Pharmacokinetics. Karim Rafaat

Similar documents
It the process by which a drug reversibly leaves blood and enter interstitium (extracellular fluid) and/ or cells of tissues.

Pharmacokinetics of Drugs. Assistant Prof. Dr. Najlaa Saadi PhD Pharmacology Faculty of Pharmacy University of Philadelphia

INTRODUCTION TO PHARMACOKINETICS

Industrial Toxicology

Unit 2b: EXCRETION OF DRUGS. Ms.M.Gayathri Mpharm (PhD) Department of Pharmaceutics Krishna Teja Pharmacy college Subject code: 15R00603 (BPPK)

Drug Distribution. Joseph K. Ritter, Ph.D., Assoc. Prof. Medical Sciences Building, Room

ADME Review. Dr. Joe Ritter Associate Professor of Pharmacology

Pharmacokinetics I. Dr. M.Mothilal Assistant professor

Excretion of Drugs. Prof. Hanan Hagar Pharmacology Unit Medical College

Principles of Drug Action. Intro to Pharmacology: Principles of Courework Drug Action Intro to Pharmacology

Basic Concepts in Pharmacokinetics. Leon Aarons Manchester Pharmacy School University of Manchester

Introduction to. Pharmacokinetics. University of Hawai i Hilo Pre-Nursing Program NURS 203 General Pharmacology Danita Narciso Pharm D

Chapter 4. Drug Biotransformation

Slide 1. Slide 2. Slide 3. Drug Action and Handling. Lesson 2.1. Lesson 2.1. Drug Action and Handling. Drug Action and Handling.

Many drugs have both lipophilic and hydrophilic chemical substituents. Those drugs that are more lipid soluble tend to traverse cell membranes more

WHY... 8/21/2013 LEARNING OUTCOMES PHARMACOKINETICS I. A Absorption. D Distribution DEFINITION ADME AND THERAPEUIC ACTION

Fundamentals of Pharmacology for Veterinary Technicians Chapter 4

Pharmacokinetics Dr. Iman Lec. 3

Determination of bioavailability

ENVIRONMENTAL TOXICOLOGY

DEPARTMENT OF PHARMACOLOGY AND THERAPEUTIC UNIVERSITAS SUMATERA UTARA

Pharmacokinetics in Drug Development. Edward P. Acosta, PharmD Professor & Director Division of Clinical Pharmacology Director, CCC PK/PD Core

Pharmacokinetic Phase

Introduction to Pharmacokinetics

Pharmacokinetic Phase

2- Minimum toxic concentration (MTC): The drug concentration needed to just produce a toxic effect.

Renal Excretion of Drugs

Toxicant Disposition and Metabolism. Jan Chambers Center for Environmental Health Sciences College of Veterinary Medicine

TOXICOKINETICS; DISPOSITION OF XENOBIOTICS (Absorption, Distribution and Excretion of xenobiotics)

Importance of drug antagonism (i) Correcting adverse effects of drugs (ii) Treating drug poisoning. e.g. Morphine with naloxone, organophosphate

Pharmacokinetics in the critically ill. Intensive Care Training Program Radboud University Medical Centre Nijmegen

PHARMACOKINETICS SMALL GROUP I:

Renal Function. 1. Glomerular filtration 2. Active tubular secretion 3. Passive tubular reabsorption 4. Excretion

Pharmacokinetics of strong opioids. Susan Addie Specialist palliative care pharmacist

Drug Dosing in Renal Insufficiency. Coralie Therese D. Dimacali, MD College of Medicine University of the Philippines Manila

BASIC PHARMACOKINETICS

Lippincott Questions Pharmacology

Chapter 1. Introduction

Basic Pharmacokinetic Principles Stephen P. Roush, Pharm.D. Clinical Coordinator, Department of Pharmacy

Chapter 3 Drug Absorption and Bioavailability

DRUG DISTRIBUTION. Distribution Blood Brain Barrier Protein Binding

Pharmacodynamics & Pharmacokinetics 1

PRESCRIBING IN LIVER AND RENAL DISEASE

Define the terms biopharmaceutics and bioavailability.

Basic Biopharmaceutics, Pharmacokinetics, and Pharmacodynamics

DRUG ELIMINATION II BILIARY EXCRETION MAMMARY, SALIVARY AND PULMONARY EXCRETION

Problem Set for Fundamentals 9 Oct 2013

B. Incorrect! Compounds are made more polar, to increase their excretion.

Metabolism. Objectives. Metabolism. 26 July Chapter 28 1

Chapter Questions. Modern Pharmacology With Clinical Applications. Sixth Edition

Drug dosing in Extremes of Weight

METABOLISM. Ali Alhoshani, B.Pharm, Ph.D. Office: 2B 84

Metabolic Changes of Drugs and Related Organic Compounds

Principles of Toxicology: The Study of Poisons

Rational Dose Prediction. Pharmacology. φαρμακον. What does this mean? pharmakon. Medicine Poison Magic Spell

Introduction to. Pharmacokinetics. University of Hawai i Hilo Pre-Nursing Program NURS 203 General Pharmacology Danita Narciso Pharm D

The ADME properties of most drugs strongly depends on the ability of the drug to pass through membranes via simple diffusion.

UNIVERSITY OF THE WEST INDIES, ST AUGUSTINE

Principles of Pharmacology. Pharmacokinetics & Pharmacodynamics. Mr. D.Raju, M.pharm, Lecturer PHL-358-PHARMACOLOGY AND THERAPEUTICS-I

Assem Al Refaei. Sameer Emeish. Dr.Alia. Hodaifa Ababneh & Abdullah Shurafa

Basic Concepts of TDM

Pharmaceutics I صيدالنيات 1. Unit 2 Route of Drug Administration

Click to edit Master title style

PHARMACOKINETICS: DRUG ABSORPTION, DISTRIBUTION, AND ELIMINATION

DEFINITIONS. Pharmacokinetics. Pharmacodynamics. The process by which a drug is absorbed, distributed, metabolized and eliminated by the body

PHARMACOLOGY-1 PHL-313. Ali Alhoshani Office: 2B 84

PHA 5127 FINAL EXAM FALL On my honor, I have neither given nor received unauthorized aid in doing this assignment.

Mechanisms of Drug Action

Pharmacokinetics PCTH 325. Dr. Shabbits September 12, C t = C 0 e -kt. Learning Objectives

The importance of clearance

Drug CHAPTER 2. Pharmacologic Principles. NDEG 26A Eliza Rivera-Mitu, RN, MSN. Pharmacology. Drug Names. Pharmacologic Principles. Drug Names (cont'd)

EVE 491/591 Toxicology. Toxicant Entry into the Body 2/19/2018. Absorption and Fate of a Toxicant

Variation in drug responses & Drug-Drug Interactions

Introduction pharmacology and drug administration

Biopharmaceutics. Tips Worth Tweeting. Contributor: Sandra Earle

Nonlinear Pharmacokinetics

Osnove farmakokinetike. Aleš Mrhar. Prirejeno po. A First Course in Pharmacokinetics and Biopharmaceutics by David Bourne,

Renal Disease and PK/PD. Anjay Rastogi MD PhD Division of Nephrology

Introduction to Pharmacokinetics (PK) Anson K. Abraham, Ph.D. Associate Principal Scientist, PPDM- QP2 Merck & Co. Inc., West Point, PA 5- June- 2017

Model Answer B.Pharm. VII sem, Examination 2013 Biopharmaceutics and Pharmacokinetics Paper code: AS-2532

Pharmacokinetics Metabolism

Chimica Farmaceutica. Pharmacokinetics and related topics

Routes of drug administration

Tamer Barakat. Abdul Aziz ALShamali. Abdul Aziz ALShamali

Definition of bilirubin Bilirubin metabolism

Orientation for New Child and Adolescent Psychiatry Residents: Module Four Pediatric Psychopharmacology

BIOPHARMACEUTICS and CLINICAL PHARMACY

Tala Saleh. Abdul Aziz ALShamali. Abdul Aziz ALShamali

General Principles of Pharmacology and Toxicology

D9G : Oro-Mucosal Dosage Forms Development Background Paper

General Pharmacology

Biology 137 Introduction to Toxicology Name Midterm Exam 1 Fall Semester 2001

BIOP211. Session 2. Pharmacology Pharmacodynamics continued Pharmacokinetics. Department of Bioscience.

Basic pharmacokinetics. Frédérique Servin APHP hôpital Bichat Paris, FRANCE

Brand and Generic Drugs. Educational Objectives. Absorption

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook with Computer Simulations

What is pharmacokinetics?

