VIVA: Pharmacology - Nervous system Flashcards
What is the mechanism of action of benzodiazepines?
- Binds to molecular components of GABA(A) receptor* in neuronal membranes in CNS* (gamma subunit of pentamer)
- This receptor is a chloride ion channel* and causes hyperpolarisation of the membrane
- The benzodiazepines do not substitute for GABA (major inhibitory neurotransmitter in the CNS), but appear to potentiate GABA’s effects without directly activating GABA(A) receptors or opening the chloride channels
- Causes an increase in the frequency (but not duration) of channel-opening events
*needed to pass
What are the organ level effects of diazepam?
- CNS:
- Sedation*
- Anxiolysis*
- Amnesia and psychomotor and cognitive depression at lower doses
- Hypnosis*
- Anaesthesia* at higher doses
- Anticonvulsant effect*
- Muscle relaxation* - Respiratory depression*
- Cardiovascular depression* (at higher doses and when hypovolaemic/CCF/chronic heart disease)
*3 to pass
What are the clinical uses of diazepam in the ED?
2 to pass:
- Anticonvulsant
- Sedation of agitated patient
- EtOH or benzodiazepine withdrawal
- Various toxidromes
What receptors do carbamazepine effect?
- Sodium channel blocker*
- Adenosine receptors antagonist
- Anticholinergic (antimuscarinic)
*needed to pass
What are the most common dose-related adverse effects of carbamazepine?
- CNS effects:
- Cerebellar effects: nystagmus, diplopia, ataxia
- Drowsiness - Anticholinergic effects:
- Dry mouth
- Tachycardia
- Blurred vision
- Delirium - Cardiovascular effects:
- Hypotension - GIT:
- GIT upset (nausea, vomiting)
- Hepatic dysfunction - Metabolic:
- Hyponatraemia, water intoxication - Haematological:
- Blood dyscrasias, including leukopaenia commonly
- Aplastic anaemic and agranulocytosis rarely - Dermatological:
- Erythematous skin rash
What important drug interactions does carbamazepine have?
- Induces CYP450 enzymes / hepatic drug metabolising enzymes* and P-glycoprotein, resulting in increased clearance of some drugs and reducing their therapeutic blood levels (e.g. OCP, warfarin, phenytoin, valproate, lamotrigine, diazepam, phenobarbitone, carbamazepine itself)
- As it induces its own metabolism, can result in breakthrough seizures
- Valproate and phenytoin may inhibit carbamazepine elimination
*needed to pass + 1 other
Outline the clinical uses of carbamazepine
- Anticonvulsant (partial and generalised tonic-clonic seizures)*
- Treatment of bipolar mood disorder
- Trigeminal neuralgia
Describe the mechanism of action of carbamazepine’s anticonvulsant activity
Blocks sodium channels:
- Inhibits high-frequency repetitive firing of neurons
- Presynaptic blocker of synaptic transmission (similar to phenytoin)
What are the pharmacokinetics of midazolam?
- Absorption:
- Water-soluble*
- Can be given PO, intranasal, buccal, PR, IV/IM/subcut
- Poor oral bioavailability* - Distribution:
- Highly protein bound*
- Crosses BBB easily at body pH - Metabolism:
- Hepatic metabolism
- Short elimination half-life* 1.5-2.5hrs - Excretion:
- Renal excretion
*2/4 to pass
What are the clinical effects of midazolam?
- Strong amnestic effect
- Anticonvulsant*
- Anxiolytic
- Sedative-hypnotic
- Antiemetic
- Reduced sensitivity to CO2 (respiratory depression)
*needed to pass + 2 others
What are the clinical indications for the use of midazolam?
- Anxiolysis
- Sedation*
- Anticonvulsant*
- Antiemetic
*needed to pass
What are the adverse effects of midazolam?
- Excess sedation*
- Respiratory depression*
- Decreased motor skills
- Impaired judgement
- Hypotension (particularly in hypovolaemic/CCF/chronic heart disease patients)
- Occasionally rash
*needed to pass
What is the mechanism of action of phenytoin?
