• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/693

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

693 Cards in this Set

  • Front
  • Back
What is potency?
the amount of drug required to produce an effect of given intensity.
Potency is measured by the ed50 which is the amount (dose) of drug required to produce 50% of the drug's maximal effect
What is efficacy?
measure of the maximum clinical respose to the drug regardless of dose
What is the ec50?
the dose at which 50% of people exhibit a quantified effect.
What is td 50?
The dose required to produce a toxic response in 50% of subjects (LD 50 has same definition but the toxic response is death)
What is the therapeutic index?
TD50/EC50 ratio
Give examples of lipid soluble ligands that cross the membrane and act on intracellular receltors?
steroids such as corticosteriods, sex steroids and vitamin D
These bind to the nucleus to stimulate transcription of genes and make new proteins- therefore the lag is 30 minutes to hours while the proteins are being made.
They persist in their effect over days when the agonist concentration goes to zero due to slow turnover of most enzymes and proteins
Give examples of substances that trigger ligand gated ion channels?
How does this work?
acetylcholine, gaba, excitatory amino acids
Receptor alters transmembrane conductance of ions and thereby alters electrical potential across the memebrane
Describe the nicotinic acetylcholine receptor?
pentamer made up of 5 polypeptide units (2 alpha, 2 beta, 1 gamma) which each cross the lipid bilayer 4 times and form a cylindrical structure. Binding of ach causes structural change that opens sodium channel. Occurs in milliseconds.
Give examples of substances that bind to a transmembrane receptor that stimulates a tyrosine kinase.
Insulin, PDGF, ANF
How do tyrosine kinase receptors work in general?
Receptor polypeptide consists of a hormone binding domain (extracellular) and an enzyme domain (cytoplasmic) which are connected through the membrane. Hormone binds with extracellular receptor, resulting conformational change that brings together the protein tyrosine kinase domains that become enzymatically active.
What are the three mechanisms by which a drug/ligand binding to a transmembrane protein can cause change?
1. The transmembrane receptor contains an ion channel which changes shape e.g. gaba, ach
2. The transmembrane receptor has a tyrosine kinase on the insidene.g. insulin, pdgf, anf
3. Transmembrane receptor stimulates a g-protein- the activated g-protein changes the activity of a receptor element (usually an enzyme or an ion channel)
What is the advantage of signalling via g-proteins?
Signalling via G proteins allows effect to persist long after the extracellular receptor has dissociated from its agonist molecule
What do Gs G-proteins do?
increase adenylyl cyclase, causing increased cAMP
Give example of Gs G-proteins?
Beta adrenergic amines, glucagons, histamine, serotonin.
What do Gi G-proteins do?
Give examples of these?
decrease adenylyl cyclase, causing decreased cAMP Open cardiac potassium channels causing decreased heart rate

Alpha2 adrenergic amines, acetylcholine (muscarinic only), opioids, serotonin
What do Gold g-proteins do
Stimulate adenylyl cyclase causing increased cAMP.
stimulated by odourants
What do Gq G-proteins do?
Give examples of these?
Increase adenylyl cyclase causing increased cAMP Increase phopholipase C resulting in increased IP3, diacylglycerol and cytoplasmic calcium.

Acetylcholine (muscarinic), serotonin
What is the structure of g-protein coupled receptors?
serpentine receptors- polypetide chains which cross the membrane 7 times.
How does cAMP work?
G protein stimulates membrane adenylyl cyclase that converts ATP to cAMP. cAMP exerts most effects by stimulating cAMP dependent protein kinases.
How does calcium and phospholipase second messaging work?
G protein stimulates membrane enzyme phospholipase that hydrolyses PIP2 to DAG and IP3. DAG is confined to the membrane and activates protein kinase C.

IP3 diffuses through the cytoplasm to trigger release of calcium from internal stores. Calcium binds to calmodulin, which regulates calcium dependent protein kinases.
How are DAG and IP3 inactivated?
DAG inactivated by phosphorylation back to phospholipid.
IP3 rapidly inactivated by dephophorylation.
How does cGMP work?
small role -mainly intestinal mucosa and vascular smooth muscle. G protein stimulates membrane guanylyl cyclase which converts GTP to cGMP. cGMP exerts most effects by stimulating cAMP dependent protein kinases.
What is the volume of distribution?
What are the units?
It relates the amount of drug in the body to its concentration in blood or plasma- depends what it is soluble in/binds to.
Vd=Amount of drug in body /concentration. Most commonly expressed in units of litres per kilogram
Give an example of a drug that is distributed in the total body water?
What is the volume of distribution?
ethanol and other small water soluble molecules
0.61L/kg (42L)
Give an example if a drug that is distributed in the extracellular water?
What is the volume of distribution?
Mannitol, gentamycin
larger water soluble molecules
0.2l/kg
14 L
What is the volume of distribution of blood?
0.08l/kg= 5.6L
Give an example if a drug that is distributed in plasma? (and explain why it is largely confined to plasma)
What is the volume of distribution?
Heparin
0.04l/kg
Bound to a plasma protein
Give an example if a drug that is distributed in fat?
What is the volume of distribution?
DDT (insecticide)
0.2-.35l/kg (14-24l)
Give an example if a drug that is distributed in bone?
What is the volume of distribution?
0.07l/kg
(4.9L)
lead and flouride
What drugs cannot be removed by dialysis?
drugs with large volumes of distribution
Give examples of drugs with large volumes of distribution? (5-10L/kg)
Antidepressants
Phenothiazines
Propanolol
Verapamil
Give examples of drugs with small volumes of distribution? (<1l/kg)
Theophylline
Salicylate
Phenobarbitone
Lithium
Phenytoin
Heparin
Warfarin
What is the half life of a drug?
Time required to change the amount of drug in the body by 1/2 during elimination or during a constant infusion
T1/2=0.7xVd / CL
Half life can refer to the drug itself or the active metabolites of the drug
What is allosteric action?
drugs that bind to the same receptor but do not prevent binding of the receptor molecule, may enhance or inhibit action
How is the action of transmembrane receptors terminated?
ligand binding often causes acellerated endoscytosis of reception followed by the degradation of the receptors and their bound ligands
Why are volumes of distribution greater than actual volumes found in the human body?
because it is the volume apparently necessary to contain the concentration found in blood, plasma or water
Drugs with very high volumes of distribution have much higher concentrations in the extravascular compartment then in the vascular compartment- i.e. not homogenously distributed.
Drugs that are completely retained within the vascular compartment have a minimum volume of distrubution
What is drug clearance?
Rate of elimination/concentration, this is additive when more than one organ clears it

i.e. CL(liver)= rate of elimination by liver/C

kidney generally excretes unchanged drug while liver metabolises
What is the rate of elimination?
CLx C when clearance is first order
calculated using the area under the time concentration curve (dose/AUC)
When is rate of elimination not first order?
capacity limited elimination e.g. phenytoin and ethanol, aspirin
also known as dose/concentration and saturatable elimination
cannot use auc to measure rate of elimination

rate of elimination = vmax x c/ km x c
What is flow-dependent elimination?
relevant for drugs that are cleared mainly on the first pass and therefore elimination depends on blood flow to the organ
What is bioavailability?
The fraction of the drug reaching the systemic circulation following administration by any route- IV has greatest bioavailability because it avoids first pass metabolism, extent of absorption (liphophilicity, reverse transporter associated with p glycoprotein)
How do your calculate the dosing rate for a drug?
CL x target concentration (TC)
How do you calculate the maintenance dose?
Drugs are usually administered in order to achieve a steady state where dosing equals elimination.
Dosing rate=CL X target concentration (TC)
Maintenance dose=dosing rate x dosing interval
When is a loading dose needed?
How does one calculate the loading dose?
Loading doses are required if the half-life of a drug is prolonged and the time taken to reach steady state would otherwise be prolonged

If the target concentration is known the clearance will determine the dosing rate
Loading dose=Vd X TC(target concentration)
Give a formula for systemic clearance of a drug?
Clearance can pertain to each organ and is additive in effect CLrenal+CLliver+CLother=CLsystemic.
What is another work for capacity limited clearance?
zero order kinetics
Describe how zero order kinetics works?
Drug elimination pathway becomes saturated at high concentration of drug. Elimination is proportional to concentration of the drug at low concentrations but at high concentrations elimination is constant.

e.g. Aspirin, Phenytoin, Ethanol
What is first order clearance?
First order clearance = clearance proportional to concentration
What are some drugs that exhibit flow dependent elimination? Give 3 examples?
Extraction is chiefly dependent on blood flow through the organ and the drug is almost completely extracted by the organ on first pass

Morphine Lignocaine Propanolol
What are some limitations to drug absorption in the gut?
Lipophilic (acyclovir) versus hydrophilic (atenolol) drugs
Bacterial metabolism within the gut (digoxin) Absorption abnormalities in small bowel.
What is the extraction ratio?
Extraction ratio defines the degree of first pass metabolism.
ER=CL liver /Q
(Q is hepatic blood flow = 90l/h)
Where does first pass elimination occur?
Can occur in gut wall, portal blood, or by excretion in bile. Most important is metabolism by liver.
What is the formula for systemic bioavailability?
Systemic bioavailability (F)
=extent of absorption (f) X (1-ER)
How do you avoid hepatic first pass metabolism?
Hepatic first pass metabolism can be avoided by sublingual, transdermal and to a lesser extent, rectal administration
What is biotransformation?
Biotransformation is the metabolism of drugs that allows for the renal excretion of lipophilic, un-ionised or partially ionised drugs that would otherwise fail to be effectively excreted and have a prolonged duration of action.

Biotransformation transforms a lipophilic molecule into a more polar and therefore more readily excretable product.
Where does biotranformation occur
Biotransformation can occur in GIT (eg clonazepam, penicillin), lungs, skin, kidneys, but most important site is liver.
What are the two phases of biotransformation reactions?
Phase 1 reactions Convert the parent drug to a more polar metabolite by introducing or unmasking a functional group such as OH, NH2, SH.

Phase 2 reactions The introduced functional group combines with an endogenous substrate to form a highly polar conjugate. Enzymes for phase 2 reactions may be located in microsomes or in the cytosol. Phase 2 reactions can sometimes precede phase 1 reactions.
How do phase 1 reactions work?
Phase 1 reactions utilise mixed function oxidases located on the ER of liver cells and other tissues.

Require oxygen and NADPH to function. Mixed function oxidases include NADPH-cytochrome P450 reductase and cytochrome P450.
How many major CYP450 isoforms are there? What is the most important one?
Different P450 isoforms are responsible for metabolism of different drugs. 7 main isoforms account for most metabolism. CYP3A4 is the largest component, responsible for 60% of clinically prescribed drugs metabolised by the liver.
Where do phase 1 and 2 reactions occur?
Whase 1 occur on the ER, Phase 2 occur in microsomes and the cytosol
What are some genetic factors that affect biotransformation?
Suxamethonium - genetic defect in
pseudocholinesterase, causes suxamethonium to remain active for prolonged periods. Other examples - oxidation of ethanol, acetylation of isoniazid
What are some environmental factors that affect biotransformation?
Enzymes may be induced or inhibited by environmental factors.
Charcoal - induction
grapefruit juice -inhibition of CYP3A4 (responsible for 60% of drug metabolism)
What dies phenytoin do to digoxin metabolism?
Phenytoin enhances digoxin metabolism
What do barbituates do to warfarin metabolism?
Barbiturates enhance warfarin metabolism
The metabolism of what drugs does cimetidine affect?
Cimetidine inhibits warfarin and diazepam metabolism
The metabolism of what drugs is affected by cardiac disease?
Cardiac disease can affect drugs that are flow-limited
e.g. Morphine, Verapamil
What are the physical barriers to drug distribution? list 4
1. Aqueous diffusion -generally determined by fixed law though if a drug is charged its flux will be influenced by electrical fields.

2. Lipid diffusion - most important limiting factor for drug permeation. Lipid:aqueous partition coefficient determines how readily the molecule moves between acid aqueous and lipid media. The abililty of weak acids and bases to move between aqueous and lipid mediums depends on pH.

3. Special carriers (active transport/facilitated diffusion) - for molecules that are too large or too insoluble - eg peptides, amino acids, glucose

4. Endocytosis and exocytosis for very large molecules -eg vit B12, iron
What is fick's law of diffusion
Passive flux of molecules down a concentration gradient equals the difference in concentration across the membrane (C1- C2) multiplied by the area of the membrane and the permeability coefficient, divided by the thickness of the membrane

conc grad x sa x perm coeff /thickness
Define weak acid?
a neutral molecule that can readily dissociate into an anion and a proton
Define weak base?
A neutral molecule that can combine with a proton and form a cation
What is the pKa?
pKa =the pH at which the concentrations of ionized and and inionized forms are equal
When are weak acids and bases more likely to be in a lipid soluble form?
More of a weak acid will be in a lipid soluble from at an acid pH.
More of a weak base will be in a lipid soluble form at an alkaline pH

This is important for excretion of drugs by the kidney.
What is the henderson hasselbach equation?
pH = pKa + log [base]/[acid]
What is the general structure of local anaesthetics?
Consist of a lipophilic group, ester or amide chain and ionisable group (usually a tertiary amine)
What are the two types of local anaesthetics?
Esters: Cocaine (procaine) (benzocaine)
Amides: Lignocaine Bupivicaine Prilocaine (etidocaine)
Define local anaesthetics?
Agents which reversibly block impulse conduction along nerve axons, thereby reducing pain sensation- usually do this by blocking voltage gated sodium channels
Where do local anaesthetics act on the sodium channel?
Blockade occurs at the intracellular end of the sodium channel
What are the pharmacodynamics of local anaesthetics?
Activated channels have higher affinity for drug therefore drug effect is more marked in rapidly firing fibres.
Progressive increase in drug concentration causes increased threshold, reduced action potential amplitude, then failure to produce an action potential.
What ion concentration changes increase and decrease the effects of local anaesthetics?
Local anaesthetic effect is increased by hyperkalaemia and decreased by hypercalcaemia
How does fibre type (size and myelination) affect how local anaesthetics work on the nerves?
Large diameter fibres less sensitive than small diameter fibres. (3 successive nodes required for blockade, nodes are further apart in large fibres).

Myelinated fibres of the same diameter as unmyelinated fibres tend to become blocked first.
Why are sensory nerves more sensitive to local anaesthetics?
Sensory fibres tend to have a fast firing rate and long action potential therefore are more sensitive to blockade.
How does the position of a nerve in the bundle affect how sensitive it is to local anaesthetics?
Peripheral nerves exposed first -motor nerves tend to be peripheral in large trunks.
What are the safe doses for lignocaine with/without adrenaline?
2mg/kg IV, 3mg/kg SC, 5mg/kg with adrenaline.
What is 1% solution of drug?
10mg/ml
What is the safe dose of bipivicaine and prilocaine?
Bupivacaine 2mg/kg SC
Prilocaine 3-5mg/kg IVRA
What are the relative potencies of the local anaesthetics, say lignocaine = 4
(if procaine = 1)
Cocaine =2
Lignocaine =4
Bupivacaine = 16
Prilocaine = 3
What is the CNS toxicity of local anaesthetics?
Drowsiness, visual and auditory disturbance, restlessness, nystagmus, shivering, convulsions, CNS depression.
How do you manage convulsions caused by local anaesthetics?
If convulsion occurs, the patient should be hyperventilated to induce respiratory alkalosis as this lowers extracellular potassium and favours rested, low affinity sodium channels
Which local anaesthetics cause the most CNS and CVS toxicity?
Bupivacaine>lignocaine> prilocaine

Bipivicaine is most cardiotoxic -Although all local anesthetics potentially shorten the myocardial refractory period, bupivacaine avidly blocks the cardiac sodium channels, thereby making it most likely to precipitate malignant arrhythmias.
What are the cardiovascular effects of local anaesthetics?
Depression of cardiac pacemaker activity, excitability and conduction. Negative inotropic effect and decreased peripheral resistance.
How is cocaine different from other local anaesthetics in its cardiovascular effect?
noradrenaline uptake blockade and subsequent vasoconstriction and hypertension.
What haemotological toxicity do local anaesthetics have?
Prilocaine in high doses liberates the 0-toluidine metabolite that causes methaemoglobinaemia.
Which drug interract with local anaesthetics?
Fentanyl and midazolam utilise same microsomal enzymes in liver

Halothane, cimetidine and beta blockers decrease hepatic blood flow and therefore reduce metabolism

Enzyme inducers such as phenytoin may increase metabolism
Are local anaesthetics acid or base?
most are weak bases, potency depends on how liphophilic it is and therefore depends on the pH of the tissue
What is the active form of local anaesthetics? Is this the form that gets into the tissues?
Most are in a charged, cationic form and this is the active form at the receptor site, but the uncharged form is required for penetration - hence poor penetration in acidic (infected) tissue
How protein bound are local anaesthetics?
bupivicaine 95%, prilocaine 50%
What affects systemic absorption of local anaesthetics?

What does local effect depend on?
dose, vascularity of site of injection, drug-tissue binding, presence of vasoconstrictors (more effective for short acting highly lipid soluble drugs), chemical properties of the drug.

Local effect is proportional to the amount of drug that penetrates the nerve fibre
What is the comparative duration of action of the common local anaesthetics?
Cocaine -medium
Lignocaine -medium
Bupivacaine - long (up to 12 hours for peripheral nerve blocks)
Prilocaine - medium.
Why do ester local anaesthetics have a very short plasma half life?
Rapidly metabolised by pseudo- cholinesterase therefore half life less than 1 minute.
How are amide local anaesthetics metabolised?
Slowly hydrolysed by liver enzymes and excreted by kidney: Dosage reduction required in liver disease and reduced hepatic blood flow

Prilocaine metabolised most rapidly, lignocaine intermediate, bupivacaine slowest: Prilocaine metabolism produces O- toluidine
What are guedel's stages of anaesthesia?
Stage of analgesia
Stage of excitement
Stage of surgical anaesthesia
Stage of medullary depression
What are the 5 factors that affect the brain uptake of an inhaled anaesthetic?
1. Partial pressure of inspired anaesthetic agent

2. Solubility (blood:gas partition coefficient)
The more soluble an agent the longer it takes for its partial pressure in blood to rise therefore the slower the onset of anaesthesia.

2. Pulmonary ventilation Increases the rate of induction of anaesthesia for drugs with high solubility - little effect on drugs with low solubility

3. Pulmonary blood flow
Increased flow decreases the rate of induction with
soluble agents, little effect with poorly soluble agents.

4. Arteriovenous concentration gradient (tissue:blood solubility coefficient)
Gradient between arterial and mixed venous blood determined by uptake of agent by highly perfused organs such as brain, heart, liver, kidneys and gut. Drugs with high tissue:blood solubility coefficient take longer to reach equilibrium.
Define the MAC (minimal alveolar concentration).

Give an example of a drug with a high MAC?
What are the limits to using MAC?
The partial pressure (% concentration) of an agent which results in immobility of 50% of patients undergoing a surgical incision.

For nitrous oxide, MAC>100% means that even if the partial pressure of nitrous oxide is 760mmHg, incomplete anaesthesia is achieved.

There may be vast individual differences and the MAC gives no indication where the other 50% lie on the curve.
Why is steady state alveolar concentration a useful measure of potency?
When steady state is achieved, the partial pressure of an inhaled anaesthetic in the brain equals that in the lung, therefore measurement of steady state alveolar concentration gives a measure of potency
How do inhaled general anaesthetic agents work?
Increase in cellular threshold to firing with subsequent decreased spontaneous and evoked neuronal activity.
Ionic basis of effect includes activation of potassium currents to cause hyperpolarisation and opening of cation channels to decrease synaptic transmission.
Research suggests that agents interact with lipid membranes to cause distortion of ion channels.
What are the cardiovascular effects of general anaesthetic agents?
Dose related variable reduction in mean arterial pressure and myocardial oxygen demand. Effects may be masked by nitrous oxide which causes sympathetic stimulation

Nitrous oxide causes minimal depressant effects

Halothane sensitises the myocardium to catecholamines and is arrhythmogenic.
What are the respiratory effects of general anaesthetics? List 5
1. Respiratory depression due to reduced TV and inadequate increased rate. (except nitrous oxide)

2. Increase apneic threshold to pCO2.

3. Decrease ventilatory response to hypoxia.

4. Decrease mucociliary function leading to atelectasis.

5. Most have bronchodilator action.
What are the CNS effects of general anaesthetics?
Increase metabolic rate and blood flow to brain due to reduced cerebrovascular resistance. Hyperventilation reduces this effect.
What are the GI and GU effects of general anaesthetics?
GUS
Reduced renal blood flow and GFR. Uterine relaxation (minimal with nitrous oxide)

GIT
Reduced hepatic blood flow.
Describe the acute and chronic toxicity of general anaesthetic agents?
Halothane -1 in 35000 cases of fatal hepatic necrosis.

Methoxyflurane -fluoride related nephrotoxicity.

Malignant hyperthermia -tachycardia, hypertension, acidosis, hyperkalaemia, muscle rigidity, hyperthermia ,more common if suxamethonium also used -treated with dantrolene.

Chronic exposure to Nitrous oxide is associated with megaloblastic anaemia
What is the MAC of the common anaesthetic agents?
Nitrous oxide >100
Isoflurane - 1.4
Halothane - 0.75
Methoxyfluorane - 0.16
What are the solubilities and the brain:blood partition coefficients of the common anaesthetic agents?
Solubility (blood:gas partition coefficient) :
Nitrous oxide - 0.47
Isoflurane -1.4
Halothane - 2.3
Methoxyfluorane - 12

Brain:blood partition coefficient Nitrous oxide ␣ 1.1 Isoflurane - 2.6
Halothane - 2.9
Methoxyfluorane - 2.0
Give some examples of IV general anaesthetics?
Barbiturates Benzodiazepines Opioids Propofol Ketamine
Give examples of barbituate general anaesthetics?
Phenobarbitone
Thiopentone -ultra short acting barbiturate intravenous anaesthetic
How do barbituates work? (3 mechanisms)
1. Barbiturates bind to components of the GABA receptor and facilitate its action by increasing the duration of chloride channel opening.
2. At high concentrations GABA may directly stimulate the receptor.
3. Also depress actions of other excitatory neurotransmitters and have non-synaptic membrane effects.
What is the structure of the GABA receptor in the CNS?
GABA receptor consists of 5 alpha, beta and gamma membrane-spanning proteins which can form different pentameric combinations.
What are the indications for barbiturates in humans?

What is the dose of thiopentone?
Phenobarbitone - neonatal seizures.
Thiopentone -anaesthesia
Dose Thiopentone 3-5mg/kg
What are the adverse effects of thiopentone?
Toxicity CVS Depression at high doses.

Dose dependent decreased blood pressure, stroke volume and cardiac output.
Respiratory depression.
Decreased cerebral metabolism and blood flow. Nystagmus.
Reduces hepatic and renal blood flow.
Dizziness, fatigue, amnesia, blurred vision. Tolerance. Dependence.
What interactions do barbituates have with other drugs?
Effects are potentiated by other sedatives such as alcohol or other sedatives-may cause fatal CVS depression.

