ACEM Pharm part 2 - Sheet1 (1) Flashcards
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?
- 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.
- Respiratory depression and apnoea.
- Metabolic acidosis
Toxicity of salycilates?
- Salicylism: tinnitus, reduced hearing, vertigo.
- Hyperventilation, fever, dehydration
- Metabolic acidosis due to salicylic acid dissociation, deranged carbohydrate metabolism and reduced renal function.
- 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?
- 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
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
- gluconeogenesis?
- glycogeniolysis?
- Lipolysis?
- Decreases lipolysis?
- B2, A
- B2, A
- B3
- A2
What adrenoceptor increases renin release at the kidney?
b1
What adrenoceptor causes
- bladder wall contraction?
- Bladder wall relaxation?
- Bladder sphincter contraction?
- Bladder sphincter relaxation
- B2
- M3
- A1
- 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?
- Glaucoma: pilocarpine, methacholine, carbachol.
- Paralytic ileus: bethanechol
- Urinary retention: bethanechol
- 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:
- Receptor sequestration -rapid and transient event decrease in receptor availability.
- Down-regulation -reduced receptors due to reduced synthesis.
- 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:
- adrenaline?
- Isoprenaline?
- Noradrenaline
- Dopamine
- 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
- Phentolamine?
- Phenoxybenzamine?
- Prazosin?
- 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) - Phenoxybenzamine: A1 selective antagonist. (also blocks Ach, H1, 5HT). Blockade of catecholamine induced vasoconstriction.
- Prazosin: Potent A1 antagonist. Decreased total peripheral resistance due to relaxation of arterial and venous smooth muscle mediated by alpha 1 blockade.
Toxicity of
- Phentolamine?
- Phenoxybenzamine?
- 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
- Phentolamine?
- Phenoxybenzamine?
- 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
- Propanolol?
- Metoprolol?
- Atenolol?
- Esmolol?
- Labetolol?
- Carvedilol?
- Propanolol: Non-selective beta antagonist No action at alpha or muscarinic receptors
- Metoprolol: B1 selective
- Atenolol: B1 selective.
- Esmolol B1 selective
- Labetolol: Mixed B1 antagonist and alpha antagonist
- 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
- metoprolol?
- atenolol?
- esmolol?
- Metoprolol: Extensive first pass metabolism
Low bioavailability, Half life 3-4 hours - Atenolol: Less extensive first pass metabolism Low bioavailability. Half life 6-9 hours
- 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:
- Mechanism?
- Organ effects?
- Clinical use?
- Centrally acting alpha agonist. Inhibition of dopa decarboxylase and depletion of noradrenaline
A2 actions greater than A1 - Centrally mediated reduction in total peripheral resistance with variable reduction in heart rate and cardiac output.
- 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:
- Absorption and distribution
- Metabolism and excretion
- Absorption Orally active. Occasional IV use occasional.
Distribution Active transport into brain - 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:
- Mechanism?
- Organ effects?
- Clinical use?
- Centrally acting partial agonist at alpha receptors, causing reduced sympathetic tone and increased parasympathetic tone. Preference for A2
- Reduction in blood pressure and mild bradycardia.
- Hypertension
For clonidine state the:
1. Toxicity and interactions
Sedation, dry mouth. Depression. Reactive hypertensive crisis if rapidly withdrawn.
For clonidine state the:
- Absorption and distribution
- Metabolism and excretion
Orally active, bioavailability 75%.
Distribution Lipid soluble and rapidly enters the brain.
Half life 8-12 hours
For acetazolamide state the:
- Mechanism?
- Organ effects?
- Clinical use?
- carbonic anhydrase inhibitor: Carbonic anhydrase most prominent in the luminal membrane of the PCT
- Profound depression of bicarbonate reabsorption in the proximal tubule
Reduced production of bicarbonate by the ciliary body - 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:
- Absorption and distribution
- Metabolism and excretion
Well absorbed orally Increased urinary pH within 30 minutes
For frusemide state the:
- Mechanism?
- Organ effects?
- Clinical use?
- 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 - Acute pulmonary oedema Hypertension Hyperkalaemia Hypercalcaemia
Dose 20-80mg PO/IV
For frusemide state the:
- 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:
- Absorption and distribution
- 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:
- Mechanism?
