pharmacology studentnotes

Upload: pasalau-daria-vasilica

Post on 10-Oct-2015

33 views

Category:

Documents


0 download

DESCRIPTION

farmaco

TRANSCRIPT

  • PHARMACOLOGY---- Student Notes

    PHARMACOLOGY LECTURE 1

    INTRODUCTION a xenobiotic is a drug that is not synthesied within the body pharmacodynamics is the effect of the drug on the body (effect of drug at

    receptor site) pharmacokinetics is the effect of the body on the drug (absorption, distribution

    and elimation of drug) toxicology deals with the side effects of drugs the therapeutic index ratio is the minimum dose that produces toxicity divided

    by the minimum dose that produces the therapeutic response (therefore you want a high therapeutic index)

    PHARMACOLOGY LECTURE 2

    RECEPTORS 1 - TARGETS FOR DRUG ACTION affinity constant = K(D) - a low KD = a high affinity

    o a K(D) of 10E-6 or more is considered too weak drugs act at the following sites:

    o cell surface receptors o ion channels o carrier pumps

    e.g. some diurectics act on the Na+/Cl- cotransporter in the distal tubules

    e.g. some diurectics act on the Na+/K+ antiporter in the collecting ducts

    e.g. some antiulcer/reflux drugs work on the H+/K+ antiporter o enzymes

    e.g. NSAIDs inhibit cyclo-oxygenase which blocks prostaglandin production

    e.g. ACE inhibitors inhibit angiotensin converting enzyme to decrease blood pressure

    e.g. HMGCoA reductase inhibitors inhibit this to reduce lipid concentration

    o nuclear receptors/RNA/DNA o intracellular structural proteins

  • Question: Aspirin and ibuprofen are NSAIDs that inhibit cyclo-oxygenase in platelets (ibuprofen - competitive; aspirin - irreversible). What are the clinical consequences of this difference in inhibition?

    o Aspirin will be longer acting. Hence, people undergoing surgery must stop taking aspirin 1 week before so clotting can occur during. whereas people must only stop taking ibuprofen 1 day before surgery

    inhibition constant = K(I) - measures degree of inhibition of an enzyme therapeutics: treatment of disease can be by different targets:

    o e.g. to treat gastric hyperacidity: neutralising drug (NaHCO3) proton pump inhibitor (omeprazole) histamine 2 receptor blocker (ranitidine)

    o some drugs act on more than one target, e.g. caffeine levels of mecahnisms of action of drugs

    o e.g. propranolol: -1. molecular: is a competitive inhibitor by binding to beta 1

    receptors -1. cellular: prevents increase in cAMP, protein phosphorylation -1. physiological: reduces cardiac heart rate and contractile force -1. therapeutic: used to treat angina

    PHARMACOLOGY LECTURE 3

    RECEPTORS 2 - INTRACELLULAR MECHANISMS (PHARMACODYNAMICS)

    there are 4 basic mechanisms for transmembrane signalling 0. ligand gated channels 1. G protein-coupled receptors/second messengers 2. intracellular receptors 3. ligand-regulated transmembrane enzymes

    1. ligand gated channels: o example: nicotinic acetylcholine receptor

    where: found on skeletal muscle function: when activated causes contraction structure: pentamer crossing lipid bilayer mechanism of action: acetycholine binds to alpha subunit -->

    conformational change --> trransient opening of central aqueous channel --> Na+ flows down concentration gradient --> depolarisation --> contraction

    2. G protein-coupled receptors/second messengers o signalling system involves:

    0. extracellular drug binds to cell surface receptor

  • 1. receptor triggers activation of a G protein on cytoplasmic face of plasma membrane

    2. activated G protein alters activity of an effector element (e.g. enzyme or ion channel)

    3. effector element changes concentration of intracellular second messenger

    o G protein structure: contains alpha, beta and gamma subunits the beta-gamma anchors the G protein to the membrane the G protein is not normally bound to the receptor but is free

    floating in the cell membrane (cytoplasmic side o how G proteins work: see Alberts Molecular Biology of the Cell, Figure

    15-23 0. when the receptor is not occupied by a drug: the G protein is in a

    resting state, where beta-gamma anchors G protein to membrane, and GDP occupies site on alpha subunit

    1. the ligand binds to the receptor, which alters the conformation of the receptor, exposing the binding site for G protein

    2. the G protein binds to the receptor, greatly weakening the affinity of the G protein for GDP

    3. GDP dissociates, allowing GTP to bind to the alpha subunit 4. this causes the alpha subunit to change conformation, it now

    dissociates from the receptor and the beta and gamma subunits 5. the free alpha subunit now changes conformation so that it can

    bind with its target enzyme 6. this target enzyme acts, e.g. converts ATP-->cAMP, ion channel

    etc. 7. hydrolysis of the GTP by the alpha subunit returns the subunit to

    its original conformation, causing it to dissociate from the target enzyme and reassociate with the beta and gamma subunits

    o Gs is a G protein that acts on adenyl cyclase, hence converts ATP-->cAMP

    o Gi is a G protein that inhibits adenyl cyclase, hence prevents ATP-->cAMP

    o note that the slower the hydrolysis of the GTP (step 8), the longer for which the G protein will act

    o SECOND MESSENGERS - commonly used second messenger systems in the body include:

    0. cAMP many drugs/hormones act by increasing or decreasing the

    adenyl cyclase which in turn increases or decreases cAMP 1. calcium-phosphoinositide pathway (IP3/DAG)

    mechanims of this pathway: 1. Gs stimulates phospholipase C (a membrane

    enzyme) -->

  • 2. hydrolysis of IP2 into DAG (diacyclglycerol) and IP3 (inositol-1,4,5-triphosphate)

    DAG activates protein kinase C --> phosphorylates intracellular proteins --> altered cellular function (e.g. contraction of smooth muscle)

    IP3 triggers release of Ca++ from storage vesicles (in smooth muscle this will increase contraction)

    3. these signals are terminated by the following: 0. dephosphorylation of IP3 1. phosphorylating DAG to arachidonic acid 2. actively removing the Ca++

    o How does ACh affect the ion channels in the heart? the vagus nerve releases ACh --> binds to receptor in cardiac

    muscle --> enhances K+ permeability --> hyperpolarisation --> decreases activity

    3. intracellular receptors - regulate gene expression o ligands for intracellular receptors include: steroids, vitamin D, thyroid

    hormone o mechanism:

    -1. ligand is lipid soluble, hence can cross plasma membrane -1. binds with intracellular receptor -1. intracellular receptors increases gene transcription by directly or

    indirectly acting on promoter region o drugs that act on intracellular receptors are slow acting because gene

    transcription takes time, but endure hours after the drug has been eliminated from the body

    4. ligand-regulated transmembrane enzymes (protein tyrosine kinases) o mechanism:

    -1. ligand (includes insulin, growth factors) binds to receptor -1. enzymes (tyrosine kinases) located in the cytoplasmic domains

    (receptors have large extracellular and intracellular domains) are activated and cause phosphorylation of certain targets

    -1. these targets are activated or inactivated by this

    SPECIFIC EXAMPLES DISCUSSED IN LECTURES, NOT ALLUDED TO ON HANDOUT

    2. SURGERY o during surgery, give curare to block nicotinic receptors (found on skeletal

    muscle) o therefore, surgeon can cut through muscle (it is relaxed) o now, the curare stops breathing, therefore, must respirate patient o after operation, give acetylcholinesterase inhibitor (stops

    acetylcholinesterase from breaking down ACh in the synaptic clefts from

  • being broken down) which will increase the amount of ACh in the synaptic clefts which is now in a great enough concentration to overcome the curare, and hence, breathing will return quickly

    2. CHOLERA o is caused by a bacterium - cholerae o bacterium secretes a toxin o toxin enters intestinal cells o toxin prevents GTP hydrolysing to GDP o this prevents G stimulatory protein from shutting itself off o therefore, intestinal cells accumate cAMP o this leads to a Cl-/Na+ imbalance (more Cl- and Na+ in the lumen) o hence, water is lost to the lumen o this results in diarrhoea

    2. CAFFEINE o this inhibits phosphoresterase o normally phosphoresterase hydrolases cAMP, which decreases cAMP o hence, caffeine causes the following effect: increased cellular cAMP in

    smooth muscle --> vasodilatation in kidney --> increases GFR --> diuresis

    PHARMACOLOGY LECTURE 4

    PHARMAKINETICS - GENERAL PRINCIPLES pharmacokinetics is the characterisation of the time course of drug/metabolite

    concentrations in the body clinical pharmacokinetics is the application of pharmacokinetic principles to the

    therapeutic management of patients disposition

    o is defined as those pharmacokinetc processes that occur after absorption of the drug into the body (i.e. dristribution and elimination, and includes:

    0. movement of drug from the site of administration to site of action 0. movement of drug to other organs 0. excretion of drug

    absorption: most drugs are administered orally, and the gastrointestinal absorption of drugs is a function of its physico-chemical properties (pKa, lipophilicty, molecular weight) and the physiology of the gastrointestinal tract (pH, gastric emptyimg)

    distribution: is a function of: o affinity of drug for tissue and plasma o blood flow rates to organs o drug's physico-chemical properties

    elimination: is a function of o biotransformation in the liver to metabolites

  • o excretion of the drug by the kidney various parameters:

    o bioavailability = F o volume of distribution = V o half-life = T(1/2) o clearance = CL

    first order kinetics = the rate of absorption, distribution and elimination is directly proportional to the concentration

    zeroth order kinetics = the rate of absorption, distribution and elimination are independent of the concetration

    the therapeutic window = the range of concentrations that give both adequate therapy without unacceptable toxicity

    PHARMACOLOGY LECTURE 5

    PHARMACOKINETIC CONCEPTS the two basic processes that modify pharmacokinetics (how the body deals with a

    drug) are: 0. clearance 0. volume of distribution

    CLEARANCE = CL

    INTRODUCTION o CL = rate of elimination/concentration (mg/h) o elimination of drug can occur at:

    CL(renal) = rate of elimination at kidneys / C CL(liver) = rate of elimination at liver / C CL(other) = rate of elimination not at the kidney or liver / C CL(total) = CL(renal) + CL (liver) + CL (other)

    o first order elimination (constant CL) for most drugs the rate of elimination is directly proportional to the

    concentration, and so clearance is constant o zeroth order elimination (variable CL) - capacity limited elimination

    some durgs do not have a constant clearance; these are capacity limited drugs (e.g. ethanol)

    for these drugs, an increase in concentration, does not see a corresponding increase in elimination because the pathways of elimination are saturated

    note: actually, most drug elimination pathways will become saturated if the concentration is high enough, and so eventually, at a high enough concentration, all drugs will become capacity

  • limited BUT most drugs are given at concentrations that aren't nearly high enough for this

    a formula describing rate of elimination is: rate of elimination = (Vmax + C) / (Km + C)

    o flow dependent elimination: for drugs that are cleared very readily, elimination is dependent on blood flow

    MEASUREMENT OF TOTAL BODY CLEARANCE: o area under concentration curve = AUC (this is the total area under the

    blood/plasma concentration time curve) - see handout for diagram o F = bioavailabilty (how much drug reaches the circulation after ingestion) o Css = steady state concentration o tau = the AUC within the dosing interval divided by the dosing interval o single dose:

    IV: CL = dose / AUC oral: CL = dose x F / AUC

    o chronic dosing: IV constant infusion into body: CL = dose rate / Css IV giving at separate intervals: CL = dose / (tau x Css) oral taken at separate intervals: CL = F x dose / (tau x Css)

    CLINICAL RELEVANCE o clearance is the parameter which determines the maintenance of dose rate

    required o rearrangement of the IV constant infusion into body formula gives:

    maintenance dose rate = desired concentration x clearance (i.e. dose rate = Css x CL)

    o clearance is an independent pharmacokinetic parameter

    VOLUME OF DISTRIBUTION (V)

    INTRODUCTION o V = amount of drug in body / concentration in plasma o example: a high V --> a low blood concentration, and therefore a high

    drug concentration in extravascular tissue MEASUREMENT OF V

    o V = dose/C(0) = dose/(AUC x terminal slope) o C(0) = plasma concentration at time 0 o C(0) can be determined by drawing a log plasma concentration vs. time

    and extending the line to time 0; note that this is most accurate using log paper since the line will probably be straight with log papers

    CLINICAL RELEVANCE o in some clinical settings, it is important to achieve the target concentration

    instantly, rather than waiting for some time - the following formula can be used

    loading dose = Css x V o volume of distribution is an independent pharmacokinetic parameter

  • HALF LIFE (t(1/2))

    is the time taken for the blood concentration to fall by 50% clearly, half life will be constant for first order drugs MEASUREMENT OF HALF LIFE

    o look at the concentration vs. time graph o OR calculate using:

    half life = ln2 x V / CL half life is a dependent pharmacokinetic parameter derived from the

    indepdent parameters CL and V CLINICAL RELEVANCE

    o for continuous dosing, steady state is said to be achieved after 5 half lives

    BIOAVAILABILITY (F)

    is defined as the frction of drug dose reaching the systemic circulation following administration by any route

    o F = 1 for IV administration o 0 < F < 1 for oral administration

    obviously therefore, F = (AUC[oral] / AUC[iv]) x (dose[iv] / dose[oral]) estimation of area under curve: bloody obvious

    PHARMACOLOGY LECTURE 6

    ADVERSE DRUG REACTIONS (ADR)

    Adverse drug reaction: noxious and unintended effect of drug, occurring at doses used for prophylaxis, diagnosis or therapy.

    CLASSIFICATION: note that it is assumed drug dosage is within therapeutic window

    Type A - explainable

    type A ADR is the result of an exaggerated, but otherwise expected pharmacological effect of drug

    are predictable and dose dependent examples:

    o antihypertensive drugs - postural hypotension o morphine - constipation o caffeine - palpitations o anti-epileptic drugs - sedation

  • some causes: -1. pharmacokinetic (what body does to drug) - common

    due to variability between patients in: bioavailability, first pass effect, metabolism and renal excretion

    predisposing factors: age pregnancy diseases affecting pharacokinetics (e.g. kidney disease)

    and/or pharmacodynamics, and pharmacogenetics (oxidation, acetylation)

    dose reduction will avoid ADR -1. pharmacodynamic (what drug does to body)

    result from altered end-organ sensitivity (e.g. changes in receptors)

    predisposing factors: eldery (impaired homeostasis; e.g. postural hypotension

    with diuretics) drug interactions unavoidable toxicity (e.g. cancer chemotherapeutic drugs

    causes bone marrow depression) -1. pharmaceutical formulation - not common

    changes in the way the drug is distributed to the body can increase concentration (e.g. by increasing bioavailability), so that the concentration is at toxic levels

    example: in 1968, changes with phenytoin increased bioavailability leading to toxicity

    Type B - unexplainable

    type B ADR is not expected from known pharmacological action of drug example:

    o skin rash - penicillin o haemolysis - antimalarials

    some causes: -1. pharmaceutical

    due to in vitro degradation of products or additives -1. pharmacokinetic

    due to formation of electrophilic drug metabolite binding covalently to tissue macromolecules

    exmple: sulphonamides - major route of metabolism is n-acetylation (non-toxic), but minor route is via cytochrome P450 metabolism (forms a reactive metabolite which binds covalently to tissue and causes cellular toxicity)

    -1. pharmacodynamic example: haemolysis with sulphonamides in patients with red

    blood cell glucose-6-phosphate dehydrogenase deficiency

  • Comparison between A and B

    feature = type A / type B pharmacology = augmented / bizarre predictable = yes / no dose dependent = yes / no morbidity = high / low mortality = low / high

    Other

    6. teratogenic, e.g. warfarin 6. carcinogenic, e.g. diethylstilboestrol 6. drug allergy

    o previous exposure - sensitisation o antigen can be drugs, drugs + protein, metabolites, impurities o 4 posible mechanisms

    0. immediate anaphylactic 0. antibody dependent cytotoxicity 0. antigen-antibody complexing 0. cell-mediated

    EPIDEMIOLOGY

    accurate informatino hard to get o 5-35% of hospital admissions involve ADR o 5% of hospital admissions are primarily due to ADR o 10-20% of inpatients suffer ADR o 0.1% of deaths on wards directly caused by ADR o communicty prevalence unknown

    FACTORS PREDISPOSING TO ADR

    1. presence of severe disease, especially renal, hepatic and heart 1. polypharmacy and prolonged treatment 1. extremes og age 1. previous ADR 1. gender: females more susceptible than males 1. atopic history (more likely to have an allergic reaction) 1. genetic polymorphism

    ADVERSE DRUG REACTION MONITORING - may be drug-oriented, disease oriented, complication oriented

    Methods

  • 7. spontaneous - case reports (only a small proportion are reported) 7. intensive hospital monitoring - research based (high costs, limited numbers of

    patients) 7. record linkage - collection of medical records of patients (confidentiality is a

    major issue in Australia) 7. mandatory/compulsory - must report by law (e.g. in Sweden) or to gain

    accreditation (e.g. Canada) 7. regulatory - manufacturer must keep record of all ADRs reported to it by doctor

    or pharmacist

    ADRs and Australia (spontaneous reporting)

    Adverse Drug Reaction Advisory Committe set up by Australian Drug Evaluation Committee

    ADRAC regularly reviews all Australian reports of suspected ADRs (5000 each year) and publishes cases and sends them to WHO Collaborating Centre for International Drug Monitorings

    ADRAC reports on suspected adverse drug reactions are sent free to all docts, dentists and pharmacists

    What to report? Anything, no matter how trivial, particularly for

    3. new drugs 3. suspected drug interactions 3. reactions signifantly affecting patient management (death, life in danger, hospital

    admission or prolongation, birth defect)

    Note that premarketing clinical trials provide no guarantee that all relatively common adverse effects will be adequately domcumated (e.g. persistent cough in 1 in 100 ACE inhibitor users)

    PHARMACOLOGY LECTURE 7

    DRUG ABSORPTION

    drug absorption = the process by which drug proceeds from the site of administration to the site of measurement (blood stream) within the body

    absorption occurs for the following drug fomrulations: 0. oral 1. sublingual (will avoid breakdown in stomach and liver) 2. transdermal (e.g. niotine) (very slow absorption)

  • 3. rectal (use when patient is vomiting) 4. intramuscular injection 5. subcutaneous injection 6. miscellaneous - inhalation, intranasal, eye, nose, ear drops

    gastrointestinal absorption = extent to which drug is absorbed from gut lumen into portal circulation

    drug absorption occurs by following 2 mechanisms: 0. passive diffusion

    rate of transfer across membrane = concentration difference x surface area x permeability constant

    permeability constant = diffusion coefficient x partition coefficient / membrane thickness

    diffusion coefficient = k / Sqrt (molecular weight) molecular weight: this is realtively constant for most drugs because

    molecular weights usually vary between about 100 and 500 membrane thickness: this is relatively constant partition coefficient:

    this is a marjor source of variation in drug diffusion drugs which are lipophilic have high partition coefficients,

    and drugs which are lipophobic have low partition coefficients

    a value above 5 indicates high lipid solubility degree of ionisation:

    only a non-ionised nonpolar drug diffuses across the membrane, hence this is an important factor

    degree of ionisation depends on pKa of drug (and pH of body fluid)

