Octanol / Water Partitioning Coefficient logp and ClogP

Similar documents
Chapter 4. Drug Biotransformation

Role of metabolism in Drug-Induced Liver Injury (DILI) Drug Metab Rev. 2007;39(1):

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

Chapter 9. Biotransformation

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

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

BIOMARKERS AND TOXICITY MECHANISMS 07 Mechanisms Metabolism & Detoxification. Luděk Bláha, PřF MU, RECETOX

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

Mechanism of Detoxification

INTRODUCTION TO PHARMACOKINETICS

MODULE No.26: Drug Metabolism

Mechanistic Toxicology

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

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

Metabolic Changes of Drugs and Related Organic Compounds

Metabolic Changes of Drugs and Related Organic Compounds

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

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

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

Introduction to Detoxification Enzymes. Evolutionary Response to Chemicals in the Environment

Controlling ADME through Chemical Design. Marty Mulvihill Chris Vulpe

Chapter Questions. Modern Pharmacology With Clinical Applications. Sixth Edition

MEDCHEM 570. First Midterm. January 30, 2015

DEPARTMENT OF PHARMACOLOGY AND THERAPEUTIC UNIVERSITAS SUMATERA UTARA

Industrial Toxicology

P450 CYCLE. All P450s follow the same catalytic cycle of;

Define the terms biopharmaceutics and bioavailability.

The importance of pharmacogenetics in the treatment of epilepsy

Click to edit Master title style

Cytochrome P 450 Unique family of heme proteins present in bacteria, fungi, insects, plants, fish, mammals and primates. Universal oxygenases (oxygen-

Biologic Oxidation BIOMEDICAL IMPORTAN

Application of Pharmacogenetics Supplementary Worksheet

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

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

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

Cytokrom P450 (CYP) Hepatic Drug Metabolism. Medicines in plasma. Plasma concentration of a medicine. Eva Brittebo Dept Pharmaceutical Biosciences

Objectives Making CYP450, Drug Interactions, & Pharmacogenetics Easy

CYTOCHROME P450: Structure-Function

Pharmacokinetics Metabolism

Variability Due to Genetic Differences

Review Session 1. Control Systems and Homeostasis. Figure 1.8 A simple control system. Biol 219 Review Sessiono 1 Fall 2016

Metabolism. Objectives. Metabolism. 26 July Chapter 28 1

Chapter 2 The Chemistry of Life Part 2

Lecture 8: Phase 1 Metabolism

PHARMACOKINETICS: DRUG ABSORPTION, DISTRIBUTION, AND ELIMINATION

PHASE I Detoxification: The First Line of Defense

BIOL 158: BIOLOGICAL CHEMISTRY II

accesalabs.c

Chemical Classification of Hormones

Helping the liver to detoxify mycotoxins

DILI: Clinical Pharmacology Considerations for Risk Assessment

MEDCH/PCEUT 527 ADVANCED DRUG METABOLISM 2019 Course Coordinators: Allan Rettie and Ken Thummel When/ Where:

Metabolism of xenobiotics FM CHE 5-6

From Atoms to Cells: Fundamental Building Blocks. Models of atoms. A chemical connection

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

Exploiting BDDCS and the Role of Transporters

Welcome to Week Oral Delivery II. Starting week four video. Multiple oral doses video. Calculating Cp for an oral drug

Organic Molecules. 8/27/2004 Mr. Davenport 1

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

Tala Saleh. Abdul Aziz ALShamali. Abdul Aziz ALShamali

Molecule - two or more atoms held together by covalent bonds. Ex. = water, H O

Name: Date: Block: Biology 12

number Done by Corrected by Doctor Nayef Karadsheh

Definition of bilirubin Bilirubin metabolism

Lujain Hamdan. Faisal Nimri

number Done by Corrected by Doctor

OBJECTIVE. Lipids are largely hydrocarbon derivatives and thus represent

number Done by Corrected by Doctor

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

Ibuprofen. Ibuprofen and Paracetamol: prescribing overview. Ibuprofen indications CYCLO-OXYGENASE (COX I) CYCLO-OXEGENASE (COX II) INFLAMMATORY PAIN

Carbohydrates, Lipids, Proteins, and Nucleic Acids

INTERACTION DRUG BODY

Chapter 26 Biochemistry 5th edition. phospholipids. Sphingolipids. Cholesterol. db=books&itool=toolbar

Constitutive Regulation of P450s by Endocrine Factors

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

Lipids and Classification:

Biomolecules. Biomolecules. Carbohydrates. Biol 219 Lec 3 Fall Polysaccharides. Function: Glucose storage Fig. 2.2

