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

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

Chapter 4. Drug Biotransformation

Industrial Toxicology

Pharmacokinetics. Karim Rafaat

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

INTRODUCTION TO PHARMACOKINETICS

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

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

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

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

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

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

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

Pharmacokinetics Dr. Iman Lec. 3

BASIC PHARMACOKINETICS

Metabolic Changes of Drugs and Related Organic Compounds

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

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

Renal Excretion of Drugs

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

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

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

Drug dosing in Extremes of Weight

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

Pharmacokinetics Metabolism

DEPARTMENT OF PHARMACOLOGY AND THERAPEUTIC UNIVERSITAS SUMATERA UTARA

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

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

Pharmacokinetics of strong opioids. Susan Addie Specialist palliative care pharmacist

Click to edit Master title style

Determination of bioavailability

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

Tala Saleh. Abdul Aziz ALShamali. Abdul Aziz ALShamali

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

DRUG DISTRIBUTION. Distribution Blood Brain Barrier Protein Binding

ENVIRONMENTAL TOXICOLOGY

Pharmacodynamics & Pharmacokinetics 1

Lippincott Questions Pharmacology

EVE 491/591 Toxicology. Toxicant Distribution 2/20/2014

ADME Review. Dr. Joe Ritter Associate Professor of Pharmacology

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

Chapter 9. Biotransformation

Pharmacokinetics I. Dr. M.Mothilal Assistant professor

MODULE No.26: Drug Metabolism

Define the terms biopharmaceutics and bioavailability.

Section 5.2: Pharmacokinetic properties

Metabolic Changes of Drugs and Related Organic Compounds

Drug Metabolism Phase 2 conjugation reactions. Medicinal chemistry 3 rd stage

Name: Class: "Pharmacology NSAIDS (1) Lecture

Definition of bilirubin Bilirubin metabolism

General Pharmacology

Helping the liver to detoxify mycotoxins

Principles of Toxicology: The Study of Poisons

Introduction to Pharmacokinetics

Lecture 1: Physicochemical Properties of Drugs and Drug Disposition

Chapter Questions. Modern Pharmacology With Clinical Applications. Sixth Edition

UNIVERSITY OF THE WEST INDIES, ST AUGUSTINE

Drug elimination and Hepatic clearance Chapter 6

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

Tamer Barakat. Abdul Aziz ALShamali. Abdul Aziz ALShamali

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

Biopharmaceutics. Tips Worth Tweeting. Contributor: Sandra Earle

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

Volume of Distribution. Objectives. Volume of Distribution

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

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

Mechanism of Detoxification

Metabolism. Objectives. Metabolism. 26 July Chapter 28 1

Basic Pharmacokinetics and Pharmacodynamics: An Integrated Textbook with Computer Simulations

PRESCRIBING IN LIVER AND RENAL DISEASE

Mechanisms of Drug Action

The importance of clearance

PHARMACOKINETICS: DRUG ABSORPTION, DISTRIBUTION, AND ELIMINATION

PHA5128 Dose Optimization II Case Study I Spring 2013

Pharmacokinetics for Physicians. Assoc Prof. Noel E. Cranswick Clinical Pharmacologist Royal Children s Hospital Melbourne

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

Pharmacokinetics: The Basics

Pharmacogenetics and Pharmacokinetics

Basic Concepts of TDM

C OBJECTIVES. Basic Pharmacokinetics LESSON. After completing Lesson 2, you should be able to:

Adjusting phenytoin dosage in complex patients: how to win friends and influence patient outcomes

BIOPHARMACEUTICS and CLINICAL PHARMACY

Metabolic Changes of Drugs and Related Organic Compounds. Oxidative Reactions. Shokhan J. Hamid. 3 rd stage/ 1 st course Lecture 6

One-Compartment Open Model: Intravenous Bolus Administration:

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

DRUG ELIMINATION II BILIARY EXCRETION MAMMARY, SALIVARY AND PULMONARY EXCRETION

What is pharmacokinetics?

Lecture 8: Phase 1 Metabolism

PHA Second Exam Fall On my honor, I have neither given nor received unauthorized aid in doing this assignment.

