Pharma1922 Lectures Introduction to Pharmacokinetics (PDF)
Document Details
Uploaded by ToughAqua3494
Tags
Summary
These lecture notes provide an introduction to pharmacokinetics (PK) and pharmacodynamics (PD). The document covers key aspects of drug action, focusing on how the body processes drugs and how drugs affect the body. It discusses factors influencing drug response.
Full Transcript
Introduction to Pharmacokinetics (PK) and Pharmacodynamics (PD) Pharmacokinetics - What the body does to the drug, the study of drug absorption, distribution, metabolism and excretion Pharmacodynamics - What the drug does to the body, biochemical and physiological effects...
Introduction to Pharmacokinetics (PK) and Pharmacodynamics (PD) Pharmacokinetics - What the body does to the drug, the study of drug absorption, distribution, metabolism and excretion Pharmacodynamics - What the drug does to the body, biochemical and physiological effects of drugs, relationship between drug concentration and drug effect - Drug action Impact of Different Routes of Administration on Drug Disposition and Action - Different routes of administration and dosage form lead to different o Concentration- time profiles o Onset of action o Duration of therapy o Location of effect o Ease of administration o Cost - Is dependent on o Solubility o Stability o Availability - Action o Both biochemical and physiological effects o Dependent on drug reaching drug receptors Role of Pharmacokinetics and Pharmacodynamics in Drug Development and Medicines Regulation - Pharmacodynamics and pharmacodynamics are the main considerations in drug development - A drug must be safe (PK), effective (PD), non-toxic, be soluble etc. - Pharmacokinetics are the biggest reason why drugs do not make it to the market Affecting Drug Response - Pregnancy - Genetics - Obesity - Disease - Age - Drug interactions - Environmental factors - Drug adherence Absorption, Distribution, Metabolism and Elimination (Pharmacokinetics) Absorption - The process where a drug enters the bloodstream - Bioavailability is the measure of absorption, how much of the given drug makes it to the bloodstream, (a fraction) - Drug formulation, route of administration etc. can impact absorption - Incomplete or erratic absorption can lead to variability in drug effects Distribution - The process where a drug is spread across the body - The drug may go to certain organs, or bind to proteins in the blood serum - The volume of distribution (Vd), is a parameter used to measure the extent of distribution - Variations in tissue distribution, blood flow etc. can lead to a different pharmacological effect Metabolism - The process where a drug is chemically altered to make it more water soluble, facilitating elimination - Primarily done in the liver where enzymes like cytochrome P450 play a big role - Genetic variance in such drug metabolising enzymes can alter how an individual metabolises drugs - Can convert an active drug to an inactive form or produce active metabolites, which may have good or bad effects - A drug’s half life plays a part in metabolism Elimination - The process where a drug or its metabolites are removed from the body - Done mainly through the kidney (urine) or liver (bile and ultimately stool) - People with liver/kidney impairment fail to effectively eliminate drugs, meaning they may have drug accumulation, leading to an increased risk of toxicity - The rate of elimination is described by the drug’s half life - Clearance (Cl); volume per time, DOSE/AUC How Pharmacokinetics Informs the Use of Different Brands of a Medicine - If two drugs achieve the same drug concentration in the body they are considered bioequivalent - Absorption rate (Cmax) and the extent of drug absorption (AUC) are the main contributors to bioequivalence Pharmacokinetics Pharmacokinetics - Examines how drug concentration in different body parts is affected by route of administration, dosage form - Important in drug development, accumulation, toxicity, distribution, etc. - Has drug safety applications, therapeutic index (TI); toxic dose/ effective dose. Key Pharmacokinetic Parameters - Clearance (L/h) (elimination/excretion) o Efficiency