PHAR 7632 Chapter 7. Table Market and Share of Pharmaceuticals by ROA Data from Viswanathan, 2004

Introducing Pharmacokinetics and Pharmacodynamics. Janice Davies Pharmacist Room 23 Maudland Building

Lecture 5: Principles of Drug Administration Drug Therapy in Special Populations Objectives

Transcription:

Pharmacokinetics Karim Rafaat

Pharmacokinetics The therapeutic effect of a drug is determined by the concentration of drug at the receptor site of action. Even though the concentration of drug that reaches the target receptor is dependent upon the dose of the drug, there are other factors that influence the delivery of the drug to the target site. These include: absorption distribution metabolism excretion.

Absorption Depends on patient compliance Depends on route of administration Small, non-ionized, lipid soluble drugs permeate plasma membranes most readily The plasma membrane is impermeable to polar, water-soluble substances

Routes of Administration Route of administratio Oral Sublingual Buccal Intra-ocular Topical Rectal Vaginal Respiratory tract Site of absorption Mouth, GI tract Under tongue Oral mucosa Eyes Skin Rectum Vagina Nasal passages or lungs

Routes of Administration Route of administration Intravenous Intramuscular Subcutaneous Epidural Intrathecal Site of absorption Directly into venous blood Muscles Into blood from skin layers Epidural space Directly into cerebrospinal fluid

Factors affecting drug absorption Compliance Food Formulation Enhance e.g. ketoconazole Impair e.g. tetracyclines, penicillin V, etidronate Enteric coated Slow release Route of administration IV = complete absorption (directly into blood stream) Oral = partial absorption Lack of specific receptor needed for absorption

Site of drug absorption If drug is absorbed from stomach, could bypass absorption site e.g. E/C preparations, PEJ tubes Malabsorption syndromes GI motility Inactivation of drug in gut or liver Pre-existing medical conditions Drug interactions e.g. cystic fibrosis e.g. rapid GI transit Crohn s disease, pro-kinetic drugs e.g. insulin destroyed by proteolytic enzymes First pass effect e.g. Acute congestive heart failure

Bioavailability The fraction of the administered dose that reaches the systemic circulation of the patient Affected by: Dosage form Dissolution and absorption of drug Route of administration Stability of the drug in the GI tract (if oral route) Extent of drug metabolism before reaching systemic circulation Presence of food/drugs in GI tract

Bioavailability Factor (F) Estimates the EXTENT of absorption Does not consider the RATE of absorption Amount of drug reaching systemic circulation = (F) x (dose)

Absorption I. Mechanisms for drug transport across membranes A. Passive (simple) diffusion 1. Rate of transfer of substances are directly proportional to the concentration gradient on both sides of the membrane 2. Rapid for lipophilic, nonionic, small molecules 3. No energy or carrier required B. Aqueous channels 1. Small hydrophilic drugs (<200 MW) diffuse along conc gradient by passing through pores (aqueous channels) 2. No energy required

Absorption C. Specialized transport 1. Facilitated diffusion drugs bind to carrier noncovalently» No energy is required 2. Active transport identical to facilitated diffusion except that ATP (energy) powers drug transport against conc gradient D. Pinocytosis and phagocytosis Engulfing of drug Ex: Vaccines

Distribution Takes place after the drug has been absorbed into the blood stream Distribution of drugs through various body compartments (to various tissues) depends on: Size of the organs (tissues) Blood flow through tissues Solubility of the drug in the tissues Binding of the drug to macromolecules in blood or tissues

Distribution The following factors influence drug distribution: 1. Protein binding Two factors determine degree of plasma protein binding:» 1. Affinity of drug for plasma protein» 2. # of binding sites available Weak acid drugs bind to albumin (phenytoin, salicylates, and disopyramide are extensively bound) Weak basic drugs bind to serum globulins α-1 acid glycoprotein (lidocaine, propranolol)

Plasma Protein Binding Many drugs bound to circulating plasma proteins such as albumin Only free drug can act at receptor site Protein-bound drug Receptor Site Free Drug

Effect of a change in plasma protein binding 90mg unbound 10mg bound Drug with low plasma protein binding

Effect of a change in plasma protein binding 90mg + 5mg unbound If the plasma protein level drops: Change in the unbound fraction is negligible 10mg - 5mg bound

Effect of a change in plasma protein binding 10mg unbound 90mg bound Drug with high plasma protein binding

Effect of a change in plasma protein binding 10mg + 5mg unbound If plasma protein level drops: 90mg 5mg bound Change in the unbound fraction is significant