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

PHA Second Exam. Fall On my honor, I have neither given nor received unauthorized aid in doing this assignment.

Polar bodies are either introduced or unmasked, which results in more polar metabolites Phase I reactions can lead either to activation or

Effects of Renal Disease on Pharmacokinetics

General Principles of Drug Action

2. List routes of exposure in the order of most rapid response.

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

MEDCHEM 570. First Midterm. January 30, 2015

PHA First Exam Fall 2003

Regulation of fluid and electrolytes balance

CLINICAL PHARMACOKINETICS INDEPENDENT LEARNING MODULE

Transcription:

It the process by which a drug reversibly leaves blood and enter interstitium (extracellular fluid) and/ or cells of tissues. Primarily depends on: 1.Regional blood flow. 2.Capillary permeability. 3.Protein binding. 4.Chemical nature of the drug.

Definition: Penetration of a drug to the sites of action through the walls of blood vessels from the administered site after absorption is called drug distribution. Drugs distribute through various body fluid compartments such as (a) plasma (b) interstitial fluid compartment (c) trans-cellular compartment. Apparent Volume of distribution (VD): The volume into which the total amount of a drug in the body would have to be uniformly distributed to provide the concentration of the drug actually measured in the plasma. It is an apparent rather than real volume.

Volume of distribution (Vd) of a drug is the volume of body fluids into which drug is distributed in same concentration as in plasma Total body fluid is about 60% of body weight i.e. about 42 L for a 70 Kg man. 2 / 3 ICF ~ 28 L 1 / 3 ECF ~ 14 L. This is divided into : 3 4 L Plasma 9 10 L Tissue fluid

In 70 kg patient : If Vd = 3-4 L, drug is mainly localized in plasma e.g. bound to plasma albumin If Vd = 10-12 L, then drug is localized in ECF i.e. it is water soluble and poorly enter cells If Vd = 20 L, then drug is lipid soluble, and partially enter into cells across cell membranes of tissues If Vd = 40 L, then drug is enough lipid soluble to be uniformly distributed in total body fluid e.g. alcohol If Vd = > 42 L e.g.100 L, drug is highly lipid soluble & is stored in some tissues e.g. Digoxin Vd = 300 L; hemodialysis is not useful to remove this drug from body in poisoning. Because of this large Vd for stored drugs, Vd is named : apparent i.e. avd

Drugs confined to the plasma compartment (plasma volume 0.05L/kg BWT) (e.g. heparin and warfarin): very large molecular weight, low lipid solubility, or binds extensively to plasma proteins. Drugs distributed in the extracellular compartment (intracellular volume 0.2L/kg) (e.g. aminoglycoside antibiotics): low molecular weight and hydrophilic

Drug distributed throughout the body water (total body water 0.55L/kg): lipid-soluble drugs that readily cross membrane. Other sites: Milk, bone, muscles. Drugs that are extremely lipid soluble (e.g. thiopental) may have unusually high volume of distribution).

1. Protein binding of drug: Many drugs circulate in the bloodstream bound to plasma proteins Albumin is a major carrier for acidic drugs and α1-acid glycoprotein (AAG) binds basic drugs The binding is usually reversible Binding of a drug to plasma proteins limits its concentration in tissues and at its site of action because only unbound drug is in equilibrium across membranes

Most drugs are bound to some extent to proteins in the blood to be carried into circulation. The protein-drug complex is relatively large & cannot enter into capillaries & then into tissues to react. The drug must be freed from the protein s binding site at the tissues.

Tightly bound released very slowly. these drugs have very long duration of action (not freed to be broken down or excreted), slowly released into the reactive tissue. Loosely bound tend to act quickly and excreted quickly Compete for protein binding sites alters effectiveness or causing toxicity when 2 drugs are given together.

The extent of plasma protein binding also may be affected by diseaserelated factors and drug-drug interactions. Hypoalbuminemia secondary to severe liver disease or nephrotic syndrome results in reduced binding and an increase in the unbound fraction The active concentration of the drug is that part which is not bound, because it is only this fraction which is free to leave the plasma and site of action.