- Sodium channel blockade* / reduced neuronal sodium conductance and prolongation of inactivated state of the sodium channel
- Reduces Ca2+ influx into cells to decrease glutamate release
- Enhances GABA release
- Inhibits generation of rapidly repetitive action potentials
*needed to pass
Describe the elimination pharmacokinetics of phenytoin and how it affects toxicity
- Phenytoin has dose-dependent elimination*
- At low serum concentration it has first order kinetics*
- Elimination becomes zero-order as serum concentration rises* with prolonged elimination and greater chance of toxicity with recurrent dosing and with even small increases in dose
*needed to pass
What are the adverse effects of phenytoin?
- Neurological (dose-related)*:
- Ataxia
- Drowsiness
- Dizziness
- Blurred vision
- Hallucinations
- Slurred speech and confusion
- Peripheral neuropathy (idiosyncratic) - Skin/soft tissue:
- Hirsutism
- Gingival hypertrophy
- Acne
- Facial coarsening - Cardiovascular*:
- Hypotension and arrhythmias with rapid IV administration
*1 of each to pass
Describe the pharmacokinetics of phenytoin
- Absorption:
- High oral bioavailability (90%), poor IMI
- Peak serum concentration 3-12hrs - Distribution:
- Highly plasma protein bound (90%)*
- Vd 45L/70kg and widely distributed (brain, liver, skeletal muscle, fat) - Metabolism:
- Metabolised to inactive metabolites by the liver*
- Dose-dependent: first order kinetics at low concentrations, zero order kinetics at higher concentrations due to saturation of hepatic enzymes (slows elimination)*
- Half-life variable (12-36hrs) dependent on serum concentration as above - Excretion:
- Renal (<2% unchanged)
*needed to pass
What is the rationale for using a loading dose of phenytoin?
Reaches target concentration dose more quickly (otherwise it takes 4 half-lives to get to steady state)
What are the risks associated with IV phenytoin administration?
- Hypotension and bradycardia with rapid infusion* (due to diluent):
- Limit rate of infusion to 50g/min maximum (30-60mins for full dose)
- Less likely with fosphenytoin - Allergic reactions
- Local necrosis if extravasation
*needed to pass
Describe the pharmacokinetics of valproate
4 to pass:
- Absorption:
- Can be administered IV or PO
- Well-absorbed orally with bioavailability >80%
- Peak blood levels within 2hrs - Distribution:
- Highly protein bound
- Low volume of distribution 0.15L/kg - Metabolism:
- Extensively metabolised in the liver
- Long half-life 9-18hrs - Excretion:
- Excreted as glucuronide conjugate in urine (30-50% of dose)
What are the adverse effects of sodium valproate?
- GIT*:
- Nausea, vomiting
- Abdominal pain
- Reflux
- Asymptomatic LFT derangement
- Weight gain, increased appetite (less commonly)
- Idiosyncratic hepatic failure (rare; risk highest <2yrs old)
- Pancreatitis - CNS*:
- Fine tremor
- Ataxia
- Sedation
- Fatal encephalopathy if there is also a genetic abnormality of urea metabolism - Skin/soft tissue:
- Alopecia
- Rash - Haematological:
- Idiosyncratic thrombocytopaenia - Metabolic:
- Hypernatraemia - Reproductive:
- Teratogenic if given in 1st trimester (e.g. neural tube defects, cardiovascular/facial/digital abnormalities) - Hypersensitivity reactions
*1 of each to pass + 2 others
Sodium valproate exhibits capacity-limited protein-binding kinetics. What is this?
- Sodium valproate is highly bound to plasma proteins (90%) at lower concentrations (75mg/L)
- This mechanism is saturated at higher concentrations (150mg/L) leading to an increase in free drug (70% protein bound)
- Results in apparent increased clearance of drug at higher doses and reduction in half-life: variable clearance
- Thus dosage is preferred as a sustained release preparation
What are the possible pharmacodynamic mechanisms of sodium valproate?
- GABA increased presynaptically by reduced GABA breakdown to succinate (ABAT/GAT1), possibly increased production (GAD)
- Direct inhibitory actions on post-synaptic sodium channel, particularly high frequency gates, and Ca2+ (membrane-stabilisation - reduced voltage-gated outflow)
- Possible blocked NMDA receptor activation effects
Describe the pharmacodynamics of amitriptyline
- Blocks reuptake of serotonin and noradrenaline*
- Blocks muscarinic, sympathetic a1, GABA(A), Na+ channel and histamine receptors
- Monoamine vs neurotrophic vs neuroendocrine theories
*needed to pass + 2 other receptors
What are the toxic effects of amitriptyline and how are they mediated?
