PEDRO AMORIM, MD PORTUGAL.

Similar documents
Drug Receptor Interactions and Pharmacodynamics

Fundamentals of Pharmacology

Life History of A Drug

INTERACTION DRUG BODY

Pharmacodynamics. OUTLINE Definition. Mechanisms of drug action. Receptors. Agonists. Types. Types Locations Effects. Definition

Receptors Families. Assistant Prof. Dr. Najlaa Saadi PhD Pharmacology Faculty of Pharmacy University of Philadelphia

Cellular Messengers. Intracellular Communication

PHRM20001: Pharmacology - How Drugs Work!

Basics of Pharmacology

STEIN IN-TERM EXAM -- BIOLOGY FEBRUARY 16, PAGE

Local Anesthetics. Xiaoping Du Room E417 MSB Department of Pharmacology Phone (312) ;

Membrane Structure and Function

Neurotransmitter Systems II Receptors. Reading: BCP Chapter 6

Review of Neurochemistry What are neurotransmitters?

INTERACTION DRUG BODY

Biosignals, Chapter 8, rearranged, Part I

Chapter 2: Cellular Mechanisms and Cognition

PHRM20001 NOTES PART 1 Lecture 1 History of Pharmacology- Key Principles

2013 W. H. Freeman and Company. 12 Signal Transduction

January 25, Introduction to Pharmacology

Receptors and Drug Action. Dr. Subasini Pharmacology Department Ishik University, Erbil

ANATOMY & PHYSIOLOGY - CLUTCH CH. 6 - CELL COMMUNICATION.

Lipids and Membranes

STEIN IN-TERM EXAM -- BIOLOGY FEBRUARY 15, PAGE

Cellular Neurophysiology I Membranes and Ion Channels

Enzymes Part III: regulation II. Dr. Mamoun Ahram Summer, 2017

Receptors. Dr. Sanaa Bardaweel

Physiology Unit 1 CELL SIGNALING: CHEMICAL MESSENGERS AND SIGNAL TRANSDUCTION PATHWAYS

Pharmacodynamics. Dr. Alia Shatanawi

Lab Results: 1. Document the initial and final egg masses. 2. Calculate the percent change

PHARMACODYNAMICS II QUANTITATIVE ASPECTS OF DRUGS. Ali Alhoshani, B.Pharm, Ph.D. Office: 2B 84

Lecture 15. Signal Transduction Pathways - Introduction

Signal Transduction: G-Protein Coupled Receptors

Introduction to Receptor Pharmacology

Which DNA sequence is most likely to form a hairpin structure? x indicates any nucleotide.

Chapter 5: Cell Membranes and Signaling

STEIN IN-TERM EXAM -- BIOLOGY FEBRUARY 18, PAGE

By the name of Allah

Plasma membranes. Plasmodesmata between plant cells. Gap junctions between animal cells Cell junctions. Cell-cell recognition

General Principles of Pharmacology and Toxicology

Electrical Properties of Neurons. Steven McLoon Department of Neuroscience University of Minnesota

Chapter 7 Nerve Cells and Electrical Signaling

Assem Al Refaei. Sameer Emeish. Sameer Emeish. Alia Shatnawi

Bear: Neuroscience: Exploring the Brain 3e

3) Most of the organelles in a neuron are located in the A) dendritic region. B) axon hillock. C) axon. D) cell body. E) axon terminals.

Comprehensive and Easy Course Notes for BIOL1040 Exams and Assessment

Pharmacology. Biomedical Sciences. Dynamics Kinetics Genetics. School of. Dr Lindsey Ferrie

number Done by Corrected by Doctor Alia Shatnawi

Ion Channels Graphics are used with permission of: Pearson Education Inc., publishing as Benjamin Cummings (

General anesthesia. No single drug capable of achieving these effects both safely and effectively.