    1. carrier mediated transport unlike passive diffusion, this exhibits saturability, specificity,

    competition, requires energy, and involves movement against a concentration gradient

    for GI absorption - facilitated diffusion is important for a few drugs based on endogenous compounds (e.g.

    levo-dopa, which is used for Parkinson's disease) and vitamins physiological factors influencing gastrointestinal absorption

    0. blood flow the blood flow is responsible for removal of drug from the other

    side of the membrane for some very lipophilic drugs, the limiting factor in

    gastrointestinal absorption is rate of blood flow (e.g. ethanol) 1. surface areas/blood flow

    the surface area of intestine is 1000 x greater than stomach blood flow of intestine is 8 x greater than stomach hence, although physico-chemical properties would dictate that

    weak acids are absorbed in stomach, and weak bases in intestine, in reality, most drugs are absorbed across intestinal wall

  • 2. gastric emptying affects the rate of drug absorption (i.e. decreased gastric emptying

    slows down absorption) gastric emptying can be decreased by drugs (opioids), disease

    (migraine, shock) and food solid oral dosage forms

    o for tablets/capsules to be absorbed, they must undergo: 0. distintegration to form granules 1. dissolution in gastrintestinal fluids 2. absorption into body

    o some special formulas are: enteric coated tablets:

    contain a coat over the tablet preventing breakdown in acid overcome

    local side effects (e.g. coated aspirin will not cause gastritic bleeding)

    degradation (e.g. coated penicillin will not undergo acid hydrolysis)

    sustained release tablets: are formulated to decrease rate of dissolution --> decrease

    rate of absorption are used to reduce the frequency of dosing (e.g. morphine)

    o hence, formulation factors can affect drug absorption o therapeutic failures have occurred when one manufacturer's brand has

    been substituted by another o the principal mechanism affecting drug absorption is dissolution - which

    can be influenced by particle size solubility fluid pH gastric emptying

    pharmacokinetic measurement of absorption: o bioavailability (F) = amount of (oral) dosewhich reaches the systemic

    circulation (between 0% and 100%) reasons why some drugs do not have bioavailability of 100%

    -1. insufficient time for absorption -1. decomposition in gut wall and lumen

    examples include: acid hydrolysis (penicillin) complexation (tetracycline with Ca++) metabolism by gut microflora metabolism in gut wall (oral contraceptive)

    -1. liver and the first pass effect = when drug is absorbed, it must first pass through portal circulation and liver

    i.e. drug absorbed across GI tract --> portal circulation --> systemic circulation

  • hence, in liver there may be metabolism of drug before it can enter the systemic circulation (e.g. morphine)

    the major determinant of oral bioavailability of high extraction drug is hepatic enzyme activity

    some drugs have low extraction (e.g. caffein), and hence high bioavailability

    some drugs have high extraction (e.g. morphine) note: F = fraction absorbed x (1-E) [E = extraction

    ratio]

    PHARMACOLOGY LECTURE 8

    DRUG DISTRIBUTION

    general concepts of distribution: o distribution = process of reversible transfer of trug to and from

    circulation o distribution rate and extent is determined by:

    delivery of drug to tissue ability of drug to pass through tissue membranes binding of drug to plasma and tissue components

    o perfusion rate limitations occur when the tissue membranes are not a barrier to distribution

    of a drug hence well perfused tissues take up the drug more rapidly than

    poorly perfused tissues (e.g. lungs, kidneys > brain, muscle > fat, bone)

    the time to convey to the tissue the amount of drug at equilibrium depends on:

    1. Kp - the equilibrium ratio (Kp = equilibrium concentration of tissue / equilibrium concentration of venous blood)

    2. perfusion rate o permeability rate limitations

    occur when the tissue membranes act as a barrier to distribution of a drug

    factors that act as barriers to distribution include: 1. tight capillaries (e.g. in brain)

    most capillaries are relatively leaky, and permeable all except very large molecules, e.g. proteins and protein bound drugs

  • however, in the brain the endothelial cells have no fenestrae and are attached by tight junctions

    2. cell membranes physico-chemical properties (molecular weight and

    solubility) determine rate at which drug penetrates cell membranes since they determine a moluecles ability to pass through:

    1. lipid matrix of membrane 2. membrane pores

    3. affinity for active transport carriers o distribution equilibrium = unbound concentrations of drug in tissue and

    plasma are equal the exception to this will be if tissue is involved in excretion or

    metabolism or where there is active transport out of the tissue plasma protein binding is an important factor in determining a drug's volume of

    distribution because only drugs not pound can be distributed outside the circulation

    o affinity of protein for a drug: [unbound drug] + [protein] [drug-protein complex] k1 is the rate constant of the reaction (drug + protein --> drug-

    protein) k2 is the rate constant of the reverse reaction (drug-protein -->

    drug + protein) Ka = k1 / k2 = association constant (a measure of affinity of the

    protein for the drug) o drugs bind predominantly to two proteins:

    0. albumin comprsises 60% of total plasma proteins acts as a transporting protein for numerous endogenous

    substances binds various anions, cations, steroids, bilirubin, many

    drugs (especially those which are acids) the drug-albumin complex usually consists of one or two

    high affinity binding sites, and many low affinity binding sites

    the 2 high affinity bindings sites are: 0. bilirubin binding site (drugs such as warfarin and

    sulphonamides bind) 1. benzodiazepine binding site (drugs such as

    benzodiazapines, tryptophan and medium change fatty acids bind)

    knowledge of which site on albumin a drug binds enables the prediction of drug-drug and drug-endogenous ligand displacement interactions

    displacement interactions and reduction in albumin concentrations may may make interpretation of total

  • (unbound + bound) concentrations difficult in therapeutic drug monitoring

    1. alpha1-acid glycoprotein is only in small concentrations in plasma, but does bind to

    many drugs (e.g. propranolol, lignocaine, imipramine) is an acute phase protein, and so in inflammatory diseases

    (including MI and cancer) its concentration may increase several fold

    binding to this protein usually occurs at one site of high affinity

    o other proteins which bind drugs are: lipoproteins (bind triglycerides, phospholipids, cholesterol, some

    drugs) transcortin (binds steroids) thyroxine binding globulin retinol binding globulin other structures (e.g. white cells bind some antimalarial drugs)

    tissue binding o tissue binding occurs at following sites:

    tissue proteins DNA (e.g. antimalarials) lipids (e.g. fat soluble drugs and environmental pollutants) skin/ocular melanin (e.g. quinacrine)

    kinetics of protein binding o fu = unbound fraction of drug in plasma = 1 / (1 + (Ka x Pu))

    Ka = association constant Pu = [unbound protein concentration] = Pt - DP

    Pt = [total protein] DP = [drug-protein]

    (note: fu is normally constant, i.e. independent of drug concentration (because only a small fraction of available binding sites are occupied))

    o fu is affected by: 0. effect of drug concentration

    if concentration of drug is so high that almost all drug binding sites are filled, then a further increase in concentration will not cause more drug-protein complexes to form, hence fu will increase (i.e. fu will no longer be constant)

    (in other words, as DP approaches Pt, fu will become variable, i.e. concentration dependent)

    this is uncommon for albumin because [albumin] is high and so large concentrations of drug are required to saturated the binding sites

  • this is more common for alpha1-glycoprotein because [alph1-glycoprotein] is low and so lower concentrations of drug are required to saturated the binding sites

    1. effect of protein concentration if total protein concentration decreases, then fu increases albumin may decrease in nephrotic syndrome or severe

    liver cirrhosis (increase fu) alpha1-acid glycoprotein may increase in many diseases

    (e.g. rheumatoid arthritis) (reudce fu) 2. effect of binding affinity:

    a decrease in affinity will increase fu this may occur because of:

    disease states displacement by endogenous ligands or other drugs

    binding and drug disposition o knowledge of degree of drug-plasma binding is important because:

    0. only unbound drug is pharmacologically active 1. is a determinant of drug's volume and distribution 2. assists in elucidating clearance mechanisms

    o volume of distribution (V) = amount of drug in body (A) / plasma concentration (C)

    a large volume of distribution indicates extensive tissue binding a simple model is:

    V = Vp + (fu x Vt / fut) Vp = plasma volume (about 3L) fu = drug's unbound fraction in plasma Vt = tissue volume fut = unbount fraction in tissue Vt can be extracellular water (15 L) or total body

    water (45 L) if a drug is polar and does not penetrate

    membrane easily, then Vt will be 15 L if a drug is lipophilic then it will distribute

    into total body water, (Vt = 45 L)

  • PHARMACOLOGY LECTURE 9

    DRUG CLEARANCE - HEPATIC METABOLISM

    Introduction

    a major route of drug clearance involves the metabolism of lipophilic drugs to hydrophilic derivatives that can be eliminated by the kidneys

    this process is called biotransformation

    Tissues invovled in drug metabolism

    most drug metabolism invovles enzymes in the liver, many of which are associated with the smooth endoplasmic reticulum of hepatocytes

    metabolism of drugs by liver directly after absorption into the portal system prior to reaching circulation is refered to as first pass metabolism

    other tissues that play a role in drug metabolism include: o gastrointestinal tract (contributes to first pass clearance) o kidneys o skins (relevenat following topical applications of drugs)