Highly Protein Bound Drugs > 95% bound Thyroxine Warfarin Diazepam Furosemide Heparin > 90% but < 95% bound Phenytoin Propranolol Sodium Valproate Changes in plasma protein binding are significant for drugs which are greater than 90% bound to plasma proteins

Factors which can increase the fraction of unbound drug: Renal impairment due to rise in blood urea Low plasma albumin levels (<20-25g/L) E.g. chronic liver disease, malnutrition Displacement from binding site by other drugs e.g aspirin, sodium valproate, sulphonamides, Saturability of plasma protein binding within therapeutic range e.g. phenytoin

Distribution 2. Membrane permeability For a drug to enter an organ (tissue), it must permeate all membranes that separate the organ from the site of drug administration A. Blood brain barrier (BBB) lipid membrane located between plasma and the extracellular space in the brain» The entry of drugs is restricted into the CNS and CSF (cerebrospinal fluid)» Lipid solubility and cerebral blood flow limit permeation of the CNS» Highly lipophilic drugs can pass the BBB (i.e. benzodiazepines)» It is difficult to tx the brain or CNS, however, the difficulty of passage into the brain can also serve as a protective barrier when treating other parts of the body

Distribution 3. Storage Depots Drugs may collect in certain body tissues A. Fat lipophilic drugs accumulate here and are released slowly (due to low blood flow)» Ex: thiopental (or other anesthetic) causes sedation in obese patients B. Bone Ca ++ binding drugs accumulate here» Ex: tetracycline can deposit in bone and teeth will cause mottling or discoloration of teeth C. Liver many drugs accumulate in the liver due to an affinity for hepatic cells» Ex: quinacrine (antimalarial agent) has higher conc (22,000 times) in the liver than in plasma due to long term administration

Distribution Redistribution after a drug has accumulated in tissue, i.e. thiopental in fatty tissues, drug is gradually returned to the plasma Equilibrium between tissue drug and plasma drug

Distribution Vd = total amt of drug in body concentration of drug in blood or plasma Drugs with very high volumes of distribution have much higher concentrations in extravascular tissue than in the vascular compartment (plasma membrane) When drugs are protein bound, it can make the apparent volume smaller

Distribution TBW = 0.6 L/Kg (42 l) ECF vol = 0.2 L/Kg (12 l) Plasma vol. = 0.05 L/Kg (3 l) Blood vol = 5.5 l Clinical prediction: If Vd = 3L, you can assume drug is distributed in plasma only If Vd = 18L, you can assume drug is distributed in plasma and tissues If Vd > 46L, the drug is likely stored in a depot because the body only contains 40-46L of fluid

Distribution drugs with low Vd are often bound to plasma proteins drugs with high Vd are often bound to tissue components (proteins or fat)

Distribution Variances: Increased adipose tissue leads to increased depot stores of a drug with large Vd Which then leads to greater stores for eventual redistribution Patients with edema, ascites, or pleural effusion offer a larger volume of distribution to hydrophilic drugs than is predicted by their smaller, ideal Vd

Distribution Variances Pediatrics Body Composition» total body water & extracellular fluid» adipose tissue & skeletal muscle Protein Binding» albumin, bilirubin, 1-acid glycoprotein Tissue Binding» compositional changes

Imagine the body was a bucket: Dose In Overflow into tissues Tissues Blood stream Excretion

Distribution Uses of Vd If a drug is highly distributed to the tissues the first few doses disappear immediately from the blood stream Loading doses are required to fill up the tissues and the plasma Important if the site of drug action is in the tissues

Uses of Volume of Distribution Imagine a bucket with a leak You give a loading dose to fill up the bucket in the first place After that you only need to give enough to replace the amount leaking out. This is the maintenance dose.

Volume of distribution can help calculate the dose needed to achieve a critical plasma concentration Loading Dose (LD) = (Vd) x (C P )

Metabolism Process of making a drug more polar and water soluble to be excreted out of the body (lead to termination) Drug metabolism often results in detoxification or inactivation of drugs where the metabolites are less active or inactive compared to the parent drug Some metabolites may be equally or even more active than the parent drug. Prodrug inactive drug that is activated by metabolism (ex: enalapril)

Metabolism Some drugs have more than one metabolite Every tissue has some ability to metabolize drugs (i.e. GI tract, lungs, skin, kidneys) however, the liver is the principal organ for drug metabolism First-pass effect some drugs go straight from the GI tract to the portal system where they undergo extensive metabolism in the liver (ex: morphine) before entering the systemic circulation