of elimination of a drug o The irreversible elimination from circulation o Volume of blood cleared per unit time o Cl (L/h) = dose (mg) / AUC (mg*h/L) o Elimination rate (mg/h) = Cl total (L/h) * [Drug] (mg/L) - Volume of distribution (L,L/kg) (distribution) o The extent of distribution, not real o The amount of drug in the body to the plasma concentration o Back calculated from initial drug concentration plasma C0 following given IV dose o V = dose (mg) / C0 (mg/L) o Lipophilic drugs can traverse membranes ▪ Apparent Vd greater than 3L o Hydrophilic drugs are trapped in the plasma ▪ Apparent Vd approximately 3L - Half-Life (metabolism) o Time to halve the amount of drug in the body o Determined by clearance and volume of distribution o K = Cl/ Vd - Bioavailability (absorption) o Fraction of drug that gets absorbed o Compared against IV infusion which is 100% bioavailable o F= AUC/100 - C max (concentration max), T max (time at concentration max), AUC (area under curve) o Calculate AUC using trapezoidal rule, Area = (t2 – t1) x c2-c1 / 2 Single Oral Dose Plasma-Time Curve Rises when absorption is higher than elimination Cmax= when they are equal Falls when elimination is higher than absorption Single IV Dose Plasma-Time Curve First Order Kinetics Zero Order Kinetics When a constant proportion of the drug is eliminated When a constant amount of the drug is eliminated One Compartment Pharmacokinetics - Treat the body like one compartment (blood) - Curve has 1 phase Two Compartment Pharmacokinetics - Treat the body like two compartments - Second peripheral compartment (e.g. tissue) - Drug may not always distribute evenly and instantaneously throughout the body - May distribute unevenly across different tissues - Curve has 2 phases Three Compartment Pharmacokinetics - Third compartment may be bone Population PK Analysis - Doing blood tests on different people at different times and combining their data to 1 population - E.g. weight, sex, age, frailty, pregnancy - Women clear aspirin better than men Absorption Drug Movement Across Membranes - Drugs must cross cell membranes to be absorbed - Cell membranes have hydrophilic heads and hydrophobic tails Characteristics That Facilitate Movement - pH - Ionisation - Surface area LogP vs LogD - LogP describes a drug’s lipophilicity and predicts movement in cell membranes - P = [compound] organic/ [compound] aqueous - LogD also describes a drug’s lipophilicity - Aqueous phase is adjusted to a specific pH Transporters Involved in Drug Disposition - Peptides - Amino acids - Glucose - Aqueous diffusion – simple diffusion, concentration gradient - Lipid diffusion - Intestinal epithelia - Hepatocytes (liver) - Kidney proximal tubules - BBB Plasma Protein Binding - After absorption, some drugs are bound to proteins in the blood (plasma) - Proteins involved in the binding of drugs include o Albumin o Alpha-1-acid glycoprotein o Some lipoproteins - Only free drug can be distributed to tissues (drugs bound to protein cannot cross the cell membrane) Blood Brain Barrier - BBB o Endothelial cells of the capillaries o Basement membrane is continuous o Limited number of small, aqueous pores - Basically a lipid barrier - Lipid soluble drugs can dissolve in the membrane and enter the brain by passive diffusion - Water soluble drugs cannot enter the brain - BBB represents significant barrier to development of CNS drugs First Pass Metabolism - When a drug is swallowed it is absorbed from the stomach/ small intestine - Enters portal circulation and carried via portal vein to liver - Liver is the major organ for drug metabolism - If a drug has a high first pass metabolism, a higher dose is required - Avoiding first pass metabolism is considered a good thing - Sublingual, rectal, transdermal, injections avoid first pass metabolism, such routes of administration are absorbed into the veins which drain into the heart without entering the portal system (liver) Routes of Administration Oral - Convenient, relatively safe, no need for sterility - Affected by o Gut content (fed vs fasted) o Gastrointestinal motility o Splanchnic blood flow o Particle size and formulation Sublingual (under the tongue) - Convenient, relatively