollipop”) epidural morphine
h. Continued included with local
provides rapid onset of (DepoDur) is
absorption from the anesthetics in epidural
analgesia and sedation administered as a
dermal reservoir infusions. Morphine in
in patients who are single epidural dose
accounts for persisting doses between 0.1 and
not good candidates (5–15 mg), the eff ects
measurable serum 0.5 mg and
for conventional oral, of which persist for 48
levels many hours aft hydromorphone in
intravenous, or h.
er patch removal. doses between 0.05
intramuscular dosing Fentanyl patches are and 0.2 mg provide
of opioids. B. Distribution
most oft en used for 12–18 hours of
Th e low Table 10–2
outpatient analgesia aft er
molecular weight and summarizes the
management of intrathecal administrat
high lipid solubility of physical characteristics
chronic pain and are ion.
fentanyl also favor that determine
particularly Morphine and
transdermal distribution and tissue
appropriate for hydromorphone are
absorption (the binding of opioid
patients who require commonly included in
transdermal fentanyl analgesics. Aft er
continuous opioid local anesthetic
“patch”). Th e amount intravenous
dosing but cannot take solutions infused for
of fentanyl absorbed administration, the
the much less postoperative epidural
per unit of time distribution half-lives
expensive, but equally analgesia. Extended-
depends on the of all of the opioids are
effi cacious, oral release TABLE 102 fairly rapid (5–20 min).
surface area of skin agents such as Physical Th e low fat solubility
covered by the patch methadone. characteristics of of morphine slows
and also on local skin A wide variety of opioids that passage across the
conditions (eg, blood fl opioids are eff ective determine blood–brain barrier,
ow). Th e time by oral administration, distribution. 1 however, so that its
required to establish a including oxycodone,
CHAPTER 10 195

onset of action is slow prior accumulation of


and its duration of other drugs, increased
action is prolonged. Th by a history of tobacco
is contrasts with the use, and decreased by
increased lipid concurrent inhalation
solubility of fentanyl anesthetic
and sufentanil, which administration.
are associated with a
faster onset and
shorter duration of
action when
administered in small
doses . Interestingly,
alfentanil has a more
rapid onset of action
and shorter duration
of action than fentanyl
following a bolus
injection, even though
it is less lipid soluble
than fentanyl. Th e
high nonionized
fraction of alfentanil at
physiological pH and
its small volume of
distribution ( V d )
increase the amount
of drug (as a
percentage of the
administered dose)
available for binding in
the brain.
Signifi cant
amounts of lipid-
soluble opioids can be
retained by the lungs
(fi rst-pass uptake); as
systemic
concentrations fall
they will return to the
bloodstream. Th e
amount of pulmonary
uptake is reduced by
196 SECTION II Clinical Pharmacology

Unbinding of opioid receptors and redistribution esmolol) by nonspecifi c esterases in red blood
(of drug from eff ect sites) terminate the clinical cells and tissue (see Figure 10–1), yielding a
eff ects of all opioids. Aft er smaller doses of the terminal elimination half-life of less than 10 min.
lipid-s oluble drugs (eg, fentanyl or sufentanil), Remifentanil biotransformation is rapid and the
redistribution alone is the driver for reducing duration of a
blood concentrations, whereas aft er larger doses
biotransformation becomes an important driver in 100

Time to 50% drop (min)


reducing plasma levels below those that have Fentanyl
clinical eff ects. Th us, the time required for 75
Alfentanil
fentanyl or sufentanil concentrations to decrease
by half is context sensitive; in other words, the 50
half-time depends on the total dose of drug and Sufentanil
25
duration of exposure (see Chapter 7).
Remifentanil
C. Biotransformation 0
100 200 300 400 500 600
With the exception of remifentanil, all opioids Infusion duration (min)
depend primarily on the liver for
biotransformation and are metabolized by the
FIGURE 102 In contrast to other opioids, the time
cytochrome P (CYP) system, conjugated in the
necessary to achieve a 50% decrease in the plasma
liver, or both. Because of the high hepatic
concentration of remifentanil (its context-sensitive half-
extraction ratio of opioids, their clearance time ) is very short and is not infl uenced by the duration
depends on liver blood fl ow. Th e small V d of of the infusion. (Reproduced, with permission, from
alfentanil contributes to a short elimination half- Egan TD: The pharmacokinetics of the new short-acting opioid
life (1.5 h). Morphine and hydromorphone remifentanil [GI87084B] in healthy adult male volunteers.
Anesthesiology 1993;79:881.)
undergo conjugation with glucuronic acid to form,
in the former case, morphine 3-glucuronide and
morphine 6-glucuronide, and in the latter case, remifentanil infusion has little eff ect on wake-up
hydromorphone 3-glucuronide. Meperidine is N time ( Figure 10–2 ). Th e context-sensitive
-demethylated to normeperidine, an active halftime of remifentanil remains approximately 3
metabolite associated with seizure activity, min regardless of the dose or duration of infusion.
particularly aft er very large meperidine doses. Th In its lack of accumulation remifentanil diff ers
e end products of fentanyl, sufentanil, and from other currently available opioids. Hepatic
alfentanil are inactive. Norfentanyl, the dysfunction requires no adjustment in
metabolite of fentanyl, can be measured in urine remifentanil dosing. Finally, patients with
long aft er the native compound is no longer pseudocholinesterase defi ciency have a normal
detectable in blood to determine chronic fentanyl response to remifentanil (as also appears true for
ingestion. Th is has its greatest importance in esmolol).
diagnosing fentanyl abuse.
D. Excretion
Codeine is a prodrug that becomes active aft
Th e end products of morphine and meperidine
er it is metabolized by CYP to morphine. Tramadol
biotransformation are eliminated by the kidneys,
similarly must be metabolized by CYP to O
with less than 10% undergoing biliary excretion.
-desmethyltramadol to be active. Oxycodone is
Because 5–10% of morphine is excreted
metabolized by CYP to series of active compounds
unchanged in the urine, kidney failure prolongs
that are less potent than the parent one.
morphine duration of
Th e ester structure of remifentanil makes it
susceptible to hydrolysis (in a manner similar to
CHAPTER 10 Analgesic Agents 197

of polypharmacy can include myocardial


1 action. Th e accumulation of morphine depression.
metabolites (morphine 3-glucuronide and
morphine 6-glucuronide) in patients with kidney B. Respiratory
failure has been associated with prolonged Opioids depress ventilation, particularly
narcosis and ventilatory depression. In fact, respiratory rate. Th us, monitoring of respiratory
morphine 6-glucuronide is a more potent and rate provides a convenient, simple way of
longer-lasting opioid agonist than morphine. As detecting early respiratory depression in patients
previously noted, normeperidine at increased receiving opioid analgesia. Opioids increase the
concentrations may produce seizures; these are partial pressure of carbon dioxide (Pa co 2 ) and
not reversed by naloxone. Renal dysfunction blunt the response to a CO 2 challenge, resulting in
increases the likelihood of toxic eff ects from a shift of the CO 2 response curve downward and
normeperidine accumulation. However, both to the right ( Figure 10–3 ). Th ese eff ects result
morphine and meperidine have been used safely from opioid binding to neurons in the respiratory
and successfully in patients with kidney failure. centers of the brainstem. Th e apneic threshold—
Metabolites of sufentanil are excreted in urine and the greatest Pa co2 at which a patient remains
bile. Th e main metabolite of remifentanil is apneic—rises, and hypoxic drive is decreased .
eliminated in urine, is several thousand times less Morphine and meperidine can cause histamine-
potent than its parent compound, and thus is induced bronchospasm in susceptible
unlikely to produce any clinical opioid eff ects.
2 patients. Rapid administration of larger doses
Eff ects on Organ Systems of opioids (particularly fentanyl, suf-
A. Cardiovascular entanil, remifentanil, and alfentanil) can induce
chest wall rigidity severe enough to prevent
In general, opioids have few direct eff ects on the
adequate bag-and-mask ventilation . Th is
heart. Meperidine tends to increase heart rate (it
centrally
is structurally similar to atropine and was
in which case drugs such and H 2 antagonists,
originally synthesized as an atropine
as sufentanil and or both. Th e end eff
replacement), whereas larger doses of morphine,
fentanyl can be ects of histamine
fentanyl, sufentanil, remifentanil, and alfentanil
associated with reduced release can be
are associated with a vagus nerve– mediated
cardiac output. Bolus reversed by infusion
bradycardia. With the exception of meperidine
doses of meperidine, of intravenous fl uid
(and only then at very large doses), the opioids do
hydromorphone, and and vasopressors.
not depress cardiac contractility provided they are
morphine evoke Intraoperative
administered alone (which is almost never the
histamine release in hypertension during
circumstance in surgical anesthetic settings).
some individuals that largedose opioid
Nonetheless, arterial blood pressure oft en falls as
can lead to profound anesthesia or nitrous
a result of bradycardia, venodilation, and
drops in systemic oxide–opioid
decreased sympathetic refl exes, sometimes
vascular resistance and anesthesia is
requiring vasopressor support. Th ese eff ects are
arterial blood pressure. common. Such
more pronounced when opioids are administered
Th e potential hazards of hypertension is oft
in combination with benzodiazepines, stability
histamine release can be en attributed to
provided by opioids is greatly diminished in actual
minimized in susceptible inadequate
practice when other anesthetic drugs, including
patients by infusing anesthetic depth,
nitrous oxide, benzodiazepines, propofol, or
opioids slowly or by thus it is
volatile agents, are typically added. Th e end result
pretreatment with H 1 conventionally
198 30 SECTION II Clinical Pharmacology

25
Alveolar ventilation (L/min)

— but transient and however, some of


20 almost certainly these apparent
unimportant— increases seizures have been
15
in cerebral artery blood retrospectively
fl ow velocity and diagnosed as severe
intracranial pressure opioid-induced
10
following opioid boluses muscle rigidity. EEG
in patients with brain activation and
5
tumors or head trauma. seizures have been
If combined with associated with the
40 50 60 70 hypotension, the meperidine
PaCO2
resulting fall in cerebral metabolite
treated by the addition response to airway perfusion pressure normeperidine, as
of other anesthetic stimulation such as could be deleterious to previously noted.
agents occurs during patients with abnormal Stimulation of
(benzodiazepines, tracheal intubation. intracranial pressure– the medullary
propofol, or potent volume relationships. chemoreceptor
inhaled agents). If depth Nevertheless, the trigger zone is
C. Cerebral
of anesthesia is important clinical responsible for
Th e eff ects of
adequate and message is that any opioid-induced
opioids on cerebral
hypertension persists, trivial opioid-induced nausea and
perfusion and
vasodilators or other increase in intracranial vomiting. Curiously,
intracranial pressure
antihypertensives may pressure would likely be nausea and vomiting
must be separated
be used. Th e inherent much less important are more common
from any eff ects of
cardiac than the much more following smaller
opioids on Pa co2 . In
FIGURE 103 likely large increases in (analgesic) than very
general, opioids
Opioids depress intracranial pressure large (anesthetic)
reduce cerebral
ventilation. This is associated with doses of opioids.
oxygen
graphically displayed intubation that might be Prolonged oral
consumption,
by a shift of the CO 2 observed in an dosing of opioids or
curve downward and cerebral blood fl ow,
inadequately infusion of large
to the right. cerebral blood
anesthetized patient doses of remifentanil
mediated muscle volume, and
(from whom opioids during general
contraction is eff intracranial pressure,
were withheld). Opioids anesthesia can
ectively treated with but to a much lesser
usually have almost no produce the
neuromuscular blocking extent than
eff ects on the phenomenon of
agents. Th is problem is barbiturates,
electroencephalogram opioid-induced
rarely seen now that propofol, or
(EEG), although large tolerance. Repeated
large-dose opioid benzodiazepines. Th
doses are associated dosing of opioids will
anesthesia is less oft en ese eff ects will
with slow δ-wave reliably produce
used in cardiovascular occur during
activity. Th ere are tolerance, a
anesthesia practice. maintenance of
curious sporadic case phenomenon in
Opioids can eff ectively normocarbia by artifi
reports that large doses which larger doses
blunt the cial ventilation;
of fentanyl may rarely are required to
bronchoconstrictive however, there are
cause seizure activity; produce the same
some reports of mild
CHAPTER 10 Analgesic Agents 199

response. Th is is not the Unique among


same as physical the commonly used
dependence or opioids, meperidine
addiction, which may has minor local D. Gastrointestinal
also be associated with anesthetic qualities, Opioids slow
repeated opioid particularly when gastrointestinal
administration. administered into motility by binding
the subarachnoid to opioid receptors
3 Prolonged dosing of space. Meperidine’s in the gut and
opioids can also clinical use as a local reducing peristalsis.
produce anesthetic has been Biliary colic may
“opioid-induced limited by its result from opioid-
hyperalgesia,” in which relatively low induced contraction
patients become more potency and of the sphincter of
sensitive to painful propensity to cause Oddi. Biliary spasm,
stimuli. Infusion of large typical opioid side eff which can mimic a
doses of (in particular) ects (nausea, common bile duct
remifentanil during sedation, and stone on
general anesthesia can pruritus) at the cholangiography, is
produce acute doses required to reversed with the
tolerance, in which induce local opioid antagonist
much larger than usual anesthesia. naloxone or
doses of opioids will be Intravenous glucagon. Patients
required for meperidine (10–25
postoperative analgesia. mg) is more eff
Relatively large doses of ective than
opioids are required to morphine or
render patients fentanyl for
unconscious ( Table 10– decreasing shivering
3 ). Regardless of the in the
dose, however, opioids postanesthetic care
will not reliably produce unit and meperidine
amnesia. Parenteral appears to be the
opioids have been the best agent for this
mainstay of pain control indication .
for more than a century.
Th e relatively recent
use of opioids in
epidural and intrathecal
spaces has
revolutionized acute and
chronic pain
management (see
Chapters 47 and 48).
200 SECTION II Clinical Pharmacology

receiving long-term E. Endocrine anesthetics. Although is incompletely


opioid therapy (eg, for much discussed, the understood. (Th e
cancer pain) usually 4 Th e actual clinical outcome results of failure to
become tolerant to neuroendocrin benefi t produced by appreciate this drug
many of the side eff ects e stress attenuating the stress interaction in the
but rarely to response to response, even in high- celebrated Libby
constipation. Th is is the surgical risk cardiac patients, Zion case led to
basis for stimulation is remains speculative changes in work
TABLE 103 Uses and doses of common opioids.
Agent Use Route 1 Dose 2

