You are on page 1of 10

INTRAVENOUS ANESTHESIA AND OPIOIDS ANESTHESIOLOGY | MIDTERMS (1st Sem)

„‟ Dr. Jayber L. Tagacay

OUTLINE PHARMACOKINETICS
I. INTRODUCTION IV. OPIOID AGONISTS  Primarily metabolized by the liver and subsequently its inactive
II. INTRAVENOUS A. Morphine and water- soluble metabolites are excreted by the kidneys
ANESTHETICS B. Meperidine  Clearance rate: 20-30mL/kg/min (approx.1.5L/min)
A. Propofol C. Fentanyl
B. Etomidate D. Sufentanil
 Initial distribution half- life: 1-8 min
C. Ketamine E. Alfentanil  Secondary slow distribution half- life: 30-70 min
D. Dexmedetomidine F. Remifentanil  Elimination half- life: 2-24 hrs
E. Benzodiazepines G. Codeine  Infusion duration of up to 8hrs maintains a reliable context-
F. Barbiturates H. Hydromorphone
G. New Intravenous I. Oxymorphone sensitive half- life of 40 min or less.
Anesthetics J. Oxycodone
III. OPIOID AGONISTS AND K. Hydrocodone PHARMACODYNAMICS
ANTAGONISTS L. Methadone  Primary mechanism: GABA- A receptor agonist
A. Introduction M. Tramadol
B. Terminologies N. Heroin (Diacetyl  Alteration of the central cholinergic transmission by propofol may
C. Classification morphine) also play a role in achieving a state of unconsciousness.
D. Mechanism of Action V. OPIID AGONISTS-  Initial low doses: sedation
E. Opioid Receptors ANTAGONISTS
F. Common Opioid Side A. Pentazocine
 Increased doses: state of paradoxical excitation may occur
Effect B. Butarphanol o Disinhibition
C. Nalbuphine o Unpredictable movement
VI. OPIOID ANTAGONISTS o Broken speech
A. Naloxone
o Not readily arousable
B. Naltrexone
 Further increase: loss of consciousness, apnea, relative
relaxation of muscles, necessitates airway support
I. INTRODUCTION
General Principles: SYSTEMIC EFFECTS
 Intravenous anesthetics are now a key component of modern  CNS
anesthesia practice. o Lower sedative doses may cause changes in EEG
pattern
Properties of the Ideal Intravenous Anesthetic o Induction dose:
Pharmacodynamic/ Pharmacokinetic Properties  Initial stages of genera; anesthesia are
 Hypnosis and amnesia reached
 Rapid onset (time of one arm – brain circulation)  β-wave activity decreases, with simultaneous
 Rapid metabolism to inactive metabolites increase in α and δ activity
 Minimal cardiovascular and respiratory depression  Burst suppression: commonly employed as
 No histamine release or hypersensitivity reactions neuroprotective measure prior to aneurysm
 Non- toxic, nonmutagenic, noncarcinogenic clipping, attained at concentrations of 8ug/mL
 No untoward neurologic effects, such as seizures, o Lower cerebral metabolic oxygen consumption rate
myoclonus, analgesia, neurtoxicity (CMRO2)
 Other beneficial effects: analgesia, antiemetic, o Decreased intracranial pressure primarily by lowering
neuroprotection, cardioprotection cerebral blood flow
 Pharmacokinetic- based models to guide accurate dosing o Cerebral perfusion pressure may also be lowered
 Ability to continuously monitor delivery o Generally considered an anticonvulsant
Physiochemical Properties o Loss of consciousness
 Water- soluble
 Stable formulation, nonpyrogenic  Cardiovascular
 Non-irritating: painless on intravenous injection o Characteristic drop in systolic and diastolic blood
 Small volume needed for induction pressure without the expected increase in heart rate.
 Inexpensive to prepare and formulate o Decrease in cardiac output, stroke volume, and
 Antimicrobial preparation systemic vascular resistance (SVR).
o Decreased sympathetic activity- indirect arterial
II. INTRAVENOUS ANESTHETICS vasodilation and venodilation.
A. PROPOFOL o Suppression of supraventricular tachycardia.
 One of the most frequently used IV anesthetics on the market  Respiratory
today. o Apnea is relatively common with a higher induction
 Desirable rapid onset dose
 Predictable context- sensitive half- time o typical maintenance dose of propofol results in
 Rapid emergence from anesthesia diminished tidal volumes and increased respiratory rate.
 Wide spectrum of uses: o Blunted response to hypoxia that may be a direct effect
o Induction and maintenance of general anesthesia on chemoreceptors.
o Intensive care unit sedation o Decreased respiratory response to hypercarbia
o Sedative- hypnotic in a variety of outpatient procedures o Potent bronchodilator  direct effects on intracellular
 The lipid emulsion comes in a familiar milky white consistency Calcium homeostasis.
and can be stored at room temperature without any significant
degradation.

ARAO 1 of 10
Midterms (1st Sem) Intravenous Anesthesia and Opioids
Dr. Jayber L. Tagacay
ANESTHESIOLOGY· December 5, 2020