safe, no need for sterility, avoids first pass metabolism and acid/enzymes in the stomach, tastes important Eye Drops - Must be sterile, localised treatment e.g. eye infection, glaucoma Injection - Must be sterile, avoids first pass metabolism, faster onset of action, difficult to administer, adverse reactions, can be subcutaneous, intramuscular, intravenous Distribution Drug Distribution - For a drug to have any effect, it must be distributed to a site of action - Most drugs are distributed via passive diffusion - Lipophilic drugs can traverse membranes - Hydrophilic drugs are trapped in the plasma - Factors that affect drug distribution are o logP, between 0 and 5 is good o Molecular weight, less than 500 is good o Water solubility o Lipid solubility o Molecular size of water-soluble drugs o Whether the drug is carried by transporters o Protein binding o Blood flow to the tissue o Specialised barriers within the body e.g. BBB Volume of Distribution (Vd) - Body water is divided into 3 distinct groups o Plasma (3L) o Interstitial (9L) o Intracellular (28L) - Lipophilic drugs can traverse membranes (Vd>3L) - Hydrophilic drugs are trapped in the plasma (Vd=3L) Tissue Affinity - Some drugs accumulate in certain tissues or cell types for which they have affinity - Drugs with an affinity for plasma proteins remain in the blood longer - Drugs with an affinity for melanin remain in pigmented tissues for longer etc. Metabolism Drug Metabolism - Increases the water solubility of a drug facilitating elimination - The liver is the main drug metabolising organ - Occurs via faeces or urine - While it facilitates elimination it may also increase toxicity or pharmacological effect - Metabolism results in metabolites, they are less active and more water soluble (polar) than the parent compound Phase I Metabolism Reactions - Catabolic - E.g., oxidation, reduction and hydrolysis - Occurs mainly in the liver - It is the chemical conversion of a chemical into something more water soluble (polar) - Cytochrome P450 enzymes are the main enzymes involved in phase I reactions, CYP450 - Different CYPs metabolise different drugs - Sometimes the drug is metabolised to be more toxic, sometimes to be more pharmacological - Some drugs are inactive until they have been metabolised - E.g. codeine o Converted to morphine by CYP2D6 o Genetic polymorphisms exist, some will have poor analgesic effect, others will have opioid effect o Dependent on race, genetics Phase II Metabolism Reactions - Anabolic/ conjugation - Making the molecule bigger and more polar; more likely to be excreted - Polar group is attached to the drug by a “handle” - E.g., glucuronidation, sulphation, glutathione, acetylation - Glucuronidation o Most common phase II o Catalysed by UGTs - Glutathione o In the liver o Non-enzyme o For paracetamol metabolism, paracetamol overdose Excretion and Elimination Renal Elimination/Excretion - Glomerular filtration o Removal of free drug at the glomerulus o Important for most drugs o Only molecules small enough will pass through filtration slit - Active tubular secretion o Energy dependent process by which drug is transported into urine o In proximal tubule o Subject to competition by drugs with same charge o High elimination rate o E.g. penicillin - Tubular reabsorption o Passive process in which drug in urine diffuses back into blood o Drug must be uncharged o Drug gets concentrated in urine, then follows gradient back out into blood - Affected by pH of the urinary matrix, you can alter your own pH o Ammonium chloride for acidification o Sodium bicarbonate for alkalinisation - If drug is unionised, it can cross cell membranes and will be reabsorbed - If drug is ionised, it will be trapped in filtrate and excreted in urine - Fraction unchanged (fu), an fu of 1 implies a drug which has been totally cleared, renally unchanged ▪ Fu = renal clearance / total clearance - Estimate renal clearance through urine samples and noncompartmental estimation Faecal Elimination - Drugs appear in faeces by two major mechanisms o By not being absorbed into systemic circulation o By being absorbed, excreted in bile and deposited back into small