Drugs Flashcards

1
Q

Describe the A-E effects of propofol

A

A - loss of laryngeal reflexes and airway muscle tone

B - respiratory depression, reduced response to hypoxia/ hypercapnia, bronchodilator by attenuating vagal bronchoconstriction

C - balanced arterial and venous vasodilation. Not a negative inotrope at clinically relevant doses, unclear whether it reduces cardiac output - traditional stance is that it does.

D - Sedation, anti-epileptic, reduces cerebral metabolic demand and ICP, anti-emetic (D2 receptor antagonism), can cause myoclonus/ opisthonus induction/ emergence phenomena

E - heat loss due to vasodilation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Describe the structure of propofol

A

A phenol ring with two propyl groups attached to 2nd and 6th carbons (2,6-diisopropylphenol)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Describe the pharmacodynamics and kinetics of propofol

A

Comes in a soybean emulsion for two reasons: raw propofol freezes at 19 degrees, and it is very insoluble in water

pKA 11.1, highly lipophillic, a weak acid

Its bolus VoD is 4L/kg, and steady state VoD is 2-10L/kg

It is highly protein-bound in plasma, but is even more lipophillic and so the vast majority of it distributed to fatty tissues such as the brain.

Clearance is hepatic via CYP450, at a rate of 30-60ml/kg/min. All metabolites are inactive and excreted renally

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

What is the mechanism of action of propofol?

A

GABA-A agonist, activating the cell membrane channels which allows chloride influx and hyperpolarizes the membrane

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

Describe the effects of opioids on respiration

A

There are three main effects:

1) Direct blunting of inherent respiratory drive by action at Bötzinger’s complex in the ventral medulla

2) Blunting of response at medullary chemoceptors to acid-base changes caused by hypercapnia

3) Blunting of response at carotid/aortic chemoceptors to hypoxia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

What is the generic receptor action of opioids?

A

They activate G-protein coupled receptors (mostly Mu) which inhibits adequate cyclase, reducing cAMP. This closes voltage-gated calcium channels and open potassium channels to cause potassium efflux and hyperpolarisation. The overall effect is inhibitory

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Describe the adverse effects of suxamethonium

A

Fasiculations/ post-op muscle pain
Relatively high risk of anaphylaxis
Malignant hyperthermia
Bradycardia
Raised compartmental pressure (eye, cranium but only 2-3mmHg, and abdomen which is more concerning for patients with reflux risk)
Hyperkalaemia (mass efflux of potassium due to synapse depolarisation)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

What is the structure of suxamethonium?

A

Two molecules of acetylcholine joined together by their acetyl groups

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

What are the two structural classifications of neuromuscular blockers?

A

Aminosteroids/ “curonium”drugs

Benzylisoquinolinium diesters/ “curium” drugs

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Compare rocuronium reversal with neo/glyo and sugammadex

A

Glyco/neo can’t reverse a high grade block (only works if at least 2 twitches present), and if longer acting NMBs are used there is a risk the still-circulating NMB will re-block
Sugammadex is rapid onset, able to reverse any degree of block
There is a risk of anaphylaxis (0.3%) and a small chance of bradycardia but sugammadex is otherwise free of side-effects. Glyco/neo causes tachycardia in addition to anti-muscarinic side effects, and takes a few minutes to work. However its risk of anaphylaxis is negligible
It effectively prevents use of rocuronium again if re-paralysis is required within a short time

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Why do more potent agents have slower onset of action?

A

The effective dose is lower, hence the concentration gradient between plasma and effector site is lower and diffusion slower.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

List 3 factors that would prolong neuromuscular blockade

A

Advanced age (for rocuronium this could equate to 50 minutes for block to wear off instead of 25)
Hepatic impairment (renal impairment for pancuronium)
Myasthenia gravis
Aminoglycosides, furosemide, volatile anaesthetics, calcium channel blockers (all decrease acetylcholine concentration at synapse, and therefore decrease competition with blocker)
Hypermagnasaemia, hypocalcaemia, and hypokalaemia all prolong the block
Acidaemia and hypothermia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Which of the following regarding NMBs is true?

A) 50% of AChR must be occupied for NMB to be evident
B) Neo/glyco can reverse a total block
C) DBS provides more accurate visual/tactile assessment of fade than ToF
D) 80% AChR blockade abolishes all twitches
E) A ToF ratio of 0.5 gives visually appreciable fade

A

C) DBS provides more accurate visual/tactile assessment of fade than ToF

DBS gives two sequences of supramaximal stimulation in quick succession, and provides a more accurate visual/tactile assessment of fade than ToF. However quantitative measures such as an accelerometer are much more accurate.

At least 70% of ACh receptors must be occupied by a NMB for the block to be evident.

Neo/glyco will have no effect until two twitches are present i.e. it can’t reverse a total block.

There is a lot of redundancy in AChR numbers - total block requires >90% of receptors to be blocked.