CLINICAL USES CLINICAL USES


 IV Sedative Hypnotic  Hemodynamically stable induction medication with relatively large
o Rapid and smooth induction and emergence from safety margin.
anesthesia  For induction of anesthesia in cardiac operating rooms
o Induction dose in a healthy adult (approx. 1-2.5mg/kg)  Used for trauma patients who are hemodynamically unstable and
o Loss of consciousness (close to 3ug/kg) are often hypovolemic.
o Elderly patients prolonged effects and increased  Reasonable choice for MAC sedation
sensitivity to propofol o Depresses airway reflexes less than Propofol (unless
o Children larger than average volume of distribution co- administered with another sedative/ analgesic
and quicker clearance agent)
o Patients with chronic alcohol abuse increased o Relaxes the smooth musculature of the pulmonary
induction dose requirement. vasculature system to a similar degree as propofol.
 Maintenance of General Anesthesia Key Pharmacology
o Infusions between 100 and 200ug/kg/min  GABA- A receptor agonist
o TIVA with propofol alone, or together with opioids as  Hemodynamically stable
part of a balanced anesthetic, has been utilized  Suppression
successfully for all types of surgery.  Postoperative nausea and vomiting
 Major Benefit Key Clinical Uses
o Prevention of post-operative nausea and vomiting  Hemodynamically stable induction
 Sedation  Cardiac, trauma, and hypovolemic patients
o Sedation for minor procedures, outpatient surgery, and
off- site anesthetics, as well as ICU sedation of SIDE EFFECTS
mechanically ventilated patients  Adrenocortical Supression
o Most significant adverse effect of Etomidate
SIDE EFFECTS o Inhibits the activity of the enzyme 11β- hydroxylase and
 Pain on injection prevents the conversion of cholesterol to cortisol.
o 60-70% of patients when administered peripherally o Pretreatment with Dexamethasone to curtail this effect
alone o Most practitioners, in an effort to limit this possibility, will
 Propofol Infusion Syndrome (PrIS) not administer repeat doses or continuous infusions.
o Extremely rare but deadly side effect of propofol  Vasoconstrictor
o Unexplained metabolic acidosis o Potent vasoconstrictor that reduces CBF, ICP, and
o Hyperkalemia CMRO2.
o Hyperlipidemia  Neurodepressant
o Rhabdomyolysis o Despite ite neurodepressant properties at high doses,
o Hepatomegaly Etomidate is often associated with epileptogenic
o Renal failure activity (excitatory spikes) on EEG.
o ECG changes, Arrythmia, Progression to Cardiac o Undesirable induction agent for patients undergoing
failure neurosurgical procedures.

B. ETOMIDATE C. KETAMINE
 Introduced into anesthetic practice as an induction agent  Emergence from Ketamine anesthesia was associated with:
 Gained much popularity because of its safe hemodynamic o Emergence delirium
profile. o Hallucinations
 Increased reports of adrenal suppression, pain on injection, o Alterations in mood and affect
thrombophlebitis, PONV, myoclonus, and hiccups.  NMDA receptor has been found to play a key role in nociception.
 The use of Etomidate is the result of a risk/benefit analysis. o Low dose Ketamine has an opiate sparing effect in the
management of acute pain.
PHARMACOKINETICS  Effects of Ketamine related to pain can be best described as
 Imidazole derivative (the D(+) enantiomer) and is not stable in antihyperalgesic, antiallodynic, or toleranceprotective.
neutral pH solutions  Has gained interest I the treatment of major depression, however
 Quick onset of action (“vein to brain”) its clinical effects are of short duration.
 Fast resolution of effect secondary to redistribution
 Metabolized in the liver and excreted predominantly by the PHARMACOKINETICS
kidneys (approx. 80%) and in bile (approx. 20%)  Analog of Phencyclidine that is a chiral compound and is a
 Largely protein bound (approx. 75%) racemic mixture of S and R enantiomers.
Initial distribution half- life 2.7 mins  Routes of administration (Bioavailability)
Redistribution half- life 29 mins o Intramuscular (93%)
Elimination half- life 2.9 – 5.3 hrs o Transnasal (25-50%)
Volume of Distribution 2.5 – 4.5L/kg o Rectal or Oral (16%)
Induction Dose 0.2 – 0.3mg/kg  High lipid solubility and low protein binding (20%)
o Allow for a rapid uptake of Ketamine in the brain, as
PHARMACODYNAMICS well as a fairly rapid redistribution.
 Binds as an agonist to the GABA-A receptors and thus has an IV onset 30 – 60 sec
inhibitory influence on the brain Duration 10 – 15 mins
 Neurodepressant properties IV induction dose 0.5 – 2 mg/kg
 Potent vasoconstrictor that reduces CBF, ICP, and CMRO2. IM induction dose 4 – 6 mg/kg
Peak plasma levels 0.75 mg/mL
CSF levels 0.2 mg/mL 1hr after dosing

ARAO 2 of 10
Midterms (1st Sem) Intravenous Anesthesia and Opioids
Dr. Jayber L. Tagacay
ANESTHESIOLOGY· December 5, 2020