(a) Free drug leave plasma to site of action (b) binding of drugs to plasma proteins assists absorption (c) protein binding acts as a temporary store of a drug and tends to prevent large fluctuations in concentration of unbound drug in the body fluids (d) protein binding reduces diffusion of drug into the cell and there by delays its metabolic degradation e.g. high protein bound drug like phenylbutazone is long acting. Low protein bound drug like thiopental sodium is short acting.

2. Plasma concentration of drug (PC): It represents the drug that is bound to the plasma proteins (albumins and globulins) and the drug in free form. It is the free form of drug that is distributed to the tissues and fluids and takes part in producing pharmacological effects. The concentration of free drug in plasma does not always remain in the same level e.g. i) After I.V. administration plasma concentration falls sharply ii) After oral administration plasma concentration rises and falls gradually. iii) After sublingual administration plasma concentration rise sharply and falls gradually.

3. Clearance: Volume of plasma cleared off the drug by metabolism and excretion per unit time. Protein binding reduces the amount of drug available for filtration at the glomeruli and hence delays the excretion, thus the protein binding reduces the clearance.

4. Physiological barriers to distribution: There are some specialized barriers in the body due to which the drug will not be distributed uniformly in all the tissues. These barriers are: a) Blood brain barrier (BBB) through which thiopental sodium is easily crossed but not dopamine. b) Placental barrier: which allows non-ionized drugs with high lipid/water partition coefficient by a process of simple diffusion to the foetus e.g. alcohol, morphine.

5. Affinity of drugs to certain organs: The concentration of a drug in certain tissues after a single dose may persist even when its plasma concentration is reduced to low. Thus the hepatic concentration of mepacrine is more than 200 times that of plasma level. Their concentration may reach a very high level on chronic administration. Iodine is similarly concentrated in the thyroid tissue.

Is the irreversible loss of drug from the body It occurs by two processes: Excretion & Metabolism Kidney and liver are the most common organs of drug elimination The kidney is the most important organ for excreting drugs and their metabolites Three fundamental processes account for renal drug excretion: glomerular filtration, active tubular secretion, passive tubular reabsorption

Drugs are chemical substances, which interact with living organisms and produce some pharmacological effects and then, they should be eliminated from the body unchanged or by changing to some easily excretable molecules. The process by which the body brings about changes in drug molecule is referred as drug metabolism or biotransformation.

Involves enzymic conversion of one chemical entity to another within the body The liver is the major site for drug metabolism Specific drugs may undergo biotransformation in other tissues, such as the kidney and the intestine

Enzymes responsible for metabolism of drugs: Microsomal enzymes: Present in the smooth endoplasmic reticulum of the liver, kidney and GIT e.g. glucuronyl transferase, dehydrogenase, hydroxylase and cytochrome P450. Non-microsomal enzymes: Present in the cytoplasm, mitochondria of different organs.e.g. esterases, amidase, hydrolase.

The chemical reactions involved in biotransformation are classified as phase-i and phase II (conjugation) reactions. In phase-i reaction the drug is converted to more polar metabolite. If this metabolite is sufficiently polar, then it will be excreted in urine. Some metabolites may not be excreted and further metabolised by phase II reactions.

The enzyme systems for drug metabolic biotransformation reactions can be grouped into two categories: Phase-I: Oxidation, reduction and hydrolysis. Phase-II: Glucuronidation, sulfate conjugation, acetylation, glycine conjugation and methylation reactions.

Excretion of drugs means the transportation of unaltered or altered form of drug out of the body. The major processes of excretion include renal excretion, hepatobiliary excretion and pulmonary excretion. The minor routes of excretion are saliva, sweat, tears, breast milk, vaginal fluid, nails and hair. The rate of excretion influences the duration of action of drug. The drug that is excreted slowly, the concentration of drug in the body is maintained and the effects of the drug will continue for longer period.

Usually convert the parent drug to a more polar metabolite by introducing a functional group (-OH, -NH2, -SH). Phase I metabolism may increase, decrease, activate (prodrug, e.g. enalapril) or leave unaltered the drug s pharmacologic activity. Phase I reactions are catalyzed by the cytochrome P450 (CYP450) system functional group ( -OH, - NH2, -S H).