ToF ratio/ fade is not visually appreciable above 0.4.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

Why is suxamethonium dangerous in patients with: burns, severe trauma, prolonged immobility/ ICU stay, spinal cord transection, and neuropathies?

A

These conditions feature up-regulation of ACh receptors, therefore when sux is given and there is potassium efflux, it may be more pronounced in these populations so the risk of cardiac events is increased.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

What is the risk of anaphylaxis with NMB drugs?

A

1 in 6000-20,000

Suxamethonium is generally accepted to have the highest risk at 1:9000 (for context the risk of sux apnoea is 1:1800)
Rocuronium has the highest risk of the non-depolarising NMBs, some large studies even have it as higher risk than sux (NAP6 said 1:17,000)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Compare the following characteristics of neuromuscular block between depolarising and non-depolarising agents:

ToF ratio
Post-tetanic potentiation

A

ToF ratio for a one-off dose of suxamethonium should be >0.7/ there should be no fade. Fade and post-tetanic potentiation are features of phase 2 depolarising block which occurs after prolonged or repeated use. However both of these features occur after one-off doses of non-depolarising blockers

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

Summarise the key pharmacokinetic differences between fentanyl and alfentanil

A

Fentanyl and alfentanil are both synthetic opioids with rapid onset used during induction of anaesthesia and maintenance of sedation in ITU.

The key differences lie in their pKa, lipophilicity and hence VoD, potency, and elimination.

Alfentanil has a pKA of 6.5 and fentanyl has a pKA of 8.4. At physiological pH this means alfentanil is 90% unionised compared to fentanyl which is 9% unionised. This means alfentanil crosses the BBB faster despite being less lipophillic. Additionally alfentanil is only 20% as potent as fentanyl, so it has a faster onset.

Fentanyl is 580 times more lipid soluble than morphine, whereas alfentanil is only 90 times. I.e. fentanyl is ~6 times more lipid soluble than alfentanil. This is reflected in VoD: 0.4-1L/kg for alfentanil, but 3-5L/kg for fentanyl.

Clearance of alfentanil is much higher than that of fentanyl: 409ml/kg/min vs. 1ml/kg/min.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Summarise the general pharmacokinetics of aminosteroid NMBs, including metabolism and excretion for rocuronium, vecuronium, and pancuronium

A

Nil oral absorption

All have poor lipid solubility, the bisquaternary more than the mono quaternary, and so a small volume of distribution (essentially just the extracellular fluid) and don’t cross the BBB or placenta.

Metabolism and excretion are as follows:

  • Pancuronium is very insoluble in lipid and so 60% is excreted in urine. the rest is metabolised in the liver w biliary excretion. 5-10% of the dose is metabolised to an active hydroxy metabolite with 50% of the potency
  • Vecuronium is mostly excreted in bile, with up to 30% being excreted unchanged in urine. A further 20% is deacetylated in the liver, with a very small amount of active metabolites being produced (with 80% vec’s potency)
  • Rocuronium is similar to vecuronium in that most undergoes biliary excretion and ~30% is excreted in urine unchanged. However very little of it is metabolised in the liver, and the small amount of active metabolites produced has 5% the potency of roc.
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

Describe the mechanism of action of non-depolarising NMBs

A

Aminosteroid NMBAs compete with acetylcholine for binding to the alpha-subunit of the nicotinic receptor

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

Which aminosteroid NMB causes vagal suppression and sympathetic stimulation?

A

Pancuronium

it inhibits noradrenaline reuptake post-synaptically, and suppresses vagal post-ganglionic fibres

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What is the intubating dose and half-life of atracurium?

A

0.5mg/kg
Onset ~2 minutes, though peak may be more like 3

Half-life is 50 minutes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What is the unique feature of atracurium’s elimination?

A

Its metabolism is split 45:45 (10% is renally excreted unchanged):

45% spontaneously degrades to inactive metabolites at normal body temperature and pH (Hoffman elimination)

45% undergoes hydrolysis by tissue esterases

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

How does cisatracurium differ from atracurium?

A

Cisatracurium is one of 10 possible isomers of atracurium, and has the C1-R-N-cis for each end of the molecule. It comprises around 15% of atracurium.

It is a little more potent so slower onset but produces only 10% as much laudonasine metabolite
Similar half-life
75% undergoes Hoffman elimination, 15% renally excreted, minimal hydrolysis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

Why do patients with significant liver failure require higher doses of non-depolarising NMBs?

A

Hepatic failure causes fuild retention which increases the volume of distribution of NMBs

These patients are also at risk of prolonged block when using NMBs that are metabolised by plasma cholinesterases as these are produced in the liver

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
How does mivacurium differ from atracurium/ cisatracurium?
MIvacurium is the most potent of the three and so has the slowest onset, while also having a very rapid offset. This is because its metabolised by plasma cholinesterases, similarly to suxamethonium
24
Which of the following would increase the speed of onset of a volatile anaesthetic agent? A) High cardiac output B) High functional residual capacity C) High blood:gas partition coefficient D) High potency E) Co-administration of nitrous oxide
E) Co-administration of nitrous oxide Speed of onset of volatiles is related to how quickly an alveolar:plasma concentration gradient can be built up, as a higher concentration gradient is the most influential factor causing fast diffusion. High CO will delay build-up of volatile in the alveoli, and a high FRFC provides a large reservoir into which volatile distributes thereby diluting its alveolar concentration. A high blood:gas partition coefficient reflects higher water-solubility essentially. This leads to slower diffusion into target site as less of a concentration gradient builds up.
25
What is the difference between blood:gas partition coefficient and oil:gas coefficient?
blood:gas partition coefficient - describes the solubility of a particular gas in blood, which is essentialy hydrophilicity oil:gas coefficient - essentially just lipophilicity which, for volatile anaesthetics, equates to potency
26
What are the properties of an ideal volatile anaesthetic agent?
Physical properties: Liquid at room temperature Stable and inert Cheap Environmentally safe Pharmacological properties: No airway irritation or respiratory depression No cardiovascular instability Analgesic, not epileptogenic, doesn't increase ICP Low blood:gas partition coefficient High oil:gas coefficient/ potency Not metabolised
27
Define the following terms: Latent heat of vaporisation Sustained vapour pressure Critical temperature
Latent heat of vaporisation - the energy required for a molecule to transition from liquid to gas without an increase in temperature Sustained vapour pressure - the pressure at which (for a given temperature) a vapour will be in equilibirum of movement between states with its liquid form Critical temperature - the temperature at which a substance is completey gaseoues irrespective of pressure
28
Summarise malignant hyperthermia
MH is an autosomal dominant condition with variable penetrance that causes mass metabolic activation and muscle breakdown leading invariably to death in the absence of treatment. Incidence is 1 in 10,000, and the mechanism is activation of the ryanodine receptor leading to uncontrolled calcium release from the sarcoplasmic reticulum of myocytes. This can be triggered by any volatile anaesthetic agent, or suxamethonium. Initial signs are rising EtCO2 and tachycardia with muscle rigidity. The patient will develop an oxygen requirement from the increased metabolic demand, and will eventually develop pyrexia and a mixed acidosis Treatment is: Stopping the causative agent and hyperventilate with high flow pure oxygen via a clean circuit. Maintain anaesthesia with TIVA and end the surgery ASAP, achieve muscle relaxation with a non-depolarising NMB. Actively cool, treat electrolyte abnormalities/ arrhythmias. Bolus dantrolene at 2.5mg/kg (9 vials for the average 70kg male). Each vial of 20mg should be reconstituted in 60ml sterile water. Repeat boluses at 1mg/kg up to a maximum of 10mg/kg NB previous uneventful anaesthesia doesn't rule out MH.
29
Give the blood:gas partition coefficient and oil:gas coefficient for each of: sevofluorane, isofluorane, and desflurane
Sevo: 0.65 - 47 Iso: 1.4 - 91 Des: 0.45 - 26
30
Describe the A-E effects of ketamine
A - preservation of airway reflexes, hypersalivation so may provoke laryngospasm especially in children B - broadly preserves respiratory drive though may cause transient apnoea on induction, Useful as a bronchodilator e.g in I&V asthma exacerbations C - increase in sympathetic drive leads to increased preload, SVR, BP, and cardiac output. Is a direct negative inotrope but its dose-dependent sympathomimetic properties almost always outweigh this. D - produces dissociative anaesthesia and is an excellent analgesic. Increases cerebral blood flow and metabolic rate. Traditionally considered to increase ICP - this is now believed to be untrue or a small rough effect as to be irrelevant. Causes emergence phenomenae in ~20% of patients.
31
Describe the effects of ketamine in chronic opioid users
It reduces down regulation of opioid receptors, and so reduces opioid tolerance and hyperalgesia. This is accomplished by disrupting interactions between NMDAr and PSD-93 & 95 scaffolding proteins, which are part of a NOS synthase->protein kinase C pathway that further up-regulates NMDAr and leads to opioid tolerance.
32
Give 3 contraindications to flumazenil
Chronic benzo use Epilepsy/ propensity to seizures Possible mixed overdose with TCAs, as this increases risk of cardiac dysrhythmia with flumazenil
33
Describe the mechanism of action of benzodiazepines