 Metabolized primarily in the liver by cytochrome P450 enzymes  Cardiovascular


demethylation to its principal metabolite Norketamine. o Associated with increased heart rate and blood
 Norketamine is eliminated by renal excretion pressure.
 Due to its lipophilicity, Ketamine is only partially removed by  Causes activation of the sympathetic
dialysis. nervous system
o Increased SVR and therefore it is an attractive option
CLINICAL USES for the induction of anesthesia in patients who are
 Anesthesia hemodynamically unstable.
o Less likely to cause a seizure and may in fact have  Respiratory
CNS protective properties. o Hypoxia can occur but can easily treated with
o Associated with profound analgesia that occurs at supplemental oxygen.
subanesthetic levels. o Ketamine is also a Bronchodilator
o Associated with increase in heart rate and blood  A secondary agent to consider for the
pressure. management of severe bronchospasm or
 Good choice for anesthetic induction in th status asthmaticus.
hemodynamically unstable patient. o Caused increased salivation that may result in
o Causes dissociative amnestic sate laryngospasm.
 Unconscious with eyes open, maintain
spontaneous respiration, and do not react to D. DEXMEDETOMIDINE
painful or noxious stimuli.  α2- adrenergic agonist
o Increase in PVR associated with Ketamine may make it  introduced into clinical practice in 1999
an unsuitable choice in patients with severe right heart o ICU sedation in mechanically ventilated patients
dysfunction. o Procedural sedation
o Management of acute postoperative pain o Component of general anesthesia
o Lower pain scores and decrease opiate requirements in  Is unique as sedative
postoperative patients Key Pharmacology
o Analgesic effects are achieved at subanesthetic blood  α2- adrenergic agonist
concentrations.  Sedation with minimal respiratory depression
o Reduces opiate requirements for postsurgical pain, but  Mimics normal sleep pattern on ecg
it cannot replace opiates altogether.  Provides analgesia at the spinal cord level
 Sedation  Administration: bolus 0.5-1ug/kg over 15mins, followed by
o Burn patients during wound care 0.3-0.7ug/kg/hr infusion
 Include analgesia as well as maintenance of Key Clinical Uses
spontaneous respiration and airway reflexes.  ICU sedation in mechanically ventilated patients
o Sedation or anesthesia in uncooperative or hostile  Procedural sedation
patients  Pediatrics: preoperative anxiolysis and emergence
o Used in pediatric patients for painful procedures delirium
such as reduction and casting of bone fractures in the
emergency department. PHARMACOKINETICS
 Chronic Pain  Following intravenous administration:
o Chronic Regional Pain Syndrome (CRPS) o Distribution hal-life: 6mins
 Yields a decrease in pain scores and opiate o Terminal elimination half- life: 2hrs
consumption o Steady state volume of distributions: 118L
 These effects are only of limited duration  Linear pharmacokinetics: dose range from 0.2 to 0.7ug/kg/hr
after Ketamine administration  Average protein binding: 94%
o Cancer patients with severe pain  Almost complete biotransformation  involves both direct
o CNS side effect and lack or oral administration limit the glucoronidation and cytochrome P450- mediated metabolism
application of Ketamine in patients with chronic pain.
 Depression PHARMACODYNAMICS
o Possible treatment for Major Depression  Acts on the α2- adrenergic receptors in the spinal cord
o Decreased depression symptoms and suicidal ideation  Effects are primarily at the locus coeruleus
within 1hr of administration.  EEG pattern resembles that of non-REM sleep
o Antidepressant effects of Ketamine may not be related  Cardiovascular effects: bradycardia and hypotension
to its NMDA antagonism, but to its effect on other CNS o 7mmHg decrease in systolic blood pressure
receptors (Dopamine, adrenergic). o 1 to 8 bpm decrease in mean heart rate
o Duration of Ketamine’s antidepressant effect is short
after single administration. CLINICAL USES
 Sedation for mechanically ventilated patients in the ICU
SIDE EFFECTS  Used for procedural sedation
 CNS  Pediatrics: pre-operative anxiolysis and emergence delirium
o Psycogenic reactions (major side effect)
 Hallucinations and out-of-body experiences SIDE EFFECTS
that have been described as frightening  Main adverse reactions
 These symptoms can be reduced with co- o Hypotension, bradycardia, dry mouth, nausea, and
administration of Benzodiazepines. hypertension
o Ketamine also causes patients to have a lateral gaze  Bradycardia
nystagmus. o Typically occurs after a loading dose
o Management: atropine, ephedrine, or volume
administration

ARAO 3 of 10
Midterms (1st Sem) Intravenous Anesthesia and Opioids
Dr. Jayber L. Tagacay
ANESTHESIOLOGY· December 5, 2020

o Omitting the loading dose decreases the incidence o Manufactured as an acidic formulation that may
 Hypotension produce mild local tissue and vein irritation.
o After a 1ug/kg bolus, blood preassure decreases at  Benzodiazepines bind to specific receptor sites that are part of the
23%. GABA-A receptor complex