- 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:
- Toxicity and interactions
Toxicity: Hypokalaemic metabolic alkalosis Hyperuricaemia
Hyperlipidaemia
Hyponatraemia
Allergic reaction
For thiazide state the:
- Mechanism?
- Organ effects?
- Clinical use?
- Toxicity and interactions
- Absorption and distribution
- 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:
- Mechanism?
- Organ effects?
- Clinical use?
- Spironolactone: Competitive antagonism of aldosterone at mineralocoticoid receptors
Triamterene, amiloride: Direct inhibition of sodium reabsorption in the CD
- Decreased sodium reabsorption Decreased potassium excretion
- 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:
- Toxicity and interactions
Toxicity: Hyperkalaemia Hyperchloraemic metabolic acidosis Gynaecomastia
Interactions: Triamterene and indomethacin have caused acute renal failure
For potassium sparing diuretics state the:
- Absorption and distribution
- 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:
- Mechanism?
- 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:
- Toxicity
Extracellular volume expansion -may exacerbate pulmonary oedema of CCF
Dehydration
For Hydralazine state the
- Mechanism?
- Organ effects?
- Clinical use?
- Direct acting vasodilator
Mechanism unknown -may interfere with calcium entry into the cell and cause electromechanical decoupling. - Arterial dilator - little effect on veins.
Lowers systemic vascular resistance to cause decreased blood pressure. - Severe hypertension.
Severe pre- eclampsia
Dose Oral : 50- 200mg/day in divided doses IV ␣ 20-40mg slowly
For Hydralazine state the
- 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
- Absorption and distribution
- Metabolism and excretion
- Oral or intravenous preparations
- 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
- Mechanism?
- Organ effects?
- Clinical use?
- Direct acting vasodilator.
Acts by activation of guanylyl cyclase resulting in increased cGMP which relaxes smooth muscle.
No effect on other smooth muscle - Arterial and venous dilator (more active on veins)
Lowers peripheral vascular resistance and venous return to cause decreased blood pressure - 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
- 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
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- Clinical use?
- Direct acting vasodilator Mechanism unclear
May act by opening potassium channels in smooth muscle membrane.
May have a calcium antagonist action - 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. - Hypertensive emergencies.
Intractable hypoglycaemia
Dose 5mg/kg
For Diazoxide state the
- Toxicity and interactions
CVS
Excessive hypotension. Reflex tachycardia - may provoke angina. Increased plasma renin activity.
Endocrine: Inhibits insulin release.
For Diazoxide state the
- Absorption and distribution
- Metabolism and excretion
Intravenous
Extensive albumin binding.
Metabolised in liver and excreted in urine.
For minoxidil state the
- Mechanism?
- Organ effects?
- Clinical use?
- 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. - Severe hypertension refractory to other antihypertensives
Dose 5-40mg/day
For minoxidil state the
- Toxicity and interactions
Headache, nausea, anorexia, palpitations. Reflex tachycardia - may provoke angina.
For minoxidil state the
- Absorption and distribution
- Metabolism and excretion
- Well absorbed orally Does not enter CNS
- Metabolised by liver and excreted in urine
Half life 4 hours.
For ACEI state the
- Mechanism?
- Organ effects?
- Clinical use?
- 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. - 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. - 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
- 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
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- Clinical use?
Glyceryl trinitrate, Isosorbide mononitrate, Isosorbide dinitrate
Nitric or nitrous acid esters of polyalcohols.
- 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. - 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.
- Stable and unstable angina. LVF. Hypertension.
Dose SL 300mcg TD 5-10mg/24hrs IV 10-400mcg/min
For nitrates state the
- 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
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- Clinical use?
- 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 - 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
- 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
- Abosrption and distribution
- 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
- Mechanism and structure?
- Organ effects?
- 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
- 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. - 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
- 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
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- Clinical use?
- 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) - Increased QRS and QT intervals
- Atrial flutter/fibrillation. Ventricular tachycardia
For quinidine state the
- 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
- Absorption and distribution
- Metabolism and excretion
- Well absorbed orally. 80% bound to plasma proteins.
- Metabolised in liver. 20% excreted unchanged in urine - enhanced by acidic urine.
Half life 6 hours.
For procanimide state the
- Mechanism?
- Organ effects?
- Clinical use?