They bind to a site on the GABA receptor adjacent to GABA. This increases the affinity of GABA for its receptor, leading to more activation and an increased frequency of opening of the chloride channel
34
Describe the A-E effects of midazolam
A - reduces upper airway tone and suppresses reflexes B - respiratory depression and blunted response to hypercarbia C - obtunds hypertensive response to laryngoscopy, arterial and venous dilatation, reduced cardiac output D - anxiolysis->sedation->hypnosis, anterograde amnesia, anticonvulsant, muscle relaxation (effect on dorsal horn of spinal cord), reduced REM sleep E - reduces hepatic and renal flow
35
Describe the pharmacokinetics of ketamine
Ketamine is presented as a colourless solution available as 1%, 5%, or 10%. It contains a mixture of S and R optical stereoisomers. It has poor oral bioavilaibilty (20-25%) but can be given orally. It is used IV and IM for anaesthesia, sedation, and analgesia. The dose for anaesthesia is 0.5-2mg/kg IV or 4-10mg/kg IM. The dose for analgesia is 0.25mg/kg IV, and the dose for sedation is up to 0.75mg/kg IV Ketamine is a non competitive NMDA receptor antagonist. Its VoD is reasonably large as 3L/kg. It is very rapid in onset owing to reasonable lipophilicity, pKA of 7.5, and small molecule size. Metabolism is hepatic to active metabolites, which are then conjugated and excreted in urine. Distribution half life is 10 minutes but elimination half life is 2-3 hours.
36
List the A-E effects of thiopentone
A - laryngeal reflexes are generally not suppressed, so there is a risk of laryngospasm when using SADs B - causes apnoea and the standard suppression of response to hypoxia and hypercarbia. However patients will breathe more quickly after thiopentone than propofol, making it more ideal for RSI in the event of failure. C - mostly causes venodilation and reduced preload -> reduced cardiac output. SVR is generally preserved, and at high doses it is a direct negative inotrope. Will cause a reflex tachycardia, and negative inotropy is more common with high rapid doses in the elderly, critically unwell, hypovolaemic, hypoalbuminaemic, and those with ischaemic/valvular heart disease. D - provides extremely quick anaesthesia (one arm brain circulation time). It is less protein bound than propofol which contributes to it having faster onset because concentration between central compartment and effect site equilibrates faster. Reduced ICP and cerebral metabolic rate. Cerebal blood flow and CO2 is preserved by autoregulation. Anticonvulsant and proven neuroprotective in focal ischaemia. E - causes enzyme induction after just one dose. Can cause tissue damage if it extravasates due to alkaline pH, and is catastrophic if given arterially.
37
Summarise the pharmacokinetics of thiopentone
A lipophillic anaesthetic agent given IV at a dose of anywhere from 3-7mg/kg. 80% protein bound so lower doses needed in hypoalbuminaemia. It has a VoD of 2L/kg and acts as a GABA agonist and non-NMDA glutamate receptor antagonist. Its pKA is 7.6 and is the fastest acting induction agent. Offset of action is due to redistribution to fatty tissue, and metabolism is actually very slow as the responsible enzymes are quickly saturated and it starts to obey zero order kinetics. After a single bolus the offset time is 5-10 minutes. Metabolism is hepatic, to inactive metabolites. Induction of CYP450 occurs after a single dose
38
Describe the A-E effects of etomidate
A - preservation of laryngeal reflexes B - brief hyperventilation followed by brief apnoea - minimal net effect. No histamine release so no bronchospasm C - famously cardiostable, may produce slight tachycardia and hypertension D - produces sedation/anaesthesia in 30-60 seconds, reduces cerebral blood flow and metabolic rate, however reduces ICP by 50% so maintains or increases cerebral perfusion pressure. Causes myoclonus on induction and prolongs seizures (hence is useful in potentiation ECT) E - dose dependent reversible inhibition of 11B hydroxylase leading to reduced cortisol production. Unclear how significant this is after one dose, and whether its more significant in the critically ill
39
Describe the dose and mechanism of action of etomidate
Action is positive modulation of the GABA A receptor at low doses, and independent agonism at higher doses. Dose is 0.2-0.6mg/kg
40
Give 5 side effects of opiates
N&V Sedation Bradycardia and hypotension Respiratory depression Constipation Urinary retention Delirium Dysphoria & hallucinations Itching Tolerance & addiction
41
Which of the following statements is true of morphine? A) VoD is ~0.5L/kg B) M3G accumulates in renal failure leading to toxicity C) It has an oral bioavialibility of ~25% D) It has a lower pKa than alfentanil E) It is a histamine receptor agonist
C) It has an oral bioavialibility of ~25% Morphine is an opioid used for acute and chronic pain. It has 25% oral bioavailability, and is used IV as a slow-onset agent (~10 minutes) at a dose of 0.1-0,2mg/kg. VoD is 3.5L/kg, pKa is 7.9, and metabolism is mostly through glucoronidation in the liver and gut excretion. The main metabolite (70%) is M3G which is inactive, but M6G is also produced which is more potent and accumulates in renal failure; in chronic use M6G is even responsible for the majority of activity. Elimination half-life is very variable, 3 hours is an average estimate.
42
Which of the following is true of diamorphine? A) It has higher affinity for mu receptors than morphine B) It is metabolised to M6G C) It is a prodrug metabolised in the liver D) It has a similar onset time to morphine E) It is much more lipid soluble than morphine
E) It is much more lipid soluble than morphine Diamorphine is a very lipid soluble prodrug with no inherent activity at opioid receptors. It is 3,6-diacetyl morphine and is rapidly converted to the active metabolite 6-MAM, then to morphine by plasma and tissue esterases. Both it and 6-MAM are much more lipid soluble than morphine and crosses the blood-brain barrier quickly. Because of these differences, diamorphine, in clinical use, has twice the potency of morphine despite having no inherent action at opioid receptors.
43
Which of the following is true of remifentanil? A) It is twice as potent as fentanyl B) It depends on plasma cholinesterase metabolism C) A Cet of 2ng/ml in combination with propofol will obtund the pressor response to larngoscopy D) It can produce lethal bradycardia and hypotension if bolused E) It produces active metabolites with 10% potency
D) It can produce lethal bradycardia and hypotension if bolused Remifentanil has a similar potency and speed of onset to fentanyl, but is metabolised by tissue esterases (not plasma cholinesterases so not affected by anti-cholinergic drugs, and not subject to genetic variation in metabolism). It produces an active metbaolite but it only has 0.1% the potency so is clinically irrelevant. In response to C), a Cet of 5ng/ml in the setting of Cet propofol 4mg/ml will obtund the pressor response to larngoscopy.
44
Which of the following is true of tramadol? A) It forms a racemic mixture with different stereoisomers activiating different receptors B) It has a similar profile to other opioids in overdose C) It is safer in renal impairment than codeine D) It is a diphenylpropylamine E) It has cholinergic effects
A) It forms a racemic mixture with different stereoisomers activiating differen receptors One stereoisomer activates opioid receotors and inhitibs serotonin reuptake , the other inibits noradrenaline reputake presynaptically. This variety of action explains why tramadol is only partly reversed by naloxone. Tramadol is more dangerous than other opioids in overdose, causing seizures in 15% of cases, though it does produce less respiratory depression. It produces active metabolites that accumulate in renal failure similarly to codeine and morphine. It is classed as a phenylpiperidine. It is a nicotinic and muscarinic antagonist.
45
Arrange the following opioids in order of pKA (ascending): Alfentanil, fentanyl, morphine, remifentanil
Alfentanil - 6.5 Remifentanil - 7.1 Morphine - 7.9 Fentanyl - 8.4
46
Which of the following statements on opioids is false? A) Fentanyl and morphine have a similar elimination half-life B) 1% of people have an absent CYP2D6 resulting in codeine inefficacy C) Pethidine belongs to the same structural group as alfentanil D) Tramadol produces less respiratory depression E) De-methylation produces the active metabolite of codeine
B) 1% of people have an absent CYP2D6 resulting in codeine inefficacy ~10% of people have an absence of this enzyme, resulting in codeine inefficacy.
47
Which of the following is true of opioid partial agonists/ antagonists? A) Buprenorphine can precipitate withdrawal if given to a patient on opioids B) Buprenorphine should be used with caution in renal failure C) Naloxone has a duration of action of ~4 hours D) Buprenorphine overdose can be reliably reversed using naloxone E) Naloxone reduces sympathetic tone
A) Buprenorphine can precipitate withdrawal if given to a patient on opioids Buprenorphine is a partial agonist with very high affinity for mu opioid receptors. Consequently it out-competes other opioids while not stimulating the receptor much, which can cause acute withdrawal. The high affinity for its receptor also explains why naloxone can be ineffective against higher doses of buprenorphine. Buprenoprhine undergoes hepatic metabolism to an active 'nor' metabolite which is excreted in bile. Nalozone is a competitive opioid antagonist, but only has a short duration of action (30-45 minutes) and so may need repeat doses or an infusion. Initial dose is often 100-400mcg, but may need more like 2mg in some cases, and can be repeated up to 10mg total. Nalaxone increases sympathetic tone which can cause hypertension, arrhythmia, and rarely pulmonary oedema.
48
Which of the following is true of local anaesthetics? A) An amide linkage makes local anaesthetics shorter-acting B) Cocaine triggers catecholamine release from pre-synpatic vesicles C) Bupivicaine can be used as a treatment for VT D) Prilocaine metabolites ionise iron in haemoglobin to increase its avidity for oxygen E) Levobupivicaine was developed to be less reno-toxic
D) Prilocaine metabolites ionise iron in haemoglobin to increase its avidity for oxygen This change ion affinity means the oxygen does not unload oxygen in hypoxic tissues - causing methaemoglobinaemia
49
Which of the following is false of local anaesthetics? A) LAs cause conformational change to block Na channels B) Ametop cream contains an amide LA C) Ropivicaine is quick onset and long action D) LAs are mostly ionised at pH 7.4 E) Altered taste, perioral tingling, and tinnitus are early signs of LA toxicity
B) Ametop cream contains an amide LA Ametop contains tetracaine (amethocaine) which is an ester LA along with cocaine, procaine, and benzocaine.
50
Explain why isoprenaline is a better chronotrope than dobutamine
Isoprenaline is very beta-selective, so its chronotropy is not offset by reflex from vasoconstriction. However dobutamine stimulates both beta1 and alpha1 so its vasoconstriction offsets some of its chronotropy.
51
Describe the mechanism of action of milrinone
Milrinone is a PDE III inhibitor. phosphodiesterases are enzymes that break down cAMP and so are part of the de-coupling of beta adrenergic reeptors that occurs with sustained stimulation. Inhibiting this breakdown potentiates beta receptors and so is particularly useful in chronically sick hearts, which will exhibit this de-coupling. Milrinone also binds to calcium channels on the sarcoplasmic reticulum, and so potentiate CICR to provide another mechanism for inotropy.
52
Which of the following is false of ephedrine & catecholamines? A) Alpha 2 receptors also inhibit ACh release B) Most norad released is taken up post synaptically and metabolised by COMT C) Ephedrine's efficacy is reduced by cocaine & amitryptilline D) Tachyphylaxis is a result of norad store depletion
B) Most norad released is taken up post synaptically and metabolised by COMT Norad is recycled pre synaptically due to very high affinity of NET for norad, where it is recycled into vesicles by VMAT or metabolised by MAO. COMT also metabolises catecholamines but is less important for norad.
53
Why is adductor policis brevis used to monitor NMB block resolution?
Because it has a comparatively poor blood supply as it is peripheral, so will recover from block later than central sites such as the diaphragm. Hence this provides a margin of safety, especially as the larynx and pharynx recover more slowly than the diaphragm.
54
Describe the two ways large volumes of N2O Co-administration with sevofluorane increase speed of onset.
Concentration effect: large volumes of a non-potent carrier gas are given alongside sevo. The carrier gas is more soluble than the other gases in the mix and so rapidly diffuses into the bloodstream. This causes loss of volume within the lungs and an increase in concentration of other gases within the mix as N2O is disproportionately absorbed and distributed. Second gas effect: almost the same thing. It refers to the augmented ventilation i.e. creation of a concentration/ pressure gradient which the anaesthetic and carrier gases then diffuse down into the alveoli. This increases the anaesthetic gradient at the alveolus:capillary interface and so speeds up diffusion.
55
What would be the advantage of using atracurium as a muscle relaxant in ITU?
Renal dysfunction is common in the critically unwell population. Atracurium's metabolism relies 45% on Hoffman elimination (depends only on pH and temp) and 45% on non-specific ester hydrolysis. Therefore it is eliminated normally even in organ dysfunction. Variations in temp and pH do affect its elimination, but not at values within a realistic clinical range. Concerns over laudanosine accumulation and seizures are purely theoretical and don't seem to have ever been substantiated. Even if this was a concern, Cisatracurium could be used as it produces 90% less laudanosine, and is 75% metabolised by Hoffman elimination with 15% urinary excretion.
56
Which of the following is false of benzodiazepines? A) They are unsafe in pregnancy and breastfeeding women B) They bind to specific receptors between the α and γ subunits C) Apart from lorazepam and oxazepam, all benzos have active metabolites D) Midazolam is highly bioavailable and quickly cleared E) GABA receptors are pentamers, mostly found on the post-synaptic membrane
D) Midazolam is highly bioavailable and quickly cleared This is half true - midazolam is cleared much faster than diagram or lorazepam. However midazolam has oral bioavailability of 44%, much lower than diazepam or lorazepam. Benzos are positive allosteric modulators of the GABAa receptor. They bind to one of two receptor subtypes (BLZ1 or 2) at a site between the alpha and gama subunits of the GABAa receptor. This increases the affinity of GABA receptors for GABA, and leads to increased frequency of opening of the central chloride channel and inhibitory hyperpolarisation of the cell membrane. Benzos are unsafe in pregnant women as they can cause cleft palate, respiratory distress, and in breastfeeding women they are secreted in breast milk and may sedate the baby. Most benzos have active metabolites, the longer acting ones (diazepam and chlordiazepoxide) produce a series of active metabolites that partly explains their long duration of action.
57
Describe the mechanism of action of neostigmine
As an intermediate-duration acetylcholinesterase inhibitor, it binds to both the anionic and esteratic enzyme sites. This prevents ACh from binding and being cleaved. The enzyme is not available to clear ACh until the neostigmine-AChE complex has been hydrolysed.
58
Which of the following is least likely to prolong the action of suxamethonium? A) Renal failure B) Hepatic cirrhosis C) Myasthenia gravis on treatment D) Untreated Alzheimer's dementia E) Cancer
D) Untreated Alzheimer's There is a link between Alzheimer's and increased AChE and plasma cholinesterase activity, hence the duration of action of sux would be less. This doesn't necessarily hold true if the patient is taking treatment, which for Alzheimer's can consist of AChE +/- BChE (butyrylcholinesterase). Factors that would prolong a suxamethonium block include wasting states (cancer, malnutrition, chronic infection), hepatic dysfunction (reduced cholinesterase manufacture), and renal disease. However a 75% decrease in AChE activity is required to be clinically significant, so this is not commonly clinically significant.
59
What is suxamethonium apnoea?
Sux apnoea describes a markedly prolonged action of suxamethonium from 15 minutes to ~8 hours. It is the result of an autosomal recessive condition resulting in deficiency of plasma cholinesterases which metabolise suxamethonium, mivacurium, and ester local anaesthetics.
60
Which of the following is least consistent with sux apnoea? A) Being homozygous for the k-variant B) ToF fade and post-tetanic potentiation C) No family history of similar issues D) An EsEs phenotype E) A heterozygous genotype at position 3q26.1-2
E) A heterozygous EuEa genotype at position 3q26.1-2 Sux apnoea generally requires two dysfunctional alleles for sufficient impairment. Phenotypically: Eu is the normal allele, Ea and Ef produce altered enzymes, and Es produces an inactive enzyme. Being homozygous for the k-variant is one of the most common genotypes in sux apnoea, along with presence of two of the Dib variants. ToF fade and post tetanic potentiation are features of either non-depolarising block or phase 2 depolarising block. In sux apnoea, suxamethonium lingers at the synapse and so causes the same phase 2 block that would be expected with repeated doses/ infusions. As sux apnoea is autosomal recessive, there may well be no family history. EsEs is the most severe phenotype in sux apnoea and is more likely in cases where recovery is more prolonged.
61
Match the following inotropes/pressors to their receptors: A) Adrenaline B) Noradrenaline C) Dopamine D) Dopexamine E) Dobutamine 1) B2, D1, B1 2) B1, B2, A1 3) A1, B2, B1 4) B1, D1, A1 5) B1, A1
A-3 B-5 C-4 D-1 E-2 It's important to remember that adrenaline is a little more potent at A1 and a little less at B1 than norad despite classic teaching. The difference in effect between the two is explained by adrenaline having a significantly greater affinity for B2 giving it inodilator properties.
62
Which of the following is false of ketamine? A) It is presented as a racemic mixture, with the S+ enantiomer being most potent B) It may cause lacrimation, mydriasis, and nystagmus C) It competes at glutamate and glycine binding sites D) It is an opioid agonist, and influences opioid tolerance/hyperalgesia via NMDA antagonism E) It antagonises a range of receptors including NMDA, muscarinic, monoamine, cholinergic, and adrenergic
C) It competes at glutamate and glycine binding sites Ketamine is a non competitive antagonist, and acts at NMDA receptors by binding at phenylcyclidine binding sites sites situated within the ion channel. Ketamine is presented as a racemic mixture, and the S enantiomer is three times more potent than the R. Due to its antagonism of a wide range of receptors (monoamine, adrenergic, cholinergic, muscarinic, nicotinic, neuronal sodium channels, purinic, and in the thalamo-neocortical projection system) it has a range of clinical effects. It also seems to be an opioid agonist.
63
Which of the following is false of ketamine? A) It disrupts PSD protein interactions with NMDAr, preventing NMDAr upregulation and opioid tolerance B) An IM dose of 4mg/kg may produce either anaesthesia or sedation C) The active metabolite has 20% potency and is conjugated and excreted in urine D) Clearance is ~1L/min, and elimination half-life is 2-3 hours E) Action would be prolonged by phenytoin
E) Action would be prolonged by phenytoin Ketamine is metabolised by cytochrome P450 and one of its metabolites (norketamine) is active with 20% potency. Phenytoin is a CYP450 inducer and so would shorten ketamine's duration of action. Ketamine does disrupt interactions between NMDAr and PSD proteins, and through this inhibits a pathway involving nitric oxide synthase and protein kinase C that is involved in opioid tolerance. 4mg/kg IM is the top end of the sedation range (2mg/kg-4mg/kg) and the bottom end of the anaesthetic range (4mg/kg-10mg/kg). Clearance is rapid, and elimination half life is likely only as long as it is because of the large volume of distribution.
64
Briefly describe activation of the NMDA receptor
The unusual feature is that it is both ligand and voltage gated - it requires both to activate. The ligand opens the channel, but magnesium and zinc ions block the channel and are only dislodged when the neuron depolarises.
65
Which of the following is false of thiopentone? A) It will precipitate in combination with morphine, rocuronium, or calcium B) It is unsuitable for RSI in respiratory failure due to risk of bronchospasm from histamine release C) It has the largest VoD and most protein-binding of any of the induction agents D) SVR is generally preserved E) Patients self-ventilate faster after thiopentone than propofol
C) It has the largest VoD and most protein-binding of any of the induction agents Thiopentone has the lowest VoD of the induction agents at 2.2, though is reasonably heavily protein-bound at 80% (propofol is 96%). Thiopentone will precipitate in combination with acidic or oxidised solutions including roc, vec, calcium chloride, morphine, and lidocaine. Thiopentone is contraindicated in respiratory disease because it triggers histamine release which could cause bronchospasm and worsen the respiratory failure. Thiopentone causes venodilation which reduces cardiac output, without decreasing afterload. These effects are more concerning in patients with hypovolaemia, pre-existing cardiac disease, or who are frail or critically unwell