E. BENZODIAZEPINES MIDAZOLAM DOSING BY CLINICAL USE


MIDAZOLAM Premedication: anxiolysis, 0.02-0.04mg/kg IV/IM; 0.4-
 Anxiolysis, anterograde amnesia, sedation, and anterograde amnesia 0.8mg/kg PO
hypnosis Induction: hypnosis, amnesia 0.1-0.2mg/kg IV
 Muscle relaxants and anticonvulsants sedation
 Quick onset, short elimination half- life, anterograde Infusion (in conjuction with 0.25-1ug/kg/min
amnestic effect, and minimal side- effect profile. volatile anesthetics): hypnosis,
 Administered intravenously, intranasally, orally, amnesia
rectally, and intramuscularly  Anticonvulsants  first line therapy in the management of
 Also used as an infusion in the ICU setting seizures
Key Pharmacology
 Can also have a profound effect on the respiratory system
 GABA-A receptor agonist
 In induction doses, depress SVR and decrease BP
 Minimal respiratory depression
 Minimal cardiovascular depression
SIDE EFFECTS
 Large therapeutic window
 Reversible with Flumazenil  Anaphylaxis (extremely rare)
Key Clinical Uses  Pain or Thrombophlebitis
 Anxiolysis o More frequently described following IV injection
 Anterograde amnesia (especially Diazepam)
 Sedation o Midazolam may also cause burning with injection
 Induction of anesthesia, hemodynamically stable secondary to its acidic formulation.
 Anticonvulsant
F. BARBITURATES
PHARMACOKINETICS  2 major classes of Barbiturates:
 Highly protein bound and highly lipophilic 1. Thiobarbiturates
 Metabolism: hepatic cytochrome P450 system via oxidation and a. Thiopental
glucuronic conjugation b. Thiamylal
 Excretion: renal 2. Oxybarbiturates
a. Methohexital
 Midazolam
o Active metabolite (1- hydroxymidazolam) contributes
PHARMACOKINETICS
minimally to its clinical effects.
 Primary metabolism: hepatic; eliminated in urine and bile
Volume of distribution 1 to 3.1L/kg after a single bolus
dose  Most common form of metabolism: oxidation of Thiopental an
Elimination half- life 1.8 – 2.6 hrs Methohexital to their respective hydroxyl derivatives.
Total clearance 6.4 – 11mL/kg/min Elimination Half-life Clearance rate
Thiopental 12 hrs 3 ml/kg/min
BENZODIAZEPINE METABOLISM AND CLEARANCE Methohexital 4 hrs 11 ml/kg/min
DRUG DURATION METABOLITE HEPATIC
CLEARANCE PHARMACODYNAMICS
Midazolam Short 1-hydroxymidazolam High  Involves cortical and brainstem GABA inhibitory pathways,
(mild CNS leading to loss of consciousness, as well as respiratory and
depressant) cardiovascular depression.
Diazepam Intermediate Onazepam and Slow  Hypnotic effects are likely enhanced by the inhibition of central
Desmethyldiazepam excitatory pathways, specifically those mediated by glutamate via
Lorazepam Long Inactive Intermediate NMDA receptors and acetylcholine.
 Central Nervous System Effects
BENZODIAZEPINE PHARMACOKINETICS o Thiopental
DRUG INDUCTI DURATIO HALF PROETEI VOL. OF CLEARANCE ELIM  Generally considered an anticonvulsant
ON
DOSE
N
ACTION
OF LIFE N
BINDING
DIST HALF-
LIFE
 Used successfully for treatment of status
Midazolam 0.1- 15-20min 7- 94% 1- 6.4- 1.8- epilepticus
0.3mg/kg 15min 1.3L/kg 11mL/kg?min 2.6hr
Diazepam 0.3- 15-30min 10- 98% 0.7- 0.2- 20-
 Smaller concentrations noted to have
0.6mg/kg 15min 1.7L/kg 0.5mL/kg/min 50hr proconvulsant properties
Lorazepam 0.03- 60-120min 3- 98% 0.8- 0.8- 11-
0.1mg/kg 10min 1.3L/kg 1.8mL/kg/min 22hr o Methohexital
 Considered to have proconvulsant effects in
PHARMACODYNAMIC AND CLINICAL USES patients with epilepsy
 3 most commonly used parenteral Benzodiazepines:  Agent of choice for induction of anesthesia
o Lorazepam prior to electroconvulsive therapy.
o Diazepam o Decrease in CMRO2
o midazolam o Increase in cerebrovascular resistnce, leading to
 Lorazepam and Diazepam decreased CBF and ICP
o Not suitable in water and often cause vein irritation due o Neuroprotective properties
to the propylene glucol admixture  Anticonvulsant properties
 Improved blood flow to ischemic parts of the
 Midazolam
brain (reversal steal effect)
o Water-soluble and undergoes conformational change in
 Free- radical scavenging
the bloodsream, becoming more lipophilic.

ARAO 4 of 10
Midterms (1st Sem) Intravenous Anesthesia and Opioids
Dr. Jayber L. Tagacay
ANESTHESIOLOGY· December 5, 2020

 Attenuation of excitatory neurotransmitter Key Clinical Uses


release and pathways  Induction of general anesthesia
 Membrane stabilization  Methohexital used for sedation, premedication, and
 Cardiovascular Effects electroconvulsive therapy
o Thiopental  Barbiturate coma (thiopental)
 Decrease in both MAP and cardiac output  Thiopental intra-arterial injection can lead to tissue
 Negative inotropic effects necrois
o Methohexital
 Smaller decrease in MAP and larger increase G. NEW INTRAVENOUS ANESTHETICS
in heart rate. REMIMAZOLAM
 Respiratory System Effects  Benzodiazepine with a structure and onset time similar to
o Apnea midazolam
 After a typical induction dose, apnea is  Takes advantage of hypnotic and amnestic effects of midazolam
commonly noted after 1 to 1.5 mins, with with a speed and mode of metabolism similar to Remifentanil.
ventilator response to carbon dioxide  Binds as an agonist to the GABA receptor
returning to baseline in approximately 6 mins  Quick onset and offset
o Respiratory depression  Initially developed for procedural sedation during procedure such
 Can cause respiratory depression in a dose- as colonoscopy
dependent manner, leading to central apnea  Considered for ICU sedation
at deeper stages of anesthesia.  Alternative to Propofol by avoiding accumulation of drug leading
to prolonged sedation.
CLINICAL USES
 Thiopental FOSPROPOFOL
o Ideal induction agent  Prodrug of Propofol produced to reduce or eliminate the averse
o Typical induction dose: 2.5-5mg/kg effects of the current Propofol emulsion.
o Light stages of anesthesia in 15-30sec and deeper  Water- based solution that yields less pain on injection, less
stages in 30-40sec that last for approximately 1min. hyperlipidemia, and less risk for bacteremia.
o Rapid onset and emergence  Sedative- hypnotic approved for MAC sedation
o Patient variability:  Associated with an increase in pruritus and paresthesia.
 Unpremedicated healthy children: 5-6mg/kg
induction dose III. OPIODS AGONISTS AND ANTAGONISTS
 Infants: higher dose requirement A. INTRODUCTION
 Elderly: lower induction dose requirement  Mainstay of modern perioperative care and pain management
 Obesity: lower induction requirement on a per  Modern word “opium” is derived from the Greek word opion
kilogram basis (poppy juice).
 Comorbid states: decrease in induction dose  The term narcotics is derived from the Greek word for stupor
requirement
and traditionally has been refer to morphine-like analgesics with
 Methohexital the potential to produce physical dependence.
o Used for induction anesthesia with typical IV induction  The German pharmacist Sertuener isolated what he called the
dose of 1-2mg/kg oporific principle in opium in 1806.
o Used as Sedative and is administered rectally in
 1817 named it morphine, after the Greek god of dreams,
solution form in the pediatric population, with
Morpheus.
recommended dose of 25mg/kg
 The development of synthetic drugs with morphine-like properties
o Maintenance of sedation and general anesthesia at
has led to the use of the term opioid to refer to all exogenous
infusion doses of 50-150ug/kg/min.
substances, natural and synthetic, that bind specifically to any of
several subpopulations of opioid receptors and produce at least
SIDE EFFECTS
some agonist (morphine-like) effects.
 Discomfort on injection sites
o Can cause discomfort if injected outside the vein into B. TERMINOLOGIES
subcutaneous tissue  Opiate
 Tissue necrosis o Drugs derived from opium, including morphine, its
o Inadvertent arterial administration can lead to semisynthetic derivatives, and codeine.
decreased blood flow due to vasospasm and thrombus  Opioid
formation, pain at the arterial site, and possible tissue o All drugs, both natural and synthetic, with morphine-like
necrosis. properties, including endogenous peptides.
o Management o Opioid can be:
 Arterial administration of Papaverine  Agonist: produce the maximum possible
 Heparinization effect of binding with the receptor
 Arteriolar dilation by appropriate regional  Antagonist: produces no direct effect when it
anesthetic technique binds the receptor
Key Pharmacology  Partial Agonist: has a dose-effect ceiling
 GABA-A receptor agonist that is lower than the maximum possible
 Prolonged context- sensitive half time effect
 CNS depressant, neuroprotective, anticonvulsant,  Mixed agonist- antagonist: acts as an
decreases CMRO2, CBF, and ICP agonist (or partial agonist) at one receptor
 EEG burst suppression (thiopental)
and an antagonist at another
 Cardiovascular: decreases mean arterial pressure,
 Narcotic
venous vascular tone, and cardiac output
o Morphine and morphine-like analgesics.
 Pulmonary: dose-dependent respiratory depression, does
not cause bronchodilation

