Pharmacology Flashcards
Give an overview of the pharmacology of drugs as organic molecules and relevance to anaesthesia.
Recognize the structure and nomenclature of common organic groups
Explain structure-activity relationships in anaesthetic practice
Organic molecules may be aliphatic or aromatic.
Aliphatic molecules consist of a root, plus various functional groups and side chains
Presence and position of functional groups determines the physical, chemical and pharmacological properties of a drug
The structure of organic molecules determines solubility, protein binding, potency and stability
Minor modifications to the chemical structure of a drug can have dramatic effects on its clinical activity
Give an overview of the aliphatic compounds and functional groups relevant to anaesthesia.
Each group of atoms that becomes attached to the carbon chain has its own characteristic set of reactions. The functional groups are commonly found in anaesthetic practice.
What is the name of this molecule?
2, 6-di-isopropyl phenol
Propofol
The number of bonds an atom has, in its uncharged state, is the valency. For example, hydrogen has a valency of 1 (Fig 1). Looking at the other atoms in Fig 1, in each case, what is the valancy? 1, 2, 3, or 4
A. Bromine
B. Carbon
C. Chlorine
D. Fluorine
E. Nitrogen
F. Oxygen
Is this molecule an acid or base?
Carboxylic acid is an acid, as it donates the ‘H’ from the COOH group.
Is this molecule an acid or base?
Amine is a base. The lone pair of electrons on the nitrogen can form a co-ordinate bond with a free hydrogen to form a positively charged ammonium group.
Is this molecule an acid or base?
Phenols are weak acids and can give up a hydrogen.
Which functional groups are present in Tetracaine?
Possible answers:
A. Amide
B. Amine
C. Aromatic group
D. Ester
E. Halide
F. Alcohol
G. Acid
A. Incorrect.
B. Correct.
C. Correct.
D. Correct.
E. Incorrect
F. Incorrect.
G. Incorrect.
Which functional groups are present in Lidocaine?
Possible answers:
A. Amide
B. Amine
C. Aromatic Group
D. Ester
E. Halide
F. Alcohol
G. Acid
A. Correct.
B. Correct.
C. Correct.
D. Incorrect.
E. Incorrect.
F. Incorrect.
G. Incorrect.
Select the drug that has greater potency.
Bupivicaine
Increasing the bulk of the side chain on the amine (a butyl group rather than a methyl group), increases the molecules lipid solubility and hence potency
Correctly identify the three molecules shown below?
A) 2-methylbutane
B) 2, 3-dimethylbutene
C) 3-chloro-2, 2-dimethylpropanol
Give an overview of the interaction between molecules and the role that it has on clinical anaesthesia.
Describe the forces that hold molecules together
Explain the concept of polarity
Explain the relevance of bonding to molecular properties
The physical and chemical properties of a drug are determined by the molecular bonds
Intramolecular bonds may be:
ionic due to transfer of electrons (water soluble)
covalent due to shared electrons (lipid soluble)
Intramolecular forces such as hydrogen bonds or Van der Waals’ forces also affect solubility and protein binding
Covalent bonds may be pure or polar
With regard to the properties of covalent compounds…
A. They have high melting and boiling points
B. They are soluble in water
C. They conduct electricity
D. Their bonds consist of shared electrons
A. False. The forces between the molecules are weak requiring little energy to break them. At room temperature they are likely to be gases or volatile liquids.
B. False. They usually dissolve in non-polar or organic liquids.
C. False.
D. True. In covalent bonding each atom donates an electron to a shared electron pair.
Is the oxygen atom in this molecule a negative or positive pole?
Possible answers:
A. Negative pole
B. Positive pole
A. Correct.
B. Incorrect.
Oxygen atoms have a higher electro-negativity than carbon or hydrogen, hence are the more negative pole of the molecules.
Place each type of bond in order of decreasing strength of attraction, starting with the strongest bond.
Give an overview of ionization and the role it plays on clinical anaesthesia.
Identify anaesthetic drugs that are weak acids and weak bases
Discuss the relationship between the pKa of a drug and the degree of ionization
Derive the Henderson-Hasselbalch equation for the bicarbonate-carbonic acid buffer pair
Describe the importance of pKa in determining pharmacokinetic behaviour of drugs
Many anaesthetic agents are weak acids and weak bases
Weak acids are ionized above their pKa and weak bases are ionized below their pKa
The pharmacokinetic behaviour of a drug is influenced by its tendency to ionize at plasma pH
Buffering is an important means of minimising pH changes both intracellularly and in the plasma
A buffer pair works most efficiently at its pKa
The bicarbonate-carbonic acid buffer system is important because it is open-ended: carbon dioxide can be excreted through the lungs
Many of the drugs we use are organic molecules with added functional groups that make them weak electrolytes (Fig 1a).
Question: Which two functional groups are most important in allowing small organic molecules to act as weak electrolytes?
The -COOH (carboxyl) and -NH2 (amine) groups (Fig 1b). Other groups may also allow dissociation, but not always at a pH relevant to human physiology.
Can you identify two groups that often dissociate outside the pH range 6.5-8.5?
A phenolic hydroxyl group, i.e. with an aromatic ring -OH, and an aldehyde group, i.e. R-CH = O; often called a keto-group (Fig 2).
Carboxyl (-COOH) and amine (-NH2) functional groups are of particular importance in allowing partial dissociation of small organic molecules in an aqueous environment.
Question: Can you identify the two commonly used analgesic drugs shown in Fig 1a?
A is aspirin and B is morphine (Fig 1b).
Which is a weak acid and which is a weak base? Related to this, can you identify the functional groups responsible?
Aspirin is a weak acid and has a carboxyl group; morphine is a weak base and has an amine group (Fig 1c).
When a weak acid, such as aspirin, dissociates it gives up a proton, i.e. a hydrogen ion, to its aqueous environment. In the case of aspirin, i.e. acetylsalicylic acid, the carboxyl group dissociates:
R - COOH ⇔ R - COO- + H+
The -COOH group is a proton donor. -COO- is the proton acceptor (Fig 1a).
Question: What two factors determine the degree of dissociation?
The pH of the environment and the dissociation constant for the above reaction (Fig 1b).
What is pKa?
pKa is the pH at which the concentration of the proton donor form is equal to that of the proton acceptor form.
Look at the drug structure. What is the single best explanation for why this is a weak acid?
A. It is an induction agent
B. It has two isopropyl groups
C. It is an aromatic molecule
D. It has a phenolic hydroxyl group
E. There are no amine groups
A. Incorrect.
B. Incorrect.
C. Incorrect.
D. Correct. This drug is propofol. The phenolic hydroxyl group can dissociate, but only at a high pH.
E. Incorrect.
The pH of a 2.5% solution of sodium thiopental is 11. What is the single best explanation of why this is necessary?
A. Thiopental is a weak acid
B. Sodium thiopental can only form at this pH
C. The pH is high enough to prevent precipitation of thiopentoic acid
D. Isomerism of thiopental to the thiol form occurs at this pH
E. Thiopental is highly lipid-soluble except at high pH
A. Incorrect.
B. Incorrect.
C. Correct. Thiopental acid has a pKa of 7.6 in water but the unionized form is insoluble, and so would precipitate out at a pH near 7.6. At pH 11 the ratio of ionized to unionized form is about 5000:1 so acid precipitation does not occur.
D. Incorrect.
E. Incorrect.
Which of these anaesthetic drugs are weak acids?
A. Aspirin
B. Etomidate
C. Fentanyl
D. Propofol
E. Thiopental
A. True.
B. False. Etomidate is a weak base.
C. False. Fentanyl is a weak base.
D. True.
E. True.
We have previously seen that:
[H+] = Ka([proton donor]/[proton acceptor])
This equation can be transformed to help us predict the relative proportion of a drug in its ionized form.
Question: What does the equation look like if we take logarithms of both sides and make both sides negative?
-log[H+] = -log(Ka) - log ([proton donor]/[proton acceptor])
or, more familiarly:
-log[H+] = -log(Ka) + log ([proton acceptor]/[proton donor])
-log[H+] = pH
If we now substitute this into our equation and allow the concentration of proton donor and proton acceptor forms to be equal, we get: pH = -log(Ka). The negative logarithm of the acid dissociation constant is known as the pKa: it is the pH at which the concentration of proton donor equals that of the proton acceptor.
The equation we have just derived can be written:
pH = pKa + log ([proton acceptor]/[proton donor])
What is the weak organic acid compound shown in Fig 1?
Carbonic acid.
It is a proton donor.
The proton acceptor form is Bicarbonate: HCO3- (Fig 2).
The Henderson-Hasselbalch equation describes how the pH of the environment influences the equilibrium between carbonic acid and bicarbonate.
We can calculate how the ratio of unionized (carbonic acid) to ionized (bicarbonate) form varies within a range of pH values around plasma pH using the Henderson-Hasselbalch equation (Fig 1).
Question: If we know that the pKa of a system is 6.1, what is the proportion of ionized to unionized form at each of the following pH values: 6.1, 7.1 and 8.1?
You should have calculated the following ratios: 1, 10 and 100.
The carbonic acid-bicarbonate system is just one of the buffer systems in the body. Proteins contain amino acids, some of which have functional groups that can ionize at body pH. Proteins are important intracellular buffers. This is particularly true of haemoglobin within the red blood cell.
Question: Can you name an intracellular buffer system that is not a protein?
The phosphate buffer system.
HPO42- + H+ ⇔ H2PO4-
The pKa of this system, 6.8, is closer to intracellular pH than the bicarbonate system is to plasma pH. Since intracellular pH is lower than plasma pH this buffer system is quite efficient.
The kidney is involved in the excretion of hydrogen ions and retention of bicarbonate in the carbonic acid-bicarbonate buffer system (Fig 1). We produce 30-40 mmol H+ daily and in order to eliminate this amount of acid the renal tubules need the assistance of urinary buffers.
Question: Which weak base is an essential part of urine buffering when excess hydrogen ions need eliminating?
Ammonia: ammonia-ammonium ion is a buffer pair, with ammonia production occurring optimally at low pH: NH3 + H+ ⇔ NH4+
In addition to bicarbonate and ammonia, which other buffer system is also important in renal excretion of hydrogen ions?
The phosphate buffer system (Fig 2). Ammonia and phosphate buffers are produced in the renal tubular cells. Excretion of ammonium ions and dihydrogen phosphate allow net loss of acid from the blood.
Morphine has a pKa of 7.9. Regarding the ionization of morphine:
A. At pH 7.4 the ratio of the ionized to the unionized form of morphine is approximately 30:1
B. At physiological pH morphine is more ionized than fentanyl
C. Morphine has a tertiary amine group
D. Morphine is a weak base that is 50% ionized at pH 7.9
E. In a patient who is severely acidaemic, more morphine exists in the unionized form than is found in a normal person
A. False. The ratio is approximately 3:1.
B. False. Fentanyl has a pKa of 8.4 so is more ionized than morphine at pH 7.4.
C. True.
D. True.
E. False. If plasma pH drops below normal, as is seen in acidaemia, then a greater proportion of morphine exists in the ionized form.
A drug needs to reach its site of action within the body. For many anaesthetic drugs this involves injection of a liquid containing the drug followed by transport within the blood to the brain.
Question: Which two chemical properties of a drug determine how rapidly an anaesthetic agent reaches the brain?
Lipid solubility and pKa.
Lipid solubility is important for crossing the blood-brain barrier but pKa determines what proportion of the unbound drug is in the unionized form.
The unionized form passes the blood-brain barrier (BBB) more rapidly.
The ionized form of a drug is more water-soluble than the non-ionized form, so ampoule contents are at a pH that favours that form, although high and low pH solutions can be very irritant to veins.
If the unionized form of the drug is not water-soluble, then the drug cannot be prepared as an aqueous solution and requires other means to solubilize it. Induction agents need to reach the brain quickly and so need to be very lipid-soluble, but for rapid onset they need to be given IV.
Question: What methods are used to produce an intravenous preparation of induction agents?
Propofol is prepared as an emulsion in intralipid; egg phosphatide is used as the emulsifying agent
Etomidate is solubilized with polyethylene glycol or intralipid
Sodium thiopental is produced as a powder under nitrogen
Indirect access to the bloodstream requires absorption through a body barrier. The non-ionized form of the drug is more lipid-soluble than the ionized form. Paracetamol, aspirin, ibuprofen and morphine liquid are all drugs that can both ionize and be given orally.
Question: What is the pH of the stomach and the small intestine?
In the stomach, pH is low at around 2-3; in the small intestine pH is high, closer to 8-9.
Can you order the drugs mentioned above (Paracetamol, aspirin, ibuprofen and morphine liquid) according to the proportion of unionized form, from the smallest to largest proportion, in (a) the stomach and (b) the small intestine?
Stomach: morphine, aspirin, ibuprofen, paracetamol. Small intestine: aspirin, ibuprofen, morphine, paracetamol.
Aspirin, ibuprofen and paracetamol are all weak acids and so are ionized above their respective pKa (3.5, 4.9, 9.4). Morphine is a weak base, and so is ionized below its pKa (7.9).
At pH 3 all the acids are unionized: the higher their pKa, the higher the unionized fraction. At this pH morphine is largely ionized, and so has the lowest proportion in the unionized form. The order in the stomach is therefore: morphine, aspirin, ibuprofen, paracetamol.
At pH 8 morphine is very slightly more unionized than ionized, but aspirin and ibuprofen are both mainly ionized, aspirin more so than ibuprofen. However, paracetamol is still largely unionized. The order in the small intestine is therefore: aspirin, ibuprofen, morphine, paracetamol.
Many anaesthetic drugs need rapid access to the CNS for their activity. Three important factors govern the speed of passive movement of a drug across the BBB:
Lipid solubility: Highly lipid-soluble drugs enter the CNS more rapidly than poorly lipid-soluble ones
Degree of protein binding: A high proportion of unbound drug creates a favourable concentration gradient for movement into the CNS
pKa: The unionized form favours passage across the BBB
Therefore drugs with high lipid solubility, low protein-binding and a low proportion in the ionized form have a more rapid onset of CNS activity. Morphine has a pKa of 7.9 but alfentanil has a pKa of 6.4. Both are weak bases and so are unionized at a pH above their pKa. Morphine is about 40% protein bound and alfentanil is about 90%.
Question: Which has the faster onset of action?
Alfentanil. Although it has a lower proportion of free drug than morphine, this difference is much smaller than the difference between the proportion of unionized drug in the bloodstream. Alfentanil is 100 times less ionized than morphine.
The pKa of a drug may also influence its duration of action, particularly when given by continuous infusion. Propofol and fentanyl are commonly given by continuous infusion. Both are very lipid-soluble but fentanyl has a pKa of 8.5, which is very much closer to physiological pH than the pKa of propofol. Propofol is effectively unionized in the body.
Question: When given by continuous infusion for several hours, how does the offset time compare for the two drugs, assuming they are given at therapeutic doses?
Propofol has a much more rapid offset time than fentanyl. Part of the reason is because of the difference in ionization.
Fentanyl is a weak base and is ionized below its pKa, and so is largely ionized at physiological pH, i.e. 10:1 in favour. This form is water-soluble and redistributes from lipophilic stores to maintain an effective plasma concentration. Propofol, on the other hand, is not ionized: once in a favoured lipid environment it does not rapidly re-enter the plasma, and so its effects wear off more quickly.
Regarding the buffer system:
H2CO3 ⇔ HCO3- + H+
Select true or false for each option, then select Submit.
A. This is the only buffer system present in red blood cells
B. Carbonic acid is the weak acid and bicarbonate the conjugate base
C. The pKa of this system is less than 0.5 pH units from normal plasma pH
D. This is the most abundant extracellular buffer system
E. Carbonic anhydrase in the plasma is essential for this system to operate
A. False. Haemoglobin is present in red blood cells and can also act as a buffer.
B. True.
C. False. The pKa of this system is around 6.4 compared with a normal plasma pH of 7.4.
D. True.
E. False. Carbonic anhydrase is present in the red blood cell not the plasma.
These drugs are weak acids:
A. Remifentanil
B. Naproxen
C. Meperidine
D. Diclofenac
E. Meloxicam
A. False. Opioids are weak bases.
B. True. NSAIDs are all weak acids.
C. False. Opioids are weak bases.
D. True.
E. True.
These drugs are 50% or more ionized at physiological pH:
A. Paracetamol
B. Etomidate
C. Remifentanil
D. Ibuprofen
E. Ketamine
A. False. Paracetamol is a weak acid with a pKa of 9.4, so is not ionized at pH 7.4.
B. False. Etomidate is a weak base with a pKa of 4.2, so is not ionized at pH 7.4.
C. False. Remifentanil is a weak base with a pKa of 7.1.
D. True.
E. True.
Remember that bases are ionized below the pKa.
Acids are ionized above their pKa.
Give an overview of isomers, and their relevance to anaesthesia.
Define the categories of isomers
Describe the properties of different isomers
Explain their relevance in anaesthesia
Isomers are molecules which have the same molecular formula but different arrangements of their atoms
Structural isomers may have very different chemical structures, resulting in drugs with different physical, chemical and pharmacological properties
Stereoisomers have the same chemical structure but a different spatial configuration. They have the same physical and chemical properties but rotate the plane of polarised light in opposite directions
Isomers are relevant to medical practice as their different actions may produce therapeutic results or unwanted side-effects
What is an isomer?
An isomer is a chemical compound with the same molecular formula as another compound but a different arrangement of atoms.
Structural isomers have the following properties (Fig 1):
The same molecular formula, i.e. the same numbers of hydrogen, carbon, oxygen, nitrogen atoms
Different chemical structures (different arrangements of the atoms)
They may have similar or completely different pharmacological properties
Question: Which common anaesthetic drugs are structural isomers?
Isoflurane and enflurane have the same molecular formulae but different chemical structures (Fig 2). Their structures are similar as they are both halogenated ethers and they both have similar physical, chemical and pharmacological properties.
What is the name of the compound shown in Fig 1?
Sodium thiopental.
This is a form of structural isomerisation called tautomerisation. Tautomerisation can be called dynamic structural isomerisation. It occurs when two structural isomers exist in equilibrium with each other.
Sodium thiopental is prepared buffered to a pH of 10.5: this allows ionisation of the thiol (-SH group) to form the sodium salt which is readily water soluble. However, at the more acidic physiological pH of 7.4 the sulphide anion attracts hydrogen ions to form the unionised thiol molecule (Fig 2). This structure rapidly undergoes tautomerisation (a hydrogen ion is transferred from the sulphur atom to the nitrogen atom). The resulting thione (-C=S) (Fig 3) is very lipid soluble and rapidly crosses the blood-brain barrier.
The structure of midazolam is also modified by changes in acidity (Fig 1).
It contains a primary amine group which at acid pH is ionized and water soluble. Once injected into plasma, pH-dependant ring closure occurs to form a benzodiazepine ring. The resulting molecule is lipid soluble and readily crosses the blood-brain barrier.
Question: Why is this not true isomerisation?
A molecule of water (H2O) is eliminated when the benzodiazepine ring is formed, the two forms consequently do not contain the same number of atoms and hence are not in fact isomers, however in many texts it is given as an example of tautomerisation.
Geometric isomers occur in compounds which have a carbon-carbon double bond.
Question: What is a –C=C- functional group called (Fig 1)?
A compound with carbon-carbon double bond is an alkene.
Whilst there is free rotation around a carbon-carbon single bond, there is no rotation about a double bond. This may result in two geometric isomers:
In the cis isomer (Fig 2), functional groups exist on the same side of the double bond
In the trans isomer (Fig 3), they are on opposite sides of the double bond
What is the relevance of isomerisation in anaesthetics?
Isomerisation is relevant in medicine because one isomer will often produce the desired effect in the patient whereas the other isomer may have no effect or even produce unwanted side-effects (Table 1).
Two enantiomers will have very different conformational relationships with a chiral receptor, which may result in different:
Potencies
Intrinsic activities, i.e. one isomer may be an agonist at a particular receptor while the other is an antagonist
Pharmacological responses, e.g. levorphanol is an opioid analgesic whereas the enantiomer dextromorphan is a cough suppressant
Understanding of isomerization has resulted in drugs now being produced as a single isomer to optimise the desired actions and minimise unwanted effects.
- Muscle relaxants
Aminosteroids: vecuronium and rocuronium contain many chiral centres but are synthesised as single isomers.
Isoquinolones: atracurium and mivacurium each have 4 chiral centres and 16 possible stereoisomers.
Atracurium is produced as a mixture of 10 isomers each with different pharmacokinetics and potencies.
One isomer, cisatracurium, has various clinical advantages, including:
Three times increase in potency
Minimal autonomic effects
Minimal histamine release
Reduced laudanosine levels
It is produced as a single enantiomer and available commercially
- Local anaesthetics
Mepivacaine, prilocaine, bupivacaine and ropivacaine all exist as pairs of optical isomers.
The S enantiomer is the most pharmacologically useful due to the following advantages:
Increased vasoconstriction resulting in prolonged duration of action and less systemic absorption
Reduced cardiotoxicity
Reduced motor blockade
As a result ropivacaine and bupivacaine are now produced commercially as single enantiomers.
Are the β-blockers practolol and atenolol, examples of structural isomerization or stereoisomer isomerization? Select the correct box.
Structural isomerisation.
Atenolol and practolol are structural isomers of each other. At the left-hand end of the molecule as drawn, the amine and alkyl groups have switched position.
However, both can also exist as two stereoisomers, as the carbon marked * has four different groups attached, and hence is chiral.
Select the type of isomerisation that is found in halothane.
Stereoisomer isomerisation
Stereoisomerization is found in halothane. Halothane can exist as two non-superimposable mirror images due to the presence of a chiral carbon.
Correctly label the two sets of structures as either cis isomers or trans isomers.
A. CIS-but-2-ene. CIS-butenedioic acid.
B. TRANS-but-2-ene. TRANS-butenedioic acid.
Select the volatile agent that does not exist as stereoisomers.
Sevofluorane.
Sevoflurane is not a stereoisomer
Select the chiral centre in each of the following four structures.
Halothane - 2nd carbon
Enflurane - 3rd carbon
Isoflurane - 2nd carbon
Desflurane - 2nd carbon
With regard to structural isomers:
A. They have the same molecular formula
B. They have the same structural formula
C. They have the same physical properties
D. They have the same pharmacological properties
E. Enflurane is a structural isomer
A. True. This is the definition of an isomer.
B. False. Structural isomers differ in the arrangement of their atoms – the building blocks of their chemical structure.
C. False. Structural isomers may be very different compounds with different physical and chemical properties.
D. False. Although some structural isomers have similar pharmacological properties, many do not.
E. True.
Sort the enantiomeric form of the following drugs into the most and least clinically useful?
Give an overview of mechanisms of specific and non-specific drug action, and the relevance to anaesthesia.
List examples of simple non-specific drug actions with reference to their physicochemical properties
Describe the key mechanisms by which drugs exert their highly specific action, including receptor, enzyme and voltage-gated ion channels interaction
Outline the key principles that govern drug-receptor interaction including affinity, specificity, efficacy, agonism and antagonism
Give examples of commonly-used anaesthetic drugs outlining their specific mechanism of action with regards to receptor binding
A drug must first bind to its target, most commonly a receptor, in order to initiate its physiologic effect
Non-specific drug actions are governed by physicochemical interaction and tend to be less potent. Specific drug action results from the binding of a protein target, most commonly a receptor. This typically leads to a predictable cellular response
Drugs display varying degrees of specificity for receptors with many being stereoselective
Receptors are complex proteins found within or on the cell membrane and can be categorised into groups accordingly to their molecular structure. These include G-protein coupled receptors, ion channels, enzymes and nuclear receptors. They are intrinsic to the process of cell-signalling which ultimately leads to an alteration in cell function
Receptor-effector coupling is often a multi-step process with both positive and negative feedback mechanisms
Key concepts that describe this drug-receptor interaction include affinity, specificity, efficacy, agonism and antagonism
Drug binding is not equivalent to drug effect. The ability for a drug-receptor complex to elicit its effect is governed by its efficacy and can be displayed using concentration-response curves
Drugs act to either increase or decrease the various functions of a biological system. They can be classified according to their concentration-effect relationships
Regarding the mechanism of drug action:
A. A drug is an exogenous chemical substance that is used to alter a physiological system
B. Physicochemical forces that form the basis of simple non-specific drug action and tend to be less potent
C. Sugammadex is a gamma cyclodextrin that is used to reverse the effects of the steroidal neuromuscular blocking agent rocuronium via neutralisation
D. The osmotic diuretic mannitol acts to reduce intracranial pressure by lowering the plasma osmolality
A. True. Drugs can be identical to an endogenous compound. Drugs can exert their effect in a number of different ways ranging from simple non-specific action to highly specific actions on protein targets, namely receptors. The majority of the drugs we use in anaesthesia act in a highly tissue specific manner.
B. True. Important physicochemical forces include covalent bonding, ionic bonds, hydrogen bonds and van der Waals’ forces.
C. False. Sugammadex is an encapsulating agent and is an example of chelation. Antacids and protamine are examples of neutralisation.
D. False. Mannitol is an alcohol derivative of the sugar mannose. It is freely filtered at the glomerulus acting as an osmotic diuretic resulting in intravascular depletion. It increases plasma osmolality which leads to a reduction in extracellular brain water and thus decrease intracranial pressure.
Regarding receptors:
A. Receptors are only found on the cell surfaces and display variable expression throughout the body
B. G protein-coupled receptors represent the largest superfamily of receptor. They are located on the cell surface and present a common binding site for hormones and neurotransmitters
C. Voltage-gated ion channels are ionotropic, being responsible for rapid synaptic transmission
D. Tyrosine kinases are receptors that are linked to cytoplasmic enzymes and mediate the first steps in the transduction of signals carried by insulin and a wide variety of growth factors
E. Cytoplasmic soluble guanylate cyclase is activated by nitric oxide resulting in vasoconstriction and platelet aggregation via cyclic GMP production in smooth muscle cells
A. False. At a cellular level, receptors can be located on the cell surface, be linked to cytoplasmic enzymes or be found within the cell nucleus. The activation of cell surface receptors typically leads to rapid, short-term effects, whereas those involving nuclear transcription factors tend to induce a slower, long-term physiological response.
B. True. Over 500 families of GPCR proteins have been identified to date. These are intrinsically linked to a diverse array of second-messenger molecules that modulate the function of downstream proteins.
C. False. Ionotropic receptors are ligand-gated, not voltage-gated, ion channels that open in the presence of an extracellular ligand. Voltage-gated ion channels open and close in response to changes in the voltage across the cell membrane.
D. True. Tyrosine kinases are part of a receptor class that are intrinsically linked to cytoplasmic enzymes. They have an extracellular domain that binds a specific ligand and a cytoplasmic domain that contains a protein tyrosine kinase. Receptor binding activates the tyrosine kinases resulting in auto-phosphorylation of enzyme domains.
E. False. Soluble guanylate cyclase activity triggers an increase in cyclic GMP production, leading to smooth muscle relaxation and subsequent vasodilatation.
Regarding the key principles that govern drug-receptor interaction including affinity, specificity, efficacy, agonism and antagonism:
A. Drug potency is governed by both its affinity for a given receptor and its efficacy
B. Affinity is probability to which a drug binds to its receptor and is reflected by its dissociation constant
C. Half-maximal effect concentration (EC50) is the most common measure of drug efficacy
D. Efficacy is an inherent property of an agonist and reflects its ability to activate the receptor and produce a maximal biological response
E. The inhibitory effect of a non-competitive antagonist is surmountable with the additional agonist
A. True. The overall effect for a given drug is dictated by both the fraction of receptors it is able to occupy and its ability to produce a biological effect.
B. True. A drug that avidly binds to a receptor is said to have a high affinity. The dissociation constant represents the concentration at which 50% of receptors are occupied. A drug with a low dissociation constant has a greater affinity. This results in a shift of the log dose-response curve to the left.
C. False. Half maximal affect concentration is the most common measure of potency. It represents the concentration at which a drug produces 50% of its maximal possible response. The more potent a drug the lower the EC50.
D. True. Efficacy is a property of the drug, not the receptor. Drugs that bind to a receptor but produce less than maximal activation are partial agonists whereas an inverse agonist displays negative efficacy.
E. False. Only the inhibitory effects of a competitive antagonist can be overcome by increasing the dose of an agonist. This is because increasing the agonist concentration effectively displaces the antagonist from its binding site. Non-competitive antagonists either bind irreversibly to the agonist binding site or to a distant site, reducing agonist binding via an allosteric mechanism. An example is ketamine with glutamate at the NMDA receptor with the CNS.
Regarding the actions of certain drugs:
A. The action of lidocaine is dependent on the blockade of the sodium voltage-gated ion channel
B. Noradrenaline is an endogenous neurotransmitter that causes vasoconstriction by stimulating alpha-1 adrenoceptors and positive inotropic / chronotropic effects via beta-1 adrenoceptors
C. Propofol is a non-barbiturate hypnotic intravenous agent that binds to the beta-subunit of the GABAA receptor resulting in cell hyperpolarisation via a reduction in chloride channel conductance
D. Opioid receptors are inhibitory G protein-coupled receptors that act to reduce pain transmission and are subject to desensitisation with chronic stimulation
E. Benzodiazepines modulate the effects of GABAA at GABAB receptors
A. True. Lidocaine crosses the phospholipid cell membrane in its unionized, lipid-soluble form and binds to the internal surface of a sodium channel, preventing it from leaving the inactivated state.
B. True. Noradrenaline activates Gq-proteins (via alpha-1 adrenoceptors) and Gs-proteins (via beta-1 adrenoceptors). This results in an increase in intracellular calcium and cAMP respectively.
C. False. The GABAA receptor is a ligand-gated chloride ion channel that is anion selective and inhibitory. Activation results in an increase in channel opening allowing increase chloride ion entry and cell hyperpolarisation.
D. True. Opioid receptors are G protein-coupled receptors linked to inhibitory G proteins. When stimulated by an opioid agonist, hyperpolarisation of the cell membrane and reduced levels of cyclic AMP result in the inhibition of neurotransmitter release
E. False. Benzodiazepines act on specific allosteric binding sites on the ionotropic GABAA receptor (ligand-gated ion channel) exerting their anxiolytic or sedative effects. GABAB receptors are metabotropic (acting via G protein and second messengers) and when stimulated increase potassium conductance, thereby hyperpolarising the neuronal membrane. The antispasmodic baclofen is an example of a GABAB receptor agonist.
Give an overview of agonists and antagonists, and their relevance to anaesthesia.
define the terms affinity, potency and efficacy
define the terms agonist and antagonist
describe the concepts of full and partial agonism
discuss inverse agonism
describe the concepts of competitive, non-competitive, reversible and irreversible antagonism
The law of mass action is fundamental in the relationship between drug and receptor. It states that the rate of the reaction is proportional to the concentrations of the reacting elements (drug, receptors, and drug-receptor complexes).
Affinity describes how avidly a drug will bind to its receptor. The dissociation constant (KD) refers to the tendency of the drug-receptor complex to dissociate back to its component parts. A drug with high affinity will have a low KD.
Potency describes the quantity (either concentration or dose) of a drug required to produce a maximal effect. Higher potency will result in lower values for EC50 and ED50.
Efficacy describes the maximum effect produced by a drug once it has bound to the receptor. Intrinsic activity provides a means of quantifying this on a scale from -1.0 to 1.0.
An agonist is a drug with affinity for a receptor that has intrinsic activity once bound. An antagonist is a drug that has affinity for a receptor but has no intrinsic activity once bound.
Regarding affinity:
A. Affinity describes how avidly a drug binds to its receptor.
B. The dissociation constant reflects the strength of the drug-receptor bond.
C. The dissociation constant has the symbol KD.
D. If a drug has a low affinity the KD will be small.
E. KD is the concentration of a drug when 50% of its receptors are occupied.
A. True.
B. False.
C. True.
D. False.
E. True.
The affinity constant reflects the strength of the drug-receptor bond. The dissociation constant reflects the tendency of the drug-receptor to dissociate back to its drug and receptor components. The dissociation constant is represented as KD. If a drug has a high affinity the KD will be small.
Regarding potency:
A. Potency describes the quantity of drug needed to activate a receptor.
B. The EC50 and ED50 refer to the same concept.
C. The EC50 is the concentration of a drug that produces 50% of the maximal response.
D. The ED is the dose of a drug that produces a response in 50% of the population.
E. A more potent drug will have a lower ED50.
A. False.
B. False.
C. True.
D. True.
E. False.
Potency refers to the quantity of drug needed to produce a maximal effect. Potency is compared using concentration (EC50) or dose (ED50). These two concepts are similar, but subtly different. A drug with a lower EC50 will have a higher potency.
Regarding efficacy:
A. The Emax is the maximum effect that can be expected from a drug.
B. Further doses of the drug will increase the Emax.
C. Intrinsic activity = Emax of a full agonist/Emax of the drug.
D. An intrinsic activity of 0 would represent an antagonist.
E. An intrinsic activity of 0.7 would represent a partial antagonist.
Submit
A. True.
B. False.
C. False.
D. True.
E. False.
The maximum effect that can be generated by a drug once it has bound to the receptor is the Emax, accordingly, further doses are unable to produce a greater effect.
Intrinsic activity is the drug’s maximal efficacy as a fraction of the maximal efficacy produced by a full agonist, i.e. intrinsic activity = Emax of the drug/Emax of a full agonist. It may lie between 1 (a full agonist) and -1 (an inverse agonist).
An intrinsic activity of 0.7 would represent a partial agonist.
Regarding agonists and antagonists:
A. An inverse agonist is a drug that produces an effect opposite to an endogenous agonist
B. Agonists can be either competitive or non-competitive
C. Increasing the concentration of the agonist will overcome the inhibitory effect of an irreversible competitive antagonist
D. Non-competitive antagonists bind at different sites to the receptor
E. Increasing the concentration of the agonist will not overcome the inhibitory effect of a non-competitive antagonist
A. True.
B. False.
C. False.
D. True.
E. True.
Antagonists may be described as either competitive or non-competitive. Competitive antagonists may be either reversible or irreversible. In the presence of a reversible competitive antagonist, increasing the concentration of the agonist will overcome any inhibitory effect.
The term that best describes the dose of a drug needed to produce a maximum effect is:
A. Affinity
B. Potency
C. Efficacy
D. Intrinsic activity
E. Full agonism
A. Incorrect. Affinity describes how avidly a drug binds to its particular receptor. Numerically it is the reciprocal of the equilibrium dissociation constant KD and represents the drug concentration at which 50% of the receptor population are occupied.
B. Correct. Potency describes the quantity of drug needed to produce a maximal effect. It is compared using concentration (EC50) or dose (ED50). The potency of a drug partly depends on its affinity for receptors and partly upon the efficiency with which it produces a response. Drugs that have high affinity for a particular receptor tend to produce a response at a lower dose, i.e. have a higher potency.
C. Incorrect. Efficacy, or Emax , is the maximum effect that can be expected from a drug once it has bound to the receptor. The maximum effect of a drug is reached when a further increase in the drug dose will not produce any greater magnitude of effect.
D. Incorrect. Intrinsic activity (IA) is the drug’s maximal efficacy as a fraction of the maximal efficacy produced by a full agonist. If no response is produced then the drug has an IA of 0, if it produces a maximal response it has an IA of 1.
E. Incorrect. A full agonist is a drug that produces a maximal response when bound to its specific receptor, i.e. it has an Emax of 1.0.
Fill in the blanks:
Once a receptor is occupied by a drug it may or may not result in activation of that receptor. Activation means that the receptor is affected by the bound drug in such a way as to elicit a tissue response, i.e. it has intrinsic activity. If the drug binds and results in activation it is an agonist. If, on the other hand, the drug binds but fails to result in activation it is an antagonist.
Please label the following dose-response curve:
A full agonist will produce a maximal (100%) response, i.e. have an Emax of 1.0. An agonist that, despite affinity for a receptor, produces a sub-maximal response is a partial agonist and will have an Emax that lies between 0 and 1.0.