Adenosine triphosphate (ATP)

Chapter 2 Transport Systems

Cysteine Peptide Scientific Review, Dr. S. Dudek, DMV International

Pharmacokinetics: The Basics

MEDCHEM 562. First Midterm (KEY) October 15, 2012

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

MITOCW watch?v=xms9dyhqhi0

Oxidative Phosphorylation

Hormones, Receptors and Receptor-Hormone Interactions

BASIC PHARMACOKINETICS

Porphyrins: Chemistry and Biology

Oxidative phosphorylation & Photophosphorylation

Fatty acid breakdown

Good Afternoon! 11/30/18

CH 3. Lipids CHAPTER SUMMARY

Lecture 12 Enzymes: Inhibition

UNIVERSITY OF GUELPH CHEM 4540 ENZYMOLOGY Winter 2005 Quiz #2: March 24, 2005, 11:30 12:50 Instructor: Prof R. Merrill ANSWERS

Biopharmaceutics Drug Disposition Classification System (BDDCS) and Its Application in Drug Discovery and Development

Moh Tarek + Faisal Massad. Tala Saleh ... Naif

Lecture 36: Review of membrane function

Chemical Energy. Valencia College

Chapter 6: Estrogen Metabolism by Conjugation

Metabolism of pentoses, glycogen, fructose and galactose. Jana Novotna

Transcription:

Lipid solubility and the brain uptake of compounds Higher the Kow, the greater the brain uptake Specific carriers can facilitate brain uptake (glucose, lucine, dopa) Binding to plasma proteins can reduce uptake (phenytoin) Octanol / Water Partitioning Coefficient logp and ClogP The logp is an important physicochemical parameter for oral absorption http://www.biobyte.com/ Measure for the lipophilicity of a compound. ClogP is the calculated log octanol/water partition coefficient, It relates to solubility and influences the ability of a compound to permeate cell membranes Too hydrophilic compounds (negative logp) cannot permeate membranes Too lipophilic compounds (high logp) are insoluble, poorly permeate through membranes (get stuck in the lipophilic bilayer). 1

Predicting Favorable ADME in Drug Development - Lipinski s Rule of Five Lipinski's rule: An orally active drug has no more than one violation of the following : 1. No more than 5 hydrogen bond donors (N or O with >1 H atom) 2. Not more than 10 hydrogen bond acceptors (N or O atoms) 3. A molecular mass < 500 daltons 4. An octanol-water partition coefficient (log P) < 5 A rule of thumb to evaluate: If a chemical compound with a pharmacological /biological activity has properties that may make it a likely orally active drug. Rule formulated by Chris Lipinski in 1997, based on the observation that most medication drugs are relatively small and lipophilic molecules * As with all rules, many exceptions! Predicts that poor absorption and permeability of potential drug candidates will occur if: 1) Number of H-bond donors > 5 2) Number of H-bond acceptors > 10 3) Molecular Weight > 500 4) ClogP > 5 Lipinski s rule does not predict if a compound is pharmacologically active. Rule is important to keep in mind during drug discovery when a lead structure is optimized step-wise to increase the activity and selectivity. Candidate drugs that conform to the RO5 tend to have lower attrition rates during clinical trials Drug Metabolism / Biotransformation Learning objectives Distinguish the differences between Phase I and Phase II reactions Recognize the enzymes involved in these processes and that various factors (age, genetic predisposition/polymorphisms, co-exposure to other drugs, diet, illness) may affect these processes Be familiar with at least one example of one of the following types of Phase I biotransformation reactions: Oxidation, Reduction, Hydrolysis. The same goes for the following types of Phase II biotransformation reactions: Conjugation with glucuronide, glutathione, sulfate. Recognize that one of the possible consequences of biotransformation is the conversion of prodrugs to active drug, or active drug to reactive intermediates, and that depending upon their chemical nature, these intermediates can modify molecular targets through different mechanisms 2

Drug Metabolizing Enzyme System Phase I and Phase II Biotransformation Phase I metabolism Functionalization Addition or exposing of a reactive functional group such as -OH, -SH, -NH2 or COOH In general, Phase I metabolism prepares the xenobiotic for subsequent Phase II reactions Phase II Reactions Enzymatic reactions that conjugate large watersoluble, charged (polar) biomolecules to xenobiotics The Truck-Hitch-Trailer Analogy to Xenobiotic Biotransformation Foreign Chemical (xenobiotic) Phase 1 enzymes add or expose a functional group Phase 2 enzymes conjugate (transfer) endogenous molecules* to the functional group lipophilic not charged not water soluble poorly excretable * sugars, amino acids, sulfates, acetyl groups HITCH Phase 1 enzymes add or expose a functional group still lipophilic possibly reactive poorly water soluble poorly excretable catalyzed by P450s TRAILER not lipophilic usually not reactive water soluble products Excretable catalyzed by transferases 3

