ABCD - Pharmacology (theory) Flashcards
Explain the concept of pharmacokinetic modeling of single and multiple compartment models
Syllabus
A pharmacokinetic compartment is a mathematical concept which describes a space in the body which a drug appears to occupy. It does not necessarily correspond to any specific anatomical space or physiological volume.
The single compartment model:
- The drug administered instantly and completed disperses to every corner of the compartment and is homogenously distributed throughout the volume (of distribution - Vd)
- It is eliminated from this volume at a constant concentration-dependent rate
- The rate of elimination is given by the rate constant for elimination, k (eg. If k = 0.5, then 50% of the drug is eliminated per unit time)
- The clearance (Cl) of the drug from this single compartment is described by the equation (k x Vd)
- Highly hydrophilic drugs which are confined to body water have single compartment pharmacokinetics (eg. Aminoglycosides)
The two-compartment model:
- The drug administered disperses homogenously into the central compartment
- The drug diffuses in and out of the central and peripheral compartments until an equilibrium is reached (assuming that there is no elimination)
- If the drug is being cleared from the central compartment at a concentration-dependent rate, the concentration/time graph will have two distinct phases of change in drug concentration:
- Distribution phase - initial rapid decline in serum drug concentration
- Elimination phase - slow decline in drug concentration, sustained by redistribution of drug from tissue stores
- If the drug is being cleared from the central compartment at a concentration-dependent rate, the concentration/time graph will have two distinct phases of change in drug concentration:
The three-compartment model:
- Three compartments (blood, lean tissues, fat)
- For a highly fat-soluble drug, a rapidly given bolus distributes rapidly into all tissues, however lean muscle contains little fat and is therefore a poor storage reservoir for the drug & drug is eliminated from this compartment at the same rate as blood
- The phases in the concentration/time graph are as follows:
- Distribution - initial rapid decline in serum concentration. This phase ends with the concentrations reaching their peaks in the peripheral compartments
- Elimination - period during which the main effect on drug concentration is elimination. This phase ends when the slow compartment concentration becomes higher than the blood concentration and the total clearance is slowed down by the gradual redistribution of the drug into the blood compartment
- Terminal phase - elimination is slow as the concentration of drug in the central compartment is minimal. The major defining factor affecting drug concentration during this phase is the redistribution of stored drug out of the slow compartment.
- Mathematically, this model will produce a polyexponential graph, which can be described by equation attached
Representing the three-compartment model in a diagram:
- Ve = effect site
- Kinetics of distribution are usually described as Kxy, where x is the compartment from which the drug is distributing and y is the compartment to which the drug is going (ie - bolus from V1 to V2 is K12)
Describe absorption and factors that will influence it
Syllabus
Absorption is the movement of a drug from its site of administration to the central compartment. Absorption may be oral, through the mucosa, transdermal, inhalational, intramuscular or subcutaneous.
