Past Q completed study Q Flashcards
Define isomer
molecules with the same atomic formula however they have different structures/chemical bonds
Divisions of isomers
They can be broken up into structural isomers where the order of atomic bonds differs or steroisomers with the same chemical constituents and bond structure but different confirguration.
Stereoisomer divisions?
- Optical isomers have one or more chiral centre which is a carbon atoms or quaternary nitrogen surrounded by 4 different chemical groups - those with a single chiral centre have 2 confirmations and are referred to as enantiomers. Optical isomers with more than one chiral centre are diastereoisomers
- Geometric isomers are where a molecule has dissimilar groups attached to 2 atoms often carbon linked either by a double bond or ring structure and free rotation is restricted
Enantiomer define? Types
Enantiomer - An optical stereoisomer that is a mirror image of another but not superimposable with rotation - has a single chiral centre. Same bond structure, atomic make up and 3D shape. Nomenclature is to use R (rectus) and S(sinister) to describe configuration, or dextrose (+) or Levo (-) where optical activity is known but absolute structure is not
Why does being an isomer affect pharmacodynamics? Provide example?
- Pharmacodynamics - Pharmacodynamic profile will be affected according to the mechanism of action of the drug
◦ Receptor mediated MOA - potency and affinity for receptors are effected by confirmational change so minor structural modifications including mirror image enantiomers can have significant effects on MOA either increasing or decreasing affinity, as well as potency
‣ E.g. The relative potency may be markedly different such as in etomidate R vs S
‣ The relative potencies may also be similar e.g. isofluorane
‣ The property of biological molecules to show stronding preference for one enantiomer over another is called stereoselectivity
◦ Ketamine
‣ S Ketamine enantiomer has more potent dissociative effects and anaesthetic potency; however the R isoforms may be a better antidepressant
◦ Physicochemical MOA - drug effect is likely to be unaffected
How does enantiomers and isomers affect pharmaceutics
◦ Molecular and physical properties are identical e.g. boiling point , pH
◦ However they do rotate polarised light in different directions; may appear distinct under X-ray crystallography and different reaction kinetics with chiral catalysts.
◦ Enantiopure preparations significantly add to the manufacturing cost
How does enantiomerism or isomerism affect pharmacokinetics
◦ Dose reduction may be possible
◦ Absorption largely depends on physical properties and will usually be the same unless there is receptor mediated pinocytosis or receptor mediated absorption e.g. L Dopa vs D Dopa; or methotrexate D enantiomer is poorly absorbed as does not receive active transport. Additionally first pass metabolism may differ
◦ Distribution - identical unless receptor uptake or protein binding is affected
◦ Metabolism - may have different metabolism as a result of enzyme affinity for metabolism e.g. Warfarin, as R warfarin is metabolised via 3 different CYP3A4/CYP2C19/CYP1A2; whereas S enantiomers is metabolised by CYP2C9 only
◦ Excretion - excretion of isomers may differ under a number of different conditions
‣ Where metabolism to inactive metabolites occurs differently due to stereoselectivity of enzymes
‣ Differential reuptake in renal tubules
‣ Differential transport in biliary clearance or reuptake in bowel wall (recycling)
‣ Differential protein binding afffecting renal clearance
* Toxicity
What factors affect diffusion coefficient
molecular size, lipid solubility, temperature, viscocity, molecular properties of the solution
Absorption rate
Ficks law
Pharmaceutic factors affecting drug absorption
◦ Drug dose - higher concentrations will have faster rates of absorption due to Fick’s laws of diffusion
◦ Molecular size - reduced molecular size increases drug passive absorption via diffusion
◦ Lipid solubility vs ionic —> pKa
‣ Acidic drugs are unionised in the stomach allowing for rapid absorption
* Acidic drugs generally have higher oral bioavailability, poorer hepatic clearance, higher protein binding and smaller volume of distribution
* Basic drugs - poorer protein bringing, larger volumes of distribution, better CNS penetration, decreased receptor selectivity, sequestered in acidic organelles e.g. mitochondria and absorbed better in the stomach
‣ Salts or permanently ionised drugs remain ionised at all times and are not absorbed
◦ Site of administration
‣ PEG, oral vs rectal
◦ Preparation specific factors
‣ Modified release drugs are released more slowly
‣ High surface area drugs - dissolved more rapidly
Luminal factors affecting drug absorption
◦ Co-administered medications interacting intra-luminal
◦ Bile salts - emulsifying effect critical for absorption of some substances inc. fat soluble vitamins
◦ Gut bacteria - deactivate or activate drugs
◦ diet factors e.g. binding to substances in food or co-administration with a drug that it combines with e.g. tetracyclines and milk
Gut wall metabolism affecting absorption
◦ Enterohepatic recirculation - some drugs are eliminated in the bile and reabsorbed in the jejunum and this can be modified or altered
◦ Metabolism in the gut wall - preceding first pass metabolism absorption into plasma may be modified prior to transfer by gut enzymes, injured enterocytes may lose this function
Absorption vs motility
◦ Gastric motility - gastric emptying is a major determinant of absorption
‣ The rate of gastric emptying affects all drugs even those which are well absorbed in the stomach - drug absorption is generally poor in the stomach due to the thick layer of protective mucous but also small surface area and therefore drug absorption is generally slow even if drugs sit in the stomach for a prolonged period.
