General Anesthetics

Download Report

Transcript General Anesthetics

General Anesthetics
Yacoub M. Irshaid, MD, PhD, ABCP
Department of Pharmacology
General Anesthetics
• General anesthesia is typically a
state of analgesia, amnesia, loss of
consciousness, inhibition of
sensory and autonomic reflexes,
and skeletal muscle relaxation.
• This is achieved by a combination
of intravenous and inhaled drugs.
General Anesthetics
Types of General Anesthesia:
A. Intravenous agents used alone, or in
combination with other anesthetic
agents, to achieve an anesthetic state
or sedation. These drugs include:
1. Barbiturates: Thiopental, methohexital.
2. Benzodiazepines: Midazolam,
diazepam.
3. Propofol.
General Anesthetics
4. Ketamine.
5. Opioid analgesics: Morphine, fentanyl,
sufentanil, alfentanil, remifentanil.
6. Miscellaneous sedative-hypnotics:
Etomidate, dexmedetomidine.
B. Inhaled anesthetics which include:
1. Volatile liquids: Halothane, isoflurane,
desflurane, enflurane, methoxyflurane,
and sevoflurane.
2. Gases: Nitrous oxide.
General Anesthetics
Balanced Anesthesia:
Although general anesthesia can be
produced by only intravenous or only inhaled
anesthetic agents, modern anesthesia
typically involves a combination of:
1. IV agents for induction of anesthesia.
2. Inhaled agents for maintenance of
anesthesia.
3. Muscle relaxants.
4. Analgesics.
5. Cardiovascular drugs to control autonomic
responses.
Inhaled Anesthetics
Pharmacokinetics:
• An adequate depth of anesthesia
depends on achieving therapeutic
concentrations in the central nervous
system.
• The rate at which an effective brain
concentration is achieved (time to
induction of anesthesia) depends on
multiple pharmacokinetic factors that
influence brain uptake and tissue
distribution of the anesthetic agent:
Inhaled Anesthetics
1. Uptake and distribution of inhaled
anesthetics:
Achievement of a brain concentration
of an inhaled anesthetic to provide
adequate anesthesia requires transfer
of the anesthetic from the alveolar air
to the blood, and from the blood to the
brain.
The rate of achievement of such a
concentration depends on:
Inhaled Anesthetics
A. Solubility of the anesthetic:
• The blood:gas partition coefficient is a
useful index of solubility, and defines
the relative affinity of the anesthetic
for the blood compared with that of
inspired gas.
• The partition coefficients for
desflurane and nitrous oxide, which
are relatively insoluble in blood, are
extremely low.
Inhaled Anesthetics
• Thus, when such agents diffuse from the
lung into the arterial blood, relatively few
molecules are required to raise its partial
pressure, and therefore the arterial
tension rises rapidly.
• Conversely, for anesthetics with
moderate-to-high solubility (halothane,
isoflurane), more molecules dissolve
before partial pressure rises significantly,
and arterial tension of the gas increases
less rapidly.
Inhaled Anesthetics
• Nitrous oxide and desflurane (and to a
lesser extent sevoflurane), with low
solubility in blood, reaches high arterial
tensions rapidly, which in turn results
in rapid equilibration with the brain and
faster onset of action.
Why induction of anesthesia is slower with more soluble anesthetic gases. In this schematic
diagram, solubility in blood is represented by the relative size of the blood compartment (the more
soluble, the larger the compartment). Relative partial pressures of the agents in the compartments
are indicated by the degree of filling of each compartment. For a given concentration or partial
pressure of the two anesthetic gases in the inspired air, it will take much longer for the blood partial
pressure of the more soluble gas (halothane) to rise to the same partial pressure as in the alveoli.
Since the concentration of the anesthetic agent in the brain can rise no faster than the concentration
in the blood, the onset of anesthesia will be slower with halothane than with nitrous oxide.
Tensions of three anesthetic gases in arterial blood as a function of time after
beginning inhalation. Nitrous oxide is relatively insoluble (blood:gas partition
coefficient = 0.47); methoxyflurane is much more soluble (coefficient = 12); and
halothane is intermediate (2.3).