3 effects + receptor responsible:
1. Anticholinergic:
- Blurred vision
- Dry mouth
- Tachycardia
- Urinary retention
- Delirium
2. Antihistamine:
- Sedation
3. Alpha adrenergic blockade:
- Hypotension
4. Na+ channel blockade:
- Widened QRS
- Bradycardia
5. Direct central effects:
- Seizures
What are the adverse and/or toxic effects of lithium?
- Neurological:
- Tremor
- Choreoathetosis
- Ataxia*
- Dysarthria
- Hyperactivity
- Confusion*
- Withdrawal - Endocrine:
- Reversible hypothyroidism* - Renal:
- Nephrogenic diabetes insipidus (polyuria, polydipsia)*
- Chronic interstitial nephritis
- Nephrotic syndrome - Cardiovascular:
- Oedema
- Worsening of sick sinus syndrome
- 3/4 to pass
Describe the pharmacokinetics of lithium
- Absorption:
- Oral absorption* peaks at 0.5-2hrs, complete at 6-8hrs - Distribution:
- Distributes in TBW*
- Therapeutic concentration 0.6-1.4mmol/L - Metabolism:
- Half-life 20hrs* - Excretion:
- Unchanged in urine*
*needed to pass
How can you assess lithium toxicity and how is it treated?
- Measure levels 10-12hrs post last dose*
- > 2mmol/L should be considered toxic
- Treatment is supportive and haemodialysis
What is the mechanism of action of tricyclic antidepressants?
- Inhibition of serotonin and noradrenaline reuptake*:
- Increases amount of serotonin and noradrenaline in certain parts of the brain (cortex and limbus; “monoamine hypothesis” for depression”) and spinal cord (ascending corticospinal tract - useful in neuropathic pain) - Also blocks:
- Na+ channels
- K+ channels
- Muscarinic (M1) receptors (anticholinergic)
- Histaminic (H1) receptors
- Alpha-1 adrenergic receptors (peripheral post-synaptic)
*needed to pass + 1 other
What clinical manifestations would be seen in an overdose of tricyclic antidepressants?
- Cardiovascular*:
- Tachycardia
- Hypotension (due to alpha blockade, impaired contractility)
- ECG changes: PR prolongation, QRS widening (Na+ blockade), prolonged QT (K+ blockade), VT, VF - CNS*:
- Drowsiness
- Delirium (anticholinergic)
- Seizures
- Coma - Anticholinergic*:
- Agitation, delirium
- Mydriasis
- Dry, warm, flushed skin
- Urinary retention
- Ileus
- 1 example from each
What factors determine the volume of distribution of a drug?
- Drug factors*:
- Lipid solubility
- pKa
- pH
- Protein binding - Patient factors*:
- Age
- Gender
- Comorbid disease (e.g. oedema, ascites)
- Body fat
- Blood flow to tissues
- 2 from each group
What therapies for tricyclic toxicity might reduce their tissue distribution?
Alkalinisation (e.g. with bicarbonate or hyperventilation) increases plasma protein binding of free drug, removing it from the tissues and reducing its toxicity
Describe the volume of distribution of tricyclic antidepressants. What factors contribute to this, and how does their volume of distribution influence their toxicity?
TCAs have a large Vd* (5-30L/kg), with high lipid solubility and high tissue protein binding
Tissue concentrations are high* in toxicity especially in well-perfused organs such as the brain and heart*
- needed to pass
By what routes can olanzapine be administered?
- PO*, sublingual
- Parenteral* (IV, IM, depot IM)
*needed to pass
What dose and what route of olanzapine would you use for sedation in an agitated patient?
Dose 10-20mg regardless of route
What are the advantages of olanzapine over older “typical” antipsychotics?
- Less extrapyramidal effects*
- Less hypotension
- Less tachycardia
- Less effect on prolactin
- More effective for both negative and positive psychotic symptoms, and cognition
- Multiple routes of administration
- High clinical potency
*needed to pass
What are the some of the clinical disadvantages of olanzapine?
2 to pass:
- Anticholinergic effects
- Lowered seizure threshold
- Weight gain
- Diabetes mellitus
- Hyperlipidaemia
- Expense