    Classification of drug metabolism pathways

    phase I reactions o (functionalisation = introduction of a new functional group) o phase I reactions are functionalisation reactions, frequently adding a new

    oxygen atom (e.g. -OH group) or exposing an existing functional group (e.g. hydrolysis of ester linkage)

    o are usually catalsed by oxidases located in endoplasmic reticulum, mainly cytochome P450 enzymes

    o CYP450s are mainly in hepatocytes, but also cells of the kidney, lung, intestine, skin, testis, brain

    o each CYP450 oxidises many substrates (with some selectivity for classes of chemicals)

    o nomenclature of CYP450s: example P4502E1

    first number (i.e. "2") refers to CYP family (>40% homology)

    capital letter (i.e. "E") refers to subfamilies (>60% homology)

    second number (i.e. "1") refers to a particular P450 isoform

  • o major human cytochrom P40s (also called hepatic mono-oxygenases) CYP1A2

    is expressed universally in liver tissue is further induced by smoking, ingestion of grilled meats metabolises drugs such as: caffeine, theophylline,

    phenacetin by dealkylations also activates aromatic amino procarcinomages

    CYP2C9/10 and CYP2C18/19 (important) collectively, the 2C family is very important in drug

    metabolism, e.g. taxol, phenytoin, diclofenac, piroxicam, diazepam, cycloguanil, imipramine

    a genetic deficeincy (polymorphism) for CYP2C9 was identified with tolbutamide

    a genetic polymorphism for CYP2C19 was identified with mephenytoin

    CYP2D6 (important) expressed mainly in liver important in drug metabolism, e.g. codein, debrisoquine,

    propranolol, captopril, dextromethorphan, nortryptiline a genetic polymorphism (identified with sparteine) affects

    10% of Caucasions, 2% of Mongoloids and Negroes CYP2E1

    expressed mainly in liver, and also peripheral lymphocytes induced by ethanol (major role in ethanol metabolism in

    alcoholics) minor role in drug metabolism, e.g. paracetamol,

    chlorzoxazone important in metabolism of a range of industrial chemicals,

    e.g. benzene, styrene, vinyl chloride, carbon tetrachloride, chloroform, ethyl carbamate

    CYP3A4 (important) most abundant human isoform (liver, intestine) induced by barbiturates, rifampicin, glucocorticoids most important CYP450 in drug metabolism, e.g.

    nifedipine, quinidine, taxol, erythromycin, contraceptives, warfarin, cyclosporin, midazolam, lidnocaine

    phase II reactions o phase II reactions are biosynthetic reactions (conjugation reactions) o involve conjugation of a hydrophilic endogenou cofactor with a drug o the conjugate is usually more polar than metabolites formed via phase I

    oxidation, thus phase II conjugates are readily cleared by kidney or bile o major phase II reactions include (primarily occur in liver)

    glucuronidation - transfer of glucuronic acid to drug enzyme involved: UDP-glucuronosyl transferase cofacter required: UDP-glucuronic acid

  • functional group required in drug: -OH, -COOH, -NH2, -NH, -SH

    sulphation - transfer SO3- to drug enzyme involved: sulfotransferase cofacter required: PAP-sulphate functional group required in drug: aromatic -OH or -NH2

    acetylation (transfer of acetyl group to drug) enzymes involved: N-acetyltransferase cofacter required: acetyl CoA functional group required in drug: aromatic or aliphatic -

    NH2 glutathione conjugation (add glutathione to drug)

    enzyme involved: glutathione-S-transferase cofacter required: glutathione functional group required in drug: epoxides, organic

    halides

    Integration of drug metabolism

    for a drug to be metabolised by phase II conjugation it requires an appropriate function group

    such functional groups may have been introduced by a phase I enzyme thus, phase I and phase II reactions are tightly integrated:

    o drug in plasma --> oxidised metabolite --> conjugated metabolite o either the product of the phase I reaction (oxidised metabolite) or of the

    phase II reaction (conjugated metabolite) may be excreted via urine or bile

    Pharmacological implcations of drug metabolism

    modification by phase I and phase II reactions usually alters the pharmacological properties of the agent

    pharmacological deactivation o most drug metabolites exhibit less affinity for relevant receptors, thus

    biotransformation usually implies pharmacological deactivation o biotransformation also typically generates metabolites that are more easily

    cleared from the body, further dimishing the duration of a drug's action o example: amphetmine

    is a CNS stimulant, reversing fatigue symptoms inactivated by CYP450-catalysed oxidation (deamination)

    o example: diazepam is a hypnosedative used in control of anxiety, insomnia undergoes oxidation by CYP2C19 to an inactive metabolite (N-

    demethylation) pharmacological activation

    o for a few drugs, inactive prodrugs undergo metabolism within the body to active drugs

  • o example: sulindac is a NSAID used in treatment of rheumatoid arthritis, gout and

    moderate pain converted to an active silphide metabolite by reductive phase I

    metabolism occurring in the gut (catalysed by bacterial reductases in gut contents, not gut wall)

    resulting metabolite is 500 times more potent inhibitor of cyclooxygenase than parent

    o example: codeine is an inactive prodrug that required CYP2D6-catalysed O-

    demethylation to form morphine, which is much more active a mu opioid receptors

    in a subsequent reaction, morphine is converted to a potent analgesic (morphine-6-glucuronide) via glucuronidation (note that it is unusual for phase II metabolites to be active)

    Role of drug metabolism in drug interactions

    knowing the metabolic fate of drug is important because: 3. it can help avoid adverse effects due to drug interactions during tretment

    with 2 or more drugs example: ketoconazole is metabolised by a specific CYP450 (3A4)

    and can inhibit the clearance of terfenadine which is a substrate for the same CYP450 (hence cardiac toxicity)

    3. prolonged exposure to some drugs can cause overexpression of CYP450s (induction) causing a loss of potency of other drugs that are metabolised via that isoform

    example: barbiturates induce specific CYP, and eththinylestradiol (contraceptive) is metabolised by this CYP, and so is metabolised too quickly (therefore ineffective) if patient is taking barbiturates

  • PHARMACOLOGY LECTURE 10

    RENAL EXCRETION

    Introduction

    renal excretion is the final step in most drug pathways (water-soluble xenobiotics and lipophilic compounds converted to water soluble metabolites are excreted)

    fraction excreted unchanges = fe = amount in urine / dosage = renal clearance / total clearance

    CL(total) = CL(renal) + CL (hepatic) + CL (other) {assume that CL (other) is minimal}

    thus, if drug's total clearnace and fe are known, renal and hepatic clearances can be calculated

    for 70% of drugs, CL(renal) < CL(nonrenal)

    Mechanisms

    1. filtration o 20% of renal flow forms glomerular filtrate (typically, GFR = 120 ml/min) o protein binding of a drug allows only unbount portion (fu) to filter o rate of filtration of a drug from plasma is related directly to plasma

    concentration (C) o equation: rate at which drug is filtered at glomerulus = C x

    CL(glomerular) = C x fu x GFR 2. active transport in the proximal tubule

    o reabsorption: the proximal tubule performs active reabsorption of glucose and

    amino acids, but drugs are not carried by these transporters o secretion:

    the proximal tubule has 2 active transport excretory systems (accepting organic cations and anions respectively) which are non-selective and excrete a wide variety of xenobiotics and endogenous substrates

    these systems: are energy dependent can be saturated at high concentrations (hence substances

    handled have a tubular maximum secretion rate [Tm]) show competition between drugs for the same transporter

    natural substrates for these transporters include metabolites conjugated with sulphage, glucuronide and glycine, and metabolic products such as creatinine (which is also filtered)

    3. distal tubule and collecting duct

  • o as the tubular concentration of drug rises progressively along the nephron due to reabsorption of water, hence a concentration gradient develops which favours the passive diffusion of drug from the tubule into the peritubular capillary

    o reabsorption varies from negligible to compelte, denedning on the physico-chemical properties of drug (molecular weight, pKa, lipophilicity)

    o two physiological variables are important in this process -1. urine flow rate

    this influences the drug concentration concentration gradient

    low tubular flwo rate will be accompanied by a higher drug concentration in distal tubular fluid, and more complete reabsorption of a lipid-soluble drug

    -1. distal tubular pH varies between 4.5 and 8 changes within this range alter degree of ionisation of a

    weak acid or base since non-ionic form only will be reabsorbed, a chnage in

    urine pH can alter tubular reabsorption example: urate excretion can be enhanced by alkalinising

    the tubular fluid (uric acid is a weak acid) since there will be more ionised urate

    overall: CL (renal) = CL(GFR) + CL (secretion) + CL (reabsorption) {note that CL (reabsorption) < 0}

    Significance

    renal disease: o renal disease alters all forms of renal excretion o renal clearance, tubular secertion etc. are reduced in direct proportion to

    the reduction in GFR o this will be clinically relevant if the major pathway of excretion is renal o example:

    lipiophobic beta-blocker atenolol has > 90% renal excretion and an intermediate therapeutic index

    in renal failure, toxicity is common unless dose is reduced estimating drug excretion:

    o normal renal function varies widely with age, body weight etc. o plasma creatinine is not sufficiently reliable o 24 hour urine collections to directly measure creatinine clearance is

    impractical o if a high degree of precision is needed, creatining clearance (GFR) is

    estimated from plasma creatinine, body weight, age, and sex a good method is the Cockkroft and Gout formula

    estimated creatinine clearance (ml/min) = (140-age) x weight / (serum creatine (mmol/L) x 815)

  • multiply this value by 0.85 for females other estimation formulae exist these methods are not valid if renal function is not stable, or on

    extremely high or low protein diets, or at extremes of age o for the few drugs which have a narrow therapeutic index and are mainly

    excreted renally (e.g. aminoglycosides and digoxin), individual GFR should be estimated and then the individual's total clearance for the drug derived; from this the correct dosage (loading and maintenance and timing) can be calculated; therapeutic drug monitoring should also occur

    o example: for digoxin, total clearance in normal man is 130 mil/min, fe = 0.6 by how much should the normal maintenance dose rate (250

    micrograms/day) be altered to achieve the same Css in a patient with a GFR of 30 ml/min?