Chapter 15: Signal transduction

Sarah Jaar Marah Al-Darawsheh

Pharmacodynamics. Prof. Dr. Öner Süzer Cerrahpaşa Medical Faculty Department of Pharmacology and Clinical Pharmacology

Communication Between

Learning Objectives. How do drugs work? Mechanisms of Drug Action. Liam Anderson Dept Pharmacology & Clinical Pharmacology

Chapter 11 Introduction to the Nervous System and Nervous Tissue Chapter Outline

General Anesthesia. Mohamed A. Yaseen

Ligand-Gated Ion Channels

Dania Ahmad. Tamer Barakat + Dania Ahmad. Faisal I. Mohammed

Chapter 2 Transport Systems

Cell Signaling part 2

Zool 3200: Cell Biology Exam 4 Part II 2/3/15

Lecture 9: Cell Communication I

- Biosignaling: Signal transduction. References: chapter 8 of Lippincots chapter 1 3 of Lehningers

Ion Channels (Part 2)

PHSI3009 Frontiers in Cellular Physiology 2017

Cell Signaling (part 1)

Bio 111 Study Guide Chapter 11 Cell Communication

Pharmacologic Principles. Dr. Alia Shatanawi

Goals and Challenges of Communication. Communication and Signal Transduction. How Do Cells Communicate?

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

Lojayn Salah. Razan Aburumman. Faisal Muhammad

Cell Biology Lecture 9 Notes Basic Principles of cell signaling and GPCR system

Bio 111 Study Guide Chapter 5 Membrane Transport and Cell Signaling

Basic Pharmacology. Understanding Drug Actions and Reactions

Chapter 2. The Cellular and Molecular Basis of Cognition Cognitive Neuroscience: The Biology of the Mind, 2 nd Ed.,

Concentration of drug [A]

STEIN IN-TERM EXAM -- BIOLOGY APRIL 19, PAGE

Chapter 20. Cell - Cell Signaling: Hormones and Receptors. Three general types of extracellular signaling. endocrine signaling. paracrine signaling

Tala Saleh. Ahmad Attari. Mamoun Ahram

It s Not Just Serotonin: Neurosignaling in Mental Illness

G-Protein Coupled Receptors GPCRs. GPCRs

Synaptic transmission

BIPN100 F15 Human Physiology 1 Lecture 3. Synaptic Transmission p. 1

QUIZ/TEST REVIEW NOTES SECTION 7 NEUROPHYSIOLOGY [THE SYNAPSE AND PHARMACOLOGY]

Session ID: 1001 June 14, 2012

Chapter 17: Functional Organization of the Endocrine System

Cell Membranes Valencia college

Chapter 25. General Anesthetics

Model Answer. M.Sc. Zoology (First Semester) Examination Paper LZT 103 (Endocrinology)

STEIN IN-TERM EXAM -- BIOLOGY APRIL 18, PAGE

Chapter 9 - Biological Membranes. Membranes form a semi-permeable boundary between a cell and its environment.

Psych 181: Dr. Anagnostaras

Cell Communication. Cell Communication. Communication between cells requires: ligand: the signaling molecule

Lecture 1 and 2 ONE. Definitions. Pharmacology: the study of the interaction of drugs within living systems

Transport through biological membranes. Christine Carrington Biochemistry Unit Apr 2010

I. Fluid Mosaic Model A. Biological membranes are lipid bilayers with associated proteins

MOLECULAR AND CELLULAR NEUROSCIENCE

1. Which of the following statements about passive and primary active transport proteins is FALSE?

G-Protein Signaling. Introduction to intracellular signaling. Dr. SARRAY Sameh, Ph.D

Transcription:

PEDRO AMORIM, MD Department of Anesthesiology, Intensive Care and Emergency Medicine Hospital Santo António Centro Hospitalar do Porto University of Porto PORTUGAL pamorim@vianw.pt

Drug Pharmacology Any exogenous non-nutritive substance that affects bodily functions This happens through physical interactions (antacids), changes in enzymatic activity or binding to molecular structures in or on the cell, that affect cellular function Pharmacokinetics Pharmacodynamics

Pharmacodynamics Pharmacodynamics for anesthesiologists taking the EDA diploma exams References/sources of information?