intestine - Bile o Excretes cholesterol o Absorbs lipids o Stimulates intestinal motility o Produced in liver and stored in gall bladder Enterohepatic Recycling - Reabsorption of drug or metabolite, bile excreted and reabsorbed by small intestine, returned to liver - May produce a secondary therapeutic effect Pulmonary Elimination - Elimination of gases via the lungs - E.g. alcohol Breast Milk Elimination - Small unionised drugs Sweat - THC, cocaine, morphine Hair - Amphetamines Saliva - Antiepileptics, amphetamines Pharmacodynamics Pharmacodynamics is what the drug does to the body - The relationship between drug concentration and effect - Drugs are also called; ligand Drug Actions - A drug must bind to one or more cell constituents to produce a pharmacological response - The drug and its binding site must be complimentary o Charge attracts the drug to binding site o Active site is called the orthosteric binding site - These sites include o Receptors ▪ Protein sensors for chemical communication ▪ Macromolecular complex which binds to a drug with high specificity o Ion Channels ▪ Protein molecules that permit flow of specific ions along their concentration gradient Target Mechanism ▪ Two types Receptor Agonist or Antagonist Ligand-gated ion channels Ion Channels Blocker or modulator Voltage-gated ion channels Transporter Uptake inhibitors ▪ Either blockers or modulators Enzymes Inhibitor o Transporters ▪ Aka carrier molecules ▪ Allow the movement of ions against their concentration gradient o Enzymes ▪ Decreases the energy required for a chemical reaction - Once the drug has bound, the effect on a particular system can be: o Activating e.g. agonist o Enhancing e.g. positive modulator o Attenuating e.g. negative modulator or inverse agonist o Interfering e.g. antagonist: no effect on receptor itself but lowers response to other agents Orthosteric and Allosteric - Orthosteric binding site o Recognition site of the endogenous molecule on the receptor, agonists and antagonists bind here - Allosteric binding site o The “other” binding site on the receptor, modulators bind here Drug Binding - Generally, the bonds between a drug target and receptor are weak and reversible o Ionic bonds o Hydrogen bonds o Dipole o Van der Waals force o Sometimes covalent Affinity - Refers to the drugs ability to bind to the target, strength of relationship between ligand and cell constituents - Quantified as concentration of drug required to occupy 50% of target proteins o Drugs with higher affinity only need a very small concentration to bind to the target. - A drug can have affinity for more than one target, it can bind to more than just one type of protein o But it can only bind to those targets for which it has affinity Selectivity - Ability of a given drug concentration to produce one effect over another (e.g. therapeutic effect over side effect) - For a drug to be effective it must act selectively at a particular target, no drug is entirely specific to a target, hence why we get side effects - Since drugs are likely to have higher affinity for some targets compared with other, we can choose a concentration of the drug that binds to one target but not others, is selective for a particular target over other targets at the concentration chosen - Selectivity is concentration-dependent, as you increase concentration you increase the probability for the drug to bind to other sites Intrinsic Efficacy - Measure of the ability of a drug to elicit a response - Defined as the maximal effect a drug can produce on a specific tissue, expressed as a proportion of the maximal effect of a full agonist on that tissue o Full agonists = intrinsic efficacy = 1 o Partial agonists = intrinsic efficacy = 0 to 1 o Antagonists = intrinsic efficacy = 0 o Inverse Agonists = intrinsic efficacy = Raf -> MAP - Jak/Stat pathway o Involved in response to cytokines o Phosphorylated stat migrates to nucleus and activates gene expression o Jak -> stat - Kinase inhibitors are drugs that stop phosphorylation occurring, helpful in cancer o 7 different types o Type I, interacts with active conformation of kinase, e.g. crizitonib o Type II, targets DFG loop (inactive), e.g. imatinib o Type III, bind to a site in the ADP pocket (deep), e.g. trametinib o Type IV, substrate directed inhibitors (allosteric), e.g. everolimus o Type V, bivalent irreversible inhibitors (allosteric), e.g. N/A o Type VI, covalent binders (permanent), e.g afatinib o Type VII, nonclassical allosteric inhibitors, e.g. SSR128129 Corticosteroids (adrenal system) - Hormones produced by the adrenal system - Produced as drugs for their anti-inflammatory and immunosuppressive properties - Adrenal system (two systems) o Inner Medulla ▪ Catecholamines e.g. dopamine o Outer Cortex ▪ Aldosterone ▪ Testosterone ▪ Glucocorticoids - Glucocorticoids o Carbohydrate and protein metabolism ▪ Increase glucose production ▪ Increase glycogen storage ▪ Decrease uptake and utilisation of glucose ▪ Decrease protein synthesis = increased protein breakdown = tissue wasting ▪ Reduces calcium absorption in GI tract = osteoporosis o Anti-inflammatory actions ▪ Decreases release of neutrophils from blood vessels ▪ Decreases activation of T-helper cells o Too much glucocorticoids = Cushing’s syndrome o Too much mineralocorticoid =Conn’s syndrome o Not enough glucocorticoids = Addison’s disease ▪ Glucocorticoid replacement therapy Cancer - Uncontrolled cell growth - Caused by mutations in DNA which control cell growth and division o Grows in absence of signals o Ignores signals for stopping division or apoptosis - Genes affected o Proto-oncogenes o Tumour suppressor genes o DNA repair genes - Treatments o Cytotoxic drugs ▪ E.g platinum o Kinase inhibitors ▪ E.g. imatinib o Monoclonal Antibodies ▪ E.g. rituximab o Immune Checkpoint Inhibitors ▪ New and upcoming ▪ Tricks the immune system into turning itself off (make cancer cells kill themselves) Overview of Preclinical Pharmacology Pre-Clinical Pharmacology - Drug Discovery o Chemical synthesis, formulations, identifying the lead drug - Pre-Clinical in vitro testing o Molecular, cellular or organ testing - Pre-Clinical In vivo testing o Testing in animals Core Framework Pharmacodynamic Profiling - Done in vitro or in silico - Requires good understanding of normal physiology or pathology of the drug’s function - E.g. o Testing organs in organ baths o Bruton tyrosine kinase inhibitor – cancer drug - Following a positive result in vitro, we move onto animal testing (in vivo) o Does the drug do what it did in vitro o Does the drug reach the target o Do we use a healthy/sick animal, old/young etc. o A lot of considerations ▪ Mode of Action ▪ Selectivity ▪ Specificity Pharmacokinetic Profiling - Considerations o Toxicity, safety, dosing regimens, route of administration, starting dose o Must think about ADME Drug Regulation Regulatory System in Australia - Since 1958 there has been regulation in Australia - TGA is currently the biggest player, updated regularly since its inception 1989 - Since the 1990s the biggest changes have been to allow for more streamline drug approval - Therapeutics Good Act o Underpinned by regulations, orders, laws, guidances and standards o Provide a national framework for the regulation of therapeutic goods so as to ensure their quality, safety, efficacy and timely availability o Established the TGA o Established the ARTG o Applies to all therapeutic goods o Licensing of medicines o Importing of medicines etc. o Pre market evaluation o Post market surveillance/ maintenance o Risk based approach to regulation Therapeutic Goods - A good which is represented in any way to be, or likely to be taken to be, for therapeutic use o Prescription medicines o OTC medicines o Complementary medicines o Sunscreen o Medical devices o Biologicals o Blood and blood components - All therapeutic goods must be on the ARTG to be supplied in, imported to or exported from Australia Pre-Market Evaluation - Rigorous trialling, Phase I to IV etc., classification/scheduling, dossier, ARTG approval, market - Engagement between authorities and drug companies Post Market Product Maintenance - Continuous monitoring for identification of safety issues - An effective system to report any safety concerns - TGA checks to ensure ongoing adherence to safety standards Future Directions of Drug Design and Discovery Cryogenic