- 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. - Action potential and refractory period are lengthened
Increased QRS and QT intervals - Atrial flutter/fibrillation. Ventricular tachycardia
For procanimide state the
- 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
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- Clinical use?
- 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. - Action potential and refractory period are shortened
- Ventricular tachycardia and fibrillation after defibrillation.
Local anaesthetic.
For Lignocaine state the
- 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
- Absorption and distribution
- Metabolism and excretion
- 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)
- Metabolised in liver and plasma.
Clearance dependent on hepatic blood flow
Excreted in urine.
Half life 1.5 hours
For Flecanide state the
- Mechanism?
- Organ effects?
- 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
- Toxicity and interactions
Safe drug with few side effects.
Pro arrhythmogenic
For Flecanide state the
1. Mechanism?
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- 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.
- 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. - 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
- 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
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- Clinical use?
class II and III Non-selective beta blocker Lengthens the action potential and causes increased refractory period
- 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
- 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
- Absorption and distribution
- Metabolism and excretion
Well absorbed orally
Largely excreted unchanged by kidney therefore dosage adjustment is required in renal impairment
For adenosine state the
- Mechanism?
- Organ effects?
- 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
- 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
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- 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
- Toxicity and interactions
- 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
- Absorption and distribution
- Metabolism and excretion
- 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 - 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
- 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
- Absorption and distribution
- Metabolism and excretion
- 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. - 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
- Mechanism?
- Organ effects?
- 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
- 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
- Absorption and distribution
- Metabolism and excretion
Intravenous administration, remains intravascular
Half life 23 minutes
For TPA (tenecteplase) state the
- Mechanism?
- 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
- 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
- Absorption and distribution
- 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
- Mechanism?
- 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
- Absorption and distribution
- 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
- Mechanism?
- Toxicity?
- 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
- Mechanism?
- Toxicity?
- ADME?
- Inhibition of ADP pathway of platelets
- Bleeding Leukopenia. Nausea, dyspepsia, diarrhoea.
- orally active
For Abciximab state the
- Mechanism?
- Organ effects?
- 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
- Toxicity and interactions
Bleeding Thrombocytopenia Multiple minor toxicities
For Abciximab state the
- Absorption and distribution
- Metabolism and excretion
Intravenous bolus dose or infusion
For Dipyridamole state the
- Mechanism?
- Organ effects?
- 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
- Toxicity and interactions
Potent vasodilator therefore caution in severe coronary artery disease
Headache Nausea, vomiting
For Dipyridamole state the
- Absorption and distribution
- Metabolism and excretion
Orally active
Widely distributed
Metabolised in the liver Excreted in bile
For Clopidogrel state the
- Mechanism?
- Organ effects?
- Clinical use?
Inhibits binding of ADP to GpIIb/IIIa and therefore inhibits platelet aggregation
Antithrombotic
Dose 75mg daily
For Clopidogrel state the
- Toxicity and interactions
Toxicity Bleeding
Interactions Aspirin, heparin -additive effect
For Clopidogrel state the
- Absorption and distribution
- Metabolism and excretion
Orally active Widely distributed
Metabolised in the liver
Active metabolite is responsible for clinical effect Excreted in bile
Vitamin K
- subtypes?
- Mechanism?
- 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
- Synthesis
- Mechanism?
- Organ effects?
- Clinical use?
- Toxicity and interactions
- Absorption and distribution
- 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 ?
- Toxicity and interactions
- Absorption and distribution
- Metabolism and excretion
- Hypoglycaemia Insulin allergy and resistance Lipodystrophy
- 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. - 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
- Mechanism?
- Organ effects?
- Clinical use?
(second generation agents) Glibenclamide Glimepiride
- 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
- 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
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- Clinical use?
- Unclear mechanism Direct stimulation of glycolysis Reduced gluconeogenesis Decreased glucose absorption
Reduced plasma glucagon - Dose 500mg-3g/day
For biguanides (metformin) state the
- 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
- Absorption and distribution
- Metabolism and excretion
Well absorbed orally Not bound to plasma proteins
Excreted unchanged in urine
Half life 1-3 hours
For glucagon state the
- Mechanism?
- 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
- Mechanism?
- Organ effects?
- 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
- Toxicity and interactions
Nausea, vomiting Rash (10%) Fever Marrow depression (0.5%) Jaundice
For carbimazole state the
- Absorption and distribution
- Metabolism and excretion
Well absorbed orally
Accumulates in thyroid gland
Half life 6 hours Excreted in urine
For Iodine state the
- Mechanism?