Biotransformation Enzyme-Containing Cells in Various Organs Organ Cell(s). Liver Parenchymal cells (hepatocytes) Kidney Proximal tubular cells (S3 segment) Lung Clara cells, Type II alveolar cells Intestine Mucosa lining cells Skin Epithelial cells Testes Seminiferous tubules, Sertoli cells Mixed Function Oxidase System (MFO) (Phase I Metabolism) MFOs Contain: Cyt. P450, 2 flavoproteins (P450 reductases), Cytochrome b5 4

CYP families in humans Humans have 57 genes and more than 59 pseudogenes divided among: 18 families of cytochrome P450 genes 43 subfamilies Function Family Members Names CYP1 drug and steroid (especially estrogen) metabolism 3 subfamilies, 3 genes, 1 pseudogene CYP1A1, CYP1A2, CYP1B1 CYP2 drug and steroid metabolism CYP2A6, CYP2A7, CYP2A13, CYP2B6, CYP2C8, CYP2C9, CYP2C18, CYP2C19, 13 subfamilies, 16 genes, 16 pseudogenes CYP2D6, CYP2E1, CYP2F1, CYP2J2, CYP2R1, CYP2S1, CYP2U1, CYP2W1 CYP3 drug and steroid (including testosterone) metabolism 1 subfamily, 4 genes, 2 pseudogenes CYP3A4, CYP3A5, CYP3A7, CYP3A43 CYP4 arachidonic acid or fatty acid metabolism 6 subfamilies, 11 genes, 10 pseudogenes CYP4A11, CYP4A22, CYP4B1, CYP4F2, CYP4F3, CYP4F8, CYP4F11, CYP4F12, CYP4F22, CYP4V2, CYP4X1, CYP4Z1 CYP5 thromboxane A 2 synthase 1 subfamily, 1 gene CYP5A1 CYP7 bile acid biosynthesis 7-alpha hydroxylase of steroid nucleus 2 subfamilies, 2 genes CYP7A1, CYP7B1 CYP8 varied 2 subfamilies, 2 genes CYP8A1 (prostacyclin synthase), CYP8B1 (bile acid biosynthesis) CYP11 steroid biosynthesis 2 subfamilies, 3 genes CYP11A1, CYP11B1, CYP11B2 CYP17 steroid biosynthesis, 17-alpha hydroxylase 1 subfamily, 1 gene CYP17A1 CYP19 steroid biosynthesis: aromatase synthesizes estrogen 1 subfamily, 1 gene CYP19A1 CYP20 unknown function 1 subfamily, 1 gene CYP20A1 CYP21 steroid biosynthesis 2 subfamilies, 2 genes, 1 pseudogene CYP21A2 CYP24 vitamin D degradation 1 subfamily, 1 gene CYP24A1 CYP26 retinoic acid hydroxylase 3 subfamilies, 3 genes CYP26A1, CYP26B1, CYP26C1 CYP27 varied 3 subfamilies, 3 genes CYP27A1 (bile acid biosynthesis), CYP27B1 (vitamin D3 1-alpha hydroxylase, activates vitamin D3), CYP27C1 (unknown function) CYP39 7-alpha hydroxylation of 24-hydroxycholesterol 1 subfamily, 1 gene CYP39A1 CYP46 cholesterol 24-hydroxylase 1 subfamily, 1 gene CYP46A1 CYP51 cholesterol biosynthesis 1 subfamily, 1 gene, 3 pseudogenes CYP51A1 (lanosterol 14-alpha demethylase) Cytochrome P450 Characteristics Broad substrate specificity - Most drug metabolizing CYPs are promiscuous re: substrate, but some CYPs are highly specific to substrate, e.g., Cyp 19 (aromatase w/ testosterone substrate). CYP enzyme levels induced by exposure to xenobiotics Broad substrate specficity = Low catalytic efficiency (Kcat). Drugs in general have t1/2 of 3 30 hrs, while endogenous substrates are much shorter (sec or min) reflects lower Kcat of drug CYPs. Broad substrate specificity of many CYPs accounts for much of reason for drug drug interactions. Multiple drugs may compete for binding site, altering metabolism of one or more of the drugs, thereby increasing t1/2 and plasma levels (Dixogin example). 5