Morphine Postoperative analgesia IM 0.05–0.2 mg/kg


IV 0.03–0.15 mg/kg

Hydromorphone Postoperative analgesia IM 0.02–0.04 mg/kg


IV 0.01–0.02 mg/kg

Fentanyl Intraoperative anesthesia IV 2–50 mcg/kg


Postoperative analgesia IV 0.5–1.5 mcg/kg

Sufentanil Intraoperative anesthesia IV 0.25–20 mcg/kg

Alfentanil Intraoperative anesthesia


Loading dose IV 8–100 mcg/kg
Maintenance infusion IV 0.5–3 mcg/kg/min

Remifentanil Intraoperative anesthesia


Loading dose IV 1.0 mcg/kg
Maintenance infusion IV 0.5–20 mcg/kg/min
Postoperative analgesia/sedation IV 0.05–0.3 mcg/kg/min
1 IM, intramuscular; IV, intravenous.
2 Note: The wide range of opioid doses refl ects a large therapeutic index and depends upon which other anesthetics are simultaneously
administered. For obese patients, dose should be based on ideal body weight or lean body mass, not total body weight. Tolerance can develop rapidly
(ie, within 2 h) during IV infusion of opioids, necessitating higher infusion rates. Dose correlates with other variables besides body weight that need to
be considered (eg, age). The relative potencies of fentanyl, sufentanil, and alfentanil are estimated to be 1:9:1/7.
the recent development measured in (and possibly rules for house offi
of the peripheral opioid terms of the nonexistent). cers in the United
antagonists secretion of specifi c States.)
methylnaltrexone and hormones, including Propofol,
Drug Interactions
alvimopan, and for their catecholamines, barbiturates,
Th e combination of
salutary eff ects in antidiuretic benzodiazepines,
meperidine and
promoting motility in hormone, and and other central
monoamine oxidase
patients with opioid cortisol. Large doses nervous system
inhibitors should be
bowel syndrome, those of opioids (typically depressants can
avoided as it may result
receiving chronic opioid fentanyl or have synergistic
in hypertension,
treatment of cancer sufentanil) block the cardiovascular,
hypotension,
pain, and those release of these respiratory, and
hyperpyrexia, coma, or
receiving intravenous hormones in sedative eff ects with
respiratory arrest. Th e
opioids aft er abdominal response to surgery opioids.
cause of this
surgery. more completely Th e
catastrophic interaction
than volatile biotransformation of
CHAPTER 10 Analgesic Agents 201

alfentanil may be part responsible for unique in that it from surgical


impaired following selective COX-2 irreversibly inhibits COX- procedures.
treatment with inhibition. Agents 1 by acetylating a serine
erythromycin, leading to that inhibit COX residue in the enzyme. Structure–
prolonged sedation and nonselectively (eg, Th e irreversible nature
respiratory depression. aspirin) will control of its inhibition underlies
Activity
fever, infl ammation, the nearly 1-week Relationships Th
pain, and duration of its clinical eff e COX enzyme is
CYCLOOXYGEN thrombosis. COX-2 ects (eg, return of inhibited by an
ASE INHIBITORS selective agents (eg, platelet aggregation to unusually diverse
acetaminophen normal) aft er drug group of compounds
Mechanisms of [paracetamol], discontinuation. that can be grouped
Action celecoxib, etoricoxib) Th e fi rst relatively into salicylic acids
Many over-the-counter can be used selective COX-2 agent to (eg, aspirin), acetic
nonsteroidal antiinfl perioperatively be developed was acid derivatives (eg,
ammatory agents without concerns acetaminophen ketorolac), propionic
(NSAIDs) work through about platelet (paracetamol). acid derivatives (eg,
inhibition of inhibition or Curiously, this agent, ibuprofen),
cyclooxygenase (COX), gastrointestinal while eff ective for heterocyclics (eg,
the key step in upset. Curiously, analgesia, produces celecoxib), and
prostaglandin synthesis. while COX-1 almost no eff ects on infl others. Th us a
COX catalyzes the inhibition decreases ammation relative to conventional
production of thrombosis, selective other COX-2 selective discussion of
prostaglandin H 1 from COX-2 inhibition agents. With few structure to potency
arachidonic acid. Th e increases the risk of exceptions, the COX (and other factors) is
two forms of the heart attack, inhibitors are oral not useful for these
enzyme, COX-1 and thrombosis, and agents. Acetaminophen chemicals, other
COX-2, have diff ering stroke. and ketorolac are than to note that the
distribution in tissue. Aspirin, the fi available in an heterocyclics tend to
COX-1 receptors are rst of the so-called intravenous form for be the compounds
widely distributed NSAIDs, formerly perioperative use. with the greatest
throughout the body, was used as an Multimodal selectivity for the
including the gut and antipyretic and analgesia typically COX-2 rather than
platelets. COX-2 is analgesic. Now it is includes the use of COX COX-1 form of the
produced in response to used almost inhibitors, regional or enzyme.
infl ammation. exclusively for local anesthesia
COX-1 and COX-2 prevention of techniques, and other
enzymes diff er further thrombosis in approaches aimed at Pharmacokineti
in the size of their susceptible reducing the
binding sites: the COX-2 individuals or for requirement for opioids cs
site can accommodate treat- in postoperative A. Absorption
larger molecules that patients. Th e hope is All COX inhibitors
are restricted from 5 ment of acute that reduced exposure (save for ketorolac)
binding at the COX-1 myocardial to opioids will hasten are well absorbed aft
site. Th is distinction is in infarction. Aspirin is and improve recovery er oral
202 SECTION II Clinical Pharmacology

administration and all D. Excretion 1 inhibition is Myles PS, Power I:


will typically achieve Nearly all COX gastrointestinal upset. In Clinical update:
their peak blood its most extreme form Postoperative
inhibitors are
concentrations in less this can cause upper analgesia. Lancet
excreted in urine aft
2007;369:810.
than 3 hours. Some COX er gastrointestinal
Parvizi J, Miller AG,
inhibitors are biotransformation. bleeding. Both Gandhi K:
formulated for topical complications result Multimodal pain
application (eg, as a gel Eff ects on from direct actions of management aft er
to be applied over joints the drug, in the former total joint
or as liquid drops to be Organ Systems case, on protective eff arthroplasty. J Bone
instilled on the eye). A. Cardiovascular ects of prostaglandins in Joint Surg Am
COX inhibitors do the mucosa, and in the 2011;93:1075.
B. Distribution not act directly on latter case, on the
Aft er absorption, COX the cardiovascular combination of mucosal
inhibitors are highly system. Any eff ects and inhibition of
bound by plasma cardiovascular eff platelet aggregation.
proteins, chiefl y ects result from the Acetaminophen
albumin. Th eir lipid actions of these abuse or overdosage is a
solubility allows them to agents on common cause of
readily permeate the coagulation. fulminant hepatic failure
blood– brain barrier to Prostaglandins resulting in need for
produce a central maintain the hepatic transplantation
analgesia and patency of the in western societies.
antipyresis, and to ductus arteriosus,
penetrate joint spaces thus prostaglandin
SUGGESTED
to produce (with the inhibitors have been READING
exception of administered to Brunton LL, Chabner
acetaminophen) an neonates to BA, Knollmann BC
antiinfl ammatory eff promote closure of a (Eds): Goodman &
ect. persistently patent Gilman’s Th
ductus arteriosus. e Pharmacological
C. Biotransformation Basis of Th
B. Respiratory
Most COX inhibitors erapeutics , 12th ed.
At appropriate
undergo hepatic McGraw-Hill, 2010:
clinical doses, none Chaps 18, 34.
biotransformation. Th e
of the COX inhibitors Chu LF, Angst MS, Clark D:
agent with the most
have eff ects on Opioid-induced
notable metabolite is
respiration or lung hyperalgesia in humans:
acetaminophen which at
function. Aspirin Molecular mechanisms
toxic, increased doses
overdosage has and clinical
yields suffi ciently large considerations. Clin J
complex eff ects on
concentrations of N Pain 2008;24:479.
acid–base balance
-acetylp -benzoquinone Jahr JS, Lee VK:
and respiration.
imine to produce Intravenous
hepatic failure. C. Gastrointestinal acetaminophen.
Th e classical Anesthesiol Clin
2010;28:619.
complication of COX-
C H A P T E R

Neuromuscular

Blocking Agents
KEYCONCEPTS
11
1 It is important to realize that muscle tripled, patients with homozygous atypical relaxation does not
ensure unconsciousness, enzyme will have a very long blockade amnesia, or analgesia. (eg,
4–8 h) following succinylcholine
2 Depolarizing muscle relaxants act as administration.
acetylcholine (ACh) receptor agonists, 7 Succinylcholine is considered whereas
nondepolarizing muscle relaxants contraindicated in the routine management function as
competitive antagonists . of children and adolescents because of the
3 Because depolarizing muscle relaxants are risk of hyperkalemia, rhabdomyolysis, and not
metabolized by acetylcholinesterase, cardiac arrest in children with undiagnosed they diff use away
from the neuromuscular myopathies . junction and are hydrolyzed in the 8 Normal muscle releases
enough plasma and liver by another enzyme, potassium during succinylcholine-induced
pseudocholinesterase (nonspecifi c depolarization to raise serum potassium cholinesterase, plasma
cholinesterase, or by 0.5 mEq/L. Although this is usually butyrylcholinesterase). insignifi cant in
patients with normal baseline
4 With the exception of mivacurium, potassium levels, a life-threatening potassium nondepolarizing
agents are not signifi cantly elevation is possible in patients with metabolized by either
acetylcholinesterase burn injury, massive trauma, neurological or pseudocholinesterase. Reversal of
their disorders, and several other conditions . blockade depends on redistribution, gradual 9
204 SECTION II Clinical Pharmacology

Doxacurium, pancuronium, vecuronium, metabolism, and excretion of the relaxant by and


pipecuronium are partially excreted by the body, or administration of specifi c reversal the kidneys,
and their action is prolonged in agents (eg, cholinesterase inhibitors) that patients with renal failure .
inhibit acetylcholinesterase enzyme activity . 10 Cirrhotic liver disease and chronic renal
5 Muscle relaxants owe their paralytic failure often result in an increased volume properties to
mimicry of ACh. For example, of distribution and a lower plasma
succinylcholine consists of two joined ACh concentration for a given dose of watermolecules .
soluble drugs, such as muscle relaxants.
6 Compared with patients with low enzyme On the other hand, drugs dependent on levels or
heterozygous atypical enzyme hepatic or renal excretion may demonstrate in whom
blockade duration is doubled or prolonged clearance. Thus, depending on
—Continued next page

199

Continued— catecholamine release from adrenergic nerve


endings .
the drug, a greater initial dose—but smaller
maintenance doses—might be required in these 13 Long-term administration of vecuronium to
diseases . patients in intensive care units has resulted in
prolonged neuromuscular blockade (up to
11 Atracurium and cisatracurium undergo degra several days), possibly from accumulation of its
dation in plasma at physiological pH and active 3-hydroxy metabolite, changing drug
temperature by organ-independent Hofmann clearance, or the development of a
elimination. The resulting metabolites (a monoq polyneuropathy.
uaternary acrylate and laudanosine) have no
intrinsic neuromuscular blocking eff ects . 14 Rocuronium (0.9–1.2 mg/kg) has an onset of
action that approaches succinylcholine (60–90
12 Hypertension and tachycardia may occur in s), making it a suitable alternative for rapid-
patients given pancuronium. These sequence inductions, but at the cost of a much
cardiovascular eff ects are caused by the longer duration of action .
combination of vagal blockade and
Skeletal muscle relaxation can be produced by nerve’s action potential depolarizes its terminal,
deep inhalational anesthesia, regional nerve block, an infl ux of calcium ions through voltage-gated
or neuromuscular blocking agents (commonly calcium channels into the nerve cytoplasm allows
called muscle relaxants ). In 1942, Harold Griffi th storage vesicles to fuse with the terminal plasma
published the results of a study using an extract of membrane and release their contents
curare (a South American arrow poison) during
anesthesia. Following the introduction of
succinylcholine muscle fi ber are separated by a
narrow (20-nm) gap, the synaptic cleft . As a
CHAPTER 11 Neuromuscular Blocking Agents 205

as a “new approach to muscular relaxation,” these (Reproduced, with permission, from Drachman DB: Myasthenia gravis.
N Engl J Med
agents rapidly became a routine part of the 1978;298:135.)
anesthesiologist’s drug arsenal. However, as noted (acetylcholine [ACh]). Th e ACh molecules diff use
by Beecher and Todd in 1954: “[m]uscle relaxants across the synaptic cleft to bind with nicotinic
given inappropriately may provide the surgeon cholinergic receptors on a specialized portion of
with optimal [operating] conditions in . . . a patient the muscle membrane, the motor end-plate. Each
[who] is paralyzed but not anesthetized—a state neuromuscular junction contains approximately 5
[that] is million of these receptors, but activation of only
about 500,000 receptors is required for normal
1 wholly unacceptable for the patient.” In other muscle contraction.
words, muscle relaxation does not ensure Th e structure of ACh receptors varies in diff
un- erent tissues and at diff erent times in
consciousness, amnesia, or analgesia. Th is chapter development. Each ACh receptor in the
reviews the principles of neuromuscular neuromuscular junction normally consists of fi ve
transmission and presents the mechanisms of protein subunits; two α subunits; and single β, δ,
action, physical structures, routes of elimination,
and ε subunits. Only the two identical α subunits
recommended dosages, and side eff ects of
are capable of binding ACh molecules. If both
several muscle relaxants.
binding sites are occupied by ACh, a
conformational change in the subunits briefl y (1
ms) opens an ion channel in the core of the
Neuromuscular Transmission receptor ( Figure 11–2 ). Th e channel will not
Association between a motor neuron and a open if ACh binds on only one site. In contrast to
muscle cell occurs at the neuromuscular junction ( the normal (or mature) junctional ACh receptor,
Figure 11–1 ). Th e cell membranes of the neuron another isoform contains a γ subunit instead of
and the ε subunit. Th is isoform is referred to as the
Axon
fetal or immature receptor because it is in the
form initially expressed in fetal muscle. It is also
V M oft en referred to as extrajunctional because,
Release site unlike the mature isoform, it may be located
Nerve anywhere in the muscle membrane, inside or
terminal ACh
outside the neuromuscular junction when
expressed in adults.
ACh JF Cations fl ow through the open ACh receptor
receptors
AChE
channel (sodium and calcium in; potassium out),
generating an end-plate potential . Th e contents
End plate of a single vesicle, a quantum of ACh (10 4
molecules per quantum), produce a miniature
Muscle
end-plate potential. Th e number of quanta
released by each nerve impulse, normally at least
FIGURE 111 The neuromuscular junction. V, 200, is very sensitive to extracellular ionized
transmitter vesicle; M, mitochondrion; ACh, acetylcholine; calcium concentration; increasing calcium
AChE, acetylcholinesterase; JF, junctional folds. concentration increases the
206 SECTION II Clinical Pharmacology

number of quanta released. When enough myasthenia gravis (decreased number of


receptors are occupied by ACh, the end-plate receptors).
potential will be suffi ciently strong to depolarize ACh is rapidly hydrolyzed into acetate and
the perijunctional membrane. Voltage-gated choline by the substrate-specifi c enzyme
sodium channels within this portion of the muscle acetylcholinesterase . Th is enzyme (also called
membrane open when a threshold voltage is specifi c cholinesterase or true cholinesterase) is
developed across them, as opposed to end-plate embedded into the motor end-plate membrane
receptors that open when ACh is applied ( Figure immediately adjacent to the ACh receptors. Aft er
11–3 ). Perijunctional areas of muscle membrane unbinding ACh, the receptors’ ion channels close,
have a higher density of these sodium channels permitting the end-plate to repolarize. Calcium is
than other parts of the membrane. Th e resulting resequestered in the sarcoplasmic reticulum, and
action potential propagates along the muscle the muscle cell relaxes.
membrane and T-tubule system, opening sodium
channels and releasing calcium from the
sarcoplasmic reticulum. Th is intracellular calcium Distinctions Between
allows the contractile proteins actin and myosin to
interact, bringing about muscle contraction. Th e
Depolarizing & Nondepolarizing
Blockade
A
Upper gate

Neuromuscular blocking agents are divided into


Membrane two classes: depolarizing and nondepolarizing (
Table 11–1 ). Th is division refl ects distinct
Lower gate differences in the mechanism of action, response
Na+ to peripheral nerve stimulation, and reversal of
B block.