Induction of liver enzymes - decreases effect of warfarin, anticonvulsants, digoxin.
What are the contraindications for barbiturates?
Precaution with liver failure. Readily crosses placenta and enters breast milk. Porphyrias
What is the absorption and distribution of thiopentone and phenobarbitone?
Phenobarbitone -Orally active, rapidly absorbed. 50% protein bound.
Thiopentone - intravenous
Highly lipid soluble and rapidly distributed to brain, then redistributed to other tissues. Redistribution from CNS to skeletal muscle and adipose tissue is important process that contributes to termination of CNS effects.
Initial redistribution to brain and viscera, then to lean tissues and then to fat
Metabolised slowly in liver to water- soluble inactive metabolites that are excreted in urine.
What are the half lives of thiopentone and phenobarbitone?
Phenobarbitone plasma half life 4 hours, excretion half life 4-5 days (therefore has a tendency to accumulate)

Thiopentone Produces hypnosis in one circulation time. Plasma:brain equilibrium occurs rapidly (< 1 minute) due to high lipid solubility. Rapid redistribution is responsible for short acting effect of 20-30minutes
How much of thiopentone and phenobarbitone is excreted unchanged?
Phenobarbitone 20% excreted unchanged.
Thiopentone - 1% excreted unchanged, metabolised at 12- 16% per hour.
What is propofol?
Phenol derivative.
Short acting intravenous anaesthetic agent.
Presented as an oil in water emulsion.
How does propofol work?
Potentiates the action of inhibitory neurotransmitters including GABA and glycine.
What is the dosage of propofol?
Sedation 0.5-1mg/kg Usually given in 20mg increments

Induction of anaesthesia 2-2.5mg/kg Usually given in 40mg increments.
Dose can be repeated as required.
What are the adverse effects and contraindications of propofol?
1. Marked hypotension (15- 25%) due to reduced peripheral vascular resistance (direct effect of propofol). Potent negative inotropic effects. No compensatory increase in heart rate.
2. Potent respiratory depression. Brief apnoea common. Infusion produces decreased tidal volume. Some bronchodilation due to direct effect on smooth muscle.
3. Rapid, smooth induction and clear headed recovery. Cerebral blood flow and ICP decrease slightly.
4. General Pain in injection in 25%.

Contraindications: Acidosis and possible neurological sequelae in children therefore contraindicated under 3 years.

Given as an intravenous fat emulsion - previous vehicle cause hypersensitivity reactions.
Onset, distribution and elimination of propofol?

What is the volume of distribution and why?
Rapid onset (30 seconds) and recovery

Distribution half life (t1/2 alpha) : 2-8 minutes,

Elimination half life (t1/2 beta): 30- 60 minutes.

90% protein bound. Small volume of distribution.
Where and how quickly is propofol metabolised?

What determines the distribution of effect of propofol?
Rapidly metabolised in liver (10 times faster than thiopentone) and excreted in urine.
Duration of effect largely determined by redistribution.

Less than 1% excreted unchanged.
What is ketamine and how does it work?
Short acting non-barbiturate intravenous anaesthetic agent. Chemically related to PCP

Action may involve blockade of glutamic acid (NMDA) receptors.
Mechanism of action largely unknown
What kind of anaesthesia does ketamine produce?
Characteristic
dissociative anaesthesia :amnesia, profound analgesia, normal or slightly increased muscle tone without loss of consciousness or loss of protective reflexes.
What is the IV dose of ketamine?
1-4mg/kg Administer slowly over 60 seconds to avoid respiratory depression and pressor response
What is the onset and offset of ketamine?
Onset 30 seconds, duration of action 5- 10 minutes. Additional doses can be given without accumulation

(Intramuscular 6.5-13mg/kg
Onset 2 minutes, duration 20 minutes)
What are the organ effects and toxicity of ketamine?
1. CVS: Increased heart rate, blood pressure and cardiac output via inhibition of noradrenaline reuptake -peaks 2-4 minutes after injection, declines after 20 minutes.
2. RS: May increase or decrease respiratory rate for several minutes. Upper airway tone is maintained.
3. CNS Marked increase in cerebral blood flow and intracranial pressure. Nystagmus

4. Emergence phenomena in 12% -disorientation, sensory and perceptual illusions and vivid dreams. Less incidence in children and elderly.
Reduced by minimising verbal, tactile and visual stimuli during recovery Interactions

Avoid hypertensive agents
Contraindications Uncontrolled hypertension, severe cardiovascular disease
Where does ketamine distrubute to and how is the effect of ketamine terminated?
Distribution Highly lipid soluble.
Rapid distribution to all tissues.
Termination of effect due to redistribution from brain to peripheral tissues
What is the metabolism of ketamine?
Metabolised by liver to 4 different metabolites including norketamine which has one sixth the potency of ketamine
Metabolites excreted in urine.
What is the structure of muscle relaxants?
What are the 2 types of muscle relaxants?
Structure: All bear a structural resemblance to acetylcholine

Depolarising Suxamethonium= 2 acetylcholine molecules linked end-to-end.

Non- depolarising (NDPMRs) Isoquinolone, tubocurarine, atracurium.
Steroid: pancuronium, vecuronium, rocuronium.
How do depolarising muscle relaxants work?
1. Binds with the nicotinic receptor at the neuromuscular junction to cause the sodium channel to open and the end plate to depolarise. This results in generalised disorganised contraction. Suxamethonium is not metabolised effectively at the synapse, therefore depolarised membranes remain depolarised and unresponsive to subsequent impulses.
2. Phase 2 block (Desensitising): depolarisation gradually decreases and the membrane becomes repolarised. Membrane cannot become repolarised while suxamethonium is present- essentially this is the same as non-depolarising blockade.
How do non-depolarising muscle relaxants work?
Reversible blockade, act predominantly at nicotinic receptors. Prevents opening of the sodium channel, may also enter ion pore at higher does and cause blockade.
What are the doses of suxamethonium and verocuronium?
Suxamethonium 1-1.5mg/kg (children relatively resistant) May be given IM
Vecuronium 0.1mg/kg
What are the adverse effects of depolarising and non-depolarising muscle relaxants?
1. CVS: Pancuronium causes a moderate increase in heart rate and cardiac output due to vagolytic action.
Vecuronium, rocuronium (and most others) have little or no cardiovascular effects
2. Suxamethonium stimulates all autonomic cholinoceptors to some extent.
Bradycardia and negative inotropic effects at low doses and especially after a second dose.
Prevented by premedication with atropine and by giving a minimum dose.
3. Hyperkalaemia Due to exaggerated release of potassium from extra- junctional nicotinic receptors. There is an exaggerated release with burns, renal failure.
4. Raised intraocular pressure Raised intragastric pressure Raised intracranial pressure
5. Muscle pain This is the most common side effect

Malignant hyperthermia: Tachycardia, tachypnoea, rigidity
Contraindications to muscle relaxants?
Personal or family history of malignant hyperthermia Muscular dystrophy
Absorption and distribution of muscle relaxants?
All are highly polar and inactive orally.


Non-depolarising : Rapid initial distribution, small volume of distribution.
Metabolism and excretion of depolarising muscle relaxants?

What defects in excretion are there and what effect can this cause? Is there a test for this?
Onset of action 30s, duration of action 5-10 minutes due to rapid hydrolysis by pseudo- cholinesterase.

This limits the amount of drug reaching the synaptic cleft. Very little plasma cholinesterase at end plate neuromuscular blockade is terminated by diffusion away from the endplate into extracellular fluid.

95% of the population will have a normal pseudo- cholinesterase response. 5% will have prolonged apnoea up to 10 minutes. <<1% have profound apnoea lasting several hours - this may be treated with fresh frozen plasma.

Plasma cholinesterase may also be reduced in pregnancy, cardiac or renal failure, liver disease, hypoproteinaemia, and thyrotoxicosis -this will result in prolonged action.
Metabolism and excretion of non depolarising muscle relaxants?
Onset of action 2-3 minutes Route of elimination correlates with duration of action.

Renal excretion is slow, hepatic excretion fast.

Steroid -metabolised to 3 hydroxy, 17 hydroxy and 3,17 hydroxy metabolites that also have clinical effect and may persist.

Vecuronium - duration of action 20-35 minutes, minimal cardiovascular effects, hepatic elimination - 85% eliminated into bile.

Pancuronium -duration of action 35minutes - 1 hour, mainly excreted by kidney.

Rocuronium - very rapid onset of action.
What is the fastest onset non-depolarising muscle relaxant?
rocuronium
Mechanism of action of dantrolene?
Acts on the sarcoplasmic reticulum of skeletal muscle
Causes reduced release of calcium from the sarcoplasmic reticulum
What is malignant hyperthermia?
Malignant hyperthermia can be triggered by general anaesthesia and neuromuscular blockade and is a hereditary impairment of the ability to sequester calcium in the sarcoplasmic reticulum
Trigger results in massive release of calcium with prolonged muscle contraction, lactic acidosis and hyperthermia
What are the classes of antipychotics?
1. Phenothiazine derivatives: Chlorpromazine
2. Thioxanthene: Thiothixene
3. Butyrophenone derivatives : Haloperidol
4. Miscellaneous: Clozapine, Rispiradone, Olanzapine
Mechanism of action of antipsychotics?
Dopamine antagonist ␣ antipsychotic action related to dopamine receptor blockade in mesolimbic and mesofrontal systems.

Chlorpromazine Alpha1=5H T2>D2>D1
Haloperidol D2>D1=D4>Alpha1> 5H T2
Clozapine Alpha1 =D4 >5HT2> D2=D1
Rispiridone D2=5HT2
Olanzapine 5HT2>D2=D1=Alpha1> H1
Potency of various antipsychotics?
Chlorpromazine -low
Thiothixene - high
Haloperidol - high
Clozapine - medium
Rispiradone - high
Olanzapine - high
Dosing of antipsychotics?
Haloperidol Oral 1-15mg/day in divided doses. IM - 2-30mg IV - 1-5mg
Chlorpromazine PO/IM 25-50mg tds
Olanzapine 10mg PO/IM
CNS Toxicity of antipsychotics?
Toxicity CNS Extrapyramidal effects Extrapyramidal toxicity related to high D2 affinity; manifest as parkinsonism, akathisia, acute dystonic reactions, tardive dyskinesia.

Chlorpromazine -high
Thiothixene - medium
Haloperidol - very high
Clozapine - very low
Rispiradone - low
Olanzapine - very low

Sedation: esp chlorpromazine

Clozapine -2% incidence of seizures (can also occur with other antipsychotics)
ANS Toxicity of antipsychotics?
Autonomic effects Antimuscarinic actions. Loss of accommodation, dry mouth, difficulty urinating, constipation : Alpha1 antagonist actions.
CVS Toxicity of antipsychotics?
Tachycardia, reduced stroke volume, decreased peripheral resistance, orthostatic hypotension

Thioridazine causes T wave abnormalities and is associated with prolonged QT, ventricular arrhythmias and sudden death

most- chlorpromazine, least: olanzapine and haloperidol
What is NMS and how is it treated?
Neuroleptic malignant syndrome Muscle rigidity, reduced sweating, fever, autonomic instability, leukocytosis

Marked increases in CK may result in renal failure -use bromocriptine
Endo, haem effects of antipsychotics?
Amenorrhoea, galactorrhoea, increased or decreased libido, impotence -secondary to blockade of dopamine induced tonic inhibition of prolactin secretion.

Clozapine, chlorpromazine may cause agranulocytosis

Chlorpromazine - corneal and lens deposits
Absorption and distribution of antipsychotics?
Orally active. Readily but incompletely absorbed.

Chlorpromazine Significant first pass metabolism :bioavailability 30%. Half life 30 hours.

Others- Moderate first pass metabolism : bioavailability 65%

95% protein bound, lipid soluble, high distribution volumes
Presentation of antipsychotic overdose?
Drowsiness, coma, neuromuscular excitability, convulsions. Miosis and loss of deep tendon reflexes. Hypotension and hypothermia.
Activated charcoal effective.
Supportive therapy.

Avoid adrenaline and lignocaine.
Mechanism of action of lithium? 3 broad effects
1. Closely related to sodium -inhibits sodium exchange across membranes but no effect on sodium/potassium or sodium/calcium exchange.

2. Effects on neurotransmitters Enhances the action of serotonin. Decreases noradrenaline and dopamine turnover. Augments the synthesis of acetylcholine.

3. Effects on second messengers. Inhibits enzymes responsible for recycling of inositol compounds, resulting in depletion of PIP2, IP3 and DAG.
Usual maintenance dose of lithium
dose 500mg-1g/day
Interractions of lithium?
Renal clearance reduced by diuretics and some NSAIDs.
Increased extrapyramidal effects when used with antipsychotics (except newer drugs) May increase the duration of muscle relaxnants.
Lithium toxicity?
Tremor (alleviated by propanolol), choreoathetosis, motor hyperactivity, ataxia, dysarthria, aphasia.

Mental confusion, drowsiness and seizures at higher levels.

Endocrine: Reversible reduction in thyroid function.

GUS: Nephrogenic diabetes insipidus, chronic interstitial nephritis, minimal change glomerulonephritis.

CVS: Nodal depression and T wave flattening.

General Oedema and weight gain.
Lithium in pregnancy?
Clearance increases during pregnancy and falls following delivery.
??dysmorphogenesis - unsettled.

Lithium toxicity in newborns characterised by lethargy, cyanosis, poor suck and reflexes
Absorption and distribution of lithium?
Orally active, completely absorbed in 6-8 hours, peak levels 30 minutes. 100% bioavailability.

Distribution Total body water, slow entry into intracellular compartment. Some sequestration into bone.
Excretion of lithium?
Not metabolised. Excreted into urine at 20% of the rate of creatinine clearance.
Elimination half- life 20 hours.
Presentation and management of lithium overdose?
anorexia, nausea, vomiting, diarrhoea, muscle weakness, lack of cooordination

No specific antidote.

Clearance increased by osmotic diuresis and urinary alkalinisation. Readily removed by haemodialysis.
Give four examples of antidepressant classes?
tricyclincs- amytriptilline, imipramine
Heterocyclics:
SSRI:
MAOI's

Tricyclics Imipramine
Amitriptyline
Heterocyclics

Second generation: Maprotiline (tetracyclic) ,Buproprion

Third generation: venlafaxine

SSRIs
fluoxetine

MAOs: Phenelzine, Moclobamide
How do tricyclic antidepressants work?

Adverse effects?
Blockade of amine (serotonin >>noradrenaline) reuptake pumps.

Antimuscarinic actions Alpha1, H1, H2 antagonist

>1000mg toxic
CNS Sedation, seizures, psychosis, coma Antimuscarinic effects very common Tremor, insomnia
CVS Orthostatic hypotension Tachycardia and minor T/ST changes very common Conduction defects (long PR, wide QRS>0.1s, long QT and ST) and arrhythmias also common.
GIS Nausea, raised liver enzymes
How do heterocyclic antidepressants work?

Adverse effects?
Buproprion alters noradrenaline neurotransmission by an unknown mechanism. Third generation heterocyclics have additional antagonist of 5HT receptors.

Same as TCAs though less pronounced.
How do MAOI's work?

Adverse effects?
Blockade of MAO mediated amine degradation. MAO-A metabolises noradrenaline and serotonin, MAO-B metabolises dopamine.

Initial increase in amine leads to down-regulation of receptors.
Adverse effects of SSRI?
Anxiety, insomnia, tremor, nausea, rash (may lead to severe vasculitis), decreased libido. SSRI use in combination with MAO may lead to toxic build up of serotonin and serotonin syndrome
What is serotonin syndrome?
TCAs and SSRIs Serotonin syndrome if characterised by hyperthermia, muscle rigidity, myoclonus.
Antidepressant interactions with other drugs?
Phenothiazines displace TCAs from protein binding site and potentiate action.

Nefazodone can inhibit P450-3A4 and block the metabolism of terfenadine and cisapride.

F luoxetine and paroxetine are potent inhibitors of P450-2D6 -desipramine, nortriptiline and flecainide are dependent on same enzyme system for clearance.

MAOs : Accumulation of tyramine (fermented foods and drinks) results in hypertension. Moclobamide is relatively short acting compared with older drugs therefore this effect is rare
Absorption and distribution of tricyclics?
Incompletely absorbed. Slow gastric emptying due to antimuscarinic effect. Significant first pass metabolism (bioavailability 30- 70%)

80-90% protein bound. High lipid solubility. Large Vd
Absorption and distribution of tetracyclics?
Incompletely absorbed. Significant first pass metabolism (bioavailability 30- 70%)
80% protein bound. High lipid solubility. Large Vd
Absorption and distribution of SSRI's?
Well absorbed orally. Moderate first pass metabolism (bioavailability 50- 70%) 95% protein bound.
High lipid solubility. Large Vd
Absorption and distribution of MAOI's?
Well absorbed orally.
Metabolism of tricyclics, tetracyclics, MAOI's?
Tricyclics: Metabolised in liver to active metabolites. Half life 10-40 hours

Heterocyclics: Metabolised in liver to active metabolite. Half life 10-40 hours

SSRIs Metabolised in liver. Fluoxetine forms an active metabolite, nil with paroxetine. Half life 24-96 hours.

MAOs: Metabolised in liver. Acetylation of phenelzine varies between individuals and may persist for several weeks.
Antidepressant overdose?
Severe antimuscarinic response is common Extension of toxic effects listed plus respiratory depression, metabolic acidosis, heart failure, cardiac arrest
Give 4 examples of anticonvulsants that are sodium channel blockers/membrane stabilisers?
Phenytoin
Carbamazepine Sodium valproate Lamotrigine
Give 4 examples of anticonvulsants that are GABA modulators?
Benzodiazepines Gabapentin Vigabatrine Phenobarbitone
What kind of antiarrhythmic is phenytoin?
class 1b
Mechanisms of phenytoin?
Membrane stabiliser - preferentially binds to inactivated sodium channel and maintains inactivated state and therefore blocks sustained high frequency repetitive firing of action potentials.

Reduces calcium permeability therefore inhibits calcium related secretory processes.
May potentiate the effects of GABA

Inhibits release of serotonin and noradrenaline, promotes uptake of dopamine.

Inhibits monoamine oxidases.
Uses of phenytoin?
Prophylaxis and treatment of partial seizures and generalised tonic clonic seizures.
Fast atrial and ventricular arrhythmias resulting from digoxin toxicity.
Trigeminal neuralgia.
Dosing of phenytoin?
10-15mg/kg not exceeding 50mg/minute then 100mg orally every 8 hours

(slow administration due to propylene glycol diluent which may induce cardiac arrhythmias
Toxicity of phenytoin?
Dose related CNS: Nystagmus and loss of smooth ocular pursuit. Diplopia and ataxia. Sedation.

CVS: Cardiovascular collapse (diluent effect if rapid administration)

Idiosyncratic: Chronic use frequently leads to gingival hyperplasia and hirsutism, coarsening of facial features, diminished tendon reflexes and osteomalacia.
Rash, fever, rare agranulocytosis.
Interractions of phenytoin with other drugs?
Protein binding : Phenytoin is displaced by highly protein bound drugs such as sulphonamides, calcium channel blockers. Hypoproteinaemia causes increased free drug.

May confuse thyroid function tests due to affinity for TBG.

Enzyme inducers: Phenytoin induces liver enzymes and affects the metabolism of other drugs such as warfarin, opioids, neuromuscular blockers, beta blockers

Other inducers may reduce phenytoin levels - anticonvulsants, rifampicin, ciprofloxacin
Enzyme inhibitors Erythromycin, cimetidine
Absorption and distribution of phenytoin?
Orally active. Intramuscular absorption unpredictable. 90% bound to plasma proteins.
Maximal effect after IV dose occurs after 30-60 minutes and may persist for 24 hours
Accumulates in endoplasmic reticulum of brain, liver, muscle and fat.

Therapeutic concentration 10- 20ug/l
Where is phenytoin metabolised and what kind of kinetics does it display?
Metabolised by liver, excreted in bile, reabsorbed and excreted in urine.

Variable order kinetics: Elimination rate is dose dependent - at low doses there is first order kinetics but metabolism is saturated at therapeutic concentrations and small increases in dose quickly lead to toxicity.
Half life and steady state of phenytoin?
Half life 12-36 hours -much higher at high concentrations.

5-7 days to reach steady state.
Overdose of phenytoin
Overdose Toxicity varies between individuals CNS effects predominate though bradycardia and heart block can also occur
Mechanism of carbamazepine?
Membrane stabiliser - preferentially binds to inactivated sodium channel and maintains inactivated state and therefore blocks sustained high frequency repetitive firing of action potentials.
Also acts pre-synaptically to decrease synaptic transmission.

Inhibits reuptake and release of noradrenaline.
Interacts with adenosine receptors - ?significance
Toxicity of carbamazepine?
Skin rash common.
Diplopia, ataxia - often mild and reversible.
Drowsiness at high concentration.

Hyponatraemia and water intoxication rarely.

Rare agranulocytosis , leukopenia is common and just requires monitoring.
Interractions of carbamazepine?
Induces liver enzymes and affects own metabolism : half-life is typically halved from 40 to 20 hours with continuous therapy.

Phenytoin, carbamazepine, barbiturates and lamotrigine induce liver enzymes and reduce levels of each other.

Reduces effectiveness of benzodiazepines, pethidine, and warfarin.
Pharmacokinetics of carbamazepine?
Orally active

Rate of absorption variable but complete
Slow distribution. 70% protein bound.

Metabolised in liver -metabolite may have some clinical activity.
What is the active constituent of sodium valproate?
Fully ionised at body pH therefore active constituent is the valproate ion.
Mechanism of sodium valproate?
Membrane stabiliser - preferentially binds to inactivated sodium channel and maintains inactivated state and therefore blocks sustained high frequency repetitive firing of action potentials.

Increases levels of GABA - likely insignificant
Increases membrane potassium conductance
Toxicity of sodium valproate?
Dose related nausea vomiting and abdominal pain.
Sedation (particularly if used with phenobarbitone)
Tremor, weight gain, hair loss.

Idiosyncratic hepatotoxicity -may be fatal, more common in children under 2 years, usually occurs within 4 months of starting the drug.
Inerractions of sodium valproate?
Dose related nausea vomiting and abdominal pain.
Sedation (particularly if used with phenobarbitone)
Tremor, weight gain, hair loss.

Idiosyncratic hepatotoxicity - may be fatal, more common in children under 2 years, usually occurs within 4 months of starting the drug.
Absorption and distribution of sodium valproate?
Orally active. 80% bioavailability. 90% plasma protein bound. Distribution confined to extracellular water due to ionised status and protein binding.
Metabolism and excretion of sodium valproate?
Clearance is dose dependant - it inhibits its own metabolism at low doses. At higher doses there is increased free valproate.

20% excreted as a direct conjugate of valproate. 80% metabolised in liver and excreted in urine.
Half life 9-18 hours.
What does lamotrigine resemble in structure?
Phenytoin
Mechanism of lamortigine
Membrane stabiliser - preferentially binds to inactivated sodium channel and maintains inactivated state and therefore blocks sustained high frequency repetitive firing of action potentials.
Indication for lamictal
Partial seizures. Usually used in add- on therapy but increasingly used alone.
What is valproate best for?
Absence seizures. Generalised tonic- clonic seizures
Inerractons of lamortigine
`Phenytoin, carbamazepine, barbiturates and lamotrigine induce liver enzymes and reduce levels of each other.
Reduces effectiveness of benzodiazepines, pethidine, warfarin.
Side effects of lamrtigine
rash- if rash then cease lamortigine as linked to SJS, aseptic meningitis, leukopaenia, dizzyness, headache diplopia
Absorption and excretion of lamotrigine?
Orally active. 50% protein bound.

Linear kinetics. Metabolised by liver and excreted by urine.
Half life 24 hours, reduced to 15 hours if taking enzyme inducing drugs.
Mechanism of vigabatrine?