Electron Microscopy (Cryo-EM) - Allows for the determination of large protein structures, specifically membrane proteins - Method o A sample of the membrane is embedded in a “nano disc”, which acts as the cell membrane o The chamber is cooled down to 4K o The membrane is coated in a very thin layer of ice o Electron beams bombard the membrane o The results are collected and a 3D map of the protein is made - Protein structures of full length are produced, enables visualisation of antibodies which was previously difficult Molecular Dynamics - Simulation of atoms over a time, not just a picture - Considerations o Bonded energy ▪ Covalent bonds ▪ Bond angle, length o Non-bonded energy ▪ Electrostatics ▪ Van der Waals o Temperature o Pressure - Method o Minimise energy o Equilibrate the system o Simulation, on GPU - Used for o Ligand binding affinity ▪ Docking score o Binding site determination ▪ Best binding site Artificial Intelligence (Alphafold) - The way that proteins are built follow fundamental rules of nature, if AI knows this it can predict protein structure - First alphafold was poor, alphafold 2 was good - Method o Takes protein sequence from databases o Develop a multiple sequence alignment o Data is processed and informs bond angles o A “relaxed” 3D image is produced - Used for o Proteins that are impossible to characterise in vitro - Limitations o Cannot predict ligand and apo states o Cannot predict DNA or RNA o Struggles to predict whether ion channels are opened or closed o Need supercomputers Organ on a Chip (OoC) - Tissue models based in microfluidic chips, mimic human physiology - Include tiny channels which allow for the uptake of small volumes - Drugs or other compounds can be added to test the response from cells Personalised Medicine - Using a person’s genetic information to inform prevention, diagnosis and treatment of disease - E.g. o Imatinib ▪ Patients are screened for a mutation, if they have it, they are given imatinib treatment, cancer Safety and Toxicology Drug Toxicity Categories - Pharmacophore based toxicity o Primary pharmacology of the agent o What does it actually do - Chemotype based toxicity o Structural features or physiochemical properties o Noncovalent bonding interactions - Metabolism based toxicity o Some drugs harbour toxic metabolites Safety Studies to Detect Toxicity in Pre-Clinical Phase - Single Dose Toxicity o 2 mammalian species, at least 1 non rodent o One route is the proposed clinical route, second route is parenteral o Dose should exceed the anticipated clinical dose, main source of PK data - Repeat Dose Toxicity o Detect longer term chronic effects o Generally, just the proposed clinical route o Dose should be up to maximum tolerated dose - Genotoxicity o Bacterial mutation (Ames) test, in vitro and in vivo o OR o Bacterial mutation (Ames) test, in vivo test for genotoxicity with two different tissues - Carcinogenicity o Rat or mouse o Given drug daily, orally at differing doses o Animal is examined for weight gain, clinical signs, tumour incidence - Reproductive toxicity - Safety pharmacology - Pharmacokinetics studies Reasons for Withdrawal from Market - Hepatic; hepatic failure, liver injury - Cardiovascular; stroke, arrythmia - Neurological; neurotoxicity, depression - Renal; renal failure - Etc. etc. Adverse Drug Reactions Augmented Adverse Drug Reactions (Type A) - Dose related - E.g. digoxin o Narrow therapeutic index o Administered orally or IV o Absorption from stomach and upper small intestine (80%) o 20% bound to plasma proteins ▪ Symptoms Lethargy Confusion GI symptoms Impaired vision Cardiac arrythmias o Digoxin toxicity is prevented by tailoring dosage to ▪ Renal function ▪ Clinical response ▪ Concentration monitoring o Digoxin toxicity is treated by ▪ Antidote (digifab) Bizarre Adverse Drug Reactions (Type B) - Not dose related - Hypersensitivity reactions - Type I o Immediate and rare o E.g. anaphylaxis, hives - Type IV o Delayed and more common o E.g. rashes, Steven Johnson Syndrome - E.g. sulphonamide; o fever, dyspnoea, cough, rash, Steven Johnson Syndrome Chronic Adverse Drug Reactions (Type C) - Dose and time related, using too much for too long - E.g. corticosteroids o Used for patients with adrenal failure o Anti-inflammatory/immunosuppressive therapy o Adverse reactions ▪ Adrenal suppression due to suppression of corticotrophin sectretion ▪ Oral candidiasis due to weakening the immune system in the mouth and throat Delayed Adverse Drug Reactions (Type D) - Time related, take long time to manifest - Teratogenesis (causes birth defects) o E.g. ▪ Thalidomide Originally a tranquiliser Marketed as reducing anxiety, tension and morning sickness Never tested on pregnant women Children had horrible birth defects - Carcinogenesis (cancer) o E.g. ▪ Smoking End of Use Adverse Drug Reactions (Type E) - Abrupt cessation of drug where there is physical dependence may lead to withdrawal syndrome - Dependence comes from repeated administration of a drug - Physical dependence- when withdrawal of the drug will cause adverse physiological effects - E.g. o Opioid withdrawal; crying, sweating, yawning, anxiety, restlessness, GI issues o Benzodiazepine withdrawal; anxiety, dysphoria, irritability, sweating, insomnia o Alcohol withdrawal; - Withdrawal is managed by medical (substituting drugs) and psychological care (behaviour management) Failure of Therapy Adverse Drug Reactions (Type F) - Failure of therapy due to insufficient drug o Enzyme inducers decreasing exposure to therapy o Prevention of absorption from gut o Overly cautious administration of chemotherapy - E.g. contraceptive pill o Lower dose meant less side effects o CYP3A4 – increases rate of metabolism – reduces plasma concentration of pill – reduces efficacy – pregnancy Clinical Trials Methodological Approaches to Understanding the Efficacy and Toxicity of New Drugs in Humans - Pre-clinical Analysis o Confirm good PK properties o Confirm mechanism of action in vitro and in vivo, identify biomarkers of response - Safety Analysis o Complete full toxicological assessment in 2-3 animal species o NOAEL calculations in 2-3 animal species o Determine safety factor o Calculate MRSD Clinical Trials - Intervention studies (I,II,III and IV) OR descriptive studies (case reports/studies, cohort studies) - Confirm absence of toxicity and establish efficacy - Identify Type A (augmented) adverse reactions – dose related - They are either o Preventive ▪ Where patients are disease free ▪ Does drug reduce risk of developing disease ▪ E.g. IBIS II trial – breast cancer prevention o Therapeutic ▪ Where patients have disease ▪ Dose drug reduce mortality due to disease ▪ E.g. salmeterol reduce bronchoconstriction in asthmatic episodes Phase I – will it harm the patient – safety - Testing an experimental drug on sick patients or healthy volunteers, up to 100 people - Open label, single arm study - Testing different doses sequentially - Identify toxicity, side effects, safe and effective dosage, pharmacokinetics, route of administration (oral, IV, inhalation etc.), dosage frequency, mechanism of action - Informs phase II - 50% of drugs have failed by now – not safe Phase II – will it make the patient better – safety + efficacy - Testing effectiveness on sick patients, up to 1000 patients - Open label, single arm study OR double blind, randomised, placebo trial - Measure treatment response, beneficial physiological change - Identify a confirmed efficacy, - Informs phase III - 66% of drugs have failed by now – do not do what they need to do Phase III – is it better than existing treatment - efficacy vs. existing treatment - Testing if it is better than existing treatments, 1,000 to 10,000 people with defined condition - Double blind, randomised clinical trial, drug vs existing drug vs placebo - It is randomised to reduce bias, random error, population variability - Identify that mechanism of action matches phase II data, no type A ADRs, better and/or safer than existing drug - Informs phase IV - 75% of drugs have failed by now Phase IV – does it work in the community – safety, efficacy - >10,000 people with the disease are given the drug in off label prescribing - Usually after the drug is approved - Further evaluation of long-term safety and effectiveness, serious ADRs - Post approval safety - Most of such systems require voluntary reporting,