You are on page 1of 37

Drug Metabolism for Master Program

Victor Shengkan Jin Associate Professor Pharmacology Department jinsh@umdnj.edu

The Journey of Drugs in Human Body


Drug administered Absorption distribution Drug in systemic circulation Drug in tissues of distribution

Drug at sites of action

Pharmacologic effect

Clinical Response

Toxicity

Efficacy

How are drugs eliminated from the body?


Major route I:
Excreted through kidney (renal excretion) unchanged. Renal excretion plays a pivotal role in terminating the biologic activity of some drugs, particularly those that have small molecular volumes or possess polar characteristics

HOWEVER:
Most drugs are relatively lipophilic, which facilitates passage through biological membranes as well as access to sites of action. Renal excretion of lipophilic compounds is poor due to reabsorption through the tubular membranes.

How are drugs eliminated from the body? Major Route II


Biotransformation of drugs or xenobiotics into more hydrophilic metabolites is critical for termination of biological activity and elimination from the body.

Drug metabolism:
Biotransformation of drugs to polar and hence more readily excretable products

Lecture Outline:

1. Drug metabolism enzymatic systems, and how drugs are metabolized by these systems; 2. Factors that can affect the drug metabolism systems and consequently affect the properties of drugs.

II. Drug Metabolism Enzymatic Systems

A. Metabolism of Xenobiotics
We humans are exposed to foreign compounds, or xenobiotics; Endogenous enzymatic systems for biotransformation and elimination of xenobiotics are critical for survival; The same xenobiotic-metabolizing system metabolizes prescribed drugs

B. Drug Metabolizing Enzymes Grouped into Phase I and II Enzymes


1. 2. Enzymes catalyze PHASE I or Functionalization Reactions Enzymes catalyze PHASE II or Conjugation (Biosynthetic) Reactions

Phase I reactions introduce or expose a polar functional group (e.g., OH, COOH, NH2, or SH) on the parent compound. Phase II reactions typically involve the conjugation of endogenous compounds (e.g., glucuronic acid or glutathione) to phase I products to yield highly polar conjugates.

Examples of Phase I Reactions


Aliphatic hydroxylation to alcohol
-minor metabolite of phenobarbital

Aromatic hydroxylation to phenol


-major metabolite of phenytoin, p-HPPH

Oxidation at S (on N)
- chlorpromazine to sulfoxide

Examples of Phase I and Phase II Reactions


Phase I Phase II

C. Sites of Drug Metabolism/Biotransformation (Tissue Level)

The liver is the principal organ of drug metabolic activity, an activity that is generally enzymatic in nature.

Other tissues also display metabolic activities, including the GI tract, lungs, skin, and the kidneys.

In the GI tract, drugs may be metabolized by microorganisms, digestive enzymes, and gastric acids.

C. Sites of Drug Metabolism/Biotransformation (Subcellular Level)


At the subcellular level most drug metabolizing activity occurs in the endoplasmic reticulum (ER) and the cytosol. Enzymes involved in Phase I reactions are typically located in the ER, while those involved in Phase II reactions are generally cytosolic. ER drug metabolizing enzymes are often called microsomal enzymes, as they are found in the microvesicles formed by ER membrane fragments (termed microsomes).

ER

Microsomes

IV. Cytochrome P450 Mixed Function Oxidase System


Major enzymatic system for Phase I drug metabolism reactions, metabolizes large number of environmental chemicals, food toxins, and drugs. Oxidative reactions require: Enzymes: cytochrome P450 and NADPH-cytochrome P450 reductase; Cofactor: NADPH (a reducing agent); and molecular oxygen (O2).