Absorption and excretion of vigabatrine?
Reversible inhibitor of GABA aminotransferase which degrades GABA. GABA levels are therefore increased. Indicated for partial seizures.

Orally active. Renally excreted. Short (5-8 hour) half life)
Toxicity of vigabatrine?
Dizziness, drowsiness and weight gain. Agitation and confusion rare
Structure and action of gabapentin?

Indications in epilepsy?
Amino acid analogue of GABA.

Despite structural relationship to GABA, does not act on GABA receptors but may alter GABA metabolism.

Partial seizures. Usually used in add- on therapy but increasingly used alone.
Interractions of gabapentin?
Does not induce liver enzymes. No significant interactions.
Absorption and excretion of gabapentin?
Orally active. Not protein bound.

Renally excreted. Short (5-8 hour) half life)
Mechanism of action of benzodiazepines?
Bind to GABA receptor in CNS

Benzodiazepines bind to the gamma subunit and in association with GABA, triggers chloride channel opening and subsequent hyperpolarisation.

Benzodiaepines increase the efficacy of GABAergic synaptic transmission -they do not cause chloride channel opening alone.
Structure of the GABA receptor in the CNS
GABA receptor consists of 5 alpha, beta and gamma membrane-spanning proteins which can form different pentameric combinations.
Dose of diazepam and midazolam used for sedation:
Diazepam Oral, IV or PR
Adult 5-40mg Child 0.1-0.3mg/kg

Midazolam
IV/IM 0.03-0.2mg/kg (higher dose used for induction of anaesthesia)
Does tolerance occur to resp depression with benzo's?
no
Do benzodiazepine cross breast milk and placenta?
yes
What interracts with benzodiazepines?
Effects potentiated by other sedatives such as alcohol - may cause fatal CVS and respiratory depression.
What does benzo absorption depend on?

Which benzodiazepines are best and worst absorbed?
Orally active. Absorption dependent on lipid solubility.

Diazepam most lipid soluble. Oxazepam, lorazepam and temazepam least lipid soluble. They are highly protein bound and CNS uptake dependent is also dependent on lipid solubility.
What is the timeline of sedation by temazepam
Peak sedation 15 minutes after IM injection, 4 minutes after IV injection
How are benzo effects terminated?
Redistribution from CNS to skeletal muscle and adipose tissue is important process that contributes to termination of CNS effects.
How are benzodiazepines metabolised?
Metabolised in liver to water-soluble metabolites that are excreted in urine.
Metabolites may be more active than parent drug (diazepam forms desmethyldiazepam which is transformed to oxazepam and temazepam)
What are the half lives of diazepam, temazepam and midazolam?
Diazepam -20-80 hours (metabolite persist much longer)
Temazepam -10-40 hours.
Midazolam 2-4 hours

Elimination may increase up to 6 times in elderly patients in ICU
What is the action of flumazenil?
Reversal of benzodiazepine induced clinical effect.
Reversal of respiratory depression is unpredictable.
Side effects of flumazenil?
Agitation, confusion, dizziness, nausea May precipitate severe withdrawal syndrome if existing physiological dependence.

Very likely to induce convulsions if patient has a known seizure disorder or in TCA overdose.
Absorption of flumazenil?
Undergoes significant first pass metabolism if administered orally.

Intravenous administration. Rapid onset.
50% protein bound. Small volume of distribution.
Metabolism of flumazenil?
Rapid hepatic metabolism to inert metabolites and excreted in urine.

Short half life 0.7- 1.3 hours. Infusion may be necessary.
What are the spinal and supraspinal sites of pain transmission?
Spinal
Dorsal horn pain transmission neurons of
spinal cord.

Pain transmission:
Ventral caudal thalamus
Diencephalon

Pain modulation:
Cortex
Midbrain periaqueductal gray area
Rostral ventral medulla
What is the mechanism of mu opioid receptors?

What are the subtypes?
Receptor binding causes closure of voltage gated calcium channels on presynaptic neurons and decreased transmitter release. Also causes opening of potassium channels on post synaptic neurons resulting in hyperpolarisation and formation of IPSPs

mu1, mu2
Action of mu opioid receptors?
Analgesia
Euphoria
Sedation
Respiratory depression
Tolerance and dependence
Agonists of mu opioid receptors
F ull: Morphine Pethidine Fentanyl
Partial: Codeine
Delta opioid receptors types and mechanisms?
delta 1 delta 2

Receptor binding causes closure of voltage gated calcium channels on presynaptic neurons and decreased transmitter release. This causes spinal analgesia.
Agonists of delta opioid receptors?
morphine and codeine are partial agonists
Subtypes and action of kappa opioid receptors
kappa 1, kappa 2 , kappa 3

Receptor binding causes closure of voltage gated calcium channels on presynaptic neurons and decreased transmitter release.
What are morphine, codeine and heroin classified as?
Phenanthrenes
What are the Phenylheptylamines?
methadone
What are Phenylpiperidines?
pethidine, fentanyl, loperamide
Can endogenous opioids cause release of exogenous opioids?
yes
What receptors does morphine act on?
mu, delta, kappa
What is paperveretum?
mixture of 253 parts morphine, 23 parts papaverine, 20 parts codeine - sometimes used as a premedication. Essentially identical actions as morphine though greater sedation and relief of anxiety.
Endocrine effects of opioids?
stimulates ADH, prolactin and somatotropin, inhibits LH

Histamine release- itching and urticaria
General effects of opioids on smooth muscles?
increase tone e.g.increased biliary smooth muscle contraction, increased GI smooth muscle contraction
Action of morphine in APO?
Reduced preload and afterload, reduced anxiety, reduced dyspnoea
Respiratory effects of opioids
respiratory depession
truncal rigidity- reduces thoracic compliance and increases work of breathing- likely supraspinal effect
Withdrawal to opioids?
rhinorrhea, lacrimation, yawning, chills, piloerection, hyperventilation, hyperthermia, mydriasis, muscular aches, vomiting, diarrhoea, and anxiety.

Onset 6-10 hours, Peak 36-48 hours, Lasts 5 days
Absorption of opioids?
Rapid absorption orally but high (and unpredictable) first pass metabolism therefore low bioavailability.

Variable protein binding.
Concentrates in highly perfused organs.

Structure determines how readily the drug crosses the blood/brain barrier -codeine and fentanyl cross quite readily, morphine less so.
Excretion of opiates?
Metabolised in liver and excreted in urine.
Morphine, codeine - metabolised in liver to active metabolites that may have greater activity than parent drug.

Excreted in urine. Small amount excreted in bile.
Metabolites may accumulate in renal failure.
Codeine mechanism?
Less efficacious - partial agonist at mu and delta receptors only.

Few side effects due to low potency. Constipation is prominent.
Absorption and metabolism of codeine?
The structure of codeine protects it from first pass effects and increases ability to cross the blood brain barrier.

10% is metabolised to morphine
Potency of pethidine?
One tenth as potent as morphine.
Metabolism of pethidine?

What interracts with this?
Metabolite 50% as active as parent drug - and is associated with seizures.

Halothane and enflurane decrease clearance by 50%.
Mechanism of fentanyl
highly selective mu agonist only
Toxicity of fentanyl:
CNS toxicity: Little hypnosis or sedation
Potent respiratory depression
Truncal rigidity
Marked muscle rigidity at high doses.

Peripheral toxicity:
Bradycardia with little effect on cardiac output or blood pressure.
Obtunds the cardiovascular response to intubation.
Minimal histamine release.

Halothane and enflurane decrease clearance by 50%.
Fentanyl potency, onset of action, duration of action?
IV or transdermal
33% bioavailability given PO

More lipid soluble than morphine therefore crosses the blood/brain barrier more readily.
Rapid onset of action.
50-80 times more potent than morphine.

Small dose has a shorter duration of action (1-1.5 hours) - larger doses 4-6 hours.
What is heroin metabolised to?
converted to morphine by tissue esterases
Tramadol mechanism?
Centrally acting analgesic with some opioid properties.
Not structurally related to opioids

Weak mu agonist actions
Noradrenaline inhibition and serotonin reuptake inhibition.
Dose of tramadol:
Mild to moderate pain
Dose PO/IV 50-100mg tds- rapidly orally absorbed
Side effects of tramadol?
Similar CNS and peripheral side effects

No respiratory depression except at high doses
No cardiac effects
No histamine release
Does not suppress opioid withdrawal symptoms
Increased risk of seizures

Avoid with SSRIs and MAOs due to actions on serotonin
Increased tramadol metabolism with enzyme inducers such as tramadol
Methadone potency?
Orally active, equivalent potency, longer duration of action

Tolerance and dependence develop and resolve more slowly and symptoms are milder.
Naloxone mechanism of action?

How does it act at the organ level?
Antagonist action at opioid receptors. More potent action at mu receptors than delta or kappa.

Inert if no agonist present. Antagonist action results in reversal of opioid induced effects in 1-3 minutes.
Duration of action of naloxone vs naltrexone?
naloxone: Extensive first pass metabolism therefore IV only. Short half life 1-2 hours (duration of action usually 20 minutes) therefore frequent dosing required in overdose

naltrexone: Longer half life than naloxone (10 hours) and will block the effects of injected heroin for 48 hours
Mechanism of ethanol?
No specific receptor.

Affects broad range of molecular processes including neurotransmission, enzyme action, electron transport chain, ion transport.

Enhances the action of GABA
Inhibits the action of glutamate
Oran effects of ethanol?
Intoxication. Myocardial depression. Smooth muscle relaxation.
Liver and GI toxicity of ethanol?
Increased alcohol metabolism leads to increased NADH/NAD+ ratio, resulting in reduced gluconeogenesis, hypoglycaemia, ketoacidosis and promotion of TG synthesis from FFA.
Accumulation of acetaldehyde promotes inflammation.

Nutritional deficiency is due to reduced intake and reduced absorption due to small bowel injury - reduces free radical scavengers such as glutathione
Increased gastric and pancreatic secretion, alteration of mucosal barriers resulting in gastritis.
Non-GI toxicity of ethanol?
CNS:
Nystagmus. Generalised symmetrical peripheral neuropathy. Gait disturbance and ataxia. Dementia. Wernicke-Korsacoff syndrome - paralysis of external ocular muscles, ataxia, acute confusion, memory loss.

CVS toxicity
Dilated cardiomyopathy, ventricular hypertrophy, fibrosis. Atrial and ventricular arrhythmias. Hypertension.

Blood:
Mild anaemia due to folate deficiency.

Immune system: Higher rates of infection

Foetal alcohol syndrome: Poor growth, microcephaly, poor coordination, flattened facial features, minor joint abnormalities, congenital heart defects.

Cancer: Mouth, pharynx, larynx, oesophagus, liver.

Interactions: Additive effect with other sedatives.

Acute use tends to inhibit enzymes, chronic use induces. Additive effect with hypoglycaemics and vasodilators.
Absorption and distribution of ethanol?
Small, water soluble molecule rapidly absorbed from GIT. Vd approximates TBW. Concentration in CNS rises quickly due to large blood flow.
Metabolism of ethanol?
90% metabolised by liver, 10% excreted by lungs and urine.
Metabolism follows zero order kinetics at approximately 1 standard drink per hour.
Ethanol converted to acetaldehyde by alcohol dehydrogenase in liver and stomach and by mixed function oxidase.

MFO activity increases in alcoholism.
Acetaldehyde converted to acetate by aldehyde dehydrogenase.

Acetete converted to carbon dioxide and water.
Mechanism of disulfram and the effect this causes with alcohol?
Inhibition of aldehyde dehydrogenase therefore aldehyde accumulates.

Flushing, throbbing, headache, nausea, vomiting, sweating, hypotension, confusion within a few minutes of alcohol consumption.
Interractions of disulfram with other drugs?
Interactions Inhibits the metabolism of phenytoin, warfarin, isoniazid.

Other drugs including metronidazole, cephalosporins may have disulfiram like effects.
Absorption and excretion of disulfram?
Orally active. Rapidly and completely absorbed

Slow elimination
How does methanol affect the human body?
No specific receptor. Affects broad range of molecular processes including neurotransmission, enzyme action, electron transport chain, ion transport.
Enhances the action of GABA. Inhibits the action of glutamate.
Symptoms of methanol consumption?
Visual disturbance predominates -may take 30 hours to be present. Formaldehyde may be detectable on breath.
Treatment of methanol intake?
Charcoal ineffective Supportive Intravenous ethanol- higher affinity for alcohol dehydrogenase therefore less formate.
Haemodialysis effective
Toxicity of methanol
Toxicity is probably related to the formation of formate. Visual disturbances (snowstorm sensation) is typical. CVS and CNS depression, metabolic acidosis.
Distribution of methanol?
Small, water- soluble molecule rapidly absorbed from GIT.

Vd approximates TBW. Concentration in CNS rises quickly due to large blood flow.
Treatment of ethylene glycol overdose?
Charcoal ineffective. Supportive.
Intravenous ethanol -higher affinity for alcohol dehydrogenase.
Consider alcohol dehydrogenase inhibitor - 4- methylpyrazole.
Haemodialysis effective.
Toxicity of ethylene glycol poisoning?
Transient excitation followed by CNS depression. Severe high anion gap metabolic acidosis after 4-12 hours.
Renal insufficiency due to deposition of oxalate.
Metacolism of ethylene glycol?
Converted to toxic aldehydes and oxalate by alcohol dehydrogenase.
Mechanisms of sodium chromoglycate?

Toxicity?
Alteration of function of chloride channels in cell membranes
Inhibition of mast cell degranulation
Likely inhibits mediator release from other cells
Used for asthma prevention through MDI or inhaler

toxicity: Reversible dermatitis, myositis or gastroenteritis in 2%
Theophilline structure and mechanism
Inhibits phosphodiesterase at high concentrations resulting reduced degradation of cAMP and high intracellular cAMP.
Increased intracellular cAMP leads to smooth muscle relaxation.
Increased cAMP in the heart leads to calcium influx and positive chronotropic and inotropic action
Inhibition of respiratory mucosal adenosine receptors which cause contraction of smooth muscle and histamine release
Blockade of cardiac adenosine receptors leading to increased catecholamine release
Possible anti-inflammatory action.
Dosing of theophilline in asthma?
Loading dose should be given slowly as rapid loading results in transient toxic plasma levels.
Dose Orally 3-4mg/kg tds
Tends to be avoided IV due to toxicities but can be given as an intravenous loading dose followed by infusion
Toxicity and interractions of theophilline?
Therapeutic and toxic effects closely related to blood level.
Therapeutic range 5-20mg/l

Anorexia, nausea, vomiting, headache at 15mg/l
Seizures or arrhythmias at >40mg/l

Interactions : Clearance enhanced by liver enzyme inducers -phenytoin, phenobarbitone.

Clearance reduced by propanolol, macrolides, cimetidine, OCP, calcium channel blockers.
Absorption and distribution of theophilline?
Absorption Well absorbed orally.
Distribution Volume of distribution is proportional to lean body weight.

Rapid intravenous loading causes transient toxic plasma levels
Metabolism of theophilline?

What type of kinetics does it display?
Metabolised by liver
(therefore reduced dose in liver disease, heart failure).

Variable order kinetics Elimination rate is dose dependent -at low doses there is first order kinetics but metabolism is saturated at therapeutic concentrations and small increases in dose quickly lead to toxicity.

Children clear theophylline most rapidly -poor in neonates and young infants
Mechanism of salbutamol at the molecular and cellular level?
B2 receptor agonists
Stimulation of adenylyl cyclase results in increased cAMP
Inhibits release of mediators

Bronchodilation due to smooth muscle relaxation. Inhibition of mediator release from mast cells. Inhibition of microvascular leakage. Increase mucociliary transport.
IV dosing of salbutamol?
Intravenous/SC
Adults Loading dose 200ug then 5-20ug/min
Children Loading dose 7.5ug/kg then 5ug/kg/hr
Toxicity of salbutamol
Predictable beta agonist actions
Tachycardia, palpitations, nausea, dizziness, tremor are common
May cause an initial reduced pO2 due to ventilation/perfusion mismatch

Hypokalaemia - especially high doses in combination with methylxanthines and diuretics
Hyperglycaemia and possible ketoacidosis in diabetics
Excessive use may result in tolerance
Metabolism of salbutamol
Metabolised by liver.
28% excreted unchanged by kidney
Half life 4 hours.
Salmeterol has long duration of action due to high lipid solubility rather than long duration of action
Mechanism of ipratropim
Muscarinic antagonist
Comtetitively inhibits the action of acetylcholine at muscarinic receptors
Toxicity of ipratropium
Predictable antimuscarinic effects though poor systemic absorption minimises this
Absorption and excretion of ipratropium
Absorption Poor systemic absorption
Quaternary ammonium structure leads to poor penetration into CNS.
10% of dose reaches airways, 90% swallowed.
Onset 1-3 minutes. Peak effect 1-2 hours.

90% excreted unchanged in faeces
Half life 3 hours.
List the common air pollutants
Carbon monoxide 52%
Sulphur oxides 14%
Hydrocarbons 14%
Nitrogen oxides 14%
Particles 4%
Toxicity of carbon monoxide?
Colourless, tasteless, odourless by product of incomplete combustion
Combines reversibly with haemoglobin to form carboxyhaemoglobin 220 times more avid binding than oxygen

An individual breathing air containing 0.1% CO (1000ppm) would have a carboxyhaemoglobin level of 50%
Treatment of carbon monoxide poisoning?
Removal of source ABC Oxygen
Room air at 1atm, elimination half life of CO 320 minutes

100% oxygen at 1atm elimination half life 80 minutes
100% oxygen at 2atm elimination half life 20 minutes
List 3 classes of insecticides and give examples?
Chlorinated hydrocarbons:
DDT Lindane

Organophosphates and carbamates:
Parathion Malathion

Naturally-derived insecticides:
Pyrethrum
Give an example of a herbicide
Paraquat
Give examples of commercially available cholinesterase inhibitors? list 3 types with examples

(Indirect-acting cholinoceptor stimulants)
Alcohols - edrophonium.
Carbamates - neostigmine, physostigmine
Organo- phosphates - parathion, malathion.

Interact with acetylcholinesterase and therefore blocks the hydrolysis of acetylcholine.
What are the mechanisms of the 3 types of cholinesterase inhibitors?

How long does the effect last for each of the 3 classes?
Alcohols - short lived reversible binding lasting 2- 10 minutes.

Carbamates -2 step hydrolysis to form a covalent bond, lasts 30mins-6hours

Organophosphates - initial hydrolysis results in a phosphorylated active site. This undergoes aging that involves strengthening of the phosphorus-enzyme bond that may last for hundreds of hours.
What are the effects of cholinesterase inhibitors at a molecular level?
Effects are similar to direct acting cholinomimetics. At the NMJ, therapeutic effects prolong and intensify the physiological action of acetylcholine resulting in increased strength of contraction.

Higher does may result in fibrillation.
What are the clinical indication for the 3 types of cholinesterase inhibitors?
Alcohols (edrophonium):
Diagnosis of myasthenia gravis = tensilon test.

Carbamates (neostigmine, physostigmine):
Reversal of non- depolarising neuromuscular blockade. Myasthenia gravis Glaucoma
Paralytic ileus Urinary retention

Organo-phosphates (parathion, malathion): Glaucoma
Side effects of cholinesterase inhibitors (describe muscarinic and nicotinic effects separately)
Muscarinic agonists: nausea, vomiting, diarrhoea, salivation, sweating, vasodilation, bronchoconstriction.

Nicotinic agonists: CNS stimulation, convulsions, coma, flaccid paralysis, hypertension and cardiac arrhythmias.

Organophosphates may also cause delayed neurotoxicity
Absorption and distribution of
1) carbamates?
2) organophosphates?
Carbamates - absorption poor due to permanent charge and lipid insolubility. Physostigmine has better absorption due to tertiary amine group.

Organophosphates - well absorbed from skin, lung, gut, conjunctiva and distributed into CNS.

Distribution:

Carbamates - distribution in CNS negligible. Physostigmine is widely distributed.

Organophosphates - widely distributed including the CNS.
Metabolism and excretion of
1) carbamates?
2) organophosphates?
Carbamates: majority of the dose is excreted in the urine.

Organo-phosphates: Malathion is rapidly metabolised to inactive products and therefore relatively safe. Parathion is metabolised less effectively and is therefore more toxic.
What are 2 naturally occuring antimuscarinic agents?
Atropine: found in Atropa belladonna (deadly nightshade) and datura stramonium

Hyoscine: found in hyoscyamus niger
What are the 2 types of synthetic antimuscarinic agents?
Tertiary amines - pirenzepine, tropicamide

Quaternary amines - propantheline, glycopyrrolate, ipratropium, benztropine.
How does atropine work?
Competitive antagonist of acetylcholine at muscarinic receptors. Reversible blockade.

No distinction between M1,2 and 3

Salivary, bronchial and sweat glands are the most sensitive.
What are the organ effects of atropine?
Mydriasis
Cycloplegia
Decreased lacrimal secretion
Tachycardia (note - may cause initial bradycardia at low dose)
Increased contractility
Arterial constriction (Dilation - high dose direct effect)
Venoconstriction (Dilation - high dose direct effect) Bronchiolar smooth muscle relaxation
Decreased mucus secretion
Gut relaxation
Contraction of sphincters
Decreased salivary, gastric and pancreatic secretion.
Bladder wall relaxation
Bladder sphincter contraction Uterus smooth muscle relaxation
Decreased sweating
Drowsiness, confusion, hallucinations, dysarthria
Clinical uses of antimuscarinic agents?
Bradycardia due to increased vagal tone, including cardiac arrest -Atropine

Cholinomimetic (direct or indirect) poisoning -Atropine

Mydriasis -Tropicamide, Atropine

Motion sickness -Hyoscine

Bronchodilation -Ipratropium

Diarrhoea - Atropine

Urinary urgency -Oxybutynin
Toxicity of antimuscarinic agents?
In adults, toxic effects are an extension of the clinical effect.
Children are sensitive to hyperthermic effects that are centrally mediated.

Produce very similar effects to LSD in high doses though delusions tend to be bizarre. Effects are very long acting (several days).

Delerium, fluctuating level of awareness, difficulty in thinking and marked loss of memory are particularly characteristic.
Contraindications Glaucoma.
Absorption and distribution of antimuscarinic agents?
Absorption
Naturally occurring agents: Well absorbed from gut, skin and conjunctival membranes.

Synthetic agents (quaternary amines) : only 10-30% absorbed orally.

Distribution:
Naturally alkaloid esters of tropic acid - widely distributed.
Synthetic antimuscarinic agents (quaternary amines) - mostly peripheral distribution.
Metabolism and excretion of antimuscarinic agents?
Metabolised in liver and excreted in urine
Half life 4 hours
What is pralidoxime?
Cholinesterase regenerator:
The Acetylcholinesterase enzyme has two parts to it. An acetylcholine molecule bound at both ends to both sites of the enzyme, is cleaved in two to form acetic acid and choline. In organophosphate poisoning, an organophosphate binds to just one end of the acetylcholinesterase enzyme [ the esteric site ], blocking its activity. Pralidoxime is able to attach to the other half [ the unblocked, anionic site ] of the acetylcholinesterase enzyme.It then binds to the organophosphate, the organophosphate changes conformation, and loses its binding to the acetylcholinesterase enzyme. The conjoined poison / antidote then unbinds from the site, and thus regenerates the enzyme, which is now able to function again.