Metabolism First-pass effect This can limit the bioavailability of certain drugs It can be greatly reduced by giving drug by other route of administration Extraction ratio an expression of the effect of first-pass hepatic elimination on bioavailability. Basically, how much drug is removed Hepatic Clearance = Hepatic blood flow x hepatic extraction ratio

Metabolism Drugs with high extraction ratios: clearance depends on hepatic blood flow Morphine Drugs with low extraction ratios: clearance depends on metabolic capacity of liver Diazepam phenytoin

Metabolism General pathways of drug metabolism Phase I reaction (oxidation, reduction, hydrolysis) Generally, the parent drug is oxidized or reduced to a more polar metabolite by introducing or unmasking a functional group (-OH, -NH 2, -SH) The more polar the drug, the more likely excretion will occur This reaction takes place in the smooth endoplasmic reticulum in hepatocytes

Metabolism Phase I reaction The smooth microsomes are relatively rich in enzymes responsible for oxidative drug metabolism Important class of enzymes mixed function oxidases (MFOs) The activity of these enzymes requires a reducing agent, NADPH and molecular oxygen (O 2 ) Two microsomal enzymes play a key role:» 1. NADPH-cytochrome P450 reductase, a flavoprotein» 2. Cytochrome P450, a hemoprotein, the terminal oxidase

Metabolism Phase I reaction Cytochrome P450 Is a family of isoenzymes Drugs bind to this enzyme and are oxidized or reduced Can be found in the GI epithelium, lung and kidney Cyp3A4 alone is responsible for more than 60% of the clinically prescribed drugs metabolized by the liver

Metabolism Phase II reaction This involves coupling the drug or it s polar metabolite with an endogenous substrate (glucuronic acid, sulfate, glycine, or amino acids) The endogenous substrates originate in the diet, so nutrition plays a critical role in the regulation of drug conjugation Drugs undergoing phase II conjugation reactions (glucuronidation, acetylation, methylation, and glutathione, glycine, and sulfate conjugation) may have already undergone phase I transformation Some parent drugs may already possess a functional group that may form a conjugate directly

Metabolism Enterohepatic recirculation some drugs, or their metabolites, which are concentrated in the bile then excreted into the intestines, can be reabsorbed into the bloodstream from the lower GI tract

Metabolism Variations in drug metabolism: Generally, men metabolize faster than women (ex: alcohol) Diseases can affect drug metabolism hepatitis, cardiac ( blood flow to the liver) Biotransformation in the neonate liver and metabolizing enzymes are underdeveloped also have poorly developed blood brain barrier Genetic differences Ex: Slow acetylators (autosomal recessive trait mostly found in Europeans living in the high northern latitudes and in 50% of blacks and whites in the US)

Excretion Elimination of unchanged drug or metabolite from the body terminating its activity Drugs may be eliminated by several different routes: exhaled air Sweat Saliva Tears Feces Urine Urine is the principle route of excretion

Three mechanisms for renal excretion: 1. Glomerular filtration passive diffusion Small nonionic drugs pass more readily. Drugs bound to plasma proteins do not Clearance by filtration = f u X GFR 2. Tubular secretion - drugs which specifically bind to carriers are transported (ex: penicillin) 3. Tubular reabsorption Small nonionic drugs pass more readily (ex:diuretics)

Excretion filtration reabsorption secretion filtration free drug only, not protein bound reabsorption passive, lipid soluble form only (ph) secretion active, acids and bases, saturable Plasma ph is constant; urine ph varies from 5.0-8.0

Excretion Elimination by the kidneys depends on Renal blood flow GFR Urine flow rate and ph, which influence Passive reabsorption Active secretion

Excretion Glomerular filtration matures in relation to age, adult values reached by 3 yrs of age Neonate = decreased renal blood flow, glomerular filtration, & tubular function yields prolonged elimination of medications

Adding it up

Enzyme processes obey Michaelis-Menton kinetics V = Vmax [Substrate] Km + [Substrate] Vmax Velocity ½ Vmax Km [Substrate] Vmax is the maximum rate of the process Km is the michaelis-menton constant, the concentration at which the rate is ½ of the maximum rate, Vmax.