Cytochrome P450 s Limted number of CYPs (15) in families 1 3 are mostly responsible for drug metabolism. In humans, 12 CYPs in particular important: CYP1A1, 1A2, 1B1, 2A6, 2B6, 2C8, 2C9, 2C19, 2D6, 2E1, 3A4, 3A5. The most active in drug metabolism are the 2C, 2D, and 3A subfamiles, with CYP3A4 involved in metabolism of ~50% of clinically used drugs. Cytochrome P450 Substrate Binding P450s possess active HEME group: 2 types of substrate binding to P450 Type 1 Binding: The substrate binds to a protein moiety near the active site. This brings the substrate within the region of the catalytic activity of the Heme-Fe. Fe in Low Spin Type II Binding: The substrate binds directly to the Heme-Fe. Fe in High Spin Can determine whether Type I or Type II binding spectrophotometrically -Type I binding of Cyt. P450 to the substrate gives a characteristic A MAX of around 390nm. -Type II binding of Cyt. P450 to the substrate gives a characteristic A MAX of around 420nm. 6

Systems Involved in Phase II Metabolism Four primary enzymes: 1. Glucuronosyltransferase glucuronic acid 2. Sulfotransferase sulfate 3. Glutathione-S-transferase glutathione (GSH) 4. Acetyltransferase acetyl enzymes are, for the most part, in the cytosol of cells In all cases (except GSH) requires conjugate activated to an Electrophilic Activated Donor Phase II Enzymes: Examples Glucuronidation and Sulfation of a Hydroxyl Group 7

UDP Conjugation - Phenytoin Phenytoin Common antiepileptic Marketed as Phenytek, Dilantin Insoluble in water Stabilizes inactive state of voltage-gated Na channels Induces CYP3A4, 2C19 Interactions: Coumarin and Warfarin increase serum phenytoin levels and prolong the serum half-life of phenytoin by inhibiting its metabolism Pharmacokinetics can be saturable, leading to highly variable plasma levels across minor dose changes Bioactivation and UDP Conjugation: Irinotecan Irinotecan cancer drug (mostly colon cancer). Topoisomerase 1 inhibitor leads to inhibition of DNA replication Administered as ProDrug Carboxyesterase activates to active drug (SN-38) 8

Gilbert s Syndrome Drug Polymorphism interaction Generally benign condition present in ~10% of pop n. Diagnosed clinically because circulating bilirubin levels are 60 70% elevated. Most common genetic polymorph associated with Gilbert s is mutation in UGT1A1 gene promoter, leading to reduced expression of UGT1A1. If drug undergoes Phase II with UGT1A1, it will compete with bilirubin metabolism conjugation, leading to reduced bilirubin elimination and build up in plasma. Glutathione-S-Transferase (GST) Thiol strong nucleophile, also susceptible to electrophilic attack +ve S H GST - Conjugates glutathione (GSH) onto substrates Tripeptide (cysteine/glutamine amide bond through glutamate R group). Present in all cells high in hepatocytes ~ 5mM 9

Factors that Affect Drug Biotransformation Species, strain, and genetic variation Age Diet Exposure to other drugs / xenobiotics Species Differences in the Duration of Action and Metabolism of Hexobarbital Factors that Affect Xenobiotic Biotransformation Substantial genetic (polymorphisms) exist in humans that impact drug/xenobiotic metabolism Clin Transplant. 2009 Aug-Sep;23(4):490-8. Epub 2009 Apr 17. Influence of genetic polymorphisms in GSTM1, GSTM3, GSTT1 and GSTP1 on allograft outcome in renal transplant recipients. Singh R, Manchanda PK, Kesarwani P, Srivastava A, Mittal RD. Department of Urology and Renal Transplantation, Sanjay Gandhi Post Graduate Institute of Medical Sciences, Uttar Pradesh, India. Breast Cancer Res Treat. 2009 Dec 25. [Epub ahead of print] Four polymorphisms in cytochrome P450 1A1 (CYP1A1) gene and breast cancer risk: a meta-analysis. Sergentanis TN, Economopoulos KP. School of Medicine, National University of Athens, Athens, Greece, tsergentanis@sni.gr. J Hum Genet. 2009 Oct;54(10):557-63. Epub 2009 Aug 21. Association between polymorphisms in glutathione S-transferase Mu3 and IgG titer levels in serum against Helicobacter pylori. Tatemichi M, Iwasaki M, Sasazuki S, Tsugane S. Department of Hygiene and Preventive Medicine, School of Medicine, Showa University, Shinagawa-ku, Tokyo, Japan. tatemichi@med.showa-u.ac.jp 10

Bimodal Distribution of Patients: Those who Rapidly Inactivate Isoniazid and Those who Slowly Metabolize It Isoniazid First-line antituberculosis medication in prevention and treatment Metabolized in the liver via acetylation. Two forms of the enzyme responsible for acetylation - Some patients metabolize the drug more quickly than others 11

