You are on page 1of 36

CHAPTER 8

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 .
5 The unitary hypothesis proposes that all
inhalation agents share a common
CHAPTER 8 155
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, potent inhalation


chloroform, and agent, but its high
ether were the fi rst solubility and low
universally accepted vapor pressure
general anesthetics. yielded longer
Methoxyfl urane inductions and
and enfl urane, two emergences. Up to
potent halogenated 50% of it was
agents, were used metabolized by
for many years in cytochrome P-450
North American (CYP) enzymes to
anesthesia practice. free fl uoride (F − ),
Methoxyfl urane oxalic acid, and
was the most other nephrotoxic
156 SECTION II Clinical Pharmacology
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
electroencephalogra well. Regardless of
phic 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. elimination of these
Five inhalation agents.
agents continue to Because of
be used in clinical their unique route
anesthesiology: of administration,
CHAPTER 8 157
inhalation concentration in the
anesthetics have central nervous
useful system (CNS). Th
pharmacological ere are many steps
properties not in between the
shared by other anesthetic vaporizer
anesthetic agents. and the anesthetic’s
For instance, deposition in the
administration via brain ( Figure 8–1 ).
the pulmonary
circulation allows a
more rapid
appearance of the
drug in arterial
blood than
intravenous
administration.

Pharma
cokineti
cs of
Inhalati
on
Anesthe
tics

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
depends on
attainment of a
therapeutic tissue
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% Pharma
oxygen for 5–10 min
aft er discontinuing codyna
nitrous oxide. Th e mics of
rate of recovery is
usually faster than
Inhalati
induction because on
tissues that have Anesthe
not reached
equilibrium will
tics
continue to take up
anesthetic until the
alveolar partial THEORIE
pressure falls below S OF
the tissue partial
pressure. For ANESTHE
instance, fat will TIC
continue to take up
anesthetic and ACTION
hasten recovery General anesthesia
until the partial is an altered
pressure exceeds physiological state
the alveolar partial characterized by
pressure. Th is reversible loss of
redistribution is not consciousness,
as useful aft er analgesia, amnesia,
prolonged and some degree of
anesthesia (fat muscle relaxation.
partial pressures of Th e multitude of
anesthetic will have substances capable
come “closer” to of producing
arterial partial general anesthesia
pressures at the is remarkable: inert
time the anesthetic elements (xenon),
was removed from simple inorganic
fresh gas)—thus, compounds (nitrous
the speed of oxide), halogenated
recovery also hydrocarbons
depends on the (halothane), ethers
length of time the (isofl urane, sevofl
anesthetic has been urane, desfl urane),
administered. and complex
170 SECTION II Clinical Pharmacology
organic structures nonspecifi c
(propofol). A manner, thereby aff
unifying theory ecting the
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
CHAPTER 8 171
horn interneurons
that are involved in 5 Past
pain transmission. understanding of
Diff ering aspects of anesthetic action
anesthesia may be attempted to
related to diff erent identify a unitary
sites of anesthetic hypothesis of
action. For example, anesthetic eff ects.
unconsciousness Th is hypothesis
and amnesia are proposes that all
probably mediated inhalation agents
by cortical share a common
anesthetic action, mechanism of
whereas the action at the
suppression of molecular level. Th
purposeful is was previously
withdrawal from supported by the
pain may be related observation that the
to subcortical anesthetic potency
structures, such as of inhalation agents
the spinal cord or correlates directly
brain stem. One with their lipid
study in rats solubility (Meyer–
revealed that Overton rule). Th e
removal of the implication is that
cerebral cortex did anesthesia results
not alter the from molecules
potency of the dissolving at specifi
anesthetic! Indeed, c lipophilic sites. Of
measures of course, not all lipid-
minimal alveolar soluble molecules
concentration are anesthetics
(MAC), the (some are actually
anesthetic convulsants), and
concentration that the correlation
prevents movement between anesthetic
in 50% of subjects or potency and lipid
animals, are solubility is only
dependent upon approximate (
anesthetic eff ects Figure 8–4 ).
at the spinal cord Neuronal
and not at the membranes contain
cortex. a multitude of
hydrophobic sites in
172 SECTION II Clinical Pharmacology
their phospholipid aff ect neuronal
bilayer. Anesthetic transmission and
binding to these away from the
sites could expand critical volume
the bilayer beyond a hypothesis.
critical amount, General
altering membrane anesthetic action
function (critical could be due to
volume hypothesis). alterations in any
Although this theory one (or a
is almost certainly combination) of
an oversimplifi several cellular
cation, it explains an systems, including
interesting voltage-gated ion
phenomenon: the channels, ligand-
reversal of gated ion channels,
anesthesia by second messenger
increased pressure. functions, or
Laboratory animals neurotransmitter
exposed to elevated receptors. For
hydrostatic pressure example, many
develop a resistance anesthetics enhance
to anesthetic eff GABA inhibition of
ects. Perhaps the the CNS.
pressure is Furthermore, GABA
displacing a number receptor agonists
of molecules from seem to enhance
the membrane or anesthesia, whereas
distorting the GABA antagonists
anesthetic binding reverse some
sites in the anesthetic eff ects.
membrane, Th ere seems to be a
increasing strong correlation
anesthetic between anesthetic
requirements. potency and
However, studies in potentiation of
the 1980s GABA receptor
demonstrated the activity. Th us,
ability of anesthetics anesthetic action
to inhibit protein may relate to
actions, shift ing binding in relatively
attention to the hydrophobic
numerous ion domains in channel
channels that might proteins (GABA
CHAPTER 8 173
receptors). ongoing for many
Modulation of GABA years,
function may prove
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
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


Inhalation Anesthetics temperature and ambient pressure. It can be
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 contractility in TABLE 85 Advantages and
NMDA receptor antagonist. disadvantages of xenon (Xe) anesthesia.
4
Percentage of absorbed anesthetic undergoing metabolism.

Eff ects on Organ Systems


A. Cardiovascular
Nitrous oxide has a tendency to stimulate the vitro, arterial blood pressure, cardiac output, and
sympathetic nervous system. Th us, even though heart rate are essentially unchanged or slightly
nitrous oxide directly depresses myocardial elevated in vivo because of its stimulation of
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

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

You might also like