Species and strain variations exist in amount and activity of cytochrome P- 450 isoforms. Isoforms are classified into families and further into subfamilies. Nomenclature : e.g. CYP3 A4 : CYP : capital letters indicating the human enzyme, Cytochrome P450, designated as CYP is a superfamily of heme-containing isozymes that are located in most cells, but primarily in the liver and GI tract 3 : numeral indicating the family number A : capital letter indicating the subfamily 4 : numeral that indicates the isoform number in the subfamily Although they are large in number (hundreds), mainly isoforms of 3 families of CYP-450 are important for drug metabolism in man

CYP450 is composed of many families of isoenzymes known as isoforms Six isoforms are responsible for the vast majority of CYP450- catalyzed reactions: CYP3A4, CYP2D6, CYP2C9/10, CYP2C19, CYP2E1, and CYP1A2 Variability in the activity of CYP450 enzymes is linked to a range of factors including genetic, environmental, and developmental

Lead to the formation of a covalent linkage between a functional group on the parent compound or phase I metabolite and endogenously derived glucuronic acid, sulfate, glutathione, amino acids, or acetate The highly polar conjugates generally are inactive and are excreted rapidly in the urine and feces Nenates are deficient in this conjugation system, making them particularly vulnerable to drugs such as cholamphenicol (gray baby syndrome)

Clearance: Volume of plasma cleared off the drug by metabolism and excretion per unit time. The main PK parameter describing elimination. It is the most important concept to consider when designing a rational regimen for long-term drug administration. Drug clearance from the organ of elimination can be described as: Q: blood flow to the organ of elimination ER: Extraction ratio CpA: arterial drug concentration CpV: venous drug concentration

Total body (systemic) clearance,cltotal, is the sum of the clearance from various drug metabolizing (mainly the liver) and drug excreting organs (mainly the kidney) [Additive process]: CLtotal = CLhepatic + Clrenal + CLpulmonary + Clother Units of clearance are volume/time (e.g. L/h or ml/min)

Is the elimination of drugs from the body 35

Inducers: The CYP450-dependent enzymes are an important target for pharmacokinetic drug interactions. Xenobiotics (chemicals not normally produced or expected to be present in the body, for example, drugs or environmental pollutants) may induce the activity of these enzymes. This results in increased biotransformation of drugs and can lead to significant decreases in plasma concentrations of drugs metabolized by these CYP isozymes, with concurrent loss of pharmacologic effect.

Inhibitors: Inhibition of CYP isozyme activity is an important source of drug interactions that lead to serious adverse events. The most common form of inhibition is through competition for the same isozyme. Numerous drugs have been shown to inhibit one or more of the CYP-dependent biotransformation pathways of warfarin. For example, omeprazole is a potent inhibitor of three of the CYP isozymes responsible for warfarin metabolism. If the two drugs are taken together, plasma concentrations of warfarin increase, which leads to greater anticoagulant effect and increased risk of bleeding.

First-order kinetics: The metabolic transformation of drugs is catalyzed by enzymes, and most of the reactions obey Michaelis-Menten kinetics. In most clinical situations, the concentration of the drug, [C], is much less than the Michaelis constant, Km, and the Michaelis-Menten equation reduces to

That is, the rate of drug metabolism and elimination is directly proportional to the concentration of free drug, and first-order kinetics is observed. This means that a constant fraction of drug is metabolized per unit of time (that is, with each half-life, the concentration decreases by 50%). First-order kinetics is also referred to as linear kinetics.

Zero-order kinetics: The enzyme is saturated by a high free drug concentration, and the rate of metabolism remains constant over time. A constant amount of drug is metabolized per unit of time. The rate of elimination is constant and does not depend on the drug concentration.

Is based on the assumption that there is a target concentration that will produce the desired therapeutic effect The intensity of a drug's effect is related to its concentration above a minimum effective concentration, whereas the duration of this effect reflects the length of time the drug level is above this value By considering drug s PKs, it is possible to individualize the dose regimen to achieve the target concentration

Half-Life (t 1/2 ) : it is the time required for the plasma concentration or the amount of drug in the body to change by one-half (i.e. 50%) The half-life is a derived parameter that changes as a function of both CL and Vd: