Past Questions

From Ethans Wiki
Revision as of 05:20, 13 February 2024 by 14.203.236.70 (talk) (Created page with "<span id="cicm-first-part-exam---ethans-answers"></span> = CICM First Part Exam - Ethan's answers = <span id="high-yield-topics"></span> === High yield topics === <blockquote>Topics with 5 or more past questions that are identical / very similar </blockquote> <span id="physiology"></span> === Physiology === * CNS ** CSF production, regulation, flow, content, physiological role (pretty much every year) * CVS ** Myocyte vs pacemaker action potentials * Renal/fluids...")
(diff) ← Older revision | Latest revision (diff) | Newer revision → (diff)

CICM First Part Exam - Ethan's answers

High yield topics

Topics with 5 or more past questions that are identical / very similar


Physiology

  • CNS
    • CSF production, regulation, flow, content, physiological role (pretty much every year)
  • CVS
    • Myocyte vs pacemaker action potentials
  • Renal/fluids/acids
    • Renal blood flow + autoregulation
    • Buffer systems of the body
  • Resp
    • Oxygen and carbon dioxide carriage in blood
    • Control of breathing
    • Respiratory/pulmonary compliance
    • Functional residual capacity (FRC): define it, describe the factors effecting it, consequences of reducing it, measurement of it
  • GIT
    • Outline the functions of the liver


Pharmacology

  • Amiodarone
  • Vasopressors, inotropes
  • GTN
  • Pharmacology of NMBs (vec, sux, roc) - essentially a question every year-sitting
  • Frusemide
  • Ketamine
  • Midazolam
  • Magnesium sulfate
  • Drugs to treat asthma (overview and MOA)
  • Fractionated vs unfractionated heparin


Measurement

  • Arterial lines and invasive BP
  • Pulse oximetry / co-oximeter
  • ETCO2 (differences between PaCO2, measurement, sources of error)



2022 (1st sitting)

Question 1

Question

Outline the effects of critical illness on drug pharmacokinetics, including examples


Example answer

Absorption

  • Oral
    • Decreased CO > decreased GIT blood flow > decreased absorption PO drugs
    • Ileus + uraemia > decreased gastric emptying > decreased absorption of PO drugs
    • Diarrhoea > fast transit time > decreased absorption
    • Change in gastric pH (e.g. with PPI) alters drug absorption
    • Decreased GIT blood flow (vasoconstrictors, barbiturates) > decreased PO absorption
  • Topical/IM/SC
    • Vasoconstriction > poor tissue perfusion > decreased/slow absorption
  • Inhalational
    • Decreased MV / TV > decreased delivery of aerosolised medications


Distribution

  • Altered Vd
    • Decreased CO (e.g. shock) > slower redistribution
    • Increased CO (e.g. hyperdynamic sepsis) > faster residistribution
    • Hypervolaemia (e.g. renal, cardiac, liver failure) > increased Vd (vice versa)
    • Critical illness > muscle wasting > alter lean mass percentage (alters Vd)
  • Protein binding
    • Decreased protein synthesis (e.g. decreased albumin) > increased unbound fraction of drug > increased Vd and drug activity
    • Acid-base disturbances will alter free drug levels depending on drug pKa and the pH
  • Inflammation > impairs barrier function (e.g. BBB) > increased penetration of meds (e.g. penicillins)


Metabolism

  • Decreased CO > decreased hepatic/renal blood flow > decreased metabolism (e.g. propofol)
  • Liver dysfunction > Impaired phase 1 and 2 reactions and reduced 1st pass effect > (e.g. labetalol, metoprolol)
  • Renal dysfunction > decreased renal metabolism > prolonged drug effect (e.g. morphine)
  • Hypothermia > decreased metabolism > Prolonged effect (e.g midazolam)
  • Resp dysfunction > Decreased resp metabolism of drugs (e.g. opioids) > prolonged effect


Elimination

  • Decreased CO (e.g. cardiogenic shock) = decreased GFR / HBF > decreased clearance (e.g. gentamicin)
  • Increased CO (e.g. hyperdynamic sepsis) > increased GFR > increased clearance
  • Liver dysfunction > impaired biliary excretion of drugs (e.g. vecuronium, rifampicin)
  • Decreased GFR (e.g. AKI) > decreased renal elimination drugs (e.g. Gentamicin, milrinone)
  • Reduced MV > decreased / slower clearance of volatile anaesthetics > prolonged effect


Examiner comments


Online resources for this question

  • Jenny's Jam Jar
  • CICM Wrecks
  • Deranged physiology


Similar questions

  • Question 13, 2013 (1st sitting)



Question 2

Question

Explain the mechanisms of transport of substances across cell membranes including appropriate examples (75%). Outline the structure of the Na+/K+-ATPase pump (25%)


Example answer

Transport across cell membranes

Mode Mechanism Energy expenditure Electrochemical gradient Example Factors affecting
Passive (simple) diffusion Molecule passes through cell membrane No (passive) With/down CO2 and pulmonary vascular endothelium Fick's law of diffusion
Facilitated diffusion Molecule crosses membrane via transmembrane protein No (passive) With/down Glucose with the GLUT transporter Ficks law diffusion and number of carrier proteins
Ion channels Membrane spanning proteins (voltage, ligand or mechanical gated) > conformational change > opening of ion channel No (passive) With/down nACHR (Ach is ligand) with Na/K as ions Concentration gradient, number of channels
Primary active transport Molecule crosses membrane via carrier proteins Yes (active) requires ATP Against Na/K ATPase pump Availability of carrier, substrate and ATP
Secondary active transport (symport or antiport) Molecule crosses membrane via a carrier protein, with the energy being provided for the transport of another molecule Yes (but not directly) Against Na / H antiporter in principle cells of renal collecting ducts Availability of carrier, substrate and ATP
Endocytosis Cell membrane invaginates around a large molecule > engulfs it > contained within vesicle Yes Usually against Phagocytosis Poorly understood. ATP
Exocytosis Vesicle (containing molecule) fuses with cell membrane > release of molecule Yes Usually against Exocytosis of ACh at the pre-synaptic cleft of the NMJ Poorly understood. ATP
File:Https://derangedphysiology.com/main/sites/default/files/sites/default/files/CICM Primary/E Cellular Physiology/cell transport across membranes2.JPG


Na/K-ATPase pump

  • Membrane bound protein pump
  • Structure
    • Transmembrane protein, consists of two globular proteins
      • Large alpha subunit (MW = 105kDa)
      • Small beta subunit (MW 55kDa)
    • Three binding sites for Na on internal surface, two binding sites for K on external surface
    • ATPase activity at the Na binding site
  • Function/Features
    • Antiport --> pumps 3 Na out and 2 K in
    • Energy dependant (requires ATP)
  • Process
    • Once Na/K binding sites full, ATPase cleaves ATP > release energy > conformational change > 3na out and 2K in
  • Function
    • Controls cell volume (prevents Gibbs Donnan equilibrium)
    • Electrogenic (contributes to RMP)


Examiner comments


Online resources for this question

  • Jenny's Jam Jar
  • CICM Wrecks
  • Deranged physiology


Similar questions

  • Question 10, 2012 (2nd sitting)



Question 3

Question

Define respiratory compliance, include its components and their normal values (25% marks). Explain the factors that affect respiratory compliance (75% of marks)


Example answer

Respiratory compliance

  • <math display="inline">Compliance \; = \; \frac{\Delta \;volume}{\Delta \; pressure}</math>
  • Compliance in the respiratory system (CRS) is a function of lung (Clung) and chest wall (CCW) compliance
    • <math display="inline">\frac {1}{C_{RS}}\; = \; \frac {1}{C_{Lung}} \; + \; \frac {1}{C_{CW}}</math>
    • Chest wall and lung compliance are roughly equal in healthy individual (~200mls.cmH2O)
    • Thus normal compliance of the respiratory system is ~100mls.cmH2O
  • Static compliance
    • Compliance of the respiratory system at a given volume when there is no flow
  • Dynamic compliance
    • Compliance of the system when there is flow (respiration)
    • Will always be less than static compliance due to airway resistance
    • At a normal RR is approximately equal to static compliance
  • Specific compliance
    • The compliance of the system divided by the FRC
    • Allows comparisons between patients which are independent of lung volumes


Factors effecting compliance

Chest wall

  • Increased
    • Collagen disorders (e.g. Ehlers-Danlos syndrome)
    • Cachexia
    • Rib resection, open chest
  • Decreased
    • Obesity
    • Kyphosis / scoliosis / Pectus excavatum
    • Circumferential burns
    • Prone positioning

Lung compliance

  • Increased

    • Normal ageing

    • Emphysema

    • Upright posture

    • Lung volume (highest compliance at FRC)

  • Decreased

    • Loss of surfactant (E.g. ARDS, hyaline membrane disease)

    • Loss of functional lung volume (e.g. pneumonia, lobectomy, pneumonectomy, atelectasis)

    • Pulmonary venous congestion (pHTN) and interstitial oedema (APO)

    • Reduced long elasticity (e.g. Pulmonary fibrosis)

    • Positioning (e.g. supine positioning)

Examiner comments


Online resources for this question

  • Jenny's Jam Jar
  • CICM Wrecks
  • Deranged physiology


Similar questions

  • Question 17, 2019 (2nd sitting)
  • Question 14, 2017 (1st sitting)
  • Question 15, 2014 (1st sitting)
  • Question 7, 2011 (2nd sitting)
  • Question 13, 2007 (1st sitting)



Question 4

Question

Describe the mechanisms of action and potential adverse effects of inhaled nitric oxide and prostacyclin


Example answer

Nitric oxide MOA (inhaled)

  • Pulmonary vasodilator
  • Diffuses into smooth muscle cell > activates guanyl cyclase > increased conversion of GTP to cGMP > decreased intracellular calcium > relaxation of smooth muscle > vasodilation
  • As it is inhaled it selectively vasodilates well ventilated alveoli, which leads to improved V/Q matching > decreased work of breathing and increased oxygenation


Prostacyclin MOA (inhaled)

  • Pulmonary vasodilator
  • Binds to prostacyclin receptor (IP receptor) > active GPCR > increased conversion of ATP to cAMP > decreased intracellular calcium > relaxation of smooth muscle > vasodilation
  • Improves V/Q matching by the same mechanism of NO


Adverse effects (NO)

  • May exacerbate left ventricle heart failure
    • Decreased pulmonary pressures > increased RV SV > increased LV preload
    • Can overwhelm an impaired LV > pulmonary oedema / worsening HF
  • Vasodilatory effects
    • Leads to flushing, headache, hypotension
    • Less pronounced with inhaled therapy
  • Rebound pulmonary hypertension and hypoxia
    • Occurs following abrupt cessation
    • Build up of vasoconstrictive molecules during therapy which are unopposed following cessation
  • Thrombocytopaenia (up to 10%)
  • Methaemaglobinaemia
    • Relatively rare
    • NO reacts with OxyHb to produce MetHb and nitrates
    • Significant amounts >5% only occurs at high doses e.g. > 20ppm
  • Tachyphylaxis (over days)
  • AKI
    • Usually with high doses > 20ppm
    • Relatively rare


Adverse effects (Prostacyclin)

  • Inhibits platelet aggregation > increased risk of bleeding
  • Rebound pulmonary hypertension and hypoxia (same mechanism as above)
  • Exacerbate LV heart failure (same mechanism as above)
  • Vasodilatory effects > flushing , headache, hypotension (less pronounced with inhaled)


Examiner comments


Online resources for this question

  • Jenny's Jam Jar
  • CICM Wrecks
  • Deranged physiology


Similar questions

  • Question 17, 2020 (2nd sitting)
  • Question 14, 2011 (1st sitting)



Question 5

Question

Write short notes on the pharmacology of labetalol and esmolol, highlighting their differences


Example answer

Name Labetalol Esmolol
Class Alpha and beta blocker Beta blocker
Indications Hypertension Short term management of tachycardia/hypertension in hospital settings (e.g. OT)
Pharmaceutics 100/200mg tablets

Clear solution (5mg.ml, 10ml ampoules)
Mixture of stereoisomers

Clear/slight yellow colourless solution 100mg/10ml
Routes IV, PO IV only
Dose IV: 5-20mg boluses, infusion 1-2mg.min

PO: 100-800mg BD

10mg boluses / infusion titrated to effect
Pharmacodynamics
MOA a1 adrenergic antagonist

Nonspecific Beta antagonist

B1 selective antagonist
Effects CVS: decreased chronotropy, inotropy, dromotropy (beta effects), decreased SVR and afterload (alpha effects) > decreased myocardial oxygen consumption + decreased BP

RENAL: B1 blockage at JG cells > decreased renin release > decreased BP

Same as labetalol (except no alpha mediated vasodilation)
Side effects CVS: bradycardia, hypotension (esp orthostatic), heart block

RESP: dyspnoea, bronchospasm
CNS: dizziness
GIT: nausea, raised LFTs

CVS: hypotension, bradycardia, heart block (B1 effects)

RESP: NO bronchospasm or dyspnoea (no B2 effects)
CNS: dizziness
GIT: Nausea

Pharmacokinetics
Onset 1-2 hours (PO), <5 mins (IV) Immediate (seconds-mins)
Absorption PO bioavailability 25%
(extensive 1st pass metabolism)
0% oral bioavailability
Distribution 50% protein bound

VOD = 8L/kg

60% protein bound

VOD 3.5L/kg

Metabolism Hepatic (extensive)

Glucuronide conjugation
Inactive metabolites

Rapid metabolism

- Hydrolysis by RBC esterases
Inactive metabolites

Elimination Renal (50%) / faecal (50%) elimination

Inactive metabolites
T 1/2 - 6 hours

Renal

Inactive metabolites
T 1/2 = 10 mins (parent)


Examiner comments


Online resources for this question

  • Jenny's Jam Jar
  • CICM Wrecks
  • Deranged physiology


Similar questions

  • Question 9, 2021 (2nd sitting)
  • Question 14, 2019 (2nd sitting)



Question 6

Question

Describe the cardiovascular changes seen throughout pregnancy


Example answer

Factor Response during pregnancy Comments
HR Increases by up to 25%

Peaks in 3rd trimester

SV Increases by up to 25%

Predominately in 1st trimester

CO Increases - Up to 50%

- Due to increased blood volume, increased HR/SV, increased VR and decreased SVR
- Majority > placenta (nutrient/gas to foetus), kidneys (80% increase, waste excretion), skin (regulates temp)

BP Decreases - By about 10%

- Due to decreased SVR

Systemic vascular resistance Decreases - By up to 30%

- Due to increased progesterone, PGs and downregulation of alpha receptors
- Occurs predominately in 1st trimester

Blood volume Increases - By up to 40%

- Due to stimulation of RAAS (oestrogen) and increased erythropoiesis (increased renal EPO)
- Plasma volume increased > RBC volume increase > decreased HCT (dilutional anaemia)

CVP Unchanged
Oxygen delivery Increased Due to increased CO
Aorto-caval compression Increased Due to weight of the gravid uterus > decreased VR
Colloid osmotic pressure Decreases Predisposes to oedema
Factor Response during delivery Comments
BP/VR/CO Uterine contraction By up to 50%

Squeezes blood back into maternal circulation > increases VR > increased CO > increased BP

Factor Response post partum Comments
CO Increased (immediately post) By up to 80%

Due to autotransfusion of blood normally directed toward placenta/foetus

CO/ BP Decrease Return to normal ~2 weeks post delivery


Examiner comments


Online resources for this question


Similar questions

  • Question 7, 2016 (first sitting)

  • Question 19, 2013 (second sitting)

  • Question 4, 2010 (second sitting)


Question 7

Question

Write notes comparing the use of serum creatinine and creatinine clearance in the assessment of renal function in the critically ill


Example answer

  • Creatinine

    • Product of muscle metabolism

    • Freely filtered and not reabsorbed (but is partially secreted)

      • Given this, it can be used to approximate GFR (decreased GFR is indicative of worsening renal function)

Creatinine clearance (CrCl)

  • CrCl = the volume of plasma cleared of creatinine per unit time

  • Measuring CrCl

      1. Measure the plasma creatinine concentration

      1. 24 hour urine collection

      1. Use the fick principle to calculate the CrCl

      • CrCl = urine volume x urine concentration / plasma concentration

  • Estimating CrCl

    • Measuring urine concentrations is cumbersome

    • Can be estimated using various formula

      • e.g. the Cockcroft-Gault equation and MDRD formulas

      • Uses Age, weight, gender and height to calculate CrCl from serum Cr

      • Not as accurate and makes several assumptions


Creatinine concentration

  • Decreased renal function > decreased CrCl > increased Cr
  • Therefore increased Cr is indicative of worsening renal function
  • However
    • The relationship is non linear
    • Plasma creatinine concentration only begins to rise when ~50% of the renal function (GFR) is lost
    • Thus there is a significant decrease in GFR before a noticeable rise in Cr
Normal serum creatinine measurements do not exclude serious loss of...
Download Scientific Diagram


Limitations of serum creatinine as biomarker of renal function

  • Creatinine is partially secreted > overestimates GFR
  • Creatinine fluctuates
    • Increased with: increased protein consumption, muscle injury, steroid use
    • Decreased in: fasting, patients with low muscle mass, increased plasma volume (dilutional)
  • Creatinine takes time to accumulate / only rises significantly following >50% of loss of renal function
  • Therefore
    • Fluid resus or oedema > falsely low creatinine > falsely improving kidney injury
    • Patients with low muscle mass may have low Cr > masked kidney injury
    • Extremes of age / weight / muscle mass > unreliable results
    • Cr and CrCl detect kidney injury late
  • The level of inaccuracy increases with extremes of renal function



Examiner comments


Online resources for this question


Similar questions

  • Question 11, 2013 (second sitting)



Question 8

Question

Describe the regulation of body water


Example answer

Overview

  • Water intake
    • Approximately 25-30ml/kg of water is needed to be ingested for fluid/body homeostasis
      • ~2-2.5L per day for an average person
    • Approximately half comes from drinking fluids, half from food and metabolic processes
  • Water is lost through numerous ways
    • Urine
      • ~1 - 1.5L / day
      • Obligatory loss is ~500mls to cover solute/waste clearance
    • Insensible losses (skin, lungs etc)
      • ~900mls / day
    • Faeces
      • ~100mls / day
  • The body tightly regulates water balance to preserve plasma osmolality and intravascular volume status, but also allow waste clearance
    • Note: Preservation of blood volume takes precedence over plasma osmolality


REGULATION

  • Sensor
    1. Osmoreceptors in hypothalamus detect increased (>290mosm/L) osmolality with dehydration (major)
    2. Low pressure baroreceptors (RA, great vessels) detect reduced pressure (stretch) with dehydration
    3. High pressure baroreceptors (carotid sinus, aortic arch) detect reduced pressure (stretch) with dehydration
    4. Macula densa (kidneys) detect reduced GFR (Na/Cl delivery) with dehydration
  • Integrator
    • Hypothalamus (anterior and lateral regions, predominately)
  • Effector/effects
    1. Release of ADH
      • Synthesised in hypothalamus, transported to posterior pituitary for release
      • ADH acts on collecting ducts in the kidney in to increase aquaporins on luminal wall --> increased water reabsorption
      • Released in response to increase osmolality and activation of RAAS
    2. ANP/BNP
      • Decreased stretch > decreased ANP/BNP secretion --> increased water reabsorption
    3. RAAS
      • Decreased baroreceptor activation --> increased renin release
      • Decreased GFR sensed by macula sensa > increased renin release
      • Renin > activation of RAAS > increased water reabsorption
    4. Thirst centre (hypothalamus)
      • Activation of thirst centre in the lateral hypothalamus (due to increased osmolality) > behavioural change to increase water intake
  • Feedback
    • The above systems work predominately on a negative feedback system


Examiner comments


Online resources for this question

  • Jenny's Jam Jar
  • CICM Wrecks
  • Deranged physiology


Similar questions

  • Question 1, 2021 (2nd sitting)
  • Question 8, 2008 (1st sitting)
  • Question 4, 2015 (1st sitting)
  • Question 9, 2018 (2nd sitting)



Question 9

Question

Describe the pharmacology of 4% albumin


Example answer

Name Albumin
Class Colloid (human plasma protein suspended in crytaloid sollution)
Indications Intravascular volume replacement

Hypoalbuminaemia
Plasma exchange
Hepatorenal syndrome, pancreatitis, burns

Pharmaceutics 4% Albumin = 40g/L

Contains Na 140mmols, Cl 128mmols, Octanoate 6.4mmols
Hypotonic (260mOsm)
-Albumin collected by blood donation (Whole blood > plasmpheresis > fractionated > pasteurised > partitioned > stored)
Stored at room temp (<30 degrees)

Routes of administration IV
Pharmacodynamics
MOA Related to volume of fluid (i.e. volume expansion)

Increase albumin concentration > increase oncotic pressure / restores transport/drug binding function

Side effects No risk of bacteria/parasite infections (destroyed during processing), but risk of blood borne viruses (HIV, HepB, HCV) remains.

Allergy, fluid overload, pulmonary oedema.

Pharmacokinetics
Absorption IV only (0% oral bioavailability)
Distribution Rapid distribution within intravascular space.

Small Vd - about 5% leaves per hour

Metabolism Cellular proteolysis by cysteine protease
Elimination Degradation by liver and reticuloendothelial system

Half life of ~20 days

Special points - Likely worsens outcomes in TBI

- No need for blood cross matching


Examiner comments


Online resources for this question

  • Jenny's Jam Jar
  • CICM Wrecks
  • Deranged physiology


Similar questions

  • Question 12, 2020 (2nd sitting)
  • Question 1, 2015 (2nd sitting)
  • Question 1, 2009 (2nd sitting)




Question 10

Question

Describe the determinants of intracranial pressure (80% marks) and outline how it can be measured (20% marks)


Example answer

Intracranial pressure (ICP)

  • ICP = The pressure within the cranium, relative to atmospheric pressure
  • Normal ICP is 5- 15 mmHg
  • Governed by the Monro-Kellie doctrine (below)
  • There is rhythmic variation in ICP due to variations in respiration and blood pressure


Monro-Kellie doctrine

  • The skull is a rigid container of fixed volume

  • The skull contents include: brain (~85%), CSF (~5-8%), blood (~5-8%)

  • Therefore any increase in volume of one substance must be met by a decrease in volume of another, or else there will be rise in the ICP

image-20211012142839178
image-20211012142839178

Determinants of ICP

  • Brain tissue
    • No capacity to alter volume under physiologically normal circumstances
    • Increased volume in pathology: e.g. tumours, cerebral oedema > increased ICP
  • CSF
    • Constantly produced (24mls/hr) and reabsorbed (24mls/hr), thus volume remains the same
    • With increased ICP
      • CSF can be displaced from the cranium into the spinal subarachnoid space (as the spinal meninges have better compliance) > decrease ICP
      • Increased ICP > increased driving pressure for CSF reabsorption
    • However if there is obstruction to CSF flow then there can be accumulation (e.g. hydrocephalus) > increased ICP
  • Blood
    • Increased blood in cranium (e.g. cerbral vasodilation or haemorrhage) > increased ICP
    • With increased ICP > Compression of the dural venous sinuses > displace venous blood from the cranium > lower ICP
    • Blood flow is extensively autoregulated and effected by
      • MAP
      • PCO2
      • PO2
      • CMRO2


Measurement of ICP

  • Clinical

    • Cannot be directly measured clinically

    • Though increased ICP may result in

      • Headaches, nausea, vomiting

      • Papilledema

      • Decreased LOC

      • Cushing's reflex (critically high ICPs, very late)

    • Optic nerve sheath diameter

      • on US

    • Lumbar puncture opening pressure

      • Can approximate but not directly measure ICP

      • high CSF opening pressure may indicate high ICP

      • Numerous reasons why there would be a discrepancy in patients with pathology

  • Devices

    • EVD (gold standard)

      • Catheter which sits in the lateral ventricle

      • Pressure transmitted to wheatstone bridge via fluid filled tubing

      • Zeroed to atmospheric pressure

    • Codmans / Intraparenchymal pressure monitor

      • Sits in brain parenchyma (~2cm deep)

      • Pisoelectric strain gauge pressure sensor connected to a monitor via a fibreoptic cable

      • Only measures local ICP and cannot be re-zeroed

Examiner comments


Online resources for this question

  • Jenny's Jam Jar
  • CICM Wrecks
  • Deranged physiology


Similar questions

  • Question 7, 2020 (2nd sitting)
  • Question 15, 2018 (1st sitting)
  • Question 18, 2016 (2nd sitting)
  • Question 14, 2016 (1st sitting)
  • Question 18, 2010 (1st sitting)




Question 11

Question

Outline the structure and function of the NMDA receptor (25% marks). Discuss the pharmacology of ketamine (75% marks)


Example answer

NMDA receptor

  • Structure
    • Tetrameric (4 subunits), ligand gated, transmembrane receptor
  • Location
    • Abundant in the CNS (brain, spinal cord)
  • Ion permeability
    • Ca, Na, K
  • Activated by
    • Glutamate (excitatory neurotransmitter) and glycine
    • Activation leads to removal of central Mg plug > Na/Ca in, K out > EPSP
  • Blocked by
    • Ketamine, Mg, memantidine


Name Ketamine
Class Anaesthetic (phencyclidine derivative)
Indications induction GA, conscious sedation, analgesia,
Pharmaceutics 10/50/100mg/ml.

Clear colourless solution.
Racemic mixture of S and R enantiomers, or S+ enantiomer alone.
Water soluble.

Routes of administration IV/IM/PO/SC/PR
Dose 0-0.25mg/kg/hr (analgesia), 1-2mg/kg (GA), 0.5mg/kg (sedation)
pKa 7.5
Pharmacodynamics
MOA NMDA antagonism, weak opioid receptor agonism, weak Ca ch inhibition
Effects CNS: dissociative anaesthesia and analgesia.

CVS: increased HR/BPN (SNS stimulation), decreased pulmonary and systemic vascular resistance,
Resp: bronchodilation

Side effects CNS: emergence reactions including hallucinations, unpleasant dreams. may increase ICP in non ventilated patients

CVS: may increase HR/BP, increased myocardial O2 req.
GIT: Nausea, vomiting, increased salivation
RESP: apnoea

Pharmacokinetics
Onset 30s IV, duration of effect 10-20mins
Absorption Lipid soluble > readily absorbed. But poor OBA (16%) due to 1st pass metabolism
Distribution Large (~3L/kg) VOD

Small protein binding (~30%).
Crosses placenta.

Metabolism Hepatic (CYP450)

Demethylation > norketamine (30% potent) and inactive metabolites

Elimination Elimination T1/2 = 2 hours.

Kidneys (95%), faeces (5%)

Special points


Examiner comments


Online resources for this question

  • Jenny's Jam Jar
  • CICM Wrecks
  • Deranged physiology


Similar questions

  • Direct copy of Question 15, 2019 (1st sitting)
  • Ketamine
    • Question 4, 2018 (2nd sitting)
    • Question 22, 2015 (1st sitting)
    • Question 16, 2011 (2nd sitting)
    • Question 7, 2010 (2nd sitting)



Question 12

Question

Describe the process of excitation-contraction coupling and relaxation in smooth muscle


Example answer

Excitation contraction coupling

  • The process linking depolarisation (generated by an action potential) and initiation of muscle contraction


Process

  • Calcium influx
    • Voltage (from action potential) or ligands (e.g hormone, neurotransmitter) can open Ca channels > calcium enters the cell
    • The initial increase in calcium > further release of Ca from the SR (calcium induced calcium release)
    • Hormones/Neurotransmitters can also directly release calcium from the SR via IP3-DAG 2nd messenger pathway
  • Calcium-calmodulin complex
    • Calcium binds to calmodulin forming a complex
    • The complex then activates myosin light chain kinase (MLCK)
  • Activation of MLCK
    • MLCK phosphorylates (activates) the myosin heads (requires ATP)
    • Allows cross bridge formation between actin/myosin
  • Relaxation
    • Decrease in Ca (taken up in the SR by SERCA) leads to relaxation (no further MLCK activation)
    • Myosin phosphatase dephosphorylates already active MLCK


image-20220531131819978
image-20220531131819978



Smooth muscle ECC vs skeletal/cardiac muscle ECC

  • There are no t-tubules
    • The AP propagates through gap junctions (unlike cardiac/skeletal muscle)
  • There is no troponin
    • They facilitate cross bridge formation via calmodulin instead (unlike cardiac/skeletal muscle)
  • Smooth muscle cells have poorly developed sarcoplasmic reticulum
    • Calcium influx is mainly from ECF and not from the SR (unlike cardiac/skeletal muscle)
  • There is DHPR/ ryanodine receptor
    • Calcium induced calcium release from the SR (unlike cardiac/skeletal muscle)
  • 10x slower, lasts 30x longer


Examiner comments


Online resources for this question

  • Jenny's Jam Jar
  • CICM Wrecks
  • Deranged physiology


Similar questions



Question 13

Question

Compare and contrast the pharmacology of suxamethonium and rocuronium


Example answer

Name Suxamethonium (succinylcholine) Rocuronium
Class Depolarising NMB Aminosteroid NMB /

Non depolarising NMB

Indications Facilitate endotracheal intubation during anaesthesia (i.e. RSI) NMB (e.g. intubation, assist with difficult mechanical ventilation)
Pharmaceutics Clear colourless solution (50mg/ml)

Refrigeration (4°C) - 2/52 at room temp
Precipitates with thiopentone

Clear colourless solution (10mg/ml, 5ml vial)

Refrigeration (4°C) - 3/12 at room temp

Routes of administration IV, IM IV
Dose (RSI) 1-2 mg/kg (IV), 2-3 mg/kg (IM)

Cant be given as infusion due to phase 2 block

0.6 - 1.2mg/kg

Can be given as an infusion but variable offset

ED95 0.3mg/kg 0.3 mg/kg
Pharmacodynamics
MOA Binds to the nACh receptor on motor end plate > depolarisation. Cannot be hydrolysed by Acetylcholinesterase in NMJ > sustained depolarisation (i.e. Na channels remain in open-inactive state) > muscle relaxation Inhibit the action of ACh at the NMJ by competitively binding to alpha subunit of nAChR on post junctional membrane
Effects NMB - paralysis. NMB - paralysis
Side effects Major: anaphylaxis, suxamethonium apnoea, malignant hyperthermia

Minor: hyperkalaemia, myalgia, bradycardia/arrhythmia
Pressure: increased IOP, ICP, intragastric pressure.

CVS: tachycardia (in high doses, rare otherwise)

IMMUNE: anaphylaxis (<0.1%)

Pharmacokinetics
Onset/Duration Onset: 30s - 60s

Duration <10 mins

Onset: 45-90s

Duration: ~30-45 mins

Absorption - -
Distribution Protein binding = 30%

Vd = 0.02 L/Kg

Protein binding = 10%

VOD = 0.2 L /kg

Metabolism Rapid hydrolysis by plasma and liver pseudocholinesterase's (~20% reaches NMJ) Minimal hepatic metabolism (<5%)
Elimination Minimal renal elimination (due to rapid metabolism)

T 1/2 = 2 mins

Bile 70%, Renal 30%

Excreted unchanged
T 1/2 - 1 - 1.5hrs

Special points May have prolonged duration of action with congenital or acquired (e.g. liver, renal, thyroid disease) pseudocholinesterase deficiency

Treatment of malignant hyperthermia is with dantrolene (+ cooling + supportive care)

Reversible with sugammadex and anticholinesterases (e.g. neostigmine)


Examiner comments


Online resources for this question

  • Jenny's Jam Jar
  • CICM Wrecks
  • Deranged physiology


Similar questions

  • Rocuronium
    • Question 13, 2018 (2nd sitting)
  • Suxamethonium
    • Question 6, 2011 (1st sitting)
    • Question 2, 2012 (2nd sitting)
    • Question 1, 2013 (2nd sitting)
    • Question 10, 2018 (1st sitting)
    • Question 10, 2020 (2nd sitting)
  • Various other questions relating to properties of NMB more broadly



Question 14

Question

Describe the neural integration of vomiting, highlighting the site and mechanism of action of antiemetics


Example answer

Vomiting

  • Involuntary, forceful and rapid expulsion of gastric contents via the mouth


Stages of vomiting

  • Deep inspiration
  • Closure of the nasopharynx and glottis
  • Large retrograde contraction of intestines > forces content into stomach
  • Relaxation of the oesophagus, lower oesophageal sphincter and body of stomach
  • Contraction of abdominal and thoracic muscles (including diaphragm)
  • Increased intrabdominal pressure > forces gastric contents into oesophagus and through the mouth


Neural integration

image-20220610194545783
image-20220610194545783

Anti-emetics

Class Example MOA
Serotonin antagonists Ondansetron Central and peripheral 5-HT3 receptor antagonism > reduced afferent input to vomiting centre in medulla
Corticosteroids Dexamethasone MOA unclear. May involve: decreased peripheral 5HT release, PG antagonism
Dopamine antagonists Metoclopramide, Droperidol Central D2 antagonism at chemoreceptor trigger zone > reduced afferent input to vomiting centre in medulla
Antihistamine Cyclizine Competitive H1 antagonism. Also has
NK1 antagonist Aprepitant Blocks action of substance P in brainstem vagal complexes involved in regulation of vomiting
Others Canabinoids Thought to act within the vomiting centre


Examiner comments


Online resources for this question

  • Jenny's Jam Jar
  • CICM Wrecks
  • Deranged physiology


Similar questions

  • Question 3, 2014 (1st sitting)




Question 15

Question

Describe the sequence of haemostatic events following injury to a blood vessel wall until clot stabilisation


Example answer

Examiner comments


Online resources for this question

  • Jenny's Jam Jar
  • CICM Wrecks
  • Deranged physiology



Similar questions

  • Question 10, 2019 (1st sitting)



Question 16

Question

Outline the impact of sedative agents on thermoregulation (40% marks) and describe the physiological effects of a low body temperature (60% marks)


Example answer

Core body temperature

  • 37 degrees +/- 0.4 degrees


Effects of anaesthesia

  • The interthreshold range

    • The range of core temps at which no autonomic thermoregulatory responses occurs

    • Normally 37 +/- 0.2 degrees

    • With general anaesthesia this is widened to 37 +/- 2 degrees

  • Responses to general anaesthetic

    • Behavioural responses completely abolished (seek warmth, clothing etc)

    • Volatile anaesthetics, opioids (e.g. morphine), IV sedatives (e.g. propofol) > vasodilation > increased heat loss.

    • Decreased responsiveness to temperature change (widened interthreshold range)

    • Opioids also decrease sympathetic outflow > impaired vasoconstriction

    • If adjunct muscle relaxant used -> shivering also prevented (decreased heat generation)

    • Therefore the only thermoregulatory responses available to anaesthetised/paralysed patient with hypothermia are vasoconstriction and non shivering thermogenesis. Hence body temperature changes passively in proportion to the difference in heat production and heat loss

Mild hypothermia

  • 34-36.5
  • Common during anaesthesia


Effects of hypothermia

  • METABOLIC

    • BMR drops 6% for every 1 degree in core temp --> Decreased VO2

    • Hyperglycaemia (decreased cell uptake)

  • CVS

    • Decrease inotropy and chronotropy > decreased CO

    • Arrhythmias (AF, VF)

    • QT prolongation, J wave

    • Resistance to DCCV

    • Peripheral vasoconstriction and blood redistribution

    • Increased risk of myocardial ischaemia

  • CNS

    • Confusion and decreasing LOC

    • Shivering

    • Seizures (increased seizure threshold)

  • RESP

    • Decreased RR

    • Left shift of O2 dissociation curve

  • GIT

    • Ileus

    • Decreased hepatic drug metabolism and clearance (slows enzymatic reactions, decreased blood flow)

  • HAEM

    • Increased HCT and blood viscosity

    • Coagulopathy,

    • Platelet dysfunction and sequestration

  • RENAL

    • Cold diuresis (decreased vasopressin synthesis)

  • ENDO

    • Decreased ACTH, TSH, vasopressin

  • ACID-BASE

    • Increased pH


Examiner comments

33% of candidates passed this question.

Sedation reduces body temperature by interfering with heat production and increasing heat loss, along with widening of hypothalamic inter-threshold range. This portion of the question was generally well answered. The question asked to "outline" the answer. Many candidates actually "described" the thermoregulation process in general but were unable to relate those with the impact of sedation. The second part of the question (physiological effect of low body temperature) was answered by most of the candidates with the structure of organ-system wise description. A few candidates scored extra marks by relating these effects with degree of hypothermia and by describing how thermogenesis responses (including shivering) can influence those effects. Some candidates restricted their answers to the effect of thermogenesis in response hypothermia and did not include the overall physiological consequences of low body temperature. Better answers displayed an understanding of core temperature regulation, inter-threshold range and the effects of sedatives on thresholds for thermogenic responses, although only a few mentioned gain and maximal response. Better answers included specific detail (mentioned bradyarrhythmia, slow AF, VF, prolonged PR/QRS / J waves rather than just stating arrhythmia) across several organ systems. Marks were not awarded for generic statements such as 'decreased liver function' without some additional detail. Inadequate depth of knowledge was main reason behind overall poor scores.


Online resources for this question

  • Jenny's Jam Jar
  • CICM Wrecks
  • Deranged physiology



Similar questions

  • Question 9, 2021 (1st sitting)




Question 17

Question

Write notes on:

  • The principles of ultrasound
  • Transducer properties and image resolution
  • The doppler effect


Example answer

Ultrasound = sound waves at higher frequencies then can be heard by humans (>20kHz)


Sound wave generation

  • Produced by piezoelectric effect
    • Electrical voltage applied to quartz (piezoelectric) crystal > vibrates > sound emitted (Converts electrical energy to sound energy)
  • Frequency of sound wave
    • Different probes emit different frequencies of sound waves:
      • Liner = 15-6 MHz
      • Curved = 8-3 MHz
      • Cardiac = 5-1 MHz
    • Effects
      • Higher frequency = shallower depth, better resolution
      • Lower frequency = better depth, less resolution


Sound wave effected by tissue and may be

  • Absorbed
    • Sound is absorbed, lost as heat
  • Reflected
    • Sound reflected off objected back to probe sensor
    • Reflection occurs at interfaces of tissues with different densities (impedance)
  • Refracted
    • Change in direction (bending) of sound wave
  • Scattered
    • Sound reflected from tissue but not received by probe sensor


Detection of sound

  • The probe spends 1% time emitting sound, 99% time listening for sound
  • The crystals are vibrated by returning sound waves (echos) and generates a voltage (converse piezoelectric effect)


Processing

  • Amplitude of the wave determines echogenicity
  • Time taken for echo's to return determines depth
  • Output mode
    • B mode (brightness mode)
      • 2D crossection through tissue
      • Largest amplitude = brightest, smallest amplitude = darkest
    • M mode (motion mode)
      • Movement of structures over time
  • Resolution
    • Spatial resolution
      • Dependant on axial (parallel to beam) and lateral (perpendicular to beam) resolution
      • Enhances by pulse wave and focusing
    • Contrast resolution
      • Distinguish between two regions of similar echogenicitiy
    • Temporal resolution
      • Distinguish change over time
      • Improved by framerate


Doppler effect

  • The change in frequency of a wave in relation to an observer that is moving relative to the wave source.
  • In medical ultrasound
    • The change in frequency of sound waves reflected from moving tissue (e.g. erythrocytes)
      • Away from probe (lower frequency = blue colour)
      • Towards probe (higher frequency = red colour)


Examiner comments


Online resources for this question

  • Jenny's Jam Jar
  • CICM Wrecks
  • Deranged physiology



Similar questions




Question 18

Question

Describe the anatomy of the left subclavian vein


Example answer

Course

  • Continuation of the axillary vein (commences at the lateral border of the 1st rib)
  • Travels medially, first arching cephalad before heading caudally toward the sternal notch
  • Terminates by joining the internal jugular vein (IJV) and forming the brachiocephalic (innominate) vein behind the sternoclavicular joint


Tributaries

  • External jugular vein
    • Drains into the subclavian at the lateral border of anterior scalene muscle
  • Thoracic duct
    • Drains into either subclavian or innominate vein behind the sternoclavicular joint


Relationships

  • Cephalad
    • Skin, subcutaneous tissue, platysma
  • Anterior
    • Skin, clavicle, subclavius muscle
  • Posterior
    • Anterior scalene muscle which separates from the subclavian artery
  • Posterior-inferiorly
    • 1st rib, pleura, phrenic nerve
  • Medial
    • Brachiocephalic vein, mediastinal structures (vagus, trachea, aorta)
  • Lateral
    • Axillary vein, brachial plexus


Surface anatomy (infraclavicular approach)

  • Needle is placed in the deltopectoral groove, inferior and lateral to the middle third of the clavicle.
  • The needle is inserted at a shallow angle, passing under the middle third of the clavicle aiming at the sternal notch.




Examiner comments


Online resources for this question

  • Jenny's Jam Jar
  • CICM Wrecks
  • Deranged physiology



Similar questions




Question 19

Question

Describe the physiological factors that affect PaCO2


Example answer

Overview

  • PaCO2 is normally 40mmHg +/- 3mmHg
  • PaCO2 is a balance between production and elimination


CO2 production

  • Metabolism
    • CO2 is the by-product of mitochondrial respiration via TCA cycle
    • Increased metabolism > increased CO2 production
      • Metabolic rate is increased with
        • Exercise
        • Increased temp (e.g. infection)
        • Youth
        • Male sex
        • Pregnancy
        • Eating
  • Energy source
    • RQ = Ratio of Co2 produced: O2 consumed
      • Effected by the energy source utilised
      • Normally
        • Fats 0.7
        • Ketones/alcohols 0.7
        • Proteins 0.8
        • Carbohydrates 1.0
    • Hence carbohydrates Increase CO2 production compared to lipids/proteins (though hot chips are delicious)


CO2 elimination

  • Alveolar ventilaiton
    • CO2 is a ventilation limited gas
    • Increase minute ventilation (RR, TV) > increase CO2 elimination via respiration > decreased PaCO2
    • Physiological factors that increase RR: Pain, anxiety, pregnancy, Hypoxia
  • Tightly regulated
    • Minute ventilation is increased by increased PaCo2
      • MV linearly increases 2L/min for every 1mmmHg increase in PaCo2
      • Due to the chemoreceptor responses
        • Central chemoreceptors
          • Located: Ventral medulla
          • CO2 diffuses across BBB > increased H+ > decreased pH > detected by chemoreceptor > stimulate dorsal resp group > Increase MV
        • Peripheral chemoreceptors
          • Located: Carotid bodies, aortic bodies
          • Increased PaCo2 or decreased PaO2 > stimulate the respiratory centre > increased MV


Examiner comments

33% of candidates passed this question.

Candidates who scored well generally defined PaCO2 and proceeded to describe factors in terms of those related to production and elimination. Good answers described the key production factor as being rate of production through aerobic metabolism which is in turn influenced by substrate and BMR. Those who scored well described elimination as being dependent upon minute ventilation, which in turn is influenced by CO2 detection by chemoreceptors, specifically detailing the difference between peripheral and central. Many candidates detailed pathophysiological factors which unfortunately did not gain any marks.


Online resources for this question

  • Jenny's Jam Jar
  • CICM Wrecks
  • Deranged physiology



Similar questions


Question 20

Question

Describe the physiological control of systemic vascular resistance (SVR)


Example answer

Examiner comments


Online resources for this question

  • Jenny's Jam Jar
  • CICM Wrecks
  • Deranged physiology



Similar questions

  • Question 7, 2020 (1st sitting)





2021 (2nd sitting)

Question 1

Question

Describe the regulation of body water


Example answer

Overview

  • Water intake
    • Approximately 30ml/kg of water is needed to be ingested for fluid/body homeostasis
      • Approx 2-2.5L per day for an average person
    • Approximately half comes from drinking fluids, half from food and metabolic processes
  • Water is lost through numerous ways
    • Urine
      • Approx 1 - 1.5L / day
      • Obligatory loss is ~500mls to cover solute/waste clearance
    • Insensible losses (skin, lungs etc)
      • Approx 900mls / day
    • Faeces
      • Approx 100mls / day
  • The body tightly regulates water balance to preserve plasma osmolality and intravascular volume status, but also allow waste clearance
    • Preservation of blood volume takes precedence over plasma osmolality


REGULATION

  • Sensors
    1. Osmoreceptors in hypothalamus detect increased (>290mosm/L) osmolality with dehydration (major)
    2. Low pressure baroreceptors (RA, great vessels) detect reduced pressure (stretch) with dehydration
    3. High pressure baroreceptors (carotid sinus, aortic arch) detect reduced pressure (stretch) with dehydration
    4. Macula densa (kidneys) detect reduced GFR (Na/Cl delivery)
  • Integrator
    • Hypothalamus (anterior and lateral predominately)
  • Effector/effects
    1. Release of ADH
      • Synthesised in hypothalamus transported to posterior pituitary for release
      • ADH acts on collecting ducts in the kidney in to increase aquaporins on luminal wall --> increased water reabsorption
      • Released in response to increase osmolality and activation of RAAS
    2. ANP/BNP
      • Decreased stretch > decreased ANP/BNP secretion --> increased water reabsorption
    3. RAAS
      • Decreased baroreceptor activation --> increased renin release
      • Decreased GFR sensed by macula sensa > increased renin release
      • Renin > activation of RAAS > increased water reabsorption
    4. Thirst centre (hypothalamus)
      • Activation of thirst centre in the lateral hypothalamus (due to increased osmolality) > behavioural change to increase water intake
  • Feedback
    • The above systems work predominately on a negative feedback system


Examiner comments

28% of candidates passed this question.

This is a level 1 topic. An understanding as to how the body regulates water is crucial to the daily practice of critical care, this topic is well described in the major texts. This type of question lends itself to the basic template of sensor mechanisms, central processing and integration with effector limbs and feedback loops. However, high scoring answers require a quantification of responses and an introduction into how these processes are integrated and fine-tuned.


Online resources for this question

  • CICM Wrecks
  • Deranged Physiology
  • Jenny's Jam Jar


Similar questions

  • Question 8, 2008 (1st sitting)
  • Question 4, 2015 (1st sitting)
  • Question 9, 2018 (2nd sitting)



Question 2

Question

Describe the pharmacology of lidocaine


Example answer

Name Lidocaine (lignocaine)
Class Amide anaesthetic / Class 1b antiarrhythmic
Indications Local/regional/epidural anaesthesia, ventricular dysrhythmias, IV analgesia,
Pharmaceutics Clear colourless solution (1%, 2%, 4%). Can come with/without adrenaline. Also available as cream/spray
Routes of administration SC, IV, epidural, inhaled
Dose Regional Use: Toxic dose 3mg/kg (without adrenaline), 7mg/kg (with adrenaline)

IV use: 1mg/kg initially, then ~1-2mg/kg/hr

pKA 7.9 (25% unionised at normal body fluid pH)
Pharmacodynamics
MOA Class 1b anti-arrhythmic: blocks Na channels, raising threshold potential + reducing slope of Phase 0 of action potential, shortened AP

Local anaesthetic: binds to, and blocks, internal surface of Na channels

Effects Analgesic, anaesthetic, anti-arrhythmic
Side effects CNS: headache, dizziness, confusion, paraesthesia, reduced LOC, seizures

CVS: hypotension, bradycardia, AV Block, arrhythmia
CC/CNS ratio = 7 (lower number = more cardiotoxic)

Pharmacokinetics
Onset Rapid onset (1-5 minutes)
Absorption IV > Epidural > subcut.

Oral bioavailability 35%

Distribution 70% protein bound,

Vd ~1L/kg.
Crosses BBB

Metabolism Extensive hepatic metabolism with some active metabolites
Elimination Metabolites excreted in urine.

Half life ~90mins. Increased with adrenaline (SC). Reduced in cardiac/hepatic failure.

Special points


Examiner comments

71% of candidates passed this question.

The answers for this question were generally of a good standard. Lidocaine is a core drug in intensive care practice and thus a high level of detail was expected. This question was best structured using a standard pharmacology template (pharmaceutics, pharmacokinetics and pharmacodynamics). A small number of answers omitted any pharmaceutic elements. Another common error was the use of vague and imprecise statements. For example, many answers stated that the maximum dose (without adrenaline) is 3 mg/kg, without specifying that this is subcutaneous. The concept of the ratio of the dose required to produce cardiovascular collapse to that required to induce seizures (CC/CNS ratio) was often mentioned. However, in many cases this was conveyed simply as an abbreviated statement without any additional explanation leaving the examiner unsure as to whether the candidate understood the concept (and thus unable to award any additional marks). In addition, many candidates confused the order of this ratio (incorrectly referring to it as a CNS/CC ratio of 7). Lastly, few answers made specific mention of the narrow therapeutic index and the associated implications for use in the ICU.


Online resources for this question


Similar questions

  • Question 17, 2014 (1st sitting)
  • Question 1, 2019 (1st sitting)



Question 3

Question

Discuss the physiological determinants of cardiac output


Example answer

Cardiac output

  • The volume of blood ejected from the heart per unit time
  • <math display="inline">CO = HR \; \times SV</math> and <math display="inline">CO \; = \; VR</math>
  • CO is approximately 5L/min in the average person


Factors affecting CO


Heart rate

  • If stroke volume remains the same, then increasing HR will increase CO
  • However, in a healthy person within physiological HRs (60-150), if there is no increase in physiological demand, altering HR has limited effects on CO as the stroke volume reduces
  • In extremes of HR, with increased metabolic demand, or pathology (e.g. poor contractility), altering HR will impact cardiac output


Stroke volume (SV)

  • Stroke volume = EDV - ESV, Normally ~70mls
  • Increased stroke volume = increased CO
  • Factors affecting stroke volume include
    • Preload
      • Myocardial sarcomere length just prior to contraction.
      • Cannot be measured, so approximated by EDV
      • Increased preload > incre
      • Factors effecting preload include
        • Ventricular compliance
        • Venous return
        • Valvular disease
        • Heart rate
        • Myocardial wall thickness
        • Atrial contractility
    • Afterload
      • External force required to be generated before the mycoardial sarcomere begins to shorten
      • Reduced afterload > increased SV > increased CO
      • Factors affecting afterload include:
        • Systemic vascular resistance
        • Outflow tract impedance
        • Transmural pressure
        • Myocardial wall thickness
    • Contractility
      • Intrinsic ability of the myocardial fibres to shorten/contract
      • Increased contractility = increased SV = increased CO
      • Factors effecting contractility include
        • Bowditch effect
        • Anrep effect
        • Tone
        • Heart rate
        • Ischaemia/drugs.


Examiner comments

65% of candidates passed this question.

Although the pass rate for this question was reasonably high the examiners commented on a lack of detailed knowledge within most answers for such a core component of our daily practice. Several candidates failed to provide a normal value and only few provided anything other than 5l/min. There was a general lack of detail, and at times, some confusion about the Frank Starling effect. Most candidates outlined the major determinants of stroke volume, although many were light on the determinants of each or incorporated incorrect facts. Several candidates did not mention HR as a determinant of CO


Online resources for this question


Similar questions

  • Question 8, 2011 (1st sitting)
  • Question 13, 2011 (2nd sitting)
  • Question 19, 2014 (1st sitting)



Question 4

Question

Compare the pharmacology of fluconazole and amphotericin


Example answer

Name Fluconazole Amphotericin
Class Azole / antifungal Polyenes / antifungal
Indications Systemic fungal infections, prophylaxis fungal infections for immunocompromised Systemic fungal infections
Pharmaceutics Tablet (PO), White powder which is clear and colourless in solution (water, saline). Powder for reconstitution and injection
Routes of administration PO, IV IV, lozenges, inhalation
Dose Generally 200-800mg daily for systemic infections, reduced dose local infections or prophylaxis (e.g 50-200mg daily) ~1-5mg/kg daily
Pharmacodynamics
MOA Fungicidal. Disrupts ergosterol production (essential for cell membrane formation) leading to increased permeability. Fungicidal. Binds directly to ergosterol > creates transmembrane channels > permeability > death
Coverage Covers: Candida albicans, Cryptococcus

Doesnt: Most other fungi/yeast, aspergillus

Good activity against almost all fungi/yeasts (inc. aspergillus, candida, crypto)
Side effects Liver: Potent CYP450 inhibitor > many drug interactions, LFT derangement

CNS: headache
CVS: QT prolongation
Derm: rash, alopecia
GIT: N/V, diarrhoea, abdo pain

Nephrotoxicity, hypokalaemia, infusion reactions , RTA
Pharmacokinetics
Onset Peak concentrations 1-2hours
Absorption Great oral bioavailability (>90%) Poor oral bioavailability (hence only given IV)
Distribution Vd close to that of water ~0.7L/Kg.

Good CSF, tissue, fluid penetration.
Poorly protein bound (10%)

Highly protein bound (90%). Negligible CSF/urine distribution.

VOD = ~1L /Kg

Metabolism Not metabolised Minimal hepatic metabolism
Elimination Renal (unchanged 80%).

T 1/2 ~30 hours

Renal/faecal elimination.

T 1/2 = 15 days.

Special points Monitoring: LFTs, drug interactions Monitoring: renal function


Examiner comments

6% of candidates passed this question.

This question exposed an area of the syllabus neglected by the candidates. Answers were generally vague in detail with lots of incorrect facts and generally displayed a very limited knowledge. Antifungal agents are regularly used in critically ill patients either as treatment or prophylaxis. An understanding of the aspects of these drugs with respect to spectrum of activity, mechanism of action, specific PK and PD properties as well as potential side effects would have been the basis for this compare and contrast question. Examiners want to be convinced that the candidates understand the strengths and weaknesses of each drug and in which circumstances one agent might be used in preference to the other.


Online resources for this question


Similar questions

  • ?Nil



Question 5

Question

Write detailed notes on angiotensin, including its synthesis, role within the body and regulation


Example answer

Synthesis and regulation

  • Angiotensinogen
    • Peptide hormone continuously synthesised in the liver and is a precursor to angiotensin
    • Increased release in response to corticosteroids, oestrogens, thyroid hormones, AGT2 levels
  • Renin converts angiotensinogen to Angiotensin I
    • Renin is produced, stored, secreted from JG cells in kidney
    • Stimulated by reduced GFR, decreased Na/CL delivery to MD, sympathetic innervation
    • Inhibited by Angiotensin II (negative feedback)
  • Angiotensin converting enzyme (ACE) converts Angiotensin I to Angiotensin II
    • ACE is present in the capillary endothelial cells in the lungs (and renal endothelium)
  • There is also angiotensin III and IV which are the product of further cleavage by peptidases
    • These have similar effects to angiotensin II (but reduced potency)
  • The renin-angiotensin-aldosterone system (RAAS) exerts negative feedback on the release of renin, additionally increased BP and Na/Cl delivery will decrease renin secretion


Effects of angiotensin

  • Angiotensin I
    • Thought to be physiologically inactive, but acts as a precursor to Angiotensin II
  • Angiotensin II
    • Renal effects
      • Increases Na-H antiporter activity in PCT > increased Na/Water reabsorption
      • Vasoconstriction of afferent + efferent arterioles + contraction mesangial cells > reduced GFR + urine output
    • CVS effects
      • Binds AT1 receptors > vasoconstriction >increased SVR > inc. BP
    • Neurohormonal effects
      • Increases sensation of thirst through activation of hypothalamus > increased blood volume
      • Increases the release of ADH from the pituitary gland
        • ADH Increases water reabsorption by inserting aquaporins in the collecting ducts
      • Increases production and release of aldosterone from adrenal cortex
        • Aldosterone increases blood volume: Increases Na/Water reabsorption in DCT
        • Aldosterone increases blood pressure: by increasing blood volume, but also by direct vasopressor effects


Examiner comments

24% of candidates passed this question.

This question provided headings for the answer template. Good answers integrated the required facts from the appropriate chapters of the major texts. Most answers lacked detail surrounding the factors that increase or decrease angiotensin activity. Few answers provided any detail as to all the mechanisms through which angiotensin exerts it effects. A lot of answers focussed singularly on the vascular effects of angiotensin. Overall, there was often a paucity of detail, with vague statements and incorrect facts


Online resources for this question


Similar questions

  • Question 18, 2009 (2nd sitting)
  • Question 6, 2011 (2nd sitting)



Question 6

Question

Describe the functions of the placenta (80% marks). Outline the determinants of placental blood flow (20% marks).


Example answer

FUNCTIONS OF PLACENTA


Nutrient/gas exchange

  • The foetus relies on maternal transfer of gasses, nutrients and wastes

  • Nutrients/wastes

    • Active transport e.g Amino acids, calcium, some vitamins/minerals

    • Facilitated diffusion e.g. glucose (GLUT1 and GLUT3)

    • Simple diffusion e.g. Na, Cl, urea, creatinine based on Fick Principle

  • Gasses

    • Oxygen

      • Passive diffusion

      • Facilitated by higher oxygen carrying capacity and affinity of foetal Hb as well as the Bohr/Double bohr effects

    • Carbon dioxide

      • Passive diffusion

      • Facilitated by the Haldane and double Haldane effects

Immunological function

  • Foetus is genetically distinct with a non functioning immune system
  • Trophoblast cells
    • Lose MHC molecules and become coated in mucoprotein > less immunogenic
  • Chorionic cells
    • Prevent maternal T cells and most immunoglobulins (except IgG) from entering > less immunogenic
    • Barrier to some bacteria/viruses and allows IgG across > some immune protection
  • Yolk sac
    • a-fetoprotein and progesterone are immunosuppressive > less immunogenic


Endocrine function

  • Syncytiotrophoblast of placenta produces
    • B-HCG - prolongs corpus luteum (prevents early miscarriage)
    • Oestrogen - increases uteroplacental blood flow, stimulates uterine growth
    • Progesterone - uterine relaxation, development of lactation glands
    • hPL - maternal lipolysis, breath growth/development


PLACENTAL BLOOD FLOW


Flow

  • Blood flow to the uterus in a non pregnant woman is normally around 200mls/min (~4% of CO)
  • In a pregnant woman at term this increases to up to 750mls/min (~15% of CO)
  • Majority of this > placenta (~600mls/min), with some supplying the hypertrophied uterus.
  • Foetal blood flow is approx half placental blood flow (~300mls/min, ~60%CO)


Determinants of flow

  • No autoregulation of uteroplacental blood flow
  • Most important factor governing flow is therefore perfusion pressure
    • Increased uteroplacental perfusion pressure > increase flow
  • Uterine perfusion pressure is therefore effected by
    • Maternal MAP
      • Effected by positioning (e.g. aortocaval compression), cardiac output, systemic vascular resistance
    • Intrauterine pressure
      • Effected by contractions > increased intrauterine pressure > decreased flow
    • Uterine vascular resistance
      • The radial arteries of the myometrium are modestly effected by exogenous vasopressors, catecholamines
  • Compensates for the lack of autoregulation by increasing oxygen extraction


Examiner comments

49% of candidates passed this question.

There was a wide range of marks for this question with a few candidates scoring excellent marks. Those answers that scored well provided a comprehensive list of functions as well as an explanation as to the what, how and/or why of these functions. Poorer answers omitted some of the functions or failed to elaborate on them by providing only a limited list. The second component of the question was generally well outlined, most candidates provided some estimate of normal values at term and a simple elaboration regarding the factors that affect placental blood flow.


Online resources for this question


Similar questions

  • Question 9, 2018 (1st sitting)




Question 7

Question

Outline how the measurement of the following can be used in the assessment of liver function (25% marks of each): 1) Albumin 2) Prothrombin time 3) Glucose 4) Ammonia


Example answer

  1. Albumin

    • Albumin is a protein synthesized in the liver (half life ~3 weeks)

    • Normally 34-45g/L in the blood

    • With chronic liver dysfunction there is reduced synthesis > low albumin

    • More commonly related to other diseases

      • Malnutrition

      • Protein loss (e.g. nephrotic syndrome)

      • Physiological dilution (e.g. pregnancy)

      • Inflammation/stress (negative acute phase reactant)

  2. Prothrombin time

    • Measures the rate of conversion of prothrombin to thrombin

    • Normal PT = 10-13 seconds

    • Most coagulation factors are synthesised by the liver

    • If synthetic function of the liver is impaired (e.g. by severe cirrhosis) there would be a prolonged prothrombin time.

    • If synthetic function of the liver is normal, but prothrombin time is prolonged, this would imply drug effect (eg. warfarin), consumptive coagulopathy, or VitK deficiency


  1. Glucose

    • Essential energy substrate

    • Normal BGL = 4-6 for most people (physiologically varies with diet/time)

    • Liver is important for glucose homeostasis including glycolysis, glycogenolysis and gluconeogenesis

    • Liver failure may lead to both diabetes as well as hypoglycaemia

    • Blood glucose levels or neither sensitive, nor specific for liver dysfunction

    • Hypoglycaemia may be causes by numerous other conditions including pancreatic disorders, stress, drugs, diet/malnutrition, GIT absorption issues etc.

  2. Ammonia

    • Ammonia is a nitrogenous waste product

      • Produced by amino acid metabolism, urea hydrolysis in the GIT and renal synthe

    • Normal level <30ug/L in adults

    • Normally transported to liver for conversion to urea via urea cycle > excreted kidneys

    • If liver is unable to metabolise ammonia > accumulates

    • Hyperammonaemia is relatively specific to cirrhotic liver disease (90% of cases)

    • Other causes include

      • Haematological disorders (e.g. myeloma)

      • Congenital defects in urea-cycle function

      • Drugs: e.g. valproate


Examiner comments

54% of candidates passed this question.

This was a new question and overall, most candidates provided some detail on each component as requested. Those answers that used a simple template for each section generally scored better than those who wrote in a paragraph style for each section. Areas expected to be covered included the following; a definition of the variable to provide context, a normal value and the range of influences that effect the variable both related to liver function and or extrinsic to the liver (attempting to introduce the concepts of sensitivity and specificity for each test). Stronger answers provided some context as to whether the variable was sensitive to acute or chronic changes in liver function and which synthetic/metabolic component of the liver the variable represented


Online resources for this question

  • CICM Wrecks
  • Deranged Physiology
  • Jenny's Jam Jar


Similar questions

  • Nil



Question 8

Question

Describe the anatomy of the internal jugular vein including surface anatomy landmarks relevant to central venous line insertion.


Example answer

Internal jugular vein

  • Originates at the jugular bulb (confluence of the inferior petrosal and sigmoid sinus')
  • Exits skull via the jugular foramen with CN IX, X, XI
  • Descends inferolaterally in the carotid sheath (initially posterior > lateral to carotid artery with descent)
  • Terminates behind the sternal end of the clavicle where it joins with the subclavian vein to form the brachiocephalic vein
  • Tributaries: facial, thyroid, pharyngeal, lingual veins
  • Right IJV usually larger then left


Relations

  • Anterior to IJV: SCM, lymph nodes, CN XI
  • Posterior to IJV: scalene muscles, lung pleura, lateral mass C1, vagus (poster-medial)
  • Inferior/at termination: pleura (extends 2cm above clavicle)
  • Medial: vagus, carotid artery


Variations

  • Stenosis, complete occlusion, aneurysms, absence
  • Variation in relation to carotid (e.g. anterior) in up to 1/4 cases


Ultrasound anatomy

  • Often lateral to carotid (not always) and often larger than carotid
  • Unlike carotid: Non pulsatile, thin walled, compressible
  • Doppler flows can also be helpful.


Surface anatomy

  • Identify triangle between the clavicle and two heads of SCM
  • Palpate carotid
  • Puncture lateral to carotid artery at 30 degree angle
  • Aim caudally towards ipsilateral nipple


Examiner comments

38% of candidates passed this question.

The overall pass rate for this question was poor considering how relevant this area of anatomy is in our daily practice. Better scoring answers used a template including a general description, origin, course and relations, tributaries and as requested in this question, the surface anatomy. Many answers that scored poorly only provided the briefest detail, were vague in their descriptions and incorrect with respect to the facts presented or imprecise with respect to the terminology used


Online resources for this question


Similar questions

  • Question 23, 2017 (2nd sitting)



Question 9

Question

Outline the classification and effects of beta-blocking drugs, including examples (50% marks). Compare and contrast the pharmacokinetics of metoprolol with esmolol (50% marks).


Example answer

Classification of beta blockers

  • All beta blockers are competitive antagnoists
  • Can be classified according to
    • Receptor selectivity
      • Non selective (B1 and B2) e.g. sotalol, propranolol
      • B1 selective e.g. metoprolol, esmolol, atenolol
      • A and B effects: labetalol, carvedilol
    • Membrane stabilising effects (inhibit AP propagation)
      • Stabilising e.g. Propanolol, metoprolol
      • Non stabilising e.g. atenolol, esmolol, bisoprolol
    • Intrinsic sympathomimetic activity
      • ISA e.g. labetalol, pindolol
      • Non ISA e.g. metoprolol, sotalol, propranolol, esmolol


Effects of beta blockers

  • B1 antagonism
    • Heart: decreased inotropy and chronotropy (decreased BP), decreased myocardial oxygen consumption, decreased AV nodal conduction
    • Kidneys: decreased renin release > decreased RAAS activation > decreased BP
  • B2 antagonism
    • Respiratory: bronchoconstriction
    • Circulation: vasoconstriction
    • Skeletal muscle: reduced glucose uptake
    • Eye: decreased aqueous humour production
  • B3 antagonism
    • Adipose tissue: reduced lipolysis


Compare/contrast metoprolol and esmolol pharmacokinetics

Name Metoprolol Esmolol
Pharmacokinetics
Onset Immediate when IV Immediate (only given IV)
Absorption 95% absorption, 50% oral bioavailability (1st pass effect) 0% oral bioavailability
Distribution VOD 5L/kg

10% Protein bound
High lipid solubility, readily crosses BBB

VOD 3L/kg

60% protein bound
High lipid solubility, can cross BBB

Metabolism - Hepatic CYP450

- Significant 1st pass metabolism.
- Inactive metabolites

- Blood

- Hydrolysis by RBC esterase

Elimination Renal excretion

T 1/2 approx 4 hours

Minimal renal excretion
T 1/2 = 10 mins


Examiner comments

59% of candidates passed this question.

This was a two-part question with marks and thus timing of the answers given as a percentage. There are generally many ways to classify drugs within the same class. These are usually well described in the relevant recommended pharmacological texts. Receptor distribution throughout the body and the effect of the drug-receptor interaction are useful ways to organise systemic pharmacodynamic responses, as opposed to a list of organ systems with associated vague statements of interaction


Online resources for this question


Similar questions

  • Question 14, 2019 (2nd sitting)



Question 10

Question

Describe the ventilation / perfusion (V/Q) relationships in the upright lung according to West’s zones (40%). Explain the physiological mechanisms responsible for these relationships (60%)


Example answer

West zones

  • A way of describing the regional differences in alveolar, arterial and venous pressures in the lung
  • Initially Zones 1-3 described, with a 4th later added


West zone 1

  • Pressure alveolus (PA) > arterial pressure (Pa) > venous pressure (Pv)
  • Alveolus compresses arterial and venous flow. Hence there is ventilation but no perfusion
  • Leads to infinite V/Q (dead space)
  • Generally does occur under physiological conditions but can when
    • Alveolar pressure is very high (very high PEEP)
    • Arterial pressure is very low (shock)


West Zone 2

  • Pa > PA > Pv
  • Intermittent blood flow throughout cardiac cycle. PA acts as starling resistor
  • Seen in the lung apex > rib ~3
  • V/Q As high as 3.0


West Zone 3

  • Pa > Pv > PA
  • Blood flows continuously throughout the cardiac cycle
  • Majority of lung below ~Rib 3
  • V/Q approaches 0.3


West zone 4

  • Pa > Pi (intersitital fluid) > Pv > PA
  • With interstitial fluid acting as a starling resistor
  • Seen in pathological states e.g. pulmonary oedema


V/Q ratio (in upright person)

  • Perfusion (Q)
    • Increases from apex > base of lung
    • Due to the effects of gravity > increased hydrostatic pressure
  • Ventilation
    • Increases from apex > base of lung
    • Because of the vertical gradient of pleural pressure (-10cmH2O apex, -3cm base) the apices get less ventilation than the bases at normal lung volumes as they are less compliant
  • V/Q ratio
    • Because blood is denser than air, the effect of gravity is greater on perfusion than ventilation
    • At about the level of rib 3: V/Q ratio is approx 1.
    • Above rib 3 (West zone 1/2): V/Q > 1.0
    • Below rib 3 (West zone 3): V/Q < 1.0


Examiner comments

47% of candidates passed this question.

This is a core aspect of respiratory physiology, and a detailed understanding of this topic is crucial to the daily practise of intensive care. As such the answers were expected to be detailed. Strong answers included precise descriptions of the zones of the lung as described by West and related these to the V/Q relationship in the upright lung. Generally, most candidates scored well in this section. Diagrams were of varying value. However, an impression from the examiners was that candidates spent too much time on this first section and ran out of time for a detailed answer in the second section. The answers to the second section seemed rushed and were often lacking in detail with many incorrect facts. This question highlights the importance of exam technique preparation in the lead up to the written paper


Online resources for this question


Similar questions

  • Nil



Question 11

Question

Provide a detailed account of the side-effects of amiodarone.


Example answer

Overview

  • Amiodarone is class III antiarrhythmic drug, which also has class I, II and IV activity
  • It is predominately used for treatment of tachyarrhythmias
  • It can be given PO or IV
  • It has numerous side effects which increase in likelihood and severity with duration/dose of therapy.
  • More than 50% patients will experience side effects with long term use.
  • Most side effects are reversible if treatment is stopped


Side effects by organ/body system

  • CNS
    • Peripheral neuropathy and myopathy
    • Sleep disturbance (10%)
  • CVS
    • Bradycardia and hypotension (esp. if given rapidly)
    • QT prolongation
  • RESP
    • Pneumonitis, pulmonary fibrosis, pleuritis - all dose related
    • High FiO2 requirement at time of therapy seems to be a risk factor
    • Mortality for amiodarone induce lung toxicity is 10%
  • GIT
    • Can cause cirrhosis, hepatitis (<5%), LFT derangement (15%), GIT upset
  • OPHTHALMIC
    • Corneal microdeposits (90%) > blurring (10%)
  • ENDOCRINE:
    • Can cause both hypothyroidism (~5%) and hyperthyroidism (~1%)
  • DERM:
    • Photosensitivity (50%), skin discolouration (<10%)
  • PHARM
    • Amiodarone can potentiate/interact with numerous other drugs, by displacing them from proteins, increasing their free fraction (e.g. phenytoin, warfarin)
  • PREGNANCY
    • Neurodevelopmental abnormalities
    • Risk of congenital hypothyroidism


Examiner comments

17% of candidates passed this question.

The question asked for a detailed account of the side effects of amiodarone, hence those candidates that just provided a list or outline scored less well. It was expected that candidates provide some detail of the side effect. Answers that scored well prioritised those relevant to ICU clinical practice. Many provided disorganised outlines of the side effects and frequently the cardiovascular side effects were poorly explained. Many candidates omitted the important drug interactions of amiodarone use and few candidates related the side effect profile to the duration of treatment


Online resources for this question


Similar questions

  • Question 5, 2008 (2nd sitting)
  • Question 22, 2010 (2nd sitting)
  • Question 14, 2014 (2nd sitting)
  • Question 11, 2016 (1st sitting)
  • Question 2, 2018 (2nd sitting)



Question 12

Question

Explain the physiological factors that affect airway resistance


Example answer

Airway resistance

  • Equal to the pressure difference between alveoli and the mouth divided by the flow rate
  • Expressed as pressure per unit flow (cm.H2O.s)


FACTORS AFFECTING AIRWAY RESISTANCE


  1. Type of flow
    • Laminar flow produces less airway resistance than transitional or turbulent flow
    • The type of flow depends on Reynolds number (Re)
      • <math display="inline">Re \; = \; \frac {2 \; r \; v \; p} {n}</math> Where r = radius, v=velocity, p=density, n=viscosity
    • Hence
      • Increase in density, velocity or radius = increased Reynolds number = more likely turbulent
      • Increased viscosity (n) = decreased Re = more likely to be laminar flow
    • Laminar flow occurs typically with Re <2000


  1. Vessel (airway) radius

    • Based off Hagen-Poiseuille equation (<math display="inline">Resistance \; = \; \frac {8nl}{\pi r^4}</math>) the smaller the calibre of the airway the increased resistance.

    • Factors which effect vessel radius

      • Lung volume:

        • increased lung volume = increased radius (radial traction pulling open bronchi)

      • Smooth muscle tone

        • Increased tone (e.g. bronchospasm or increased PSNS tone) narrows radius

      • Decreased internal diameter

        • E.g. due to sputum plugging/aspiration > decreased effective radius

      • External compression

        • E.g. tumour, pneumothorax > decreased radius

  2. Length

    • Based off the H-P equation, increased length = increased resistance

    • Not seen in physiological conditions but can be altered for example with artificial ventilation

  3. Dynamic airway compression

    • With forced expiration > increased intrapleural pressure

    • If IP pressure > airway pressure > collapse > decreased radius


Examiner comments

31% of candidates passed this question.

It was expected candidates cover the breadth of the factors that affect airway resistance. Generally, as a concept the type of flow (laminar vs turbulent) was answered well by most candidates, however many failed to mention the other factors that affect airway resistance. Airway diameter as a primary determinant of airway resistance was commonly omitted. Better answers which covered the factors affecting airway diameter classified them broadly and included examples such as physical compression/external obstruction, broncho-motor tone and local cellular mechanisms. Some answers did not explain these factors in enough detail and often with incorrect facts


Online resources for this question


Similar questions

  • Question 18, 2009 (1st sitting)

  • Question 23, 2013 (2nd sitting)

  • Question 6, 2016 (2nd sitting)



Question 13

Question

Describe the factors that affect mixed venous oxygen saturation


Example answer

Mixed venous oxygen saturation (SmvO2)

  • The oxygen saturation of haemoglobin when measured in the pulmonary artery (after venous mixing in the right ventricle)

  • Measured using a pulmonary artery catheter

  • Normally ~75%

  • Provides better idea of whole body venous O2 sats (blood from SVC, IVC and coronary sinus)

Factors affecting SmvO2

  • SmvO2 is a balance between oxygen delivery and oxygen consumption
    • Oxygen delivery (DO2) = cardiac output (CO) x oxygen content of arterial blood (CaO2)
    • CaO2 is dependant on the arterial oxygen saturation, partial pressure of oxygen and the loading ability of Hb (thus the PCO2, temperature, H+ concentration)
  • Cardiac output
    • Increased CO = increased oxygen delivery = increased SmvO2 (vice versa)
  • Hb concentration
    • Increased Hb = increased DO2 = increased SmvO2`
  • Saturation of Hb
    • Decreased arterial Hb saturation > decreased DO2 > decreased SmvO2
  • Loading of Hb with O2
    • Left shift O2-Hb dissociated curve (Decreased H+, decreased PCO2, decreased temp) = increased SmvO2
  • Oxygen consumption
    • Increased oxygen consumption = decreased SmvO2
    • Physiological conditions
      • e.g. exercise = increasing consumption = Decreased SmvO2
    • Pathological conditions
      • e.g. fever/burns = increased consumption = decreased SmvO2
      • e.g. cyanide toxicity, hypothermia = decreased consumption/utilisation = increased SmvO2


Evidence

  • No evidence to support targeting ScvO2 or SmvO2 saturations at present


Examiner comments

49% of candidates passed this question.

Mixed venous oxygen saturation is used as a surrogate marker for the overall balance between oxygen delivery and oxygen consumption. A good answer stated this, described the importance of where it is measured and went on to describe the various factors that affect oxygen delivery and consumption. Descriptions of the factors that affect oxygen saturation of haemoglobin, partial pressure of oxygen in the blood and position of oxygen-haemoglobin dissociation curve were necessary to score well. Important omissions were factors that increased and decreased oxygen consumption. While many candidates were able to correctly write the equations for oxygen content and oxygen flux, they then failed to describe how the variables within these equations were related to mixed venous oxygen saturation.


Online resources for this question

  • CICM Wrecks
  • Deranged Physiology
  • Jenny's Jam Jar


Similar questions

  • Question 10, 2008 (1st sitting)
  • Question 19, 2017 (1st sitting)
  • Question 8, 2019 (1st sitting)



Question 14

Question

Describe the production, action and regulation of thyroid hormones.


Example answer

Overview

  • Thyroid gland produces and secretes two hormones
    • T4 (thyroxine) = 93%
    • T3 (tri-iodothyronine) = 7%


Production and secretion

  • T3/T4 synthesised in thyroid follicles
  • Iodide is taken into thyroid follicles via secondary active transport and oxidised to iodine by thyroperoxidase
  • Thyroglobulin is synthesized in the follicular cell and is secreted into follicular cavity where it combines with iodine to form DIT and MIT, which subsequently couple to form T3 or T4
  • T3/T4 are secreted from the vesicles (thyroglobulin is cleaved off in the process)


Regulation

  • Increased production
    • Increased TSH (from anterior pituitary) > increased T3/T4 production and release (from thyroid)
    • TSH is increased by TRH (produced by paraventricular nucleus in hypothalamus)
    • TRH is stimulated by numerous factors including low T3/4, cold, hypoglycaemia, pregnancy
  • Decreased production
    • Secretions are controlled via negative feedback loop on the hypothalamic-pituitary-thyroid axis
    • Thus increased T3/T4 > decreased TSH (from pituitary) and decreased TRH (from hypothalamus)


Transport / half life

  • Transported in blood bound to albumin, thyroxine binding globulin
  • Both are >99% protein bound
  • T3 has half life 24 hours
  • T4 has half life 7 days


Functions

  • T3/T4 act on thyroid receptors in the cell nucleus > increased gene transcription + protein synthesis
  • T3 is 3-5x more active than T4 (though less abundant)
  • Effects on organ system
    • CVS
      • Increased HR, inotropy, CO
      • Decreased SVR
    • RESP
      • Increased minute ventilation (due to increased CO2 production)
    • CNS
      • Increased: neuroexcitability, tremors
      • Decreased: depression, psychosis
    • MSK
      • Increased osteoblast activity
    • GIT
      • Increased GIT motility
    • METABOLIC
      • Increased basal metabolic rate
      • Increased carbohydrate, fat and protein metabolism



Examiner comments

81% of candidates passed this question.

This question was divided in three sections to help candidates formulate an answer template, which for the most part was answered well. Most answers included a detailed description of the production and regulation of thyroid hormones, including the importance of negative feedback. A brief description of the action of thyroid hormones on intracellular receptors, and a system-based description of physiological effects, including CHO, protein and fat metabolism was expected.


Online resources for this question


Similar questions

  • Question 17, 2016 (1st sitting)


Question 15

Question

Classify and describe the mechanisms of drug interactions with examples.


Example answer

Classification of drug-drug interactions Example
BEHAVIOURAL
- One drug alters behaviour of patient for another - A depressed patient taking an antidepressant may be more compliant with other medications for unrelated conditions
PHARMACEUTIC
- Formulation of one drug is altered by another before administration - Precipitation of thiopentone (basic) and vecuronium (acidic) in a giving set
PHARMACOKINETIC
Absorption Bioavailability of bisphosphonates is reduced when co-administered with calcium as the drugs interact to form insoluble complexes
Distribution Valproate and phenytoin compete for the same transport protein binding sites > decreased protein binding phenytoin > increased effect
Metabolism Macrolides reduce metabolism of warfarin by outcompeting it for similar metabolic pathways (CYP450 enzymes) > increased duration of effect
Elimination Probenecid decreases the active secretion of B-lactams and cephalosporins in renal tubular cells by competing for transport mechanisms > decreased elimination of B-lactams / cephalosporins
PHARMACODYNAMIC
Homodynamic effects Drugs bind to the same receptor site (e.g. naloxone reverses the effects of opioids by outcompeting for the opioid receptor sites)
Allosteric modulation Drugs bind to the same receptor (GABA) but at different sites (e.g. barbiturates and benzodiazepines) > increased effect
Heterodynamic modulation drugs bind to different receptors but affect the same second messenger system (e.g. glucagon reverses the effects of B-blockers by activating cAMP)
Drugs with opposing physiological actions (but different biological mechanisms) e.g. GTN vasodilates via guanyl cyclase-cGMP mediated vasodilation, while noradrenaline vasoconstricts via <math display="inline">\alpha</math> agonism


Examiner comments

54% of candidates passed this question.

This question has been asked previously, the answer template expected some description rather than a list of drug interactions. Generally, examples were provided for each type of interaction. The examiners reported too many vague, factually incorrect descriptions of the mechanisms and in some cases a very limited classification.


Online resources for this question


Similar questions

  • Question 3, 2017 (1st sitting)
  • Question 9, 2015 (1st sitting)



Question 16

Question

Classify the anti-psychotic drugs (25% marks). Outline the pharmacology of haloperidol (75% marks).


Example answer

Classification of antipsychotics

  • First generation (typical) antipsychotics
    • Higher affinity for D2 receptors
    • Leads to better effect on 'positive' symptoms (hallucinations, delusions, hyperactivity)
    • Greater incidence of EPSE and less metabolic side effects
    • Examples: haloperidol (Butyrophenones), chlorpromazine (Phenothiazines)
  • Second generation (atypical) antipsychotics
    • Block D2 as well as 5HT2A
    • Greater effect on negative symptoms (apathy, lethargy etc)
    • Fewer EPSE, but increased metabolic side effects (weight gain, diabetes, hyperChol etc)
    • Examples: olanzapine, quetiapine, clozapine


Haloperidol

Name Haloperidol
Class Antipsychotic (1st generation)
Indications Behavioural emergencies, psychosis, intractable nausea/vomiting
Pharmaceutics PO tablets Clear solution for injection
Routes of administration PO, IM, IV
Dose 1-5mg IV, 2-30mg IM, 1-10mg PO
Pharmacodynamics
MOA Antipsychotic actions thought to be mediated by blockade of dopamine (D2 > D1) receptors particularly in the limbic system. Also demonstrate weak antagonism of H1, mACh receptors
Effects CNS: apathy, decreased agitation, sedation

CVS: QT prolongation / TdP
GIT: anti-emetic
MET: weight gain, diabetes, hyperChol
Other: neuroepileptic malignant syndrome, extrapyramidal side effects (dystonia, akathisia, parkinsonism, TD)
HAEM: leukopaenia
RESP: respiratory depression in large enough doses

Pharmacokinetics
Onset Peak effects after 3 hours (PO)
Absorption 80% PO bioavailability
Distribution >90% protein bound
VOD = 20L/kg
Metabolism Hepatic > inactive metabolites
Elimination Renal (major) and faecal (minor) excretion of metabolites

T 1/2 = ~24 hours (longer in IM, shorter in PO)

Special points


Examiner comments

28% of candidates passed this question.

Excellent answers were able to provide a classification of antipsychotics based on either typical/atypical or first/second generation categories, provide examples of each and identify key differences in mechanism and effects. They also distinguished between butyrophenones and phenothiazines within the typical antipsychotic group. Haloperidol was identified as a butyrophenone, with description of pharmaceutics, dose and route, as well as pharmacodynamics and pharmacokinetics. Key adverse effects were detailed, focusing on those specific to haloperidol, including a description of different types of extrapyramidal symptoms and QT prolongation/ torsades de pointes.


Online resources for this question


Similar questions

  • Question 21, 2014 (2nd sitting)



Question 17

Question

Explain the components of an ECG (electrocardiogram) monitor (70% marks). Outline the methods employed to reduce artefact (30% marks).


Example answer

The ECG

  • Myocyte action potentials sum to produce a voltage which can be measured as a potential difference between two electrodes on the skin
  • ECG is therefore a graphical recording of the electrical events of the cardiac cycle
    • P wave = atrial depolarisation
    • P-R interval = av nodal conduction
    • QRS = ventricular depolarisation
    • T wave = ventricular repolarisation
  • Useful in diagnosis of a range of cardiac conditions including arrhythmias, infarction, hypertrophy etc.


Components of ECG

  1. Electrodes
    • Sicky + conducting gel to ensure adequate skin contact
    • Silver chloride electrode to detect electrical potential differences
    • Earth lead - reduces interferance
    • Bipolar leads: 1, II and II. Unipolar: aVR, aVL, aVF, Praecordial V1-6
  2. Physical leads/cables (insulated)
    • Transmit the electrical signal
    • Insulation reduces interference / risk of harm from electrocution
  3. Processor/Amplifier
    • Processes augmented leads (creates 6 ecg leads from 3 physical limb leads)
    • Amplifies the low signal (~1mV) through differential amplification
    • Filters out noise/artefact
      • High input impedance - filters out EMG signal and mains interference
      • Low pass filtering - eliminates movement artefact
  4. Monitor/output device
    • Displays/prints/records the trace


Artefact and error

  • Machine
    • Incorrect filtering settings
      • ECG monitoring mode: Strong filter setting to focus on rhythm, reduces artefact
      • Diagnostic mode: Lower filtering setting to allow for subtle changes in ST segments (greater resolution at expense of noise)
  • Cabling/Circuit
    • Incorrect lead placement --> errors with augmented leads, and interpretation --> correct
    • Interference with electronics (e.g. ventilators, dialysis machines) --> limit exposure
    • Damaged/broken cables --> replaced
  • Patient
    • Excessive movement or motion artefact (movement, shivering, seizure)
      • Rewarm, low pass filtering, place over bony prominences
    • Poor contact due to things such as hair, lotions, etc.
      • Cleaned with alcohol wipe / shaved to improve contact



Examiner comments

46% of candidates passed this question.

Excellent answers described the function of the ECG device and its components. Components include electrodes, which form leads (unipolar and bipolar), the amplifier and an output device. The process of amplification and filtering (e.g., high and low pass filters), as well as monitoring and diagnostic ECG modes were described. A comprehensive list of ways to reduce artefacts, including strategies to address both patient and equipment factors was generally provided.


Online resources for this question


Similar questions

  • Question 5, 2011 (2nd sitting)
  • Question 9, 2016 (1st sitting)



Question 18

Question

Outline the neural pathways for the pupillary light, corneal, oculomotor and gag reflexes. The anatomical course of nerves is NOT required.


Example answer

Pupillary light reflex

  • Receptors: photoreceptors in retina
  • Afferent: cranial nerve II
  • Integrator/controller: pretectal nucleus in midbrain > Edinger-Westphal nuclei
  • Efferent: Cranial nerve III (preganglionic) > ciliary ganglion
  • Effector: iris via short ciliary nerves (postganglionic)
  • Effect: direct + consensual pupillary constriction to light


Corneal reflex

  • Receptors: free nerve endings / stretch receptors in epithelium of cornea
  • Afferent: Ophthalmic division of cranial nerve V
  • Integrator/controller: trigeminal nucleus > facial nucleus
  • Efferent: cranial nerve VII
  • Effector: orbicularis oculi muscle
  • Effect: ipsilateral eyelid movement (early response) followed by bilateral blink (late response)


Oculomotor reflex (Vestibulo–ocular reflex)

  • Sensation: head rotation (angular acceleration)
  • Sensor: semi-circular canals and otoliths in inner ear
  • Afferent: cranial nerve VIII
  • Integrator: vestibular nuclear complex (medulla and pons)
  • Efferents: cranial nerves III, IV and VI
  • Effect: activation of recti muscles (depending on rotation) to maintain visual focus


Gag reflex

  • Stimulus: sensation to posterior pharyngeal wall
  • Afferent: cranial nerve IX
  • Integrator: NTS > nucleus ambiguus
  • Efferent: CN X
  • Effects: Contraction of pharyngeal muscles



Examiner comments

43% of candidates passed this question.

This is a fact-based question with little integration of knowledge required. Those candidates who synthesised their knowledge into a succinct and precise description of afferent and efferent pathways with a description of the various sensor and integrator components scored very high marks. A good working knowledge of all the cranial nerve reflex pathways are crucial to the practise of intensive care medicine. Marks were not awarded for any anatomical description related to these pathways.


Online resources for this question

  • CICM Wrecks
  • Deranged Physiology
  • Jenny's Jam Jar


Similar questions

  • ?Nil



Question 19

Question

Outline the process of fibrinolysis (40% marks). Write short notes on the indications, mechanism of action, pharmacokinetics and side effects of tranexamic acid (60% marks).


Example answer

Fibrinolysis

  • Process by which fibrin (within blood clots) is broken down by plasmin into fibrin degradation products
  • Normal physiological process as part of wound healing and is important for keeping vessels patent
  • Steps
    • Plasminogen (b-globulin) is produced in the liver
    • Plasminogen is trapped in fibrin meshwork during initial clot formation
    • Plasminogen can be converted by plasminogen activators (serum protease) into plasmin
    • Plasmin subsequently cleaves fibrin > fibrin degradation products
  • Activation
    • Intrinsic: Main physiological activator is tissue plasminogen activator which is released from injured endothelial cells (but is a slower process than coagulation to allow healing to take place)
    • Extrinsic: urokinase, streptokinase, recombinant tPA can increase activation of plasminogen > plasmin > increased fibrinolysis


Name Tranexamic acid
Class Antifibrinolytic
Indications Trauma (within 3 hours), cardiac/obstetric/orthopaedic surgery, haemorrhage
Pharmaceutics 500mg Tablets (PO)
Clear colourless solution (100mg/ml) for injection (IV)
Routes of administration PO, IV, IM
Dose 0.5 - 1 g (slow IV push), followed up by infusion of 1g over 8 hrs if needed
pKA
Pharmacodynamics
MOA Competitive inhibition of the activation of plasminogen into plasmin by binding to lysine binding sites on plasminogen
Effects Decreased fibrinolysis > decreased bleeding
Side effects HAEM: Prothrombotic complications in those patients with risk factors

GIT: nausea, vomiting CNS: seizures and dizziness (dose related)

Pharmacokinetics
Onset Immediate (IV), 1 hour (IM), 2 hours (PO)
Absorption PO bioavailability = 50%, IM bioavailability 100%
Distribution Protein binding: very low (<5%)

VOD = 0.3L / kg

Metabolism Minimal hepatic metabolism
Elimination Renal elimination of active drug (95% unchanged)
T 1/2 = 2hrs (IV), 10 hrs (PO)
Special points Dose reduce in renal failure


Examiner comments

30% of candidates passed this question.

The relative allocation of marks and thus time to be spent on each component was delineated by the relative percentages in the question. The first part of the question required a step-by-step outline of the fibrinolytic pathway with mention of the regulatory processes. Tranexamic acid is an important drug in the practice of intensive care and the question provided the headings under which to answer the question. The detail surrounding the keys aspects of this drug with respect to its use in critical care were often vague and underappreciated.


Online resources for this question


Similar questions

  • Question 4, 2013 (1st sitting)
  • Question 19, 2015 (2nd sitting)



Question 20

Question

Describe the physical principles of haemodialysis and haemofiltration, including the factors affecting clearance (80% marks). Outline the key components of renal replacement fluids (20% marks).


Example answer

Dialysis

  • Separation of particles in a liquid, based on their differential ability to pass through a membrane
  • Main mechanisms: haemodialysis, hemofiltration, combination of above
  • Main indications: acidosis, electrolyte derangement, intoxication, fluid overload, ureamia,


Haemodialysis

  • Utilises principle of diffusion
    • Spontaneous movement of a substance from area of high > low concentration
    • Movement is dependant on Fick's law (thus temp, size, concentration, distance etc)
  • Process
    • Blood is pumped through an extracorporeal dialysis circuit
    • Dialysate flows in a counter-current direction (maintains concentration gradient)
    • Blood is separated from dialysate via semipermeable membrane (does not mix)
    • Movement of molecules then diffuses according to Ficks law.
  • Useful for clearance of small molecules, cannot clear larger molecules


Haemofiltration

  • Uses principle of convection and solvent drag
    • Elimination of materials is via bulk flow and independent of concentration
    • Clearance is dependant on starling forces
  • Process
    • Blood is pumped through extracorporeal dialysis circuit
    • A transmembrane pressure is applied to the blood side of a semi-permeable membrane
    • Plasma is filtered across membrane and solutes (via drag) are eliminated as effluent.
    • Renal replacement fluid is added to patient blood to restore volume, buffers, and normal haematocrit
  • Can clear small-medium sized molecules


Factors effecting clearance

  • Drug factors

    • Protein binding

      • Small molecules bound to large proteins (e.g. aspirin bound to albumin) cannot be cleared

    • Size/molecular weight

      • Smaller molecules are more readily dialysed

    • Volume of distribution

      • Drugs with large Vd (e.g. barbituates) cannot readily be cleared as they rapidly redistribute

  • Dialysis factors

    • Haemodialysis

      • Blood / dialysate flow rate

      • Dialysate composition

    • Haemofiltration

      • Blood / effluent flow rate

      • Transmembrane pressure

      • Prefilter dilution

      • Sieving coefficient

Renal replacement fluids

  • 5000ml bag, warmed to body temperature
  • Contains
    • Electrolytes
      • Na = isotonic
      • K, Mg, Phos, Ca = variable
    • Buffers
      • Bicarbonate (predominately)
      • Can also use lactate, citrate
    • Nutrients (i.e. gluc)
    • Sterile water
  • Osmolarity ~285
  • Dose varied depending on degree of fluid removal desired


Examiner comments

28% of candidates passed this question.

A brief description of the underlying mechanisms of dialysis and hemofiltration was required. Diffusion, the predominant mechanism in haemodialysis, involves movement of solute down the concentration gradient across the semipermeable membrane. This concentration gradient is generated and maintained by counter current movement of dialysate and blood. In hemofiltration the predominant mechanism is convection and solvent drag of the solute across the semipermeable membrane by application of transmembrane pressure. The filtrate is then replaced by replacement fluid. Small molecules are effectively removed by dialysis whereas hemofiltration can remove small and middle molecules. Various factors that impact clearance in haemodialysis and haemofiltration were expected separately. Constituents of replacement fluid should have included three broad headings of electrolytes, buffer and sterile water. Many answers lacked the details of how counter current mechanisms help, the difference in the two modalities in regard to clearance of molecules, how clearance is impacted by protein binding and volume distribution, sieving coefficient of the membrane and flow rates of blood and dialysate (or effluent) flow. The constituents of replacement fluid lacked details of various types of electrolytes, the common buffers and the strong ion difference.


Online resources for this question


Similar questions

  • Question 24, 2011 (1st sitting)



2021 (1st sitting)

Question 1

Question

Describe the pharmacology of adrenaline.


Example answer

Name Adrenaline
Class Naturally occurring catecholamine
Indications Haemodynamic support, anaphylaxis, bronchoconstriction/airway obstruction
Pharmaceutics Clear solution, light sensitive (brown glass), 1:1000 or 1:10,000
Routes of administration IV, IM, INH, ETT, Topical, subcut
Pharmacodynamics
MOA Non-selective adrenergic receptor agonist.

At low doses B effects dominate, at high doses alpha dominate.
Adrenaline > a-1 receptor > increased IP3 (2nd messenger) > increased Ca
Adrenaline > B1,B2,B3 receptors > increased cAMP (second messenger)

Effects CVS: vasoconstriction (high doses), vasodilation (low doses), increased inotropy + chronotropy

RESP: bronchodilation, increased minute ventilation
METABOLIC: hyperglycaemia (glycogenolysis, lipolysis, gluconeogenesis)
CNS: increased MAC
GIT: decreased intestinal tone/secretions

Side effects Extravasation > tissue necrosis, pHTN due to increased PVR, hyperglycaemia, tachyarrhythmias,
Pharmacokinetics
Absorption Zero oral bioavailability due to GIT inactivation. variable/erratic ETT absorption.
Distribution Poor lipid solubility, doesn't cross BBB, crosses placenta
Metabolism Metabolised by MAO (mitochondria) and COMT (liver, blood, kidney) to VMA and metadrenaline
Elimination T 1/2: ~2 mins (due to rapid metabolism)

Metabolites (above) are excreted in the urine


Examiner comments

90% of candidates passed this question.

Adrenaline is a level 1 drug and is commonly used in intensive care. A comprehensive explanation of the drugs MOA, PK, PD and side effect were expected. Candidates who scored well generally provided a factually accurate, detailed and well-structured answer. Overall, the quality of answer provided for this question was of a high standard.


Online resources for this question


Similar questions

  • Question 2, 2018 (Paper 1)
  • Question 15, 2017 (Paper 2)
  • Question 18, 2012 (Paper 2)
  • Question 8, 2012 (Paper 1)





Question 2

Question

Describe the work of breathing and its components.


Example answer

Work of breathing

  • Energy used by the respiratory muscles during respiration
  • Work of breathing (joules) = pressure x volume
  • Normally ~0.35 J/L (approx 2% BMR)
  • Can be represented using pressure-volume curves


Components

  • Elastic work (~70%)

    • Force required to overcome the elastic forces of the chest wall, lung parenchyma, and alveoli surface tension

    • Elastic resistance increases with increasing tidal volumes

    • Energy is stored as elastic potential energy and used on expiration

    • Factors increasing elastic work:

      e.g. obesity, chest wall deformities, circumfrential burns etc.

      e.g. loss of surfactant in ARDS

  • Non elastic (resistive) work (~30%)

    • Derived from airway resistance (majority; 80% of non elastic work) and viscous tissue resistance (e.g. lung sliding over chest wall)

    • Airway resistance increases with increased RR (frequency dependence of work of breathing) or smaller airway diameter (e.g. bronchospasm)

    • Energy is lost as heat


Work of breathing during a tidal volume breath (ref to fig below)

img
img




Examiner comments

24% of candidates passed this question.

This is a core topic within respiratory physiology. There was a very low pass rate for this question. Expected components of the answer included: a definition of WOB as a product of pressure and volume or force and distance including the units of measurement; followed by a detailed explanation of the following three broad components – elastic resistance, viscous resistance and airflow resistance. Further marks were awarded to situations where the energy for expiration increases beyond stored potential energy as well as the impact of respiratory rate and tidal volume on different aspects of the WOB. For example, the changes in TV will have relatively greater impact on the elastic component, whereas RR will impact the resistance component. Additional marks were awarded for describing the efficiency of breathing. A common area where candidates missed out on marks was producing a diagram of WOB without a description; many diagrams were often incorrectly drawn or had no axes labelled. There were many incorrect definitions or respiratory equations provided without any link to the written answer. Factual inaccuracy and limited depth of knowledge were also prevalent in poorly performing answers. Marks were not awarded for a description of the control of breathing.


Online resources for this question


Similar questions

  • Nil



Question 3

Question

Outline the formation, structure, and function of the platelet.


Example answer

Formation/fate

  • Produced via thrombopoiesis
    • Within the bone marrow, common myeloid progenitor cells differentiate into megakaryocytes
    • Megakaryocytes are the largest cell of the bone marrow (50-150um), have multiple (up to 8) nuclei. They produce pro-platelets in their cytoplasm which break up into numerous smaller functional platelets. Each megakaryocyte produces ~5000 platelets each
    • Thrombopoiesis takes ~10 days
    • Thrombopoietin (produced primarily in the liver) stimulates the differentiation and release of plts
    • Process regulated by negative feedback loop based on platelet count
  • Normally 150-400 x 10^9/L
  • Lifespan is 7-10 days
  • Utilised during clotting or removed by the reticuloendothelial system in the spleen or liver


Structure

  • Not true cells, but fragments of the Megakaryocyte cytoplasm
  • 1-4 um in size, Irregular, No nucleus.
  • External glycocalyx later
  • Contain:
    • Mitochondria
    • Dense granules (ATP, ADP, calcium)
    • Alpha granules (vWF, thrombin)
    • Contractile elements (microtubules)


Function

  • Main function of platelets is haemostasis
  • Platelets are important for formation of the platelet plug (primary haemostasis)
    • Platelet adhesion
      • Damage to blood vessel wall exposes vWF in the subendothelium
      • Glycoprotein receptor complex (GP1b-IX) on platelets bind to vWF (adhesion)
    • Platelet activation
      • When exposed to Tissue Factor, collagen, and vWF the platelets become activated
      • Activated platelets
        • Change shape (Swell, become irregular, develop pseudopods) to enable aggregation
        • Release molecules (Thromboxane A2, ADP, Serotonin) which activates further platelets + vasoconstricts
      • Platelet aggregation
        • Activated platelets bind to fibrinogen and vWF to form a soft platelet plug
  • Platelets are also central to the cell based model of secondary haemostasis
    • Initiation (less relevant to platelets)
    • Amplification
      • Surface of activated platelets is primed with factors V, VIII, XI
      • Small amount of thrombin produced during initiation activates V, VIII and XI
      • XIa activates IX to IXa, which leads to formation of tenase complexes (accelerate thrombin production at platelet surface)
    • Propagation
      • Begins with formation of tenase complexes on platelet surfaces (IXa-VIIIa)
      • Leads to increased rate of Factor X activation
      • The large amounts of Xa interacts with factor Va forming prothrombinase complex (Va-Xa)
      • Va-Xa catalyses the conversion of prothrombin to thrombin



Examiner comments

79% of candidates passed this question.

This question was divided in three sections to help candidates formulate an answer template. The first section required a brief outline of the formation of platelets from pluripotent stem cells via megakaryocytes. The second section required an outline of platelet structure highlighting the special features such as, an absence of a nucleus, the presence of an external glycocalyx layer, specific surface receptors, contractile proteins, dense tubular system and granules. The third section was about platelet function where the expected focus was on the role of platelets in haemostasis. This required outlining the mechanism of platelet plug formation by adhesion-activation-aggregation, interactions with the coagulation cascade and role of platelets in clot contraction as well as fibroblast invasion. Although many candidates were able to answer the first section reasonably well, there was a noticeable knowledge deficit in the latter two sections. A significant proportion of answers had missing information on platelet structure and lack of structure in outlining platelet function


Online resources for this question


Similar questions

  • Question 6, 2016 (1st sitting)
  • Question 20, 2011 (2nd sitting)


Question 4

Question

Outline the dose (10% marks), composition (60% marks) and side effects (30% marks) of total parenteral nutrition (TPN).


Example answer

Overview

  • TPN is the delivery of nutrients into the venous circulation to replace enteral requirements

Indications

  • Patients who are unable to be fed via enteral route for prolonged periods of time (e.g. >72 hours)
  • May include patients who fail trial of enteral feeding or other contraindications (e.g. GI obstruction, severe pancreatitis, short gut syndromes)


Daily nutritional requirements (major)

Nutrient Requirement (kg/day)
H2O 30mls
Energy 25-30 kcal
Protein 1g (higher in critically ill, 1.5g)
Glucose 2g
Lipids 1g
Na 1-2 mmol
K 1mmol
Ca / Mg 0.1mmol
PO4 0.4 mmol

Note

  • Requirements vary according to physiological (e.g. age, gender, body size, activity levels) and pathological (e.g. burns, sepsis, renal failure, hepatic failure) factors

Composition of TPN (note many formulations of this)

  • Glucose

    • Typically supplies around 60-70% of daily caloric needs (~1400KCal)

    • Typically 50% dextrose used (3.4 kCal/mil - 824mls)

  • Lipid

    • Typically supplies around 30-40% daily caloric needs (~600Kcal)

    • Can be olive oil, soybean, fish oil based

  • Protein (Amino acids)

    • Will contribute to energy source + provides essential amino acids

    • L-amino acids used only

    • Typically 1.5g/kg/day in critically ill (~100g protein / day)

  • Electrolytes (Na, K, Mg, Ca, Cl, PO4)

    • TPN solutions can come with/without electrolytes and adjusted

  • Vitamins, trace elements

    • Micronutrients are added in appropriate amounts to the bag for adequate daily intake

    • Thiamine, folic acid and vitK are vulnerable to depletion and additional may be needed

  • Water

    • In solution, though insufficient for daily requirements

    • Hyperosmolar solution due to above nutrients

Side effects / complications

  • Delivery device / vascular access related
    • Infection, pneumothorax, thrombosis, air embolism
  • Fluid/elextrolyte disturbances
    • Fluid overload
    • Electrolyte derrangements, shifts and refeeding syndrome
    • Acid base disturbances
  • Metabolic disturbances
    • Hypoglycaemia, hyperglycaemia (dose related issues due to over/under feeding)
    • Hyperlipidaemia


Examiner comments

59% of candidates passed this question.

The pharmacology of enteral and parenteral nutrition is a level 1 topic in the first part syllabus. The TPN dose in terms of daily calorie and other nutritional requirements were key expectations in first part of the question. A detailed list of all macro and micronutrients was required under TPN composition. Expected information about macronutrients were their forms in the TPN solution (e.g., carbohydrate in the form of glucose, protein in the form amino acids), their relative calorie contributions and their essential components (e.g., the names of the essential amino acids). Identification of potential variability in composition and dose based on specific patient factors scored extra marks. Side effects included metabolic derangements (refeeding syndrome, over or under-feeding, hyperglycaemia, hyperlipemia), biochemical disturbances (fluid and electrolyte imbalances), organ injury (liver, pancreas) and vascular access related complications. Limited breadth and depth of information as well as incorrect facts were prevalent in the answers that scored lower marks.


Online resources for this question

Similar questions

  • 2015 Fellowship paper


Question 5

Question

Outline the factors that determine central venous pressure (60% marks) and explain how it is measured (40% marks).


Example answer

Overview

  • CVP is the venous blood pressure (of the great veins) measured at or near the right atrium
  • Normally 0-6mmHg in spontaneously breathing non ventilated patient


Measurement

  • Non invasive
    • Echocardiography can provide non invasive estimations off the CVP
    • Visual inspection of the height of the JVP can provide some bedside clinical insight
  • Invasive
    • Most commonly measured using central line
    • Central line tip sits at or near the right atrium
    • CVC is connected to a pressure transducer via incompressible tubing with flush solution
    • Transducer is zeroed to the atmospheric pressure and levelled at the height of the right atrium


Factors determining CVP

  • <math display="inline"> \Delta CVP \; = \; \frac {\Delta Volume}{\Delta Compliance}</math>
  • Thus factors determining CVP related to those that influence compliance and volume
  • Central venous blood volume
    • Increased total blood volume (e.g. renal failure) = increased CVP
    • Decreased CO (e.g. LV failure) > blood backs up > increased thoracic blood volume > increased CVP
  • Central venous vascular compliance
    • Increased vascular tone central veins (e.g. noradrenaline) > decreased compliance > increased CVP
    • Decreased myocardial/pericardial compliance > increased CVP
    • Decreased pulmonary arterial compliance (e.g. PAH) > increased CVP
  • Tricuspid valve function
    • TV regurg > increased CVP (retrograde transmission of RV systolic pressure)
    • TV stenosis > increased CVP (increased resistance to RV inflow)
  • Intrathoracic pressure
    • ITP is transmitted to the central venous compartment
    • Thus, increased PEEP, IPPV, or a tension pneumothorax will lead to increased CVP
  • Measurement technique
    • Level of the transducer will clearly influence the CVP measured


Examiner comments

57% of candidates passed this question.

This question examined a core area of cardiac physiology and measurement. Considering this, candidates overall, scored poorly in this section. There was a common misunderstanding around the relationship between cardiac output and CVP. A decrease in cardiac output (e.g. due to either decreased stroke volume or heart rate) will cause an increase in CVP as blood backs up in the venous circulation, increasing venous volume as less blood moves through to the arterial circulation; the resultant increase in thoracic volume increases central venous pressure. Several candidates confused the direction of their arrows, for example "increased right atrial compliance increases CVP". Double negatives were used by several candidates which then resulted in the incorrect relationship described. (e.g., "arrow down compliance and arrow down CVP"). The measurement section should have included an explanation of the components of an invasive pressure monitoring system relevant to the measurement of CVP.


Online resources for this question


Similar questions

  • Question 6, 2019 (2nd sitting)



Question 6

Question

Describe the pharmacology of vecuronium, including factors that prolong its action of neuromuscular blockade.


Example answer

Name Vecuronium
Class Aminosteroid
Indications Muscle relaxant; intubation, control of ICP, assist ventilation,
Pharmaceutics Potentially unstable in solution

Comes in powder (10mg), dissolved in water (5ml) for use

Routes of administration IV
Dose Intubation: 0.1mg/kg
ED95
Pharmacodynamics
MOA Non depolarising muscle relaxant;

Competitive antagonism of ACh at N2 receptors on PSM of NMJ

Effects MSK: NMJ blockage (paralysis)

CVS: nil
RESP: Apnoea

Side effects MSK: prolonged use can lead to myopathy

Rare for histamine release (anaphylaxis, hypotension)

Pharmacokinetics
Onset/duration 90-120s; 30-45 minutes
Absorption IV only
Distribution Doesn't cross BBB; VD 0.23L/kg
Metabolism 20% hepatic de-acetylation
Elimination 70% biliary, 30% urinary
Reversal Can be reversed with sugammadex


Factors prolonging action

  • Electrolyte disturbances
    • Hypokalaemia, Hypermagnesemia and hypocalcaemia potentiates non depolarising NMBA
  • Acidosis
    • Increased affinity for ACh receptor
  • Hypothermia
    • Reduced metabolism of muscle relaxant > prolonged effects
  • Hepatic/renal disease
    • Prolonged metabolilsm/elimination of active metabolites
  • Drug interactions
    • e.g. lithium, diuretics, volatile anaesthetics, aminoglcosides


Examiner comments

13% of candidates passed this question.

Vecuronium is a commonly available and regularly used amino-steroid neuromuscular blocking agent. It is a level 1 drug in the 2017 syllabus. A simple template utilising the headings; pharmaceutics, PK, PD, uses in ICU and adverse reactions with associated relevant important facts would have scored well. Expected information regarding the factors prolonging neuromuscular blockade included electrolyte abnormalities, drug interactions and patient factors. Overall, the level of understanding and knowledge demonstrated in the answers was below an expected standard for a level 1 drug.


Online resources for this question


Similar questions

  • Question 1, 2008 (2nd sitting)
  • Question 3, 2009 (2nd sitting)
  • Question 7, 2015 (2nd sitting)
  • Note: there has essentially been a question every year in relation to the pharmacology of one of the NMB drugs (sux, vec, roc) in one form of another.



Question 7

Question

Outline the anatomy of the blood supply (arteries and veins) of the gastrointestinal system (oesophagus to anus)


Example answer

Arterial supply

  • The aorta (and its branches) supplies the entire arterial supply to the GIT
  • The oesophagus is supplied by various arterial branches
    • Cervical portion- inferior thyroid artery
    • Thoracic portion - bronchial arteries
    • Abdominal portion - left gastric, inferior phrenic arteries
  • The abdominal aorta then has three main branches which supply the remainder of the GIT
    • Celiac trunk
      • Arises from abdominal aorta immediately below aortic hiatus at T12/L1
      • Divides into left gastric artery, splenic artery, common hepatic artery
        • Left gastric a. (supplies stomach)
        • Splenic a. (supplies spleen, pancreas)
        • Common hepatic, divides into
          • Hepatic a. proper (supplies liver)
          • Gastroduodenal (supplies pancreas, duodenum, stomach)
          • Right gastric (supplies stomach)
    • Superior mesenteric artery (SMA)
      • Arises from abdominal aorta immediately inferior to coeliac trunk (L1)
      • Multiple branches (15-20) which join in an arcade
      • Supplies the midgut structures (from duodenum to 2/3 transverse colon)
    • Inferior mesenteric artery (IMA)
      • Arises from abdominal aorta ~L3
      • Multiple branches (including Left colic, sigmoid, superior rectal arteries), join in arcade
      • Supplies the hindgut (distal 1/3 transverse colon - rectum)


Venous drainage

  • For the most part, the venous drainage of the GIT is via veins which accompany the arterial system

  • They return via the portal vein

    • Portal vein

      • Combination SMV and splenic vein

      • Receives drainage from forgut structures

    • Splenic vein

      • Travels along with the splenic artery + drains corresponding regions (foregut)

      • Combines with SMV to form portal vein

    • Superior mesenteric vein (SMV)

      • Travels along with the SMA + drains corresponding regions (midgut)

      • Combines with splenic vein to form portal vein

    • Inferior mesenteric vein (IMV)

      • Travels along with the IMA + drains corresponding regions (hindgut)

      • Drains into the splenic vein

Examiner comments

48% of candidates passed this question.

This question was answered best if the main arteries and veins were discussed first and then their corresponding supply outline in reasonable detail. Very few candidates were able to achieve this. Listing the names of vessels with no context and in a random non-sequential order did not attract many marks. The physiology of the blood supply to the liver also did not attract marks.


Online resources for this question


Similar questions

  • Question 7, 2018 (1st sitting)



Question 8

Question

Describe renal handling of potassium (60% marks), including factors that may influence it (40% marks).


Example answer

Renal handling of potassium

  • Potassium is freely filtered at the glomerulus
    • Serum K = 4.2mmols/L, with 180L filtered / day (assuming GFR 125mls/min)
    • Thus most of filtered K needs to be reabsorbed in the kidney
  • Proximal convoluted tubule (PCT)
    • ~60% of K reabsorbed
    • Reabsorbed passively by solute drag (coupled to water reabsorption) + concentration gradient
    • Water absorption is driven by the Na/K ATPAse on basolateral membrane > drives Na reabsorption
  • Loop of henle (LOH)
    • 30% reabsorbed in thick ascending LOH
    • NK2Cl cotransporter drives transcellular reabsorption (via basolateral K channels) + paracellular reabsorption (due to negative charge generated by Cl reabsorption)
    • Active/secondary active transport
  • DCT + CD
    • 0-10% reabsorbed
    • Secretion and absorption
      • Principle cells in DCT and CD: secrete potassium
      • Type A intercalated cells: reabsorb potassium
    • Net effect depends on the K state at the time
      • With normal intake or excessive intake, net effect is secretion
      • With low intake the net effect is reabsorption


Regulation / factors influencing renal handling K

  • Aldosterone
    • Increases Na-K ATPase activity primarily in the principle cells > increasing secretion of K
  • Vasopressin
    • Increases ROMK channels, increasing K secretion (balanced by decreased urinary flow rate)
  • Acid base disturbances
    • Metabolic alkalosis
      • Potassium secretion increases (increased Na/K ATPase activity due to low H+)
    • Metabolic acidosis
      • Potassium secretion decreases (opposite of above)
  • K intake
    • Increased intake > Increases ROMK channels > increasing K secretion


Examiner comments

33% of candidates passed this question.

This question covers a core physiology topic. The detail required is well described in the recommended reference texts. Generally, this question was poorly answered. From an answer template perspective, a "describe question" in this context involves both the stating the relevant potassium handling mechanism and then giving a description of how it occurs and how this system is regulated. Many answers that scored poorly simply listed sites of potassium handling but excluded the details surrounding the specific receptors and channels involved as well as the processes that exist to perpetuate and regulate these biological processes. Simple identification as to whether the potassium was being secreted or reabsorbed as well as the location as to where this may occur within the nephron, were often not specifically detailed or used interchangeably. Such answers scored poorly


Online resources for this question


Similar questions

  • Question 16, 2010 (2nd sitting)
  • Question 12, 2013 (1st sitting)



Question 9

Question

Outline the mechanisms by which normal body temperature is maintained and regulated


Example answer

Overview

  • Human 'core temperature' is the 'deep body' temperature of the internal organs and viscera
    • Core temperature ~ 37°C <math display="inline">\pm</math> 0.4°C, despite changes in ambient temperature
    • Rectal, bladder, oesophageal, central vascular temperatures are often used as approximations.
  • Peripheral temperatures are variable and generally less than the core temperature
  • Significant hypothermia (e.g. <35°C) or hyperthermia (e.g. >40°C) can lead to multi-organ dysfunction
  • Humans have multiple thermoregulatory mechanisms to resist change in core body temperature
    • In general, heat is lost by 4 mechanism: conduction, convection, evaporation, radiation
    • In general, heat is gained by 5 mechanisms: conduction, convection, evaporation, radiation, metabolism



Thermoregulatory system & regulation

  • Sensor
    • Peripheral:
      • Skin thermoreceptors (cold= bulbs of Krause; warm=bulbs of Ruffini)
      • Travels via spinothalamic tract to hypothalamus
    • Central:
      • Thermoreceptors (hypothalamus and spinal cord)
  • Integrator/controller
    • Hypothalamus
      • Functions as the thermostat (temperature kept around a thermoneutral zone, TNZ)
      • Stimulation of anterior hypothalamus leads to heat loss (temp above TNZ)
      • Stimulation of the posterior hypothalamus leads to heat conservation/generation (below TNZ)
  • Effector/Response
    • Response to cold
      • Shivering -> involuntary muscle contractions that generate heat (ATP hydrolysis)
      • Peripheral vasoconstriction (ANS) --> decreased cutaneous blood flow --> decreased heat transfer from ambient air
      • Increase metabolic rate, thyroid hormone secretion, Non shivering thermogenesis (brown fat paeds, skeletal muscle adults) -> increased heat generation
      • Behavioural changes (seek warmth)
      • Piloerection (unimportant in humans)
    • Response to heat
      • Peripheral vasodilation (ANS) --> increased cutaneous blood flow > increased heat loss
      • Sweating --> evaporative heat loss
      • Behavioural changes (seek cool)


Examiner comments

59% of candidates passed this question. This question was relatively well answered by most candidates. There was significant variation in the temperatures expressed as normal and few candidates mentioned CORE temperature as a concept. Several candidates gave a detailed description of thermo-neutrality for which there were no marks.


Online resources for this question

Similar questions

  • Question 6, 2007 (1st sitting)
  • Question 4, 2009 (1st sitting)
  • Question 19, 2018 (1st sitting)



Question 10

Question

How does warfarin exert its pharmacological effect (40% marks)? Write brief notes on the pharmacology of the agents that can be used to reverse the effects of warfarin (60% marks).


Example answer

Name Warfarin
Class Oral anticoagulant
Indications Systemic anticoagulation, e.g. in prophylaxis/treatment of thromboembolism
Pharmaceutics Racemic mixture of two enantiomers R and S, with the S isomer more biologically active.
Routes of administration Oral
Dose Varies; titrated to INR generally
Pharmacodynamics
MOA Inhibits the synthesis of vitamin K dependant clotting factors (II, VII, IX, X).

Specifically, inhibits vitamin K epoxide reductase (VKORC1) from converting VitK from the oxidised to reduced form, which prevents carboxylation (activation) of clotting factors listed above (as well as protein C and S)

Effects Anticoagulation
Side effects Haemorrhage, teratogenicity (1st trimester), foetal haemorrhage (3rd trimester), drug interactions
Pharmacokinetics
Onset Peak onset is 72 hours (as existing clotting factors not affected by warfarin)
Absorption 100% oral bioavailability
Distribution 99% protein bound, small VD 0.14L/Kg
Metabolism Complete hepatic metabolism
Elimination Renal elimination of metabolites
Reversal Vitamin K, FFP, Prothrombinex, cessation+time


Reversal (in more detail)

  • Cessation + time
    • Stopping warfarin, will lead to normalisation of the INR generally in 4-5 days (but varies according to initial INR, comorbidities etc)
    • Mechanism: drug washout
    • Con: slow, risk of bleeding
    • Pro: decreased risk thrombotic events from over/rapid correction
  • Vitamin K
    • can be given IV/IM/PO
    • Higher doses, IV doses can reverse more rapidly
    • Mechanism: replenishes the substrate
  • Fresh frozen plasma
    • Mechanism: Contains all necessary clotting factors - hence rapid reversal
    • Blood product, with all the risks associated with this (fluid overload, infection, allergic responses)
    • Dose: 2-4 units (varies)
    • Con: require crossmatch, time for thawing etc
  • Prothrombinex
    • Mechanism: Contains factors II, IX, X (Aus) 500IU each- hence immediate reversal
    • Dose: 25-50 u/kg
    • Pro: immediate effects, smaller fluid volume, immediately available for use
    • Con: Factor 7 absent, expensive


A general approach, adapted from Red Cross Blood Service

  • Not bleeding
    • INR not too high (<4.5) and/or low bleeding risk = expectant management
    • INR high >4.5 and/or moderate-high bleeding risk = oral/IV Vit K
  • Bleeding
    • Life threatening: IV Vit K 10mg , Prothrombinex 50u/kg (or FFP if not available)
    • Clinically significant: IV VitK 5-10mg, Prothrombinex 25u/kg (or FFP if not available)
    • Minor bleeding: IV VIt K


Examiner comments

43% of candidates passed this question.

Warfarin is listed as a level 1 drug in the 2017 syllabus and as such a detailed knowledge of its mechanism of action would be expected from candidates sitting the exam. The reversal agents for warfarin are collectively classed as level 2 drugs and hence the knowledge required would be at a write short notes level. The following topics were expected: what drugs may be used, how they work, in what dose, any common side effects, why/when would one be used in preference to others etc. The use of reversal agents for warfarin is a common practice in ICU. Generally, answers demonstrated a lack of a precise and detailed knowledge with respect to warfarin’s mechanism of action and had a very superficial knowledge with incorrect facts regarding the reversal agents


Online resources for this question


Similar questions

  • Question 21, 2014 (1st sitting)
  • Question 8, 2015 (1st sitting)
  • Question 10, 2016 (1st sitting)


Question 11

Question

Describe the buffer systems in the body


Example answer

Buffers

  • A solution consisting of a weak acid and its conjugate base
  • Main function is to resist change to pH, with the addition of stronger acids/bases, through reversible binding of H+ ions
  • Effectiveness depends on the buffer pKa, the pH of the solution, the amount of buffer present, whether the system is open or closed
  • All buffers participate in equilibrium with each other in defence of pH (Isohydric principle)


Main buffering systems in ECF

  • Bicarbonate-carbonic acid buffering system
  • Protein buffering system (includes Hb buffering system)
  • Phosphate buffering system


Bicarbonate-carbonic acid system

  • pKa of 6.1
  • Weak acid (H2CO3) and base (HCO3 salt)
  • Via reaction: <math display="inline"> CO_2 \; + H_2O \; \leftrightarrow \; H_2CO_3 \; \leftrightarrow \; HCO_3^- \; + H^+</math>
  • Increased acid > increased CO2 (excreted via lungs)
  • Increased base > increased HCO3 (excreted via kidneys)
  • OPEN system - hence most important - responsible for 80% of the ECF buffering


Protein buffering system

  • Include haemoglobin (150g/L) and plasma proteins (70g/L)
    • Hb has pKa of 6.8. Weak acid (HHb) and weak base (KHb)
  • H+ binds to the histadine residues on imadazole side chains, the HCO3 diffuses down concentration gradient into ECF
  • Hb is quantitatively 6 times more important than plasma proteins, as the concentration is double and there are three times as many histadine residues in Hb

Phosphate buffering system

  • Overall pKa 6.8
  • Tribasic (HPO4, H2PO4, H3PO4) though only the H2PO4 has a physiological pKa to be useful
  • Overall contribution is minimal to the blood due to the low concentration of phosphate. However more important in the urine where the concentration is higher
  • closed system


Examiner comments

57% of candidates passed this question.

This is a core physiology topic; a detailed knowledge of buffering and the available buffer systems is crucial to ICU practice. A candidate presenting for the first part exam should have a detailed understanding of all aspects of the buffer systems. Higher scoring answers provided both technical details of the buffer systems, the context for their normal function and their relative importance. Efficient answers dealt with the buffers by chemical rather than by site, but many answers categorising buffers by site also scored well. Many low scoring answers simply failed to provide detail, some provided incorrect information. Very few candidates demonstrated an understanding of the isohydric principle


Online resources for this question


Similar questions

  • Question 3, 2008 (2nd sitting)
  • Question 19, 2011 (1st sitting)
  • Question 22, 2014 (1st sitting)
  • Question 6, 2018 (1st sitting)


Question 12

Question

Describe the pharmacology of oxycodone.


Example answer

Name Oxycodone
Class Semi-synthetic opioid (phenanthrene)
Indications Analgesia
Pharmaceutics White tablet (IR, MR) +/- naloxone - various conc

Oral solution (1mg/ml)
Clear, colourless, solution (10mg/ml)

Routes of administration PO, IV, SC, IM
Dose Depends

- Example: PO 5-10mg PRN 4hrly, IV 1mg 5 minutes PRN

Morphine equivalence 1.5 x morphine

(10mg oxycodone = 15mg morphine )

Pharmacodynamics
MOA MOP receptor (Gi PCR) in cerebral cortex, basal ganglia, periaqueductal grey

Weak KOP / DOP receptor activity

Effects CNS: Analgesia, sedation, euphoria, dyspho

CVS: bradycardia, hypotension
RESP: respiratory depression (reduces chemoreceptor sensitivity to CO2), depressed cough reflex
GIT: decreased peristalsis > constipation, nausea, vomiting
MSK: pruritis, muscle rigidity
GU: urinary retention
EYE: miosis

Pharmacokinetics
Onset Peak 5 mins, duration 4 hrs
Absorption 80% oral bioavailability
Distribution ~50% protein bound

VOD = 3L/Kg
Crosses placenta

Metabolism Hepatic metabolism (CYP3A4) - extensive

Oxidation > demethylation
Active metabolites: noroxycodone, oxymorphone

Elimination Renal elimination

Active metabolites
T 1/2 = 2-4hrs (IR)

Reversal Naloxone (100mcg IV boluses, PRN 3 minutely)


Examiner comments

54% of candidates passed this question.

There were many exceptional answers which provided extensive detail on the drug. The best of these gave context for the drug characteristics, such as by referring to oxycodone relative to other opioid drugs that might be chosen, or to considerations for safe and effective administration. Some answers, however, provided generic information on opioid drugs, which could not gain all the available marks.


Online resources for this question


Similar questions

  • Question 12, 2017 (1st sitting)


Question 13

Question

List the cell types in the anterior pituitary gland. Outline their secretions, control and target organ effects.


Example answer

Anterior pituitary connected to the hypothalamus via the hypophyseal portal system


Anterior pituitary cell types

  • Chromophils (granular secretory cells)
    • Acidophils (80%)
      • Red staining
      • Includes lactotropes (prolactin) and somatotropes (GH)
    • Basophils (20%)
      • Blue staining
      • Includes gonadotropes (FSH, LH), thyrotropes (TSH) and corticotropes (ACTH)
  • Chromophobes (agranular secretory cells)
    • inactive/degranulated secretory cells
    • Weakly staining, no longer secrete hormones


Anterior pituitary hormones and their effects

Hormone Increased release (stimulation) Decreased release (inhibition) Effects
ACTH Corticotrophin releasing hormone (CRH): stress, catecholamines, ADH Cortisol (negative feedback) Acts on adrenal cortex to increase release and synthesis of glucocorticoids and androgens
TSH Thyrotropin releasing hormone (TRH) from hypothalamus Negative feedback (T3/T4), somatostatin Increased synthesis and secretion of T3/T4 from thyroid cells

Hyperplasia and hypertrophy of thyroid follicular cells

GH (somatotropin) Somatotropin releasing hormone from hypothalamus. Stress, starvation, hypoglycaemia - Somatostatin

- Negative feedback (IGF-1)

- Releases IGFs (anabolic action; growth and differentiation of cells)

- Increases protein, fat, carbohydrate metabolism/utilisation in liver, adipose/muscle tissues.

FSH, LH Gonadotropin releasing hormone from hypothalamus Negative feedback (FSH, LH) LH: ovulation (females), testosterone secretion (males)

FSH: ovarian follicle development (females), spermatogenesis (males)

Prolactin Thyrotropin releasing hormone (TRH), suckling Prolactin inhibiting hormone, dopamine, negative feedback Promotes mammary gland and ductal development (during pregnancy)

Promotes lactation, amenorrhoea (following delivery)



Examiner comments

40% of candidates passed this question.

Few candidates described cell types as chromophils and chromophobes. There were many errant references to chromaffin cells which are found mainly in the adrenal medulla, and to staining on H&E. Chromophil cells stain by absorbing chromium salts. Few candidates mentioned that the hormones secreted by the anterior pituitary are peptides. Most candidates outlined the hypophyseal-portal system well. Knowledge of TSH and ACTH control and target organ effects were good. Similar knowledge for LH, FSH, PRL and GH was much more sporadic.


Online resources for this question


Similar questions

  • None?


Question 14

Question

Describe the pharmacology of sodium bicarbonate.


Example answer

Name Sodium Bicarbonate
Class
Indications Severe NAGMA, alkalinisation of urine (salicylate toxicity), hyperkalaemia, TCA overdose (Na channel blocking effects)
Pharmaceutics Tablet (varying doses e.g. 300mg)

Clear colourless solution (various concentrations e.g, 4.2%, 8.4%) which can be given hypertonic or isotonic

Routes of administration PO, IV
Dose 1mmol/kg IV = 1ml/kg of 8.4% (cardiac arrest due to hyperK)
Pharmacodynamics
MOA Dissociates into Na and HCO3. The HCO3 functions as a buffer in the bicarbonate-carbonic acid buffering system (raising pH). The Na increases the strong ion difference in plasma (raising pH)
Effects Increases pH, alkalinisation of urine
Side effects Hypokalaemia, hypocalcaemia, hypernatremia

Fluid overload (Large doses IV)
Extravasation tissue injury (IV)
Metabolic alkalosis (overdose)

Pharmacokinetics
Onset Immediate
Absorption NA
Distribution Intravascular space
Metabolism <math display="inline"> CO_2 \; + H_2O \; \leftrightarrow \; H_2CO_3 \; \leftrightarrow \; HCO_3^- \; + H^+</math>
Elimination Renal (bicarbonate), lungs (as CO2)
Special points Incompatible with calcium /magnesium salts (precipitates)


Examiner comments

29% of candidates passed this question.

This question was best answered with a structured approach as per any pharmacology question. It nonetheless required good understanding of various aspects of physiology. Many candidates failed to gain marks by omitting to mention facts which could have been prompted by a defined structure. A good response mentioned the pharmaceutic features including formulation and the hypertonicity of IV bicarbonate, pharmacodynamics including indications for use, mode of action, adverse effects (systemic and local), pharmacokinetics and dose. Pleasingly a few candidates stated that sodium bicarbonate’s mechanism of action to cause alkalosis involved increasing the strong ion difference in plasma. Credit was also given for stating the mechanism of action as providing bicarbonate ions to augment the extracellular buffer system


Online resources for this question


Similar questions

  • None?


Question 15

Question

Explain perfusion limited and diffusion limited transfer of gases in the alveolus.


Example answer

Gas diffusion

  • Rate of diffusion of gasses is given by Fick's Law

<math display="block">Diffusion = \frac {A \times D \; \times \Delta P}{T }</math> Where: A= lung area, D = diffusion constant of the gas, <math display="inline">\Delta</math>P = partial pressure gradient of gas, T=thickness of membrane. Diffusion constant is influence by the Temperature of the gas, the density of the gas and the size of the molecules

  • Gas diffusion at the level of the alveolus can either be perfusion or diffusion limited.


Perfusion limited gases

  • Rapidly equilibrates between alveolus and capillary
  • Equilibration time is less than the capillary transit time
  • Thus for the majority of the RBCs time travelling through the capillary, there is no further diffusion
  • As a result this gas is 'perfusion limited' because increasing the blood flow (perfusion) will increase gas transfer, but increasing the rate of diffusion will not
  • Examples
    • Oxygen (under normal conditions)
      • Due to the large partial pressure gradient (100 > 40)
      • Equilibrates within 0.25s (pulmonary capillary transit time 0.75s)
    • Carbon dioxide
      • While a smaller partial pressure gradient (46 > 40), the diffusivity of CO2 is 20 X greater than O2
      • Equilibrates within 0.25s
      • Actually ventilation limited - as you need to blow off CO2 to ensure gradient
    • Nitrous oxide
      • Relatively insoluble and doesn't bind to Hb, therefore struggles to equilibrate


Diffusion limited gasses

  • Does not rapidly equilibrate between alveolus and capillary
  • Equilibration time is greater than the capillary transit time
  • Thus for the entirety of the RBCs time in the capillary there is ongoing diffusion occurring
  • As a result this gas is 'diffusion limited' because increasing the rate of diffusion will increase the rate of gas transfer, but increasing the blood flow (perfusion) will not.
  • Examples
    • Carbon monoxide
      • Slowly diffuses
      • CO binds to Hb so avidly that there is virtually none in the plasma
      • Therefore the equilibrium is never reached and further gas exchange could occur with a greater diffusivity
    • Oxygen
      • Typically perfusion limited under normal circumstances.
      • Under extreme conditions it may become diffusion limited
        • Increase altitude > decreased PAO2
        • High cardiac output > reduced capillary transit time
        • Alveolar membrane disease > decreases rate of diffusion


Examiner comments

36% of candidates passed this question.

This question required detail on those factors affecting gas exchange at the level of the alveolus. A description of the components of the Fick equation was expected - and how this related to oxygen and carbon dioxide transfer at the alveolar capillary membrane. The rapid rate of equilibration (developed tension) was the limiting factor in of blood/alveolar exchange that rendered some gases perfusion limited (examples - N2O, O2 under usual conditions but not all) and the slower rate of others diffusion limited (examples CO and O2 under extreme conditions e.g., exercise, altitude). Estimates of time taken for each gas to equilibrate relative to the time taken for the RBC to travel across the interface was also expected for full marks. CO2 despite rapid equilibration and higher solubility was correctly described as perfusion limited (unless in disease states). Better answers described CO2 as ventilation limited. Some answers also correctly included the component of interaction with the RBC and haemoglobin. Ventilation/perfusion inequalities over the whole lung were not asked for and scored no marks


Online resources for this question


Similar questions

  • None


Question 16

Question

Describe the pharmacology of piperacillin-tazobactam


Example answer

Name Piperacillin-tazobactam
Class Semi-synthetic penicillin (piperacillin)

B-lactamase inhibitor (tazobactam)

Indications Pseudomonal infection

Broad spectrum antimicrobial cover of severe infections/sepsis

Pharmaceutics Powder, reconstitutes in water/NaCl/glucose
Routes of administration IV/IM
Dose 4g/0.5g 8hrly

4g/0.5g 6hrly (pseudomonas cover); dose reduced renal failure

Pharmacodynamics
MOA Piperacillin: bactericidal - inhibits cell wall synthesis by preventing cross linking of peptidoglycans by replacing the natural substrate (D-ala-D-ala) with their B-lactam ring

Tazobactam: B lactamase inhibitor (prevents piperacillin degradation)

Antimicrobial cover Broad spectrum coverage of gram positive bacteria, gram negative bacteria, anaerobes. Covers pseudomonas.

Doesn't cover: MRSA, VRE, ESBL, atypical

Side effects GIT: diarrhoea, nausea, vomiting

Renal: AKI
Allergy (up to 10%), rash most common, skin eruptions/SJS and anaphylaxis (<1/10,000)

Pharmacokinetics
Absorption Minimal oral absorption > IV

Peak concentrations immediately after dose.

Distribution Very good tissue penetration (minimal CNS without active inflammation)

Low protein binding (<30%)

Metabolism Piperacillin: not metabolised

Tazobactam: metabolised to M1, an inactive metabolite

Elimination Renal (80% unchanged)
Special points Removed by haemodialysis


Examiner comments

62% of candidates passed this question.

Most candidates used a structured approach with the usual major pharmacology headings. Mechanism of action was well described by most, with better answers including mechanisms of resistance. Higher scoring candidates included an explanation as to the combination of the drugs. Likewise, better answers included detailed information on spectrum of activity beyond “gram positive and gram negative”, including relevant groups of organisms which are not covered. There also seemed to be some confusion about coverage for anaerobes, which piperacillin tazobactam covers well. Specific detail about adverse reactions, other than ‘allergy, rash, GI upset, phlebitis, etc’, is expected for commonly used antibiotics.


Online resources for this question


Similar questions

  • Question 20, 2019 (2nd sitting)


Question 17

Question

Describe the principles of measurement of arterial haemoglobin O2 saturation using a pulse oximeter (60% marks). Outline the limitations of this technique (40% marks).


Example answer

Definition

  • Non invasive spectrophotometric technique to measure O2 saturation in arterial blood
  • Normal: typically 95-100% (for young health individuals)


Components

  • Two light sources (LEDs): emit light at 660nm and 940nm
  • Light detector (photodiode)
  • Opaque housing unit (minimises ambient light)
  • Signal amplifier, noise filter
  • Microprocessor
  • Connectors , user interface and alarm system


Physical principles

  1. Utilises the principles of the Beer-Lambert law: <math display="inline">A = ε \; l \; c</math>
    • Absorption (A) of light passing through a substance is directly proportional to
      • The optical path length (Lambert's law; l)
      • The concentration of attenuating species within the substance (Beers Law; c)
      • The absorptivity of the attenuating species ( ε )
  1. Utilises the different absorption spectra of Oxy- and deoxy-Hb
    • Deoxy-Hb absorbs far more light in the red spectra (660nm)
    • Oxy-Hb absorbs far more light in the near-infrared spectra (940nm)


How it works

  1. Pulsatile blood (arterial) is isolated and Hb saturation calculated
    • During pulsatile flow, there is expansion and contraction of the blood vessels
    • This alters the optical distance (Lamberts law), changing the absorption spectra
    • Non pulsatile elements (e.g. venous blood) are excluded from the pulsatile elements (arterial blood) by creating a ratio of absorbances (R)
    • Whereas the ratio of absorbances at different spectra (660nm vs 940nm) utilised to calculate saturation of Hb

<math display="block">R = \frac {Pulsatile_{660} \; / \; Non-pulsatile_{660}}{Pulsatile_{940} \; / \; Non-pulsatile_{940}}</math>

  1. Empirical correlation with SaO2
    • The relationship between R and SpO2 was derived empirically by comparing arterial oxygen saturations (from ABGs) at different R values in healthy volunteers
  2. There are important corrections in modern pulse oximeters
    • Correction for Hb concentration using isosbestic points
    • Correction for ambient light using rapid cycling of the light source (up to 1000 hz)


Limitations

  • Requires detectable pulsatile flow (shock, poor perfusion, hypothermia, ECMO, CPB)
  • Bodily movements (e.g. shivering, seizure) confound readings
  • Not accurate nor calibrated at low saturations (progressive decline in accuracy as SaO2 decreases)
  • Not all devices are created equal (device accuracy ranges; generally within 1-5% of ABGs)
  • Ambient light contamination (effects minimal due to rapid cycling as described above)
  • Interference: nail polish, oedema, intravascular dyes (methylene blue)
  • False readings: carbon monoxide poisoning, MetHb
  • Racial bias in pulse oximetry: tested predominately on white population. A study demonstrated 3x as many black patients had occult hypoxemia compared to white patients.


Examiner comments

74% of candidates passed this question.

Most candidates provided a reasonable structured sequence of how a pulse oximeter generates a value. Nearly all candidates described the Beer-Lambert laws correctly, but few specifically described the basic principles of absorption spectrophotometry. Most candidates had a reasonable list of extrinsic factors that can interfere with pulse oximeter performance, but few described the intrinsic/inherent limitations of the device that can cause SpO2 to be different to SaO2, such as functional versus fractional saturation.


Online resources for this question

Similar questions

  • Question 3, 2008 (1st sitting)
  • Question 5, 2013 (2nd sitting)
  • Question 7, 2014 (1st sitting)
  • Question 8, 2018 (1st sitting)


Question 18

Question

Outline the pharmacology of intravenous magnesium sulphate


Example answer

Name Magnesium sulphate
Indications HypoMg, eclampsia/pre-eclampsia, severe asthma, arrhythmias (including TdP), analgesia
Pharmaceutics Clear colourless solution, various concentrations
Routes of administration IV, IM
Dose 5mmol bolus (torsade's).

4g (16mmols) bolus > 1g/hr thereafter (eclampsia, PET)

Pharmacodynamics
MOA Essential cation
- Essential cofactor in hundreds of enzymatic reactions
- Necessary in several steps of glycolysis (ATP production)
- NMDA receptor antagonism (increasing seizure threshold)
- Inhibits Ach release at NMJ (muscle relaxation)
- Smooth muscle relaxation (Inhibits Ca L-type channels)
Effects CNS: anticonvulsant (NMDA effect)

Resp: Bronchodilation (CCB effect > SM relaxation)
CVS: Anti-arrhythmic ( decreased conduction velocity due to CCB effect)

Side effects Related to speed of administration + degree of HyperMg

Toxicity generally occurs > 4mmol/L
CVS: Hypotension, bradycardia
CNS/MSK: hyporeflexia, muscle weakness, CNS depression, potentiates NMBs
RESP: respiratory depression
GIT: Nausea, vomiting

Pharmacokinetics
Onset Immediate
Absorption N/A
Distribution 30% protein bound
Metabolism Not metabolised
Elimination Urine; clearance is proportional to GFR and plasma concentration
Special points Incompatible with calcium salts > precipitation

Drug interaction with NMB agents (potentiation)


Examiner comments

57% of candidates passed this question.

The best answers appropriately addressed the pharmacology of magnesium sulphate, rather than diverting into physiology. They noted that the question concerned intravenous magnesium sulphate and did not discuss other routes. They included pharmaceutics, important examples of the wide-ranging indications, listed potential modes of action and considered the full range of body systems affected including potential adverse effects. Drug interactions, such as potentiation of neuromuscular blocking agents, and pharmacokinetics (including stating that magnesium is not metabolised) were described


Online resources for this question


Similar questions

  • Question 10, 2011 (2nd sitting)
  • Question 20, 2014 (2nd sitting)
  • Question 10, 2015 (2nd sitting)
  • Question 4, 2019 (2nd sitting)


Question 19

Question

Describe the adult coronary circulation (50% marks) and its regulation (50% marks)


Example answer

Vascular anatomy

  • Coronary arteries arise from the sinuses of valsalva at the aortic root
    • Left coronary artery (LCA)
      • LAD
        • Branches: D1, D2 arteries
        • Supplies: anterior 2/3 of the IV septum, anterior LV, LA
      • LCx
        • Branches: OM1, OM2
        • Supplies: inferolateral LV wall, SA node (40%), AV node (20%)
    • Right coronary artery
      • Branches: Right marginal branches
      • Supplies: RA, RV, SA node (60%), AV node (80%)
    • PDA
      • Continuation of RCA (~70%), LCx (~15%), or both (~15%)
      • Supplies: posterior inferior LV
  • Coronary veins
    • Majority (85%) of venous drainage is via the coronary sinus
      • Great cardiac vein (follows LAD)
      • Middle cardiac vein (follows PDA)
      • Small cardiac vein (follows RCA)
    • Remainder (15%)
      • Anterior cardiac veins --> RA
      • Thebesian veins (drains into cardiac chamber directly)


Coronary blood flow

  • CBF ~250mls/min (5% CO)
  • Oxygen extraction near maximal (70%) --> Increased CBF is needed for increased O2 demand.
  • RCA: blood flow is constant, pulsatile and higher flow rate during systole
  • LCA: blood flow is intermittent, pulsatile, and higher flow rate during diastole


Regulation of flow

  • Autoregulation
    • Metabolic autoregulation
      • Anaerobic metabolism > increased vasoactive substances (lactate, adenosine, CO2, NO) > vasodilation > increased flow
      • Predominant means of autoregulation
    • Myogenic autoregulation
      • CBF is autoregulated over a wide range of BPs (perfusion pressure 50-120mmhg)
      • Increased transmural pressure > vasoconstriction > flow reduction
      • Modest means of autoregulation
  • Direct autonomic control
    • Weak effect
    • a1 activation > vasoconstriction; B/muscarinic activation > vasodilation
  • Indirect autonomic control
    • Increase / decrease HR to alter time in diastole/systole which will lead to increased/decreased flow
    • i.e. Increased PSNS activity > decreased HR > increased diastolic time > increased CBF


Examiner comments

62% of candidates passed this question.

Good candidates described normal blood flow to the coronary circulation, including differences between the right and left ventricles. Coronary artery anatomy was outlined, including the regions of the heart supplied and the concept of dominance. In addition to epicardial vessels, strong answers also outlined penetrating arteries, subendocardial supply and venous drainage. Regulation of coronary blood flow required an explanation of flow-dependence of the heart given its high oxygen extraction rate. Metabolic autoregulation and its mediators needed to be described, along with the physical factors driving coronary blood flow. Less important mechanisms such as the role of the autonomic nervous system were also described, with an emphasis on indirect effects over direct effects.


Online resources for this question


Similar questions

  • Question 11, 2008 (2nd sitting)

  • Question 10, 2014 (2nd sitting)

  • Question 11, 2018 (1st sitting)

Question 20

Question

Outline the physiological factors that influence cerebral blood flow


Example answer

Cerebral blood flow (CBF)

  • ~15% CO (~750mls/min) under normal resting conditions
  • High demand due to high metabolic rate
  • Brain is very sensitive to interruptions (due to high demand and inability to store energy)


Factors effecting CBF

  • Related to blood pressure, vessel characteristics, rheological factors per Hagan-Poiseuille eqn.
    • <math display="inline">CBF \; = \; \frac {\Delta P \pi R^4}{8 \eta L}</math>
    • Where: ΔP represents the driving pressure (i.e. CPP), R is the radius of the blood vessels, η is blood viscosity, L is the length of the tube.


  1. Pressure effects

    • Cerebral perfusion pressure (CPP) = MAP - ICP (or CVP whichever is higher)

    • Normally CBF is kept constant across a wide range of CPP (approx 50-150mmHg)

    • Able to do this via cerebral autoregulation

      • Predominant means is myogenic autoregulation

        • Increased CPP > Inc. stretch > inc. wall tension > vasoconstriction > decreased CBF

    • Outside autoregulatory ranges (e.g. MAP <50mmhg or >150mmHg)

      • CBF becomes pressure passive (any increase in CPP = increase CBF, vice versa)

  2. Vessel radius effects

    • Myogenic autoregulation (per above)

    • PaCO2

      • CO2 is a cerebral vasodilator = increased CBF

      • Linear increase in CBF for increase CO2 between 20-80mmHg

    • PaO2

      • Within normal physiological limits does not effect CBF

      • Exponential increase in CBF with hypoxia (e.g. PaO2 <50-60) due to vasodilation

    • Metabolism/metabolic autoregulation

      • Linear increase in CBF for any increase in mebtaolism (flow metabolism coupling)

      • Controlled by local vasoactive mediators (H+, adenosine, NO)

    • Neurohormonal

      • Minimal impact of hormones (e.g. adrenaline) on vessel radius

      • Minimal impact of ANS mediated vasoconstriction

  3. Rheological factors

    • Minimal effect, does not rapidly change

    • Mostly dependant upon HCT

    • Increased HCT = increased resistance = reduced blood flow

  4. Vessel length

    • Would not change = no effect


Examiner comments

19% of candidates passed this question. Overall, this question was poorly answered with a high failure rate. A good answer gave a normal value, iterated that CBF is held relatively constant by autoregulation, and proceeded to divide factors affecting CBF into categories with an explanation/description of each. Those factors with the greatest influence were expected to have more accompanying information (e.g., pressure/myogenic autoregulation, metabolic). Systemic factors such as MAP, O2, CO2 were expected to be mentioned with detail of the impact (i.e., key values, relationships demonstrated with a description and/or labelled graph). Local factors within the brain such as H+ concentration/pH, metabolic activity (including the impact of temperature, inclusion of mediators, regional variation based on activity & grey versus white matter) were also expected to be mentioned. Few answers mentioned impact of pH change independently of CO2. Few answers mentioned how CO2 changes the pH of CSF and that over time, this impact is buffered/reduces. The role of the sympathetic nervous system was required to be mentioned although not explored in detail (although many answers overstated the importance of the SNS on CBF or gave a simplistic concept such as increased SNS activity increases CBF). Many answers focussed on descriptions of the Monro-Kelly doctrine and ICP to the exclusion of the aforementioned factors or included detail on factors influencing MAP which were not required (and irrelevant when within the autoregulation range). Many answers were simplistic: e.g., increase MAP increase CPP therefore increase CBF, or by stating CO2/O2 without mentioning a relationship or the limits/patterns of the relationship. Many answers failed to separate the effect of systemic PaO2 and PaCO2 from metabolic autoregulation.


Online resources for this question


Similar questions

  • Question 11, 2009 (1st sitting)
  • Question 5, 2008 (1st sitting)



2020 (2nd sitting)

Question 1

Question

Describe and compare the action potentials from cardiac ventricular muscle cells and the sino-atrial node.


Example answer

Pacemaker vs myocyte action potential

Myocyte Pacemaker
Resting potential -90 mV No set RMP but minimum potential is around -60 mV
Threshold potential -70 mV -40 mV
Phases of AP
Phase 4 - Resting membrane potential (-90mV)

- Maintained by inward rectifying K current

- Slow depolarisation (drift) to threshold (-40mv)

-Funny current Na influx, slowing K efflux

Phase 0 - Rapid depolarisation at threshold (-70mV)

- Fast voltage gated Na opens (influx) > depolarisation
-Peak around +40mV

- Depolarisation (slower relative)

- L type Ca channels open (influx)
-Peak around +20mV

Phase 1 - Partial repolarisation

- Na close (stops influx), K opens (efflux)

No Phase 1
Phase 2 - Long Plateau (100-200ms)

- L type Ca channels opens (influx) which balances K efflux

No phase 2, no plateau
Phase 3 - Rapid repolarisation to membrane potential (~-90mv)

- L type Ca channels close, continued K efflux

- Rapid repolarisation

-K open (efflux), Ca close (stops influx)

File:Https://derangedphysiology.com/main/sites/default/files/sites/default/files/old image pile/Neurocritical-care/images/comparison of ventricular myocyte and pacemaker action potentials 3.jpg

image-20220202114143450image-20220202114127372


Examiner comments

72% of candidates passed this question.

This question details an aspect of cardiac physiology which is well described in multiple texts. Comprehensive answers included both a detailed description of each action potential and a comparison highlighting and explaining any pertinent differences. The question lends itself to well-drawn, appropriately labelled diagrams and further explanations expressed in a tabular form. Better answers included a comparison table with points of comparison such as the relevant RMP, threshold value, overshoot value, duration, conduction velocity, automaticity, ion movements for each phase (including the direction of movement) providing a useful structure to the table. Incorrect numbering of the phases (0 – 4) and incorrect values for essential information (such as resting membrane potential) detracted from some responses


Online resources for this question


Similar questions

  • Question 23, 2010 (2nd sitting)
  • Question 19, 2013 (1st sitting)
  • Question 11, 2016 (2nd sitting)
  • Question 21, 2017 (2nd sitting)



Question 2

Question

Define functional residual capacity (10% marks). Outline the functions (70% marks) of the functional residual capacity and the factors affecting it (20% marks).