Inhaled Anesthetics
B. Anesthetic concentration in the
inspired air:
The concentration of an inhaled
anesthetic in the inspired gas mixture
has direct effects on both the maximum
tension in the alveoli and the rate of
increase in its tension in the arterial
blood.
Inhaled Anesthetics
• Increases in the inspired anesthetic
concentration increases the rate of
induction of anesthesia.
• Advantage is taken of this effect in
anesthetic practice. For example, a
high concentration of isoflurane (1.5%)
is used for an increased rate of
induction, which is then reduced (0.751%) for maintenance of anesthesia.
Inhaled Anesthetics
• Similarly, moderately soluble
anesthetics are often administered in
combination with a less soluble agents
to reduce the time needed for loss of
consciousness and achievement of a
surgical depth of anesthesia. (nitrous
oxide + halothane).
Inhaled Anesthetics
C. Pulmonary ventilation:
The rate of rise of anesthetic gas tension
in arterial blood is directly dependent on
both the rate and depth of ventilation. The
magnitude of the effect depends on
blood:gas partition coefficient.
• An increase in pulmonary ventilation is
accompanied by only a slight increase in
arterial tension of an anesthetic with low
blood solubility, but can significantly
increase tension of agents with moderateto-high blood solubility.
Inhaled Anesthetics
• Fore example, a 4-fold increase in
ventilation rate almost doubles arterial
tension of halothane during the first 10
minutes of anesthesia but increases
the arterial tension of nitrous oxide by
only 15%.
Ventilation rate and arterial anesthetic tensions. Increased ventilation (8 versus 2
L/min) has a much greater effect on equilibration of halothane than nitrous oxide.
Inhaled Anesthetics
• Therefore, hyperventilation increases
the speed of induction of anesthesia
with inhaled anesthetics that would
normally have a slow onset.
• Depression of respiration by opioid
analgesics slows the onset of
anesthesia of inhaled anesthetics if
ventilation is not manually or
mechanically assisted.
Inhaled Anesthetics
D. Pulmonary blood flow:
Changes in blood flow to and from the
lungs influence transfer processes of
anesthetic gases.
An increase in pulmonary blood flow
slows the rate of rise in arterial tension,
particularly for agents with moderateto-high blood solubility.
Inhaled Anesthetics
• Increased pulmonary blood flow
exposes a large volume of blood to the
anesthetic; thus, blood capacity
increases and the anesthetic tension
rises slowly.
• A decrease in pulmonary blood flow
has the opposite effect, increasing the
rate of rise of arterial tension of inhaled
anesthetics.
Inhaled Anesthetics
• In patients with circulatory shock, the
combined effect of decreased cardiac
output and increased ventilation will
accelerate induction of anesthesia with
halothane and isoflurane. This is less
likely with less soluble agents such as
nitrous oxide and desflurane.
Inhaled Anesthetics
E. Arteriovenous concentration gradient:
• The anesthetic concentration gradient
between arterial and mixed venous blood
is dependent mainly on the uptake of
anesthetic by the tissue
• Venous blood returning to the lungs may
contain significantly less anesthetic than
arterial blood.
• The greater this difference, the more time
it will take to achieve equilibrium with
brain tissue.
Inhaled Anesthetics
2. Elimination of inhaled anesthetics:
• The time of recovery from inhalation
anesthesia depends on the rate of
elimination of the anesthetic from the
brain.
• Many of the processes of anesthetic
transfer during recovery are simply
the reverse of those that occur
during induction of anesthesia.
Inhaled Anesthetics
• The blood:gas partition coefficient of
the anesthetic is one of the most
important factors governing recovery,
which include pulmonary blood flow,
ventilation magnitude, and tissue
solubility of the anesthetic.
• Two features of recovery are different
from what happens during induction:
Inhaled Anesthetics
1. Although the transfer of the anesthetic
from the lungs to the blood can be
enhanced by increasing its
concentration in inspired air, the reverse
can not be enhanced, because the
concentration in the lung can not be
reduced below zero.
2. At the beginning of recovery, the
anesthetic gas tension in different
tissues may be variable. In contrast, with
induction the initial anesthetic tension is
zero in all tissues.
Inhaled Anesthetics
• Inhaled anesthetics that are relatively
insoluble in blood (low blood:gas
partition coefficient) and brain (?) are
eliminated at faster rates than more
soluble anesthetics. The washout of
nitrous oxide, desflurane, and
sevoflurane occurs at a rapid rate 
more rapid recovery from their
anesthetic effect compared to
halothane and isoflurane.
Inhaled Anesthetics
• Halothane is ~ twice as soluble in brain
tissue and 5X more soluble in blood
than nitrous oxide and desflurane 
more slow elimination and less rapid
recovery from halothane anesthesia.
• The duration of exposure to the
anesthetic can have a marked effect on
recovery time, especially for more
soluble anesthetics.
Inhaled Anesthetics
• Accumulation of (isoflurane) in muscle,
skin and fat increases with prolonged
inhalation, and blood tension may
decline slowly during recovery.
• When the exposure is short, recovery
may be rapid even with the more
soluble agents.
• Clearance of the inhaled anesthetics by
the lungs is the major route of
elimination from the body.
Inhaled Anesthetics
• Hepatic metabolism may also
contribute to the elimination of
halothane (~ 40% during an average
anesthetic procedure).
• Oxidative metabolism (CYP2E1) of
halothane results in formation of
trifluoroacetic acid and release of
chloride and bromide ions.
Inhaled Anesthetics
• Under conditions of low oxygen
tension, halothane is metabolized to
the chlorotrifluoroethyl free radical
which is capable of reacting with
hepatic cell membrane and producing
halothane hepatitis.
• < 10% of enflurane is metabolized.
• Isoflurane and desflurane are the least
metabolized of fluorinated anesthetics.
Inhaled Anesthetics
• The metabolism of methoxyflurane
(70%) results in elevation of renal
fluoride levels and nephrotoxicity.
• Enflurane and sevoflurane metabolism
leads to formation of fluoride ions but
do not reach toxic levels.
• Nitrous oxide is not metabolized by
human tissues, but can be metabolized
by bacteria in the GIT.
Inhaled Anesthetics
• Sevoflurane is degraded by contact
with the carbon dioxide absorbent
(soda lime = Ca(OH)2 (about 75%), H2O
(about 20%), NaOH (about 3%), KOH
(about 1%)) in anesthesia machines
yielding a vinyl ether which can cause
renal damage if high concentrations are
absorbed.
General Anesthetics
Pharmacodynamics:
• Both the inhaled and intravenous
anesthetics can depress
spontaneous and evoked activity
of neurons in many regions of the
brain, with several potential
molecular targets for anesthetic
actions.
General Anesthetics
• Interaction of the anesthetics with
specific nerve membrane
components results in
modification of ion currents,
particularly the ligand-gated ion
channel family.
General Anesthetics
• A primary molecular target of general
anesthetics (halogenated inhalational
agents, propofol, barbiturates,
etomidate, ..) is the GABAA receptorchloride channel, a major mediators of
inhibitory synaptic transmission. Either
it is directly activated or facilitated.
General Anesthetics
• Glycine receptor is another target
for inhaled anesthetics.
• Inhalational agents enhance the
capacity of glycine to activate
glycine-gated chloride channels 
inhibitory neurotransmission in
spinal cord and brain stem.
General Anesthetics
• Propofol and barbiturates, but not
etomidate and ketamine, also
potentiate glycine-gated currents.
• The only general anesthetics that do
not have significant effects on
GABAA or glycine receptors are
nitrous oxide and ketamine, which
act on calcium selective NMDA
glutamate receptor.
General Anesthetics
• Neuronal nicotinic acetylcholine receptors
inhibition by inhalational agents do not
mediate anesthetic effect but mediate
analgesia and amnesia.
• Certain inhalational anesthetics may
cause membrane hyperpolarization by
activation of potassium channels.
• Inhalational agents can produce
presynaptic inhibition of neurotransmitter
release in the hippocampus contributing
to the amnesic effect of these agents.
Inhaled Anesthetics
Organ System Effects of Inhaled
Anesthetics:
A. Effects on the Cardiovascular System:
• Halothane and enflurane reduce
arterial pressure by reduction of
cardiac output.
• Isoflurane, desflurane, and
sevoflurane reduce arterial blood
pressure by decreasing systemic
vascular resistance.
Inhaled Anesthetics
• Halothane may cause bradycardia
probably because of direct vagal
stimulation.
• Desflurane and isoflurane increase heart
rate.
• All depress myocardial function, including
nitrous oxide.
• Halothane, and to a lesser effect
isoflurane sensitize the myocardium to
circulating catecholamines  ventricular
Inhaled Anesthetics
B. Effects on the Respiratory System:
• All except nitrous oxide decrease tidal
volume and increase respiratory rate
• All volatile anesthetics are respiratory
depressants and reduce the response
to increased levels of carbon dioxide.
• All volatile anesthetics increase the
resting levels of PaCO2.
Inhaled Anesthetics
• The respiratory depressant effect is
overcome by assisted or controlled
ventilation.
• Inhaled anesthetics depress mucociliary
function of airways  pooling of mucus
 atelectasis and postoperative
respiratory infection.
• Halothane and sevoflurane have
bronchodilating action (?).
• Airway irritation with desflurane.
Inhaled Anesthetics
C. Effects on the Brain:
• Decrease metabolic rate of the brain.
• Increase cerebral blood flow by
decreasing cerebrovascular resistance
(not desirable in patients with increased
intracranial pressure). Nitrous oxide is the
least likely to do so.
• If the patient is hyperventilated before the
volatile agent is administered, the
increase in ICP can be minimized (by
inducing hypocapnoeic vasoconstriction).
Inhaled Anesthetics
• Nitrous oxide has analgesic and
amnesic properties.
D. Effects on the Kidney:
• Decrease GFR and renal blood flow,
and increase the filtration fraction.
• Impair autoregulation of RBF.
E. Effects on the Liver:
• Reduce hepatic blood flow.
Inhaled Anesthetics
F. Effects on Uterine Smooth Muscle:
• Nitrous oxide has little effect.
• Halogenated anesthetics are potent
uterine muscle relaxants.
Inhaled Anesthetics
Toxicity:
1. Hepatotoxicity:
• Potentially life-threatening in subjects
previously exposed to halothane.
• Incidence is 1:20,000 – 35,000.
• Obese patients are most susceptible.
• Mechanism is unclear, but may be:
a. Direct hepatocellular damage by
reactive metabolites (free radicals).
Inhaled Anesthetics
b. Initiation of immune-mediated responses
by reactive metabolites. Serum of patients
with halothane hepatitis contain a variety
of autoantibodies against hepatic
proteins.
Trifluoroacetylated proteins in the liver
could be formed in hepatocytes during
halothane biotransformation. They are
also found in the sera of patients who did
NOT develop hepatitis after halothane
anesthesia.
Inhaled Anesthetics
2. Nephrotoxicity:
• Prolonged exposure to
methoxyflurane and enflurane leads
to formation of fluoride ions
intrarenally by the renal enzyme βlyase  changes in renal
concentrating ability (? proximal
tubular necrosis).
Inhaled Anesthetics
3. Malignant hyperthermia:
• Is an autosomal dominant genetic
disorder of skeletal muscle that occurs
in individuals undergoing general
anesthesia with volatile agents +
succinylcholine.
• It consists of rapid onset of tachycardia
and hypertension, severe muscle
rigidity, hyperthermia, hyperkalemia, and
acidosis.
• It is rare but is an important cause of
anesthetic morbidity and mortality.
Inhaled Anesthetics
• Associated with increased calcium
concentration in skeletal muscle cells
(from the sarcoplasmic reticulum).
Reduced by dantrolene.
4. Prolonged exposure to nitrous oxide
decrease methionine synthase activity
and can potentially cause
megaloblastic anemia in inadequately
ventilated operating room personnel.
Intravenous Anesthetics
• Are commonly used for induction of
general anesthesia because of more
rapid onset than inhaled agents.
• Recovery is rapid and permits their use
for short procedures.
Intravenous Anesthetics
Barbiturates
• Thiopental is the barbiturate that is
commonly used for induction of
anesthesia.
• Thiamylal is similar in pharmacokinetics
and pharmacodynamics.
• Methohexital is shorter-acting.
• Very highly lipid soluble.
• After an IV bolus injection, thiopental
rapidly crosses the blood-brain barrier,
and can produce hypnosis in one
circulation time. Blood:brain equilibrium
occurs rapidly (< 1 min).
Barbiturates
• Thiopental rapidly diffuses out of the brain
and other highly vascular tissues and is
redistributed to muscle and fat  a brief
period of unconsciousness.
• 12-16% of the dose is metabolized.
• With large doses, or a continuous infusion,
thiopental produces dose-dependent
decreases in arterial blood pressure, stroke
volume, and cardiac output. Most likely due
to myocardial depression and increased
venous capacitance.
Barbiturates
• Thiopental is also a potent respiratory
depressant  transient apnea and
lowering the sensitivity of the medullary
respiratory center to carbon dioxide.
• Cerebral metabolism and oxygen
utilization are decreased after barbiturate
administration in proportion to the degree
of cerebral depression. Cerebral blood
flow is decreased but less than oxygen
consumption.
Barbiturates
• Thiopental does not increase
intracranial pressure and volume
(unlike volatile anesthetics), and is
desirable for patients with cerebral
swelling.
• Methohexital can cause central
excitatory activity (myoclonus), but it
also has anti-seizure activity.
• Occasionally these agents precipitate
porphyric crisis during induction in
susceptible individuals.
Benzodiazepines
• Diazepam, lorazepam, and midazoloam
are used in anesthesia primarily as
premedications, because of their sedative,
anxiolytic and amnestic properties, and to
control acute agitation.
• Compared with IV barbiturates, these
drugs produce a slower onset of CNS
depression with a depth inadequate for
surgical anesthesia.
• Large doses that achieve deep sedation
prolong postanesthetic recovery period
and can produce anterograde amnesia.
Opioid Analgesics
• Highly potent agents include fentanyl,
sufentanil, and remifentanil.
• Remifentanyl is an extremely short-acting
opioid, and has been used to minimize
residual ventilatory depression.
• Awareness during anesthesia and
unpleasant postoperative recall can
occur.
• Large doses can produce chest wall and
laryngeal rigidity, thereby acutely
impairing ventilation and produce
tolerance  increasing postoperative
opioid requirements.
Opioid Analgesics
• Have been used in premedications as well
as adjunct to both IV and inhalational
anesthesia to provide perioperative
analgesia.
• The shorter-acting alfentanil and
remifentanil have been used as coinduction agents with IV sedativehypnotic anesthetics.
• Remifentanil is rapidly metabolized by
esterases in blood (not plasma
cholinesterase) and muscle tissue 
extremely rapid recovery.
Opioid Analgesics
• Can be administered in very low doses
into epidural and subarachnoid space to
produce excellent postoperative
analgesia.
• Fentanyl and droperidol (related to
haloperidol) are administered together to
produce analgesia and amnesia
(neuroleptanalgesia), and combined with
nitrous oxide to produce
neuroleptanesthesia.
Propofol
• The most popular IV anesthetic.
• Its rate of onset of action is similar to IV
barbiturates but recovery is more rapid
and patient ambulation is earlier.
• The patient subjectively feel better in
the immediate postoperative period
because of the reduction in
postoperative nausea and vomiting.
• It is the agent of choice for ambulatory
surgery.
Propofol
• It is used for both induction and
maintenance of anesthesia as part of total
intravenous or balanced anesthesia.
• It is effective in producing prolonged
sedation in patients in critical care
setting, but cumulative effect can lead to
delayed arousal.
• Prolonged administration of conventional
emulsion formulation can raise serum
lipids.
Propofol
• When used in critically ill young
children for sedation, it has caused
severe acidosis in the presence of
respiratory infection and to possible
neurologic sequelae upon
withdrawal.
• After IV administration, the
distribution half-life is 2-8 minutes
and the redistribution half life is ~ 3060 minutes.
Propofol
• It is rapidly metabolized in the liver and
excreted in urine as glucuronide and
sulfate conjugates.
• Extrahepatic mechanisms may be
involved in elimination.
• Less than 1% of the drug is excreted
unchanged in urine.
• It produces depression of central
ventilatory drive and apnea.
Propofol
• Produces a marked decrease in blood
pressure during induction of anesthesia
through arterial and veno dilation.
• It has the greatest direct negative
inotropic effect than other IV
anesthetics.
• Pain at the site of injection is the most
common adverse effect after IV bolus
administration (reduced by admixture
with lidocaine).
Propofol
• Muscle movements, hypotonus and
rarely tremors have been reported after
prolonged use.
Etomidate
• It is used for induction of anesthesia in
patients with limited cardiovascular
reserve, because it causes minimal
cardiovascular and respiratory
depression and minimal hypotension.
• It produces rapid loss of
consciousness.
• It has no analgesic effects.
• Recovery is less rapid than that of
propofol.
Etomidate
• Distribution of etomidate is rapid, with a
biphasic plasma concentration curve
showing initial and intermediate
distribution half-lives of 3 & 29 minutes,
respectively.
• Redistribution of the drug from the brain
to highly perfused tissues is responsible
for the short duration of action.
• It is extensively metabolized in the liver
and plasma and only 2% of the drug is
excreted unchanged in urine.
Etomidate
•
1.
2.
3.
4.
Adverse effects:
High incidence of pain on injection.
Myoclonic activity.
Postoperative nausea and vomiting.
Inhibition of steroidogenesis with
decreased plasma levels of cortisol
and hypoadrenalism hypotension,
electrolyte imbalance and oliguria.
Ketamine
• It produces a “dissociative anesthetic
state” characterized by catatonia
(muscular rigidity and mental stupor,
sometimes alternating with great
excitement and confusion), amnesia
and analgesia, with or without loss of
consciousness.
• It is chemically related to
phencyclidine, a psychoactive drug
with high abuse potential.
Ketamine
Mechanism of Action:
May involve blockade of the membrane
effects of the excitatory neurotransmitter
glutamic acid at the NMDA receptor
subtype.
Pharmacokinetics:
• It is highly lipid soluble and rapidly
distributed into well-perfused organs,
including brain, liver, and kidney.
• It is then redistributed to less well
perfused tissues, with hepatic metabolism
followed by hepatic and biliary excretion.
Ketamine
Pharmacodynamics:
• It is the only IV anesthetic that have
both analgesic properties and the
ability to produce dose-related
cardiovascular stimulation.
• It stimulates the central sympathetic
nervous system and, to a lesser extent,
inhibits the reuptake of norepinephrine
at sympathetic nerve terminals.
Ketamine
• It increases heart rate, cardiac output
and arterial blood pressure which reach
a peak in 2-4 minutes and decline back
to baseline over the next 10-20 minutes.
• It increases cerebral blood flow, oxygen
consumption, and intracranial
pressure. Thus, it is potentially
dangerous in patients with elevated
intracranial pressure.
Ketamine
• It decreases respiratory rate but upper
airway muscle tone is well maintained
and airway reflexes are usually
preserved.
• Its use has been associated with
postoperative disorientation, sensory
and perceptual illusions, and vivid
dreams (called emergence
phenomena).
Ketamine
• These effects can be reduced by
premedication with a
benzodiazepine (diazepam,
midazolam).
• It is specially useful in patients
undergoing painful procedures
such as burn dressing.
Dexmedetomidine
• Sedative effects of the intravenous
anesthetic dexmedetomidine are
produced via actions in the locus
ceruleus.
• It stimulates α2-adrenergic receptors at
this site and reduces central sympathetic
output, resulting in increased firing of
inhibitory neurons.
• In the dorsal horn of the spinal cord it
modulates release of substance P 
analgesic effects.