Professors Jennie Hunter and Ronald Miller

Pharmacodynamics Pharmacodynamics describes the relationship between plasma drug concentration and pharmacologic effect. Simply stated: pharmacokinetics describes what the body does to the drug, whereas pharmacodynamics describes what the drug does to the body

outline transduction of biologic signals (receptor theory and structure) developments in molecular pharmacology clinical evaluation of drug effects

Drug Receptor a component of a cell that interacts selectively with an extracellular compound to initiate a cascade of biochemical events that culminate in the pharmacologic effects of the compound receptors are now recognized as discrete excitable proteins

binding between drug and receptor (1) the quantitative relationship between a given dose of a drug and the resulting effect (2) the selectivity of a given drug's activity and effect (3) the pharmacologic activity of receptor agonists, antagonists, and inverse agonists Receptors therefore serve to mediate and amplify the effect of a drug on the biologic system

Claude Bernard (1813-1878) receptor theory ligated vessels leading to one hind limb of a frog while leaving nerve input intact. He then administered intravenous curare. Pinching the paralyzed hind limb produced reflex movements in the opposite unparalyzed vessel-ligated hind limb. These experiments and others demonstrated for the first time the separation between the sensory and motor nervous systems and also revealed that circulating substances produce selective effects on organ systems, a concept important in the development of receptor theory.

drug receptor interaction law of mass effect Combination of drug (ligand) and receptor depends on the concentrations of each The amount of drug-receptor complex formed determines the magnitude of the response

Pharmacokinetics involves the use of many formulas Pharmacodynamics also involves using some formulas

binding of a ligand Binding of a ligand, L, to its receptor, R, follows the laws of mass action and can be summarized by the relationship [L] + [R] K OFF K ON [LR] kon is the rate constant for binding of the ligand to the receptor koff is the rate constant for the dissociation of the ligand from the receptor [L] is the concentration of the ligand [R] is the concentration of the unbound receptor [LR] is the concentration of the bound receptor Note that the units for kon are time-1. units of [L] -1. The units of L are typically nanomoles per liter. The units for koff are time-1.

The rate of formation of [LR] is: At steady state, which is nearly instantaneous, the net rate of formation is 0, and thus [L] [R] kon = [LR] koff.

dissociation constant - Kd The term Kd, or dissociation constant, defines the characteristics of ligand/receptor interactions at equilibrium.

Drug binding Strength, duration and type of drug receptor interaction represent the affinity: Size and shape of drug molecule Types, number and spatial arrangement of binding sites (stereochemistry) Intramolecular forces between drug and binding sites: Van de Walls forces = weak bonds andn transient reversible effects Hydrogen bonds = intermidiate and transient reversible Covalent bonds = strong and long-lasting or irreversible

drug binding Binding of drugs involves mostly weak bonds The drug-receptor complex is not static Continuous association and dissociation of the drug The equilibrium dissociation constant Kd is a measure of this process Kd also represents the drug concentration necessary to bind 50% of the total receptor population

Free energy maps Ligand receptor interactions can be studied by energy free maps Binding affinities between propofol and HES were evaluated by binding free energy approximation (HGb, kj.mol-1) Aura Silva, Pedro Amorim et al

Selectivity of drug responses The cell responds only to the drugs that exhibit affinity for the receptors it expresses The greater the extent a drug molecule exhibits affinity for only one receptor, the more selective the drug s actions with lower potential for side effects The higher the affinity the lower the amount of drug necessary to exert an effect

Selectivity of drug responses Few drugs are entirely specific for one receptor Selectivity is relative As the concentration of drug increases it will combine with receptors for which it has lower affinity Off target side effects are generated Example: Beta- adrenergic receptor agonists and cardiovascular selectivity (asthma patients )

Tissue distribution of receptors

RECEPTOR AGONISTS AND ANTAGONISTS Drugs that are agonists mimic endogenous hormones or neurotransmitters when bound to a receptor. This effect may be excitatory or inhibitory.