MECHANISM OF ACTION
Similar to ACh, all neuromuscular blocking agents
are quaternary ammonium compounds whose
C positively charged nitrogen imparts an affi nity to

amount of ACh released and the number of


receptors subsequently activated will normally far
exceed the minimum required for the initiation of
an action potential. Th e nearly 10-fold margin of
safety is lost in Eaton–Lambert myasthenic
syndrome (decreased release of ACh) and
CHAPTER 11 Neuromuscular Blocking Agents 207

FIGURE 113 Schematic of the sodium channel. The sodium ACh receptors; this is called a phase I
channel is a transmembrane protein that can be conceptualized block. Aft er a period of time, prolonged
as having two gates. Sodium ions pass only when both gates are end-plate depolarization can cause poorly
open. Opening of the gates is time dependent and voltage understood changes in the ACh receptor
dependent. The channel therefore possesses three functional that result in a phase II block, which
states. At rest, the lower gate is open but the upper gate is closed
clinically resembles that of
( A ). When the muscle membrane reaches threshold voltage
depolarization, the upper gate opens and sodium can pass ( B ).
nondepolarizing muscle relaxants.
Shortly after the upper gate opens the timedependent lower gate Nondepolarizing muscle relaxants
closes ( C ). When the membrane repolarizes to its resting bind ACh receptors but are incapable of
voltage, the upper gate closes and the lower gate opens ( A ). inducing the conformational change
Depolarizing Nondepolarizing
necessary for ion channel opening.
Because ACh is prevented from binding to
Short-acting Short-acting
its receptors, no end-plate potential
Succinylcholine Gantacurium 1
Intermediate-acting develops. Neuromuscular blockade occurs
Atracurium even if only one α subunit is blocked.
Cisatracurium
Vecuronium 2 Th us, depolarizing muscle relaxants act
Rocuronium
Long-acting as ACh receptor agonists, whereas
Pancuronium nondepolarizing
TABLE 111 Depolarizing and nondepolarizing muscle relaxants function as competitive
muscle relaxants. antagonists. Th is basic diff erence in
nicotinic ACh receptors. Whereas most agents have mechanism of action explains their varying
two quaternary ammonium atoms, a few have one eff ects in certain disease states. For
quaternary ammonium cation and one tertiary amine example, conditions associated with a
that is protonated at physiological pH. chronic decrease in ACh release (eg,
Depolarizing muscle relaxants very closely muscle denervation injuries) stimulate a
resemble ACh and readily bind to ACh receptors, compensatory increase in the number of
generating a muscle action potential. Unlike ACh, ACh receptors within muscle membranes.
however, these drugs are not metabolized by Th ese states also promote the expression
acetylcholinesterase, and their concentration in the of the immature (extrajunctional) isoform
synaptic cleft does not fall as rapidly, resulting in a of the ACh receptor, which displays low
prolonged depolarization of the muscle end-plate. channel conductance properties and
Continuous end-plate depolarization causes prolonged open-channel time. Th is up-
muscle relaxation because opening of perijunctional regulation causes an exaggerated
sodium channels is time limited (sodium channels response to depolarizing muscle relaxants
rapidly “inactivate” with continuing depolarization) (F (with more receptors being depolarized),
igure 11–3) . Aft er the initial excitation and opening (F but a resistance to nondepolarizing
igure 11–3B) , these sodium channels inactivate ( Figure relaxants (more receptors that must be
11–3C ) and cannot reopen until the end-plate blocked). In contrast, conditions
repolarizes. Th e end-plate cannot repolarize as long as associated with fewer ACh receptors (eg,
the depolarizing muscle relaxant continues to bind to down- regulation in myasthenia gravis)
demonstrate a resistance to depolarizing
1 Not yet commercially available in the United States.
208 SECTION II Clinical Pharmacology

relaxants and an increased sensitivity to pseudocholinesterase (nonspecifi c


nondepolarizing relaxants. cholinesterase, plasma cholinesterase, or
butyrylcholinesterase). Fortunately, this is
a fairly rapid process, because no specifi c
OTHER MECHANISMS agent to reverse a depolarizing blockade is
available.
OF NEUROMUSCULAR BLOCKADE
Some drugs may interfere with the function of the ACh 4 With the exception of the discontinued
receptor without acting as an agonist or antagonist. Th drug mivacurium, nondepolarizing
ey interfere with normal functioning of the ACh agents are not
receptor binding site or with the opening and closing of metabolized by either acetylcholinesterase
the receptor channel. Th ese may include inhaled or pseudocholinesterase. Reversal of their
anesthetic agents, local anesthetics, and ketamine. Th e blockade depends on unbinding the
ACh receptor–lipid membrane interface may be an receptor, redistribution, metabolism, and
important site of action. excretion of the relaxant by the body, or
Drugs may also cause either closed or open administration of specifi c reversal agents
channel blockade. During closed channel blockade, the (eg, cholinesterase inhibitors) that inhibit
drug physically plugs up the channel, preventing acetylcholinesterase enzyme activity.
passage of cations whether or not ACh has activated Because this inhibition increases the
the receptor. Open channel blockade is use dependent, amount of ACh that is available at the
because the drug enters and obstructs the ACh neuromuscular junction and can compete
receptor channel only aft er it is opened by ACh with the nondepolarizing agent, clearly,
binding. Th e clinical relevance of open channel the reversal agents are of no benefi t in
blockade is unknown. Based on laboratory reversing a depolarizing block. In fact, by
experiments, one would expect that increasing the increasing neuromuscular junction ACh
concentration of ACh with a cholinesterase inhibitor concentration and inhibiting
would not overcome this form of neuromuscular pseudocholinesterase-induced metabolism
blockade. Drugs that may cause channel block in the of succinylcholine, cholinesterase
laboratory include neostigmine, some antibiotics, inhibitors can prolong neuromuscular
cocaine, and quinidine. Other drugs may impair the blockade produced by succinylcholine . Th
presynaptic release of ACh. Prejunctional receptors e ONLY time neostigmine reverses
play a role in mobilizing ACh to maintain muscle neuromuscular block aft er succinylcholine
contraction. Blocking these receptors can lead to a is when there is a phase II block (fade of
fading of the train-of-four response. the train-of-four) AND suffi cient time has
passed for the circulating concentration of
succinylcholine to be negligible.
REVERSAL OF Sugammadex, a cyclodextrin, is the fi
rst selective relaxant-binding agent; it
NEUROMUSCULAR BLOCKADE exerts its reversal eff ect by forming tight
Because succinylcholine is not metabolized by complexes in a 1:1 ratio with steroidal
acetylcholinesterase, it unbinds the receptor and diff nondepolarizing agents (vecuronium,
uses away from the neuromuscular junction to be rocuronium,). Th is drug has been in use in
hydrolyzed in the plasma and liver by another enzyme, the European Union for the past few years,
CHAPTER 11 Neuromuscular Blocking Agents 209

but is not yet commercially available in the United (blockade of ACh mobilization). Adequate
States. clinical recovery correlates well with the
Th e newer neuromuscular blocking agents, such absence of fade. Because fade is more
as gantacurium, which are still under investigation, obvious during sustained tetanic
show promise as ultrashort-acting nondepolarizing stimulation or double-burst stimulation
agents; they undergo chemical degradation by rapid than following a train-of-four pattern or
adduction with L-cysteine. repeated twitches, the fi rst two patterns
are the preferred methods for determining
adequacy of recovery from a
RESPONSE TO PERIPHERAL NERVE nondepolarizing block.
STIMULATION Th e ability of tetanic stimulation
Th e use of peripheral nerve stimulators to monitor during a partial nondepolarizing block to
neuromuscular function is discussed in Chapter 6. increase the evoked response to a
Four patterns of electrical stimulation with subsequent twitch is termed posttetanic
supramaximal square-wave pulses are considered: potentiation. Th is phenomenon may
Tetany: A sustained stimulus of 50–100 Hz, usually relate to a transient increase in ACh
lasting 5 sec. mobilization following tetanic stimulation.
Single twitch: A single pulse 0.2 ms in duration. In contrast, a phase I depolarization
block from succinylcholine does not exhibit
Train-of-four: A series of four twitches in 2 s (2-Hz
fade during tetanus or train-of-four;
frequency), each 0.2 ms long.
neither does it demonstrate posttetanic
Double-burst stimulation (DBS): Th ree short (0.2
potentiation. With longer infusions of
ms) high-frequency stimulations separated by a 20-ms
succinylcholine, however, the quality of
interval (50 Hz) and followed 750 ms later by two (DBS
the block will sometimes change to
3,2 ) or three (DBS 3,3 ) additional impulses.
resemble a nondepolarizing block (phase II
Th e occurrence of fade, a gradual diminution of
block).
evoked response during prolonged or repeated nerve
Newer quantitative methods of
stimulation, is indicative of a nondepolarizing block (
assessment of neuromuscular blockade,
Table 11–2) , or of a phase II block if only
such as acceleromyography, permit
succinylcholine has been administered. Fade may be
determination of exact train-of-four ratios,
due to a prejunctional eff ect of nondepolarizing
as opposed to subjective interpretations.
relaxants that reduces the amount of ACh in the nerve
Acceleromyography may reduce the
terminal available for release during stimulation
incidence of
TABLE 112 Evoked responses during depolarizing (phase I and phase II) and
nondepolarizing block.
210 SECTION II Clinical Pharmacology

unexpected postoperative residual neuromuscular Physical Structure


blockade.
5 Succinylcholine—also called
diacetylcholine
or suxamethonium—consists of two joined
Depolarizing ACh molecules ( Figure 11–4 ). Th is structure underMuscle Relaxants lies
succinylcholine’s mechanism of action, side eff ects, and metabolism.
CHAPTER 11 Neuromuscular Blocking Agents 211

SUCCINYLCHOLINE Metabolism & Excretion


Th e only depolarizing muscle relaxant in clinical use Th e popularity of succinylcholine is due to its today
is succinylcholine. rapid onset of action (30–60 s) and short duration
212 SECTION II Clinical Pharmacology

FIGURE 114 Chemical structures of neuromuscular blocking agents.


CHAPTER 11 Neuromuscular Blocking Agents 213

of action (typically less than 10 min). Its rapid all neuromuscular blockers, has a small volume onset of
action relative to other neuromuscular of distribution due to its very low lipid solubility, blockers is largely
due to the relative overdose and this also underlies a rapid onset of action. As that is usually
administered. Succinylcholine, like succinylcholine enters the circulation, most of it is
rapidly metabolized by pseudocholinesterase into Echothiophate Organophosphate use for
succinylmonocholine. Th is process is so effi cient glaucoma
that only a small fraction of the injected dose ever Neostigmine Cholinesterase inhibitors
reaches the neuromuscular junction. As drug Pyridostigmine
levels fall in blood, succinylcholine molecules diff
Phenelzine Monoamine oxidase inhibitor
use away from the neuromuscular junction,
limiting the duration of action. However, this Cyclophosphamide Antineoplastic agent
duration of action can be prolonged by high doses, Metoclopramide Antiemetic/prokinetic agent
infusion of succinylcholine, or abnormal
metabolism. Th e latter may result from Esmolol β-Blocker
hypothermia, reduced pseudocholinesterase Pancuronium Nondepolarizing muscle relaxant
levels, or a genetically aberrant enzyme.
Hypothermia decreases the rate of hydrolysis. Oral contraceptives Various agents

Reduced levels of pseudocholinesterase patients with low enzyme levels or heterozygous


(measured as units per liter) accompany atypical enzyme, patients with homozygous
pregnancy, liver disease, renal failure, and certain atypical enzyme will have a very long blockade
drug therapies (Table 11–3 ). Reduced (eg, 4–8 h) following administration of
pseudocholinesterase levels generally produce succinylcholine. Of the recognized abnormal
only modest prolongation of succinylcholine’s pseudocholinesterase genes, the dibucaine-
actions (2–20 min). resistant (variant) allele, which produces an
One in 25-30 patients of European extraction enzyme with 1/100 of normal affi nity for
is a heterozygote with one normal and one succinylcholine, is the most common. Other
abnormal (atypical) pseudocholinesterase gene, variants include fl uoride-resistant and silent (no
resulting in a slightly prolonged block (20–30 min). activity) alleles.
Even fewer (1 in 3000) patients have two copies of Dibucaine, a local anesthetic, inhibits normal
the most prevalent abnormal gene (homozygous pseudocholinesterase activity by 80%, but inhibits
atypical) that produce an enzyme with little or no atypical enzyme activity by only 20%. Serum from
affi nity for an individual who is heterozygous for the atypical
enzyme is characterized by an intermediate 40%
6 succinylcholine. In contrast to the doubling or to 60% inhibition. Th e percentage of inhibition of
tripling of blockade duration seen in pseudocholinesterase activity is termed the
dibucaine number . A patient with normal
pseudocholinesterase has a dibucaine number of
80; a homozygote for the most common abnormal
TABLE 113 Drugs known to decrease allele will have a dibucaine number of 20. Th e
pseudocholinesterase activity. dibucaine number measures pseudocholinesterase
Drug Description function, not the amount of enzyme. Th erefore,
adequacy of pseudocholinesterase can be
determined in the laboratory quantitatively in
214 SECTION II Clinical Pharmacology

units per liter (a minor factor) and qualitatively by


dibucaine number (the major factor). Prolonged
paralysis from succinylcholine caused by
abnormal pseudocholinesterase (atypical
cholinesterase) should be treated with continued
mechanical ventilation and sedation until muscle
function returns to normal by clinical signs. Such
unsedated patients do NOT appreciate
unnecessary, repetitive use of nerve stimulation
when all members of a department come by to
confi rm the diagnosis .

Drug Interactions
Th e eff ects of muscle relaxants can be modifi ed
by concurrent drug therapy ( Table 11–4 ).
Succinylcholine is involved in two interactions
deserving special comment.