‣ Gastric motility is affected by
* Food - especially fat
* Viscosity of stomach contents
* Size of tablet/capsule
* Sedation
* Mobility
* Abdominal compartment pressure
* Gastric perfusion
* Autonomic and hormonal activity
* Baseline stomach emptying is minutes when empty, and many hours when full
‣ SMall intestine has a significantly larger surface area and contributed the most to drug absorption
◦ intestinal motility can increase or decrease absorption - slow transit may increase amount of an ingested substance absorbed
Absorption potential of the gut wall is affected by (4)
◦ Surface area - microvilli increase the surface area, if there is mucosal breakdown and lost microvilli or resection of bowel then surface area for absorption is reduced —> reduced absorption
◦ Thickness - Increased bowel wall thickness e.g. oedema will reduce absorption
◦ Active transport
‣ Some drugs may have active absorption via pinocytosis, active transport which result in increased drug absorption compared to passive processes only e.g. amino acid absorption or vitamins e.g. thiamine
‣ Genetic - Congenital malabsorption of specific substances
◦ GI blood supply - shock
‣ Reduced mucosal perfusion but increased mucosal permeability due to loss of barrier function with ischaemia. Delayed gastric emptying due to poor perfusion, intestinal wall oedema and altered gut microbiome may affect the processing of drugs
‣ Gut perfusion is the rate limiting step for rapidly absorbed drugs, as it slows it becomes more important for all drugs
Define protein binding
2 baseline characteristics
- Protein binding is a reversible, saturatable interaction between proteins and drug compounds in either the plasma or interstitial or intracellular fluid whose dissociation at baseline varies depending on the drug and protein involved occurring in an equilibrium. Only unbound drug is free to cross to sites of effect or sites of metabolism/excretion. Rate occurs rapidly
Examples of proteins that bind
number and characteristics of binding vary with pH
* Albumin - neutral or acidic drugs. 6 total binding sites .e.g barbituates/Benzos/ibuprofen (Sudlow site 2), warfarin (Sudlow site 1)
◦ Binding at one site does affect binding of other sites
* Globulins
◦ alpha 1 acid glycoprotein binds basic drugs e.g. morphine, lignocaine
◦ Alpha 2 globulin copper
‣ ceruloplasmin binds vitamins ADEK
◦ Beta 1 globulin iron
* Other
◦ Haemoglobin - phenytoin
◦ Lipoproteins - cyclosporine
Factors determining protein binding (4)
- Concentration of the drug itself
- Environmental modifiers
◦ PH
◦ Temperature
◦ Endogenous ligands for the same binding site - Binding affinity
◦ Affinity of drug for protein /association constant
◦ Lipid solubility of drug - increased lipid solubility = increased binding
◦ PKa - Ionised drugs do not bind to proteins - Available binding sites
◦ Number of available binding sites e.g. albumin has multiple additional binding sites, high capacity; whereas glycoproteins generally have low capacity and are more specific
◦ Competition for the same binding sites with other drugs
◦ Concentration of binding proteins - e.g. inflammation or chronic liver disease reducing albumin
What influence does protein binding have on pharmacodynamics
generally only of significance if protein binding >90% where small changes in bound fraction produce large changes in unbound drug concentration.