After some time though, some inhibitors can develop a permanent bond with cholinesterase, known as aging, where oximes such as pralidoxime can not reverse the bond
What is pralidoxime?
Hydrolysis of phosphorylated acetylcholinesterase
Slows aging process
Pralodoxime is only effective if aging has not occurred
Dosing of pralidoxime?
Pralidoxime is initially administered intravenously in a dose of 1 to 2 g. Signs of recovery appear rapidly. If the symptoms reappear, then an infusion of 2.5% is infused at a rate of 0.5 g/hour.
What is paraqat and why is it toxic?
herbicide

Paraquat forms a potent free radical that accumulates in the lung: Oedema, alveolitis, progressive fibrosis
Lethal dose of paraqat
50-500mg/kg
Toxidrome of paraqat?
Initial gastrointestinal symptoms Delayed onset respiratory distress Death may occur after several weeks Oxygen aggravates pulmonary effects
How does arsenic affect tissues and what effects does this cause?
Inhibition of enzymes of oxidative phosphorylation

Shock Arrhythmias Encephalopathy Peripheral neuropathy Pancytopenia
How is arsenic absorbed and excreted?
absorbed: All mucosal surfaces Widely distributed
Excreted by kidney
How does lead affect tissues and what effects does this cause?
inorganic oxides: Inhibition of enzymes, interferes with essential cations, alters membrane structure

Inorganic oxides and salts: Anaemia, peripheral neuropathy, nephropathy, hypertension
Organic: Encephalopathy
How is lead absorbed and excreted?
Inorganic oxides and salts: Gastrointestinal and respiratory tracts
Organic: All mucosal surfaces

Excretion:
Organic -Metabolised by liver to lead, excreted in urine and faeces
How does mercury affect tissues and what effects does this cause?
Inhibition of enzymes and membrane alterations

Elemental:
Behavioural disturbance (erethism) Gingivostomatitis
Peripheral neuropathy
pneumonitis

Inorganic: (Hg+ less toxic than Hg2+)
ATN

Organic:
CNS effects, birth defects
How is mercury absorbed?
Elemental: Respiratory tract

Inorganic:
Gastrointestinal tract
Skin

Organic:
All mucosal surfaces
Tend to concentrate in soft tissues, especially kidney
How is mercury excreted?
Elemental: Converted to Hg2+
Excreted in urine and faeces

Inorganic: Excreted in urine
Symptoms of iron toxicity
Nausea Epigastric pain Abdominal cramps Constipation Diarrhoea
Black stools
Symptoms of iron overdose?
Seen commonly in young children (10 tablets can be lethal)
Necrotising gastroenteritis with vomiting, abdominal pain and bloody diarrhoea
Shock

Improvement followed by severe metabolic acidosis, coma and death
Treated with desferrioxamine
What is desferrioxamine?
Used for iron poisoning

Avidly binds to iron
Competes for iron binding with haemosiderin and ferritin

Iron-chelator complex is excreted in urine
Side effects of desferroxamine?
Flushing
Gastrointestinal symptoms
ARDS
Toxic dose of tricyclic antidepressants?
>1000mg
Toxic side effects of tricyclic antidepressants?
1. CNS: Psychosis, sedation, seizures, coma
Antimuscarinic
Sympathomimetic: Tremor, insomnia

2. CVS: Orthostatic hypotension
Conduction defects (long PR, wide QRS>0.1s, long QT and ST), arrhythmias.

3. Respiratory depression and apnoea.

4. Metabolic acidosis
Toxicity of salycilates?
1. Salicylism: tinnitus, reduced hearing, vertigo.
2. Hyperventilation, fever, dehydration
3. Metabolic acidosis due to salicylic acid dissociation, deranged carbohydrate metabolism and reduced renal function.
4. Respiratory alkalosis due to central stimulation of respiratory centre Eventual renal and respiratory failure
Norml anion gap?
12-16meq/L

(Na+ + K+) - (HCO3- + Cl-)
What causes an increased anion gap metabolic acidosis?
Methanol
Ethylene glycol
Lactic acid
Cyanide
Carbon monoxide
Salicylates
Metformin
What is the osmolar gap and what are some causes of increased osmolar gap?
Alcohols:
ethanol intoxication
methanol ingestion
ethylene glycol ingestion
acetone ingestion
isopropyl alcohol ingestion

Sugars:
mannitol
sorbitol
glucose (in those with insulin resistance, such as diabetics)

Lipids:
Hypertriglyceridemia

Proteins:
Hypergammaglobinemia (M. Waldenström)
What is the calculated osmolality?
2 x [Na mmol/L] + [glucose mmol/L] + [urea mmol/L]
Causes of increased anion gap metabolic acidosis?
Causes include:
lactic acidosis
ketoacidosis
chronic renal failure (accumulation of sulfates, phosphates, urea)

intoxication:
organic acids (salicylates, ethanol, methanol, formaldehyde, ethylene glycol, paraldehyde, INH)
sulfates, metformin (Glucophage)
massive rhabdomyolysis
Causes of normal anion gap metabolic acidosis?
U - ureterosigmoidostomy
S - saline administration (in the face of renal dysfunction)
E - endocrine (Addisons, spironolactone, triamterene, amiloride, primary hyperparathyroidism)
D - diarrhea
C - carbonic anhydrase inhibitors
A - ammonium chloride
R - renal tubular acidosis
P - pancreatitis
Is peritoneal dialysis an option for getting rid of drugs?
no, not effective for most drugs
What determines whether a substance is dialisable?
Molecular weight
Water solubility
Protein binding
Endogenous clearance
Volume of distribution (small)
What drugs is dialysis ineffective for?
Amphetamines, cocaine
Benzodiazepines
Phenothiazines
Digoxin
Opioids
Quinidine
Tricyclics
What drugs is dialysis effective for?
Ethylene glycol
Methanol
Salicylate
Theophylline
Procainamide
Features of benzo and barbiturate withdrawal?
Withdrawal - agents with short half lives produce rapidly evolving severe withdrawal. Longer half life produces gradual, less severe withdrawal. Withdrawal similar to alcohol - agitation, nausea and vomiting reduced seizure threshold, delirium, psychosis.
Mechanism of caffeine as a stimulant?
Inhibits phosphodiesterase at high concentrations resulting reduced degradation of cAMP in high intracellular cAMP. Possible inhibition of adenosine receptors.
Mechanism of nicotine?
Causes release of catecholamines from central and peripheral nerves.
Produces insidious onset central euphoriant effect. Withdrawal characterised by pronounced and long lasting craving.
Mechanism of cocaine?
Inhibition of dopamine and noradrenaline reuptake Produces marked increased mental alertness and euphoria then may progress to delusions and psychosis.
Short acting compared with amphetamines but magnified effect.
Mechanism of amphetamine?
Cause central increased catecholamine neurotransmitter release Produces marked increased mental alertness and euphoria then may progress to delusions and psychosis

Withdrawal - lethargy, increased appetite, depression

Methamphetamine - speed
Methyeledoxymethamphetamine - ecstasy
Mechanism and effects of LSD?
The psychedelic effects of LSD are attributed to its strong partial agonist effects at 5-HT2A receptors but exact mechanism unknown. LSD affects a large number of the G protein-coupled receptors, including all dopamine receptor subtypes, and all adrenoreceptor subtypes, as well as many others.

Mechanism unknown ␣ interacts with several serotonin receptor subtypes. Produces dizziness, weakness, tremors, nausea and prominent visual illusions and other perceptive abnormalities. Also causes pupillary dilation, tachycardia and increased blood pressure.
PCP mechanism and effects?
Related to ketamine.

May act at opioid, dopamine or glutamate receptors. NMDA antagonist, D2 partial agonist, nAchR antagonist.

Produces detachment, disorientation, distortion of body image, nystagmus (vertical and horizontal), sweating, tachycardia, hypertension. Overdose can be fatal (in contrast with LSD).
Effect of antimuscarinics (in drug abuse setting)?
Produce very similar effects to LSD in high doses though delusions tend to be bizarre.

Effects are very long acting (several days). Delirium, fluctuating level of awareness, difficulty in thinking and marked loss of memory are particularly characteristic.
Pharmacological profile of marijuana?
Where does it act?
Contains several cannaboids including tetrahydrocannabinol (THC). Stimulates a specific receptor located in basal ganglia, substantia nigra, globus pallidus, hippocampus and brain stem. May also have a non-specific membrane effect.
Produces euphoria and characteristic uncontrollable laughter, alteration of time sense, sharpened vision followed by extreme relaxation and dream like states. Tachycardia and conjunctival reddening are characteristic.

Therapeutic use likely to increase due to antiemetic and
analgesic actions
How does activated charcoal work as a decontaminant?
Large surface area available to adsorb many drugs and poisons. 1 gram has a surface area of 1000m2 Manufactured by heating charcoal under pressure. Most effective in a 10:1 ratio of charcoal to poison.
What substances is activated charcoal not effective for?
does not bind:

Ions - lithium, potassium, cyanide
Heavy metals - iron Hydrocarbons
Acids/alkalis
Alcohols: ethanol, methanol
What is whole bowel irrigation and what substances is it useful for?
Balanced polyethylene glycol-electrolyte solution: used for iron, enteric coated medications, foreign bodies.
Antidote to paracetamol?
acetylcysteine
Antidote for anticholinesterases? (organophosphates and carbamates)?
pralidoxime
Antidotes of tricyclic antidepressants and quinine?
bicarbonate
Antidote for iron salts?
desferrioxamine
Antidote for digoxin?
digibind, digoxin antibodies
Antidote for methanol and ethylene glycol?
rthanol
Antidote for benzodiazepines?
flumazenil
Antidote for beta blockers?
glucagon
Antidote for opioids?
naloxone
Antidote for carbon monixide?
oxygen
Antidote for antimuscarinics?
physostigmine
Mechanism of aspirin?
Reduced synthesis of eicosanoid mediators:
Irreversible inhibition of cyclooxygenase
Reduced synthesis of thromboxane A2 Reduced synthesis of prostaglandins

Central blockade of CNS response to IL1 in causing fever
Effects of aspirin and how long does it last?
Antiplatelet
action lasts for the lifespan of the platelet - thromboxane A2 stimulates platelet aggregation and granule release

Antiinflammatory Analgesic Antipyretic
Effects of aspirin at therapeutic, anti-inflammatory and toxic range?
1. Therapeutic range
0-10mg/kg
Gastritis, ulceration Impaired haemostasis

2. Anti-inflammatory range 50mg/kg
Salicylism: tinnitus, reduced hearing, vertigo.

3. Toxic range 50-150mg/kg hyperventilation, fever, dehydration, metabolic acidosis

4. Serious intoxication >150mg/kg
Respiratory alkalosis due to central stimulation of respiratory centre
Renal compensation for respiratory alkalosis. Metabolic acidosis due to salicylic acid dissociation, deranged carbohydrate metabolism and reduced renal function.
Eventual renal and respiratory failure
Interactions of aspirin with other drugs?
Displaces from protein binding (phenytoin, methotrexate)
Decreased activity of spironolactone
Decreased tubular secretion of penicillin
Absorption and distribution of aspirin?
Orally active Rapidly absorbed. Acidity of stomach keeps aspirin in nonionised form that is more readily absorbed

Bound to albumin in low doses. As serum concentration rises, increasing fraction is unbound
Metabolism and excretion of aspirin?
Hydrolysed to acetic acid and salicylate by blood and tissue esterases.
Salicylate conjugated by liver and excreted by kidney.
Demonstrates variable order kinetics ␣ metabolism is saturable and small further increases in aspirin dose results in large rise in salicylate levels.
Half life 3-5 hours at low dose, 12 hours at anti- inflammatory doses

Alkalinisation of the urine increases rate of excretion of free salicylate
Haemodialysis indicated in severe toxicity
Mechanism of nsaids?
Reduced synthesis of eicosanoid mediators
Reversible inhibition of cyclooxygenase (COX1, COX2 or both)
Reduced synthesis of thromboxane A2 Reduced synthesis of prostaglandins

Prostaglandins are important mediators of inflammation

Inhibition of mediator release from leukoctes
Decreased sensitivity of vessels and pain sensors to bradykinin and histamine
Central blockade of CNS response to IL1 in causing fever
Toxocity of ibuprofen?
Gastritis, ulceration and minor gastrointestinal disturbance
Impaired haemostasis
Nephrotoxicity and reduced renal function in those with renal disease

Hepatotoxicity and increased liver enzymes in those with hepatic disease
Oedema, especially if pre- existing heart failure
Visual disturbances
Aseptic meningitis
What does ibuprofen interact with?
Warfarin: risk of fatal haemorrhage due to displacement from albumin
Lithium/digoxin: increased plasma levels Antihypertensives: reduced effect
Contraindications of ibuprofen?
NSAID/aspirin sensitive asthma
3rd trimester of pregnancy: may cause closure of the fetal ductus arteriosus, fetal renal impairment, inhibition of platelet aggregation and delay labour and birth
Absorption of ibuprofen?
Orally active
Rapidly absorbed.
Absorption slowed by food
Highly protein bound
Metabolism and excretion of ibuprofen?
Metabolised by liver to inactive metabolites.
Excreted in urine and bile
Half life 2 hours
Dising of naproxen and toxicity of naproxen?
10mg/kg in 2 divided doses slow release formulation available

may impair feritlity
Excretion of naproxen
Mostly excreted unchanged Half life 12 hours
Mechanism of indomethasin, ketoprofen and diclofenac?
Indomethacin, ketoprofen and diclofenac inhibit lipoxygease and therefore reduce formation of leukotrienes

ketoprofen Half life 2 hours
Mechanism of mefanamic acid?
May antagonise the actions of prostaglandins PGE2 and PGF2alpha at uterine receptors
Is indomethacin more or less potent than aspirin?

Most effective absorption?
Potent prostaglandin inhibitor 28 times more potent than aspirin

PR more rapid
T 1/2 4 hours
Indications for indomethacin?
Patent ductus arteriosus
Gout
Preterm labour (though may cause closure of the ductus, renal toxicity, delayed labour, impaired haemostasis

Dose 50-200mg/day in 2- 3 divided doses
Indications and dosing for kerolac?
Short-term management of post operative pain
Equally effective as 10mg morphine/100mg pethidine IM or panadeine forte
Dose 10-30mg IM every 6 hours for a maximum of 5 days
Mechanism of paracetamol?
Para-aminophenol derivative
Weak prostaglandin inhibitor
Probably has COX3 antagonist actions in the CNS
Symptoms of paracetamol toxicity/overdose?
What doses typically cause this?
Toxic symptoms include vomiting, abdominal pain, hypotension, sweating, central stimulation with exhilaration and convulsions in children, drowsiness, respiratory depression, cyanosis and coma.
Hypokalaemia and ECG changes have also been noted
In adults, hepatotoxicity may occur after ingestion of a single dose of paracetamol 10 to 15 g
25 g is potentially fatal.
Symptoms during the first two days of acute poisoning by paracetamol do not reflect the potential seriousness of the intoxication.

Major manifestations of liver failure such as jaundice, hypoglycaemia and metabolic acidosis may take at least three days to develop.
Medications which interact with paracetamol?
Alcohol and enzyme inducers :increased risk of toxicity
Absorption of paracetamol?
Orally active
Peak blood levels 30- 60 minutes Food intake delays paracetamol absorption. Partially protein bound
Metabolism of paracetamol?
Metabolised in liver In adults at therapeutic doses, paracetamol is mainly conjugated with glucuronide or sulfate.
Also metabolised to a toxic metabolite (cyp3a4 and cyp2a1) that is detoxified by conjugation with glutathione
Excreted in the urine

Elimination half- life varies from one to three hours.
Overdose Activated charcoal IV fluids
If 15g or more ingested, acetylcysteine
Mechanism of paracetamol toxicity?
Paracetamol is metabolised in the liver, mainly by conjugation with glucuronide and sulfate. It is also metabolised by cytochrome P450 to form a reactive, potentially toxic, metabolite.
This metabolite is normally detoxified by conjugation with hepatic glutathione, to form nontoxic derivatives.
In paracetamol overdosage, the glucuronide and sulfate conjugation pathways are saturated, so that more of the toxic metabolite is formed.

As hepatic glutathione stores are depleted, this toxic metabolite may bind to hepatocyte proteins, leading to liver cell damage and necrosis.
Mechanism of NAC in paracetamol toxicity?
Acetylcysteine is a sulfydryl (SH) group donor, and may protect the liver from damage by restoring depleted hepatic reduced glutathione levels, or by acting as an alternative substrate for conjugation with, and thus detoxification of, the toxic paracetamol metabolite.
Dosing of NAC in paracetamol overdose?
8 hours or less since overdose ingestion.
Initial dose 150 mg/kg over 15 minutes, followed by continuous infusion of 50 mg/kg in glucose 5% 500 mL over four hours and 100 mg/kg in glucose 5% 1 L over 16 hours.

If more than eight hours have elapsed since the overdosage was taken, the antidote may be less effective.

Rumack-Matthew nomogram gives indication of likelihood of toxicity as a function of time since ingestion and plasma levels
NAC toxicity and contraindications?
Nausea and vomiting
Allergic reactions
Tachycardia, chest pain

Contraindications: Asthma, renal and hepatic failure - administer with caution
Mechnism of colchicine?
Binds to intracellular tubulin, therefore inhibiting leucocyte migration and phagocytosis (also anti- mitotic)
Inhibits formation of leukotriene B4
Inhibits urate crystal deposition
Dosing of colchicine?
Acute gouty arthritis
Dose 0.5-1mg then 0.5mg every 2 hours until pain is relieved or diarrhoea occurs
Do not exceed 8mg
Toxicity of colchicine?
Diarrhoea (80% in 8-12 hours)
Nausea and vomiting
Bone marrow suppression (especially in overdose) inhibition of B12 absorption
Contraindications of colchicine?
Hepatic or renal disease
Cardiac disease
Gastrointestinal disease
Toxicity of colchicine? Describe the dose and the two phases of toxicity
Toxic dose >0.5mg/kg

Latent period- 2-12 hours:
Burning throat pain, bloody diarrhoea, dehydration

Second phase 24-72 hours: Shock, renal failure, muscular weakness and ascending paralysis, bone marrow suppression, DIC, multiorgan failure
Absorption and distribution of colchicine?
Rapidly absorbed
Bioavailability 25- 50%
Metabolism of colchicine?
Metabolised by liver, excreted in bile and urine.
Some enterohepatic circulation
Half life 4 hours
Management of colchicine overdose
Activated charcoal
Supportive measures
Mechanism of alloprinol?
Xanthine oxidase inhibitor
Urate formed from amino acids and purines
Xanthine oxidase is required for formation of urate therefore its inhibition will decrease production
Toxicity of allopurinol?
May precipitate acute gout unless given with colchicine or probenicid
Rash
Nausea and vomiting
Bone marrow depression
Impaired renal function
Impaired hepatic function
Absorption of allopurinol?

Metabolism of allopurinol?
80% absorbed not bound to plasma proteins

Metabolised by xanthine oxidase which it also inhibits
Half life 1-2 hours
Describe the anatomy of sympathetic pre and post ganglionic fibres?
Sympathetic preganglionic fibres terminate in ganglia in paravertebral chains.
Post ganglionic fibres then pass to the organs
Describe the anatomy of parasympathetic pre and post ganglionic fibres?
Parasympathetic fibres: some terminate in parasympathetic ganglia (ciliary, pterygopalatine, submandibular, otic, pelvic), majority terminate in organs.
Where is acetylcholine found as a neurotransmitter?
All pre-ganglionic autonomic
All parasympathetic post-ganglionic
All somatic motor
How is acetylcholine made? Where do the components come from?

What can block this production?
Acetylcholine synthesised in cytoplasm from acetyl-CoA (made in mitochondria) and choline (transported from extracellular fluid by sodium dependent carrier) - enzyme=choline acetyltransferase.

Hemicholiniums block choline carrier
Where is acetycholine stored?

What can block the storage?
Acetylcholine transported from cytoplasm to storage vesicle by proton antiporter.

Vesamicol blocks antiporter.
What causes acetylcholine release?
What proteins are involved?
What blocks this?
Calcium influx causes the vesicle to fuse with the terminal membrane and release acetylcholine into the synaptic cleft.

Synaptobrevin, SNAP and syntaxin required.

Botulinum toxin blocks release by enzymatic removal of 2 amino acids from synaptobrevin.
What terminates the action of acetylcholine?
Acetylcholine degraded by acetylcholinesterase into acetate and choline.
Where is noradrenaline found?
Most post-ganglionic sympathetic
How is noradrenaline synthesised?

What inhibits this?
Tyrosine carried into cell by sodium dependent carrier.
Converted to DOPA by tyrosine hydroxylase.
DOPA converted to Dopamine by DOPA decarboxylase.

Metyrosine inhibits action of tyrosine hydroxylase.
What blocks the storage of dopamine in vesicles?
Dopamine transported into vesicle by a carrier. Reserpine blocks carrier.
What is the mechanism of noradrenaline release into the synapse?

What drugs potentiate this? What drugs block this?
Calcium influx causes the vesicle to fuse with the terminal membrane and release noradrenaline into the synaptic cleft. ATP and dopamine beta hydroxylase are also released into the cleft.

Tyramine and amphetamines are capable of noradrenaline release by a displacement process that is not calcium dependent.
Release can be blocked by bretylium and guanethadine
What terminates the action of noradrenaline at the synapse?

What stops this termination?
Noradrenaline diffuses away from the cleft or is transported back into the cytoplasm or into the post-junctional cell.
Noradrenaline is then metabolised by MAO and COMT.
Reuptake is blocked by cocaine and tricyclic antidepressants.
Where are muscarinic M1 receptors found?
CNS, sympathetic postganglionic, some pre-synaptic
What is the result of M1 muscarinic receptor binding?
IP3, DAG, increased calcium
Where are M2 muscarinic receptors found?
heart, smooth muscle, endothelium, some pre- synaptic
What is the binding result if M2 muscarinic receptors?
Inhibit adenylyl cyclase, open potassium channels, stimulates release of EDRF.
Where are M3 muscarinic receptors found and what is the result of receptor binding at M3 receptors?
Exocrine glands, vessels, eye, lungs, GIT, bladder

IP3, DAG, increased calcium
Where are nicotinic N receptors bound and what does binding result in?
Postganglionic neurons, some pre-synaptic cholinergic terminals

Open sodium and potassium channels, depolarisation
Where are nicotinic M receptors found and what is the result of activation of these receptors?
Skeletal muscle endplates
Open sodium and potassium channels, depolarisation
Where are alpha 1 adrenoceptors found and what is the result of binding to these?
mostly smooth muscle IP3, DAG, increased calcium
Where are alpha 2 adrenoceptors found and what is the result of binding to these?
pre-synaptic terminals, platelets, lipocytes, smooth muscle inhibition of adenylyl cyclase, reduced cAMP
Where are beta 1 adrenoceptors found and what is the result of binding to these?
heart, brain, lipocytes plus presynaptic stimulation of adenylyl cyclase, increased cAMP
Where are beta 2 adrenoceptors found and what is the result of binding to these?
heart, lungs, smooth muscle stimulation of adenylyl cyclase, increased cAMP
Where are beta 3 adrenoceptors found and what is the result of binding to these?
lipocytes stimulation of adenylyl cyclase, increased cAMP
What receptor dilates the pupil

i.e. constricts radial (pupillary dilator) muscle of iris?
A1
What receptor constricts pupil?

i.e. constricts circular (pupillary constrictor) muscle
M3
What receptor contracts the ciliary muscle?
M3
What receptor increases aqueous humour?
B
What receptors increase and decrease heart rate at the sa node?
B1, B2 (acellerate)
M2 (decellerate)
What receptors increase cardiac contractikity?
B1, A1, B2 (increase)
M2 (decrease)
What receptor contracts Skin, splanchnic blood vessels?
A1
What receptor relaxes skeletal muscle blood vessels?
B2
What adrenoceptor aggregates platelets?
A2
What adrenoceptors relax and contract gut wall?
relax a2, b2
contracts M3

sphincters contract a1, relax m3
What adrenoceptor increases secretions (salivary, gastric, pancreatic)?
M3
What adrenoceptor causes
1. gluconeogenesis?
2. glycogeniolysis?
3. Lipolysis?
4. Decreases lipolysis?
1. B2, A
2. B2, A
3. B3
4. A2
What adrenoceptor increases renin release at the kidney?
b1
What adrenoceptor causes
1. bladder wall contraction?
2. Bladder wall relaxation?
3. Bladder sphincter contraction?
4. Bladder sphincter relaxation
1.B2
2. M3
3. A1
4. M3
What adrenoceptor causes uterine contraction and relaxation?
relax: b2
contract: A, M3
What adrenoceptor causes
1) piloerection in the skin?
2. Increased sweating?
1) A1
2) A, M
Describe the 2 classes of cholomimetic drugs with examples?