ARAO 5 of 10
Midterms (1st Sem) Intravenous Anesthesia and Opioids
Dr. Jayber L. Tagacay
ANESTHESIOLOGY· December 5, 2020

 Efficacy  k - Receptor
o Range in magnitude of an effect produced by a drug o k - Receptor–mediated analgesia may be less effective
receptor combination relative to the maximum possible for high-intensity painful stimulation than µ-opioid–
effect mediated
 Potency o Opioid agonist–antagonists often act principally on k-
o The relative dose required to achieve an effect, and is receptors
related to receptor affinity

C. CLASSIFICATION

AGONISTS AGONISTS- ANTAGONISTS


ANTAGONISTS
 Morphine  Pentazocine  Naloxone
 Morphine-6-  Butorphanol  Naltrexone
glucuronide  Nalbuphine  Nalmefene
 Meperidine  Buprenorphine
 Sufentanil  Nalorphine
 Fentanyl  Bremazocine
 Alfentanyl  Dezocine
 Remifentanil  Meptazinol
 Codeine
F. COMMON OPIOID SIDE EFFECT
 Hydromorphon
e  Cardiovascular System
 Oxymorphone o Morphine, even in large doses, given to supine and
 Oxycodone normovolemic patients is unlikely to cause direct
 Hydrocodone myocardial depression or hypotension.
 Propoxyphene o The same patients changing from a supine to a
 Methadone standing position, however, may manifest orthostatic
 Tramadol hypotension and syncope, presumably reflecting
 heroin morphine-induced impairment of compensatory
sympathetic nervous system responses
o Morphine can decreases in systemic blood pressure
D. MECHANISM OF ACTION due to drug-induced bradycardia (stimulation of the
 Act as agonists at specific opioid receptors at presynaptic and vagal nuclei in the medulla, direct depressant effect on
postsynaptic sites in the central nervous system (mainly the the sinoatrial node) or histamine release
brainstem and spinal cord) as well as in the periphery. o In contrast to morphine, the infusion of fentanyl does
 These opioid receptors normally are activated by three not cause release of histamine in any patient.
endogenous peptide opioid receptor ligands known as o During anesthesia, opioids are commonly administered
enkephalins, endorphins, and dynorphins. (Opioids mimic the with inhaled or intravenous anesthetics to ensure
actions of these endogenous ligands by binding to opioid amnesia.
receptors, resulting in activation of pain-modulating  The combination of an opioid agonist such
[antinociceptive] systems.) as morphine or fentanyl with nitrous oxide
o The principal effect of opioid receptor activation is a results in cardiovascular depression which
decrease in neurotransmission that occurs largely by does not occur when either drug is
presynaptic inhibition of neurotransmitter release administered alone.
(acetylcholine,dopamine, norepinephrine, substance P).  Decreases in systemic vascular resistance
o The intracellular biochemical events initiated by and systemic blood pressure may
occupation of opioid receptors with an opioid agonist accompany the combination of an opioid and
are characterized by increased potassium conductance a benzodiazepine, whereas these effects do
(leading to hyperpolarization), calcium channel not accompany the administration of either
inactivation, or both, which produce an immediate drug alone.
decrease in neurotransmitter release. o Opioids recognized as playing a role in protecting the
 Opioid receptors are coupled with inhibitory G-proteins and their myocardium from ischemia.
activation has a number of actions:  Ventilation
o Closing of voltage sensitive calcium channels o All opioid agonists produce dose-dependent and
o Stimulation of potassium efflux leading to gender-specific depression of ventilation, primarily
hyperpolarization and reduced cyclic adenosine through an agonist effect at mu2 receptors, leading to a
monophosphate (cAMP) production direct depressant effect on brainstem ventilation
 Overall, the effect is a reduction in neuronal cell excitability centers
leading to reduced transmission of nociceptive impulses. o Opioid-induced depression of ventilation is
characterized by decreased responsiveness of these
E. OPIOID RECEPTORS ventilation centers to carbon dioxide as reflected by an
 µ - Receptors increase in the resting Paco2 and displacement of the
o Principally responsible for supraspinal and spinal carbon dioxide response curve to the right
analgesia. o Death from an opioid overdose is almost invariably due
o Respiratory depression characteristic of µ - receptor to depression of ventilation.
activation is less prominent with k-receptor activation, o Clinically, depression of ventilation produced by opioids
although dysphoria and diuresis may accompany manifests as a decreased frequency of breathing that
activation of these receptors is often accompanied by a compensatory increase in
tidal volume