RECEPTOR AGONISTS AND ANTAGONISTS Full agonist - completely activates a receptor Partial agonist - partially activates a receptor, even at very high concentrations Neutral antagonist - has no activity of its own Inverse agonist - superantagonists because they decrease receptor responses to less than the baseline

Flumazenil, Bretanazil, Flumazenil and RO 19-4063

Drug-receptor occupancy curves law of mass action Relationship is not linear but parabolic (Linear scale)

Drug-receptor occupancy curves law of mass action Logarithmic scale

Drug-receptor occupancy curves law of mass action Logarithmic scale

Dose-response relationships it is necessary to understand the relationship between the amount of drug given and the anticipated effect in the patient This relationship is described quantitatively by the dose-response curve There are two basic types of dose-response curve: Graded Quantal

Dose-response curves - relationships Measure an effect that is continuous such that, in theory, any value is possible in a given range (0% through 100%). Have a sigmoidal shape similar to the drug receptor occupancy curves, because the biologic response to a drug is determined by the interaction of a drug with a receptor or molecular target Exhibit a dose beyond which no further response is achieved (maximal effect; Emax) Emax is a measure of the pharmacologic efficacy of the drug Show the dose that produces 50% of the Emax (ED50). ED50 is an index of the potency of the drug Agonists with higher potency will have lower ED50 values

Dose-response curves - relationships The relationship between the drug-receptor binding event and the ultimate biologic effect is complex Quite often in experimental settings, the KD (concentration causing 50% receptor occupancy) does not correspond to a 50% maximal response from the test tissue or organism In many cases half-maximal tissue responses are obtained at drug concentrations below the KD, suggesting that amplification of drug response occurs.

potency from a therapeutic perspective, potency is often defined in terms of the dose-versus-response relationship. However, from a pharmacologic perspective, potency is best described in terms of the concentration versus-response relationship potency should be defined in terms of a specific drug effect (e.g., 50% of maximal effect). This is particularly important if the two drugs have different Hill coefficients or efficacies (Emax)

efficacy Efficacy is a measure of the intrinsic ability of a drug to produce a given physiologic or clinical effect The scale used to describe intrinsic efficacy at a given receptor ranges from 0 to 1 Efficacy is 1.0 for full agonists, 0 for neutral antagonists, and between 0 and 1.0 for partial agonists In contrast, the term potency refers to the quantity of drug that must be administered to produce a specific effect

graded dose-response curves A is the most potent (less ED 50) B & C equipotent (same ED 50) B more efficacy (higher Emax)

Quantal dose-response curves Quantify responses for variables that are all or none For example seizure or no seizure Describe the relationship between drug dosage and the frequency with which a biologic effect occurs For example, in individuals administered an anticonvulsant medication, the percentage of individuals not experiencing a convulsive episode at any given dose is plotted in cumulative fashion Represent a cumulative frequency distribution for a given response Provide an ED50 value that reflects the dose of drug at which 50% of the patients respond also called the median effective dose Provide an Emax value that is the dose at which all of the patients respond to the drug.

Quantal dose-response curves % patients responding vs log drug dose

Therapeutic Index Therapeutic index (TI) or therapeutic ratio is the ratio of the LD50 and ED50 Large values of Therapeutic Index are desirable because they indicate that the doses that produce death are much greater than those that produce a therapeutic effect

Therapeutic window Therapeutic window is a loosely defined term that generally refers to the range of doses that produce therapeutic effects with minimal toxic effects It can be viewed as the lack of overlap between the quantal dose-response curves for therapeutic and toxic or lethal effects There are several indices of the degree of this overlap.

safety Certain safety factor: The certain safety factor is the dose of drug that produces a lethal effect in 1% of the population (LD 1 ), divided by the dose that produces a therapeutic effect in 99% of the population (ED 99 ). So: (LD 1 /ED 99 ) Protective index: The protective index is calculated as the dose for an undesirable effect in 50% of patients (TD50) divided by the ED50 for the desired effect (TD50/ED50)

Lethal dose

Potency and efficacy and variability

Drug receptors Receptors are ubiquitous in the cell, being present in cell membranes, cytoplasm, intracellular organelle membranes, and the nucleus The overall physical structure of a receptor depends on the type of receptor and its location Cytoplasmic and nuclear receptors bind ligands that readily traverse cell membranes These ligands must therefore contain significant hydrophobic (lipophilic) components.