Centrally mediated alerting action, tremor, emesis, convulsions.
Choline esters: Acetylcholine (both), Methacholine (muscarinic), Carbachol (both), Bethanecol (muscarinic)

Alkaloids: Pilocarpine (muscarinic), Nicotine (nicotinic), Lobeline (nicotinic), Muscarine (muscarinic).
Indications for cholomimetic drugs?
1. Glaucoma: pilocarpine, methacholine, carbachol.
2. Paralytic ileus: bethanechol
3. Urinary retention: bethanechol
4. Antimuscarinic drug intoxication.
Absorption and distribution of choline esters and alkaloids?
Choline esters: Poorly absorbed orally.
Alkaloids: Well absorbed orally and transcutaneously.

Distribution: Choline esters Hydrophilic therefore poorly distributed in CNS
Alkaloids: Nicotine very lipid soluble therefore widely distributed in CNS
Metabolism and excretion of choline esters and alkaloids?
Choline esters: Hydrolysed in GIT (acetylcholine more than others, carbachol and methacholine negligible)

Alkaloids: Renally excreted, increased by acidification of the urine.
What are the 2 types of direct acting sympathetomimetics?

Give examples of these?
catecholamine and non-catecholamine

Catecholamines:
Adrenaline, Noradrenaline, Isoprenaline, Dopamine Dobutamine

Non-catecholamines:
Ephedrine
Phenylephrine, Amphetamine

Indirect acting sympathomimetics:
Cocaine
Tyramine
Give an example of an alpha 1 agonist and antagonist.
agonist: Phenylephrine A1>A2>>>>>B
antagonist: prazocin
Give an example of an alpha 2 agonist and antagonist?
Agonist: Clonidine A2>A1>>>>>B
antagonist: Yohimbine
Give an example of an beta 1 agonist and antagonist?
agonist: Dobutamine B1>B2>>>>A
antagonist: Betaxolol
Give an example of an beta 2 agonist and antagonist?
agonist: Terbutaline/Salbutamol (B2>>B1>>>>A)
antagonist: Butoxamine
What is a mixed beta agonist and antagonist?
agonist: isoprenaline
antagonist: propranolol
What receptors does dopamine activate?
D1=D2>>B>>A
How are adrenoceptors regulated and what is the mechanism of this?
Number and function of adrenoceptors may be regulated to modify physiological response. Desensitisation may occur after exposure over a period of time and results in lesser response to further stimulation.

Mechanisms:
1. Receptor sequestration -rapid and transient event decrease in receptor availability.
2. Down-regulation -reduced receptors due to reduced synthesis.
3. Receptor phosphorylation - resulting in impaired binding
What is the general structure of sympathomimetic drugs?

What happens if you substitute at the
1) benzeine ring?
2) amino group?
3) alpha carbon?
Sympathomimetic drugs are based on a benzene ring structure with an ethylamine side chain. Substitutions made on the terminal amino group, benzene ring or the alpha or beta carbons modify the affinity of binding at specific receptors.

Catecholamine formed by substitution with hydroxy groups on the benzene ring this increase potency but decreases bioavailability and decreases duration of action by making the drug subject to inactivation by COMT.

Substitution on the amino group increases beta receptor activity. Substitution at the alpha carbon blocks oxidation by MAO. Substitution on the beta carbon is important for storage.
What are the catecholamine sympathomimetics?
Adrenaline, Noradrenaline, Isoprenaline, Dopamine, Dobutamine.
What are the non-catecholamine sympathomimetics?
Ephedrine, Phenylephrine, Amphetamine.
Mechanism of:
1. adrenaline?
2. Isoprenaline?
3. Noradrenaline
4. Dopamine
5. Dobutamine
Adrenaline: Non-selective alpha and beta- adrenergic agonist.

Noradrenaline: Non-selective alpha agonist, B1 greater than B2.

Isoprenaline: Selective beta agonist Potent beta agonist, little alpha agonist effect.

Dopamine: D1 and D2 agonist with beta and alpha actions at high doses.

Dobutamine: B1 selective agonist.
(Dobutamine is a racemic mixture -the positive isomer has B1 agonist and A1 antagonist actions, the negative isomer has A1 agonist actions - the net effect is positive inotrope action with little peripheral effect hence less reflex tachycardia)
Phenylephrine mechanism?
Relatively pure alpha agonist with limited beta action
Ephedrine mechanism?
Some direct non-selective action on adrenoceptors, also causes release of stored catacholamines.
Amphetamine mechanism?
Causes release of stored catecholamines
Methyldopa and clonidine mechanism?
Methyldopa - centrally acting A2 agonist.
Clonidine - centrally acting A2 agonist).

causes centrally mediated reduction in TPR (clonidine also causes bradycardia)
Cardiovascular effects of adrenaline and noradrenaline?
Adrenaline: Rise in systolic blood pressure due to positive inotropic and chronotropic effects via B1 receptors. Total peripheral resistance and hence diastolic blood pressure may fall due to vasodilation mediated by B2 receptors. Bolus doses tend to have mainly peripheral vasoconstrictor effect whereas infusions have the effect described above

Noradrenaline: Rise in systolic blood pressure due to positive inotropic and chronotropic effects via B1 receptors. Total peripheral resistance and diastolic pressure also increase due to alpha effect and lack of B2 mediated vasodilation
Cardiovascular effects of isoprenaline and dopamine?
Isoprenaline: Marked increase in cardiac output due to positive inotropic and chronotropic effect mediated by B1 receptors. Total peripheral resistance and diastolic blood pressure fall due to vasodilation mediated by beta receptors. Systolic pressure typically falls by a small amount though may rise.

Dopamine: Reduction in TPR mediated by D1 receptors on blood vessels and pre-synaptic D2 receptors that result in reduced noradrenaline secretion. Most important effects are renal vasodilation. At higher doses, dopamine acts on beta receptors then alpha receptors to cause vasoconstriction and has an adrenaline-like action.

Dose: Renal 0.5-2ug/kg/min
Beta 2-10ug/kg/min
Alpha >10ug/kg/min
Cardiovascular effects of dobutamine?
Increased stroke volume and cardiac output mediated by B1 receptors. Less chronotropic effects. Mild vasodilation, sometimes vasoconstriction.
Cardiovascular effects of phenylephrine and ephedrine and adrenaline?
Phenylephrine: Marked increase in peripheral vascular resistance and decrease in venous capacitance mediated by alpha receptors and resulting in hypertension, and reflex vagally-induced mild bradycardia.

Ephidrine: Mainly used as a nasal decongestant, mild central stimulant effect.

Amphetamine: Marked central stimulant effect on mood and alertness. Appetite suppressant. Some efficacy in ADHD. Most have marked central stimulant effect, especially amphetamine.
Catecholamine absorption, distribution and excretion?
Poor bioavailability after oral administration due to catechol structure and subsequent inactivation by COMT found in gut and liver

Poor distribution to CNS due to catechol structure.
Metabolised by COMT and MAO and excreted in the urine.
Non catecholamine sympathomimetic absorption, distribution and excretion?
Orally active, longer duration of action.
Readily enters CNS, especially amphetamine.
Significant fraction excreted unchanged.
Weak base therefore excretion enhanced by acidification of the urine.
Give an example of an indirectly acting sympathomimetic?
What does this do
Cocaine

Inhibition of noradrenaline reuptake at noradrenergic synapses

Widely distributed and readily enters CNS, short duration of action
Give 3 examples of alpha antagonists and their structure?
Phentolamine imidazoline derivative.
Phenoxy- benzamine
Prazosin (Piperazinyl quiazoline)
What are the receptor selectivities of
1. Phentolamine?
2. Phenoxybenzamine?
3. Prazosin?
1. Phentolamine: Non selective mixed A1 and A2 antagonist.
Reduction in peripheral vascular resistance mediated by blockade of alpha receptors. This may result in a reflex tachycardia. Antagonism of pre- synaptic A2 receptors may cause noradrenaline release. (Also inhibits response to 5HT and H1/H2)

2. Phenoxybenzamine: A1 selective antagonist. (also blocks Ach, H1, 5HT). Blockade of catecholamine induced vasoconstriction.

3. Prazosin: Potent A1 antagonist. Decreased total peripheral resistance due to relaxation of arterial and venous smooth muscle mediated by alpha 1 blockade.
Toxicity of
1. Phentolamine?
2. Phenoxybenzamine?
3. Prazosin?
Phentolamine: Reflex tachycardia due to greater release of noradrenaline and its action on beta receptors

Phenoxybenzamin: Postural hypotension and reflex tachycardia. Inhibition of ejaculation, fatigue, sedation.

Prazosin: Less reflex tachycardia
1st dose hypotension
Tends to cause salt and water retention
Dizziness Palpitations Headache No effect on lipids
Absorption and distribution of
1. Phentolamine?
2. Phenoxybenzamine?
3. Prazosin?
Phentolamine: Poorly absorbed orally. Duration of action determined by half life and rate of dissociation from the receptor.

Phenoxybenzamine: Well absorbed orally.

Prazosin: Well absorbed orally Highly protein bound
Metabolism and excretion of prazocin?
Extensively metabolised by liver
50% bioavailability. Half-life 3 hours.
Mechanism of
1. Propanolol?
2. Metoprolol?
3. Atenolol?
4. Esmolol?
5. Labetolol?
6. Carvedilol?
1. Propanolol: Non-selective beta antagonist No action at alpha or muscarinic receptors
2. Metoprolol: B1 selective
3. Atenolol: B1 selective.
4. Esmolol B1 selective
5. Labetolol: Mixed B1 antagonist and alpha antagonist
6. Carvedilol: Mixed non-selective beta blocker and alpha antagonist
Which beta blockers also have local anesthetic actions?
The following beta blockers also act as membrane stabilisers by sodium channel blockade
Acebutolol Betaxolol Labetolol Metoprolol Pindolol Propanolol
CVS, respiratory, eye and metabolic effects of beta blockers?
CVS
Negative inotropic and chronotropic effect on the heart. (including slowed AV conduction and increased PR interval)
Increased peripheral vascular resistance due to unopposed alpha effects. Antagonism of the release of renin (B1) resulting in reduced TPR

RS
Increased airway resistance (largely avoided by B1 selective agents but not completely).

Eye
Decreased aqueous humour production leading to reduced intraocular pressure.

Metabolic and endocrine: Inhibition of catecholamine induced lipolysis and glycogenolysis via B2 receptors.
Impair the recovery from hypoglycaemia as this is usually mediated by catecholamines.
Masking of clinical signs of hypoglycaemia and impaired recovery from hypoglycaemia
Increased VLDL, decreased HDL
Metoprolol and propranolol CNS side effects?
Sedation, sleep disturbance, depression, psychotic episodes.
What do beta blockers interract with?
Calcium antagonists - leading to severe hypotension, bradycardia, congestive cardiac failure
Absorption and distribution of beta blockers?
Absorption Well absorbed orally.

Distribution Large volume of distribution.

Propanolol: Lipid soluble Readily crosses BBB
Metoprolol: Moderate lipid solubility
Atenolol: Low lipid solubility
Esmolol: Low lipid solubility
Metabolism and excretion of propranolol?
Extensive first pass metabolism Excreted in urine Variable between individuals
Low bioavailability
Half life 3-6 hours
Dose reduction required in hepatic failure
Metabolism and excretion of
1. metoprolol?
2. atenolol?
3. esmolol?
1. Metoprolol: Extensive first pass metabolism
Low bioavailability, Half life 3-4 hours
2. Atenolol: Less extensive first pass metabolism Low bioavailability. Half life 6-9 hours
3. Esmolol: Rapidly hydrolysed by esterases in red blood cells. Half life 10 minutes.
What does Trimethaphan do?
ganglion blocking agent: Competitive antagonist at nicotinic cholinoceptors on sympathetic and parasympathetic postganglionic neurons.
Causes pooling of blood in capacitance vessels.

Abandoned due to side effects ␣ sympathoplegia (excessive orthostsaic hypotension, sexual dysfunction) and parasympathoplegia (constipation, urinary retention, glaucoma, blurred vision, dry mouth)

IV administration
Mechanism of Guanethidine?
Inhibitor of noradrenaline release from postganglionic sympathetic neurons: Guanethidine is transported across the nerve membrane by uptake 1. Concentrated in transmitter vesicles where it replaces noradrenaline causing depletion of noradrenaline stores.

Causes reduced cardiac output due to bradycardia and relaxation of capacitance vessels.

Postural hypotension common. Overdosage may result in severe hypotension or shock. May induce hypertensive crisis in those with phaechromocytoma due to release of noradrenaline. Effects are attenuated when TCAs are coadministered due to their effect on blocking reuptake.

Very large volume of distribution and long half life.
Mechanism of reserpine?
Reserpine blocks carrier mediated transport of dopamine into the vesicle. Results in depletion of dopamine, noradrenaline, and serotonin in central and peripheral neurons.

Causes antihypertensive effect by reduction in cardiac output and total peripheral resistance.

Effects are largely irreversible and last for several days.
For methyldopa state the:
1. Mechanism?
2. Organ effects?
3. Clinical use?
1. Centrally acting alpha agonist. Inhibition of dopa decarboxylase and depletion of noradrenaline
A2 actions greater than A1
2. Centrally mediated reduction in total peripheral resistance with variable reduction in heart rate and cardiac output.
3. Mild to moderate hypertension.
Pregnancy induced hypertension
Dose 0.5-3g/day in divided doses 0.25-1g IV over 20 minutes.
For methyldopa state the:
1. Toxicity and interactions
CVS
May cause postural hypotension and bradycardia.

CNS
Sedation and loss of concentration Depression Nightmares.

Endocrine: Lactation -mediated by inhibiting action on dopaminergic mechanisms in the hypothalamus.

Other: Positive Coombs test is 25%.
Interactions May result in lithium toxicity if co-administered
For methyldopa state the:
1. Absorption and distribution
2. Metabolism and excretion
1. Absorption Orally active. Occasional IV use occasional.
Distribution Active transport into brain
2. Extensive first pass metabolism.
Metabolised in liver ␣ likely production of an active metabolite.
Most is renally excreted.
Antihypertensive effect in 4-6 hours, clinical effect may last 24 hours due to active metabolite.
For clonidine state the:
1. Mechanism?
2. Organ effects?
3. Clinical use?
1. Centrally acting partial agonist at alpha receptors, causing reduced sympathetic tone and increased parasympathetic tone. Preference for A2
2. Reduction in blood pressure and mild bradycardia.
3. Hypertension
For clonidine state the:
1. Toxicity and interactions
Sedation, dry mouth. Depression. Reactive hypertensive crisis if rapidly withdrawn.
For clonidine state the:
1. Absorption and distribution
2. Metabolism and excretion
Orally active, bioavailability 75%.
Distribution Lipid soluble and rapidly enters the brain.
Half life 8-12 hours
For acetazolamide state the:
1. Mechanism?
2. Organ effects?
3. Clinical use?
1. carbonic anhydrase inhibitor: Carbonic anhydrase most prominent in the luminal membrane of the PCT
2. Profound depression of bicarbonate reabsorption in the proximal tubule
Reduced production of bicarbonate by the ciliary body
3. Glaucoma Urinary alkalinisation Metabolic alkalosis Acute mountain sickness
Epilepsy
Dose 250mg-1g/24hours
For acetazolamide state the:
1. Toxicity and interactions
Toxicity: Hyperchloraemic metabolic acidosis
Renal stones
Renal potassium wasting
Skin reactions

Interactions :
Salicylates - increased risk of metabolic acidosis Phenytoin - reduced excretion of phenytoin resulting in toxic levels

Contraindications:
Hepatic failure - acetazolamide will decrease the urinary loss of ammonia.
For acetazolamide state the:
1. Absorption and distribution
2. Metabolism and excretion
Well absorbed orally Increased urinary pH within 30 minutes
For frusemide state the:
1. Mechanism?
2. Organ effects?
3. Clinical use?
1. Inhibition of the Na/K/2Cl cotransporter in the thick ascending limb of the loop of Henle resulting in reduced reabsorption of sodium chloride
Indirect increase in calcium and magnesium excretion
3. Acute pulmonary oedema Hypertension Hyperkalaemia Hypercalcaemia
Dose 20-80mg PO/IV
For frusemide state the:

1. Toxicity and interactions
Toxicity Hypokalaemic metabolic acidosis
Dose related ototoxicity
Hyperuricaemia
Hypomagnesaemia
Allergic reactions

Hyperbilirubinaemia -displaces bilirubin from albumin

Contraindications: Known hypersensitivity to sulphonamides (cross- sensitivity may occur) ARF
For frusemide state the:

1. Absorption and distribution
2. Metabolism and excretion
Absorption: Well absorbed orally Diuretic response in less than 5 minutes with intravenous use

Distribution: Bound to albumin
Half life 2-3 hours

Diuretic response is proportional to their excretion in the urine
One third is filtered, two thirds is secreted by the proximal tubule
For thiazide state the:
1. Mechanism?
2. Clinical use?
Inhibition of sodium/chloride cotransporter in the early DCT

Hypertension Congestive cardiac failure Urolithiasis due to idiopathic hypercalciuria Nephrogenic diabetes insipidus
Dose 25-50mg/day
For thiazide state the:

1. Toxicity and interactions
Toxicity: Hypokalaemic metabolic alkalosis Hyperuricaemia
Hyperlipidaemia
Hyponatraemia
Allergic reaction
For thiazide state the:
1. Mechanism?
2. Organ effects?
3. Clinical use?
1. Toxicity and interactions
1. Absorption and distribution
2. Metabolism and excretion
Absorption Well absorbed orally
Distribution Bound to albumin
Eliminated rapidly by the kidney
Half life 5-14 hours
For potassium sparing diuretics state the:
1. Mechanism?
2. Organ effects?
3. Clinical use?
1. Spironolactone: Competitive antagonism of aldosterone at mineralocoticoid receptors

Triamterene, amiloride: Direct inhibition of sodium reabsorption in the CD

2. Decreased sodium reabsorption Decreased potassium excretion
3. Most useful when there is a mineralocorticoid excess such as Conn syndrome or secondary aldosteronism due to congestive heart failure
For potassium sparing diuretics state the:

1. Toxicity and interactions
Toxicity: Hyperkalaemia Hyperchloraemic metabolic acidosis Gynaecomastia

Interactions: Triamterene and indomethacin have caused acute renal failure
For potassium sparing diuretics state the:

1. Absorption and distribution
2. Metabolism and excretion
Spironolactone: Metabolised by liver
Triamterene: Metabolised by the liver and excreted by the kidney
Amiloride: Excreted unchanged in urine
For osmotic diuretics state the:
1. Mechanism?
2. Clinical use?
Mannitol :
Filtered
Not metabolised Not secreted Not reabsorbed
Maintains osmotic gradient in the lumen of the tubule and therefore preventing absorption of water

Increase urine volume in haemolysis or rhabdomyolysis
Reduction in intracranial and intraoccular pressure
Dose 1-2g/kg
For osmotic diuretics state the:

1. Toxicity
Extracellular volume expansion -may exacerbate pulmonary oedema of CCF
Dehydration
For Hydralazine state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
1. Direct acting vasodilator
Mechanism unknown -may interfere with calcium entry into the cell and cause electromechanical decoupling.
2. Arterial dilator - little effect on veins.
Lowers systemic vascular resistance to cause decreased blood pressure.
3. Severe hypertension.
Severe pre- eclampsia
Dose Oral : 50- 200mg/day in divided doses IV ␣ 20-40mg slowly
For Hydralazine state the

1. Toxicity and interactions
CVS
Reflex tachycardia - may provoke angina. (can be used in combination with a beta blocker) Increases plasma renin activity

CNS
Increased cerebral blood flow
Others Headache Nausea Flushing
For Hydralazine state the

1. Absorption and distribution
2. Metabolism and excretion
1. Oral or intravenous preparations
2. Extensive first pass metabolism. (variable between individuals due to acetylation status)
Low bioavailability.
Half life 2-4 hours.
Mostly excreted in urine.
For sodium nitroprusside state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
1. Direct acting vasodilator.
Acts by activation of guanylyl cyclase resulting in increased cGMP which relaxes smooth muscle.
No effect on other smooth muscle
2. Arterial and venous dilator (more active on veins)
Lowers peripheral vascular resistance and venous return to cause decreased blood pressure
3. Hypertensive crisis Aortic dissection prior to surgery
Dose 0.5-10ug/kg/min if blood pressure is not controlled at maximum rate in 10 minutes the infusion should be terminated
500ug/kg of SNP produces toxic amounts of cyanide
For sodium nitroprusside state the

1. Toxicity and interactions
CVS
Compensatory tachycardia. Compensatory increase in catecholamines and renin.

RS
May causes reduced PaO2 due to attenuation of hypoxic pulmonary vasoconstriction

CNS: Increased cerebral blood flow and increased intracranial pressure

Endocrine: Delayed hypothyroidism can occur due to take up by thyroid tissue.

Other: Cyanide combines with cytochrome C and leads to impairment of aerobic metabolism. Accumulation can cause metabolic acidosis, arrhythmias and hypotension.
Cyanide toxicity related to rate of infusion rather than dose.