* Pharmacodynamics - only unbound fraction is available free for pharmacological effect
◦ When there are abrupt changes to bound fraction there will be changes in unbound drug concentration which has a subsequent effect on drug availability at receptor sites and action
What effect does protein binding have on Vd
highly plasma protein bound drugs have a smaller volume of distribution, whereas drugs with strong tissue binding have a very large volume of distribution e.g. amiodarone
Metabolism and how it is affected by protein bidning
◦ Generallly only free fraction is available for clearance and metabolism
‣ Some drugs have a higher affinity for their metabolic mechanisms than proteins and therefore this does not apply
‣ E.g. Furosemide which is bound to albumin is stripped off the albumin on the basal side of the PCT before furosemide is transported into the cell. IN albumin deficiency there is a decreased delivery of the drug to this site
◦ generally with changes in bound fraction and subsequent changes in unbound drug concentration this has an effect on drug metabolism proportionately and a new steady state is found. For drugs such as phenytoin with near saturated metabolic pathways plasma concentration increases if unbound fraction increases
Define first and zero order kinetics
- First order kinetics - the rate of drug elimination at any given time is directly proportional to its plasma concentration at that time
- Zero order kinetics - the rate of drug elimination is independent of the concentration of the drug
Draw a graph representing first order and zero order kinetics? Describe the mathematical appearance of the graph? State in words what the graph represents
e.g. of a drug subject to this
Provide an example of a drug relevant to intensive care practice that is a good demonstration of first and zero order kinetics
◦ Phenytoin metabolism becomes saturated within the upper limit of the normal range
◦ This results in a transformation from 1st order kinetics to zero order kinetics
◦ Therefore small increases in dose around this level can have large effects on plasma levels —> toxicity
‣ This is especially important in critical care where absorption of phenytoin given PO/NG/NJT can be variable and highly influenced by coadministered feeds and medications leading to fluctuations in bioavailability —> which when combined with its metabolism profile can rapidly lead to toxicity
‣ Additionally it is highly protein bound and plasma free component will increase in states of low albumin! Meaning when in the higher range of total drug concentration it has a risk of toxicity
◦ During zero order kinetics there is no steady state - if the drug delivery exceeds the rate of excretion plasma levels will continue to rise infinitely until ingestion stops or toxicity causes death
◦ Additionally to add to the complexity Amiodarone, erythromycin, fluconazole all inhibit the enzyme metabolising phenytoin (CYP2CP and CYP2C19) meaning first order kinetics will change to zero order kinetics at lower levels; while fluoroquinolones and folate supplements do the opposite
What is half life
- Hlaf life is the time taken for the amount of drug in the body (or plasma concentration) to fall by half
A constant proportion of drug is eliminated per unit of time - what does this represent
FIRST order kinetics
Exponential decline in drug concentraiton over time
What do you log in a concentration vs time curve?
Concentration
Concentration at any time in relation to a concentration time curve of linear kinetics is?
Concentration at any time is = Intiial concentration x e ^ -kt
Where k is the elimination rate constant = proportion eliminated per units of time (/hr) (= clearance / volume of distribution)
T = time
What relationship does half time have to pharmacokinetics
Rearranged: t 1/2 = 0.693 x volume of distribution / clearance
0.693 is the logarithm of 2 (exponential rate of elimination if there is first order kinetics)
Volume of distribution =
◦ V = total amount of drug in the body / plasma drug concentration
◦ V = plasma volume + fraction unbound in plasma / fraction unbound in tissue x tissue volume
What effect does Vd have on half life
◦ Half life is increased by increasing volume of distribution
◦ The larger the volume of dsibtuion the more drug is concentrated in tissues compared to blood, and therefore not exposed to metabolism or clearance mechanisms based in plasma
What are the 4 factors affecting Vd
‣ Properties of the drug - molecular size, charge, pKa, protein binding, tissue bidning,lipid water partition coefficient
‣ Properties of the patient - volume status, protein content, body fluid pH, competitive drug for binding sites
‣ Pathophysiology - age, gender, obesity,pregnancy, oedema
‣ Extracorpereal sites of distribution
Clearance is
the volume of blood cleared of drug per unit time (litre/hr)
Clearance in relation to half life is
Half life increased by reduced clearance
Clearance in relation to blood flow
◦ Clearance in each organ or each site is a reflection of blood flow to the organ and efficiency of irreversible drug extraction.
◦ Clearance can be anywhere between 0 and the total blood flow to the organ
Elimination rate has what relationship to clearance
◦ Elimination rate = clearance x plasma drug concentration
What factors affect clearance 6
‣ Concentration of the drug
‣ Susceptibility to bio transformation -clearance by metabolism eg. Hepatic
‣ Susceptibility to removal by filtration or diffusion e.g. lungs, renal
‣ Active secretion
‣ Functional status of clearance organs
‣ Blood delivery to organs of clearance
Why does half life even matter
- Duration of action after a single dose
- Time required to reach steady state with chronic dosing - 3-5 half lives
- Dosing frequency requirted to avoid large fluctuations in plasma concentration