    you know that F x dose rate = CL x Css therefore, to keep Css constant, need to adjust dose rate in direct

    proportion to Cl CL(nonrenal) = 130 x (1 - fe) = 130 x 0.4 = 50 ml/min (for normal

    man and patient since this involves liver) in normal man, CL(renal) = 130 x 0.6 = 80 ml/min in patient, CL(renal) = 30 x 0.6 = 20 ml/min therefore in patient, CL(total) = CL(nonrenal) + CL (renal) = 50 +

    20 = 70 ml/min hence, need to reduce maintenance dose to 70/130 = 55% of

    normal (125 micrograms/day) altered tubular secretion - potential drug interaction

    o anion pathway: handles many drugs such as penicillins, cephalosporins,

    probenecid, salicylates, diuretics (PAH (para-amino-hipppurate) is transported avidly, and clearance

    by the healthy kidney > 90% of total renal plasma flow (600 ml/min) - hence can be used to determine renal blood flow; fortunately most drugs aren't transported so efficiently)

    o cation pathway: handles drugs such as cimetidine, procainamide, amiloride,

    pindolol, ranitidine o drugs compete for transport, thus introducing one can reduce the renal

    clearnace of another example: probenecid has been used to reduce excretion of

    penicillin, and thus reduce the dose required (by up to 80%) example: cimetidine can produce a significant rise in plasma

    procainamide due to competition for common transport mechanism o note: in neonates, tubular secretory function is very poor, but develops

    after birth - caution is needed in drug treatment distal tubule

    o effect of urine volume

  • drug nephrotoxicity can be worse when urine volume is low (e.g. gentamicin toxicity)

    in drug overdoses: high fluid intake and therefore urine output often aids recovery

    o effect of urine pH for drugs that are weak acids (pKa 3-7) or weak bases (pKa 6-12),

    the degree of ionisation in tubular fluid is dependent upon pH example: methamphetamine

    is a weak base (pKa = 10) renal excretion is 4 x faster in an acid than an alkaline urine

    (because obviously a lower pH means lower ionisation and hence less reabsorption and hence greater excretion)

    example: phenobarbital is a weak acid (pKa = 7.4) renal excretion is 7 x faster in alkaline urine (however renal

    clearance is still only a small fraction of total clearance) clinically important example: salicylic acid

    is a weak acid (pKa = 3.5) at physiologic pH it is mainly ionised at pH of 5.0, the amount of non-ionised form is 25 x that

    present at pH of 7.4 in serious aspirin overdose, a systemic and tubular acidosis

    occurs, which enhances tubular reabsorption (and therefore prolongs half life of elimination)

    this can be reversed by giving systemic alkali and fluids and producing an alkaline pH urine with high flow

    (note that haemodialysis is actually used in life-threatening poisoning)

    renal handling of drug metabolites o for a few drugs, the metabolite is toxic or active, examples include:

    clofibrate is a cholesterol lowering drug is metabolised in liver to clofibric acid glucuronide which

    is excreted renally however, in renal failure, clofibrate treatment produced an

    unexpected syndrome of muscle pain and inflammation it turned out that clofibric acid is a selective muscle toxin at

    the plasma levels reached in renal failure morphine

    in renal failure, morphine has much longer duration of action than plasma half life (the plasma half lfie is not prolonged in renal failre)

    morphine is metabolised in part to morphine-6-glucuronide which is renally excreted

    M-6-g has opioid properties, and elevated levels of m-6-g account for this difference in renal failure

  • hence give morphine in less frequent doses in patients with failure

    procainamide dose of procainamide should be reduced in renal failure

    because its excretion is reduced, and the major metabolite N-acetyl-procainamide (renally excreted) is antiarrhythmic

    it is usual to use TDM and measure procainamide and N-acetyl-procainamide levels to control treatment

    drugs acting on kidneys o some drugs depend on concentration mechanisms in renal tubule for their

    therapeutic effect and selectivity action o examples:

    urinary tract antibiotics (nitrofurantoi, nalidixic acid) are present in urine at 100 x the plasma concentration

    loop acting (freusemide) and distal vonvoluted tubule acting (thiazides) diuretics depend on tubular concentration for their selective effects on the luminal surface of the tubular cells

    o these drugs become less effective when the tubular concentrating process is impaired in renal disease

    PHARMACOLOGY LECTURE 11

    INTERRELATIONS Diagrams on handout are useful

    Introduction

    usually, there is a functional and reversible relationship between the concentration of a drug at its site of action and the intensity and duration of response produced

    because it's impossible to measure concentration of drug at site of action and response at site of action, inferences have to be made by studying plasma drug concentrations and clinical responses

    Concentration and response

    response increases with increasing concentration (see diagrams - note that the shapes of 1a and 1b are different because 1a has a linear x axis scale, and 1b has a logarithmic x axis scale) - note that this is not a linear relationship (i.e. doubling the dose does not double the effect)

    Hill equation:

  • o intensity of effect = (Emax x c^n) / (EC50^n + c^n) o Emax = maximum effect o EC50 = concnetration needed to produce 50% of Emax o c = concentration of drug in plasma o n = shape (also called slope) factor that accommodates the shape of the

    curve a higher n defines a steeper curve example: if n = 1 then EC20 = 0.25EC50 and EC80 = 4EC50 example: if n = 2 then EC20 = 0.5EC50 and EC80 = 2EC50 most drugs have an n value between 1 and 3

    examination of 1b shows that: between about 20% and 80% of maximum effect, response is approximately directly proportional to the logarithm of the concentration

    Time delays

    drug effect often lags behind plasma concentrations example: digoxin

    o used for treatment of heart failure and some arrhythmias o has a positive inotropic effect (i.e. it increases force of heart muscle

    contraction) o the effect lags behind plasma concentrations by several hours after dosing o this delay is related to the time taken for the drug concentration to

    equilibriate between the plasma and the site of action (myocardium) o hence TDM during the distribution phase is highly misleading

    see figure 2 o the diagram is divided into 4 parts:

    1. large increase in concentration, small increase in effect 2. small increase in concentration, large increase in effect 3. small decrease in concentration, no change in effect 4. decrease in concentration, decrease in effect

    causes for the time delay in effect following concentration include: 0. time taken for drug concentration to equilibriate between plasma and site

    of action 1. drug produces active metabolite(s) 2. observed effect is indirect measure of true effect

    example: allopurinol used to prevent gout acts to inhibit xanthine oxidase (the enzyme that catalyses

    the formation of uric acid) 2. drug reduces the concentration of an endogenous substance, whose

    concentration must fall below a critical value to produce observed response

    example: warfarin is an anticoagulant inhibits formation of several clotting factors

  • 2. drug acts irreversibly at site of action example: aspirin

    inhibits enzyme cyclooxygenase

    Onset-duration-intensity relationships

    following asumptions are made: o response can be characterised by Hill equation o only the drug acts to produce response and does not have active

    metabolites o the drug does not influence its own kinetics (for example, by altering liver

    blood flow or fu or CL) onset (see figure 3)

    o onset occurs when concentration at site of action reaches critical value (Cmin = minimum concentration at site of action, Amin = minimum amount in the body needed to achieve effect)

    o onset is governed by factors such as: dosage form (e.g. rapid release or slow release tablets) dose size route of administration absorption rate distribution kinetics concentration-response relationships (EC50, n)

    duration (see figure 3) o duration of effect will last as long as Cmin is exceeded at site of action o is a function of dose and rate of removal (elimination or redistribution) o if a drug is eliminated by first order kinetics: doubling a single dose, will

    obviously increase the duration of effect by one half life o there is a limit to how many dose doublings can be achieved because of

    toxicity o hence multiple dosings are usually essentially

    Integration of concentration-time-intensity relationships

    see sheet; it is very basic and obvious, and hence not worth writing about

  • PHARMACOLOGY LECTURE 12 AND 13

    VARIABILITYIN DRUG RESPONSES

    Description of variability

    dose(min) - dose effective at the 5 centile (of the population) dose(max) - dose effective at the 95 centile typically, dose(min):dose(max) = 1:4

    o example: penicillin is effective for 5% of population at dose z, and is effective for 95% of population as dose 2z, hence dose(min):dose(max) = 1:2

    sources of variability are: o genetic o drug interactions o food interactions o drug formulation and route o age, weight and gender o disease o environmental factors o in practice, behavioural factors are common (non compliance and dosing

    errors)

    Classification of variability in response

    pharmacokinetic factors o absorption

    0. formulation of drug this determines rate and extent of dissolution an unreliable oral formulation can lead to incomplete and

    variable absorption examples where this especially a problem: digoxin,

    phenytoin, cyclosporin 1. route of administration

    patient response varies with route example: lignocaine

    is an antiarrhythmic well absorbed orally, but because of high

    presystemic metabolism, therapeutic plasma levels are not reached, hence has to be given intravenously

    also has a neurotoxic metabolite, methylxylidide 2. other drugs: may interfer with absorption 3. food may alter absorption, examples include:

  • fat delays gastric emptying and therefore prolongs the T1/2 of absorption - this may delay Tmax but does not usually affect bioavailability (although a poorly absorbed drug such as griseofulvin (antifungal) has a higher F after food)

    dietary Ca++ forms non-absorbed chelates with tetracycline (these should therefore be taken on an empty stomach)

    absorption of L-dopa is less reliable after food because of competition for its active transport system (in gut wall and blood-brain barrier) by amines in food

    o bioavailability for lipid soluble (well absorbed) drugs, F is mainly determined by

    degree of presystemic hepatic metabolism (F = 1 - E(H)) E(H) is dependent on hepatic enzymes and shows marked

    interindividual variation clearly, for drugs with a large E(H), only a small variation in E(H)

    will lead to a large change in F example: propranolol

    has an average E(H) = 70%, therefore average F = 30%

    a subject showing a 50% reduction in Vmax will half E(H), hence F will become 65% and less than one half the average oral dose will be required

    this is not true for the majority of drugs that have a negligble first pass effect (E(H))

    o volume of distribution for a given drug, V(D) is mainly proportional to body weight

    (although for non-fat soluble drugs, lean body weight is a better predictor) (not true for neonates)

    V(D) determines the loading dose, so body weight is necessary in calculating loading doses for drugs with narrow TIs

    example: loading dose of gentamicin and tobramicin is 2 mg/kg BW

    o clearance -1. renal excretion

    renal clearance correlates directly with GFR renal function is proportional to body surface area - weight

    relates closely to this for the few drugs with a narrow therapeutic range and

    predominantlly renal excretion, maintenance dose is calculated from body weight (unless renal disease is present)

    example: usual adult dose of gentamicin is 1 mg/kg BW (8-hourly) for adults, 1.5 mg/kg BW for children (reflects higher body surface area)

    -1. hepatic metabolism

  • is major cause of variation in clearance of most drugs and is hard to predict, but associated with:

    genetic traits (most unknown and polygenic) smoking (enhance clearance of diazepam,

    chlorpromazine, theophylline) chronic alcohol intake (induces CYP450s and

    increases clearnaces of diazepam, phenytoin) acute alcohol intake (acts as a transient phase I

    enzyme inhibitor) living in a city or rurual area (city dwellers have

    higher hepatic clearance, ascribed to enzyme induction from environment exposure to chemicals)

    example: theophylline (see figure c) shows wide and unpredictable variation between

    individuals (T1/2 between 4 and 14 hours) is due to difference in hepatic clearance (but F does

    not alter (low E(H))) example: propranolol (see figure a and b)

    shows wide variation between individuals (T1/2 between 2 and 6 hours)

    E(H) varies in parallel (so a subject with impaired hepatic metabolism on propranolol will show both increased bioavailability and reduced drug clearance)

    2 non-linear kinetic examples: phenytoin and theophylline Km is close to therapeutic levels, hence a small

    change in either enzyme activity or dosage will lead to a large change in plasma steady state levels

    note: these have very high 1st pass CL Ca++ channel blockers beta blockers tetracyclic (?) antidepressants

    Pharmacodynamic factors

    poorly understood, although many drugs have a greater dynamic than kinetic variability

    monitoring drug plasma levels of these drugs will not be useful in predicting response

    for drugs that compete at site of action with endogenous ligands, plasma concentration of ligand may determine drug response:

    o example: hypotensive effect of ACE inhibitors correlates with plasma angiotensin levels; hence may cause severe hypotension in dehydration

    o example: anticoagulant effect of warfarin is partly determined by vitamin K intake (broccoli)

    or, the pathophysiologic state may determine response

  • o example: BP fall after nifedipine is proportional to original blood pressure

    Clinical importance of variability

    5. standard dose forms o example: theoretical drug

    T1/2 = 12 hours dose(min) = 50 mg (12 hourly) dose(min) : dose(max) = 1 : 4 if the drug was marketed in 50mg, 100mg and 200mg tablets then

    90% of population could take a 100 mg tablet, twice a day the 10%:

    low clearance group take the 50 mg daily (T1/2 will be longer)

    high clearance group take the 200 mg, 3 times a day (T1/2 will be shorter)

    6. starting treatment -1. CL determines maintenance dose -1. for drugs that have common dose-related side effects, treatment is started

    with dose(min) (with or without a loading dose) -1. in a few subjects, an altered response can be predicted and starting must be

    modified -1. dose-escalation is then performed as appropriate - genearlly by allowing

    concentrations to reach steady state before assessing response (although the time to response for some drugs differs from the T1/2)

    -1. for drugs that have minimal dose-related side effects, or treatment is urgent, the dose(max) is generally used for simplicity (although cost implications)

    Predicting drug responses in special population groups

    in people who need treatment, pharmacokinetics/dynamics often differ greatly from that seen in normal (because of age and disease)

    example: o peak incidence of heart failure is age > 70 o drug absorption, distribution and effects differ with old age and with heart

    failure the young

    o in kids, adjustment of dose for body weight/body surface area is usually adequate

    o however, in neonates (especially premature), drug absorpiton, distributio and clearance differ

    o premature infants: 85% of body weight is water (adult 55%) and body fat is 1% or

    less: a lipid-soluble drug will need a lower loading dose (per kg BW compared with adults)

  • hepatic metabolic pathways and renal excretion are immature (30-50% of adult values on a BW basis), maintenance drug dosage is reduced accordingly

    o examples: chloramphenicol (antibiotic) caused cardiovascular collapse - the

    'grey' baby syndrome in neonates when give in standard dosage; the drug accumulated due to reduced hepatic clearance

    penicillin: active renal tubular secretion is absent at birth, the clearance/BW of penicillin is 20% of adult values (but can be induced by drug exposure)

    phenobarbitone: is an effectve (but out-dated) treatment when given to near-term women to reduce foetal haemolytic jaundice; it crosses the placenta and induces foetal hepatic enzymes which have low activity normally; this increases conjugation and excretion of bilirubin

    the elderly o renal function:

    age-related reduction in GFR and renal blood flow is universal toxicity from standard dose of drugs eliminated mainly by kidney

    (especially digoxin and gentamicin) would be expected estimation of GFR and dose adjustement is necessary (GFR varies

    positively with 140 - age) o hepatic function:

    hepatic clearance declines (especially phase I metabolism) - but cannot be assessed clinically

    hence, T1/2 for many drugs increases with age (e.g. for diazepam) cautious drug sue, smaller doses and longer inter-dosing intervals

    are recommended o pharmacodynamic factors: elderly appear to be more sensitive to sedative

    effects of diazepam even when differences in clearance have been accounted for

    pregnancy o drug use must be minimised because of risk to foetus o drug distribution changes by 14th week because plasma and ECF volumes

    increase by 30%, and plasma proteins, including albumin, fall by 20% o also, GFR increases and some hepatic phase I metabolism is increases

    (probably induced by high levels of progesterone) o problems arise when treatment is essential, example: phenytoin (for

    epilepsy contorl) increased metabolism, hence phenytoin levels fall but, fall in albumin means that f(u) of this highly protein-bound

    drug is greater since TDM measures total drug concentrations, increasing dose to

    reach a plasma level that is in the (non-pregnant) therapeutic range will result in toxicity due to high concentrations of unbound drug

  • instead, a lower therapeutc range is desirable, and regular TDM is necessary

    after delivery, the dose needs to be rapidly reduced to avoid toxicity

    disease o low cardiac output

    in general: in shock and severe left heart failure, blood flow to most

    organs is decreased intestinal, subcutaneous and intramuscular drug absorption

    is slow and unreliable in shock therefore example: diabetics with hypotensive shock

    when given subcutaneous insulin, often do not respond until the circulation improves, when delayed hypoglycaemia may occur

    it is necessary to give insulin intravenously to ensure a prompt response

    toxicity in brain and heart: blood flow to brain and heart is better maintained than to

    other organs in shock therefore, a rapid intravenous bolus will produce very high

    peak local concentrations and increased risk of toxicity in these organs

    example: there is an increased risk of cardiac arrhythmias with intravenous digoxin in heart failure

    this is minimised by giving drugs by slow intravenous infusion of minutes

    impaired renal and hepatic function due to cardiac disease is discussed below

    o renal disease for a drug with mainyl renal excretion, clearance will be prolonged

    in renal disease example (taken from renal clearance lecture)

    for digoxin, total clearance in normal man is 130 mil/min, fe = 0.6

    by how much should the normal maintenance dose rate (250 micrograms/day) be altered to achieve the same Css in a patient with a GFR of 30 ml/min?

    you know that F x dose rate = CL x Css therefore, to keep Css constant, need to adjust dose rate in

    direct proportion to Cl CL(nonrenal) = 130 x (1 - fe) = 130 x 0.4 = 50 ml/min (for

    normal man and patient since this involves liver) in normal man, CL(renal) = 130 x 0.6 = 80 ml/min in patient, CL(renal) = 30 x 0.6 = 20 ml/min

  • therefore in patient, CL(total) = CL(nonrenal) + CL (renal) = 50 + 20 = 70 ml/min

    hence, need to reduce maintenance dose to 70/130 = 55% of normal (125 micrograms/day)

    note: if there are active metaabolites which are renally excreted,

    these may accumulate example: morphine-6-glucuronide

    Vd is sometimes lower in renal failure example: for digoxin

    non-renal clearance may vary in renal failure pharmacodynamics may differ

    despite the above points, a general rule of thumb is that if Fe < 30% (i.e. renal clearance is less than 30%) then no dose reduction is needed even in severe renal disease

    o liver disease bioavailability of drugs with high hepatic extraction is increased in

    severe liver enzyme because either: depressed gepatic enzymes

    or shunting of blood from portal to systemic venous

    circulations many drugs, particularly those invovled with phase I clearance,

    have decreased clearance in liver disease (but the relationship with severity of liver disease is poor)

    drugs that undergo mainly phase II metabolism do not show impaired clearance unless liver failure is extreme

    also, Vd may be increased for drugs with low Fu when albumin levels are low

    specific examples: see handout for more details o hepatic disease

    cirrhosis theophylline (bronchodilator) - reduced CL (slower fall in

    plasma concentration) propranolol (beta blocker) - F increased (higher peak

    plasma concentration) viral hepatitis

    warfarin (anticoagulant) - CL reduced (excessive response) o cardiovascular disease

    congestive cardiac failure lignocaine (anti-arrhythmic) - CL and V reduced (elevated

    plasma concentration) o renal disease

    uraemia gentamicin (antibiotic) - reduced CL

  • thiopental (anaesthetic) - prolonged anaesthesia (unknown reason)

    o gastrointestinal disease coeliac disease

    ferrous sulphate (haematinic) - absorption reduced (anaemia does not respond)

    Crohn's disease propranolol (beta blocker) - increased plasma binding:

    elevated alpha1-acid glycoprotein o respiratory disease

    emphysema morphine (analgesic) - increased sensitivity to respiratory

    depression (unknown reason) chest infection

    theophylline (bronchodilator) - metabolic CL decreased (elevated plasma concentrations)

    o endocrine disease thyroid disease

    digoxin (cardioactive agent) - altered pharmacodynamics (dimished response in hyperthyroidism; increased response in myxoedema)

    o fever quinine (antimalarial) - impaired metabolism (plasma

    concentration elevated)

  • PHARMACOLOGY LECTURE 14

    THERAPEUTIC DRUG MONITORING

    What is therapeutic drug monitoring (TDM)?

    = measurement of drug in plasma to assist in indivualising dosage regimen for a particular patient (especially when therapeutic effect is not easily measured)

    What is the theoretical basis for TDM?

    major assumption is that there is a direct and predicatble relationship between: o concentration of drug in plasma o unbound concentration of drug in plasma o tissue concentration of drug (inc. concentration at site(s) of action) o pharmacological effect, either efficacy and toxicity

    What is the therapeutic concentration rage?

    = range of concentrations associated with a high degree of efficacy and a low risk of dose related predictable toxicity derived from populations of patients

    note: a small proportion of patients will show excellent responses be;pw the lower limit of the range, and others will derive benefit only a concentration above the range

    When is TDM useful?

    TDM is only useful when there is a known relationship between plasma drug concentration and pharacological response

    for some drugs, the therapeutic response is difficult to measure (e.g. prophylactic agents such as anticonvulstants, cardiac anti-arrhythmic agents)

    some drugs have unpredictable relations between dose and plasma concentration (e.g. dose dependnet or "non linear" kinetic drugs such as phenytoin)

    some drugs have very narrow therapeutic index (e.g. digoxin, aminoglycosides, anticonvulsants, anti-arrhytmic agents, theophylline, lithium, salicylic acid)

    where poor patient compliance is suspected for some drugs, adverse effects of drug may mimic disease state (e.g. digoxin can

    cause arrhythmias, cyclosporin) when there is suspected toxicity when there is reason to suspect there has been a change in pharmacokinetics

    (renal function, hepatic function etc.)

    What do you need to know to intrepret the TDM results?

  • exact knowledge of sampling time of plasma in relation to time of dose administered; in general, it is bets to take a pre-dose (trough) sample, because this is the most easily predictable part of the concentration-time curve (i.e. you know that this will be the minimum value)

    knowledge of the sampling time in relation to "steady state" (how many half lives have elapsed since the dose was started or changed)

    an understanding of the drug's (and the formulation) plasma concentraion versus time profile (e.g. immediate or slow release)

    knowledge of any possible non-linear kinetics of the drug o example: protein binding - valproate, salicylate o example: clearance - phenytoin

    knowledge of patient specific changes, examples include o pregnancy o extremes of age o genetics o renal, hepatic, cardiac disease o hypoalbuminaemia o concomitant drug therapy

    What is the proper clinical role of TDM?

    plasma drug concentrations must always be seen in a clinical context

    CASE 1

    20 year old has had epilepsy since age 12, but been seizure free since age 14 over this 6 year period, he has taken phenytoin (260 mh daily) and

    carrbamazepine (400 mg daily) he attended his family doctor, who ordered TDM of both drugs, giving these

    values: o plasma phenytoin concentration = 24 umol/L (therapeutic range 40-80) o plasma carbamazepine concentration = 12 umol/L (therapeutic range 20-

    50 umol/L) on the basis of these, doctor increased dose of phenytoin to 400 mg daily and

    carbamazepine to 800 mg daily one week later, the patient required admission to hospital because of marked

    drowsiness, ataxia, vomiting, nystagmus plasma concentrations were:

    o phenytoin = 130 umol/L o carbamazepine = 24 umol/L

    COMMENTS o phenytoin- nonlinear kinetics o carbamazepine - linear kinetics o there was no need to increase dosage because he was seizure free; if

    anything, the dose should have been reduced because the doctor should have considered that the epilepsy may have left

  • o note, even if the doses should have been increased, phenytoin dose should have only been increased slightly, because of its non-linear kinetics

    CASE 2

    66 year old man admitted to hospital due to severe breathlessness and swollen ankles over past week

    has been heavy smoker and has ischaemic herat disease and COAD at time of admission, there is evidence of congestive heart failure, and his airways

    disease appears to be no worse than usual he has been on theophylline at home, receiving 800 mg daily on admission, he complains of severe nausea, and shakes plasma theophylline concentration is 39 mg/L (therapeutic range 10-20 mg/L) his heart failure is treated and he improves dose of theophylline is reduced to 400 mg daily on 5th day he becomes breathless, and is found to have extensive bronchospasm,

    but no evidence of heart failure his plasma theophylline concentration is now 6 mh/L COMMENTS

    o theophylline has linear kinetics in this range o hence, halving dose normally would have plasma levels o but perhaps - congestive heart failure --> decreased liver function

    (reduced liver flow and liver congestion) --> decreased theophylline clearance --> theophylline rises during CCF

    o also, smoking induces P450s (maybe he recently began smoking)

    CASE 3

    74 year old woman, admitted for inguinal hernia repair found to have atrial fibrillation (ventricular rate at rest of 105 beats/minute) digoxin is started at a dose of 125 micron daily (with no loading dose) a very conscientious intern orders a serum digoxin concentration on the next day the result if 0.4 nmol/L (therapeutic range 0.6 - 2.3 nmol) COMMENTS

    o should have given loading dose (because her surgery will be soon and there will not be enough time to give for 5 half-lives)

    o not until 1 week will steady state be reached (half life = 24 hours for digoxin)

    o note: 0.4 is a trough level after 1 half life, therefore is too high, therefore should decrease dosage

  • PHARMACOLOGY LECTURE 15

    INTEGRATION OF PHYSIOLOGICAL CONCEPTS AND PHARMACOKINETICS

    "~" means approximately

    CLEARANCE BY THE LIVER

    hepatic extraction ratio (E) o = fraction of dose entering liver from blood which is irreversibly

    eliminated (metabolised) during one pass through the liver o rate of entry = flow x concentration in = Q x C(in) o rate of exit = flow x concentration out = Q x C(out) o rate of elimination = rate of entry - rate of exit = Q(Cin - Cout) o extraction ratio = E = (how much is extracted / how much goes in)

    = (Cin - Cout) / Cin o i.e. E = 1 --> complete extraction, whereas E = 0 --> no extraction o note also, for CL

    rate of elimination = CL x Cin CL = rate of elimination / Cin CL = Q(Cin - Cout) / Cin CL = Q x E (i.e. clearnance is a function of blood flow rate and extraction ratio

    - this applies to any organ) o the hepatic extraction ratio can also be described physiologically by:

    E = (fu x CLint) / (Q + (fu x CLint)) fu = unbound fraction in blood (since only unbound can diffuse

    into liver cells; there are exceptions) CLint = intrinsic clearance of liver (the ability of liver to

    metabolise the drug in the absence of restrictions imposed by blood flow or binding)

    o note: F = 1 - E o i.e. F = Q / (Q + (fu x CLint) o hence, it can be seen that hepatic clearance and bioavailability are

    described in terms of: Q fu CLint

    o there are two limiting cases: low hepatic extraction ratio drugs - when Q >>> fu x CLint

  • i.e. the unbound fraction x the intrinsic clearance is very small (therefore, using logic, bioavailability is very high)

    i.e. F ~ Q / (Q + 0) i.e. F ~ 1

    by the same logic, hepatic CL will be very low, regardless of flow rate

    the clearance of such drugs depends directly on the degree of binding and the activity of the drug's metabolising enzymes

    these drugs have a very low first pass, and bioavailability will be close to 100%

    examples: caffeine ibuprofen

    increasing or decreasing rate of supply by altering Q will make very little difference to hepatic clearance

    also called capacity limited drugs summary: these drugs will have a high bioavailability, and

    clearance will vary depending on hepatic enzyme activity and plasma binding

    high hepatic extraction ratio drugs - when Q

  • SUMMARY: bioavailability and first pass effect o F = fraction absorbed x (1 - E) o some drugs have low E (e.g. caffeine) --> high F (a change in E therefore

    has a minor effect on F) o some drugs have high E (e.g. morphine) --> low F (a change in E therefore

    has a major effect on F) o the major determinant of oral bioavailability of high extraction drugs is

    hepatic enzyme activity EXERCISE: determine for some low and high extraction drugs, the effect of

    changes in drug binding on o clearance o volume of distribution o half life o bioavailability o unbound stead state plasma concentrations on chronic dosing (IV and oral)