Exacerbated by B12 deficiency, hypothermia, renal and hepatic impairment. Thiocyanate toxicity may occur after several days and cause weakness, disorientation and psychosis.
For sodium nitroprusside state the

1. Absorption and distribution
2. Metabolism and excretion
Given by IV infusion. Sensitive to light therefore IV line covered in foil.
Distribution Extracellular fluid volume

Rapidly metabolised by uptake into red cells
Effectively combine with methaemaglobin to form cyanomethaemaglobin
One molecule of SNP combines with haemoglobin to form cyanmethaemoglobin and 4 molecules of cyanide
Cyanide ions combine with methaemoglobin to form cyanomethaemoglobin.
Cyanide also reacts with thiosulphate in the plasma to form thiocyanate.
Saturation of the above mechanisms leads to the accumulation of cyanide

Free cyanide reacts with cytochromes and prevents their participation in oxidative metabolism, resulting in severe metabolic acidosis
For Diazoxide state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
1. Direct acting vasodilator Mechanism unclear
May act by opening potassium channels in smooth muscle membrane.
May have a calcium antagonist action
2. Arteriolar dilator ␣ little effect on veins. Lowers systemic vascular resistance to cause decreased blood pressure.
Effect is rapid and profound and is associated with significant reflex tachycardia.
3. Hypertensive emergencies.
Intractable hypoglycaemia
Dose 5mg/kg
For Diazoxide state the

1. Toxicity and interactions
CVS
Excessive hypotension. Reflex tachycardia - may provoke angina. Increased plasma renin activity.
Endocrine: Inhibits insulin release.
For Diazoxide state the

1. Absorption and distribution
2. Metabolism and excretion
Intravenous
Extensive albumin binding.
Metabolised in liver and excreted in urine.
For minoxidil state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
1. Arterial dilator - little effect on veins. Lowers systemic vascular resistance to cause decreased blood pressure. Tends to have a greater effect than hydralazine.
Direct acting vasodilator - acts by opening potassium channels in smooth muscle membrane.
2. Severe hypertension refractory to other antihypertensives
Dose 5-40mg/day
For minoxidil state the

1. Toxicity and interactions
Headache, nausea, anorexia, palpitations. Reflex tachycardia - may provoke angina.
For minoxidil state the

1. Absorption and distribution
2. Metabolism and excretion
1. Well absorbed orally Does not enter CNS
2. Metabolised by liver and excreted in urine
Half life 4 hours.
For ACEI state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
1. Inhibition of angiotensin converting enzyme that converts angiotensin I to angiotensin II and inactivates bradykinin.
Angiotensin II is a potent vasoconstrictor. Bradykinin is a potent vasodilator.
2. CVS
Reduced peripheral vascular resistance due to:
Inhibition of renin- angiotensin system
Stimulation of the kallikrein-kinin system.
Cardiac output and heart rate are unaffected.
Most effective in individuals with high plasma renin activity.
Effective in improving intrarenal haemodynamics due to reduction in intraglomerular capillary pressure. Subsequent reduction in proteinuria.
3. Hypertension. Diabetes.
Congestive cardiac failure.
Post myocardial infarction to reduce the risk of heart failure
Dose Captopril 25mg tds Enalapril 10-20mg od Lisinopril 10-40mg od
For ACEI state the

1. Toxicity and interactions
Toxicity: First dose hypotension - profound hypotension especially if hypovolaemic.
Dry cough and angioedema - due to effects of bradykinin.
Acute renal failure -can occur with all ACE inhibitors particularly associated with renal artery stenosis. due to dependence on renin- angiotensin system for renal function

Hyperkalaemia (more likely in renal failure or diabetes) -important consideration with potassium sparing drugs.

Mild hepatitis, rare hepatic failure
Altered taste. Rash.

Interactions: NSAIDs should be avoided as they inhibit prostaglandin synthesis that is crucial to the bradykinin mechanism for inducing vasodilation

Contraindications: Pregnancy (2nd and 3rd trimester) due to fetal malformations, fetal hypotension and fetal renal abnormalities.
For ACEI state the

1. Absorption and distribution
2. Metabolism and excretion
Captopril Rapidly absorbed.
Bioavailability 70%
Wide distribution except CNS.

Lisinopril: Slowly absorbed

Captopril 70% bioavailability.
Metabolised to disulphide conjugates and excreted in urine
Dose reduction required in renal failure
Half life 3 hours.

Enalapril - prodrug that is converted to enalaprilat.
Lisinopril - a lysine derivative of enalaprilat
All others are prodrugs converted to active agents by hydrolysis in the liver
Long half lives - up to 12 hours
For nitratesstate the
1. Mechanism?
2. Organ effects?
3. Clinical use?


Glyceryl trinitrate, Isosorbide mononitrate, Isosorbide dinitrate
Nitric or nitrous acid esters of polyalcohols.
1. Cause release of NO from vascular smooth muscle endothelium.
Nitric oxide causes activation of guanylyl cyclase and an increase in cGMP
cGMP causes dephosphorylation of myosin light chain to result in relaxation of muscle.
2. Direct effects: Relaxation of smooth muscle. Veins respond at a lower drug concentration than arteries. Arterioles and pre- capillary sphincters have a lesser response due to local reflex responses and decreased ability to secrete NO.
Venodilation causes marked increase in venous capacitance, decreased preload and in most cases reduced cardiac output, thereby reducing myocardial oxygen demand.
Systolic BP decreases more than diastolic.
Arterial dilation also reduces afterload.
There is dilation of the coronary arteries that may reduce spasm.
Nitrates tend to causes bronchiolar and gut smooth muscle relaxation also.

Indirect effects: Compensatory responses include sympathetic tachycardia and increased contractility.
Also retention of salt and water via renin angiotensin system.
Overall effect is decreased myocardial oxygen requirement.
Relaxation of bronchial and gastrointestinal smooth muscle.
Decreased platelet aggregation.
Nitrate converted in small quantities to nitrite which reacts with haemoglobin to form methaemoglobin.

3. Stable and unstable angina. LVF. Hypertension.
Dose SL 300mcg TD 5-10mg/24hrs IV 10-400mcg/min
For nitrates state the

1. Toxicity and interactions
CVS
Extension of clinical effect : hypotension, tachycardia, headache.
Tolerance - smooth muscle develops tolerance after continuous exposure. Tolerance may occur at a cellular level but may also result from compensation (indirect) actions of the drug itself. Clinically important tolerance does not occur with IV infusion.
Other Formation of nitrosamines by combination of nitrate/nitrite with amines - potentially carcinogenic.

Coronary Steal Syndrome: If two branches of a coronary artery have different levels of obstruction, the more obstructed branch will have relative arteriolar dilation at rest due to local metabolites. If an arteriolar vasodilator is introduced, both most effect will be to the less obstructed arteriole. Resistance in this vessel will fall and steal blood from the more obstructed vessel worsening the angina.
Therefore, drugs such as nitrates that do not have a profound effect on arterioles are most effective in angina.
For nitrates state the

1. Absorption and distribution
2. Metabolism and excretion
Absorption: High capacity hepatic nitrate reductase removes nitrate groups and therefore limits bioavailability after oral administration to 10%

Sublingual route bypasses first pass metabolism. If given orally, much larger does required.
Transdermal route also effective.
Distribution 60% protein bound

metabolised in the liver and red cells by reduction to dinitrates, mononitrates and nitrites.
Half-life of short- acting sublingual GTN is 4-8 minutes though clinical effect may persist to 20-30 minutes.
Active metabolites can persist for up to 3 hours.
Excretion is via the kidney.
For dihydropyridine and non-dihydropyridine calcium channel blockers state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
1. Dihydropyridine and miscellaneous calcium channel blockers bind to slightly different receptors associated with the (L type) calcium channel.
Drugs act from the inner side of the membrane.
Binding is more effective to channels in depolarised membranes. (although drug binds to activated and inactivated channels)
Binding results in the channel failing to open in response to depolarisation.
Decrease in calcium current causes smooth muscle relaxation, reduced contractility and slowing of pacemaker cells.
Blockade can be partially reversed by sympathomimetic that increase transmembrane flux of calcium
Vascular smooth muscle is most sensitive, but other smooth muscle is partially affected
Verapamil and diltiazem may also causes sodium channel blockade
Verapamil may have some anti-adrenergic effect
2. Smooth muscle Most types of smooth muscle are affected ␣ vascular smooth muscle is particularly sensitive.
Arterioles are more sensitive than veins
Results in reduced peripheral vascular resistance
Also may reduce coronary artery tone
Dihydropyridines have a greater vascular selectivity. Nimodipine more effective in cerebral vessels.
Cardiac muscle

Verapamil and diltiazem demonstrate cardiac selectivity
Calcium channel blockade is more marked in those tissues that fire frequently and depend solely on calcium current such as SA and AV nodes. Atrioventricular nodal conduction and refractory period are prolonged.
Verapamil is effective in suppressing early and late delayed after potentials
Negative inotrope.

Skeletal muscle unaffected by calcium channel blockers as it utilises intracellular pools of calcium. Verapamil may have a specific anti-adrenergic effect.
For dihydropyridine and non-dihydropyridine calcium channel blockers state the

1. Toxicity and interactions
CVS: Extensions of therapeutic effects ␣ hypotension, bradycardia, AV block, cardiac depression.
Hypotension and VF can occur if given for VT.
All cause worsening of heart failure
Verapamil has greatest cardiac depressant effects followed by diltiazem
Dihydropiridines have less cardiac depressant effects and tend to cause a reflex tachycardia

Endocrine: Verapamil inhibits insulin release (not significant) and may be useful in cancer chemotherapy
General: Flushing Dizziness Nausea Constipation

Interactions: Dangerous cardiodepressant effects can occur if co- administered with beta blockers.
Verapamil may cause increased blood levels of digoxin.
Contraindications: Nifedipine has been associated with increased risk of myocardial infarction when used for hypertension in patients with unstable angina
For dihydropyridine and non-dihydropyridine calcium channel blockers state the
1. Abosrption and distribution
2. Metabolism and excretion
Absorption Orally active
Distribution >90% protein bound

All extensively metabolised
Variable bioavailability -generally greater than 50% except verapamil (20%)
Diltiazem excreted in faeces.
Verapamil metabolised by liver and mostly excreted by kidney, remainder by GIT.
Half lives Amlodipine 30-50 Nifedipine 4 Felodipine 11-16 Nimodipine 1-2
Diltiazem 3-4 Verapamil 6
For digoxin state the
1. Mechanism and structure?
2. Organ effects?
3. Clinical use?
Steroid nucleus (lipophilic) attached to a lactone ring (hydrophilic).
No ionisable group therefore solubility is not pH dependant.
Found in foxglove, oleander, lily of the valley, toads

1. Inhibition of Na+/K+ ATPase
Several binding sites are present and digoxin binds to the alpha subunit. Very low concentrations of drug may stimulate the enzyme. Toxic doses ␣ sympathetic effects.
2. Direct membrane actions: Increase in intracellular sodium concentration.
Increased intracellular sodium results in increased function of the sodium/calcium exchanger, causing a rise in intracellular calcium.
Increased free calcium results in increased intensity of actin/myosin interaction, resulting in increased force of contraction.
Increased free calcium also reduces potassium conductance and causes shortening of the action potential. (though initial lengthening of the action potential is characteristic)
Inhibition of Na+/K+ ATPase, as well as causing increased intracellular sodium, results in decreased intracellular potassium (toxic effect) and reduced resting membrane potential.
More toxic concentrations of digoxin lead to overloading of calcium stores and fluctuation in concentration, resulting in oscillatory depolarising after potentials that may result in ectopic beats ␣ sometimes resulting in bigeminy/VT/VF.
Autonomic actions: Low dose -parasympathomimetic effects via central vagal stimulation, sensitisation of baroreceptors and facilitation of muscarinic transmission.
For digoxin state the

1. Toxicity and interactions
CVS:
Arrhythmias - due to direct membrane effect and autonomic effect: AV junctional, ventricular ectopics, bigeminy, VT, VF.

GIT:
Anorexia, nausea, vomiting, diarrhoea.

CNS:
Disorientation, hallucinations, visual disturbance of colour perception, convulsions.

Endocrine: Gynaecomastia.

Interactions: Potassium and digoxin inhibit each others binding to Na/K ATPase
Hyperkalaemia therefore reduces digoxin binding and reduces toxicity
Hypokalaemia (and hypomagnesaemia) may result in digoxin toxicity (therefore care with diuretics)
Hypercalcaemia and hypomagnesaemia increases toxic effects of digoxins as they accelerate cellular calcium overload

Quinidine displaces digoxin from binding site but more importantly markedly reduces digoxin clearance

Contraindications WPW syndrome: digoxin increases the probability of conduction through alternative pathways.
Increased sensitivity of the myocardium in elderly patients
For digoxin state the

1. Absorption and distribution
2. Metabolism and excretion
Well absorbed orally. 10% have gut bacteria that inactivate digoxin therefore require higher dose Care when these people are then given antibiotics as they may become digitoxic
Distribution Widely distributed in all tissues including CNS
Tends to accumulate in heart, kidney and liver due to propensity to bind tissue proteins in these organs

2/3 excreted unchanged by kidneys.
Renal clearance proportional to creatinine clearance
Dose adjustment required in renal insufficiency
Half life 40 hours
Loading dose is given
Narrow therapeutic window.
Dosing of digoxin?
Non-emergency administration Oral 125-250ug/day

Rapid oral digitalisation Adult 750-1500ug Elderly 500-750ug as a single dose or 3-4 divided doses, then maintenance

Emergency parenteral digitalisation
Adult 500-1000ug Elderly 250-500ug as a single dose or 3-4 divided doses, then maintenance
What determines the pacemaker rate? (i.e. what slows it down)
More negative potential in phase 4 (ie resting potential)
Reduction in the slope of phase 4
More positive threshold potential
Prolongation of the action potential duration.
What are early depolarisations?
Occur in phase 3 Arise from plateau Exacerbated by slow heart rates Contribute to development of QT related arrhythmias.
What do class 1a, 1b, 1c, 2, 3, 4 antiarrhythmics do?
1 are sodium channel blockers
1a: Quinidine Procainamide Disopyramide TCAs- shorten action potential duration
1b- shorten action potential duration Lignocaine Tocainide, Mexilitine, Phenytoin
1c- No effect or may minimally lengthen action potential duration Flecainide
class 2 - beta blockers
Class 3 potassium channel blockers
Class 4 calcium channel blockers
For quinidine state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
1. Class 1a antiarrhythmic: Binds to and blocks activated sodium channels. Block results in: Reduced rate of rise of phase 0 of the cardiac action potential, causing lengthening of the action potential
Lengthening of the refractory period
Alpha adrenoceptor properties (may be prominent with IV injection)
2. Increased QRS and QT intervals
3. Atrial flutter/fibrillation. Ventricular tachycardia
For quinidine state the

1. Toxicity and interactions
Pro arrhythmogenic
Antimuscarinic actions: May inhibit vagal effects - this may result in increased sinus rate and increased atrioventricular conduction
syncope
Prolonged QT may lead to VF or torsades de pointes in 2-8%

Cardiac depression: May cause asystole, more likely at high concentrations and hyperkalaemia.

Preceded by 30% increase in QRS duration.

Extracardiac: Nausea, vomiting and diarrhoea. Rash, fever, hepatitis

Quinidine only Cinchonism - headache, dizziness, tinnitus. Angioneurotic oedema.

Interactions: Reduces digoxin clearance therefore increases levels
Increased effect with hyperkalemia
Inhibition of metabolism of TCAs, beta blockers
For quinidine state the

1. Absorption and distribution
2. Metabolism and excretion
1. Well absorbed orally. 80% bound to plasma proteins.
2. Metabolised in liver. 20% excreted unchanged in urine - enhanced by acidic urine.
Half life 6 hours.
For procanimide state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
1. Binds to and blocks activated sodium channels.
Block results in: Reduced rate of rise of phase 0 of the cardiac action potential, causing lengthening of the action potential Lengthening of the refractory period
Blocks potassium channels and reduces outward repolarising current. Lengthens action potential duration (reflected by a lengthening of QT interval) - reduces the time spent in diastole and therefore makes more activated sodium channels available for blockage.
2. Action potential and refractory period are lengthened
Increased QRS and QT intervals
3. Atrial flutter/fibrillation. Ventricular tachycardia
For procanimide state the

1. Toxicity and interactions
Pro arrhythmogenic
Antimuscarinic actions: Mild compared with quinidine
syncope
Prolonged QT may lead to VF or torsades de pointes in 2-8%
Cardiac depression May cause asystole, more likely at high concentrations and hyperkalemia. Preceded by 30% increase in QRS duration.

Ganglion blocking actions Can cause hypotension with IV use though usually not a problem with oral use.
Extracardiac: Nausea, vomiting and diarrhoea. Rash, fever, hepatitis
0.5% risk of serious blood dyscrasia

Drug-induced lupus Interactions
Increased effect with hyperkalemia
Amiodarone causes increased effect
For procanimide state the

1. Absorption and distribution
2. Metabolism and excretion
Oral or intravenous Widely distributed
Metabolised in liver. Major metabolite N acetyl procainamide (NAPA) has class 3 actions.
Metabolism reduced in slow acetylators
Renal excretion important for NAPA - therefore dose adjustment is required in renal failure.
Half life of procainamide 3-4 hours.
Half life of NAPA is considerably longer and therefore may accumulate.
For Lignocaine state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
1. Blocks both activated and inactivated sodium channels.
Shortens the action potential thereby increasing diastole.
Lignocaine suppresses activity of depolarised arrhythmogenic tissue but has minimal effect on normal tissue.
Most cells become drug free in diastole.
2. Action potential and refractory period are shortened
3. Ventricular tachycardia and fibrillation after defibrillation.
Local anaesthetic.
For Lignocaine state the

1. Toxicity and interactions
CVS
Pro arrhythmogenic
Depression of cardiac pacemaker activity, excitability and conduction.
Negative inotropic effect and decreased peripheral resistance.
Exacerbates ventricular arrhythmias in <10%.
May causes hypotension.

CNS
Drowsiness, visual and auditory disturbance, restlessness, nystagmus, shivering, convulsions, CNS depression.

Interactions: Drugs that decrease liver blood flow (propanolol, cimetidine) reduce lignocaine clearance.
For Lignocaine state the

1. Absorption and distribution
2. Metabolism and excretion
1. Widely distributed after intravenous administration to highly perfused organs.
Extensive first pass metabolism - 3% bioavailability.

(note - Tocainide and mexiletine are cogeners of lignocaine that are resistant to first-pass metabolism and have similar effects)

2. Metabolised in liver and plasma.
Clearance dependent on hepatic blood flow
Excreted in urine.
Half life 1.5 hours
For Flecanide state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Sodium channel blocker
Decreases rate of rise pf phase 0 of the cardiac action potential
Little effect on action potential duration
Lengthens refractory period
Greatest action in the His-Purkinje system
SVT Pre-excitation syndromes Ventricular arrythymias
For flecanide state the

1. Toxicity and interactions
Safe drug with few side effects.
Pro arrhythmogenic
For Flecanide state the
1. Mechanism?

1. Absorption and distribution
2. Metabolism and excretion
Well absorbed orally.
Half life 20 hours, usually given twice daily. Metabolised by liver and excreted by the kidney.
For Amiodarone state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
1. Potassium channel blockade - class 3 action Results in lengthening of the action potential even at high heart rates.
Increased refractory period

Sodium channel blockade - class 1 action
Acts on inactivated channels only
Most pronounced in tissues that have long action potentials, frequent action potentials or less negative diastolic potentials.

Calcium channel blockade - class 4 action

Non competitive beta blockade - class 2 action
Also causes partial inhibition of beta receptors.

2. CVS
Slows sinus rate. Slows AV nodal conduction. Markedly prolongs QT interval.
Prolongs QRS duration.
Increases refractory period duration.
Increases coronary blood flow and reduces myocardial oxygen demand
Extra-cardiac effects: Peripheral vascular dilation due to beta and calcium blocking effects.
3. Supraventricular and ventricular tachyarrhythmias including in cardiac arrest protocols.
Dose Intravenous 5mg/kg usually administered as an infusion over 20 minutes but can be given as a bolus.
Oral 200mg tds reducing to 100- 200mg od after a week.
For Amiodarone state the

1. Toxicity and interactions
CVS: Bradycardia or heart block in those with existing nodal disease. May precipitate heart failure.

Respiratory effects: Pulmonary fibrosis.
Eye effects: Corneal deposits
Skin effects: Skin deposits and photodermatitis. Grey skin discolouration with chronic use in 5%.
Neurological effects: Peripheral neuropathy, paresthesia, tremor, ataxia, headaches.
Thyroid effects: Hypo or hyperthyroidism can occur in 5%.
Gastrointestinal effects: Constipation, hepatocellular necrosis.
Contraindications: Porphyria
For Amiodarone state the

1. Absorption and distribution
2. Metabolism and excretion
Bioavailability 20- 80%
95% protein bound.
Small volume of distribution.
Active metabolite may accumulate.
Excreted in bile, faeces and urine.
Half-life highly variable 14-110 days.
15-30 days required for loading.
Toxic effects may continue after drug is ceased.
For sotalol state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
class II and III
Non-selective beta blocker
Lengthens the action potential and causes increased refractory period

2. CVS: Negative inotropic and chronotropic effect on the heart. (including slowed AV conduction and increased PR interval)
Increased peripheral vascular resistance due to unopposed alpha effects. Antagonism of the release of renin.
Increase in length of action potential and refractory period

RS:
Increased airway resistance (largely avoided by B1 selective agents but not completely).

Eye: Decreased aqueous humour production leading to reduced intraocular pressure

Metabolic and endocrine Inhibition of lipolysis and glycogenolysis via B2 receptors.

used: Supraventricular and ventricular arrhythmias.
For sotalol state the

1. Toxicity and interactions
CVS: Prolongation of repolarisation may lead to torsades de points - risk greatest at high concentrations.
Myocardial decompensation in pre-existing abnormal myocardial function.
Decreased perfusion in severe peripheral vascular disease.

RS:
Airway obstruction in pre- existing asthma/reactive airways.

Endocrine: Masking of clinical signs of hypoglycaemia

Interactions: Calcium antagonists - leading to severe hypotension, bradycardia, congestive failure
For sotalol state the

1. Absorption and distribution
2. Metabolism and excretion
Well absorbed orally
Largely excreted unchanged by kidney therefore dosage adjustment is required in renal impairment
For adenosine state the
1. Mechanism?
2. Organ effects?
3. Clinical use
Adenosine
Naturally occurring nucleoside
Miscellaneous
Acts via adenosine receptors
Enhanced potassium conductance and inhibition of cAMP-induced calcium influx.
Results in marked hyperpolarisation and suppression of calcium- dependent action potentials.
2. Inhibits AV nodal conduction and increases AV nodal refractory period.
Mild effects on SA node.
3. SVT
Diagnosis of narrow or broad complex tachyarrhythmias
Dose 3,6,9,12mg with rapid flush
For adenosine state the

1. Toxicity and interactions
Transient arrhythmias in >50% including bradycardia and ventricular standstill Generally short lived due to short half life
Flushing in 20% Hypotension <1%

Shortness of breath and chest burning in 10%.

May cause bronchospasm therefore caution in asthmatics.

Interactions: Less effective in the presence of adenosine receptor blockers such as caffeine and theophylline.
Contraindications 2nd or 3rd degree AV block, sick sinus syndrome Specific risk of torsades in long QT interval
For adenosine state the

1. Absorption and distribution
2. Metabolism and excretion
inactive when administered orally. Absorbed into red blood cells and vascular endothelium where it is phosphorylated or deaminated.
Half life less than 10 seconds.
For heparin state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Binds to endothelial cell surfaces
Biological activity dependent on the presence of antithrombin III that inhibits clotting factor proteases by forming complexes with them.
Heparin binds to antithrombin III and causes a conformational change resulting in more rapid interaction with clotting factors IX, X, XI and XII
Binding to clotting factors is accelerated 1000 fold.
In particular, inactivation of factor X results in reduced conversion of prothrombin to thrombin
Higher doses of heparin also inhibit thrombin and therefore reduce conversion of fibrinogen to fibrin
One third of molecules in heparin have biological effect. Heparin is not consumed in the binding process.
HMW fractions (5000- 30000) have a marked affinity for thrombin.
LMW fractions (<9000) inhibit activated factor X but have a lesser effect on thrombin.
2. Prevention and treatment of thromboembolic disease.
Treatment of DIC and fat embolism.
Priming of vascular catheters -haemodialysis, cardiac bypass machines etc.
Treatment of unstable angina and non-Q wave MI
Dose: Heparin 5000u bd 1000u/hr
Enoxaparin
For heparin state the
1. Toxicity and interactions
2. reversal (how does this work?)
Bleeding.: Higher risk in elderly and renal failure.
Long term use associated with osteoporosis and fractures.
Transient thrombocytopenia in 25% of patients. Severe thrombocytopenia in 5%.
Can cause paradoxical thromboembolism due to heparin induced platelet aggregation.

Contraindications: Active bleeding.
Clotting disorders - haemophilia, thrombocytopenia, purpura.
Severe hypertension.
Recent neurosurgery, eye surgery or LP. A ny condition where the likelihood of uncontrolled bleeding exists.

Reversal: Protamine - highly basic peptide that combines with heparin to form a complex that has no anticoagulant activity. Excess protamine also has an anticoagulant effect.
For heparin state the

1. Absorption and distribution
2. Metabolism and excretion
1. Absorption Heparin Peak plasma levels 2- 4 hours after subcutaneous injection Highly bound to plasma proteins
LMW heparin Increased bioavailability from subcutaneous site.
Distribution Vascular compartment
2. Largely unknown Probable metabolism by the liver
For Warfarin state the
1. Mechanism?
Vitamin K dependent oral anticoagulant
Racemic mixture of 2 stereoisomers
S isomer is 4 times more potent than R isomer

Blocks gamma carboxylation of several glutamate residues in factors II, VII, IX, X and protein C resulting in biologically inactive molecules.
Carboxylation is physiologically coupled to Vitamin K metabolism and Vitamin K is oxidised to an inactive form.
In order for carboxylation to occur, Vitamin K must be reduced back to an active form.
Warfarin inhibits Vitamin K epoxide reductase and therefore prevents formation of active vitamin K.
For Warfarin state the

1. Toxicity and interactions
Bleeding. Toxic to fetus - causes haemorrhagic defects and bone malformations. Cutaneous necrosis due to depression of protein C synthesis.
Interactions

Increased INR:
Metronidazole Fluconazole Trimethoprim - sulfamethoxazole (inhibition of metabolism of S isomer) Cimetidine
Amiodarone (Inhibition of metabolism of both isomers)

Pharmcodynamic Aspirin (additive effect) 3rd generation cephalosporins (elimination of bacteria in the GIT that produce Vitamin K) Heparin
Hepatic disease Hyperthyroidism (increased turnover of clotting factors)

Decreased INR:
Pharmacokinetic Barbiturates
Rifampicin
(enzyme induction)

Pharmcodynamic Diuretics (increased clotting factor concentration) Vitamin K
Hereditary resistance
Hypothyroidism (decreased turnover of clotting factors)
For Warfarin state the

1. Absorption and distribution
2. Metabolism and excretion
1. Absorption Orally active with 100% bioavailability.
8-12 hour delay in effect as pre- synthesised factors must degrade - this takes up to 60 hours in the case of factor II.
Maximal clinical effect takes 1-3 days.
Distribution Small volume of distribution as tightly bound to albumin.

2. Half life in plasma is 36 hours.
Metabolised in the liver and conjugated to glucuronide. Metabolites excreted in urine and faeces.
For streptokinase state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Fibrinolytic
Protein synthesised by group c beta haemolytic streptococci.
Combines with plasminogen.
Streptokinase/plasminogen complex catalyses conversion of further plasminogen to active plasmin.
(plasmin is protected from plasma antiplasmins while it is within the clot)
Fibrinolysis
Fibrinolytic effect lasts a few hours.
APTT elevated for approximately 24 hours

Acute myocardial infarction PE Proximal DVT Arterial thromboembolism
Dose 1.5 million units over 30-60 minutes
For streptokinase state the

1. Toxicity and interactions
Transient hypotension
Arrhythmias -1-10%
Haemorrhage (clotting factors return to normal after 24 hours
Pyrexia
Hypersensitivity.

Contraindications: Clotting disorder Recent (10/7) major gastrointestinal haemorrhage Recent (2/12) CVA or neurosurgery
Recent (10/7) major surgery, trauma or CPR. Uncontrolled hypertension. Pregnancy.
Previous use >5/7 <12/12
Recent streptococcal infection (complex is inactivated by streptococcal antibodies)
For streptokinase state the

1. Absorption and distribution
2. Metabolism and excretion
Intravenous administration, remains intravascular
Half life 23 minutes
For TPA (tenecteplase) state the
1. Mechanism?
2. Organ effects?
t-P A Tenecteplase
recombinant tissue plasminogen activator
Preferentially activates plasminogen that is bound to fibrin therefore theoretically more specific to formed thrombus.

Dose Weight dependent 30-50mg
For TPA (tenecteplase) state the

1. Toxicity and interactions
Transient hypotension
Arrhythmias - 1-10%
Haemorrhage (clotting factors return to normal after 24 hours
Pyrexia
Hypersensitivity.

Contraindications: Recent major haemorrhage Clotting disorder Recent (2/12) CVA or neurosurgery Recent (10/7) major surgery. Uncontrolled hypertension. Pregnancy.
For TPA (tenecteplase) state the

1. Absorption and distribution
2. Metabolism and excretion
Intravenous administration, remains intravascular
Binds to heptic receptors and is catabolised to small peptides
Kidney not involved with clearance
For aspirin state the
1. Mechanism?
2. Organ effects?
Reduced synthesis of eicosanoid mediators
Irreversible inhibition of cyclooxygenase
Reduced synthesis of
thromboxane A2 Reduced synthesis of prostaglandins
Central blockade of CNS response to IL1 in causing fever

Antiplatelet
action lasts for the lifespan of the platelet - thromboxane A2 stimulates platelet aggregation and granule release
Antiinflammatory Analgesic Antipyretic
For aspirin state the
1. Toxicity and interactions
Therapeutic range 0-10mg/kg Gastritis, ulceration Impaired haemostasis

Anti-inflammatory range 50mg/kg Salicylism ␣ tinnitus, reduced hearing, vertigo.

Toxic range 50-150mg/kg hyperventilation, fever, dehydration, metabolic acidosis

Serious intoxication >150mg/kg Metabolic acidosis due to salicylic acid dissociation, deranged carbohydrate metabolism and reduced renal function.
Respiratory alkalosis due to central stimulation of respiratory centre
Renal compensation for respiratory alkalosis.
Eventual renal and respiratory failure
Interactions
Displaces from protein binding (phenytoin, methotrexate) Decreased activity of spironolactone Decreased tubular secretion of penicillin
For aspirin state the
1. Absorption and distribution
2. Metabolism and excretion
Orally active Rapidly absorbed. Acidity of stomach keeps aspirin in nonionised form that is more readily absorbed
Bound to albumin in low doses. As serum concentration rises, increasing fraction is unbound
Hydrolysed to acetic acid and salicylate by blood and tissue esterases.
Salicylate conjugated by liver and excreted by kidney.
Demonstrates variable order kinetics ␣ metabolism is saturable and small further increases in aspirin dose results in large rise in salicylate levels.
Half life 3-5 hours at low dose, 12 hours at anti- inflammatory doses
Alkalinisation of the urine increases rate of excretion of free salicylate
For tirofiban state the
1. Mechanism?
2. Toxicity?
3. ADME?
Gp IIb/IIIa antagonist
Potent inhibition of platelet function
Antithrombotic in unstable angina and myocardial infarction
2. Nausea, dyspepsia, diarrhoea. Haemorrhage. Leukopenia.
3. Guven as IV infusion. Metabolised by liver and excreted in urine
Forticlopidine state the
1. Mechanism?
2. Toxicity?
3. ADME?
1. Inhibition of ADP pathway of platelets
2. Bleeding Leukopenia. Nausea, dyspepsia, diarrhoea.
3. orally active
For Abciximab state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Monoclonal antibody that binds to GpIIb/IIIa receptors and prevents binding of fibrinogen and vWF

Antithrombotic Used in patients undergoing angioplasty or stent placement
For Abciximab state the

1. Toxicity and interactions
Bleeding Thrombocytopenia Multiple minor toxicities
For Abciximab state the
1. Absorption and distribution
2. Metabolism and excretion
Intravenous bolus dose or infusion
For Dipyridamole state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Inhibition of uptake of adenosine into red blood cells and potentiation of NO

Antiplatelet action
Also has vasodilator actions
Antithrombotic
Often combined with aspirin as a slow-release preparation
For Dipyridamole state the

1. Toxicity and interactions
Potent vasodilator therefore caution in severe coronary artery disease
Headache Nausea, vomiting
For Dipyridamole state the

1. Absorption and distribution
2. Metabolism and excretion
Orally active
Widely distributed

Metabolised in the liver Excreted in bile
For Clopidogrel state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Inhibits binding of ADP to GpIIb/IIIa and therefore inhibits platelet aggregation

Antithrombotic
Dose 75mg daily
For Clopidogrel state the

1. Toxicity and interactions
Toxicity Bleeding
Interactions Aspirin, heparin -additive effect
For Clopidogrel state the
1. Absorption and distribution
2. Metabolism and excretion
Orally active Widely distributed
Metabolised in the liver
Active metabolite is responsible for clinical effect Excreted in bile
Vitamin K
1. subtypes?
2. Mechanism?
3. ADME
Fat soluble - K1 found in food, K2 found synthesised in intestine by bacteria.
Necessary for formation of factors II, VII, IX and X

Orally active - requires bile slats for absorption. IV should be given slowly. Clinical effect delayed for 6 hours
For Insulin state the
1. Synthesis
2. Mechanism?
2. Organ effects?
3. Clinical use?
1. Toxicity and interactions
1. Absorption and distribution
2. Metabolism and excretion
Polypeptide containing 2 chains (A and B) of amino acids linked by disulfide bridges. Synthesised as part of preproinsulin. Removal of a peptide leader sequence forms proinsulin. Connecting peptide is removed in the granules prior to release

Insulin binds with an insulin transmembrane receptor that binds and stimulates a protein tyrosine kinase. Exposure to increased insulin down regulates receptor concentration and affinity.
What are the 4 types of glucose transporters and where are they?
Glucose enters cells by facilitates diffusion through glucose transporters

GLUT1: brain, red cells, placenta, many other organs GLUT2 : B cells, liver, intestine, kidney -transport glucose out of the cell
GLUT4 - adipose tissue and muscle

A pool of GLUT4 transporters is maintained in the cytoplasm of insulin sensitive cells and when these cells are exposed to insulin the transporters move to the cell membrane. Secondary active transport (intestine and kidney) Utilise sodium dependent glucose transporters - SGLT1 and SGLT2.
What are the tissue effects of insulin?
Rapid (seconds) Increased transport of glucose, amino acids and potassium into insulin sensitive cells.

Intermediate (minutes) Stimulation of protein synthesis Inhibition of protein degradation Activation of glycolytic enzymes and glycogen synthase Inhibition of phophorylase and gluconeogenic enzymes

Delayed (hours) Increase in mRNAs for lipogenic and other enzymes.

Liver: Decreased glucose output due to decreased gluconeogenesis and increased glycogen synthesis.
Increased protein synthesis
Increased lipid synthesis
Decreased ketogenesis

Fat:
Increased glucose entry
Increased fatty acid synthesis
Increased triglyceride deposition
Activation of lipoprotein lipase
Increased potassium uptake

Muscle:
Increased glucose entry
Increased glycogen synthesis Increased amino acid uptake
Increased protein synthesis
Decreased protein catabolism
Decreased release of gluconeogenic amino acids Increased ketone uptake
Increased potassium uptake

Increased cell growth
What are the types of insulin used for diabetes?
Ultra short acting: Clear solution at neutral pH. Contains small amounts of zinc to improve stability and shelf life

Short acting: Clear solution at neutral pH. Contains small amounts of zinc to improve stability and shelf life. Used for DKA.

Intermediate: Neutral pH with protamine or phosphate buffer or zinc in acetate buffer. This delays absorption and prolongs duration of action.

Long acting: Neutral pH with protamine or phosphate buffer or zinc in acetate buffer. This delays absorption and prolongs duration of action.

Insulin can be derived from beef, pork or from humans.
For commonly used insulins describe the ?
1. Toxicity and interactions
2. Absorption and distribution
3. Metabolism and excretion
1. Hypoglycaemia Insulin allergy and resistance Lipodystrophy
2. Ultra short acting Short acting Effect within 30 minutes, lasts 5-7 hours. Intermediate Neutral pH with protamine or phosphate buffer or zinc in acetate buffer. Long acting
Neutral pH with protamine or phosphate buffer or zinc in acetate buffer.
3. Half life 5 minutes. Binds to insulin receptors and is internalised. Destroyed by insulin protease. 80% is degraded in liver and kidney. (this ratio is reversed in diabetics on insulin)
For sulphonylureas state the
1. Mechanism?
2. Organ effects?
3. Clinical use?


(second generation agents) Glibenclamide Glimepiride
1. Increased release of insulin from B cells
Inhibition of glucagon release
Potentiation of insulin action on target tissues

Dose Gliclazide 80-160mg/day in divided doses Glimepiride 1-8mg/day in single dose
For sulphonylureas state the

1. Toxicity and interactions

(second generation agents) Glibenclamide Glimepiride
Hypoglycaemia (alcohol predisposes)
Tachyphylaxis - B cells may become refractory to response
Blurred vision
Rash
Blood dyscrasias
Hepatic toxicity
Renal insufficiency

Contraindications: Severe renal or hepatic impairment
For sulphonylureas state the

1. Absorption and distribution
2. Metabolism and excretion

(second generation agents) Glibenclamide Glimepiride
Well absorbed orally
Half life 5 hours Metabolised by liver and excreted in urine
For biguanides (metformin) state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
1. Unclear mechanism Direct stimulation of glycolysis Reduced gluconeogenesis Decreased glucose absorption
Reduced plasma glucagon
2. Dose 500mg-3g/day
For biguanides (metformin) state the

1. Toxicity and interactions
Nausea, vomiting, diarrhoea Reduced B12 absorption Lactic acidosis - increased risk with renal insufficiency, age and alcohol (does not cause hypoglycaemia) Contraindications Alcoholism Renal or hepatic disease
For biguanides (metformin) state the

1. Absorption and distribution
2. Metabolism and excretion
Well absorbed orally Not bound to plasma proteins
Excreted unchanged in urine
Half life 1-3 hours
For glucagon state the
1. Mechanism?
2. Indications
Binds with transmembrane receptor protein that stimulates a GTP-binding signal transducer protein (G protein) that in turn generates an intracellular second messenger. Second messengers include cAMP, calcium and phosphoinositides.
Glycogenolysis (liver but not muscle) Gluconeogenesis Lipolysis
Ketogenesis
Secretion of growth hormone, insulin and pancreatic somatostatin.
Potent inotropic and chronotropic effect on the heart independent of metabolic effects and without requiring functioning adrenoceptors
Profound relaxation of smooth muscle at high doses

Severe hypoglycaemia
Beta blocker overdose
Oesophageal foreign body
Dose
For carbimazole state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Inhibition of thyroid peroxidase resulting in reduced formation of thyroid hormones
Inhibition of peripheral deiodination of T4

Reduced thyroid hormone synthesis
Onset of effect may take several weeks due to stored hormone
For carbimazole state the

1. Toxicity and interactions
Nausea, vomiting Rash (10%) Fever Marrow depression (0.5%) Jaundice
For carbimazole state the

1. Absorption and distribution
2. Metabolism and excretion
Well absorbed orally
Accumulates in thyroid gland
Half life 6 hours Excreted in urine
For Iodine state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Inhibition of organification and release of thyroid hormones
Decreases size and vascularity of the gland
Pre-operative decrease in gland size and vascularity
Thyroid storm
How does radioactive iodine work?
Concentrated by thyroid, emission of beta rays results in parenchymal destruction
What drugs are used to treat hypercalcaemia?
Saline diuresis
Bisphosphonates (inhibition of bone resorption) Etidronate
Calcitonin Gallium (inhibition of bone resorption) Phosphate
What glucocorticoids are available for pharmacological use?
What is prednisone?
Natural agents Hydrocortisone (cortisol)
Synthetic agents: Prednisolone, methylprednisolone Betamethasone, Dexamethasone
Prednisone is a prodrug of prednisolone
For glucocorticoids state the
1. Mechanism?
Steroid enters the cell as a free molecule
Binds to intracellular receptor that is bound to stabilising heat shock protein
Heat shock protein is released and the steroid- receptor complex enters the nucleus and bind to glucocorticoid response element on the DNA

This causes regulation of transcription and production of appropriate protein
For glucocorticoids state the
1. Toxicity and interactions
Cushings syndrome
Excessive acid and pepsin secretion by the stomach
Myopathy Psychosis Adrenal suppression
Contraindications Uncontrolled infection
For glucocorticoids state the
1. Absorption and distribution
2. Metabolism and excretion
Oral Topical Intramuscular Intravenous
Well absorbed orally
Bound to corticosteroid binding globulin and albumin

Hydrocortisone, prednisolone and methylprednisolone are short acting agents

Betamethasone and dexamethasone are long acting

Half life of hydrocortisone is 60-90 minutes
Metabolised by liver to active and inactive metabolites and excreted in the urine

Dose should be increased for patients on long term therapy during times of stress
Organ effects of glucocorticoids?
Intermediary metabolism:
Protein catabolism Gluconeogenesis and decreased peripheral glucose utilisation Ketogenesis

Permissive action effects: Required for glucagon and catecholamines to exert their effects
Inhibition of ACTH secretion
Facilitation of effective water excretion (unknown mechanism)

Therapeutic effects: Anti-inflammatory and immunosuppressant effects

Due to effects on concentration, distribution and function of peripheral white cells.
Increased circulating neutrophils, platelets and red blood cells
Reduced lymphocytes, eosinophils, monocytes and basophils
Reduced secretion of cytokines
Increased neutrophils results in decreased neutrophils at site of inflammation

Inhibition of leucocyte function, especially macrophages due to decreased mediator production

Reduced synthesis of prostaglandins and leukotrienes via action on phospholipase A2 and COX2
Vasoconstriction
Reduced capillary permeability
What are the relative potencies of the glucocorticoids?
Hydrocortisone - 1
Prednisolone - 5
Betamethasone - 25
Dexamethasone - 30

Hydrocortisone has mixed glucocorticoid and mineralocorticoid actions, prednisolone has minor mineralocorticoid actions, nil for betamethasone and dexamethasone
For ranitidine, cimetidine, famotidine state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Competitive antagonism at H2 receptors.
H2 receptors found in gastric enterochromaffin-like cells, cardiac muscle, mast cells and brain

Reduced gastric acid secretion
90% reduction in basal, food-stimulated and nocturnal secretion of gastric acid after a single dose
Reduced volume of gastric secretion and concentration of pepsin

Little effect on heart or blood pressure.

Peptic ulceration Oesophagitis and gastritis. Zollinger Ellison syndrome. Prevention of stress ulceration in critically ill.
Dose Ranitidine Oral 150mg twice daily

Cimetidine 400mg twice daily
Famotidine 20mg twice daily
For ranitidine, cimetidine, famotidine state the

1. Toxicity and interactions
Minor toxicities: Diarrhoea, dizziness, headache, rash.

GIT
May mask gastric malignancy Reversible cholestasis and hepatitis

CNS
acute confusion (cimetidine >> ranitidine)
Antiandrogen effects Gynecomastia, galactorrhoea. (cimetidine only)

Marrow
Blood dyscrasias (cimetidine >> ranitidine)
(Famotidine free of above effects)

Interactions Inhibition of cytochrome P450 and reduction of liver blood flow. Care with warfarin, phenytoin, beta blockers, benzodiazepines, tricyclics, calcium channel blockers, antiarrhythmics, alcohol. (cimetidine >> ranitidine)
Histamine antagonists reduce the absorption of ketoconazole
Inhibition of renal clearance of drugs (cimetidine >> ranitidine)

Contraindications: Porphyria
For ranitidine, cimetidine, famotidine state the
1. Absorption and distribution
2. Metabolism and excretion
20% plasma bound. 70% bioavailability.
Half life 1-3 hours
Mostly renal metabolism.
For proton pump inhibitors state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
?Reversible inhibition of parietal cell proton pump
For proton pump inhibitors state the

1. Toxicity and interactions
May mask gastric malignancy
May increase the risk of gastrointestinal infection
Nausea, vomiting, abdominal pain
Headache

Interactions:
Diazepam - decreased clearance Contraindications Severe hepatic dysfunction
For proton pump inhibitors state the

1. Absorption and distribution
2. Metabolism and excretion
Well absorbed orally Highly protein bound
Metabolised by liver and excreted in urine
For octreotide state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Somatostain analogue
Reduced portal pressure in chronic liver disease resulting in decreased risk of variceal haemorrhage
For prochlorperazine state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Phenothiazine Strong antiemetic and antipsychotic actions
Antidopamine action - therapeutic effect and unwanted effects including extrapyramidal disorders and endocrine disturbances.
Alpha-adrenoreceptor antagonism, which contributes to cardiovascular side effects, e.g. Orthostatic hypotension and reflex tachycardia.
Potentiation of noradrenaline by blocking its reuptake into nerve terminals.
Weak anticholinergic action, weak antihistamine action. Weak serotonin antagonism.
Prochlorperazine also has an effect on temperature control and blocks conditioned avoidance responses.
For prochlorperazine state the

1. Toxicity and interactions
Sedation, Hypotension
Anticholinergic effects, especially in elderly, especially constipation, dry mouth, blurred vision Severe acute dystonic reactions in children
Tardive dyskinesia
Neuroleptic malignant syndrome

Hypo/hyperthermia

Interactions: Enhances the effect of other CNS depressants

Contraindications: Hypotension (alpha- adrenoreceptor antagonism)

Drowsiness, coma, neuromuscular excitability, convulsions.
Miosis and loss of deep tendon reflexes. Hypotension and hypotermia.
Activated charcoal effective.
Supportive therapy.
Avoid adrenaline and lignocaine.
For prochlorperazine state the
1. Absorption and distribution
2. Metabolism and excretion
Orally active.
Metabolised by liver and excreted in faeces and urine.
For metoclopramide state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Dopamine antagonist
Increased upper gastrointestinal motility
Dose Oral-10mg tds
Intravenous -10mg tds, slowly Rapid injection leads to feelings of anxiety

Daily dose should not exceed 0.5mg/kg
For metoclopramide state the
1. Toxicity and interactions
Restlessness, fatigue, drowsiness in 10% Dystonic reaction ␣ more common in children and young adults Tardive dyskinaesia in chronic use Neuroleptic malignant syndrome
Interactions May affect absorption of other drugs due to effects on motility

Contraindications Phaechromocytoma ␣ causes increased release of catecholamines

Epilepsy - may increase the frequency of seizures Porphyria, Parkinson's
For metoclopramide state the

1. Absorption and distribution
2. Metabolism and excretion
Orally active
Metabolised in liver and excreted in urine
For ondansetron state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
5HT3 antagoni
For ondansetron state the
1. Toxicity and interactions
Dizziness Muscle pain Chest pain Seizures

Contraindications: IHD
Severe hepatic impairment
For ondansetron state the

1. Absorption and distribution
2. Metabolism and excretion
Orally active Protein bound
Metabolised by liver
For promethazine state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
First generation H1 antagonist
Phenothiazine derivative
Other agents: Diphenhydramine Chlorpheniramine
Competitive antagonism at H1 receptors. (note that some cardiovascular effects of histamine are mediated by H2 receptors)
also have antimuscarinic, anti-adrenergic, antiserotonin actions

Anaphylaxis Allergic reactions Motion sickness and vestibular dysfunction. Nausea (can also be used as a local anaesthetic as has some sodium blocking actions)
Dose 10-25mg orally, intravenous or intramuscular
For promethazine state the
1. Toxicity and interactions
Marked sedation. Antimuscarinic actions. Orthostatic hypotension Excitation and convulsions in children. Postural hypotension.
For promethazine state the

1. Absorption and distribution
2. Metabolism and excretion
Orally active. Widely distributed. Readily enter the CNS
Extensively metabolised by liver and excreted in urine.
For loratidine state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Second generation H1 antagonist
Other agents Terfenadine (terfenadine is a prodrug of fexofenadine that lacks cardiotoxic effects)
Competitive antagonism at H1 receptors. (note that some cardiovascular effects of histamine are mediated by H2 receptors)
also have anticholinergic, anti-adrenergic, antiserotonin actions
For loratidine state the

1. Toxicity and interactions
Mild toxicities compared with first generation agents
Interactions Terfenadine and astemizole metabolised by CYP3A4 which is inhibited by grapefruit, ketoconazole and macrolides ␣ associated with QT prolongation and potentially toxic arrhythmias

Fexofenadine is the metabolite of terfenadine and lacks cardiotoxic effects
For loratidine state the
1. Absorption and distribution
2. Metabolism and excretion
Widely distributed except CNS
Extensively metabolised by liver and excreted in urine.
For Penicillin V state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Inhibition of cell wall synthesis Penicillins bind to penicillin binding proteins and inhibit transpeptidation in peptidoglycan synthesis and therefore formation of cross- links in the cell wall that confer rigidity.