A. Cholinesterase Inhibitors
Although cholinesterase inhibitors reverse
nondepolarizing paralysis, they markedly prolong
a depolarizing phase I block by two mechanisms.
By inhibiting acetylcholinesterase, they lead to a
CHAPTER 11 Neuromuscular Blocking Agents 215

4 Potentiation (+) and resistance (−) of neuromuscular blocking agents by other drugs.
Eff ect on Eff ect on
Depolarizing Nondepolarizing
Drug Blockade Blockade Comments

Antibiotics + + Streptomycin, aminoglycosides, kanamycin, neomycin, colistin,


polymyxin, tetracycline, lincomycin, clindamycin

Anticonvulsants ? − Phenytoin, carbamazepine, primidone, sodium valproate

Antiarrhythmics + + Quinidine, calcium channel blockers

Cholinesterase + − Neostigmine, pyridostigmine


inhibitors

Dantrolene ? + Used in treatment of malignant hyperthermia (has


quaternary ammonium group)

Inhalational + + Volatile anesthetics


anesthetics

Ketamine ? +

Local anesthetics + + High doses only

Lithium carbonate + ? Prolongs onset and duration of succinylcholine

Magnesium sulfate + + Doses used to treat preeclampsia and eclampsia of pregnancy

higher ACh concentration at the nerve Dosage


terminal, which intensifi es depolarization. Because of the rapid onset, short duration,
Th ey also reduce the hydrolysis of and low cost of succinylcholine, many
succinylcholine by inhibiting clinicians believe that it is still a good choice
pseudocholinesterase. Organophosphate for routine intubation in adults. Th e usual
pesticides, for example, cause an irreversible adult dose of succinylcholine for intubation
inhibition of acetylcholinesterase and can is 1–1.5 mg/kg intravenously. Doses as small
prolong the action of succinylcholine by 20– as 0.5 mg/kg will oft en provide acceptable
30 min. Echothiophate eye drops, used in intubating conditions if a defasciculating
the past for glaucoma, can markedly prolong dose of a nondepolarizing agent is not used.
succinylcholine by this mechanism. Repeated small boluses (10 mg) or a
B. Nondepolarizing Relaxants succinylcholine drip (1 g in 500 or 1000 mL,
In general, small doses of nondepolarizing titrated to eff ect) can be used during
relaxants antagonize a depolarizing phase I surgical procedures that require brief but
block. Because the drugs occupy some ACh intense paralysis (eg, otolaryngological
receptors, depolarization by succinylcholine endoscopies). Neuromuscular function
is partially prevented. should be frequently monitored with a
nerve stimulator to prevent overdosing and
If enough depolarizing agent is
to watch for phase II block. Th e availability
administered to develop a phase II block, a
of intermediate-acting nondepolarizing
nondepolarizer will potentiate paralysis.
muscle relaxants has reduced the popularity
of succinylcholine infusions. In the past,
216 SECTION II Clinical Pharmacology

TABLE 11
these infusions were a mainstay of to those at the neuromuscular junction. Th e
ambulatory practice in the United States. entire parasympathetic nervous system and
Because succinylcholine is not lipid parts of the sympathetic nervous system
soluble, it has a small volume of distribution. (sympathetic ganglions, adrenal medulla,
Per kilogram, infants and neonates have a and sweat glands) depend on ACh as a
larger extracellular space than adults. Th neurotransmitter.
erefore, dosage requirements for pediatric Succinylcholine not only stimulates
patients are oft en greater than for adults. If nicotinic cholinergic receptors at the
succinylcholine is administered neuromuscular junction, it stimulates all ACh
intramuscularly to children, a dose as high as receptors. Th e cardiovascular actions of
4–5 mg/kg does not always produce succinylcholine are therefore very complex.
complete paralysis. Stimulation of nicotinic receptors in
Succinylcholine should be stored under parasympathetic and sympathetic ganglia,
refrigeration (2–8°C), and should generally and muscarinic receptors in the sinoatrial
be used within 14 days aft er removal from node of the heart, can increase or decrease
refrigeration and exposure to room blood pressure and heart rate. Low doses of
temperature. succinylcholine can produce negative
chronotropic and inotropic eff ects, but
higher doses usually increase heart rate and
Side Eff ects & Clinical contractility and elevate circulating
Considerations catecholamine levels. In most patients, the
Succinylcholine is a relatively safe drug— hemodynamic consequences are
assuming that its many potential inconsequential in comparison to the eff
complications are under- ects of the induction agent and
laryngoscopy.
7 stood and avoided. Because of the risk of Children are particularly susceptible to
hyperkalemia, rhabdomyolysis, and cardiac profound bradycardia following
arrest in children with undiagnosed administration of succinylcholine.
myopathies, succinylcholine is considered Bradycardia will sometimes occur in adults
relatively contraindicated in the routine when a second bolus of succinylcholine is
management of children and adolescent administered approximately 3–8 min aft er
patients. Most clinicians have also the fi rst dose. Th e dogma (based on no real
abandoned the routine use of evidence) is that the succinylcholine
succinylcholine for adults. Succinylcholine is metabolite, succinylmonocholine, sensitizes
still useful for rapid sequence induction and muscarinic cholinergic receptors in the
for short periods of intense paralysis sinoatrial node to the second dose of
because none of the presently available succinylcholine, resulting in bradycardia.
nondepolarizing muscle relaxants can match Intravenous atropine (0.02 mg/kg in
its very rapid onset and short duration. children, 0.4 mg in adults) is normally given
prophylactically to children prior to the fi rst
A. Cardiovascular and subsequent doses, and usually before a
Because of the resemblance of muscle second dose of succinylcholine is given to
relaxants to ACh, it is not surprising that adults. Other arrhythmias, such as nodal
they aff ect cholinergic receptors in addition
CHAPTER 11 Neuromuscular Blocking Agents 217

bradycardia and ventricular ectopy, have Guillain-Barré syndrome


been reported. Severe Parkinson’s disease
Tetanus
Prolonged total body immobilization
B. Fasciculations Ruptured cerebral aneurysm
Th e onset of paralysis by succinylcholine is Polyneuropathy
usually signaled by visible motor unit Closed head injury
contractions called fasciculations. Th ese can Hemorrhagic shock with metabolic acidosis
Myopathies (eg, Duchenne’s dystrophy)
be prevented by pretreatment with a small
dose of nondepolarizing relaxant. Because
this pretreatment usually antagonizes a
depolarizing block, a larger dose of
Following denervation injuries (spinal
succinylcholine is required (1.5 mg/kg).
cord injuries, larger burns), the immature
Fasciculations are typically not observed in
isoform of the ACh receptor may be
young children and elderly patients.
expressed inside and outside the
neuromuscular junction (up-regulation). Th
C. Hyperkalemia
ese extrajunctional receptors allow
Normal muscle releases enough succinylcholine to eff ect widespread
8
depolarization and extensive potassium
potassium during succinylcholine-
release. Life-threatening potassium release
induced depolariza-
is not reliably prevented by pretreatment
tion to increase serum potassium by 0.5 with a nondepolarizer. Th e risk of
mEq/L. Although this is usually insignifi cant hyperkalemia usually seems to peak in 7–10
in patients with normal baseline potassium days following the injury, but the exact time
levels, it can be life- threatening in patients of onset and the duration of the risk period
with preexisting hyperkalemia. Th e increase vary. Th e risk of hyperkalemia from
in potassium in patients with burn injury, succinylcholine is minimal in the fi rst 2 days
massive trauma, neurological disorders, and aft er spinal cord or burn injury.
several other conditions ( Table 11–5 ) can
be large and catastrophic. Hyperkalemic D. Muscle Pains
cardiac arrest can prove to be quite Patients who have received succinylcholine
refractory to routine cardiopulmonary have an increased incidence of
resuscitation, requiring calcium, insulin, postoperative myalgia. Th e effi cacy of
glucose, bicarbonate, and even nondepolarizing pretreatment is
cardiopulmonary bypass to support the controversial. Administration of rocuronium
circulation while reducing serum potassium (0.06–0.1 mg/kg) prior to succinylcholine
levels. has been reported to be eff ective in
5 Conditions causing preventing fasciculations and reducing
susceptibility to succinylcholine- postoperative myalgias. Th e relationship
induced hyperkalemia. between fasciculations and postoperative
Burn injury
myalgias is also inconsistent. Th e myalgias
Massive trauma are theorized to be due to the initial
S evere intraabdominal infection unsynchronized contraction of muscle
Spinal cord injury groups; myoglobinemia and increases in
Encephalitis serum creatine kinase can be detected
Stroke
218 SECTION II Clinical Pharmacology

TABLE 11
following administration of succinylcholine. some of the signs and symptoms of
Perioperative use of nonsteroidal antiinfl neuroleptic malignant syndrome (NMS)
ammatory drugs may reduce the incidence resemble those of malignant hyperthermia,
and severity of myalgias. the pathogenesis is completely diff erent
and there is no need to avoid use of
E. Intragastric Pressure Elevation succinylcholine in patients with
Abdominal wall muscle fasciculations NMS.
increase intragastric pressure, which is off
set by an increase in lower esophageal I. Generalized Contractions
sphincter tone. Th erefore, despite being Patients affl icted with myotonia may
much discussed, there is no evidence that develop myoclonus aft er administration of
the risk of gastric refl ux or pulmonary succinylcholine.
aspiration is increased by succinylcholine. J. Prolonged Paralysis
As discussed above, patients with reduced
F. Intraocular Pressure Elevation levels of normal pseudocholinesterase may
Extraocular muscle diff ers from other have a longer than normal duration of
striated muscle in that it has multiple motor action, whereas patients with atypical
end-plates on each cell. Prolonged pseudocholinesterase will experience
membrane depolarization and contraction markedly prolonged paralysis.
of extraocular muscles following
administration of succinylcholine transiently K. Intracranial Pressure
raise intraocular pressure and theoretically Succinylcholine may lead to an activation of
could compromise an injured eye. However, the electroencephalogram and slight
there is no evidence that succinylcholine increases in cerebral blood fl ow and
leads to worsened outcome in patients with intracranial pressure in some patients.
“open” eye injuries. Th e elevation in Muscle fasciculations stimulate muscle
intraocular pressure is not always prevented stretch receptors, which subsequently
by pretreatment with a nondepolarizing increase cerebral activity. Th e increase in
agent. intracranial pressure can be attenuated by
maintaining good airway control and
G. Masseter Muscle Rigidity
instituting hyperventilation. It can also be
Succinylcholine transiently increases muscle
prevented by pretreating with a
tone in the masseter muscles. Some diffi nondepolarizing muscle relaxant and
culty may initially be encountered in
administering intravenous lidocaine (1.5–2.0
opening the mouth because of incomplete mg/kg) 2–3 min prior to intubation. Th e eff
relaxation of the jaw. A marked increase in
ects of intubation on intracranial pressure
tone preventing laryngoscopy is abnormal far outweigh any increase caused by
and can be a premonitory sign of malignant
succinylcholine, and succinylcholine is NOT
hyperthermia. contraindicated for rapid sequence
H. Malignant Hyperthermia induction of patients with intracranial mass
lesions or other causes of increased
Succinylcholine is a potent triggering agent
intracranial pressure.
in patients susceptible to malignant
hyperthermia, a hypermetabolic disorder of
skeletal muscle (see Chapter 52). Although
CHAPTER 11 Neuromuscular Blocking Agents 219

L. Histamine Release
Slight histamine release may be observed
following succinylcholine in some patients.

Nondepolarizing
Muscle Relaxants

Unique Pharmacological
Characteristics
In contrast to depolarizing muscle relaxants,
there is a wide selection of nondepolarizing
muscle relaxants ( Tables 11–6 and 11–7 ).
Based on their chemical structure, they can
be classifi ed as benzylisoquinolinium,
steroidal, or other compounds. It is oft en
said that choice of a particular drug depends
on its unique characteristics, which are oft
en related to its structure; however, for
most patients, the differences among the
intermediate-acting neuromuscular blockers
are inconsequential. In general, steroidal
compounds can be vagolytic, but this
property is most notable with pancuronium
and clinically unimportant with vecuronium
or rocuronium. Benzylisoquinolines tend to
release histamine. Because of structural
similarities, an allergic history to one muscle
relaxant strongly suggests the possibility of
allergic reactions to other muscle relaxants,
particularly those in the same chemical
class.
A. Suitability for Intubation
None of the currently available
nondepolarizing muscle relaxants equals
succinylcholine’s rapid onset of action or
short duration. However, the
TABLE 116 A summary of the pharmacology of nondepolarizing muscle relaxants.
220 SECTION II Clinical Pharmacology

TABLE 11
Chemical Primary Histamine Vagal
Relaxant Structure 1
Metabolism Excretion Onset 2
Duration 3
Release 4 Blockade 5

Atracurium B +++ Insignificant ++ ++ + 0

Cisatracurium B +++ Insignificant ++ ++ 0 0

Pancuronium S + Renal ++ +++ 0 ++

Vecuronium S + Biliary ++ ++ 0 0

Rocuronium S Insignificant Biliary +++ ++ 0 +

Gantacurium C +++ Insignificant +++ + + 0

7 Clinical characteristics of nondepolarizing muscle relaxants.


ED 95 for Adductor Maintenance Maintenance
Pollicis During Nitrous Intubation Onset of Action Duration of Dosing by Dosing by
Oxide/Oxygen/Intravenous Dose for Intubating Intubating Boluses Infusion
Drug Anesthesia (mg/kg) (mg/kg) Dose (min) Dose (min) (mg/kg) (µg/kg/min)

Succinylcholine 0.5 1.0 0.5 5–10 0.15 2–15 mg/min

Gantacurium 1
0.19 0.2 1–2 4–10 N/A —

Rocuronium 0.3 0.8 1.5 35–75 0.15 9–12

Mivacurium 2
0.08 0.2 2.5–3.0 15–20 0.05 4–15

Atracurium 0.2 0.5 2.5–3.0 30–45 0.1 5–12

Cisatracurium 0.05 0.2 2.0–3.0 40–75 0.02 1–2

Vecuronium 0.05 0.12 2.0–3.0 45–90 0.01 1–2

Pancuronium 0.07 0.12 2.0–3.0 60–120 0.01 —

Pipecuronium 2
0.05 0.1 2.0–3.0 80–120 0.01 —

Doxacurium 2
0.025 0.07 4.0–5.0 90–150 0.05 —
1 Not commercially available in the United States. times the ED 95 or twice the dose that
2 No longer available in the United States.
produces 95% twitch depression is usually
used for intubation. Although a larger
onset of nondepolarizing relaxants can be intubating dose speeds onset, it exacerbates
quickened by using either a larger dose or a side eff ects and prolongs the duration of
priming dose. Th e ED 95 of any drug is the eff blockade. For example, a dose of 0.15 mg/kg
ective dose of a drug in 95% of individuals. of pancuronium may produce intubating
For neuromuscular blockers, one oft en conditions in 90 sec, but at the cost of more
specifi es the dose that produces 95% twitch pronounced tachycardia—and a block that
depression in 50% of individuals. One to two may be irreversible (by neostigmine) for
1 B, benzylisoquinolone; S, steroidal; C, chlorofumarate.
2 Onset: +, slow; ++, moderately rapid; +++, rapid.
3 Duration: +, short; ++, intermediate; +++, long.
4 Histamine release: 0, no eff ect; +, slight eff ect; ++, moderate eff ect; +++, marked eff
ect. 5
Vagal blockade: 0, no eff ect; +, slight eff ect; ++, moderate eff ect.
CHAPTER 11 Neuromuscular Blocking Agents 221

more than 60 min. Th e consequence of a important during intubation—recover from


long duration of action is the ensuing diffi blockade more quickly than the adductor
culty in completely reversing the blockade pollicis, which is commonly monitored by
and a subsequent increased incidence of the peripheral nerve stimulator.
postoperative pulmonary complica tions. As
a general rule, the more potent the B. Suitability for Preventing
nondepolarizing muscle relaxant, the slower Fasciculations To prevent fasciculations
its speed of onset; the “explanatory dogma” and myalgias, 10% to 15% of a
is that greater potency necessitates a nondepolarizer intubating dose can be
smaller dose, with fewer total drug administered 5 min before succinylcholine.
molecules, which in turn, decreases the rate When administered only shortly before
of drug binding opportunities at the succinylcholine, myalgias, but not
neuromuscular junction. fasciculations, will be inhibited. Although
Th e introduction of short- and most nondepolarizers have been
intermediate- acting agents has resulted in successfully used for this purpose,
the greater use of priming doses. Th tubocurarine and rocuronium have been
eoretically, giving 10% to 15% of the usual most popular (precurarization);
intubating dose 5 min before induction will tubocurarine is no longer available in the
occupy enough receptors so that paralysis United States.
will quickly follow when the balance of C. Maintenance Relaxation
relaxant is administered. Use of a priming
Following intubation, muscle paralysis may
dose can produce conditions suitable for
need to be maintained to facilitate surgery,
intubation as soon as 60 sec following
(eg, abdominal operations), to permit a
administration of rocuronium or 90 sec
reduced depth of anesthesia, or to control
following administration of other
ventilation. Th ere is great variability among
intermediate-acting nondepolarizers. A
patients in response to muscle relaxants.
priming dose does not usually lead to
Monitoring neuromuscular function with a
clinically signifi cant paralysis, which requires
nerve stimulator helps to prevent over- and
that 75% to 80% of the receptors be blocked
underdosing and to reduce the likelihood of
(a neuromuscular margin of safety). In some
serious residual muscle paralysis in the
patients, however, the priming dose
recovery room. Maintenance doses,
produces distressing dyspnea, diplopia, or
whether by intermittent boluses or
dysphagia; in such instances, the patient
continuous infusion ( Table 11–7 ), should be
should be reassured, and induction of
guided by the nerve stimulator and clinical
anesthesia should proceed without delay.
signs (eg, spontaneous respiratory eff orts or
Priming can additionally cause measureable
movement). In some instances, clinical signs
deterioration in respiratory function (eg,
may precede twitch recovery because of diff
decreased forced vital capacity) and may
ering sensitivities to muscle relaxants
lead to oxygen desaturation in patients with
between muscle groups or technical
marginal pulmonary reserve. Th ese negative
problems with the nerve stimulator. Some
side eff ects are more common in older,
return of neuromuscular transmission
sicker patients.
should be evident prior to administering
Muscle groups vary in their sensitivity
each maintenance dose, if the patient needs
to muscle relaxants. For example, the
to resume spontaneous ventilation at the
laryngeal muscles—whose relaxation is
222 SECTION II Clinical Pharmacology