- (Note: the characteristic feature of absorption is that the substance is taken up by a volume, in contrast to adsorption where the substance is deposited on a surface - eg. Eating a cake vs smooshing on face)
Bioavailability is the fraction of the dose which reaches the systemic circulation intact (and escapes first past elimination)
- ‘Absolute’ bioavailability compares one non-IV route with IV administration
- ‘Relative’ bioavailability compares one non-IV route or formulation with another
- It is measured using the area under the concentration-time curve (Dost’s Law) - “The ratio of the area beneath the blood level vs time curves after oral administration to that following intravenous administration of the same dose is a measure of the absorption of the drug administered”
- Bioavailability = –> (absolute bioavailability)
- Bioequivalence (clinical definition) - drugs are considered bioequivalent if the extents and rates of absorption of drug from them are so similar that there is likely no clinically important difference between their effects
FACTORS INFLUENCING ABSORPTION
Drug factors
- Concentration - large drug dose will be absorbed more rapidly because of the high concentration gradient
- Size
- Molecule size - small molecule diffuse more easily
- Particle size - smaller the particles, the greater the surface area from which the drug will be eluted
- pKa - weak acids and lipophilic drugs are better absorbed
- Tablet disintegration (for PO route) - will determine rate of dispersion
Site properties (route of administration)
- Oral (gastrointestinal)
- Destructive effects of gastric acid
- Gastric emptying & intestinal transit
- Gastric emptying is the major determinant
- Slow intestinal motility can ensure complete absorption (while rate of absorption may decrease as delivery of drug to absorptive surfaces is reduced)
- Splanchnic perfusion
- Surface area
- Small intestinal SA can be decreased in gastroenteritis (denuded villi) or surgical short gut
- Characteristics of the gut content (eg. Food - tetracyclines chelate intestinal calcium from milk & forms insoluble complex)
- Emulsifying effect of bile is critical to the absorption of fat-soluble vitamins and minerals
- Metabolism by gut organisms (they may either deactivate or activate drugs)
- Mucosal
- Irritant effects
- Transdermal
- Integrity of the dermis
- Characteristics of the subcutaenous tissue
- Inhalational
- Volatility of drug
- Size of droplets/particles
- Injection (eg. IM)
- Blood flow to site
- Solubility of the drug in the interstitial fluid
Disease states - Shock
- Globally, cardiogenic/obstructive/haemorrhagic/septic shock will cause decreased perfusion to various administration sites and therefore decreased absorption
- Anaphylactic shock will cause increased perfusion + therefore increased absorption (although there may be decreased penetration of drug to absorption site for the inhalation route due to bronchospasm
- Effect on bioavailability - largely, bioavailability increases
- First pass metabolism
- Hepatic blood flow - decreases, which will decrease metabolism
- Hepatic enzyme activity - may be downregulated (eg CYP enzymes in septic shock) or abolished completely (ischaemic hepatitis)
- Shunts - portosystemic shunts may open, which allows drugs to bypass first pass metabolism
- Already absorbed drugs
- Decreased protein binding - drug binding proteins will have their production downregulated
- Decrease plasma metabolism - plasma esterase and protease synthesis will be decreased leading to diminished clearance
- First pass metabolism
Describe factors influencing the distribution of drugs
Syllabus
Factors affecting Vd:
- Drug factors
- Molecular size (larger molecules = harder to passively diffuse out of central compartment)
- Molecular charge (High ionised = more likely to be trapped in central compartment)
- pKa (degree of ionisation & therefore lipid solubility)
- Lipid solubility
- Water solubility
- Patient factors
- pH - influences degree of ionisation according to pKa & influences degree of protein binding
- Body water volume - dehydration will result in concentrated drug levels
- Protein levels - lower protein = higher unbound drug fraction (may make Vd appear smaller if measuring free drug levels)
- Displacement - drugs may be displaced by effects of pH or competition from other drugs/substances
- Age - Body water content decreases & muscle mass decreases - shrinks Vd of water-soluble drugs and decreases tissue binding
- Gender - Female Vd < male (due to total body water)
- Oedema/ascites/effusions - increases Vd
- Measurement factors
- Calculation factors
- PK model - Vd can be different if using Vinitial, Vextrap, Varea or Vss
- Free vs total drug - for highly protein bound drugs, total Vd will correspond to Vd of the binding protein rather than the drug itself
- Apparatus factors
- ECMO + dialysis tend to adsorb drugs in unpredictable fashion
- Volume expansion in extracorporeal circuits
- Calculation factors
Define volume of distribution and explain how it is calculated
Own question
Definition:
- An imaginary volume which relates plasma drug concentration to the concentration of drug at site of effect
- The apparent volume into which a drug disperses in order to produce the observed plasma concentration
- V(d) =
- Also, V(d) =
- Volume of distribution calculated by administering dose, measuring plasma concentrations and plotting on logarithmic conc. vs time graph and extrapolating plasma conc. at time 0. V(d) can then be calculated by above equation
- Different V(d) values:
- Vinitial - calculated by extrapolating line from plasma concentration measurements to time 0
- Volume of initial dispersion of the drug (represents behaviour of the drug during the first rapid phase of distribution through the central compartment)
- Generally determined by the degree of protein binding. Drugs with high protein binding will have larger Vinitial if measuring free drug levels
- Use - if you know that the drug will have no protein binding, this can be used to calculate total blood volume (eg 53Cr labeled red cells)
- Vextrap - calculated by extrapolating line from terminal elimination phase to time 0
- Represents behaviour of the drug in tissues
- Not a lot of clinical utility. Will likely give large overestimate of Vd (low plasma conc at time =0)
- Varea - non-compartmental calculation of Vd.