Active against gram positive cocci, gram negative cocci, some anaerobes
Destroyed by beta lactamases
Inactive against enterococci, some anaerobes, gram negative rods
Streptococci Meningococci Enterococci Pneumococci Staphylococci Treponema pallidum Bacillus anthracis Clostridium
Dose 10-50mg/kg/day in 3-4 doses orally or IV
For Penicillin V state the
1. Toxicity and interactions
Minor toxicities such as nausea, vomiting, diarrhoea
Important cause of type I hypersensitivity. Type III hypersensitivity can also occur.
5-8% claim penicillin allergy but only 5-10% of these will have a reaction.

High doses in renal failure can causes seizures
For Penicillin V state the
1. Absorption and distribution
2. Metabolism and excretion
Original penicillins such as penicillin G acid labile.
Penicillin V is acid stable and well absorbed orally but has poor bioavailability
Avoid administration with meals
60% protein bound.
Penetrates tissues very well except eye, prostate and CNS - though penetration is better if inflammation is present.

Renal excretion 10% by filtration, 90% by tubular secretion.
Half-life 30 minutes, increases to 10 hours in renal failure.
Dose adjustment required in renal failure
Frequent dosing required
For Flucloxacillin/dicloxacillin state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Beta lactam antibiotic with resistance to staphylococcal betalactamases.
Penicillins bind to penicillin binding proteins and inhibit transpeptidation in peptidoglycan synthesis and therefore formation of cross- links in the cell wall that confer rigidity.
Active against gram positive cocci including beta lactamase producing staphylococci
Inactive against enterococci, anaerobes, gram negative.
For Flucloxacillin/dicloxacillin state the

1. Toxicity and interactions
Minor toxicities such as nausea, vomiting, diarrhoea
Important cause of type I hypersensitivity. Type III hypersensitivity can also occur.
5-8% claim penicillin allergy but only 5-10% of these will have a reaction.
High doses in renal failure can causes seizures
Small risk of hepatitis hence introduction of dicloxacillin
For Flucloxacillin/dicloxacillin state the

1. Absorption and distribution
2. Metabolism and excretion
Acid stable and well absorbed orally.
Absorption impaired by food.
Highly protein bound.
Penetrates tissues very well except eye, prostate and CNS -though penetration is better if inflammation is present.

Hepatic metabolism and rapid renal excretion ␣ 10% by filtration, 90% by tubular secretion.
No adjustment in renal failure.
For Amoxicillin/Ampicillin/Piperacillin/Ticarcillin state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Inhibition of cell wall synthesis Penicillins bind to penicillin binding proteins and inhibit transpeptidation in peptidoglycan synthesis and therefore formation of cross- links in the cell wall that confer rigidity

Similar spectrum to penicillin but better penetration of gram negative bacteria, though still sensitive to beta lactamases

Streptococci Meningococci Pneumococci (particularly active therefore 1st choice for respiratory infection)
Staphylococci
Treponema pallidum
Bacillus anthracis
Clostridium
(not enterobacter)
Ampicillin effective for Shigella

Ampicillin not active against E coli Proteus
Haemophilus Klebsiella Pseudomonas
Enterobacter Citrobacter Serratia

Ticarcillin is also active against Pseudomonas Enterobacter

Piperacillin is also active against Klebsiella
What are some resistance mechanisms to penicillin? (name 3 )
1. Beta lactamases: Destroyed by beta lactamases produced by staphylococci, haemophilus, E coli, pseudomonas, enterobacter

2. Alteration on target penicillin binding proteins. Resistant organisms have binding sites with low affinity for binding - particularly seen with MRSA and pneumococcus

3. Poor ability to penetrate outer membrane Gram-negative organisms
For Amoxicillin/Ampicillin/Piperacillin/Ticarcillin state the
1. Absorption and distribution
2. Metabolism and excretion
Acid stable and well absorbed orally.
Highly protein bound.
Penetrates tissues very well except eye, prostate and CNS - though penetration is better if inflammation is present.
For clavulanic acid state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Resemble beta lactam molecules and protect against many beta lactamasesActive against beta lactamases produced by Haemophilus Neisseria gonorrhoea Salmonella
Shigella E coli Klebsiella Legionella Bacteroides
Not active against beta lactamases produced by Enterobacter Citrobacter
Serratia Pseudomonas
For 1st generation cephalosporins state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
4. examples
Cefadroxil Cefazolin Cephalexin Cephalothin Cephadrine
Cephalosporins bind to penicillin binding proteins and inhibit transpeptidation in peptidoglycan synthesis and therefore formation of cross-links in the cell wall that confer rigidity.

Gram positive cocci plus E coli Klebsiella
Proteus Anaerobic cocci
Peptococcus Peptostreptococcus

Not active against:
Listeria MRSA Haemophilus Pseudomonas Some proteus Enterobacter Serratia Citrobacter

Surgical prophylaxis Uncomplicated UTI, skin and soft tissue infection
For 1st generation cephalosporins state the

1. Toxicity and interactions
Cross allergy between penicillins and cephalosporins is 5-10% - withhold in anaphylaxis only.
Toxicity Local irritation.
Superinfection.
For 1st generation cephalosporins state the

1. Absorption and distribution
2. Metabolism and excretion
Well absorbed orally
Renal excretion 10% by filtration, 90% by tubular secretion.
Half-life 30 minutes, increases to 10 hours in renal failure.Gram positive cocci plus E coli Klebsiella
Proteus Anaerobic cocci
Peptococcus Peptostreptococcus
Plus extended gram negative cover against Haemophilus Some serratia
Not active against Listeria MRSA Pseudomonas Some proteus Enterobacter Some serratia Citrobacter
Dose adjustment required in renal failure
For 2nd generation cephalosporins state the
1. Examples and Mechanism?
2. Organ effects?
3. Clinical use?
Cefaclor Cefuroxime Cefoxitin
Cephalosporins bind to penicillin binding proteins and inhibit transpeptidation in peptidoglycan synthesis and therefore formation of cross-links in the cell wall that confer rigidity.
For 2nd generation cephalosporins state the
1. Toxicity and interactions
Cross allergy between penicillins and cephalosporins is 5-10% - withhold in anaphylaxis only

Local irritation. Cefaclor associated with serum-sickness like reaction
Superinfection.
For 2nd generation cephalosporins state the

1. Absorption and distribution
2. Metabolism and excretion
Renal excretion 10% by filtration, 90% by tubular secretion.
Half-life variable
Dose adjustment required in renal failure
For 3rd generation cephalosporins state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Ceftriaxone Cefotaxime Cephtazidime
Extended coverage of gram-negative organisms compared with first and second generation.
Citrobacter Serratia Haemophilus Neisseria
Particularly pseudomonas.
Less active against gram-positive organisms.
Not active against enterococci or listeria.

Treatment of serious infections by susceptible organisms.
Treatment of serious infection if organism unknown.
Especially useful for CNS infection.
Treatment of penicillin resistant infections including MRSA and gonorrhoea
Dose 10-50mg/kg/day. Ceftriaxone suitable for once daily dosing.
For 3rd generation cephalosporins state the
1. Toxicity and interactions
Allergy Cross allergy between penicillins and cephalosporins is 5-10% - withhold in anaphylaxis only.
Toxicity Local irritation
For 3rd generation cephalosporins state the
1. Absorption and distribution
2. Metabolism and excretion
Intravenous dosing. Good tissue penetration, especially into CNS.
Half-life 7-8 hours.
Metabolised by liver and excreted in bile.
No dosing adjustment required in renal failure
For Meropenem/Imipenem state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Structurally related to beta lactams
Inhibition of cell wall synthesis Binds to penicillin binding proteins and inhibit transpeptidation in peptidoglycan synthesis and therefore formation of cross- links in the cell wall that confer rigidity.

nfections due to resistant organisms
Highly active against resistant pneumococci and enterobacter
For Meropenem/Imipenem state the

1. Toxicity and interactions
Minor toxicities including nausea, vomiting, diarrhoea and skin rashes
High doses in renal failure can causes seizures
For Meropenem/Imipenem state the

1. Absorption and distribution
2. Metabolism and excretion
Inactivated by dehydropeptidases in renal tubules therefore administered with cilastatin
For Vancomycin state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Binds to peptidoglycan and inhibits transglycosylase therefore preventing peptidoglycan elongation and cross-linking that confers rigidity.
Active against gram positive bacteria (plus flavobacterium)
Bactericidal
Synergistic with gentamicin

Resistance mechanisms: Alteration of binding site.
For Vancomycin state the
1. Toxicity and interactions
Minor reactions in 10% Phlebitis Histamine release (red man/red neck syndrome)
Ototoxicity and nephrotoxicity, especially if administered with aminoglycoside
For Vancomycin state the
1. Absorption and distribution
2. Metabolism and excretion
Poorly absorbed orally -used orally for the treatment of resistant clostridium difficile
90% filtered by kidney
Dose adjustment required in renal failure
Not removed by haemodialysis
For Teicoplanin state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Binds to peptidoglycan and inhibits transglycosylase therefore preventing peptidoglycan elongation and cross-linking that confer rigidity.

Active against gram positive bacteria (plus flavobacterium)
Synergistic with gentamicin
For Teicoplanin state the

1. Toxicity and interactions
Poorly absorbed orally ␣ used orally for the treatment of resistant clostridium difficile
For Teicoplanin state the
1. Absorption and distribution
2. Metabolism and excretion
Poorly absorbed orally -used orally for the treatment of resistant clostridium difficile
90% filtered by kidney
Dose adjustment required in renal failure
For chloramphenicol state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Potent inhibition of microbial protein synthesis
Reversibly binds to 50S subunit of bacterial ribosome
Bacteristatic

Broad spectrum
Not effective for
chlamydia

Effectively obsolete as a systemic drug due to other less toxic agents
Eye infections ␣ due to broad spectrum and good tissue penetration
Not effective for chlamydia
For chloramphenicol state the
1. Toxicity and interactions
2. resistance mechanisms
1.Toxicity GIT Nausea, vomiting, diarrhoea
Bone marrow Commonly causes dose related reversible bone marrow suppression
Rare idiosyncratic aplastic anaemia (1 in 30000)
Neonates: Grey baby syndrome

Interactions: Inhibition of hepatic microsomal enzymes -prologed half life and increased concentrations of phenytoin and warfarin

2. Decreased permeability
Production of chloramphenicol acetyltransferase that inactivates the drug
For chloramphenicol state the
1. Absorption and distribution
2. Metabolism and excretion
Well absorbed orally
Widely distributed
Good tissue penetration

Metabolised by liver and excreted by kidney
Dose adjustment required in hepatic failure
For tetracyclines state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Potent inhibition of microbial protein synthesis
Reversibly binds to 30S subunit of bacterial ribosome
Enter microorganisms by diffusion and active transport

Broad spectrum
Active against Rickettsiae, Chlamydiae, Mycoplasma, Vibrio
Also active against some protozoa
For tetracyclines state the
1. Toxicity and interactions
GIT Nausea, vomiting, diarrhoea Bacterial overgrowth
Liver toxicity
ATN

Bones and teeth: Tooth discolouration due to chelation with calcium
Other Photosensitivity

Interactions: Enzyme inducers such as phenytoin and carbamazepine reduce half life by 50%
For tetracyclines state the
1. Absorption and distribution
2. Metabolism and excretion
Well absorbed orally
Absorption not impaired by food
Impaired by divalent cations and dairy products
40-80% protein bound
Widely distributed
except CNS

Metabolised by liver, excreted in urine and bile.
Bile concentration 10 times serum concentration
Dose reduction required in renal failure
Doxycycline is excreted by non- renal mechanism and therefore is drug of choice in renal failure
Half life 12-16 hours
For macrolides state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Potent inhibition of microbial protein synthesis
Reversibly binds to 50S subunit of bacterial ribosome
Concentrated in polymorphs and macrophages
Bacteristatic at low concentrations, bactericidal at high concentrations

Erythromycin
Semi-synthetic Roxithromycin Clarithromycin Azithromycin

Broad spectrum
Gram positive (strep>>staph) Gram negative
Neisseria Bordetella Rickettsia Treponema Campylobacter
Chlamydia Mycoplasma Legionella
Less active against haemophilus and staphylococcus
Clarithromycin more active against mycobacterium avium intracellulare
For macrolides state the
1. Toxicity and interactions
Nausea, vomiting, diarrhoea
Acute cholestatic hepatitis
(semi-synthetic macrolides better tolerated)

Interactions: Erythromycin and clarithromycin Inhibition of cytochrome P450 resulting in increased concentrations of theophylline, warfarin, antihistamines

Causes increased bioavailability of digoxin
Semi-synthetic macrolides relatively free of above effects due to less avid binding to P450
For macrolides state the
1. Absorption and distribution
2. Metabolism and excretion
Erythromycin base combined with stearate or ester confers acid stability
Widely distributed
except CNS

Erythromycin Metabolised by liver, excreted in bile
No adjustment necessary for renal impairment.
Half life 1.5 hours
Synthetic macrolides metabolised by liver and excreted in bile and urine therefore dose adjustment in renal failure is recommended
For aminoglycosides state the
1. Mechanism?
2. Organ effects?

Gentamycin Tobramycin Netilmycin
Aminoglycoside enters the bacteria by passive diffusion via porin channels across the outer membrane (this process is aided by penicillins)
Aminoglycoside is then actively transported into the cytoplasm
Binds to 30S subunit of bacterial ribosome
Bactericidal

Gram negative Pseudomonas
Proteus Enterobacter Klebsiella Serratia
E coli
Some gram positive activity Streptococci and enterococci are relatively resistant
No action against anaerobes
Tobramycin is more active against pseudomonas


concentration dependent killing is more important than time-dependent killing therefore once daily dosing
For aminoglycosides state the
1. Toxicity and interactions

Gentamycin Tobramycin Netilmycin
Ototoxic, vestibulotoxic and nephrotoxic ␣ more likely in prolonged use, elderly, renal insufficiency and concurrent use of other nephrotoxic substances
Ototoxicity manifests mainly as vestibular dysfunction
Gentamycin is mostly nephrotoxic and vestibulotoxic (less ototoxicity)

Nephrotoxicity occurs in 5- 25% of patients receiving drug for more than 3-5 days
Neuromuscular blockade can occur in very high doses
For aminoglycosides state the
1. Absorption and distribution
2. Metabolism and excretion

Gentamycin Tobramycin Netilmycin
Not orally active -virtually the entire oral dose is excreted in faeces
Highly polar compounds poorly distributed to CNS and eye though inflammation increases penetration.
Poor activity in low pH or low oxygen tension.

Half life 2-3 hours
Renally excreted
Dose adjustments required in renal insufficiency
Trough concentration should be less than 2ug/ml
Aminoglycoside clearance is directly proportional to creatinine clearance Cockcroft-gault formula
Resistance mechanisms and contraindications for tetracyclines?
Resistance mechanisms:
Decreased intracellular accumulation due to impaired active transport
Decreased binding to ribosome due to production of inhibitory proteins
Enzymatic inactivation
(note resistance is common)

Contraindications Children under 8 years
Resistance mechanisms to macrolides?
Decreased intracellular accumulation due to decreased permeability
Decreased binding to ribosome due to modification of the binding site by methylase (accounts for 90% of resistance)
Enzymatic inactivation by enterobacter
Resistance mechanisms to gentamycin?
Decreased intracellular accumulation due to abnormal porins or anaerobic conditions Gram positive organisms are resistant by this mechanism

Decreased binding to ribosome due to modification of the binding site

Enzymatic inactivation by transferases (most important for gram negative resistance ␣ netilmycin is relatively resistant to this)
For sulphonamides state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
A nti-folate
Sulphonamides are structural analogues of para- aminobenzoic acid that bind to dihydropteroate synthase and competitively inhibit folic acid synthesis
Bacteriostatic Bacteriocidal when given with trimethoprim

Broad spectrum
Gram positive and gram negative actions
Includes Chlamydia
Stimulates growth of Rickettsia

Used in combination with trimethoprim in the treatment of urinary tract infection, respiratory tract infections and in episodes of resistance
Resistance common Use is limited by toxicity Very cheap therefore extensive use in 3rd world
For sulphonamides state the
1. Toxicity and interactions
5% of patients have side effects
Nausea, vomiting, diarrhoea Fever Exfoliative dermatitis Photosensitivity
Stevens Johnson syndrome
GIT
Sulfonamides may precipitate in urine and may cause obstruction
Marrow
Aplastic anaemia
Contraindications Porphyria
For sulphonamides state the
1. Absorption and distribution
2. Metabolism and excretion
Orally active (slow)
Sulfamethoxazole chosen due to its similar half life to trimethoprim
Wide distribution

Metabolised in liver and excreted in urine.
Metabolised impaired in slow acetylators
Dose adjustment required in renal insufficiency
Half life 10-12 hours
For Trimethoprim state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
A nti-folate Inhibition of dihydrofolate
reductase
Synergistic effect when given with sulphonamide due to sequential action in folate synthesis

Used in combination with trimethoprim in the treatment of urinary tract infection, respiratory tract infections, skin infections



Broad spectrum
Especially E coli Enterobacter Proteus
Neisseria Salmonella Klebsiella
Haemophilus
Not active against Pseudomonas Mycoplasma Mycobacterium Treponema
For Trimethoprim state the
1. Toxicity and interactions
Toxicity Nausea, vomiting, diarrhoea

GIT
Sulfonamides may precipitate in urine and may cause obstruction

Marrow
Megaloblastic anaemia

Contraindications: Porphyria
For Trimethoprim state the
1. Absorption and distribution
2. Metabolism and excretion
Concentrated in prostate, vagina., kidney and lungs
Metabolised in liver and excreted in urine.
Dose adjustment required in renal insufficiency
Half life 10-12 hours
Difference between heparin and LMWH?
Heterogeneous mixture of sulphated muco-polysacc- harides.
Enoxaparin Low molecular weight heparin
For Quinolone state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
the important quinolones are synthetic fluorinated analogues of nalidixic acid.
Nalidixic acid Norfloxacin Ciprofloxacin Ofloxacin Sparfloxacin

DNA gyrase inhibitors
Block relaxation of positively supercoiled DNA required for normal transcription and replication of bacteria.


Active against a variety of gram negative and gram-positive bacteria.
Nalidixic acid ␣␣␣␣␣␣␣␣␣␣␣␣␣␣␣␣ systemic levels therefore only used for urinary tract infections.
Norfloxacin Least active Ciprofloxacin Particularly active against gram-negative cocci and bacilli including enterobacter, pseudomonas, neisseria, haemophilus and campylobacter.
Less effective against gram-positive organisms especially streptococci.
Ofloxacin Sparfloxacin Improved gram-positive action. Longer half-life.
Anaerobes are generally resistant though intracellular organisms are susceptible.
For Quinolone state the

1. Toxicity and interactions
Well tolerated.
General Nausea Diarrhoea Headache Rash
May damage growing cartilage therefore not recommended in children unless no other drug available or suitable
Little data on pregnancy. Excreted in breast milk.

Interactions: Enzyme inducer - increases the metabolism of phenytoin
For Quinolone state the
1. Absorption and distribution
2. Metabolism and excretion
Well absorbed orally with greater than 80% bioavailability.
Concentrates in prostate and kidney.

Half-life 3-4 hours.
Excreted renally therefore accumulates in renal failure.
For metronidazole state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Antiprotozoal agent with potent anti-anaerobic actions
Reduction of the nitro group produces toxic metabolites

Only active against obligate anerobes

Bacteroides Fusobacterium
Clostridium
Anaerobic streptococci
Trichomoniasis
Giardiasis
Amoebiasis
For metronidazole state the
1. Toxicity and interactions
General: Nausea, vomiting, diarrhoea Metallic taste Dry mouth Headache Disulfiram-like effect with alcohol Pancreatitis

CNS: Ataxia, seizures
Interactions Potentiates the effect of warfarin

Reduced half life if taken with enzyme inducers phenytoin, phenobarbitone Increased half life if taken with enzyme inhibitors cimetidine
For metronidazole state the
1. Absorption and distribution
2. Metabolism and excretion
Well absorbed orally Also given rectally and intravenously

Metabolised by liver
Half life 7 hours
May accumulate in hepatic dysfunction
For acyclovir state the
1. Mechanism?
2. Organ effects?
3. Clinical use?
Antiherpes agent
Selectively activated by phosphorylation in infected cells only.
Acyclovir triphosphate inhibits vial DNA synthesis

Active against HSV1 and HSV2 Varicella zoster
For acyclovir state the
1. Toxicity and interactions
Nausea, vomiting Headache
Rapid intravenous administration may be associated with renal insufficiency and neurological toxicity
For acyclovir state the
1. Absorption and distribution
2. Metabolism and excretion
Oral, topical and intravenous formulations
Well absorbed orally, low bioavailability, hence frequent dosing
Distributed to most tissues

Cleared by glomerular filtration and tubular secretion.
Half life 3-4 hours
Dosage adjustment required for renal impairment
Mechanism of resistance to quinolones?
Uncommon point mutation in the quinolone-binding region.
Define
1. disinfection
2. antiseptic
3. sterilisation
4. autoclaving
1. a chemical or physical process that kills micro-organisms
2. a disinfectant with sufficiently low toxicity to use on skin, mucous membranes and wounds
3. a process to remove all micro-organisms, spores and viruses
4. sterilisation using pressurised steam at 120 degrees for 20 minutes
Resistance mechanisms to sulphonamides?
Some bacteria utilise exogenous folate therefore are not susceptible
Decreased intracellular accumulation reduced permeability
Decreased binding to dihydropteroate synthase
Resistance is common
Resistance mechanisms to trimethoprim?
Resistance mechanisms Some bacteria utilise exogenous folate therefore are not susceptible
Decreased intracellular accumulation due to reduced permeability
Decreased binding to dihydrofolate reductase