- Organ effects?
- 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
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- 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
- Toxicity and interactions
Minor toxicities: Diarrhoea, dizziness, headache, rash.
GIT
May mask gastric malignancy Reversible cholestasis and hepatitis
CNS
acute confusion (cimetidine»_space; ranitidine)
Antiandrogen effects Gynecomastia, galactorrhoea. (cimetidine only)
Marrow
Blood dyscrasias (cimetidine»_space; 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»_space; ranitidine)
Histamine antagonists reduce the absorption of ketoconazole
Inhibition of renal clearance of drugs (cimetidine»_space; ranitidine)
Contraindications: Porphyria
For ranitidine, cimetidine, famotidine state the
- Absorption and distribution
- Metabolism and excretion
20% plasma bound. 70% bioavailability.
Half life 1-3 hours
Mostly renal metabolism.
For proton pump inhibitors state the
- Mechanism?
- Organ effects?
- Clinical use?
?Reversible inhibition of parietal cell proton pump
For proton pump inhibitors state the
- 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
- Absorption and distribution
- Metabolism and excretion
Well absorbed orally Highly protein bound
Metabolised by liver and excreted in urine
For octreotide state the
- Mechanism?
- Organ effects?
- Clinical use?
Somatostain analogue
Reduced portal pressure in chronic liver disease resulting in decreased risk of variceal haemorrhage
For prochlorperazine state the
- Mechanism?
- Organ effects?
- 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
- 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
- Absorption and distribution
- Metabolism and excretion
Orally active.
Metabolised by liver and excreted in faeces and urine.
For metoclopramide state the
- Mechanism?
- Organ effects?
- 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
- Absorption and distribution
- Metabolism and excretion
Orally active
Metabolised in liver and excreted in urine
For ondansetron state the
- Mechanism?
- Organ effects?
- 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
- Absorption and distribution
- Metabolism and excretion
Orally active Protein bound
Metabolised by liver
For promethazine state the
- Mechanism?
- Organ effects?
- 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
- Absorption and distribution
- Metabolism and excretion
Orally active. Widely distributed. Readily enter the CNS
Extensively metabolised by liver and excreted in urine.
For loratidine state the
- Mechanism?
- Organ effects?
- 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
- 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
- Absorption and distribution
- Metabolism and excretion
Widely distributed except CNS
Extensively metabolised by liver and excreted in urine.
For Penicillin V state the
- Mechanism?
- Organ effects?
- 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
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- 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
- 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
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- 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 )
- Beta lactamases: Destroyed by beta lactamases produced by staphylococci, haemophilus, E coli, pseudomonas, enterobacter
- Alteration on target penicillin binding proteins. Resistant organisms have binding sites with low affinity for binding - particularly seen with MRSA and pneumococcus
- Poor ability to penetrate outer membrane Gram-negative organisms
For Amoxicillin/Ampicillin/Piperacillin/Ticarcillin state the
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- 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
- Mechanism?
- Organ effects?
- Clinical use?
- 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
- 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
- Absorption and distribution
- 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
- Examples and Mechanism?
- Organ effects?
- 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
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- 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
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- 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
- 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
- Absorption and distribution
- Metabolism and excretion
Inactivated by dehydropeptidases in renal tubules therefore administered with cilastatin
For Vancomycin state the
- Mechanism?
- Organ effects?
- 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
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- 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
- Toxicity and interactions
Poorly absorbed orally ␣ used orally for the treatment of resistant clostridium difficile
For Teicoplanin state the
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- 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
- Toxicity and interactions
- 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
- Decreased permeability
Production of chloramphenicol acetyltransferase that inactivates the drug
For chloramphenicol state the
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- 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
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- 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
- Absorption and distribution
- 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
- Mechanism?
- 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
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- 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
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- 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
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- 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
- 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
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- 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
- Absorption and distribution
- 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
- Mechanism?
- Organ effects?
- 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
- Absorption and distribution
- 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
- disinfection
- antiseptic
- sterilisation
- autoclaving
- a chemical or physical process that kills micro-organisms
- a disinfectant with sufficiently low toxicity to use on skin, mucous membranes and wounds
- a process to remove all micro-organisms, spores and viruses
- 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