TABLE 11
end of the anesthetic. When an infusion is cisatracurium, vecuronium, and rocuronium,
used for maintenance, the rate should be are devoid of signifi cant autonomic eff ects
adjusted at or just above the rate that in their recommended dosage ranges.
allows some return of neuromuscular
transmission so that drug eff ects can be G. Histamine Release
monitored. Histamine release from mast cells can result
in bronchospasm, skin fl ushing, and
D. Potentiation by Inhalational hypotension from peripheral vasodilation.
Anesthetics Volatile agents decrease Both atracurium and mivacurium are
nondepolarizer dosage requirements by at capable of triggering histamine release,
least 15%. Th e actual degree of this particularly at higher doses. Slow injection
postsynaptic augmentation depends on both rates and H 1 and H 2 antihistamine
the inhalational anesthetic (desfl urane > pretreatment ameliorate these side eff ects.
sevofl urane > isofl urane and enfl urane >
H. Hepatic Clearance
halothane > N 2 O/O 2 / narcotic) and the
Only pancuronium and vecuronium are
muscle relaxant employed (pancuronium >
metabolized to any signifi cant degree by
vecuronium and atracurium).
the liver. Active metabolites likely contribute
E. Potentiation by Other to their clinical eff ect. Vecuronium and
Nondepolarizers Some combinations of rocuronium depend heavily on biliary
nondepolarizers produce a greater than excretion. Clinically, liver failure prolongs
additive (synergistic) neuromuscular pancuronium and rocuronium blockade,
blockade. Th e lack of synergism (ie, the with less eff ect on vecuronium, and no eff
drugs are only additive) by closely related ect on pipecuronium. Atracurium,
compounds (eg, vecuronium and cisatracurium, and
pancuronium) lends credence to the theory
that synergism results from slightly differing
mechanisms of action.

F. Autonomic Side Eff ects


In clinical doses, the nondepolarizers diff er
in their relative eff ects on nicotinic and
muscarinic cholinergic receptors. Some
older agents (tubocurarine and, to a lesser
extent, metocurine) blocked autonomic
ganglia, reducing the ability of the
sympathetic nervous system to increase
heart contractility and rate in response to
hypotension and other intraoperative
stresses. In contrast, pancuronium (and
gallamine) block vagal muscarinic receptors
in the sinoatrial node, resulting in
tachycardia. All newer nondepolarizing
relaxants, including atracurium,
CHAPTER 11 Neuromuscular Blocking Agents 223
mivacurium, although extensively metabolized, magnesium sulfate (or aft er intravenous
depend on extrahepatic mechanisms. Severe liver magnesium administered in the operating room),
disease does not signifi cantly aff ect clearance of potentiates a nondepolarizing blockade by
atracurium or cisatracurium, but the associated competing with calcium at the motor end-plate.
decrease in pseudocholinesterase levels may slow TABLE 118 Additional considerations in
the metabolism of mivacurium. special populations.
I. Renal Excretion Pediatric Succinylcholine – should not be used routinely
Nondepolarizing agents – faster onset
Vecuronium – long-acting in neonates
9 Doxacurium, pancuronium, vecuronium, and
pipecuronium are partially excreted by the Elderly Decreased clearance – prolonged duration,
except with cisatracurium
kidneys, and their action is prolonged in patients
with renal failure. Th e elimination of atracurium, Obese Dosage 20% more than lean body weight;
onset unchanged
cisatracurium, mivacurium, and rocuronium is
Prolonged duration, except with cisatracurium
independent of kidney function.
Hepatic Increased volume of distribution
disease Pancuronium and vecuronium – prolonged
General Pharmacological elimination due to hepatic metabolism and
Characteristics biliary excretion
Cisatracurium – unchanged
Some variables aff ect all nondepolarizing muscle Pseudocholinesterase decreased; prolonged
relaxants. action may be seen with succinylcholine in
severe disease
A. Temperature Renal Vecuronium – prolonged
Hypothermia prolongs blockade by decreasing failure Rocuronium – relatively unchanged
metabolism (eg, mivacurium, atracurium, and Cisatracurium – safest alternative
cisatracurium) and delaying excretion (eg, Critically Myopathy, polyneuropathy, nicotinic
pancuronium and vecuronium). ill acetylcholine receptor up-regulation

B. Acid–Base Balance D. Age


Respiratory acidosis potentiates the blockade of Neonates have an increased sensitivity to
most nondepolarizing relaxants and antagonizes nondepolarizing relaxants because of their
its reversal. Th is could prevent complete immature neuromuscular junctions ( Table 11-8 ).
neuromuscular recovery in a hypoventilating Th is sensitivity does not necessarily decrease
postoperative patient. Confl icting fi ndings dosage requirements, as the neonate’s greater
regarding the neuromuscular eff ects of other extracellular space provides a larger volume of
acid–base changes may be due to coexisting distribution.
alterations in extracellular pH, intracellular pH, E. Drug Interactions
electrolyte concentrations, or structural diff As noted earlier, many drugs augment
erences between drugs (eg, monoquaternary nondepolarizing blockade (see Table 11–4 ). Th ey
versus bisquaternary; steroidal versus have multiple sites of interaction: prejunctional
isoquinolinium). structures, postjunctional cholinergic receptors,
C. Electrolyte Abnormalities and muscle membranes.
Hypokalemia and hypocalcemia augment a F. Concurrent Disease
nondepolarizing block. Th e responses of patients Th e presence of neurological or muscular disease
with hypercalcemia are unpredictable. can have profound eff ects on an individual’s
Hypermagnesemia, as may be seen in response
preeclamptic patients being managed with
224 SECTION II Clinical Pharmacology
choice of muscle relaxant. In general, the
10 to muscle relaxants ( Table 11–9 ). Cirrhotic diaphragm, jaw, larynx, and facial muscles
liver disease and chronic renal failure oft en result (orbicularis oculi) respond to and recover from
in an increased volume of distribution and a lower muscle relaxation sooner than the thumb.
plasma concentration for a given dose of
TABLE 119 Diseases with altered responses to muscle relaxants.
Disease Response to Depolarizers Response to Nondepolarizers

Amyotrophic lateral sclerosis Contracture Hypersensitivity

Autoimmune disorders Hypersensitivity Hypersensitivity


Systemic lupus erythematosus
Polymyositis
Dermatomyositis
Burn injury Hyperkalemia Resistance

Cerebral palsy Slight hypersensitivity Resistance

Familial periodic paralysis (hyperkalemic) Myotonia and hyperkalemia Hypersensitivity?

Guillain–Barré syndrome Hyperkalemia Hypersensitivity

Hemiplegia Hyperkalemia Resistance on affected side

Muscular denervation (peripheral nerve injury) Hyperkalemia and contracture Normal response or resistance

Muscular dystrophy (Duchenne type) Hyperkalemia and malignant hyperthermia Hypersensitivity

Myasthenia gravis Resistance Hypersensitivity

Myasthenic syndrome Hypersensitivity Hypersensitivity

Myotonia Generalized muscular contractions Normal or hypersensitivity


Dystrophica
Congenital
Paramyotonia
Severe chronic infection Hyperkalemia Resistance
Tetanus
Botulism
water-soluble drugs, such as muscle relaxants. On Although they are a fortuitous safety feature,
the other hand, drugs dependent on hepatic or persistent diaphragmatic contractions can be
renal excretion may demonstrate prolonged disconcerting in the face of complete adductor
clearance ( Table 11-8 ). Th us, depending on the pollicis paralysis. Glottic musculature is also quite
drug chosen, a greater initial (loading) dose—but resistant to blockade, as is oft en confi rmed
smaller maintenance doses—might be required in during laryngoscopy. Th e ED 95 for laryngeal
these diseases. muscles is nearly two times that for the adductor
pollicis muscle. Good intubating conditions are
G. Muscle Groups usually associated with visual loss of the
Th e onset and intensity of blockade vary among orbicularis oculi twitch response.
muscle groups. Th is may be due to diff erences in Considering the multitude of factors infl
blood fl ow, distance from the central circulation, uencing the duration and magnitude of muscle
or diff erent fi ber types. Furthermore, the relative relaxation, it becomes clear that an individual’s
sensitivity of a muscle group may depend on the response to neuromuscular blocking agents should
CHAPTER 11 Neuromuscular Blocking Agents 225
be monitored. Dosage recommendations, room temperature. At room temperature, it
including those in this chapter, should be should be used within 14 days to preserve
considered guidelines that require modifi cation potency.
for individual patients. Wide variability in
sensitivity to nondepolarizing muscle relaxants is Side Eff ects & Clinical Considerations
oft en encountered in clinical practice. Atracurium triggers dose-dependent histamine
ATRACURIUM release that becomes signifi cant at doses above
0.5 mg/kg.
Physical Structure
Like all muscle relaxants, atracurium has a A. Hypotension and Tachycardia
quaternary group; however, a benzylisoquinoline Cardiovascular side eff ects are unusual unless
structure is responsible for its unique method of doses in excess of 0.5 mg/kg are administered.
degradation. Th e drug is a mixture of 10 Atracurium may also cause a transient drop in
stereoisomers. systemic vascular resistance and an increase in
cardiac index independent of any histamine
Metabolism & Excretion release. A slow rate of injection minimizes these
Atracurium is so extensively metabolized that its eff ects.
pharmacokinetics are independent of renal and B. Bronchospasm
hepatic function, and less than 10% is excreted Atracurium should be avoided in asthmatic
unchanged by renal and biliary routes. Two patients. Severe bronchospasm is occasionally
separate processes are responsible for seen in patients without a history of asthma.
metabolism.
C. Laudanosine Toxicity
A. Ester Hydrolysis
Laudanosine, a tertiary amine, is a breakdown
Th is action is catalyzed by nonspecifi c esterases,
product of atracurium’s Hofmann elimination and
not by acetylcholinesterase or
has been associated with central nervous system
pseudocholinesterase.
excitation, resulting in elevation of the minimum
B. Hofmann Elimination alveolar concentration and even precipitation of
A spontaneous nonenzymatic chemical seizures. Concerns about laudanosine are probably
breakdown occurs at physiological pH and irrelevant unless a patient has received an
temperature. extremely large total dose or has hepatic failure.
Laudanosine is metabolized by the liver and
Dosage excreted in urine and bile.
A dose of 0.5 mg/kg is administered intravenously D. Temperature and pH Sensitivity
for intubation. Aft er succinylcholine, Because of its unique metabolism, atracurium’s
intraoperative relaxation is achieved with 0.25 duration of action can be markedly prolonged by
mg/kg initially, then in incremental doses of 0.1 hypothermia and to a lesser extent by acidosis.
mg/kg every 10–20 min. An infusion of 5–10
mcg/kg/min can eff ectively replace intermittent E. Chemical Incompatibility
boluses. Atracurium will precipitate as a free acid if it is
Although dosage requirements do not signifi introduced into an intravenous line containing an
cantly vary with age, atracurium may be shorter alkaline solution such as thiopental.
acting in children and infants than in adults.
Atracurium is available as a solution of 10 mg/ F. Allergic Reactions
mL. It must be stored at 2–8°C, as it loses 5% to Rare anaphylactoid reactions to atracurium have
10% of its potency for each month it is exposed to been described. Proposed mechanisms include
226 SECTION II Clinical Pharmacology
direct immunogenicity and acrylate-mediated Side Eff ects & Clinical Considerations
immune activation. IgE-mediated antibody Unlike atracurium, cisatracurium does not produce
reactions directed against substituted ammonium a consistent, dose-dependent increase in plasma
compounds, including muscle relaxants, have been histamine levels following administration.
described. Reactions to acrylate, a metabolite of Cisatracurium does not alter heart rate or blood
atracurium and a structural component of some pressure, nor does it produce autonomic eff ects,
dialysis membranes, have also been reported in even at doses as high as eight times ED 95 .
patients undergoing hemodialysis. Cisatracurium shares with atracurium the
production of laudanosine, pH and temperature
sensitivity, and chemical incompatibility.
CISATRACURIUM
Physical Structure PANCURONIUM
Cisatracurium is a stereoisomer of atracurium that
is four times more potent. Atracurium contains Physical Structure
approximately 15% cisatracurium. Pancuronium consists of a steroid ring on which
two modifi ed ACh molecules are positioned (a
Metabolism & Excretion
bisquaternary relaxant). Th e steroid ring serves as
a “spacer” between the two quaternary amines.
11 Like atracurium, cisatracurium undergoes
Pancuronium resembles ACh enough to bind (but
degradation in plasma at physiological pH and
not activate) the nicotinic ACh receptor.
temperature by organ-independent Hofmann
elimination. Th e resulting metabolites (a
monoquaternary acrylate and laudanosine) have Metabolism & Excretion
no neuromuscular blocking eff ects. Because of Pancuronium is metabolized (deacetylated) by the
cisatracurium’s greater potency, the amount of liver to a limited degree. Its metabolic products
laudanosine produced for the same extent and have some neuromuscular blocking activity.
duration of neuromuscular blockade is much less Excretion is primarily renal (40%), although some
than with atracurium. Nonspecifi c esterases are of the drug is cleared by the bile (10%). Not
not involved in the metabolism of cisatracurium. surprisingly, elimination of pancuronium is slowed
Metabolism and elimination are independent of and neuromuscular blockade is prolonged by renal
renal or liver failure. Minor variations in failure. Patients with cirrhosis may require a larger
pharmacokinetic patterns due to age result in no initial dose due to an increased volume of
clinically important changes in duration of action. distribution but have reduced maintenance
requirements because of a decreased rate of
Dosage plasma clearance.
Cisatracurium produces good intubating
conditions following a dose of 0.1–0.15 mg/kg Dosage
within 2 min and results in muscle blockade of A dose of 0.08–0.12 mg/kg of pancuronium
intermediate duration. Th e typical maintenance provides adequate relaxation for intubation in 2–3
infusion rate ranges from 1.0–2.0 mcg/kg/min. Th min. Intraoperative relaxation is achieved by
us, it is more potent than atracurium. administering 0.04 mg/kg initially followed every
Cisatracurium should be stored under 20–40 min by 0.01 mg/kg.
refrigeration (2–8°C) and should be used within 21 Children may require moderately larger doses
days aft er removal from refrigeration and of pancuronium. Pancuronium is available as a
exposure to room temperature. solution of 1 or 2 mg/mL and is stored at 2–8°C but
may be stable for up to 6 months at normal room
temperature.
CHAPTER 11 Neuromuscular Blocking Agents 227
Side Eff ects & Clinical Considerations
13 ance compared with pancuronium. L ong-term
A. Hypertension and Tachycardia
administration of vecuronium to patients in
intensive care units has resulted in prolonged
12 Th ese cardiovascular eff ects are caused by
neuromuscular blockade (up to several days),
the combination of vagal blockade and
possibly from accumulation of its active 3-hydroxy
sympathetic stimulation. Th e latter is due to a
metabolite, changing drug clearance, and in some
combination of ganglionic stimulation,
patients, leading to the development of a
catecholamine release from adrenergic nerve
polyneuropathy. Risk factors seem to include
endings, and decreased catecholamine reuptake.
female gender, renal failure, long-term or high-
Large bolus doses of pancuronium should be given
dose corticosteroid therapy, and sepsis. Th us,
with caution to patients in whom an increased
these patients must be closely monitored, and the
heart rate would be particularly detrimental (eg,
dose of vecuronium carefully titrated. Long-term
coronary artery disease, hypertrophic
relaxant administration and the subsequent
cardiomyopathy, aortic stenosis).
prolonged lack of ACh binding at the postsynaptic
B. Arrhythmias nicotinic ACh receptors may mimic a chronic
Increased atrioventricular conduction and denervation state and cause lasting receptor
catecholamine release increase the likelihood of dysfunction and paralysis. Tolerance to
ventricular arrhythmias in predisposed individuals. nondepolarizing muscle relaxants can also develop
Th e combination of pancuronium, tricyclic aft er longterm use. Fortunately, the use of
antidepressants, and halothane has been reported unnecessary paralysis has greatly declined in
to be particularly arrhythmogenic. critical care units.

C. Allergic Reactions Dosage


Patients who are hypersensitive to bromides may
Vecuronium is equipotent with pancuronium, and
exhibit allergic reactions to pancuronium
the intubating dose is 0.08–0.12 mg/kg. A dose of
(pancuronium bromide).
0.04 mg/kg initially followed by increments of 0.01
mg/kg every 15–20 min provides intraoperative
VECURONIUM relaxation. Alternatively, an infusion of 1–2
mcg/kg/min produces good maintenance of
Physical Structure relaxation.
Vecuronium is pancuronium minus a quaternary Age does not aff ect initial dose requirements,
methyl group (a monoquaternary relaxant). Th is although subsequent doses are required less
minor structural change benefi cially alters side eff frequently in neonates and infants. Women seem
ects without aff ecting potency. to be approximately 30% more sensitive than men
to vecuronium, as evidenced by a greater degree
Metabolism & Excretion of blockade and longer duration of action (this has
Vecuronium is metabolized to a small extent by also been seen with pancuronium and
the liver. It depends primarily on biliary excretion rocuronium). Th e cause for this sensitivity may be
and secondarily (25%) on renal excretion. related to gender-related diff erences in fat and
Although it is a satisfactory drug for patients with muscle mass, protein binding, volume of
renal failure, its duration of action is somewhat distribution, or metabolic activity. Th e duration of
prolonged. Vecuronium’s brief duration of action action of vecuronium may be further prolonged in
is explained by its shorter elimination half-life and postpartum patients due to alterations in hepatic
more rapid clear- blood fl ow or liver uptake.
228 SECTION II Clinical Pharmacology

Side Eff ects & Clinical Considerations adequate vocal cord and diaphragmatic paralysis
A. Cardiovascular for intubation, but not until aft er 3–6 min (deltoid
injection has a faster onset than quadriceps), and
Even at doses of 0.28 mg/kg, vecuronium is
can be reversed aft er about 1 hr. Th e infusion
devoid of signifi cant cardiovascular eff ects.
requirements for rocuronium range from 5–12
Potentiation of opioid-induced bradycardia may be
mcg/kg/min. Rocuronium can produce a
observed in some patients.
prolonged duration of action in elderly patients.
B. Liver Failure Initial dosage requirements are modestly
Although it is dependent on biliary excretion, the increased in patients with advanced liver disease,
duration of action of vecuronium is usually not presumably due to a larger volume of distribution.
signifi cantly prolonged in patients with cirrhosis
unless doses greater than 0.15 mg/kg are given. Side Eff ects & Clinical Considerations
Vecuronium requirements are reduced during the
anhepatic phase of liver transplantation. 14 Rocuronium (at a dose of 0.9–1.2 mg/kg) has an
onset of action that approaches succinyl-
choline (60–90 s), making it a suitable alternative
ROCURONIUM for rapid-sequence inductions, but at the cost of a
much longer duration of action. Th is intermediate
Physical Structure duration of action is comparable to vecuronium or
Th is monoquaternary steroid analogue of atracurium.
vecuronium was designed to provide a rapid onset Rocuronium (0.1 mg/kg) has been shown to
of action. be a rapid (90 s) and eff ective agent (decreased
fasciculations and postoperative myalgias) for
Metabolism & Excretion precurarization prior to administration of
Rocuronium undergoes no metabolism and is succinylcholine. It has slight vagolytic tendencies.
eliminated primarily by the liver and slightly by the
kidneys. Its duration of action is not signifi cantly
aff ected by renal disease, but it is modestly OTHER RELAXANTS
prolonged by severe hepatic failure and Muscle relaxants, primarily of historical interest,
pregnancy. Because rocuronium does not have are either no longer manufactured or not clinically
active metabolites, it may be a better choice than used. Th ey include tubocurarine, metocurine,
vecuronium in the rare patient requiring gallamine, alcuronium, rapacuronium, and
prolonged infusions in the intensive care unit decamethonium. Tubocurarine, the fi rst muscle
setting. Elderly patients may experience a relaxant used clinically, oft en produced
prolonged duration of action due to decreased hypotension and tachycardia through histamine
liver mass. release; its ability to block autonomic ganglia was
of secondary importance. Histamine release could
Dosage also produce or exacerbate bronchospasm.
Rocuronium is less potent than most other Tubocurarine is not metabolized signifi cantly, and
steroidal muscle relaxants (potency seems to be its elimination is primarily renal and secondarily
inversely related to speed of onset). It requires biliary. Metocurine, a closely related agent, shares
0.45–0.9 mg/kg intravenously for intubation and many of the side eff ects of tubocurarine. It is
0.15 mg/kg boluses for maintenance. A lower dose primarily dependent on renal function for
of 0.4 mg/kg may allow reversal as soon as 25 min elimination. Patients allergic to iodine (eg, shellfi
aft er intubation. Intramuscular rocuronium (1 sh allergies) could exhibit hypersensitivity to
mg/kg for infants; 2 mg/kg for children) provides metocurine preparations, as they contain iodide.
Gallamine has the most potent vagolytic
CHAPTER 11 Neuromuscular Blocking Agents 229
properties of any relaxant, and it is entirely solution; therefore, it requires reconstitution prior
dependent on renal function for elimination. to administration. In preclinical trials, gantacurium
Alcuronium, a long-acting nondepolarizer with demonstrated an ultrashort duration of action,
mild vagolytic properties, is also primarily similar to that of succinylcholine. Its
dependent on renal function for elimination. pharmacokinetic profi le is explained by the fact
Rapacuronium has a rapid onset of action, minimal that it undergoes nonenzymatic degradation by
cardiovascular side eff ects, and a short duration two chemical mechanisms: rapid formation of
of action. It was withdrawn by the manufacturer inactive cysteine adduction product and ester
following multiple reports of serious hydrolysis. At a dose of 0.2 mg/kg (ED 95 ), the
bronchospasm, including a few unexplained onset of action has been estimated to be 1-2 min,
fatalities. Histamine release may have been a with a duration of blockade similar to that of
factor. Decamethonium was an older depolarizing succinylcholine. Its clinical duration of action
agent. ranged from 5-10 min; recovery can be
More recently, doxacurium, pipecuronium, accelerated by edrophonium, as well as by the
and mivacurium are no longer commercially administration of exogenous cysteine.
available in the United States. Mivacurium is a Cardiovascular eff ects suggestive of histamine
benzylisoquinolinium derivative, which is release were observed following the use of three
metabolized by pseudocholinesterase; therefore, times the ED 95 dosage.
its duration of action may be prolonged in AV002 (CW002) is another investigational
pathophysiological states that result in low nondepolarizing agent. It is a benzylisoquinolinium
pseudocholinesterase levels. Th e usual intubating fumarate ester-based compound with an
dose is 0.2 mg/kg, with the steady state infusion intermediate duration of action that undergoes
rate being 4-10 mcg/kg/ min. Mivacurium releases metabolism and elimination similar to that of
histamine to about the same degree as gantacurium.
atracurium; the resulting cardiovascular eff ects
can be minimized by slow injection. Doxacurium is
a potent long-acting benzylisoquinolinium CASE DISCUSSION
compound that is primarily eliminated by renal
excretion. Adequate intubating conditions are Delayed Recovery from
achieved in 5 min with 0.05 mg/ kg. It is essentially General Anesthesia
devoid of cardiovascular and histamine-releasing A 72-year-old man has undergone general
side eff ects. Pipecuronium, on the other hand, is a anesthesia for transurethral resection of the
bisquarternary steroidal compound similar to prostate. Twenty minutes after conclusion of the
pancuronium, without the vagolytic eff ects. Onset procedure, he is still intubated and shows no
and duration of action are also similar to evidence of spontaneous respiration or
pancuronium; elimination is primarily through consciousness .
renal (70%) and biliary (20%) excretion. Th e usual What is your general approach to this diagnostic
intubating dose ranges from 0.06-0.1 mg/kg; its dilemma?
pharmacologic profi le is relatively unchanged in
Clues to the solution of complex clinical problems
elderly patients.
are usually found in a pertinent review of the medical
and surgical history, the history of drug ingestions,
NEWER MUSCLE RELAXANTS the physical examination, and laboratory results. In
Gantacurium belongs to a new class of this case, the perioperative anesthetic management
nondepolarizing neuromuscular blockers called should also be considered .
chlorofumarates. It is provided as a lyophilized What medical illnesses predispose a patient to
powder, because it is not stable as an aqueous delayed awakening or prolonged paralysis?
230 SECTION II Clinical Pharmacology
Chronic hypertension alters cerebral blood fl ow Long-acting sedatives, such as the benzodiazepines,
autoregulation and decreases the brain’s tolerance to can delay awakening .
episodes of hypotension. Liver disease reduces Does anesthetic technique alter awakening?
hepatic drug metabolism and biliary excretion,
Preoperative medications can aff ect awakening.
resulting in prolonged drug action. Reduced serum
In particular, anticholinergics (with the exception of
albumin concentrations increase free drug (active
glycopyrrolate, which does not cross the blood–brain
drug) availability. Hepatic encephalopathy can alter
barrier), opioids, and sedatives can interfere with
consciousness. Kidney disease decreases the renal
postoperative recovery. Patients with low cardiac
excretion of many drugs. Uremia can also aff ect
output may have delayed absorption of intramuscular
consciousness. Diabetic patients are prone to
injections .
hypoglycemia and hyperosmotic, hyperglycemic, and
Intraoperative hyperventilation is a common
nonketotic coma. A prior stroke or symptomatic
cause of postoperative apnea. Because volatile agents
carotid bruit increases the risk of intraoperative
raise the apneic threshold, the Pa CO 2 level at which
cerebral vascular accident. Right-to-left heart shunts,
spontaneous ventilation ceases, moderate
particularly in children with congenital heart disease,
postoperative hypoventilation may be required to
allow air emboli to pass directly from the venous
stimulate the respiratory centers. Severe
circulation to the systemic (possibly cerebral) arterial
intraoperative hypotension or hypertension may lead
circulation. A paradoxic air embolism can result in
to cerebral hypoxia and edema .
permanent brain damage. Severe hypothyroidism is
Hypothermia decreases minimum alveolar
associated with impaired drug metabolism and,
concentration, antagonizes muscle relaxation
rarely, myxedema coma .
reversal, and limits drug metabolism. Arterial hypoxia
Does an uneventful history of general anesthesia or severe hypercapnia (Pa CO 2 > 70 mm Hg) can alter
narrow the diff erential? consciousness .
Hereditary atypical pseudocholinesterase is ruled Certain surgical procedures, such as carotid
out by uneventful prior general anesthesia, assuming endarterectomy, cardiopulmonary bypass, and
succinylcholine was administered. Decreased levels of intracranial procedures, are associated with an
normal enzyme would not result in postoperative increased incidence of postoperative neurological defi
apnea unless the surgery was of very short duration. cits. Subdural hematomas can occur in severely
Malignant hyperthermia does not typically present as coagulopathic patients. Transurethral resection of the
delayed awakening, although prolonged somnolence prostate is associated with hyponatremia from the
is not unusual. Uneventful prior anesthetics do not, dilutional eff ects of absorbed irrigating solution .
however, rule out malignant hyperthermia. Persons What clues does a physical examination provide?
unusually sensitive to anesthetic agents (eg, geriatric
Pupil size is not always a reliable indicator of
patients) may have a history of delayed emergence .
central nervous system integrity. Fixed and dilated
How do drugs that a patient takes at home aff ect pupils in the absence of anticholinergic medication or
awakening from general anesthesia? ganglionic blockade (eg, the formerly used
hypotensive agent, trimethaphan), however, may be
Drugs that decrease minimum alveolar
an ominous sign. Response to physical stimulation,
concentration, such as methyldopa, predispose
such as a forceful jaw thrust, may diff erentiate
patients to anesthetic overdose. Acute ethanol
somnolence from paralysis. Peripheral nerve
intoxication decreases barbiturate metabolism and
stimulation also diff erentiates paralysis from coma .
acts independently as a sedative. Drugs that decrease
liver blood fl ow, such as cimetidine, will limit hepatic What specifi c laboratory fi ndings would you order?
drug metabolism. Antiparkinsonian drugs and tricyclic
Arterial blood gases and serum electrolytes,
antidepressants have anticholinergic side eff ects that
particularly sodium, may be helpful. Computed
augment the sedation produced by scopolamine.
tomographic scanning may be necessary if
unresponsiveness is prolonged. Increased
CHAPTER 11 Neuromuscular Blocking Agents 231
concentrations of inhalational agent provided by
respiratory gas analysis, as well as processed
electroencephalogram (EEG) measurements, may
assist in determining if the patient is still under the eff
ects of anesthesia. Slow EEG signals can be indicative
of both anesthesia and cerebral pathology. Processed
EEG awareness monitors can also be employed with
the realization that low numbers on the bispectral
index can be caused both by anesthetic suppression
of the EEG and ischemic brain injury .

What therapeutic interventions should be


considered?
Supportive mechanical ventilation should be
continued in the unresponsive patient. Naloxone, fl
umazenil, and physostigmine may be indicated,
depending on the probable cause of the delayed
emergence, if drug eff ects are suspected and reversal
is considered both safe and desirable .
H
C A P T E R

Cholinesterase Inhibitors

& Other Pharmacologic 12


Antagonists to Neuromuscular
Blocking Agents
KEYCONCEPTS

1 The primary clinical use of cholinesterase in the duration of action of a cholinesterase inhibitors, also
called anticholinesterases, is inhibitor .
to reverse nondepolarizing muscle blockade. 6 The time required to fully reverse a
2 Acetylcholine is the neurotransmitter for nondepolarizing block depends on several the entire
parasympathetic nervous system factors, including the choice and dose of (parasympathetic
ganglions and eff ector cholinesterase inhibitor administered, the cells), parts of the
sympathetic nervous muscle relaxant being antagonized, and the system (sympathetic
ganglions, adrenal extent of the blockade before reversal .
medulla, and sweat glands), some neurons 7 A reversal agent should be routinely given to in the
central nervous system, and somatic patients who have received nondepolarizing nerves innervating
skeletal muscle . muscle relaxants unless full reversal can 3 Neuromuscular transmission is blocked be
demonstrated or the postoperative when nondepolarizing muscle relaxants plan includes continued
intubation and compete with acetylcholine to bind ventilation .
to nicotinic cholinergic receptors. The 8 In monitoring a patient’s recovery from cholinesterase
inhibitors indirectly increase neuromuscular blockade, the suggested the amount of
acetylcholine available t o end points are sustained tetanus for compete with the
nondepolarizing agent, 5 sec in response to a 100-Hz stimulus in
224 SECTION II Clinical Pharmacology
thereby reestablishing neuromuscular anesthetized patients or sustained head
transmission. lift in awake patients. If neither of these
4 In excessive doses, acetylcholinesterase end points is achieved, the patient should inhibitors
can paradoxically potentiate a remain intubated and ventilation should nondepolarizing
neuromuscular blockade. be continued.
In addition, these drugs prolong the 9 Sugammadex exerts its eff ects by forming
depolarization blockade of succinylcholine . tight complexes in a 1:1 ratio with steroidal 5 Any
prolongation of action of a neuromuscular blocking agents.
nondepolarizing muscle relaxant from renal 10 C ysteine causes inactivation of
gantacurium or hepatic insuffi ciency will probably be via metabolic degradation and adduct
accompanied by a corresponding increase formation .

223

Incomplete reversal of neuromuscular blocking the adrenergic eff ects of nor adrenaline
agents and residual post-procedure paralysis are (norepinephrine). Acetylcholine is synthesized in
associated with morbidity; therefore, careful the nerve terminal by the enzyme
evaluation of neuromuscular blockade and cholineacetyltransferase, which catalyzes the
appropriate pharmacologic antagonism are reaction between acetylcoenzyme A and choline (
strongly recommended whenever muscle Figure 12–1 ). Aft er its release, acetylcholine is
relaxants are adminis- rapidly hydrolyzed by acetylcholinesterase (true
cholinesterase) into acetate and choline.
1 tered. Th e primary clinical use of
cholinesterase inhibitors, also called 2 Acetylcholine is the neurotransmitter for the
anticholinesterases, is to reverse nondepolarizing entire parasympathetic nervous system
muscle blockade. Some of these agents are also (para-
used to diagnose and treat myasthenia gravis. sympathetic ganglions and eff ector cells), parts of
More recently, newer agents, such as the sympathetic nervous system (sympathetic
cyclodextrins and cysteine, with superior ability to ganglions, adrenal medulla, and sweat glands),
reverse neuromuscular blockade from specifi c some neurons in the central nervous system, and
agents, are being investigated with promising somatic nerves innervating skeletal muscle (
results. Th is chapter reviews cholinergic Figure 12–2 ).
pharmacology and mechanisms of Cholinergic receptors have been subdivided
acetylcholinesterase inhibition and presents the into two major groups based on their reaction to
clinical pharmacology of commonly used the alkaloids muscarine and nicotine ( Figure 12–
cholinesterase inhibitors (neostigmine, 3 ). Nicotine stimulates the autonomic ganglia and
edrophonium, pyridostigmine, and skeletal muscle receptors (nicotinic receptors),
physostigmine). It concludes with a brief whereas muscarine activates end-organ eff ector
description and mechanisms of action of some cells in
unique reversal agents.

Cholinergic Pharmacology
Th e term cholinergic refers to the eff ects of the
neurotransmitter acetyl choline , as opposed to
CHAPTER 12 225

FIGURE 121 The synthesis and hydrolysis of


acetylcholine.

bronchial smooth muscle, salivary glands, and the


sinoatrial node (muscarinic receptors). Th e
central nervous system has both nicotinic and
muscarinic receptors. Nicotinic receptors are
blocked by muscle relaxants (also called
neuromuscular blockers), and muscarinic
receptors are blocked by anticholinergic drugs,
such as atropine. Although nicotinic and
muscarinic receptors diff er in their response to
some agonists (eg, nicotine, muscarine) and some
antagonists (eg, vecuronium vs atropine), they
both respond to acetylcholine ( Table 12–1 ).
Clinically available cholinergic agonists resist
hydrolysis by cholinesterase. Methacholine and
bethanechol are primarily muscarinic agonists,
whereas carbachol has both muscarinic and
nicotinic agonist activities. Methacholine by
inhalation has been used as a provocative test in
asthma, bethanechol is used for bladder atony,
and carbachol may be used topically for wide-
angle glaucoma.
When reversing neuromuscular blockade, the
primary goal is to maximize nicotinic transmission
with a minimum of muscarinic side eff ects.

Acetyl-CoA

+ CH3
+
Choline HO CH CH NCH 3
2 2
CH3

Choline
acetyltransferase

CH3
+
Acetylcholine CH 3 C O CH CH NCH 3
Acetylcholinesterase

O
226 SECTION II Clinical Pharmacology
Acetate CH 3 CO H Cholinesterase Inhibitors & Other Pharmacologic Antagonists to Neuromuscular Blocking Agents

Choline

FIGURE 122 The parasympathetic nervous system uses acetylcholine as a preganglionic and postganglionic
neurotransmitter. sites, thereby blocking neuromuscular
transmission. Reversal of blockade depends on
gradual diff usion, redistribution, metabolism, and
excretion from the body of the nondepolarizing
relaxant ( spontaneous reversal ), oft en assisted
MECHANISM OF ACTION by the administration of specifi c reversal agents (
pharmacological reversal ). Cholinesterase
3 Normal neuromuscular transmission critically
inhibitors indirectly increase the amount of
depends on acetylcholine binding to
acetylcholine available to compete with the
nicotinic cholinergic receptors on the motor
nondepolarizing agent, thereby reestablishing
endplate. Nondepolarizing muscle relaxants act by
normal neuromuscular transmission.
competing with acetylcholine for these binding
CHAPTER 12 227

Nicotine Antagonist Nondepolarizing Antimuscarinics


s relaxants Atropine
Scopolamine
Glycopyrrolate
N covalent bonds of neostigmine and pyridostigmine
N
are longer lasting.
CH3
Organophosphates , a special class of
cholinesterase inhibitors, form very stable,
Muscarine
irreversible bonds to the enzyme. Th ey are used
HO
in ophthalmology and more commonly as
CH3
pesticides. Th e clinical duration of the
+ cholinesterase inhibitors used in anesthesia,
H3C O CH2 N CH3
CH3 however, is probably most infl uenced by the rate
of drug disappearance from the plasma. Diff
FIGURE 123 The molecular structures of nicotine and erences in duration of action can be overcome by
muscarine. Compare these alkaloids with acetylcholine dosage adjustments. Th us, the normally short
( Figure 12–1 ). duration of action of edrophonium can be partially
overcome by increasing the dosage.
Cholinesterase inhibitors are also used in the
Cholinesterase inhibitors inactivate diagnosis and treatment of myasthenia gravis.
acetylcholinesterase by reversibly binding to the Mechanisms of action other than
enzyme. Th e stability of the bond infl uences the acetylcholinesterase inactivation may contribute
duration of action. Th e electrostatic attraction to the restoration of neuromuscular function.
and hydrogen bonding of edrophonium are short- Edrophonium seems to have prejunctional eff ects
lived; the that enhance the release of acetylcholine.
Neostigmine has a direct (but weak) agonist eff ect
on nicotinic receptors. Acetylcholine mobilization
TABLE 121 Characteristics of cholinergic and release by the nerve may also be enhanced (a
receptors. presynaptic mechanism).
Nicotinic Muscarinic
4 In excessive doses, acetylcholinesterase
inhibitors paradoxically potentiate a
Location Autonomic ganglia Glands
Sympathetic Lacrimal
nondepolariz-
ganglia Salivary ing neuromuscular blockade. Standard dogma
Parasympathetic Gastric states that neostigmine in high doses may cause
ganglia Smooth muscle
Skeletal muscle
receptor channel blockade; however, clinical
Bronchial evidence of this is lacking. In addition, these drugs
Gastrointestinal
Bladder
prolong the depolarization blockade of
Blood vessels succinylcholine. Two mechanisms may explain this
Heart latter eff ect: an increase in acetylcholine (which
Sinoatrial node increases motor end-plate depolarization) and
inhibition of pseudocholinesterase activity.
Atrioventricul Neostigmine and to some extent pyridostigmine
ar node
display some limited pseudocholinesterase-
Agonists Acetylcholine Nicotine Acetylcholine inhibiting activity, but their eff ect on
Muscarine
acetylcholinesterase is much greater.
Edrophonium has little or no eff ect on
pseudocholinesterase. In large doses, neostigmine
228 SECTION II Clinical Pharmacology
can cause a weak depolarizing neuromuscular Inactivation of nicotinic acetylcholine
blockade. receptors in the central nervous system may
play a role in the action of general anesthetics.
Unlike physostigmine, cholinesterase
CLINICAL PHARMACOLOGY inhibitors used to reverse neuromuscular
blockers do not cross the blood–brain barrier.
General Pharmacological
Gastrointestinal receptors —Muscarinic
Characteristics stimulation increases peristaltic activity
Th e increase in acetylcholine caused by (esophageal, gastric, and intestinal) and
cholinesterase inhibitors aff ects more than the glandular secretions (eg, salivary).
nicotinic receptors of skeletal muscle ( Table 12– Postoperative nausea, vomiting, and
2 ). Cholinesterase fecal incontinence have been attributed
Cholinesterase Inhibitors & Other Pharmacologic Antagonists to Neuromuscular Blocking Agents

TABLE 122 Muscarinic side eff ects of to the use of cholinesterase inhibitors.
cholinesterase inhibitors. Unwanted muscarinic side eff ects are
Organ System Muscarinic Side Eff ects minimized by prior or concomitant administration
of anticholinergic medications, such as atropine
Cardiovascular Decreased heart rate,
bradyarrhythmias
sulfate or glycopyrrolate. Th e duration of action is
similar among the cholinesterase inhibitors.
Pulmonary Bronchospasm, bronchial
Clearance is due to both hepatic metabolism (25%
secretions
to 50%) and
Cerebral Diffuse excitation 1

5 renal excretion (50% to 75%). Th us, any


Gastrointestinal Intestinal spasm, increased
salivation prolongation of action of a nondepolarizing
mus-
Genitourinary Increased bladder tone
cle relaxant from renal or hepatic insuffi ciency
Ophthalmological Pupillary constriction will probably be accompanied by a corresponding
1
Applies only to physostigmine. increase in the duration of action of a
cholinesterase inhibitor.
As a rule, no amount of cholinesterase
inhibitor can immediately reverse a block that is so
inhibitors can act at cholinergic receptors of intense that there is no response to tetanic
several other organ systems, including the peripheral nerve stimulation. Moreover, the
cardiovascular and gastrointestinal systems. absence of any palpable single twitches following
Cardiovascular receptors —Th e predominant 5 sec of tetanic stimulation at 50 Hz implies a very
muscarinic eff ect on the heart is bradycardia intensive blockade that cannot be reversed.
that can progress to sinus arrest. Excessive doses of cholinesterase inhibitors may
actually prolong recovery. Some evidence of
Pulmonary receptors —Muscarinic stimulation can
spontaneous recovery (ie, the fi rst twitch of the
result in bronchospasm (smooth muscle
train-of-four [TOF]) should be present before
contraction) and increased respiratory tract
reversal is attempted. Th e posttetanic count (the
secretions.
number of palpable twitches aft er tetanus)
Cerebral receptors —Physostigmine is a generally correlates with the time of return of the
cholinesterase inhibitor that crosses the fi rst twitch of the TOF and therefore the ability to
blood– brain barrier and can cause diff use
reverse intense paralysis. For intermediate-acting
activation of the electroencephalogram by
agents, such as atracurium and vecuronium, a
stimulating muscarinic and nicotinic receptors
palpable posttetanic twitch appears about 10 min
within the central nervous system.
before spontaneous recovery of the fi rst twitch of
CHAPTER 12 229
the TOF. In contrast, for longer-acting agents, such muscle relaxants unless full reversal can be
as pancuronium, the fi rst twitch of the TOF demonstrated or the postoperative plan includes
appears about 40 min aft er a palpable posttetanic continued intubation and ventilation. In the latter
twitch. situation, adequate sedation must also be
provided.
6 Th e time required to fully reverse a A peripheral nerve stimulator should also be
nondepolarizing block depends on several used to monitor the progress and confi rm the
factors, including the choice and dose of adequacy of reversal. In general, the higher the
cholinesterase inhibitor administered, the frequency of stimulation, the greater the
muscle relaxant being antagonized, and the sensitivity of the test (100-Hz tetany > 50-Hz
extent of the blockade before reversal. For tetany or TOF > single-twitch height). Clinical signs
example, reversal with edrophonium is of adequate reversal also vary in sensitivity
usually faster than with neostigmine; large
(sustained head lift > inspiratory force > vital
doses of neostigmine lead to faster reversal
capacity > tidal volume).
than small doses; intermediate-acting
relaxants reverse sooner than long-acting 8 Th erefore, the suggested end points of
relaxants; and a shallow block is easier to
recovery are sustained tetanus for 5 sec in
reverse than a deep block (ie, twitch height
>10%). Intermediate-acting muscle relaxants response
therefore require a lower dose of reversal to a 100-Hz stimulus in anesthetized patients or
agent (for the same degree of blockade) than sustained head or leg lift in awake patients.
long-acting agents, and concurrent excretion Newer quantitative methods for assessing
or metabolism provides a proportionally recovery from neuromuscular blockade, such as
faster reversal of the short- and acceleromyography, may further reduce the
intermediate-acting agents. Th ese incidence of residual postoperative neuromuscular
advantages can be lost in conditions paralysis.
associated with severe end-organ disease
(eg, the use of vecuronium in a patient with
liver failure) or enzyme defi ciencies (eg,
Specifi c Cholinesterase
mivacurium in a patient with homozygous Inhibitors
atypical pseudocholinesterase). Depending
on the dose of muscle relaxant that has been
given, spontaneous recovery to a level
NEOSTIGMINE
adequate for pharmacological reversal may
take more than 1 hr with long-acting muscle Physical Structure
relaxants because of their insignifi cant Neostigmine consists of a carbamate moiety and a
metabolism and slow excretion. Factors quaternary ammonium group ( Figure 12–4 ). Th e
associated with faster reversal are also former provides covalent bonding to
associated with a lower incidence of residual acetylcholinesterase. Th e latter renders the
paralysis in the recovery room and a lower molecule lipid insoluble, so that it cannot pass
risk of postoperative respiratory through the blood–brain barrier.
complications.
Dosage & Packaging
7 A reversal agent should be routinely given to Th e maximum recommended dose of
patients who have received nondepolarizing neostigmine is 0.08 mg/kg (up to 5 mg in adults),
but smaller
230 SECTION II Clinical Pharmacology