Regarding inverse agonists:
A. They can have an intrinsic activity of 0.8
B. A full agonist will have a higher affinity than an inverse agonist
C. They produce an effect opposite to an endogenous agonist
D. An antagonist has an intrinsic activity of 0
E. They compete with endogenous agonists for the same receptor
A. False.
B. False.
C. True.
D. False.
E. False.
Inverse agonists will produce an effect opposite to an endogenous agonist. They will have intrinsic activity which will be expressed as a negative number. The affinity for a receptor cannot be determined based on whether it is a full or inverse agonist. Affinity refers to how easily the drug binds to a receptor, it does not provide information on the response produced.
Regarding the dose-response curve in Figure 1:
A. Agonist A has a lower Emax
B. Agonist B shows the agonist effect in the presence of a non-competitive antagonist
C. The EC50 of both agonists are identical
D. Line B shows the agonist effect in the presence of a competitive antagonist
A. False.
B. False.
C. False.
D. True.
Antagonists may be described as either competitive or non-competitive. Competitive antagonists are subdivided into those that are reversible or irreversible. In the presence of a reversible competitive antagonist, increasing the concentration of the agonist will overcome any inhibitory effect. This is not the case with irreversible antagonists. The Emax of the agonist will remain unchanged, although the EC50 will increase.
Non-competitive antagonists bind to a different site to the receptor, resulting in conformational change. Here the Emax will fall and the EC50 will increase.
Give an overview of partial agonists and inverse agonists, and the relevance to anaesthesia.
Define the terms partial agonist and inverse agonist
Draw log (dose) v response curves and relate the features of this curve according to the properties of the drug
List examples of partial and inverse agonists and recognise how their pharmacodynamics relate to their clinical use
A partial agonist is a drug which produces a sub-maximal response even when all the receptors are fully occupied. It has intrinsic activity of greater than 0 but less than 1
A partial agonist can reduce the potency of a full agonist when they act upon the same receptors and they are present simultaneously
Buprenorphine has partial agonist effects as MOP and NOP opioid receptors
Receptors are not necessarily quiescent when there is no agonist present. They can demonstrate constitutive activity
Inverse agonists suppress constitutive activity and therefore produce a negative response by the system
Inverse agonists have an intrinsic activity of less than 0
The term which best describes how well a drug binds to a receptor is:
A. Efficacy
B. Affinity
C. Potency
D. Intrinsic activity
A. Incorrect. Efficacy - describes the magnitude of receptor response produced by a drug at a defined receptor. The maximum effect of a drug is reached when a further increase in the drug dose will not produce any greater magnitude of effect.
B. Correct. Affinity - a unique, derived constant for each drug-receptor combination that is the measure of the drug’s ability to bind to that particular receptor. Numerically it is the reciprocal of the equilibrium dissociation constant (K[D]) and represents the drug concentration at which 50% of the receptor population are occupied. Receptor selectivity occurs due to differing affinity for different receptors based upon the drug structure. Selectivity is generally deemed to be favourable in drug development as it aids reduction of adverse effects caused by off-target drug actions.
C. Incorrect. Potency - the concentration (EC50) or dose (ED50) of a drug required to produce 50% of that drug’s maximal effect. The potency for a drug partly depends on its affinity for receptors and partly upon the efficiency with which it produces a response. Drugs that have high affinity for a particular receptor tend to produce a response at a lower dose, i.e. have a higher potency.
D. Incorrect. Intrinsic activity - the drug’s maximal effect presented as a fraction of the maximal efficacy (Emax) capable by a system with a full agonist acting through the same receptors under the same conditions. If no response is produced then the drug has an intrinsic activity (IA) of 0, if it produces a maximal response it has an IA of 1.
Drug A has an ED50 of 25 mg.
Drug B has an ED50 of 50 mg.
A. Drug A is half as potent as Drug B
B. Drug A is twice as potent as Drug B
C. Drug B is twice as potent as Drug A
A. Incorrect.
B. Correct. Although two drugs, A and B, may produce the same maximal system response, drug A produces a response at half the dose of drug B. Therefore drug A has twice the relative potency of drug B.
C. Incorrect.
D. Incorrect.
The graph demonstrates a log (dose) v response curve for several full agonists at a particular receptor. It demonstrates the effect of drug potency upon the curve; a less potent drug requires a higher concentration in order to produce the maximal effect (thereby shifting the curve to the right).
See if you can label this example log (dose) v response curve for full agonists:
Although all these drugs produce the same maximal response they do this at differing drug concentrations and their EC50s are different. Therefore potency is A>B>C>D.
Which statements are correct regarding the graph in Fig 1?
A. Drug B has an intrinsic activity of 0.65
B. Drug A is a partial agonist
C. If the dose of Drug B is increased it will achieve the same maximal effect as Drug A
D. Drug A is more potent than Drug
A. Correct.
B. Incorrect.
C. Incorrect.
D. Correct.
Drug B produces a response at 65% of that which is possible in the system when a full agonist is used. This equates to an IA of 0.65. Drug A has an EC50 lower than Drug B, therefore drug A is more potent than drug B.
When in the presence of a partial agonist, the full agonist log (dose) v response curve will:
A. Remain unchanged
B. Have reduced maximal effect
C. Move to the left
D. Move to the right
A. Incorrect.
B. Incorrect.
C. Incorrect.
D. Correct. This is demonstrated on the log (dose) v response graph below by the curve being pushed towards the right when both full agonist and partial agonist are present.
Regarding partial agonists:
A. A partial agonist has an intrinsic activity of 1
B. Buprenorphine shows partial agonism at MOP and NOP receptors
C. Partial agonists increase the potency of full agonists when present within the same system
D. A maximal system response can be produced by increasing the dose of the partial agonist
A. Incorrect. A partial agonist has intrinsic activity that lies between 0 and 1. It does not produce a maximal effect. A drug with an intrinsic activity of 1 is a full agonist.
B. Correct.
C. Incorrect. Partial agonists can have an antagonistic effect on full agonists, this effectively reduces the potency of the full agonist.
D. Incorrect. A partial agonist can never produce a maximal system response, regardless of dose.
Regarding inverse agonists:
A. An inverse agonist can have an intrinsic activity of 0.6
B. A full agonist will have higher affinity than an inverse agonist
C. An inverse agonist suppresses the constitutive activity in a system
D. An inverse agonist has an intrinsic activity of 0
A. Incorrect.
B. Incorrect.
C. Correct.
D. Incorrect.
Inverse agonists suppress the constitutive activity present in a system. They therefore have intrinsic activity which will be expressed as a negative number. We cannot determine which drug has greater affinity for a receptor on the basis of whether it is a full or inverse agonist. Affinity refers to how easily the drug binds to a receptor, it does not provide information on the response produced.
Label the graph, demonstrating the varying relative efficacies of an agonist, antagonist and inverse agonist.
Give an overview of Drugs Affecting Transmembrane Signalling, and the relevance to anaesthesia.
Understand the mechanisms of transmembrane signalling
Explain transmembrane signalling with regards to basic groups of drugs
Receptors use different mechanisms of transmembrane signalling
G protein-coupled receptors are affected by opioids
Ion channel receptors are affected by local anaesthetics
Insulin receptors are an example of enzyme-linked receptors
G protein-coupled receptors:
A. Constitute the largest family of transmembrane receptors
B. Depend for their activity on the dissociation of the G protein into its constitutive subunits
C. Are the predominant receptor category for anaesthetic drug action
D. Have seven transmembrane helices
E. Do not work via second messengers
A. True.
B. True.
C. False. Anaesthetic drugs predominantly target ligand-gated ion channels.
D. True.
E. False. G protein-coupled receptors synthesise second messengers, depending on which G protein is coupled to the receptor.
G protein-coupled receptors (GPCRs) are the largest family of integral membrane proteins. 50% of modern drugs target GPCRs. These include opioids.
The action of the GPCR depends on three elements:
Receptor (Fig 1a)
The receptor has seven transmembrane helices. Binding sites are in the extracellular regions or between helices. It has an intracellular binding site for the G protein.
G protein (Fig 1b)
A G protein is a heterotrimeric protein composed of three subunits – α, β and γ.
Effector molecule (Fig 1c)
The effector molecule is activated by the α subunit of the G protein. It synthesises second messengers, depending on which G protein is coupled to the receptor. For example, β adrenergic receptors stimulate production of cAMP, α1 adrenergic receptors stimulate inositol trisphosphate (IP3), diacylglycerol (DAG), and Ca2+.
Which of the following are ion channel receptors?
A. G protein-coupled receptors
B. GABA receptors
C. Sodium channel receptors
D. NMDA receptors
E. Insulin receptors
A. False. G protein-coupled receptors act via a second messenger.
B. True.
C. True.
D. True.
E. False. Insulin receptors are enzyme-linked receptors.
Ion channel receptors are multimeric proteins located in the cell membrane. Each of these proteins arranges itself to form a passage extending from one side of the hydrophobic membrane to the other. The amino acids that line the channel and the physical width of the channel determine which ions are able to pass through. Ion channels cause a much faster reaction within the cell.
Types of ion channels:
- Voltage-gated ion channels
Voltage-gated ion channels are responsive to changes in the local electrical membrane potential (Fig 1). They are critical for the function of excitable cells, such as neurons and muscle cells.
Calcium channel blockers are one example of a drug that acts on voltage-gated ion channels. Other examples are sodium channel blockers, class III antiarrhythmics and local anaesthetics.
- Ligand-gated ion channels
Ligand-gated ion channels mediate passive ion flux driven by the electrochemical gradient for the permeant ions. They are gated by the binding of a specific ligand to an orthosteric site(s) that triggers a conformational change.
They are responsible for fast synaptic transmission in the nervous system and at the somatic neuromuscular junction.
Examples of this group of ion channels are:
Nicotinic acetylcholine receptors (nAChRs)
5HT3 receptors
NMDA receptors
GABA receptors (Fig 1)
Drugs affecting these receptors include muscle relaxants and most anaesthetic induction agents.
- Other ion channels
Other ion channel families include the aquaporins (Fig 1) (which also includes aquaglyceroporins), a family of chloride channels, which includes the cystic fibrosis transmembrane conductance regulator (CFTR).
Drugs that act on these receptors are ivacaftor (CFTR potentiator) and lumacaftor (CFTR correcting).
Which receptors do the following drugs act on?
Give an overview of nuclear drug receptors, and the relevance to anaesthetics.
understand the concept of intracellular nuclear receptors
recall the main types of nuclear receptors
recall the basic mechanisms of action
explain the role these receptors play in protein synthesis and clinical response
Intracellular receptors target transcription and DNA sequencing.
They are found inside the cell membrane in the cytoplasm or nucleus of the cell.
Steroid receptors and thyroid hormone receptors are important examples of intracellular receptors.
Drugs can target these receptors causing gene expression and protein synthesis.
Which of the following are intracellular receptors?
A. G-protein coupled receptors
B. Ligand-gated channel receptors
C. Oestrogen receptors
D. Tyrosine kinase receptors
E. Thyroid hormone receptors
A. False. This is an example of a cell surface receptor.
B. False. This is an example of a cell surface receptor.
C. True. Oestrogen receptors are type 1 intracellular receptors.
D. False. This is an example of a cell surface receptor.
E. True. Thyroid hormone receptors are type 2 intranuclear receptors.
Intracellular receptors are receptors located inside the cell membrane. They can be cytosolic (in the cell cytoplasm) or intranuclear (inside the nucleus of the cell). There are 2 main types.
Type 1
Ligands bind to protein receptor in cytoplasm or nucleus (Figure 1). Some examples are:
sex hormone receptors (oestrogen, testosterone)
cortisol receptors
mineralocorticoid receptors
Type 2
Ligands bind directly to DNA proteins (Figure 2). Some examples are:
vitamin A receptors
vitamin D receptors
retinoid receptors
thyroid hormone receptors
Regarding intracellular receptors:
A. They are only found in the nucleus
B. They have intrinsic transcriptional activity
C. They have ligands that are small and lipophilic
D. They do not contain zinc loops
E. Only type 1 class intracellular receptors bind directly to DNA proteins
A. False. They can be cytosolic or intranuclear.
B. True.
C. True.
D. False. The DNA-binding domain has ‘zinc fingers’ which are phosphorylated during transcription.
E. False. Type 1 intracellular receptors bind to protein receptors in the cytoplasm or nucleus.
Intracellular receptors have intrinsic transcriptional activity (Figure 1). Structurally they have:
a transcription-activating domain
a DNA-binding domain
a ligand-binding domain
The DNA-binding domain has zinc-containing loops (‘zinc fingers’) which phosphorylate when binding occurs.
Ligands that bind to intracellular receptors are small lipophilic molecules which easily cross the phospholipid membrane, for example steroid hormones, thyroid hormones and vitamin D.
Regarding thyroid hormone receptors:
A. They are only found in the nucleus
B. They contain a DNA-binding domain
C. They are only activated by thyroxine
D. They are type 1 intracellular receptors
E. When activated, thyroid hormone receptors bind via the transcription activating domains
A. True. They are examples of intranuclear receptors.
B. True.
C. False. Thyroid receptors can be activated with or without ligand binding. Also, T4 is de-iodinated intracellularly in the cytoplasm to the more active form T3. rT3, however, is metabolically inactive.
D. False. Thyroid hormone receptors are type 2 intranuclear receptors.
E. False. The DNA-binding domain of the active TR binds via the thyroid response elements.
Thyroid hormone production is a system regulated by the hypothalamic-pituitary-thyroid axis (Figure 1). This system works via a negative feedback loop where the thyroid hormone (T3 or T4) sends a signal to the hypothalamus and anterior pituitary to decrease TRH (thyroid releasing hormone) and TSH (thyroid stimulating hormone) production respectively.
The thyroid hormone receptor (TR) is a type 2 intracellular receptor which is already inside the nucleus (Figure 2). They have an important role in the physiological functioning of the body and are responsible for growth, metabolism and development.
The effects of thyroid hormone are mediated by the changes in expression of T3 responsive genes in target tissues. These changes in gene expression can occur both with and without ligand binding, thus making these receptors unique.
When thyroid hormone binds to the TR it undergoes a conformational change. The DNA-binding domain (of the active TR) will then bind to the thyroid response elements (TREs) which are located in the promoter region of the T3-responsive gene. Subsequently, gene transcription and expression will occur (Figure 1).
When TR binds without its ligand, it recruits a co-repressor protein. This causes the gene to be silent and thus gene expression will not take place.
Regarding gene expression and protein synthesis:
A. Gene expression is a fast process
B. Gene expression and protein production occurs in minutes
C. Intranuclear receptors are important drug targets
D. Receptor activation causes protein production
E. Intranuclear receptors are important in gene transcription
A. False. It is a slow process.
B. False. It occurs in hours to days.
C. True. Many useful drugs target intranuclear receptors, for example exogenous steroids.
D. True. The ultimate aim of gene expression is the production of specific proteins.
E. TrueIt is important to understand how these receptors work, as they are frequently targeted in clinical practice.
Intranuclear receptors have a slow onset of action. This is why it can take hours to days to see clinical responses when drugs targeting these receptors are given. It takes time for gene expression and protein production to occur once the receptor is activated. Furthermore, once administered, these drugs can have longer durations of action, lasting beyond the drug’s presence in the body.
Think about the patient with an exacerbation of asthma; the steroids given will not work immediately. In order to produce the desired clinical outcome (decreased inflammatory immune response) it will take several hours to days.
Give an overview of Enzymes as Drug Targets Including Anticholinesterases, and the relevance to anaesthetics.
Describe anticholinesterases, their mechanism of action (including short- vs medium- vs long-acting) and their common uses
List carbonic anhydrase inhibitors and their uses
Identify NSAIDs and their mechanisms of action
Describe monoamine oxidase inhibitors and their uses
Enzymes are important for the body to be able to carry out fast chemical reactions, which allows complex multicellular organisms to exist
Carbonic anhydrase inhibitors are used for glaucoma and for mountain sickness. They achieves this by alkalinisation of urine to create a metabolic acidosis to cause further hyperventilation to improve oxygenation
NSAIDs are a group of anti-inflammatory agents that are used as opioid sparring pain killers
MAOi are rarely used as first-line drugs, although a certain proportion of patients are still on these and thus special attention needs to be paid to them to avoid potentially catastrophic complications
Regarding anticholinesterases:
A. They are divided into ultra-fast acting, medium-acting and long-acting subgroups
B. Tensilon test is a diagnostic test for Myasthenia Gravis
C. Neostigmine is hydrolysed in exactly the same way as acetylcholine, but takes much longer
D. Medium-acting anticholinesterases can be used as oral treatment of myasthenic crisis
A. False. Anticholinesterases are divided into short-, medium- and long-acting.
B. True.
C. True.
D. False. Myasthenic crisis is a complication of myasthenia gravis characterised by worsening of muscle weakness, resulting in respiratory failure that requires intubation and mechanical ventilation.
Anticholinesterases are agents that target the enzyme acetylcholinesterase by occupying its active site to prevent ACh breakdown (Fig 1).
They are used for a multitude of clinical scenarios. For anaesthetists they are most commonly used to reverse the effects of non-depolarising muscular blocking agents (NdMBA). By reducing the breakdown of ACh, anticholinesterases increase the competitive displacement of NdMBA from the neuromuscular junction (NMJ).
They are also used in the diagnosis and treatment of myasthenia gravis (MG) and are an ingredient in pesticides and nerve gases.
They are classified according to duration of action:
- Short-acting
Edrophonium (Tensilon) (Fig 1) is an example of a short-acting anticholinesterase:
Lasts 10-20 mins
Used to diagnose MG, where the strength of muscles improves after administration of Tensilon - Medium-acting
Neostigmine (Fig 2):
Lasts 1-2 hours
Carbamylates the active site of the enzyme and, once bonded, it is hydrolysed like ACh, but takes much longer
Inhibits the action of plasma pseudocholinesterases and so prolongs the effects of suxamethonium and mivacurium
Reverses competitive neuromuscular blockers and is used to treat constipation on the Intensive Care Unit (ICU)
Is added to glycopyrrolate to offset that drug’s parasympathetic anticholinergic effects, e.g. bradycardia, hypotension, bronchoconstriction
Pyridostigmine
Lasts 2 to 3 hours
An oral treatment of MG
Physostigmine
Lasts 30 minutes to 5 hours
Available as topical eye drops for treatment of glaucoma
- Long-acting
chothiophate (Fig 3):
Lasts weeks
Phosphorylates the active site of the enzyme in a covalent bond. The enzyme, therefore, takes weeks to hydrolyse the drug
Was historically used for treatment of glaucoma
This group also includes Sarin and VX nerve gases used in chemical warfare, and tetraethyl pyrophosphate (TEPP) which is an insecticide
In toxic doses, e.g. organophosphorus poisoning, anticholinesterases cause SLUDGE syndrome (Salivation, Lacrimation, Urination, Defecation, GI upset and Emesis) and can cause death by paralysis of the respiratory muscles. Treatment is with antimuscarinic agents such as atropine or pralidoxime
Regarding long-acting anticholinesterases:
A. They can last days, not weeks
B. They form an ionic bond with the enzyme that takes weeks to break down
C. They are used in chemical warfare and pesticides
D. SLUDGE syndrome includes sweating, lacrimation, uraemia, defecation, GI upset and erectile dysfunction
A. False. Long-acting drugs can last weeks.
B. False. They form a covalent bond that takes weeks to break down.
C. True.
D. False. Sludge syndrome is characterised by salivation, lacrimation, urination, defecation, GI upset and emesis.
Carbonic anhydrase enzymes are found in the following cells:
A. Gastrin cells of the stomach
B. Proximal convoluted tubules in the kidney
C. Red blood cells
D. Pancreatic cells
A. False. Carbonic anhydrase enzymes are found in the parietal cells of the stomach.
B. True.
C. True.
D. True.
Carbonic anhydrase is an enzyme that catalyses the following formulae:
CO2 + H2O H2CO3 ←→ H+ + HCO3
Acetazolamide is a carbonic anhydrase inhibitor which is used for the treatment of mountain sickness and as a weak diuretic, as well as for the prophylaxis and treatment of glaucoma.
These act on the proximal convoluted tubules. They are non-competitive inhibitors of the enzyme affecting the sodium/H+ exchanger, thus causing alkaline urine with metabolic acidosis.
Regarding NSAIDs:
A. NSAIDs can be classified by their structure or COX inhibition
B. They have no opioid sparing effect
C. NSAIDs all have anti-platelet activity
D. COX 2 inhibitors have no significant risks
A. True.
B. False. NSAIDs have opioid sparing effect.
C. False. Only aspirin has been used for its antiplatelet activity.
D. False. COX 2 inhibitors have less risk of bleeding, but still have significant side-effects. Some have been withdrawn due to their increased risk of thrombotic complications.
Regarding cyclo-oxygenase inhibitors:
A. COX 2 is found in most cells under normal conditions
B. They aim to reduce the inflammatory mediators produced by the COX enzyme
C. They cannot trigger an asthma attack in those susceptible
D. Prostanoids are in a fine balance with prostacyclin
A. False. COX 1 is found in most cells, COX 2 is found mainly in inflammatory cells.
B. True.
C. False. COX inhibitors are well known to cause deterioration in asthma of those susceptible to this, by increasing production of leukotrienes causing bronchospasm.
D. True.
Non-steroidal anti-inflammatory drugs (NSAIDs) exert their effects by inhibiting the action of cyclo-oxygenase (COX) enzymes, thus reducing the production of prostanoids, i.e. inflammatory mediators. There are three forms of COX:
COX-1 is constitutive and is found in most cells
COX-2 is inducible and is normally undetectable in normal tissues, but is found in abundance in macrophages and other inflammatory cells
COX-3 is an isoenzyme most likely to be a CNS variant of COX-1 (site of action of paracetamol)
Normally prostanoids are in a fine balance with prostacyclin, a vasodilator which also prevents formation of platelet plug, and thromboxane, a vasoconstrictor and potent platelet aggregator.
COX inhibitors block conversion of arachidonic acid to prostanoids and thus more of it is converted to leukotrienes, which can cause bronchospasm - this is the mechanism whereby some asthmatics may worsen on administration of NSAIDs.
COX inhibitors can be classified either by structure or by COX inhibition:
Structure:
Acetylsalicylic acid (aspirin)
Phenyloacetic acids (diclofenac)
Carboacetic acid (indomethacin)
Propionic acids (ibuprofen)
Enolic acid (piroxicam)
COX inhibition:
Non-selective, e.g. aspirin, diclofenac and ibuprofen
Selective, e.g. parecoxib
NSAIDs are used as anti-inflammatory and analgesic agents. They have opioid-sparing effects.
Aspirin has unique antiplatelet action and thus is used for the prophylaxis and treatment of arterial thrombosis.
COX-2 inhibitors cause less risk of bleeding and cause fewer ulcers, but some of these have been associated with increased incidence of thrombotic complications, especially myocardial infarction.
GI bleeding risk is due to decreased levels of circulating prostaglandins that are essential in maintaining gastric mucosal integrity.
NSAID-induced exacerbation of asthma occurs in 10-20% of asthmatics due to increase production of leukotrienes.
Regarding monoamine oxidase:
A. They are normally bound to the cell surface
B. They break down monamines in the body only from food sources
C. They can have a role in psychiatric conditions
D. The two main types are MAO-AB and MAO-RIMA
A. False. They are normally attached to mitochondria.
B. False. They help break down all monoamines, including ones from food, and thus can inadvertently lead to a hypertensive crisis.
C. True. They have been implicated in many neurological and psychiatric conditions.
D. False. They are classified as MAO-A, RIMA or MAO-B.
Monoamine oxidases are a group of enzymes that, as the name suggests, catalyse the oxidation of monoamines. This uses its oxygen molecule to remove an amine group from the drug in question. They are found in most cell types, normally bound to the mitochondria.
They breakdown monoamines in food and hence are found in the liver, aka tyramine oxidase. They are also vital in the breakdown of monoamine neurotransmitters, and as such have a big role in many neurological and psychiatric conditions, where monoamine oxidase inhibitors (MAOi) are used.
There are two types of monoamine oxidases found in humans:
MAO-A
MAO-A is found in high concentrations in the cortex, while the cingulate gyrus had an equal balance of both. These are found in areas where there is increased serotonergic neurotransmission
MAO-A is found in the liver, pulmonary vascular endothelium, GI tract and placenta
MAO-B
MAO-B is found in the striatum and Globus pallidus in high quantities, and correlates to areas of the brain where there is a high level of noradrenaline neurotransmission (norepinephrine)
There are equally high levels of both MAO in the hypothalamus and hippocampal uncus
MAO-B is found in platelets
MAO-A and MAO-B deaminate monoamines by oxidation, as described earlier, and as indicated by the name.
Oxygen is used to remove an amine group, and its adjacent hydrogen atom, from the molecule in question, resulting in a ketone, or aldehyde, and ammonia (Fig 1).
Monoamine oxidases are classified as belonging to the family of proteins known as flavin-containing amine oxidoreductases (flavoproteins). They have a covalently-bonded FAD (flavin adenine dinucleotide). Both types are very similar in structure and have hydrophobic substrate binding sites.
MAO-A breaks down:
Serotonin
Melatonin
Noradrenaline
Adrenaline
MAO-B breaks down:
Phenethylamine
Benzylamine
They both also breakdown:
Dopamine
Tyramine
Tryptamine
MAOi can be divided into non-selective (older) and selective agents, as well as Reversible Inhibitors of MAO-A (RIMAs)
Regarding MAO:
A. MAO are found in the brain, liver, lung and placenta
B. Only MAO-B is found from birth
C. They remove an amine from a molecule
D. They are part of the flavoprotein family
A. True.
B. False. Only MAO-A is found from birth
C. True.
D. True.
MAO-A breaks down:
A. Noradrenaline
B. Metformin
C. Adrenaline
D. Selegiline
A. True.
B. False.
C. True.
D. False.
A good way to remember this is: MAO-A breaks down MANS (melatonin, adrenaline, noradrenaline and serotonin).
MAOi:
A. Are used as first-line agents in depression
B. Can be used in combination with SSRI/SNRI
C. Can be used in Parkinson’s disease
D. Can sometimes be used as prophylaxis in migraines
A. False. These drugs are not routinely used as first-line agents due to their many interactions, especially with food products.
B. False. They should never be used in conjunction with SSRIs/SNRIs due to the unacceptable risk of increased neurotransmitter levels in the brain causing hypertensive crisis.
C. True.
D. True.
Monoamine oxidases are a group of enzymes that, as the name suggests, catalyse the oxidation of monoamines. This uses its oxygen molecule to remove an amine group from the drug in question. They are found in most cell types, normally bound to the mitochondria.
They breakdown monoamines in food and hence are found in the liver, aka tyramine oxidase. They are also vital in the breakdown of monoamine neurotransmitters, and as such have a big role in many neurological and psychiatric conditions, where monoamine oxidase inhibitors (MAOi) are used.
There are two types of monoamine oxidases found in humans:
MAO-A
MAO-A is found in high concentrations in the cortex, while the cingulate gyrus had an equal balance of both. These are found in areas where there is increased serotonergic neurotransmission
MAO-A is found in the liver, pulmonary vascular endothelium, GI tract and placenta
MAO-B
MAO-B is found in the striatum and Globus pallidus in high quantities, and correlates to areas of the brain where there is a high level of noradrenaline neurotransmission (norepinephrine)
There are equally high levels of both MAO in the hypothalamus and hippocampal uncus
MAO-B is found in platelets
MAO-A and MAO-B deaminate monoamines by oxidation, as described earlier, and as indicated by the name.
Oxygen is used to remove an amine group, and its adjacent hydrogen atom, from the molecule in question, resulting in a ketone, or aldehyde, and ammonia (Fig 1).
Monoamine oxidases are classified as belonging to the family of proteins known as flavin-containing amine oxidoreductases (flavoproteins). They have a covalently-bonded FAD (flavin adenine dinucleotide). Both types are very similar in structure and have hydrophobic substrate binding sites.
MAO-A breaks down:
Serotonin
Melatonin
Noradrenaline
Adrenaline
MAO-B breaks down:
Phenethylamine
Benzylamine
They both also breakdown:
Dopamine
Tyramine
Tryptamine
MAOi can be divided into non-selective (older) and selective agents, as well as Reversible Inhibitors of MAO-A (RIMAs)
MAOi can cause:
A. Red man syndrome
B. Psychosis
C. Nausea, diarrhoea and/or constipation
D. Headache, drowsiness and/or insomnia
A. False. Red man syndrome is a skin reaction most associated with by overstimulation of specific immune cells in the body in response to vancomycin. MAOi can cause serotonin syndrome.
B. True.
C. True.
D. True.
Which of the following drugs are irreversible enzyme inhibitors?
A. Penicillin
B. Diclofenac
C. Aspirin
D. Omeprazole
E. S-Warfarin
A. True. Penicillin and other beta-lactam antibiotics irreversibly inhibit transpeptidase in the bacterial cell wall, disrupting peptidoglycan cross-linking and cell wall synthesis.
B. False. Diclofenac is a reversible COX inhibitor.
C. True. Aspirin is an irreversible enzyme inhibitor of platelet cyclo-oxygenase for the platelet’s lifespan, requiring the formation of new platelets to recover their normal function.
D. True. Omeprazole is a proton pump inhibitor that irreversibly inhibits the hydrogen/potassium ATPase pump in gastric parietal cells to decrease acid secretion. The synthesis of new proton pumps is necessary to produce gastric acid again.
E. False. Warfarin inhibits vitamin K epoxide reductase, which is required to regenerate reduced vitamin K, which can then activate clotting factors.
Irreversible enzyme inhibitors either act via allosteric binding to alter the active site, or by binding directly to the active site of the enzyme.
This last mechanism is usually through the drug forming covalent bonds at the active site of the enzyme. This prevents it from binding its endogenous ligand, but also the drug-enzyme complex cannot dissociate so the degree of inhibition cannot be overcome by increasing the substrate concentration. Reversal, therefore, depends on the synthesis of new enzymes, e.g. the action of aspirin on cyclo-oxygenase.
Proton pump inhibitors, e.g. omeprazole, also irreversibly inhibit the hydrogen/potassium ATPase pump in gastric parietal cells to decrease acid secretion. The synthesis of new proton pumps is necessary to produce gastric acid.
Drugs may act as analogues to an endogenous ligand and can compete with them to transiently block the active site of the enzyme from being accessed, decreasing the rate of complex formation and reaction.
The degree of inhibition depends on the concentration of the drug relative to that of the endogenous substrate, so increasing inhibition as the drug concentration rises.
Inhibition may be overcome by increasing the amount of substrate present, therefore the Vmax eventually achieved remain the same, but are reached at a slower rate.
Most competitive inhibitiors act reversibly.
Examples of reversible competitive enzyme inhibitors include:
Acetylcholinesterase inhibitors, e.g. neostigmine
Cyclo-oxygenase inhibitors, e.g. non-steroidal anti-inflammatory drugs: ibuprofen and diclofenac
Phosphodiesterase inhibitors, e.g. theophylline
Angiotensin converting enzyme inhibitors, e.g. ramipril
GIve an overview of Enzyme Induction and Inhibition, and the relevance to anaesthesia.
Discuss enzyme kinetics
Define the drugs that target enzyme systems
Explain the processes of enzyme induction and inhibition
Describe common pharmacokinetic interactions occurring due to altered enzyme activity
Drugs may elicit their therapeutic effect through enzyme stimulation or inhibition
A number of commonly used drugs are direct enzyme inhibitors
Drug metabolism by the cytochrome P450 enzyme system is subject to Michaelis-Menten kinetics
Pharmacokinetic metabolic interactions can occur due to cytochrome P450 enzyme induction or inhibition leading to adverse drug effects
Examples of acetylcholinesterase inhibitors and their mechanism of action include:
Neostigmine and pyridostigmine
Neostigmine and pyridostigmine act as substrates to form a carbamylated enzyme complex with AChE, with a slow rate of hydrolysis over 30 minutes, preventing it from metabolising ACh.
Organophosphate compounds
Organophosphate compounds irreversibly inhibit AChE. They phosphorylate the esteratic site of AChE, making it resistant to hydrolysis and reactivation. It may be reactivated with pralidoxime. Pralidoxime binds to the anionic site of AChE, then cleaves the phosphate-ester bond. Thus the organophosphate dissociates from AChE.
Organophosphate poisoning produces a cholinergic syndrome featuring salivation, urination, diarrhoea, muscle weakness and bradycardia and requires resuscitation and organ support to allow time for enzyme recovery or synthesis, and elimination of the poison.
Edrophonium
Edrophonium acts by binding to the anionic site, reversibly inhibiting AChE. It has a fast onset of action with a short duration and is used clinically to differentiate a myasthenic crisis, where neuromuscular function should improve, from that of a cholinergic crisis.
Monoamine oxidases (MAO) are isoenzymes which deaminate neurotransmitters. Types A and B exist; MAO-A deaminates serotonin and catecholamines and MAO-B deaminates tyramine and phenyethamine. Both metabolise dopamine.
MAO inhibitors are used in the management of resistant depression, obsessive compulsive disorder, chronic pain and Parkinson’s disease.
The older non-selective monoamine oxidase inhibitors, phenelzine and tranylcypromine, act irreversibly. Moclobemide is a reversible selective MAO-A inhibitor and selegeline is an irreversible selective MAO-B inhibitor.
Question. Which antibiotic is a reversible MAO inhibitor?
Linezolid, used in the treatment of resistant infections such as vancomycin-resistant Enterococci and methicillin-resistant Staphylococcus aureus, is a reversible MAO inhibitor.
Any patient taking MAO inhibitors may exhibit pronounced hypertension and arrhythmias on administration of indirectly acting sympathomimetics which are metabolised by MAO and should be avoided. Directly acting sympathomimetic amines should also be used with caution as they may also have an exaggerated response, but they are additionally metabolised by catechol-o-methyl-transferase.
G protein-coupled receptors (GPCRs) act through second messenger systems, i.e they are metabotropic. GPCR stimulation triggers coupling with a specific G protein on the intracellular side of the membrane to activate or inhibit enzymes. G proteins Gs or Gi stimulate or inhibit respectively, the enzyme adenylyl cyclase that regulates cAMP formation (Fig 1). Gq activates phospholipase C.
Question: Can you give an example of a drug that works via a Gi protein-coupled receptor?
Morphine. Opioid receptors MOP, KOP and DOP are linked to Gi. When stimulated by opioid-receptor binding, it leads to the inhibition of adenylyl cyclase, reducing levels of cAMP, and therefore decreasing neurotransmitter release.
What are the potential effects of treating a patient taking diltiazem with clarithromycin?
Clarithromycin inhibits CYP 3A4 activity, reducing the metabolism of calcium channel blockers such as diltiazem, leading to increased serum concentrations with an increased risk of bradycardia, A-V blockade and hypotension.
In the case of pro-drugs, enzyme inhibitors reduce their conversion into active metabolites and decrease the drug’s effect. For example, inhibition of the CYP 2D6 isoenzyme that metabolises codeine results in the decreased production of codeine-6-glucuronide, norcodeine and morphine, and patients may experience less effective analgesia. This is seen with the co-administration of anti-depressants like fluoxetine.
Conversely, a patient taking an enzyme-inducer, such as carbamazepine, may demonstrate an increased analgesic effect.
Regarding mechanisms of enzyme inhibition:
A. Neostigmine and pyridostigmine competitively inhibit acetylcholinesterase
B. Non-competitive inhibitors can be overcome by increasing the substrate concentration
C. Aminophylline and theophylline are examples of selective PDE inhibitors
D. The cytochrome P450 enzyme system is not subject to dose-dependent kinetics
E. Genetic polymorphisms in CYP can influence the speed of dug metabolism and response
A. True. Neostigmine, pyridostigmine and physostigmine are reversible enzyme carbamylators that act as substrates binding to both sites of AChE to form a carbamylated enzyme complex, with a slow rate of hydrolysis over 30 minutes, preventing it from metabolising ACh.
B. False. Non-competitive inhibitors bind away from the active site of the enzyme, preventing it from being activated, inhibiting product formation. It decreases the Vmax achievable and cannot be overcome by increasing the substrate concentration.
C. False. Aminophylline and theophylline are examples of non-selective PDE inhibitors. They are used in the management of acute severe asthma and uncontrolled chronic asthma for their bronchodilatory effects.
D. False. CYP enzyme-drug reactions follow dose-dependent kinetics as described by the Michaelis-Menten equation influencing the plasma drug levels achieved.
E. True. There are poor, extensive and ultra-rapid phenotypes. Extensive and rapid phenotypes rapidly metabolise relevant drugs, leading to reduced plasma levels with loss of efficacy, necessitating increased doses to achieve a therapeutic response. However, they metabolise pro-drugs like codeine into their active metabolites faster, with a greater risk of opioid-related side-effects.
Regarding Michaelis-Menten kinetics:
A. They obey the law of mass-action
B. The Michaelis constant, Km is the concentration of substrate at which the reaction is occurring at 50% of its maximum velocity
C. They describe the affinity the enzyme has for its substrate
D. The addition of a competitive inhibitor to an enzyme reaction decreases the Vmax that can be achieved
E. They describe kinetics involving covalent bonding between enzyme and substrate
A. True. Michaelis-Menten kinetics obey the law of mass action between receptor saturation and drug concentration.
B. True. Km is the concentration of substrate at which the reaction is at 50% of its maximum velocity.
C. True. It is also a measure of the binding affinity between the enzyme and a specific substrate. A higher Km indicates a lower affinity of the enzyme for a substrate and a higher concentration of substrate is necessary to bind the enzyme to reach Vmax, which is approached more slowly.
D. False. Competitive inhibitors block the active site, therefore decreasing enzyme affinity and increasing the Km. Increasing the substrate concentration increases enzyme-substrate complex formation, so the Vmax achievable is not altered, whereas a non-competitive inhibitor decreases the Vmax that can be achieved.
E. False. Michaelis-Menten kinetics describe enzyme and substrate reactions which are weakly bonded and allow dissociation.
Enzymes are protein catalysts that increase the speed at which chemical reactions occur with substrates at physiological conditions, without being expended themselves. They lower the activation energy necessary to start a reaction and orientate molecules to allow the reaction to progress more rapidly.
They have an active site, which binds a specific substrate to form an enzyme-substrate complex. This reacts to either synthesise or release a product, subsequently leaving the enzyme intact for future reactions (Fig 1).
The rate of such chemical reactions can be described as follows:
First order reaction, whose rate depends on the concentration of the reacting substrate and is an exponential process approaching a maximum velocity
Zero order reaction; this is reached when the enzyme systems’ active sites have all become saturated. The rate then becomes constant and independent of any further changes in the substrate concentration
The Michaelis-Menten equation can be used to describe the rate or velocity of such reactions.
The Michaelis-Menten equation can be used to describe the rate or velocity of such reactions.
V = (Vmax [S]) / (Km + [S])
V is the velocity of the reaction.
Vmax is the maximum velocity of the reaction. This is reached when enzymes active sites have been saturated with substrate.
S is the substrate concentration.
Km is the Michaelis constant, specific to a single substrate-enzyme reaction. It is defined as the concentration of substrate at which the velocity of the reaction is half of the maximum velocity, Km = ½Vmax as demonstrated in Fig 1.
It is also the reciprocal of the enzymes affinity for a specific substrate. A small Km will have a high affinity for a substrate so less is required to reach ½Vmax at a faster rate.
Cytochrome P450 enzyme induction:
A. Increases the formation of new CYP enzymes
B. Produces a conformational change in the enzymes active site to increase substrate binding
C. May be caused by smoking
D. Is not associated with alcohol consumption
E. Develops immediately following drug binding
A. True. Enzyme induction involves increased transcription and production of more isoenzyme to increase metabolic activity.
B. False. There is no change in the active site. However, the presence of more enzyme increases its affinity for the substrate.
C. True. Polycyclic aromatic hydrocarbons in cigarette smoke can induce CYP1A2 and CYP2E1 expression with higher rates of metabolism of opioids and volatile agents.
D. False. Alcohol abuse strongly induces CYP2E1 enzyme expression.
E. False. Enzyme induction takes some time to develop.
Drug metabolism occurs mainly in the liver in two phases. Drugs generally undergo phase I followed by phase II reactions, but some are metabolised by phase II reactions only:
Phase I reactions include oxidation, reduction and hydrolysis and are catalysed by the cytochrome P450 (CYP) enzyme system. They are also required to convert pro-drugs such as codeine and ACE inhibitors into their active form
Phase II reactions include glucuronidation, sulfation, acetylation and methylation. These processes conjugate drugs to increase their water solubility and improve their excretion. They involve enzymes such as uridine-diphospho-glucuronosyl transferase, which have a role in the metabolism of drugs like codeine, morphine, and propofol. These may also be subject to enzyme induction
CYP is mainly found in the smooth endoplasmic reticulum of hepatocytes as well as many tissues throughout the body, including the gut mucosa, kidney and lung. They are classified into a number of families, subfamilies and isoforms, with some drugs being metabolised by more than one isoenzyme.
57 of these isoenzymes have been identified, with 30 being involved in drug metabolism, the majority of which is performed by only 6:
CYP 1A2
CYP 2C9
CYP 2C19
CYP 2D6
CYP 2E1
CYP 3A4
These are listed in Table 1, with some important drugs they metabolise.
These CYP enzyme-drug reactions follow dose-dependent kinetics as described by the Michaelis-Menten equation. Plasma levels of a drug may therefore be affected by the concurrent administration of other drugs undergoing metabolism by the same cytochrome P450 isoenzyme.
CYP2D6 is an important isoform with numerous polymorphisms and is involved in the metabolism of commonly encountered perioperative drugs such as codeine, tramadol, ondansetron, as well as antidepressants, neuroleptics, beta-blockers and anti-arrhythmics. Polymorphisms result in variable individual responses to opioids; 5-10% of Caucasians are poor metabolisers, 65-80% are extensive metabolisers and 5-10% are ultra-rapid metabolisers with a variable prevalence found globally. Ultra-rapid metabolism is seen in up to 29% of the population in Ethiopia, but only in 0.5% of the Chinese, where 30% may be poor metabolisers.
CYP3A is the most abundant subfamily and is responsible for metabolising opioids, benzodiazepines, tricyclic antidepressants, anti-arrhythmics, calcium channel antagonists and statins amongst many other drugs.
Regarding the Cytochrome P450 system:
A. Centrilobular hepatocytes contain low concentrations of CYP450
B. It is involved in phase 2 reactions
C. Drugs that are activated by CYP450 enzymes display an increased effect with enzyme induction
D. CYP450 isoenzymes metabolise specific substrates
E. The majority of all drug metabolism occurs through only 6 isoenzymes
A. False. Centrilobular hepatocytes contain high concentration of CYP 450 enzymes.
B. False. Phase I reactions include oxidation, reduction and hydrolysis and are catalysed by the cytochrome P450 (CYP) enzyme system.
C. True. Pro-drugs that require activation will demonstrate an increased effect in the presence of enzyme inducers.
D. True. Enzymes react with specific substrates.
E. True. 90% of foreign substances are metabolised by the following six isoenzymes: CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP2E1, and CYP3A4.
Drug metabolism occurs mainly in the liver in two phases. Drugs generally undergo phase I followed by phase II reactions, but some are metabolised by phase II reactions only:
Phase I reactions include oxidation, reduction and hydrolysis and are catalysed by the cytochrome P450 (CYP) enzyme system. They are also required to convert pro-drugs such as codeine and ACE inhibitors into their active form
Phase II reactions include glucuronidation, sulfation, acetylation and methylation. These processes conjugate drugs to increase their water solubility and improve their excretion. They involve enzymes such as uridine-diphospho-glucuronosyl transferase, which have a role in the metabolism of drugs like codeine, morphine, and propofol. These may also be subject to enzyme induction
CYP is mainly found in the smooth endoplasmic reticulum of hepatocytes as well as many tissues throughout the body, including the gut mucosa, kidney and lung. They are classified into a number of families, subfamilies and isoforms, with some drugs being metabolised by more than one isoenzyme.
57 of these isoenzymes have been identified, with 30 being involved in drug metabolism, the majority of which is performed by only 6:
CYP 1A2
CYP 2C9
CYP 2C19
CYP 2D6
CYP 2E1
CYP 3A4
These are listed in Table 1, with some important drugs they metabolise.
These CYP enzyme-drug reactions follow dose-dependent kinetics as described by the Michaelis-Menten equation. Plasma levels of a drug may therefore be affected by the concurrent administration of other drugs undergoing metabolism by the same cytochrome P450 isoenzyme.
CYP2D6 is an important isoform with numerous polymorphisms and is involved in the metabolism of commonly encountered perioperative drugs such as codeine, tramadol, ondansetron, as well as antidepressants, neuroleptics, beta-blockers and anti-arrhythmics. Polymorphisms result in variable individual responses to opioids; 5-10% of Caucasians are poor metabolisers, 65-80% are extensive metabolisers and 5-10% are ultra-rapid metabolisers with a variable prevalence found globally. Ultra-rapid metabolism is seen in up to 29% of the population in Ethiopia, but only in 0.5% of the Chinese, where 30% may be poor metabolisers.
CYP3A is the most abundant subfamily and is responsible for metabolising opioids, benzodiazepines, tricyclic antidepressants, anti-arrhythmics, calcium channel antagonists and statins amongst many other drugs.
Regarding cytochrome P450 enzyme inhibition:
A. Drugs may inhibit multiple classes of isoenzymes
B. Glucocorticoids are potent enzyme inhibitors
C. Competitive inhibition decreases as the drug plasma concentration falls
D. Penicillin is a reversible bacterial transpeptidase inhibitor
E. Neostigmine is a reversible competitive acetylcholinesterase inhibitor
A. True.
B. False. Dexamethasone is an inducer of CYP2D6.
C. True.
D. False. Penicillin and beta-lactam antibiotics are irreversible inhibitors of transpeptidase and inhibit peptidoglycan cross-linking, therefore weakening the bacterial cell wall.
E. True.
Drug metabolism occurs mainly in the liver in two phases. Drugs generally undergo phase I followed by phase II reactions, but some are metabolised by phase II reactions only:
Phase I reactions include oxidation, reduction and hydrolysis and are catalysed by the cytochrome P450 (CYP) enzyme system. They are also required to convert pro-drugs such as codeine and ACE inhibitors into their active form
Phase II reactions include glucuronidation, sulfation, acetylation and methylation. These processes conjugate drugs to increase their water solubility and improve their excretion. They involve enzymes such as uridine-diphospho-glucuronosyl transferase, which have a role in the metabolism of drugs like codeine, morphine, and propofol. These may also be subject to enzyme induction
CYP is mainly found in the smooth endoplasmic reticulum of hepatocytes as well as many tissues throughout the body, including the gut mucosa, kidney and lung. They are classified into a number of families, subfamilies and isoforms, with some drugs being metabolised by more than one isoenzyme.
57 of these isoenzymes have been identified, with 30 being involved in drug metabolism, the majority of which is performed by only 6:
CYP 1A2
CYP 2C9
CYP 2C19
CYP 2D6
CYP 2E1
CYP 3A4
These are listed in Table 1, with some important drugs they metabolise.
These CYP enzyme-drug reactions follow dose-dependent kinetics as described by the Michaelis-Menten equation. Plasma levels of a drug may therefore be affected by the concurrent administration of other drugs undergoing metabolism by the same cytochrome P450 isoenzyme.
CYP2D6 is an important isoform with numerous polymorphisms and is involved in the metabolism of commonly encountered perioperative drugs such as codeine, tramadol, ondansetron, as well as antidepressants, neuroleptics, beta-blockers and anti-arrhythmics. Polymorphisms result in variable individual responses to opioids; 5-10% of Caucasians are poor metabolisers, 65-80% are extensive metabolisers and 5-10% are ultra-rapid metabolisers with a variable prevalence found globally. Ultra-rapid metabolism is seen in up to 29% of the population in Ethiopia, but only in 0.5% of the Chinese, where 30% may be poor metabolisers.
CYP3A is the most abundant subfamily and is responsible for metabolising opioids, benzodiazepines, tricyclic antidepressants, anti-arrhythmics, calcium channel antagonists and statins amongst many other drugs.
The following drugs may display pharmacokinetic interactions causing adverse effects:
A. Rifampicin and warfarin
B. Paroxetine and metoprolol
C. Erythromycin and diltiazem
D. Ritonavir and methadone
E. Cimetidine and rocuronium
A. True. Rifampicin induces CYP2C9 with a risk of inadequate anticoagulation by warfarin.
B. True. Paroxetine inhibits CYP2D6, reducing the metabolism of beta-blockers.
C. True. Erythromycin inhibits CYP3A4, reducing the metabolism of diltiazem.
D. False. Ritonavir is a widespread enzyme inhibitor, but has not been shown to significantly affect methadone’s efficacy. Lopinavir induces metabolism of methadone and may lead to opioid withdrawal.
E. False. Cimetidine is an enzyme inhibitor, but does not influence rocuronium, which is largely excreted unchanged.
Give an overview of unwanted drug side effects, and the relevance to anaesthesia.
Define unwanted effects
Explain reactions that can occur in anyone and reactions that occur only in susceptible individuals
Discuss ways of categorising unwanted effects
Give examples of these unwanted drug effects
Discuss types of allergic reactions and common tests
Unwanted effects of the drugs we prescribe and administer are common and potentially life-threatening
Some of these unwanted effects can be predicted, or have their severity reduced, by careful history-taking, by having detailed knowledge of the pharmacology of the drug, and by being aware of potential issues and the rapid responses to those issues
There are several different ways to classify and define unwanted effects
Further testing, and obtaining specialist help in the case of some reactions, may be required to assess the risk of administration of a drug, e.g. if suxamethonium apnoea or malignant hyperthermia is suspected
Genetic variation between individuals and between ethnic populations can lead to unwanted drug effects.
Plasma pseudocholinesterase E1a gene
A mutation in plasma pseudocholinesterase E1a gene is associated with deficient enzyme activity and affects metabolism of suxamethonium and mivacurium.
Suxamethonium apnoea is one of the most common unwanted effects in anaesthesia.
4% of Caucasians carry this abnormal gene. This figure is higher in Asians and those from Middle Eastern descent and lower in Africans
Cytochrome P450 2D6 enzyme
Cytochrome P450 2D6 enzyme (CYP2D6) is responsible for converting codeine to its active metabolites.
Variations in this enzyme can affect the metabolism of codeine, tramadol, ondansetron and beta-blockers.
Poor metabolisers of codeine may have a poor analgesic result. There are extensive metabolisers, for example there is lower activity of the enzyme in Chinese populations compared to Caucasian populations. There are also ultrarapid metabolisers, such as the Saudi Arabian and Ethiopian populations, who metabolise these drugs faster and more completely.
Adverse Drug ReactionsReactions due to Genetic Variation
Genetic variation between individuals and between ethnic populations can lead to unwanted drug effects.
Select each label for more information.
CYP2C19 enzyme
In Chinese populations, a CYP2C19 enzyme deficiency affects response to diazepam and tricyclic antidepressants.
G6PD enzyme
G6PD enzyme deficiency results in haemolytic anaemia in susceptible individuals.
Such a deficiency is commonest in Mediterranean, African and Asian populations and it affects the use of sulphonamides and nitrofurantoin in susceptible individuals.
Porphobilinogen deaminase
Porphobilinogen deaminase deficiency causes acute intermittent porphyria.
It is commonest in Switzerland, Sweden and the Netherlands and affects barbiturates and nitrofurantoin use.
Regarding adverse drug reactions:
A. Suxamethonium apnoea is an example of a reaction that occurs in susceptible individuals
B. Suxamethonium apnoea is an example of a Type A reaction
C. Hypoglycaemia with aspirin is an example of a Type A reaction
D. Malignant hyperthermia is an example of drug idiosyncrasy
E. Anaphylaxtic reactions are mediated by IgE
A. True.
B. False. Suxamethonium apnoea is an example of a Type B (bizarre) reaction.
C. True.
D. True
E. True.
Type A (augmented)
Type A reactions make up 85-90% of all adverse drug reactions
They are often related to pharmacological effects of the drug
They are commonly dose-related
They can be further divided into:
Primary reactions due to an exaggerated response to the drug, e.g. hypoglycaemia with aspirin
Secondary reaction not related to the desired effect, e.g. tinnitus with aspirin
Type B (bizarre)
Type B reactions are bizarre, unpredicted, and not dose-related
They make up 10-15% of adverse drug reactions
Examples include suxamethonium apnoea, malignant hyperthermia and hepatic porphyria
These reaction types occur in susceptible individuals only:
A. Drug intolerance
B. Drug idiosyncrasy
C. Drug overdose
D. Drug side-effect
E. Drug interaction
F. Drug allergy
G. Pseudoallergic reaction
A. True.
B. True.
C. False. Drug overdose can occur in anyone.
D. False. Drug side-effects can occur in anyone.
E. False. Drug interactions can occur in anyone.
F. True.
G. True.
Regarding the classification of reactions:
A. Coombs’ types are a way of defining reactions by time of onset
B. Type A and Type B classification is a way of defining reactions by time of onset
C. Coombs types are a way of defining reactions by immunological type
D. DoTS classification is a way of defining reactions according to dose, timing and susceptibility
A. False. Coombs’ types defines reactions by immunological type, not by time of onset.
B. False. Type A and Type B classification is a way of defining reactions by augmented and bizarre reactions.
C. True.
D. True.
Regarding allergic reactions:
A. Pseudoallergy is immunologically medicated
B. Allergy is immunologically mediated
C. RAST is a test for diagnosis of allergy
D. Allergic reactions must happen within minutes to be a true allergy
A. False. Pseudoallergic reactions have the same clinical manifestations as an allergic reaction, due to histamine release, but are not immunologic reactions.
B. True.
C. True.
D. False. Allergic reaction can be described as immediate and delayed. Immediate reactions, i.e. Type I reactions, classically begin within one hour of the first administered dose. Delayed reactions are those appearing after one hour, although most of them begin after six hours, and typically after days of treatment. Drug-induced hypersensitivity syndrome (DiHS) can begin after weeks of continuous treatment.
The commonest triggers were:
Antibiotics (47%) - Teicoplanin comprised 12% of antibiotic exposures, but caused 38% of antibiotic-induced anaphylaxis
Muscle relaxants (33%) - Suxamethonium-induced anaphylaxis was twice as likely as with other NMBAs and mainly presented with bronchospasm. Atracurium-anaphylaxis mainly presented with hypotension
Chlorhexidine (9%)
Patent Blue dye (5%)
Commonest presenting features:
Hypotension (46%)
Bronchospasm (particularly in patients with morbid obesity and asthma) (18%)
Tachycardia (9.8%)
Oxygen desaturation (4.7%)
Bradycardia (3%)
Reduced/absent capnography trace (2.3%)
All patients were hypotensive during the episode. 15% of patients had a cardiac arrest. The elderly with cardiac disease and the obese were most at risk of cardiac arrest and death.
Immediate
This is intended to distinguish IgE-mediated, Type I reactions from other types. Type I reactions classically begin within one hour of the first administered dose. This period of one hour identifies the majority of IgE-mediated reactions, which carry the risk of anaphylaxis if the patient is re-exposed.
Delayed
Delayed reactions are those appearing after one hour, although most begin after six hours and typically after days of treatment. These reactions may be caused by several different mechanisms, but they are not IgE-mediated. Types II, III, and IV immunologic reactions are all considered delayed reactions.
Drug rash with eosinophilia and systemic symptoms (DRESS) also known as drug-induced hypersensitivity syndrome (DiHS) can begin after weeks of continuous treatment. It is characterised by fever, rash, and multiorgan involvement, with or without eosinophilia and lymphocytosis. Hepatitis and hypersensitivity myocarditis may also occur. These reactions can persist for weeks to months, even after the medication is stopped.
Diagnosis of allergic reactions may be through:
Skin prick tests
RAST (radioallergosorbent tests)
Mast cell tryptase
Provocation tests
Give an overview of drug tolerance and tachyphylaxis, with the relevance to anaesthetics.
Define the terms tolerance and tachyphylaxis
Describe a classification system for types of tolerance
Explain how tolerance and tachyphylaxis differ
List clinically relevant examples of tachyphylaxis and tolerance
Drug tolerance may be thought of as a decrease in pharmacological response following repeated or prolonged drug administration
Tachyphylaxis describes a hyper-acute form of tolerance in which rapidly increasing doses are necessary to maintain a clinical effect over a time frame
Pharmacodynamics describes how drugs interact at a molecular level to produce their effects.
When a drug is given frequently, particularly at high concentrations, a desensitisation effect can occur due to repeated binding interactions between the drug and its receptor. This is known as pharmacodynamic tolerance.
Question: What other possible mechanisms are there?
Other possible mechanisms include a reduction in receptor density, conformational changes in the receptor structure and changes to the action potential thresholds. Down-regulation of receptors and associated reduction in neurotransmitter release may also play a part. Changes typically occur over days to weeks.
Pharmacodynamic tolerance develops at different rates for different effects of any given drug, e.g. cocaine users can quickly become tolerant to the euphoric ‘high’ they experience and therefore need ever larger doses to achieve that effect again. However, tolerance to the toxic cardiovascular effects is less pronounced.
It would be beneficial if tolerance occurred quickly to the negative side-effects of any drug whilst sparing the main clinical use. However, in practice it is a variable and unpredictable phenomenon and risks accidental overdose and serious harm if care is not taken.
Regarding classification systems for types of tolerance:
A. Innate tolerance describes a lack of sensitivity to a drug the first time it is administered
B. Acquired tolerance can be subdivided into three main categories: pharmacokinetic, pharmacodynamic and behavioural tolerance
C. Pharmacodynamics is concerned with the absorption, distribution, metabolism and excretion of a drug
D. Pharmacokinetics is most commonly associated with orally ingested agents
A. Correct.
B. Correct.
C. Incorrect. Pharmacokinetics is concerned with the absorption, distribution, metabolism and excretion of a drug and is therefore sometimes referred to as ‘what your body does to the drug’.
D. Correct.
As tolerance develops, a series of both physiological and psychological behaviours occur alongside. These may take the form of compensatory responses that continue for as long as the drug is taken.
Question: What symptoms can these include?
They can include symptoms such as pain, discomfort or mood changes and are usually described as physical dependence. Intense cravings can sometimes accompany this, which in the past was felt to be psychological but has now been recognised as a complex neuro-hormonal response and the basis for addiction.
If a drug to which tolerance has developed and for which a physical dependence has resulted, is stopped abruptly, withdrawal symptoms may occur. These are often unpleasant for the patient and may include sweating, paranoia, tachycardia and mood disturbance. They usually last for days to weeks and can be avoided or at least minimalised by gradually tapering off the usual dose.
The difference between tolerance and tachyphylaxis is:
A. Tolerance is when larger doses of a drug are required to produce the same effect
B. Tachyphylaxis is when there is a rapid increase in response to repeated doses over a short time period
C. Tachyphylaxis describes a hyper-acute form of tolerance
D. Clinically noticeable tachyphylaxis can sometimes occur even between the first and second doses of a drug
A. True.
B. False. Tachyphylaxis is when there is a rapid decrease in response to repeated doses over a short time period
C. True.
D. True.
Tachyphylaxis describes a hyper-acute form of tolerance in which rapidly increasing doses are necessary to maintain a clinical effect over a time-frame that may be as short as minutes.
Clinically noticeable tachyphylaxis can sometimes occur even between the first and second doses of a drug.
It has been postulated that rapid occupation of almost all the drug binding sites after one dose may be the cause, although the exact mechanism is unknown.
Clinically relevant examples in anaesthesia include ephedrine and hydralazine.
Clinically relevant examples of tachyphylaxis and tolerance are:
A. Opiates are a good example of a drug to which tolerance occurs to the desired effects
B. Clinically relevant examples in anaesthesia for tachyphylaxis include ephedrine and hydralazine
C. Alcohol is a good example of a drug that exhibits tachyphylaxis
D. Genetic polymorphisms in the enzyme alcohol dehydrogenase play a significant role in the wide variety of tachyphylaxis demonstrated between different groups
A. True.
B. True.
C. False. Alcohol does not typically exhibit tachyphylaxis. Repeat dosing over either hours or days tends to result in progressively increasing effects such as euphoria, confusion, ataxia and vomiting. However, tolerance to these effects does develop over a period of months to years with repeated use.
D. False. Genetic polymorphisms affect an individual’s ability to metabolise alcohol. This can lead to more marked effects but not to the development of tachyphylaxis.
Opiates still form the basis of systemic analgesia for severe pain even 200 years after the discovery of morphine.
It is a good example of a drug to which tolerance occurs rapidly to the desired effect, i.e. analgesia, but much less so to the unwanted effects, e.g. nausea, itching, constipation. This leads to rapid dose escalation in chronic users with the attendant risks of chronic dependence and accidental overdose.
Opiates undergo extensive hepatic metabolism via phase 1 and 2 pathways, leading to acquired tolerance over time.
Polymorphisms in the cytochrome P450 system are an obvious source of individual variation in this process. In particular, the extremely polymorphic CYP2D6 can lead to metabolism of opiates over a wide range of time-frames. Such patients have been classified as either poor, intermediate, extensive or ultra-rapid metabolisers.
Poor metabolisers will not readily convert codeine into morphine and will therefore appear more tolerant, whilst the ultra-rapid metabolisers may experience profound clinical effects, a state that has been linked to unexpected deaths in a small group of such patients 2.
Opiates are also substrates of the efflux transporter P-glycoprotein. It is expressed in a number of tissues in which it regulates absorption or penetration of substances across cellular membranes. Up-regulation over time of this barrier transporter could limit central nervous system penetration and therefore contact with opioid receptors
A number of metabolites are derived from opiates. This is most well described with morphine, which is broken down in the liver to morphine-3-glucuronide and morphine-6-glucuronide. Whilst the latter is a potently active metabolite, the former has been associated with neuro-excitatory behavioural responses opposed to the analgesic effects of morphine at its receptors 4.
Pharmacodynamic mechanisms also contribute to opioid tolerance. These include receptor mediated changes, receptor polymorphisms and cross tolerance. The process of Mu-receptor activation for example is complex involving multiple sequential steps but is thought to differ depending on whether stimulation is acute or chronic.
Regarding tolerance and tachyphylaxis:
A. Tolerance is an increase in pharmacological response to a drug following prolonged or repeated exposure
B. Tachyphylaxis only occurs when a patient has been taking a drug for a long period of time
C. Tachyphylaxis describes an acute drop in blood pressure after administration of a drug
D. Innate tolerance describes the influence of our genetic make-up on the handling of drugs
A. False. Tolerance is a decrease in pharmacological response to a drug following prolonged or repeated exposure.
B. False. Tachyphylaxis is a rapid form of tolerance, it can occur after one dose.
C. False. Anaphylaxis describes an acute drop in blood pressure after administration of a drug.
D. True.
Tolerance can be classified in a number of ways, with the simplest initial division into
Innate tolerance
Innate tolerance results from the influence of our genetic make-up on the handling of drugs.
It describes a lack of sensitivity to a drug the first time it is administered. Put simply, everyone reacts differently to the administration of any drug but when the effects of that drug, either wanted or unwanted, are markedly reduced in an individual, then innate tolerance is said to occur.
This may be related to physical factors such as body mass index, total body fat content, the presence of frailty and acute illness, or genetic factors such as specific polymorphisms that alter drug metabolism or transport.
Acquired tolerance
Acquired tolerance can be subdivided into three main categories:
Pharmacokinetic tolerance
Pharmacodynamic tolerance
Behavioural tolerance
Give an overview of drug interactions, and the relevance to anaesthesia.
List the main types of drug interactions
Describe some classification systems for drug interactions
Give examples of some drug interactions
Describe potential interactions between drugs used in anaesthesia and commonly used herbal medications
You should be able to describe these types of drug interactions and give examples:
Physiochemical: neutralization, precipitation, chelation
Pharmacokinetic: absorption, elimination, metabolism, distribution
Pharmacodynamic interactions: summation, potentiation, synergism, and antagonism
The potential interactions between commonly used herbal medications and drugs used in anaesthesia.
Cytochrome p450: inducers and inhibitors
Pharmacokinetic interactions describe the relationship between rates of change of drug concentration in the different parts of the body. Reactions occur when absorption, metabolism or excretion of one drug is altered by the presence of another drug. It is the magnitude and duration of effect that are altered, rather than the type of effect
Absorption
Examples of absorption include:
The use of activated charcoal in poisoning cases to absorb the toxin and reduce systemic absorption
Prokinetics such as metoclopramide alter gastric emptying and therefore alter the rate of drug delivery to the absorption site and influence uptake
Muscarinic antagonists do the opposite and slow GI function, decreasing absorption
The use of local anaesthetics with adrenaline reduces local blood flow and allows larger doses to be more safely administered
Second gas effect seen with nitrous oxide and volatile agent alveolar concentrations
Distribution
Examples of distribution include:
Any drug which alters cardiac output will alter drug distribution
Beta-blockers reduce cardiac output and therefore increase the time taken for suxamethonium to reach the neuromuscular junction and take effect
Competition for binding sites alters distribution. An example would be warfarin, which is displaced by highly bound drugs such as erythromycin and amiodarone
Metabolism
Examples of metabolism include:
The liver has a hugely significant role in metabolism, and blood flow to the liver is key in this. Drugs with high intrinsic clearance, i.e. lidocaine, are flow-dependent. Increased blood flow increases clearance.
Elimination
Examples of elimination include:
Drugs affecting cardiac output affect the rate of elimination of opiates and benzodiazepines
Drugs affecting pulmonary ventilation affect the rate of elimination of inhaled agents
The water solubility of a drug is related to ionization and excretion. For example the administration of sodium bicarbonate makes urine more alkaline, which increases water solubility and excretion of weak acids, aspirin and barbiturates
Pharmacodynamics describes how a drug affects an organism. It is the relationship between drug concentration and drug response. Pharmacodynamic interactions can be described as interactions where one drug alters the sensitivity of tissues to another drug, either by having an agonistic (same) effect or an antagonistic (blocking) effect. These effects can be at the receptor level or intracellularly.
Summation
Summation is where the action of drugs is additive.
Examples include:
Premedication with a benzodiazepine and then propofol at induction. The dose of propofol required to achieve the same level of anaesthesia is lower
The use of nitrous oxide with other inhalation anaesthetics
Potentiation
Potentiation is when one drug increases the effect of another drug.
Examples include:
The potentiation of non-depolarising neuromuscular blockade by magnesium
Probenacid increasing the action of penicillin by reducing its renal excretion
Synergism
Synergism is when the combined action of two drugs is greater than the action that would be expected from summation alone.
Examples include:
Clonidine and opiates
Propofol and remifentanil
Antagonism
Antagonism is where two drugs have opposite effects.
Examples include:
Flumazenil, which is a competitive antagonist, reversing the effect of benzodiazepines
The use of neostigmine for reversal of non-depolarising muscle relaxants
Some drugs may alter electrolyte concentrations. Altered electrolyte concentrations may mediate some drug actions or alter intravascular volume
Hypokalaemia
Hypokalaemia causes increased cardiac excitability and lowers the arrhythmia threshold. Therefore it increases arrhythmia susceptibility with catecholamines and anticholinergics.
Drugs which can cause hypokalaemia include diuretics, corticosteroids and insulin.
Hyperkalaemia
Hyperkalaemia reduces cardiac automaticity.
Drugs that increase potassium concentration include suxamethonium, and potassium-sparing diuretics.
Hyponatraemia
Hyponatraemia potentiates local anaesthetics and is indicative of a depleted volume status.
Drugs causing hyponatreamia include diuretics and sulphonylurea.
Hypernatraemia
Hypernatraemia is common in the critical care population or in those unable to drink water. Common presentations include thirst, confusion and muscle spasms. This can lead to seizures.
Drugs causing hypernatraemia include mannitol, sodium bicarbonate and hypertonic saline.
Hypomagnesaemia
Hypomagnesaemia can cause cardiac arrhythmias.
Drugs that can cause hypomagnesaemia include diuretics and laxatives.
Hypermagnesaemia
Administration of magnesium can cause cardiovascular effects, prolong neuromuscular blockade and has effects on peripheral vascular tone.
The American Society of Anaesthesiologists recommends that patients cease herbal medications two weeks before surgery. This time frame probably errs on the side of caution, because some of these remedies are eliminated rapidly after discontinuation.
Echinacea
Echinacea is taken to improve the immune system.
Chronic use can result in hepatic failure, which can then enhance the hepatotoxic effects of drugs such as amiodarone, methotrexate and halothane.
Ephedra
Ephedra is used as a CNS stimulant, for weight loss and for the treatment of asthma.
Caution is advised when ephedra is used in combination with other sympathomimetic drugs. Long-term use of ephedra may deplete endogenous catecholamine stores, leading to further cardiovascular instability intraoperatively and tachyphylaxis to other sympathomimetic drugs. Fatal arrhythmias have also been reported in patients taking ephedra who were exposed to halothane anaesthesia.
Garlic is used as a treatment for hypertension, hyperlipidaemia, and atherosclerosis.
It can potentiate the anti-platelet effects of aspirin and NSAIDs.
Ginger is used as an anti-inflammatory and an antiemetic.
Caution is advised when taken in combination with NSAIDS and warfarin.
Gingko biloba
Gingko biloba is thought to be neuroprotective and to improve blood flow.
It should be avoided in combination with NSAIDs, aspirin, and warfarin.
GInseng
Ginseng is used as a mood enhancer and an aphrodisiac.
Caution is advised when used in combination with NSAIDS and warfarin.
St John’s Wort
St John’s Wort is used as an antidepressant.
It has similar precautionary measures to conventional monoamine oxidase inhibitors (MAOIs).
It is a potent inducer of hepatic cytochrome P450 CYP3A4 isoform and may significantly increase the metabolism of many concomitantly administered drugs such as alfentanil, midazolam, and lidocaine.
It induces the P450 2C9 isoform that results in the reduction in effect of warfarin and NSAIDs.
The sedative properties of St John’s Wort may potentiate or prolong the effect of anaesthetic agents.
Valerian
Valerian is used as an anxiolytic and a hypnotic.
It increases barbiturate-induced sleep times. Acutely stopping the drug may result in a withdrawal syndrome.
These are examples of physiochemical interactions:
A. Neutralization
B. Metabolism
C. Precipitation
D. Elimination
E. Chelation
F. Absorption
A. True.
B. False. Metabolism is an example of a pharmacokinetic interaction.
C. True.
D. False. Elimination is an example of a pharmacokinetic interaction.
E. True.
F. True. Direct absorption is a physicochemical interaction, although absorption can also be an example of a pharmacokinetic interaction when a substance alters the absorption interface.
Pharmaceutical incompatibility describes the situation in which two drugs are either physically or chemically incompatible. Pharmaceutical and physiochemical interactions can occur both inside and outside the body. Such interactions can cause significant harm, but can also be used for therapeutic purposes.
They can be divided into:
Neutralization - For example, heparin and protamine.
Precipitation - For example, thiopentone and suxamethonium.
Chelation - For example, sugammadex and rocuronium.
Heavy metal poisoning can be treated using chemicals which chelate and remove them from the GI tract, i.e. in Wilson’s disease penicillamine can be administered to remove copper deposits from the body.
Absorption - For example, halothane dissolving into rubber.
Pharmaceutical incompatibility can also occur between drugs and equipment. An example of this is the interaction between paraldehyde and plastic, which necessitates the use of a glass syringe for administration 1.
Regarding pharmacokinetic interactions:
A. The use of local anaesthetics with adrenaline to reduce local blood flow and allow larger doses to be given more safely is an example of absorption
B. The second gas effect is an example of distribution
C. Liver blood flow is an example of metabolism
D. Drugs affecting cardiac output, which alters the rate of elimination of opiates and benzodiazepines, is an example of elimination
A. True.
B. False. The second gas effect is an example of absorption.
C. True.
D. True.
These herbal medicines interact with NSAIDs:
A. Echinacea
B. Ephedra
C. Garlic
D. Ginger
E. Gingko biloba
F. Ginseng
G. St John’s Wort
H. Valerian
A. False. Chronic echinacea use can result in hepatic failure, which can then enhance the hepatotoxic effects of drugs such as amiodarone, methotrexate and halothane.
B. False. Ephedra is used as a CNS stimulant. Caution is advised when it is used in combination with other sympathomimetic drugs.
C. True.
D. True.
E. True.
F. True.
G. True.
H. False. Valerian is used as an anxiolytic and a hypnotic. It increases barbiturate-induced sleep times.
Regarding physiochemical interactions:
A. The reaction between heparin and protamine is an example of neutralization
B. The interaction between paraldehyde and plastic is an example of pharmaceutical incompatibility
C. The reaction between thiopentone and suxamethonium is an example of chelation
D. The reaction between sugammadex and rocuronium is an example of absorption
A. True.
B. True.
C. False. The reaction between thiopentone and suxamethonium is an example of a precipitation.
D. False. Chelation explains the reaction between sugammadex and rocuronium. An example of absorption would be the reaction when halothane dissolves into rubber.
Regarding pharmacodynamic interactions:
A. Summation is where the action of drugs is additive
B. Potentiation is when the combined action of two drugs is greater than the action than would be expected from summation alone
C. Synergism is when one drug increases the effect of another drug
D. Antagonism is where two drugs have opposite effects
A. True.
B. False. Synergism is when the combined action of two drugs is greater than the action than would be expected from summation alone.
C. False. Potentiation is when one drug increases the effect of another drug.
D. True.
Regarding electrolyte abnormalities and their possible causes:
A. Hypokalaemia can be caused by steroids
B. Hyperkalemia can be caused by insulin
C. Hyponatraemia can be caused by diuretics
D. Hypernatraemia can be caused by diuretics
E. Hypermagnesaemia can be caused by laxatives
A. True.
B. False. Drugs that increase potassium concentration include suxamethonium, and potassium-sparing diuretics.
C. True.
D. True. Drugs causing hypernatraemia include mannitol, sodium bicarbonate and hypertonic saline.
E. False. Diuretics and laxatives can cause hypomagnesaemia.
Drugs which induce the cytochrome p450 system include:
A. Rifampicin
B. Barbiturates
C. Amiodarone
D. Alcohol, in chronic use
E. Phenytoin
F. Carbamazepine
G. Itraconazole
A. True.
B. True.
C. False. Amiodarone is an inhibitor of the P450 system.
D. True.
E. True.
F. True.
G. False. Itraconazole is an inhibitor of the P450 system.
Give an overview of Absorption and Bioavailability, and the relevance to anaesthesia.
Define absorption, bioavailability and first-pass metabolism
Describe factors which can affect absorption
Calculate the bioavailability for a given drug
Absorption is the passage of a drug from its site of administration into the plasma
There are multiple factors affecting absorption, including drug formulation, route of administration, physicochemical properties of the drug and local blood flow
First-pass metabolism is a phenomenon whereby the concentration of orally administered drugs is reduced, primarily by the gut wall and liver
Bioavailability is the fraction of a drug dose that reaches the systemic circulation
Bioavailability (F) is calculated by dividing the area under a curve describing concentration versus time for a non-intravenously administered drug, by the area under the curve for the same drug administered intravenously
Local anaesthetics (LAs) are weak bases. Why do you think they are less effective when administered into inflamed tissues, e.g. around an abscess?
Inflammation produces chemicals which make tissue pH more acidic. Weak bases will become more ionized and therefore not be able to cross the membranes to their intended site of action.
Remember:
An aide memoire which might help is:
AIA - Acids ionize above (their pKa)
BIB - Bases ionize below (their pKa)
Because pH and pKa are logarithmic derivatives, a difference between the two values of 1 corresponds to 90% association or dissociation, a difference of 2 to 99% and a difference of 3 to 99.9%.
Regarding absorption:
A. Absorption is defined as the movement of a drug from its site of administration into the plasma
B. Pinocytosis is an important mechanism by which drug absorption occurs
C. Carrier-mediated transport describes the mechanism of absorption whereby a drug molecule combines with an integral membrane protein and is transported against a concentration gradient
D. If 10 mg of oral morphine is administered to a patient and first-pass metabolism reduces this by about 70%, the amount reaching the systemic circulation is 7 mg
E. Local pH has an important influence on drug absorption, mainly because most drugs are weak acids or bases
A. True.
B. False. Direct diffusion is by far the most important mechanism of drug absorption. Uncharged molecules dissolve freely across the lipid bilayer of cell membranes.
C. True. In carrier-mediated transport, a molecule is absorbed by combining with a membrane protein. This may occur down a concentration gradient in which case it is called facilitated CMT and does not require energy, or against a concentration gradient which requires energy. Energy is provided by the hydrolysis of ATP.
D. False. FPM reduces the active drug to 30% of the original dose, i.e. 0.3 x 10mg = 3mg.
E. True. pH affects the degree of ionization of weak acids and bases which in turn will affect their ability to diffuse across the non-polar lipid membranes of cells.
Cell membranes form the barriers between aqueous compartments in the body. The basic structure of a cell membrane consists of a phospholipid bilayer with embedded proteins and attached carbohydrates (Fig 1).
Epithelial surfaces, such as gastrointestinal tract (GIT) mucosa and renal tubules, consist of a layer of tightly connected cells across which molecules must traverse to pass from one side to another (Fig 2).
Vascular endothelium forms a cellular layer between intra and extravascular compartments. Its permeability varies between different tissues.
The cells lining blood capillaries have pores between them allowing relatively free passage of drug molecules (Fig 3).
In order to be absorbed, drugs commonly need to cross cell membranes, e.g. drugs administered orally have to cross the GIT epithelium.
The main mechanisms by which drugs commonly cross cell membranes are (Fig 1):
- Direct diffusion, the most important by far
Non-polar (i.e. uncharged) molecules diffuse readily across cell membranes, because they can dissolve freely in the lipid bilayer (Fig 1).
The degree and rate of absorption is determined primarily by the concentration gradient across the membrane, the surface area available for absorption, and the lipid solubility.
- Via channels that traverse the lipid bilayer
Absorption of small water-soluble drugs may occur by passive diffusion, i.e. down a concentration gradient, through aqueous channels.
This is an important mechanism of transport across capillary endothelium membranes, such as in glomerular filtration.
- Carrier-mediated transport
Drug molecules which are too large or too insoluble to diffuse through cell membranes can use integral carrier proteins that normally transport naturally occurring substances for absorption.
Drug molecules can bind with these carrier proteins, which then undergo a conformational change and release the molecules on the other side of the membrane.
This method of absorption is saturable and subject to competitive inhibition by other drugs which are able to bind to the same carrier.
Carrier-mediated transport (CMT) may be:
Primary active transport
This occurs against a concentration gradient and is energy dependent, with the energy provided by adenosine triphosphate (ATP). Examples include 5FU (fluorouracil) absorption through the gut wall, levodopa uptake into the brain, and the sodium-potassium pump (Fig 1).
Facilitated diffusion
Facilitated diffusion via carrier proteins enhances absorption of molecules with low lipid solubility (Fig 2). It occurs down a concentration gradient and is not energy dependent. It is of limited importance in drug absorption, but is the mechanism by which both vitamin B is absorbed in the GIT and glucose into red blood cells.
Another mechanism of absorption which plays a fairly insignificant role in drug transport, but is worth mentioning for completeness, is pinocytosis (Fig 1).
In this energy-dependent process, molecules are engulfed by a portion of the cell membrane forming a vesicle, which is then internalized.
Drug A is a weak acid with a pKa of 5 and drug B is a weak base with a pKa of 9.
How would each drug be affected at a pH of 3, e.g. in the stomach?
A. A would become more ionized and absorption increased
B. A would become less ionized and absorption increased
C. B would become more ionized and absorption decreased
D. B would become less ionized and absorption decreased
A. False.
B. True.
C. True.
D. False.
Remember AIA and BIB. At a low pH, weak acids are more unionized and therefore easily cross through the cell membrane, whereas weak bases become more ionized and are not able to cross by passive diffusion. At a pH of 3, drug A is about 99% unionized and drug B is completely ionized.
Regarding drug absorption:
A. Drugs with high first-pass metabolism are usually prescribed in small dosages when given orally
B. If a person becomes shocked due to massive blood loss, the intravenous route would be better for emergency drugs than the intramuscular route
C. In the treatment of angina, the onset time of action for GTN would be similar if given orally or sublingually
A. False. If a drug has a high FPM only a small amount reaches the systemic circulation, therefore a large dose is usually required.
B. True. Absorption from the IM route is dependent on an adequate blood supply locally, and this may be significantly reduced in shock.
C. False. GTN has a rapid onset of action when administered sublingually, because it is taken up rapidly into the systemic circulation and avoids FPM.
Which of these factors can influence drug absorption?
A. Route of administration
B. Drug formulation
C. Local blood flow
D. Degree of ionization of the drug
A. True.
B. True.
C. True.
D. True.
All of these factors can influence drug absorption.
Regarding bioavailability (BA):
A. BA is defined as the fraction of an administered drug that reaches the systemic circulation
B. BA relates to the percentage of a drug formulation which consists of therapeutically active molecules
C. The inhalational route of drug administration has the lowest BA
A. True. This is the definition of bioavailability and represents the amount of active drug which is available at the site of action.
B. False.
C. False. Oral administration of a drug has the lowest BA, due mainly to first-pass metabolism.
Bioavailability (BA) refers to the fraction (F) of an administered drug that reaches the systemic circulation intact and is therefore available to act at the site of action.
By definition, BA following an IV dose is 100%. For example, if x number of molecules of a drug are injected into a vein, all x molecules reach the systemic circulation.
If a drug is administered by a non-intravenous route, BA is reduced because of factors involved in the absorption process. Oral BA is reduced compared to an IV dose due to GIT absorption and first-pass metabolism.
Assessing bioavailability requires calculation of the area under a curve (AUC) which describes blood drug concentration versus time following administration via a defined route.
For drugs taken orally (PO), BA is equal to the ratio of the AUC (PO) compared to the AUC (IV).
BA (F) = AUC (PO) / AUC (IV)
BA can also be calculated for other routes of drug administration, again by comparing the measured AUC to the AUC (IV).
Referring to the given plasma concentration versus time curves for the drug in Fig 1, calculate oral bioavailability.
A. 0.2
B. 0.4
C. 0.6
D. 0.8
E. 1.0
A. Incorrect.
B. Incorrect.
C. Correct. BA = AUC (PO)/AUC (IV) = 48/80 = 0.6 or 60%.
D. Incorrect.
E. Incorrect.
When discussing body fluid, we are generally referring to intracellular fluid volume, interstitial fluid volume and plasma volume as the three major components of the overall fluid volume.
The 42 L of fluid is held in different spaces within the body:
Intracellular Fluid (ICF)
This is the fluid contained within cell walls.
Irrespective of cell type, the composition of ICF is very similar. For modelling purposes, this allows us to classify and treat it as one fluid volume, even though it is divided into the millions of individual cellular volumes.
The ICF makes up 2/3 or 28/42 L of the total body water and the physiological barrier containing it is the cell membrane.
Extracellular Fluid (ECF)
Extracellular Fluid (ECF) is the remaining 1/3 or 14/42 L, which exists outside the cell membrane. This can be further subdivided into:
Intravascular fluid (roughly 1/4 or 3 of 14 L)
Interstitial Fluid (roughly 3/4 or 10.5/14 L)
Transcellular
Interstitial Fluid
Interstitial Fluid (roughly 3/4 or 10.5/14 L).
This is the fluid which lies in the space between cells, external to both the cell membrane and the vascular endothelium.
Intravascular fluid (plasm)
Intravascular fluid (roughly 1/4 or 3 of 14 L).
The fluid component of the non-cellular contents of the vascular system, i.e. the plasma volume.
The physiological barrier containing it is the vascular endothelium.
Transcellular
Transcellular, a small remaining volume of fluid which lies between epithelial lined spaces (0.5 L).
This is typically actively produced by excretion or filtration (CSF, breast milk, pleural fluid).
Give an overview of Pharmacokinetics: Distribution, Protein Binding and Body Compartments, with relevance to anaesthetics.
Describe the different body compartments, and barriers to the movement of substances between compartments
Explain the concept of volume of distribution
Describe the factors which have an impact on the distribution of a drug within the body
Explain protein binding, and its implications for drug distribution
Distribution describes the reversible transfer of drugs from one location to another
Understanding the body fluid compartments and physiological barriers between them allows us to predict how a drug will distribute
There are numerous physicochemical properties which determine the distribution of a drug
Volume of distribution can be calculated to allow assumptions about patterns of distribution, as well as calculating loading doses
Plasma proteins can act as a drug reservoir, and in some circumstances must be taken into account when calculating drug levels and dosing
Let us use IV fluid to demonstrate an example. Think about the composition of the following 3 fluids: 5% Dextrose, NaCl 0.9% and Gelofusin. If they are administered intravenously to the intravascular compartment, can you determine where each fluid will distribute to, and how much of each fluid will pass into each compartment?
Hint: think about the contents of the fluid, and whether they would be able to penetrate the different barriers between compartments.
Place the barrier names, barrier descriptions and the fluid names in each strip.
Select the labels in the diagram for information about fluid compartments and inter-compartmental barriers.
Dextrose
The Dextrose in 5% dextrose solution is rapidly metabolised by the body, and therefore behaves similarly to free water once administered.
Free water is able to distribute evenly between all three body compartments because it contains no large molecules or charged particles, which would block its movement past the physiological barriers.
Therefore, it will disperse proportionally to the volumes of the body compartments.
Sodium chloride
Sodium chloride contains lots of charged particles – Na+ and Cl-.
The vascular endothelium will not block the movement of small polarised molecules, and so the NaCl solution can move freely out of the intravascular space into the interstitial spaces.
The cell membrane will, however, block the passage of charged particles, and so they will be unable to enter the Intracellular space. Therefore, NaCl will be distributed proportionally through the ECF volume.
Gelofusin
Gelofusin/equivalent colloid fluid contains large protein structures.
These will be blocked from passing through the vascular endothelium, and therefore the whole volume administered will be confined to the intravascular space.
What factors may mean that a drug is not evenly distributed?
There are a few factors which are not accounted for by our physiological fluid compartment model.
An example would be partitioning, e.g. into lipid stores.
Lipid can make up a large % of the body by mass and the lipid solubility of a drug is an important factor in determining drug distribution. The more lipid soluble a drug, the greater the amount that will be removed from the circulation and stored in the lipid cells.
Imagine adding a block of lipid into the fluid in our simple single container model of the body.
Now, if we add our drug to the container it has the opportunity to disperse through the fluid, and the lipid.
Scenario 1
If the drug is not lipid soluble, e.g. a very polar molecule like a muscle relaxant, then the concentration found in the fluid will be unchanged – as if the lipid was not present.
With our formula we can determine the volume of distribution:
Vd = dose/concentration
In scenario 1, non-lipid soluble drug:
Vd = 1000 mg/200 mg/L = 5 L
Scenario 2
If, however, the drug is very lipid soluble e.g. propofol, then a significant amount of the initial dose will be removed from the plasma/fluid (‘V’) and stored within the lipid. This results in a significantly lower plasma/fluid concentration of drug.
With our formula we can determine the volume of distribution:
Vd = dose/concentration
In scenario 2, lipid soluble drug:
Vd = 1000 mg/L mg/L = 1000 L
Clearly, the volume of fluid in the body/container is nowhere near 1000 L, but it allows us to make assumptions about the distribution of a drug.
Just as lipid tissue can absorb a drug and cause an increase in our calculated volume of distribution, other things can also cause partitioning effects which impact on Vd.
Other examples include:
Bone absorption: tetracyclines and bisphosphonates (increases the Vd)
Thyroid sequestration: iodine (increases the Vd)
Tissue binding: Digoxin is extensively bound to NA+/K+ ATPase. (increases the Vd)
Plasma protein binding: reduces the Vd
You calculate the loading dose of a drug by:
Loading dose = Volume of distribution x Target concentration
Question: What proportion of a drug is ionised?
Drugs which are weak acids or bases exist in equilibrium between an ionised and non-ionised form
The non-ionised/non-charged form may be able to penetrate the cell membrane or barrier whilst the charged form typically cannot pass through
One good example of this is local anaesthetics (LA):
Only the ionised form of LAs is active, but it works from inside the cell
There is no transporter for LAs and so they can only enter the cell by diffusion through the cell membrane in the unionised form
Once inside the cell, a proportion of the drug can then become ionised again and act on the internal surface of the sodium channels to block them
The pKa determines the proportion of polarised:non-polarised drug at a given pH
If the pH conditions are not favourable, e.g. in infected tissue, the pH is low/acidic, then there is minimal drug in the un-ionised form, and so it cannot enter the cell to work. Clinically, this explains why local anaesthetic does not work well in infected or inflamed tissues without the addition of alkalinising agents like bicarbonate, to raise the local pH
Whilst it takes longer for drugs to reach the lipid tissues (due to lower regional blood flow), a drug which is highly lipid soluble will gradually distribute to, and accumulate in, fat stores. This is relevant when thinking about redistribution phenomena, e.g. with propofol (Fig 1).
The initial bolus dose provides a high plasma concentration which is quickly delivered to the brain due to its high regional blood flow. The large concentration gradient results in rapid diffusion across the cell membrane (as it is lipid soluble), high neuronal concentrations, and acts to produce anaesthesia.
If plasma concentrations are measured over time, they rapidly drop to relatively low concentrations.
Question: Why is this?
Question: What is the clinical implication of this?
Propofol is a lipid soluble drug and so will cross any cell membrane.
The initial high plasma concentration will result in rapid diffusion into all cell types - the location of which is dependent on regional blood flow, resulting in a fast reduction in plasma concentration as the drug moves out of the plasma into the larger volume of the total body water.
The implication is that the concentration of propofol in the neurone is now higher than that in the plasma – a reversal of the initial concentration gradient. This results in drug diffusion out of the brain and back into the plasma, a reduction in neuronal concentration and, therefore, an anaesthetic effect.
Clinically, this can be seen as a ‘window’ of anaesthesia after a single bolus of propofol. The offset of action is not due to metabolism or excretion of drug from the body, but to re-distribution out of the plasma volume, into total body water volume and lipid stores.
Question: Some situations may arise where the amount of free drug could be significantly altered due to changes affecting protein binding. Can you think of any?
lasma albumin levels are reduced, e.g. liver injury, or renal dysfunction
Significant reduction in the overall number of available binding sites. If available binding sites are saturated then the amount of unbound drug in the plasma will rapidly rise
Competitive binding at the same site between two drugs
One drug (X) may competitively displace another (Y) from the protein binding site, resulting in a shift in the equilibrium of bound:unbound drug
This would cause an increase in plasma and tissue concentration of drug Y
Because there are fewer binding sites available to X, its plasma and tissue concentrations will also be higher than if Y were not present
So far in the models discussed we have seen that when a drug is sequestered into or soluble in extravascular structures such as lipid or bone, then the plasma concentration falls and therefore the Vd rises.
With intravascular protein binding the opposite is true. Because the drug is being held in the intravascular compartment by the plasma protein, plasma samples will show a higher concentration than if the drug was evenly distributed through the body. This means that the volume of distribution will be lower than expected.
Digoxin
Digoxin = 500 L – this molecule is extensively bound by cellular surface protein and so has a Vd significantly greater that total body water.
Warfarin
Warfarin = 8 L – note that this is smaller than ECF volume, but larger than the plasma volume, which must be due to the effect of intravascular sequestration.
Gentamicin
Gentamicin = 18 L – close to the ECF volume, which agrees with the fact that it is a small, highly charged molecule which does not cross cell membranes easily, but should distribute through the ECF.
Ethanol
Ethanol = 30-40 L – this is a non-polar water soluble small molecule that can distribute through the Total Body Water, crossing both the vascular endothelium and the cell membrane. It does not bind significantly to plasma proteins and has a Vd of around 40 L – similar to Total Body Water. This also implies that there is minimal tissue protein binding or sequestration.
Regarding SBA - volumes of distribution Qn:
A. A lipid soluble drugs will have a large volume of distribution
B. A highly protein bound drug will have a small volume of distribution
C. The volume of distribution can be many times the total volume of the body
D. The units of Vd are L/kg
E. Can be used to calculate the loading dose of a drug
A. True.
B. False. It depends on whether the drug binds to plasma proteins of tissue proteins. If the drug is highly bound to plasma protein, then the Vd will be very low as the concentration of drug in plama will be far higher than if the drug had distribute evenly through all compartments. If the drug is bound to tissue proteins (like receptors) then the Vd will be high – because most of the drug is held outside of the vascular space so plasma concentration would be lower than if the drug was distributed evenly through all compartments.
C. True.
D. True.
E. True.
When discussing body compartments:
A. The significant compartments are: Intracellular, transcellular, and intravascular compartments
B. An adult is roughly 60% water - or 42 L
C. The extracellular fluid volume is made up of the interstitial fluid, plasma volume and transcellular fluid volume
D. The vascular endothelium acts as a functional barrier between intracellular and extracellular fluid
E. A small polar molecule will be able to enter to all fluid compartments
A. False. Typically we are discussing the intracellular, interstitial and intravascular compartments. The transcellular compartment makes up an insignificant volume of the TBW.
B. True. TBW is roughly 60% of an adults mass. Though this percentage changes with age and sex.
C. True. Remember that a significant proportion of fluid within the intravascular space is actually intracellular fluid held within the RBCs.
D. False. The vascular endothelium separates intravascular fluid from interstitial fluid. The cell membrane separates intracellular from extracellular.
E. False. Small molecules can cross the vascular endothelium, but because it is charged it cannot transfer across the cell membrane therefore will only distribute through the extracellular fluid volume.
The body can be divided into a number of different fluid filled compartments, i.e. different fluid spaces which exist physiologically, separated from each other by functional barriers to the free movement of solute within the fluid.
The adult body, by mass, is 60% water, i.e. the Total Body Water of a 70 kg adult is 42 L.
The barriers to free distribution are:
Vascular endothelium
Variable depending on location, but generally blocks large molecules and cells, e.g. red blood cells, heparin or large proteins like albumin, from leaving the plasma/intravascular volume.
Cell membrane
A lipid bi-layer with multiple embedded protein structures, like receptors, channels and transport proteins.
It will allow the passage of lipid soluble molecules, e.g. thyroxine, which simply dissolve directly through the lipid membrane, or particles with specific transport carrier proteins, like glucose. However, it will mostly block the free movement of charged particles from entering the cells, e.g. sodium, unless specific channels, or transport systems, are present.
Protein binding:
A. Frequently involves strong bonds forming between drug and plasma protein
B. Can occur in the plasma, or in the tissues
C. Is a non-saturable process
D. A drug which is highly protein bound may show a clinically significant reduction in plasma concentration if a second highly plasma protein bound drug is administered
E. The main plasma proteins involved with drug binding are albumin and alpha1-acid-glycoprotein
A. False. It is a reversible process which typically involves the formation of weak bonds including Van der Waals, hydrogen and ionic bonds.
B. True.
C. False. There is a finite amount of protein and therefore number of binding sites. The system can be saturated.
D. False. Even with highly plasma protein bound drugs, it is uncommon for clinically significant changes in plasma concentration to occur due to protein binding alone. In any case, the second drug would displace the initial drug from plasma protein and raise, not lower, the plasma concentration.
E. True.
If drugs are extensively bound by protein in the plasma, then the amount of free drug is lower than it would be if it were unbound.
This, in turn means that there is a lower concentration gradient for diffusion into tissues, and so there will be slower distribution of drug.
SBA – if a dose of 8 milligrams of drug X is administered IV and the Vd of the drug is 50 L, what is the plasma concentration?
A. 16 micrograms/L
B. 1.6 micrograms/mL
C. 160 micrograms/L
D. 1.6 milligrams/L
E. 160 micrograms/mL
A. Incorrect.
B. Incorrect.
C. Correct. Concentration = mass of dose/Vd.
D. Incorrect.
E. Incorrect.
Give an overview of the Pharmacokinetics of inhalational drug administration, and the relevance to anaesthesia.
explain how the physical and chemical properties of inhalation agents influence the pharmacokinetics of these drugs
describe the pharmacokinetics of inhaled anaesthetic agents during onset, maintenance, and offset of anaesthesia
describe how the patient and equipment can affect the pharmacokinetics, including onset, maintenance and offset of anaesthesia
compare the anaesthetic inhaled drugs and draw the wash-in and the wash-out curves
explain the concentration and second gas effect
The pharmacokinetic properties of inhaled anaesthetic agents are determined by their physicochemical properties.
Speed of onset and offset of inhaled anaesthesia is predominantly determined by the blood:gas partition coefficient.
The physiological factors that increase speed of onset are increased minute ventilation and a low(er) cardiac output.
Nitrous oxide is unique in clinical practice because we can use it in sufficient concentrations to utilise the concentration and second gas effects to increase the speed of onset of anaesthesia.
Duration of inhaled anaesthesia does not affect the half-time of the drugs but does affect the 90% decrement time, which clinically slows wake-up time in long duration anaesthetics.
Most of an inhaled anaesthetic drug is exhaled unchanged, but a small proportion of the drug is oxidised in the liver by the cytochrome P450 CYP2E1 enzyme.
Inhaled anaesthetic drugs are greenhouse gases that contribute to global warming and ozone destruction and we have a responsibility to minimise the anaesthetic impact on climate change .
Pharmacokinetics is the study of the way in which the body handles administered drugs. It describes drug absorption, distribution, metabolism, and excretion.
Absorption
Inhalational drugs used in anaesthesia are said to exist in at least three compartments: alveoli, blood, and brain.
The uptake from the alveoli, across the alveolar membrane, to the arterial blood is the absorption of the drug and is highly affected by the physicochemical properties of the anaesthetic agent. Wash-in curves allow comparison of the onset of each of the anaesthetic agents.
Distribution
Once the drug enters the arterial circulation the distribution of the drug commences, to the effect site of the brain. Both the physicochemical properties of the anaesthetic drugs and the physiological parameters of the patient affect this pharmacokinetic component.
Metabolism
Metabolism is discussed later in the session but has limited effect on the offset of general anaesthesia of inhaled anaesthetic agents and is determined by the chemical structure of the drug.
Excretion
Excretion of the drug is demonstrated using wash-out curves and varies depending on the physicochemical properties of the drug and the physiology of the patient.
The chemical structures of the inhaled anaesthetic drugs determine their physicochemical properties; and the physical and chemical properties have a major effect on their kinetics.
Question: What specific properties are used when describing the kinetics of inhaled anaesthetic agents?
Properties include:
solubility: lipid for potency; water (blood) for speed of onset
partition coefficients: potency and speed of onset
chemical structure: degree of metabolism
All volatile anaesthetic agents are ethers, except for halothane - a halogenated hydrocarbon, that you are unlikely to see used in 21st century clinical practice.
Ethers are organic compounds characterised by an oxygen atom bonded by two alkyl (or aryl) groups (Fig 1).
They are large molecules that are less lipid soluble than halothane, which means halothane is more potent than all newer agents in clinical practice.
They are also unable to form hydrogen bonds, due to the lack of polarised O-H bond, and are therefore less water (blood) soluble. They can however form hydrogen bonds with other molecules using their nonbonding electron pairs in the oxygen atoms.
Changes in the chemical structure of the drugs alter the inhaled agents’ solubility, and the extent of metabolism.
Question: Can you think of some examples of these changes?
Substitutions in the chemical formula
Lowest blood:gas partition coefficient (lowest blood solubility) because fluorine is the only substitution for the hydrogen molecules in the hydrocarbon groups.
This single atom substitute increases desflurane resistance to metabolism and increases its volatility due to having the lowest boiling point of the inhaled anaesthetic agents.
Isomerism
Isoflurane and enflurane are isomers: same molecular formula, different arrangement of atoms in space.
The altered position of fluorine atoms between the molecules make isoflurane less water (blood) soluble, more lipid soluble, and less susceptible to metabolism than enflurane.
Potency is a measure of drug activity and expresses the amount of drug required to produce an effect. Highly potent drugs require small doses to produce their effect.
Potency of inhaled anaesthetic drugs is reflected in the minimum alveolar concentration (MAC) of each drug: the more potent the drug the lower the MAC.
Question: What is the definition of MAC?
MAC is defined as the minimum alveolar concentration of an inhaled anaesthetic drug at steady-state that prevents reactive movement to a standard surgical stimulus (skin incision) in 50% of non-premedicated subjects at 1 atmosphere.
In usual practice inhaled anaesthetic agents are given at 1 atmosphere and indexing MAC to atmospheric pressure may be forgotten and lead to the assumption that concentration is the key measurement, but partial pressure is the key measure of inhaled anaesthetic drugs. However, atmospheric pressure is approximately 100kPa and so when partial pressure is measured in kPa it is virtually the same as concentration, and these terms are often used interchangeably.
MAC is specific to each anaesthetic drug and is additive when drugs are used concurrently. This can be used deliberately in clinical practice, for example, sevoflurane and nitrous oxide mixture is used in obstetric general anaesthesia to reduce the dose of sevoflurane and its effect on uterine tone.
MAC correlates with lipid solubility: the more lipid soluble the drug the lower the MAC; therefore, the more lipid soluble the more potent the drug.
A potent drug does not necessarily have a fast onset of action.
Question: State the MAC of isoflurane and desflurane and describe what this means for each drug’s solubility and potency.
Isoflurane
MAC = 1.2, high lipid solubility, more potent, slower onset of action and slow to reach compartment equilibrium.
Desflurane
MAC = 6.6, low lipid solubility, less potent, faster onset of action and quick to reach compartment equilibrium.
What is a partition coefficient?
A partition coefficient is the ratio of the amount of substance present in one phase compared with another, where the two phases are of equal volume and exist in equilibrium; the temperature must also be specified.
In anaesthesia this means that the partition coefficient describes the relative solubility of a single substance (inhaled anaesthetic drug) in two different materials (lipid and water [blood]) relative to gas. It is expressed as a dimensionless number, as a ratio to 1, and allows comparison of inhaled anaesthetic drugs.
The two partition coefficients important in determining volatile anaesthetic drug behaviour are the oil:gas partition coefficient and blood:gas partition coefficient (Fig 1).
The oil:gas partition coefficient describes the relative solubility of an inhaled anaesthetic agent in lipid. This determines the potency and the MAC of the drug.
The blood:gas partition coefficient describes the relative solubility of an inhaled anaesthetic agent in blood (water). This determines the speed of onset and offset of anaesthesia.
Table 1 details the partition coefficients of inhaled anaesthetic drugs.
What physiological and pharmacological factors affect the MAC?
Inhalational drugs used in anaesthesia exist in three ‘compartments’ of the body in which they exert a partial pressure (Table 1).
Question: What is partial pressure?
Partial pressure is defined by Dalton’s law of partial pressures: in a mixture of gases the pressure exerted by each component gas is the same as that which it would exert if it alone occupied the container.
The inhaled anaesthetic drugs produce anaesthesia in the central nervous system (CNS) when their corresponding partial pressure is reached.
The partial pressure of the anaesthetic drug equilibrates between the alveoli, arterial blood, and the CNS over time, and we use the partial pressure measured in the alveoli as a surrogate for the partial pressure in the brain, i.e. the effect site.
The partial pressure required to produce the appropriate anaesthesia is the MAC and is specific to each drug (Table 2).
A highly potent drug needs a small dose to exert its effect, and for anaesthetic drugs to exert their effect they must cross the blood-brain barrier (BBB) - a lipid bilayer.
The inhaled anaesthetic drugs of higher potency are also highly lipid soluble, which allows them to cross the BBB and produce anaesthesia even with a small dose. The small dose required means their minimum alveolar concentration (MAC), and therefore minimum brain concentration, is low.
MAC decreases with increased potency.
High lipid solubility is described by the oil:gas partition coefficient and the anaesthetic drugs with a high coefficient have a high potency because they are highly lipid soluble. For example, isoflurane.
The Meyer-Overton hypothesis recognised the relationship between potency and oil:gas partition coefficient and demonstrated that the correlation between potency and oil:gas partition coefficient of an inhaled anaesthetic drugs was directly proportionate.
Fig 1 shows the correlation between MAC and the oil:gas partition coefficient, in logarithm scales.
Question: Will a higher potency cause a quicker onset of anaesthesia?
A high potency does not cause a quicker onset of anaesthesia.
The speed of onset relies on a quick build-up of anaesthetic drug partial pressure.
A highly lipid soluble drug is absorbed easily and takes longer to establish this partial pressure, therefore inhaled anaesthetic drugs with a high potency (low MAC) and high lipid solubility correlating high oil:gas partition coefficient have a slower onset of anaesthesia.
We can use the FA/FI ratio rates to compare anaesthetic drugs. These are known as wash-in curves.
Each agent has its own wash-in curve (Fig 1).
Question: What does the steepness of the graph indicate?
Question: From the graph which volatile agent has (a) the fastest and (b) the slowest onset? (excluding nitrous oxide (N2O))
The rate at which the curves approach an FA/FI ratio of 1.0, which indicates how quickly the drug reaches equilibrium between inspired concentration and alveolar concentration, and therefore how quickly it produces anaesthesia.
Note that it takes up to 6 hours (or more) to reach equilibrium.
(a) Fastest: desflurane (b) Slowest: halothane.
Fig 1 shows that the curve for nitrous oxide is steeper than the curve of desflurane despite desflurane’s lower blood:gas partition coefficient. This is because the rate of rise of FA/FI in N2O depends more on its inspired concentration (Fig 2).
Nitrous oxide is the only inhaled anaesthetic drug that can be used in such different inspired concentrations to demonstrate such considerable differences in rate of rise in FA/FI, and therefore affect onset. It also demonstrates the concentration effect and the second gas effect, which are explained on the next few pages.
Inhaled anaesthetic drugs with a low lipid solubility also have a low blood solubility. This means they have a low blood:gas partition coefficient. It also means they have a higher MAC, due to their lower potency and lipid solubility.
When anaesthetic drugs have a low blood:gas partition coefficient they can increase their partial pressure quicker, equilibrate the FA/FI quicker, and generate a partial pressure gradient.
These partial pressure gradients are created between the different compartments of the body (alveoli, arterial blood, and brain) and the lower the blood:gas partition coefficient of the drug the quicker the increase in partial pressure gradient and the greater the pressure gradient generated between compartments.
The wash-in curves previously described demonstrate the blood:gas partition coefficient effect on accumulation of the alveolar partial pressure. The concept of the wash-in curve can be applied to the different compartments of the body as the anaesthetic drug moves from high partial pressure area, such as in the arterial blood, down its gradient to the low partial pressure area, such as the brain, until equilibrium is achieved.
Fig 1 represents two anaesthetic drugs and their blood:gas partition coefficients.
Question: Which of the two volatile agents (blue or red drug) will have a faster onset of action?
The blue drug: it has a lower blood:gas partition coefficient so alveolar concentration will reach equilibrium quicker, creating a greater partial pressure gradient between blood and alveolar gas and leading to more rapid equilibration in the brain.
What is the concentration and second gas effect?
Nitrous oxide is unique in its use because we can use it at significantly higher concentrations clinically to demonstrate the concentration effect and the second gas effect, which rely on the relative increased solubility of nitrous oxide compared with oxygen and nitrogen. The linked descriptions of these demonstrate how they alter the pharmacokinetics and how this impacts on the onset of anaesthesia.
The concentration and second gas effects are phenomena only seen with nitrous oxide because the other inhaled anaesthetic drugs are unable to be used at high enough concentrations to demonstrate the same effects.
The concentration effect
Despite both N20 and N2 being relatively insoluble in blood, N20 is about 20-30 times more soluble in blood than N2 (despite its low blood:gas partition coefficient).
During induction of anaesthesia using high concentrations of N2O the initial transfer across the alveolar membrane and absorption in the blood is high. This occurs while a significantly smaller volume of N2 enters the alveoli from the blood, causing a reduction in total volume of the alveolus.
Decreasing the alveolar volume leads to a concentration of the remaining gases in the alveolus and an increase in their partial pressures and may include an increased partial pressure of other inhaled anaesthetic drugs. This is known as the concentration effect.
The second gas effect
The diminished volume of the alveoli from the concentration effect is replenished by gases from the dead space, which contains and delivers more anaesthetic drug to the alveoli.