Reaction Cycle Involving Cytochrome P450 in Drug Oxidation


RH + O2 + 2H+ + 2e ROH + H2O Examples:

Activated oxygen: a very potent oxidizing reagent Promiscuous: Low substrate specificity, due to the large and fluid of substrate binding site

Schematic P450 Reaction


RH + O2 + 2H+ + 2e ROH + H2O

P450 active site

O
substrate

OCH 3

Substrate binding site

CYTOCHROME P450
SUPERGENE FAMILY - Human: 18 families, 43 subfamilies, 57 sequenced genes RECOMMENDED NOMENCLATURE BASED ON AA SEQUENCE SIMILARITY CYP for cytochrome P450 For distinct gene families: AA sequence less than 40%

identical, designated by Arabic numerals (e.g. CYP3)


For subfamilies: 40 to 70% AA sequence similarity,

designated by Capital letters (e.g., CYP3A)


For individual genes: designated by Arabic numerals (e.g.CYP3A4)

Human Liver P450 Enzymes


CYP3A4, 2C9, 1A2, 2E1, 2D6, and 2C19 appear to be the major forms of human liver P450s. These six enzymes are responsible for the bulk of the drug and xenobiotic metabolism in the liver. CYP3A4 accounts for more than 60% of the clinically prescribed drugs that undergo hepatic metabolism.

CYTOCHROME P450: GENERAL PROPERTIES


The CYPs have a tremendous capacity to metabolize a large number of structurally diverse chemicals Large family of CYP genes Single CYP metabolizes many structurally distinct chemicals (promiscuous) A single compound can be metabolized at different positions on the molecule A single molecule can be metabolized by different CYPs (overlapping substrate specificities)
INDUCIBLE BY A VARIETY OF CHEMICALS

These Properties are Part of the Molecular Basis of Drug-Drug Interactions

Phase II (Conjugation) Reactions


Conjugation involves specific transferases, which typically are located in the cytosol or ER. Transferases catalyze the coupling of an energy-rich (activated) endogenous substance with an exogenous (e.g., a drug) or other endogenous compound. Highly polar nature of most conjugates promotes their elimination in the urine or bile. Including glucuronidation, acetylation, glutathione conjugation, sulfate conjugation, methylation and water conjugation, etc.

Glucuronidation
Quantitatively, glucuronidation is the most important conjugation reaction. Versatile: O-, N-, S- glucuronidations

The Enzymes UDP-glucuronosyltransferases Functional Groups -OH, -COOH, -NH2, -SH Endogenous Reactant UDP- Glucuronic acid
Example:

Glucuronic acid

N-glucuronidation of 4- aminobiphenyl

Glutathione conjugation
Conjugation of reactive electrophilic compounds with the tripeptide glutathione is a major detoxification pathway for drugs and carcinogens.

The Enzymes Glutathione-S-transferase Functional Groups Epoxide, organic halides Endogenous Reactant Glutathione (Glu-Gly-Cys) The genes Two supergene family Cytosolic GSTs: 16 genes Membrane GSTs: 6 genes

Glutathione

Sulphation

The Enzymes Sulphotransferase Functional Groups -OH, -NH2, -SO2NH2 Endogenous Reactant PAPS (3-Phosphoadenosine5-phosphosulfate)

The proportion of drugs metabolized by the major phase II enzymes

UGT: UDP-glucoronosyl transferase; ST: sulfotransferase; GST: glutathione S-transferase; NAT, N-acetyltransferase. (Goodman & Gilman)

Drug Biodisposition Pathways


Biotransformation reactions often occur sequentially (i.e., Phase I Phase II). Some Examples of Drug Biodisposition Pathways

but with exceptions: Metabolism of Isoniazid (INH)


(phase II reactions precede phase I reactions)

The Importance of Drug Metabolism


Drug administered Absorption distribution Drug in systemic circulation Drug in tissues of distribution

Drug at sites of action

Pharmacologic effect

Clinical Response

Toxicity

Efficacy

The Importance of Drug Metabolism


Alter Pharmacodynamic Properties of Drugs
metabolic products are often inactive or less active than the parent drugs some metabolic products may have enhanced activity
e.g. inactive prodrugs may convert metabolically to active drugs