Neostigmine Pyridostigmine
H3C H3C
+ +
N C O N(CH3)3 N C O N CH3
H3C H3C
O O

Edrophonium Physostigmine
CH3 CH3
+
HO N CH3 H3C N C O
CH3
H O
N N

CH3 CH3

FIGURE 124 The molecular structures of neostigmine, pyridostigmine, edrophonium, and physostigmine.
Cholinesterase Inhibitors & Other Pharmacologic Antagonists to Neuromuscular Blocking Agents

TABLE 123 The choice and dose of cholinesterase inhibitor determine the choice and dose of
anticholinergic.
Usual Dose of
Usual Dose of Recommended Anticholinergic per mg of
Cholinesterase Inhibitor Cholinesterase Inhibitor Anticholinergic Cholinesterase Inhibitor

Neostigmine 0.04–0.08 mg/kg Glycopyrrolate 0.2 mg

Pyridostigmine 0.1–0..25 mg/kg Glycopyrrolate 0.05 mg

Edrophonium 0.5–1 mg/kg Atropine 0.014 mg

Physostigmine 1
0.01–0.03 mg/kg Usually not necessary NA
1
Not used to reverse muscle relaxants. is mild to moderate and a dose of 0.08 mg/kg (or 5
mg) if intense paralysis is being reversed. Th e
duration of action is prolonged in geriatric
amounts oft en suffi ce and larger doses have also patients. Muscarinic side eff ects are minimized by
been given safely ( Table 12–3 ). Neostigmine is prior or concomitant administration of an
most commonly packaged as 10 mL of a 1 mg/mL anticholinergic agent. Th e onset of action of
solution, although 0.5 mg/mL and 0.25 mg/mL glycopyrrolate (0.2 mg glycopyrrolate per 1 mg of
concentrations are also available. neostigmine) is similar to that of neostigmine and
is associated with less tachycardia than is
Clinical Considerations experienced with atropine (0.4 mg of atropine per
Th e eff ects of neostigmine (0.04 mg/kg) are 1 mg of neostigmine). It has been reported that
usually apparent in 5min, peak at 10 min, and last neostigmine crosses the placenta, resulting in fetal
more than 1 hr. If reversal is not complete in 10 bradycardia. Th us, theoretically , atropine may be
min aft er 0.08 mg/kg, the time for full recovery of a better choice of an anticholinergic agent than
neuromuscular function will depend on the glycopyrrolate in pregnant patients receiving
nondepolarizing agent used and the intensity of neostigmine, but there is no evidence that this
blockade. In practice, many clinicians use a dose of makes any diff erence in patient outcomes.
0.04 mg/kg (or 2.5 mg) if the preexisting blockade
CHAPTER 12 231
Neostigmine is also used to treat myasthenia any of the cholinesterase inhibitors. Reduced
gravis, urinary bladder atony, and paralytic ileus. doses should not be used, because longer-acting
PYRIDOSTIGMINE muscle relaxants may outlast the eff ects of
edrophonium. Higher doses prolong the duration
Physical Structure of action to more than 1 hr. Edrophonium may not
Pyridostigmine is structurally similar to be as eff ective as neostigmine at reversing
neostigmine except that the quaternary intense neuromuscular blockade. In equipotent
ammonium is incorporated into the phenol ring. doses, muscarinic eff ects of edrophonium are less
Pyridostigmine shares neostigmine’s covalent pronounced than those of neostigmine or
binding to acetylcholinesterase and its lipid pyridostigmine, requiring only half the amount of
insolubility. anticholinergic agent. Edrophonium’s rapid onset
is well matched to that of atropine (0.014 mg of
Dosage & Packaging atropine per 1 mg of edrophonium). Although
Pyridostigmine is 20% as potent as neostigmine glycopyrrolate (0.007 mg per 1 mg of
and may be administered in doses up to 0.25 edrophonium) can also be used, it should be given
mg/kg (a total of 20 mg in adults). It is available as several minutes prior to edrophonium to avoid the
a solution of 5 mg/mL. possibility of bradycardia.

Clinical Considerations
Th e onset of action of pyridostigmine is slower PHYSOSTIGMINE
(10–15 min) than that of neostigmine, and its
Physical Structure
duration is slightly longer (>2 h). Glycopyrrolate
Physostigmine, a tertiary amine, has a carbamate
(0.05 mg per 1 mg of pyridostigmine) or atropine
group but no quaternary ammonium. Th erefore,
(0.1 mg per 1 mg of pyridostigmine) must also be
it is lipid soluble and is the only clinically available
administered to prevent bradycardia.
cholinesterase inhibitor that freely passes the
Glycopyrrolate is preferred because its slower
blood– brain barrier.
onset of action better matches that of
pyridostigmine, again resulting in less tachycardia.
Dosage & Packaging
Th e dose of physostigmine is 0.01–0.03 mg/kg. It
EDROPHONIUM is packaged as a solution containing 1 mg/mL.
Physical Structure
Because it lacks a carbamate group, edrophonium Clinical Considerations
must rely on noncovalent bonding to the Th e lipid solubility and central nervous system
acetylcholinesterase enzyme. Th e quaternary penetration of physostigmine limit its usefulness
ammonium group limits lipid solubility. as a reversal agent for nondepolarizing blockade,
but make it eff ective in the treatment of central
Dosage & Packaging
anticholinergic toxicity caused by overdoses of
Edrophonium is less than 10% as potent as
atropine or scopolamine. In addition, it reverses
neostigmine. Th e recommended dosage is 0.5–1
some of the central nervous system depression
mg/kg. Edrophonium is available as a solution
and delirium associated with use of
containing 10 mg/mL; it is available with atropine
benzodiazepines and volatile anesthetics.
as a combination drug (Enlon-Plus; 10 mg
Physostigmine (0.04 mg/kg) has been shown to be
edrophonium and 0.14 mg atropine per mL).
eff ective in preventing postoperative shivering. It
reportedly partially antagonizes morphine-induced
Clinical Considerations respiratory depression, presumably because
Edrophonium has the most rapid onset of action morphine reduces acetylcholine release in the
(1–2 min) and the shortest duration of eff ect of
232 SECTION II Clinical Pharmacology
brain. Th ese eff ects are transient, and repeated NONCLASSIC REVERSAL
doses may be required. Bradycardia is infrequent
in the recommended dosage range, but atropine AGENTS
should be immediately available. Because Besides cholinesterase inhibitors, two unique
glycopyrrolate does not cross the blood–brain drugs (sugammadex and L-cysteine) are currently
barrier, it will not reverse the central nervous under investigation in the United States; these
system eff ects of physostigmine. Other possible agents act as selective antagonists of
muscarinic side eff ects include excessive nondepolarizing neuromuscular blockade.
salivation, vomiting, and convulsions. In contrast Sugammadex is able to reverse aminosteroid-
to other cholinesterase inhibitors, physostigmine induced neuromuscular blockade, whereas
is almost completely metabolized by plasma cysteine has been shown to reverse the
esterases, so renal excretion is not important. neuromuscular blocking eff ects of gantacurium
and other fumarates.

OTHER CONSIDERATIONS
Recovery from neuromuscular blockade is infl
SUGAMMADEX
uenced by the depth of block at the time of Sugammadex is a novel selective relaxant-binding
antagonism, clearance and half-life of the relaxant agent that is currently available for clinical use in
used, and other factors that aff ect neuromuscular Europe. It is a modifi ed gamma-cyclodextrin (su
blockade ( Table 12–4 ), such as drugs and refers to sugar, and gammadex refers to the
electrolyte structural molecule gamma-cyclodextrin).

Physical Structure
Its three-dimensional structure resembles a
TABLE 124 Factors potentiating hollow truncated cone or doughnut with a
neuromuscular blockade. hydrophobic cavity and a hydrophilic exterior. H
Drugs ydrophobic
Volatile anesthetics
Antibiotics: Aminoglycosides, polymyxin B, neomycin, 9 interactions trap the drug (eg, rocuronium) in
tetracycline, clindamycin
Dantrolene the cyclodextrin cavity (doughnut hole), resulting
Verapamil in tight formation of a water-soluble guest–host
Furosemide complex in a 1:1 ratio. Th is terminates the
Lidocaine neuromuscular blocking action and restrains the
Electrolytes and acid–base disorders drug in extracellular fl uid where it cannot interact
Hypermagnesemia with nicotinic acetylcholine receptors.
Hypocalcemia
Sugammadex is essentially eliminated unchanged
Hypokalemia
Respiratory acidosis via the kidneys.
Temperature
Hypothermia Clinical Considerations Sugammadex has
Cholinesterase Inhibitors & Other Pharmacologic Antagonists to Neuromuscular Blocking Agents been
disturbances. In addition, some specifi c agents
with the potential of reversing the neuromuscular administered in doses of 4–8 mg/kg. With an
blocking eff ects of nondepolarizing muscle injection of 8 mg/kg, given 3 min aft er
relaxants merit brief discussion. administration of 0.6 mg/kg of rocuronium,
recovery of TOF ratio to 0.9 was observed within 2
min. It produces rapid and eff ective reversal of
both shallow and profound rocuronium-induced
CHAPTER 12 233
neuromuscular blockade in a consistent manner. Which drugs administered to this patient could
Because of some concerns about hypersensitivity explain her hypoventilation?
and allergic reactions, sugammadex has not yet Isofl urane, morphine sulfate, and vecuronium all
been approved by the US Food and Drug interfere with a patient’s ability to maintain a normal
Administration. ventilatory response to an elevated Pa CO 2 .

Why would the patient’s breathing worsen in the


L CYSTEINE recovery room?

L -cysteine is an endogenous amino acid that is oft Possibilities include the delayed onset of action
of morphine sulfate, a lack of sensory stimulation in
en added to total parenteral nutrition regimens 10 the recovery area, fatigue of respiratory muscles, and
to enhance calcium and phosphate solubility. splinting as a result of upper abdominal pain.
Th e ultrashort-acting neuromuscular Could the patient still have residual
blocker, gantacurium, and other fumarates rapidly neuromuscular blockade?
combine with L - cysteine in vitro to form less If the dose of neostigmine was not determined
active degradation products (adducts). Exogenous by the response to a peripheral nerve stimulator, or if
administration of L -cysteine (10–50 mg/kg the recovery of muscle function was inadequately
intravenously) given to anesthetized monkeys 1 tested after the reversal drugs were given, persistent
min aft er these neuromuscular blocking agents neuromuscular blockade is possible. Assume, for
abolished the block within 2–3 min; this example, that the patient had minimal or no response
antagonism was found to be superior to that to initial tetanic stimulation at 100 Hz. Even the
produced by anticholinesterases. Th is unique maximal dose of neostigmine (5 mg) might not yet
method of antagonism by adduct formation and have adequately reversed the paralysis. Because of
inactivation is still in the investigative stage, enormous patient variability, the response to
especially in terms of its safety and effi cacy in peripheral nerve stimulation must always be
humans. monitored when intermediate- or longacting muscle
relaxants are administered. Even if partial reversal is
achieved, paralysis may worsen if the patient
CASE DISCUSSION hypoventilates . Other factors (in addition to
respiratory acidosis) that impair the reversal of
Respiratory Failure in nondepolarizing muscle relaxants include intense
the Recovery Room neuromuscular paralysis, electrolyte disturbances
A 66-year-old woman weighing 85 kg is brought (hypermagnesemia, hypokalemia, and
to the recovery room following cholecystectomy.
hypocalcemia), hypothermia (temperature <32°C),
The anesthetic technique included the use of isofl
drug interactions (see T able 11–4) , m etabolic
urane and vecuronium for muscle relaxation. At
alkalosis (from accompanying hypokalemia and
the conclusion of the procedure, the
hypocalcemia), and coexisting diseases (see Table
anesthesiologist administered 6 mg of morphine
11–8 ).
sulfate for postoperative pain control and 3 mg of
neostigmine with 0.6 mg of glycopyrrolate to How could the extent of reversal be tested?
reverse any residual neuromuscular blockade. The Tetanic stimulation is a sensitive but
dose of cholinesterase inhibitor was empirically uncomfortable test of neuromuscular transmission in
based on clinical judgment. Although the patient an awake patient. Because of its shorter duration,
was apparently breathing normally on arrival in double-burst stimulation is tolerated better than
the recovery room, her tidal volume progressively tetany by conscious patients. Many other tests of
diminished. Arterial blood gas measurements neuromuscular transmission, such as vital capacity
revealed a Pa CO 2 of 62 mm Hg, a Pa O 2 of 110 mm and tidal volume, are insensitive as they may still
Hg, and a pH of 7.26 on a fraction of inspired seem normal when 70% to 80% of receptors are
oxygen (F IO 2) of 40%. blocked. In fact, 70% of receptors may remain blocked
234 SECTION II Clinical Pharmacology
despite an apparently normal response to TOF
stimulation. The ability to sustain a head lift for 5 sec,
however, indicates that fewer than 33% of receptors
are occupied by muscle relaxant .
What treatment would you suggest?
Ventilation should be assisted to reduce the
respiratory acidosis. Even if diaphragmatic function
seems to be adequate, residual blockade can lead
to airway obstruction and poor airway protection.
More neostigmine (with an anticholinergic) could
be administered up to a maximum recommended
dose of 5 mg. If this does not adequately reverse
paralysis, mechanical ventilation and airway
protection should be instituted and continued until
neuromuscular function is fully restored .

SUGGESTED READING
Naguib M: Sugammadex: another milestone in clinical
neuromuscular pharmacology. Anesth Analg
2007;104:575.
Naguib M, Lien CA: Pharmacology of muscle relaxants
and their antagonists. In: Miller’s Anesthesia , 7th
ed. Miller RD, Eriksson LI, Wiener-Kronish JP, Young
WL (editors). Churchill Livingstone, 2010.
Savarese JJ, McGilvra JD, Sunaga H, et al: Rapid chemical
antagonism of neuromuscular blockade by L-cysteine
adduction to and inactivation of the olefi nic (double-
bonded) isoquinoliniumdiester compounds
gantacurium (AV430A), CW 002, and CW 011.
Anesthesiology 2010;113:58.
Stoelting RK: Pharmacology and Physiology in
Anesthetic Practice , 4th ed. Lippincott, William &
Wilkins, 2005.
Taylor P: Anticholinesterase agents. In: Goodman and
Gilman’sPharmacological Basis of Th erapeutics ,
12th ed. Hardman JG (editor). McGraw-Hill, 2011.

You might also like