Age as Affecting Xenobiotic Biotransformation E.g., jaundice in newborns due to poorly induced glucuronidation of heme bilirubin build up Drug Metabolic Activity Schematic representation of the ontogeny of hepatic drug metabolic activity Environmental Factors Affecting Biotransformaiton Induction of Xenobiotic Metabolizing Systems 1. Many chemicals can induce the synthesis of the enzymes involved in Phase I and II xenobiotic metabolism and include chemicals found in the environment, the diet, and other drugs. 2. Inducers often exhibit specificity for the enzymes which they induce 12

Induction of Xenobiotic Metabolizing Systems Aryl Hydrocarbon Receptor System Expert Opin. Drug Metab. Toxicol. (2005) 1(1) CYP450 expression Nuclear Receptor Pregnane X Receptor (PXR) Transcriptional regulator of CYP3A4 Binds to response element of CYP3A4 promoter as a heterodimer with the 9-cis retinoic acid receptor (RXR). 13

Bioactivation and Toxicity Chemical Nature of Reactive Intermediates: Electrophiles Form covalent (irreversible) bonds with cellular nucleophiles such as GSH, proteins and DNA Free Radicals Odd or unpaired electron Can act as electrophiles Can abstract hydrogen from target molecules, such as lipids or nucleic acids Can activate molecular oxygen Acetaminophen toxicity O HN C CH 3 OH acetaminophen 15 20 tablets = overdose Severe liver centrilobular necrosis -also renal effects Can be fatal common means of suicide 14

Metabolism ~40% Glucuronidatoin Minor (CYP2E) ~60% Sulfation NABQI N-acetyl benzoquinone imine TOXIC Electrophile -forms adducts with proteins, Bioactivation of Acetaminophen Relationship between hepatic glutathione levels and covalent binding of acetaminophen to target nucleophiles (proteins) Toxicity to acetaminophen occurs when GSH is depleted by > 80% 15

Potentially Toxic Drugs / Intermediates are: Electrophiles Modeling and Prediction Software has helped identify ROS - generating candidate lead structures potentially toxic in parent for or likely to yield reactive intermediates Febrifugine Derived from the Chinese herb Chang Shan Used traditionally in the treatment of malaria Use associated with hepatotoxicity 16

Overview of Pharmacokinetics Rates of Absorption, Disposition, Metabolism, Elimination Drive Therapeutic Dosing 12 Plasma Concentration 10 8 6 4 2 TOXIC RANGE THERAPEUTIC RANGE SUB-THERAPEUTIC 0 0 1 2 3 4 5 6 7 8 9 Dose 17

Influence of Variations in Relative Rates of Absorption and Elimination on Plasma Concentration of an Orally Administered Drug Plasma concentration 14 12 10 8 6 4 2 0 Ka/Ke=10 Ka/Ke=1 Ka/Ke=0.1 Ka/Ke=0.01 0 5 10 15 20 TIME (hours) LOCUS OF ACTION RECEPTORS TISSUE RESERVOIRS Bound Free Free Bound ABSORPTION Free Drug EXCRETION Bound Drug SYSTEMIC CIRCULATION BIOTRANSFORMATION 18

Elimination Zero order: constant rate of elimination irrespective of plasma concentration. Rate of elimination = Amount constant First order: rate of elimination proportional to plasma concentration. Constant Fraction of drug eliminated per unit time. Rate of elimination = K x Amount 19

Plasma Concentration Profile after a Single I.V. Injection Plasma Concentration 10000 1000 100 10 1 Distribution and Elimination C 0 Elimination only Distribution equilibrium 0 1 2 3 4 5 6 Time lnc t = lnc o K el.t V d = Dose/C 0 t 1/2 = 0.693/K el When t = 0, C = C 0, i.e., the concentration at time zero when distribution is complete and elimination has not started yet. Use this value and the dose to calculate V d. 20

Pharmacokinetic parameters Get equation of regression line; from it get K el, C 0, and AUC Volume of distribution V d = DOSE / C 0 Plasma clearance Cl = K el.v d plasma half-life t1/2 = 0.693 / K el Bioavailability (AUC) x / (AUC) iv 21

The half-life of elimination of a drug (and its residence in the body) depends on its clearance and its volume of distribution Rate of elimination = K el x Amount in body Rate of elimination = CL x Plasma Concentration (CL in units vol/time) Therefore, K el x Amount = CL x Concentration K el = CL/V d 0.693/t1/2 = CL/V d t1/2 = 0.693 x V d /CL 22