- Calculated by Varea = , where β = terminal elimination time constant, X = drug dose and AUC = area under concentration vs time curve
- Pitfalls - will often underestimate the conc at time zero due to using the terminal elimination constant & therefore overestimate Vd
- Vss - describes Vd when there is a stable drug concentration
- Vss =
- Vinitial - calculated by extrapolating line from plasma concentration measurements to time 0
- As drug distributes around the body, (especially with IV administration), higher blood conc.s will reach highly perfused tissues earlier (eg. Brain) and slower perfused tissues later (eg. Skeletal +muscle tissue)
- Eg. Diazepam + digoxin - have similar elimination rates, so plasma conc will be similar at various time points but IV diazepam effect is quicker as effect site is in highly perfused tissue (brain) vs digoxin (cardiac tissue)
- Therefore if you want quick effect, can calculate a loading dose based on V(d) to achieve desired plasma conc by rearranging above equation:
- dose = V(d) x desired plasma conc
Outline the role of the liver in metabolism
Past question
- Liver metabolises drugs by uptake of drugs into hepatocytes from blood.
- Metabolism is dependant on 3 factors: blood flow (QH), strength of binding of drugs to proteins & the efficiency/rate at which hepatic enzymes can break down the drug when that drug is a substrate (Intrinsic clearance - CLint)
-
- Where CLH = Hepatic clearance; QH = Hepatic blood flow & EH = Hepatic extraction ratio
- But
- CLint can be further defined by:
- the maximal rate at which an enzyme can convert a drug to a metabolite
- Km - dissociation constant - ie, the lower the constant, the strong the binding of the drug to the enzyme which metabolises it)
- Combining the above 2 equations gives:
- This can be simplified by considering drugs that have a high hepatic extraction ratio (where CLint is high - ie. the efficiency of the hepatic enzymes to metabolise that drug are high).
- In this case, if CLint >>> QH, then approaches and
- Ie. For drugs that have high liver metabolism, hepatic clearance is determined by hepatic blood flow
- Examples of these drugs include: GTN, verapamil, propranolol, lignocaine, morphine, ketamine, metoprolol, propofol
- In the case of drugs that have a low hepatic extraction ratio,
- (If QH >>> CLint) then approaches QH and
- Ie. For drugs that have low liver metabolism, hepatic clearance is determined by drug-protein binding affinity and the rate at which hepatic enzymes can metabolise the drug (intrinsic clearance)
- Examples of these drugs include: diazepam, lorazepam, warfarin, phenytoin, carbamazepine, theophylline, methadone, rocuronium
- Disease states which alter QH
- Sepsis - early sepsis (hyperdynamic) will increase QH, whereas late sepsis will decrease it
- Haemorrhage and cardiogenic shock will decrease it
- Ionotropes - vasoconstrictors like norad and adrenaline will decrease hepatic blood flow, while vasodilators like clonidine will increase it
First pass clearance:
- The extent to which a drug is removed by the liver during its first passage in the portal blood through the liver to the systemic circulation
- This is a combination of :
- Metabolism by gut bacteria
- Metabolism by intestinal brush border enzymes
- Metabolism in the portal blood
- Metabolism by liver enzymes
- Drugs with high first pass clearance will:
- Have a greater difference between oral and IV doses
- Have more significant changes in oral bioavailability with changes in hepatic enzyme kinetics
How changes in liver function will influence pharmacokinetics:
- Decreased synthetic function - decrease plasma protein synthesis will influence Vd/free drug levels.