Introduction Enzyme processes obey Michaelis-Menton kinetics V = Vmax [Substrate] Km + [Substrate] Vmax Velocity ½ Vmax Linear region Km [Substrate] Drugs used at concentrations at or below their Km are in the Linear region

Introduction Enzyme processes obey Michaelis-Menton kinetics V = Vmax [Substrate] Km + [Substrate] Vmax Velocity ½ Vmax Linear region Saturated region Km [Substrate] Drugs used at concentrations at or below their Km are in the Linear region Drugs used at concentrations several times their Km are in the Saturated region

Zero-order Kinetics For a drug whose concentration is above the Saturated value At this point, [Drug] >> Km, so the eqation: Simplifies to V = V = Vmax [Drug] Km + [Drug] Vmax [Drug] [Drug] A Constant Amount of drug is eliminated/metabolized per unit time Increased [Drug] DOES NOT result in an increase in metabolism or excretion (as appropriate) Referred to as Zero-Order Kinetics

Zero-order Kinetics Referred to as Zero-Order Kinetics Rate of elimination of [Drug] is Independent of [Drug] [Drug] dc dt = Constant TIME Zero-Order Kinetics Rate of Elimination is Constant with respect to Time

Zero-order Kinetics Only 2 common drugs: Acetylsalicylic Acid (ASA) Alcohol Example: Alcohol Alcohol Dehydrogenase Acetaldehyde Enzyme 94% saturated at 0.1% Blood Ethanol At saturation metabolizes ~ 10ml Ethanol / hour (~ 1 standard drink)

First-order Kinetics For a drug at a concentration in the Linear region Rate of elimination is Proportional to concentration = First-Order Kinetics Vmax Velocity ½ Vmax Linear region Saturated region Km [Substrate] Most Drug Elimination obeys First-Order Kinetics

First-order Kinetics For a drug whose concentration is in the linear region At this point, [Drug] << Km, so the eqation: Simplifies to V = V = Vmax [Drug] Km + [Drug] Vmax [Drug] Km Rate of Elimination is Proportional to concentration - First-Order Kinetics Where Vmax/Km equals the First-Order rate constant, k e

First-order Kinetics First-Order Kinetics A constant Proportion eliminated per unit time Amount Remaining 100% 50% dc dt = -10% C / Unit Time = -0.1 C / Unit Time Note: A curve, not a straight line Units of Time

Half-Life Ke values are difficult to conceptualize Solution: Concept of Half-Life (t 1/2 ) 16 8 t 1/2 = time required for [Drug] to decline by 1/2 Log [Drug] 4 2 t 1/2 t 1/2 Time

Relationship between ke and t 1/2 Relationship between ke and t 1/2 t 1/2 = 0.693/k e

Some purposeful examples

Pediatrics Differences in Distribution Less protein binding in infants Lower total protein and albumin than adults Leads to higher free plasma levels More free drug, greater pharmacologic effect» Phenytoin, salicylate, barbituates and diazepam» Also greater potential for toxicity

Volume of Distribution Infants have a greater proportion of bodyweight in the form of water Water soluable meds thus need higher initial loading dose» Digoxin, succinylcholine and gentamicin Smaller proportion of weight as fat and muscle mass Drugs that redistribute to muscle and fat have higher peak level and more sustained blood level» Barbituates and narcotics» Small muscle mass also means lower serum levels of muscle relaxants are required for effect

Hepatic Excretion Hepatic drug metabolism less active in neonates Phase I and II reactions reach adult activity by 6 to 18 months of age Lower proportion of blood flow to liver in younger children Diazepam, thiopental and phenobarbital have much increased serum half lives in infants

Renal Excretion Renal function less efficient in infants than adults relative to body weight Fully mature GFR by 2 years of age Drugs excreted by filtration have longer half lives» Aminoglycosides and cephalosporins

Propofol Highly lipophilic with rapid distribution to organs High Vd Termination of action secondary to redistribution and rapid hepatic and extrahepatic clearance Redistribution accounts for rapid onset and offset Children require lower induction doses than adults secondary to lower redistribution to fat stores

Morphine Half life markedly prolonged in infants Secondary to decreased capacity of liver glucuronidation activity Adult activity reached by one month of age Immaturity of blood brain barrier accounts for increased sensitivity to morphine

Fentanyl Longer half life in infants and neonates Lower hepatic blood flow Lower hepatic function Lower Vd secondary to lower fat mass Low dose fentanyl termination of action is by redistribution to fat and muscle High dose termination is secondary to metabolism EXTREMELY lipid soluable quickly passes through BBB

Vecuronium Infants less than 1yo more sensitive, with a longer duration of action Plasma conc required for given effect lower Less neuromuscular junctions secondary to less muscle mass Larger Vd, combined with lower necessary plasma level, leads to a slower decrease in plasma concentration