Drug receptors - anesthetics The receptors of most interest to anesthesiologists are located on the cell membrane They include membrane receptors, ligand-gated ion channels, and voltage-gated ion channels Receptors on the cell membrane bind water-soluble ligands that are typically unable to cross lipid bilayers and mediate a cellular response via intermediary proteins (e.g., G protein coupled receptors) or by causing changes in ion flux (e.g., ligand-gated ion channels).

Membrane associated drug targets in anesthesia

Guanine Nucleotide (G) Protein Coupled Receptors G protein coupled receptors are the most abundant type of receptor known Extracellular amino terminus containing glycosylation sites, an intracellular carboxyl terminus, fatty acid attachment (usually via palmitoylation or myristoylation of a carboxylterminal cysteine residue), three extracellular loops, and four intracellular loops.

G protein coupled receptors (two-dimensional version of receptor structure ) with seven transmembrane domains, an extracellular amino (NH2) terminus (with associated glycosylation sites [Y]), an intracellular carboxyl (COOH) terminus, palmitoylated cysteine residue (the crooked line extending into the membrane), three extracellular loops, and four intracellular loops

Schematic of the three-dimensional structure of the β2- adrenergic receptor (a prototypical G protein coupled receptor)

Ion channels Some anesthetic drugs target voltage-gated ion channels. These receptors mediate neural signaling by modulating ion permeability in electrically excitable membranes in response to changes in membrane potential Voltage-gated ion channels such as the sodium channel have charged regions that span the cell membrane The formation of ion pairs between many positive and negative charges helps stabilize the ion-conducting pore Local anesthetics work by blocking voltage-gated sodium channels

Ligand gated ion channels The ligand-gated ion channel is a combination of a classic receptor protein and an ion channel Ligand gated ion channels permit certain drugs to directly alter membrane potentials Many anesthetic drugs act on ligand-gated ion channels, such as the nicotinic acetylcholine receptor and the GABAA receptor.

The GABA receptor the neurotransmitter GABA binds to its receptor within the GABAA ligand-gated ion channel complex and causes the intracellular flux of chloride ions This action results in hyperpolarization of the membrane potential, a hallmark of inhibitory neurotransmission Drugs binding to other sites on the GABAA receptor facilitate the action of the endogenous ligand GABA

GABA receptor

Ion pumps Another type of excitable membrane protein is the ion pump. The sodium-potassium ATPase pump is perhaps the most familiar ion pump to the anesthesiologist because it is inhibited by digitalis Action potentials activate sodium channels, thereby allowing sodium to flux intracellularly along a combined chemical and electrical gradient The sodium-potassium-atpase pump then rapidly pumps sodium out of the cell in exchange for potassium, thereby returning the cell to its original cation composition and electrical gradient Digitalis acts by inhibiting the sodium-potassium- ATPase pump

Second messengers Binding of a ligand to a receptor does not instantly produce its clinical effects. Instead, a series of rapid biochemical events couples receptor binding to ultimate clinical effects These biochemical events are called second messengers Many membrane receptors couple to their second messenger through G proteins, which are intermediate regulatory molecules. Coupling of G proteins to receptor-hormone complexes requires energy in the form of guanosine triphosphate (GTP).

β-adrenergic receptor signal transduction cascade. βar, β-adrenergic receptor; ATP, adenosine triphosphate; camp, cyclic adenosine monophosphate; Gsα, α subunit of the stimulatory G protein (Gs); βγ, stimulatory G protein, βγ-subunit

Stimulation of receptors and second messengers ultimately leads to physiologic effects in a given tissue The physiologic effects produced depend on the presence of specific receptor subtypes, G proteins, and second messengers within that tissue. An example of the wide-ranging cardiovascular effects produced by norepinephrine