- Liver also synthesis plasma esterases and peptidases
- Changes to secretory function - drugs and metabolite which rely on biliary excretion will be retained
- High bilirubin may result in displacement of drugs from albumin as it competes for binding sites
- Portal hypertension - leads to shunting of portal venous blood into systemic circulation (decreases first pass metabolism)
BIOTRANSFORMATION:
The liver plays an important role in the metabolism of most drugs. Most compounds are lipophilic or partially ionised and the liver converts them to hydrophilic substances that may be eliminated by the kidneys or in the bile
Biotransformation
- Phase I- increase hydrophilicity, products may be pharmacologically active
- Oxidation (CYP450, smooth ER)
- Reduction (reductase enzymes, cytoplasm)
- Hydrolysis (hydroxylase enzymes, cytoplasm)
- Phase II- conjugation, increase polarity, occurs generally in the cytoplasm, usually inactive
- Glucuronidation (glucuronosyl transferases, most common conjugation reaction)
- Sulphation
- Acetylation
Basic chemistry….
- Redox reactions
- Oxidation: loss of electrons (or gain of oxygen) during a reaction by a molecule, atom or ion
- Reduction: gain of electrons (or loss of oxygen)
- Hydrolysis: a molecule of water breaks one or more chemical bonds
- Conjugation: coupling the drug or its metabolites to another molecule
- Glucuronidation: glucuronic acid, derived from cofactor UDP-glucuronic acid, is covalently linked to a substrate
- Sulphation: addition of a sulfur (usually SO3) group
- Acetylation: acetyl group added to an organic chemical compound, usually in place of a hydrogen atom
*Sites on drugs where conjugation reactions occur include carboxy (-COOH), hydroxy (-OH), amino (NH2) and thiol (-SH) groups
Define Clearance and its calculation
Own question
- Definition: the volume of blood cleared of drug per unit time
- Refers to the efficiency of irreversible elimination of a drug from the systemic circulation
- Clearance can be calculated wrt total body or a particular organ (eg. Kidney or liver)
- Systemic clearance (Cls) = sum of all clearances
- Renal clearance (Clr) = fraction of total clearance which is handled by the kidneys
- Metabolic clearance (Clm) - fraction of total clearance which is the consequence of drug biotransformation, by whatever mechanisms
- Hepatic clearance (ClH) - fraction of total clearance which is handled by the liver (biliary excretion or metabolism)
- Intrinsic clearance (Clint) - the hepatic clearance a drug would have if it was not restricted by hepatic blood flow rate
- Intrinsic unbound clearance (Cl’int) - the intrinsic clearance a drug would have in the absence of plasma protein binding
- Measured in L/hr or mL/min
- Cl (L/hr) =
- In the case of a single dose, it can be measured by:
- Cl (L/hr) =
- Where AUC = area under curve of concentration vs time graph post single dose of drug
- This equation can be rearranged to determine maintenance dose rate:
- Note: elimination rate can be calculated from the same formula (elimination = CL X plasma drug conc)
- Note: minimum clearance = 0, maximum clearance = blood flow rate to particular organ
- Elimination rate is distinct from clearance and refers to amount of drug removed from body (measured in mg/hour)
- Refers to both removal of unchanged drug in urine/sweat/faeces etc & metabolised drug
- Does not refer to elimination of metabolites or to drug which binds to tissue and rejoins plasma
Describe renal clearance
Renal clearance:
- The magnitude of renal drug clearance is the sum of glomerular filtration and active excretion minus renal drug reabsorption
- Filtration
- Factors affecting filtration:
- Protein binding - glomerulus only filters unbound drug
- Charge - negatively charged GBM theoretically repels negatively charged drugs
- Size - molecules <30Angstroms are freely filtered at the glomerulus
- Non-drug properties - renal disease, renal blood flow
- Factors affecting filtration:
- Secretion
- Active transporters
- Weak acid transporters (substrates eg: β-lactam antibiotics, frusemide, HCT, probenecid)
- Weak base transporters (substrates: procainamide, ranitidine, trimethoprim, ethambutol)
- Nucleoside transporters (substrates: ribavarin, gemcitabine)
- P-glycoprotein transporters (substrates: digoxin, verapamil, cyclosporin)
- These mechanisms are saturable - when there are multiple suitable substrates, they will compete with each other
- The rate of clearance by secretion depends on renal blood flow
- Molecules that are too large to be filtered may still be secreted. Protein bound drugs will not be cleared this way
- Active transporters
- Reabsorption
- Passive: if a drug becomes so significantly concentrated in the tubule, it sets up a gradient which favours the diffusion of drug back into tubular cells and back out into the blood
- Therefore factors influencing reabsorption are: drug concentration, urine flow rate, urine pH, drug ionisation
- Active: mainly happens in PCT.
- Kidneys will attempt to reclaim organic acids, water soluble vitamins, ions and various metabolic substrates like lactate and glucose. Drugs with any molecular resemblance to these will be resorbed in a similar manner
- Passive: if a drug becomes so significantly concentrated in the tubule, it sets up a gradient which favours the diffusion of drug back into tubular cells and back out into the blood
- Metabolism of drugs by the kidneys
- PCT contains enzymes which are capable of digestion (breaking down filtered peptides into proteins + amino acids)
- Adjustment in renal impairment (degree of dysfunction, dose adjustment & monitoring)
- Degree of dysfunction - measure creatinine clearance (renal clearance of drugs is proportional to creatinine clearance, not matter the mechanism of elimination
- Dose adjustment = non-renal clearance + (fraction of renal clearance x fraction of residual renal function). The loading dose does not need to be adjusted & maintenance dosing can be adjusted by reducing the regular dose, changing the dosing interval, or both
- Monitoring of drugs levels - essential for drugs with narrow therapeutic index
Explain the difference and the clinical relevance, between zero and first order kinetics. (60% marks) Give an example that is relevant to intensive care practice. (40% marks)
Past question
First order elimination kinetics:
- Definition: A constant fraction of drug in the body is eliminated per unit of time
- All enzymes and clearance mechanisms are working at well below their maximum capacity, and the rate of elimination is directly proportional to drug concentration. First order kinetics has a linear upslope on a concentration (x) vs elimination (y) graph.
- Eg. gentamicin
Zero order elimination kinetics:
- Definition: A constant amount of drug in the body is eliminated per unit of time
- Increasing the concentration will have no effect on elimination. This will look like a horizontal line on a conc vs elimination graph
- Eg. Alcohol (Km for alcohol dehydrogenase is very low)
Michaelis-Menten elimination kinetics:
- (Non-linear) kinetics
- At low concentrations, the kinetics are first order. Upon saturating the enzymes, higher concentrations will result in zero-order kinetics
- The maximum rate of reaction = Vmax; The concentration required to achieve 50% of the maximum rate is called Km.
- V = Where V = velocity (rate) of reaction & S = substrate (drug) concentration
- This has relevance to clinical pharmacology. Drugs which have a therapeutic concentration range in the steep part of this curve are said to have a narrow therapeutic range (i.e. the effective dose is not too far off the toxic dose). If relatively large changes in dose produce relatively small changes in the concentration , toxic levels will be difficult to achieve and the drug is said to have a broad therapeutic index.
- As a rough rule, if the drug concentration required to produce a useful effect is above Km, then the drug will have a narrow therapeutic index
- Eg. phenytoin
- Drugs with zero-order elimination have a variable half-life, whereas drugs with first-order elimination will have a fixed predictable half life.
- Zero-order elimination may be very slow in a large overdose
- Drugs with non-linear elimination kinetics and a narrow therapeutic index require frequent plasma concentration monitoring
- First-order elimination kinetics may be seen in normal therapeutic range (i.e the therapeutic range is well below Km ) but in overdose the drug may be cleared with zero-order kinetics.