66
Describe the mechanism of action of opioid-mediated analgesia
Action sites for opioid-mediated analgesia can be divided into three: 1) Central inhibition of ascending pain pathways - Takes place in the substantia gelatinosa of the dorsal horn of the spinal cord - Opioid receptors activation reduces calcium influx and neurotransmitter release pre-synaptically, and causes potassium efflux and membrane hyperpolarisation post-synaptically 2) Central enhancement of descending inhibitory pathways - These pathways run from the periaqueductal grey matter in the pons, via the nucleus raphe magnus in the medulla, down the spinal cord as far as the dorsal horn - Opioid act on GABA-ergic interneurons in the periaqueductal grey matter. They act pre-synpatically to inhibit these neurons. This disinhibits the descending inhibitory pathway, which then releases noradrenaline and serotonin to inhibit nociceptive transmission. 3) Peripheral - Inflammation triggers up regulation of opioid receptors locally, and hence provides a non-CNS site of action for opioids.
67
Which of the following is false regarding opioids? A) Most morphine is metabolised by demethylation at the 'N' site B) Morphine's low lipid-solubility may lead to delayed respiratory depression C) Morphine's M3G metabolite features in entero-hepatic recycling D) Pethidine has anti-cholinergic effects E) Oxycodone is derived from thebaine
A) Most morphine is metabolised by demethylation at the 'N' site Most morphine (70%) undergoes glucoronidation in the liver to M3G, an inactive metabolite. Some undergoes glucoronidation to M6G which is more potent than morphine. A small amount is demthylated to normorphine at the 'N' site.
68
Which of the following is false of furosemide? A) 80% is excreted unchanged B) It inhibits the triple co-transporter on the apical membrane in the thick ascending limb C) It is highly protein bound which accounts for lack of efficacy in nephrotic syndrome D) It may cause hypercalcaemia E) It increases renal blood flow
D) It may cause hypercalcaemia Hypercalcaemia is a feature of thiazides, loop diuretics cause hypocalcaemia. Furosemide is a loop diuretics which blocks the co-transporter of sodium/ potassium/ chloride on the apical membrane of the thick ascending limb in the Loop of Henle. It is heavily protein bound and 80% is excreted unchanged, with the other 20% undergoing glucoronidation in the kidney.
69
Which of the following is true regarding benzodiazepines? A) They are modulators of a voltage gated pentamer that forms a chloride channel B) Flumazenil is excreted unchanged C) Midazolam is unionised in an acidic solution D) Chlordiazepoxide, diazepam, and tempazepam are all metabolised to oxazepam E) Diaz, loraz, and midaz have similar VoD, protein-binding, and oral bioavailability
D) Chlordiazepoxide, diazepam, and tempazepam are all metabolised to oxazepam Long-acting benzodiazepines are partly long-acting because they produce a series of active metabolites. Benzodiazepines are positive modulators of the GABA A receptor. They bind at their receptor (BZ1 and BZ2) between the alpha and gama subunits of the receptor, whereas GABA binds between the alpha and beta subunits. However the GABA receptors is ligand gated rather than voltage gated. Flumazenil is an imidazobenzodiazepine that acts as a competitive benzo antagonist. It is 40-50% protein bound, has a half life of 50 minutes, and is metabolised in the liver to inactive metabolites. Midazolam is presented in a solution with pH 3.0 which makes it ionised and water-soluble. Once it is in a solution with pH >4 then it becomes unionised and lipid-soluble. Diaz, loraz, and midaz all have similar protein binding and VoD. However midazolam is half as bioavailable orally as the others.
70
Which of the following is false of furosemide? A) Hypoalbuminaemia increases the effect of furosemide B) It must be secreted in the PCT in order to work C) It is a competitive chloride antagonist D) 50% is excreted unchanged in urine, and 50% is gluroronidated in the kidney E) It alters tubular osmolality to reduce water reabsorption in the collecting ducts
A) Hypoalbuminaemia increases the effect of furosemide Furosemide is heavily protein bound, so you would expect this to be the case. However experiments have shown that low albumin reduces furosemide delivery to the PCT. Furthermore, Co-administration of albumin has no effect to counter this.
71
Which of the following is true of furosemide? A) Furosemide can cause hyponatraemia B) Interruption of chloride absorption increases serum calcium C) Reabsorption of Na via ENaC channels is responsible for hypokalaemia D) Furosemide is lipophillic E) It may interact with warfarin via CYP450
C) Reabsorption of Na via ENaC channels is responsible for hypokalaemia Furosemide inhibits absorption of potassium, sodium, and chloride from the tubular lumen. The sodium is then reabsorbed in the distal nephron via ENaC channels (RAAS mediated). This creates a charge gradient down which potassium exits the cell into the tubule. Furosemide does cause urinary excretion of sodium, however there is enough reabsorption via ENaC channels plus concentration due to water loss that it will generally cause hypernatraemia.
72
Which benzodiazepines do not produce active metabolites?
Oxazepam, temazepam, lorazepam