Actions at different receptors Anesthesia is the practice of applied drug interactions General anesthesia is, at a minimum, a combination of hypnosis, immobility, and antinociception Potentiation or synergism may occur and we use them intentionally

Effect of fentanyl on MAC of Isoflurane

Synergism of propofol and alfentanil Influence of alfentanil on the concentration of propofol associated with a 50% probability of response to intubation and incision, as well as a 50% probability of awakening at the end of surgery

Summary of drug interactions in humans and animals for hypnosis (loss of consciousness in humans, loss of righting reflex in animals) and immobility (loss of movement response to noxious stimulation). The numbers in each cell refer to the number of papers supporting the finding

MOLECULAR PHARMACOLOGY Molecular pharmacology takes advantage of the finding that all proteins, including excitable membrane proteins, are encoded in the human genome as nucleic acids Every amino acid present in a protein is encoded by a specific combination of three nucleotides in DNA Therefore, the DNA sequence that encodes a receptor protein can be determined Encoding DNA sequences (i.e., genes) can be inserted into special cells that will express (manufacture, assemble, and deliver to the appropriate location in the cell) the receptor protein in high quantity.

MOLECULAR PHARMACOLOGY By changing (mutating) nucleotide sequences, an abnormal (synthetic or designer ) receptor can be created Naturally occurring human receptor variants have been investigated and tested for alterations in pharmacologic properties PHARMACOGENOMICS New investigational drugs can be rapidly screened for effects on a spectrum of native and variant receptors

Clinical Evaluation of Drug Effects Once pharmacologic agents have exerted their actions on a molecular level, stimulated their targets, and produced a physiologic effect, it is important to evaluate the effect on the whole organism Dose-response curves, efficacy, potency, the median effective dose (ED50), the median lethal dose (LD50), and the therapeutic index

Clinical Evaluation of Drug Effects PATIENT VARIABILITY Genetic factors Biologically active genetic polymorphisms have been found in numerous receptors, second messenger systems, and ion channels Physiologic factors Age Liver function Proteins/albumin Disease states

Drug Interactions Pharmacokinetic Interactions Pd Drug-Time Interaction: Desensitization Pd Drug-Time Interaction: Increased Receptor Sensitivity Pd Drug-Drug Interactions

Anesthesia Pharmacodynamics How do we assess what the drug does to the body? How do we administer drugs? The way we were taught! In a per kilogram of weight basis!!! We use the concept of the average patient We do that for opioids We do that for inhaled anesthetics with MAC We do that for propofol

the average patient does not exist! BHU - Varanasi, 2013

We assess the patient clinically and with monitors BHU - Varanasi, 2013

Pharmacodynamic assessement of anesthesia How do we assess adequacy of hypnosis? In individual patients? Is it important to measure anesthetic plasma drug s concentrations in real time? Can we measure plasma concentrations in real time? We can measure expired end-tidal concentrations of inhalational anesthetics and calculate MAC! What about intravenous anesthetics?

propofol anesthesia

PHARMACODYNAMICS in ANESTHESIA Is it useful to measure real time concentrations? Should we guide anesthesia administration based on desired or recommended plasma concentrations? It makes more sense to use pharmacodynamic parameters! To use monitors that use variables that represent the effect!

BHU - Varanasi, 2013 EEG based indexes like the BIS

Or State Entropy and Response Entropy BHU - Varanasi, 2013

How do we assess analgesia adequacy? Can we use objective monitors? Can we measure pain? We can objectively measure the balance between nociceptive stimuli and antinociceptive drugs?

Closed Loop Anesthesia - Analgesia BHU - Varanasi, 2013

Analgesia / Nociception Index ANI: Analgesic Nociceptive Index Mathieu JEANNE, MD, PhD Anesthesia & Intensive Care Cic-It 807 Inserm University Hospital Lille, France

ANESTHESIA Bringing pharmacokinetics and pharmacodynamics together With modern Pk modeling With modern Pd monitors With modern drugs The anesthesiologist can become A SURFER!

Closed Loop Anesthesia - Ngai Liu Combining hypnosis and analgesia closed loop control BHU - Varanasi, 2013

Thank You