Example answer

Functional residual capacity

  • The volume of gas in the lungs at end-expiration during tidal breathing
  • Typically ~30mls/kg (or ~2.1L in 70kg adult)
  • Sum of the residual volume and expiratory reserve volume
  • Represents the point at which the elastic recoil force of lung, and the expanding elastic force of the chest wall are equal


Functions/role of FRC

  • Oxygen reservoir
    • Maintains an oxygen reservoir > maintains oxygenation between breaths / periods of apnoea
    • Prevents rapid changes in PaO2
  • Maintains small airway patency
    • At FRC, the airway resistance is low
    • Normally FRC > closing capacity (prevents atelectasis)
  • Reduces work of breathing
    • At FRC, lung compliance is maximal and airway resistance is low
  • Minimises cardiac workload
    • At FRC, pulmonary vascular resistance is minimal
  • Important starting point for measuring lung volumes


Factors effecting FRC

  • Lung size
    • Increasing lung size = increasing FRC
    • Thus affected by
      • Height (Taller FRC > shorter)
      • Age (adult FRC > children)
      • Gender (Male FRC > female)
  • Respiratory compliance
    • Increase in compliance
      • e.g. emphysema, increased PEEP
      • Leads to increased FRC
    • Decrease in compliance
      • E.g. ARDS, obesity, pregnancy
      • Leads to reduction in FRC
  • Age (increasing age generally increases FRC)
  • Anaesthesia
    • Reduces FRC (multifactorial)
  • Posture
    • FRC decreases when going from erect to supine position


Thus if FRC is reduced we will get

  • Reduction in
    • Lung compliance
    • oxygen reserves
    • tidal volumes
  • Increase in
    • airway resistance
    • pulmonary vascular resistance
    • atelectasis
    • work of breathing
    • V/Q mismatch


Examiner comments

79% of candidates passed this question.

This question was in two parts with the percentage of marks allocated an indication of the relevant time or detail expected per part. The second part of the question also contained two distinct headings which should have been used in the answer. As an outline question, dot points with a brief explanation of each point were expected. Most candidates drew diagrams, few of which added value. For a diagram to add value it should be accurate, have labelled axes, a scale with numerical values and units. As a general rule, diagrams should also be explained and help to illustrate or relate to a written point.
For factors affecting FRC, to score full marks, it should be clearly stated if the factor causes an increase or decrease in FRC. This topic is well covered in the recommended respiratory texts.


Online resources for this question


Similar questions

  • Question 15, 2010 (2nd sitting)
  • Question 4, 2015 (2nd sitting)
  • Question 8, 2017 (1st sitting)
  • Question 24, 2017 (2nd sitting)


Question 3

Question

Describe the pharmacology of hydrocortisone.


Example answer

Name Hydrocortisone
Class Glucocorticoid (endogenous)
Indications Glucocorticoid insufficiency, allergy/anaphylaxis/asthma, severe septic shock, immunosuppression (e.g. transplant, autoimmune dz)
Pharmaceutics Tablet, white powder diluted in water
Routes of administration IV, PO
Dose 50-200mg QID (commonly in ICU population)
Bio-equivalence 100mg hydrocort = 25mg pred = 20mg methypred = 4mg dex
Pharmacodynamics
MOA Lipid soluble > crosses cell membrane > binds to intracellular steroid receptors > alters gene transcription > metabolic, anti-inflammatory & immunosuppressive effects in tissue-specific manner
Effects CNS: sleep disturbance, psychosis, mood changes

CVS: Increased BP (mineralocorticoid effect + increased vascular smooth muscle receptor expression to catecholamines)
RESP: decreased airway oedema, increased SM response to catecholamines
RENAL: Na + water retention (mineralocorticoid effect)
Metabolic: Hyperglycaemia, gluconeogenesis, protein catabolism, fat lipolysis and redistribution, adrenal suppression
MSK: Osteoporosis, skin thinning
Immune: immunosuppression + anti-inflammatory effects (decreased phospholipase, interleukins, WBC migration and function)
GIT: Increased risk of peptic ulcers

Pharmacokinetics
Onset Peak effect 1-2 hours, duration of action 8-12 hours
Absorption 50% oral bioavailability
Distribution 90% protein bound, small Vd (0.5L/kg)
Metabolism Hepatic > inactive metabolites
Elimination Metabolites excreted renally. Elimination T/12 = ~1 hour
Special points Risk of reactivation of latent TB / other infections


Examiner comments

69% of candidates passed this question.

Hydrocortisone is a level 1 drug in the syllabus. Most answers were well structured, many used key headings. In general, detailed information specific to hydrocortisone was lacking. Answers that focused on the mechanism of action, pharmacodynamic effects and pharmacokinetics effects which were detailed and accurate scored well. It was expected that significant detail be included in the sections with relevance to clinical practice for example, the mechanism of action and pharmacodynamic effects including the side effect profile. An indication/appreciation of the timelines of such was also represented in the marking template.


Online resources for this question


Similar questions

  • Question 10, 2017 (1st sitting)


Question 4

Question

Outline the role of the liver in the metabolism of fat (â…“ marks), carbohydrate (â…“ marks) and proteins (â…“ marks).


Example answer

Carbohydrate metabolism

  • Glycolysis
    • Metabolises glucose to generates ATP + pyruvate.
    • Pyruvate is converted to Acetyl Coa and enters the TCA cycle (aerobic) or is converted to lactate (anaerobic)
    • Catabolic role
  • Glycogenesis
    • The liver can store up to a 100g of glucose in the form of glycogen
    • Stimulated by insulin (released from the pancreas) when BSLs are HIGH
    • Anabolic role
  • Glycogenolysis
    • Liver can mobilise stored glycogen to produce glucose via glycogenolysis
    • Stimulated by glucagon (released from pancreas) when BSLs are LOW
    • Catabolic role
  • Gluconeogenesis
    • Liver can synthesise glucose from non-carbohydrate precursors (amino acids, lactate, glycerol)
    • Stimulated by glucagon (released from pancreas) when BSLs are LOW
    • Anabolic role


Fat metabolism

  • Lipid breakdown (B oxidation)
    • In the liver, free fatty acids undergo B-oxidation to Acetyl CoA
    • Acetyl Coa then enables energy production by entering Krebs Cycle
    • Catabolic role
  • Lipid synthesis
    • Lipids, including cholesterol, are synthesised in liver from Acetyl CoA
    • Anabolic role
  • Lipid processing
    • Apolipoproteins are synthesised in the liver and are responsible for processing of VLDL, LDL, HDL


Protein metabolism

  • Protein synthesis
    • Liver is responsible for synthesis of most plasma proteins (except immunoglobulins)
    • Anabolic role
  • Deamination
    • Individual amino acids have their amino groups removed by liver > a keto acids > TCA cycle
    • Catabolic role
  • Amino acid synthesis
    • Keto-acids can be transformed into non-essential amino acids by transamination, forming new amino acids.
  • Urea formation
    • Ammonia (end product of amino acid degradation) is converted to urea > excretion in urine


Examiner comments

54% of candidates passed this question.

This question relates to basic hepatic physiology and is well described in the recommended texts. The mark allocation and division of time was indicated in the question. Better answers used the categorisation in the question as an answer structure. Many candidates gave a good description of beta oxidation, the formation of Acetyl Co A and ketone synthesis. A description of the synthesis of cholesterol, phospholipids, lipoproteins and fatty acid synthesis from proteins and carbohydrates mainly using glycogen, glucose and lactate also received marks. Candidates seem to have a better understanding of fat and glucose metabolism than protein metabolism. Higher scoring candidates appreciated the anabolic and catabolic processes of each component.


Online resources for this question


Similar questions

  • Question 18, 2015 (1st sitting)


Question 5

Question

Describe the anatomy (70% marks) and effects (30% marks) of the sympathetic nervous system.


Example answer

Sympathetic nervous system (SNS)

  • Portion of the autonomic nervous
  • Provides involuntary control of many bodily functions


Anatomy of SNS

  • Preganglionic component
    • Short, myelinated, preganglionic neurons
    • Originate in the lateral horn of the spinal cord between T1 and L3 (thoracolumbar outflow)
    • Travel via ventral roots and white rami communicantes to synapse in the ganglia
    • Neurotransmitter is acetylcholine > nicotinic receptor
  • Ganglionic component
    • Two types (prevertebral ganglia and paravertebral ganglia)
      • Paravertebral ganglia form the two sympathetic chains which extend along the vertebral column
      • Prevertebral ganglia are located in abdominal cavity around branches of aorta (e.g.. coeliac ganglia)
    • Preganglionic neurons can synapse at ganglia above, below, at same level or directly to prevertebral ganglia
  • Post ganglionic component
    • Long, unmyelinated, postganglionic neurons
    • Leave the ganglia through the grey matter communicantes > effector tissue/organ
    • Neurotransmitter is noradrenaline > adrenergic receptor
    • There are exceptions e.g. Adrenal medulla: directly innervated by preganglionic neurons (ACh)


Effects of SNS

Organ SNS
Heart Increased chronotropy (B1) and inotropy (B1), increased lusitropy
Arterioles Vasoconstrict (a1, a2)
Lung Bronchodilation (B2)
GIT Inhibition of peristalsis (predominately a1,a2)
Liver Glycogenolysis (B2)
Renal Increased renin release (B1)
Pupils Dilation (a1)
Salivary glands Inhibition of salivation
Adrenal gland Release of norad and adrenaline
Bladder Detrusor relaxation (B2), sphincter contraction (a1)
Sweat gland Sweat (ACh)


Examiner comments

51% of candidates passed this question.

Most candidates had a suitable structure to their answers, those without a clear organisation of thought tended to gain fewer marks. In many cases incorrect information or limited detail, particularly around the anatomical organisation prevented higher marks.


Online resources for this question


Similar questions

  • Question 17, 2013 (2nd sitting)
  • Question 20, 2015 (1st sitting)



Question 6

Question

Classify the oral hypoglycaemic drugs (20% marks); include their mechanism of action (40% marks) and their most significant side effects (40% marks).


Example answer

Hypoglycaemic agents

Drug class Example Mechanism of action Important side effects
Commonly used
Biguanides Metformin Multiple mechanisms of action.

1) Inhibits hepatic+renal gluconeogenesis
2) increases insulin sensitivity (increases GLUT4 receptors to increase peripheral utilisation),
3) Delayed glucose uptake from GIT

Lactic acidosis (higher risk with renal/liver impairment) due to increased glycolysis and impaired gluconeogenesis leading to lactatemia

GIT upset (diarrhoea, nausea, vomiting)

Sulfonylureas Gliclazide Increase insulin secretion from pancreatic B cells , reduce insulin sensitivity Risk of hypoglycaemia, GIT upset, blood dyscrasias
DPP-4 inhibitors Sitagliptin Inhibit DPP-4 (which normally breaks down GLP-1). GLP-1 stimulates insulin release from pancreas, reduces appetite, delays gastric emptying Risk of hypoglycaemia

Risk of pancreatitis

SGLT-2 inhibitors Empagliflozin Inhibits SGLT-2 receptors > decrease glucose reabsorption in the PCT Osmotic diuresis (Polyuria, polydipsia, dehydration), euglycemic diabetic ketoacidosis, risk of hypoglycaemia, UTIs
Not commonly used
Alpha glucosidase inhibitors Acarbose Slows/prevents carbohydrate breakdown and absorption GIT upset
Thiazolidineodiones Pioglitazone Increases insulin sensitivity via PPAR receptors in fat cells Increased risk of heart failure
Meglitinides Repaglinide Similar to sulfonureas, though different receptor Hypoglycaemia, sig. interaction with antifungals > high levels > hypos


GLP-1 agonists

  • Commonly given S/C
  • New oral agents are becoming available - not yet widely used in AUS


Examiner comments

37% of candidates passed this question.

High scoring answers most often started with a strong and logical structure and focused on the requested categories of information. Many candidates gave good answers across the wide range of drugs. Several candidates could have scored more highly by giving more correct information on biguanides and sulphonylureas.


Online resources for this question


Similar questions

  • Question 6, 2013 (1st sitting)



Question 7

Question

Compare and contrast external ventricular drains and intraparenchymal fibreoptic pressure monitors.


Example answer

External ventricular drain (EVD) intraparenchymal catheter (e.g. codman)
Location/anatomy Sits in the lateral ventricle. Inserted 2-3cm lateral midline ~10cm posterior to the nasion (Kochers point) aiming away from motor cortex Sits in the brain parenchyma (2cm depth)
Method of measurement Pressure transmitted to wheatstone bridge via fluid filled non compressible tubing. Pisoelectric strain gauge pressure sensor, connected to monitor via fibreoptic cable
Calibration Yes, can be zero'd post insertion to atmosphere No, cannot be zeroed post insertion
Diagnostic (ICP) Yes, gold standard. Yes
Diagnostic (CSF sample) Yes, can sample CSF No, cannot sample CSF
Therapeutic

(drain CSF)

Yes No, cannot drain CSF
Sources of error Migration of catheter tip, blockage of EVD, incorrect levelling to tragus, damping/resonance Drift, only measures local ICP (not global ICP)
Advantages Diagnostic (CSF sample, ICP) and therapeutic (high ICP), can be re-zeroed, cheaper Easier to insert with less expertise, less complications (infection, haemorrhage)
Disadvantages Increased risk of: trauma, infection, misplacement Decreased risk of trauma, infection, haemorrhage. Not therapeutic. Cannot be recalibrated and prone to drift. More expensive. measures local ICP only


Examiner comments

22% of candidates passed this question.

This question is ideally suited to a tabular format, where candidates are expected to highlight the significant similarities and differences as well as why a certain monitor may be chosen in preference to another rather than compile two lists written next to each other. To score well in this question, a statement of what could be measured (ICP: global vs local), a description of the measurement principles, along with other measurement related information like calibration and sources of error was required. Also sought was information regarding anatomical placement (e.g., lateral ventricle for EVD) and method of placement.
Furthermore, a comparison with each other (e.g., higher infection/bleeding risk with EVD, greater risk of trauma due to size and insertion, expertise to insert, cost, therapeutic benefit, risk of blocking) was required for completion. Candidates who structured these elements into advantages and disadvantages were generally able to elucidate this information and score better.


Online resources for this question

  • Jennys Jam Jar
  • [chrome-extension://efaidnbmnnnibpcajpcglclefindmkaj/viewer.html?pdfurl=https%3A%2F%2Fcicmwrecks.files.wordpress.com%2F2021%2F05%2F2020-2-07.pdf&clen=256304&chunk=true CICM Wrecks]
  • Deranged Physiology


Similar questions

  • ?None so far as I can tell



Question 8

Question

Describe the cough reflex.


Example answer

Cough

  • Complex, sudden expulsion of air from the airways
    • Can voluntarily cough, however the cough reflex = involuntary


Purpose of cough reflex

  • Airway protective function
    • Helps clear foreign material/noxious stimuli from the airway
  • N.B: Useful clinically in brain death testing


COUGH REFLEX


Sensors

  • Rapidly adapting mechanoreceptors, slowly adapting mechanoreceptors, and c-fibres


Stimulus for cough

  • Chemical, mechanical, noxious stimuli in the airways (larynx, trachea, carina, bronchi)
    • e.g. leukotrienes, histamine, bradykinin, foreign bodies


Afferents

  • Afferents from the internal laryngeal nerve (br. of vagus nerve)


Integrator/controller

  • Vagal afferents synapse in the medullary respiratory centre (NTS)


Efferents

  • Diaphragm (via phrenic nerve)
  • Abdominal muscles (via spinal motor nerves)
  • Larynx (via laryngeal branch of vagus nerve)


Effector / mechanism

  • Coordinated action of respiratory, pharyngeal, abdominal muscles

  • Phase 1: inspiratory phase

    • Deep inspiration to near vital capacity (muscles of inspiration, including diaphragm)

  • Phase 2: compressive phase

    • Closure of the cords+epiglottis, contraction of the abdominal and intercostal muscles

    • Leads to dramatic rise in intrapleural pressure (>100cmH2O)

  • Phase 3: expulsive phase

    • Sudden partial opening of the cords and epiglottis

    • Leads to violent expiration of turbulent air removing foreign material

Examiner comments

62% of candidates passed this question.

Overall, this question was reasonably well answered. Those that performed well had suitably detailed knowledge and structured their responses which generally included a definition and purpose of the reflex as well as the identification and a description of the afferent, integrator/controller, and efferent limbs of the reflex. This structure allowed a logical platform for the elucidation of the detail required in the answer, including types of stimulus, receptors, nerves (for both limbs of the reflex) and the muscles used in the phasic response to be clearly articulated.


Online resources for this question


Similar questions

  • Question 13, 2014 (1st sitting)

  • Question 8, 2020 (2nd sitting) of the Fellowship exam


Question 9

Question

Outline the daily nutritional requirements, including electrolytes, for a normal 70 kg adult.


Example answer

Energy intake

  • Most guidelines recommend around 25-30 kcal/kg of energy / day
    • Approx 2000 kcal / day for average adult
  • Met with a combination of carbohydrates, protein, fats
  • Critically unwell patients, may need more due to increased energy expenditure.


Carbohydrates

  • Preferred substate for energy production
  • Average intake (adult) ~350g / day
    • Minimum recommended intake >2g.kg.day
  • 1g carbs = 4 kCal energy


Fats

  • Provides essential fatty acids (e.g. Omega 6 + 3 fatty acids)
  • Essential for synthesis of cell membranes and for fat soluble vitamins (A,D,E,K)
  • Recommended intake = 1g.kg.day (i.e. ~70g / day)
  • 1g fat = 9kCal energy
  • Ideal carb : fat ratio not empirically known, but in practice we use ~70:30


Protein

  • Replaces essential amino acids (e.g. phenylalanine, valine, leucine) which cannot be produced in vivo
  • Recommended intake (healthy adult) = 1g.kg.day
    • Critically ill patients will need more (1.5- 2g.kg.day - i.e. 100-140g/day)
  • 1g protein = 4 kCal energy
  • Not typically included in the resting energy expenditure


Water / electrolytes

  • Water: 30ml/kg/day
  • Sodium: 1-2mmol/kg/day
  • Potassium: 1mmol/kg/day
  • Calcium: 0.1mmol/kg/day
  • Magnesium: 0.1 mmol/kg/day
  • Phosphate 0.4mmol/kg/day


Vitamins

  • Organic compounds that the body is unable to synthesise, though needs for cellular function

    • Commonly enzyme cofactors, antioxidants, metabolic regulators

    • Required in small amounts

  • Fat soluble

    • A,D,E,K

    • Excessive intake > toxicity

    • Stored largely in liver

  • Water soluble

    • E.g. vitamin C, B1, nicotinic acid, B12, folate

    • Not readily stored - readily excreted in urine > less likely to be toxic

Trace elements

  • E.g. zinc, copper, iron, selenium, iodine
  • Needed for daily functioning in trace amounts


Examiner comments

40% of candidates passed this question.

This topic is well covered in the recommended physiology textbooks. Many answers unfortunately simply listed the various components without providing sufficient detail; outline questions require some context around the key points as opposed to just a list.
Most candidates had a good estimate for the basal energy requirements of a resting adult. Good candidates were able to outline the g/kg daily protein requirements and the distribution of remaining energy intake between carbohydrates and lipids and included how this may change during periods of stress. They also stated the energy derived per gram of each of those food groups. Few candidates mentioned the need to include essential amino acids. Similarly, with fat intake, few candidates mentioned the need for essential fatty acids. A definition of “vitamin” would have received credit. Most candidates were able to classify vitamins as water soluble or fat soluble. Most candidates mentioned trace elements (with an abbreviated list) and mentioned bone minerals. A daily intake requirement for Na and K was expected, though not for bone minerals or trace elements.


Online resources for this question


Similar questions

  • Question 2, 2017 (2nd sitting)



Question 10

Question

Describe the pharmacology of suxamethonium.


Example answer

Name Suxamethonium (succinylcholine)
Class Depolarising muscle relaxant
Indications Facilitate endotracheal intubation during anaesthesia (i.e. RSI)
Pharmaceutics Clear colourless solution (50mg/ml), needs refrigeration (4°C) or else lasts only a couple of weeks at room temp
Routes of administration IV, IM
Dose 1-2 mg/kg (IV), 2-3 mg/kg (IM)
Pharmacodynamics
MOA Binds to the nACh receptor on motor end plate > depolarisation. Cannot be hydrolyed by Acetylcholinesterase in NMJ > sustained depolarisation (i.e. Na channels remain in open-inactive state) > muscle relaxation
Effects Flaccid paralysis.
Side effects Major: anaphylaxis, suxamethonium apnoea, malignant hyperthermia

Minor: hyperkalaemia, myalgia, bradycardia/arrhythmia
Pressure: increased IOP, ICP, intragastric pressure.

Pharmacokinetics
Onset Onset 30s - 60s, duration <10 mins
Absorption -
Distribution 30% protein bound

Vd = 0.02 L/Kg

Metabolism Rapid hydrolysis by plasma cholinesterase's (~20% reaches NMJ)
Elimination Minimal renal elimination (due to rapid metabolism)
Special points May have prolonged duration of action with congenital or acquired (e.g. liver, renal, thyroid disease) pseudocholinesterase deficiency

Treatment of malignant hyperthermia is with dantrolene (+ cooling + supportive care)


Examiner comments

63% of candidates passed this question.

This was a level 1 pharmacology question, and it represents core knowledge. The mechanism of action of suxamethonium and the interactions at the neuromuscular junction as well as pharmaceutics were areas that often required further detail. Few candidates mentioned the effects of suxamethonium on the autonomic nervous system. Another common omission related to the factors that reduce plasma cholinesterase activity beyond genetic deficiency (such as liver disease, renal failure, thyrotoxicosis). Pleasingly, there was generally a good understanding of role, dosing, side effect profile, pharmacokinetics and of special situations and limitations of use pertinent to this drug.


Online resources for this question

Similar questions

  • Question 6, 2011 (1st sitting)
  • Question 2, 2012 (2nd sitting)
  • Question 1, 2013 (2nd sitting)
  • Question 10, 2018 (1st sitting)
  • Various other questions relating to properties of NMB more broadly



Question 11

Question

Describe the changes in the circulatory system that occur during exercise.


Example answer

Exercise

  • Leads to increased oxygen demand (predominately skeletal muscle) and increased metabolic waste products which need to be cleared
  • Leads to many circulatory changes:


Cardiac output

  • Increased oxygen demand > increased CO (as CO is the main modifiable component of the oxygen delivery equation - Hb, Sats, PaO2 not readily changeable)
  • Most of the increased CO goes to skeletal muscle beds
  • Due to a combination of increased HR/SV
  • Increases 5-6x - from 5L/min up to 30L/min


Heart rate

  • Increased HR due to SNS mediated chronotropy
  • Max = 220-age


Stroke volume

  • Increased SV initially is due to
    • Reduced afterload (skeletal muscle vasodilation > decreased peripheral vascular resistance)
    • Increased preload (peripheral venoconstriction > increased venous return)
    • SNS mediated inotropy
  • With increasing HR, SV will begin to decrease (due to reduced diastolic filling time)

    • Plateaus at ~50% VO2max

Redistribution of blood flow

  • Vasodilation in skeletal muscle beds
    • Mediated by local factors (hypoxia, CO2, Lactate, adenosine) which lead to vasodilation (to decrease resistance, thus increase blood flow)
    • Also mediated by autonomic factors: SNS activation > B2 stimulation > vasodilation
  • Vasoconstriction of non working tissues
    • SNS mediated vasoconstriction of GIT, Kidneys > blood flow directed to "working tissues"
  • Coronary blood flow
    • Increases by metabolic autoregulation due to increased demand from increased inotropy/chronotropy
  • Cerebral blood flow
    • Remains constant (autoregulation) - no increase in metabolic demand. Increased BP > myogenic vasoconstriction.


Increased oxygen extraction

  • Increased CO2 and H+ and temperature in working skeletal muscle beds > right shift of the oxygen-Hb dissociation curve > increased O2 extraction (Bohr effect)


Blood pressure/s

  • Increased SBP (due to increased inotropy > increased CO)
  • Decreased DBP (due to reduced SVR from skeletal vasodilation)
  • Widened pulse pressure (increased SBP, decreased DBP)
  • Overall increase in MAP (increase in CO is greater than reduction in PVR)


Other haemodynamics

  • Increased venous return > increased CVP and PCWP


Examiner comments

22% of candidates passed this question.

This is an applied physiology question. Better answers categorised the changes in some manner and included a measure of the degree of change as applicable (e.g., what increases, what decreases and what may stay the same). The question was to describe the changes so that the detail behind the mechanisms enabling these changes to occur was expected (e.g., neurohumoral, local factors). Marks were also awarded for any regional variation that occurs


Online resources for this question


Similar questions

  • ? none



Question 12

Question

Describe the physiology (50% marks) and pharmacology (50% marks) of albumin.


Example answer

Albumin physiology

  • Structure
    • Human plasma protein
    • 69kDa
  • Synthesis
    • Synthesised in the liver (~10-15g/day)
    • Decreased synthesis: liver disease, protein malnutrition, sepsis/infection (prioritises other Acute Phase Reactants)
  • Distribution
    • Accounts for ~50% plasma proteins
    • 40% intravascular, 60% extravascular (skin, muscle, liver)
  • Functions
    • Osmotic pressure - accounts for majority (80%) of plasma oncotic pressure
    • Transport / drug binding (mainly acidic drugs)
    • Acid-base buffer (protein-buffering system)
    • Detoxification role
  • Breakdown
    • Broken down by cysteine protease into amino acids
    • Half life ~20 days
  • Elimination
    • Elimination half life 16 hours
    • Increased loss with renal dysfunction (e.g. nephropathy)


Albumin Pharmacology

Name Albumin
Class colloid (human plasma protein)
Indications Intravascular volume replacement, low albumin, hepatorenal syndrome, SBP
Pharmaceutics 4% or 20% concentrations. Hypotonic

-Collected by blood donation (whole blood, plasma apheresis) > fractionated > pasteurised > partitioned > stored.

Routes of administration IV
Pharmacodynamics
MOA Related to volume of fluid (i.e. volume expansion) and role of albumin (oncotic, transport, etc)
Side effects No risk of bacteria/parasite infections (destroyed during processing), but risk of blood borne viruses (HIV, HepB, HCV) remains.

Allergy, fluid overload.

Pharmacokinetics
Absorption IV only (0% oral bioavailability)
Distribution Rapid distribution within intravascular space.

Small Vd - about 5% leaves per hour

Metabolism Cellular proteolysis by cysteine protease
Elimination Degradation by liver and reticuloendothelial system
Special points - May worsen outcomes in TBI

- No need for blood cross matching


Examiner comments

19% of candidates passed this question.

The question required an equal treatment of the physiology and pharmacology of albumin. The physiology discussion needed to include synthesis, factors affecting synthesis, distribution in the body (including the proportion divided between the plasma and interstitial space), functions, breakdown, and elimination half-life. Discussion of the pharmacology should have included available preparations (4% and 20% Albumin) and pharmaceutics, distribution, elimination (both the protein and crystalloid components), mechanism of action to expand the plasma compartment, longevity in the plasma compartment, indications, and adverse effects. Oedema, circulatory overload, immunological reactions, and relative contraindication in brain injury were important to mention. There was some confusion regarding the infectious risks of albumin. An outline of the manufacturing process from donated plasma and pasteurisation was expected.


Online resources for this question

  • Deranged Physiology
  • Jennys Jam Jar
  • [chrome-extension://efaidnbmnnnibpcajpcglclefindmkaj/viewer.html?pdfurl=https%3A%2F%2Fcicmwrecks.files.wordpress.com%2F2021%2F05%2F2020-2-12.pdf&clen=247533&chunk=true CICM Wrecks]


Similar questions

  • Question 1, 2015 (2nd sitting)
  • Question 1, 2009 (2nd sitting)


Question 13

Question

Describe the anatomical (20% marks) and physiological (80% marks) features of the pulmonary circulation.


Example answer

Anatomical features (pulmonary circulation)

  • Low pressure, low resistance, high capacitance, high flow system
    • Thus --> vessel walls are highly elastic + have less muscle + much thinner than systemic circulation
  • Circulation
    • RV > Pulmonary trunk > R/L pulmonary artery > progressively smaller pulmonary arteries > capillaries > progressively larger pulmonary veins > pulmonary veins x4 > LA
    • Arteries and veins travel with respective bronchi, nerves and lymphatics in bronchovascular bundle


Physiological features (pulmonary circulation)

  • Low pressure system

    • Normal PA

      • Systolic pressure 15-25mmHg

      • Diastolic pressure 8-15mmHg

      • Mean pressure 10-15mmHg

    • Pulmonary venous pressure ~8-10mmHg

  • Low resistance system

    • ~100-200dynes/sec/cm-5

    • ~10% of systemic circulation

    • With further flow (e.g. increased CO during exercise) can maintain low resistance by recruitment of additional capillaries

  • High flow system

    • Pulmonary arterial flow = cardiac output

    • Needs capacity to expand (highly elastic) with increasing CO

  • Volume

    • Contains ~10% circulating blood volume (~500mls)

    • Has capacity to expand (highly elastic, recruit additional capillaries)

  • Regional distribution of blood flow

    • Right lung receives 55% CO, left lung 45% CO

    • Flow distributed according to hydrostatic and alveolar pressure (west zones)

    • Hypoxic pulmonary vasoconstriction can redirect blood flow away from poorly ventilated regions

  • Regulation

    • Minimal capacity to self regulate (except for hypoxic vasoconstriction) with weak autonomic activity

    • Response to hypoxia: vasoconstriction

    • Response to hypercapnia: vasoconstriction

  • Functions

    • Main function is gas exchange: Absorbs O2, releases Co2

    • Other functions: filtration clots/debris, source of ACE, metabolism of PGs

Examiner comments

25% of candidates passed this question.

The examiners consider that an understanding of the pulmonary circulation is core area of the syllabus. In general, the anatomy section was better answered than the physiological features. As well as a description of the gross anatomy of the pulmonary circulation tracking it from the pulmonary valve to the left atrium, some mention of the microscopic anatomy was required (e.g., that the pulmonary arteries are thin walled with little smooth muscle).
For the second part of the question, a breadth of knowledge was required. Candidates were expected to address the following physiological features of the pulmonary circulation: volume, pressure, resistance, regulation and regional distribution and function. Marks were apportioned to each section, so it was important to write something on each section. Focussing on one section in detail (e.g., a very detailed description of West’s Zones) usually came at the expense of missing one or more of the other sections, most commonly the functions of the pulmonary circulation. Indeed, candidates that scored well provided information on each section and for the functions of the pulmonary circulation mentioned more than gas exchange.


Online resources for this question


Similar questions

  • Question 7, 2017 (1st sitting)



Question 14

Question

Describe the anatomy of the larynx.


Example answer

  • General anatomy
    • Located at the levels of C3- C6
    • Serves as a connection between the oropharynx and the trachea
    • Lined by pseudostratified columnar ciliated epithelium below cords, stratified square epithelium above
  • General functions
    • Respiration (conductive airway)
    • Swallowing and protection of airway from GI tract
    • Phonation
    • Cough reflex
  • Cartilages
    • 3 Paired: arytenoid, corniculate, cuneiform
    • 3 unpaired: thyroid, cricoid, epiglottis
  • Extrinsic muscles
    • Infrahyoid and suprahyoid muscles
    • move the larynx as a whole (elevates, depresses)
  • Intrinsic muscles
    • Move individual laryngeal components
    • Grouped into: adductors/abductors (e.g. cricoarytenoids, oblique arytenoids), tensors/relaxors (e.g. cricothyroid, thyroarytenoid) and the vocalis muscle (minute adjustments vocal cord)
  • Vocal ligament
    • Attaches to thyroid cartilage (ant) to arytenoid cartilage (post)
    • Opening forms the Rima Glottis
    • Produces phonation
  • relations
    • Skin/fascia (anterior)
    • Thyroid gland (anterior, lateral and inf.)
    • pharynx/oesophagus (posterior)
    • Carotid arteries (lateral), jugular veins (lateral)
    • Vagus and laryngeal nerves (lateral)
  • Innervation
    • Motor: All laryngeal muscles are supplied by the RLN except the cricothyroid which is supplied by the External branch of the superior laryngeal nerve
    • Sensory: internal branch of the superior laryngeal nerve (above cords), RLN (below cords)
  • Arterial supply
    • Upper half: Superior laryngeal artery (br. from the superior thyroid artery)
    • Lower half: Inferior laryngeal artery (br. of the inferior thyroid artery)
  • Venous drainage
    • Superior and inferior laryngeal veins which drain into respective thyroid veins
  • Lymphatics
    • Above the vocal cords: superior deep cervical LNs
    • Below the vocal cords: inferior deep cervical LNs


Examiner comments

40% of candidates passed this question.

For this question, candidates were expected to address the location of the larynx, its relations, the cartilages (single and paired), ligaments, muscles (intrinsic and extrinsic), innervation (sensory and muscular) and blood supply (including venous drainage). Marks were apportioned to each section, so whilst some detail was required, breadth of knowledge was also important. Most candidates had a grasp of the gross anatomy, the main relations and at least the innervation provided by the recurrent laryngeal nerve. However, an understanding of the functional anatomy of the cartilages was not always apparent. It should be noted that not every single muscle needed to be named (especially for the extrinsic muscles), but an understanding of the major muscle groups should have been included


Online resources for this question


Similar questions

  • Question 24, 2015 (1st sitting)



Question 15

Question

Compare and contrast the pharmacology of dobutamine and levosimendan.


Example answer

Name Dobutamine Levosimendan
Class Synthetic catecholamine (inodilator) Calcium sensitizer (Inodilator)
Indications Increase inotropy in cardiogenic shock, cardiac stress testing Increase inotropy in cardiogenic shock
Pharmaceutics Clear colourless solution (12.5mg/ml)

Diluted in water

Diluted in glucose, clear-yellow
Routes of administration IV IV, PO
Dose Infusion (0.5-20 ug/kg/min) Load, then infusion
pKA 10.4 6.3
Pharmacodynamics
MOA B1 and B2 agonist (B1>> B2) - Sensitises troponin C to calcium > increases contractility (without impairing relaxation)

- Activates ATP-sensitive K channels in smooth muscle > vasodilation

Effects CVS: increased inotropy, increased chronotropy, increased lusitropy, increased dromotropy, decreased SVR, increased BP, increased risk arrhythmias, increased myocardial oxygen requirement

RESP: bronchodilation,
CNS: Increased CBF
RENAL: Increased RBF

Increased chronotropy, increased inotropy, coronary vasodilation, decreased afterload, increased SV and CO, decreased SVR, decreased blood pressure and myocardial oxygen consumption, hypotension, arrhythmias, GIT upset, dizziness
Pharmacokinetics
Onset Immediate 1 hour
Absorption 0% oral bioavailability 85% oral bioavailability
Distribution Small Vd (0.2L/Kg)

Unknown protein binding

Small Vd (0.3L/kg)

99% protein bound

Metabolism Hepatic and tissue metabolism

COMT/MAO > inactive metabolites

By liver into inactive metabolites (95%) and active metabolites (5%)
Elimination Renal (70%) and faecal (20%) excretion of metabolites

T 1/2 = 2mins

Renal elimination of metabolites (active metabolites last as long as 80 hours)
Special points Does not require SAS approval Requires SAS approval in AUS


Examiner comments

41% of candidates passed this question.

The objective of this question was that candidates relay a detailed knowledge of both drugs with respect to their individual pharmacology highlighting the important clinical aspects of each drug (e.g., mechanism of action, metabolism, duration of effect). Then an integration of this knowledge was in the contrast section where the better candidates highlighted features of the drug that would influence when or why one may use it with respect to the second agent. Tabular answers of the pharmacology of each drug without any integration or comparison scored less well. A detailed knowledge of both agents was expected to score well.


Online resources for this question


Similar questions

  • Dobutamine
    • Question 14, 2011 (2nd sitting)
    • Question15, 2016 (2nd sitting)
  • Levosimendan
    • Question 8, 2012 (1st sitting)
    • Question 10, 2013 (2nd sitting)



Question 16

Question

Describe the formation of gastric acid (50% marks) and the regulation of gastric acid secretion (50% marks).


Example answer

Gastric acid

  • Gastric acid (HCl, pH 1.6) is one component of gastric secretions
  • Other components include: Gastrin, pepsinogen, IF, mucous
  • ~2L of gastric secretions produced per day
  • Gastric acid is important for innate immunity, pepsin activity, iron absorption etc.


Formation of gastric acid

  • Produced by the parietal cells in the stomach

  • Mechanism of HCl production

    • CO2 diffuses into parietal cells from blood

    • CO2 reacts with water to give H2CO3 (catalysed by CA)

    • H2CO3 dissociates into H+ and HCO3

    • At the basolateral membrane: HCO3 is exchanged for Cl (Cl in, HCO3 out)

    • Cl then passively diffuses down concentration gradient into secretory canaliculi

    • At the apical membrane: H-K ATPase pumps H+ into secretory canaliculi (against concentration gradient)

Stages of secretion

  • Cephalic
    • ~30% of gastric secretions as a result of this phase
    • Due to thought / taste / sight / smell of food
    • Leads to increased PSNS (vagal) activity
  • Gastric
    • ~60% of gastric secretions during this phase
    • Due to the mechanical stretch of the stomach by the food
    • Leads to increased PSNS activity and gastrin release
  • Intestinal
    • <10% of gastric secretions during this phase
    • Distention of small intestine --> release of secretin
    • Increased acid load in duodenum --> release of somatosatin


Regulation of gastric acid secretion

  • Histamine
    • Most important stimulus for gastric acid secretion
    • Synthesised and stored in neighbouring ECL cells
    • Binds to H2 receptors on parietal calls > HCl release
    • Stimuli: PSNS activity + gastrin
  • PSNS (vagal) activity
    • Vagal nerve stimulation of M3 receptors (Ach) on parietal cells > increased release HCl
    • Vagal stimulation of ECL cells > increased release histamine
  • Gastrin
    • Released from G cells
    • Indirectly leads to increased release of histamine from ECL cells
    • Activated by vagus, Inhibited by secretin
  • Somatostatin
    • Released from D cells
    • Inhibits gastrin
  • Secretin
    • Released from S cells
    • Inhibits gastrin


Examiner comments

26% of candidates passed this question.

The is question was divided into two sections offering equal marks. The first section required a description of the generation and transport of both H+ and Cl- into the stomach lumen by the parietal cell. The contributions of basolateral and luminal ion channels, the role of carbonic anhydrase and accurate description of the net flux was expected for full marks. The second section required comments on the roles of neural and endocrine regulation. Increased acid secretion via acetylcholine (via muscarinic M3), histamine (via H2) and gastrin were expected as was reduced secretion via secretin and somatostatin. Better responses were able to combine and integrate these into cephalic, gastric, and intestinal phases. The nature and function of other gastric secretions and the role of pharmacologic agents was not asked for and therefore not awarded any marks.


Online resources for this question


Similar questions

  • None



Question 17

Question

Describe the pharmacology of inhaled nitric oxide (NO).


Example answer

Name Nitric oxide
Class Inorganic gas / inhaled pulmonary vasodilator
Indications ARDS, Right heart failure, pHTN
Pharmaceutics Colourless gas (100ppm NO, 800ppm N2) in aluminium cylinders
Routes of administration Inhaled (via the inspiratory limb of an ETT)
Dose Typically 5-20ppm - titrated to minimal effective dose
Pharmacodynamics
MOA Stimulates cGMP > reduction in intracellular Ca > relaxation of SM.

As inhaled > selectively vasodilates well ventilated alveoli

Effects RESP: Inhibits HPV, improves V/Q matching,

CVS: decreased pulmonary vascular resistance,
CNS: Increased CBF

Side effects Methaemoglobinaemia

hypotension
Rebound pHTN following abrupt cessation
Thrombocytopaenia

Pharmacokinetics
Onset Seconds
Absorption Rapidly absorbed in pulmonary circulation due to high lipid solubility
Distribution Minimal systemic distribution
Metabolism Reacts with oxyHb to produce methaemaglobin and nitrates.

T 1/2 5 seconds

Elimination Metabolites (main metabolite = nitrate) are renally excreted
Special points


Examiner comments

24% of candidates passed this question.

Nitric Oxide (NO) is an inorganic colourless and odourless gas presented in cylinders containing 100/800 ppm of NO and nitrogen. Many candidates mentioned oxygen instead of nitrogen. The exposure of NO to oxygen is minimized to reduce formation of nitrogen dioxide and free radicals. Hence it is administered in inspiratory limb close to the endotracheal tube. Many candidates did not mention the contraindications/caution for NO use. Candidates generally did well in mentioning the impact on improving V/Q mismatch by promoting vasodilatation only in the ventilated alveoli and reducing RV afterload. Many candidates did not mention the extra cardio-respiratory effects. The expected adverse effects of NO were nitrogen dioxide related pulmonary toxicity, methemoglobinemia and rebound pulmonary hypertension on abrupt cessation. Pharmacokinetics of NO carried a significant proportion of marks. It was expected that the answers would involve mention of location of delivery of NO in inspiratory limb and reason behind it, the high lipid solubility and diffusion, the dose (5-20ppm), very short half-life of < 5 seconds and combination with oxyhemoglobin to produce methaemoglobin and nitrate. The main metabolite is nitrate which is excreted in urine.


Online resources for this question


Similar questions

  • Question 14, 2011 (1st sitting)



Question 18

Question

Define afterload (10% marks) and describe the physiological factors that may affect afterload on the left ventricle (90% marks).


Example answer

Afterload

  • Isolated muscle: The external force required to be generated before the myocardial sarcomere can begin to shorten in the isolated muscle.
  • Intact heart: the forces impeding ejection of blood from the ventricle during contraction


Change in afterload

  • Decreased afterload --> increased LV stroke volume --> increased CO
  • Increased afterload --> reduced LV stroke volume --> reduced CO


Factors effecting afterload

  • Broadly, the factors effecting afterload can be broken up into factors effecting
    • Mycocardial wall stress
    • Impedance to flow


MYOCARDIAL WALL STRESS (governed by the law of LaPlace)

  • Transmural pressure
    • Negative intrathoracic pressure > increased transmural pressure > increased afterload
    • e.g. inhalation (more pronounced in asthma)
  • Ventricular size
    • Increased radius of ventricle > increased wall stress > increased afterload
    • e.g. ventricular dilation
  • Myocardial wall thickness
    • Increased thickness > reduced wall stress (more sarcomeres share tension) > reduced afterload
    • e.g. LV hypertrophy


IMPEDENCE TO FLOW

  • Arterial compliance
    • Poorly compliance vessels > increased afterload
    • e.g. in pathology such as atherosclerosis
  • Arterial resistance/impedance
    • Related to (Hagen-Poiseuille equation)
      • the length of the arterial system (fixed)
      • blood viscosity (e.g. HCT, changes slowly)
      • Vessel radius (most important factor, changes readily)
    • E.g. profound vasoconstriction of capacitance vessels (e.g. norad infusion) > increased resistance > increased afterload
  • Outflow tract impedance
    • Leads to increased afterload (increased forced required for ejection)
    • E.g. valvular disease (AS), SAM, LVOT


Examiner comments

53% of candidates passed this question.

Afterload can be defined as factors resisting ventricular ejection and contributing to myocardial wall stress during systole. Most answers utilised the law of Laplace to expand upon factors affecting ventricular wall tension. Systemic vascular resistance was commonly mentioned, but less frequently defined. Aortic and left ventricular outflow tract impedance were commonly referred to. Effects of preload and neurohumoral stimuli were less well outlined. Description of factors affecting right ventricular afterload and depictions of left ventricular pressure volume loops earned no extra marks unless directly referenced to the question.


Online resources for this question


Similar questions

  • Question 7, 2009 (1st sitting)
  • Question 17, 2012 (1st sitting)
  • Question 19, 2014 (2nd sitting)



Question 19

Question

Explain how the kidney handles an acid load.


Example answer

Acid production

  • Normal biproducts of cellular function and metabolism
  • 'Fixed acids'
    • Body produces ~1mmol/kg/day
    • Fixed acids, except for lactate, are eliminated by the kidneys
  • 'volatie acids' (i.e. CO2)
    • Body produces ~15-20moles/day
    • Eliminated by the lungs


Mechanisms of acid-base regulation by kidneys


  1. Secretion of H+ / Reabsorption of HCO3
    • H+ activately secreted into the urine
      • Na-H exchanger (PCT, LOH)
      • H+ ATPase (DCT)
      • H-K ATPase (CD)
    • HCO3 is freely filtered at glomerulus (needs to be reabsorbed)
    • H+ and HCO3 combine to form H2CO3
    • H2CO3 converted to H2O and CO2 (by apical carbonic anhydrase)
    • H2O aand CO2 diffuse into cell and converted back to H2CO3 by CA
    • H2CO3 then dissociates into HCO3 and H+ (HCO3 reabsorbed, H+ is secreted once more)
    • This allows for all HCO3 to be reabsorbed


  1. Combination with titratable acids

    • Excess H+ combines with filtered buffers (e.g. phosphate, sulphate)

    • Phosphate is most important and is responsible for eliminating ~40% of excess fixed acid load / day

      • H+ combines with HPO4 > H2PO4 (ionised, not reabsorbed)

    • Minimal capacity to increase

  2. Ammonium mechanism

    • Excess H+ can bind to ammonia > excreted

      • in PCT/DCT: metabolism of glutamine > releases new HCO3 and excess NH4

      • In CD: secretion of NH3 binds to H+ > NH4 (ionised and cannot be reabsorbed)

    • Accounts for remainder of excess fixed acid load,

    • Has capacity to greatly expand when there is excess H+


Examiner comments

51% of candidates passed this question.

This question required candidates to understand the renal response to an acid load. It was expected that candidates would answer with regard to recycling of bicarbonate in the proximal tubule, excretion of titratable acid via the phosphate buffer system and generation of ammonium and its role in acid secretion. Many candidates had a good understanding of the bicarbonate system but used this to explain the secretion of new acid.


Online resources for this question


Similar questions

  • Question 12, 2014 (2nd sitting)



Question 20

Question

Describe the pharmacology of intravenous sodium nitroprusside.


Example answer

Name Sodium nitroprusside
Class Nitrate vasodilator
Indications Hypertensive emergencies (or need for strict BP control)
Pharmaceutics IV solution (50mg/2mL)

Light sensitive

Routes of administration IV only
Dose Titrated to effect (0-2mcg/kg/min)
pKA 3.3
Pharmacodynamics
MOA Prodrug

Diffuses into RBCs and reacts with Oxy-Hb to produce NO
NO diffuses into cell > incr cGMP > decreased Ca > SM relaxation

Effects CVS: decreased BP, afterload

RESP: impairs HPVC
CNS: cerebral vasodilation
GI: ileus
metabolic: acidosis

Side effects headache, hypotension, rebound hypertension (abrupt withdrawal), cyanide toxicity (high doses), metabolic acidosis, hypoxia, raised ICP
Pharmacokinetics
Onset/offset Immediate onset + offset
Absorption 0% oral bioavailability
Distribution VOD 0.25L/Kg (confined intravasc).

Nil protein binding

Metabolism Nitroprusside > cyanide > prussic acid > thiocyanate
Elimination Metabolites via urine
Special points


Examiner comments

49% of candidates passed this question.

This was a straightforward pharmacology question relating to a relatively common and archetypal intensive care medication. The structure of the question was well handled by most of the candidates; easily falling into the classic pharmaceutics, pharmacokinetic and pharmacodynamics framework. Many candidates had a superficial knowledge of the presentation and formulation of the drug, aside from its light sensitivity. Better answers detailed the drug according to the above-mentioned framework but also accurately highlighted specific points relevant to the ICU practise such as the metabolic handling of sodium nitroprusside and relating this to the consequences of the various metabolic products.


Online resources for this question


Similar questions

  • Question 11, 2015 (1st sitting)




2020 (1st sitting)

Question 1

Question

Describe the carriage of carbon dioxide in blood.


Example answer

Overview

  • CO2 is constantly produced as a by-product of metabolism and needs to be cleared
  • CO2 content of blood
    • Mixed venous: 52mls/100mls blood, at PaCO2 of ~45mmHg
    • Arterial: 48mls/100mls blood, at PaCO2 of ~40mmHg
  • CO2 is transported in three main forms in the blood
    • Dissolved
    • As bicarbonate
    • In combination with proteins (carbamino compounds)


Dissolved CO2

  • Accounts for
    • ~5% of the total carbon dioxide in the blood
    • ~10% of the CO2 evolved by the lung
  • The amount dissolved is proportional to the partial pressure (Henry's Law)
  • 20x more soluble than O2, so dissolved CO2 plays a more significant role in transport


Bicarbonate

  • Accounts for

    • ~90% of the carbon dioxide in the blood

    • ~60% of the CO2 evolved by the lung

  • Bicarbonate is formed by the following sequence

    <math display="block">CO_2 + H_{2}O \leftrightarrow H_{2}CO_{3} \leftrightarrow H^+ + HCO_3^-</math>
  • Process

    • CO2 dissolves into RBC and leads to H+ and HCO3 (per above equation)

    • HCO3 moves into plasma, H+ binds to reduced (deoxy) Hb

      • KHb + H+ <-> HHb + K+

    • Cl moves into the cell to maintain electroneutrality (chloride shift)

    • When Hb is oxygenated in the lungs, H+ dissociates and converted back to CO2 by the above equation and is exhaled

    • Haldane effect accounts for the increased capacity of Hb to carry CO2 when poorly oxygenated


Carbamino compounds

  • Accounts for
    • ~5% of the CO2 in the blood
    • ~30% of the CO2 evolved by the lung
  • Formed by the combination of CO2 with terminal amine groups in blood proteins
    • NH2 + CO2 <-> NHCOO- + H+
  • Haemoglobin is the most abundant protein and has most imadazole side chains (greatest carrier capacity)
    • The reaction occurs faster with deoxHb than oxy-Hb (Haldane effect)


Examiner comments

68% of candidates passed this question.

A detailed understanding of the carriage of carbon dioxide (CO2) in the blood is essential to the
practice of intensive care medicine. Comprehensive answers classified and quantified the
mechanisms of CO2 carriage in the blood and highlighted the differences between the arterial and
venous systems. An explanation of the physiological principles surrounding these differences and
the factors which may affect them was expected. The changes that occur at the alveolar and
peripheral tissue interfaces with a similar explanation of process was also required. Candidate
answers were often at the depth of knowledge required for an ‘outline question’ and a more
detailed explanation was required to score well.


Online resources for this question


Similar questions

  • Question 6, 2012 (1st sitting)
  • Question 1, 2012 (2nd sitting)
  • Question 13, 2015 (1st sitting)
  • Question 5, 2018 (2nd sitting)
  • Question 1, 2020 (1st sitting)


Question 2

Question

Describe the pharmacology of glyceryl trinitrate (GTN)


Example answer

Name Glyceryl trinitrate (GTN)
Class Organic nitrate
Indications Hypertension, acute pulmonary oedema, angina, ACS/LV failure,
Pharmaceutics Clear liquid (IV), Patch (transdermal), tablet (SL), spray (SL)
Routes of administration Sublingual, intravenous, transdermal, PO
Dose Patch: 5/15mg/24hr SL: 400mcg PRN IV: titrated to effect
pKA 5.6
Pharmacodynamics
MOA Prodrug, which is dinitrated to produce active nitric oxide (NO). NO diffuses into smooth muscle cell > binds to guanylyl cyclase > increased cGMP > decreased intracellular Ca > smooth muscle relaxation > vasodilation
Effects CVS: systemic vasodilation (preferentially venodilation, coronary arterial dilation) > decreased SVR > decreased BP + VR, decreased myocardial O2 consumption (decreased VR > decreased preload), reflex tachycardia CNS: Increased CBF > inc ICP, headache RESP: Bronchodilation (weak), decreased PVR OTHER: flushing, methaemaglobinaemia
Pharmacokinetics
Onset 1-3 mins (SL), <1 min (IV), Patch variable.
Absorption Oral bioavailability <5% (hepatic high first pass effect) Sublingual spray 40% Sublingual tablet 60%
Distribution 60% protein bound. Vd 3L/kg
Metabolism Hydrolysis Site: liver + RBC cell wall + vascular cell walls. Active metabolites
Elimination Renal T 1/2 B = 5 minutes (parent compound).
Special points Can develop tachyphylaxis (depletion of sulfhydryl groups)


Examiner comments

69% of candidates passed this question.

GTN is a commonly used ‘level 1’ drug. The most comprehensive answers included information
on available drug preparations, indications, mechanism of action, pharmacodynamics and
pharmacokinetics and its side-effect profile. It was expected that significant detail be included in
the pharmacodynamic section (e.g. preferential venodilation, reflex tachycardia, effects on
myocardial oxygen demand etc). Common omissions included tachyphylaxis, dosing and its
metabolism. Many answers didn’t mention the first pass effect.


Online resources for this question


Similar questions

  • Question 15, 2008 (1st sitting)
  • Question 20, 2008 (2nd sitting)
  • Question 16, 2010 (1st sitting)
  • Question 2, 2016 (2nd sitting)



Question 3

Question

Outline the potential adverse consequences of blood transfusion.


Example answer

Immunological (acute < 24 hours)

Adverse event Incidence Mechanism
Acute haemolytic transfusion reaction 1 : 75,000

(Death 1:2 mil)

E.g. ABO incompatibility. Immunological destruction of transfused cells (Type II hypersensitivity).
Febrile non-haemolytic transfusion reaction ~1% Cytokine release from stored cells causing a mild inflammatory reaction with recipient alloantibodies
Mild allergic reactions (e.g. urticaria) ~1-5% Hypersensitivity to plasma proteins in the transfused unit
Severe allergic reactions (i.e. anaphylaxis) ~ 1 : 50,000 Type I hypersensitivity reaction to plasma protein in transfused unit
Transfusion related acute lung injury (TRALI) Variable Donor plasma HLA activates recipient pulmonary neutrophils, causing fever, shock, and non-cardiogenic pulmonary oedema


Immunological (delayed > 24 hours)

Adverse event Incidence Mechanism
Delayed haemolytic transfusion reaction 1: 5,000 Alloimmunized to minor RBC antigens (kidd, duffy, Kell) during previous transfusions which is not detected due to low levels in pre-transfusion screening. Reaction if re-exposed
Transfusion associated graft versus host disease Rare Transfused lymphocytes recognise host HLA as positive causing marrow aplasia (rare now with leukodepletion)
Alloimmunisation ~1-10% Previous sensitisation leading to antibody production on re-exposure.


Non-immunological (acute < 24 hours)

Adverse event Incidence Mechanism
Non immune mediated haemolysis Rare Due to physicochemical damage to RBCs
Transfusion transmitted bacterial infections 1:250,000 - 2.5 million Contamination during collection or processing. Most common organisms are those which use iron as a nutrient and reproduce at low temperatures, e.g. Yersinia Pestis.
Transfusion associated circulatory overload 1% Rapid increase in intravascular volume in patients with poor circulatory compliance or chronic anaemia. May result in pulmonary oedema and be confused with TRALI.
Others Variable Hypothermia, coagulopathies, electrolytes disturbance, metabolic derangements


Non-immunological (delayed > 24 hours)

Adverse event Incidence Mechanism
Iron overload Rare (unless massive transfusion e.g. >20 units) Each unit of PRBC contains ~250mg of iron, whilst average excretion is 1mg.day-1.
Infections Less than 1:1 million From donor


Examiner comments

43% of candidates passed this question.

As only an outline was asked for, a brief statement about each complication was sufficient. Better answers were structured using a classification of: Acute Immunological, Acute Non- Immunological, Delayed Immunological and Delayed Non-immunological. Examples of expected detail would include the following: E.g. Bacterial infection – a statement outlining the incidence of bacterial infection, a common causative organism or why bacterial infections are more commonly associated with platelet transfusions than red cells would have scored the marks allocated to ‘bacterial infection’. E.g. Acute Haemolytic Transfusion Reaction – a statement about red cells being destroyed due to incompatibility of antigen on transfused cells with antibody of the recipient and an approximate incidence scored the marks allocated to AHTR. An excellent resource is the Australian Red Cross transfusion website as listed in the suggested reading section of the syllabus.


Online resources for this question


Similar questions

  • Question 13, 2013 (2nd sitting)

  • Question 16, 2017 (1st sitting)


Question 4

Question

Explain the counter-current mechanism in the kidney.


Example answer

Purpose

  • The counter current mechanism of the kidney is important for establishing the osmotic gradient necessary for forming concentrated urine (thus preserving water)


Formation

  • Formed by the loop of henle
  • Assuming a naïve system. Iso-osmolar fluid (300mosm) arrives at the aLOH.
    • This is because water/solutes are absorbed in equal amounts in the PCT.
  • In the aLOH the Na-K-2Cl transporter reabsorbs these ions. Water is impermeable. Interstitium becomes hyperosmotic (now 400mosm)
  • When the next iso-osmotic fluid arrives (300sm), there is a concentration gradient (water leaves through permeable dLOH) leading to hypertonic filtrate
  • The hypertonic filtrate then arrives in the aLOH and the Na-K-2Cl pump works again.
  • The interititum becomes further hypertonic (e.g. 400mosm).
  • Process repeats until a maximal concentration gradient of around 600mosm exists between inner medulla and cortex
  • Finally, through urea trapping from the collecting ducts (facilitated diffusion via UTA1 and UTA3 receptors), the osmotic gradient in the inner medulla is increased to approximately 1200mosm


Maintenance

  • Vasa recta is organised in such a way that it does not wash away the established concentration gradient (close proximity, in parallel, opposite direction of flow)
  • Achieves this by also looping down into the inner medulla (water lost, solute gained) then back up (water gained, solute lost)
  • The slow nature of this flow in combination with its anatomy (parallel + close proximity) prevents the washing away of the concentration gradient
  • This is known as the counter current exchanger


Examiner comments

63% of candidates passed this question.

Higher scoring candidates described the counter-current multiplier mechanism, the countercurrent
exchanger and the contribution of urea cycling to the medullary osmotic gradient. Detailing
the mechanisms as to how they may be established, maintained and or regulated. Descriptions
of the multiplier (LOH) alone did not constitute a passing score. Values for osmolality at the cortex
& medulla and within the different parts of the LOH was required. A description of the countercurrent
exchanger system where inflow runs parallel to, counter to and in close proximity to the
outflow was expected. This could have been achieved by describing the anatomical layout of the
loop of Henle and the vasa recta.


Online resources for this question


Similar questions

  • Question 9, 2011 (1st sitting)
  • Question 22, 2015 (2nd sitting)



Question 5

Question

Outline the mechanisms of antimicrobial resistance (50% of marks). Briefly outline the pharmacology of ciprofloxacin (50% of marks).


Example answer

Antimicrobial resistance

  • Occurs when the maximal level of drug tolerated in insufficient to inhibit growth
  • Broadly occurs via genetic alteration or changes to protein expression


Mechanisms of resistance

  1. Prevent access to drug target
    • Decrease permeability
      • E.g. pseudomonas resistance to carbapenems due to reduction in porins
    • Active efflux of drug
      • Efflux pumps > extrude antibiotics e.g. fluoroquinolone resistance
  2. Alter antibiotic target site
    • e.g. VRE - Alteration to Peptidoglycan binding site protein, reducing affinity of drug.
  3. Modification / inactivation of drug
    • E.g. ESBL and penicillins/cephalosporins whereby b-lactamases hydrolyse B-lactam rings
  4. Modification of metabolic pathways
    • E.g. Bactrim resistance. Metabolic pathways bypass site of antibiotic action


Name Ciprofloxacin
Class Quinolone
Indications Prostatitis, complicated UTIs, bone/joint infections
Pharmaceutics Oral tablet, light-yellow power for injection (water diluent)
Routes of administration IV, PO
Dose 250-750mg BD (PO), 200-400mg BD/TDS (IV)
Pharmacodynamics
MOA Bactericidal; inhibit bacterial DNA synthesis by blocking DNA gyrase and topoisomerase IV.
Spectrum Broad spectrum (GN+ MSSA). Effective against pseudomonas + anthrax (lol)

Effective against some atypicals (legionella).
No anaerobe cover.

Side effects GIT: nausea, vomiting

CNS: dizziness, headache
CVS: prolonged QT interval, arrhythmias
MSK: Myopathy, tendonitis + rupture, arthropathy

Pharmacokinetics
Onset 1-2 hours (PO) for peak effect. Immediately (IV)
Absorption Oral bioavailability 70%
Distribution Vd 2.5L/kg. Protein binding 25%, good tissue penetration (except for poor CSF penetration)
Metabolism Partially hepatic
Elimination Renal excretion of metabolites. T1/2 3-5 hours.
Special points Increasing world wide resistance to quinolones.


Examiner comments

71% of candidates passed this question.

Most candidates had a structured answer to mechanisms of resistance that covered the major categories (alter target protein, prevent entry, efflux, degrade drug) and provided an example of a bacteria and the affected antibiotic, as was required to answer the question in full. Ciprofloxacin, whilst perhaps not a first line drug in the ICU, was not well known by many candidates. Better answers included a brief outline of class, mechanism of action (action on DNA gyrase to inhibit replication), spectrum (Gram negatives particularly mentioning Pseudomonas, lesser Gram positive cover, not anaerobes, some atypical), PK (with correct dose, wide penetration into tissues including bone/prostate etc., predominantly renal excretion), side effects/toxicity (common or specific to cipro e.g. QT, tendinitis, arthropathy) and an example of resistance.


Online resources for this question


Similar questions

  • Question 9, 2019 (1st sitting)



Question 6

Question

Outline how the respiratory system of a neonate differs from that of an adult.


Example answer

Anatomical differences in neonates (with respect to adults)

  • Head
    • Larger head and occiput
    • Smaller mandible
    • Larger tongue, tonsils, adenoids.
  • Neck
    • Shorter neck with higher laryngeal position
    • Larger, less rigid epiglottis
  • Larynx/trachea/bronchi
    • Shorter, narrower and softer trachea
    • Main Bronchi angle equal L-R for neonate (adults right is more vertical)
    • Reduced bronchial smooth muscle > less bronchospasm (but bronchodilators less effective)
  • Alveoli
    • Fewer (and more immature) alveoli
  • Reduced type 1 muscle fibres in diaphragm (25% neonates, 55% in adults) - more susceptible to fatigue


Physiological differences in neonates (with respect to adults)

  • Volumes/capacities
    • Similar FRC (30ml/kg), and TV (7mls/kg) to adults
    • Lower vital capacity (45mls/kg) and TLC (65mls/kg)
    • Increased minute ventilation (generated by increased RR, as TV proportionally similar)
    • Increased physiological deadspace (3mls vs 2 mls)
    • Increased closing capacity > increased shunt
  • Compliance
    • Increased chest wall compliance (proportionally more cartilage)
    • Decreased lung compliance (less surfactant)
  • Resistance
    • Respiratory resistance is increased at birth: bronchi are smaller and lung volumes are smaller
  • Mechanics
    • Obligate nose breathers
    • More susceptible to fatigue (Reduced type 1 muscle fibres in diaphragm)
    • Increased work of breathing overall (increased MV, increased dead space, increased shunt)
  • Gas exchange
    • Increased oxygen consumption (6mls/kg/min)
    • Increased shunt (10-25%, due to patent ductus arteriosus)
    • Foetal haemoglobin = increased oxygen affinity (left shift oxy curve)
  • Control
    • Immature respiratory centre - decreased response to hypercapnia, periodic apnoea,


Examiner comments

20% of candidates passed this question.

This question required an outline of the anatomical, mechanical and functional differences. It was expected that factors leading to an increased work of breathing and oxygen cost would be mentioned. The mechanics of expiration were not often included in candidates’ answers. Immaturity of the alveoli and peripheral chemoreceptors were common omissions. Inaccuracies regarding upper airway anatomy and compliance of the chest wall cost some candidates marks. The question did not call for an explanation of the relative difficulty of intubation. Discussion of pathophysiology due to airway obstruction, causes of central apnoea or sensitivity to drugs was not required. Many answers included inaccurate information. Points which were often missed were difference in bronchial angles, number of alveoli, number of type 1 fibres in diaphragm, ciliary function and peripheral chemoreceptors.


Online resources for this question


Similar questions

  • Question 7, 2017 (1st sitting)



Question 7

Question

Describe the physiological control of systemic vascular resistance (SVR).


Example answer

Overview

  • SVR is the impediment to flow generated by the systemic vasculature (excluding pulmonary) and can be defined according to ohms law (SVR = (MAP-CVP) / CO)
  • The main determinants of SVR is conceptualised by the Hagen-Poiseuille equation
    • <math display="inline">SVR = { 8l. \eta \over \pi r^4}</math>
    • Length (l), and viscosity of blood (n) does not readily change, hence the most significant determinant of SVR is the vessel radius (R)
  • Control of radius
    • Majority of this control occurs at the level of the arteriole (sig. amount of smooth muscle in wall - can readily alter calibre)


Systemic control of vessel radius

  • SNS
    • Activation of the SNS (pain, emotion, exercise, fear etc) > release of NA from the post ganglionic neurons > activates alpha-1 receptors > vasoconstriction > increased SVR
    • Alpha-1 receptors are plentiful in the skin, kidneys, GIT (but minimal in the heart and brain, leading to preferential flow to these organs)
  • PSNS
    • Much less important (external genitalia)
    • Activation leads to vasodilation (decreased SVR)
  • Arterial baroreflex control
    • Increased BP > increased arterial wall stretch > increased firing of aortic and carotid sinus baroreceptors > decreased sympathetic tone > vasodilation > decreased SVR (vice versa)
  • Chemoreceptor reflex
    • Peripheral and central chemoreceptors activated by hypoxia > increased SVR
  • Hormonal control
    • Numerous endocrine mediators affect SVR
    • E.g. Angiotensin (AT1 receptors) and vasopressin (V1 receptors) increase SVR
  • Temperature
    • Heat causes vasodilation > decreased SVR (and vice versa)


Regional/local control of vessel radius

  • Myogenic autoregulation
    • e.g. brain, kidneys, can alter vessel radius via myogenic means. Less important peripherally.
    • Incr. pressure > incr. stretch > release of vasoactive mediators > constriction > increased SVR
  • Metabolic autoregulation
    • e.g. brain, coronary vasculature
    • Decreased oxygen delivery / increased utilisation > increased metabolites (CO2, H, lactate, NO, adenosine) > vasodilation > decreased SVR


Examiner comments

21% of candidates passed this question.

This question invited a detailed discussion of the physiological control mechanisms in health, not
pathophysiology nor drug-mediated effects. The central and reflex control mechanisms that regulate SVR over time are distinct from the local determinants of SVR. There was often confusion between dependent and independent variables. Cardiac output is generally depended upon SVR, not vice versa, even though SVR can be mathematically calculated from CO and driving pressures. The question asked about systemic vascular resistance and did not require a discussion of individual organs except for a general understanding that local autoregulation versus central neurogenic control predominates in different tissues. Emotional state, temperature, pain and pulmonary reflexes were frequently omitted. Peripheral and central chemoreceptors and low-pressure baroreceptors were relevant to include along with high pressure baroreceptors.


Online resources for this question


Similar questions

  • None so far as I can tell..



Question 8

Question

Describe the production, metabolism and role of lactate.


Example answer

Production

  • Lactate is a product of anaerobic metabolism
    • Glucose is converted to pyruvate via glycolysis
      • Aerobic metabolism: Pyruvate is converted to Acetyl Coa and enters the TCA cycle > oxidative phosphorylation (38 ATP per glucose)
      • Anaerobic metabolism: pyruvate is unable to be converted to Acetyl CoA and enter the TCA cycle. Instead it is converted into lactate (producing 2 ATP and regenerates NAD+ to allow glycolysis to continue)
  • Lactate is produced mainly in skin, muscle, RBCs, brain, intestines
  • Normal plasma levels are ~0.5-2mmols
  • Increased lactate (>2 mmols) may be due to numerous causes
    • Physiological causes: E.g. exercise
    • Hypoxaemia: e.g. Shock, anaemia, CO poisoning, hypoxia
    • Disease: e.g. Sepsis, liver failure, thiamine def.
    • Drugs/toxins: e.g. adrenaline, salbutamol, ethanol, biguanides, cyanide
    • Congenital errors in metabolism: e.g. G6PD deficiency


Metabolism/fate

  • Lactate produced intracellulary diffuses out of the cell
  • Majority (80%) of circulating lactate is then metabolised in the liver via the cori-cycle
    • Lactate is converted back to glucose via gluconeogenesis (consumes 6ATP), and can undergo glycolysis again
  • Lactate can also be used as a fuel source, for example in the heart


Role

  • Lactate sink
    • Allows a period of ongoing ATP production from glycolysis during periods of hypoxia, TCA inhibition, pyruvate accumulation
  • Lactate shuttle hypothesis
    • Lactate is produced under aerobic+anaerobic conditions and may shuttle intra-cellularly and inter-cellularly to be used as sources of energy via gluconeogenesis
  • Signalling molecule
    • Emerging evidence that lactate
      • alters gene expression
      • may be involved in redox signalling
      • Mediate control of lipolysis


Examiner comments

16% of candidates passed this question.

Better answers used the categorisation in the question as a structure for their answer. Many candidates gave a good description of lactate production from glycolysis, increasing with accumulation of NADH and pyruvate, when these are unable to enter Krebs cycle. There were however, many vague and incorrect descriptions as to what lactate is and its physiological role. Many candidates suggested that its presence is abnormal or pathological. Most answers demonstrated a superficial understanding and physiological detail of lactate’s role as an energy currency in times of oxygen debt. Higher scoring candidates often mentioned non-hypoxic causes of pyruvate accumulation which include; circulating catecholamines, exercise, sepsis or lack or mitochondria (RBCs). Mention of the relative ATP production of the two fates of pyruvate was also noted in more complete answers. The Cori cycle was generally superficially described. A key role of lactate is the ‘lactate sink’, allowing a period of ongoing ATP production from glycolysis when cells become oxygen deplete or the Kreb’s cycle is inhibited; few candidates detailed or highlighted this.


Online resources for this question


Similar questions

  • Question 5, 2010 (1st sitting)
  • Question 6, 2015 (2nd sitting)



Question 9

Question

Outline the changes to drug pharmacokinetics and pharmacodynamics that occur at term in pregnancy.


Example answer

Pharmakokinetics

  • Absorption
    • Oral
      • Nausea and vomiting in early preg > reduced PO absorption
      • Increased intestinal blood flow (due to increased CO) > increased PO absorption
      • Decreased gastric acid production > increased pH > unionised drugs absorbed more
      • Delayed gastric emptying peri-labour may increase/decrease absorption depending on drug
    • IM / SC / Transdermal
      • Increased absorption due to increased CO + increased skin/muscle blood flow
    • IV
      • Faster IV onset due to increased CO
    • Neuraxial
      • Decreased peridural space > decreased dose required
  • Distribution
    • Volume of distribution
      • Increased total body water > increased Vd for hydrophilic drugs
      • Increased body fat > increased Vd for lipophilic drugs
    • Plasma proteins
      • Decreased protein binding (increased free fraction) due to reduced concentrations albumin and a-1 glycoprotein
  • Metabolism
    • Liver
      • Some metabolic enzymes reduced / some increased (due to progesterone/oestrogen ratio)
      • Leads to variable drug responses
        • E.g. increased metabolism of midazolam, phenytoin, but decreased caffeine.
    • Placenta metabolises some drugs (?sig of effect)
    • Decreased plasma cholinesterase (though no change in Succinylcholine effect)
  • Elimination
    • Renal
      • Increased clearance due to increased GFR (e.g. cefazolin)
    • Hepatobiliary
      • Decreased clearance due to cholestatic effects of oestrogen (e.g. rifampacin)
    • Resp
      • Increased volatile washout due to increased minute ventilation


Pharmacodynamics

  • Increased sensitivity to volatile anaesthetics (decreased MAC)
  • Increased sensitivity to IV anaesthetics
  • Increased sensitivity to local anaesthetics
  • Changed therapeutic indices due to risk of teratogenicity / fetal damage


Examiner comments

7% of candidates passed this question.

Answers framed around absorption, distribution, metabolism and excretion performed better. Some brief comments on physiology are required as the basis for pharmacokinetic change, but discussion of physiology that was not then specifically related to pharmacology did not score marks. Specific ‘real life’ examples necessitating change in practice or prescribing were well regarded e.g. reduction in spinal/epidural local anaesthetic dosing. Vague statements about possible or theoretical changes were less well regarded.


Online resources for this question


Similar questions

  • Question 11, 2011 (1st sitting)
  • Question 16, 2016 (2nd sitting)


Question 10

Question

Compare and contrast the pharmacology of noradrenaline and vasopressin.


Example answer

Name Noradrenaline Argipressin
Class Endogenous catecholamine Endogenous nonapeptide
Indications Vasopressor (Hypotension/shock) Platelet dysfunction (vWD), hypotension/shock (catecholamine sparing), CNS diabetes insipidus
Pharmaceutics Clear solution. 1:1000. Brown ampule (prevent light oxidation). Diluted in dextrose. Clear colourless solution
Routes of administration IV only (central vein) IV infusion (central vein) for vasopressor support. Given IN/SC for other indications.
Dose Infusion titrated to effect (generally 0 - 0.5 mcg/kg/min) 2.4 units/hr (for vasopressor support)

- At lower doses has predominant V1 activity, V2 activity at higher doses

pKA
Pharmacodynamics
MOA Predominately Alpha 1 agonism. Some beta adrenergic receptor agonism. a1 > B1 > B2 Physiologically secreted by PVN of hypothalamus > stored in posterior pituitary > secreted in response to hypovolaemia + increased osmolality

→ V1 receptor (blood vessels) agonism > Vasoconstriction > increased SVR > increased BP
→ V2 receptor (collecting ducts of nephrons) agonism > increased water reabsorption > increased BP
→ V2 receptor (endothelial cells) agonism > increased vWF release and Factor VIII activity

Effects CVS: peripheral vasoconstriction > increased SVR > inc. BP, reflex bradycardia, increased afterload (from SVR) > decreased CO (minor), increased myocardial O2 consumption, no sig. change in dromotropy, lusitropy or inotropy

CNS: decreased CBF (depending on BP), headache
RESP: increased PVR, bronchodilation
GIT/RENAL/uterine: vasoconstriction > decreased BF
MSK: extravasation > necrosis

CVS: ACS, angina, arrhythmias

HAEM: Excessive platelet aggregation / thrombosis
RENAL: Hyponatraemia (increased water reabsorption > Na reabsorption)
GIT: abdominal pain , nausea, vomiting
DERM: Ischaemia from vasoconstriction
Allergic reactions (bronchospasm, urticarial rash, anaphylaxis)

Pharmacokinetics
Onset Immediate Fast (not as fast as noradrenaline)
Absorption IV only (0% oral bioavailability) IV only (0% oral bioavailability)
Distribution Does not cross BBB.

Vd = 0.1L/kg
Protein binding = 25%

20% protein bound, Vd 0.2L/Kg
Metabolism Readily metabolised by MAO and COMT into inactive metabolites (VMA, normetadrenaline). 25% taken up in lungs. Extensive hepatic and renal metabolism by serine proteases and oxido-reductase enzymes > inactive metabolites
Elimination Excreted in urine as inactive metabolites (>85%).

Half life ~2 mins

Renal elimination

T 1/2 <10 minutes

Special points Tachyphylaxis (slow)

Effect exaggerated in patients taking MAOI (less breakdown)


Examiner comments

49% of candidates passed this question.

These are both level 1 drugs regularly used in intensive care. Significant depth and detail of each
drug were expected. Overall knowledge was deemed to be superficial and lacked integration.
Better answers identified key points of difference and overlap in areas such as structure, pharmaceutics, pharmacokinetics, pharmacodynamics, mechanism of action, adverse effects
and contraindications. A tabular list of individual drug pharmacological properties alongside each
other did not score as well as answers which highlighted key areas of difference and similarities.


Online resources for this question


Similar questions

  • None directly comparing these. Some a while back on norad or vaso individually


Question 11

Question

Describe the structure and function of adult haemoglobin


Example answer

Haemoglobin

  • Metalloprotein found within erythrocytes (RBCs)
  • 200-300 million molecules of Hb within each RBC


Structure

  • Hb molecules are tetramer's consisting of four globular protein subunits
    • Majority of adult blood contains 2x alpha and 2x beta globular subunits (HbA)
    • Various other forms of Hb: HbA2 (adult), HbF (fetal), HbS (Sickle cell disease), etc.
    • The make up of these subunits effects their capacity to bind + transport O2
  • Each globular subunit is attached to one Haem group
  • Each haem group contains:
    • A protoporphyrin ring
    • A central ion molecule in ferrous state (Fe2+)


Function

  • Oxygen transport

    • Reversibly binds to oxygen and transports it around the body in the blood

    • One haem group can bind one O2 molecule (each Hb molecule binds four O2 molecules), exhibits positive cooperativity.

    • Amount of binding is related to PAO2 (98% at 100mmHg, 75% at 40mmhg)

    • 98% of oxygen in the blood is carried by Hb

  • Carbon dioxide transport

    • Reversibly binds to carbon dioxide to transport it away from the tissues to the lungs

    • Hb contributes to the CO2 transport by 2 mechanisms

      • By directly forming carbo-amino compounds (30% CO2 evolved from lung)

      • As a proton acceptor for the RBC bicarbonate transport system (60% of CO2 evolved from lung)

  • Buffer

    • Haemoglobin is the primary protein buffering system in the blood

    • It exists as a weak acid (HHb) and Base (KHb)

    • Buffers by binding excess H+ ions to the imidazole side chains of the histidine residues

  • Nitric oxide regulation

    • Hb is important in regulating NO function

    • Hb readily binds NO and can inactivate or transport it, thus regulating its activity.

Examiner comments

57% of candidates passed this question.

Marks were awarded for the two components of this question – structure and function. The
structure component was often only briefly described with a cursory overview provided; however,
this component contributed around half of the available marks. Many candidates were unable to
accurately describe the structural components of the haemoglobin molecule. The functional
component was handled better – however much time was wasted with detailed drawings of the
oxyhemoglobin curve (not many marks awarded for this). The basic function of haemoglobin
carriage of oxygen and carbon dioxide was known, but detail was often missing about its role as
a buffer or its role in the metabolism of nitric oxide.


Online resources for this question


Similar questions

  • Question 11, 2014 (1st sitting)

  • Question 14, 2016 (2nd sitting)


Question 12

Question

Explain resonance and its significance and the effects of damping on invasive arterial blood pressure measurement.


Example answer

Resonance

  • Increase in the oscillations of a vibrating system when energy is applied to the system in harmonic proportions to the natural frequency of the system


Natural frequency

  • Frequency at which a system oscillates when not subjected to repeated/continuous external forces, in the absence of damping


Resonance and IABP

  • An arterial waveform is the composite of many wave forms of increasing frequencies (harmonics)

  • At least 8 harmonics must be analysed to have sufficient resolution in the waveform

  • We do not want the arterial line system to oscillate at a frequency close to the heart rate

  • Commonly measured HR ranges 30 to 180/min = 0.5Hz to 3Hz

  • To minimise effects of resonance, the natural system of our arterial system must therefore be 8 harmonics above the frequency we are measuring (3Hz)

  • If 3Hz is the fastest HR we are measuring then 8x3 = 24Hz. Thus our system must be >24Hz

  • To increase the natural frequency of the arterial line system we can use a short, wide, stiff catheter with no bubbles in tube

Damping

  • Loss of energy in the system, which gradually reduces amplitude of oscillations
  • Dampening is used to prevent large amplitude changes due to resonance when the natural frequency of the system is close to the transducers natural frequency
  • There is an optimal level of damping (damping coefficient 0.64)which maximises frequency responsiveness
  • Degree of damping can be assessed using the square wave (fast flush) test


Overdamped (coefficient >0.7)

  • Falsely low SBP
  • Falsely high DBP
  • Loss of fine waveforms
  • MAP remains fairly accurate


Underdamped (coefficient <0.6)

  • Falsely high SBP
  • Falsely low DBP
  • MAP remains fairly accurate


Examiner comments

23% of candidates passed this question.

Many candidates gave detailed answers that involved the set up and components of the arterial
line system that was not asked for in the question and did not attract marks. There was confusion
around the correct use of the terms natural frequency, resonance frequency and harmonics –
candidates that were able to describe these frequencies correctly went on to achieve a good mark
– the graphs and discussion around optimal dampening, over and underdamped traces were
often drawn poorly or without sufficient detail, and at times were not used within in the context of
the answer. Descriptions of the clinical effect seen with over / under dampened traces on blood
pressure was well described.


Online resources for this question

  • Intensive Blog

  • Deranged Physiology and Deranged Physiology

  • [chrome-extension://efaidnbmnnnibpcajpcglclefindmkaj/viewer.html?pdfurl=http%3A%2F%2Fwww.anaesthesia.uct.ac.za%2Fsites%2Fdefault%2Ffiles%2Fimage_tool%2Fimages%2F93%2F05-Arterial%2520Transducers%2520and%2520Damping%2520%2528G%2520Davies%2529.pdf&clen=592839&chunk=true Anaesthesia course]

  • Jenny's Jam Jar

  • [chrome-extension://efaidnbmnnnibpcajpcglclefindmkaj/viewer.html?pdfurl=https%3A%2F%2Fcicmwrecks.files.wordpress.com%2F2020%2F07%2F2020-1-12.pdf&clen=388609&chunk=true CICM wrecks] and [chrome-extension://efaidnbmnnnibpcajpcglclefindmkaj/viewer.html?pdfurl=https%3A%2F%2Fcicmwrecks.files.wordpress.com%2F2017%2F04%2F2015-1-12-resonance-and-damping.pdf&clen=226337&chunk=true CICM Wrecks]

Similar questions

  • Question 24, 2012 (1st sitting)

  • Question 12, 2015 (1st sitting)

  • Question 17, 2017 (2nd sitting)

Question 13

Question

Explain the control of breathing


Example answer

Normal respiration

  • 12-20 / min
  • Normal tidal volumes ~6-8mls/kg (~500mls in 70kg adult)
  • Normal minute ventilation ~6-10L/min


Voluntary control of respiration

  • Mediated via the cerebral cortex via the corticospinal tracts and motor neurons of the muscle of respiration


Involuntary control of respiration

  • Adjusts ventilation to adapt to the needs of the body


Sensors + afferents

  • Peripheral chemoreceptors
    • Located in carotid and aortic bodies
    • Stimulated by fall in PaO2, rise in PaCO2, or fall in pH
    • Afferents CN IX (carotid body) and CN X (aortic body)
  • Central chemomreceptors
    • Located at ventral medulla near the respiratory centre
    • Detects change in CSF pH (due to CO2 diffusion across BBB)
  • Mechanoreceptors
    • Slow stretch receptors in the bronchial and lung tissue
    • Activated by stretch
    • Afferent is CN X
  • Other inputs:
    • Temperature (thalamus)
    • Emotion (limbic system)
    • Hormones (adrenaline)
    • Lung water (J receptors in the lung)
    • Baroreceptor reflex


Integrator/controller + efferents

  • Respiratory centre in the medulla and pons
  • Nucleus retroambiugualis (controls expiratory muscle group via UMN)
  • Nucleus parambigualis (controls inspiratory muscle group via UMN)
  • Nucleus ambigualis (pharyngeal muscle dilator function)
  • Pre-botsinger complex (respiratory pacemaker > phrenic nerve)
  • Pontine respiratory group (prevents over expansion of the lung)


Effectors

  • Muscles of respiration
    • Pharyngeal muscles - dilate airway
    • Laryngeal muscles - abduct vocal cords
    • External intercostals - elevate ribs, move sternum forward > increase AP + lateral diameter thoracic cavity
    • Diaphragm = main inspiratory muscle > increases intrathoracic volume
    • Accessory muscles = SCM, pecs, scalene, abdominal muscles


Examiner comments

53% of candidates passed this question.

Most candidates provided a structured answer based around a sensor / central integration /
effector model with appropriate weighting towards the sensor / integration component. Better
answers provided an understanding of details of receptor function, roles of the medullary and
pontine nuclei and how these are thought to integrate input from sensors. Marks were awarded
to PaCO2 ventilation and PaO2 ventilation response when accurate, correctly labelled diagrams
or descriptions were provided.


Online resources for this question


Similar questions

  • Question 21, 2013 (1st sitting)

  • Question 2, 2015 (1st sitting)

  • Question 13, 2015 (2nd sitting)


Question 14

Question

Describe the pharmacology of frusemide.


Example answer

Name Furosemide
Class Loop diuretic
Indications Oedema/fluid overload, renal insufficiency, hypertension
Pharmaceutics Tablet, clear colourless solution (light sensitive),
Routes of administration IV, PO,
Dose Varies (~40mg daily commonly used for well patients, can be sig. increased)
pKA 3.6 (highly ionised; poorly lipid soluble)
Pharmacodynamics
MOA Binds to NK2Cl transporter in the thick ascending limb LOH, leads to decreased Na,K, Cl reabsorption > decreased medullary tonicity + Inc Na/Cl delivery to distal tubules > decreased water reabsorption > diuresis
Effects Renal: diuresis

CVS: hypovolaemia, arteriolar vasodilation + decreased preload (=mechanism for improvement of dyspnoea before diuretic effect)
Renal: increase in RBF

Side effects CVS: hypovolaemia, hypotension

Renal/metabolic: Metabolic alkalosis, LOW Na, K, Mg, Cl, Ca, increased Cr
Ototoxicity, tinnitus, deafness

Pharmacokinetics
Onset 5 mins (IV), 30-60 mins (PO), Effect lasts 6 hours.
Absorption Bioavailability varies person-person (40-80%)
Distribution Vd = 0.1L/Kg, 95% protein bound (albumin)
Metabolism < 50% metabolised renally into active metabolite
Elimination Renally cleared (predominately unchanged). T1/2 ~90 mins.
Special points Deafness can occur with rapid adminsitration in large doses


Examiner comments

51% of candidates passed this question.

Most candidates presented a well-structured answer and provided a basic understanding.
Answers that provided accurate indications and details of the mechanism underlying the actions
of frusemide attracted more marks. Those recognising the increased delivery of sodium and
chloride to the distal tubule (exceeding resorptive capacity) were awarded more marks that those
answers that attributed the diuretic action solely to reduction in the medullary gradient. Frusemide
has many potential adverse effects and a reasonable list was expected. Conflicting information
was common (e.g. highly bound to albumin – Vd 4 L/kg) and better answers avoided this.


Online resources for this question


Similar questions

  • Question 20, 2013 (1st sitting)
  • Question 16, 2018 (1st sitting)
  • Question 18, 2019 (1st sitting)


Question 15

Question

Define bioavailability (10% of marks). Outline the factors which affect it (90% of marks).


Example answer

Bioavailability

  • The fraction of the drug dose reaching the systemic circulation, compared to an equivalent dose given intravenously.
  • Can be calculated from the area under the concentration time curves for an identical bolus dose given non-intravenously (e.g. orally) and intravenously at the same time.

<math display="block">{Bioavailability} = \frac{AUC_{oral}}{AUC_{IV}}</math>

File:C:\Users\ethan\AppData\Roaming\Typora\typora-user-images\image-20210305141509389.png


Factors influencing bioavailability

  • Preparation of the drug
    • Preparation (e.g. solution > capsule > tablet > coated tablet)
  • Drug properties
    • Molecular size (increase = decreased absorption)
    • Degree of ionisation (non ionised - increased bioavailability)
    • Lipid solubility (increased solubility = increased bioavailability)
  • Route of administration
    • Oral, transdermal, Subcutaneous, intramuscular, intranasal, inhaled etc.
  • Drug interactions
    • Drugs/food may interact/inactivate/bind to the drug
    • e.g. absorption of tetracyclines reduced with concurrent administration of calcium such as in milk
  • Patient factors
    • Oral: Malabsorption syndromes (e.g. coeliac disease), gastric stasis (e.g. postop),
    • IM/SC/Topical: Degree of tissue perfusion
    • Pregnancy - alters gastric pH, intestinal motility
    • Pharmacogenetic differences in absorption, metabolism of drugs (e.g. isoniazid)
  • First pass metabolism
    • Drugs absorbed via GIT pass via portal vein to liver and are subject to first pass metabolism (metabolised prior to reaching systemic circulation).
    • May be impaired with hepatic insufficiency (increased bioavailability)


Examiner comments

49% of candidates passed this question.

Many candidates spent time defining and describing aspects of pharmacokinetics which were not
relevant to the question. E.g. clearance, volume of distribution and half-life. Candidates who
scored well utilised a structure which incorporated the headings of the factors which affect the
bioavailability of medications with a simple description as to the nature of the effect. These factors
included: the physical properties of the drug, the preparation, patient factors, the route of
administration and metabolism amongst others.


Online resources for this question

Similar questions

  • Question 22, 2017 (2nd sitting)


Question 16

Question

Outline the formation, circulation and functions of cerebrospinal fluid


Example answer

CSF

  • ECF located in the ventricles and subarachnoid space
  • ~2ml/kg
  • Divided evenly between the cranium and spinal column


Formation

  • Constantly produced
  • ~550ml produced per day (~24mls/hr)
  • Produced by
    • Choroid plexus (70%) - located in ventricles of brain
    • Capillary endothelial cells (30%)
  • Produced by a combination of ultrafiltration (via fenestrated choroidal capillaries) and active secretion
    • Na actively transported out. Gradient drives co-transport of HCO3 + Cl
    • Glucose via facilitated diffusion, water by osmosis


Circulation

  • Circulation is driven by

    • Ciliary movement of ependymal cells

    • Respiratory oscillations and arterial pulsations

    • Constant production and absorption

  • CSF flows from

    • Lateral ventricles > foramen of Monro > 3rd ventricle > Sylvian aqueduct > 4th ventricle > cisterna magna (via foramen megendie and luschka) > spreads between spinal/cranial subarachnoid spaces

  • Reabsorption by the arachnoid villi

    • Rate of ~24mls/hr

    • Located predominately in the dural walls of the sagittal + sigmoid sinuses

    • Function as one way valves, with driving pressure leading to absorption.

Functions

  • Mechanical protection
    • The low specific gravity of CSF > decreased effective weight of the brain (1500g > 50g)
    • With the reduced weight
      • Less inertia = less acceleration/deceleration forces
      • Suspended > no contact with the rigid skull base
  • Buffering of ICP
    • CSF can be displaced / reabsorbed to offset any increase in ICP
  • Stable extracellular environment
    • Provides a constant, tightly controlled, ionic environment for normal neuronal activity
  • Control of respiration
    • The pH of CSF is important in the control of respiration (CO2 freely diffuses into CSF and can activate central chemoreceptors)
  • Nutrition
    • Provides a supply of oxygen, sugars, amino acids to supply the brain


Examiner comments

81% of candidates passed this question.

This is a three-part question and was marked as such. The circulation and functions of CSF was
generally well answered. Formation of CSF, however, was answered poorly, with many candidates listing its composition instead. The examiners were looking for an understanding of the physiological processes of formation not the composition


Online resources for this question


Similar questions

  • Question 22, 2007 (1st sitting)
  • Question 6, 2008 (2nd sitting)
  • Question 2, 2013 (1st sitting)
  • Question 16, 2015 (1st sitting)
  • Question 24, 2017 (1st sitting)
  • Question 15, 2018 (2nd sitting)
  • Question 11, 2019 (1st sitting)


Question 17

Question

Discuss the advantages and disadvantages of the use of an intravenous infusion of fentanyl in comparison to morphine.


Example answer

Distribution

  • Fentanyl
    • Widely and rapidly distributed in tissues
    • Thus will accumulate in tissues with sustained infusions
  • Morphine
    • Relatively less widely distribute and thus less likely to accumulate in tissues


Context sensitive half time (CSHT)

  • due to the differences in distribution, fentanyl has an increased CSHT relative to morphine
  • Therefore, the effects of morphine are less likely to be effected by the duration of infusions, whereas with fentanyl, increasing infusions will lead to longer time to wear off and in a less predictable manner


Metabolism

  • Morphine
    • Metabolised hepatically (hepatic insufficiency - prolonged effect)
    • Active metabolites (accumulate in renal failure - prolonged effect)
  • Fentanyl
    • Metabolised by liver (thus hepatic insufficiency = prolonged effect)
    • Does not have active metabolites


Lipid solubility

  • Morphine = poor lipid solubility = prolonged CNS effect
  • Fentanyl = good lipid solubility = reduced CNS effect


Protein binding

  • Fentanyl has 90% protein binding, morphine 30%
  • Thus low protein in critical ilness = increased effect of fentanyl (morphine less effected)


Other pharmocodynamics

  • Morphine is a vasodilator (helpful in CCF, less so in septic shock)
  • Fentanyl has no direct cardiovascular effects
  • Fentanyl has a fast onset of action


Other

  • Fentanyl more expensive than morphine

Examiner comments

27% of candidates passed this question.

These are both level 1 drugs commonly used as an infusion in daily practice. This question specifically asked the candidates to frame their answers around an intravenous infusion of fentanyl in comparison to morphine. A tabular listing of general properties of the two drugs highlighting the differences between the drugs would not score well. The question asks for a considered response that should focus on context sensitive half-life, compartments and metabolism, instead many focused on the speed of onset and potency, which are minor considerations when drugs are given for long periods by infusion. Candidates often demonstrated a superficial knowledge of key pharmacokinetic concepts with limited application of these principles in the context of an intravenous infusion. Better answers also related the above to various relevant pharmacodynamic influences such as age, liver and renal impairment.


Online resources for this question


Similar questions

  • Question 16, 2011 (1st sitting)
  • Question 12, 2016 (2nd sitting)
  • Question 13, 2017 (2nd sitting)


Question 18

Question

Describe the respiratory changes that occur throughout pregnancy


Example answer

Anatomical changes

  • Diaphragm

    • Ascends progressively (up to 4cm) throughout pregnancy due to mass effect from the foetus

  • Chest wall

    • Increased AP + Lateral chest wall diameters

    • Thoracic circumfrence increases

    • Due to effect of relaxin

  • Increased oropharyngeal oedema (increased oestrogen)

Lung volumes

  • Lung volume changes occur after trimester 1
  • Mostly due to mass effect of pregnancy
    • TLC decreases ~5%
    • FRC decreases ~20%
    • IC increases by 10%
    • FRC decreased by 20%
    • No change to closing capacity


Breathing + Mechanics

  • Minute ventilation
    • Increases by 50% (Due to increased TV and RR) - trimester 1
    • Due to left shift of PaCO2 curve by progesterone
    • Increases during labour due to pain
  • Compliance
    • chest wall compliance decreases due to increased abdominal contents (Trimester 1)
    • Lung compliance stays the same
  • Resistance
    • Increased upper airway resistance due to mucosal oedema (due to oestrogen, progesterone)


Gas exchange / tension

  • PaO2 increases
  • PaCO2 decreases (increased minute ventilation) due to progesterone induced sensitivity to CO2
  • Leads to compensated respiratory alkalosis (progesterone)


VO2

  • Increased oxygen consumption (~20%) due to increased body mass + fetus
  • Increases by up to 60% in labour


Post delivery

  • FRC and TV return to normal within 5 days


Examiner comments

31% of candidates passed this question.

The question asked for a description of the respiratory changes throughout pregnancy, which includes labour. Simple lists of changes did not score highly. A straightforward structure including; first, second and third trimester delineation would have elevated many answers from below par to a pass. Many good answers gave succinct detail on both mechanical respiratory changes and the hormonal mechanisms behind them. Higher scoring answers also described the overall effect of individual changes to spirometry, geometry or respiratory control.


Online resources for this question


Similar questions

  • Question 4, 2014 (1st sitting)


Question 19

Question

Discuss the determinants of venous return to the heart.


Example answer

Venous return

  • Rate of blood flow back to the right atrium
  • In healthy state: venous return = cardiac output (else pathological pooling of blood occurs)
  • Can be defined by
    • VR = MSFP - RAP / resistance to venous return
  • Therefore factors effecting venous return are those that affect
    • MSFP
    • RAP
    • Resistance to venous return
    • Cardiac output


Cardiac output

  • Increased CO = increased venous reutn
  • CO is effected by
    • Afterload (reduced afterload = increased cardiac output = increased VR)
    • Contractility (increased contractility = increased CO = increased VR)


MSFP

  • Normally ~7mmHg
  • Increased MSFP = increased VR
  • Affected by venomotor tone and blood volume
  • Increased VR (= increased blood volume and increased venomotor tone)


RAP

  • Increased RAP = reduced driving pressure = reduced venous return
  • Factors which increase RAP
    • Positive intrathoracic pressure (e.g. PPV)
    • Reduced pericardial compliance (e.g. effusion)
    • Reduced RA compliance/contractility (e.g. AF)
    • TVR


Resistance to venous return

  • Increased RVR = reduced VR (due to ohms law)
  • Factors effecting RVR
    • Autonomic tone
    • Intrabdominal pressure
    • IVC Obstruction (e.g. pregnancy) reduces VR
    • Posture (decreased VR with erect posture)
    • Vasoactive drugs
    • skeletal muscle pump


Examiner comments

67% of candidates passed this question.

The factors that influence VR are captured in 2 formulae; VR = CO, and VR = (MSFP-RAP) / Venous Resistance. Candidates that used these as the backbone structure of their answer scored well. Quite a few candidates failed to consider factors that affect left heart CO also effect VR. Recognising that CO does = VR appeared to elude some candidates.


Online resources for this question


Similar questions

  • Question 6, 2018 (2nd sitting)


Question 20

Question

Outline the distribution, absorption, elimination, regulation and physiological role of phosphate.


Example answer

Absorption

  • Normal intake = ~0.5mmols/kg/day
  • Absorbed in the intestine (duodenum, jejunum)
    • Passive mechanism = paracellular = not regulated
    • Active mechanism = cotransport with sodium = regulated


Distribution

  • 85% = stored in bone/teeth
  • 14% = intracellular
  • 1% = extracellular fluid (half ionised, other half forms complexes/proteins)
    • Normal serum level = 0.8-1.2 mmol/L


Elimination

  • Renal
    • Freely filtered in kidney
    • Most is reabsorbed in proximal and distal tubules
    • 2/3 of phosphate that is lost, is lost renally
  • Stool
    • 1/3 lost in stools


Regulation

  • Calcitriol

    • Increased bone reabsorption

    • Increased Intestinal absorption

    • Increased Renal reabsorption

  • PTH

    • Decreased renal reabsorption

    • Increased bone resorption

    • Net effect =decrease in serum phosphate

  • Thyroxine

    • Increased renal reabsorption

  • Glucocorticoids

    • Decreased renal reabsorption

Role

  • Structural role
    • bone and teeth formation
    • Phospholipids of cell membranes, DNA, RNA
  • Regulatory role
    • Second messenger (IP3)
  • Metabolic role
    • Synthesis of ATP
    • Acid base regulation (urinary and intracellular buffering)
    • cofactor in oxygen transport (2-3 DPG)


Examiner comments

29% of candidates passed this question.

The answer structure should have utilized the headings provided in the question. Many candidates described the physiology of calcium, which while related, did not attract marks. The distribution section required not only the sites of distribution but also the percentages found in each. The regulation should have included both primary and secondary mechanisms and an outline on the factors affecting renal excretion, intestinal absorption and release from bone etc. An outline of the physiological role of phosphate required a broad knowledge of physiological processes.


Online resources for this question


Similar questions

  • ?None




2019 (2nd sitting)

Question 1

Question

Describe the physiological consequences of the oral ingestion of 1 litre of water in a young adult.


Example answer

Handling of oral water ingestion

  • Absorption
    • Near complete absorption of water occurs in the proximal small intestine (85%), with 10% in large bowel, 5% in rectum.
    • Most of the diffusion is transcellular and driven by osmosis (due to active absorption of other electrolytes, including sodium)
  • Distribution
    • Absorbed water distributes equally amongst all body fluid compartments, proportional to size
      • ~66% into the ICF (~667mls)
      • ~33% into the ECF (~333mls)
        • ~75% of which is interstitial fluid
        • ~21% of which is intravascular
        • ~4% of which is transcellular fluid
  • Elimination
    • Water is eliminated predominately by renal excretion
    • Filtered water at the glomerulus is highly regulated


Physiological consequences of oral water ingestion

  • Decrease in osmolality
    • ~2.5% decrease in osmolality for 1L of oral water
    • Sensed by osmoreceptors (hypothalamus) which have sensitivity of ~2% > decrease in secretion of vasopressin from the posterior pituitary gland
    • Decreased vasopressin > decreased luminal aquaporin channel insertion in collecting ducts of nephrons > decreased water reabsorption > diuresis
  • Decrease in plasma Na concentration
    • Leads to release of angiotensin and aldosterone > increased Na reabsorption in nephron
  • Small increase in blood volume
    • For 1L oral ingestion of water > leads to ~70mls of intravascular water (33% of 1L goes to ECF, 21% of which is intravascular)
    • This change is below the sensitivity threshold of the cardiovascular regulatory reflexes > no change in blood pressure/HR of a normal healthy individual


Examiner comments

28% of candidates passed this question.

It was expected candidates would provide details the consequences of water ingestion from its rapid absorption in the small intestine to the resultant impact on plasma osmolarity and the minimal impact of plasma volume of this volume. Some detail on the mechanisms of absorption (transcellular vs osmosis) was expected and the distribution of water across body fluid spaces. Many candidates accurately described the small drop in plasma osmolarity that is sufficient to trigger osmoreceptors with better answers providing details of the locations and mechanisms involved. The physiological consequences of inhibition of ADH, including the renal effects of decreased water permeability in distal renal tubules and collecting ducts. The volume load after distribution would be lower than the plasma volume triggers for the circulatory reflex responses.


Online resources for this question


Similar questions

  • ? None



Question 2

Question

Describe renal blood flow and its regulation (80% of marks). Outline the impact of adrenoreceptor agonists on renal blood flow (20% of marks).


Example answer

Renal blood flow (RBF)

  • Approximately 20-25% of cardiac output (1-1.25L/min)
  • Majority of blood flow is distributed to the renal cortex (95%) compared to renal medulla (5%)
  • Renal blood flow far exceeds metabolic requirements --> to support the filter function of th ekidney


Anatomy of RBF

  • Renal arteries > interlobar arteries > arcuate arteries > interlobular arteries > afferent arterioles > glomerulus > efferent arterioles > peritubular capillaries > venous system
  • Venous system similarly named in reverse


Regulation of RBF

  • Autoregulation
    • Kidneys have the capacity to autoregulate (cortical nephrons can, juxtamedullary nephrons cant)
    • Can maintain a constant RBF across a wide range in MAP (70-170mmHg)
    • Two main mechanisms: myogenic autoregulation, tubuloglomerular feedback
    • Myogenic autoregulation
      • Intrinsic contraction of the afferent arterioles in response to increased transmural pressures via release of vasoactive mediators
    • Tubuloglomerular feedback
      • Increased RBF > increased GFR > increased Na/Cl sensed by macula densa > releases adenosine > constriction of afferent arterioles > decreased RBF
      • Decreased RBF > decreased GFR > decreased Na/Cl at macula densa > releases NO > dilation > increased RBF
  • SNS
    • Activation of adrenoreceptors > constriction of arterioles > decreased RBF
  • Hormonal response
    • Renin is released by B1 stimulation and decreased GFR
    • AG2 constricts afferent and efferent arterioles > decreased flow


Impact of adrenoreceptor agonists on RBF

  • As mentioned, kidneys are innervated by SNS (adrenergic receptors)
  • Massive SNS stimulus (e.g. shock, high dose adrenergic agonists) can override autoregulation
  • Efferent arterioles constrict greater than afferent arterioles > decrease in RBF, but the GFR is proportionally less effected (greater perfusion pressure)
  • Effect of alpha adrenergic agonists
    • Will act as renal vasoconstrictors > decrease renal blood flow / GFR
    • Examples: phenylephrine, metaraminol
  • Effect of beta adrenergic agonists
    • Will lead to increased RBF (vasodilator)
    • Example: isoprenaline
  • Non-selective adreneergic agonists
    • Greater proportion of alpha > beta receptors.
    • Mixed agonists (e.g. adrenaline) will predominately lead to decreased flow (alpha predominance)


Examiner comments

64% of candidates passed this question.

This question was well answered by most candidates. The description of renal flow involves a brief comment of the anatomy including interlobar, arcuate, interlobular arteries, then afferent and efferent arterioles – 2 sets of capillaries and then corresponding veins and better answers made the distinction better cortical and medullary flow and went on to detail the consequence of this. Renal blood flow is autoregulated and most candidates describe well the various mechanisms around myogenic and tubuloglomerular feedback. Additional marks were gained with by discussing renal vascular resistance and how this may be varied. The impact of adrenoreceptor agonists is varied but generally sympathomimetic agents will vasoconstrict and therefore increase renovascular resistance and result in a decrease renal blood flow. The relative impact on afferent vs efferent arteriolar tone may alter glomerular perfusion pressure.


Online resources for this question


Similar questions

  • Question 4, 2019 (1st sitting)
  • Question 18, 2017 (1st sitting)
  • Question 3, 2015 (2nd sitting)
  • Question 11, 2012 (1st sitting)
  • Question 21, 2011 (2nd sitting)
  • Question 12, 2008 (2nd sitting)



Question 3

Question

Describe the relationship between muscle length and tension (50% of marks). Outline the physiologic significance of this relationship in cardiac muscle (50% of marks).


Example answer

Length-tension relationship

  • The tension generated within a single muscle fibre is related to its length

  • Total tension = passive tension + active tension

  • Passive tension

    • Increases with increasing muscle length (modelled as a non-linear spring)

  • Active tension

    • Also varies with muscle length, but is described by the sliding filament model and has an optimal length at which maximal tension is generated

    • The physiological basis of this is due to different number of actin-myosin cross bridges formed at the different muscle lengths

    • The optimal myocardial sarcomere length is ~2.2 um (greatest overlap of actin-myosin filaments)

  • The resting muscle length is often close to the optimal length for active tension

Cardiac muscle

  • The muscle length-tension relationship forms an important part of the Frank-Starling law (strength of myocardial contraction is dependant on the initial muscle fibre length)
  • With increase in diastolic filling of the heart > increase stretch (preload) > increased muscle length (increased cross bridge formation) > increased force of contraction > increased stroke volume
    • Note: this mechanism is within limits. When the muscle length is too great, there is actually a reduction in cross bridge formation > decreased SV
  • Importance
    • Ensures venous return = cardiac output (else pooling would occur)
    • Allows beat-beat adjustments to variation in preload


Examiner comments

41% of candidates passed this question.

Some detail was expected on a general description that tension is variable with the length of muscle. It was expected answers would describe that there is a resting length at which tension developed on stimulation is maximal. Many candidates omitted that differences exist between muscle types with smooth muscle behaving differently. Additional credit was given for the distinction about active tension vs resting tension. It was expected a description of the potential mechanism would be included with discussion of sliding filament theory, overlapping fibres and optimal sarcomere length. Some candidates utilised a diagram effectively to convey understanding and more detail was rewarded with additional marks.
The second half of the question involved describing how this relationship is particularly important in cardiac muscle and underpins the Frank Starling relationship and all the cardiac physiology that follows. Initial length of fibres is determined by the diastolic filling of the heart, so pressure developed is proportionate to the total tension developed. The developed tension increases as diastolic volume increases to a maximum (the concept of Heterometric regulation). Better answers appreciated that the physiology may be different for a whole heart rather than isolated muscle fibres.


Online resources for this question


Similar questions

  • ? None



Question 4

Question

Outline the pharmacology of intravenously administered magnesium sulphate


Example answer

Name Magnesium sulphate
Indications HypoMg, eclampsia/pre-eclampsia, severe asthma, arrhythmias (including TdP), analgesia
Pharmaceutics Clear colourless solution, various concentrations
Routes of administration IV, IO
Dose 10-20mmols
Pharmacodynamics
MOA Essential cation
- Essential cofactor in hundreds of enzymatic reactions
- Necessary in several steps of glycolysis (ATP production)
- NMDA receptor antagonism (increasing seizure threshold)
- Inhibits Ach release at NMJ
- Smooth muscle relaxation (Inhibits Ca L-type channels)
Effects CNS: anticonvulsant

Resp: Bronchodilation
CVS: Anti-arrhythmic

Side effects Related to speed of administration + degree of HyperMg (dose dependant)

CVS: Hypotension, bradycardia
CNS/MSK: hyporeflexia, muscle weakness, CNS depression
RESP: respiratory depression
GIT: Nausea, vomiting

Pharmacokinetics
Onset Immediate
Absorption N/A
Distribution 30% protein bound
Metabolism Nil
Elimination Urine; clearance is proportional to GFR and plasma concentration
Special points Incompatible with calcium salts > precipitation

Drug interaction with NMB agents (potentiation)


Examiner comments

55% of candidates passed this question.

Overall answers were well structured. However, a lack of detail and inaccurate pharmacokinetics was common. Better answers included a discussion of the mechanism of action of Mg++ including Ca++ antagonism, presynaptic cholinergic effects and NMDA receptor antagonism. Adverse effects were not discussed in detail by many candidates and contraindications were commonly omitted.


Online resources for this question


Similar questions

  • Question 10, 2011 (2nd sitting)
  • Question 20, 2014 (2nd sitting)
  • Question 10, 2015 (2nd sitting)
  • Question 18, 2021 (1st sitting)



Question 5

Question

Describe the anatomical course and relations of the trachea and bronchial tree (to the level of the segmental bronchi).


Example answer

Trachea

  • 10-12cm long fibromuscular tube
  • Continuation of the larynx (at ~C6) and divides into the left and right main bronchi at the level of the carina (~T5)
  • Relations
    • Superior: larynx
    • Anterior: manubrium, thyroid, brachiocephalic trunk, thymus
    • Posteriorly: oesophagus, recurrent laryngeal nerve
    • Right lateral: thyroid, right common carotid a., right vagus nerve, azygous vein. Eventually the right lung and pleura,
    • Left lateral: thyroid, left common carotid, arch of aorta, left subclavian artery, left recurrent laryngeal nerve. Eventually the left lung and pleura,


Bronchi

  • Left main bronchi
    • 5cm long, courses leftward
    • More horizontal and smaller diameter than right main bronchi
    • Relations: azygos vein, right pulmonary artery (ant), pulmonary veins, oesophagus (post)
  • Right main bronchi
    • 2.5cm long, courses rightward
    • More verticle and larger in diameter than left main bronchi
    • Relations: pulmonary hilum - aortic arch, descending aorta, left pulmonary artery, left pulmonary veins


Lobar bronchi

  • Each main bronchi gives rise to lobar bronchi
    • Right: upper, middle and lower lobe bronchi
    • Left: upper and lower lobe bronchi
  • Each lobar bronchi gives rise to segmental bronchi
    • 10 segmental bronchi on each side (left / right) corresponding to the 'bronchopulmonary segments'
    • Left
      • LUL: Apical, Superior, Inferior, Anterior
      • LLL: Anterior, Lateral, Posterior, Superior
    • Right
      • RUL: Apical, Posterior, Anterior
      • RML: Lateral, Medial,
      • RLL: Superior, Medial, Anterior, Lateral, Posterior
  • Relations: predominately alveoli, pleura and accompanying pulmonary arteries/veins at this stage


Addit: mnemonics for remembering bronchopulmonary lung segments

  • Right lung
    • 'A PALM Seed Makes Another Little Palm'
      • RUL: Apical, Posterior, Anterior
      • RML: Lateral, Medial,
      • RLL: Superior, Medial, Anterior, Lateral, Posterior
  • Left lung
    • 'ASIA ALPS'
      • LUL: Apical, Superior, Inferior, Anterior
      • LLL: Anterior, Lateral, Posterior, Superior

Examiner comments

24% of candidates passed this question.

Better answers included details of the significant structures related to the cervical and mediastinal trachea and bronchi. The lobar branches and bronchopulmonary segments requiring naming to attract full marks. Many answers lacked sufficient detail or contained inaccuracies regarding vertebral levels and key structural relations. Some candidates discussed the general anatomy of the airway, including the larynx, structure of the airways, blood supply and innervation. This did not attract marks.


Online resources for this question


Similar questions

  • Question 24, 2016 (2nd sitting)



Question 6

Question

Outline the factors that determine central venous pressure and explain how it is measured.


Example answer

Overview

  • CVP is the venous blood pressure measured at or near the right atrium
  • Normally 0-6mmHg in spontaneously breathing non-ventilated patient
  • Measurement taken at end-expiration


Measurement

  • Most commonly measured using central venous catheter (CVC)
    • CVC tip sits at or near the right atrium
    • CVC is connected to a pressure transducer via incompressible tubing with flush solution
    • Transducer is zeroed to the atmospheric pressure and levelled at the height of the right atrium
  • Echocardiography can provide non-invasive estimations off the CVP
  • Visual inspection of the height of the JVP can provide some bedside clinical insight


Factors determining CVP

  • Central venous blood volume
    • Increased total blood volume (e.g. renal failure) = increased MSFP > increased CVP
    • Decreased CO (e.g. LV failure) > blood backs up > increased thoracic blood volume > increased CVP
  • Central venous vascular compliance
    • Increased vascular tone central veins (e.g. noradrenaline) > decreased compliance > increased CVP
    • Decreased myocardial/pericardial compliance > increased CVP
    • Decreased pulmonary arterial compliance > increased CVP
  • Tricuspid valve function
    • TV regurg > increased CVP (retrograde transmission of RV systolic pressure)
    • TV stenosis > increased CVP (increased resistance to RV inflow)
  • Intrathoracic pressure
    • ITP is transmitted to the central venous compartment
    • Thus, increased PEEP, PPV, or a tension pneumothorax will lead to increased CVP
  • Measurement technique
    • Level of the transducer will clearly influence the CVP measured
    • Timing of the measurement with respiratory cycle


Examiner comments

18% of candidates passed this question.

It was expected that answers include central venous blood volume, central venous vascular compliance, intrathoracic pressure and tricuspid valvular function. Good answers outlined how each of these factors determine CVP and whether it was increased or decreased. Many candidates incorrectly described the effect of venous return.


Online resources for this question


Similar questions

  • Question 5, 2021 (1st sitting)



Question 7

Question

Define closing capacity (10% of marks). Describe the factors that alter it (30% of marks), its clinical significance (30% of marks) and one method of measuring it (30% of marks).


Example answer

Closing capacity

  • The point in expiration when the small airways begin to collapse
  • Small airway closure occurs because the elastic recoil of the lung overcomes the negative intrapleural pressure keeping the airway open.
  • More likely to occur in dependant parts of the lung where airways are smaller (due to effects of gravity).
  • Closing capacity = residual volume + closing volume


Significance of closing capacity

  • If closing capacity exceeds the FRC then small airway collapse occurs during tidal respiration
    • This leads to shunt > V/Q mismatch > impairs oxygenation > hypoxaemia
    • This also leads to gas trapping > reduced compliance
  • High CC will therefore also
    • Decrease the effect of anaesthetic pre-oxygenation
    • Increases dependant atelectasis.
      • Cyclic opening/closing of airways due to increased atelectasis > lung injury


Factors affecting closing capacity

  • Age
    • Normally CC is less than FRC at a young age
    • Increasing age > increasing closing capacity
      • At age 44, supine FRC = closing capacity
      • At age 66, erect FRC = closing capacity
  • Expiratory gas flow
    • Increase flow > increased closing capacity
  • Pathology
    • Parenchymal/airway disease (e.g. COPD) > loss of tissue available for radial traction > (closing capacity > FRC)
    • Decreased surfactant > increased surface tension > increasing collapsing pressure > increased CC
    • Increased pulmonary blood volume (e.g. CCF) > increased weight compressing small airways > increased CC


Measurement of CC

  • Closing capacity = closing volume + residual volume
  • Closing volume
    • Measured using the single breath nitrogen washout test
    • Subject exhales to residual volume
    • Pure oxygen inhaled to TLC
    • Subject breaths out through a nitrogen sensor (records N2 concentration vs time curve)
    • Phase 4 of this curve indicates the closing volume.
  • Residual volume
    • Cannot be directly measured
    • FRC is first calculated by body plethysmography (or other methods)
    • ERV measured using spirometry
    • Residual volume is the difference between FRC and ERV


Examiner comments

49% of candidates passed this question.

Many candidates confused the factors that affect closing capacity (CC) with factors which affect functional residual capacity (FRC). Some candidates confused airway closure with expiratory flow limitation secondary to dynamic airway compression. A good answer would have included the following:
Small airway closure occurs because the elastic recoil of the lung overcomes the negative intrapleural pressure keeping the airway open. Thus, airway closure is more likely to occur in dependant parts of the lung where airways are smaller. Normally closing capacity is less than FRC in young adults but increases with age. Closing capacity becomes equal to FRC at age 44 in the supine position and equal to FRC at age 66 in the erect position. Closing capacity is increased in neonates because of their highly compliant chest wall and reduced ability to maintain negative intrathoracic pressures. In addition, neonates have lower lung compliance which favours alveolar closure. Closing capacity is also increased in subjects with peripheral airways disease due to the loss of radial traction keeping small airways open.
The consequences of airway closure during tidal breathing include shunt and hypoxaemia, gas trapping and reduced lung compliance. In addition, cyclic closure and opening of peripheral airways may result in injury to both alveoli and bronchioles. Closing volume (CV) may be measured by the single breath nitrogen washout test or by analysis of a tracer gas such as xenon during a slow exhaled vital capacity breath to residual volume. Residual volume (RV) cannot be measured directly but is calculated as follows: the FRC is measured using one of three methods: helium dilution, nitrogen washout or body plethysmography. The expiratory reserve volume (ERV) may be measured using standard spirometry. Using the measured FRC and ERV we may calculate RV from the equation:
RV = FRC – ERV. Then CC = RV + CV.


Online resources for this question


Similar questions

  • None



Question 8

Question

Outline the pharmacology of drugs used to treat asthma.


Example answer

Oxygen

  • Increases FiO2 > increased SaO2 (by increasing PAO2 as per Alveolar gas equation).
  • Given by numerous devices (nasal prongs, masks, NIV, ETT)
  • Dose titrated to SaO2
    • Hypoxemia is harmful (but optimal target SaO2 unclear)
    • Generally titrated to Sats 94-98% (with caveats for some subgroups of patients)
    • Hyperoxia may lead to hypercapnia, worsening of V/Q mismatch (through alteration of HPVC), lung damage


Beta-adrenergic agonists

  • Long acting B2 selective agonists (e.g. salmeterol) are used in prevention
  • Short acting B2 selective agonists (e.g. salbutamol) are preferred first line therapy for exacerbation
  • Nonselective adrenergic agonists (e.g. adrenaline) can also be used in severe exacerbations
  • SABAs can be given inhaled (via spacer), nebulised or via IV infusion (if unresponsive to inhaled)
  • Example: Salbutamol
    • Short acting B2 agonist
    • MOA: Acts on B2 receptors (Gs protein coupled receptors) in bronchial smooth muscle cells > activates activates adenyl cyclase-CAMP system > increase cAMP > decreased intracellular Ca > SM relaxation / bronchodilation
    • Side effects: Tachycardia, Anxiety, tremor, Hypokalaemia, lactic acidosis


Anticholinergics

  • Example: ipratropium bromide
  • Routes: Inhaled, nebuliser
  • MOA: Competitive antagonism of muscarinic ACh receptors > bronchodilation + decreased secretions
  • Side effects: dry mouth, N/V, headache, blurred vision


Corticosteroids

  • Examples: hydrocortisone (IV), prednisone (PO), budesonide (inhaled)
  • Systemic corticosteroids should be given to all mod-severe asthma > improve outcomes
  • MOA: bind to cytoplasmic glucocorticoid receptors > change in gene transcription > down-regulates the synthesis of proinflammatory cytokines/mediators
  • Effects: increased B receptor responsiveness, decreased inflammation, decreased mucus secretion
  • Side effects: numerous! Depends on dose/duration. Examples:
    • Short term: hyperglycaemia, hypokalaemia, immunosuppression, insomnia/confusion/psychosis,
    • Long term: cushings, osteoporosis, skin thinning, weight gain, immunosuppression


Other potential treatment options (and MOA)

  • Magnesium sulphate > inhibits L type calcium channels > bronchodilation/SM relaxation
  • Ketamine >inhibits L type calcium channels > Bronchial smooth muscle relaxation
  • Aminophylline > PDEI > SM relaxation / bronchodilation
  • Heliox > Improves laminar airflow > may improve ventilation


Examiner comments

29% of candidates passed this question.

Answers should have included the most important aspects of the pharmacology of the most commonly used drugs e.g. class, mechanism of action, pharmacodynamics and important adverse reactions. More information on beta-agonists and corticosteroids (mainstays of management) was expected than drugs like magnesium, ketamine and other adjunctive treatments.


Online resources for this question


Similar questions

  • Question 12, 2019 (1st sitting)
  • Question 4, 2016 (2nd sitting)
  • Question 11, 2014 (2nd sitting)
  • Question 9, 2010 (1st sitting)



Question 9

Question

Compare and contrast the pharmacology of propofol and midazolam.


Example answer

Name Midazolam Propofol Notes/comparisons
Class Benzodiazepine (sedative) Phenolic derivative (IV anaesthetic) -
Indications Anaesthesia, sedation, treatment of seizures, anxiolysis Anaesthesia, sedation -
Pharmaceutics IV: clear solution, pH 3.5. High water solubility White, opaque, liquid emulsion. Contains soybean oil, egg lecithin, glycerol. Poor water solubility Midaz clear, propofol distinctive white colout
Routes of administration IV, IM, S/C, intranasal, buccal, PO IV
Dose Dose depends on many pt. factors. 1-5mg premedication. 2.5-10mg seizures. Infusions. RSI 1-2mg/kg. Infusion (4-12mg/kg/hr)
pKa 6.5 11 (almost completely unionised) Propofol has higher pKa
Pharmacodynamics
MOA Allosteric modulator of GABAA receptors (ionotropic ligand gated channel) in the CNS. Binds to distinct site from GABA. Leads to Cl enters > hyperpolarisation. Propofol binds to B subunit of GABAA receptor > Cl enters > hyperpolarisation Midaz binds to site distinct from GABA
Effects CNS: sedation, amnesia, anticonvulsant effects, decreased cerebral O2 demand CNS: depression, anti-epileptic, decreased CMRO2/CBF/ICP

RESP: depression, apnoea

Both cause sedation and respiratory depression
Side effects CVS: bradycardia, hypotension

CNS: confusion, restlessness
RESP: respiratory depression/ apnoea

RENAL: green urine

CNS: depression, pain injection site
CVS: decreased SVR > hypotension, bradycardia
MET: high lipids

Both cause cardiovascular instability and respiratory depression
Pharmacokinetics
Onset peak effect 2-3 minutes (IV) Seconds Propofol has faster onset / offset
Absorption ~40% oral bioavailability

Absorbed well, but sig. 1st pass metabolism

IV Only (high first pass metabolism) Propofol is IV only
Distribution 95% protein bound, very lipid soluble

Vd = 1L / kg

98% protein bound

VOD 2-10L/kg
Readily crosses BBB

Both highly protein bound.
Metabolism Hepatic metabolism by hydroxylation

Active (1-a hydroxymidazolam) and inactive metabolites

Hepatic > inactive metabolites (glucuronides and sulphates) Both metabolised by liver. Midaz has active metabolites
Elimination Renal excretion

T 1/2 = 4 hours

Renal excretion

Bolus T1/2 - 120s.

Special points Flumazenil - antagonist (reversal agent) No reversal agent


Examiner comments

77% of candidates passed this question.

Highlighting important similarities and differences between the drugs scored higher marks than listing the pharmacology of each drug separately. More pharmacokinetic information was required than simply stating both drugs “are metabolized in the liver and excreted by the kidney”.


Online resources for this question


Similar questions

  • Midazolam
    • Question 7, 2019 (1st sitting)
    • Question 9, 2019 (2nd sitting)
    • Question 4, 2018 (2nd sitting)
    • Question 24, 2016 (1st sitting)
    • Question 2, 2008 (2nd sitting)
  • Propofol
    • Question 11, 2017 (2nd sitting)
    • Question 14, 2015 (2nd sitting)
    • Question 21, 2013 (2nd sitting)
    • Question 5, 2012 (1st sitting)
    • Question 20, 2008 (2nd sitting)



Question 10

Question

Describe the principles of capnography, including calibration, sources of error and limitations.


Example answer

Capnography

  • The graphical display of expired CO2 concentration over time
  • This is different to ETCO2 (which is the CO2 concentration at end-expiration) and capnometry (which is the measurement of CO2 concentration)


Measurement / Principles

  • CO2 concentrations (capnometry) is measured in clinical practice using infrared spectroscopy and applying the principles of the Beer-Lambert Law

    • The concentration of CO2 is measured by exploiting the differences in CO2 absorption of light in the NIR spectrum. With the degree of absorption related to concentration of substance.

  • Components: light emitting diode, photosensor, circuitry, microprocessor, output device

  • Can be monitored using 'side stream' (sampling line with sensor) or 'inline' (sensor directly inline with breathing system) methods

  • Calibration: capnometers are zeroed to room air

Limitations + Sources of error

  • Cannot distinguish Nitrous Oxide (N2O shares similar absorption spectra to CO2)
  • Not diagnostic
    • Waveforms are helpful in assessment (e.g. of bronchospasm, oesophageal intubation) but not diagnostic
    • Patients may have mixed disorders which lead to increased and decreased ETCO2 which are cancelled out and appear falsely normal
  • Side stream monitoring have short delay in measurement and small air leak
  • In line monitoring devices increase dead space (more relevant in paeds)
  • Sensor is susceptible to blockage by secretions
  • Incorrect calibration
  • Water condensation > absorbs IR light > overestimates CO2


Examiner comments

31% of candidates passed this question.

Answers that scored well followed the structure outlined in the question and explained the principles of each component of the question.


Online resources for this question


Similar questions

  • Question 9, 2015 (2nd sitting)



Question 11

Question

Outline the composition of plasma (50% of marks). Describe the functions of albumin (50% of marks).


Example answer

Plasma

  • Cell free liquid component of blood
  • Constitutes ~55% of circulating blood volume
  • Components
    • 92% water
    • 7% proteins (Albumin, globulin, fibrinogen)
    • ~1% other (carbohydrates, lipids, gases, hormones, electrolytes)


Proteins (~7% plasma)

  • 60-80g/L in blood
  • Albumin
    • Majority of plasma protein (35-45g/L)
  • Globulins
    • Second largest component (25-35g/L)
    • Subtypes
      • <math display="inline">\alpha</math>1 globulin (<math display="inline">\alpha</math>1 antitripsin, <math display="inline">\alpha</math>-lipoproteins)
      • <math display="inline">\alpha</math>2 globulin (Haptoglobin, prothrombin, <math display="inline">\alpha</math>2 macroglobulin)
      • <math display="inline">\beta</math>-globulins (CRP, transferrin)
      • <math display="inline">\gamma</math> globulins (Immunoglobulins)
  • Fibrinogen
    • 1-5 g/L
  • Clotting factors


Other solutes (~1% plasma)

  • Carbohydrates (i.e. glucose)
  • Lipids (e.g. LDL, VLDL, HDL, TGs)
  • Gases (e.g. oxygen, carbon dioxide)
  • Hormones (e.g. thyroxine, cortisol, IGF-1)
  • Electrolytes (e.g. Na, Cl, HCO3, K, Mg, Ca)


Albumin (functions)

  • Osmotic pressure
    • Responsible for 80% of the plasma colloid osmotic pressure
    • Helps keep fluid intravascularly
  • Transport function
    • Transports hormones (e.g. thyroxine), fatty acids, electrolytes (e.g. calcium) and drugs
  • Extracellular acid-base buffer
    • Imidazole side chains can bind hydrogen ions and can buffer against change to pH
  • Anticoagulant
    • Has heparin like activity. Low albumin attenuates fibrinolysis
  • Protein store:
    • ~50-60% of plasma protein
  • Anti-oxidant effect
    • Abundant in thio groups which readily scavenge reactive oxygen and nitrogen species
  • Inflammatory marker
    • Negative phase protein (decreases in response to inflammation)


Examiner comments

30% of candidates passed this question.

A good answer began with a definition of plasma and then listed the components - water, albumin, globulins, fibrinogen and other proteins before mentioning the lipid content, nutrient content, wastes and electrolytes. Frequently the breakdown of the globulin component was inaccurate. A common omission was dissolved gas components. Descriptions of the calculation of oncotic pressure and GFR were not asked and hence did not attract marks.
The functions of albumin may be subdivided into: Osmotic pressure, transport function, acid-base buffer, anti-oxidant, anticoagulant effect, protein store, metabolism and 'other'.


Online resources for this question


Similar questions

  • Question 16, 2012 (2nd sitting)



Question 12

Question

Define pain. Outline the processes by which pain is detected in response to a peripheral noxious stimulus


Example answer

Pain

  • "An unpleasant sensory and emotional experience associated with, or resembling that associated with, actual or potential tissue damage" (IASP, 2020)
  • Can be broadly classified by duration (e.g. acute vs chronic) or aetiology (e.g. visceral vs neuropathic)


PAIN PATHWAY


Nociceptors

  • Free unmyelinated nerve endings which convert noxious stimuli into action potentials (APs)
  • Activation
    • Activated by thermal, mechanical, and chemical stimuli
    • Sensitised by inflammatory mediators (e.g bradykinin, histamine, Substance P)
  • Leads do conformational change in nociceptor > ion channel opening > depolarisation
  • Relay APs from the nociceptor receptor to the dorsal horn of the spinal cord (primary afferent)
    • Neuron may travel up/down the tract of Lissauer 1-2 levels prior to synapsing in the dorsal horn
  • Two types of nociceptor neurons
    • Type A<math display="inline">\delta</math> fibres
      • Impulses from mechanical and thermal stimuli
      • Large, myelinated, fast (40m/s)
    • Type C fibres
      • Impulses from thermal, mechanical and chemical stimuli
      • Small, unmyelinated, slow (2m/s)


Second order neurons (afferent)

  • Synapse with primary afferents in Dorsal horn
  • Decussates in the anterior commissure, ascends in the spinothalamic tract, synapses in the thalamus with third order neurons


Third order neuron (afferent)

  • Relays information from the thalamus to the cerebral cortex for central processing of pain


PAIN MODULATION

  • Descending inhibition

    • Neurons from periaqueductal grey matter project to the spinal cord

    • Noradrenaline and serotonin are main neurotransmitters

    • Have inhibitory effects on the synapse of 1st/2nd order neurons

  • Segmental inhibition

    • Initially thought to be due to gate control theory

    • Now other mechanisms though to be responsible

  • Endogenous opioid system

    • (e.g. endorphins) which can bind to opioid receptors > reduced nociception

Examiner comments

33% of candidates passed this question.

Starting with the WHO definition of pain, followed by a brief description of the nature of noxious stimuli (thermal, mechanical, chemical) then proceeding to mention the nature of the cutaneous receptors would have been a very good start to this question. Following this, a description of the various substances involved in pain (K, prostaglandins, bradykinin, serotonin, substance P) and outlining the types of nerve fibres involved in pain transmission and how they synapse in the spinal cord and cortex was expected. The presence and nature of the descending inhibitory pathways was mentioned by very few.


Online resources for this question


Similar questions

  • Question 1, 2014, (1st sitting)
  • Question 22, 2013 (2nd sitting)
  • Question 15, 2011 (2nd sitting)



Question 13

Question

Describe the exocrine functions of the pancreas.


Example answer

Exocrine function of pancreas

  • ~1L - 1.5L of exocrine pancreatic secretions are produced per day
  • Pancreatic (exocrine) secretions are
    • Isotonic
    • Alkaline (pH ~8) - Rich in HCO3
    • Main cation is Na
  • Pancreatic secretions are important for
    • Reducing acidity of contents from stomach (rich in HCO3)
    • Assisting in completion of digestion (contains enzymes)


Main components of exocrine secretions

  • Bicarbonate
    • Produced by ductal cells (up to 150mmol/L)
    • Indirect process driven by Na/H exchanger in basolateral membrane
      • CO2 dissolves into ductal cell. Converted to HCO3 and H+
      • H+ is pumped back out (NA/H exchanger) to maintain gradient
      • HCO3 > facilitated diffusion into lumen
  • Digestive enzymes
    • Produced by rough ER in acinar cells
    • Stored in zymogen granules as pro-enzymes while awaiting release
    • Enzymes
      • Proteases
        • Includes trypsinogen and chromotripsinogen (converted to active form by enterokinase in duodenum)
        • Hydrolyse peptide bonds between amino acids
      • Amylase
        • Secreted in active form
        • Hydrolyses glycogen, starch other carbohydrate complexes > disaccharides
      • Lipases
        • Lipase and phospholipase
        • Hydrolyses TGs to glycerol and fatty acids
      • Other enzymes
        • Elastase (breaks down eleastic tissue)
        • Ribonuclease/deoxyribonuclease (break down RNA/DNA)
  • Water and electrolytes


Control of exocrine secretions

  • Neural and hormonal control
  • Cephalic phase
    • Thought, taste, sight, smell food > increased PSNS (vagal activity)
    • Vagal (ACh mediated) efferents innervate the acinar cells
    • Leads to release of pancreatic juice (~20%)
  • Gastric phase
    • Mechanical stretch of stomach by food
    • Leads to increased PSNS (Vagal activity) + Gastrin release (from G cells in stomach/duodenum)
    • Leads to release of pancreatic juice (~10%)
  • Intestinal phase
    • Acidification of duodenum (from stomach acid) > increased pancreatic exocrine secretion
    • Mediated by secretin (released from S cells of duodenum) and CCK (enteroendocrine cells in duodenum)
    • Major factor which leads to increased secretion of pancreatic juice.
  • Inhibitory factors
    • Glucagon
    • Somatostatin


Examiner comments

33% of candidates passed this question.

Most candidates were able to mention some pancreatic enzymes, though often in insufficient detail to attract full marks. The amount, type, pH, etc. of pancreatic secretions was often not included. Many candidates did not describe the stimuli for pancreatic secretion. Better answers described the cephalic, gastric and intestinal phases of pancreatic secretion.


Online resources for this question


Similar questions

  • None



Question 14

Question

Outline the classification and effects of beta-blocking drugs with examples (50% of marks). Compare and contrast the pharmacokinetics of metoprolol with esmolol (50% of marks).


Example answer

Classification of beta blockers

  • All beta blockers are competitive antagonists
  • Can be classified according to
    • Receptor selectivity
      • Non selective (B1 and B2) e.g. sotalol, propranolol
      • B1 selective e.g. metoprolol, esmolol, atenolol
      • A and B effects: labetalol, carvedilol
    • Membrane stabilising effects
      • Stabilising e.g. Propanolol, metoprolol, labetalol
      • Non stabilising e.g. atenolol, esmolol, bisoprolol
    • Intrinsic sympathomimetic activity
      • ISA e.g. labetalol, pindolol
      • Non ISA e.g. metoprolol, sotalol, propranolol, esmolol


Effects of beta blockers

  • B1 antagonism
    • Heart: decreased inotropy and chronotropy (decreased BP), decreased myocardial oxygen consumption + increased supply (increased diastolic time), decreased dromotropy
    • Kidneys: decreased renin release > decreased RAAS activation > decreased BP
  • B2 antagonism
    • Respiratory: bronchoconstriction
    • Circulation: vasoconstriction
    • Skeletal muscle: reduced glucose uptake
    • Eye: decreased aqueous humour production > decrease
  • B3 antagonism
    • Adipose tissue: reduced lipolysis


Compare/contrast metoprolol and esmolol pharmacokinetics

Name Metoprolol Esmolol
Pharmacokinetics
Onset Immediate when IV Immediate (only given IV
Absorption 95% absorption, 50% oral bioavailability (1st pass effect) 0% oral bioavailability
Distribution VOD 5 L/kg

10% Protein bound
High lipid solubility, readily crosses BBB

VOD 3L/kg

60% protein bound
High lipid solubility, can cross BBB

Metabolism - Hepatic CYP450

- Significant 1st pass metabolism.
- Inactive metabolites

- Blood

- Hydrolysis by RBC esterase > inactive metabolites

Elimination Renal excretion

T 1/2 approx 4 hours

Renal excretion
T 1/2 10 mins


Examiner comments

47% of candidates passed this question.

Beta-blocking drugs were generally well classified. Selectivity, membrane stabilising activity and ISA should have been mentioned. Many candidates omitted or poorly answered the ‘effects’ of beta blockers. Candidates who performed well answering the pharmacokinetics of metoprolol and esmolol provided a table of the two drugs. Superficial statements such as “hepatic metabolism and renal excretion” attracted minimal marks. The mechanism of action of beta blockers was not requested


Online resources for this question


Similar questions

  • Question 9, 2021 (2nd sitting)



Question 15

Question

Define clearance and hepatic extraction ratio (30% of marks). Describe the role of the liver in drug clearance with examples (70% of marks).


Example answer

Clearance

  • The volume of plasma that is cleared of a drug per unit time (ml/min)

  • <math display="inline">Clearance = \frac {elimination \ rate}{plasma \ concentration}</math>

Hepatic Extraction Ratio (HER)

  • The fraction of drug entering the liver in the blood that is irreversibly removed as it is filtered through during one pass
  • Can be expressed by

<math display="block">ER (Hepatic) = \frac {FU \times Cl_{int} } {Q_H \; + \; FU \; \times Cl_{int}}</math>

  • Whereby
    • FU = fraction of unbound drug in plasma (drug bound to protein cannot be cleared)
    • <math display="inline">Cl_{int}</math> = intrinsic hepatic enzymatic capacity
    • <math display="inline">Q_H</math> = hepatic blood flow


Hepatic clearance

  • The two major determinants of hepatic clearance are the HER and the hepatic blood flow

<math display="block">Clearance_{Hepatic} = Q_H \times ER_{Hepatic}</math>

  • Effect of hepatic blood flow changes in relation to HER
    • For drugs with low ER (e.g. diazepam, warfarin) increasing QH leads to:
      • Minimal increase in clearance (capacity limited)
      • Decreased hepatic ER (more pronounced relatively)
    • For drugs with high ER (e.g. propofol and GTN) increasing QH leads to:
      • Marked increase in clearance (flow limited)
      • Decreased hepatic ER (less pronounced relatively)
  • Role of liver in drug clearance
    • Liver is heavily involved in drug metabolism
      • Phase 1 metabolic reactions
        • Involves oxidation (loss of electrons), reduction (gain of electrons) and hydrolysis (addition of H2O molecule)
        • Driven predominately by the Cytochrome p450 system and esterases in liver
        • E.g. metabolism of Propofol, benzodiazipines, opioids, volatiles anaesthetics
        • There can be significant variability in activity of CYP enzymes which leads to variability in drug response (e.g. CYP2C19 polymorphism > variability in phenytoin and clopidogrel metabolism)
  • Phase 2 reactions

    • Involves conjugation (increasing water solubility)

      • Typically occurs in hepatic endoplasmic reticulum

      • Includes glucuronidation (e.g. morphine), sulfation (e.g. quetiapine), acetylation (e.g. hydralazine), methylation (e.g. catecholamines)

Examiner comments

70% of candidates passed this question.

Clearance was generally well answered. It is the volume of plasma cleared of a drug per unit time, not the mass of drug cleared. An equation was helpful in identifying the relevant components of hepatic clearance.
ClHep=QH X ERHep
ERHep= FU x ClInt / QH + FU x ClInt
QH = hepatic blood flow
ERHep = hepatic extraction ratio
FU = fraction of drug unbound in plasma
ClInt = hepatic enzymatic capacity
Many candidates did not describe the effects of hepatic blood flow and intrinsic clearance on drugs with high and low hepatic extraction ratios. Some discussion of Phase I and II reactions was also expected.


Online resources for this question


Similar questions

  • Question 20, 2016 (1st sitting)


Question 16

Question

Compare the structure, function and coronary circulation of the right and left ventricles


Example answer

Structure/blood supply

Right ventricle left ventricle
Shape More triangular More conical
Valves Tricuspid (3 valve leaflets + papillary muscle)

Pulmonary (3 cusps: right, left, anterior)

Mitral (2 valve leaflet + papillary muscle)

Aortic (3 cusps: Right, left, posterior)

Wall thickness Relatively thinner (2-5mm) Relatively thicker (5-10mm)
Mass Lighter (25g) Heavier (100g) - due to increased afterload on LV
Position in chest Right/anterior Left/posterior
Arterial supply Predominately RCA Predominately LAD, LCx, PDA
Venous drainage Small + anterior cardiac veins Great and middle cardiac veins
Blood flow Constant, maximal flow rate during systole (majority still occurs in diastole due to increase duration) Intermittent (no flow in early systole), maximal flow rate + total flow occurs diastole


Function/Physiology

Right ventricle Left ventricle
Function Receive deoxygenated blood from systemic circulation > pump to pulmonary circulation Receive oxygenated blood from pulmonary circulation and pump to the systemic circulation
Blood flow RA > tricuspid valve > RV > pulmonary valve > pulmonary trunk LA > mitral valve > LV > aortic valve > aorta
C VO2 Lower Higher
EDV Higher Lower
ESV Higher Lower
Stroke volume Equal Equal
Systolic pressure Lower (~15-25mmhg) Higher (~120mmHg)
Diastolic pressure Lower (0-5mmhg) Higher (5-15mmHg)
ESPVR (contractility) Lower Higher
EDPVR (elastance) Lower Higher
Ea (afterload) Lower Higher
Work Lower Higher


Examiner comments

27% of candidates passed this question.

The question sought information on the structure (anatomy), function (physiology) and vascular supply of the right and left ventricle. Good answers provided detail in each section e.g. values for ventricular pressure rather than simply stating “high- and low-pressure systems”.
Many marks may be gained by a simple anatomical description & labelled PV loop for each ventricle. Many candidates focussed solely on the coronary circulation, to which only a proportion of the marks were allocated.


Online resources for this question


Similar questions

  • None



Question 17

Question

Explain respiratory compliance and outline the factors that affect it.


Example answer

Respiratory compliance

  • <math display="inline">Compliance \; = \; \frac{\Delta \;volume}{\Delta \; pressure}</math>
  • Compliance in the respiratory system (CRS) is a function of lung (Clung) and chest wall (CCW) compliance
    • <math display="inline">\frac {1}{C_{RS}}\; = \; \frac {1}{C_{Lung}} \; + \; \frac {1}{C_{CW}}</math>
    • Chest wall and lung compliance are roughly equal in healthy individual
  • Normal compliance of the respiratory system is approximately 200mls.cmH2O
  • Static compliance
    • Compliance of the respiratory system at a given volume when there is no flow
  • Dynamic compliance
    • Compliance of the system when there is flow (respiration)
    • Will always be less than static compliance due to airway resistance
    • At a normal RR is approximately equal to static compliance
  • Specific compliance
    • The compliance of the system divided by the FRC
    • Allows comparisons between patients which are independent of lung volumes


Factors effecting compliance

Chest wall

  • Increased
    • Collagen disorders such as Ehlers-Danlos syndrome
    • Cachexia
    • Rib resection
  • Decreased
    • Obesity
    • Kyphosis / scoliosis / Pectus excavatum
    • Circumferential burns
    • Prone positioning

Lung compliance

  • Increased

    • Normal ageing

    • Emphysema

    • Upright posture

    • Lung volume (highest compliance at FRC)

  • Decreased

    • Loss of surfactant (E.g. ARDS, hyaline membrane disease)

    • Loss of functional lung volume (e.g. pneumonia, lobectomy, pneumonectomy, atelectasis)

    • Pulmonary venous congestion (pHTN) and interstitial oedema (APO)

    • Reduced long elasticity (e.g. Pulmonary fibrosis)

    • Positioning (e.g. supine positioning)

Examiner comments

51% of candidates passed this question.

Answers were generally well structured. Better answers described lung and chest wall compliance and the pressures which are used to calculate compliance. Better answers displayed an understanding of dynamic, static and specific compliance and provided a reasonably comprehensive list of the physiological factors affecting chest and lung compliance.


Online resources for this question


Similar questions

  • Question 14, 2017 (1st sitting)
  • Question 15, 2014 (1st sitting)
  • Question 7, 2011 (2nd sitting)
  • Question 13, 2007 (1st sitting)



Question 18

Question

Compare and contrast the pharmacology of metaraminol and noradrenaline


Example answer

Name Noradrenaline Metaraminol
Class Endogenous catecholamine Synthetic non-catecholamine
Indications Vasopressor (Hypotension/shock) Vasopressor (hypotension/shock)
Pharmaceutics Clear solution. 1:1000. Brown ampule (prevent light oxidation). Diluted in dextrose. Clear solution. Typically 0.5mg/ml syringes, or 10mg/ml vials.
Routes of administration IV only (central vein) IV, IM
Dose Infusion titrated to effect (generally 0 - 0.5 mcg/kg/min) 0.5mg boluses, infusion
pKA 8.85 8.79
Pharmacodynamics
MOA Predominately Alpha 1 agonism. Some beta adrenergic receptor agonism. a1 > B1 > B2 Direct and indirect alpha-1 agonism (very weak B agonism)
Effects CVS: peripheral vasoconstriction > increased SVR > inc. BP CVS: peripheral vasoconstriction > increased SVR > increased BP. Also increased PVR. Reflex bradycardia.
Side effects CVS: Hypertension, reflex bradycardia, increased afterload

Extravasation > necrosis
Renal, hepatic vasoconstriction > decreased blood flow

CVS: Increased afterload > worsen heart failure, bradycardia
Pharmacokinetics
Onset Immediate Immediate
Absorption IV only (0% oral bioavailability) IV (though some oral bioavailability)
Distribution Does not cross BBB. 25% taken up in lungs. VOD = 4L/kg

45% protein bound

Metabolism Readily metabolised intro adrenaline by MAO and COMT Not metabolised
Elimination Excreted in urine as inactive metabolites (>85%).

Half life ~2 mins

Minutes, renal elimination
Special points Tachyphylaxis (slow)

Effect exaggerated in patients taking MAOI (less breakdown)

Tachyphylaxis (fast( with infusion))


Examiner comments

71% of candidates passed this question.

Marks were distributed across pharmaceutics, uses, dose & administration, mechanism of action, Pharmacokinetcs and Pharmacodynamics. Common omissions were doses/rates of infusion, effects other than on heart/SVR (e.g. splanchnic, renal blood flow), indirect effect of metaraminol, receptor effect of noradrenaline other than alpha 1 and tachyphylaxis.


Online resources for this question


Similar questions

  • Noradrenaline
    • Question 12, 2009 (2nd sitting)
    • Question 7, 2007 (1st sitting)
    • Question 23, 2011 (1st sitting)
    • Question 10, 2020 (1st sitting)
    • Question 15, 2016 (2nd sitting)
  • Metaraminol
    • New!



Question 19

Question

Describe the pharmacology of atropine.


Example answer

Name Atropine
Class Naturally occurring tertiary amine. Muscarinic antagonist.
Indications Bradycardia

Organophosphate poisoning
Counteract muscarinic effects of anticholinesterases
drying of secretions

Pharmaceutics Clear colourless solution. 600mcg/ml. Racemic mixture with the L-isomer being active
Routes of administration IV, IM, topical (eye)
Dose 600mcg - repeated administration can be given
Pharmacodynamics
MOA Competitive antagonism of muscarinic anticholinergic receptors
Effects CVS: increased HR (and CO), decreased AV conduction time

RESP: bronchodilation
GIT: Drying of secretions

Side effects CNS: Hallucinations, confusion, amnesia, delirium, central anticholinergic syndrome

GIT: dry mouth, delayed GIT motility
CVS: may cause initial transient bradycardia when given slowly
MSK: inhibits sweating

Pharmacokinetics
Onset Seconds. Duration 2-3hours
Absorption IV
Distribution 50% protein bound. VOD=3L/kg. Crosses blood brain barrier and placenta
Metabolism Extensive hepatic hydrolysis into tropine and tropic acid
Elimination Renal elimination of metabolites. T 1/2 approx 2 hours
Special points


Examiner comments

53% of candidates passed this question.

Most candidates used a good structure to compose their answer. Better candidates understood that CNS effects occur as atropine is a tertiary amine that crosses the blood brain barrier. The mechanism of action was required. Indications for use should have included bradycardia, organophosphate poisoning, drying of secretions etc. Reasonably extensive details regarding pharmacodynamics was expected, including potential toxic effects. There was limited knowledge regarding pharmacokinetics.


Online resources for this question


Similar questions

  • Question 3, 2011 (1st sitting)



Question 20

Question

Compare the pharmacology of piperacillin-tazobactam and ciprofloxacin


Example answer

Name Piperacillin-Tazobactam Ciprofloxacin
Class Semi-synthetic penicillin (piperacillin)

B-lactamase inhibitor (tazobactam)

Fluroquinolone
Indications Pseudomonal infection

Broad spectrum antimicrobial cover of severe infections/sepsis

Effective for many infections (skin, joint, gastro, UTI, LRTI)
Pharmaceutics Powder, reconstitutes in water/NaCl/glucose Tablet (250-750mg) or yellowish powder for dilution.
Routes of administration IV/IM IV, PO
Dose 4g/0.5g 8hrly or 4g/0.5g 6hrly (pseudomonas cover)

Dose reduced renal failure

250-750mg BD (PO), 200-400mg BD/TDS (IV)
Pharmacodynamics
MOA Piperacillin: bactericidal - inhibits cell wall synthesis by preventing cross linking of peptidoglycans by replacing the natural substrate (D-ala-D-ala) with their B-lactam ring

Tazobactam: B lactamase inhibitor (prevents piperacillin degradation)

Bactericidal - Inhibits DNA gyrase and topoisomerase IV > inhibits DNA synthesis
Antimicrobial cover Broad spectrum coverage of gram positive bacteria, gram negative bacteria, anaerobes. Covers pseudomonas.

Doesn't cover: MRSA, VRE, ESBL, atypical

Broad spectrum (GN > GP).

Effective against pseudomonas + anthrax. Effective against some atypicals (legionella, mycoplasma).
No anaerobe cover.

Side effects GIT: diarrhoea, nausea, vomiting

Renal: AKI
Allergy (up to 10%), rash most common, skin eruptions/SJS and anaphylaxis (<1/10,000)

MSK: tendon rupture, arthritis, myalgia

CNS: peripheral neuropathy, headache
GIT: nausea, vomiting, abdominal pain, dyspepsia
CVS: Qtc prolongation, arrhythmias
RENAL: AKI, nephritis

Pharmacokinetics
Absorption Minimal oral absorption > IV

Peak concentrations immediately after dose.

70% oral bioavailability
Distribution Very good tissue penetration (minimal CNS without active inflammation)

Low protein binding (<30%)

Low protein binding (25%). Great tissue penetration. VOD 2.5L/kg.
Metabolism Piperacillin: not metabolised

Tazobactam: metabolised to M1, an inactive metabolite

Limited hepatic metabolism (15%)
Elimination Renal (80% unchanged)

T 1/2 2 hrs

Renal excretion of metabolites. T1/2 3-5 hours.
Special points Removed by haemodialysis Worldwide resistance to quinolones is increasing


Examiner comments

58% of candidates passed this question.

This question was most effectively answered using a tabular format. Only a minority of candidates demonstrated a comprehensive knowledge of these level 1 drugs and very few candidates compared the two in areas which lent themselves to comparison. The spectrum of activity generally lacked detail. Few candidates mentioned that piperacillin-tazobactam had superior gram-positive cover, both have extensive gram-negative cover including Pseudomonas. Piperacillin-tazobactam is effective against anaerobes; whilst ciprofloxacin has some atypical cover against Mycoplasma.
The mechanism of action was generally well described for piperacillin; many candidates incorrectly stated the mechanism of action for ciprofloxacin, confusing the drug with a macrolide. Better answers included time- dependant and concentration-dependent killing. The concept of half-life was frequently confused with the dosing interval.
Minimal marks were awarded for “allergy” and “gastrointestinal side-effects”. Better candidates mentioned Liver function derangement, neutropenia, interstitial nephritis for piperacillin and tendonitis for ciprofloxacin.


Online resources for this question


Similar questions

  • PipTaz: Question 16, 2021 (1st sitting)
  • Cipro: Question 5, 2020 (1st sitting)



2019 (1st sitting)

Question 1

Question

Describe the pharmacology of lignocaine.


Example answer

Name Lidocaine (lignocaine)
Class Amide anaesthetic / Class 1b antiarrhythmic
Indications Local/regional/epidural anaesthesia, ventricular dysrhythmias
Pharmaceutics Clear colourless solution (1%, 2%, 4%). Can come with/without adrenaline. Also available as cream/spray
Routes of administration SC, IV, epidural, inhaled
Dose Regional Use: Toxic dose 3mg/kg (without adrenaline), 7mg/kg (with adrenaline)

IV use: 1mg/kg initially, then ~1-2mg/kg/hr

pKA 7.9, 25% unionised at normal body fluid pH
Pharmacodynamics
MOA Class 1b anti-arrhythmic: blocks Na channels, raising threshold potential + reducing slope of Phase 0 of action potential, shortened AP

Local anaesthetic: binds to, and blocks, internal surface of Na channels

Effects Analgesic, anaesthetic, anti-arrhythmic
Side effects CNS: headache, dizziness, confusion, paraesthesia, reduced LOC, seizures

CVS: hypotension, bradycardia, AV Block, arrhythmia
CC/CNS ratio = 7 (lower number = more cardiotoxic)

Pharmacokinetics
Onset Rapid onset (1-5 minutes)
Absorption IV > Epidural > subcut. Oral bioavailability 35%
Distribution 70% protein bound, Vd 0.9L/kg. Crosses BBB
Metabolism Hepatic, some active metabolites
Elimination Metabolites excreted in urine. Half life ~90mins. Increased with adrenaline (SC). Reduced in cardiac/hepatic failure.
Special points


Examiner comments

16% of candidates passed this question.

Comprehensive answers included uses (including antiarrhythmic action and a role in analgesia), physical properties and preparations, pharmacodynamics and pharmacokinetics. Its mode of action should also have been described. Many candidates focussed on toxicity and its management but provided little information on pharmacodynamics and pharmacokinetics, commonly omitting factors which affect its systemic absorption. Other common omissions were the dose required for its local anaesthetic effect and for its antiarrhythmic effect


Online resources for this question


Similar questions

  • Question 17, 2014 (1st sitting)

  • Question 2, 2021 (2nd sitting)


Question 2

Question

Outline the components required to measure blood pressure from an intra-arterial catheter (75% of marks). What information (other than blood pressure) may be gained from an arterial line trace (25% of marks)?


Example answer

Components

  • Intra-arterial catheter
    • narrow (generally 18-22G) and relatively short - minimises resonance
    • Provides conduit to transmit blood pressure wave to circuit
  • Fluid filled tubing
    • Permits hydraulic coupling of mechanical signical
    • Non compressible fluid filled (usually saline) tubing minimises damping
  • Counterpressure bag with flush system
    • pressure of~300mmHg - counteracts arterial pressure
    • Fluid provides slow continuous infusion to maintain catheter patency (prevent clots etc)
    • Can be used for diagnostic/trouble shooting purposes with 'fast flush test'
  • An electrical transducer
    • Usually a Wheatstone bridge peizoresistive transducer
    • Movement of the diaphragm caused by arterial pressure changes leads to stretching/compression of the strain gauges and is converted into an electrical signal
    • Placed at phlebostatic axis and requires calibration
  • Microprocessor + amplifier
    • Processor uses Fourier analysis to break down the waveform into component sine waves which are reconstructed with 8-10 harmonic sine waves
    • Amplifies signal
  • Cabling
    • To transmit the information/electrical signals
  • Monitor
    • To visualise the information (including pressures and waveform)
  • 3 way tap
    • Allows sampling of arterial blood for diagnostic purposes
    • Allows 'zeroing' to atmosphere for calibration


Information gained

  • Heart rate
  • Heart rhythm - regular or irregular
  • Blood pressures - Systolic pressure, diastolic pressure, mean arterial pressures, pulse pressures
  • Pulse pressure variation
  • Issues with system - Dampened trace may indicate kinks, bubbles, clots in the circuit
  • Cardiac output, stroke volume, stroke volume variation by pulse contour analysis (e.g. FloTrac)
  • Waveform may indicate underlying pathology with modest accuracy (e.g. collapsing wave in AS)


Examiner comments

44% of candidates passed this question.

Most of the marks were allocated to the components of the measuring system (as detailed in the question), hence a level of detail was required. An explanation of how the various components work was required; e.g. hydraulic coupling and transducers. Some candidates forgot to include heart rate as a piece of information derived from the trace.


Online resources for this question


Similar questions

  • Previous questions on damping/resonance - nil on arterial line setup.



Question 3

Question

Compare and contrast fresh frozen plasma and prothrombin complex concentrate


Example answer

Name FFP Prothrombinex
Description Human plasma including all coagulation factors Human plasma derivative containing a concentrate of specific clotting factors
Preparation 1) Separation of whole blood or apheresis

2) Frozen and stored
3) Rethawed in water bath prior to use

1) Separation of whole blood or apheresis

2) Separation of clotting factors II, IX and X via ion exchange chromatography
3) Freeze dried powder

Indications Coagulopathy

Plasma exchange
ACE-I angioedema, suxamethonium apnoea

Warfarin reversal

Correction of coagulopathy from factor II, IX or X deficiency

Pharmaceutics 250-300ml bags, labelled with donor blood types Glass vial with powdered concentrate for reconstitution with water. Generally 500U per vial

Contains small amount of heparin

Storage Stored for 12 months Stored for 6 months
Routes of administration IV IV
Dose 2-4 units (varies) 25-50 u/kg
Contraindications ABO incompatibility DIC, HITS (contains heparin), liver failure
Contents/factors All clotting factors (except fibrinogen) Contains factors II, IX, X (500 units each)
Adverse effects Blood product, with all the risks associated with this (fluid overload, infection, allergic responses) Allergic or anaphylactic reactions

Thrombosis in predisposed individuals

Pros Contains all necessary clotting factors

Less expensive than PTX

Does not need group/crossmatch (therefore available for immediate use)

Smaller fluid volume

Cons Requires ABO grouping

Requires time for thawing etc
More fluid, more side effects

Factor 7 absent

More expensive than FFP


Examiner comments

10% of candidates passed this question.

Very few answers included details on prothrombin complex concentrate which meant it was difficult to score well. Useful headings included preparation and administration, dose, indications and adverse effects. Not many candidates knew the dose of FFP, and few were able to describe the preparation/production of the product. Few candidates knew the factors available from either product. Commonly missed was the need for ABO typing for FFP and that Prothrombin complex concentrate did not require this.


Online resources for this question


Similar questions

  • ? None



Question 4

Question

Outline the functional anatomy of the kidney (40% of marks). Outline the regulation of renal blood flow (60% of marks)


Example answer

Gross anatomy of kidney

  • Gross anatomy
    • Paired retroperitoneal organ
    • Sits at ~ T12-L3 (right lower than left)
  • Structure
    • Outer fibrous capsule > Outer 'cortex' > Inner 'medulla' > renal pelvis
  • Blood supply
    • Arterial: renal arteries from abdominal aorta
    • Venous: renal veins > IVC
  • Innervation
    • SNS (T9-13)


Functional anatomy

  • The nephron is the basic functional unit of kidney (~1-2 million nephrons per kidney)
    • 85% of nephrons are predominately located in cortex (cortical nephrons)
    • 15% are juxtamedullary and extend deep into the medulla.
  • Nephron structure
    • Renal corpuscle (filtration): with glomerulus + bowman's capsule
    • Juxtaglomerular apparatus (adjustments to GFR): contains macula densa, JG cells, mesangial cells
    • Tubular system (reabsorption/regulation): PCT > LOH > DCT
    • Collecting duct system
  • Vascular
    • Afferent arteriole: blood supply to individual nephron
    • Efferent arteriole: carries blood away from individual nephron


Renal blood flow regulation

  • Normal RBF = ~20% of CO (~1L / min), predominately distributed to cortex > medulla
  • Kidneys are able to autoregulate (maintain constant blood flow) across a wide range of MAP (~70-170mmhg)
  • Myogenic regulation
    • Intrinsic constriction of afferent arterioles in response to increased transmural pressure of vessel wall (e.g. increased BP)
  • Tubuloglomerular feedback
    • Regulated by macula densa
    • Increased perfusion pressure > Increased Na/Cl sensed by macula densa > adenosine released > constriction > decreased GFR. Vice versa (except NO is released with decreased Na/Cl delivery to vasodilate and increased GFR)
  • Neuronal control
    • SNS activation > afferent and efferent arteriole constriction > decreased flow
  • Hormonal control
    • Renin-angiotensin system: Renin released (SNS stimulation, hypotension, decreased Na at JGA) > increased AG1 > increased AG2 > constriction of arterioles > decreased RBF/GFR


Examiner comments

71% of candidates passed this question.

It was expected that answers include sections on the blood supply, the nephron (including the difference between the cortical and juxta-medullary nephrons) and innervation. A number of candidates failed to quantify renal blood flow and to define autoregulation. The concept that it’s the flow that’s regulated was not described by some. Tubuloglomerular feedback was generally described correctly but a reasonable number had the blood flow increasing when an increased sodium was sensed at the macula densa.


Online resources for this question


Similar questions

  • Renal blood flow and autoregulation
    • Question 18, 2007 (1st sitting)
    • Question 12, 2008 (2nd sitting)
    • Question 11, 2012 (1st sitting)
    • Question 3, 2015 (2nd sitting)
  • Functional anatomy of kidneys
    • Question 21, 2011 (2nd sitting)
  • Functional anatomy + autoregulation of blood flow
    • Question 18, 2017 (1st sitting)



Question 5

Question

Define volume of distribution (15% of marks). Outline the factors affecting volume of distribution (60% of marks) and explain how it may be measured (25% of marks).


Example answer

Volume of distribution

  • The theoretical volume in which an amount of drug would distribute to produce an observed plasma concentration
  • Note: Does not correspond to any real volume. Can often exceed total body water


Measurement

  • Assumes that

    1. The drug is evenly distributed (often not the case)

    2. Metabolism and elimination have not taken place (often not the case)

  • Requires drug dose to be given, then plasma samples to be taken

  • Semilogarithmic plasma concentration vs time curve plotted.

  • In a single compartment model, VOD can then be calculated as VOD = dose given / plasma concentration at time 0 (which is back extrapolated on the curve from 1st time point.)

    File:C:\Users\ethan\AppData\Roaming\Typora\typora-user-images\image-20220804131048030.png

Factors affecting volume of distribution

  • Patient factors
    • Age: decreasing water content with age = decreased Vd (water soluble drugs)
    • TBW: decreases with age = decreased Vd (water soluble drugs)
    • Fat %: increased body fat = increased Vd of lipophilic drugs
    • Gender: women generally have lower Vd (water soluble drugs) due to lower TBW but higher Vd for fat soluble drugs
  • Drug factors
    • Molecular size: decreased size = increased Vd
    • Lipid solubility: increased lipophilicity = increased Vd
    • pKa: basic drug in low pKa = increased Vd
    • Protein binding: increased protein binding = decreased Vd
    • Charge: ionised molecules > may be traped in central compartment > decreased Vd
  • Logistical factors
    • Timing of measurement
    • Modelling used (e.g. single compartment)
  • Pathological factors
    • Renal failure/hepatic failure: may lead to low protein = decreased Vd
    • Oedema, ascites - reservoir for water soluble drugs


Examiner comments

51% of candidates passed this question.

The first two parts of the question were reasonably done. Most candidates had well-structured answers which included drug factors and patient factors. In addition to listing the factors it was expected candidates state how these factors affect volume of distribution. Explaining how volume of distribution is determined was not so well done.


Online resources for this question


Similar questions

  • Question 12, 2015 (second sitting)



Question 6

Question

Outline the physiology of the adrenal gland (70% of marks). Describe the actions of aldosterone (30% of marks).


Example answer

Adrenal gland

  • Paired organs, immediately superior to kidneys
  • Broken up into an outer adrenal cortex (80%) and an inner adrenal medulla (20%)


Adrenal cortex

  • Three zones of cells
  • Zona glomerulosa
    • Outermost zone of cortex
    • Synthesises mineralocorticoids (e.g. aldosterone)
    • Important for regulation of electrolytes (Na, K) and water balance
    • Regulated by ACTH from anterior pituitary, angiotensin II and plasma potassium levels
  • Zone fasiculata
    • Secretes glucocorticoids (e.g. cortisol ~95% of activity)
    • Widely important, particularly for metabolism and cardiovascular function (HR, BP etc)
    • Regulated by ACTH from the anterior pituitary gland
  • Zona reticularis
    • Innermost zone of cortex
    • Secretes androgen precursors (e.g. DHEA) which get converted into testosterone and oestrogen
    • Regulated by ACTH and androgen stimulating hormones


Adrenal medulla

  • Innermost portion of the adrenal gland
  • Responsible for producing catecholamines (adrenaline, noradrenaline)
  • Chromaffin cells (modified neuroendocrine cells) are responsible for storing/synthesising the catecholamines
  • Regulated by SNS activity from T5-T11 (thus stress, hypoglycaemia, etc can activate)


Aldosterone

  • Primary mineralocorticoid hormone from adrenal gland (90% of activity)

  • Actions of aldosterone

    • Increases reabsorption of Na in the DCT and CD (principle cells)

    • Increases secretion of potassium in the DCT and CD (principle cells)

    • Increased Na reabsorption in sweat glands, salivary glands and GIT

    • Increases ECF volume (by increased H2O reabsorption by osmosis with the Na)

    • Increased H+ excretion in DCT (leads to Cl reabsorption and metabolic alkalosis)


Examiner comments

43% of candidates passed this question.

Lack of breadth and detail were in many of the answers. Physiology of the adrenal gland includes an outline of the adrenal medulla, the types of chromaffin cells, hormones secreted and how secretion is stimulated. The three zones of the adrenal cortex should have been outlined including substances secreted, their function and again how their secretion is stimulated. The actions of aldosterone should have been described; a comment on sodium and water excretion was insufficient to attain many marks for this section. The extra-renal actions of aldosterone were missing from most answers.


Online resources for this question


Similar questions

  • None ?



Question 7

Question

Compare and contrast the pharmacokinetics and pharmacodynamics of midazolam and dexmedetomidine.


Example answer

Name Midazolam Dexmedetomidine Notes
Class Benzodiazepine (sedative) Central alpha agonist (sedative) Diff. classes
Indications Anaesthesia, sedation, treatment of seizures, anxiolysis Short term sedation and anxiolysis Dexmed = short term and no anticonvulsant / amnesic properties
Pharmaceutics IV: clear solution, pH 3.5. Diluted in water. Clear colourless isotonic solution. Or white powder for dilution
Routes of administration IV, IM, S/C, intranasal, buccal, PO IV only in AUS Midaz has more routes available
Dose Dose depends on many pt. factors. 1-5mg premedication. 2.5-10mg seizures. Infusions. Infusion (though loading boluses can be given)
pKa 6.5 7.1
Pharmacodynamics
MOA Midazolam (BZD) binds to GABAA receptors (ionotropic ligand gated channel) in the CNS. Cl enters > hyperpolarisation. Selective central a2 agonism (predom. at the locus coeruleus and spinal cord) Different receptors
Effects CNS: sedation, amnesia, anticonvulsant effects, decreased cerebral O2 demand CNS: Sedation, anxiolysis No amnesia with dexmed
Side effects CVS: bradycardia, hypotension

CNS: confusion, restlessness
RESP: respiratory depression/ apnoea

CVS: hypotension, bradycardia

Other: hyperthermia, confusion, dry mouth

Bradycardia worse with dexmed. No Resp depression with dexmed.
Pharmacokinetics
Onset peak effect 2-3 minutes (IV) ~30 mins (without bolus) Midaz = quicker onset
Absorption ~40% oral bioavailability

Absorbed well, but sig. 1st pass metabolism

IV only in Aus. Low PO bioavailability Midaz has greater PO bioavailability
Distribution 95% protein bound, very lipid soluble

Vd = 1L / kg

95% protein bound, very lipid soluble

Vd = 1.3L/kg

Similar
Metabolism Hepatic metabolism by hydroxylation

Active (1-a hydroxymidazolam) and inactive metabolites

Biotransformation (direct glucuronidation and CYP450 metabolism) > inactive metabolites Midas has active metabolites
Elimination Renal excretion

T 1/2 = 4 hours

Renal excretion (5% stool)

t 1/2 = 2 hours

Similar
Special points Flumazenil - antagonist (reversal agent) Atipamezole = antagonist (reversal agent)


Examiner comments

27% of candidates passed this question.

Most candidates used the effective tabular format presenting pharmacokinetics and pharmacodynamics of each drug side by side. Many answers demonstrated a lack of correct detail with respect to the pharmacokinetics and pharmacodynamics of these two level 1 drugs. Many included pharmaceutics which attracted no marks as it was not asked.


Online resources for this question


Similar questions

  • Midazolam
    • Question 9, 2019 (2nd sitting)
    • Question 4, 2018 (2nd sitting)
    • Question 24, 2016 (1st sitting)
    • Question 2, 2008 (2nd sitting)
  • Dexmed
    • Question 22, 2015 (1st sitting)
    • Question 5, 2012 (1st sitting)
    • Question 2, 2008 (2nd sitting)



Question 8

Question

Compare and contrast the measurement (40% of marks) and interpretation (60% of marks) of both central venous and mixed venous oxygen saturations.


Example answer

Central venous oxygen saturations (ScvO2)

  • The oxygen saturation of haemoglobin at the cavo-atrial junction
  • Normally measured using a CVC
  • ScvO2 is normally ~70% (slightly lower than SmvO2 in well patients due to higher oxygen extraction from upper body)
  • ScvO2 is used as a surrogate for SmvO2 as it is more accessible for most ICU patients as need a CVC not PAC


Mixed venous oxygen saturation (SmvO2)

  • The oxygen saturation of haemoglobin when measured in the pulmonary artery (after venous mixing in the right ventricle)
  • Measured using a pulmonary artery catheter
  • SmvO2 is normally ~75%
  • SmvO2 provides better idea of whole body venous O2 sats (blood from SVC, IVC and coronary sinus)
  • Can be used to estimate the cardiac output via the modified fick equation
    • CO = oxygen consumption / (Oxygen content arterial blood - oxygen content mixed venous blood)


Measurement

  • Both can be measured using the same methods
    • Intermittent sampling
      • ABG: derivation of the SvO2 value from the PO2, pH and pCO2, using the oxygen-haemoglobin dissociation curve.
      • Co-oximetry: measures absorption of near-IR light by haemoglobin species, and the use of the Beer-Lambert law to calculate the concentrations of oxyhaemoglobin and deoxyhaemoglobin
    • Continuous monitoring
      • Using a CVC or PAC with with fibre optic reflectance spectrophotometer
      • Near IR light reflectance strength used to determine ratio of Oxy and deoxy Hb


Interpretation of ScvO2 / SmvO2

  • Increased saturation
    • Anaesthesia
    • Septic shock
    • Cyanide toxicity
    • High output cardiac failure
    • Hypothermia
    • Severe liver disease
  • Decreased saturation
    • Cardiogenic shock
    • Septic shock
    • Hyperthermia
  • Importantly, not sensitive to regional hypoxia/dysoxia


Evidence

  • No evidence to support targeting ScvO2 or SmvO2 saturations at present


Examiner comments

8% of candidates passed this question.

Many candidates did not appreciate that ScvO2 refers to SVC / RA junction venous oximetry and not femoral or peripheral venous oximetry. Methods of measurement such as co-oximetry and reflectance spectrophotometry needed to be explained. Marks were awarded for the normal values. Discussion of the relationship between ScvO2 and SmvO2 and changes during shock attracted marks. Better answers quoted the modified Fick equation and related this to cardiac output and factors affecting oxygen consumption versus delivery.


Online resources for this question


Similar questions

  • ? None



Question 9

Question

Classify antibiotics with respect to their mechanism of action (50% of marks). Outline the mechanisms of antimicrobial resistance (50% of marks). Give specific examples of each.


Example answer

Classification of antibiotics

  • Inhibitors of cell wall synthesis
    • Beta-lactams: e.g. flucloxacillin
    • Cephalosporins: e.g. ceftriaxone
    • Carbapenems: e.g. meropenem
    • Monobactams: eg. aztreonam
    • Glycopeptides e.g. vancomycin
  • Inhibitors of cytoplasmic membrane function
    • Polymyxins: e.g. colistin
    • Lipopetides e.g daptomycin
  • Inhibitors of nucleic acid synthesis
    • Quinolones e.g ciprofloxacin
    • Rifamycins e.g. rifampicin
    • Nitroimidazoles e.g. metronidazole
  • Folate metabolism inhibitors
    • e.g. trimethoprim
  • Inhibitor of protein synthesis
    • Aminoglycosides: e.g. gentamycin
    • Tetracyclines: doxycycline
    • Lincosamides: e.g clindamycin
    • Macrolides: e.g. erythromycin


Antimicrobial resistance

  • Occurs when the maximal level of drug tolerated in insufficient to inhibit growth
  • Broadly occurs via genetic alteration or changes to protein expression


Mechanisms of resistance

  1. Prevent access to drug target
    • Decrease permeability
      • E.g. pseudomonas aeruginosa resistance to carbapenems due to reduction in porins
    • Active efflux of drug
      • Efflux pumps > extrude antibiotics (eg. fluoroquinolone resistance with E.coli)
  2. Alter antibiotic target site
    • Alteration to Peptidoglycan binding site protein, reducing affinity of drug.
    • E.g. Vancomycin and VRE (e.g. E. faecium)
  3. Modification / inactivation of drug
    • E.g. ESBL and penicillin's/cephalosporins whereby b-lactamases hydrolyse B-lactam rings
  4. Modification of metabolic pathways
    • Metabolic pathways bypass site of antibiotic action
    • E.g. Bactrim resistance (synthesise their own folic acid)


Examiner comments

70% of candidates passed this question.

This question was well answered. Marks were awarded for correct pairing of mechanism of action and resistance with examples of drug class. Few mentioned the mechanisms by which resistance is present; acquired or generated.


Online resources for this question


Similar questions

  • Question 5, 2020 (1st sitting)



Question 10

Question

Outline the sequence of haemostatic events after injury to a blood vessel wall (50% of marks). Discuss the role of naturally occurring anticoagulants in preventing clot formation in-vivo (50% of marks).


Example answer

  1. Vascular constriction
    • Occurs instantly, lasts a few minutes
    • Due to
      • Local myogenic spasm
      • Nervous reflexes
      • Release of local vasoconstrictors (e.g. Thromboxane A2)
    • Limits the amount of haemorrhage and creates environment suitable for clot formation


  1. Primary haemostasis (platelet plug formation)

    • Platelet adhesion

      • Exposed vWF in endothelium binds to glycoprotein receptor complex on platelets

      • Platelet GP1a binds to subendothelial collagen fibres by vWF bridge

    • Platelet activation

      • Activated following exposure to tissue factor, vWF and collagen

      • Results in them

        • Changing shape (large, more irregular, pseudopod formation) > assists with clot formation

        • Release molecules (Thromboxane A2, ADP, serotonin) > vasoconstricts + activates platelets

    • Platelet aggregation

      • Activated platelets bind fibringoen, vWF and fibronectin forming a soft platelet plug

  2. Secondary haemostasis (clot formation)

    • Two main models: classical (in vitro) model and modern cell based (in vivo) model

    • Cell based model

      • Initiation

        • Vessel damage exposes plasma to tissue factor

        • tissue factor binds to and activates factor VII

        • TF-Factor VIIa complex activates factor X

        • Factor X activates prothrombin > thrombin (small amounts)

      • amplification

        • This causes local activation of platelets (via vWF), Factor V, Factor VIII and factor XI

        • This greatly accelerates the production of thrombin around the surface of the platelets

      • Propagation

        • Begins with formation of tenase complexes on platelet surfaces (IXa-VIIIa) which greatly increases the rate of Factor X activation

        • The large amounts of Xa interacts with factor Va forming prothrombinase complex (Va-Xa) which catalyses the conversion of prothrombin to thrombin

        • Positive feedback loop


Natural anticoagulants --> prevent unnecessary coagulation

  • Antithrombin 3
    • Inactivates IIa and Xa
  • Protein C
    • Inactivates Va and VIIa
  • Protein S
    • Cofactor for upregulating protein C
  • Thrombomodulin
    • Bound to the endothelial membrane
    • Binds thrombin and activates protein C
  • Heparan
    • Activates AT-3, which in turn inactivates thrombin


Examiner comments

40% of candidates passed this question.

This question was best answered in a chronological manner. Many candidates omitted initial vasoconstriction and its mechanism. The platelet plug and formation of the clot should have then been described followed by the fate of the clot, including in-growth of fibroblasts. Strictly, fibrinolysis is a system for repairing / limiting clot propagation after the fact. Anticoagulants refer to antithrombin III, heparin, thrombomodulin and protein C and S. An explanation of the interaction of these naturally occurring anticoagulants was expected. The clotting factors that are specifically inhibited was expected as part of the discussion. The glycocalyx and vessel wall also plays a role in preventing coagulation.


Online resources for this question


Similar questions

  • Question 4 (part 2), 2009 (1st sitting)
  • Question 17, 2011 (1st sitting)



Question 11

Question

Describe the physiology of cerebrospinal fluid (CSF) (60% of marks). Describe the anatomy relevant to performing a lumbar puncture (40% of marks).


Example answer

CSF

  • ECF located in the ventricles and subarachnoid space
  • volume: ~2ml/kg
  • Divided evenly between the cranium and spinal column


Formation

  • Constantly produced (~24mls/hr)
  • Produced by choroid plexus (70%) and capillary endothelial cells (30%)
  • Produced by a combination of ultrafiltration (via fenestrated choroidal capillaries) and active secretion


Composition relative to plasma

  • Similar: Na, osmolality, HCO3
  • Increased: Cl, Mg, CO2
  • Decreased: pretty much everything else


Circulation

  • CSF flows from lateral ventricles > foramen of Monro > 3rd ventricle > Sylvian aqueduct > 4th ventricle > cisterna magna (via foramen megendie and luschka) > spreads between spinal/cranial subarachnoid spaces


Reabsorption

  • Reabsorption by the arachnoid villi located predominately in the dural walls of the sagittal + sigmoid sinuses (one way valves)

  • Reabsorbed at ~24mls/hr

Functions

  • Mechanical protection: low specific gravity of CSF > decreased effective weight of brain > no contact with skull base + less inertia forces
  • Buffering of ICP - CSF can be displaced / reabsorbed to offset increase in ICP
  • Stable extracellular environment for neuronal activit
  • Control of respiration: pH regulates respiration via central chemoreceptors
  • Nutrition: supply of oxygen, sugars, amino acids to supply the brain


Anatomy of LP

  • Positioning:
    • lateral decubitus or sitting position
  • Level:
    • L2-5 possible (below conus medullaris)
    • L3/4 or L4/5 are recommended (safety).
  • Surface landmarks:
    • Line between iliac crests (Tuffiers line) = L4/5
    • Line between PSISs = L3/4
    • Central positioning by spinous processes'
  • Angle of needle
    • Toward umbilicus (~15 degrees)
  • Order of tissues/structures passed through by needle
    • Skin
    • Subcut tissue
    • Supraspinous ligament
    • Interspinous ligament
    • Ligamentum flavum
    • Epidural space
    • Dura mater
    • Arachnoid mater
    • Subarachnoid space = CSF


Examiner comments

86% of candidates passed this question.

Better answers had a structure with headings such as function, formation, circulation, absorption and composition with dot point facts under each heading. The second part of the question lent itself to a diagram with labelling which scored well. Precise surface anatomy and mentioning all layers from the skin to the sub-arachnoid space scored well


Online resources for this question


Similar questions

  • Question 16, 2020 (1st sitting)

  • Question 15, 2018 (2nd sitting)

  • Question 9, 2017 (2nd sitting)

  • Question 24, 2017 (1st sitting)

  • Question 16, 2015 (1st sitting)

  • Question 2, 2013 (1st sitting)

  • Question 22, 2007 (1st sitting)

  • Question 6, 2008 (2nd sitting)


Question 12

Question

Compare and contrast the pharmacology of salbutamol and ipratropium bromide.


Example answer

Name Salbutamol Ipratropium bromide Notes
Class Short acting B2 agonist (synthetic sympathomimetic amine) Anticholinergic (quaternary ammonium derivative of atropine)
Indications Bronchoconstriction, hyperkalaemia, tocolytic Bronchoconstriction
Pharmaceutics Clear solution for neb, solution for IV (post dilution), powder / aerosol for inhalation, PO tablets Aerosol for inhalation, clear colourless solution for neb
Routes of administration Neb, IV, INH, PO Neb, INH Salbutamol can be given IV
Dose 2.5mg-5mg PRN (Neb)

200-400mcg PRN (INH)
0.5mch/kg.min (infusion)

Neb: 100-500mcg QID

INH: 100-500mcg BD

Salbutamol given more regularly
Pharmacodynamics
MOA B2 agonism > increased cAMP > decreased Ca > bronchial smooth muscle relaxation Competitive antagonism of muscarinic ACh receptors > bronchodilation + decreased secretions Can be used synergistically (different MOA)
Side effects CNS: anxiety, tremor

RESP: reverses HPVC
CVS: tachycardia
MET: hypoK (stimulates Na/K ATPAse), lactic acidosis

RESP: dry mouth, N, V

CNS: headache, blurred vision

Pharmacokinetics
Onset/duration Immediate, fast offset (mins) Peak effect 1-2 hours, lasts 6 hours Ipratropium has slow onset, longer duration of effect
Absorption 50% bioavailability 5% inhaled absorbed systemically Salbutamol can be given PO
Distribution VOD: ~150L/kg

10% protein bound
Can cross placenta

VOD: 4-5L/kg

Very weak protein binding

Salbutamol crosses placenta
Metabolism Metabolised in liver > inactive + active metabolites Metabolised in liver by CYP450 > inactive Salbutamol has active metabolites
Elimination Metabolites via urine + faeces
T 1/2: 4 hours
Metabolites via urine + faeces
Elimination half life 3 hours
Similar
Special points


Examiner comments

46% of candidates passed this question.

Overall candidates had a superficial knowledge of these level 1 drugs. To pass candidates needed to identify points of difference and overlap in various areas such as structure, pharmaceutics, pharmacokinetics, pharmacodynamics, mechanism of action, adverse effects and contraindications.


Online resources for this question


Similar questions

  • None directly
  • Bronchodilators more broadly have been asked for in
    • Question 4, 2016 (2nd sitting)
    • Question 11, 2014 (2nd sitting)
    • Question 9, 2010 (1st sitting)



Question 13

Question

Classify circulatory shock and provide examples (40% of marks). Outline the cardiovascular responses (60% of marks).


Example answer

Shock

  • Life threatening, generalised maldistribution of blood flow resulting in failure to deliver and/or utilise oxygen, leading to tissue dysoxia.


Classifications

  • Hypovolaemic
    • Caused by intravascular volume depletion
    • Includes haemorrhage, fluid loss (e.g. dehydration) and fluid shifts (e.g. pancreatitis)
  • Cardiogenic
    • Caused by cardiac pump failure or dysfunction
    • Includes: cardiomyopathy, ACS, arrhythmia, valve failure
  • Distributive
    • Caused by significant peripheral vascular dilation leading to fall in PVR
    • Includes: sepsis, inflammation (e.g. post cardiopulmonary bypass), anaphylaxis, neurogenic (e.g. high spinal cord injury)
  • Obstructive
    • Caused by circulatory obstruction/impedance
    • Includes: tamponade, tension pneumothorax, pulmonary embolism


Cardiovascular response to circulatory shock

Stimulus Sensor Integrator Effector
Hypotension Baroreceptors Nucleus of the solitary tract (NTS) - CNX inhibition (increased HR) - SNS activation (vasoconstriction, redistribution of BV, increased CO) - RAAS activation
Decreased VO2 Aortic arch chemoreceptors NTS As above
Decreased circulatory volume Atrial myocytes - Decreased release ANP
Decreased circulatory volume Baroreceptors Hypothalamus Increased release of vasopressin
Decreased circulatory volume Renal JG cells - Increased release of renin, RAAS activation
Inadequate tissue perfusion vascular SM and endothelium - Autoregulatory vasodilation


Examiner comments

83% of candidates passed this question.

Answers should have included the various types of shock and provided clear examples. Cardiovascular responses including sensor, integrator, effector mechanisms were necessary to pass.


Online resources for this question

Similar questions

  • ? None



Question 14

Question

Compare and contrast the mechanism of action, pharmacokinetics and adverse effects of digoxin and sotalol.


Example answer

Name Digoxin Sotalol Notes
Class Cardiac glycoside (antiarrhythmic) B-blocker Different class
Indications tachyarrhythmias (e.g. AF, SVT), heart failure tachyarrhythmias (e.g. AF, SVT) and ventricular arrhythmias Sotalol can be used for ventricular arrhythmias
Pharmaceutics 62.5mcg/250mcg (PO tablets), 25/250 mcg/ml (IV) 80+160mg PO tablets. IV in racemix mixture of enantiomers (through SAS). Digoxin available IV
Routes of administration IV and PO PO, IV (via SAS)
Dose Generally load with 250-500mcg, then 62.5-125mcg daily thereafter 40-160mg PO BD No loading for sotalol
pKA 7.2 9.8
Pharmacodynamics
MOA Direct cardiac: inhibits Na/K ATPase > increased Ca > positive inotropic effect + increased refractory period

Indirect cardiac: increased PSNS release of ACh at M receptors > slowed conduction at AV node/bundle

1) Non selective B-blocker (class II) > decreased chronotropy and inotropy

2) Class III activity (K channel blocker) > prolonged refractory period + repolarisation > slow AV conduction and lengthens QT

Dig = increased inotropy and short QT

Sotalol = decreased inotropy + prolonged QT

Side effects CVS: May worsen arrhythmia (lead to VF), AV block, shortened QT interval, scooped ST, TWI, bradycardia

GIT: nausea, anorexia, vomiting
CNS: dizziness, drowsiness

CVS: precipitation of tDP, bradycardia, prolonged QT int, bradycardia, hypotension

Resp: bronchospasm
CNS: dizziness, drowsiness

Dig shortens Qt, sotalol prolongs it.
Pharmacokinetics
Onset/duration 2-3 hours (PO), 10-30mins (IV), duration of action 3-4 days 2-3 hours (PO) Similar onset
Absorption 80% oral bioavailability 95% oral bioavailability Both have good OBA
Distribution Protein binding 25%

VOD 6-7L/kg

No protein binding

VOD 1-2L/kg

Dig = larger VOD and protein binding
Metabolism minimal hepatic metabolism (15%) Nil
Elimination T 1/2 48 hours

urine excretion (70% unchanged)

T 1/2 12 hours

Urine excretion (unchanged)

Dig lasts longer in system
Special points Reduce dose in renal failure, monitor with dig level. not removed by dialysis Reduce dose in renal failure

Requires SAS for IV

Both require renal adjustment


Examiner comments

19% of candidates passed this question.

Good answers listed class and the multiple mechanisms of action for both these antiarrhythmics, briefly outlining relevant downstream physiological effects and contrasting effects on inotropy. Knowledge of specific pharmacokinetic parameters of these agents was generally lacking. Clinically relevant adverse effects were frequently omitted (e.g. prolonged QT/Torsades for sotalol, hypokalaemia potentiating toxicity of digoxin).


Online resources for this question


Similar questions

  • Digoxin
    • Question 2, 2018 (2nd sitting)
    • Question 10, 2013, (2nd sitting)
    • Question 22, 2010 (2nd sitting)



Question 15

Question

Describe the physiology of the NMDA (N-Methyl D-aspartate) receptor (40% of marks). Outline the pharmacology of ketamine (60% of marks).


Example answer

NMDA receptor

  • Structure
    • Tetrameric, ligand gated, transmembrane receptor
  • Location
    • Abundant in the CNS (brain, spinal cord)
  • Ion permeability
    • Ca, Na, K
  • Activated by
    • Glutamate (excitatory neurotransmitter) and glycine
    • Activation leads to removal of central Mg plus (Na/Ca in, K out) > EPSP
  • Blocked by
    • Ketamine, Mg, memantidine


Name Ketamine
Class Anaesthetic (phencyclidine derivative)
Indications induction GA, conscious sedation, analgesia,
Pharmaceutics 100mg/ml. Clear colourless solution. Racemic mixture of S and R enantiomers, or S+ enantiomer alone. Water soluble.
Routes of administration IV/IM/PO/SC/PR
Dose 0-0.25mg/kg/hr (analgesia), 1-2mg/kg (GA), 0.5mg/kg (sedation)
pKa 7.5
Pharmacodynamics
MOA NMDA antagonism, weak opioid receptor agonism, weak Ca ch inhibition
Effects CNS: dissociative anaesthesia and analgesia.

CVS: increased HR/BP, decreased pulmonary and systemic vascular resistance,
Resp: bronchodilation

Side effects CNS: emergence reactions including hallucinations, unpleasant dreams. may increase ICP in non ventilated patients

CVS: may increase HR/BP, increased myocardial O2 req.
GIT: Nausea, vomiting, increased salivation
RESP: apnoea

Pharmacokinetics
Onset 30s IV, duration of effect 10-20mins
Absorption Lipid soluble > readily absorbed. But poor OBA (16%) due to 1st pass metabolism
Distribution Large (5L/kg) VOD. small protein binding (25%). Crosses placenta.
Metabolism Metabolised by CYP450 > majority inactive metabolites (norketamine active)
Elimination Elimination T1/2 = 2 hours. Kidneys (95%), faeces (5%)
Special points


Examiner comments

49% of candidates passed this question.

The NMDA receptor is a ligand gated voltage dependent ion channel located on post synaptic membranes throughout the CNS, with glutamate, an excitatory neurotransmitter, its natural ligand. A brief description of its structure, roles of glycine and magnesium, ions conducted, result of activation, role in memory and learning and agonists/antagonists was expected. Detail on structure and functions of the receptor were a common omission.
Ketamine, a phencyclidine derivative, is a non-competitive antagonist at the NMDA receptor. It is presented as a racemic mixture or as the single S(+) enantiomer (2-3 X potency). Administration routes and doses scored marks. Pharmacodynamics were generally well covered including CVS (direct and indirect effects), CNS (anaesthesia, analgesia, amnesia, delirium, effects on CBF and ICP) respiratory (bronchodilator with preservation of airway reflexes) GIT effects (salivation, N and V). Knowledge of specific pharmacokinetic parameters was less well covered, including low oral bioavailability and protein binding and active metabolite (norketamine).


Online resources for this question


Similar questions

  • Ketamine

    • Question 4, 2018 (2nd sitting)

    • Question 22, 2015 (1st sitting)

    • Question 16, 2011 (2nd sitting)

    • Question 7, 2010 (2nd sitting)


Question 16

Question

Describe the role of carbon dioxide in the control of alveolar ventilation


Example answer

SENSORS

Peripheral chemoreceptors

  • Located in the carotid body
    • Sense a rise in PaCO2 (as well as a fall in PaO2 or pH)
    • Afferent nerve = CN IX
  • Located in the aortic body
    • Sense a rise in PaCO2 (or fall in PaO2)
    • Afferent nerve CN X


Central chemoreceptors

  • Located in the ventral medulla near the respiratory centre
  • Stimulated by a fall in pH of the CSF
  • The most important mediator of the change in pH is PaCO2 which freely diffuses across the blood-brain-barrier and dissociates into H+
  • The reduced buffering capacity (HCO3 cannot pass the BBB) make these receptors very sensitive to change in CSF pH


CENTRAL PROCESSOR

  • Respiratory centre in the medulla and pons
    • Nucleus retroambigualis --> Expiratory muscle control via UMN
    • Nucleus parambigualis --> Inspiratory muscle control via UMN
    • Nucleus ambigualis --> Pharyngeal/laryngeal muscles via CN 9/10
    • pre-Botzinger complex --> respiratory pacemaker


EFFECTORS

  • Muscles of respiration (diaphragm, intercostals, accessory muscles etc)


Ventilatory response to CO2 change

  • Linear response (increase in PaCO2 = increase in minute ventilation)
    • Left shift: metabolic acidosis, hypoxia
    • Right shift: sleep, anaesthesia, opiates
File:Https://derangedphysiology.com/main/sites/default/files/sites/default/files/CICM Primary/F Respiratory system/ventilatory response to CO2 under different conditions.jpg


Examiner comments

57% of candidates passed this question.

Better answers considered the role of CO2 in the control of alveolar ventilation in terms of sensors, central processing and effectors - with an emphasis on sensors. Features of central and peripheral chemoreceptors should have been described in detail. The PCO2/ventilation response curve is best described using a graph, with key features of the curve identified (including gradient and axes). Various factors affecting the gradient of this curve and how CO2 affects the response to hypoxic drive should be described.


Online resources for this question


Similar questions

  • None the same, but broadly related to

    • Question 21, 2013 (1st sitting)

    • Question 1, 2015 (1st sitting)

    • Question 13, 2015 (2nd sitting)


Question 17

Question

Explain the physiology of neuromuscular transmission


Example answer

Neuromuscular junction (NMJ)

  • Synapse between a motor neuron and a muscle cell
  • Components
    • Terminal bouton of nerve axon
    • Synaptic cleft
    • Junctional folds
    • Motor end plate
  • The neurotransmitter of the NMJ is acetylcholine (Ach) which is synthesised in the nerve axoplasm
File:C:\Users\ethan\AppData\Roaming\Typora\typora-user-images\image-20220304095424224.png


Neuromuscular transmission

  • Action potential depolarised nerve terminal
  • Voltage gated calcium channels open & calcium enters
  • Calcium influx, triggers synaptic vesicles to release Ach into the synaptic cleft via exocytosis
  • ACh diffuses across the synaptic cleft and binds to post-synaptic nicotinic receptors
    • Nicotinic ACh receptors (nAChR) are transmembrane ligand gated, ion channel linked receptors
  • Activation of nAChR leads to Na influx, which depolarises the cell (excitatory post synaptic potential)
  • Muscle contraction occurs via muscle excitation-contraction coupling
  • ACh is subsequently metabolised by acetylcholinesterase (into Acetyl CoA and choline) and the NMJ returns to its resting state
File:Https://cdn.kastatic.org/ka-perseus-images/c2792e65f78b25734f78a5f34cd296104a2e5d86.png


Examiner comments

60% of candidates passed this question.

Description of sequential events from axon conduction to detail at the neuromuscular junction was required. Well-constructed answers defined neuromuscular transmission, elucidated the structure of the neuromuscular junction (best done with a detailed diagram), described the central importance of acetylcholine, including synthesis, storage, receptors, and degradation. An ideal answer also described both pre-synaptic (e.g. voltage-gated calcium channels, exocytosis of vesicles) and post-synaptic events (acetylcholine receptors, end plate potentials, and the events that lead to excitation-contraction coupling in skeletal muscle).


Online resources for this question


Similar questions

  • ? None



Question 18

Question

Describe the pharmacology of frusemide


Example answer

Name Furosemide
Class Loop diuretic
Indications Oedema/fluid overload, renal insufficiency, hypertension
Pharmaceutics Tablet, clear colourless solution (light sensitive),
Routes of administration IV, PO,
Dose Varies (~40mg daily commonly used for well patients, can be sig. increased)
pKA 3.6 (highly ionised; poorly lipid soluble)
Pharmacodynamics
MOA Binds to NK2Cl transporter in the thick ascending limb LOH, leads to decreased Na,K, Cl reabsorption > decreased medullary tonicity + Inc Na/Cl delivery to distal tubules > decreased water reabsorption > diuresis
Effects Renal: diuresis

CVS: hypovolaemia, arteriolar vasodilation + decreased preload (=mechanism for improvement of dyspnoea before diuretic effect)
Renal: increase in RBF

Side effects CVS: hypovolaemia, hypotension

Renal/metabolic: Metabolic alkalosis, LOW Na, K, Mg, Cl, Ca, increased Cr
Ototoxicity, tinnitus, deafness

Pharmacokinetics
Onset 5 mins (IV), 30-60 mins (PO), Effect lasts 6 hours.
Absorption Bioavailability varies person-person (40-80%)
Distribution Vd = 0.1L/Kg, 95% protein bound (albumin)
Metabolism < 50% metabolised renally into active metabolite
Elimination Renally cleared (predominately unchanged). T1/2 ~90 mins.
Special points Deafness can occur with rapid administration in large doses


Examiner comments

13% of candidates passed this question.

The majority of answers were well structured, some using tables and others using key headings. In general, for a commonly used drug that is listed in the syllabus as Level 1 of understanding, detailed information was lacking. In particular, mechanism of action, dose threshold and ceiling effect and pharmacokinetics lacked detail and/or accuracy.




Question 19

Question

Describe the effects of ageing on the respiratory system.


Example answer

Age relate changes Effects of change
Airway

- Increased airway reactivity
- Decreased ciliary number/activity
- Diminished airway reflexes

- Increased risk of bronchospasm

- Reduced clearance of secretions
-Increased propensity towards pharyngeal collapse

Chest wall

- Calcification of costal ligaments
-Reduced vertebral body height
-Kyphosis

- Decreased chest wall compliance

- Reduced vital capacity
- Increased RV and FRC

Respiratory muscles

- Decreased muscle mass/strength
-Decreased proportion fast-twitch fibres

- Decreased FEV1

- Fatigue develops faster

Lungs

- Senile emphysema (hyperinflation)
- Degradation of elastic fibres and supporting tissues

- Increased lung compliance

- Increased dead space
- Decreased elastic recoil
-Increased closing volume

Gas exchange

-Increased alveolar-capillary membrane thickness
-Senile emphysema

- Decline in DLCO

- Decreased surface area for gas exchange
- Increased shunt / V/W mismatch

Control of ventilation

- Decrease in efferent neural output to respiratory muscles
- Minute volume remains similar

- Reduction in response to hypoxia and hypercarbia

-Decrease in Vt --> increase in RR to maintain MV

Work of breathing Overall increased due to the net effect of the above changes


Examiner comments

5% of candidates passed this question.

Answers should have included the effects of ageing on the efficiency of gas exchange, how the expected PaO2 changes with age, and its causation. Anatomical changes should have been included as should changes in lung volumes, particularly the significance of an increased closing volume. Marks were not awarded for the effects of disease states.


Online resources for this question


Similar questions

  • None :(



Question 20

Question

Describe the cardiovascular effects of positive pressure ventilation on a patient who has received a long acting muscle relaxant.


Example answer

Overview

  • Normal spontaneous ventilation generates negative intrapleural pressure
  • PPV has numerous cardiovascular implications


Effects of PPV

  • Left ventricle
    • Decreased preload
    • Decreased afterload
  • Right ventricle
    • Decreased preload
    • Increased afterload


Mechanism of PPV effects

  • Right heart
    • Increased intrathoracic pressure (ITP) is transmitted to central veins + right atrium (RA)
      • Leads to increased RA pressure > impairs venous return > decreased RV preload
      • Leads to increased pulmonary vascular resistance > increased RV afterload
    • Increased RV afterload + reduced RV preload > decreased RV stroke volume
    • Increased RV afterload leads to increased RV end diastolic pressure
      • If RVEDP is greater than LVEDP > bulging of IV septum into LV > ventricular interdependence
  • Left heart
    • Decreased preload
      • Due to reduced RV stroke volume and ventricular interdependence (explained above)
    • Decreased afterload
      • PPV > reduction in LV end systolic transmural pressure > decreased afterload (Law of LaPlace)


Net effect on cardiac output

  • If the patient has normal LV
    • Net decrease in CO
    • Decreased preload has overall greater impact compared to decreased afterload
  • If the patient has impaired LV
    • Net increase in CO
    • Decrease in afterload has overall greater impact, compared to decreased preload


Examiner comments

33% of candidates passed this question.

Structured answers separating effects of positive pressure on right and left ventricle, on preload and on afterload were expected. Overall there was a lack of depth and many candidates referred to pathological states such as the failing heart. Simply stating that positive pressure ventilation reduced right ventricular venous return and/or left ventricular afterload, without some additional explanation was not sufficient to achieve a pass level.


Online resources for this question


Similar questions

  • None the same
  • Broadly related to physiological changes of PEEP/PPV
    • Question 17, 2009 (1st sitting)
    • Question 3, 2014 (2nd sitting)
    • Question 2, 2016, (1st sitting)



2018 (2nd sitting)

Question 1

Question

Describe the surface anatomy of the anterior neck (30% of marks) and the underlying structures relevant to performing a tracheostomy (70% of marks).


Example answer

Structure

  • Fibromuscular tube ~10cm long
  • Supported by 16-20 incomplete cartilaginous rings which joined by fibroelastic tissue and are connected posteriorly by smooth muscle (the trachealis)
  • Divided into cervical and thoracic parts


Course

  • Trachea begins approximately C6 where it is continuous with the larynx
  • Trachea travels inferoposteriorly
  • Enters thoracic cavity through the superior thoracic aperture, at the level of the jugular notch
  • Ends approximately at level of sternal angle (T4/5) where it divides into left and main bronchi


Relations

  • Posterior: oesophagus
  • Anterior: thyroid gland (isthmus), cervical fascia, manubrium, thymus remnants,
  • Right lateral: thyoid gland (lobe), carotid sheath ( common carotid, vagus, IJV), RLN
  • Left lateral: thyroid gland (lobe), carotid sheath ( common carotid, vagus, IJV), RLN


Neurovascular supply

  • SNS: sympathetic trunks
  • PSNS: recurrent laryngeal and vagus nerves
  • Arterial supply: Branches from inferior thyroid arteries
  • Venous drainage: Inferior thyroid veins


Surface anatomy of anterior neck (superior --> inferior)

  • Hyoid bone (C3)
  • Thyroid cartilage
  • Cricothyroid membrane
  • Cricoid cartilage (C6)
  • Thyroid gland
  • Sternohyoid muscle just lateral to the midline structures, overlies sternothyroid and thyrohyoid


Layers of dissection in tracheostomy (from anterior --> posterior)

  • Skin
  • Subcutaneous tissue
  • Fat
  • Pretracheal fascia
  • Fibroelastic tissue between tracheal cartilage rings
  • Trachea


Examiner comments

79% of candidates passed this question.

Answers required a description of the surface anatomy outlining the midline structures including
the hyoid bone and cartilages. The tissue layers should have been mentioned as should the
relevant tracheal anatomy. The anterior, posterior and lateral relations of the trachea should
also have been included along with the relevant nerves and blood vessels. Diagrams were not
essential but could have been included.
Candidates should note that marks were not awarded for a description of how to perform a
tracheostomy.


Online resources for this question


Similar questions

  • Question 11, 2017 (1st sitting)



Question 2

Question

Compare and contrast amiodarone and digoxin.


Example answer

Name Digoxin Amiodarone
Class Cardiac glycoside (antiarrhythmic) Antiarrhythmic (Class III), though other class (I, II, IV) activity
Indications tachyarrhythmias (e.g. AF, SVT), heart failure Tachyarrhythmias (e.g. SVT, VT, WPW)
Pharmaceutics 62.5mcg/250mcg (PO tablets)

25/250 mcg/ml (IV)

100-200mg tablets

Clear solution in ampoules (150mg) for dilution in dextrose

Routes of administration IV and PO IV and PO
Dose Generally load with 250-500mcg, then 62.5-125mcg daily thereafter. Digoxin level (0.7 - 1.0) for most conditions. IV: 5mg/kg, then 15mg/kg infusion / 24hrs.

Oral: 200mg TDS (1/52) > BD (1/52) > daily

pKA 7.2 6.6 (highly lipid soluble)
Pharmacodynamics
MOA Direct cardiac: inhibits Na/K ATPase > increased Ca > positive inotropic effect + increased refractory period

Indirect cardiac: increased PSNS release of ACh at M receptors > slowed conduction at AV node/bundle

- Blocks K channels (Class III effects) prolonging repolarisation and therefore refractory period.

- Decreases velocity of Phase 0 by Blocking Na channels (Class I effects)
- Non-competitive inhibition of Ca channels prolonging depolarisation + AV nodal conduction time (Class IV effects)
- Slows AV/SA nodal conduction via anti-adrenergic activity (Class II effects)

Side effects CVS: May worsen arrhythmia (lead to VF), AV block, shortened QT interval, scooped ST, TWI, bradycardia

GIT: nausea, anorexia, vomiting
CNS: dizziness, drowsiness

Side effects worsen w. time!

RESP: pneumonitis, fibrosis
CVS: bradycardia, QT prolongation
CNS: peripheral neuropathy
Thyroid: hypo/hyperthyroidism
LIVER: cirrhosis, hepatitis
DERM: photosensitivity, skin discolouration

Pharmacokinetics
Onset 2-3 hours (PO), 10-30mins (IV), duration of action 3-4 days Immediate (IV), 4 hours (PO)
Absorption 80% oral bioavailability PO bioavailability 40-60%
Distribution Protein binding 25%

VOD 6-7L/kg

Highly protein bound (>95%)

VOD: ~70L /kg

Metabolism Minimal hepatic metabolism (15%) Hepatic (CYP3A4) with active metabolites (desmethylamiodarone)
Elimination T 1/2 48 hours

urine excretion (70% unchanged)

T 1/2 = 1 month
Faces, urine, skin
Special points Reduce dose in renal failure, monitor with dig level. not removed by dialysis Amiodarone increases digoxin level (by preventing renal excretion and lowering protein binding)


Examiner comments

82% of candidates passed this question.

Most candidates had a good structure for answering this question; a table was commonly used.
Marks were awarded for indications and an explanation of the mechanism of action of both
drugs, which was generally well explained. The pharmacodynamic effects were often listed in a
general manner and more detail would have achieved a higher mark, including a list of the ECG
effects. Some detail on the pharmacokinetics and adverse effects of the drugs was expected
and this section was generally well answered. Better answers noted digoxin levels and potential
drug interactions.


Online resources for this question


Similar questions

  • Amiodarone
    • Question 5, 2008 (2nd sitting)
    • Question 22, 2010 (2nd sitting)
    • Question 14, 2014 (2nd sitting)
    • Question 11, 2016 (1st sitting)
    • Question 11, 2021 (2nd sitting)
  • Digoxin
    • Question 14, 2019 (1st sitting)
    • Question 10, 2013, (2nd sitting)
    • Question 22, 2010 (2nd sitting)



Question 3

Question

Explain the causes of the differences between measured end tidal and arterial partial pressures of carbon dioxide (CO2).


Example answer

ETCO2 - PaCO2 gradient

  • There is normally a gradient between PaCO2 and ETCO2 of 0-5mmHg (where ETCO2 is lower)
  • The difference between the values is due to alveolar dead space
    • Alveolar dead space is due to alveoli which are ventilated but not perfused (e.g. west zone 1 lungs)
    • These alveoli do not participate in gas exchange (there is no perfusion), thus contain very little CO2 and a lot of O2 (the same amount as in inspired air)
    • This relatively CO2 deplete gas mixes with the rest of the expired gas, diluting the ETCO2 reading, thus leading to an observed discrepancy
    • Note: It is not due to anatomical dead space as this gas has already been washed out in the early stages of exhalation and thus does not contributed to ETCO2
  • Healthy/awake patients have near zero alveolar dead space, so near zero gradient


Factors affecting ETCO2 - PaCO2 gradient

  • Changes in pulmonary perfusion

    • Global reduction in pulmonary perfusion

      • e.g. pHTN, heart failure, Cardiac arrest, Severe shock

    • Regional decreases in pulmonary perfusion

      • e.g. pulmonary embolism, fat embolism

  • Changes in ventilation

    • Excessively high PEEP --> increased West Zone 1

  • Measurement error

    • Inline HME filters

    • Timing of measurement (measuring before end-expiration)

    • Poor / loss of ETCO2 calibration

    • Interference from other gases (e.g. N2O and collision broadening)

  • Physiological factors

    • Increasing age > increased gradient

Examiner comments

29% of candidates passed this question.

The answer required an explanation of the causes of the difference between the PaCO2 and ETCO2. This required recognising how the end point of phase 3 of the capnograph trace corresponds with end tidal CO2. The difference is caused by the alveolar dead space. The difference is normally very small in healthy adults with the ETCO2 being lower than the PaCO2. It is increased with increasing alveolar dead space. Many incorrectly attributed anatomical dead space as a contributor to the PaCO2-ETCO2 gradient. Discussion of the various types of dead space did not score marks. Marks were awarded for the processes that cause an increased gradient e.g. low cardiac output and pulmonary embolism. Recognising physiological factors such as increasing gradient with increasing age scored marks. Marks were not awarded for descriptions on how dead space is measured.


Online resources for this question


Similar questions

  • Question 21, 2017 (1st sitting)
  • Question 9, 2009 (1st sitting)
  • Question 24, 2007 (1st sitting)



Question 4

Question

Compare and contrast ketamine and midazolam.


Example answer

Name Midazolam Ketamine Notes
Class Benzodiazepine (sedative) Anaesthetic (phencyclidine derivative)
Indications Anaesthesia, sedation, treatment of seizures, anxiolysis induction GA, conscious sedation, analgesia,
Pharmaceutics IV: clear solution, pH 3.5. Diluted in water. 100mg/ml. Clear colourless solution. Racemic mixture of S and R enantiomers, or S+ enantiomer alone. Water soluble.
Routes of administration IV, IM, S/C, intranasal, buccal, PO IV/IM/PO/SC/PR
Dose Dose depends on many pt. factors. 1-5mg premedication. 2.5-10mg seizures. Infusions. 0-0.25mg/kg/hr (analgesia), 1-2mg/kg (GA), 0.5mg/kg (sedation)
pKa 6.5 7.5
Pharmacodynamics
MOA Midazolam (BZD) binds to GABAA receptors (ionotropic ligand gated channel) in the CNS. Cl enters > hyperpolarisation. NMDA antagonism, weak opioid receptor agonism, weak Ca ch inhibition - Ketamine has weak GABA effects
Effects CNS: sedation, amnesia, anticonvulsant effects, decreased cerebral O2 demand CNS: dissociative anaesthesia and analgesia.

CVS: increased HR/BP, decreased pulmonary and systemic vascular resistance
RESP: bronchodilation

- Ketamine has analgesic and bronchodilator properties

- Midaz has anticonvulsant properties

Side effects CVS: bradycardia, hypotension

CNS: confusion, restlessness
RESP: respiratory depression/ apnoea

CNS: emergence reactions including hallucinations, unpleasant dreams. May increase ICP in non vent. pts.

CVS: may increase HR/BP, increased myocardial O2 req.
GIT: Nausea, vomiting, increased salivation

- Ketamine does not cause respiratory depression and preserves airway reflexes
Pharmacokinetics
Onset peak effect 2-3 minutes (IV) 30s IV, duration of effect 10-20mins
Absorption ~40% oral bioavailability

Absorbed well, but sig. 1st pass metabolism

Lipid soluble > readily absorbed. But poor OBA (16%) due to 1st pass metabolism Both have poor PO bioavailability
Distribution 95% protein bound, very lipid soluble

Vd = 1L / kg

Large (5L/kg) VOD.

Small protein binding (25%). Crosses placenta.

- Midaz will rapidly accumulate with infusions, ketamine will not
Metabolism Hepatic metabolism by hydroxylation

Active (1-a hydroxymidazolam) and inactive metabolites

Metabolised by CYP450 > majority inactive metabolites (norketamine active 33% potency) Similar
Elimination Renal excretion

T 1/2 = 4 hours

Elimination T1/2 = 2 hours. Kidneys (95%), faeces (5%) Both predominately renal excretion
Special points Flumazenil - antagonist (reversal agent) Nil reversal agent No reversal agent for ketamine

- Midaz exhibits tolerance, withdrawal, dependence, ketamine does not.


Examiner comments

62% of candidates passed this question.

In addition to the key PK and PD properties of each drug, a clear comparison was required to score well (why choose one drug over the other?). When a table was used the addition of a comparison column was helpful. A good answer covered the following: ketamine has analgesic properties whilst midazolam does not; ketamine preserves airway reflexes and does not cause respiratory depression unlike midazolam; whilst ketamine increases cerebral blood flow and CMRO2, midazolam decreases t; ketamine has a direct myocardial depressant effect which is often offset by an increase in sympathetic tone, whilst midazolam has no direct cardiac depressant effects but may reduce BP due to reduced SVR; midazolam has anticonvulsant properties, ketamine does not; ketamine is a bronchodilator; both drug effects are offset by redistribution; midazolam is lipophillic at body pH and will accumulate with prolonged infusions, ketamine will not; both are metabolised in the liver; midazolam can be reliably reversed by flumazenil, whereas there is no reliable complete reversal of ketamine; midazolam exhibits tolerance, dependence and withdrawal, whereas patients will only experience tolerance to the analgesic properties of ketamine. “Drugs in Anaesthesia and Intensive care” chapters on midazolam and ketamine outline the key facts to include in this answer; interpretation and comparison of these facts will help achieve a good mark.


Online resources for this question


Similar questions

  • Midazolam
    • Question 7, 2019 (1st sitting)
    • Question 9, 2019 (2nd sitting)
    • Question 24, 2016 (1st sitting)
    • Question 2, 2008 (2nd sitting)
  • Ketamine
    • Question 15, 2019 (1st sitting)
    • Question 22, 2015 (1st sitting)
    • Question 16, 2011 (2nd sitting)
    • Question 7, 2010 (2nd sitting)



Question 5

Question

Describe the carriage of carbon dioxide (CO2) in the blood.


Example answer

Overview

  • CO2 is constantly produced as a by-product of metabolism and needs to be cleared

  • CO2 content of blood

    • Mixed venous: 52mls/100mls blood, at PaCO2 of ~45mmHg

    • Arterial: 48mls/100mls blood, at PaCO2 of ~40mmHg

CO2 is transported in three main forms in the blood:


Dissolved CO2

  • Accounts for
    • ~5% of the total carbon dioxide in the blood
    • ~10% of the CO2 evolved by the lung
  • The amount dissolved is proportional to the partial pressure (Henry's Law)
  • 20x more soluble than O2, so dissolved CO2 plays a more significant role in transport


Bicarbonate

  • Accounts for

    • ~90% of the carbon dioxide in the blood

    • ~60% of the CO2 evolved by the lung

  • Bicarbonate is formed by the following sequence

    <math display="block">CO_2 + H_{2}O \leftrightarrow H_{2}CO_{3} \leftrightarrow H^+ + HCO_3^-</math>
  • Process

    • CO2 dissolves into RBC and leads to H+ and HCO3 (per above equation)

    • HCO3 moves into plasma, H+ binds to reduced (deoxy) Hb

    • Cl moves into the cell to maintain electroneutrality (chloride shift)

    • When Hb is oxygenated in the lungs, H+ dissociates and coverted back to CO2 by the above equation and is exhaled

    • Haldane effect accounts for the increased capacity of Hb to carry CO2 when poorly oxygenated


Carbamino compounds

  • Accounts for
    • ~5% of the CO2 in the blood
    • ~30% of the CO2 evolved by the lung
  • Formed by the combination of CO2 with terminal amine groups in blood proteins
  • Haemoglobin is the most abundant protein and has most imadazole side chains (greatest carrier capacity)
    • The reaction occurs faster with deoxHb than oxy-Hb (Haldane effect)


Examiner comments

65% of candidates passed this question.

A definition of arterial and venous CO2 content (mls and partial pressure) and an outline of the 3 forms of CO2 in the blood and their contribution to the AV difference, followed by a detailed explanation of each form of carriage was required for this question. A good answer included a table of the contribution of each form of carriage to arterial and venous content and the AV difference; explained the concepts of chloride shift when describing carriage as HCO3 -; detailed the Haldane effect and its contribution to carbamino carriage and referenced Henry’s law when describing dissolved CO2. West’s Chapter 6 on gas transport details the key information to score well on this question


Online resources for this question


Similar questions

  • Question 6, 2012 (1st sitting)
  • Question 1, 2012 (2nd sitting)
  • Question 13, 2015 (1st sitting)
  • Question 5, 2018 (2nd sitting)
  • Question 1, 2020 (1st sitting)



Question 6

Question

Outline the determinants of venous return to the heart


Example answer

Venous return

  • Rate of blood flow back to the right atrium
  • In healthy state: venous return = cardiac output (else pathological pooling of blood occurs)
  • Can be defined by following eqns:
    • VR = CO
    • VR = MSFP - RAP / resistance to venous return
  • Therefore factors effecting venous return are those that affect
    • MSFP
    • RAP
    • Resistance to venous return
    • Cardiac output


Cardiac output

  • Normally ~5L/min
  • Increased CO = increased venous reutn
  • CO is effected by
    • Afterload (reduced afterload = increased cardiac output = increased VR)
    • Contractility (increased contractility = increased CO = increased VR)


MSFP

  • Normally ~7mmHg
  • Increased MSFP = increased VR
  • Affected by venomotor tone and blood volume
  • Increased VR (= increased blood volume and increased venomotor tone)


RAP

  • Normally 2-6mmHg
  • Increased RAP = reduced driving pressure = reduced venous return
  • Factors which increase RAP
    • Positive intrathoracic pressure (e.g. PPV)
    • Reduced pericardial compliance (e.g. effusion)
    • Reduced RA compliance/contractility (e.g. AF)
    • TVR


Resistance to venous return

  • Increased RVR = reduced VR (due to ohms law)
  • Factors effecting RVR
    • Autonomic tone
    • Intrabdominal pressure
    • IVC Obstruction (e.g. pregnancy) reduces VR
    • Posture (decreased VR with erect posture)
    • Vasoactive drugs
    • skeletal muscle pump


Examiner comments

31% of candidates passed this question.

Answers should have included a description of the need for a pressure gradient for flow and a discussion on right atrial pressure, mean systemic filling pressure and resistance to blood flow. The discussion of each of these factors included definitions, normal values, factors affecting them and the direction of change on venous return. Diagrams were not essential, but their use assisted some candidates in explaining the effects of RAP on venous return.


Online resources for this question


Similar questions

  • Question 19, 2020 (1st sitting)



Question 7

Question

Describe protein binding and its significance in pharmacology.


Example answer

Protein binding

  • Drugs in blood can exist in two forms (protein bound, protein unbound)

  • Protein binding of drugs involves the formation of reversible drug-protein complexes

    • Protein + drug <-> protein-drug complex

  • Drugs vary greatly in degree of plasma protein binding

    • e.g. warfarin and phenytoin which are >95% protein bound

    • e.g. rocuronium which is approx. 10% protein bound

  • Types of proteins

    • Drugs can bind to proteins in the plasma (e.g. albumin, globulins) or tissue

    • Albumin is the most sig. drug binder and binds neutral/acidic drugs (e.g. barbiturates)

    • a-1 glycoproteins and globulins bind basic drugs (e.g. morphine)

    • Haemoglobin can bind some drugs e.g. phenytoin

  • Effect of protein binding

    • Only unbound fraction exerts can interact with receptors and exert its pharmacologic effect

    • Only unbound drug in plasma can freely cross cell membranes

    • Only unbound drugs can undergo filtration or metabolism

    • For drugs which are highly protein bound (>90%), small changes in degree of protein binding can have sig. clinical effects. I.e. protein binding from 99% to 98% doubles to unbound (active) drug concentration (from 1% to 2%)

    • Highly tissue bound drugs have long duration of action, high volume of distribution and readily build up in the body

  • Protein binding is affected by

    • Protein factors

      • Concentration of protein (decreased protein > increased unbound drug)

      • Number of available protein binding sites

    • Drug factors

      • Protein affinity

      • Concentration of drug - higher drug concentration > saturation of protein > higher unbound (free) drug

    • Patient factors

      • temperature and pH

      • Inflammation, infection, surgery > increased acute phase reactants > increased protein binding

      • Age

Examiner comments

19% of candidates passed this question.

Descriptions of protein binding were generally too brief (e.g. a statement saying that drugs and hormones bind to proteins in the plasma rather than a description of usually reversible binding with a drug-protein equilibrium). It was expected that the factors which determine protein binding would be described. Marks were attributed if proteins, along with characteristics of the drugs they bind, were named. Candidates achieved better marks if they named the pharmacological parameters affected by protein binding and explained how and why change occurs along with the significance of those changes. Few candidates differentiated between tissue and plasma protein binding and the different effects on the volume of distribution.


Online resources for this question


Similar questions

  • Nil



Question 8

Question

Describe gastric emptying (40% of marks) and outline its regulation (60% of marks).


Example answer

Gastric emptying

  • The coordinated emptying of chyme from the stomach to the duodenum
  • Regulated by food, local mechanical, neural, hormonal and drug factors
  • Mechanism
    • During fasting
      • Migrating motor complexes (Slow peristaltic waves that originate in fundus) sweep through stomach at regular intervals
      • Role is to keep the stomach empty of secretions and food debris
      • Interrupted by food consumption
    • During fed state
      • Receptive relaxation of stomach following swollowing
      • Tonic contraction / peristalsis > propelling food towards pylorus > mixing
      • Small food particles <2mm are pushed through pyloric sphincter at a stable rate
        • Half time of solids is ~2 hours
      • Liquids empty more rapidly and the rate of emptying is dependant on the antral-duodenal pressure gradient.
        • Half time of liquids <30 minutes


Regulation

  • Food factors
    • Fluids have half time of 30 mins, solids have half time of 2 hours
    • Carbohydrates (fastest) > proteins > fatty acids (slowest)
    • Tonicity: increased tonicity = decreases emptying rate
  • Local factors
    • Increased gastric volume > increased gastric emptying
    • Duodenal stretch / wall irritation / acidity >reflex inhibition > decreased gastric emptying
  • Neural factors
    • Increased SNS stimulation > decreased contractility + gastric emptying
    • Increased PSNS (vagal) activity > increased contractility + gastric emptying
  • Hormonal factors
    • secretin (stimulated by low duodenal pH) > decreased emptying
    • Cholecystokinin (stimulated by fatty acids) > decreased emptying
    • Somatostatin > decreased emptying
    • Gastrin (stimulated by stretch, amino acid content) > increased emptying)
    • Motilin: stimulates migrating motor complex > increased emptying
  • Drugs factors
    • e.g. opioids > decreased empyting
    • eg. metoclopramide > increased emptying


Examiner comments

24% of candidates passed this question.

Candidates were required to provide a description of gastric emptying (40% marks). Although
the question showed the allocation of marks, many candidates did not provide sufficient detail
for this section. This required some description of what gastric emptying is (the co-ordinated
emptying of chyme from the stomach into the duodenum).
Better answers provided detail regarding the process of gastric emptying in the fed and fasted
state and differentiated between liquids, solids, carbohydrate, protein and fats. Factors
regulating emptying included an outline of peristaltic waves, the basal electrical rhythm and its
modulation, the migratory motor complex (MMC) and its modulation, neural input, stretch and
hormonal control.
Many candidates erred by answering the question "the regulation of gastric secretions" rather
than the question (the regulation of gastric emptying). Although they scored well for hormonal
control, they missed out on marks for the other factors relevant to the regulation of gastric
emptying.


Online resources for this question


Similar questions

  • Question 21, 2010 (2nd sitting)
  • Question 8, 2012 (2nd sitting)
  • Question 24, 2014 (2nd sitting)



Question 9

Question

Describe the renal handling of water including the modulation of water excretion


Example answer

Renal handling of water

  • Glomerulus
    • Water is freely filtered at the glomerulus (~180L / day)
    • The amount filtered will depend on the GFR and starlings forces
  • Proximal convoluted tubule (PCT)
    • Approximately 60-70% of the filtered water is reabsorbed
    • Secondary active transport of Na in the PCT creates an osmotic gradient which allows passive absorption of water via osmosis
  • Loop of Henle (LOH)
    • Approximately 10-15% of the filtered water is reabsorbed in the descending LOH
      • Iso-osmotic absorption due to the increased medullary concentration gradient
    • The ascending LOH is relatively water impermeable
  • Distal convoluted tubule (DCT)
    • Approximately 0-5% water reabsorbed in DCT
    • Relatively impermeable
  • Collecting duct (CD)
    • Reabsorbs 5-20% of the remaining water (depending on the level of ADH)
    • ADH inserts luminal aquaporins in collecting duct cells which allows increased reabsorption of water down the osmotic concentration gradient


Regulation

  • There is an obligatory water loss of ~500mls a day needed for waste clearance

  • The body also needs to maintain fluid and osmolality homeostasis

  • The main site of water regulation in the nephron is in the collecting ducts via the action of ADH

  • Mechanism

    • Primary:

      • Osmoreceptors in hypothalamus detect increased osmolality > increased production of ADH > increased release of ADH from posterior pituitary > increased luminal aquaporins in CD > increased water reabsorption

    • Secondary

      • Baroreceptors detect reduced blood pressure > increased ADH secretion

      • ANP/BNP secretion is reduced with decreased BP (stretch) > decreased GFR + activation of RAAS

Examiner comments

37% of candidates passed this question.

This question required a brief introduction of the role the kidney plays in water balance; a more
detailed description of how water is handled as it passes through the various segments of the nephron (glomerulus, PCT, Loop of Henle, DCT and Collecting Duct); the modulation of water excretion by the kidney due to ADH (vasopressin) and how this operates; and the stimuli (osmotic and non-osmotic) for ADH secretion. Although worth mentioning in the context of the effect they have on water movement through the kidney, detailed explanations of Starling's forces in the glomerulus, and of the operation and maintenance of the counter-current mechanism, were not required. More important was describing the control of water reabsorption in the collecting ducts (and thus modulation of water excretion by the kidney) under the influence of ADH


Online resources for this question


Similar questions

  • Question 8, 2008 (1st sitting)
  • Question 4, 2015 (1st sitting)



Question 10

Question

Compare and contrast the pharmacology of vancomycin and flucloxacillin.


Example answer

Name Vancomycin Flucloxacillin
Class Glycopeptides (antibiotic) Penicillins (antibiotic)
Indications Severe gram positive infections,MRSA, C.diff Gram positive infections (particularly staph)
Pharmaceutics White powder for reconstitution Capsule, tablet or white power for reconstitution
Routes of administration PO, IV, PR, intrathecal PO, IV,
Dose Dose/interval adjusted according to desired peak/trough levels 250-1g, every 6 hrs
Pharmacodynamics
MOA Inhibits cell wall synthesis by binding to D-ala-D-Ala portion of growing cell wall Beta-lactam ring binds to penicillin binding protein > prevents crosslinking > impaired cell wall synthesis
Microbial coverage Gram positives, including MRSA. C diff coverage Narrow spectrum

Gram positive bacteria
Does

Side effects CNS: ototoxicity

RENAL: nephrotoxicity
HAEM: thrombocytopaenia, leukopenia
IMMUNO: red man syndrome

GIT: diarrhoea, nausea, cholestatic hepatitis

IMMUNO: penicillin allergy
CNS: neurotoxicity
Haem: blood dyscrasias

Pharmacokinetics
Absorption PO bioavailability <1%. Only given orally for C. diff infections. PO bioavailability 70%
Distribution Poor CSF penetration (requires higher dosing)

VOD = 0.5L / kg
50% protein bound

95% protein bound

VOD = 0.3 L /kg
CNS penetration with meningitis only

Metabolism No metabolism Hepatic metabolism
Elimination Unchanged in the urine

T 1/2 = 6 hrs

Renal elimination (predominately unchanged)

T 1/2 = 1 hour

Monitoring Renal function Monitor LFTs, renal function
Resistance Cannot treat VRE (VanA/B resistance genes) Can treat b-lactamase producing bacteria, but not MRSA (mecA gene)


Examiner comments

49% of candidates passed this question.

Most candidates structured their answers well. Expected information included: the class of antibiotic of each agent, their respective pharmaceutics, pharmacodynamics, pharmacokinetics, indications and adverse effects. Better answers provided pharmacodynamic and pharmacokinetic information relevant to each drug rather than generic statements. Good answers also included the common resistance mechanisms for both agents.


Online resources for this question


Similar questions

  • Question 22, 2016 (1st sitting)



Question 11

Question

Describe the anatomy relevant to the insertion of an intercostal catheter.


Example answer

Surface anatomy

  • Lateral approach
    • ICC is inserted in the 'triangle of safety' based off surface landmarks
      • Anterior border: lateral border of the pectoralis major
      • Posterior Border: lateral border of latissimus dorsi
      • Inferior border: 5th intercostal space
      • Superior: base of axilla
  • Anterior approach
    • Second intercostal space, midclavicular line


Layers of dissection / path of needle

  • Skin
  • Subcutaneous tissue
  • Pectoralis muscle (in anterior approach only)
  • External intercostal muscle
  • Internal and innermost intercostal muscles
  • Parietal pleura
  • Pleural space


Important anatomical considerations

  • Intercostal neurovascular bundle
    • Sits in the inferior aspect of the rib, between innermost and internal IC muscles
    • Vein > artery > nerve (from superior to inferior)
    • Care to avoid this by aiming for the rib below, and guiding over the top of the inferior rib
  • Anterior approach
    • Variable degrees of breast/subcutaneous tissue
    • Will also contain the pectoralis major muscle (variable thickness) between subcutaneous tissue and intercostal muscles
  • 5th intercostal space
    • The reason it is important to place above the 5th intercostal space as this reduces of inadvertently placing the ICC into intrabdominal structures (e.g. liver, spleen) or penetration of the diaphragm (as the diaphragm can go as high as 5th intercostal space during expiration / pregnancy)
  • Deeper structures
    • Beneath pleural space is the visceral pleura and lung parenchyma, which should be avoided..obviously
  • Internal mammary artery / lymphatic ducts
    • Too far medial on anterior approach risks damage to these structures


Examiner comments

56% of candidates passed this question.

An anatomy question expects the use of anatomical nomenclature to describe relationships. Good answers defined the “safe triangle” for the lateral approach, soft-tissue layers passed through from skin to pleura and relationship of the neurovascular bundle to the ribs and intercostal muscles. Additional marks were gained for describing the anterior approach and related structures. Common omissions included description of deeper structures (relations) including intrathoracic and intra-abdominal organs and level of the diaphragm with regard to rib space. No marks were awarded for a description of intercostal catheter insertion.


Online resources for this question


Similar questions

  • ? Nil



Question 12

Question

Outline the control of blood glucose.


Example answer

Overview

  • Normal blood glucose levels (BGLs) are ~4-6mmol/L
  • BGL will rise following carbohydrate consumption
  • Regulation of BGL is via short and long term mechanisms
  • Insulin and glucagon are the main regulatory hormones


High BGL

  • Increased BGL (>6.0mmols) is sensed directly by the pancreas
  • The increased glucose is taken up by GLUT receptors > undergoes glycolysis > increased ATP/ADP ratio > depolarisation > exocytosis of insulin from pancreatic B-islet cells
  • There is an initial rapid release, followed by a prolonged slow release
  • The increased insulin results in
    • Increased glucose uptake into cells and Glycogenesis (liver)
    • Decreased gluconeogenesis, glycogenolysis and lipolysis
  • The net effect is reduced BGL


Low BGL

  • Decreased BGL (or during times of fasting) is sensed by pancreas
  • Leads to
    • Increased glucagon secretion from a-islet cells in pancreas (<3.0 mmols)
    • Decreased insulin secretion from the B-islet cells in pancreas (<4.0 mmols)
  • Glucagon acts via GPCR (Gs) to
    • Increased glycogenolysis and gluconeogenesis in the liver
    • Increased lipolysis and ketoacid formation
  • Hypoglycaemia also directly stimulates the hypothalamus (with prolonged hypoglycaemia, starvation)
    • Stimulates GHRH release > decreased glucose uptake + increased fat utilisation
    • Stimulates ACTH release > increased cortisol > decreased glucose uptake + increased fat utilisation
    • Stimulates TRH release > increased TSH > increased GIT absorption of glucose
    • Stimulates "hunger" centre in the lateral hypothalamus > seek food
    • Direct SNS stimulation of adrenal medulla > increased adrenaline > increased catabolism
  • The net effect is increased BGL


Other factors

  • BGL control is interconnected to liver function
    • Involved in glycogenolysis/glycogenesis functions regulated by insulin/glucagon
    • Hence liver dysfunction can impair its glucostat function and BSL control
  • BGL control is interconnected to renal function
    • Can help modulate BGL control through control of absorption of glucose
  • Other
    • Insulin and glucagon also affected by: cholecystokinin, somatostatin, food intake


Examiner comments

53% of candidates passed this question.

A definition of normal glucose levels was expected, mentioning how it is regulated despite variable intake. Most answers incorporated the roles of insulin/glucagon and the glucostat function of the liver. Sufficient detail regarding the mechanism of insulin release was often lacking. Extra marks were awarded for description of the role of the satiety centre in the hypothalamus, glucokinase and processes in fasting and starvation that maintain blood glucose levels. Marks were not awarded for describing effects of insulin and glucagon unrelated to glucose control.


Online resources for this question


Similar questions

  • ? None



Question 13

Question

Compare and contrast rocuronium and cisatracurium


Example answer

Name Rocuronium Cisatracurium
Class Aminosteroid NMB Benzylisoquinolinium derivative (NMB)
Indications NMB (e.g. RSI) NMB (i.e. RSI)
Pharmaceutics Clear colourless solution (50mg/5ml vials) Shelf life increased in fridge. Clear colourless solution (10mg/5ml vials) stored at 4 degrees
Routes of administration IV (can also be given IM) IV (can also be given IM))
Dose 0.6 - 1.2mg/kg (RSI dose) 0.15-0.2mg/kg (RSI) Used more commonly as an infusion (titrated to desired TOF)
Pharmacodynamics
MOA Non depolarising NMB Inhibits the action of ACh at the NMJ by competitively binding to alpha subunit of nAChR on pre and post junctional membrane Non-depolarising NMB Inhibits the action of ACh at the NMJ by competitively binding to alpha subunit of nAChR on pre and post junctional membrane
Effects NMB > paralysis NMB > muscle paralysis
Side effects Histamine release: none ANS: vagolytic (inc HR) OTHER: anaphylaxis (<0.1%), pain on injection Histamine release: none ANS: no vagolysis OTHER: anaphylaxis (very rare)
Pharmacokinetics
Onset Onset: 45-90s Duration: ~30 mins Onset: 1-3 minutes Duration: 30-45 minutes
Absorption IV only IV only
Distribution VOD = 0.2 L /kg Protein binding = 10% Doesn't cross BBB VOD = 0.15 L/kg Protein binding = 15%
Metabolism Minimal hepatic metabolism (<5%) Organ independent Hoffman elimination (70-90%) > laudanosine and acrylate
Elimination Bile 70%, Renal 30% elimination Unchanged drug T 1/2 = 90 mins Renal and biliary elimination (10-30%) Inactive metabolites T 1/2 - 30 mins
Special points Reversible with sugammadex Not reversible with sugammadex

Examiner comments

32% of candidates passed this question.

This question was best answered using a tabular format outlining class of drug, pharmaceutics, pharmacokinetics, reversibility and side effects. Better answers commented on the significance of the differences between the two agents and its relevance to ICU practice. Many candidates confused these muscle relaxants with each other and with depolarising muscle relaxants.


Online resources for this question


Similar questions

  • None




Question 14

Question

Explain the detection and response to hypoxaemia


Example answer

Hypoxaemia

  • Abnormally low concentration of oxygen in arterial blood
  • Usually defined clinically as a PaO2 < 60mmhg or SaO2 < 0.9


Hypoxia

  • Oxygen deficiency at the tissues which is typically, but not always, due to hypoxaemia
  • Prolonged hypoxaemia may often result in hypoxia


Detection of hypoxaemia

  • Stimulus
    • Decreased PaO2
  • Sensors
    • Peripheral chemoreceptors located in the carotid body and aortic arch
  • Afferents
    • CN IX (carotid body receptors)
    • CN X (aortic arch receptors)
  • Integrator/controller
    • Medullary and pontine respiratory control centres
    • Includes nucleus retroambiualis, parambigualis, ambigualis, PreBotzinger and Botziner complexes
  • Efferents and effectors
    • Phrenic nerve (diaphragm) - predominant
    • UMN nerves to the other muscles of respiration
  • Effector muscles
    • Diaphragm and intercostal muscles
    • Accessory muscles of respiration (SCM, pectoral, scalene, pharyngeal, abdominal muscles )


Response to hypoxaemia

  • Ventilatory response
    • Increased minute ventilation (hyperbolic relationship with rapidly increasing MV when PaO2 <50-60)
  • Cardiovascular response
    • Hypoxic vasoconstriction of pulmonary circulation
    • Hypoxic vasodilation of systemic circulation
  • Autonomic response
    • Relative increase in sympathetic tone
      • Leads to tachycardia, increased CO, increased SVR
      • BP stable / slight increase
  • Metabolic changes
    • If concurrent hypoxia there will be a switch from aerobic to anaerobic metabolism
  • Hypoxia inducible factors (HIF)
    • With tissue hypoxia, hypoxia inducible transcription factors are no longer broken down.
    • HIFs > increased erythropoiesis (increased EPO), cell differentiation and angiogenesis


Examiner comments

34% of candidates passed this question.

A logical approach to answering this question included a definition of hypoxaemia and then a
discussion of the sensors, integrators and effectors involved. It was expected that candidates
would cover the peripheral chemoreceptor response (including the respiratory, cardiovascular
and autonomic effects), time course of the ventilatory response, hypoxia-inducible factors,
vascular effects (hypoxic vasoconstriction in the pulmonary circulation and vasodilatation in the
systemic circulation) and metabolic changes (switch to anaerobic metabolism). No marks were
awarded for discussing the causes of hypoxaemia. Many candidates incorrectly stated that
hypoxaemia is detected by the central chemoreceptors.


Online resources for this question


Similar questions

  • ? None



Question 15

Question

Outline the production / absorption (30% of marks), composition (30% of marks) and function of cerebrospinal fluid (CSF) (40% of marks).


Example answer

CSF

  • ECF located in the ventricles and subarachnoid space
  • ~2ml/kg of CSF
  • Divided evenly between the cranium and spinal column


Production

  • Constantly produced
  • ~550ml produced per day (~24mls/hr)
  • Produced by
    • Choroid plexus (70%) - located in ventricles of brain
    • Capillary endothelial cells (30%)
  • Produced by a combination of ultrafiltration (via fenestrated choroidal capillaries) and active secretion
    • Na actively transported out. Gradient drives co-transport of HCO3 + Cl
    • Glucose via facilitated diffusion, water by osmosis


Composition relative to plasma

  • Similar: Na, osmolality, HCO3
  • Increased: Cl, Mg, CO2
  • Decreased: pretty much everything else (protein, potassium, calcium, glucose, pH)


Circulation

  • Circulation is driven by

    • Ciliary movement of ependymal cells

    • Respiratory oscillations and arterial pulsations

    • Constant production and absorption

  • CSF flows from

    • Lateral ventricles > foramen of Monro > 3rd ventricle > Sylvian aqueduct > 4th ventricle > cisterna magna (via foramen megendie and luschka) > spreads between spinal/cranial subarachnoid spaces

Reabsorption

  • Rate of ~24mls/hr

  • By the arachnoid villi

    • Located predominately in the dural walls of the sagittal + sigmoid sinuses

    • Function as one way valves, with driving pressure leading to absorption.

Functions

  • Mechanical protection
    • The low specific gravity of CSF > decreased effective weight of the brain (1500g > 50g)
    • With the reduced weight
      • Less inertia = less acceleration/deceleration forces
      • Suspended > no contact with the rigid skull base
  • Buffering of ICP
    • CSF can be displaced / reabsorbed to offset any increase in ICP
  • Stable extracellular environment
    • Provides a constant, tightly controlled, ionic environment for normal neuronal activity
  • Control of respiration
    • The pH of CSF is important in the control of respiration (CO2 freely diffuses into CSF and can activate central chemoreceptors)
  • Nutrition
    • Provides a supply of oxygen, sugars, amino acids to supply the brain


Examiner comments

71% of candidates passed this question.

This question was generally well answered. Better answers noted production including an
amount, site and mechanism. Similarly, absorption included the site, the rate and factors which
affect the rate. The electrolyte and pH and how they compare to extracellular fluid should have
been included in the section on composition.


Online resources for this question


Similar questions

  • Question 16, 2020 (1st sitting)

  • Question 11, 2019 (1st sitting)

  • Question 9, 2017 (2nd sitting)

  • Question 24, 2017 (1st sitting)

  • Question 16, 2015 (1st sitting)

  • Question 2, 2013 (1st sitting)

  • Question 22, 2007 (1st sitting)

  • Question 6, 2008 (2nd sitting)


Question 16

Question

Describe the forces that result in fluid exchange across capillary membranes


Example answer

Fluid exchange across membranes

  • Bulk flow of fluid across a semi-permeable membrane is a balance of Starling forces

  • Can be expressed using the following formula

    • <math display="inline">Bulk \: flow \: = \: \kappa[(P_c \; - \; P_{if}) \; - \; \delta(\pi_p \; - \pi_{if})]</math>

  • Where:

    • Kappa = membrane filtration constant

      • Accounts for membrane permeability and surface area

    • Delta = reflection constant

      • Takes into consideration protein leakage

      • Values range from 0-1

    • Capillary hydrostatic pressure (Pc)

      • Main factor determining bulk flow under physiological conditions

      • Normally ~35mmhg at arterial end, 15mmHg at venous end of capillary.

      • Determined by

        • The ratio of resistances between pre/post capillary arterioles

        • Arterial and venous blood pressure and gravity

    • Interstitial hydrostatic pressure(Pif)

      • Normally ~0mmhg (there is minimal interstitial fluid which is draining away)

      • Affected by anything that modifies lymphatic drainage (e.g. immobility, tourniquet)

    • Plasma oncotic pressure (Ï€p)

      • The osmotic pressure attributed to by large insoluble proteins (e.g. albumin) within plasma

      • Normally ~28mmHg. Does not rapidly change

      • Affected by plasma protein concentrations and intravascular fluid status

    • Interstitial oncotic pressure (Ï€if)

      • Osmotic pressure attributed to by small amounts of insoluble proteins which have leaked into interstitial space

      • Normally ~3mmHg. Does not rapidly change

      • Affected by membrane integrity

  • Using the above values at venous/arterial ends, it is demonstrated that bulk flow occurs

    • OUT of the vessel at arterial end

    • IN to the vessel at venous end

  • Main factor as described is the Capillary hydrostatic pressure gradient (Pc - Pif)


Examiner comments

57% of candidates passed this question.

The expected answer included a clear explanation of Starling’s forces, including an understanding of the importance of the relative difference along the length of the capillary, with approximate values and examples of factors that influence them. Some explanation of what contributed to the hydrostatic or osmotic pressure gained more marks than merely stating there was a pressure. Several candidates digressed to Fick’s law of diffusion or intracellular flow of ions which was not directly relevant to capillary flow.


Online resources for this question


Similar questions

  • Question 19, 2016 (2nd sitting)
  • Question 18, 2011 (1st sitting)



Question 17

Question

Describe ketone bodies including their synthesis and metabolism


Example answer

Ketone Bodies

  • Water soluble molecules, derived from fatty acids, that contain ketone groups
  • Three main compounds: acetoacetate, 3-β-Hydroxybutyrate, and acetone
  • Normal plasma level is <0.6mmol/L


Ketogenesis

  • Ketone bodies can only be produced in the liver
  • β-oxidation of fatty acids in the liver produces acetyl-CoA
  • Acetyl-CoA usually enters the citric acid cycle to produce ATP
  • When large amounts of acetyl CoA are produced they condense to form acetoacetate
  • Acetoacetate is then reduced in the mitochondria to 3-β-hydroxybutyrate (majority) or acetone (minority).


Regulation

  • The body constantly produces small amounts of ketone bodies (even during fed states)
  • When carbohydrate stores are available the main pathway for energy utilisation is glycogenolysis
  • Ketogenesis is accelerated by decreased insulin levels and increased glucagon levels (e.g. in times of starvation or carbohydrate restriction). This leads to increased activity of hormone sensitive lipase and acetyl Coa Carboxlyase which drive ketogensis
  • As the lack of insulin is the main driver of ketogenesis, it explains why Type 1 diabetics develop diabetic ketoacidosis


Metabolism/utilisation

  • Ketone bodies can be used as an energy substrate by
    • Kidney, skeletal muscle and cardiac muscle cells (under physiological conditions)
    • Nervous tissue (during times of starvation)
  • Process
    • Ketone bodies enter mitochondria
    • Ketone body reconstituted to Aceto-acetyl CoA (by SCOT)
    • Cleavage of acetyl group by MAT to form Acetyl CoA
    • Acetyl CoA enters the Citric acid cycle


Examiner comments

35% of candidates passed this question.

Whilst most candidates understood that ketones provided an alternative source of substrate for energy production, many lacked a basic understanding of their synthesis and metabolism Important facts included what ketone bodies are, where they were synthesised, where they were taken up and used as fuel, under what circumstances they are used and the integral role of insulin. Many candidates accurately reproduced the glycolytic and/or the TCA cycle, but this was not being examined, and did not score additional marks. Many candidates incorrectly stated that ketone production was the result of anaerobic metabolism.


Online resources for this question


Similar questions

  • ? None




Question 18

Question

Describe the factors affecting left ventricular function


Example answer

Not 100% if this answer is exactly what they want tbh.


Factors affecting LV systolic function

  • Preload
    • Frank starling mechanism
      • Increased preload > increased sarcomere length > increased force of LV contraction > increased SV
  • Afterload
    • LV is afterload independent (due to compensatory mechanisms)
  • Contractility
    • Anrep effect
      • Means of autoregulating contractility with changes in preload
      • Increase in afterload > increased ESV > increased sarcomere stretch > increased force of subsequent contraction > increased SV
    • Bowditch effect
      • A means of compensating for decreased diastolic filling time with fast heart rates
      • Increased HR > decreased time to expel intracellular calcium > accumulation > increased inotropy
    • Integrity of myofilaments
      • Damaged myocardial tissue > impaired LV contraction (e.g. in ischaemia/infarction)
    • Coordinated depolarisation
      • Suboptimal myocardial depolarisation in the LV > impaired coordination of LV contraction
      • e.g. in heart block, sinus node dysfunction
    • Substrate supply
      • Adequate supply of ATP (derived from glucose,fat, protein) to ensure ability of LV to function as needed
    • Hormones
      • e.g. catecholamines --> increased inotropy/chronotropy/lusitropy
    • Autonomic tone
      • Increased SNS activity / decreased PSNS activity > increased chronotropy and inotropy
    • Drugs
      • E.g. B agonists --> increased chronotropy and inotropy
    • Electrolytes
      • E.g. Calcium: too little = impaired systolic function, too much = impaired diastolic function


Factors affecting LV diastolic function

  • LV diastolic function is determined by it compliance
    • LV systolic function
      • Poor LV systolic function > high end systolic volume > impedes diastolic filling
    • Heart rate
      • Increased HR > shorter time in diastole > reduced compliance; filling is time dependant
    • Lusitropic properties of the ventricle
      • Increased by SNS tone and catecholamines
    • Wall thickness
      • Increased thickness = reduced compliance


Examiner comments

12% of candidates passed this question.

Candidates often misinterpreted the question and described determinants of cardiac output. The answer should have focussed on factors affecting/contributing to normal LV function - not pathological states. Some answers showed a lack of appreciation that normal left ventricular function is afterload independent, due to compensatory reflexes. Answers needed to consider intrinsic and extrinsic factors affecting LV function - the latter (e.g. SNS, PSNS, hormones, drugs) was often left out. Answers needed to consider both systolic and diastolic function. An excellent answer included physiological phenomena such as the Treppe effect, Anrep effect and baroreceptor and chemoreceptor reflexes. Mention of normal conduction and pacing as well as blood supply limited by diastole scored additional marks.


Online resources for this question


Similar questions

  • ? None directly




Question 19

Question

Describe toxicity of local anaesthetic agents


Example answer

Local anaesthetic toxicity

  • Typically occurs ~3mg/kg (without adrenaline) ~6-7mg/kg (with adrenaline) when used regionally
  • CNS and CVS side effects are most evident
  • CNS effects occurring at lower plasma drug concentrations
  • CNS side effects
    • Lower doses: Visual disturbances, perioral numbness, tremors
    • Higher doses: Slurred speech, confusion, decreased level of consciousness
    • Highest doses: Seizures, coma, apnoea
  • CVS side effects
    • Lower doses: Hypertension, tachycardia
    • Higher doses: Hypotension, bradycardia,
    • Highest doses: Cardiovascular collapse, arrhythmias
  • Other effects
    • Methemoglobinemia
    • Allergy


Factors affecting toxicity

  • Patient factors
    • Acidosis: decreases protein binding > increased unbound fraction
    • Increased age: decreased clearance
    • Pregnancy: decreased protein levels > increased unbound fraction
    • Hyperkalaemia: decreased dose required for toxicity
    • Hepatic dysfunction: reduced metabolism > increased risk of toxicity
    • Renal dysfunction: reduced clearance > increased risk of toxicity
  • Drug factors
    • Increasing dose = increased risk of toxicity
    • Type of local anaesthetic
      • e.g. bupivacaine has lower CC/CNS ratio than lidocaine (more likely to be cardiotoxic than CNS toxic)
    • Site of administration: more vascular areas > higher risk
    • Coadministration with vasoconstrictors (e.g. adrenaline) > slower absorption > reduced risk toxicity
    • Drug interactions: displacement from protein binding sites by highly protein bound drugs e.g. phenytoin > increased unbound fraction > increased risk of toxicity


Management of local anaesthetic toxicity

  • Alkalinise
    • Decreases the unbound (active) fraction of the drug
  • Give intralipid
    • Increases the lipid bound fraction (decreases active unbound fraction)


Examiner comments

28% of candidates passed this question.

Most questions lacked a systematic approach to the question and specific detail. The relationship between systemic toxicity (CNS and CVS) and plasma levels should be described. Many candidates did not clearly state that CNS toxicity occurs at lower plasma levels that CVS toxicity. Factors that affect toxicity (e.g. drug factors, patient factors, interactions) needed to be elaborated with some detail. Patient factors such as age, pregnancy, acidosis, hyperkalaemia, hepatic failure were often omitted. Finally, marks were also awarded for noting methaemoglobinaemia as possible toxicity and the existence of specific therapy (intralipid).


Online resources for this question


Similar questions

  • Question 22, 2012 (2nd sitting)
  • Question 3, 2009 (1st sitting)




Question 20

Question

Describe the pharmacology of heparin highlighting important differences between unfractionated and fractionated (low molecular weight) heparin


Example answer

Name HMWH (heparin) LMWH (enoxaparin)
Class Anticoagulant Anticoagulant
Indications Prophylactic and therapeutic anticoagulation (e.g. AF, DVT, PE, ACS etc) Prophylactic and therapeutic anticoagulation (e.g. AF, DVT, PE, ACS etc)
Pharmaceutics MW = 5,000-25,000 Da

Clear solution for injection

MW = 5,000 Daltons

Clear solution for injection

Routes of administration IV, SC SC (main), can also be given IV
Dose Prophylactic: 5,000 IU BD-TDS Therapeutic: infusion (APTT target) Therapeutic: 1mg/kg BD or 1.5mg/kg OD

Prophylactic: 20-40mg OD

pKA
Pharmacodynamics
MOA Heparin binds to antithrombin 3 > conformational change > increases affinity for inactivating thrombin (factor IIa) and Factor Xa Enoxaparin binds to AT-3 > conformational change > increases affinity for inactivating factor Xa (and weakly factor IIa - 4x less activity)
Effects Anticoagulation Anticoagulation
Side effects HAEM: increased risk of haemorrhage, bruising, HITTS (higher than LMWH) HAEM: increased risk of haemorrhage, bruising, HITTS (lower than HMWH)
Pharmacokinetics
Onset Immediate (IV), 30 mins (SC) Peak effect 3-4 hrs post SC injection
Absorption PO bioavailability - 0%

Variable SC absorption

PO bioavailability - 0%
>90% bioavailability post SC injection
Distribution VOD = 0.1L/kg

Lipid solubility: low
Protein binding: high
Does not cross BBB / placenta

VOD = 4.3L

Protein binding: does not bind to heparin binding proteins

Metabolism Reticuloendothelial system Minimal hepatic metabolism
Elimination Renal elimination (very minimal) - hence preferred in renal failure

T 1/2 = 1 hrs

Renal elimination of active and inactive metabolites
T 1/2 = 6-12 hours
Special points Reversal: protamine (1mg = 100IU) - 100%

Monitoring: APTT level

Reversal: protamine (<75% efficacy) Monitoring: Anti-Xa level


Examiner comments

71% of candidates passed this question.

Better answers were tabulated and included sections on pharmaceutics, indications and an explanation on how the difference in molecular weight influenced pharmacodynamics and pharmacokinetics. Knowledge of adverse effects was limited to bleeding and HITTS, often without consideration of relative risk from LMWH. Many candidates did not know the t1/2 of UFH or LMWH.


Online resources for this question


Similar questions

  • Question 5, 2017 (2nd sitting)
  • Question 8, 2009 (2nd sitting)
  • Question 2, 2008 (2nd sitting)


2018 (1st sitting)

Question 1

Question

Describe the carriage of oxygen in the blood, including total oxygen delivery per minute


Example answer

Oxygen is transported in the blood in two main forms:

  • Dissolved oxygen
  • Combined with haemoglobin (oxyhaemoglobin)


Dissolved oxygen

  • Amount of oxygen dissolved in blood is proportional to Henrys Law
  • There is 0.03ml oxygen per 1L blood for each mmHg of PO2 at 37 degrees
  • Thus for PO2 of 100 there is 3 ml dissolved oxygen per 1L blood


Oxyhaemoglobin

  • 98% of oxygen in the blood is carried by haemoglobin

  • Haemoglobin reversibly binds O2 and transports it around the body

  • One haem group binds 1 oxygen molecule. Each Hb molecule binds four O2 molecules

  • Oxygen capacity of Hb (1g of Hb carries 1.34ml Oxygen)

  • Binding of O2 to Hb

    • Hb exists in tense (unbound) and relaxed (bound states)

    • As Hb binds oxygen, it exhibits positive cooperativity (additional binding is easier), as the R state Hb has increased oxygen affinity. Explains sigmoidal shape of oxy-dissociation curve

  • Oxygen bound to Hb does not contribute to PO2 of blood - maintaining diffusion gradient

Oxygen content of blood (CaO2)

  • <math display="inline">CaO_2 = (1.34 \; \times \; [Hb] \; \times \; SaO_2) \; + \; (0.03 \times PO_2)</math>
  • Where [Hb] is the Hb concentration, 1.34 is the oxygen carrying capacity of Hb (Huffners constant), SaO2 is the percentage of Hb saturated with oxyge, 0.03 is the dissolved oxygen content of blood, and PO2 is the partial pressure of oxygen in blood


Oxygen delivery (DO2)

  • Oxygen delivery (DO2) is a function of the cardiac output and oxygen content of blood (CaO2)
    • <math display="inline">DO2 \; = \; CO \; \times CaO_2</math>
    • <math display="inline"> DO2 \; = \; CO \; \times (1.34 \; \times \; Hb \; \times \; SaO_2) \; + \; (0.03 \times PO_2)</math>
  • Assuming CO of 5L/min, 100% sats, 150g/L Hb, PO2 of 100mmHg = 1L/min


Examiner comments

32% of candidates passed this question.

Better answers divided oxygen carriage into that bound to haemoglobin and that carried in the dissolved form. A reasonable amount of detail on the haemoglobin structure and its binding of oxygen was expected, including an explanation of co-operative binding and the oxygen carrying capacity of haemoglobin. Better answers mentioned Henry’s law in the description of dissolved oxygen, along with an estimation of the amount of oxygen that is normally in the dissolved form.
It was expected that answers include the equation for oxygen delivery, a brief description of the components of that equation and the normal value, which a large number of candidates omitted.


Online resources for this question


Similar questions

  • Question 1, 2012 (2nd sitting)


Question 2

Question

Compare and contrast the pharmacology of adrenaline and milrinone


Example answer

Name Adrenaline Milrinone Comments
Class Naturally occurring catecholamine Phosphodiesterase inhibitor Different classes
Indications Haemodynamic support, anaphylaxis, bronchoconstriction/airway obstruction Haemodynamic support for acute heart failure Adrenaline has more/other uses
Pharmaceutics Clear solution, light sensitive (brown glass), 1:1000 or 1:10,000 Yellow solution, 10mg/ml ampoules,
Routes of administration IV, IM, INH, ETT, Topical, subcut IV only in AUS Milrinone only IV in Aus.
Pharmacodynamics
MOA Non-selective adrenergic receptor agonist.

At low doses B effects dominate, at high doses alpha dominate.
Adrenaline > a-1 receptor > increased IP3 (2nd messenger) > increased Ca
Adrenaline > B1,B2,B3 receptors > increased cAMP (second messenger)

PDE III inhibition > decreased cAMP breakdown > increased Ca Different MOA - can be used synergistically
Effects CVS: vasoconstriction (high doses), vasodilation (low doses), increased inotropy + chronotropy

RESP: bronchodilation, increased minute ventilation
METABOLIC: hyperglycaemia (glycogenolysis, lipolysis, gluconeogenesis)
CNS: increased MAC
GIT: decreased intestinal tone/secretions

CVS: increased inotropy, lusitropy, minimal chronotropy, vasodilation Milrinone is cardiovascularly selective.
Side effects Extravasation > tissue necrosis, pHTN due to increased PVR, hyperglycaemia, tachyarrhythmias, May precipitate an arrhythmia, hypotension (vasodilator) Milrinone is a vasodilator and may need adjunct vasopressor
Pharmacokinetics
Onset/Offset Immediate / immediate 5-10 minutes / 3 hours Adrenaline has faster onset/offset
Absorption Zero oral bioavailability due to GIT inactivation. variable/erratic ETT absorption. Readily absorbed orally (tablets not available in AUS) Milrinone readily PO absorbaable
Distribution Poor lipid solubility, doesn't cross BBB, crosses placenta Small VOD = 0.4L/kg, protein binding 80%
Metabolism Metabolised by MAO (mitochondria) and COMT (liver, blood, kidney) to VMA and metadrenaline Minimal hepatic metabolism (10%)
Elimination T 1/2: ~2 mins (due to rapid metabolism)

Metabolites (above) are excreted in the urine

Renal excretion (unchanged 80%). T1/2 = 3 hours Milrinone requires dose adjustment in renal impairment + has longer half life
Special points Dose adjust in renal failure


Examiner comments

45% of candidates passed this question.

This question was best answered using a table. Better answers included: the mechanisms of action, the pharmacokinetics and pharmacodynamics, indications for use and adverse effects. To complete the answer, the two drugs should have been compared and contrasted. There are many areas which could be contrasted e.g. different indications, different mechanisms of action, different half-lives and duration of action, different metabolism and different pharmacodynamic effects, in particular the effects on the cardiovascular system and the pulmonary circulation. Similarities should also have been highlighted.


Online resources for this question


Similar questions

  • Milrinone
    • Question 14, 2011 (2nd sitting)
  • Adrenaline
    • Question 1, 2021 (1st sitting)
    • Question 8, 2012 (1st sitting)



Question 3

Question

Define dead space and its components (30% of marks). Explain how these may be measured (35% of marks) and describe the physiological impact of increased dead space (35% of marks).


Example answer

Dead space

  • The fraction of the tidal volume that does not participate in gas exchange
  • Made up of
    • Apparatus dead space
      • Related to artificial breathing circuits/equipment (e.g. NIV)
    • Physiological dead space (sum of alveolar and anatomical dead space)
      • Alveolar dead space
        • Volume of gas in poorly perfused lung units (West Zone 1)
      • Anatomical deadspace
        • Volume of gas in conducting airways
        • Approx 2ml/kg


Measurement of dead space

  • Physiological deadspace
    • Calculated using the modified version (Enghoff) of the Bohr Equation
      • <math display="inline">\frac {V_D}{V_T} = \frac {P_aCO_2 - P_ECO_2}{P_aCO_2}</math>
  • Anatomical deadspace
    • Can be calculated using Fowlers method
      • Subject exhales to residual volume. Pure oxygen is inhaled to total lung capacity. Subject breathes out through a nitrogen sensor. A nitrogen concentration vs volume can be generated
      • The midpoint of phase 2 = anatomical dead space
  • Alveolar dead space
    • Equals the difference between physiological and anatomical dead space


Impact of increased dead space

  • Increasing dead space has the same effects on gas exchange as decreased tidal volumes
    • Reduced CO2 clearance
    • Decreased oxygenation (due to increased CO2)
  • This results in decreased efficiency of ventilation
    • For any given minute volume, CO2 clearance is reduced
    • Leads to increased minute ventilation > increased work of breathing


Examiner comments

59% of candidates passed this question.

Some candidates failed to provide a correct definition of dead space. An outline of anatomical, alveolar and physiological dead space was expected. The Bohr equation was commonly incorrect, and many did not comment on how to measure the components of the Bohr equation. Fowler’s method was generally well described though some plotted the axes incorrectly.
The impact of increased dead space was not often well explained. Very few people stated the major impact of increased dead space is reduced minute ventilation and how this would affect CO2.


Online resources for this question


Similar questions

  • Question 19, 2010 (1st sitting)




Question 4

Question

Describe the renal handling of sodium


Example answer

Renal handling of sodium

  • Sodium is freely filtered in the glomerulus
  • PCT:
    • ~60-70% reabsorbed
    • Driven predominately by Na/K ATPase pump on the basolateral membrane which creates an electrochemical gradient for Na to flow down into.
  • dLOH
    • Nil reabsorbed (impermeable)
  • aLOH
    • ~25% reabsorbed
    • Driven by the Na-K-2Cl co-transporter
  • DCT
    • ~5% Na reabsorbed
    • Driven by the Na-Cl co-transporter
  • Collecting duct
    • <5% Na reabsorbed
    • Driven by ENaC channels


Regulation

  • Tubuloglomerular feedback (release of renin in response to reduced Na/flow sensed at the JGA)
  • Aldosterone
    • Increases ENaC and Na/K ATPase activity in the DCT and collecting ducts
  • Angiotensin II
    • Increases Na/K ATPase activity on basolateral membrane - creates electrochemical gradient
    • Increases Na-H reabsorption on luminal membrane in PCT
  • ANP
    • Inhibits ENaC (in collecting ducts)
  • Pharmacological agents


Examiner comments

46% of candidates passed this question.

A description of filtration and reabsorption, including amounts was required. Better answers described sodium handling in a logical sequence as it progressed through the nephron including the percentages reabsorbed in each segment. In addition to the amounts reabsorbed, the mechanisms of transport across the tubular luminal and basolateral membranes into interstitial space should have been described.


Online resources for this question


Similar questions

  • Question 22, 2009 (2nd sitting)
  • Question 23, 2014 (2nd sitting)



Question 5

Question

Compare and contrast the pharmacokinetics and pharmacodynamics of IV fentanyl and IV remifentanil (60% of marks). Discuss the concept of context sensitive half-time using these drugs as examples (40% of marks).


Example answer

Name Fentanyl Remifentanyl
Class Opioid

Synthetic phenylpiperidine derivative

Opioid

Synthetic phenylpiperidine derivative

Indications Analgesia Analgesia
Pharmaceutics Colourless solution (50ug/ml) Crystalline white powder for reconstitution
pKa 8.4 7.3
Routes of administration SC, IM, IV, epidural, intrathecal, transdermal IV, intranasal
Pharmacodynamics
MOA Mu-opioid receptor agonist > hyperpolarisation Mu-opioid receptor agonist > hyperpolarisation
Effects Analgesia Analgesia
Side effects CVS: bradycardia

Resp: respiratory depression, blunted cough reflex
GIT: decreased GI motility, nausea/vomiting
CNS: Dysphoria, confusion,

CVS: bradycardia + hypotension

Resp: respiratory depression
GIT: Decreased GI motility
MSK muscle rigidity at high doses

Pharmacokinetics
Onset/Offset Rapid onset (2-5 mins)

Rapid offset (30mins)

Rapid onset (1 mins)

Rapid offset (5-10mins)

Absorption PO bioavailability (33%). Mucosal absorption is poor PO Bioavailability (0%). Mucosal absorption is rapid
Distribution VOD high = 6L / kg

Highly protein bound (90%)
Good lipid solubility

VOD low = 0.1L/kg

Highly protein bound (70%)
Very lipid solubility

Metabolism Hepatic metabolism > demethylation > inactive metabolites Ester hydrolysis by plasma and tissue esterases > inactive metabolites
Elimination T 1/2 = 4 hours, prolonged with infusions.

Excreted in urine

T 1/2 5 mins. No CSHT

Excreted in urine


Context sensitive half time (CSHT)

  • CSHT is the time required for 50% decrease in central compartment drug concentration after an infusion of the drug is ceased (context refers to the duration of infusion)
  • Drugs with high VOD and minimal metabolism (e.g. fentanyl) will have different CSHT depending on the duration of infusion
    • Short infusion = short CSHT. Long infusion = long CSHT
  • Drugs with small VOD and extensive metabolism (e.g. remifentanil) has a CSHT which is independent of duration of infusion


Examiner comments

66% of candidates passed this question.

Well-constructed answers were presented in a table to compare pharmacokinetics and pharmacodynamics with a separate paragraph to discuss the concept of context sensitive half-time. Important pharmacokinetic points included: the differences in lipid solubility, ionised fractions and onset, and differences in metabolism. Marks were awarded for a definition of context-sensitive half-time. A discussion of these two drugs’ context-sensitive half-times should have included the differences in re-distribution into other compartments and rates of elimination.


Online resources for this question


Similar questions

  • Fentanyl and remifentanyl
    • Question 16, 2011 (1st sitting)
  • Fentanyl
    • Question 12, 2016 (2nd sitting)
    • Question 13, 2017 (2nd sitting)



Question 6

Question

Define a buffer (25% of marks). Describe how acid and base shifts in the blood are buffered (75% of marks).


Example answer

Buffers

  • A solution consisting of a weak acid and its conjugate base
  • Main function is to resist change to pH, with the addition of stronger acids/bases, through reversible binding of H+ ions
  • Effectiveness depends on the buffer pKa, the pH of the solution, the amount of buffer present, whether the system is open or closed
  • All buffers participate in equilibrium with each other in defence of pH (Isohydric principle)


MAIN BUFFER SYSTEMS


Bicarbonate-carbonic acid system

  • pKa of 6.1
  • Consists of weak acid (H2CO3) and base (HCO3 salt)
  • Via reaction: <math display="inline"> CO_2 \; + H_2O \; \leftrightarrow \; H_2CO_3 \; \leftrightarrow \; HCO_3^- \; + H^+</math>
  • Increased acid > increased CO2 (excreted via lungs)
  • Increased base > increased HCO3 (excreted via kidneys)
  • OPEN system - hence most important - responsible for 80% of the ECF buffering


Protein buffering system (including Hb)

  • Includes haemoglobin (150g/L) and plasma proteins (70g/L)
  • Proteins buffer by binding H+ to imidazole side chains of their histidine residues
  • Hb is quantitatively 6 times more important than plasma proteins, as the concentration is double and there are three times as many histidine residues in Hb compared to plasma proteins
  • Hb has pKa of 6.8. Weak acid (HHb) and weak base (KHb)
  • Mechanism: H+ binds to the histidine residues on imidazole side chains, the HCO3 diffuses down concentration gradient into ECF


Phosphate buffering system

  • Overall pKa 6.8
  • Tribasic (HPO4, H2PO4, H3PO4) though only the H2PO4 has a physiological pKa to be useful
  • Overall contribution is minimal to the blood due to the low concentration of phosphate. However more important in the urine where the concentration is higher
  • closed system


Examiner comments

45% of candidates passed this question.

Few candidates defined a buffer making it difficult to award 25% of the marks for this question. The three main buffers in blood should have been described: bicarbonate system, haemoglobin and proteins. The pKa, the buffering mechanism and the capacity of the system should have been described. The Henderson Hasselbach equation was sometimes incorrect. Marks were only awarded for buffers in blood and unfortunately some candidates described non-blood buffers.


Online resources for this question


Similar questions

  • Question 11, 2021 (1st sitting)
  • Question 22, 2014 (1st sitting)
  • Question 19, 2011 (1st sitting)
  • Question 3, 2008 (2nd sitting)



Question 7

Question

Outline the blood supply to the gastrointestinal system (arteries and veins).


Example answer

Arterial supply

  • The aorta (and its branches) supplies the entire arterial supply to the GIT
  • The oesophagus is supplied by various arterial branches
    • Cervical portion- inferior thyroid artery
    • thoracic portion - bronchial arteries
    • Abdominal portion - left gastric, inferior phrenic arteries
  • The abdominal aorta then has three main branches which supply the remainder of the GIT
    • Celiac trunk
      • Arises from abdominal aorta immediately below aortic hiatus at T12/L1
      • Divides into left gastric artery, splenic artery, common hepatic artery
        • Left gastric a. (supplies stomach)
        • Splenic a. (supplies spleen, pancreas)
        • Common hepatic, divides into
          • Hepatic a. proper (supplies liver)
          • Gastroduodenal (supplies pancreas, duodenum, stomach)
          • Right gastric (supplies stomach)
    • Superior mesenteric artery (SMA)
      • Arises from abdominal aorta immediately interior to coeliac trunk (L1)
      • Multiple branches (15-20) which joint in an arcade
      • supplies the midgut structures (from duodenum to 2/3 transverse colon)
    • Inferior mesenteric artery (IMA)
      • Arises from abdominal aorta ~L3
      • Multiple branches (including Left colic, sigmoid, superior rectal arteries), join in arcade
      • Supplies the hindgut (distal 1/3 transverse colon - rectum)


Venous drainage

  • For the most part, the venous drainage of the GIT is via veins which accompany the arterial system

  • They return via the portal vein

    • Portal vein

      • Combination SMV and splenic vein

      • Receives drainage from forgut structures

    • Splenic vein

      • Travels along with the splenic artery + drains corresponding regions (foregut)

      • Combines with SMV to form portal vein

    • Superior mesenteric vein (SMV)

      • Travels along with the SMA + drains corresponding regions (midgut)

      • Combines with splenic vein to form portal vein

    • Inferior mesenteric vein (IMV)

      • Travels along with the IMA + drains corresponding regions (hindgut)

      • Drains into the splenic vein

Examiner comments

7% of candidates passed this question.

An outline of the blood supply from the oesophagus down to the anus was expected. Very few candidates knew the branches of the main 3 arteries and which portion of the gastrointestinal system they supplied. Concepts related to control of blood flow and autoregulation of blood flow were not asked and therefore marks were not awarded for this information.


Online resources for this question


Similar questions

  • Question 7, 2021 (1st sitting)



Question 8

Question

Outline the principle of co-oximetry (40% of marks), describe what a co-oximeter is able to measure (30% of marks), and compare its limitations to those of a pulse oximeter (30% of marks).


Example answer

CO-oximetry

  • A laboratory device that uses spectrophotometry to measure relative blood concentrations of Hb species
  • Principle
    • Blood sample is heparinised, heated to 37 degrees, haemolysed by vibations
    • Multiple wavelengths of light are then passed through the sample and the absorption spectra is assessed, using principles of Beer-Lambert law
    • There is no need for pulsatile flow
  • There is becoming available pulse co-oximetry which is similar to pulse-oximeters though can detect some of the other Hb species (e.g. COHb)


Measured indices

  • SaO2 %
  • Total [Hb]
  • Met Hb %
  • Sulpha Hb %
  • CO Hb %
  • Most co-oximetry machines can also obtain all the regular blood gas tensions/values


Interpretation

High pulse oximetry Low pulse oximetry
High Co-oximetry Reflects normal SpO2 Reflects normal SpO2

Differences possibly due to:
1) Poor tissue perfusion + shock
2) Dyes (e.g. methylene blue)
3) Tricuspid regurgitation
4) Poor probe contact
5) Contamination (ambient light)

Low Co-oximetry Reflects low SpO2

Differences possibly due to:
1) Carboxyhaemaglobinaemia
2) Methaemaglobinaemia
3) Radiofrequency interference

Reflects low SpO2


Co-oximeter vs Pulse-oximeter

  • Advantages of co-oximetry
    • More accurate sats (i.e. low reading = low sats) as accounts for other Hb species
    • Not confused by ambient light, poor tissue perfusion, dyes etc
    • Does not require pulsatile flow
  • Disadvantages of co-oximetry
    • More invasive (requires blood sample) - though pulse co-oximetry becoming available
    • Heavy machinery, requiring calibration, less accessible
    • Not continuous measurements


Examiner comments

32% of candidates passed this question.

Most candidates confused co-oximetry with other methods of measuring oxygenation of blood. Whilst there were a number of excellent descriptions of pulse oximetry, these attracted no marks for the first two sections. Structuring the answer based on the three parts asked, would have improved answers ensuring all aspects of the question were addressed.


Online resources for this question


Similar questions

  • None



Question 9

Question

Describe the functions of the placenta (80% of marks). Outline the determinants of placental blood flow (20% of marks).


Example answer

FUNCTIONS OF PLACENTA


Nutrient/gas exchange

  • The foetus relies on maternal transfer of gasses, nutrients and wastes

  • Nutrients/wastes

    • Active transport e.g Amino acids, calcium, some vitamins/minerals

    • Facilitated diffusion e.g. glucose (GLUT1 and GLUT3)

    • Passive diffusion e.g. Na, Cl, urea, creatinine

  • Gasses

    • Oxygen

      • Passive diffusion

      • Facilitated by higher oxygen carrying capacity and affinity of foetal Hb as well as the Bohr/Double bohr effects

    • Carbon dioxide

      • Passive diffusion

      • Facilitated by the Haldane and double Haldane effects

Immunological function

  • Foetus is genetically distinct with a non functioning immune system
  • Trophoblast cells
    • Lose MHC molecules and become coated in mucoprotein > less immunogenic
  • Chorionic cells
    • Prevent maternal T cells and most immunoglobulins (except IgG) from entering > less immunogenic
    • Barrier to some bacteria/viruses and allows IgG across > some immune protection
  • Yolk sac
    • a-fetoprotein and progesterone are immunosuppressive > less immunogenic


Endocrine function

  • Syncytiotrophoblast of placenta produces
    • B-HCG - prolongs corpus luteum (prevents early miscarriage)
    • Oestrogen - increases uteroplacental blood flow, stimulates uterine growth
    • Progesterone - uterine relaxation, development of lactation glands
    • hPL - maternal lipolysis, breath growth/development


PLACENTAL BLOOD FLOW


Flow

  • Blood flow to the uterus in a non pregnant woman is normally around 200mls/min (~4% of CO)
  • In a pregnant woman at term this increases to up to 750mls/min (~15% of CO)
  • Majority of this > placenta, with some supplying the hypertrophied uterus.


Determinants of flow

  • No autoregulation of uteroplacental blood flow

  • Most important factor governing flow is therefore perfusion pressure

    • Increased uteroplacental perfusion pressure > increase flow

  • Uterine perfusion pressure is therefore effected by

    • Maternal MAP

      • Effected by positioning (e.g. aortocaval compression), cardiac output, systemic vascular resistance

    • Intrauterine pressure

      • Effected by contractions > increased intrauterine pressure > decreased flow

    • Uterine vascular ressistance

      • Modestly effected by exogenous vasopressors, catecholamines

  • Compensates for the lack of autoregulation by increasing oxygen extraction

Examiner comments

32% of candidates passed this question.

Many candidates provided a broad overview of functions of the placenta but lacked detail. Placental blood flow has maternal and foetal components, though most only considered the maternal circulation to the placenta and didn't mention the foetal vessels. Many were not specific as to what blood vessels were described.
Many stated that uterine blood flow is not autoregulated, however went on to describe myogenic and neuro-humoral mechanisms of autoregulation.


Online resources for this question


Similar questions

  • Question 6, 2021 (2nd sitting)



Question 10

Question

Outline the advantages (15% of marks) and disadvantages (85% of marks) of the clinical use of suxamethonium.


Example answer

Suxamethonium

  • Depolarising muscle relaxant

  • Used to facilitate endotracheal intubation during anaesthesia (i.e. RSI)

  • MOA: Binds to the nACh receptor on motor end plate > depolarisation. Cannot be hydrolysed by Acetylcholinesterase in NMJ > sustained depolarisation > muscle relaxation

Advantages

  • Cheaper than other NMB agents

  • Pre-mixed

  • Can be IV or IM

  • Rapid onset (<1 min)

  • Rapid offset (< 10 mins)

  • Safe in pregnancy/neonates

  • Not end-organ dependant for metabolism (plasma cholinesterase)

Disadvantages

  • Needs to be stored at 4 degrees
  • Numerous side effects
    • Major: anaphylaxis, suxamethonium apnoea, malignant hyperthermia

Minor: hyperkalaemia, myalgia, fasiculations, bradycardia/arrhythmia
Pressure related: increased IOP, ICP, intragastric pressure.

  • Numerous contraindications
    • Hyperkalaemic patients and those at risk (renal failure, sepsis, burns)
    • Burns patients
    • Personal/family history of malignant hyperthermia or plasma cholinesterase deficiency
    • Muscular dystrophies, myasthenia gravis
    • Penetration eye injury
  • Issues with repeat dosing
    • Repetitive dosing may lead to phase 2 (depolarising) block > requiring reversal


Examiner comments

46% of candidates passed this question.

This commonly used drug should be very well-known. The question asked for an outline, hence long explanations of various aspects of pharmacology (e.g. pseudocholinesterase deficiency) were unnecessary.
Headings should have included: advantages (e.g. rapid onset, rapid offset, short acting, IV or IM administration, not end organ dependent for metabolism, premixed, safe in pregnancy and neonates). The disadvantages section should have included the following headings: pharmaceutical, adverse drug reactions (including several potentially fatal ones), numerous contraindications, unpleasant side-effects and potential problems with repeat dosing.


Online resources for this question


Similar questions

  • Question 6, 2011 (1st sitting)
  • Question 2, 2012 (2nd sitting)
  • Question 1, 2013 (2nd sitting)
  • Question 10, 2020 (2nd sitting)



Question 11

Question

Describe the regulation of the coronary circulation


Example answer

Coronary blood flow

  • Normal resting coronary artery blood flow (CBF) is ~250mls/min (5% CO)
  • RCA: blood flow is constant, pulsatile and higher flow rate during systole
  • LCA: blood flow is intermittent, pulsatile, and higher flow rate during diastole
  • Oxygen extraction is high (70%) and near maximal - increased CBF is needed for increased O2 demand.


Regulation of flow

  • Autoregulation
    • CBF is autoregulated over a wide range of BPs (perfusion pressure 50-120mmhg)
    • Metabolic autoregulation
      • Anaerobic metabolism > increased vasoactive substances (lactate, adenosine, CO2, NO) > vasodilation > increased flow
      • Predominant means of autoregulation
    • Myogenic autoregulation
      • Increased transmural pressure > vasoconstriction > flow reduction
      • Modest means of autoregulation
  • Direct autonomic control
    • Weak effect
    • a1 activation > vasodilation; B/muscarinic activation > vasoconstriction
  • Indirect autonomic control
    • Increase / decrease HR to alter time in diastole/systole which will lead to increased/decreased flow
    • i.e. Increased PSNS activity > decreased HR > increased diastolic time > increased CBF
  • External (e.g drugs)
    • Nitrates (dilate)
    • BBlockers (reduce HR > reduced O2 use and increased diastole time)
    • CCB (coronary vasodilation)


Examiner comments

46% of candidates passed this question.

Some answers suffered from listing things rather than describing things as the question required.
Better answers included a description of metabolic, physical and neuro-humoral factors and the relative importance of each.
Many described detailed anatomy which was not necessary.


Online resources for this question


Similar questions

  • Question 11, 2008 (2nd sitting)
  • Question 10, 2014 (2nd sitting)
  • Question 19, 2021 (1st sitting)



Question 12

Question

Briefly describe the cardiac events that occur during ventricular diastole.


Example answer

Cardiac cycle

  • Can be broken into two phases: systole and diastole
  • Diastole is the phase corresponding to ventricular relaxation
  • Diastole can be broken into 4 stages (below; using LV as example)


Isovolumetric relaxation

  • Aortic valve closes (producing 2nd heart sound) ending systole, beginning diastole
  • LV begin to relax without any change in volume > decreasing LV pressure
  • There is ongoing LA filling leading to increased LA pressure and the V wave.
  • Corresponds with the peak of the T wave


Early diastolic (rapid ventricular) filling

  • When LV pressure < LA pressure the mitral valve opens
  • This leads to increased LV volume and reduced LA pressure (the y descent on CVP waveform)
  • With continued ventricular relaxation there is ongoing decrease in LV pressure
  • This corresponds to the 3rd heart sound and isoelectric baseline on ECG
  • There remains no further aortic flow
  • Coronary blood flow is highest


Late diastolic (reduced ventricular or diastasis) filling

  • Ongoing slow ventricular filling leading to gradual rise in atrial, ventricular and venous pressures as well as ventricular volume
  • Corresponds to isoelectric baseline on ECG just prior to P wave


Atrial systole

  • Begins just after the start of the P wave on ECG and finish before Q wave
  • Leads to atrial contraction which increases atrial pressure, and leads to further ejection of blood into the ventricles (increasing LV volume and pressure).
  • Atrial contraction produces the a wave on the CVP trace
  • Fourth heart sound heard here: caused by oscillation of blood into ventricles following atrial systole


Examiner comments

29% of candidates passed this question.

Many answers lacked structure and contained insufficient information. Better answers defined diastole and described the mechanical events in the 4 phases of diastole. A common error was the ECG events in diastole. The electrical events and coronary blood flow should have been mentioned.


Online resources for this question


Similar questions

  • Question 21, 2011 (1st sitting)



Question 13

Question

Explain the difference between viscosity and density (10% of marks). Describe the effects of changes in viscosity and density on the flow of gases and liquids (90% of marks).


Example answer

Viscosity (n)

  • Liquids/gas internal resistance to flow


Density (p)

  • Mass of a substance per unit volume


Flow (Q)

  • The volume of liquid/gas moved per unit time
  • Can be laminar, turbulent or transitional - determined by Reynolds number


Reynolds number

  • <math display="inline">Re \; = \; \frac {2 \; r \; v \; p} {n}</math>
  • Where r = radius, v=velocity, p=density, n=viscosity
  • If Reynolds number is
    • Re <2000 = laminar flow
    • Re 2000-4000 = transitional flow
    • Re >4000 flow is predominately turbulent
  • Increased density (p) = increased Reynolds number = more likely to be turbulent flow
  • Increased viscosity (n) = decreased Re = more likely to be laminar flow
  • Density is a more important determinant of Re


Laminar flow

  • Smooth flow of gas in layers that do not mix
  • Flow is proportional to driving pressure, linear relationship
  • Flow (Q) rate can be calculated using the Hagen-Poiseuille equation
  • <math display="inline">Q = \frac {\pi r^4 \Delta P}{8nl}</math>
  • <math display="inline">Resistance (R) = \frac {8nl} {\pi r^4}</math>
  • Therefore viscosity (n), not density, affects the laminar pressure-flow relationship
    • Increased viscosity = increased resistance = decreased flow


Turbulent flow

  • Eddies and swirls of gas that mix layers of gas

  • Flow is proportional to the square root of driving pressure, non linear relationship

  • Resistance increases in proportion to flow rate, but cannot be described using the Hagen-Poiseuille equation but instead by the Fanning Equation

  • Density (p), not viscosity, affects the turbulent pressure-flow relationship

Examiner comments

46% of candidates passed this question.

Whilst most candidates defined density correctly, there was a lot of uncertainty regarding viscosity. Most candidates recognised that flow may be laminar, turbulent or transitional. Most accurately recounted Reynolds number and applied this correctly. Additionally, the Poiseuille equation was correctly stated by most candidates and correctly related to laminar flow. Few candidates recalled the equation describing turbulent flow.


Online resources for this question


Similar questions

  • ? None



Question 14

Question

Classify anticholinesterase drugs according to chemical interaction with an example of each (30% of marks). Outline the pharmacodynamic effects of anticholinesterase drugs and their clinical indications (70% of marks).


Example answer

Anticholinesterase drugs

  • Bind to and inhibit the action of Acetylcholinesterase (AChE)
  • AChE is an enzyme found in synaptic clefts which hydrolyses acetylcholine (ACh) into choline and acetate, terminating synaptic transmission


Anticholinesterase drugs classification (according to method of inhibition)

  • Reversible antagonists (via electrostatic binding)
    • E.g. edrophonium
  • Reversible antagonist (via covalent bonding, susceptible to hydrolysis)
    • e.g. neostigmine, physostigmine
  • Irreversible antagonist (via covalent bonding, resistant to hydrolysis)
    • e.g. organophosphates, insecticides, nerve gases


Pharmacodynamic effects

  • Anticholinesterase drugs inhibit AChE at both muscarinic and nicotinic ACh receptors
  • Nicotinic effects (nAChR) - "target"
    • Reversal of non-depolarising NMBs
  • Muscarinic effects (mAChR) - "off target"
    • CVS: bradycardia, hypotension
    • RESP: bronchoconstriction/spasm
    • CNS: miosis, cholinergic syndrome (confusion, agitation, nausea)
    • GIT: hypersalivation, increased GIT motility, N/V
    • GUT: urination/incontinence
    • OTHER: diaphoresis, lacrimation


Clinical indications

  • Reversal of non-depolarising neuromuscular blockers
    • Increased synaptic ACh competes with non-depolarising NMBs for nAChR > reversal of NMB (e.g. neostigmine, plus atropine/glycopyrrolate to offset AEs)
  • Diagnosis + treatment of myasthenia gravis
    • Increased synaptic ACh > competes with myasthenia autoantibodies for nACHR > increased muscle strength (e.g. pyridostigmine)
  • Treatment of neurodegenerative disorders
    • Increased synaptic ACh > increased cholinergic transmission (e.g. donepezil)
  • Treatment of glaucoma
    • Increased ACh > mAChR > miosis > decreased IOP (e.g. physostigmine)
  • Treatment of anticholinergic syndrome
    • features: delirium, tachycardia, dilated pupils, agitation, seizures
    • Drugs: antiparkinsons, atropine, anti histamines, antispasmodics
    • Management: physostigmine > increase ACh


Examiner comments

32% of candidates passed this question.

Many candidates who scored poorly confused anticholinesterase drugs with anticholinergic drugs. Some described pharmacokinetics when it was not asked. Similarly, treatment of organophosphate poisoning and/or cholinergic crisis was not asked for in the question.


Online resources for this question


Similar questions

  • Question 21, 2015 (1st sitting)



Question 15

Question

Describe the physiological regulation of intracranial pressure


Example answer

Intracranial pressure (ICP)

  • ICP = The pressure within the intracranial space, relative to atmospheric pressure
  • Normal ICP is < 15 mmHg
  • Governed by the Monro-Kellie doctrine
  • There is rhythmic variation in ICP due to variations in respiration and blood pressure


Monro-Kellie doctrine

  • The skull is a rigid container of fixed volume
  • The skull contents include: brain (~1400ml), CSF (~150ml), blood (~150ml)
  • Therefore any increase in volume of one substance must be met by a decrease in volume of another, or else there will be rise in the ICP


Physiological regulation of ICP

  • Brain tissue
    • No capacity to alter volume under physiologically normal circumstances
  • CSF
    • CSF can be displaced from the cranium into the spinal subarachnoid space (as the spinal meninges have better compliance)
    • With increased ICP there is also increased driving pressure for CSF reabsorption
  • Blood
    • Compression of the dural venous sinuses can displace venous blood from the cranium


Pressure volume relationship

  • Hyperbolic relationship - indicating that there is limited capacity to buffer increased volume, before large increases in ICP

image-20211012142839178
image-20211012142839178


Examiner comments

45% of candidates passed this question.

A definition and a normal value were expected. A description of the Monro-Kellie doctrine was expected. Better answers divided into the various components of the cranium with the answer focussing on cerebral blood volume and CSF volume as the brain tissue as no capacity to change its volume.


Online resources for this question


Similar questions

  • Question 18, 2016 (2nd sitting)
  • Question 14, 2016 (1st sitting)
  • Question 18, 2010 (1st sitting)



Question 16

Question

Compare and contrast the pharmacology of furosemide (frusemide) and acetazolamide


Example answer

Name Frusemide Acetazolamide Notes
Class Loop diuretic Carbonic anhydrase inhibitor Different class
Indications Oedema/fluid overload, renal insufficiency, hypertension Metabolic alkalosis, glaucoma, altitude sickness Different indications
Pharmaceutics Tablet, clear colourless solution (light sensitive), White scored tablets (250mg), colourless solution -
Routes of administration IV, PO, PO, IV Both available IV and PO
Dose Varies (~40mg daily commonly used for well patients, can be sig. increased) 125mg-1g, up to 4 hourly -
pKA 3.6 (highly ionised; poorly lipid soluble) pKa 7.2 Acetazolamide more lipid solu
Pharmacodynamics
MOA Binds to NK2Cl transporter in the thick ascending limb LOH, leads to decreased Na,K, Cl reabsorption > decreased medullary tonicity + Inc Na/Cl delivery to distal tubules > decreased water reabsorption > diuresis Inhibits carbonic anhydrase in PCT > decreased reabsorption of filtered HCO3 Different MOA
Effects Renal: diuresis

CVS: hypovolaemia, arteriolar vasodilation + decreased preload (=mechanism for improvement of dyspnoea before diuretic effect in APO)
Renal: increase in RBF

CNS: decreased IOP by decrease aqueous humour

RENAL: diuresis, decreased HCO3 reabsorption (metabolic acidosis),

Both lead to diuresis/hypovolaemia. Acetazolamide has extra-renal effects (e.g. IOP effects)
Side effects CVS: hypovolaemia, hypotension

Renal/metabolic: Metabolic alkalosis, LOW Na, K, Mg, Cl, Ca, increased Cr
Ototoxicity, tinnitus, deafness

CNS: paraesthesia, fatigue, drowsiness

RENAL: hypoNa, HypoK, HyperCl
GIT: Nz/Vz/Dz

Both lead to electrolyte disturbances (hypoNa and HypoK). Frusemide > metabolic alkalosis, Acetazolamide > metabolic acidosis
Pharmacokinetics
Onset 5 mins (IV), 30-60 mins (PO), Effect lasts 6 hours. Onset 1-2hrs Frusemide has faster onset
Absorption Bioavailability varies person-person (40-80%) PO bioavailability 60% Similar
Distribution Vd = 0.1L/Kg, 95% protein bound (albumin) 95% protein bound, VOD 0.3L/kg Both have small VOD and are highly protein bound
Metabolism < 50% metabolised renally into active metabolite Nil metabolism Acetazolamide not metabolised
Elimination Renally cleared (predominately unchanged). T1/2 ~90 mins. Renal clearance, T 1/2 = 6hrs Acetazolamide has longer T1/2
Special points Deafness can occur with rapid administration in large doses


Examiner comments

30% of candidates passed this question.

The use of a table assisted with both clarity and the ability to compare the two drugs. Writing separate essays about each makes it difficult to score well. It was expected that candidates would follow a standard pharmacology format and discuss pharmaceutics, pharmacokinetics, pharmacodynamics and adverse drug reactions. Both of these drugs are ‘Level A’ in the syllabus and a suitable level of detail was expected.
It was expected candidates would discuss in detail the mechanism of action, electrolyte and acid-base effects. Pharmacokinetic values were poorly answered. Qualitative terms such as ‘moderate, good and some’ are vague and should be avoided. Only correct numerical values (or ranges) attracted full marks.


Online resources for this question


Similar questions

  • Frusemide + acetazolamide
    • Question 20 , 2013 (1st sitting)
  • Frusemide
    • Question 18, 2019 (1st sitting)
    • Question 14, 2020 (1st sitting)



Question 17

Question

Define the osmolality and tonicity of an intravenous fluid (20% of marks). Compare and contrast the pharmacology of intravenous Normal Saline 0.9% and 5% Dextrose (80% of marks).


Example answer

Osmolality

  • The measure of solute concentration per unit mass of solvent.
  • Measured in osmoles / kg solvent


Tonicity

  • The measure of the osmotic pressure gradient between two solutions separated by a semi permeable membrane
  • Only influenced by the solutes which cannot cross the semi-permeable membrane
  • Can be hypotonic, isotonic, hypertonic


Normal saline vs 5% dextrose

Name 0.9% normal saline (IV) 5% dextrose (IV)
Class Crystalloid fluid Crystalloid fluid
Pharmaceutics Clear solution, various volume bags (e.g. 100ml, 500mls, 1L)
Clear solution, various volume bags (e.g. 1L, 500mls)
Osmolality 308 mOsm / Kg (calculated)

286 mOsm/kg (measured)

278 mOsm/kg
Tonicity Isotonic Hypotonic (dextrose rapidly metabolised)
Contents 9g NaCl / 1L solution 50g dextrose / 1L solution
Pharmacodynamics
MOA Expands the ECF volume and changes biochemistry of body fluids Expands ECF volume and changes body fluid biochemistry
Effects Increased ECF volume Increased ECF volume

Glucose replacement

Side effects Fluid overload, hyperchloraemic metabolic acidosis, electrolyte imbalances Fluid overload, cerebral oedema, hyperglycaemia, vein irritation, electrolyte imbalances (e.g. HypoNa)
Pharmacokinetics
Onset Immediate (IV) Immediate (IV)
Absorption IV bioavailability = 100% IV bioavailability = 100%
Distribution VOD = 0.2 L/Kg

> 25% intravascular
> 75% interstitial

VOD = 0.6L/Kg

> 5% intravascular
> 25% interstitial
> 70% intracellular

Metabolism Not metabolised Metabolised by all body tissues (esp liver) into water and CO2
Elimination Renal Water eliminated renally, CO2 eliminated by lungs


Examiner comments

29% of candidates passed this question.

Most candidates gave an adequate definition of osmolality and tonicity. A single concise sentence for each attracted full marks. Some candidates drew diagrams & equations, which added few marks. Some candidates confused osmolarity (mOsm/L) and osmolality (mOsm/kg).
Tonicity was best defined as the number of ‘effective’ osmols (those that cannot cross the cell membrane) in a solution relative to plasma. The use of a table greatly facilitated the comparison of 0.9% saline and 5% dextrose solutions. Values for composition, osmolarity and osmolality were poorly done. Some manufacturers state calculated values and some approximate values on the bags – both were accepted.
No candidate correctly pointed out the fluids respectively have 9g NaCl and 50g dextrose per litre.


Online resources for this question


Similar questions

  • None



Question 18

Question

Compare and contrast non-invasive oscillometric and invasive arterial blood pressure monitoring.


Example answer

Non-invasive oscillometric vs invasive arterial BP monitoring

Invasive arterial BP Oscillometric (non invasive) BP
Equipment - Arterial catheter

- Incompressible tubing
- Pressure transducer
- Counterpressure fluid
- Microprocessor

- Inflatable cuff

- Cuff manometer
- Release valve
- Microprocessor

Method/ principles 1) Pressure wave of the arterial blood is transmitted via a fluid column to a transducer

2) Pressure changes converted to resistance changes in Wheatstone bridge transducer
3) Converted to electrical signal > transmitted to microprocessor
4) Microprocessor uses Fourier analysis to breakdown waves

1) counterpressure (cuff) applied to limb over artery (e.g. brachial)

2) cuff inflated above SBP
3) cuff deflated slowly, measuring amplitude of the pulse pressure which is transmitter to cuff
4) Maximal amplitude of PP = MAP
5) SBP and DBP are then derived

Advantages - Gold standard BP measurement (all variables directly derived)

- Can measure continuously
- Can derive other variables (e.g. CO)
- Can be used to generate waveform which can be used clinically

- Non invasive

- Relatively cheap
- Convenient and fast to obtain
- Reusable

Disadvantages - More expensive

- More invasive
- Takes time / less portable
- All the risks associated with arterial puncture (infection, thrombosis etc)
-Not re-usable

- Less accurate

- Not continuous

Sources of error - Incorrect position of transducer

- Incorrect calibration
- Counterpressure bag not adequately inflated
- Damping and resonance

- Wrong cuff size

- Movement
- Arrhythmias
- Faint pulse (e.g hypotension and peripheral vascular disease)


Examiner comments

52% of candidates passed this question.

There were some good answers, though invasive BP measurement was better answered than oscillometry. Many candidates provided extensive detail in one area i.e. the workings of a Wheatstone bridge, to the detriment of a balanced answer.
Few seemed to have a structure consisting of "equipment, method, sources of error, advantages, disadvantages" or similar and missed providing important information as a result. Several described auscultatory non-invasive blood pressure measurement, rather than oscillometry, which although related in principle is a different process.


Online resources for this question


Similar questions

  • None



Question 19

Question

Explain the mechanisms by which normal body temperature is maintained and regulated


Example answer

Overview

  • Temperature: the average kinetic energy of the atoms/molecules that make up a substance

  • Human 'core temperature' is the 'deep body' temperature of the internal organs and viscera

  • Humans maintain a core temperature of 37°C <math display="inline">\pm</math> 0.4°C, despite changes in ambient temperature

    • Rectal, bladder, oesophageal, central vascular temperatures are often used as approximations.

  • Peripheral temperatures are variable and generally less than the core temperature

  • Significant hypothermia (e.g. <35°C) or hyperthermia (e.g. >41°C) can lead to multi-organ dysfunction

  • Humans have multiple thermoregulatory mechanisms to resist change in core body temperature

  • In general, heat is lost by 4 mechanism: conduction, convection, evaporation, radiation

  • In general, heat is gained by 5 mechanisms: conduction, convection, evaporation, radiation, metabolism

Thermoregulatory system & regulation

  • Sensor
    • Peripheral: Skin thermoreceptors (cold= bulbs of Krause; warm=bulbs of Ruffini)
      • Travels via spinothalamic tract to hypothalamus
    • Central: Hypothalamic thermoreceptors
  • Integrator/controller
    • Hypothalamus
      • Functions as the thermostat
      • Stimulation of anterior hypothalmus leads to heat loss
      • Stimulation of the posterior hypothalamus leads to heat conservation/generation
  • Effector/Response
    • Response to cold
      • Shivering -> involuntary muscle contractions that generate heat (ATP hydrolysis)
      • Peripheral vasoconstriction (ANS) --> decreased cutaneous blood flow --> decreased heat transfer from ambient air
      • Increase metabolic rate, thyroid hormone secretion, Non shivering thermogenesis (paeds) -> increased heat generation
      • Behavioural changes (seek warmth)
      • Piloerection (unimportant in humans)
    • Response to heat
      • Peripheral vasodilation (ANS) --> increased cutaneous blood flow > increased heat loss
      • Sweating --> evaporative heat loss
      • Behavioural changes (seek cool)


Examiner comments

52% of candidates passed this question.

The best answers were systematic, using a sensor, integrator, effector approach, while also mentioning physiological variations i.e. diurnal, with ovulatory cycle etc.
Few candidates raised the concept of central and peripheral compartments. The differentiation of the concepts of set point, interthreshold range and thermoneutral zone was often confused.


Online resources for this question


Similar questions

  • Question 6, 2007 (1st sitting)
  • Question 4, 2009 (1st sitting)
  • Question 9, 2021 (1st sitting)



Question 20

Question

Outline the structure (20% of marks) and function (80% of marks) of the hypothalamus.


Example answer

Overview

  • Small (4g) almond shaped structure
  • Posterior: mammillary bodies. Anterior: anterior commissure. Superior: thalamus. Inferior: pituitary
  • Can be broken up into four functional regions, with discrete nuclei with various functions


Structure and function

Region Nuclei Function
Anterior hypothalamus - Supraoptic nuclei (ADH, oxytocin)

- Paraventricular nuclei (TRH, CRH)

- PSNS activity (increased)

- Thermoregulation (leads to heat loss)
- Water balance (ADH production / release)
- Sleep/wake cycle (promotes sleep)

Medial hypothalamus - Ventromedial nuclei

- Dorsomedial nuclei

- Sexual function (release of GnRH)

- Energy balance (BGL)
- Satiety centre (inhibits appetite)

Lateral hypothalamus - Tuberal nuclei

- Forebrain bundle

- Behaviour/emotions (inc. punishment/reward)

- Regulation of body water (thirst centre)
- Regulation of hunger (increased)

Posterior hypothalamus - Mammillary nuclei - SNS activity (increased HR, BP, constriction)

- Vasomotor control
- Thermoregulation (heat gain)
- Sleep wake cycle (wakefulness)


Regulation of pituitary function

  • Hypothalamus exerts control of pitutiary gland via two mechanisms
    • Anterior lobe of pituitary
      • Controlled by secretion of hypothalamic hormones along the portal vein
        • TRH > TSH release
        • CRH > ACTH release
        • GHRH > GH release
        • GnRH > TSH/FSH release
        • PRH > prolactin
    • Posterior lobe pituitary
      • Controlled by direct neural connections from the anterior hypothalamus > pituitary
      • Pituitary hormones (ADH, oxytocin)


Examiner comments

21% of candidates passed this question.