Alter Pharmacokinetic Properties of Drugs


often dramatically increase clearance and shorten half lives

e.g. lipophilic barbiturates such as thiopental and pentobarbital would have extremely long half-lives if it were not for their metabolic conversion to water soluble compounds
Affect Toxicity Properties of Drugs --Usually Detoxification --but some metabolic products have enhanced toxic properties (Acetaminophen )

The Importance of Drug Metabolism (contd)


Drug metabolism affects: drug efficacy and safety Factors related to drug metabolism play an important role in adverse drug response (ADR), which leads to a cost of $100 billion and over 100,000 death annually (Goodman and Gilmans, ed. 11, p88). Mandated by FDA, before a new drug application (NDA) is filed the route of metabolism and the enzymes involved in the metabolism must be known. the variables that are likely to significantly alter the metabolism of the drugs you prescribe, especially those have a narrow therapeutic index. You should be sure that the patients rate of metabolism is neither so slow that the drug reaches toxic concentrations nor so fast that the drug never achieves an effective concentration.

Factors Affecting Drug Metabolism


Genetic Factors
-genetic defects -genetic polymorphisms

Non-Genetic Factors:
Age and Sex -diseases

Environmental Determinants:
Dietary factors: cigarette smoking, alcohol consumption, etc. Environmental factors: pollutants, pesticides, etc. Drug-Drug interactions: inhibition or induction of drug
metabolism

Drugs and Endogenous Compounds interactions

Genetic Factors in Drug Metabolism: Personalized medicine? Genetic factors contribute to large interindividual differences in metabolic rate.
An Example: N-Acetylation of Isoniazid

Fast Acetylators

Slow Acetylators

Underlying Mechanism for Slower Acetylators: polymorphism of NAT2 (N-acetyltransferase 2) gene

patients with 2 alleles of slow NAT2 exhibit slower acetylator phenotype adverse response resembles a drug overdose thus determine the polymorphism of NAT2 gene is important reducing dosage or increasing dosing interval is recommended.
Personalized medicine

Dietary and Environmental Factors Induction (increasing the amount of enzymes):


Induce cytochrome P450 gene (making more of the enzyme), and increase metabolism rates of some drugs: -Charcoal-broiled food, -Cruciferous vegetables, induces CYP1A: e.g. warfarin -Cigarette smoking

Inhibition (inhibiting the activity of enzymes):


-Grapefruit juice (CYP3A4): e.g. cyclosporine -Alcohol

Drug-drug Interactions: Induction of Metabolizing Enzymes


examples:
CYP3A4: oral contraceptives and St. Johns wort. St. Johns Wort: a herbal medication which has effect on antidepression, strong inducer of CYP3A4 Reducing the concentration of oral contraceptives and other drugs coadministered.

Drug-drug Interactions: Inhibition of Metabolizing Enzymes

Inactivation of CYP enzymes by certain drugs Consequence: May impair elimination of the more slowly
metabolized drug, prolong or potentiate pharmacological effects, and increase incidence of drug-induced toxicity.

Diseases
Acute and chronic diseases that affect liver function or architecture markedly diminish the metabolism of some drugs. Alcoholic or Viral-Induced Hepatitis Alcoholic or Biliary Cirrhosis Cancer (e.g., Hepatocarcinomas)
t1/2 of chlordiazepoxide and diazepam Impaired oxidative metabolism of aminopyrine

Age and Gender


Very young and very old patients exhibit increased susceptibility to drug activities (both pharmacologic and toxic) compared with young adults. Phase I and II reactions occur in newborns, but generally at a slower rate than in adults. Age related decreases in liver mass, hepatic enzyme activity, and hepatic blood flow decrease the overall metabolic activity (particularly P450) of the elderly. Decreased oxidation of estrogens and benzodiazepines has been reported in females relative to males, but the generality of such genderspecific effects remains to be assessed

How are drugs eliminated from the body?

The Journey of Drugs in Human Body


Drug administered Absorption distribution Drug in systemic circulation Drug in tissues of distribution

Drug at sites of action

Pharmacologic effect

Clinical Response

Toxicity

Efficacy

You might also like