    APPENDIX o variability in response - for high extraction drugs, a small change in E will

    lead to a large change in F, leading to more variability in plasma concentrations after oral vs IV doses

    o dosage (oral vs. IV) - for a drug with high hepatic extraction, an oral dose will need to be much higher than an IV dose to elicit the plasma concentrations

    o other routes - to avoid some first pass effect, use sublingual, transdermal, inhalation, rectal

    o drug interactions - other drugs may induce or inhibit hepatic enzymes, and so for high extraction drugs, this can lead to significant changes in bioavailability

    o liver disease - if shunts exist because of liver disease, bioavailability will increase because blood flow through liver will be less; therefore, for high extraction drugs given to patients with liver disease, there is the potenital for increases adverse effects

  • PHARMACOLOGY LECTURE 16

    PHARMACOGENETICS

    genetic polymorphism: trait determined by a single gene, occurring in normal population in a least two phenotypes, neither of which is rare (i.e. at least 1%)

    extensive metabolisers - these people metabolise the drug normally poor metabolisers - these people do not metabolise the drug at the same rate as

    extensive metabolisers

    GENETICALLY DETERMINED VARIATION IN PHARMACOKINETICS

    Debrisoquine-type polymorphism of oxidative metabolism

    4. History o was observed that excretion of unchanged debrisoquine in urine was very

    variable o extensive metabolisers could be heterzygos or homozygous for the normal

    allele o poor metaboliser phenotype occurs in 10% of Caucastion population;

    lower frequency in other populations 5. Metabolic defect

    o impaired drug hydroxylation by CYP 2D6 o no enzyme protein is detectable in homozygotes o detection:

    phenotype - enzyme function tested by ratio of parent drug to hydroxylated metabolite in urine after standard dose of drug

    genotype - gene sequences 6. Clinical relevance

    o poor metabolisers have impaired metabolism of a number of drugs, therefore:

    0. more prone to adverse drug reactions if given standard doses 1. reduces drug effect if metabolite is active (e.g. codeine -->

    morphine) o there seems to be a possible link with other diseases

    poor metabolisers have lower rate of cancers of lung, liver and gastrointestinal tract

    poor metabolisers may have a higher rate of Parkinson's disease 6. Other drugs of clinical relevance (common examples only; there are > 50)

    o beta-adrenoceptor antagonists, e.g. metoprolol o tricyclic antidepressants, e.g. amitriptyline o anti-anginal vasodilator - perhexiline (can have bery severe adverse effects

    including irreversible peripheral neuropathy)

  • Polymorphism of drug acetylation

    2. History o observed bimodal distribution of plasma concentration after standard dose

    of isoniazid (used to treat TB) --> higher risk of peripheral neuropathy o slow acetylators are homozygous for mutant allele

    2. Metabolic defect o reduced activity of hepatic N-acetyltransferase o detection:

    phenytype by measuring plasma concentration of isoniazid or caffeine after standard dose (slow acetylators have higher concentration)

    frequency in different populations varies widely: about 10% in Eskimos, 20% in Asians, 50% in Caucasions, 80% in Egyptians

    2. Clinical relevance o reduced clearance and increased risk of adverse effects from drugs which

    are usually acetylated o epidemiological associated with other diseases:

    slow acetylators - bladder cancer, Gilbert's disease (gemetic problem in liver), SLE (autoimmune against various organs)

    fast acetylators - breast cancer, diabetes mellitus 2. Some examples of drugs of clinical relevance (slow acetylators at ncreased risk of

    ADR) o vasodilators - hydralazine --> lupus-like syndrome (auto-immune disease) o anti-arrhythmic - procainamide --> lupus-like syndrome o some benzodiazepines - e.g. nitrazepam --> sedation

    Atypical plasma pseudocholinesterase (a rare genetic trait)

    4. Metabolic defect o there are many variants of plasma pseudocholinesterase (each under

    separate genetic control) these have much lower than normal affinity for succinylcholine

    (muscle relaxant used in general anaesthesia) people with variant enzyme have much longer duration of action

    following usual doses (50-100 mg) of the drug (hours vs. minutes) o enzyme activity can be tested in vitro, but in practice, most cases are

    detected following anaesthetia o incidence in Caucasians is about 0.05%; not found in Japanese, Eskimos

    or South American Indians 4. Clinical relevance

    o succinylcholine produces muscle paralysis (including respiratory muscles) and is often used during induction of anaesthesia to allow tracheal intubation

  • o effect usually lasts only a few minutes, but may last for hours in patients with atypical cholinesterase - these patients wake up paralysed and require prolonged mechanical ventilation

    GENETICALLY DETERMINED VARIATION IN PHARMACODYNAMICS

    Drug induced haemolytic anaemia (glucose-6-phosphate dehydrogenase deficiency)

    2. Clinical problem o sometimes there is an unexpected occurrence of haemolytic anaemia after

    treatment with oxidant drugs o such drugs can produce haemolytic crises in about 20% of males in some

    populations 2. Mechanism

    o normally, glutathione is involved in the mopping up of free radicals (it can chemically detoxify H2O2)

    o in this disorder, haemolysis caused because there is less reduced glutathione (G-SH) within red cell due to a deficiency of enzyme G6PD

    o lack of reduced glutathione makes cell vulnerable to oxidation, with damage to cell membrane and lysis; therefore drugs that in someway increase this oxidation can lead to haemolytic anaemias

    o G6PD occurs in many different abnormal variants and genetic transmission is X-linked

    o (note heterozygotes are more resistant to malaria than homozygotes of normal allele)

    o about 200 million people are affected world-wide 2. Some drugs involved (should be avoided in people with G6PD deficiency)

    o urinary antiseptic - nitrofurantoin o anti-malarial drugs - primaquine, chloroquine o analgesics - aspirin

  • PHARMACOLOGY LECTURE 17

    INTRODUCTION TO TOXICOLOGY

    BIOACTIVATION OF FOREIGN COMPOUNDS TO TOXIC METABOLITES

    normally, phase I and II reactions dimish biological properties but, sometimes bioactivation occurs and products are more toxic toxic metabolites cause cellular dysfunction by reacting with components such as

    DNA or proteins note that toxicity only occurs in organs that possess the necessary enzymes that

    cause the bioactivation (unless the metabolites are stable enough to migrate to other organs via the blood)

    hence most toxic drugs tend to preferentially affect a limited numbers of organs if the metabolite causes DNA damage, then the patient is at a greater risk of

    cancer general outcomes:

    o cleared via urine/bile

    DNA damage --> cancer protein and cell damage --> organ pathology

    CHEMICALLY INDUCED ORGAN DAMAGE: THE LIVER AS A TARGET

    liver is vulnerable to damage by ingested chemicals because: o of its close proximity to blood supply from digestive dract o it actively concentrates foreign chemicals o it plays a major role in biotransformation

    a number of toxic responses are known within the liver, including hypersensitivity reactions (e.g. drug induced hepatitis), cirrhosis, intrahepatic cholestasis and necrosis

    PARACETAMOL-INDUCED HEPATOTOXICITY o is an intrinsic hepatotoxicant, i.e. produces a predictable, dose-dependent

    toxic response o is very safe at therapeutic doses o normally:

    90% is metabolised via phase II pathways (sulphation and glucuronidation)

    10% is oxidised via phase I (hepatic CYP2E1, CYP1A2, CYP3A4) to a chemically reactive quinoneimine toxic metabolite (NAPQI), which reacts with glutathione to form a non-toxic metabolite which is excreted in the urine

  • o biochemistry under overdose conditions: conjugative phase II metabolism is saturated and used up, and

    therefore NAPQI overproduction occurs this results in depletion of glutathione in absence of glutathione, NAPQI reacts with nucleophilic sites on

    critical cellular proteins, interfering with cell function and initiating hepatocellular necrosis

    o symptoms, signs, tests and treatment of paracetamol-induced hepatotoxicity:

    symptoms: early symptoms of poisoning include nausea and vomiting

    (requires 10-30 g in adults) some patients progress to fulminant hepatic necrosis,

    involving progressive jaundice with hepatic encephalopathy (headache, drowsiness, confusion are early signs of the rapid onset of cerebral oedema [which requires CNS monitoring])

    signs: liver tenderness may appear afer 12 hours, and may persist

    for up to 72 hours serum enzymes:

    hepatic necrosis is evident after 3 days when huge (up to 500 fold) elevations in serum hepatic enzymes occur (e.g. aspartate and alanine transaminases)

    serum enzymes peak on day 3, and return to normal after 7-21 days in survivors

    clotting: liver toxicity is accompanied by impaired hepatic

    production of clotting factors (esp. III, V, VII), increasing the prothrombin time and prothrombin time ratio

    treatment: thiol therapy (oral or IV N-acetylcysteine [NAC])

    ameliorates toxicity by replenishing hepatic glutathione stores, facilitating NAPQI detoxification

    NAC therapy is most effective if administered within 8 10 hours of ingestion

    effective management requires knowledge of plasma paracetamol concentrations

    note that anaphylaxis occurs in 5% of IV NAC recipients o see diagram on sheet

    CHEMICALLY INDUCED ORGAN DAMAGE: THE BRAIN AS A TARGET

    blod brain barrier protects brain against toxic ionised and bulky water soluble agents

  • but, some toxicants can penetrate CNS, and can lead to diffuse encephalopathy with global or dysfunction or a localised encephalopathy

    MTTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) o in California in 1983, unusual motor defecits resembling Parkinson's

    disease were seen in drug addicts using MPPP, a designer drug analogue of meperidine opioid

    o symptoms: developed within hours to days included difficulty in initiating and terminating movement, resting

    tremors, rigidity (which sometimes progressed to immobility) o biochemistry

    toxicity was due to a synthetic by-product in the drug, MPTP 0. MPTP crosses blood brain barrier 0. is activated by monoamine oxidase B in astrocytes to

    MPP+ which is a toxic metabolite (note that MPP+ couldn't enter CNS, and therefore only the MPP+ produced in the