The increased alveolar partial pressure of the inhaled anaesthetic drug(s), from both the concentration effect and the increased delivery from the dead space, leads to a higher FA/Fi ratio and induces a faster onset of anaesthesia.
This is the second gas effect.
We can take advantage of the second gas effect when performing a gas induction using O2, N2O and a volatile anaesthetic drug: the presence of nitrous oxide significantly increases the speed of induction.
There are several physiological parameters that affect the uptake and onset of inhaled anaesthetic drugs.
Question: What two physiological factors have the most influence on the onset time of inhaled anaesthetic drugs?
The two most influential physiological factors are:
minute ventilation (MV)
cardiac output
Other physiological factors that influence the onset of anaesthesia are functional residual capacity (FRC) and cerebral blood flow (CBF).
FRC is the lung volume remaining at the end of expiration when breathing normally (Fig 1). (FRC = ERV + RV)
A capacity is the sum of two or more lung volumes, and the FRC is the sum of the expiratory reserve volume (ERV) and the residual volume (RV).
The FRC acts as an oxygen reservoir when we pre-oxygenate the patient before the induction of anaesthesia, particularly important during a rapid sequence induction of anaesthesia.
FRC is altered in many clinical circumstances.
Question: Can you name at least two factors that increase FRC and two factors that decrease FRC?
Question: What is the effect of a high FRC on the speed of onset of the inhaled anaesthetic agent?
A high FRC is a high volume of gas to dilute the inhaled anaesthetic agent. This therefore dilutes the alveolar concentration (lowers the alveolar partial pressure) of anaesthetic drug and may slow the onset of anaesthesia.
A low FRC will dilute the anaesthetic gas less and increase the alveolar concentration of drug, increasing the partial pressure gradient, and increasing the speed of onset of anaesthesia.
Choosing an appropriate MV will help offset the dilutional effect of the FRC.
A lower cardiac output is associated with a faster onset of inhaled anaesthesia, and therefore a higher cardiac output is associated with a slower onset.
This is because a lower cardiac output causes a slower removal of anaesthetic drug from the alveoli and permits the alveolar partial pressure of the drug to increase quicker: the fraction of alveolar concentration (FA) equilibrates quickly with the fraction of inspired anaesthetic drug (FI).
The increased alveolar concentration maintains a larger partial pressure gradient between the alveoli and the blood, and between the blood and the brain, creating a faster rise in brain partial pressure down a steeper gradient, and increasing the speed of onset of anaesthesia.
Question: What is the other reason low cardiac output is associated with an increased onset time of anaesthesia?
Blood flow to the brain is well preserved in low cardiac output states. As proportionately more blood will flow to the brain it results in a proportionately higher flow of anaesthetic-containing blood, increasing the onset of anaesthesia.
Increases in CBF will speed onset of anaesthesia, as more agent is delivered to the brain per unit time.
CBF is autoregulated and tends to be well maintained with even moderate falls in blood pressure and cardiac output as discussed above but can also be manipulated if required.
Question: Can you name a respiratory factor that can influence CBF and alter onset-time for anaesthesia
Hyperventilation or hypoventilation.
CO2 is a cerebral vasodilator.
Hyperventilation reduces carbon dioxide tension and reduces CBF through vasoconstriction, thus slowing onset time.
Hypoventilation increases CO2 tension, causing vasodilation, therefore increasing CBF and speeding onset time.
Volatile agents usually cause cerebral vasodilatation, resulting in increased CBF. This may aid onset of anaesthesia but can also increase intracranial pressure (ICP). They should, therefore, be used judiciously in patients with known or suspected increased ICP but are normally safe at standard clinical doses with careful maintenance of blood pressure and ventilation to avoid hypercarbia.
Wash-out curves are drawn to show the progressive decline in the alveolar concentration of inhaled anaesthetic drugs over time that occurs after the vaporiser has been switched off.
Mathematically washout curves exhibit a negative exponential process. This means the rate of change in the y-axis decreases as the x-axis increases, and is the same mathematical process as the wash-in curves because even though the actual value on the y-axis is increasing in the wash-in curve the rate of change is decreasing.
Question: Why is the y-axis description in the wash-out graph (FA/FAE) not the same as the wash-in graph (FA/FI)?
After the vaporiser has been switched off FI (inspired concentration of inhaled anaesthetic agent) is zero so FA/FI, i.e. FA/0, is infinite, or undefined. The y-axis can be plotted as alveolar partial pressure (usually % concentration) against time but for a comparison of drugs we use FA/FAE, where FAE is the partial pressure in the lungs when the vaporiser is turned off.
In practice the gas reaching the alveoli from the inspiratory limb of breathing circuit may still contain some anaesthetic drug, i.e. a small FI, and is most pronounced when using a circle breathing circuit. Increased fresh gas flows should be used at the end of the anaesthetic to minimise the effects of the any residual anaesthetic agent, especially when using a circle system.
Inhalational anaesthetic drugs are distributed in a multi-compartment model and therefore could be considered able to be described in similar pharmacokinetic principles as the intravenous maintenance drugs: where multi-compartment models are subject to context-sensitive half-time.
Question: What is context-sensitive half-time?
Context-sensitive half-time (CSHT) is defined as the time taken for the plasma concentration of a drug to fall by half after the cessation of an infusion designed to maintain a steady plasma concentration, where context refers to the duration of infusion.
In clinical practice the time taken for the inhaled anaesthetic drugs to fall by half (50% decrement time) varies little (~5 min) between the drugs, regardless of duration. Therefore, CSHT offers a poor comparison of these drugs.
Wake-up occurs at much lower concentrations of inhaled drug, much less than at 50% decrement in alveolar concentration, towards 90% decrement, and there is a big difference in 90% decrement times between the inhaled anaesthetic drugs, particularly with increasing duration of anaesthesia. This means the context is sensitive for 90% decrement-time, rather than half-time.
The differences in 90% decrement time are a result of the drug’s physicochemical properties
Desflurane has the shortest decrement time (Fig 1) due to its low lipid solubility (oil:gas partition coefficient) and low blood:gas partition coefficient.
The low coefficients allow the desflurane effect site concentration to reduce by 90% quickly because it doesn’t saturate the additional lipid tissues (no additional ‘lipid’ compartment to a multi compartment model) and doesn’t generate a drug reservoir to alter the partial pressure gradient between the brain and blood.
A patient is anaesthetised for day case hysteroscopy. Intravenous induction with propofol is chosen followed by sevoflurane maintenance via a laryngeal mask.
Ten minutes after induction the MAC on the machine reads 1.2 but the patient moves at the start of the operation.
Question: Why did the patient move when the MAC is 1.2?
What strategies can be used to overcome this problem in clinical practice?
The end tidal concentration of sevoflurane is measured by the anaesthetic machine, and the MAC is calculated based on the end tidal concentration - a surrogate measure of alveolar concentration.
The anaesthetic effect of the inhaled anaesthetic drug relies on the concentration (or partial pressure) of the drug at the effect site, i.e. the brain.
The equilibrium of concentration of anaesthetic drug between alveoli, arterial blood and brain takes time, different for each anaesthetic drug, and the patient moved because the concentration of sevoflurane in her brain was too low.
Setting a higher inspired anaesthetic drug concentration (on the vaporiser dial) than is required for anaesthesia, i.e. dialling higher than the MAC for that anaesthetic drug. This is known as over-pressuring.
Deliberately setting a higher inspired concentration of inhaled anaesthetic drug will increase the partial pressure in the alveoli and increase the concentration gradient between the alveoli and the arterial blood, and therefore increase the gradient between the blood and the brain. This allows the CNS concentration of anaesthetic drug to increase more quickly, and therefore produce anaesthesia at the effect site quicker as it reaches the MAC of anaesthetic drug sooner.
When using inhaled anaesthetic drugs:
A. Measured end-tidal alveolar concentration is a good surrogate for brain concentration at the beginning of the case
B. Desflurane is a good drug for inhalational induction of anaesthesia because of its speed of onset
C. Recovery from anaesthesia is faster with sevoflurane than isoflurane
D. Desflurane is extensively metabolised
E. When the vaporiser is dialled to 4% sevoflurane it means that exactly 4% sevoflurane is delivered to the patient using a circle system
A. False. End-tidal concentration is the same as brain concentration at equilibrium, which takes time to achieve. Therefore it is not a good surrogate at the beginning of the case.
B. False. Desflurane does have a quick onset but is too pungent and irritant for awake patients to breathe.
C. True. Sevoflurane is less soluble in blood than isoflurane (low blood:gas partition coefficient), therefore it has a faster offset as well as onset.
D. False. Desflurane is 0.02% metabolised.
E. False. Vaporiser output is diluted by gas already in the breathing system, and monitoring of inspired and end-tidal concentration of volatile agent is mandatory and allows adjustments of vaporiser concentration as required.
Minute ventilation:
Increasing alveolar ventilation and FGF during offset increases the removal of drug from the lungs.
The increased drug removal from a higher respiratory rate (or tidal volume) increases the partial pressure gradient between the (pulmonary arterial) blood and alveolus, and speeds transfer to the lungs for removal.
The higher FGF ensures low to no residual anaesthetic drug remains in the inspiratory arm of the circuit and prevents it being re-delivered to the alveoli to reduce the pressure gradient.
If the PCO2 becomes low because of the increased minute volume it can cause cerebral vasoconstriction. This can delay the offset of anaesthesia by reducing blood flow to the brain, therefore reducing the transfer of drug from the brain to the blood.
A low PCO2 may also delay the return of spontaneous breathing because PCO2 is a trigger for breathing via the central chemoreceptors in the medulla. If reliant on spontaneous ventilation for anaesthetic drug removal this will delay offset of anaesthesia.
Other drugs commonly used in anaesthesia affect spontaneous ventilation, for example opioid analgesic drugs reducing respiratory rate, may also slow patient wake-up by delaying offset of anaesthesia through low minute ventilation.
Rank the following according to which agent reaches equilibrium between alveolar and inspired concentrations most rapidly.
Nitrous oxide reaches equilibrium more quickly at high concentrations because of the concentration effect.
Regarding the chemical properties and metabolism of inhaled anaesthetic drugs:
A. The ethers are more lipid soluble than hydrocarbons
B. Only nitrous oxide is used at a high enough concentration to produce the concentration effect
C. Sevoflurane is metabolised to trifluoroacetic acid
D. The blood:gas partition coefficient of desflurane is lower than the coefficient of nitrous oxide
E. The solubility of nitrous oxide in blood is lower than the solubility of nitrogen
A. False. The ethers are less lipid soluble than halothane, a halogenated hydrocarbon; halothane is the most potent volatile agent in use and has the highest oil:gas partition coefficient.
B. True.
C. False. This is the only anaesthetic drug that is not metabolised to trifluoroacetic acid; it is metabolised to hexafluoroiso-propanol. It also forms the degradation product Compound A when used with CO2 absorbers.
D. True.
E. False. Nitrous oxide is more soluble than nitrogen in blood and this property helps explain the alveolar concentration effect that occurs during induction when a volatile agent is administered with nitrous oxide and oxygen.
Regarding inhaled anaesthetic drugs:
A. MAC correlates with drug solubility in blood
B. Partition coefficients have no units
C. A more potent drug has a quicker onset of action
D. Halothane is the most potent of the halogenated inhaled anaesthetic drugs
E. Inhaled anaesthetic drugs have a constant 90% decrement time
A. False. MAC is a measure of potency and correlates with lipid solubility (and therefore oil:gas partition coefficient).
B. True. They are a ratio of two solubilities, therefore have no units.
C. False. A potent drug has a high lipid solubility and requires a small dose to cause its effect, but in inhaled anaesthetic drugs it takes a potent drug a longer time to produce its anaesthetic effect. Drugs with a quicker onset of action have a low blood solubility.
D. True. It has the lowest MAC of the drugs discussed but is not used in clinical practice in the UK. The most potent drug in current use in the UK is isoflurane.
E. False. The 90% decrement time varies between agents but is longer for those with both a higher blood:gas partition coefficient and oil:gas partition coefficient.
The following physiological changes slow the onset of anaesthesia using inhaled anaesthetic drugs:
A. Term pregnancy
B. Reduced alveolar ventilation
C. Reduced cardiac output
D. Emphysema
E. Hypovolaemic shock
A. False. There are several physiological changes in pregnancy: hyperventilation and reduced FRC will favour a faster onset, but an increased cardiac output would tend to oppose this. Overall the respiratory effects predominantly increase the speed of onset of anaesthesia.. but an inhalational induction at term pregnancty is generally contraindicated because of aspiration risk.
B. True. This reduces delivery of the agent to the alveoli.
C. False. Low cardiac output results in increased alveolar partial pressure of agent, therefore faster induction. Cerebral autoregulation preserves blood flow to the brain, so a higher proportion of cardiac output goes to the brain.
D. True. Emphysema increases the FRC volume and dilutes the agent in the alveoli.
E. False. This causes a low cardiac output.
Give an overview of Pharmacokinetics: Elimination, and the relevance to anaesthetics.
Describe the pathways of biotransformation
Explain factors influencing drug biotransformation
Describe different routes of drug excretion
Describe the dynamics of drug clearance
Know the difference between zero and first order kinetics
Drug elimination involves the processes of drug metabolism and excretion and is carried out mainly by the liver and kidneys including other organs.
Biotransformation of drugs is either by phase 1 and/or phase 2 metabolism leading to the excretion of the drug. Liver is the major organ for drug metabolism but there are certain drugs which undergo organ independent metabolism like Hoffman’s degradation for atracurium and cisatracurium, esterases metabolism for suxamethonium and remifentanil.
Excretion mainly occurs through kidney via glomerular filtration, active tubular secretion and tubular reabsorption.
Clearance is the volume of the plasma which is cleared of the drug per unit time and is related to the elimination rate constant (Ke).
Drug clearance occurs either via first order or zero order kinetics.
Drugs which are eliminated via first order kinetics, a constant fraction of the drug is eliminated per unit time. Most of the drugs follow first order kinetics.
Drugs following zero order kinetics have a constant amount of drug which is eliminated per unit time, for example Thiopentone, phenytoin.
Michaelis-Menten kinetics involves the interaction between the enzymes and the drug where the velocity of the reaction (v) increases as the drug/substrate concentration (S) increases until all the enzymes are occupied by the drug causing saturation and reaching the maximal rate of reaction. This is then followed by conversion of the first order kinetics to zero order kinetics where V = Vmax, i.e. the velocity of the reaction does not increase with increase in substrate concentration 3.
Regarding oxidation reactions:
A. A substrate gains electrons from an oxidation reaction
B. It is the most common Phase 1 reaction
C. Dehydrogenation is a type of oxidation reaction
D. Thiopentone is metabolised by oxidation
A. False. Oxidation is loss of electrons while reduction is gain of electrons.
B. True.
C. True. Oxidation reactions involve oxygenation or dehydrogenation or electron transfer.
D. True. Thiopentone is metabolised in the liver by oxidation to thiopental carboxylic acid, hydroxythiopental and pentobarbital.
Phase 1 metabolism is characterised as a functionalisation reaction, either adding or unmasking a functional group (-OH, -SH, -NH2) on the parent drug molecule on which phase 2 reactions can occur (Fig 1). The three types of reactions are:
oxidation (most common)
reduction
hydrolysis
Phase 1 metabolism can convert the drug to either an active or an inactive metabolite. However, phase 1 metabolism usually converts the drug into intermediate metabolites, which may be highly reactive and toxic.
Oxidation involves loss of electrons or gain of oxygen (removal of hydrogen).
Phase 1 metabolism is usually mediated by microsomal cytochrome p450 mono-oxygenases with the CYTP3A4 being the most common (responsible for metabolism of > 60% of the drugs).
Barbiturates, benzodiazepines, paracetamol, ropivacaine and omeprazole are some drugs metabolised by oxidation. Halothane undergoes mainly oxidative metabolism.
Which of the following enzymes are involved in Phase 1 reactions?
A. Monoamine oxidases
B. Glucuronyl transferases
C. Esterases
D. Monooxygenases
A. True.
B. False. Glucuronyl transferase catalyses the attachment of a sugar moiety to a functional polar group and is a type of phase 2 reaction (conjugation).
C. True.
D. True.
Reduction involves the gain of electrons and is the converse of oxidation. It can be mediated by CYT P450 reductase and usually takes place under anaerobic conditions, such as the reduction of prodrug prednisone to active drug prednisolone.
Halothane may also be metabolised by reduction reactions to more toxic metabolites, especially when the liver is hypoxic (Fig 1).
Hydrolysis involves the cleavage of drug molecule using a molecule of water, and catalysed by esterases and amidases. It usually occurs in liver, plasma, intestines and other tissues, for example hydrolysis of amides and esters (lidocaine, prilocaine, meperidine, oxytocin) (Fig 1).
There are some non-P450 phase 1 reactions:
Mitochondrial enzyme monoamine oxidase is involved in metabolism of monoamines like adrenaline, noradrenaline and dopamine.
Esterases metabolise esters like aspirin, remifentanil, etomidate and atracurium in liver and muscles.
Alcohol dehydrogenase present in cytoplasm metabolises ethanol to acetaldehyde and further to acetic acid.
Angiotensin-converting enzyme in the lung is responsible for conversion of AT1 to AT2.
Hoffmann’s degradation is a pH- and temperature-dependent process in plasma that breaks down atracurium and cisatracurium.
Both of these reactions are phase 1 reactions.
Which of the following are Phase 1 reactions?
A. Conjugation to alcohols
B. Oxidation of monoamines
C. Ester hydrolysis
D. Acetylation
A. False.
B. True.
C. True.
D. False.
Phase 1 reactions involve oxidation, reduction and hydrolysis.
Regarding phase 1 reactions:
A. They involve the addition of a polar molecule to a functional group already present on the drug, or its metabolite
B. Sulfation is an example of phase 1 reaction
C. They involve the addition of a polar group to the drug
D. All inhalational agents are metabolised by phase 1 reactions
E. Most phase 1 reactions inactivate the drug
F. They occur primarily in the gut wall and blood
A. False. Phase 1 metabolism is characterised as adding or unmasking a functional group (-OH, -SH, -NH2) on the parent drug molecule on which phase 2 reactions can occur.
B. False.
C. True.
D. False.
E. False.
F. False. Phase 1 reactions can occur in the gut wall or blood supply, but do not occur exclusively at these sites. Most of phase 1 reactions happen in the liver.
Phase 2 metabolism involves the addition of a polar molecule to a functional group placed by phase 1 metabolism. Most phase 2 reactions inactivate the drug or active metabolite formed from phase 1 reactions.
Regarding extraction ratios:
A. Hepatic clearance of a drug with high extraction ratio depends on the drug protein binding
B. The hepatic extraction ratio becomes equal to 1 (unity) when the drug that reaches the liver is completely extracted
C. When no drug is extracted by the liver, the extraction ratio is zero
D. Propofol, ketamine, etomidate and morphine all have a high extraction ratio
A. False.
B. True.
C. True.
D. True.
Hepatic clearance for drugs with high ER is independent of plasma protein binding. When hepatic extraction ratio approaches 1, clearance equals hepatic plasma flow. All intravenous induction agents have high extraction ratios.
The hepatic extraction ratio (HER) is the fraction of the drug which is removed (extracted) during one pass of the blood through the liver. It is the rate-limiting factor in first order kinetics.
HER = (Ci - Co) / Ci
Where:
Ci = Drug concentration in blood entering the organ
Co = Drug concentration in blood leaving the organ
Hepatic Clearance = Hepatic Blood Flow x Hepatic Extraction Ratio
Drugs with high HER (HER>0.7) are rapidly cleared from the blood by the liver and clearance depends mainly on hepatic blood flow (flow limited or high clearance drugs).
Drugs with low HER (<0.3) are not efficiently cleared by the liver and their clearance is dependent on the metabolising capacity of the liver and free fraction of the drug and is independent of hepatic blood flow (capacity limited or low clearance drugs).
Regarding phase 2 reactions:
A. They usually involve conjugation reactions
B. They are less likely to reach saturation (Vmax) levels than phase 1 reactions
C. They requires molecular oxygen (O2) and NADPH as cofactors
D. Phase 2 reactions are detoxification pathways
A. True.
B. False.
C. False.
D. True.
Glucuronide conjugation usually decreases toxicity. Elderly patients usually metabolise drugs via phase 2 metabolism and have decreased phase 1 metabolism
Phase 2 (synthetic phase) metabolism involves formation of a covalent linkage between a functional group on the parent compound (or on a phase 1 metabolite) with endogenously derived glucuronic acid, sulphate, glutathione, amino acids or acetate (Fig 1).
It includes:
glucuronidation (most common)
acetylation
sulphation
methylation
glutathione conjugation
Either drugs undergo phase 1 followed by phase 2 reactions, or some undergo only phase 2 reaction to metabolites, which are usually water-soluble, pharmacologically inactive and non-toxic to be excreted in urine.
Phase 2 (synthetic phase) metabolism mainly occurs in the liver, but is also found in other sites like the lungs, kidneys, GIT, red blood cells and the spleen.
The enzymes involved are transferases, which attach small polar molecules (glucuronate, glutathione, sulphate and acetate) to a drug to make it more water-soluble.
The following changes to drug metabolism occur in the elderly:
A. Reduced microsomal enzyme activity
B. Reduced hepatic blood flow
C. Decreased body fat
D. Increased protein binding
E. Increased cardiac output
A. True. There is a reduced content of phase 1 drug-metabolising enzymes.
B. True. Decreased hepatic blood flow leads to decreased clearance of high extraction ratio drugs (morphine, lidocaine) in elderly.
C. False. There is a 10-15% increase in total body fat.
D. False. There is decreased serum albumin leading to increased free fraction of albumin bound drugs.
E. False.
Regarding renal excretion:
A. Drugs which are weak acids are excreted faster in alkaline urinary pH
B. Drugs which are weak bases are excreted faster in alkaline urinary pH
C. Reabsorption occurs mainly in the proximal convoluted tubule of the nephron
D. Renal clearance of drugs is affected by drug secretion rate and presence of renal disease
A. True. Weak acids are excreted faster in alkaline urine as it promotes the dissociation of the weak acids into ions and lead to increased excretion in urine. The major driving force for glomerular filtration in glomerular capillaries is the hydrostatic pressure. Diet may have an influence on urinary pH, for example a high-protein diet results in acidic urine.
B. False.
C. True.
D. True.
The kidneys are the main route of excretion of all the water soluble drugs and this occurs via three mechanisms:
- Glomerular filtration
Glomerular filtration
Blood enters through the afferent artery and glomerular hydrostatic pressure drive the process of filtration across glomerular capillaries. Glomerular capillaries have large pores which allows free drug (non-protein bound) whether lipid soluble or insoluble to be filtered into the Bowman’s capsule and then enter the PCT. It is a passive process.
Factors affecting glomerular filtration:
Renal blood flow
Molecular weight: drugs that are low molecular weight <7,000 Daltons are readily filtered
Molecular charge: negatively charged particles are repelled by negatively charged glomerular basement membrane
Plasma protein binding: drugs like heparin which are bound to plasma proteins cannot be filtered and therefore do not get excreted by glomerular filtration. Propofol has a low molecular weight but has 98% protein binding capacity and therefore is not actively filtered
Age and renal disease: renal drug excretion decreases progressively after the age of 50 years and GFR is low in renal failure
- Tubular reabsorption
Tubular reabsorption occurs after glomerular filtration into the kidney tubules and can be active or passive, but mostly occurs via passive diffusion.
Factors affecting passive tubular reabsorption include drug concentration, lipid solubility, ionisation and urine pH.
Most of the lipophilic and non-ionized drug which was filtered at the glomerulus will be reabsorbed in the PCT. Tubular reabsorption and secretion are immature at birth and mature by the end of the first year of life. Therefore, the duration of action of many drugs is prolonged in neonates.
- Active tubular secretion
Renal excretion is the sum total of these processes.
It is responsible for the elimination of many drugs.
It is an active carrier-mediated process where drugs are transported against their concentration gradient to be secreted into urine.
This energy-dependent process is capacity limited (saturable) for each drug type, i.e. maximum clearance of one basic drug leads to reduced clearance of another basic drug, but it can be also blocked by metabolic inhibitors.
It involves two carrier systems:
Organic acidic carrier or anions for acidic drugs like penicillin, aspirin
Organic base transport or cations for basic drugs like lidocaine, dopamine, amiloride.
Renal excretion = (glomerular filtration + tubular secretion) - reabsorption.
Regarding elimination half-life:
A. Half-life is increased by an increase in the volume of distribution
B. Half-life is increased by an increase in the rate of clearance
C. In renal failure with oedema, Vd and renal clearance decreases leading to an unchanged half-life
D. It takes 4 half-lives for 87.5% of the drug to be eliminated
A. True.
B. False. t½ = 0.693*Vd/Cl. Therefore half-life is directly proportional to Vd and inversely proportional to Clearance.
C. False. In patients with renal failure with oedema Vd increases and renal clearance decreases.
D. False. It takes 3 half-lives for 87.5% of the drug to be eliminated.
Elimination half-life is the time taken for the plasma concentration of the drug to drop by 50% of its original value in a single compartment model or the time taken by the drug concentration to fall to half by processes such as redistribution, elimination, and metabolism (Fig 1).
Many drugs have an initial redistribution phase with a short half-life followed by elimination phase with a longer half-life.
A process is said to be complete after 4-5 half-lives.
The concentration drops to:
50% drug left after 1 half-life
25% drug left after 2 half-lives
12.5% drug left after 3 half-lives
And finally, after 5 half-lives 3.125% left or it takes about 5 half-lives for a drug to be roughly 97% eliminated.
Therefore it will take ~4-5 elimination half-lives of a drug for a constant-rate infusion to reach its final concentration.
t1/2 = 0.693 x (Vd / Cl)
Regarding Michaelis-Menten kinetics:
A. The substrate is the limiting factor
B. The enzyme is the limiting factor
C. If the enzyme concentration is reduced the reaction velocity at each substrate concentration will be reduced
D. Higher Km means higher substrate affinity for the enzyme
A. False. The substrate is not the limiting factor.
B. True. The enzyme concentration is negligible compared to the substrate concentration.
C. True. Velocity of the reaction or product formation is proportional to the enzyme concentration.
D. False. Km is a measure of the enzyme’s affinity for the substrate therefore low Km equates to higher substrate affinity.
The Michaelis-Menten kinetics equation relates the enzyme-mediated rate of reaction to drug concentration for non-linear drug pharmacokinetics.
It describes enzymatic reactions where 100% enzyme saturation is reached and therefore transition occurs from first order to zero order, such as with thiopentone, phenytoin or ethanol.
It has been described by the Michaelis-Menten equation where Vmax is the maximum rate of reaction and Km is the concentration required to achieve 50% of this maximum rate. It is given by the equation opposite.
The curve is rectangular hyperbola.
V = (Vmax x S) / (Km + S)
Where:
V = velocity of reaction
Vmax = the maximum rate of reaction
S = Substrate concentration
Km = substrate concentration at half max velocity
The velocity of product formation no longer depends on the substrate concentration, but depends on the enzyme present and hence V = Vmax and therefore becomes zero order.
Therefore if a substrate has high affinity for the enzyme then the Km will be lower.
Regarding zero order kinetics:
A. The rate of elimination of a drug is dependent on the initial drug concentration
B. The half-life is given by the following equation t1⁄2 = 0.693/k
C. The half-life of the drug is directly proportional to the initial concentration of the drug
D. The plot of drug concentration against time is a straight line with a gradient of 1⁄K
A. True.
B. False.
C. True.
D. False.
In zero order kinetics the rate of degradation is independent of the concentration of the drug. The half-life of first order kinetics is constant.
With zero order kinetics, the rate of elimination is constant irrespective of drug concentration, which means that a constant amount of drug is excreted per unit time.
For example, if we start with 100mg of drug in a given volume and, say, elimination is 20mg/hr, then after 1 hour we will be left with 80mg of the drug. Because the elimination rate is constant, we will be left with 60mg of the drug after another hour.
Clearance is variable in zero order kinetics because a constant amount of drug is eliminated per unit time (Fig 1) and (Fig 2).
The half-life in zero order kinetics depends on initial concentration of the drug (C0).
Consider the graph.
Question: Assuming a zero order reaction, how would you label the the x and y axis?
Question: What does the intercept on y axis correspond to?
Question: How would you determine the rate constant?
How would you plot the first order elimination kinetics on the same graph?
The y axis is drug concentration and the x axis is time.
The intercept corresponds to initial drug concentration C0.
The rate constant is given by the -ve of the slope.
In zero order kinetics, the same amount of drug is eliminated per unit time while in first order kinetics variable amount of drug is elimimated per unit time as a constant proportion or fraction of the drug is eliminated.
Regarding first order kinetics:
A. Concentration falls exponentially with time
B. Half-life is equal to 3 time constants
C. Rate constant = clearance x vol of distribution
D. Rate of elimination is inversely proportional to the concentration of the drug
A. True.
B. False. Half-life is the time taken for C to decline to half its initial value. Time constant is the time required for C to fall to 1/e of its former value. For an exponential decline C = C0e-k.t where C0 is the concentration at time t = 0 and c is the concentration at time t. Time constant is the time taken for the concentration to fall to 37% of its original value (1/e).
Therefore half-life = 0.693. In first order kinetics rate of metabolism is directly propotional to the drug concentration.
C. False. Clearance = Vol of distribution (Vd) x elimination rate constant (Ke).
D. False. Clearance = Rate of elimination/ C (drug conc).
First order kinetics can be defined as “a constant proportion or fraction of the drug is eliminated per unit time”.
The rate of elimination is not constant and is proportional to the amount of drug present in the body (plasma concentration).
We can demonstrate by the following example:
If the drug follows first order kinetics then, say, if we start with 100mg of the drug in a unit volume of 1L and the half-life is 1 hour, then after 1 hour we will have 50mg of the drug remaining.
The rate of elimination is 100-50/1 = 50mg/hr.
Because a constant fraction, or proportion, of the drug is eliminated per unit time, which in this case is 50% over 1 hour, it drops to half, i.e. 25mg from 50mg, in another 1 hour and, hence, the rate of elimination will be 50-25/1 = 25mg/hr.
Hence, we see here that as the plasma drug concentration drops the rate of elimination also drops (Fig 1) and (Fig 2).
t1/2 = ln 2 / k = 0.693 / k
Therefore the half-life of first order reaction is constant.
In the graph below please label the elimination rate constant K1, K2 and K3 if K1< K2 < K3 assuming first order kinetics.
t1⁄2 = 0.693⁄K and hence t1⁄2 ∝ 1⁄K
Therefore the lower the values of K slower is the elimination of the drug.
Following an IV bolus of a drug:
If it achieves a plasma concentration of 12mg/L after the 1st dose, then what would be the plasma level before the 5th dose, given both half-life and the dosing interval is 8 hours.
A. 11.25mg/L
B. 18mg/L
C. 21mg/L
D. 32.5mg/L
A. Correct.
After the 1st dose (8 hours), the drug concentration is going to be 6mg/L. And then adding a second dose would make it 6 + 12 = 18mg/L.
After another 8 hours drug conc is going to be 9 + 12 = 21mg/L with the 3rd dose.
It will become 10.5 + 12 = 22.5mg/L in another 8 hours with the 4th dose.
It will reduce to 11.25 in another 8 hours, before the 5th dose.
Therefore, with multiple IV dosings, drug tend to accumulate and reach steady state which is considered to be achieved when concentrations are within 10%.
B. Incorrect.
C. Incorrect.
D. Incorrect.
Fig 1 is a drug concentration versus time graph, showing the pharmacokinetics when an infusion of thiopentone is started and continued for up to 48 hours in a patient with status epilepticus. Which parts of the graph represent first order and zero order kinetics?
It is well-established that thiopental shows nonlinear (Michaelis-Menten or saturable) kinetics when administered in high doses for prolonged periods.
Initially the plasma concentration follows first order kinetics and therefore an exponential rise followed by a transition to zero order kinetics when all the enzymes are saturated and then the plasma concentration rises.
Give an overview of the Pharmacokinetics of drug metabolism and excretion, and the relevance to anaesthesia.
Pharmacokinetics describes what happens to a drug in the body. The processes which pharmacokinetics can broadly be split into are called ADME:
A - absorption
D - distribution
M - metabolism
E - excretion
The principles of A.D.M.E. are shown in Fig 1.
Pharmacodynamics describes the actions produced by the drug on the body. Therefore, the effects of a drug result from a combination of its pharmacokinetic and pharmacodynamic characteristics in that individual.
define drug metabolism
describe where metabolism occurs in the body
classify the phases of metabolism
describe the phases of metabolism
describe factors affecting metabolism
explain the clinical importance of drug metabolism with relevant examples
Drugs are in general lipophilic and poorly hydrophilic, making them difficult to excrete.
In order to make them easier to excrete, they are metabolised.
Metabolism takes place throughout the body, but the liver is the main organ of metabolism.
Metabolism is broadly split into phase 1 and 2 reactions.
The purpose of phase 1 reactions is to add functional groups which allow conjugation (phase 2) reactions to occur.
Phase 1 reactions include oxidation, reduction and hydrolysis. The most common is oxidative, catalysed by the cytochrome P450 enzyme system of the liver.
Phase 2 reactions are conjugation reactions, which involve a bond being created with the new functional site created by phase 1.
Phase 2 reactions generally create larger, more hydrophilic, more polar molecules which are excreted more readily in the urine or bile.
There are many factors, physiological and pathophysiological, affecting metabolism.
What is true regarding renal clearance of drugs?
A. It involves zero order kinetics
B. It can occur via glomerular filtration, which depends on drug protein binding and the glomerular filtration rate
C. It is not affected by carrier saturation
D. Dissociation of drug in the urine favours reabsorption
Submit
A. Incorrect.
B. Correct. Renal drug clearance is the sum of glomerular filtration and active secretion minus drug absorption. Only the free fraction of the drug is filtered in glomerular filtration and is affected by the renal blood flow, age and renal disease. Renal clearance can occur via active secretion, which is an active carrier-mediated transport process and hence saturable. Lipid-soluble drugs undergo passive diffusion, being reabsorbed into the bloodstream.
C. Incorrect.
D. Incorrect.
Question: What is drug metabolism?
Drug metabolism, or “biotransformation”, is the breakdown of drugs by living organisms, usually through specialised enzymatic systems.
Drugs can undergo one of several pathways on entry into the body:
they can change spontaneously into other compounds (for example, atracurium)
they can be excreted unchanged by the body (for example, vancomycin)
or they can be metabolised by enzymes to different compounds to facilitate excretion
The majority of drugs fall into this last pathway.
Many drugs need to be lipid-soluble in order to cross cell membranes to access their target site. Indeed, as you may have learnt in previous sessions, anaesthetic agent potency is related to lipid solubility. This, however, makes these drugs relatively insoluble in water, and therefore difficult to excrete. The overall aim of metabolism is to produce a more water-soluble compound to facilitate the excretion of the drug in body fluids such as urine and bile. Very few drugs are excreted without being metabolised.
Question: Consider if the following statement is true or false:
Metabolism of a drug always decreases its therapeutic effect.
It is false. Metabolism can increase or decrease a drugs therapeutic effect, or create metabolites with similar effect.
Metabolism of a drug generally decreases its therapeutic effect. Some drugs, however, are metabolised firstly into active intermediate metabolites before further metabolism to inactive compounds. Some drug metabolites have significant activity similar to the parent compound, for example; morphine and its metabolite morphine-6-glucuronide.
Question: What is a prodrug?
Drugs called prodrugs have no intrinsic activity before metabolism, for example; diamorphine, codeine, enalapril and prednisone. They are only active after initial metabolism into an active compound.
In biochemistry reduction refers to a chemical reaction in which the substrate gains electrons (Fig 1).
Frequently, the resulting amino compounds are oxidised which forms toxic metabolites. Some compounds can be reduced to free radicals, which are reactive with biological tissues. Reduction reactions therefore frequently result in activation of a drug rather than detoxification.
Reduction is often achieved by the addition of hydrogen to a molecule or the removal of oxygen from a molecule.
Reduction reactions are the second most common type of phase 1 reactions. They are also catalysed by the CYP450 system and often take place under anaerobic conditions. There are fewer specific reduction reactions than oxidizing reactions.
Question: Can you think of any of the specific reduction reactions?
The nature of these reactions is also described by their name. Some reducing reactions include:
azo reduction
dehalogenation
disulfide reduction
nitro reduction
n-oxide reduction
sulfoxide reduction
Match the following descriptions to the relevant part of the body.
Gut
Enzymes within the gut wall also contribute to “first-pass” metabolism after oral administration of some drugs.
Metabolism in the intestinal lumen of drugs and their metabolites (excreted via bile) can also reverse the effects of hepatic microsomal enzymes, rendering compounds less polar and therefore available for reabsorption. This leads to the establishment of “enterohepatic recycling”.
For example, the glucuronides of morphine are hydrolysed back to the parent drugs, which may then contribute to further activity after reabsorption.
Bacterial enzymes play a significant role in this process, and the anaerobic environment in which they exist encourages reductive reactions, which again tend to offset the largely oxidative changes of the CYP450.
Renal
The renal system has a well described role as a major excretory organ for drugs and their metabolites. Its involvement in drug metabolism, however, is relatively poorly understood.
There is an increasing body of evidence demonstrating that the kidney is metabolically active. The kidneys express an array of enzymes variably involved in metabolism, both CYP450 enzymes, and those involved in phase 2 reactions such as UDP-glucuronosyltransferases.
Studies have demonstrated that the maximal rate of propofol glucuronidation by renal microsomes is 3 to 4 times greater than that of liver microsomes, and unsurprisingly, total systemic clearance of propofol in humans can exceed hepatic blood flow. The kidneys have been identified as contributing almost one-third of total systemic clearance.
Similarly, studies have demonstrated that the systemic clearance of morphine in humans can exceed hepatic clearance by 38%. In the absence of evidence of gut wall metabolism, the authors concluded that the most likely site of extrahepatic metabolism is the kidney.
Lungs
The lungs are principally concerned with the uptake of oxygen, and excretion of carbon dioxide. The lungs are the major excretory organ for some compounds, notably the inhalation anaesthetics.
They are involved in the modification or uptake of many endogenous substances including serotonin, prostaglandins and noradrenaline.
The lungs also have a limited capacity for drug metabolism. The lung is particularly suited to function as a “chemical filter” as it receives 100% of cardiac output and has the largest capillary endothelial surface in the body.
Propofol and local anaesthetics can be metabolised in the lungs, but this probably does not contribute significantly to total body metabolism. Other drugs that are metabolised include budesonide, salmeterol, fluticasone, and theophylline.
Blood
The red blood cell (RBC) contains moderate CYP450-like activity, in addition to the ability to catalyse various other reactions across a range of drugs. Whilst there is no evidence demonstrating the RBC’s capacity for the more important phase II reactions (glucuronidation, sulphation), they are capable of some of the less common ones (methylation, acetylation).
Several commonly used IV agents used by anaesthetists are rapidly metabolised by enzymes found within the blood.
Suxamethonium, mivacurium, procaine and other related local anaesthetics are substrates of pseudocholinesterase, found mainly in plasma. Esmolol is metabolised by red cell esterases.
Remifentanil is rapidly degraded by red cell esterases, principally to a carboxylic acid derivative, remifentanil acid. This organ-independent elimination and predictable, rapid metabolism makes it a useful agent in the ICU setting.
Liver
The most metabolically active organ in the body, the liver is strategically placed between the port of entry for many drugs (the orogastric route) and the rest of the body. Following absorption from the GI tract extensive “first-pass” metabolism of drugs occurs here after uptake via the portal vein, significantly reducing their systemic availability. This phenomenon explains why many drugs have an oral bioavailability less than the equivalent parenteral dose.
The large size and proportion of received cardiac output also contribute to the liver’s capacity for metabolism. Most importantly, the liver has very high concentrations of drug metabolising enzymes, relative to other organs.
The smooth endoplasmic reticulum of the hepatocyte is the principal site of metabolism in the liver, with enzymes being contained within the microsomes. The cytochrome P450 system is the largest family of membrane-bound, nonspecific, mixed-function enzymes.
Non-cytochrome P450 enzymes in the liver are also involved in metabolism, including esterases and flavin-containing mono-oxygenase enzymes.
In biochemistry oxidation is a chemical reaction in which a substrate loses electrons.
There are a number of reactions that can achieve the removal of electrons from the substrate (Fig 1). The addition of oxygen, or oxygenation, was the first of these reactions discovered and thus the reaction was named oxidation.
However, many of the oxidising reactions do not involve oxygen. The simplest type of oxidation reaction is dehydrogenation, which is the removal of hydrogen from the molecule.
Another example of oxidation is electron transfer that consists simply of the transfer of an electron from the substrate.
Oxidation is the most common of the phase 1 reactions and usually involves the initial insertion of a single oxygen atom onto the drug molecule.
Question: Can you think of any of the specific oxidizing reactions?
The specific oxidising reactions and oxidising enzymes are numerous and are described by the name of the reaction or enzyme involved. Some of these oxidising reactions include:
alcohol dehydrogenation
aldehyde dehydrogenation
alkyl/acyclic hydroxylation
aromatic hydroxylation
deamination
desulfuration
n-dealkylation
n-hydroxylation
A drug that has undergone a phase 1 reaction is now an intermediate metabolite that contains a reactive group such as hydroxyl (-OH), amino (-NH2), and carboxyl (-COOH). Many of these intermediate metabolites are not hydrophilic enough to permit elimination, and therefore must undergo additional metabolism as a phase 2 reaction.
Phase 2 metabolism includes enzymatic reactions that conjugate (join together) the modified drug with another substance. The conjugated products are larger molecules than the substrate and generally polar in nature (water soluble). Thus, they can be readily excreted from the body. Conjugated compounds also have poor ability to cross cell membranes, reducing their bioactivity.
Phase 2 conjugation reactions involve the attachment of ionised groups to the drug and it is this process that significantly increases their water solubility, allowing excretion in the bile and urine. These reactions include:
glucuronidation
sulfation
acetylation
methylation
Question: Consider if the following statement is true or false:
Phase 2 reactions always result in a metabolically inactive compound.
This is false. This is most often the case, but there are some notable exceptions, such as morphine-6-glucuronide which is pharmacologically active.
Glucuronide conjugation is one of the most important and common phase 2 reactions (Fig 1). It is an important metabolic pathway for many anaesthetic drugs.
Glucuronic acid, derived from glucose, is used in this reaction - added directly to the drug or its phase 1 metabolite. The sites of glucuronidation reactions are substrates that have an oxygen, nitrogen, or sulphur bond. This applies to a wide array of drugs and endogenous substances, such as bilirubin, steroid and thyroid hormones.
The UDP-glucuronosyltransferases (UGTs) catalyse this reaction which requires UDP-glucuronic acid (UDPGA) as its group-donating co-substrate.
Currently there are 24 identified human UGT genes. One important member of this group of enzymes is the UGT2B7 variant, which plays a role in the metabolism of a range of endogenous substances including steroid hormones and bile acids well as a number of commonly used drugs such as the NSAIDs, morphine and codeine.
Question: What does glucuronide conjugation usually do?
Glucuronide conjugation usually decreases toxicity (although there are some notable exceptions). The glucuronide conjugates are generally quite hydrophilic and are excreted by the kidney or bile, depending on the size of the conjugate.
Each substrate is converted by one or several of the UGT enzyme forms. Therefore, competition for glucuronidation may occur for drugs; as yet there is little evidence that this plays an important role clinically.
Volatile anaesthetics are thought to decrease the hepatic UDPGA, and subsequently may impair glucuronidation of drugs and endogenous substances.
Propofol is rapidly metabolised in the liver by conjugation to glucuronide and sulphate, producing water-soluble compounds which are excreted mainly by the kidneys. Clearance of propofol is extremely high (greater than hepatic blood flow), suggesting additional, extrahepatic, metabolism
Opioids are metabolised mainly in the liver to both active and inactive compounds that are excreted in urine and bile. Morphine is partially excreted in bile as glucuronide conjugates. In the gut, these glucuronides are metabolised by the normal gut flora to the parent opioid compound and reabsorbed (entero-hepatic recirculation).
Metabolism occurs in the gut wall and the liver via glucuronidation to morphine-3-glucuronide (M3G) (70%), morphine-6-glucuronide (M6G) (10%). Some sulfation reactions occur. M6G is 13x more potent than morphine and is normally excreted in urine
Match the following reactions to the correct phase.
The purpose of drug metabolism is to:
A. Increase lipophilicity of a drug
B. Decrease potency of a drug
C. Increase polarity of a drug
D. Add a functional group to a drug
E. Make a drug easier to excrete
A. False. The purpose is to increase hydrophilicity.
B. False. Although this is a desirable by-product.
C. True. This makes it easier to excrete.
D. True. This is a phase 1 reaction, which allows phase 2 to occur.
E. True. This is the purpose of metabolism.
The following drugs have active metabolites:
A. Morphine
B. Pethidine
C. Diazepam
D. Atracurium
E. Pancuronium
A. True. Morphine is metabolised by hepatic glucuronidation to morphine-3-glucuronide and morphine-6-glucuronide, the latter of which is clinically active.
B. True. Pethidine is demethylated by the liver to norpethidine, which has half the analgesic potential of pethidine, but a longer elimination half-life.
C. True. Diazepam is metabolised to desmethyldiazepam, which is active.
D. True. Atracurium is metabolised primarily by Hoffman degradation, of its metabolites, laudanosine, can lower seizure trigger thresholds and cause seizure activity.
E. True. Pancuronium is largely excreted unchanged (80%), but its deacetylated metabolites (20%) are pharmacologically active.
The following drugs are mainly eliminated from the body by hepatic metabolism:
A. Isoflurane
B. Morphine
C. Atracurium
D. Vancomycin
E. Remifentanil
A. False. Hepatic metabolism of isoflurane is <1%, the vast majority is excreted unchanged by the lungs.
B. True. Morphine is metabolised by hepatic glucuronidation to morphine-3-glucuronide and morphine-6-glucuronide, the latter of which is clinically active.
C. False. Atracurium is an interesting drug who’s metabolism is dependent on ester hydrolysis in plasma and Hoffman degradation, making it a useful drug in liver and renal failure.
D. False. Vancomycin is excreted by the kidneys unchanged.
E. False. Remifentanil is rapidly degraded by non-specific red cell esterases.
The following are prodrugs:
A. Suxamethonium
B. Diamorphine
C. Captopril
D. Paracetamol
E. Enalapril
F. Prednisolone
A. False. Suxamethonium is an active drug.
B. True. Diamorphine (diacetyl morphine) is a prodrug with no intrinsic activity, it is rapidly de-acetylated in a two step process to form morphine – the active drug.
C. False. Captopril is an active drug.
D. False. Paracetamol is an active drug.
E. True. Enalapril is a weak inhibitor of ACE that is metabolically transformed to an active metabolite, enaloprilat, in the liver. Enaloprilat is 10–20 times more potent than enalapril.
F. False. Prednisolone is the metabolite of a prodrug – prednisone.
Give an overview of the pharmacokinetics of the cytochrome P450 system
define what the cytochrome P450 system is (location, structure and physical properties)
identify families of the cytochrome P450 system
describe its role in pharmacology and physiology
relate the effects of specific drugs on the cytochrome P450 enzymes and the significance in clinical practice (induction, inhibition and genetic variation)
Cytochrome P450 enzymes are a superfamily of haemoprotein enzymes which can be categorised according to their genetic similarities.
They have a multisystem role which extends into the synthesis and metabolism of endogenous compounds.
They are key to the metabolism of exogenous substrates, and can be altered by certain drugs, which may result in adverse or beneficial effects depending on the drug and its metabolites.
Inhibitors and inducers of the CYP450 enzymes are important to recognise, in order to anticipate interactions which may result during polypharmacy.
Here are some examples of the physiological roles of the enzymes according to their location.
Adrenals
11-beta-hydroxylase is a CYP450 enzyme required for the synthesis of cortisol and aldosterone, and therefore ion transport, fluid balance and glucose metabolism.
CNS
High concentrations of CYP450 enzymes are present in the brainstem and medulla. Roles involved in:
the processing of neurotransmitters, such as dopamine and serotonin
peptide hormone release from the hypothalamus and the pituitary
cerebrovascular tone via arachidonic acid metabolites
GIT or liver bile acid production
70% isoform 3A4 found in mucosa of small intestine.
Contributes to reducing the bioavailability of drugs.
Kidneys
Key location is the brush border of PCT cells.
Here, they have a role in the metabolism of arachidonic acid to metabolites which affect renal vascular tone, glomerular permeability and sodium transport (via Na/K/Cl and Na/K ATPase channels).
Lung
CYP1A has a protective function in hyperoxic lung injury.
Multiple CYP450 enzymes, expressed throughout lung tissue, are involved in bioactivation of carcinogens in tobacco smokers, which contribute to pulmonary malignancies and COPD.
What are the cytochrome P450 enzymes and why are they important?
The cytochrome P450 enzyme system is a super family of mono-oxygenase enzymes, which holds an important role in physiological and pharmacological systems.
They are primarily found in the liver, bound to the lipid bilayer of the endoplasmic reticulum of hepatocytes. Importantly, they are also found in the kidneys and adrenals.
The enzymes are fundamental to the biotransformation and metabolism of exogenous and endogenous compounds, as well as the synthesis of physiologically active substances.
In the liver, they have a pivotal role in phase one metabolism of drugs: by oxidising substrates, they produce more hydrophilic metabolites for excretion.
Altered function may result in unwanted and adverse effects of drugs, and must be considered in certain disease states and when co-administering certain drugs.
The CYP2 family is one of the largest families and is responsible for the majority of drug metabolism.
CYP2C Subfamily — CYP2C9 isoform
Metabolises s-warfarin, NSAIDS, phenytoin and prodrug (to active form) losartan.
Inhibited by fluconazole and amiodarone.
So patients taking amiodarone and warfarin will be at increased risk of bleeding
CYP2C Subfamily — CYP2C19 isoform
Metabolises diazepam, phenytoin and omeprazole.
Inhibited by PPIs, particularly omeprazole, antifungals, for example, ketoconazole, cimetidine.
CYP2D Subfamily — Isoform CYP2D6 (accounts for 25% of drug metabolism!)
Metabolises:
analgesics, for example, codeine
tramadol beta blockers, for example, atenolol
metoprolol antidepressants, for example, TCA and SSRIs
other, for example, flecainide, ondansetron
Inhibited by SSRIs, for example:
fluoxetine and paroxetine
quinidine
amiodarone
cimetidine
methadone. This means that administering drugs such as tramadol and codeine which are metabolised to morphine by CYP2D6, can prevent their beneficial effects. It can potentially lead to toxic levels of other substrates of this enzyme
CYP2E subfamily — CYP2E1 isoform (only isoform)
Metabolises volatile anaesthetic agents (for example, sevoflurane, enflurane, isoflurane), ethanol, benzodiazepines and paracetamol.
Induced by isoniazid and chronic alcohol.
Inhibited by disulfiram and acute alcohol.
So acute intoxication may result in prolonged effects of BZDs.
Disulfiram is used in the treatment of alcohol dependence. It inhibits CYP2E1, preventing the normal metabolism of ethanol (ETOH), leading to accumulation of metabolites which results in unpleasant side effects when ETOH is ingested.
The cytochrome P450 enzyme system is a super family of mono-oxygenase enzymes, which holds an important role in physiological and pharmacological systems. In the human body they are found in:
A. the heart, liver, and kidneys
B. the bloodstream
C. the liver, kidneys, and adrenals
D. only the liver
A. Incorrect.
B. Incorrect.
C. Correct. They are primarily found in the liver, bound to the lipid bilayer of the endoplasmic reticulum of hepatocytes. Importantly, they are also found in the kidneys, and adrenals.
D. Incorrect.
The function of the cytochrome P450 enzymes is:
A. the biotransformation and metabolism of exogenous and endogenous compounds
B. the synthesis of physiologically active substances
C. to oxidise substrates to produce more hydrophilic metabolites for excretion
D. phase II metabolism
A. True. The enzymes are fundamental to the biotransformation and metabolism of exogenous and endogenous compounds, as well as the synthesis of physiologically active substances.
B. True.
C. True. In the liver, they have a pivotal role in phase one metabolism of drugs: by oxidising substrates, they produce more hydrophilic metabolites for excretion. Altered function may result in unwanted and adverse effects of drugs, and must be considered in certain disease states and when co-administering certain drugs.
D. False.
The CYP3 family is a large family which metabolises endogenous and exogenous compounds. Exogenous (drugs) include erythromycin, lidocaine, diazepam and midazolam, fentanyl and alfentanil, nifedipine, chlorphenamine.
CYP3A3
3A3 and 3A4 are the most important in metabolism of drugs.
CYP3A4
CYP3A4 is the most abundant CYP enzyme in the liver. Induced by: rifampicin, phenytoin and phenobarbitone. Inhibited by erythromycin, ketoconazole, cimetidine and grapefruit juice.
Give an overview and introduction to Pharmacokinetics and Modelling, and the relevance to anaesthetics.
Identify the four phases of drug disposition in the body
Describe routes of drug adminstration
Define biovailable fraction, volume of distribution, clearance and time constant
Describe drug behaviour in a simple one-compartment model
Discuss why simple models do not fit observed drug behaviour
The one compartment model has a single volume to consider, with volume of distribution Vd
The equation describing how plasma concentration changes with time is a negative exponential
The rate at which drug is eliminated from this volume is the product of the concentration and the clearance
Clearance is the ratio of the volume of distribution and the time constant
The time constant is the inverse of the rate constant for elimination
The half-life is shorter than the time constant by a ratio of 0.693, i.e. ln2, to 1
It takes approximately five half-lives or three time constants to reach steady state - either for elimination or for infusion
The pharmacokinetic behaviour of very few drugs used clinically can be described by such a simple model
Regarding anaesthetic drugs and the CYP450 enzymes:
A. CYP3A4 is involved in the metabolism of fentanyl
B. Chronic alcohol use may increase the requirements of some volatile agents, due to inhibition of CYP450 enzymes
C. CYP2D6 displays genetic polymorphism in 1% of Caucasian population which can limit the effects of tramadol
D. Fentanyl and midazolam demonstrate competitive inhibition of CYP3A3
A. True.
B. False. It is due to inducing the CYP2E1 isoform, so it metabolises substrates, such as volatile gases more quickly, increasing the amount required.
C. False. 5-10% of Caucasian population. CYP2D6 is the best-known polymorphic isoform; slow metabolisers who express the mutation may experience a reduced effect from tramadol and codeine, which require metabolism to produce morphine for their analgesic benefit.
D. False. Fentanyl and midazolam demonstrate competitive inhibition of CYP3A4.
Analgesics
Analgesics, for example, codeine may not be effective in patients who are slow metabolisers.
Benzodiazepines
Benzodiazepines, for example, midazolam, when co-administered with fentanyl may result in prolonged activity of benzodiazepines.
ETOH
Depending on if intake is acute or chronic, the anaesthetic effect and degree of metabolism of volatiles may be different from that expected by age alone.
Anticoagulants
If warfarin is co-administered with inhibitors such as amiodarone or omeprazole there is an increased bleeding risk, and the dose may need to be reduced.
Liver failure
Patients with liver failure may have reduced synthetic function, meaning they have reduced CYP450 enzymes, and therefore reduced metabolic capacity, leading to accumulation of drugs.
There are many possible routes for drug administration.
Question: Before reading any further, can you make a list of ten or more of these routes?
Question: Can you think of two examples of routes of drug administration where the drug is not meant to reach the systemic circulation?
Here is a list of some possible routes - you may have thought of others too:
Inhalation
IV
Oral
IM
Intranasal
Sublingual
Buccal
SC
Intrathecal
Epidural
Spinal
Rectal
Intraosseous
Transdermal
Topical
Inhaled bronchodilators in asthma
Topical steroids for eczema
Intranasal decongestants (vasoconstrictors)
Which of the following correctly describe relationships between the parameters of a one-compartment model:
A. Clearance = Vd/k
B. Vd = dose/C0 for a single dose
C. τ = 0.693 . t1/2
D. Rate of elimination = clearance x plasma concentration
E. k = 1/τ
A. False. Clearance = Vd.k
B. True.
C. False. t1/2 = ln2 . τ = 0.693τ
D. True
E. True
The simple one compartment model assumes that the body is homogeneous and that all tissues behave the same as plasma.
Drug enters this single compartment and is then eliminated (Fig 1). Observation tells us that the rate of elimination is directly proportional to plasma concentration, so the rate at which plasma concentration falls must also decline with time.
This can be represented mathematically by a negative exponential relationship between plasma concentration (C) and time (t).
The relationship is written as: C = C0e-kt
The important elements are:
The size of the compartment - the volume of distribution
The type of input - bolus doses or infusions
The output - representing loss of drug from the plasma in an exponential fashion with k being the rate constant for elimination.
Drug elimination is described as “linear” or “first order” because the ratio between plasma concentration and rate of elimination is constant.
For the mathematically minded, rate of elimination can be written as a differential equation that can be integrated to give the equation introduced before: C = C0e-kt
Differential equation
The differential equation is: dC/dt = -kC
This is then integrated with respect to time t from 0 to infinity, knowing that only the exponential function integrates to itself, C must be expressed as an exponential function of time.
C = Ae-kt where A is a constant
Using the condition that at time t = 0, C = C0 this tells us that A = C0 and we get the exponential relationship: C = C0e-kt
After a single IV bolus dose the plasma concentration v time curve is modelled by a single negative exponential function:
C = C0e-kt
The features are that the intercept on the concentration axis is C0 and the constant of proportionality describing the decline is k (the rate constant of elimination).
This relationship can be expressed in a different way in order to find the constant of proportionality.
If we take natural logarithms of both sides and re-arrange then the above equation can be written as:
lnC = lnC0 - kt
This is the equation of a straight line with intercept on the lnC axis of lnC0, from which we can find Vd, and the slope of the line will be -k (Fig 1).
There are many factors that influence bioavailability of a particular drug.
Can you think of three factors that might influence bioavailability?
Can you think of three routes of administration that will bypass hepatic metabolism?
The type of preparation: for example enteric coated tablets prevent acid in the stomach from breaking down some compounds
The route of administration is important: avoiding pre-systemic metabolism will increase bioavailability.
Co-administered drugs may alter pre-systemic metabolism and either increase or decrease bioavailability
Inhalation, IV, IM, sublingual and rectal routes will avoid the first-pass metabolism associated with oral administration. You may have thought of others as well.
Patient factors are also important. Genetic factors and co-administered drugs may influence metabolism and hence bioavailability. The health of the patient is also important: hepatic failure in particular.
Using a different route for drug administration will result in a different profile of plasma concentration over time compared with the standard route.
The ‘gold standard’ route for access to the systemic circulation is IV. Drugs given by routes other than this may encounter enzymes that restrict the proportion of administered drug that can reach the systemic circulation.
The proportion (fraction) of drug, administered by a route other than IV, that reaches the systemic circulation is known as the bioavailable fraction (BF). If given IV the bioavailable fraction is considered to be 1.
Bioavailability is often quoted as a percentage: if given IV bioavailability is considered to be 100%: other routes may have a considerably lower bioavailability.
Table of drug and bioavailability fraction:
Naproxen
0.99
Isosorbide mononitrate (ISMN)
0.93
Paracetamol
0.9
Phenytoin
0.9
Ibuprofen
0.85
Nifedipine
0.5
Amiodarone
0.5
Morphine
0.25
Isosorbide dinitrate (ISDN)
0.2
In the ln(C) v time graph, the gradient of the straight line is -k.
Question: What is the gradient when logarithms to base 10 are used? (Hint: remember the equation for this straight line)
We saw that the equation for the ln(C) v time graph is:
lnC = lnC0- kt
We have just shown that ln(x) = 2.303.log(x), so:
2.303.logC = 2.303 logC0- kt
logC = logC0 - kt/2.303
The gradient is therefore -k/2.303, which makes it less steep than when natural logarithms are used.
If 100 mg of a drug is given intravenously a one-compartment model predicts it will have an initial plasma concentration of 50 mcg/ml. Which one of the following is the value of its volume of distribution?
A. 2 L
B. 5 L
C. 20 L
D. 50 L
E. 200 L
A. Correct.
B. Incorrect.
C. Incorrect.
D. Incorrect.
E. Incorrect.
Volume of distribution is given by the relationship: Dose/concentration at time t = 0
Here we have: 100 000/50 ml = 2000 ml = 2 L
In many texts the semi-logarithmic plot of concentration against time is done on a logarithmic scale to base 10. It is important to appreciate the differences between these two plots, although both can be used to find the constants Vd and k.
Question: What is the relationship between logarithms to base 10 and natural logarithms? (Hint: think how x can be expressed in both systems)
x = 10y = ez, where y is log(x) and z is ln(x)
Taking logarithms to base 10 this gives:
log(x) = y = log(ez) = zlog(e) = ln(x).0.434
log(x) = 0.434.ln(x)
Alternatively:
ln(x) = 2.303.log(x)
The plasma concentration of a drug is 20 mcg/ml five minutes after IV injection and 10 mcg/ml after 105 minutes. If it has a clearance of 70 ml/min which of the following is the closest approximation to its volume of distribution?
A. 500 ml
B. 1 L
C. 5 L
D. 10 L
E. 50 L
A. Incorrect.
B. Incorrect.
C. Incorrect.
D. Correct.
E. Incorrect.
The half life is 100 min, since this the time it takes the plasma concentration to halve.
The relationship between clearance (Cl) and Vd is:
Cl = Vd/τ
= Vd/(t1/2/0.693) = 0.693Vd/t1/2
Rearrangement gives:
Vd = Cl. t1/2 /0.693
So Vd approximates to 70 x 100/0.7 = 10 000 ml or 10 L
Which of the following are an essential part of the mathematical model describing a drug given IV in a simple one-compartment model:
A. Volume of distribution
B. Bioavailability
C. Plasma protein binding
D. Clearance
E. Time constant
A. True.
B. False. This is important for routes other than IV.
C. False. This may affect the value of the volume of distribution.
D. False. This is the ratio of the two important parameters: volume of distribution and time constant.
E. True.
Which one of the following is the clearance of a drug in a simple one-compartment model if it has an initial concentration of 5 mcg/ml after a dose of 50 mg and a time constant of 50 min?
A. 100 ml/min
B. 200 ml/min
C. 500 ml/min
D. 750 ml/min
E. 1000 ml/min
A. Incorrect.
B. Correct.
C. Incorrect
D. Incorrect.
E. Incorrect.
Clearance (Cl) is the ratio of volume of distribution (Vd) to time constant (τ).
Cl = Vd/τ
Vd = dose/C0, where C0 is the initial concentration
Vd = 50 000/5 ml or 10 000 ml
Cl = 10 000/50 ml/min
Cl = 200 ml/min
Instead of multiple doses, we can use an IV infusion to reach a steady plasma concentration. How can we predict what the plasma concentration will be when steady state is reached?
Question: What principle will help us answer this question?
At steady state input = output.
We know that the rate of drug elimination in the one-compartment model is given by: plasma concentration x clearance - this is the output in mg/min. Input is the infusion rate, in mg/min.
Which of the following can affect bioavailability:
A. Route of administration
B. Drug concentration
C. Plasma protein binding
D. Renal failure
E. Co-administered drugs
A. True.
B. False. This affects rate of absorption.
C. False. This affects free drug concentration.
D. True.
E. True.
Using a different route for drug administration will result in a different profile of plasma concentration over time compared with the standard route.
The ‘gold standard’ route for access to the systemic circulation is IV. Drugs given by routes other than this may encounter enzymes that restrict the proportion of administered drug that can reach the systemic circulation.
The proportion (fraction) of drug, administered by a route other than IV, that reaches the systemic circulation is known as the bioavailable fraction (BF). If given IV the bioavailable fraction is considered to be 1.
Bioavailability is often quoted as a percentage: if given IV bioavailability is considered to be 100%: other routes may have a considerably lower bioavailability.
Bioavailability is particularly important when oral medications can no longer be given by mouth because of illness, especially in the ICU or during anaesthesia.
When given intravenously, maximum drug concentration is reached very rapidly and then declines over time as drug is metabolised and/or excreted.
When given orally, drug may be metabolised by the gut wall before reaching the intestinal venous circulation, which then enters the portal circulation to the liver where further metabolism occurs.
From the table (Table 1) we see that NSAIDs and paracetamol have high bioavailability compared with morphine. Drug selection is important; the mononitrate ISMN has a high bioavailability but ISDN, the dinitrate, is extensively metabolised in the gut wall and has low bioavailability.
When changing from an oral to an intravenous route we need to know the bioavailable fraction. This can be measured by an AUC (area under the curve) method (Fig 1).
Give an overview of the pharmacokinetics of Two- and Three-Compartment Models, and the relevance to anaesthesia.
Describe the two and three-compartment models
Discuss the movement of drug between compartments during infusions
Compare the volumes of distribution for models of different drugs
Discuss drug clearance
Describe the three-compartment model for propofol
Compare models representing opioid pharmacokinetics
Most drugs require a two or three-compartment model to describe pharmacokinetic behaviour
The 2-C and 3-C models are linear, just as the 1-C model, since the rate at which plasma concentration falls with time is proportional to plasma concentration itself
In a 2-C model there is one and in a 3-C model there are two inter-compartmental clearances and each has just one elimination clearance
The values for compartment volumes and clearances are determined by many factors: physicochemical properties as well as rate of metabolism
Volume of distribution at steady state for propofol and fentanyl is many times greater than total body water, emphasizing that volume of distribution does not correspond to any physiological volume
3-C models are used to programme target-controlled infusion devices for propofol and remifentanil
We have values for volume of distribution and clearance for propofol: clearance is about 1.9 L/min for a 70 kg person and volume of distribution is about 230 litres.
Fig 1 shows what is observed clinically compared with what would be predicted from a 1-C model that used the known Vd and Cl.
Question: What is the most obvious difference between these two graphs?
Question: How can we model what is actually happening?
The initial rate at which plasma propofol concentration falls with time is very much faster than predicted by the simple model.
Introduce additional compartments into which drug moves rapidly.
The simple model does not explain the behaviour of most drugs, not just propofol. We know that physiologically the body is not homogeneous and that fat and water content of and blood supply to different tissues varies markedly. It is not surprising that a single compartment is insufficient to describe drug disposition in plasma.
An extension of the simple model uses a second compartment, V2, with movement between the compartments described by rate constants for distribution (k12) and re-distribution (k21). It is still assumed that drug enters the central compartment, now called V1, and is eliminated from the body only from this central compartment with a rate constant for elimination of k10.
This model will be referred to as a 2-C model, compared with the simple one-compartment (1-C) model.
Question: What factors determine the values of k12 and k21?
The physicochemical properties of the drug: its lipid and water solubilities and its pKa in particular. Blood supply and the presence of transport process
We saw with a 1-C model that a fixed-rate infusion reached a steady-state concentration that was determined by the input rate and the clearance of the drug.
In a 2-C model, the only difference is that as drug enters the central compartment some will be eliminated and some will distribute. The rate of distribution slows as the peripheral compartment fills and steady state is reached when the concentration in both compartments is equal. At this point there is no overall distribution/redistribution.
Question: What can we calculate from the concentration at steady state (Css) and the infusion rate?
Clearance from the body using the principle:
INPUT = OUTPUT
This is done in the same way as for the 1-C model:
Clearance = INPUT/Css
INPUT has units of mg/min, Css has units of mg/ml, consistent with clearance having units of ml/min.
In a 2-C model a hydraulic analogy can demonstrate what happens when we stop an infusion that has reached steady state.
Initially the concentrations in both compartments is the same. The only thing that can happen now is that the concentration in the central compartment will fall as drug is eliminated. This creates a concentration gradient between the peripheral and central compartments, favouring redistribution.
Question: What factors determine how long it takes for plasma concentration to fall below an effective level?
It depends on:
The difference between the steady state concentration and the minimum effective concentration
The relative values of the inter-compartmental and the elimination clearances
If the inter-compartmental clearance is low and the elimination clearance high, then recovery from drug effect may be rapid (Fig 1).
If inter-compartmental clearance is high and elimination clearance low, then recovery will be prolonged because of rapid redistribution (Fig 2).
Using the hydraulic model, think about what will happen when we stop an infusion before reaching steady state.
Question: If the infusion is stopped before steady-state is reached what happens initially?
he concentration in V1 falls: some drug will distribute into V2 and some will be eliminated. This continues until the concentration in V2 approaches that of V1. From this point on, the system behaves as described for stopping the infusion after steady state has been reached.
The extent of this initial distribution depends on how long the infusion has been running and the ratio of inter-compartmental to elimination clearance values. If elimination clearance is very fast compared with inter-compartmental clearance, then elimination will predominate and the distribution phase will be relatively short.
Which of the following correctly identify the relationship among parameters of a two-compartment model?
A. There are three exponential processes
B. Elimination clearance is given by V1.k10
C. Inter-compartmental clearance is given by V2.k12
D. The peripheral compartment must have a larger volume than the central compartment
E. The volume of distribution is that of the central compartment only
A. Incorrect. There are two exponential processes.
B. Correct.
C. Incorrect. Inter-compartmental clearance is given by V1.k12 or V2.k21.
D. Incorrect. It is very common for this to be the case, but the model does not demand this.
E. Incorrect. The volume of distribution at steady state is the sum of both the peripheral and central compartment volumes.
Label the two compartment model.
Which of the following are true of a three-compartment model?
A. There are three exponential processes
B. Elimination clearance is given by V3.k31
C. There are three inter-compartmental clearances
D. Terminal elimination half-life is the same as elimination clearance
E. The volume of distribution is the sum of the three compartment volumes
A. True.
B. False. This is the inter-compartmental clearance between the central and third compartments. The elimination clearance is given by V1.k10.
C. False. There are two inter-compartmental clearances, not three.
D. False. It is related to elimination and re-distribution rate constants.
E. True.
Which of the following are true of a two-compartment model?
A. It is a second order process
B. Clearance occurs between compartments
C. The two exponential processes have half-lives of ln2/k12 and ln2/k21
D. Inter-compartmental clearance is always faster than elimination clearance
E. Elimination of drug from the model can occur from both compartments
A. False. It is a first order process because the rate of decline of plasma concentration is still proportional to plasma concentration.
B. True.
C. False. The two exponential processes have half-lives of t1/2α and t1/2β, which are both dependent on all rate constants in a complex way.
D. False. It may be true but the model does not demand this.
E. False. Elimination occurs only from the central compartment.
In the 1-C model we saw that the half-life of the single exponential process could be found from the gradient of a semi-logarithmic plot of concentration against time.
Question: The 3-C model has three exponential processes. How many half-lives can we describe?
Question: What are the three half-lives and what do they represent?
Three: related to the rate constants α, β, and δ. These are the gradients of the three lines that comprise the semi-logarithmic plot of concentration against time.
Rapid initial distribution t1/2α = ln2. 1/α;
Intermediate elimination and distribution t1/2 β = ln2. 1/β
Terminal elimination half-life t1/2δ = ln2. 1/δ, which represents re-distribution and elimination.
The inter-compartmental clearance between the central and third compartments is, by convention, slower than that between the central and second compartments.
Question: How do we use the rate constants and volumes to describe the inter-compartmental clearance between the central and third compartments (Cl13)?
Question: What form do you expect the mathematical equation that describes the 3-C model to take?
Cl13 = V1.k13 = V3.k31
This is exactly analogous to the inter-compartmental clearance between the central and peripheral compartment in the 2-C model.
The sum of three exponential processes:
C = Ae-αt + Be-βt + De-δt
Although the 2-C model is useful for predicting the behaviour of many drugs, it does not work well for many anaesthetic drugs that are given by continuous infusion for several hours in the operating theatre or even for several days in an intensive care setting.
Propofol, in particular, is an extremely lipid soluble drug that has a very large volume of distribution. Although a 2-C model can be used for predicting plasma concentration after bolus dose administration or for short-duration infusions, it is not adequate in the context of longer infusions.
A three-compartment (3-C) model has been shown to be the best predictor of plasma propofol levels.
The principles underlying a 3-C model are very similar to those of a 2-C model. The major differences are the existence of a third compartment and another inter-compartmental clearance (Fig 1).
The arrangement of compartments is called ‘mammillary’ since both peripheral compartments are in communication with the central compartment.
The 3-C model identifies three volumes that behave differently: the central compartment represents the behaviour of plasma, the second and third compartments represent peripheral tissues. In general the third compartment is very large for lipid soluble drugs.
The model is still a linear model since all elimination rates are linearly dependent on plasma concentration just the same as in the 1-C, and 2-C models met before.
The equation describing the concentration in the central compartment is now:
C1 = Ae-αt + Be-βt + De-δt
The slow, terminal phase has a rate constant δ, which is a hybrid rate constant as we described for the two rate constants for the 2-C model.
In anaesthetic practice the 3-C model is used to programme target-controlled infusion pumps for propofol and remifentanil. This will be covered in a later session.
Match the values to a parameter to construct the Marsh model for propofol.
A three compartment model is used to model the behaviour of propofol.
Which of the following correctly describe such a model:
A. Propofol elimination occurs only from the third compartment
B. Propofol moves directly between the second and the third compartments
C. The central compartment volume is smaller than the second compartment volume
D. The third compartment has a volume equivalent to total body water
E. Clearance by elimination is faster than both inter-compartmental clearances
A. False. Elimination occurs only from the central compartment, V1.
B. False. A mammillary model provides the best fit where drug moves from second to third compartments indirectly through the central compartment.
C. True.
D. False. The volume of the third compartment is in excess of 200 L, whereas total body water is only 42 L.
E. True. The inter-compartmental clearance from central to second compartment is almost as fast (1.78 L/min compared to 1.9 L for a 70 kg person).
Alfentanil, remifentanil, morphine and fentanyl are commonly used by infusion in theatre and in the ICU. They are all opioid agonists but have very different physicochemical properties and as a result require quite different models to predict their behaviour.
Question: What are the pKas of these opioids and how might this influence kinetic behaviour?
Question: How will this be reflected in the model?
Alfentanil 6.5; remifentanil 7.1; morphine 8.0; fentanyl 8.4. The lower the pKa the higher the proportion of unionized drug, which is associated with faster access to the CNS across the highly lipid blood-brain barrier and also into adipose tissue.
If lipid solublity and metabolism were all equal, then this difference in pKa would be associated with faster inter-compartmental clearances for lower pKa.
Which of the following are true of compartmental models for drug behaviour?
A. Most drugs behave according to a one-compartment model
B. Volume of distribution at steady state is the sum of all the compartment volumes
C. The second compartment volume is always smaller than the third compartment volume
D. The third compartment has a smaller inter-compartmental clearance than the second compartment
E. Inter-compartmental clearance between the central and second compartment may be greater than elimination clearance
A. False. Most drugs require a 2-C or 3-C model to describe their behaviour.
B. True.
C. False. This is often the case, as for fentanyl and propofol, but is not necessarily true - as for remifentanil.
D. True. This is the convention, as drug distributes more quickly to the second than the first compartment of a 3-C model.
E. True. This is true for fentanyl.
Which of the following are true of pharmacokinetic models for opioid drug behaviour?
A. Morphine has a lower volume of distribution than remifentanil
B. Alfentanil has a faster inter-compartmental clearance, Cl12, than elimination clearance
C. Fentanyl and morphine both have steady-state volumes of distribution greater than 3 ml/kg
D. The volume of the third compartment directly reflects lipid solubility
E. Morphine has a faster elimination clearance than fentanyl
A. False. Remifentanil has the smallest volume of distribution of the four opioid drugs described in this session.
B. True.
C. True.
D. False. Morphine is much less lipid soluble than fentanyl, but it has a similar volume of distribution. Lipid solubility can be important, but the model parameters are all highly inter-related and prediction of their relative values is impossible: the model will reflect observed behaviour.
E. True.
Before comparing the models for the opioids, consider the information in Table 1 and how it might influence the compartment volumes and inter-compartmental clearances.
Which of the following describe the pharmacokinetics of a drug that distributes according to a two-compartment (2-C) model?
A. It is a linear model
B. The rate at which plasma concentration falls with time is constant
C. Inter-compartmental clearance is given by V1.k12
D. At steady state, the elimination clearance can be calculated if plasma concentration is known
E. Only drugs given intravenously can be modelled this way
A. True.
B. False. There is a constant proportionality between concentration and rate at which concentration falls: dC/dt = -kC.
C. True.
D. True. Input is known from infusion rate and concentration of solution being infused; output is the product of clearance and concentration at steady state.
E. False. As long as bioavailability is known, then drug behaviour can be modelled.
Give an overview of the pharmacokinetics of Clearance and Volume of Distribution, and the relevance to anaesthesia.
Discuss drug clearance and its relationship to metabolism
Describe the non-compartmental method for measuring clearance
Discuss the variability in volume of distribution for anaesthetic drugs
Identify methods for measuring volume of distribution
Clearance is the elimination of parent drug from plasma. It may be due to excretion unchanged or metabolism or a combination of these two processes
In a one-compartment model, clearance is the ratio of volume of distribution to time constant
In a multi-compartment model clearance is found using the area under the concentration-time curve (AUC) method
Volume of distribution varies widely among drugs used in anaesthesia and intensive care
The physicochemical properties of a drug influences its volume of distribution and the physiological spaces into which it can distribute
The best estimation of volume of distribution is the ratio of the bioavailable administered drug dose to the clearance found from the AUC method
Comparing the pharmacokinetic behaviour of fentanyl and propofol:
A. Propofol is more lipid soluble than fentanyl
B. For fentanyl, inter-compartmental clearances are greater than for propofol
C. Propofol has a lower pKa than fentanyl
D. Both fentanyl and propofol distribute rapidly to peripheral tissues after a bolus dose
E. After constant-rate infusions lasting several hours, propofol and fentanyl both show a rapid decline in central compartment concentration
A. True.
B. True.
C. False. Propofol has a high pKa but, unlike fentanyl, it is a weak acid, so mainly unionized at plasma pH.
D. True.
E. False. Fentanyl redistributes very rapidly after long infusions, which maintains plasma levels much higher than for propofol.
Propofol and fentanyl are both very lipid soluble drugs with rapid metabolism. Both have rapid offset of effects after a single dose.
They might therefore be expected to have a similar pharmacokinetic model to describe their behaviour. However they differ in one important physicochemical property: fentanyl has an ionisable group and is largely in the ionized form in plasma whereas propofol is effectively unionized. In addition, propofol is more heavily protein bound, which accounts for a larger central compartment volume.
When comparing the models it can be seen that fentanyl has much greater inter-compartmental clearances than propofol, which reflects this difference in ionization. In addition, the clearance of propofol is greater than that of fentanyl. These two differences mean that, despite a larger central compartment and even after prolonged infusions, the plasma level of propofol will fall more quickly than for fentanyl.
In a later session we will expand on this context-sensitive behaviour of the two drugs.
Clearance represents elimination of drugs from the body. It may be due to metabolism or excretion unchanged or a combination of these. It has units of ml/min or L/h.
Question: In a simple one-compartment (1-C) model, how is clearance defined and found?
It is the ratio of volume of distribution (Vd) to time constant (τ).
Cl = Vd/τ
Both Vd and τ are found from the semi-logarithmic plot of concentration against time, as seen previously.
As we have noted in a previous session, few drugs behave as if they distribute into a single volume and the concentration vs time curve can be described by the sum of two or three exponential processes.
The concentration v time curve for most drugs can be described by the sum of two or three exponential processes.
The area under the concentration v time curve represents the clearance of the entire dose reaching the systemic circulation. The theory underlying this comes from non-compartmental models and applying the concept of statistical moments. You do not need to know about this in any detail.
Clearance can be measured from a bolus dose given intravenously and measuring the area under the curve, assuming all the dose given reaches the systemic circulation.
Cl = dose/AUViv
When given orally, only part of the dose will reach the systemic circulation.
Question: What do we call that fraction of drug taken orally that reaches the systemic circulation?
Question: How can we use bioavailable fraction (BF) to find clearance after an oral drug dose?
The bioavailable fraction. We can still calculate clearance from an orally administered dose, as long as we know the bioavailable fraction.
BF = AUCo/AUCiv
AUCiv = AUCo/BF
The clearance can then be calculated from:
Cl = dose x BF/AUCo
For a small number of drugs the KM is close to clinically relevant concentrations. In this case a small increase in substrate concentration will increase activity a small amount but a larger rise in plasma concentration may cause the substrate concentration to exceed that at which enzyme activity is maximal.
Question: What happens if substrate concentration exceeds that for maximal metabolic activity?
If this is the only enzyme that can metabolise the substrate then the rate of metabolism remains constant, at maximum capacity, independent of substrate concentration. This is the phase of zero order kinetics (Fig 1 and Fig 2).
When substrate concentration falls below that at which maximum metabolism occurs first order kinetics is re-established.
Thiopental, when given by infusing for prolonged periods, will saturate its enzyme system and display zero kinetics. Similarly phenytoin can also saturate enzyme capacity leading to adverse clinical effects that can be prolonged.
For most drugs first order kinetics prevail over the whole range of plasma concentrations. For some drugs the KM is many orders of magnitude above plasma concentration; hepatocyte concentration of drug is maintained effectively at zero.
Question: What limits rate of drug metabolism under these conditions?
Question: What anaesthetic drug is metabolised so rapidly that its metabolism is determined by hepatic blood flow?
Delivery of substrate: hepatic blood flow.
Metabolism of propofol is very dependent on hepatic blood flow: it has a very high extraction ratio. Fentanyl also has a high hepatic extraction ratio (ER).
Phenytoin, and other drugs where the enzymes are working at concentrations of substrate above their KM, has a low ER since the intracellular concentration is close to plasma.
Clearance represents elimination of drug from the body.
Question: How many ways can drug be eliminated?
Question: What can alter clearance?
Metabolism, particularly hepatic, excretion through the kidneys, lungs and other routes.
Changes in elimination and volume of distribution. People differ in their metabolic and excretory rates, which accounts for inter-individual variation. Illness will also alter clearance by altering volume of distribution. Drug interactions will alter metabolism and therefore clearance.
We will return to the effects of hepatic and renal failure later in this session.
Not all drugs need to be metabolised to be eliminated from the body.
Highly polar drugs will be excreted unchanged through the kidneys by filtration. Only unbound drug is filtered, unlike the hepatocyte, where capilliary sinusoids are freely permeable.
Many drugs are weak acids and weak bases and can be secreted into the proximal tubule by secondary transporters.
Renal elimination of drugs is therefore complex.
Question: What other route of excretion is of importance in anaesthesia?
The inhalational route for volatile agents.
The degree of protein binding differs widely among clinically useful drugs. Drugs compete for binding sites on albumin and globulins and one drug may displace another.
Question: What may happen if competition occurs?
It depends on the ER of a drug and its therapeutic ratio. Drugs with a high ER would still be entirely metabolised, so changes in plasma binding have no effect.
For most drugs with moderate ER, an increase in free drug concentration would increase hepatic uptake and enzyme activity increases so as to maintain effective plasma levels.
For a small number of very highly bound drugs with very low extraction ratios, such as warfarin and phenytoin, a small change in protein binding results in a very large increase in free drug concentration. Since both ER and intrinsic clearance are low, plasma concentrations will rise and the toxic threshold may be exceeded.
For drugs that are only moderately plasma protein bound, a small change in binding has little effect on plasma concentration and metabolism increases just a little to maintain levels.
These effects are best illustrated using Blaschke’s triangle (Fig 1).
In the 1-C model there is a single volume that represents the apparent volume into which the drug disperses.
Question: How do the two- and three-compartment models differ from the 1-C in terms of volume of distribution?
There is a central volume of distribution and one or two peripheral compartments, each of which has a volume that will contribute to the overall volume into which drug can disperse.
This reminds us that drug is initially given into the central compartment and then distributes to the peripheral compartment(s) later.
The central compartment volume represents the initial volume of distribution.
Question: Why might it be important to know the value of this initial volume of distribution?
Question: Can you think of any drugs where loading doses are used then smaller, maintenance doses keep plasma levels within therapeutic range?
To calculate a loading dose. There are several considerations when determining how to load a patient with a drug, particularly if the therapeutic window is small.
Warfarin and digoxin require loading doses for oral administration and amiodarone for intravenous use. TCI propofol also uses a loading dose. You may have thought of others.
You know that the initial volume of distribution of propofol is about 16 L; if you wish to establish an initial plasma concentration of 6 μg/ml, what loading dose would you use?
A. 13 ml of a 2% solution
B. 4.8 ml of a 1% solution
C. 18.2 ml of a 2% solution
D. 9.6 ml of a 1% solution
E. 26 ml of a 1% solution
A. Incorrect.
B. Incorrect.
C. Incorrect.
D. Correct.
E. Incorrect.
The initial volume of distribution (Vd) is 16 000 ml.
The required plasma concentration (Cp) is 6 μg/ml.
The total amount of propofol required to fill the initial volume of distribution is:
Cp x Vd
= 16, 000 x 6 μg
= 96 000 μg or 96 mg
A 1% solution of propofol has 10 mg/ml and a 2% solution 20 mg/ml.
The correct answer would either be 9.6 ml of 1% or 4.8 ml of 2% propofol.
A drug behaves according to a single-compartment model. Experimentally you find that it has a half-life of 70 min and a clearance of 100 ml/min. Which one of the following is its approximate volume of distribution?
A. 7 L
B. 10 L
C. 70 L
D. 100 L
E. 1000 ml
Submit
A. Incorrect.
B. Correct.
C. Incorrect.
D. Incorrect.
E. Incorrect.
In a simple one-compartment model clearance is the ratio of volume of distribution (Vd) and time constant (τ).
Cl = Vd/τ
Time constant is related to half-life:
t 1/2 = ln2.τ = 0.693 τ
⇒ Vd = Cl.τ = Cl.t1/2 /0.693
Vd is approximately 100 x 70/0.7 ml = 10,000 ml = 10 L
When a drug is given it will distribute through the body, carried in blood. Its physicochemical properties determine how easily it passes passively through lipid barriers and membrane.
When considering physiological volumes, there is the intravascular space and the interstitial space, which are part of the extracellular volume, as well as the total body water.
Question: If a drug was restricted to the vascular volume, what would you expect its volume of distribution to be?
The vascular space has a volume of 70-80 ml/kg, so a drug restricted to this space would have a small volume of distribution less than 10 L.
It is often said that highly protein-bound drugs have a small volume of distribution: this is not necessarily true. Only unbound drug can distribute, so highly protein bound drugs will simply distribute more slowly. Propofol is more highly bound than fentanyl (98% compared with 83%) but both distribute throughout a large volume of distribution, fentanyl more rapidly than propofol.
The intravascular space is small compared with total body water (42 L).
Question: What characteristics would you expect of a drug that is restricted to the intravascular volume?
Question: Can you think of a commonly used drug that has these characteristics?
A large, polar molecule that cannot pass through the endothelial gaps in the capillary vessels. It may be highly protein bound.
Heparin is a good example, it has a volume of distribution of about 40-70 ml/kg - very similar to blood volume.
The extracellular space (14 L) is about one third the total body water (42 L). Some drugs can distribute from the capillaries into the extracellular space, but not into the total body water.
Question: What characteristics would you expect of a drug that is restricted to the extravascular volume?
Question: Can you think of a class of drugs used commonly in our anaesthetic practice that has these characteristics?
A small, polar molecule that cannot pass through cell membranes.
Muscle relaxants are permanently charged molecules that do not pass through cell membranes and exert their action extracellularly. They have low protein binding and can easily pass out out of the capillaries, but not into cells - particularly not into adipocytes. Their volumes of distribution are around 14 L.
Total body water is around 42 L in an adult. Some drugs can distribute from the capillaries into the extracellular space and then pass into the cell.
Question: What characteristics would you expect of a drug that can access the intracellular volume?
Question: Can you think of a commonly abused drug that has these characteristics?
Low molecular weight drugs that can readily pass through cell membranes.
Ethanol can distribute through the total body water. Deuterium (‘heavy water’) can be used to measure this volume as it distributes freely.
If a drug can be described by a 1-C model, then volume of distribution is easily measured from the concentration-time curve.
Question: What is the best graph to plot for finding the volume of distribution in a 1-C model?
Question: How do you find the volume of distribution from this initial concentration?
A semi-logarithmic plot, which gives a straight line; the intercept on the y-axis is the concentration at time t=0.
Vd = dose/concentration, with units of mL or L.
Total body water is around 42 L in an adult. Some drugs have volumes of distributions many times this value.
Question: Can you think of two or more reasons why drugs could have large volumes of distribution?
Very lipophilic drugs that are poorly water soluble will preferentially enter adipose cells and be retained, such as propofol. Some drugs are actively transported into cells and concentrated by physiological processes, such as iodine. Some drugs enter cells and are then bound strongly to protein or nucleic acids within the cell, such as chloroquine.
This wide variation in volume of distribution for drugs is summarised in Table 1.
In hepatic failure there is a change in the distribution of body water with the development of ascites.
Question: How might a large volume of ascites affect clearance and hence drug administration?
The initial volume of distribution generally increases so loading dose needs to increase.
There is also a reduction in synthesis of plasma proteins that bind drugs, such as albumin, which complicates calculation of loading dose. Metabolism will decline, so maintenance dose will be reduced.
Loss of metabolic function is not equal for all drugs: not all drugs are metabolised in the liver. Moderate liver failure may not affect metabolism of all drugs requiring hepatic transformation equally, this depends on intrinsic clearance and hepatic blood flow.
Severe, end-stage hepatic failure is associated with poor drug metabolism in general for all drug biotransformed in the liver.
Earlier in the session we identified several factors that can influence how individuals vary in their handling of drugs. There can be very wide differences in clearance even among healthy people.
Question: Can you think of four factors, other than hepatic or renal failure, that could influence inter-individual variability in drug clearance?
Age - this alters the proportion of body water
Gender - lean body mass depends on gender and affects volume of distribution
Genetic disposition - possession of a different isoform of an enzyme may alter metabolism and clearance
Co-administration of other drugs: drugs can induce or inhibit enzyme activity
In renal failure, as in hepatic failure, there is a change in the distribution of body water.
Question: How might renal failure affect clearance?
Question: What drugs are excreted unchanged by the kidney?
The initial volume of distribution generally increases, since there is an increase in extracellular volume. A loading dose needs to be greater.
Hepatic metabolism is not necessarily affected, but some drugs are dependent upon renal excretion.
Those drugs that are highly polar: permanently charged drugs, such as muscle relaxants, may partly be excreted unchanged. Active metabolites that have been initially biotransformed in the liver will be poorly excreted, which might be important - for example morphine-6-glucuronide, which is more potent than morphine.
The clearance of a drug is 1.4 L/min. The initial half-life is 10 min. What loading dose is required to give a plasma concentration of 10 mcg/ml?
A. 100 μg
B. 200 μg
C. 200 mg
D. 10 g
E. 200 g
A. Incorrect.
B. Incorrect.
C. Correct.
D. Incorrect.
E. Incorrect.
The loading dose is the product of the concentration required and the volume of distribution. The initial volume of distribution is found from clearance:
Cl = Vd/τ and t1/2 = 0.693 τ
Vd = Cl. t1/2/0.693
Vd = 1.4 x 10/0.7 (approximately)
Vd = 20 L
Dose = Vd x concentration
= 20 000 x 10 μg = 200 000 μg = 200 mg
The initial volume of distribution of a drug is 20 L.The effective dose is 5 μg/ml. What loading dose is required?
A. 100 μg
B. 1 mg
C. 10 mg
D. 100 mg
E. 1 g
A. Incorrect.
B. Incorrect.
C. Incorrect.
D. Correct.
E. Incorrect.
The loading dose is the product of the concentration required and the volume of distribution.
Dose = Vd x concentration
= 20 000 x 5 μg = 100 000 μg = 100 mg
Which of the following correctly describe the pharmacokinetics of drug distribution?
A. In a 1-C model, clearance equals volume of distribution divided by half-life
B. In a 3-C model, clearance is dependent upon plasma concentration
C. In a 2-C model, terminal elimination half-life can be used to estimate volume of distribution
D. In a 2-C model the slower terminal rate constant represents elimination
E. Non-compartmental kinetics can be used to find clearance
A. False. Clearance is the ratio of volume of distribution to time constant, not half-life.
B. False. Clearance is independent of plasma concentration.
C. True.
D. False. The terminal rate constant, β, is a hybrid rate constant and depends, in a complex way, upon elimination and inter-compartmental transfer rate constants.
E. True.
Regarding pharmacokinetics:
A. Half-life is longer than time constant
B. Non-compartmental kinetics can be used to calculate bioavailable fraction
C. Terminal elimination half-life is dependent only on excretion
D. In a 2-C model inter-compartmental clearance is the product of V2 and k21
E. When measuring clearance after an oral dose, bioavailable fraction must be known
A. False. Time constant is longer than half-life by a factor of 1.443 since t1/2 = 0.693τ.
B. True.
C. False. It is also dependent upon re-distribution.
D. True.
E. True.
Give an overview of Target Controlled Infusions, and the relevance to anaesthetics.
Identify the difference between targeting plasma and effect-site
Describe the three-compartment models for propofol and remifentanil
Discuss the implications of duration of infusion on recovery times
Identify ideal properties for a drug to be used in a TCI system
Microprocessor driven infusion pumps can be used to induce and maintain anaesthesia with propofol and remifentanil
It is possible to target either plasma or effect-site
The Marsh model was designed to target plasma propofol concentration
The Schnider model was designed to target effect-site propofol concentration: this model is very different from the Marsh model
The time it takes for plasma concentration to fall to half of its value at the end of a targeted infusion is known as the context sensitive half-time (CSHT)
The context of the CSHT is the duration of the infusion
Propofol has a maximum CSHT of just under 20 min
Remifentanil has an almost constant CSHT due to its rapid metabolism
Fentanyl has a very long CSHT if infused for many hours
In an earlier session the three-compartment model (3-C) was introduced. This model can be used to predict the behaviour of both propofol and remifentanil.
Question: What are the pharmacokinetic parameters needed to describe a 3-C model?
Question: How can this model be applied to clinical use?
: A central volume of distribution and clearance together with two peripheral compartments and two inter-compartmental clearances.
An infusion pump can be controlled by a microprocessor that calculates the volume of drug to be delivered.
Which of the following can alter pharmacokinetic behaviour of a given drug in a particular patient?
A. Increasing age
B. Co-administered drugs
C. Onset of renal failure
D. Increase in body mass index (BMI)
E. Developing hepatic failure
A. True.
B. True.
C. True.
D. True.
E. True.
The first 3-C model described for use in an infusion device was the Marsh model.This allowed the plasma concentration to be targeted, with a pump delivering the calculated amount of propofol.
The plasma concentration required for induction of anaesthesia is around 6 μg/ml. The concentration to maintain anaesthesia varies between 3-10 μg/ml, depending on how stimulating the surgery is.
Question: What information does the pump need to deliver the appropriate amount of propofol?
In the Marsh model the initial volume of distribution is 15.9 L for a 70 kg patient.
Question: What loading dose will be delivered to achieve a plasma concentration of 6 μg/ml?
Question: If the pump delivers at a maximum rate of 1200 ml/h, for how long will the pump run before delivering this amount of propofol if a 1% solution is used?
The weight of the patient and the plasma concentration required.
Loading dose is the product of Vd and concentration so loading dose is:
15 900 x 6 = 95 400 μg or 95.4 mg
95.4 mg is 9.54 ml of a 1% solution. The pump runs at 1200 ml/h or 20 ml/min so this volume should be delivered in 9.54/20 min or just under 30 seconds. In practice a small amount more is given to compensate for the elimination and distribution taking place over this short time.
Consider a target-controlled infusion of propofol maintaining a plasma concentration of 6 μg/ml:
A. After a 30 min infusion the context sensitive half-time is 5 min
B. If the CSHT is 8 min then it will take 16 min for the concentration to fall to 1.5 mcg/ml after stopping the infusion
C. After an infusion lasting 10 h the CSHT is constant
D. At steady state infusion rate is equal to elimination rate
E. The context of CSHT is the duration of the infusion
A. True.
B. False. It will take longer for the concentration to fall from 3 μg/ml to 1.5 μg/ml than from 6 to 3 μg/ml.
C. False. It takes around 70 h before the CSHT is constant.
D. True.
E. True.
An ideal anaesthetic agent suitable for use by TCI will have the following properties:
A. It will behave according to a one-compartment model
B. It must be much more water than lipid soluble
C. It will be eliminated more rapidly than it is re-distributed
D. It will be cardiovascularly stable
E. It will have no active metabolites
A. False. Very few drugs behave this way, certainly not anaesthetic agents.
B. False. It needs to be lipid soluble to cross the blood-brain barrier.
C. True.
D. True.
E. True. This allows its effects to be predicted from its plasma concentration.
A patient on the ICU is receiving a constant-rate infusion of fentanyl:
A. The plasma concentration of fentanyl will not become constant until the infusion has been running for an hour
B. If the infusion has been running for less than 30 min then fentanyl has a short CSHT
C. The volume of distribution of fentanyl is more than 300 L
D. If a TCI infusion is used, then after a 6 h infusion the CSHT will be shorter than for a similar duration of infusion of propofol
E. If the infusion is stopped, the duration of its hypnotic effect depends only on the CSHT
A. False. It takes around 60 hours for steady state to be reached.
B. True.
C. True.
D. False. Propofol will have a much shorter CSHT after a 6 h infusion.
E. False. It also depends on the infusion rate: if low, then the effect may wear off before the concentration in plasma has halved. Patient pathology also influences pharmacodynamic response.
A patient is being anaesthetised with TCI propofol and remifentanil, both targeting the effect site:
A. On induction, the bolus dose of propofol will be larger than if an identical plasma concentration had been targeted
B. Remifentanil reaches steady-state at the same time as propofol
C. The effect-site is not appropriate to target when using the Minto model for remifentanil
D. The Schnider model would be appropriate to use for propofol in TCI effect mode
E. The CSHT of remifentanil is always shorter than that for propofol when they are used together
A. True.
B. False. Remifentanil has a smaller volume of distribution and reaches steady-state more rapidly than propofol.
C. False. The Minto model was designed to target the effect-site.
D. True.
E. True.
Which of the following statements are true for pharmacokinetics in older patients?
A. Oral absorption of most drugs in older patients is altered due to reduced gastric emptying
B. Absorption in older patients is the same as it is for other adults
C. The increase in total body water affects the distribution of drugs in older patients
D. Plasma and tissue concentrations are increased due to reduction in volume of distribution of water soluble drugs
E. The reduction of hepatic blood flow leads to increased systematic bioavailability of drugs dependent on hepatic clearance
F. The decrease in glomerular filtration rate and tubular function prolongs the elimination of drugs dependent on this route of elimination
A. False. Ageing is associated with changes in gastric pH, slowing of gastric emptying, and reduced small bowel surface area.
B. False. Drug absorption in older patients is slower than other adults.
C. False. There is an overall decrease in total body water in older patients. Younger patients have higher total body water content.
D. True.
E. True.
F. True.
Depending on the route of administration, the absorption of some drugs may be reduced in the elderly.
The decrease in body water affects the distribution and serum concentration of water soluble drugs. The half-life of lipid soluble drugs is increased and unbound drug concentration is increased due to changes in body fat and serum protein content.
The decrease in liver blood flow affects the rate of drug clearance. Re-accumulation of drugs normally eliminated by the kidneys can occur due to decreased renal function.
The relative reduction in body protein corresponds with loss of muscle bulk. This is also associated with reduced muscle blood flow and reduced metabolism of certain drugs such as remifentanil.
Which of the following statements are true for pharmacokinetics in neonates and infants?
A. Absorption is slower because of longer gastric emptying time
B. Hepatic enzyme activity is very similar to adults
C. Adult rates of metabolism can be reached with most drugs 6 weeks postnatally
D. Glomerular filtration rate is 20-40% of adult rate
E. Scaling down an adult dose of a water-soluble drug in proportion to body weight can result in a lower plasma concentration in neonates
A. True.
B. False. This is false for neonates and infants but may be true for older children.
C. False. Adult rates of metabolism for some drugs, such as barbiturates and phenytoin, can occur 2-4 weeks postnatally.
D. True.
E. True.
As with other special population groups, absorption is a key factor requiring consideration in neonates and infants.
Absorption will depend on the route of administration. Tissue sites of drug administration may lead to unreliable absorption due to vasomotor instability.
There are three different routes of administration which must be considered:
- Enteral route
There are several key facts to remember about absorption via the enteral route of administration in neonates:
Rate of absorption is slower because of the prolonged gastric emptying time and the increased intestinal transit time
Gastric pH is less acidic in neonates
Contact time - because of the longer gastric emptying time, there is greater contact time with the mucosa and thus an increased amount of drug absorption
- Rectal route
The site of placement of the drug within the rectal cavity may influence absorption. This is because of the difference in venous drainage systems. The rectal route of administration should be considered less reliable than IV.
The rectal route may be appropriate in certain out-of-hospital emergencies such the treatment of status epilepticus with rectal diazemuls.
- Transdermal route
In neonates the transdermal route of administration may be associated with rapid absorption because the stratum corneum is thin.
The ratio of body surface area to weight is much greater in neonates and infants than for older children and adults.
Total body water (TBW) plays an important role in distribution of drugs in neonates and infants (Fig 1).
TBW is 80% of total body weight in a preterm baby.
Doses of water soluble drugs based on scaling down adult doses in proportion to body weight can result in lower tissue concentrations.
There is a marked difference in the body fat content of neonates and infants, older children, adults (Fig 2) and older patients (Fig 3). This will influence the distribution of drugs.
Lower body fat and increased permeability of the blood-brain barrier can lead to increased concentrations of lipid soluble drugs in the brain.
There is a decrease in protein binding of drugs causing increased availability of the unbound drug. This leads to enhanced pharmacological action.
Metabolism of drugs in neonates depends on the size of the liver and activity of microsomal enzyme systems.
In neonates and infants:
Enzyme activity is immature
Phase I metabolism activity is reduced in neonates but increases progressively during the first 6 months of life and can exceed adult rates by the first few years for some drugs. It slows again during adolescence, and usually attains adult rates by late puberty
Adult rates of metabolism, however, may be achieved for some drugs 2-4 weeks postnatally
Phase II metabolism varies considerably
The glomerular filtration rate in neonates and infants is 20-40% of adult rate and therefore drugs removed by this route are eliminated slowly.
How would you expect the rate of absorption, distribution, metabolism and elimination to change in older patients when compared with neonates?
A. These factors would remain unchanged
B. These factors would change due to decrease in body water content
C. There would be some changes but generally they would be the same as for younger adults
A. Incorrect.
B. Correct.
C. Incorrect.
There is a marked difference between neonates and older adult patients in all aspects of pharmacokinetics. A number of factors differ in older people when compared with neonates. These include:
Total body water
Enzyme activity
Renal function
Hepatic blood flow
Level of body fat
Protein binding
Give an overview of Pharmacokinetic Principles in Different Patient Groups, and discuss the relevance to anaesthesia.
Define the concept of pharmacokinetics with reference to absorption, distribution, metabolism and elimination
Explain the absorption, distribution, metabolism and elimination of anaesthesia in key patient groups including neonates and infants, the elderly, the obese, pregnant patients and the critically ill
Neonates
Geriatric
Pregnant
Obese
Critically-ill
Which of the following statements best defines the concept of pharmacokinetics?
A. The study of the biochemical and physiological effects of drugs on the body
B. The study of the interactions that occur between the body and a drug following administration
C. The study of how the body absorbs and metabolises drugs, how long this takes and how the drugs are eliminated from the body
A. Incorrect. This is the definition of pharmacodynamics.
B. Incorrect. This is the definition of pharmacology.
C. Correct.
Pharmacokinetics can be defined as the study of the mechanisms of absorption and distribution of an administered drug, the rate at which a drug action begins and the duration of the effect, the chemical changes of the substance in the body and the routes of elimination.
The literature on basic pharmacokinetics may convey an impression that it is an exact science governed by rules and definitions. If the underlying laws can be elucidated, the disposition of a drug in any patient should be predictable. There is, however, a very wide interindividual variability in the way anaesthetic agents are dealt with by the body. This session will look at how pharmacokinetics varies in different patient groups and why.
Which of the following factors would you expect to influence drug absorption, distribution, metabolism and elimination in neonates and infants?
A. Total body water
B. Enzyme activity
C. Renal function
D. Hepatic blood flow
E. Amount of body fat
F. Protein binding
A. Correct.
B. Correct.
C. Correct.
D. Correct.
E. Correct.
F. Correct.
All of these factors influence pharmacokinetics in extremely young patients.
Which of the following factors would you expect to be relevant in the pharmacokinetics of a pregnant woman?
A. Hepatic blood flow
B. Hormone levels
C. Placental enzymes
D. Cardiac output
E. Protein binding
A. Correct.
B. Correct.
C. Correct.
D. Correct.
E. Correct.
All of the following factors are pertinent when considering pharmacokinetics in pregnant patients:
Hepatic blood flow
Hormone levels
Placental enzymes
Cardiac output
Protein binding
This part of the session will discuss how they are relevant.
A lady in her 39th week of pregnancy has gestational diabetes. Which, of the following statements, is correct regarding pharmacokinetics in this patient?
A. Gastric motility is unaffected due to an increase in serum progesterone levels
B. A decrease in serum albumin levels increases the free fraction of local anaesthetic drugs
C. Substances produced by the placenta metabolise hormones and drugs
D. Clearance of all drugs is enhanced by an increase in renal blood flow
A. Incorrect.
B. Incorrect.
C. Correct.
D. Incorrect.
Absorption in pregnant patients is affected by gut motility and has been related to increased progesterone levels and changes in the relative amounts of gastric acid and mucous secretion.
Gastric emptying is also delayed and this leads to increased uptake of drugs absorbed in the stomach and decreased uptake of drugs absorbed in the upper part of the intestines.
An increase in total body water and fat stores increases the volume of distribution. This may lead to lower total plasma concentrations with pre-pregnancy doses of drugs.
Protein binding of drugs is variable during pregnancy. Albumin levels are reduced due to plasma dilution. The binding of acidic drugs to albumin is also affected by the increase in free fatty acids which compete for alkaline binding sites. This leads to a larger free fraction of the drug.
Alkaline drugs, such as local anaesthetics and opioids, bind to α-acid glycoprotein whose concentrations are fairly consistent during pregnancy.
Metabolism
Metabolism in pregnant patients is affected by the increase in cardiac output. The clearance of many drugs may be enhanced because of this, as well as the increase in hepatic blood flow. The increase seen in enterohepatic circulation may lead to potentiation of certain drugs. The enzymes produced by the placenta are responsible for metabolism of various neurotransmitters and endogenous compounds.
Of note, is the production of placental lactogen which degrades insulin causing decreased sensitivity to insulin.
Elimination
The increased volume of distribution seen with certain drugs leads to prolongation of their half-lives, as clearance remains unchanged.
When considering pharmacokinetics in obese patients, which of the following is most important?
A. The ratio of adipose tissue to total body mass
B. The relative reduction in total body water
C. The relative reduction in muscle mass
A. Correct.
B. Incorrect.
C. Incorrect.
All these are true, but the most important is the increase in the percentage of adipose tissue.
Which of the following describe changes in pharmacokinetics in the obese population?
A. Drug absorption is increased in obesity
B. Volume of distribution of lipophilic drugs is increased
C. Serum albumin level decreases
D. Body mass index differentiates between adipose tissue and muscle mass
E. Considering actual body weight leads to overestimation of clearance
A. Incorrect.
B. Correct.
C. Incorrect.
D. Incorrect.
E. Correct.
The variation in drug response seen in this population is partly due to changes in pharmacokinetics.
The following links provide further details:
Absorption
Changes in absorption in obese patients could be expected because of:
Increased body surface area
Increased cardiac output
Increased gut perfusion 2
However, despite the theoretical rationale for altered absorption in obese patients, there is no difference when comparing oral drug absorption in obese patients with non-obese patients
Distribution
There are changes in drug distribution in obese patients because:
The increase in adipose tissue mass will increase the volume of distribution of drugs with lipophilic properties 4
The kinetics of hydrophilic drugs is also affected by an increase of organ mass, lean body mass and blood volume in obesity
Sub-therapeutic or supra-therapeutic concentrations can occur due to the physiologic changes which influence the volume of distribution of administered drugs
Metabolism
It is worth considering Phase I and Phase II metabolism, as well as liver function in obese patients:
Phase I - metabolism can remain unchanged or can increase
Phase II - drug biotransformations are increased leading to suboptimal serum concentrations of drugs metabolised by this route. There is an increased metabolism of lorazepam and oxazepam which undergo conjugation which leads to decreased sedation
Liver function - changes in liver function are not routinely seen despite fatty changes in the liver observed in obese patients
Elimination
Three factors to consider are:
Renal clearance correlates with creatinine clearance which is altered compared with non-obese patients
Use of conventional clearance equations may be inaccurate in the obese
Overestimation or underestimation of clearance can occur in obesity when considering actual body weight versus ideal body weight respectively
Which of the following statements are correct with regard to absorption in critically ill patients?
A. Absorption of drugs by the enteral route is decreased in a state of shock
B. Intestinal enzyme activity is always maintained
C. Absorption of enterally administered drugs is enhanced by the use of opioids
D. Phenytoin bioavailability is decreased when administered together with enteral nutrition
A. Correct.
B. Incorrect.
C. Incorrect.
D. Correct.
The bioavailability of orally or enterally administered drugs is highly variable, and usually significantly reduced in the critically ill.
Usually, the preferred route of administration is intravenous because of predictable bioavailability.
The pathophysiological changes caused by critical illness include:
Perfusion abnormalities
Decreased perfusion of the gastrointestinal system occurs due to redistribution of blood flow to vital organs.
Thus there is a reduction in the absorption of drugs from the periphery and intestines.
Intestinal atrophy
Intestinal atrophy can occur in the critically ill patient as a result of starvation 5.
The enzymatic activity may also be reduced thus further impairing absorptive function.
Motility dysfunction
Gut dysmotility can be due to early hypoperfusion. The use of opioid analgesics may further impair GI motility.
This leads to impaired absorption of enterally administered drugs.
Which of the following statements is correct regarding absorption of drugs?
A. The intramuscular route is reliable in neonates
B. Obesity markedly decreases enteral absorption of drugs
C. The intravenous route of administration is preferred in the critically ill as it achieves 90% bioavailability
D. Hyperperfusion of gut in septic shock leads to increased absorption
E. Opioid analgesics may lead to impaired absorption of enterally administered drugs
F. The transrectal route of drug administration is a reliable route
A. Incorrect.
B. Incorrect. There is no difference between obese and non-obese population.
C. Incorrect. Intravenous route achieves 100% bioavailability.
D. Incorrect. Hypoperfusion occurs in septic shock.
E. Correct.
F. Incorrect.
Which of the following statements is correct regarding the metabolism of drugs?
A. Phase I reactions are decreased in obesity
B. Changes in hepatic blood flow in critically-ill patients affects metabolism
C. Hepatic enzyme activity is increased in the critically-ill
D. Enzyme activity is mature in a term neonate
E. Phase II metabolism in a neonate is unchanged for all the drugs
A. Incorrect. Phase I reactions remain unchanged.
B. Correct.
C. Incorrect. Enzyme activity may vary in the critically-ill.
D. Incorrect. Enzyme activity is immature at birth.
E. Incorrect.
A 45-year-old female patient is admitted to the intensive care unit in septic shock. Which of the following statements best describes pharmacokinetics in this patient?
A. Delayed enteral feeding causes intestinal mucosal hypertrophy
B. Starting stress ulcer prophylaxis does not interfere with drug absorption
C. Increase in capillary permeability decreases the volume of distribution
D. Decreased protein leads to an increase in unbound fraction of all drugs
E. Hepatic blood flow decreases in the hyperdynamic stage of shock
F. Acute phase proteins decrease the hepatic enzyme activity
A. Incorrect. Delayed feeding causes atrophy.
B. Incorrect. pH changes in the stomach can affect absorption.
C. Incorrect. Increase in capillary permeability leads to an increase in volume of distribution of hydrophilic drugs.
D. Incorrect. Decreased protein leads to an increase in unbound fraction of protein bound drugs.
E. Incorrect. Hepatic blood flow increases in the hyperdynamic state.
F. Correct.
pH changes
The change in pH seen in the critically-ill patient affects the ionized state of drugs.
The non-ionized fraction of the drug is known to penetrate cellular membranes more effectively and the concentration of this fraction of the drug can affect the extent of distribution of the drug.
Fluid shifts
Administration of fluids in the critically-ill can cause leakage of large volumes into the interstitium.
The contributing factors can be an increased capillary permeability and decreased oncotic pressure which are seen in septic states. This increases the volume of distribution of hydrophilic drugs such as aminoglycosides.
The changes in blood concentration seen as a result emphasizes the need to monitor drugs, with narrow therapeutic indices, closely.
Protein binding
The concentration of plasma proteins may decrease in the critically-ill.
The unbound fraction of a protein bound drug increases as the protein concentration decreases.
This increases the volume of distribution of the drug.
Hepatic metabolism is dependent upon:
Hepatic blood flow (HBF)
Enzyme activity
Protein binding
Hepatic extraction ratio is dependent on these variables and knowledge of the ratio is useful in predicting changes in drug metabolism.
The metabolism of drugs dependent on hepatic blood flow (extraction ratio >0.7) is affected by the status of the cardiovascular system. An increase in hepatic blood flow, as seen in the early phases of sepsis, increases clearance. The reverse is true in conditions with poor perfusion 6.
Drugs with low hepatic extraction ratio are dependent on metabolic enzyme activity for their clearance. The enzyme activity is decreased by the cytokines and acute phase proteins. A diet with increased protein is known to increase enzyme activity and hence clearance.
Many drugs and their metabolites are eliminated by the renal route.
Renal dysfunction is common in critically ill patients, which has clinical implications for drugs or active metabolites that are eliminated through the kidneys.
The dose modifications for patients with renal dysfunction are widely available (e.g. manufacturer’s information, British National Formulary). Frequent blood concentration monitoring of drugs is required for drugs which need minimum drug levels to be effective (such as antimicrobials) or have narrow therapeutic indices.
Which of the following is correct regarding the distribution of drugs?
A. The volume of distribution of lipophilic drugs is increased in obesity
B. A relatively low volume of total body water in neonates affects distribution
C. Concentration of ionized drug changes in all critically-ill patients
D. Decreased protein concentration increases the unbound fraction of drug in the obese
E. There is a decrease in the free fraction of the drug in pregnancy
A. Correct.
B. Incorrect. There is an increase in body water.
C. Incorrect. The ionized drug concentration changes with change in plasma pH.
D. Incorrect. Changes in protein concentration are not noted commonly in the obese.
E. Incorrect. There is an increase in the free fraction of the drug.
A 35-year-old male patient with a body mass index of 40 kg/m2 is to undergo laparoscopic cholecystectomy. Which of the following statements best describes the pharmacokinetics of this patient?
A. The absorption of oral premedication is enhanced
B. The volume of distribution of fentanyl is decreased
C. Fatty changes in the liver routinely affects liver function
D. Volume of distribution of propofol is unchanged
E. Intravenous fluids administered during the procedure leads to delay in elimination of lipid soluble drugs
F. Increased metabolism of lorazepam leads to decreased sedation
A. Incorrect.
B. Incorrect. There is an increase in the volume of distribution of lipid soluble drugs.
C. Incorrect. Fatty changes have minimal effect on liver function.
D. Incorrect.
E. Incorrect. Intravenous fluids have no effect on elimination of lipid soluble drugs.
F. Correct.
A 60-year-old patient is admitted to the intensive care unit with acute renal failure as a result of septic shock. Which of the following statements is correct regarding drug distribution in this patient?
A. pKa of administered drugs is affected
B. The ionized fraction of the drug affects distribution
C. Lipophilic drugs have an increased volume of distribution due to ‘third spacing’
D. Decreasing albumin levels decrease volume of distribution
A. Incorrect. pKa is not influenced by external factors.
B. Correct.
C. Incorrect.
D. Incorrect.
Give an overview of the Pharmacokinetics of Intravenous Anaesthesia, and the relevance to anaesthesia.
Describe the mechanism of ‘target control infusion’ (TCI) and the various pharmacokinetic models used
Pharmacokinetics of target controlled infusions (TCI)
Various pharmacokinetic models used in TCI practice
Practical aspects of TCI
Which of the following statements best describe a pharmacokinetic model?
A. A pharmacokinetic model is a mathematical model that can be used to predict the blood concentration profile of a drug after a bolus dose or an infusion of varying duration
B. A pharmacokinetic model is a model predicting the rates of metabolism of induction agents
C. A pharmacokinetic model is a table of infusion rates corrected for age and gender and is useful for total intravenous anaesthesia
A. Correct.
B. Incorrect.
C. Incorrect.
A pharmacokinetic model is a mathematical model that can be used to predict the blood concentration profile of a drug after a bolus dose or an infusion of varying duration.
Arterial or venous blood/plasma concentrations of a drug are measured after a bolus or infusion in a group of patients or volunteers.
Standardized statistical approaches and software are used to estimate model parameters in that population.
For most anaesthetic agents in common use there are several published models. Each model describes the:
Number of compartments and their volumes
Rate of drug metabolism or elimination
Rate of drug transfer between the different compartments
The following pages provide further information on the various pharmacokinetic models.
Which of the following statements are true regarding pharmacokinetic models used in target controlled infusion (TCI)?
Select one or more options from the answers below.
A. Marsh model incorporates age as a covariate
B. The Schnider model has a fixed central compartment volume
C. The Minto model can be used for both remifentanil and propofol
D. The Domino model is used for thiopentone TCI on ICU
E. The central compartment volume in the Schnider model is a linear function of weight of the patient
F. The Marsh model can be used on any infusion pump
G. Lean body mass is used as a covariate in the Marsh model
A. Incorrect. The Schnider model incorporates age as a covariate.
B. Correct.
C. Correct.
D. Incorrect. There is no clinical model for thiopentone TCI.
E. Incorrect. The central compartment volume is fixed in the Schnider model.
F. Incorrect. A microprocessor with an appropriate model is needed for TCI.
G. Incorrect. Lean body mass is used as a covariate in the Schnider model to calculate clearance.
Match the most appropriate TCI model to each agent:
Maitre developed a three compartmental model that is the most commonly used model for TCI alfentanil systems. The model incorporates weight, age and gender as covariates.
The model developed by Shafer is most commonly used for TCI fentanyl administration. This model has no covariates.
The three compartment Domino model is the model used for TCI ketamine administration.
For propofol, the Marsh model requires actual body weight; the more recent Schnider model requires age, height, and total body weight to calculate lean body mass.
The model most commonly used for remifentanil is a three compartment model described by Minto. Covariates include weight, height, gender and age.
Appropriate effect site targets to set initially for a spontaneously breathing 40 year old man undergoing a GA for a minor procedure are:
A. Propofol 4 ng/mL
B. Remifentanil 4 mcg/mL
C. Propofol 5 mcg/mL
D. Remifentanil 8 ng/mL
E. Propofol 2 mcg/mL
F. Remifentanil 3 ng/mL
A. False. This is the correct number, but the wrong units.
B. False. This is the correct number, but the wrong units.
C. True. Although this should then be adjusted to clinical effect and appropriate processed EEG value.
D. False. This is likely to be too high to maintain spontaneous breathing.
E. False. This target is more appropriate for a more elderly patient.
F. True. Although this should then be adjusted to clinical effect and appropriate processed EEG value.
Give an overview of Interindividual Variation in Drug Response, and the relevance to anaesthesia.
Describe the underlying mechanisms of genetic variation
Describe the genetic variations of plasma cholinesterase
Identify which cytochrome P450 enzymes show significant genetic variability
Discuss the implications of gene multiplication
Identify receptors for which genetic variation has clinical significance
Identify those drugs for which genetic polymorphism contributes significantly to therapeutic efficacy
Regarding human chromosomes:
A. 50% of DNA codes for genes
B. Pre-mRNA from introns is spliced to form mRNA
C. The human karyotype has 22 pairs of autosomes
D. The ‘start’ codon also codes for methionine
E. Each amino acid is coded for by more than one codon
A. False. The 30 000 genes are coded for by just 20% of DNA.
B. False. Pre-mRNA from exons is spliced to form mRNA.
C. True. Humans have 23 pairs of chromosones, 22 autosomal pairs and one pair of sex chromosomes.
D. True. There is just one ‘start’ codon.
E. False. Methionine and tryptophan are both coded for by just one codon.
We all have 23 pairs of chromosomes, 22 autosomal pairs and the pair that determine sex (Fig 1). This complete diploid set of chromosomes is known as the human karyotype. There are about 30 000 genes encoded across these chromosomes.
On each chromosome are many genes; each gene is represented on both chromosomes so an individual has two representations of each gene: one is inherited from each biological parent (Table 1). For each gene we therefore have two alleles. There may be several variants of these alleles. The combined expression of the two alleles determines our phenotype for that gene.
Both alleles are usually transcribed when a gene is activated. Frequently, the expression of one allele dominates and determines the inherited phenotype (the dominant allele): the other allele is then referred to as recessive. If both alleles contribute equally to phenotype, then the term co-dominant is applied to the gene.
Some of these alleles may contain abnormalities. If the gene is autosomal dominant and just one allele is abnormal (heterozygous) then the phenotype will be normal, only if both alleles are abnormal (homozygous) will the phenotype be abnormal.
In your anaesthetic practice you may encounter genetic differences that can alter a patient’s response to the drugs we use routinely.
Question: Can you think of one very important inherited condition that affects your choice of anaesthetic drugs during emergency surgery?
Question: What enzyme is affected and where is the gene coding for it located?
Succinylcholine (suxamethonium) apnoea occurs with an incidence of around 1 in 2500 for the commonest homozygous abnormality.
Plasma (or pseudo-) cholinesterase, more correctly called butyrylcholinesterase (BChE), is coded by the BCHE gene located on the long arm of chromosome 3.
In addition to pseudocholinesterase, there are other genetic polymorphisms affecting enzymes that can alter a patient’s response to the drugs we use routinely.
Question: Can you think of two examples of enzymes demonstrating genetic variation?
Question: Which family of CYP450 enzymes have particularly important genetic variants?
Two good examples are:
The enzyme that acetylates hydralazine
Certain cytochrome P450 (CYP450) enzymes
The CYP2 family: CYP2C9, CYP2C19 and CYP2D6 have significant genetic polymorphisms.
Fig 1a shows the clotting cascade.
There is very strong evidence that the considerable variation in patient sensitivity to warfarin has a genetic basis. Two of the enzymes involved in warfarin activity and metabolism show genetic variability.
Question: Which cytochrome is responsible for metabolism of S-warfarin?
Question: How does warfarin block the activity of vitamin K-dependent factors in the coagulation cascade?
CYP2C9.
Warfarin inhibits the enzyme vitamin K epoxide reductase (VKOR), specifically subunit 1 (VKORC1). This enzyme is essential for recycling vitamin K, which is an essential part of post-transcriptional carboxylation of factors II, VII, IX and X and proteins S and C (Fig 1b).
Which of the following CYP450 enzymes demonstrate important pharmacogenetic variation?
A. CYP1B1
B. CYP1C19
C. CYP2C9
D. CYP2D6
E. CYP2E1
A. Incorrect. CYP1B1 does have pharmacogenetic variants, but they are associated with susceptibility to ovarian and other cancers.
B. Incorrect. CYP2C19 shows pharmacogenetic variation but CYP1C19 does not.
C. Correct.
D. Correct.
E. Incorrect. Although it belongs to the CYP2 family of enzymes, there are no important pharmacogenetic variants.
Which of the following drugs are metabolized mainly by enzymes that demonstrate important pharmacogenetic variation?
A. Losartan
B. Mivacurium
C. S-warfarin
D. Tramadol
E. Vecuronium
A. Correct. Losartan is a pro-drug so may not be effective in patients with abnormal CYP2C9.
B. Correct. Mivacurium is metabolized by plasma cholinesterase, which shows significant genetic variation.
C. Correct. S-warfarin (the more potent enantiomer) is metabolized by CYP2C19.
D. Correct. Tramadol is converted to an active metabolite that works through opioid receptors by CYP2D6.
E. Incorrect. Vecuronium is metabolized by CYP3A4.
Regarding pharmacogenomics:
A. 75% of patients with malignant hyperthermia have a SNP in the gene for the ryanodine receptor
B. The wild-type gene for the metabolism of S-warfarin is CYP2C9*10
C. Ultrafast metabolizers of codeine have multiple copies of the CYP2D6 gene
D. Splice variants of the μ-opioid receptor are found in different areas of the CNS
E. Hypertensive patients who are hetereozygous for SNPs in both amino acids at positions 49 and 389 respond to metoprolol
A. False. About 25% of MH patients have a demonstrable SNP in the ryanodine receptor.
B. False. The wild-type always has 1 as its final designation, so CYP2C91 is the wild-type.
C. True.
D. True. Splice variants are less widespread than the normal gene product and appear to be more localized.
E. False. Those who respond to metoprolol can be heterozygous for a SNP at just one of the two contributing amino acid positions, but not both.
All of the drugs below show pharmacogenetic variation. Match the drugs to the enzymes that are responsible for their metabolism.
An 19-year-old university student presents with severe abdominal pain and is scheduled for an emergency appendicectomy. He has not eaten for 48 hours and has not been vomiting. He gives a vague history of his father having had a ‘bad reaction’ to anaesthesia and needing care on an ICU. He knows no details, but it was before he was born. His parents cannot be contacted and the surgeon wants him to go to theatre now.
A. Cancel surgery until his parents can be contacted
B. Go ahead with surgery but avoid succinylcholine
C. Arrange a blood test to look at his plasma cholinesterase activity before proceeding
D. Go ahead with surgery using a volatile-free anaesthetic machine and total intravenous anaesthesia
E. Go ahead with surgery but avoid muscle relaxants and volatile agents
Submit
You should go ahead with surgery but avoid muscle relaxants and volatile agents.
It is not uncommon for patients to report ‘reactions’ to anaesthesia and in an emergency there is always pressure to get on with surgery. The most common adverse reactions requiring ICU admission are:
Anaphylaxis, particularly to muscle relaxants, which is idiosyncratic
Malignant hyperthermia and
Succinylcholine apnoea
Malignant hyperthermia and succinylcholine apnoea are both inherited conditions. You should always contact your senior for advice.
The safest option, if considering just the history of an adverse reaction in a close family member, is to delay surgery but in this case there is a significant risk of peritonitis.
By avoiding all muscle relaxants and using total IV anaesthesia (TIVA) in a careful way, with senior assistance at hand, the risk of a significant adverse reaction is minimized.
This case went ahead as discussed, with no adverse consequences. It later transpired that the student’s father had had succinylcholine apnoea but his mother had never been tested. The entire family was subsequently offered screening and his mother found to be heterozygous for the silent gene.
Concerning genetics:
A. mRNA has just a single helix
B. tRNA is formed from A,G,T and C nucleotides
C. The result of a SNP is always a single aminoacid substitution
D. μ-opioid receptors are formed from several genes
E. Monozygotic twins have the same genotype for plasma cholinesterase
A. True. DNA is a double helix, but mRNA just a single helix.
B. False. In tRNA, as in mRNA, T is replaced by U.
C. False. SNPs can have three possible outcomes: no change in amino acid sequence, a single amino acid substitution or a ‘stop’ codon may result with premature truncation of the amino acid sequence.
D. False. There is a single gene: MOR1, but more than one promotor and several possible splice variants.
E. True. Identical (monozygotic) twins have the same genotype.
Give an overview of Halogenated Volatile Agents and Unwanted Effects of Volatile Agents, and reference and application to anaesthetic.
List the important pharmacokinetic properties of anaesthetic agents and concepts such as MAC and oil:gas solubility
Describe the structure of the halogenated volatile agents
List the pharmodynamic properties of the agents including the unwanted clinical effects
Explain the synergistic effects of anaesthetic agents and the influence of differing ages on the clinically observed effects
Halogenated volatile agents are liquids at room temperature, which easily vaporise and have a low boiling point and high saturated vapour pressure
MAC is inversely related to potency and MAC increase is inversely proportional to the oil:gas solubility
The blood:gas partition coefficient describes the solubility of an inhaled gas in blood and the lower the solubility the faster the speed of onset of the agent
The volatile agents all have some adverse physiologically effects and all pose the rare but significant risk of the patient developing malignant hyperthermia
Question: From the knowledge that the boiling point of xenon is -108°C what can you deduce about the nature of this element at room temperature?
It will all be in the vapour phase and with the additional information that the critical temperature is 16.5°C and the critical pressure is 5.84 MPa, at standard room temperature and pressure, it will all be gas.
The critical temperature of a substance is the temperature at and above which vapor of the substance cannot be liquefied, no matter how much pressure is applied.
The critical pressure of a substance is the pressure required to liquefy a gas at its critical temperature.
Which of these volatile agents is the most potent?
A. Isoflurane (MAC 1.1)
B. Sevoflurane (MAC 2.0)
C. Desflurane (MAC 6.6)
Isoflurane is the most potent; potency is inversely related to MAC and is a measure of dose of drug required to exert a clinical effect.
MAC is the minimum alveolar concentration of an anaesthetic agent that prevents movement in response to a standard skin incision in standard conditions in 50% of subjects, when breathing in 100% oxygen and in the absence of other analgesic or anaesthetic agents.
It is useful to allow comparisons between different agents and as a guide for the percentage anaesthetic required for anaesthesia. It is also a measure of potency and can be compared to the ED50, i.e. the dose of a drug which produces a biological effect.
MAC is inversely related to potency.
Potency is the ability of a drug to produce an effect. The more potent the drug the smaller the dose required.
So, considering the values in Table 1, you can see that halothane is the most potent agent, i.e. requires the smallest concentration to produce a clinical effect.
MAC is also useful clinically as it can readily be measured by anaesthetic gas analysers.
It is far from clear how anaesthetic agents work, however with the observation that MAC increase was inversely proportional to the oil:gas solubility, Meyer and Overton hypothesised that inhalational agents must work non-specifically on the lipid rich neuronal cells of the central nervous system. They also proposed that potency increases with oil:gas solubility.
More recent research suggests the theory that agents work on CNS receptors such as potentiation at GABAA, glycine receptors and potentially inhibition at MNDA receptors as well as newer evidence for the role of two-pore domain potassium channels.
A 12-year-old patient is having an elective orthopaedic procedure to her ankle, requiring a general anaesthetic. She is given an IV induction agent, a supra-glottic airway is inserted and anaesthesia is maintained on sevoflurane at a MAC of 1 (ET Sevo of 2.0).
Question: Why did the anaesthetist choose sevoflurane? Should they have chosen a different vapour?
Question: During a stimulating part of the operation she moves and coughs, why might this be?
Sevoflurane is a liquid at room temperature and can be readily vaporised in a plenum vaporiser, with a relatively high SVP of 21 kPa.
It has the advantage of being non-irritant to the airways and has a pleasant odour so is suitable for use with an LMA and because of this, it can also be used for inhalational induction. A further factor facilitating this is the low blood:gas solubility resulting in a rapid speed of onset of anaesthesia.
Arguments for the use of a different agent would be that you could consider isoflurane, which has a similarly low MAC and high SVP but is somewhat cheaper. It does however have a more pungent odour and is a respiratory suppressant causing coughing and breath holding, so is not suitable for gaseous induction.
After checking the delivery of the anaesthetic agents and finding no issues it was felt that the level of anaesthesia was insufficient. The MAC value of any of the agents is affected by numerous variables. Specific to this case MAC is affected by young age (needing an increased MAC).
MAC is additive within the group of inhalational agents, so a mixture of agents with a cumulative alveolar concentration of 1 MAC has the same effect as 1 MAC of a single agent. This is most commonly seen with the use of nitrous oxide but also apples to xenon, e.g. If 60% nitrous oxide (MAC 105%), is added to a volatile agent in 40% oxygen the volatile agent requirement is reduced by 57%.
Regarding important pharmacodynamic properties of the volatile agents:
A. The MAC required for a neonate is higher than that for a 5-year-old
B. A higher MAC is required for an acutely intoxicated patient
C. MAC is inversely related to potency
D. The use of midazolam preoperatively decreases the MAC requirement
A. False. As per the table both the elderly and neonates require a lower MAC.
B. False. Higher MAC is required for patients who have a chronic consumption of alcohol whereas an acutely intoxicated patient will have a lower requirement.
C. True.
D. True. Midazolam is a sedative drug and will therefore decrease the MAC requirement.
Properties of an ideal anaesthetic agent include:
A. Low MAC
B. High latent heat of vaporisation
C. Boiling point around room temperature
D. Low blood:gas coefficient
E. Low oil:gas coefficient
A. True. A low MAC allows delivery with a high concentration of oxygen. Compare sevoflurane (MAC 2) with N2O (MAC 104).
B. False. A low latent heat of vaporisation facilitates easy vaporisation.
C. False. The inhalational agent must be predictably vaporised at room temperature to ensure reliable delivery of gas concentration, e.g. desflurane’s boiling point is at around room temperature. Therefore, it needs its own specialised vaporiser to ensure 100% saturation of the gas flow.
D. True. A low blood:gas coefficient allows for a rapid onset and offset of anaesthesia.
E. False. A high oil:gas coefficient relates to a high drug potency and therefore a more efficacious anaesthetic agent.
Which of the agents in Table 1 would you expect to be the least potent?
A. Sevoflurane
B. Isoflurane
C. Desflurane
D. N20
E. Xenon
N20 has the lowest oil:gas solubility.
Isoflurane has the highest oil:gas solubility therefore using Meyer-Overton hypothesis this is the most potent of these agent.
The agents are inhaled by the patient via the breathing circuit and uptake is via the alveoli of the lungs, where the small volatile molecules readily pass through the alveolar membranes to enter the bloodstream. Another important concept in the speed of onset of an anaesthetic agent relates to its solubility in blood.
The blood:gas partition coefficient describes the solubility of an inhaled gas in blood (Table 1).
The blood:gas coefficient is the ratio of the concentration of agent in one solvent compared to another solvent at equilibrium.
It would be easy to assume that the greater the solubility in blood the faster the speed of onset, however the converse is true as it is the partial pressure of agent in the blood and subsequently the brain (PB) which produces a more rapid onset of action (and offset).
Simply put, the anaesthetic agents exert their effects on brain tissue and the less soluble the agent is in blood the greater the partial pressure of gas which is available to exert its effect on the brain.
The higher the blood:gas coefficient the higher the uptake of gas that is required, i.e. there will be a longer induction time. Thus, it follows that the lower the blood:gas coefficient, the faster the onset of action.
A wash-in curve (Fig 1) illustrates that the lower the blood:gas coefficient, the faster the volatile agent exerts its clinical effect. Note clinical effect would be expected at 1.0 MAC.
Whereas the wash-out curve (Fig 2) shows the converse; the lower the blood:gas coefficient the faster the offset time and the quicker the patient will regain consciousness.
Place these anaesthetic agents at the correct point on the graph.
Regarding malignant hyperthermia:
A. Susceptibility to MH is carried by an autosomal recessive gene with variable penetrance
B. It can only be triggered in susceptible patients by volatile agents
C. The initial dose of dantrolene in the immediate management of MH is 2.5 mg/kg by IV bolus
D. It is carried on a gene on the X chromosome
E. The first clinical sign is a steady rise in the patient’s temperature
A. False. Autosomal dominant.
B. False. It can also be triggered by suxamethonium
C. True. And in a 70 kg man this requires 9 vials for the initial dose; each being 20 mg and being mixed with 60 ml of sterile water for injection.
D. False. It is carried on a gene on chromosome 19.
E. False. The first clinical signs are tachycardia, increasing ETCO2 and increased oxygen consumption. Pyrexia is a late sign.
Give an overview of Nitrous Oxide and Xenon, and the relevance to anaesthetics.
Describe the production, purification and storage of N2O
Define the relevant physicochemical properties of both N2O and xenon
Describe the Concentration Effect and the Second Gas Effect
List the side-effects, potential for toxicity and contraindications of N2O
State the similarities and differences between N2O and xenon
Both N2O and xenon are general anaesthetic agents
The values of the physicochemical properties for N2O should be committed to memory
The Concentration Effect accounts for the more rapid rise of FA/FI for higher as opposed to lower concentrations of N2O
The Second Gas Effect is caused by the Concentration Effect
Xenon is produced by fractional distillation of liquid air, a by-product of oxygen manufacture
Xenon costs approximately 2000 times that of N2O, hence its slow introduction into mainstream anaesthetic practice
N2O is most commonly produced by heating ammonium nitrate (NH4NO3) to 250oC.
Question. N2O is not the sole product derived from using this heating process. Can you name other ones?
The other products derived from this process are:
NO (nitric oxide)
NO2 (nitrogen dioxide)
NH3 (ammonia)
N2 (nitrogen)
HNO3 (nitric acid)
H2O (water)
The temperature of the reaction has to be very carefully controlled to minimise N2O contamination. These toxic impurities are removed by cooling and passing the raw products through alkaline gas washes. During this process, NO and NH3 are also removed but this occurs by acidic washes.
Regarding critical temperature:
A. This is the temperature above which it is not possible to return N2O to a liquid, regardless of the pressure
B. This definition should be quoted at one atmosphere
C. Above 36.5 oC N2O is defined as a vapour
A. Correct. Well done. This is the definition of critical temperature.
B. Incorrect. Critical temperature is the temperature above which it is not possible to return N2O to a liquid regardless of the pressure.
C. Incorrect. Above its critical temperature N2O is a gas. A vapour can be compressed to a liquid but a gas cannot.
For a saturated vapour pressure of 5200 kPa:
A. The gauge pressure of a cylinder of N2O should therefore be 5.2 atmospheres
B. This value falls in line with temperature
C. This may be defined as the pressure of a vapour at equilibrium that exists above its liquid and is independent of temperature
A. Incorrect. It should be 52 atmospheres or 52 bar at 20oC. (100 kPa = 1 bar = 1 atmosphere).
B. Correct. SVP is usually quoted at 20oC. The value will alter with temperature.
C. Incorrect. It is dependent on temperature – otherwise this definition is correct.
Critical temperature
This is the temperature above which it is not possible to return a substance into a liquid regardless of the pressure applied. Above its boiling point (-88.5oC) N2O is a vapour and with sufficient pressure may be ‘squashed’ into a liquid state but once the critical temperature (36.5oC) is exceeded the liquid state becomes impossible. Of academic note, N2O is therefore inhaled as vapour but exhaled as a gas.
Saturated vapour pressure
This is the pressure that exists above its liquid phase at equilibrium and is dependent on temperature, but independent of other gases present. The pressure gauge on a cylinder of N2O indicates its saturated vapour pressure which is constant while the cylinder contains N2O liquid.
The filling ratio is the ratio of the mass of liquid in the cylinder compared with the mass of water that the cylinder can hold. In temperate regions this is 0.75, while in the tropics it is 0.67. Why is the filling ratio higher in temperate regions?
Select one answer from the options, then select Submit.
Possible answers:
A. The cylinders are smaller
B. The temperature in temperate regions never rises above the critical temperature of N2O
C. The cylinders are made to a higher standard and can cope with the higher filling ratio
A. Incorrect. The size of the cylinders has nothing to do with the filling ratio.
B. Correct. If a full cylinder of N2O with a filling ratio of 0.75 reaches >36.5 ° C, all the N2O would convert to gas and exceed the pressure that the cylinder is designed to cope with. This would lead to an explosion. When the filling ratio is reduced to 0.67, even at temperatures >36.5 ° C, the cylinders are able to withstand the pressure of N2O gas.
C. Incorrect. The cylinders used in the tropics are made to the same exacting standards.
Regarding N2O:
A. It is not possible to anaesthetize anyone with N2O alone
B. 103 kPa of N2O prevents patients moving to a surgical stimulus
C. One MAC of N2O prevents patients from waking up during a simple operation
D. N2O is not very potent
A. Incorrect. It is not possible to anaesthetize anyone with N2O alone at one atmosphere, but in a pressurized chamber it is possible.
B. Incorrect. 103 % of N2O prevents 50 % of patients moving to a standard surgical stimulus.
C. Incorrect. The effects of one MAC refer to movement, not waking up.
D. Correct. Well done. A high MAC value indicates low potency while a low MAC value indicates high potency, e.g. isoflurane’s MAC is 1.17 % at one atmosphere.
MAC = 103 %
MAC is defined as the minimum alveolar concentration required to prevent 50 % of patients moving to a standard surgical incision, at one atmosphere.
The MAC value of N2O is 103 %. At first glance it therefore appears impossible to produce anaesthesia with N2O alone because using 100 % N2O is clearly a hypoxic mixture, never mind the extra 3 % required.
However, thinking in ‘percentages’ can result in missing the point because you should be thinking in ‘percentages of one atmosphere’ which leads to partial pressure - the key. Partial pressure is independent of atmospheric (or ambient) pressure so that for N2O it can be seen that 103 kPa are required for anaesthesia.
So it is possible to produce anaesthesia with N2O alone, you just need a hyperbaric chamber to allow 103 kPa of N2O and some additional oxygen.
Remember, for any anaesthetic agent, anaesthesia results from exposure to a partial pressure (which is independent of atmospheric or ambient pressure) rather than a percentage (which varies with atmospheric or ambient pressure).
Regarding blood: gas solubility coefficient:
A. The blood: gas solubility coefficient of N2O is much lower than other commonly used inhaled agents
B. A high blood: gas solubility coefficient results in a fast onset of action because agent gets into the blood quickly and can therefore get to the brain quickly
C. Ether has a blood: gas solubility coefficient of about 12. This means that at equilibrium the amount of ether in the blood will be 12 times that in the gas phase
A. Incorrect. Its value is 0.47, which is only slightly lower than other commonly used agents.
B. Incorrect. Agents with a high blood: gas solubility take a long time to generate a sufficient partial pressure so therefore have a slow onset of action.
C. Correct. Ether’s blood: gas solubility coefficient is high (12) resulting in the proportions as described.
Blood: gas solubility coefficient = 0.47
The blood: gas solubility coefficient is defined as the ratio of the amount of anaesthetic in blood and gas when the two phases are of equal volume, pressure and in equilibrium at 37 ° C. So in relation to N2O, roughly half as much exists in the blood phase compared to the gas phase.
A value of 0.47 may be regarded as low (however all modern anaesthetic agents have low values).
A low value results in a rapid onset of action as it equilibrates rapidly with the pulmonary capillaries and what little enters the blood generates a high partial pressure causing a rapid effect.
Agents with high values may enter the blood rapidly but the blood acts like a bottomless pit and so it takes a long time for the agent to reach a sufficient partial pressure to have an effect.
Entonox ® is a 50:50 mixture of N2O and O2. It takes on certain properties that mark it out as distinct from the properties of its constituents. This is called the Poynting effect.
The temperature below which Entonox ® separates back into N2O and O2 is pressure dependent and is known as the pseudocritical temperature.
It is stored at 137 bar in cylinders, but may also be delivered via pipes in hospitals at 4.1 bar.
Question: If a cylinder of Entonox ® were to fall below its pseudocritical temperature, would the gas initially withdrawn contain more O2 than N2O or more N2O than O2?
It would initially contain more O2 than N2O. This is because N2O would come out of Entonox ® as a liquid, while the O2 would remain as a gas. As time progresses, the mixture will contain less oxygen and may then eventually become hypoxic.
In which of the following situations is Entonox ® a useful analgesic?
A. Labour
B. Dressings change
C. Post major joint replacement
D. Fractured ribs
E. Removal of drains
A. True. This is due to its fast onset.
B. True. It is useful on wards to assist in painful procedures of short duration.
C. False.
D. False. There are two reasons Entonox ® is not suitable. There may be underlying pneumothorax which would get worse. Secondly, fractured rib pain is not short-term.
E. True. As with dressings change, Entonox ® can be used to assist pain relief.