Most candidates understood the endocrine functions of the hypothalamus, and to some degree its interactions with the pituitary. Fewer candidates mentioned the importance of the hypothalamus as an integrator for the autonomic nervous system, or its roles in arousal/emotions.
Many candidates had only a vague idea of the structure of the hypothalamus, while the best candidates were able to relate function to structure quite accurately.


Online resources for this question


Similar questions

  • ? None




2017 (2nd sitting)

Question 1

Question

Compare and contrast the pharmacology of ibuprofen and paracetamol.


Example answer

Examiner comments

65% of candidates passed this question.

This was a standard compare and contrast question of common analgesic pharmacology and it
was generally well answered. The use of a table ensured all areas were covered including
class, indications, pharmaceutics, mode of action, pharmacodynamics, pharmacokinetics and
adverse effects. The uncertain nature (and possibilities) of the mechanism of action of
paracetamol was alluded to in better responses.

Details of the comparative pharmacokinetics were often lacking. Answers should have included
a comment on first-pass effect, the significance of the difference in protein binding and the
details of metabolism, particularly paracetamol. Metabolism limited to "hepatic metabolism and
renal excretion” gained no marks as better responses were more detailed and clearer about the
differences between the two drugs. Knowledge of metabolism at therapeutic doses and the
effect of overdose were expected. Better answers included potential interactions with other
drugs (e.g. warfarin) and contraindications to the use of these drugs.


Online resources for this question


Similar questions

  • ? None



Question 2

Question

Outline the daily nutritional requirements, including electrolytes, for a normal 70 kg adult.


Example answer

Examiner comments

21% of candidates passed this question

The provision of nutrition is a core skill in ICU. An understanding of its key elements enables prescription and modification. However, most answers lacked detailed information which is available in the standard texts. Better responses outlined the caloric requirements including each major element (water, carbohydrate, fat and protein) along with the caloric values and potential sources. Essential amino acids, fatty acids, fat and water-soluble vitamins were expected. A list of the requirements for major electrolytes and some of the trace elements were expected. Some candidates seemed to confuse calories, kilocalories and kilojoules.

Some answers did not provide the nutritional requirements, as asked, but instead discussed the
fate of the nutrients; hence did not score marks. Candidates are reminded to read the question
carefully.


Online resources for this question


Similar questions

  • Question 9, 2020 (2nd sitting)




Question 3

Question

Describe the factors that determine the filtered load of a substance at the renal glomerulus.


Example answer

Examiner comments

67% of candidates passed this question

A good place to start was with the correct equation for a filtered load and a description of the
components. Better answers described the components and how they differ and change over
the glomerulus. Many candidates usefully based answers around the Starling forces.

A summary of factors including the role of plasma concentration, protein binding, molecular size
and charge was required to pass. Many answers gave examples for the effects of size and
charge and relate endocrine responses to specific alterations in arteriolar tone and how this
affected filtration. A detailed discussion of cardiovascular and endocrine responses to
hypovolaemia was not required.

Some candidates confused clearance with filtered load. Candidates are reminded to write
legibly - especially where subscripts and Greek letters are used. Directional arrows (if used)
should correlate with text.


Online resources for this question


Similar questions

  • Question 13, 2009 (1st sitting)
  • Question 4, 2016 (1st sitting)



Question 4

Question

Describe how interstitial fluid recirculates to the vascular system.


Example answer

Examiner comments

10% of candidates passed this question

Candidates had a limited understanding of this area of the syllabus. It was expected that answers would describe important concepts including the anatomy of venous structures, valves and lymphatics, permeability and factors which influence permeability. A description of hydrostatic forces, other pressures involved, and the role of osmotic and electric forces were required.


Online resources for this question


Similar questions

  • ? None




Question 5

Question

Compare and contrast unfractionated heparin with low molecular weight heparin.


Example answer

Name HMWH (heparin) LMWH (enoxaparin)
Class Anticoagulant Anticoagulant
Indications Prophylactic and therapeutic anticoagulation (e.g. AF, DVT, PE, ACS etc) Prophylactic and therapeutic anticoagulation (e.g. AF, DVT, PE, ACS etc)
Pharmaceutics MW = 5,000-25,000 Da Clear solution for injection MW = 5,000 Daltons Clear solution for injection
Routes of administration IV, SC SC (main), can also be given IV
Dose Prophylactic: 5,000 IU BD-TDS Therapeutic: infusion (APTT target) Therapeutic: 1mg/kg BD or 1.5mg/kg OD Prophylactic: 20-40mg OD
pKA
Pharmacodynamics
MOA Heparin binds to antithrombin 3 > conformational change > increases affinity for inactivating thrombin (factor IIa) and Factor Xa Enoxaparin binds to AT-3 > conformational change > increases affinity for inactivating factor Xa (and weakly factor IIa - 4x less activity)
Effects Anticoagulation Anticoagulation
Side effects HAEM: increased risk of haemorrhage, bruising, HITTS (higher than LMWH) HAEM: increased risk of haemorrhage, bruising, HITTS (lower than HMWH)
Pharmacokinetics
Onset Immediate (IV), 30 mins (SC) Peak effect 3-4 hrs post SC injection
Absorption PO bioavailability - 0%

Variable SC absorption - less predictable response

PO bioavailability - 0%
>90% bioavailability post SC injection
Distribution VOD = 0.1L/kg Lipid solubility: low Protein binding: high Does not cross BBB / placenta VOD = 4.3L

Protein binding: does not bind to heparin binding proteins

Metabolism Reticuloendothelial system Minimal hepatic metabolism
Elimination Renal elimination (very minimal) - hence preferred in renal failure
T 1/2 = 1 hrs
Renal elimination of active and inactive metabolites
T 1/2 = 6-12 hours
Special points Reversal: protamine (1mg = 100IU) - 100% Monitoring: APTT level Reversal: protamine (<75% efficacy) Monitoring: Anti-Xa level


Examiner comments

68% of candidates passed this question.

This question was generally well answered and lent itself well to a tabular format. Expected information included an approximation of the molecular weights / significance of the differences in size and therefore its mechanism of action. Other pertinent areas to mention included pharmacokinetic differences and its use in renal failure, side effect profiles, monitoring, predictability of response and reversibility for the two agents.


Online resources for this question


Similar questions

  • Question 20, 2018 (2nd sitting)
  • Question 2, 2008 (1st sitting)
  • Question 20, 2009 (2nd sitting)



Question 6

Question

Describe the effects of Ventilation/Perfusion (V/Q) inequality on the partial pressure of oxygen (PaO2) in arterial blood.


Example answer

Examiner comments

48% of candidates passed this question

Overall answers lacked sufficient detail on a core area of respiratory physiology. Answers expected included a description of V/Q ratios throughout the lungs and an explanation of how V/Q inequality lowers PaO2.


Online resources for this question


Similar questions

  • Question 6, 2008 (1st sitting)
  • Question 5, 2014 (2nd sitting)



Question 7

Question

Compare and contrast the sympathetic and parasympathetic nervous systems.


Example answer

Examiner comments

75% of candidates passed this question

This question was generally well answered A table or diagram lent structure to the answer. More complete answers included details on the function, anatomy, a description of the pre- and post-ganglionic fibres, ganglia, receptors and neurotransmitters involved.

Whilst most commented on ‘fight or flight’ for the SNS and ‘rest and digest’ for the PNS, no candidate observed that the SNS is a diffuse physiological accelerator and that the PNS acts as a local brake. No candidate included the fact that the SNS supplies viscera and skin whilst the PNS only supplies the viscera. Many candidates failed to make reference to the fact that the postganglionic SNS receptor is G protein coupled and the PNS postganglionic receptor is Gcoupled on muscarinic receptors but operates an ion channel when nicotinic.

Candidates may have scored higher if they had provided a little more detail in their answers.


Online resources for this question


Similar questions

  • ? None



Question 8

Question

Classify calcium channel antagonists and give one example of each class (30% of marks). Describe the pharmacology of Nimodipine including important drug interactions (70% of marks).


Example answer

Examiner comments

19% of candidates passed this question

The classification was done well. Most candidates demonstrated that they had a structure for a “drug” question, but were often challenged to fill in the detail of that structure and failed to deliver enough content to secure a pass. Many candidates wrote a generic answer for calcium channel blockers instead of the specifics of nimodipine.

Frequently the pharmacokinetic data recounted was incorrect. Candidates failed to distinguish between absorption and bioavailability. The difference between oral and intravenous dosing was often omitted. Few answered the section on important drug interactions.


Online resources for this question


Similar questions

  • Question 17, 2011 (2nd sitting)
  • Question 2, 2014 (1st sitting)




Question 9

Question

Briefly outline the formation, absorption, distribution, role and composition of cerebrospinal fluid


Example answer

Examiner comments

44% of candidates passed this question

The question spelt out very specific areas of CSF physiology to outline and the marks were evenly distributed among these areas. The candidates who did not pass this question usually did not provide enough detailed information. Details of the production and absorption of CSF were commonly lacking. The majority of candidates correctly described the composition of CSF; indicating whether a particular variable was higher or lower than in plasma, scored less marks than more specific information.


Online resources for this question


Similar questions

  • Question 22, 2007 (1st sitting)
  • Question 6, 2008 (2nd sitting)
  • Question 2, 2013 (1st sitting)
  • Question 16, 2015 (1st sitting)
  • Question 24, 2017 (1st sitting)
  • Question 15, 2018 (2nd sitting)
  • Question 11, 2019 (1st sitting)
  • Question 166, 2020 (1st sitting)



Question 10

Question

Compare and contrast two methods of measuring cardiac output.


Example answer

Examiner comments

35% of candidates passed this question

Good answers began with a definition of cardiac output. For each method, it was expected that
candidates discuss the theoretical basis, equipment, advantages and disadvantages / sources
of error and limitations. Additional marks were awarded when an attempt was made to compare
and contrast the two methods (often helped by the use of a table).


Online resources for this question


Similar questions

  • ? None




Question 11

Question

Describe the pharmacology of propofol


Example answer

Examiner comments

76% of candidates passed this question.

A structured approach proved a good basis to answer this question. It was expected candidates
would outline the uses such as anaesthesia, more prolonged sedation or possible additional roles in patients with seizures or head injuries. Discussion of the presentation and pharmaceutics, including a comment on antibacterial preservatives or lack thereof was expected. The mechanism of action should have been described. It was expected candidates could provide an indication of the usual dose (and how it differs in the more unwell / elderly patient population). A maximal rate and possible toxicity was expected.

A discussion on the pharmacodynamics by major organ systems was expected and credit was given for additional comments about hyperlipidaemia, urine colour changes or metabolic alterations. It was expected that candidates would mention propofol infusion syndrome at some point in their answer with some mention of clinical features or pathophysiology.

The important aspects of its pharmacokinetics should have been mentioned (high protein binding, large volume of distribution, termination of effect by redistribution, hepatic metabolism, context sensitive half life). A mention of adverse effects would complete the answer.


Online resources for this question


Similar questions

  • Question 14, 2015 (2nd sitting)
  • Question 21, 2013 (2nd sitting)
  • Question 5, 2012 (2nd sitting)
  • Question 10, 2008 (2nd sitting)



Question 12

Question

Compare and contrast aspirin and clopidogrel


Example answer

Examiner comments

68% of candidates passed this question

Both of these commonly used agents are level A in the syllabus and thus a high level of detail was expected. Marks were awarded in the following areas - pharmaceutics, mechanism of action, pharmacokinetics (PK) and side effects. For the PK parameters a general description rather than exact values was sufficient (i.e. ‘high protein binding’ rather than ‘98% protein bound’). It was expected that candidates would mention the fact that clopidogrel is a pro-drug and the factors which influence its conversion to the active form. Additional marks were awarded for well-structured answers which attempted a comparison between the two drugs (helped by the use of a table).


Online resources for this question


Similar questions

  • Question 3, 2013 (2nd sitting)
  • Question 20, 2015 (2nd sitting)



Question 13

Question

Compare and contrast the pharmacology of intravenous fentanyl and morphine


Example answer

Examiner comments

68% of candidates passed this question

Good candidates produced a well-structured answer and highlighted the differences between the two drugs. It was important to include the dose, potency, time course of effect of both agents, and differences in pharmacokinetic and pharmacodynamic effects. Candidates should have specific knowledge of these important drugs. Many candidates failed to focus the question on intravenous fentanyl and intravenous morphine as asked. No marks were given for information about other routes of administration.


Online resources for this question


Similar questions

  • Question 12, 2016 (2nd sitting)
  • Question 16, 2011 (1st sitting)



Question 14

Question

Explain the mechanisms responsible for the cell resting membrane potential (60% of marks) and describe the Gibbs Donnan effect (40% of marks)


Example answer

Examiner comments

35% of candidates passed this question.

A good answer included a definition of the resting membrane potential and a clear description of
the factors that determine it. Explanation of these factors should have included a detailed description of the selective permeability of the membrane, electrochemical gradients and active transport mechanisms. Answers should demonstrate awareness of the Nernst equation and the Goldman-Hodgkin-Katz equation. These were often confused, sometimes with the GibbsDonnan effect. Descriptions of the Gibbs-Donnan effect generally lacked detail and understanding. The better answers included a definition and discussed in detail the influence of non-diffusible ions (intracellular proteins) on the distribution of diffusible ions


Online resources for this question


Similar questions

  • ? none



Question 15

Question

List the properties of an ideal inotrope (50% of marks). How does adrenaline compare to these ideal properties (50% of marks)?


Example answer

Examiner comments

98% of candidates passed this question

Many candidates scored very highly on this core topic. It was expected information be included
on pharmaceutics, cost, availability and compatibilities. Relevant pharmacokinetics (onset/offset, titratability) and pharmacodynamics (including relevant receptors, nuances of haemodynamic effects e.g. effect on diastolic pressure and regional perfusion) should have been detailed. Adverse effects and safety profile (e.g. use in pregnancy, therapeutic index) should also have been included. Good answers were structured and highlighted differences with specific facts and data


Online resources for this question


Similar questions

  • Question 18, 2012 (2nd sitting)



Question 16

Question

Classify and describe adverse drug reactions with examples of each.


Example answer

Adverse drug reaction

  • Noxious or unintended effect associated with drug administration at a normal dose
    • This is different from an adverse drug event which is the occurrence of any untoward event following administration of a drug (thus all ADRs are ADEs)
  • Risk factors: extremes of age, polypharmacy, genetic variability (e.g. CYP enzymes), concurrent illness (renal, liver, cardiac impairment), pregnancy


Classification of adverse drug reactions

Reaction type Mechanism Features Example/s Management
Type A

'Augmented'

Related to the pharmacological action of the drug (dose related) - Common

- Predictable
- Low mortality

- Bleeding related to administration of anticoagulants (e.g. heparin) Reduce or withhold
Type B

'Bizarre'

Non-dose related (any exposure > reaction) - Rare

- Unpredictable
- Not related to action of drug
- High mortality

Anaphylaxis to penicillin's Withhold and avoid future use
Type C

'Chronic'

Due to the cumulative dose received (dose and time related) - Uncommon Adrenal suppression with prolonged course of corticosteroids. Reduce or withhold (may need to happen over time)
Type D

'Delayed'

Does not appear for a prolonged period after exposure (time related) - Uncommon

- Usually also dose related

Tardive dyskinesia from long term use of typical antipsychotics Can be intractable
Type E

'End of treatment'

Withdrawal reactions from drug cessation - Uncommon

- Fast onset

Seizures from abrupt withdrawal of benzodiazepines or alcohol Reintroduce + withdraw slowly
Type F

'Failure'

Unexpected failure or decrease in efficacy - Common

- Dose related

Ineffectiveness of clopidogrel (non metabolisers) Increase dosage / alternative therapy


Examiner comments

44% of candidates passed this question.

Candidates should have provided a definition of adverse drug reactions and then a classification. There are at least two widely accepted systems for classification, either was acceptable; though candidates often switched between both which led to a less structured answer. The WHO classification is comprehensive and logical, though both Rang and Dale and Goodman and Gilman also have sections on this topic. Common errors were the citing of examples with the incorrect mechanism, describing only drug interactions rather than all adverse reactions and focussing the answer on the 4 hypersensitivity reactions which could only score a low mark. Some candidates confused drug errors with adverse reactions


Online resources for this question


Similar questions

  • Question 14, 2012 (2nd sitting)



Question 17

Question

Define and explain damping, resonance, critical damping and optimum damping.


Example answer

Examiner comments

25% of candidates passed this question

Concise definitions were required with a clear explanation of the underlying physical principles.
The response time of the system, degree of overshoot, effect on amplitude, noise and ability to
faithfully reproduce frequencies relative to the natural resonant frequency were important considerations.
Many candidates interpreted the question as relating to arterial lines and a detailed discussion
of the components and characteristics of an intra-arterial catheter and transducer system did not
attract marks.


Online resources for this question


Similar questions

  • Question 12, 2020 (1st sitting)
  • Question 24, 2012 (2st sitting)
  • Question 12, 2015 (1st sitting)



Question 18

Question

Draw and numerically label, on a spirogram, the lung volumes and capacities of a 30 kg child.


Example answer

Examiner comments

87% of candidates passed this question.

This core respiratory physiology topic was well answered by most candidates. Candidates generally were able to draw a spirogram. A common omission was inspiratory capacity.


Online resources for this question


Similar questions

  • ? none



Question 19

Question

Describe the physiology of a vasovagal syncope


Example answer

Examiner comments

41% of candidates passed this question

Generally, there was a lack of knowledge about this topic with many candidates confusing vasovagal syncope with a Valsalva or orthostatic hypotension. A “vasovagal” is from excessive autonomic reflex activity in contrast to orthostatic hypotension which is a failure of the autonomic reflex response.
A good place to start was with a description of vasovagal syncope, also known as neurocardiogenic syncope. It is benign, self-limiting and caused by an abnormal or exaggerated autonomic response to various stimuli (which should have been listed). The mechanism should have been described including the various receptors involved


Online resources for this question

Similar questions

  • ? None



Question 20

Question

Outline the functions of the liver


Example answer

Examiner comments

86% of candidates passed this question

This is a very straightforward question testing breadth of knowledge rather than depth. It was
well answered by the majority of candidates.


Online resources for this question


Similar questions

  • Question 22, 2017 (1st sitting)

  • Question 4, 2013 (2nd sitting)

  • Question 12, 2011 (2nd sitting)


Question 21

Question

Describe and compare the action potentials from cardiac ventricular muscle and the sinoatrial node.


Example answer

Examiner comments

95% of candidates passed this question

This topic was well understood and answered by most candidates. Some candidates had a good knowledge base but missed out on potential marks by failing to compare and contrast. A diagram outlining the various phases was a useful way to approach the question.


Online resources for this question


Similar questions

  • Question 1, 2020 (2nd sitting)
  • Question 23, 2010 (2nd sitting)
  • Question 19, 2013 (1st sitting)
  • Question 11, 2016 (2nd sitting)



Question 22

Question

Define bioavailability. Outline the factors which affect it.


Example answer

Examiner comments

33% of candidates passed this question

Many candidates did not specify that bioavailability describes the proportion/fraction of drug
reaching the systemic circulation (to differentiate from the portal circulation). Some candidates considered only factors impacting absorption from the GI tract or stated that bioavailability related only to orally administered drugs. Candidates failed to provide an equation, or got equations or graphs wrong. Nearly all candidates failed to provide a comprehensive list of factors affecting bioavailability.


Online resources for this question


Similar questions

  • Question 15, 2020 (1st sitting)



Question 23

Question

Outline the anatomy of the internal jugular vein relevant to central venous line cannulation (80% of marks). Include important anatomical variations (20% of marks).


Example answer

Internal jugular vein

  • Originates at the jugular bulb (confluence of the inferior petrosal and sigmoid sinus')
  • Exits skull via the jugular foramen with CN IX, X, XI
  • Descends inferolaterally in the carotid sheath (initially posterior > lateral to carotid artery with descent)
  • Terminates behind the sternal end of the clavicle where it joins with the subclavian vein to form the brachiocephalic vein
  • Tributaries: facial, thyroid, pharyngeal, lingual veins
  • Usually larger on the right


Relations

  • Anterior to IJV: SCM, lymph nodes, CN XI
  • Posterior to IJV: scalene muscles, lung pleura, lateral mass C1, vagus (poster-medial)
  • Inferior/at termination: pleura (which extends ~2cm above clavicle)
  • Medial: vagus, carotid artery


Variations

  • Stenosis, complete occlusion, aneurysms, absence
  • Variation in relation to carotid (e.g. anterior) in up to 1/4 cases


Ultrasound anatomy

  • Often lateral to carotid (not always) and often larger than carotid
  • Unlike carotid: Non pulsatile, thin walled, compressible
  • Doppler flows can also be helpful.


Surface anatomy

  • Identify triangle between the clavicle and two heads of SCM
  • Palpate carotid
  • Puncture lateral to carotid artery at 30 degree angle
  • Aim caudally towards ipsilateral nipple


Examiner comments

14% of candidates passed this question.

Good answers were structured including origin, termination, tributaries, relationships, surface
anatomy and common variations.
Factual inaccuracies were common and there was confusion about the relations of the internal
jugular vein. Many candidates did not mention the changing relationship between the internal
jugular and the carotid artery as they travel through the neck or the changes that result from
repositioning for insertion. Many candidates also forgot to mention surface anatomy and a
number talked about ultrasound and views used for insertion of central lines. Common
omissions included the origin, tributaries, relationship with the correct cranial nerves and the fact
that it is usually larger on the right. Almost nobody mentioned the relationship to the pleura.


Online resources for this question


Similar questions

  • Question 8, 2021 (2nd sitting)
  • Question 18, 2014 (1st sitting)



Question 24

Question

What is functional residual capacity (30% of marks)? Describe two methods of measuring functional residual capacity (70% of marks).


Example answer

Examiner comments

59% of candidates passed this question

Most candidates could state 2 methods of measuring FRC. Some candidates (especially for
nitrogen wash out) failed to provide enough information e.g. statements such as "if the amount
of nitrogen is measured then FRC can be derived" were insufficient to score many marks.


Online resources for this question


Similar questions

  • Question 2, 2020 (2nd sitting)
  • Question 15, 2010 (2nd sitting)
  • Question 4, 2015 (2nd sitting)
  • Question 8, 2017 (1st sitting)
  • Question 24, 2017 (2nd sitting)



2017 (1st sitting)

Question 1

Question

Outline the anatomy and physiology of the parasympathetic nervous system.


Example answer

PSNS

  • Division of the autonomic nervous system
  • Important for physiological regulation of our organ systems
  • Broadly speaking, there are pre + post ganglionic neurons
    • Preganglionic neurons
      • CN 3,7,9,10, as well as S2-4 (craniosacral outflow)
      • Long and synapse close to the effector organ
      • Neurotransmitter is ACh > nicotinic receptor
    • Postganglionic neurons
      • Short
      • Neurotransmitter is ACh > muscarinic receptor


Anatomy + effects (based on "target" organ system)

Target Organ Pre- Ganglionic fibre origin Pre- Ganglionic nerve Ganglion Post- Ganglionic Receptor Effect
Heart Vagal nucleus in Medulla CN X Cardiac plexus ganglia M2 Decreased inotropy and chronotropy
Lung Vagal nucleus in Medulla CN X Pulmonary plexus ganglia M3 Bronchoconstriction
Pupils Oculomotor nucleus CN III Ciliary ganglion M3 Constriction
Salivary glands Superior and inferior salivary nuclei CN VII (mandibular, maxillary)

CN IX (parotid)

-Submaxillary ganglion

- Otic ganglion

M3 Salivation
GIT Vagal nucleus

Spinal cord

CN X

S2,3,4 nerves

Gastric and hypogastric plexus M3 Increased peristalsis
Bladder, Penis Spinal cord S2,3,4 nerves Hypogastric plexus M3 Contraction of bladder, erection
Adrenal gland - - - - No effect
Arterioles - - - - No effect
Sweat gland - - - - No effect


Examiner comments

32% of candidates passed this question

An efficient way to answer this question was to describe the anatomy and physiology of both cranial and sacral sections together. High scoring answers included an outline of the relevant nerves, the various ganglia, neurotransmitters and physiological effects. Some candidates described the cellular basis of Nicotinic, Muscarinic and M1-M5 receptors which didn't attract marks.


Online resources for this question


Similar questions

  • Question 4, 2014 (2nd sitting)
  • Question 7, 2017 (2nd sitting)



Question 2

Question

Outline the components of dietary fat (20% of marks). Describe their possible metabolic fates (80% of marks).


Example answer

Examiner comments

21% of candidates passed this question

Almost all candidates interpreted "metabolic fate" to mean absorption, digestion and transport of
fat. Hence a lot of time was spent on this and little on the fate of fat once it enters the blood stream. The processes of neither beta oxidation, nor lipogenesis were not well understood. Ketone body production was better understood.


Online resources for this question


Similar questions

  • Question 9, 2014 (2nd sitting)



Question 3

Question

Classify and describe the mechanisms of drug interactions with examples


Example answer

Classification of drug-drug interactions Example
BEHAVIOURAL
- One drug alters behaviour of patient for another - A depressed patient taking an antidepressant may be more compliant with other medications for unrelated conditions
PHARMACEUTIC
- Formulation of one drug is altered by another before administration - Precipitation of thiopentone (basic) and vecuronium (acidic) in a giving set
PHARMACOKINETIC
Absorption Bioavailability of bisphosphonates is reduced when co-administered with calcium as the drugs interact to form insoluble complexes
Distribution Valproate and phenytoin compete for the same transport protein binding sites > decreased protein binding phenytoin > increased effect
Metabolism Macrolides reduce metabolism of warfarin by outcompeting it for similar metabolic pathways (CYP450 enzymes) > increased duration of effect
Elimination Probenecid decreases the active secretion of B-lactams and cephalosporins in renal tubular cells by competing for transport mechanisms > decreased elimination of B-lactams / cephalosporins
PHARMACODYNAMIC
Homodynamic effects Drugs bind to the same receptor site (e.g. naloxone reverses the effects of opioids by outcompeting for the opioid receptor sites)
Allosteric modulation Drugs bind to the same receptor (GABA) but at different sites (e.g. barbiturates and benzodiazepines) > increased effect
Heterodynamic modulation drugs bind to different receptors but affect the same second messenger system (e.g. glucagon reverses the effects of B-blockers by activating cAMP)
Drugs with opposing physiological actions (but different biological mechanisms) e.g. GTN vasodilates via guanyl cyclase-cGMP mediated vasodilation, while noradrenaline vasoconstricts via <math display="inline">\alpha</math> agonism


Examiner comments

44% of candidates passed this question

Candidates with a well organised answer scored highly. A list of drug interactions was not sufficient to pass, as the question asked to 'describe' the mechanism of drug interactions. Some candidates described the interaction but did not give examples. Common mistakes included using incorrect examples for a particular mechanism and describing the mechanism of action of drugs instead of drug interactions


Online resources for this question


Similar questions

  • Question 15, 2021 (2nd sitting)
  • Question 9, 2015 (1st sitting)



Question 4

Question

Describe the endocrine functions of the kidney.


Example answer

The kidneys are involved in hormone production, modification and clearance


Production

  • Erythropoietin (EPO)
    • Production:
      • 90% produced in kidneys (~10% in liver) from fibroblast like interstitial cells
    • Function:
      • Stimulates the development of proerythroblasts from haematopoietic stem cells in the bone marrow and increases the speed of their maturation
    • Regulation:
      • Released in response to hypoxia, low HCT and hypotension.
      • Decreased in renal failure, increased HCT, inflammation
  • Renin
    • Production:
      • Produced, stored, secreted from JG cells in kidney
    • Function:
      • Activation of the renin-angiotensin-aldosterone system leading to increased sodium and water reabsorption, increased vasoconstriction and blood pressure
    • Regulation:
      • Stimulated by reduced GFR, direct B1 SNS activation, decreased Na/Cl delivery to JGA
      • Inhibited by negative feedback
  • Thrombopoietin
    • Production
      • Predominately liver, small amount in kidneys (PCT)
    • Function
      • Stimulate megakaryocytes to produce platelets
    • Regulation
      • Stimulated by thrombocytopaenia and inflammatory cytokines
      • Inhibited by negative feedback loop
  • Urodilatin
    • A natriuretic peptide secreted by DCT in response to increased Na delivery


Modification

  • Vitamin D
    • 25-hydroxy vitamin D3 converted into calcitriol in the PCT
    • Leads to increased Ca absorption from GIT, increased liberation of Ca from bone, increased reabsorption of Ca from DCT in kidney
    • Stimulated by hypocalcaemia, low vitamin D, high parathyroid hormone levels
    • Inhibited by low PTH, hypercalcaemia, high calcitriol


Clearance

  • Gastrin
    • 90% cleared in the kidney in the PCT
  • Insulin
    • 30% cleared by the kidney in the PCT


Examiner comments

39% of candidates passed this question

It was expected that candidates would discuss the major hormones produced (or activated) by the kidney. These included erythropoeitin, renin and calcitriol. Good answers included the following: the area where the hormone is produced or modified; stimuli for release; factors which inhibit release; and the subsequent actions / effects. Marks were not awarded for hormones that act on the kidney


Online resources for this question


Similar questions

  • ? None



Question 5

Question

Describe the regulation of plasma calcium concentration.


Example answer

Calcium distribution

  • Total body calcium ~400mmol/kg
  • Over 99% is locked up in bone/teeth (hydroxyapatite, phosphates etc) and not readily redistributable
  • Less than 1% of body calcium is located in the ECF (~2.4mmol/L)
    • Approximately 45% is unbound (ionised calcium ~1.1mmol/L) and therefore in active form
  • There is almost no calcium in the intracellular fluid


Calcium homeostatis

  • Maintained as a balance between
    • Intake:
      • Dietary intake
      • GIT absorption (passive during normocalcaemia, active during hypocalcaemia)
    • Exchange:
      • With bone (balance between osteoclast and osteoblast activity)
    • Loss:
      • Faecal (80% of daily losses), renal (20%)
  • Principally regulated by three hormones


Calcium regulation

Parathyroid hormone Calcitriol (active VitD) Calcitonin
Production - Secreted by parathyroid gland - Metabolic product of vitamin D - Secreted from parafollicular cells (thyroid)
Stimulating factors - Hypocalcaemia

- Hypophosphatemia

- Hypocalcaemia

- Hypophosphatemia
- PTH

- Hypercalcaemia

- Gastrin

Inhibitory factors - Calcitriol

- Hypermagnesemia
- Hypercalca

- Hypercalcaemia

- Dec. sun exposure
- Dec. renal function

- Hypocalcaemia

- Somatostatin

Effect on Calcium Increases calcium Increases calcium Decreases calcium
Mechanism of effect on Ca - Increased renal reabsorption (DCT)

- Increased osteoclast activity
- Increased production of calcitriol

- Increased GIT absorption (ileum)

- Increased renal reabsorption (DCT)
- Increased osteoclast activity

- Decreased renal reabsorption (DCT)

- Inhibition of osteoclast activity


Examiner comments

51% of candidates passed this question

High scoring answers discussed the three major hormones involved in calcium regulation - parathyroid hormone, vitamin D and calcitonin. For each of these it was expected that candidates include: site of production, stimulus for release, inhibitory factors and actions. In the case of renin it was expected that candidates also include the actions of angiotensin and aldosterone. Very few answers discussed inhibitory factors or negative feedback loops.


Online resources for this question


Similar questions

  • Question 1, 2016 (2nd sitting)
  • Question 7, 2008 (1st sitting)



Question 6

Question

Explain the meaning of the components of a Forest plot.


Example answer

Removed from primary syllabus


Examiner comments

65% of candidates passed this question

To score full marks candidates needed to describe each feature of the forest plot provided. This included: odds ratio on the x axis; line of no effect; individual studies on the y axis; point estimate for each study (box position); weighting of the study (box size); pooled effect estimate (diamond position); size of the diamond; and the 95% confidence intervals and their interpretation.


Online resources for this question

  • Removed from primary syllabus


Similar questions

  • Removed from primary syllabus




Question 7

Question

Compare and contrast the systemic circulation with the pulmonary circulation


Example answer

Category Pulmonary circulation Systemic circulation
Anatomical features Thin vessel, minimal smooth muscle, elastic Thick vessel, abundant smooth muscle
Blood volume ~500mls (70kg adult) or 10% total volume ~4.5L (70kg adult) or 90% volume
Blood flow = cardiac output (~5L/min) = cardiac output (~5L/min)
Blood pressure PAP normally ~25/8mmhg (mPAP ~10-15mmHg) BP normally ~120/80mmHg (MAP ~90mmHg)
Circulatory resistance PVR ~ 100 dynes.sec.cm-5 ~10% of SVR SVR approx 1000 dynes.sec.cm-5
Circulatory regulation Minimal capacity to self regulate (except hypoxic pulmonary vasoconstriction) Regional blood flow readily regulated at the level of arterioles
Regional distribution of blood flow Flow affected by gravity, alveolar recruitment, hypoxic vasoconstriction Significant organ dependant variation in flow (often demand dependant) with minimal affect from gravity. Organs have capacity to autoregulate flow.
Response to hypoxia Vasoconstriction Vasodilation
Response to hypercapnia Vasoconstriction Vasodilation
Gas exchange function Absorbs O2, releases CO2 Absorbs CO2, releases O2
Metabolic function Metabolism of PGs and substrates for ACE Delivers metabolic substrates, removes metabolic waste
Synthetic function Source of thromboplastin and heparin Source of nitric oxide and anticoagulants/procoagulants
Filter function Filters emboli >8um Filters arterial blood in renal and hepatic vascular beds


Examiner comments

26% of candidates passed this question

This question encompasses a wide area of cardiovascular physiology. As a compare and contrast question this question was well answered by candidates who used a table with relevant headings. Comprehensive answers included: anatomy, blood volume, blood flow, blood pressure, circulatory resistance, circulatory regulation, regional distribution of blood flow, response to hypoxia, gas exchange function, metabolic and synthetic functions, role in acid base homeostasis and filter and reservoir functions. A frequent cause for missing marks was writing about each circulation separately but comparing. For example: many candidates stated 'hypoxic pulmonary vasoconstriction', but did not contrast this to 'hypoxic vasodilation' for the systemic circulation. Frequently functions of the circulations were limited to gas transport / exchange.


Online resources for this question


Similar questions

  • ? None comparing



Question 8

Question

Describe the physiological consequences of decreasing the functional residual capacity (FRC) in an adult by 1 litre.


Example answer

Examiner comments

70% of candidates passed this question

High scoring answers began with a definition and normal values, followed by a detailed list of the consequences of decreasing the FRC. Some candidates included descriptions of the normal function of FRC, conditions that decrease FRC and ways of improving reduced FRC. These were not required and did not attract marks. Diagrams require correctly labelled axes, values & units.


Online resources for this question


Similar questions

  • Question 15, 2010 (2nd sitting)



Question 9

Question

Outline how the following tests assess coagulation:

a. Prothombin Time (PT)

b. Activated Partial hromboplastin Time (APTT)

c. Activated Clotting Time (ACT)

d. Thromboelastography (TEG or ROTEM)


Example answer

test PT APTT ACT TEG/ROTEM
Pathway Extrinsic + common Intrinsic + common Intrinsic + common Clot formation to lysis
Use Warfarin monitoring

Screening for coagulopathy

Heparin monitoring

Screening for coagulopathy

Dosing/reversal of heparin in extracorporeal circuits Guide to product replacement
POC/LAB Lab Lab POC POC
Sample Plasma (post centrifuge) Plasma (post centrifuge) Whole blood Whole blood
Principle Tissue factor added to plasma > activates extrinsic pathway > wait until clot formation Phospholipid added to plasma (+ activation agent) > stimulates intrinsic pathway > wait until clot formation Blood added to kaolin clotting activator > stimulates intrinsic pathway > wait until clot formed Blood distributed into cuvettes. Pin immersed in blood and either cuvette (TEG) or pin (ROTEM) spins. As blood clots > resists movement. TEG: toque exerted on the pin. ROTEM: impedance to rotation detected by optical system.
Normal 11-13 seconds

(INR 0.8-1.2)

30-40 seconds 100-130 INTEM: similar to APTT

EXTEM: similar to PT
CT = time until 2mm amplitude
A10 = amplitude at 10 mins
MCF = time until maximal clot firmness
ML: maximal lysis

Prolonged Warfarin / vitamin K deficiency / factor II, VII, IX, X deficiency

Liver disease
Consumptive coagulopathy

Heparin

Factor deficiency (II, IX, X, XI, XII) Liver disease
Consumptive coagulopathy

Any coagulopathy (non specific) including systemic heparinisation Hyperfibrinolysis

Factor deficiency / inhibition
Platelet deficiency / dysfunction
Fibrinogen deficiency

Errors Different thromboplastins in lab give different PT times > INR standardises Inadequate mixing of blood

Inadequate blood:citrate ratio

Underfilling shortens ACT

Overfilling, prolongs ACT

Calibration of machine


Examiner comments

61% of candidates passed this question.

Many candidates incorrectly stated that the PT assessed the intrinsic system and that the APTT assessed the extrinsic system. This led to subsequent errors in relating a coagulation test to the appropriate coagulation factors that it assessed. Some candidates produced elaborate diagrams of the coagulation cascade in isolation without relating it to the question.


Online resources for this question


Similar questions

  • Question 8, 2014 (2nd sitting)




Question 10

Question

Describe the pharmacology of hydrocortisone.


Example answer

Name Hydrocortisone
Class Glucocorticoid (endogenous)
Indications Glucocorticoid insufficiency, allergy/anaphylaxis/asthma, severe septic shock, immunosuppression (e.g. transplant, autoimmune dz)
Pharmaceutics Tablet, white powder diluted in water
Routes of administration IV, PO
Dose 50-200mg QID (commonly in ICU population)
Bio-equivalence 100mg hydrocort = 25mg pred = 20mg methypred = 4mg dex
Pharmacodynamics
MOA Lipid soluble > crosses cell membrane > binds to intracellular steroid receptors > alters gene transcription > metabolic, anti-inflammatory & immunosuppressive effects in tissue-specific manner
Effects/side effects CNS: sleep disturbance, psychosis, mood changes

CVS: Increased BP (mineralocorticoid effect + increased vascular smooth muscle receptor expression to catecholamines)
RESP: decreased airway oedema, increased SM response to catecholamines
RENAL: Na + water retention (mineralocorticoid effect)
Metabolic: Hyperglycaemia, gluconeogenesis, protein catabolism, fat lipolysis and redistribution, adrenal suppression
MSK: Osteoporosis, skin thinning
Immune: immunosuppression + anti-inflammatory effects (decreased phospholipase, interleukins, WBC migration and function)

GIT: Increased risk of peptic ulcers
Pharmacokinetics
Onset Peak effect 1-2 hours, duration of action 8-12 hours
Absorption 50% oral bioavailability, 100% IV
Distribution 90% protein bound

Small Vd (0.5L/kg)

Metabolism Hepatic > inactive metabolites
Elimination Metabolites excreted renally.

Elimination T/12 = ~1 hour

Special points Risk of reactivation of latent TB / other infections


Examiner comments

54% of candidates passed this question

Hydrocortisone is listed as a Class A drug in the syllabus and as such knowledge of its
pharmacokinetics is expected. No marks were awarded for generic pharmacokinetic statements such as: "average bioavailability", "moderate protein binding", "bioavailability 100% for IV preparation" etc


Online resources for this question


Similar questions

  • Question 3, 2020 (2nd sitting)



Question 11

Question

Outline the anatomical relations of the trachea relevant to performing a percutaneous tracheostomy.


Example answer

Structure

  • Fibromuscular tube ~10cm long, approx 2.5cm wide, ~2cm deep
  • Supported by 16-20 incomplete cartilaginous rings which joined by fibroelastic tissue and are connected posteriorly by smooth muscle (the trachealis)
  • Divided into cervical and thoracic parts


Course

  • Trachea begins approximately C6 where it is continuous with the larynx
  • Trachea travels inferoposteriorly
  • Enters thoracic cavity through the superior thoracic aperture, at the level of the jugular notch
  • Ends approximately at level of sternal angle (T4/5) where it divides into left and main bronchi


Relations

  • Posterior: oesophagus
  • Anterior: thyroid gland (isthmus), cervical fascia, manubrium, thymus remnants,
  • Right lateral: thyroid gland (lobe), carotid sheath ( common carotid, vagus, IJV)
  • Left lateral: thyroid gland (lobe), carotid sheath ( common carotid, vagus, IJV)


Neurovascular supply

  • SNS: sympathetic trunks
  • PSNS: recurrent laryngeal and vagus nerves
  • Arterial supply: Branches from inferior thyroid arteries
  • Venous drainage: Inferior thyroid veins


Surface anatomy of anterior neck (superior --> inferior)

  • Hyoid bone (C3)
  • Thyroid cartilage
  • Cricothyroid membrane
  • Cricoid cartilage (C6)
  • Thyroid gland
  • Sternohyoid muscle just lateral to the midline structures, overlies sternothyroid and thyrohyoid


Layers of dissection in tracheostomy (from anterior --> posterior)

  • Skin
  • Subcutaneous tissue
  • Fat
  • Pretracheal fascia
  • Fibroelastic tissue between tracheal cartilage rings
  • Trachea


Examiner comments

44% of candidates passed this question

Many candidates described how to perform a tracheostomy or the structure of the trachea rather than the relevant anatomical relations. It was expected that answers include anterior, posterior and lateral relations at the correct tracheal level including relevant vascular structures.


Online resources for this question


Similar questions

  • Question 1, 2018 (2nd sitting)




Question 12

Question

Describe the pharmacology of oxycodone


Example answer

Name Oxycodone
Class Semi synthetic opioid
Indications Analgesia
Pharmaceutics White tablet (IR, MR), colourless solution (10mg/ml)
Routes of administration PO/IV
Dose PO 5-10mg PRN 4hrly, IV 1mg 5 minutes PRN
Morphine equivalence 1.5 x morphine (10mg oxycodone = 15mg morphine )
Pharmacodynamics
MOA Mu receptor activity, weak Kappa/Delta activity
Effects CNS: Analgesia, sedation, euphoria

CVS: bradycardia/hypotension
RESP: respiratory depression
GIT: decreased peristalsis. N/V. constipation
MSK: pruritis

Side effects Everything listed above that is is not analgesia
Pharmacokinetics
Onset / duration 15 mins (PO), 4-6 hours (PO)
Absorption 70% oral bioavailability, pKa 8.5
Distribution ~50% protein bound,

VD = 3L/Kg,
crosses placenta and BBB

Metabolism Hepatic metabolism (CYP) to noroxycodone, oxymorphone
Elimination Half-life 2-4hrs, excreted in urine
Reversal Naloxone (100mcg IV boluses, PRN 3 minutely)


Examiner comments

53% of candidates passed this question

Few candidates covered the pharmacokinetic aspect of the question sufficiently.
No marks were awarded for generic comments such as hepatic metabolism and renal excretion


Online resources for this question


Similar questions

  • Question 12, 2021 (1st sitting)




Question 13

Question

Outline the anatomy relevant to the insertion of a Dorsalis Pedis arterial cannula (50% of marks). Explain the differences between blood pressure measurement at this site compared to measurement at the aortic arch (50% of marks)


Example answer

Examiner comments

30% of candidates passed this question

The anatomy component of answers frequently lacked required detail. Many candidates listed
the observed differences in the waveforms however an explanation for these differences was
required.


Online resources for this question


Similar questions

  • ? None



Question 14

Question

Define respiratory compliance (20% of marks). Describe the factors that affect it (80% of marks).


Example answer

Examiner comments

54% of candidates passed this question.

This question was generally well answered with good structure.


Online resources for this question


Similar questions

  • Question 17, 2019 (2nd sitting)
  • Question 15, 2014 (1st sitting)
  • Question 7, 2011 (2nd sitting)
  • Question 13, 2007 (1st sitting)



Question 15

Question

Outline the cardiovascular changes associated with morbid obesity.


Example answer

Obesity

  • Multisystem disorder defined by an increased BMI
    • Overweight: BMI 25-30
    • Obese: BMI >30
    • Morbidly obese: BMI >35


Effects of obesity on CVS

  • Oxygen demand / utilisation
    • Increased due to the increased body mass (both adipose and lean body mass)
    • Must be met by increased DO2 (in form of increased CO) to prevent ischaemia
  • Cardiac output (including HR, SV)
    • Increased to meet oxygen demand (~1L for every 12.5 BMI points)
    • Predominately due increased stroke volume as HR remains stable/slight increase
  • Stroke volume
    • Increased
    • Due to the increased preload and the frank-starling mechanism (see below)
  • Heart rate
    • Mostly stable / slight increase
    • The required increase in CO predominately comes from the increased stroke volume
  • Blood volume
    • Increased
    • Due to neurohormonal changes associated with obesity
    • Adipocytes secrete leptin > increased activation of renin-angiotensin-aldosterone system (RAAS) > increased Na and water reabsorption
  • Blood pressure
    • Often increased (>60%)
    • Due to neurohormonal changes (leptin activation of RAAS) and LV remodelling (see below)
  • Preload
    • Generally increased
    • Due to the increased blood volume which increases mean systemic filling pressure and thus venous return
  • Afterload
    • May be decreased or increased
  • Cardiac remodelling
    • LV hypertrophy (from hypertension and increased afterload and leptin)
    • Chamber dilation (from chronic volume overload) > increased
    • Fatty infiltration > increases risk of arrhythmias
    • Fibrosis > leads to diastolic dysfunction
  • Pulmonary artery pressures
    • Increased
    • Due to hypoxic pulmonary vasoconstriction (obesity hypoventilation syndrome) and LV diastolic dysfunction from cardiac remodelling


Examiner comments

42% of candidates passed this question

Many candidates did not include enough detail in their answers. Higher scoring answers included more depth such as the following: blood volume, left ventricular changes, arterial blood pressure, pulmonary artery pressures, risks of ischaemia, arrhythmias etc.


Online resources for this question


Similar questions

  • Question 17, 2008 (2nd sitting)
  • Question 8, 2015 (2nd sitting)



Question 16

Question

List the potential problems resulting from blood transfusion and methods used to minimise them


Example answer

Examiner comments

53% of candidates passed this question

This question required a broad answer. It was generally well answered. Those candidates who scored well had a good structure to their answers e.g. grouping potential electrolyte disturbances together, and infectious risks together etc. and including methods used to minimise these risks in appropriate detail.


Online resources for this question


Similar questions

  • Question 3, 2020 (1st sitting)
  • Question 13, 2013 (2nd sitting)



Question 17

Question

Compare and contrast the pharmacology of phenytoin and levetiracetam


Example answer

Name Phenytoin Levetiracetam
Class Anticonvulsant Anticonvulsant
Indications - Seizure prophylaxis

- Epilepsy (simple-complex and focal-generalised)
- Status epilepticus

- Seizure prophylaxis

- Focal partial seizures (monotherapy)
- Partial/generalised seizures (adjunct)
- Status epilepticus

Pharmaceutics Capsules, syrup, clear IV solution for injection Oral tablet or liquid. Clear solution for injection
Routes of administration PO, IV, IM PO, IV
Dose 15-20mg/kg load

Target plasma level of 10-20mcg/ml

Load = 60mg/kg (Status epilepticus)

500-2000mg BD (maintenance)

Pharmacodynamics
MOA Stabilises Na channels in their inactive state, thereby inhibiting the generation of further action potentials.

Also decreases Ca entry > increased GABA activity.

Exact MOA unclear. May modulate neurotransmission by binding to synaptic vesicle protein 2A
Effects Prevents propagation of seizure activity Prevent hypersynchronization of epileptiform burst firing and propagation of seizure activity
Side effects CVS: hypotension, heart block

GIT: Nausea, vomiting
CNS: ataxia, confusion, nystagmus, visual disturbance
Other: acne, hirsutism, blood dyscrasias, gingival hypertophy
Pregnancy = teratogenic
Many drug-drug interactions

CNS: irritability, agitation, anxiety, drowsiness, dizziness, headache, ataxia

MSK: weakness/fatigue
Other: allergy, angioedema, SJS

Pharmacokinetics
Onset Slow oral absorption (1-3 hours onset) < 1 hour (PO)
Absorption PO bioavailability = 90% Nearly 100% PO bioavailability
Distribution VOD= 1L/kg

Protein binding >90%

VOD = 0.5 L / Kg

Protein binding = < 5%

Metabolism - Hepatic hydroxylation by CYP450 system (saturable)

- Wide patient variation (10% population are slow hydroxylators)

Enzymatic hydrolysis ~30% of dose to inactive metabolites
Elimination Renal elimination of metabolites

T 1/2 = 12 hours

Renal excretion
- unchanged drug (70%) and metabolites (30%) T 1/2 = 6 hours
Monitoring Phenytoin level 10-20ug/ml Nil


Examiner comments

35% of candidates passed this question

A table was useful to answer this question. Comparing and contrasting the pharmacology was required to score well rather than listing various aspects of pharmacology. The key properties of the drugs which demonstrate their importance to ICU was required.


Online resources for this question


Similar questions

  • Phenytoin alone
    • Question 3, 2012 (1st sitting)
    • Question 2, 2010 (1st sitting)
  • Phenytoin vs Keppra
    • Question 2, 2014 (2nd sitting)
    • Question 17, 2017 (1st sitting)
  • Keppra alone
    • Nil



Question 18

Question

Outline the functional anatomy of the kidneys (40% of marks). Outline the regulation of renal blood flow (60% of marks).


Example answer

Examiner comments

67% of candidates passed this question

Candidates who scored well weighted their answers according to the marks allocation outlined in the question and adopted a good structure. A number of candidates confused the roles of tubuloglomerular feedback and the renin angiotensin aldosterone pathway.


Online resources for this question


Similar questions

  • Question 4, 2019 (1st sitting)

  • Question 21, 2011 (2nd sitting)


Question 19

Question

Define mixed venous PO2 (20% of marks). Outline the factors that affect this value (80% of marks).


Example answer

Examiner comments

37% of candidates passed this question

This question was in two parts – the first part was worth 20% and candidates were expected to
provide a definition of mixed venous blood as well as the partial pressure of oxygen in mixed
venous blood (including normal range). Good answers also provided the varying PO2 from
different tissue beds that make up mixed venous blood, where the ‘mixing’ occurs (the right
ventricle) and where it is sampled (pulmonary artery).
For the second part of the question, worth 80% of the marks, good answers included the
relationship between mixed venous PO2 and mixed venous O2 content (including the shape and
position of the HbO2 dissociation curve); the variables encompassed in the modified Fick
equation; arterial oxygen content and its determinants; oxygen consumption (VO2); and cardiac
output (CO). Including an outline of how each affects the value of mixed venous PO2.
A number of candidates wrote about mixed venous oxygen saturation. Other common errors
were: missing a number of key factors that affect PO2; and using an incorrect form and/or
content of the modified Fick equation.


Online resources for this question


Similar questions

  • Question 10, 2008 (1st sitting)
  • Question 8, 2019 (1st sitting)



Question 20

Question

Describe the pharmacology of vasopressin (70% of marks) and its analogues (30% of marks).


Example answer

Name Vasopressin (argipressin)
Class Endogenous nonapeptide
Indications Hypotension/shock (catecholamine sparing)
Pharmaceutics Clear colourless solution (20units/ml)
Routes of administration IV infusion (central vein)
Dose 2.4 units/hr (for vasopressor support)

- At lower doses has predominant V1 activity, V2 activity at higher doses

pKA 10.3
Pharmacodynamics
MOA / effects Physiologically secreted by PVN of hypothalamus > stored in posterior pituitary > secreted in response to hypovolaemia + increased osmolality

→ V1 receptor (blood vessels) agonism > Vasoconstriction > increased SVR > increased BP
→ V2 receptor (collecting ducts of nephrons) agonism > increased water reabsorption > increased BP
→ V2 receptor (endothelial cells) agonism > increased vWF release and Factor VIII activity

Side effects CVS: reflex bradycardia, splanchnic vasoconstriction (possible ischaemia)

HAEM: Excessive platelet aggregation / thrombosis
RENAL: Hyponatraemia (increased water reabsorption > Na reabsorption)
GIT: abdominal pain / GIT ischaemia

Pharmacokinetics
Onset Fast (not as fast as noradrenaline)
Absorption IV only (0% oral bioavailability as inactivated by trypsin)
Distribution No (or very minimal) protein binding

Vd 0.2L/Kg

Metabolism Extensive hepatic and renal metabolism by serine proteases and oxido-reductase enzymes > inactive metabolites
Elimination Renal elimination of changed and unchanged drug

T 1/2 15 minutes


Vasopressin analogues

  • Desmopressin (DDAVP)
    • Indications: central diabetes insipidus, vWD, slowing correction of hyponatraemia
    • Route: IV, IN, SC, PO, IM
    • MOA: predominately V2 mediated effects (limited V1 effects) > increased H2O reabsorption + increased vWF and Factor VIII activity. Minimal effects on vasoconstriction activity
    • Similar PK to argipressin except it is not metabolised, has a longer T 1/2
  • Terlipressin
    • Indication: variceal bleeding, hepatorenal syndrome
    • Route: IV
    • MOA: predominately V1 mediated effects > splanchnic vasoconstriction > decreased portal venous pressure. Minimal effects of platelet aggregation or fluid absorption
    • Similar PK to argipressin except it is not metabolised, has a longer T1/2


Examiner comments

28% of candidates passed this question

A pharmacology answer template outlining pharmacokinetics and dynamics was required. Candidates failed to score marks for describing the physiology of vasopressin secretion. A number of answers demonstrated limited knowledge about its indications for use and its potential adverse effects.


Online resources for this question


Similar questions

  • Vasopressin alone
    • Question 22, 2013 (1st sitting)
  • Norad vs vasopressin
    • Question 7, 2007 (1st sitting)
    • Question 23, 2011 (1st sitting)
    • Question 10, 2020 (1st sitting)



Question 21

Question

Explain the potential causes of a difference between the measured end tidal CO2 and the arterial partial pressure of CO2.


Example answer

ETCO2 - PaCO2 gradient

  • There is normally a gradient between PaCO2 and ETCO2 of 0-5mmHg (where ETCO2 is lower)
  • The difference between the values is due to alveolar dead space
    • Alveolar dead space is due to alveoli which are ventilated but not perfused (e.g. west zone 1 lungs)
    • These alveoli do not participate in gas exchange (there is no perfusion), thus contain very little CO2 and a lot of O2 (the same amount as in inspired air)
    • This relatively CO2 deplete gas mixes with the rest of the expired gas, diluting the ETCO2 reading, thus leading to an observed discrepancy
    • Note: It is not due to anatomical dead space as this gas has already been washed out in the early stages of exhalation and thus does not contributed to ETCO2
  • Healthy/awake patients have near zero alveolar dead space, so near zero gradient


Factors affecting ETCO2 - PaCO2 gradient

  • Changes in pulmonary perfusion
    • Global reduction in pulmonary perfusion
      • e.g. pHTN, heart failure, Cardiac arrest, Severe shock
    • Regional decreases in pulmonary perfusion
      • e.g. pulmonary embolism, fat embolism
  • Changes in ventilation
    • Excessively high PEEP --> increased West Zone 1
  • Measurement error
    • Inline HME filters
    • Timing of measurement (measuring before end-expiration)
    • Poor / loss of ETCO2 calibration
    • Interference from other gases (e.g. N2O and collision broadening)
  • Physiological factors
    • Increasing age > increased gradient


Examiner comments

30% of candidates passed this question.

Many candidates didn’t distinguish between the different types of dead space. In general this topic was not well understood.


Online resources for this question


Similar questions

  • Question 3, 2018 (2nd sitting)
  • Question 24, 2007 (1st sitting)
  • Question 9, 2009 (1st sitting)



Question 22

Question

Outline the functions of the liver


Example answer

Examiner comments

56% of candidates passed this question

Most candidates attempted a structure however did not expand the answers within the categories: e.g. a passing mention of glucose homeostasis is insufficient to score full marks for the carbohydrate metabolism category.


Online resources for this question


Similar questions

  • Question 20, 2017 (2nd sitting)

  • Question 23, 2009 (2nd sitting)

  • Question 12, 2011 (2nd sitting)

  • Question 4, 2013 (2nd sitting)


Question 23

Question

Draw and label a left ventricular pressure volume loop in a normal adult (40% of marks). List the information that can be obtained from this loop (60% of marks).


Example answer

File:C:\Users\ethan\AppData\Roaming\Typora\typora-user-images\image-20210320173717612.png

Information obtained

  • Volumes
    • End diastolic volume
    • End systolic volume
    • Stroke volume
    • Ejection fraction
  • Pressures
    • Systolic BP
    • Diastolic BP
    • Pulse pressure
    • End systolic pressure
  • Pressure-volume relationships
    • End diastolic pressure-volume relationship (EDPVR)
      • Describes elastance, Non linear
    • End systolic pressure-volume relationship (ESPVR)
      • Describes contractility, Linear
    • Arterial elastance
      • Approximation of afterload
      • Line between EDV and ESP
  • Areas
    • Total mechanical work (combination of stroke and potential work)
      • Stroke work (inside PV loop)
      • Stored potential work (outside loop, under ESPVR line)


Examiner comments

65% of candidates passed this question.

Many candidates lost marks for poor quality diagrams with inaccurate labelling. An accurate diagram was required. Many answers lacked sufficient detail regarding contractility and afterload.


Online resources for this question


Similar questions

  • Question 24, 2008 (1st sitting)



Question 24

Question

Outline the physiology of cerebral spinal fluid (CSF).


Example answer

CSF

  • ECF located in the ventricles and subarachnoid space
  • ~2ml/kg of CSF
  • Divided evenly between the cranium and spinal column


Production

  • Constantly produced

  • ~550ml produced per day (~24mls/hr)

  • Produced by

    • Choroid plexus (70%) - located in ventricles of brain

    • Capillary endothelial cells (30%)

  • Produced by a combination of ultrafiltration (via fenestrated choroidal capillaries) and active secretion

Composition relative to plasma

  • Similar: Na, osmolality, HCO3
  • Increased: Cl, Mg, CO2
  • Decreased: pretty much everything else (protein, potassium, calcium, glucose, pH)


Circulation

  • Circulation is driven by

    • Ciliary movement of ependymal cells

    • Respiratory oscillations and arterial pulsations

    • Constant production and absorption

  • CSF flows from

    • Lateral ventricles > foramen of Monro > 3rd ventricle > Sylvian aqueduct > 4th ventricle > cisterna magna (via foramen megendie and luschka) > spreads between spinal/cranial subarachnoid spaces

Reabsorption

  • Rate of ~24mls/hr

  • By the arachnoid villi

    • Located predominately in the dural walls of the sagittal + sigmoid sinuses

    • Function as one way valves, with driving pressure leading to absorption.

Functions

  • Mechanical protection
    • The low specific gravity of CSF > decreased effective weight of the brain (1500g > 50g)
    • With the reduced weight
      • Less inertia = less acceleration/deceleration forces
      • Suspended > no contact with the rigid skull base
  • Buffering of ICP
    • CSF can be displaced / reabsorbed to offset any increase in ICP
  • Stable extracellular environment
    • Provides a constant, tightly controlled, ionic environment for normal neuronal activity
  • Control of respiration
    • The pH of CSF is important in the control of respiration (CO2 freely diffuses into CSF and can activate central chemoreceptors)
  • Nutrition
    • Provides a supply of oxygen, sugars, amino acids to supply the brain


Examiner comments

67% of candidates passed this question

Better answers included details on CSF production (amount, site), reabsorption and factors which influences CSF and its circulation.


Online resources for this question


Similar questions

  • Question 22, 2007 (1st sitting)
  • Question 6, 2008 (2nd sitting)
  • Question 2, 2013 (1st sitting)
  • Question 16, 2015 (1st sitting)
  • Question 24, 2017 (1st sitting)
  • Question 15, 2018 (2nd sitting)
  • Question 11, 2019 (1st sitting)
  • Question 16, 2020 (1st sitting)




2016 (1st sitting)

Question 8

Question

Describe the characteristics of a drug that influence its excretion by the kidneys


Example answer

Renal excretion of drugs is related to factors which affect

  • Filtration at the glomerulus
  • Secretion into the tubules
  • Reabsorption in the tubules


Factors affecting glomerular filtration

  • GFR
    • Increased GFR (e.g. increased CO) will lead to increased filtration and clearance of hydrophilic drugs
  • Drug size
    • Increasing drug size = decreased renal clearance
    • Only drugs <7kDa (weight) or <30 Angstrom units (width) are able to pass the capillary BM
  • Protein binding
    • Only unbound drugs can pass the glomerular BM (hence highly protein bound drugs are poorly filtered)
  • Charge
    • Negatively charged molecules cannot readily pass BM (as it is also negatively charged)


Factors affecting drug secretion

  • Active process
  • Protein binding, renal blood flow (GFR) as per above
  • Concentration: Increased concentration = increased secretion (until transporters are saturated)
  • Concomitant drugs - competition for receptors


Factors affecting drug reabsorption

  • Can be active or passive (most are passive)
  • Also depends on charge (ionised drugs cannot pass through BM) and become trapped in the urine
    • Ionisation depends on pH urine, pKa drug (acidic drugs are ionised in alkaline urine)
  • Concentration (as passive diffusion depends on concentration gradient)
  • Urine flow rate
    • Increased urine flow rate > reduces concentration of drug in urine > increased concentration gradient + elimination


Examiner comments

29% of candidates passed this question.

Drug characteristics that might influence the renal excretion processes include charge, size, solubility, and binding to specific structures or protein. Whether the drug is unchanged versus metabolised can influence these factors. This question tests core knowledge of pharmacology principles and should be answered with equations, graphs or simple clear descriptions of physical and chemical principles. Extended examples and hedged statements about “influencing” without the direction, magnitude and necessary conditions for the influence did not score marks.



Question 13

Question

Describe the cardiovascular effects of a sudden increase in afterload.


Example answer

Afterload

  • Force that must be overcome prior to the sarcomere being able to shorten during contraction (i.e. the forces opposing ventricular ejection)


CVS effects

  • HR
    • If the increase in afterload is associated with increase in carotid pressure > baroreceptor reflex activation > compensatory decrease in HR
  • SV
    • The increased afterload > earlier closure of the AV valve (increased diastolic pressure) and decrease in velocity of myocyte shortening > increased end-systolic volume (and pressure) > decreased stroke volume > decreased CO
  • Preload
    • Increase in afterload > decrease in SV > increase in LV end systolic volume (and pressure) > Increase in EDV (preload)
  • Contractility
    • Increases due to the Anrep effect
      • Increase in afterload > increased LV EDP > frank starling effect > Calcium accumulation > increased contractility > increased SV
  • Cardiac output
    • Decreases in short term
    • Ideally recovers quickly if the compensatory mechanisms work
    • If the LV is impaired, or the body is unable to compensate > LV heart failure
  • Myocardial oxygen consumption
    • Increases due to increased contractility and increased work to overcome afterload
  • Coronary blood flow
    • Remains stable due to autoregulation




File:C:\Users\ethan\AppData\Roaming\Typora\typora-user-images\image-20220713192800402.png



Examiner comments

21% of candidates passed this question.

It was expected the answer would start with a definition of afterload and then proceeded to indicate what effects this increase in afterload would have on ventricular end-systolic pressure, ventricular end-diastolic pressure, left atrial pressure, cardiac output, myocardial oxygen demand and myocardial work, coronary blood flow and systemic blood pressure.
Most candidates who failed to pass this question submitted answers that were just too brief, only including a small subset of the material required. Very few candidates included any mention of myocardial oxygen demand or myocardial work or the impact upon the cardiac output. A number of candidates included a detailed description of the Sympathetic Nervous System and the Renin-Angiotensin system, material which was not asked for. There were quite a number of incorrect perceptions about what effect a sudden increase in afterload would have on the systemic blood pressure. Candidates who mentioned the baroreceptor response and the stretch receptor response where rewarded with additional credit.


Online resources



Question 20

Question

Outline the role of the liver in drug pharmacokinetics


Example answer

Absorption

  • The liver will affect the bioavailability of drugs which are subject to first pass metabolism

  • Hence, oral absorption, high PR, > subject to hepatic extraction and metabolism

  • Drugs which are not subject to first pass metabolism e.g. inhalation, intravenous, IM ,> higher bioavailability if hepatically metabolism/cleared

  • Liver dysfunction > alter first pass metabolism

Distribution

  • The liver is responsible for producing majority of the proteins that drugs bind
  • Hence, for highly protein bound drugs (e.g. warfarin), small changes in protein levels, can lead to large changes in the proportion of unbound (active drug)
  • Liver dysfunction > alter protein binding


Metabolism

  • The liver is a primary organ of drug metabolism and biotransformation
  • Phase 1 reactions
    • Hydrolysis, Reduction, Oxidation
    • Small increase in hydrophilicity
  • Phase 2 reactions
    • Glucuronidation, sulfation, conjugation, methylation
    • Significantly increased hydrophilicity
  • Liver dysfunction can alter biotransformation
    • E.g. liver damage > unable to process paracetamol > increased toxicity


Elimination

  • The hepatic system is important for drug elimination
  • Drugs with a high hepatic extraction ratio, will also be dependant on the hepatic blood flow. Drugs with a low hepatic extraction ratio will depend on the function of the liver
  • For hydrophilic drugs, that are highly protein bound, decreased proteins related to hepatic dysfunction will increase elimination
  • Biliary section
  • Drugs which rely on biliary exertion will be retained in liver dysfunction
  • Portal hypertension > shunting of blood > decreased first pass metabolism


Examiner comments

62% of candidates passed this question.

Most candidates structured their answer to this question well – they were aware of first pass metabolism and the effect of protein synthesis upon volume of distribution of drugs. Knowledge concerning Phase I and Phase II reactions was frequently inadequate. Many candidates were aware that these processes as well as inactivating or activating drugs resulted in increased water solubility to aid excretion via bile or urine. Few candidates discussed the significance of the large blood flow to the liver or the implications of high and low extraction ratios especially in relation to liver blood flow



Question 24

Question

Describe the ideal sedative agent for an Intensive Care patient (50%). How does midazolam compare to this (50%)?


Example Answer

Name Midazolam Ideal sedative agent
Class Benzodiazepine (sedative) -
Pharmaceutics Clear colourless solution

pH 3.5.
Diluted in water.

- Water soluble

- Chemically stable with long shelf life (various temperatures)
- Does not need reconstitution.

- Compatible w. all drugs / IVF

- Enantiopure preparation
- No additives

Routes of administration IV, IM, S/C, intranasal, buccal, PO Multiple routes of administration available
pKa 6.5 -
Dose Variable Predictable response for a given weight based dosing regime
Pharmacodynamics
MOA Midazolam (BZD) binds to GABAA receptors (ionotropic ligand gated channel) in the CNS. Cl enters > hyperpolarisation. Known MOA with specific and targeted receptors
Effects CNS: sedation, amnesia, anxiolysis, hypnosis, anticonvulsant effects, decreased cerebral O2 demand, MSK: muscle relaxant CNS: sedation, amnesia, anxiolysis, decreased cerebral O2 demand
Side effects CVS: bradycardia, hypotension

CNS: confusion, restlessness
RESP: respiratory depression/ apnoea

No side effects, including no cardiorespiratory depression or emergence delirium
Pharmacokinetics
Onset peak effect 2-3 minutes (IV), offset variable Rapid onset / offset
Absorption ~40% oral bioavailability

- Absorbed well, but sig. 1st pass metabolism

Absorbed well from all routes, including oral and inhaled with minimal first pass metabolism
Distribution Vd = 1L / kg

95% protein bound

Vd = <0.3L/Kg

Minimal protein binding (decreases availability)

Metabolism Hepatic metabolism by hydroxylation

Active (1-a hydroxymidazolam) and inactive metabolites

Either no metabolism or organ independent metabolism with inactive metabolites (prevents accumulation)
Elimination Renal excretion

T 1/2 = 4 hours

Rapidly cleared with a short and predictable half life and small CSHT
Reversal Flumazenil - antagonist (reversal agent) Readily reversible with no rebound/side effects


Examiner comments

60% of candidates passed this question.

Candidates who had a structured approach (i.e. pharmaceutical, pharmacokinetic, pharmacodynamic) provided more content and scored higher. Candidates who also approached pharmacodynamic effects in an organ system based approach scored higher. Relating a pharmacokinetic property of midazolam (e.g. volume of distribution or half-life) to a un/desirable attribute e.g. offset of action and accumulation displayed a greater understanding of the question. For many candidates, the description of an ideal drug contained more detail and candidates were not able to adequately state how midazolam compares.


Online resources





2016 (2nd sitting)

Question 4

Question

Categorise the drugs used in the treatment of asthma, give examples and outline their mechanism of action


Example Answer

Oxygen

  • Increases FiO2 > increased SaO2 (by increasing PAO2 as per Alveolar gas equation).
  • Given by numerous devices (nasal prongs, masks, NIV, ETT)
  • Dose titrated to SaO2
    • Hypoxemia is harmful (but optimal target SaO2 unclear)
    • Generally titrated to Sats 94-98% (with caveats for some subgroups of patients)
    • Hyperoxia may lead to hypercapnia, worsening of V/Q mismatch (through alteration of HPVC), lung damage


Beta-adrenergic agonists

  • Long acting B2 selective agonists (e.g. salmeterol) are used in prevention
  • Short acting B2 selective agonists (e.g. salbutamol) are preferred first line therapy for exacerbation
  • Nonselective adrenergic agonists (e.g. adrenaline) can also be used in severe exacerbations
  • SABAs can be given inhaled (via spacer), nebulised or via IV infusion (if unresponsive to inhaled)
  • Example: Salbutamol
    • Short acting B2 agonist
    • MOA: Acts on B2 receptors (Gs protein coupled receptors) in bronchial smooth muscle cells > activates activates adenyl cyclase-CAMP system > increase cAMP > decreased intracellular Ca > SM relaxation / bronchodilation
    • Side effects: Tachycardia, Anxiety, tremor, Hypokalaemia, lactic acidosis


Anticholinergics

  • Example: ipratropium bromide
  • Routes: Inhaled, nebuliser
  • MOA: Competitive antagonism of muscarinic ACh receptors > bronchodilation + decreased secretions
  • Side effects: dry mouth, N/V, headache, blurred vision


Corticosteroids

  • Examples: hydrocortisone (IV), prednisone (PO), budesonide (inhaled)
  • Systemic corticosteroids should be given to all mod-severe asthma > improve outcomes
  • MOA: bind to cytoplasmic glucocorticoid receptors > change in gene transcription > down-regulates the synthesis of proinflammatory cytokines/mediators
  • Effects: increased B receptor responsiveness, decreased inflammation, decreased mucus secretion
  • Side effects: numerous! Depends on dose/duration. Examples:
    • Short term: hyperglycaemia, hypokalaemia, immunosuppression, insomnia/confusion/psychosis,
    • Long term: cushings, osteoporosis, skin thinning, weight gain, immunosuppression


Other potential treatment options (and MOA)

  • Magnesium sulphate > inhibits L type calcium channels > bronchodilation/SM relaxation
  • Ketamine >inhibits L type calcium channels > Bronchial smooth muscle relaxation
  • Aminophylline > PDEI > SM relaxation / bronchodilation
  • Heliox > Improves laminar airflow > may improve ventilation
  • Inhaled anaesthetics (e.g. sevoflurane)
  • Montelukast (leukotriene receptor antagonist used in children)


Examiner comments

71% of candidates passed this question.

Asthma drugs are typically categorised according to mechanism of action. A reasonable alternative is to categorise by clinical use, e.g. short acting, long acting, preventer, rescue etc.
A lot of emphasis in marking was placed on an understanding of the beta-adrenergic pathway, its secondary messenger system and how this medicates smooth muscle relaxation. Candidates whose answers had structure as well those who described the wide range of drugs used to treat asthma scored well.




Question 9

Question

Describe the immunology and drug treatment of anaphylaxis.


Answer

Anaphylaxis

  • Life threatening systemic hypersensitivity reaction
  • May be immune mediated (IgE or non IgE) or non immune mediated


IgE immune mediated anaphylaxis (a Type-1 hypersensitivity reaction)

  • Initial contact between a B-cell and an antigen (allergen) leads to the formation of a IgE against it
  • The specific IgE then binds to Fc receptors on mast cells (in tissues) and basophilis (in circulation)
  • Further exposure of the antigen leads to formation of cross links between IgE-Fc complex and the antigen which leads to activation and release of pre-synthesised mediators
    • Mediators
      • Histamine -> vasodilation, increased vascular permeability, increased chronotropy
      • Leukotrienes -> bronchoconstriction, increased vascular permeability
      • Serotonin -> SM contraction
      • Tryptase -> activates complement, coagulation and Kallikrein-kinin pathways
      • Platelet activating factor --> platelet activation
  • This manifests as
    • CVS: Hypotension/cardiovascular collapse, flushing
    • RESP: Bronchospasm, airway oedema, angioedema, dyspnoea, stridor, hypoxaemia
    • DERM: pruritis, urticaria, angioedema,
    • GIT: abdominal pain, nausea, vomiting, diarrhoea


DRUG TREATMENT


Oxygen

  • Increase FiO2 > improved oxygenation whilst there is bronchoconstriction/airway oedema


Fluids

  • Increase in MSFP > increase VR > increased CO > increased BP


Adrenaline

  • Mainstay of treatment for anaphylaxis

    • Treats cardiovascular collapse, bronchospasm and prevents further degranulation of mast cells

  • Dose is 0.3-0.5mg IM (adults), 0.01mg/kg IM (children), every 5-15 mins (or infusion as needed)

  • Effects

    • Alpha 1 mediated vasconstriction > increases SVR > increases BP

    • B1 mediated increase in inotropy > increase in CO > increase in BP

    • B2 mediated bronchodilation and mast cell/basophil stabilisation

Supplemental drug treatment


Bronchodilators

  • E.g. salbutamol, adrenaline
  • Supportive management for severe bronchospasm
    • B2 agonism > bronchodilation > decrease airways resistance > improve WOB and oxygenation
  • Does not alter the course of the illness


Glucocorticoids

  • e.g. methylpred, pred, hydrocort

  • binds to intracellular steroid receptors > alters gene transcription > anti-inflammatory & immunosuppressive effects

  • Do not alter acute course of illness

  • May prevent biphasic responses / prolonged course of illness

Antihistamines

  • e.g. loratadine, premethazine
  • Symptomatic treatment strategy in mild disease
    • May provide some relief from pruritis/rash (via blocking H1 receptor)
    • Minimal effects on systemic mast cell and basophil degranulation
    • No effects on outcomes
  • Not recommended for severe disease


Glucagon

  • For patients who have taken b-blockers (and thus have reduced responsiveness to adrenaline)



Examiner comments

32% of candidates passed this question.

It was expected candidates would detail the process of IgE mediated type I hypersensitivity reaction with some discussion of the mediators (Histamine / tryptase and others) and their consequences. Some detail describing time frame of response and the pre-exposure to Antigen (or a similar Antigen) was expected. Drug treatments would include oxygen and fluids as well as more specific agents such as adrenaline and steroids. Adrenaline is the mainstay of therapy and some comment on its haemodynamic role and prevention of ongoing mast cell degranulation was required.
Better answers noted steroids take time to work and some also discussed the role of histamine blocking agents


Online resources






Question 16

Question

Outline the influence of pregnancy on pharmacokinetics


Example answer

Absorption

  • Oral
    • Nausea and vomiting in early preg > reduced PO absorption
    • Increased intestinal blood flow (due to increased CO) > increased PO absorption
    • Decreased gastric acid production > increased pH > unionised drugs absorbed more
    • Delayed gastric emptying peri-labour may increase/decrease absorption depending on drug
  • IM / SC / Transdermal
    • Increased absorption due to increased CO + increased skin/muscle blood flow
  • IV
    • Faster IV onset due to increased CO
  • Neuraxial
    • Decreased peridural space (venous engorgement) > decreased dose required


Distribution

  • Volume of distribution
    • Increased total body water > increased Vd for hydrophilic drugs
    • Increased body fat > increased Vd for lipophilic drugs
  • Plasma proteins
    • Decreased protein binding (increased free fraction) due to reduced concentrations albumin and a-1 glycoprotein


Metabolism

  • Hepatic
    • Some metabolic enzymes reduced / some increased (due to progesterone/oestrogen ratio)
    • Leads to variable drug responses
    • E.g. increased metabolism of midazolam, phenytoin, but decreased caffeine.
  • Placenta metabolises some drugs (COMT and MOA enzymes > metabolises catecholamines)
  • Decreased plasma cholinesterase (though no change in Succinylcholine effect)


Elimination

  • Renal
    • Increased clearance due to increased GFR (e.g. gentamycin)
  • Hepatobiliary
    • Decreased clearance due to cholestatic effects of oestrogen (e.g. rifampicin)
  • Resp
    • Increased volatile washout due to increased minute ventilation


Examiner comments

47 % of candidates passed this question.

Most candidates divided the answer into effects on absorption, distribution, metabolism and elimination, which is a good way of presenting the answer. However, the good candidates also mentioned effects on the foetus due to ion trapping caused by the more acidic foetal blood.
Many candidates forgot to include effect on epidural administration of drugs in pregnancy caused by engorged epidural veins during labour.
Candidates lost marks for omitting the effect of increased cardiac output on the rate of distribution of IV drugs to effector sites, the effect of increased hepatic blood flow on drugs with high intrinsic clearance, the increased clearance of drugs with renal clearance due to increased GFR & renal plasma flow



Question 23

Question

Compare and contrast the mechanism of action, spectrum of activity and adverse effects of benzyl penicillin and fluconazole.


Answer

Benzylpenicillin Fluconazole
Mechanism of action Penicillin antibiotic

Both disrupt cell wall synthesis (but different mechanisms): Binds to penicillin binding proteins > inhibits peptidoglycan cross linking > bactericidal

Azole antifungal

Inhibits the fungal CYP450 enzyme responsible for ergosterol production (needed for fungal cell membrane synthesis) > cell death

Spectrum of activity Narrow spectrum penicillin

Covers: Most GPC (staph, strep, enterococci), some GPB (e.g. listeria), very few GNC (e.g. Neisseria sp).
Does not cover: MRSA, CRE, VRE, manty other GN bacteria + anaerobes, all fungi/yeasts, all parasites/ protozoans

Narrow spectrum azole

Covers: Candida and cryptococcal species
Does not cover: some candida sp (e.g. krusei), aspergillus and most other fungi/yeast, all bacteria (GP and GN), all parasites/protozoa

Adverse effects CNS: confusion, coma, seizure

CVS: Nil major
RESP: Nil
GIT: Raised LFTs, nausea and vomiting and abdominal pain, pseudomembranous colitis
HAEM: agranulocytosis
RENAL: Interstitial nephritis
IMMUNO: rash, allergy, anaphylaxis
OTHER: less drug interactions, not a teratogen

CNS: headache

CVS: Prolonged QTc
RESP: Nil
GIT: Raised LFTs, nausea, vomiting, abdo pain
HAEM: thrombocytopaenia, leukopaenia
RENAL: Nil
IMMUNO: rash, allergy, alopecia, anaphylaxis
OTHER: Drug interactions (CYP450), teratogen


Examiner comments

8% of candidates passed this question.

To pass this question each of the three components needed to be compared and contrasted for both agents. A tabulated answer helped in this regard but was not essential.
Some answers included information that could not gain marks, as it was not directly relevant to the question asked (e.g. presentation and dose).
In spectrum of activity, as well as what important organisms the agents were effective against, marks were also given for the important organisms that they were not effective against (e.g. MRSA and beta-lactamase producing organisms for penicillin G; and aspergillus for fluconazole).
In general, of the two agents, fluconazole was the least well answered. For example, a common omission either in mechanism of action or in adverse effects was that fluconazole inhibits microsomal P450 enzymes.
Some candidates confused fluoroquinalone with fluconazole.


Online resources







2015 (1st sitting)

Question 10

Question

Compare and contrast the pharmacology of mannitol and hypertonic saline.


Example Answer

Name Mannitol Hypertonic saline
Class Osmotherapy agent / osmotic diuretic Osmotherapy agent / concentrated electrolyte
Indications Temporary reduction in ICP / IOP

Diuresis

Temporary reduction in ICP / IOP

Hyponatraemia

Pharmaceutics Clear colourless solution (10-25% conc)

- 10% = 10g/100ml
Precipitates at low temperatures

Clear colourless solution (various concentrations)

- 3% saline = 513 mmols Na + Cl (= osmolarity 1026)

Routes of administration IV IV (central)

- Risk phlebitis, necrosis

Dose 0.25-1g/kg bolus (max 100g)

repeated 3 hourly

3ml/kg (3% saline) bolus over 10 mins. Can be repeated to target Na 145-155
pKa 12.6 3.1
Pharmacodynamics
MOA ↑ osmolality of ECF > ↓ volume of ICF (through osmotic shift) > ↓ cerebral volume > ↓ ICP.

Also --> freely filtered at glomerulus (but not reabsorbed) > acts osmotically to ↓ H2O reabsorption.

Increases osmolality of ECF > decreases volume of ICF (through osmotic shift) > decreases cerebral volume > decreases ICP
Effects/side effects RENAL: osmotic diuresis, electrolyte disturbances (variable)

CNS: increased osmolality of ECF > osmotic fluid shifts out of cells > decreased ICP/IOP
CVS: initial rise in MSFP>preload>BP (fluid load) which then decreases with diuresis > hypotension
RESP: Pulmonary oedema (increased in ECF volume)

Renal: Increases Na, Cl, NAGMA, osmolality

MSK: necrosis/phlebitis if given peripherally/extravasates
CVS: increased ECF > overload
RESP: pulmonary oedema (fluid overload)

Pharmacokinetics
Onset / duration Onset <15 mins

Duration = 4-6 hours

Onset <15 mins

Duration = 1 hour

Absorption Given IV only (PO bioavailability - 0%) IV only
Distribution Does not cross BBB

VOD = 0.2L / Kg
75% becomes interstitial fluid, 25% intravascular

Does not cross BBB

VOD = 0.2L / Kg
75% becomes interstitial fluid, 25% intravascular

Metabolism Nil (negligible hepatic metabolism) Nil
Elimination Renal elimination (unchanged)

T 1/2 = 2-3 hours

Renal elimination (unchanged)

T 1/2 =

Monitoring Monitoring (osmolality, 320) Monitoring (Na 145-155)
Advantages Relatively cheap, as effective as HT saline Cheap, stable, small volumes

No diuretic effect > no hypoT
Easily monitored

Disadvantages Unstable at low temps, leads to diuresis, more cumbersome to monitor Needs central access, can cause hypernatraemia,


Examiner comments

8 % of candidates passed this question.

A structured approach is important and a table worked best for most candidates, although a few attempted this in free text. Despite attempting a structured answer very few candidates provided information in regards to preparation, dose, monitoring of osmolarity, adverse effects or contraindications. Understanding of the action of these drugs was expected and factual inaccuracies were common with many candidates suggesting hypertonic saline acts as an osmotic diuretic. Better answers mentioned other potential mechanisms of action of mannitol. Many candidates failed to appreciate the impact on raised intracranial pressure.


Online resources



2015 (2nd sitting)

Question 24

Question

Compare and contrast the pharmacology of valproic acid and carbamazepine


Example Answer

Name Sodium valproate (valproic acid) Carbamazepine
Class Anticonvulsant Anticonvulsant
Indications - Migraine

- Epilepsy (simple-complex and focal-generalised)
- Status epilepticus

- Epilepsy

- Trigeminal neuralgia
- Bipolar disorder

Pharmaceutics Enteric coated tablets, oral liquid

Powder for reconstitution

IR and MR tablets

Oral liquid

Routes of administration IV, PO PO
Dose 15-30mg/kg in divided doses 400mg-1.2g in 2/3 divided doses
Pharmacodynamics
MOA Stabilises Na channels in their inactive state, thereby inhibiting the generation of further action potentials. Also by stimulating GABAergic inhibitory pathways Stabilises Na channels in their inactive state, thereby inhibiting the generation of further action potentials. Also by stimulating GABAergic inhibitory pathways
Effects CNS: Anticonvulsant, drowsiness, dizziness, ataxia

GIT: Nausea, dyspepsia, liver failure, pancreatitis
HAEM: thrombocytopaenia, neutropoenia
Other: teratogen, hair loss

CNS: Anticonvulsant, drowsiness, dizziness, ataxia, headache, diplopia

GIT: Nz, Vz, Dz, raised LFTs
HAEM: neutropoenia, thrombocytopaenia
OTHER: severe skin reactions, teratogen

Pharmacokinetics
Onset TMax 2 hrs (PO), immediate (IV) TMax 1.5 hours PO
Absorption PO bioavailability = 90% PO bioavailability = 80%
Distribution Protein binding 90%

VOD = 0.2L / Kg

Protein binding = 75%

VOD = 1L/kg

Metabolism Hepatic metabolism (glucuronidation)

Active and inactive metabolites

Hepatic (98%)

CYP3A4
Active metabolites

Elimination Renal elimination of metabolites (85%)

T 1/2 = 12 hours

Renal (70%) and faecal (30%) elimination

T 1/2 = 14 hours (metabolites 30 hours)

Special points Monitor LFTs first 6 months given risk of liver failure


Examiner comments

6% of candidates passed this question.

Both these agents are listed as “level B” in the syllabus pharmacopeia and as such a general understanding of each class and relevant pharmacokinetics and pharmacodynamics was expected. Most candidates had better knowledge of valproate than carbamazepine. Some description of the toxicological features for intensive care practitioners was expected.


Online resources





2014 (2nd sitting)

Question 13

Question

Outline the pharmacology of amiodarone


Answer

Name Amiodarone
Class Antiarrhythmic (Class III)

- However, also has class I, II, and IV activity

Indications Tachyarrhythmias (e.g. SVT, VT, WPW)
Pharmaceutics 100-200mg tablets

Clear solution in 150mg ampoules for dilution in dextrose

Routes of administration IV and PO
Dose IV: 5mg/kg, then 15mg/kg infusion / 24hrs.

Oral: 200mg TDS (1/52) > BD (1/52) > daily thereafter

pKA 6.6
Pharmacodynamics
MOA - Blocks K channels (Class III effects) prolonging repolarisation and therefore refractory period.

- Decreases velocity of Phase 0 by Blocking Na channels (Class I effects)
- Non-competitive inhibition of Ca channels prolonging depolarisation + AV nodal conduction time (Class IV effects)
- Slows AV/SA nodal conduction via anti-adrenergic activity (Class II effects)

Effects Rhythm / rate control of tachyarrhythmias
Side effects Side effects worsen/increase with duration of therapy!

RESP: pneumonitis, fibrosis
CVS: bradycardia, QT prolongation
CNS: peripheral neuropathy
Thyroid: Hypo/hyperthyroidism
LIVER: cirrhosis, hepatitis
DERM: photosensitivity, skin discolouration

Pharmacokinetics
Onset / TMax Immediate (IV), 4 hours (PO)
Absorption PO bioavailability 40-60%
Distribution Highly protein bound (>95%)

VD ~70L /kg

Metabolism Hepatic (CYP3A4) with active metabolites (desmethylamiodarone)
Elimination T1/2 = 1 month

Faces, urine, skin elimination

Special points Many drug-drug interactions (e.g. digoxin and warfarin) due to high PPB and enzymatic system


Examiner comments

77% of candidates passed this question.

This was a repeat question and was generally answered well. Some candidates lost marks for being too approximate on the pharmacokinetics.








Question 17

Question

Describe the pharmacology of Oxygen


Answer

Name Oxygen
Class Naturally occurring gas (atomic number 8, atomic weight 16)
Indications Supplementation (i.e hypoxia)

Prophylaxis (e.g. prior to intubation)
CO poisoning
Pneumothorax
Decompression sickness

Pharmaceutics Diatomic gas, normally present at 21% in atmosphere

Colourless, tasteless, odourless
Stored in cylinders (various forms), flammable

Routes of administration Inhaled (variety of delivery devices)

Intravenous (i.e. ECMO)
External (hyperbaric oxygen therapy)

Dose 0.21 - 1.0 FiO2 (generally targeting SaO2 >94%; or 88-92% on CO2 retainers; though exact target not entirely evidenced based)
Pharmacodynamics
MOA Oxygen is delivered to tissues for aerobic metabolism via oxidative phosphorylation
Effects RESP: improved oxygen saturations (may also improve DO2), decreased respiratory drive (very minimal), pulmonary toxicity (free radical generation), may worsen V/Q mismatch (impairs HPVC), drying of mucous membranes

CVS: decreased pulmonary vascular resistance (vasodilation) due to reversal of HPV, increased HR/SV/SVR if hypoxic (via chemoreceptor reflex) > increased CO and BP
CNS: anxiety, nausea, visual changes (neonates), seizures (hyperbaric hyperoxia), decreased CBF (vasoconstriction)
MET: oxidative phosphorylation > ATP production

Pharmacokinetics
Absorption Diffusion across the alveolar capillary membrane.

Rate of diffusion is governed by Fick's Law and is therefore proportional to the lung area, gas diffusion constant, partial pressure gradient and inversely proportional to membrane thickness

Distribution Bound to plasma Hb (98%)

Dissolved in plasma (<2%) - related to Henrys Law

Metabolism Metabolised in mitochondria during the Citric acid cycle, to produce ATP and generate CO2 and H2O
Elimination Exhalation of CO2 via lungs
Special points


Examiner comments

35% of candidates passed this question.

Use of a general "pharmacology" structure to answer this question would help avoid significant omissions such as only discussing pharmacokinetics or only discussing pharmacodynamics. Oxygen has a well described list of pharmacodynamics effects that includes, cardiovascular, respiratory and central nervous system effects. Candidates’ knowledge of the pharmaceutics was limited for a routine drug. It was expected candidates would mention the potential for oxygen toxicity including a possible impact on respiratory drive in selected individuals, retrolental fibroplasia and seizures under some circumstances. Many candidates did not answer the question asked, and instead focussed on the physiology of oxygen delivery and binding of oxygen to haemoglobin


Online resources



2014 (1st sitting)

Question 5

Question

Describe the pharmacological effects of paracetamol (40% marks). Outline its toxic effects and their management (60% marks).


Example Answer

Pharmacological effects

  • MOA
    • Not entirely understood
    • Analgesic effect thought to be related to
      • Decreased central prostaglandin synthesis by inhibition of COX-3
      • Modulation of 5-HT pathways (increased descending inhibition)
      • Activation of endocannabinoid (CB1) and capsaicin (TRPV1) receptors
    • Antipyretic effect thought to be due to decreased PG synthesis in hypothalamus by COX-3
  • Effects/side effects
    • CNS: analgesia, antipyretic
    • CVS: Hypotension (IV preparation, related to excipients)
    • GIT: deranged LFTs, liver failure/damage in high doses/toxicity (below)
    • Other: hypersensitivity reactions


Toxic effects

  • Mechanism
    • Paracetamol is normally hepatically metabolised
      • Principally it is conjugated (glucuronide and sulfate) > eliminated
      • Small amounts undergo oxidation > toxic metabolites (NAPQI)
    • In regular amounts, the NAPQI can be neutralised by hepatic glutathione (anti-oxidant)
    • In excess/toxicity, the conjugative pathways are saturated and there is increased oxidation > increased NAPQI. The hepatic glutathione is exhausted > build up of NAPQI > liver damage
  • Effects of toxicity:
    • GIT: Liver failure / hepatitis, abdominal pain, nausea, vomiting
    • HAEM: coagulopathy (related to liver failure)
    • MET: impaired glucose homeostasis, lactataemia
    • CVS: peripheral vasodilation > shock
  • Management
    • Immediately post ingestion
      • activated charcoal
    • N-acetylcysteine infusion
      • Converted to glutathione > replenishes stores
      • Increased inactivation of the toxic metabolites (NAPQI) > reduced liver damage
      • 200mg/kg over first 4 hours, then 100mg/kg over next 16 hours
    • Supportive care
      • Fluids, antiemetics, etc
      • Complications of acute liver failure
      • Dialysis, ventilation etc


Examiner comments

63% of candidates passed this question.

This question was generally well answered with narrow variance; very few candidates discussed factors predisposing to hepato-toxicity or renal toxicity. Discussion of pharmacokinetics only gained marks when relevant to toxicity.


Online resources



Question 17

Question

Classify local anaesthetic agents and give examples (30% marks). Describe the pharmacology of lignocaine (70% marks).


Example Answer

Local anaesthetics

Classified according to the linkage between the hydrophilic and lipophilic groups

Esters Amides
Link Ester link Amide link
Examples Cocaine, tetracaine, procaine Lidocaine, bupivacaine, ropivacaine
Stability in solution Unstable More stable
Metabolism Plasma esterase's Hepatic (CYP450) dealkylation
Onset Slow Faster
Duration Shorter Longer
Toxicity Less likely More likely
Allergy Possible Very rare


Lignocaine/Lidocaine

Name Lidocaine (lignocaine)
Class Amide local anaesthetic / Class 1b antiarrhythmic
Indications Local/regional/epidural anaesthesia, ventricular dysrhythmias, pain
Pharmaceutics Clear colourless solution (1%, 2%, 4%).

Can come with/without adrenaline. Also available as cream/spray

Routes of administration SC, IV, epidural, inhaled, topical, PO
Dose Regional: Toxic dose 3mg/kg (without adrenaline), 7mg/kg (with adrenaline)

IV use: 1mg/kg initially, then ~1-2mg/kg/hr

pKA 7.9, 25% unionised at normal body fluid pH
Pharmacodynamics
MOA Class 1b anti-arrhythmic: blocks Na channels, raising threshold potential + reducing slope of Phase 0 of action potential, shortened AP

Local anaesthetic: binds to, and blocks, internal surface of Na channels

Effects Analgesic, anaesthetic, anti-arrhythmic
Side effects CNS: headache, dizziness, confusion, paraesthesia, reduced LOC, seizures, tinnitus, burred vision

CVS: hypotension, bradycardia, AV Block, arrhythmia
CC:CNS ratio = 7 (lower number = more cardiotoxic)
Other: allergy, anaphylaxis, methaemaglobinemia,

Pharmacokinetics
Onset Rapid onset (1-5 minutes)
Absorption IV > Epidural > subcut .

Oral bioavailability ~35%.
S/C Dependant on site of injection, blood flow, use of adrenaline.

Distribution 70% protein bound

Vd ~0.9L/kg.
Crosses BBB

Metabolism Hepatic (CYP450 dealkylation)

Some active metabolites

Elimination Renal excretion (98%) of metabolites

Half life ~90mins --> Increased with adrenaline (SC).
Reduced in cardiac/hepatic failure.

Special points Intralipid can be used in LA toxicity


Examiner comments

71% of candidates passed this question.

The first part of this question was answered well by most candidates.
Generally, the second part of the question was poorly organised by many candidates, the consequence being that many opportunities for picking up marks were lost. A brief statement as to what lignocaine is, its presentations and dose, some facts about PD and PK followed by a few lines on toxicity (CC/CNS ratio) was mostly what was required. Only a few candidates mentioned lignocaine toxicity.


Online resources




Question 20

Question

Describe the factors affecting drug absorption from the gastrointestinal tract


Example answer

Drug factors

  • Concentration of drug
    • Increased concentration gradient = more rapid absorption
  • Physical form of drug
    • Liquid drug > faster gastric transit time
    • Ability to dissolve (e.g. enteric coatings) > delays time
    • MR preparations > delayed absorption
  • pKa of drug
    • Weaker acids better absorbed
  • Lipophilicity of the drug
    • Lipophilic drugs better absorbed
  • Size of the drug
    • Smaller = faster/more readily absorbed
  • Drug-drug interactions
    • E.g. vitamin c increases absorption of iron
    • activated charcoal prevents absorption of some drugs through chelation


Patient factors

  • Gastric emptying time
    • Diarrhoea, constipation, ileus will all influence this > slow/fasten absorption
  • Food intake
    • Drugs can interact with food (e.g. iron absorbed better with orange juice due to vitamin C)
  • Altered surface area of the GIT
    • E.g. chrons or surgical short gut > decreased absorption
  • GIT blood flow
    • Reduced blood flow > decreased rate of absorption
  • Biliary function
    • Emulsifying effect of bile important for absorption of fat soluble vitamins and steroids
  • Pancreatic function


Examiner comments

45% of candidates passed this question.

This is a very broad and open question. While a structured approach was useful, a sound knowledge of first principles or even being able to “think on the fly” would have provided candidates with enough opportunities to generate a pass.



2013 (1st sitting)

Question 1

Question

List the different mechanisms of drug actions with examples


Answer

Classification Mechanism Example
NON RECEPTOR
Physiochemical actions Drug exerts its effects due to its physiochemical composition Antacids (basic) which neutralise gastric acid > decreased GORD symptoms
Colligative properties Drug exerts its effect due to the concentration of solute, not the identity of the solute Mannitol > increased plasma osmolality > diuresis
Actions on enzyme systems Decreased concentration of the substrate or product of the enzyme system ACE inhibitors > decreased concentration of angiotensin II
Prodrugs Converted from inactive drug > active drug following administration Levodopa > dopamine
Alteration of a carrier protein Alter the normal function of a carrier protein Frusemide which inhibits NaK2Cl co transporter in LOH > diuresis
Voltage gated ion channels Activated by changes in membrane potential near the ion channel Local anaesthetics > block voltage gated Na channels
RECEPTOR
Binding to intracellular receptors Lead to changes in cell function by altering DNA/RNA transcription Steroids (nuclear receptor)
Binding to ionotropic receptors Lead to changes in cell function by allowing flow of ions down a concentration/electrical gradient GABAA receptor: GABA binds > Cl channel opens > hyperpolarisation > inhibitory post synaptic potential
Binding to metabotropic receptors Bind to G protein coupled receptors and lead to changes in cell function through chemical second messenger systems Adrenaline binds to Gs PCR in myocardium > activation of cAMP 2nd messenger pathway > increased inotropy
Binding to enzyme coupled receptors Lead to changes in cell function through activation of an intracellular enzyme system Tyrosine kinase receptor (class II): insulin binds > activates tyrosine kinases on intracellular domain > phosphorylates IRS > cellular cascade


Examiner comments

A good answer to this question required candidates to think broadly about how drugs act and have a system for classifying their actions. One possible classification is action via receptors or non-receptor actions. Many candidates used categories such as physiochemical, receptor and enzymes. Common problems were failure to mention a whole class of drug actions e.g. drugs acting via voltage-gated ion channels or gene transcription regulation. Candidates also gave far too much detail in some sections e.g. a description of zero order and first order kinetics is not required. Candidates often did not give examples of the drug action they described.


Online resources


Question 4

Question

Describe the pharmacology of tranexemic acid.


Answer

Name Tranexamic acid (TXA)
Class Antifibrinolytic
Indications Trauma (within 3 hours)

Cardiac/obstetric/orthopaedic/dental surgery
Haemorrhage/Coagulopathy
Hereditary angioedema
Heavy menstrual bleeding
Epistaxis

Pharmaceutics 500mg Tablets (PO)

Clear colourless solution (100mg/ml) for injection (IV)

Routes of administration PO, IV, nebulised, topical, IM
Dose Trauma: 1 g (slow IV push) -->infusion of 1g over 8 hrs (if needed)

1g TDS/QID (PO) for most other conditions

pKA 10.2
Pharmacodynamics
MOA Competitive inhibition of plasminogen activation

-> binds to lysine binding sites of plasminogen
-> prevents the activation of plasminogen > plasmin
-> decreases fibrinolysis

Effects HAEM: Decreased fibrinolysis > prothrombotic complications in those patients with risk factors

GIT: nausea, vomiting, diarrhoea
CNS: seizures, headache, dizziness (dose related)
CVS: hypotension (rapid administration)
DERM: allergic skin reactions

Pharmacokinetics
Onset / duration Immediate (IV), 1 hour (IM), 2 hours (PO)

Duration = 18-24 hours

Absorption PO bioavailability = 50%

IM/IV bioavailability 100%

Distribution Protein binding: very low (<5%)

VOD = 0.3L / kg

Metabolism Minimal (<5%) hepatic metabolism

Inactive metabolites

Elimination Renal elimination of active drug (95% unchanged)
T 1/2 = 2hrs (IV), 12 hrs (PO)
Special points Dose reduce in renal failure


Examiner comments

Tranexamic acid is a drug used to reduce bleeding in trauma or surgery. It is also used for hereditary angioedema and menstrual bleeding. It is being increasingly used in critically ill patients. As a Level B listed drug within the Primary Syllabus candidates would be expected to know it in some depth. Often basic information such as mechanism of action, pharmacokinetics and adverse effects was lacking.


Online resources





Question 13

Question

Outline the effects of critical illness on drug pharmacokinetics


Example answer

Absorption

  • Oral
    • Decreased CO > decreased GIT blood flow > decreased absorption PO drugs
    • Ileus + uraemia > decreased gastric emptying > decreased absorption of PO drugs
    • Diarrhoea > fast transit time > decreased absorption
    • Change in gastric pH (e.g. with PPI) alters drug absorption
  • Topical/IM/SC
    • Vasoconstriction > poor tissue perfusion > decreased/slow absorption
  • Inhalational
    • Decreased MV / TV > decreased delivery of aerosolised medications


Distribution

  • Altered Vd
    • Decreased CO (e.g. shock) > slower redistribution
    • Increased CO (e.g. hyperdynamic sepsis) > faster residistribution
    • Hypervolaemia (e.g. renal, cardiac, liver failure) > increased Vd (vice versa)
    • Critical illness > muscle wasting > alter lean mass percentage (alters Vd)
  • Protein binding
    • Decreased protein synthesis > increased unbound fraction of drug > increased Vd and activity
    • Acid-base disturbances will alter free drug levels depending on drug pKa and the pH
  • Inflammation > impairs barrier function (e.g. BBB) > increased penetration of meds (e.g. penicillins)


Metabolism

  • Decreased CO > decreased hepatic/renal blood flow > decreased metabolism (e.g. propofol)
  • Liver dysfunction > Impaired phase 1 and 2 reactions and reduced 1st pass effect > (e.g. labetalol, metoprolol)
  • Renal dysfunction > decreased renal metabolism > prolonged drug effect (e.g. morphine)
  • Hypothermia > decreased metabolism > Prolonged effect (e.g midazolam)
  • Resp dysfunction > Decreased resp metabolism of drugs (e.g. opioids) > prolonged effect


Elimination

  • Decreased CO (e.g. cardiogenic shock) = decreased GFR / HBF > decreased clearance (e.g. gentamicin)
  • Increased CO (e.g. hyperdynamic sepsis) > increased GFR > increased clearance
  • Liver dysfunction > impaired biliary excretion of drugs (e.g. vecuronium)
  • Decreased GFR (e.g. AKI) > decreased renal elimination drugs (e.g. Gentamicin, milrinone)
  • Reduced MV > decreased / slower clearance of volatile anaesthetics > prolonged effect


Examiner comments

Most candidates answered the question under the subheadings absorption, distribution, metabolism and elimination. However, they didn’t give any details of the direction or mechanism of change, often used vague statements without specifically addressing the question and failed to give examples. The impact of the shock state on different kinetic parameters including absorption from skin, tissue, muscles, enteral absorption and inhalational was often overlooked. Similarly, the consequences of changes in volume of distribution, protein binding (e.g. albumin and globulin, ionisation) was poorly understood as was alteration in liver and kidney function. Although this topic is very broad candidates were asked to only outline the details of this topic




Question 23

Question

How do chemical messengers in the extracellular fluid bring about changes in cell function? Give an example of a chemical messenger for each mechanism noted


Example Answer

Chemical messengers (ligands) bind to receptors to elicit a response.

Receptors may be located on the cell surface or within the cell.


Intracellular receptors

  • Proteins located in the cytosol or cell nucleus
  • Activated by lipid soluble ligands (as they must be able to penetrate the lipid bilayer)
  • Lead to changes in cell function by altering DNA/RNA transcription
  • Example:
    • Steroids (nuclear receptor) and milrinone (cytosolic receptor)
    • Specific effects depend on the ligand and the receptor location


Ionotropic receptors

  • Membrane spanning proteins
  • Lead to changes in cell function by allowing flow of ions down a concentration/electrical gradient
  • Examples:
    • GABAA receptor: GABA binds > Cl channel opens > hyperpolarisation > inhibitory post synaptic potential (drug example = benzos)
    • nAChR receptor: acetylcholine binds > non selective cation channel opening - Na/K/Ca > depolarisation > excitatory post synaptic potential (drug example is sux, which is agonist)
    • NMDA receptor: glutamate binds > non selective cations > Na/K/Ca > depolarisation > excitatory post synaptic potential (drug example = ketamine which is an antagonist)


Metabotropic (G protein coupled) receptors

  • Transmembrane proteins with 7 regions

  • Lead to changes in cell function through chemical second messenger systems

    • Activation of the extracellular domain > conformation change in intracellular domain > activation of G proteins > second messenger pathway

  • Three subtypes

    • Gs (stimulatory; increased cAMP)

      • Example: adrenaline (beta-1 receptor) > increased inotropy

    • Gi (inhibitory; decreased cAMP)

      • Example: clonidine (alpha-2 receptor) > decreased SNS outflow

    • Gq (stimulatory; increased IP3)

      • Example: noradrenaline (alpha-1 receptor) > vasoconstriction

Enzyme coupled receptors

  • Transmembrane protein receptor linked to an intracellular receptor
  • Lead to changes in cell function through activation of an intracellular enzyme
  • Example
    • Tyrosine kinase receptor (class II): insulin binds > activates tyrosine kinases on intracellular domain > phosphorylates IRS > cellular cascade


Examiner comments

Overall answers lacked structure and depth, to what is a very fundamental topic. This topic is generally covered within the opening chapters of most physiology texts. Common errors were not answering the question, writing lists rather than describing and explaining, and poor categorisation. Candidates were expected to mention and give example for mechanisms such as hormones binding to cytoplasmic or intra-nuclear receptors, binding to transmembrane receptors coupled to G proteins, cAMP, cGMP, tyrosine kinase, etc.


Online resources



Question 24

Question

Describe the mechanism of action and side effects of 3 classes of drugs that increase uterine tone and 3 classes of drugs that decrease uterine tone.


Answer

INCREASE TONE

Class Oxytocin derivative Ergot derivative Prostaglandin
Example Syntocin Ergometrine Carboprost
MOA Binds to GqPCR in uterus > IP3/DAG pathway > uterine contraction Not fully understood Synthetic PGF2a analogue > binds to PG receptor > myometrial contraction
Effects - Uterine SM contraction

- Weak antidiuretic effect

- Uterine SM contraction - Uterine SM contraction
Side effects IMMUNO: Allergic reactions

CVS: transient hypotension > reflex tachycardia, arrhythmias, flushing
CNS: headache
GIT: nausea, vomiting

CVS: Hypertension

GIT: nausea, vomiting, abdominal pain

CVS: Severe hypertension

RESP: bronchospasm (rare)
GIT: nausea, vomiting, abdominal pain
OTHER: fever


DECREASE TONE

Class Beta agonist CCB NSAIDs
Example Salbutamol Nifedipine Indometacin
MOA Activate B2 receptors (GsPCR), ↑ cAMP > activates protein kinase A > inhibition of MLCK > relaxation Block L-type Ca2+ channels, causing relaxation of SM Inhibit prostaglandin synthesis (via inhibition of COX1/2) > decreased uterine contraction
Effects Decrease uterine tone Decrease uterine tone Decrease uterine tone
Side effects CNS: headacge, hyperactivity

CVS: tachycardia, palpitations
CNS: Anxiety, tremor
RENAL: hypokalaemia
HAEM: lactatemia, hyperglycaemia

CVS: hypotension, flushing, pulmonary oedema

CNS: headache, dizziness
GIT: nausea, vomiting

Mother: gastritis, nausea, vomiting, platelet dysfunction, AKI

Baby: premature closure of ductus arteriosus


Examiner comments

Candidates often appeared to have a sufficient awareness of the choice of drugs (e.g. oxytocin analogues, ergot alkaloids, beta-receptor agonists, calcium channel blockers, etc.), but then failed to produce sufficient depth of knowledge to adequately describe their mechanisms of action in respect to uterine tone. Candidates are reminded that if asked to mention side effects, mentioning side effects of greatest relevance to intensive care (e.g. bronchospasm) in addition to the more generic side effects (e.g. rash).


Online resources




2013 (2nd sitting)

Question 6

Question

Describe the pharmacology of short acting insulin (actrapid).


Answer

Name Short acting insulin (e.g. actrapid)
Class synthetic polypeptide hormone
Indications Diabetes / hyperglycaemia

Hyperkalaemia (in conjunction with dextrose)
B-blocker toxicity (high dose insulin therapy)

Pharmaceutics Clear colourless solutions (generally 100IU/ml)
Routes of administration SC, IV
Dose Variable, titrated to effect (generally bSL)
Pharmacodynamics
MOA The same pharmacodynamic profile of endogenous insulin

> Insulin binds to the alpha subunit of the insulin receptor (tyrosine kinase receptor). Leads to activation of tyrosine kinases on intracellular domain > phosphorylates IRS > cellular cascade

Effects Increased: glucose uptake in cells, glycogenesis, protein synthesis

Decreased: BSL, gluconeogenesis, lipolysis, proteolysis
Cellular shift of potassium (intracellular) due to increased Na/K activity

Side effects Hypoglycaemia (excessive dosing) --> decreased LOC, seizures, death

Hyperglycaemia / DKA (inadequate dosing)
Hypokalaemia --> arrhythmias
Weight gain (long term)

Pharmacokinetics
Profile (SC admin) Onset: 15-30 mins

Peak: 1-2 hours
Duration: 6-8 hours

Absorption No oral absorption (inactivated by GIT enzymes)

SC administration is close to 100%

Distribution Protein binding <10%

VOD = < 0.1 L/Kg

Metabolism Hepatic proteases
Elimination Renal elimination of inactive metabolites

T 1/2B = 90 mins

Monitoring BSL levels (frequency depends on indication, glycaemic stability, route etc)


Examiner comments

In general candidates lacked a sufficient depth of knowledge for this commonly used drug. Some candidates confused actrapid with novo rapid. A structured approach (e.g. pharmaceutics, mode of action, pharmacokinetics, etc.) was expected.


Online resources



2012 (2nd sitting)

Question 5

Question

How does liver failure affect the pharmacology of drugs?


Example answer

Absorption

  • Drugs which are absorbed orally are subject to first pass metabolism by the liver

  • Liver failure > decreased first pass metabolism > increased bioavailability > potential for toxicity

  • oedema (from liver failure) > impair subcut absorption

Distribution

  • The liver is responsible for producing majority of the proteins that drugs bind to in plasma
  • Therefore liver failure > decreased plasma proteins
  • Highly protein bound drugs (e.g. warfarin) are greatly affected by reduced plasma proteins
    • Small decrease in plasma protein levels > large change in the proportion of unbound (active) drug


Metabolism

  • The liver is a primary organ of drug metabolism and biotransformation
    • Phase 1 reactions by liver
      • Hydrolysis, Reduction, Oxidation
      • Small increase in hydrophilicity
    • Phase 2 reactions
      • Glucuronidation, sulfation, conjugation, methylation
      • Significantly increased hydrophilicity (for renal excretion)
  • Liver damage > impaired metabolism / biotransformation > accumulation > toxicity (e.g. diazepam in liver failure)
  • Portal hypertension > shunting of blood > decreased first pass metabolism > accumulation


Elimination

  • Liver failure > decreased hepatic blood flow > decreased elimination of drugs with high hepatic extraction ratio
  • Liver failure > decreased elimination of drugs with low hepatic extraction ratio (regardless of HBF)
  • Liver failure > decreased plasma proteins > increased unbound fraction drug > increased renal elimination of hydrophilic drugs
  • Liver failure > decreased elimination on lipophilic drugs excreted in biliary system


Examiner comments

59% of candidates passed this question.

Good answers were structured using pharmacokinetic and pharmacodynamics headings.
They included some mention of changes in absorption, volume of distribution (an increase
in Vd in liver failure), altered protein binding, altered metabolism and thus change in
clearance, and changes in excretion (decreased biliary excretion of drugs). In respect to
pharmacodynamics candidates could have mentioned increased sensitivity and prolonged
action of sedative drugs, oral anticoagulants, etc. Good candidates also differentiated for
acute (often hepatocellular dysfunction) and chronic liver failure (cirrhosis and changes in
liver blood flow). Common problems were not using a logical structure to answer the
question and stating an effect but not describing how this affected pharmacology. For
example stating decreased albumin production but then not stating the consequence of this
on drug distribution. Primary examination questions may often require candidates to
integrate knowledge from across different sections of the syllabusor apply basic
physiological or pharmacological principles.


Question 9

Question

Classify the anti-arrhythmic drugs using the Vaughan-Williams classification (30% of marks). Compare and contrast the electrophysiological effects of Class 1 anti-arrhythmics (70% marks).


Answer

Class Ia Ib Ic II III IV
Mechanism Blocks Na channels Blocks Na channels Blocks Na channels \beta-adrenergic blockade Blocks K+ channels Blocks Ca channels
Example Procainamide Lidocaine Flecainide Propranolol Esmolol, Atenolol Sotalol Amiodarone (also I,II,IV effects) Sotalol Verapamil Diltiazem
Effects on
Phase 0 ↓ - ↓ - - -
Conduction velocity ↓ - ↓ ↓ ↓ -
ERP ↑ ↓ ↑ ↓ ↑ -
APD ↑ ↓ - ↑ ↑ ↓
QRS duration ↑ - ↑ - ↑ -
QTc ↑ ↓ ↑ ↓ ↑ -
Drugs not included
  • Digoxin
  • Adenosine
  • Magnesium


Examiner comments

Most candidates displayed a basic knowledge of the Vaughan-Williams classification and gave an example of each class. The remainder of the question lent itself very well to a tabular format. Better answers included the effect on the action potential (diagrams were useful here), channel dissociation kinetics (this was frequently omitted) and examples from each class of drug. There is an excellent table in Stoelting which answers this question nicely. Marks were not awarded for clinical effects. Overall, this question was generally well answered.



Question 13

Question

Describe the effects of obesity on drug pharmacology (70% of marks). Give examples of those drugs that illustrate those effects (30% of marks)


Example answer

Obesity

  • BMI > 30
  • Alters all aspects of pharmacology to varying degrees


PHARMACOKINETICS


Absorption

  • Increased gastric emptying > increased absorption
  • Decreased subcutaneous blood flow (increased adiposity, no increase in vascularity) > slow rate of SC absorption
  • Difficulty with IM administration due to tissue (may lead to inadvertent SC injection)
  • Increased CO > delayed onset of inhalation anaesthetics


Distribution

  • Increased body adiposity > Increased volume of distribution of lipid soluble drugs (e.g. benzodiazipines, thiopentone)
  • Small (relative to body fat) increased Vd for hydrophilic drugs e.g. gentamicin (due to increased blood volume, total body water).
  • Generally lipid soluble drugs dosed on actual body weight, hydrophilic drugs on ideal body weight


Metabolism

  • Increased hepatic blood flow (due to increased CO) = increased clearance of high extraction ratio drugs (flow dependant extraction) e.g. propofol
  • Decreased hepatic blood flow (due to dysfunction, fatty infiltration) > decreased hepatic extraction and metabolism
  • Increased activity of plasma and tissue esterases > increased metabolism/clearance of drugs using these systems (e.g. remifentanil)
  • Increased pseudocholinesterase levels in obesity (sux to be doses on total body weight)


Elimination

  • Increased GFR (due to increased CO) – increased renal clearance of hydrophilic drugs (e.g vancomycin)
  • Decreased GFR due to diabetic nephropathy = decrease renal clearance
  • Due to distribution, lipid soluble drugs may have increased elimination half life


PHARMACODYNAMICS

  • Receptor resistance (e.g. insulin resistance in obesity)


Examiner comments

36 % of candidates passed this question.

This question could be approached by describing the effects of obesity on drug distribution,
binding and elimination. Candidates that took this approach generally did better than those
with a less structured approach. With obesity, fat body mass increases relative to the
increase in lean body mass leading to an increased volume of distribution particularly for
highly lipid soluble drugs, e.g. midazolam. However, the dosing of non-lipid soluble drugs,
e.g. non-depolarising muscle relaxants, should be based on ideal body weight. An increase in
blood volume and cardiac output associated with obesity may require an increased loading
dose to achieve a therapeutic effect, e.g. thiopentone. Plasma protein binding of drugs may
be decreased due to an increased binding of lipids to plasma proteins, resulting in an
increased free fraction of drug. A reduction in plasma protein concentration due to an
increase in acute phase proteins may also result in decreased plasma protein drug binding
and increased free fraction of drug. Pseudocholinesterase levels are increased in obesity and
therefore the dose of suxamethonium should be based on total body weight. Plasma and
tissue esterase levels are increased resulting in the increased clearance of drugs by these
enzymes e.g. remifentanil. Hepatic clearance is usually normal but may be impaired in liver
disease caused by obesity. Renal clearance is usually increased due to increased body
weight, increased renal blood flow and increased glomerular filtration rate. Renal clearance
may be impaired in renal disease caused by obesity related diseases, e.g. diabetes. Insulin
doses may be increased due to peripheral insulin resistance in type 2 diabetes caused by
obesity. Most answers were deficient in examples of drugs to illustrate the effects of obesity
on drug pharmacology.



Question 20

Question

What are drug enantiomers? (20% of marks). Explain the clinical relevance of enantiomers (60% marks). Give clinically relevant example (20% of marks)


Example answer

Enantiomers

  • A stereoisomer which has identical chemical formula and bond structure, but the relative positions of the functional groups in 3D space differ such that the molecules are not superimposable (they form mirror images of each other)
  • Named according to their absolute configurations in 3D space
    • R (rectus) atomic numbers descend clockwise
    • S (sinister) atomic numbers descend anticlockwise


Example

  • Ketamine is typically presented in a racemic mixture
  • However, R- is less effective and has a higher incidence of adverse effects compared to S+ ketamine enantiomer.


Relevance

  • Enantiomers, due to their different configurations in space, interact differently with receptors, transport proteins and enzymes > differing pharmacokinetics and dynamics
  • Pharmaceutics
    • Enantiopure preparations are more expensive (hence racemic mixtures more common)
  • Pharmacodynamics
    • Will interact with receptors differently > differing degrees of agonism/antagonism + also compete for receptors > variable effects (R-ibuprofen 100X more portent inhibitor COX than S ibuprofen)
    • Enantiopure preparations are more likely to include the most active or least toxic isomer (e.g. s-ketamine)
  • Pharmacokinetics
    • Absorption
      • No change in passive absorption
      • Active transport may favour one enantiomer over another (e.g methotrexate)
    • Distribution
      • Stereoselectivity in protein binding which will also affect the Volume of distribution and proportion of active drug (e.g. propranolol)
    • Metabolism
      • Stereoselectivity in metabolism due to varying degrees of interaction with enzymes (e.g. warfarin and CYP450)
    • Elimination
      • Stereoselectivity in elimination (e.g. ibuprofen)


Examiner comments

41% passed

Enantiomers refer to isomeric molecules with centres of asymmetry in 3 dimensions that are mirror images of each other but not superimposable. Enantiomers may be distinguished by the direction in which polarised light is rotated. Interactions involving weak drug-receptor bonds feature a dependence upon recognition of shape, i.e. stereochemical structure is often important. Frequently one enantiomer may bind to a given receptor more avidly than the other, thus pharmacodynamics, pharmacokinetics and toxicity may vary between enantiomers. Many drugs are supplied as racemic mixtures, the components of which have different activity. Clinically relevant examples that candidates could have mentioned, included bupivacaine, ropivacaine, ketamine and carvedilol.



2011 (1st sitting)

Question 14

Question

Describe the mechanisms of action and adverse effects of pulmonary vasodilators that are administered via the inhalational route.


Answer

Oxygen

  • A pulmonary vasodilator in hypoxic patients (reverses hypoxic pulmonary vasoconstriction)
  • MOA
    • Initial phase HPVC: Decreased O2 (hypoxia) > altered redox state in mitochondria of SM muscles > inhibition of K channels > depolarisation > Ca influx > vasoconstriction
    • Prolonged hypoxia: maintains HPVC through decreased NO release +release of endothelin derived vasoconstrictors
    • O2 therefore reverses this process of HPVC
  • Effects
    • RESP: improved oxygen saturations (may also improve DO2), decreased respiratory drive (minor), pulmonary toxicity (free radical generation), may worsen V/Q mismatch, absorption atelectasis, hypercapnoea (in chronic CO2 retainers)
    • CVS: decreased pulmonary vascular resistance (vasodilation) due to reversal of HPV, increased HR/SV/SVR in setting of hypoxia (via chemoreceptor reflex) > increased CO and BP, coronary vasoconstriction
    • CNS: anxiety, nausea, seizures (hyperbaric hyperoxia)
    • MET: oxidative phosphorylation > ATP production


Nitric oxide

  • Class:
    • Pulmonary vasodilator / Inorganic gas
  • MOA:
    • Binds to guanyl cyclase > Increases cGMP > reduction in intracellular Ca > relaxation of SM.
    • As inhaled > selectively vasodilates in regions of well ventilated alveoli
  • Effects
    • RESP: pulmonary artery vasodilation > improves V/Q matching > dec. WOB
    • CVS: decreased pulmonary VR, decreased mPAP, decreased RHS, hypotension, rebound pHTN following cessation
    • CNS: Increased CBF
    • HAEM: thrombocytopaenia, methemoglobinemia


Prostacyclin

  • Class
    • Pulmonary vasodilator / prostacyclin analogue
  • MOA:
    • Binds to prostacyclin receptor (IP receptor) > Activates GPCR > increases cAMP > decreased platelet activation and increased SM relaxation.
    • If given inhaled > local effects in regions of well ventilated alveoli only
  • Effects
    • RESP: pulmonary arterial vasodilation > improve V/Q matching + oxygenation in patients with ARDS if inhaled (goes to ventilated regions only), but may worsen it if given intravenously (goes to all pulmonary blood vessels > worsening shunt)
    • HAEM: inhibition of platelet aggregation > increased risk bleeding
    • CVS: flushing, hypotension, reflex tachycardia, decreased pulmonary vascular resistance and mPAP > decreased RV afterload (may improve CO in RHF)
    • CNS: headache, increased CBF


Examiner comments

Many candidates neglected to include oxygen which is also a drug with significant pulmonary vasodilating properties. Accurate detail concerning the receptor and second messenger effects of drugs was expected. The importance of V/Q matching and reduction in systemic effects via inhalational administration needed to be stated. Better answers included discussion of serious adverse effects such as methaemoglobinaemia, acute lung injury, systemic hypotension, rebound phenomena and heart failure.


Online resources




2010 (2nd sitting)

Question 5

Question

List the antiplatelet drugs and outline their mechanism of action, adverse effects, mode of elimination and duration of action


Answer

Class Example Mechanism of action Elimination Reversibility Duration of antiplatelet effect Adverse effects
COX inhibitors Aspirin Inhibits COX on platelets > ↓ thromboxane A2 > ↓ platelet aggregation and activation Renal (100%) Irreversible inhibition Life of platelet (~7 days) -Haemorrhage

- GIT ulcers
- Allergy, angioedema, bronchospasm
- AKI

ADP receptor antagonists Clopidogrel Binds to P2Y12 subtype of the ADP receptor on platelets > ↓ GP IIb/IIIa activation > ↓ platelet activation Renal (50%) Faecal (50%) Irreversible inhibition Life of platelet (~7 days) - Haemorrhage

- Non responder (CYP2C19 polymorphism)
-CYP450 drug interactions
- aplastic anaemia, thrombocytopaenia, anaemia,
- GIT ulcers
- Rash, urticaria, angioedema, TTP

Prasugrel As above Renal (70%)

Faecal (30%)

Irreversible inhibition Life of platelet (~7 days) - Haemorrhage

- Rash, urticaria, angioedema, TTP

Ticagrelor As above (but binds to a different binding site) Faecal (70%)

Renal (30%)

Reversible inhibition 2-3 days - Haemorrhage

- Dyspnoea
- Rash, urticaria, angioedema, TTP

GP IIb/IIIa receptor antagonists Abciximab Directly bind to GP IIb/IIIa and block the final common pathway of platelet aggregation Renal Reversible inhibition 1-2 days - Haemorrhage

- ↓ PLTs

Tirofiban As above Renal (70%)

Faecal (30%)

Reversible inhibition 4-6 hours - Haemorrhage

- ↓ PLTs, TTP
- Allergy

Phospho-diesterase inhibitors Dipyridamole Inhibits platelet adhesion to walls (by inhibiting adenosine uptake). Also inhibits phosphodiesterase activity > increased cAMP > decreased calcium > inhibition of platelet aggregation Faecal Reversible inhibition 1-2 days - Haemorrhage

- Hypotension
- GIT upset (nausea, vomiting, diarrhoea)
-Rash, urticaria

Prostacyclins Epoprostenol Binds to IP receptors > increased cAMP > ↓ calcium > ↓ platelet aggregation Renal (70%), Faecal (15%) Reversible inhibition < 5 mins - Hypotension, headache, flushing, Haemorrhage
File:Https://www.mja.com.au/sites/default/files/issues/178 11 020603/han10033 fm-1.gif

Examiner comments

67% of candidates passed this question

Most candidates did reasonably well by including aspirin, ADP receptor blockade and glycoprotein 2b/3a blockade in their answers. The best approach to answer this type of question was to use a table with each anti-platelet agent within a column and headings for the rows such as mechanisms of action, adverse effects, mode of elimination and duration of action.
Common omissions included the irreversibility of the blockade of the platelet function by many of these agents, renal toxicity and bronchospasm as side effects of aspirin, bone marrow toxicity of ADP receptor blockers, and dipyridamole as an anti-platelet agent. Some candidates classified clopidogrel as a glycoprotein 2b/3a blocker incorrectly and thought clopidogrel has a relative short duration of action on platelet function because of its half-life. Clopidogrel as a prodrug requiring activation by cytochrome P450 and hence significant potential drug interactions were not mentioned by any candidates.


Online resources




Question 20

Question

Outline the pharmacokinetic consequences of old age. Illustrate your answer with examples


Example answer

Absorption

  • Decreased cutaneous blood flow > slower/reduced absorption of transdermal (GTN patch) and subcut routes (e.g. heparin)
  • Decreased intestinal absorptive capacity with age > decreased PO absorption (e.g. digoxin)
  • Decreased gastric emptying rate > decreased PO absorption (e.g. digoxin)
  • Decreased acid secretion > increased pH gastric > decreased absorption strong acids (e.g. amoxicillin)


Distribution

  • Decreased TBW > decreased Vd of hydrophilic drugs > increased effect (e.g. ethanol, gentamicin)
  • Increased fat / decreased muscle mass > increased Vd of lipophilic drugs > prolonged effect (e.g. amiodarone, diazepam)
  • Decreased plasma proteins (e.g. albumin) > increased unbound (active) drug > increased redistribution and potency (e.g. phenytoin, warfarin)
  • Reduced CO > altered redistribution


Metabolism

  • Decreased portal blood flow = increased oral bioavailability (e.g. labetalol)
  • Decreased hepatic blood flow = decreased clearance (e.g. morphine) and phase 1 metabolism (e.g. ibuprofen)
  • Decreased hepatic tissue mass = decreased Phase 1 reaction (e.g. ibuprofen)


Elimination

  • Decreased renal mass / nephrons with age > decreased GFR > reduced clearance > prolonged effects (e.g. vancomycin) or increased toxicity (e.g. gentamicin)
  • Decreased hepatic blood flow / tissue mass > impaired liver/GIT clearance of drugs (e.g. morphine)


Examiner comments

53% of candidates passed this question.

As the general population ages, and many elderly are admitted to intensive care units and/or
encountered during intensive care ward consultations, this topic is highly relevant. Unfortunately
candidate performance generally lacked sufficient depth and breadth in this area. Good answers
were expected to mention changes in body compartments (eg total body water, lean body mass
decrease, etc), consequences of changes in organ function (eg deteriorating glomerular filtration
rate, reduced liver blood flow, etc), alterations in protein levels and binding, increased likelihood of
drug interactions and the influence of disease states.


Question 24

Question

Classify anti-hypertensive agents by their mechanism of action with a brief outline of each mechanism and an example of a drug in each class.


Answer

Sympatholytic's

Class Example MOA
Alpha blockers Prazosin <math display="inline">\alpha</math>1 antagonist > arterial and venous vasodilation > ↓ SVR > ↓ BP
Beta blockers Metoprolol <math display="inline">\beta</math>1 antagonist > ↓ inotropy and ↓ chronotropy > ↓ BP
Centrally acting Clonidine Central <math display="inline">\alpha</math>2 agonist > ↓ SNS tone (via ↓ NA release) > ↓ BP


RAAS inhibitors

Class Example MOA
ACE inhibitors Ramipril Block the conversion of angiotensin I to angiotensin II by ACE > decreased AG2 > ↓ SVR and ↑ natriuresis > ↓ BP
ARBs Candesartan Same as ACEI (above) but blocks AG2 directly.


Calcium channel blockers

Class Example MOA
Dihydropyridine Amlodipine Blocks L-Type calcium channels in SM > ↓ intracellular Ca > vasodilation > ↓ SVR > ↓ BP
Non-dihydropyridine Verapamil Same as dihydropyridines, but additionally preferentially acts on cardiac cells > ↓ HR and ↓ contractility > ↓BP


Diuretics

Class Example MOA
Loop diuretic Frusemide Blocks to NK2Cl transporter in the aLOH> ↓ Na,K, Cl reabsorption > ↓ medullary tonicity + ↑ Na/Cl delivery to distal tubules > diuresis > ↓ BP. N.B Direct vasodilation effect - MOA unclear
Thiazide diuretic Hydrochlorothiazide Blocks Na/Cl cotransporter in DCT > ↓ Na+ and Cl- reabsorption > diuresis > ↓ BP
Potassium sparing diuretic Spironolactone Competitive aldosterone antagonist > ↓ Na reabsorption in DCT > diuresis > ↓ BP


Vasodilators

Class Example MOA
Nitrates GTN Dinitrated to NO > diffuses into SM > binds to guanylyl cyclase > ↑ GMP > ↓ intracellular Ca > vasodilation > ↓ BP
Hydralazine Hydralazine Not fully understood. Though to also activate guanylyl cyclase > ↑ GMP > ↓ intracellular Ca > arteriolar vasodilation > ↓ BP



Examiner comments

67% of candidates passed this question.

There are many valid lists that can be used as a template to answer this question. One such list might broadly classify antihypertensive agents into sympatholytic agents, vasodilators, calcium channel antagonists, renin-angiotensin inhibitors and diuretics. Within each of these categories are a variable number of sub classes, for example diuretics might include thiazides, loop diuretics and potassium sparing diuretics. A good answer would include such a listing with a brief description of the mechanism of action with respect to the antihypertensive effect and the name of a typical drug that acts in the manner described. Most candidates were able to generate such a list and populate it as required by the question, thus being rewarded with good marks. Poorer answers lacked any logical classification system and were merely a random list of antihypertensive drugs and their actions. Candidates are reminded that organisation within an answer helps in answering the question and achieving marks.


Online resources




2009 (2nd sitting)

Question 5

Question

Outline the kinetic characteristics and the mode of action of digoxin (75% marks). List the cardiovascular effects of digoxin (25% marks).


Answer

Pharmacokinetics

  • Onset/duration
    • Onset; 2-3 hours (PO), 10-30mins (IV)
    • Duration of action: 3-4 days
  • Absorption
    • Well absorbed from GIT
    • 80% oral bioavailability
  • Distribution
    • Protein binding ~25%
    • VOD 6-7L/kg
    • High lipid solubility
  • Metabolism
    • Minimal hepatic metabolism (15%)
      • Oxidation and conjugation
      • Active and inactive metabolites
  • Elimination
    • Renal elimination (70% unchanged)
    • Small amounts of faecal/biliary elimination <15%
    • T 1/2 = 48 hours
    • Not readily dialysable


Cardiovascular effects

  • Positive inotropy
    • Inhibits Na/K ATPase > Increased Na > impairs Na/Ca exchanger > increased intracellular Ca > increased inotropy > increased CO
  • Negative chronotropy and dromotropy
    • Increased PSNS release of ACh at M receptors > decreases SA node firing (chronotropy) + prolongs AV conduction (dromotropy) > increased diastolic filling time > increased preload > increased SV > increased CO + BP
    • Can also lead to bradycardia, AV block, bradyarrhythmia's
  • Increased excitability
    • Increases slope of phase 4 > enhances automaticity of atrial, junctional, ventricular tissue > arrhythmias
      • Not nodal tissue (due to vagal effects)
  • ECG changes
    • Shortens phase 2 > shortened QT interval
    • AV nodal inhibition > prolonged PR interval
    • Shortened Phase 2 > repolarisation abnormalities (scooped ST, TWI)


Examiner comments

0 (0%) of candidates passed this question.

The Syllabus for the Primary examination describes an outline to be “Provide a summary of the important points.” Thus candidates were expected to briefly mention the fundamental pharmacokinetic characteristics (eg highly lipid soluble, well absorbed from small intestine, oral bioavailability of 60 - 90%, protein binding of 20 - 30%, volume of distribution, half life, etc) and mode of action. This was poorly done and candidates’ answers often lacked structure. The question outlines the distribution of marks, being 25% for listing cardiovascular effects. Thus candidates were expected to broadly list the important cardiovascular effects relating to mechanical (eg increase intensity of myocardial contraction, direct venous and arteriolar constriction, etc) and electrical ( increase phase 4 slope & automaticity, hyperpolarization, shortening of atrial action potentials, decrease AV conduction velocity and prolong AV refractory period, increase PR & QT intervals, dose and baseline autonomic activity dependent actions, etc).



Question 17

Question

Explain the difference and clinical relevance between zero and first order kinetics (60% marks). Give an example that is relevant to intensive care practice (40% marks)


Example answer

First order kinetics

  • A constant proportion of a drug is eliminated per unit time
  • Enzyme/elimination systems are working below their maximum capacity
  • Therefore, elimination is proportional to drug concentration
    • Increasing concentration of drug will increase elimination of drug
    • Exponential concentration per time graph
  • Most drugs eliminated in this way


Zero order kinetics

  • A constant amount of drug is eliminated per unit time
  • Enzyme/elimination systems are saturated/working at maximal capacity
    • Increasing concentrations will not lead to increase in elimination
    • Linear concentration vs time graph
  • The transition from first order kinetics to zero order kinetics is described in the Michalis-Menten equation
  • Only some drugs are eliminated this way
    • Example: phenytoin, ethanol, salicylates
  • Because of this, increasing concentrations > increased risk of toxicity
  • Phenytoin reaches the therapeutic range at the point at which it transitions from first to zero-order kinetics > very narrow therapeutic range > requires monitoring / dosage adjustment


Examiner comments



2008 (1st sitting)

Question

Question

Describe the role of the kidney in drug excretion and the factors affecting this. Briefly outline how you would alter the dosing of gentamicin in a patient with renal impairment


Example answer

Renal excretion/elimination

  • Principle mechanism of drug elimination

  • Renal elimination is a balance between glomerular filtration, tubular secretion and reabsorption.

Factors affecting glomerular filtration

  • GFR
    • Increased GFR = increased filtration = increased clearance of hydrophilic drugs
  • Drug size:
    • Increasing drug size = decreased renal clearance
    • Only drugs <7kDa (weight) or <30 Angstrom units (width) are able to pass the capillary BM
  • Protein binding
    • Only unbound drugs can pass the glomerular BM
      • Highly protein bound drugs are poorly filtered
  • Charge
    • Negatively charged molecules cannot readily pass BM (as it is also negatively charged)


Factors affecting drug secretion

  • Protein binding and renal blood flow as per above
  • Concentration: Increased concentration = increased secretion (until tubular transporters are saturated)
  • Multiple substrates competing for the same transporters


Factors affecting drug reabsorption

  • Can be active or passive (most are passive)
  • Affected by charge (ionised drugs cannot pass through BM) and become trapped in the urine
  • Concentration (as passive diffusion depends on concentration gradient)
  • Lipophilicity - Lipophilic drugs are often reabsorbed


Gentamicin

  • Basic pharm overview
    • Bactericidal aminoglycoside, demonstrates concentration dependant activity
    • Small volume of distribution (0.3L/kg), minimal protein binding (15%), not metabolised
    • Renally excreted (GFR limited) unchanged with a normal T 1/2 of 3 hours
    • Narrow therapeutic index
  • Adjustments
    • Loading dose
      • Loading dose is the same (though some antibiotic guidelines will recommend lower end-normal if reduced GFR)
    • Ongoing therapy (if needed)
      • If CrCl <40 strongly consider ongoing need
      • If ongoing need - stretch interval due to reduced renal clearance
      • Consider plasma concentration monitoring if therapy > 48 hours needed


Examiner comments



2008 (2nd sitting)

Question 8

Question

Compare and contrast the pharmacology of sodium nitroprusside and glyceryl trinitrate


Answer

Name Sodium nitroprusside Glyceryl trinitrate
Class Nitrate vasodilator Organic nitrate
Indications Hypertensive emergencies (or need for strict BP control) Hypertension, acute pulmonary oedema, angina, ACS/LV failure,
Pharmaceutics IV solution (50mg/2mL)

Light sensitive

Clear liquid (IV), Patch (transdermal), tablet (SL), spray (SL)
Routes of administration IV only (non PVC giving sets) Sublingual, intravenous, transdermal
Dose Titrated to effect (0-2mcg/kg/min) Patch: 5-21 mcg/hr

SL: 400mcg PRN
IV: titrated to effect

pKA 3.3 5.6
Pharmacodynamics
MOA Prodrug

- Diffuses into RBCs and reacts with Oxy-Hb to produce NO
- NO diffuses into cell > incr cGMP > decreased Ca > SM relaxation

Prodrug

- Dinitrated to produce active nitric oxide (NO).
- NO diffuses into smooth muscle cell > binds to guanylyl cyclase > increased cGMP > decreased intracellular Ca > SM relaxation > vasodilation

Effects CVS: Arterial+venous vasodilation > decreased BP + afterload

RESP: impairs HPVC
CNS: cerebral vasodilation
GI: ileus
Metabolic: acidosis

CVS: systemic vasodilation (preferentially venodilation) > decreased VR > decreased stretch > decreased O2 consumption, coronary arterial dilation

CNS: Increased CBF > inc ICP
RESP: Bronchodilation, decreased PVR

Side effects headache, hypotension, rebound hypertension (abrupt withdrawal), cyanide toxicity (high doses), metabolic acidosis, hypoxia, raised ICP CVS: reflex tachycardia, hypotension

CNS: Headache, increased ICP
Derm: flushing

Pharmacokinetics
Onset/offset Immediate onset + offset 1-3 mins (SL), <1 min (IV), Patch variable.
Absorption 0% oral bioavailability Oral bioavailability 3%

(hepatic - high first pass effect)

Distribution VOD 0.25L/Kg (confined intravasc).

Nil protein binding

60% protein bound.

Vd 3L/kg

Metabolism Nitroprusside > cyanide > prussic acid > thiocyanate

site: RBC (and liver secondarily)

Hydrolysis into inactive compounds

Site: liver + RBC cell wall + vascular cell walls.

Elimination Metabolites via urine (major)

T 1/2 = 3 mins

80% urine.

T 1/2 = 5 minutes.

Special points Can develop tachyphylaxis (depletion of sulfhydryl groups)


Examiner comments

80% of candidates passed this question

It was expected candidates would address specific aspects of pharmacology such as action, mechanism of action, half life and duration of effect, route of administration, potential toxicity and special precautions. These agents lend themselves to comparison and contrast as several distinct similarities and differences exist and credit was given for highlighting these. Specific comments should include that both agents result in blood vessel dilation with extra credit given for detailing the differences in the balance of arterial versus venous effects between them. For both agents the effect is mediated through nitric oxide and it was expected candidates would identify that nitroprusside releases NO spontaneously and GTN requires enzymatic degradation with the resultant effects on smooth muscle mediated via c GMP. They are both short acting agents when used intravenously and require careful titration to measured blood pressure for effect. Extra credit was given for mentioning that routes other than IV are available for GTN (topical / oral) but not for nitroprusside. Comments on special precautions such as Nitroprusside should be protected from light and GTN given via non PVC giving sets gained additional marks. In addition to the well described adverse effects of each agent, it was expected candidates would mention the potential for cyanide toxicity with nitroprusside and extra marks were awarded for an indication of usual doses.




2007 (2nd sitting)

Question 2

Question

Outline the sites and mechanisms of action of diuretics. Give one example of drug acting at each site and list two side effects of each drug.


Example Answer

Site of action Example Mechanism of diuresis Side effects
Entire Mannitol Freely filtered at glomerulus (but not reabsorbed). Acts osmotically to ↓ H2O reabsorption - ↓ Na, K, Cl

- Hypotension, hypovolaemia

PCT Acetazolamide Inhibits carbonic anhydrase in PCT > ↓ reabsorption of filtered HCO3 + Na > ↑ tubular osmolality > diuresis - Metabolic acidosis (↓ HCO3)

- ↓ Na, K, Cl

LOH Frusemide Binds to NK2Cl transporter in the thick ascending limb LOH > ↓ Na,K, Cl reabsorption > impairs counter current multiplier + ↓ medullary tonicity - ↓ Na, K, Cl

- Metabolic alkalosis (↓ K, Cl)
- Hypovolaemia, Hypotension

DCT HCT Inhibit Na+ and Cl- reabsorption (Na/Cl cotransporter) > ↓ H2O reabsorption - ↓ K, Na, Cl

- ↑ BSL, lipids
- Metabolic alkalosis

CD Spironolactone Competitive aldosterone antagonist > inhibition of ENaC > ↓ Na reabsorption (and ↓K excretion) > diuresis -↑ K and metabolic acidosis

- Anti-androgen effects (decreased libido, menstrual irregularities, gynecomastica)

CD Amiloride Blocks ENaC > ↓ Na/Water reabsorption - HypoNa (blocked ENaC)

- HyperK (ENaC drives ROMK channels)


Examiner comments

Good answers to this question were those that had a tabular format to the structure of the answer — for example columns headed mechanism, sites, drug and side effects. Most common omissions were not to further describe how the different mechanisms of action of diuretics increased urine output, e.g. "disruption of the counter current multiplier system by decreasing absorption of ions from the loop of Henle into the medullary interstitium, thereby decreasing the osmolarity of the medullary interstitial fluid". There was often little mention of increased urine solutes and the effect the electro chemical effect had in promoting a diuresis. Examples of drugs were well done


Online resources




Misc / not previously examined

Question a

Question

Outline with examples the role of excipients in drug formulations.


Example answer

Excipient

  • Components of a drug preparation that do not exert the pharmacological effect
  • Function: assists with optimal delivery of the active ingredient
  • Ideally: nontoxic, inactive, and don’t interact with active ingredient


Preservatives

  • Prevent/inhibit growth of microorganisms in the drug preparation
  • Generally weak acids (pKa 4-5)
  • Example: benzyl alcohol


Antioxidants

  • Prevent/limit the degree of chemical breakdown due to oxidative reactions
  • Example: ascorbic acid


Solvents

  • A liquid (usually) substance which can dissolve another substance
  • Water is the most common solvent (most drugs are water soluble to an acceptable degree)
  • For non-water-soluble drugs, or drugs unstable in water, non-aqueous solvents are used (e.g. mannitol, propylene glycol)


Buffer

  • A solution consisting of a weak acid and its conjugate base
  • Maintains the pH of a drug preparation to maximise stability and/or maintain solubility
  • Example: acetic acid / sodium acetate


Emulsifying agents

  • Substances that stabilise emulsions which are typically unstable
  • Example: soya bean oil / egg lethicin in propofol


Diluents

  • Provide bulk and enable accurate dosing of potent ingredients
  • E.g. glucose, lactose


Binders

  • Bind tablet ingredients together for form/strength
  • Example: starches, sugars


Flavours

  • Added to increase compliance / ease of use
  • Example: aspartame


Colouring

  • Added for marketing purposes / compliance
  • E.g. beta caroteine


Coatings/film

  • Designed to make tablets easier to swallow, improve predictability of absorption, protect from environment e.g moisture
  • Example: cellulose for enteric coating to delay release of agent


Examiner comments

Not previously examined



Question b

Question

Describe the mechanism of action and effects of corticosteroid drugs with particular reference to asthma


Answer

Asthma

  • Asthma is an inflammatory condition of the airways characterised by airway narrowing, mucous secretion and expiratory airflow limitation.


Corticosteroids

  • Steroid hormones normally produced by the adrenal cortex
  • Two main classes of corticosteroids
    • Glucocorticoids (secreted from zona fasciculata and reticularis)
    • Mineralocorticoids (secreted from zona glomerulosa)
  • Glucocorticoids are used in the treatment of asthma
    • Systemic: Prednisone, Hydrocortisone
    • Inhaled: Beclomethasone, Budesonide
  • Note: prednisone and hydrocortisone also have some mineralocorticoid effect


Glucocorticoids

  • Mechanism of action
    • Lipid soluble hormone > crosses cell membrane > binds to intracellular steroid receptors > translocate to nucleus > alters gene transcription > metabolic, anti-inflammatory & immunosuppressive effects in tissue-specific manner
    • The anti-inflammatory process is mediated by suppression of phospholipase A2 > decreased arachidonic acid > decreased PGs, TXA2, Leukotrienes
    • Inhaled steroids (e.g. budesonide) at regular dosage tend to have only respiratory (local) effects, whereas systemic glucocorticoids (e.g. hydrocortisone, prednisone) exhibits effects at all sites.
  • Effects on asthma
    • RESP
      • Reduced airway oedema > bronchodilation
      • Increased SM responsiveness to catecholamines and B2 agonists > bronchodilation
      • Decreased mucous secretion > reduced mucous plugging
      • Bronchodilation + decreased mucous secretion > improved ventilation + oxygenation > decreased work of breathing
    • CVS
      • Increased BP (Due to mineralocorticoid effect on kidneys (increased H2O reabsorption) and increased alpha adrenergic responsiveness to endogenous catecholamines)
  • Other effects of systemic steroids
    • RENAL
      • Increased fluid reabsorption (Due to mineralocorticoid effect on kidneys > increased Na/H20 reabsorption in DCT > increased BP, oedema)
    • Metabolic
      • Hyperglycaemia (Due to increased gluconeogenesis, protein catabolism, lipolysis)
      • Adrenal suppression (Due to negative feedback on the pituitary (inhibits ACTH) and hypothalamus (inhibits CRH))
    • CNS
      • Sleep disturbance, mood changes, psychosis
    • IMMUNE
      • Immunosuppression (particularly mast cells, eosinophils, T cells) > decreased cytokines/pro inflammatory mediators
    • GIT
      • GIT ulceration (inhibition of COX systems)
    • MSK
      • Skin thinning and muscle wasting (due to increased protein/fat catabolism)
      • Osteoporosis



Question c

Question

Pharmacology of aminophylline


Answer

Name Aminophylline (and Theophylline)
Class Methylxanthine derivative
Indications Severe airway obstruction, including acute asthma (less commonly used nowadays)
Pharmaceutics (aminophylline) Complex of 80% theophylline (active component) and 20% ethylenediamine (improves solubility, no effect). Concentration of 25mg/ml in 10ml vials
Routes of administration IV (aminophylline) , PO (aminophylline and theophylline)
Dose (Aminophylline) Loading = 5mg/kg (slow injection)

Maintenance = 0.5mg.kg.hr

Pharmacodynamics
MOA - Non selective phosphodiesterase inhibitors > increased cAMP > decreased Calcium > SM + bronchial relaxation

- Also block adenosine receptors > decreased inflammatory response

Effects Narrow therapeutic window

RESP: Bronchodilation (via SM relaxation), increased respiratory centre sensitivity to CO2, improved diaphragm contractility
CNS: headache, irritability, tremor, seizures
CVS: palpitations, tachycardia, arrhythmia, increased inotropy/chronotropy
GIT: Nausea, vomiting, diarrhoea
RENAL: natriuresis

Pharmacokinetics
Absorption PO bioavailability > 90%
Distribution Vd = 0.5 L /kg

Protein binding = 40%

Metabolism Hepatic metabolism (90%) via CYP450 mechanisms to active and inactive metabolites. 10% unchanged
Elimination Renal elimination of active and inactive metabolites

Dialysable
T 1/2 = 6-12 hours (longer in children)

Special points Therapeutic concentration 10-20mg/ml


Examiner comments

Not previously examined



Question dnus

Question

Outline the pharmacology of metoclopramide.


Answer

Name Metoclopramide
Class Antiemetic / Prokinetic
Indications - Nausea and vomiting

- Prokinetic

Pharmaceutics Clear colourless solution (5mg/ml).

Tablet (10mg)

Routes of administration IV, PO, IM
Dose 10mg TDS (adults) for short duration (max 5 days)
Pharmacodynamics
MOA Central D2 antagonism at chemoreceptor trigger zone > reduced afferent input to vomiting centre in medulla
Effects GIT: Anti-emetic, Prokinetic (acceleration of gastric emptying)

CNS: EPSE (akathisia, dystonia, tardive dyskinesia) in children, drowsiness, dizziness, headache, worsening of Parkinson symptoms
CVS: arrhythmias

Pharmacokinetics
Onset Tmax < 1 hour (PO), <15 mins (IV)
Absorption PO bioavailability 80%
Distribution VOD = 3L / Kg

Protein binding 30%

Metabolism Minimal hepatic metabolism (conjugation)
Elimination Renal elimination (85%)

Active and inactive metabolites
T 1/2 = 4 hrs

Special points - Contraindicated in pheochromocytoma (precipitates pheo crisis), Parkinson's (Blocks Dopamine receptors), GI obstruction/perforation (prokinetic), avoid in children <20 years old (risk of EPSE)