ARAO 6 of 10
Midterms (1st Sem) Intravenous Anesthesia and Opioids
Dr. Jayber L. Tagacay
ANESTHESIOLOGY· December 5, 2020

 Central Nervous System METABOLISM:


o In the absence of hypoventilation, opioids decrease  Primarily through conjugation with glucuronic acid in hepatic and
cerebral blood flow and possibly intracranial pressure extrahepatic sites, especially the kidneys
(ICP)  about 75% to 85% of a dose of morphine appears as morphine-
o Rigidity 3-glucuronide pharmacologically inactive, major metabolite
 Rapid intravenous (IV) administration of large  5% to 10% as morphine- 6-glucuronide pharmacologically
doses of an opioid (particularly fentanyl and active, causes respiratory depression and sedation
its derivatives as used in cardiac surgery) can
lead to generalized skeletal muscle rigidity ELIMINATION:
o Sedation  Small amount of unchanged opioid is excreted in the urine
 Postoperative titration of morphine frequently  Concentrations of morphine in the colostrum of parturients
induces sedation that precedes the onset of receiving patient-controlled analgesia with morphine are low and it
analgesia is unlikely transferred to the breast-fed neonate.
 The usual recommendation for morphine
titration includes a short interval between B. MEPERIDINE
boluses (5 to 7 minutes) to allow evaluation  Is a synthetic opioid agonist at µ and k - opioid receptors
of its clinical effect  Shares several structural features that are present in local
o Nausea and Vomiting anesthetics and structurally similar to atropine (possesses a mild
 Opioid-induced nausea and vomiting are atropine-like antispasmodic effect on smooth muscle)
caused by direct stimulation of the
chemoreceptor trigger zone in the floor of PHARMACOKINETICS
the fourth ventricle  About one-tenth as potent as morphine producing equivalent
 Morphine cause nausea and vomiting by sedation, euphoria, nausea, vomiting, and depression of
increasing gastrointestinal secretions and ventilation with a duration of action of 2 to 4 hours.
delaying passage of intestinal contents
toward the colon. METABOLISM
o Placental Transfer
 Is extensive, with about 90% of the drug initially undergoing
 Opioids are readily transported across the
demethylation to normeperidine and hydrolysis to meperidinic
placenta (depression of the neonate can
acid
occur)
 Normeperidine (one-half as active as an analgesic) undergoes
o Overdose
hydrolysis to normeperidinic acid
 The principal manifestation of opioid
overdose is depression of ventilation,
CLINICAL USES
manifesting as a slow breathing frequency,
 Declined greatly in recent years because of possible
which may progress to apnea (triad of
normeperidine toxicity
miosis, hypoventilation, and coma should
suggest overdose with an opioid)  Effective in suppressing postoperative shivering (anti shivering
effects may reflect stimulation of k - receptors)
 Treatment of opioid overdose is mechanical
ventilation of the patient’s lungs with oxygen SIDE EFFECTS
and administration of an opioid antagonist  Resemble those described for morphine
such as naloxone (may precipitate acute  In contrast to morphine, rarely causes bradycardia but instead
withdrawal in dependent patients) may increase heart rate, reflecting its modest atropine-like
qualities
IV. OPIOID AGONISTS
 Large doses of meperidine result in decreases in myocardial
A. MORPHINE
contractility, which, among opioids, is unique for this drug.
 Is the prototype opioid agonist to which all other opioids are
compared, producing analgesia, euphoria, sedation, nausea, and C. FENTANYL
pruritus (especially in the cutaneous areas around the nose)
 Is a phenylpiperidine-derivative synthetic opioid agonist that is
 The cause of pain persists, but even low doses of morphine structurally related to meperidine
increase the threshold to pain and modify the perception of
 As an analgesic, fentanyl is 75 to 125 times more potent than
noxious stimulation such that it is no longer experienced as pain
morphine
(continuous, dull pain is relieved by morphine more effectively
than is sharp, intermittent pain)
PHARMACOKINETICS
 In contrast to non-opioid analgesics, morphine is effective against
 Single dose of fentanyl administered IV has a more rapid onset
pain arising from the viscera
and shorter duration of action than morphine
 Analgesia is most prominent when morphine is administered
 Produces a rapid onset
before the painful stimulus occurs
 Greater potency and more rapid onset of action reflect the across
the blood–brain barrier
PHARMACOKINETICS:
 The lungs serve as a large inactive storage site, with an estimated
 Absorbed after intramuscular (IM) administration, with onset of
75% of the initial fentanyl dose undergoing first-pass pulmonary
effect in 15 to 30 minutes, a peak effect in 45 to 90 minutes and a
uptake
clinical duration about 4 hours
 Usually administered IV in the perioperative period, thus METABOLISM
eliminating the unpredictable influence of drug absorption
 Metabolized by N-demethylation
 The peak effect after IV administration of morphine is delayed
 Pharmacologic activity of fentanyl metabolites is believed to be
compared with opioids such as fentanyl and alfentanil, requiring
minimal.
about 15 to 30 minutes

ARAO 7 of 10
Midterms (1st Sem) Intravenous Anesthesia and Opioids
Dr. Jayber L. Tagacay
ANESTHESIOLOGY· December 5, 2020

ELIMINATION HALF TIME CLINICAL USES


 Has a short duration of action, but its elimination half-time is  A single dose of sufentanil, 0.1 to 0.4 g/kg IV, produces a longer
longer than that for morphine period of analgesia and less depression of ventilation than does a
 Longer elimination half-time reflects a larger volume of distribution comparable dose of fentanyl (1 to 4 g/kg IV)
(Vd) of fentanyl due to its greater lipid solubility and thus more  Dose of 18.9 g/kg IV results in more rapid induction of anesthesia,
rapid passage into highly vascular tissues compared with the less earlier emergence from anesthesia, and earlier tracheal
lipid-soluble morphine extubation
 The plasma concentrations of fentanyl are maintained by slow  Produces bradycardia that may be sufficient to decrease cardiac
reuptake from inactive tissue sites, which accounts for persistent output
drug effects that parallel the prolonged elimination half-time
 Prolonged elimination half-time for fentanyl in elderly patients is E. ALFENTANIL
due to decreased clearance of the opioid because Vd is not  Is an analogue of fentanyl that is less potent (one-fifth to one-
changed in comparison with younger adults tenth) and has one-third the duration of action of fentanyl
 Unique advantage compared with fentanyl and sufentanil is the
CLINICAL USES more rapid onset of action (rapid effect-site equilibration) after the
 Analgesia (1-2ug/kg IV) IV administration of alfentanil.
 Adjuvant to inhaled anesthetics to blunt the response to direct
laryngoscopy or sudden changes in the level of surgical PHARMACOKINETICS
stimulation (2-20ug/kg IV); consider effect- site equilibration time  Has a short elimination half-time compared with fentanyl and
 Decrease doses of inhaled anesthetics needed to blunt sufentanil
 Despite its lesser lipid solubility, penetration of the blood–brain
sympathetic nervous system responses to surgical stimulation
barrier by alfentanil is rapid because of its large nonionized
(1.5-3ug/kg IV 5min before induction of anesthesia)
fraction at physiologic Ph
 Produce surgical anesthesia (50-150ug/kg IV)
 Analgesia for early labor (25ug intrathecal) METABOLISM
 Preoperative medication (transmucosal)  Hepatic clearance of about 96% from the plasma within 60
 Postoperative analgesia (transdermal patch) minutes of its administration
 Alfentanil clearance is markedly influenced by CYP3A activity
SIDE EFFECTS
 Cardiovascular Effects CLINICAL USES
o Even in large doses (50 g/kg IV), does not evoke the  Has a rapid onset and off set of intense analgesia, reflecting its
release of histamine very prompt effect-site equilibration.
o Bradycardia is more prominent with fentanyl than  Used to provide acute but transient analgesia
morphine and may lead to occasional decreases in  Associated with a lower incidence of postoperative nausea and
vomiting in outpatients compared with fentanyl and sufentanil
blood pressure and cardiac output.
 Seizure Activity F. REMIFENTANIL
o It is difficult to distinguish opioid-induced skeletal  Is a selective opioid agonist with an analgesic potency similar to
muscle rigidity or myoclonus from seizure activity that of fentanyl (15 to 20 times as potent as alfentanil) and a
 Intracranial Pressure blood–brain equilibration time similar to that of alfentanil
o Associated with increase (6 to 9 mm Hg) in ICP despite  Short-acting phenylpiperidine derivatives, structurally unique
maintenance of an unchanged Paco2 together with because of its ester linkage structure that is susceptible to
alfentanil hydrolysis by nonspecific plasma and tissue esterases to inactive
metabolites
DRUG INTERACTIONS  The unique pathway of metabolism leads to (a) brief action, (b)
 Fentanyl greatly potentiate the effects of benzodiazepines precise and rapidly titratable effect due to its rapid onset and off
(synergism with respect to hypnosis and depression of ventilation) set, (c) lack of accumulation, and (d) rapid recovery after
discontinuation of its administration.
 The advantage of synergy between opioids and benzodiazepines
for the maintenance of patient comfort is carefully weighed PHARMACOKINETICS
against the disadvantages of the potentially adverse depressant  Characterized by small Vd, rapid clearance, and low
effects of this combination. interindividual variability compared to other IV anesthetic drugs
 Because of its rapid systemic clearance, remifentanil provides
D. SUFENTANIL pharmacokinetic advantages in clinical situations requiring
 Is a thienyl analogue of fentanyl with an analgesic potency of predictable termination of drug effect
sufentanil that is 5 to 10 times that of fentanyl.  Combination of rapid clearance and small Vd produces a drug
with a uniquely transient effect
PHARMACOKINETICS
 Elimination half-time of sufentanil is intermediate between that of METABOLISM
fentanyl and alfentanil  Unique among the opioids in undergoing metabolism by
nonspecific plasma and tissue esterases to inactive metabolites
 Permits rapid penetration of the blood–brain barrier and onset of
 Clearance not affected by cholinesterase deficiency or
CNS effects anticholinergic
 Redistribution to inactive tissue sites terminates the effect of small  Pharmacokinetics will be unchanged by renal or hepatic failure
doses, but a cumulative drug effect can accompany large or because esterase metabolism is usually preserved
repeated doses of sufentanil
CLINICAL USES
METABOLISM  Unique pharmacokinetics which allows rapid onset of drug effect,
 Rapidly metabolized by N-dealkylation and the products are precise titration to the desired effect, the ability to maintain a
pharmacologically inactive, whereas desmethyl sufentanil has sufficient effect-site concentration to suppress the stress
about 10% of the activity of sufentanil response, and rapid recovery from the drug’s effect
 Remifentanil, 0.05 to 0.10 g/kg/min in combination with
midazolam, 2 mg IV, provides effective sedation and analgesia