Define half-life, how it is calculated and what it is used for
Own question
HALF-LIFE
- The time taken for half of the drug to be eliminated by the body
- It is an exponential curve
- =
- Ct = concentration of drug at time t; C0 = conc at time 0; k = elimination rate constant
- At the first half life, Ct = 0.5 x C0 & solving for k gives:
- k =
- Half life is also a function of clearance (CL) and volume of distribution (V)
- Substituting this into above equation gives:
- k =
- So, as volume of distribution increases, half life increases & as clearance increases, half life decreases
- In disease states, CL and V may move in the same direction, so half-life is not useful for determining duration of action of drug
- What half life is useful for:
- Calculating elimination after a single dose of a drug
- Calculating time to steady state with continuous dosing
- It usually takes ~3-5 half lives to achieve approximate steady state
- Determining frequency of dosing
- To maintain plasma concentration within therapeutic range. For drugs with short half lives, sustained release preparations may be more useful as very frequent dosing can be impractical. In this setting, plasma concentration is determined by absorption more than elimination
Explain the concepts of intravenous bolus and infusion kinetics. To describe the concepts of effect-site and context sensitive half time
Syllabus
Intravenous bolus + infusion:
- In regular doses, drug concentration achieves a steady state in steps, but plasma concentration reaches a point at which dose rate and clearance rates are equal after about 5 half-lives
- In first order kinetics, doubling the administered dose will lead to an increase in duration of action by one half-life
- Loading dose:
- Rapidly achieves the peak concentration necessary to compete with clearance so that the desired effect is achieved and maintained sooner
- Loading dose is calculated by multiplying the desired peak concentration by the volume of distribution of the drug.
- If a drug has a high volume of distribution, the loading dose to achieve steady-state concentration may be impractically large
- Dosing interval - generally, for most drugs, the dosing interval is approximately one half-life
Effect site half-time:
- A substance with extensive tissue distribution, will reach peak concentration in different tissues at different times depending on rate of distribution to the tissue. This may result in different clinical effects at different times.
- The phase of distribution into the ‘clinically interesting’ compartment is known as effect site distribution, or distribution into the biophase (biologically active phase of distribution)
- The effect site is a virtual compartment which is in some way linked to the central compartment
- As the movement of drugs between compartments is concentration driven, the rate of drug movement can be described as a first-order constant - ke0
- ke0 is calculated based on plasma concentrations and clinical effect. It reflects those drug properties that affect diffusability
- Equilibrium between the central compartment and effect-site compartment follows first-order kinetics, described by constant ke0. Using this constant, it is possible to generate a value for ke0 half-time or t1/2ke0
- t1/2ke0 = 0.693/ke0
- This value represents the time required for the effect-site compartment to reach 50% of the plasma concentration as an infusion of the drug is running to maintain a constant plasma concentration
- If a drug redistributes out of plasma to effect site quickly (ie. Lower t1/2ke0), this decreases time to peak effect and increases magnitude of peak effect for a given dose
Context sensitive half times:
- The time required for a 50% decrease in the central compartment drug concentration after drug administration has ceased, where the ‘context’ is the duration of an infusion that has maintained a steady state concentration in the central compartment
- It isnt a number, but a relationship of how long it will take for concentration to decrease by 50% as a function of the duration of infusion
- Propofol approaches a maximum CSHT of ~20minutes but fentanyl can reach an excess of ~300minutes
- Determinants of context sensitive half time:
- Volume of the central compartment
- The rate of systemic clearance of the drug from the body
Explain clinical drug monitoring with regard to peak and trough concentrations, minimum therapeutic concentration and toxicity
Syllabus
Characteristics of drugs which make therapeutic drug monitoring useful:
- Marked pharmacokinetic variability
- Therapeutic and adverse effects related to drug concentration
- Narrow therapeutic index
- Defined therapeutic concentration range
- Desired therapeutic effect difficult to monitor (eg. Lithium) or drug is being used prophylactically (to maintain the absence of a condition - eg. Seizures, manic episodes etc)
Timing of sample for monitoring:
- At the beginning of treatment, after steady state has been achieved
- After a dose adjustment has been made
- When factors affecting clearance have changed: eg. Pt develops renal failure, suspected drug interaction
- When treatment seems to be ineffective
Factors relevant to interpretation of drug levels:
- Pharmacokinetic factors:
- Protein binding - total plasma drug level may not be reflective of drug activity (eg. Phenytoin in hypoalbuminaemia - drug is highly protein bound, so lower plasma levels may produce clinical effect)
- Relationship of plasma concentration to effect site concentration - if drug penetrates variably and incompletely, it is difficult to relate serum levels to effect site levels (eg. Vancomycin in ventriculitis (CSF penetration low despite high plasma conc))
- Factors which affect plasma concentration - Vd, tissue binding, sites of metabolism
- Timing of sample in relation to dose - most drug levels are trough levels as it is the least variable point in the dosing regimen (drug concentration change the least over time)
- Pharmacodynamic factors:
- Relationship of plasma concentration to clinical drug effect - in some cases there is no such relationship (eg. Levetiracetam)
- Active metabolites makes this relationship more difficult
- Individual variability in drug response - therapeutic effect in some individuals may be different than the reported therapeutic window
- Relationship of plasma concentration to clinical drug effect - in some cases there is no such relationship (eg. Levetiracetam)
- Clinical/practical factors
- Simplicity of assay
- Convenience of sample collection
- Cost and benefit of the assay
Other important points to remember wrt therapeutic drug monitoring:
- Therapeutic ranges are classified by monitoring effect and drug concentration in small groups of people - so the minimum therapeutic concentration does not ‘switch on’ the therapeutic effect - this is often a continuous spectrum
Describe the pharmacokinetics of drugs in the epidural and subarachnoid space
Syllabus
Pharmacokinetics of epidural drug administration
- Administration - about 40mL of fluid can be injected into the epidural space
- Absorption
- Can occur via 4 different routes:
- Exit the intervertebral foramina to reach the paraspinous muscle space
- Distribute into epidural fat
- Diffuse into ligaments
- Diffuse across the spinal meninges and into the CSF
- Diffusion is the main influence on drug absorption - this will be dependent on properties in Fick’s law - eg. Concentration, surface area (largely determined by volume of infused drug), lipid solubility, protein binding
- CSF flow and volume is important as it will affect the concentration gradient
- Can occur via 4 different routes:
- Distribution - systemic distribution follows a 2 compartment model (note: lung water tends to accumulate local anaesthetics due to its low pH)
Pharmacokinetics of subarachnoid drug administration
- Absorption
- High CSF concentration is achieved rapidly, hence absorption into spinal tissue is very rapid
- With fewer barriers to diffusion (no meninges), required dose is much smaller & onset is more rapid
- Bypassing the BBB is much easier
- Systemic absorption is slower - there is no rapid distribution phase (perfusion of subarachnoid space is less extensive & systemic distribution is slower)
- Increased half life of particularly hydrophilic drugs
- Distribution
- Baricity refers to the comparative density of spinal anaesthetic solution in comparison to CSF (density of solution divided by density of CSF, which is ~1.003mg/mL)
- Isobaric solutions stay roughly at the level they are injected, while hyperbaric solutions sink
Metabolism (relevant to both)
- The meninges do not perform any major metabolic function & most of the dose enters the systemic circulation unchanged
- Metabolism is therefore dependent on the same pathways as other sites of administration
Define and explain dose-effect relationships of drugs, including dose-response curves with reference to graded and quantal response
Syllabus
Graded and quantal response:
- Graded dose-response relationships demonstrate the response to a drug against its concentration
- 4 main parameters describe a graded dose-response curve
- Potency (EC50 - relates to a drugs’ affinity for its receptor)
- Slope - shallow slope suggests greater chance of overlap between desired effects and side-effects. A slope which is too steep suggests that it will be difficult to achieve precise control of the effects
- Maximum (Emax - max effect achievable by that drug)
- Threshold dose - lowest dose at which a drug effect is seen. May be at the zero point but doesn’t have to be (There may be some baseline pre-drug effect eg. NA and arterial tone)
- 4 main parameters describe a graded dose-response curve
- Quantal dose-response relationships demonstrate the rate of an outcome occurring in a population against the drug dose. A quantal dose response is a defined drug effect which is either present or absent
- “ordered” dose-response relationships describe a series of quantal relationships which are displayed on the same graph - where the increase in the dose of a drug produces a progression of effects which can each be set as the end-point of a quantal relationship (eg. Stages of anaesthesia)
- This graph describes the cumulative percentage of a population which can be expected to achieve a response at a given dose
Define and explain dose-effect relationships of drugs, including dose-response curves with reference to therapeutic index
Syllabus
Therapeutic index:
- The median therapeutic dose (ED50) in quantal dose-response curves is the dose at which 50% of individuals exhibit the specified quantal effect.
- It is entirely dependent on the definition of the quantal response
- It does not necessarily represent a clinically relevant dose
- The median toxic dose (TD50) - the dose at which some clinically relevant toxic effect is reported in 50% of the tested population. This also depends on definition of toxic. Problems:
- Toxicity may develop after years of post-marketing data (eg. Carcinogenesis)
- Toxicity may be unrelated to dose
- The median lethal dose (LD50) - the dose required to produce a lethal effect in 50% of the population. Same as TD50, however the toxic effect is death. Note: the pharmacokinetics change the LD50 (eg phenytoin administered rapidly may be fatal, whereas the same dose administered slowly may have no toxic effects
- The therapeutic index is the ratio of the TD50 to the ED50 for some therapeutically relevant effect. It is a numeric measure of the selectivity of the drug for its desired effect - the larger the value, the safer the drug. However, it can misrepresent risk (slope of dose-response curves may be different)
- The therapeutic window (or range) is the dose range over which drug is effective and relatively non-toxic in most of the population
Define and explain dose-effect relationships of drugs, including dose-response curves with reference to potency and efficacy, competitive and non-competitive antagonists & partial agonists, mixed agonist-antagonists and inverse agonists
Syllabus
Potency and efficacy:
- Potency is defined as the concentration (EC50) or dose (ED50) of a drug required to produce 50% of that drug’s maximal effect
- Two drugs may have the same efficacy but one may achieve the effect at a lower dose
- Efficacy - the degree to which different agonists produce varying responses, even when occupying the same proportion of receptors
- Efficacy can be expressed numerically - as a ratio of the drug’s maximal efficacy to the maximal efficacy of some known potent agonist (aka intrinsic activity or maximal agonist effect)
Competitive and non-competitive antagonists:
- Competitive antagonists - binding of antagonists precludes binding of the agonist (ie binding of the agonist and antagonist is mutually exclusive)
- In the presence of a competitive antagonist, the Emax of the agonist is unaffected, but the potency is reduced
- Non-competitive antagonists - the antagonist opposes the action of the agonist but does so without competing with it for the binding site
- An insurmountable antagonist reduces the Emax of the agnoist
Partial agonists, mixed agonist-antagonists and inverse agonists
- Agonists - a ligand that binds to a receptor and alters the receptor state resulting in a biological response
- An allosteric modulator is a ligand that increases or decreases the action of a primary or orthosteric agonist or antagonist by combining with a distinct site on the receptor macromolecule
- Full agonist - a drug which is capable of producing a maximum response that the target system is capable of
- Spare receptors - refers to unbound receptors when a full agonist has produced the maximal effect on binding a fraction of the total receptor population. If these receptors were inactivated by an antagonist, the initial drug would still be able to produce the maximal effect, but at reduced potency
- Partial agonists - will never achieve the maximal response that the system is capable of because its efficacy is lower than that of a full agonist
- Inverse agonists - a ligand which, by binding to a receptor, produces the opposite of the effect which would be produced when an agonist binds to the same receptor. When given in the absence of any other drug, it will have the opposite effect to the normal expected effect of receptor activation
- Inverse agonists are described as having negative efficacy. This is distinct to ‘neutral’ antagonists, which bind to the receptor and inactivate them (ie produce 0% response)