ARAO 8 of 10
Midterms (1st Sem) Intravenous Anesthesia and Opioids
Dr. Jayber L. Tagacay
ANESTHESIOLOGY· December 5, 2020

during monitored anesthesia care in otherwise healthy adult  Compared with morphine, parenteral heroin has a
patients a. more rapid onset
b. less opioid-induced nausea
SIDE EFFECTS c. greater potential for physical dependency
 The short recovery period may be considered a disadvantage if
the infusion is stopped suddenly V. OPIOID AGONIST- ANTAGONISTS
 Nausea and vomiting, depression of ventilation, and mild  These drugs bind to receptors, where they produce limited
decreases in systemic blood pressure and heart rate responses (partial agonists) or no effect (competitive antagonists)
 Hyperalgesia  The side effects are similar to those of opioid agonists, and, in
o Patients receiving large doses of remifentanil addition, these drugs may cause dysphoric reactions
intraoperatively are often surprisingly high, suggesting  The advantages of opioid agonist–antagonists are the ability to
remifentanil may be associated with acute opioid produce analgesia with limited depression of ventilation and
tolerance a low potential to produce physical dependence
 In general, agonist–antagonist drugs should be reserved for
G. CODEINE patients who are unable to tolerate a pure agonist.
 Ss the result of the substitution of a methyl group for the hydroxyl
group on the number 3 carbon of morphine A. PENTAZOCINE
 The presence of this methyl group limits first-pass hepatic  Possesses opioid agonist actions (antagonized by naloxone) as
metabolism and accounts for the efficacy of codeine when well as weak antagonist actions (sufficient to precipitate
administered orally withdrawal symptoms when administered to patients who have
 About 10% of administered codeine is demethylated in the liver been receiving opioids on a regular basis).
to morphine, which may be responsible for the analgesic effect
of codeine, although codeine-6-glucuronide may also exert an PHARMACOKINETICS
analgesic effect.  Well absorbed after oral or parenteral administration
 Effective at suppressing cough at oral doses  First-pass hepatic metabolism is extensive, with only about 20%
 Maximal analgesia, equivalent to that produced by 650 mg of of an oral dose entering the circulation.
aspirin, occurs with 60 mg of codeine
 When administered IM, 120 mg of codeine is equivalent in CLINICAL USES
analgesic effect to 10 mg of morphine  10 to 30 mg IV or 50 mg orally, is used for the relief of moderate
 Included in medications as an antitussive or is combined with pain.
nonopioid analgesics for the treatment of mild to moderate pain.  An oral dose of 50 mg is equivalent in analgesic potency to 60 mg
of codeine
H. HYDROMORPHONE  Useful for treatment of chronic pain when there is a high risk of
 Derivative of morphine that is about five times as potent as physical dependence
morphine but has a slightly shorter duration of action
 Hydromorphone is an effective alternative to morphine in the SIDE EFFECTS
treatment of opioid-responsive moderate to severe pain  The most common side effect of pentazocine is sedation
 Nausea and vomiting are less common than with morphine
I. OXYMORPHONE  Dysphoria, including fear of impending death, is associated with
 About 10 times as potent as morphine and seems to cause more high doses.
nausea and vomiting  Pentazocine, 20 to 30 mg IM, produces analgesia, sedation, and
 Has a great potential for physical dependence depression of ventilation similar to 10 mg of morphine.

J. OXYCODONE B. BUTARPHANOL
 Commonly used orally for treating acute pain (about twice as  Compared with pentazocine, butorphanol’s agonist effects are
potent as oral morphine and has a similar duration of analgesic about 20 times greater, whereas its antagonist actions are 10 to
action) 30 times greater
 Abuse potential is but new, abuse-resistant formulations that are  Rapidly and almost completely absorbed after IM injection
not easily solubilized for IV injection are now widely marketed  In postoperative patients, 2 to 3 mg IM produces analgesia and
depression of ventilation similar to 10 mg of morphine.
K. HYDROCODONE
 A commonly used oral opioid for treating acute pain and is similar SIDE EFFECTS
in potency to oral morphine and has a similar duration of  Common side effects include sedation, nausea, and diaphoresis
analgesic action (high abuse potential)  Dysphoria is infrequent

L. METHADONE C. NALBUPHINE
 Synthetic opioid agonist that produces analgesia in the setting of  Phenanthrene opioid derivative
chronic pain syndromes and is highly effective by the oral route  The most common side effect was drowsiness
 The efficient oral absorption, prompt onset of action, and  Used to antagonize the ventilatory depressant effects of µ opiod
prolonged duration of action render this an attractive drug for receptor agonist while still providing analgesia by K receptor
suppression of withdrawal symptoms in physically dependent stimulation.
persons such as heroin addicts
VI. OPIOID ANTAGONISTS
M. TRAMADOL  Minor changes in the structure of an opioid agonist can convert
 A centrally acting analgesic that is 5 to 10 times less potent than the drug into an opioid antagonist at one or more of the opioid
morphine in volunteers receptor sites
 Naloxone antagonized only an estimated 30% of the effect of  Naloxone, naltrexone, and nalmefene are pure opioid receptor
tramadol. antagonists with no agonist activity
N. HEROIN (DIACETYLMORPHINE) A. NALOXONE
 A synthetic opioid produced by acetylation of morphine  Nonselective antagonist at all three opioid
 There is rapid penetration of heroin into the brain,where it is  1 to 4 g/kg IV reverses opioid-induced analgesia and depression
hydrolyzed to the active metabolites monoacetylmorphine and of ventilation
morphine

ARAO 9 of 10
Midterms (1st Sem) Intravenous Anesthesia and Opioids
Dr. Jayber L. Tagacay
ANESTHESIOLOGY· December 5, 2020

 Duration of action (30 to 45 minutes) is presumed to be due to its B. NALTREXONE


rapid removal from the brain  In contrast to naloxone, is highly effective orally, producing
sustained antagonism of the effects of opioid agonists for as long
SIDE EFFECTS as 24 hours.
 Depression of ventilation accompanied by an inevitable reversal
of analgesia
 Titrate the dose of naloxone such that depression of ventilation is References
partially but acceptably antagonized to also maintain partial  Dr. Tagacay‟s ppt
analgesia
 Nausea and vomiting related to the dose and speed of injection
 Cardiovascular stimulation increased sympathetic nervous system
activity manifest as tachycardia, hypertension, pulmonary
edema, and cardiac dysrhythmia.

ARAO 10 of 10

You might also like